1
|
Watanabe K, Takayama S, Yamada T, Hashimoto M, Tadano J, Nakagawa T, Watanabe T, Fukusaki E, Miyawaki I, Shimma S. Novel mimetic tissue standards for precise quantitative mass spectrometry imaging of drug and neurotransmitter concentrations in rat brain tissues. Anal Bioanal Chem 2024; 416:5579-5593. [PMID: 39126505 PMCID: PMC11493812 DOI: 10.1007/s00216-024-05477-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/18/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Understanding the relationship between the concentration of a drug and its therapeutic efficacy or side effects is crucial in drug development, especially to understand therapeutic efficacy in central nervous system drug, quantifying drug-induced site-specific changes in the levels of endogenous metabolites, such as neurotransmitters. In recent times, evaluation of quantitative distribution of drugs and endogenous metabolites using matrix-assisted laser desorption/ionization (MALDI)-mass spectrometry imaging (MSI) has attracted much attention in drug discovery research. However, MALDI-MSI quantification (quantitative mass spectrometry imaging, QMSI) is an emerging technique, and needs to be further developed for practicable and convenient use in drug discovery research. In this study, we developed a reliable QMSI method for quantification of clozapine (antipsychotic drug) and dopamine and its metabolites in the rat brain using MALDI-MSI. An improved mimetic tissue model using powdered frozen tissue for QMSI was established as an alternative method, enabling the accurate quantification of clozapine levels in the rat brain. Furthermore, we used the improved method to evaluate drug-induced fluctuations in the concentrations of dopamine and its metabolites. This method can quantitatively evaluate drug localization in the brain and drug-induced changes in the concentration of endogenous metabolites, demonstrating the usefulness of QMSI.
Collapse
Affiliation(s)
- Kenichi Watanabe
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Sayo Takayama
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Toichiro Yamada
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Masayo Hashimoto
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Jun Tadano
- Research Planning & Coordination, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Tetsuya Nakagawa
- Research Planning & Coordination, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Takao Watanabe
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Eiichiro Fukusaki
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Osaka University Shimadzu Omics Innovation Research Laboratory, Osaka University, Osaka, Japan
| | - Izuru Miyawaki
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Shuichi Shimma
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
- Osaka University Shimadzu Omics Innovation Research Laboratory, Osaka University, Osaka, Japan.
| |
Collapse
|
2
|
Moore AM, Bowman A, Wali SN, Weigand MR, Wagner D, Yang J, Laskin J. Quantitative Analysis of Drugs in a Mimetic Tissue Model Using Nano-DESI on a Triple Quadrupole Mass Spectrometer. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024. [PMID: 39485739 DOI: 10.1021/jasms.4c00345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mass spectrometry is a powerful analytical technique used at every stage of the pharmaceutical research process. A specialized branch of this method, mass spectrometry imaging (MSI), has emerged as an important tool for determining the spatial distribution of drugs in biological samples. Despite the importance of MSI, its quantitative capabilities are still limited due to the complexity of biological samples and the lack of separation prior to analysis. This makes the simultaneous quantification and visualization of analytes challenging. Several techniques have been developed to address this challenge and enable quantitative MSI. One such approach is the mimetic tissue model, which involves the incorporation of an analyte of interest into tissue homogenates at several concentrations. A calibration curve that accounts for signal suppression by the complex biological matrix is then created by measuring the signal of the analyte in the series of tissue homogenates. Herein, we use the mimetic tissue model on a triple quadrupole mass spectrometer (QqQ) in multiple reaction monitoring mode to demonstrate the quantitative abilities of nanospray desorption electrospray ionization (nano-DESI) and compare these results with those obtained using atmospheric pressure matrix-assisted laser desorption/ionization (AP-MALDI). For the tested compounds, our findings indicate that nano-DESI achieves lower standard deviations than AP-MALDI, resulting in superior limits of detection for the studied analytes. Additionally, we discuss the limitations of the mimetic tissue model in the quantification of certain analytes and the challenges involved with the implementation of the model.
Collapse
Affiliation(s)
- Alyssa M Moore
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Andrew Bowman
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Syeda Nazifa Wali
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Miranda R Weigand
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - David Wagner
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Junhai Yang
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Julia Laskin
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
3
|
Xie F, Gales T, Ringenberg MA, Wolf AI, Groseclose MR. Characterizing the Distribution of a Stimulator of Interferon Genes Agonist and Its Metabolites in Mouse Liver by Matrix-Assisted Laser Desorption/Ionization Imaging Mass Spectrometry. Drug Metab Dispos 2024; 52:1181-1186. [PMID: 37884391 DOI: 10.1124/dmd.122.001076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
A STING (stimulator of interferon genes) agonist GSK3996915 under investigation in early discovery for hepatitis B was orally dosed to a mouse model for understanding the parent drug distribution in liver, the target organ. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was used to quantify the distribution of GSK3996915 in liver collected from mice administered a single oral dose at 90 mg/kg. GSK3996915 was detected with a zonal distribution localized in the portal triad and highly concentrated in the main bile ducts, indicating clearance through biliary excretion. High spatial resolution imaging showed the distribution of the parent drug localized to the cellular populations in the sinusoids, including the Kupffer cells. Additionally, a series of drug-related metabolites were observed to be localized in the central zones of the liver. These results exemplify the potential of utilizing MALDI IMS for measuring not only quantitative drug distribution and target exposure but also drug metabolism and elimination in a single suite of experiments. SIGNIFICANCE STATEMENT: An integrated imaging approach utilizing matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) complemented with immunohistochemistry (IHC) and histology was used to address the question of target exposure at the cellular level. Localized quantification of the parent drug in the target organ and identification of potential metabolites in the context of tissue histology were also achieved in one experimental suite to support characterization of pharmacokinetic properties of the drug in the early discovery stage.
Collapse
Affiliation(s)
- Fang Xie
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Tracy Gales
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M A Ringenberg
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Amaya I Wolf
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M Reid Groseclose
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| |
Collapse
|
4
|
McDonald H, Li Q, Ashaduzzaman M, Zhao C, Pan S, Szulczewski GJ, Liang Q. Quantitative MALDI-MS and Imaging of Fungicide Pyrimethanil in Strawberries with 2-Nitrophloroglucinol as an Effective Matrix. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1272-1281. [PMID: 38687954 DOI: 10.1021/jasms.4c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
This work explores the use of 2-nitrophloroglucinol (2-NPG) as a matrix for quantitative analysis of the fungicide Pyrimethanil (PYM) in strawberries using matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) and imaging. 2-NPG was selected for PYM analysis for optimum sensitivity and precision compared to common matrices α-cyano-4-hydroxylcinnamic acid (CHCA) and 2,5-dihydroxybenzoic acid (DHB). PYM-sprayed strawberries were frozen 0, 1, 3, and 4 days after treatment and sectioned for MALDI imaging. The remaining part of each strawberry was processed using quick easy cheap effective rugged and safe (QuEChERS) extraction and analyzed by MALDI-MS and ultraperformance liquid chromatography multireaction-monitoring (UPLC-MRM). MALDI-MS showed comparable performance to UPLC-MRM in calibration, LOD/LOQ, matrix effect, and recovery, with the benefit of fast analysis. The MALDI imaging results demonstrated that PYM progressively penetrated the interior of the strawberry over time and the PYM concentration on tissue measured by MALDI imaging correlated linearly with MALDI-MS and UPLC-MRM measurements and accounts for 79% MALDI-MS and 85% UPLC-MRM values on average. Additionally, quartz crystal microbalance (QCM) was introduced as a new approach to determine strawberry tissue mass per area for MALDI imaging absolute quantitation with sensitive, direct, and localized measurements. This work demonstrates the first example of absolute quantitative MALDI imaging of pesticides in a heterogeneous plant tissue. The novel use of the 2-NPG matrix in quantitative MALDI-MS and imaging could be applied to other analytes, and the new QCM tissue mass per area method is potentially useful for quantitative MALDI imaging of heterogeneous tissues in general.
Collapse
Affiliation(s)
- Heather McDonald
- Department of Physical Sciences, University of West Alabama, Livingston, Alabama 35470, United States
| | - Qi Li
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Md Ashaduzzaman
- Department of Chemistry & Biochemistry, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Chao Zhao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Shanlin Pan
- Department of Chemistry & Biochemistry, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Greg J Szulczewski
- Department of Chemistry & Biochemistry, University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Qiaoli Liang
- Department of Chemistry & Biochemistry, University of Alabama, Tuscaloosa, Alabama 35487, United States
| |
Collapse
|
5
|
Tressler CM, Wadsworth B, Carriero S, Dillman N, Crawford R, Hahm TH, Glunde K, Cadieux CL. Characterization of Humanized Mouse Model of Organophosphate Poisoning and Detection of Countermeasures via MALDI-MSI. Int J Mol Sci 2024; 25:5624. [PMID: 38891812 PMCID: PMC11172367 DOI: 10.3390/ijms25115624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Organophosphoate (OP) chemicals are known to inhibit the enzyme acetylcholinesterase (AChE). Studying OP poisoning is difficult because common small animal research models have serum carboxylesterase, which contributes to animals' resistance to OP poisoning. Historically, guinea pigs have been used for this research; however, a novel genetically modified mouse strain (KIKO) was developed with nonfunctional serum carboxylase (Es1 KO) and an altered acetylcholinesterase (AChE) gene, which expresses the amino acid sequence of the human form of the same protein (AChE KI). KIKO mice were injected with 1xLD50 of an OP nerve agent or vehicle control with or without atropine. After one to three minutes, animals were injected with 35 mg/kg of the currently fielded Reactivator countermeasure for OP poisoning. Postmortem brains were imaged on a Bruker RapifleX ToF/ToF instrument. Data confirmed the presence of increased acetylcholine in OP-exposed animals, regardless of treatment or atropine status. More interestingly, we detected a small amount of Reactivator within the brain of both exposed and unexposed animals; it is currently debated if reactivators can cross the blood-brain barrier. Further, we were able to simultaneously image acetylcholine, the primary affected neurotransmitter, as well as determine the location of both Reactivator and acetylcholine in the brain. This study, which utilized sensitive MALDI-MSI methods, characterized KIKO mice as a functional model for OP countermeasure development.
Collapse
Affiliation(s)
- Caitlin M. Tressler
- The Johns Hopkins University Applied Imaging Mass Spectrometry Core and Service Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin Wadsworth
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD 21010, USA
| | - Samantha Carriero
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD 21010, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Natalie Dillman
- The Johns Hopkins University Applied Imaging Mass Spectrometry Core and Service Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel Crawford
- The Johns Hopkins University Applied Imaging Mass Spectrometry Core and Service Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tae-Hun Hahm
- The Johns Hopkins University Applied Imaging Mass Spectrometry Core and Service Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kristine Glunde
- The Johns Hopkins University Applied Imaging Mass Spectrometry Core and Service Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - C. Linn Cadieux
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Gunpowder, MD 21010, USA
| |
Collapse
|
6
|
Sorokin AA, Pekov SI, Zavorotnyuk DS, Shamraeva MM, Bormotov DS, Popov IA. Modern machine-learning applications in ambient ionization mass spectrometry. MASS SPECTROMETRY REVIEWS 2024. [PMID: 38671553 DOI: 10.1002/mas.21886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024]
Abstract
This article provides a comprehensive overview of the applications of methods of machine learning (ML) and artificial intelligence (AI) in ambient ionization mass spectrometry (AIMS). AIMS has emerged as a powerful analytical tool in recent years, allowing for rapid and sensitive analysis of various samples without the need for extensive sample preparation. The integration of ML/AI algorithms with AIMS has further expanded its capabilities, enabling enhanced data analysis. This review discusses ML/AI algorithms applicable to the AIMS data and highlights the key advancements and potential benefits of utilizing ML/AI in the field of mass spectrometry, with a focus on the AIMS community.
Collapse
Affiliation(s)
- Anatoly A Sorokin
- Laboratory of Molecular Medical Diagnostics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Stanislav I Pekov
- Mass Spectrometry Laboratory, Skolkovo Institute of Science and Technology, Moscow, Russia
- Translational Medicine Laboratory, Siberian State Medical University, Tomsk, Russia
- Department for Molecular and Biological Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Denis S Zavorotnyuk
- Laboratory of Molecular Medical Diagnostics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Mariya M Shamraeva
- Laboratory of Molecular Medical Diagnostics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Denis S Bormotov
- Laboratory of Molecular Medical Diagnostics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Igor A Popov
- Laboratory of Molecular Medical Diagnostics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Translational Medicine Laboratory, Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
7
|
Paulson AE, Larson EA, Lee YJ. Mobilized Electrospray Device for On-Tissue Chemical Derivatization in MALDI-MS Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:205-213. [PMID: 38147682 DOI: 10.1021/jasms.3c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Applying solutions of a matrix or derivatization agent via microdroplets is a common sample preparation technique for matrix-assisted laser desorption/ionization-mass spectrometry imaging (MALDI-MSI) experiments. Mobilized nebulizer sprayers are commonly used to create a homogeneous matrix or reagent layer across large surfaces. Electrospray devices have also been used to produce microdroplets for the same purpose but are rarely used for large tissues due to their immobility. Herein, we present a movable electrospray device that can be used for large tissue sample preparation through a simple modification to an automatic commercial nebulizer device. As demonstrated for on-tissue chemical derivatization (OTCD) with Girard's reagent T using a mimetic tissue model, the sprayer has the additional benefit of being able to investigate reaction acceleration in OTCD when comparing electrostatically charged spray to electrostatically neutral spray. Finally, MALDI-MSI of fatty aldehydes is successfully demonstrated in rat brain tissues using this device for both OTCD and matrix application.
Collapse
Affiliation(s)
- Andrew E Paulson
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | - Evan A Larson
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| | - Young Jin Lee
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
8
|
Rajbhandari P, Neelakantan TV, Hosny N, Stockwell BR. Spatial pharmacology using mass spectrometry imaging. Trends Pharmacol Sci 2024; 45:67-80. [PMID: 38103980 PMCID: PMC10842749 DOI: 10.1016/j.tips.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/07/2023] [Accepted: 11/11/2023] [Indexed: 12/19/2023]
Abstract
The emerging and powerful field of spatial pharmacology can map the spatial distribution of drugs and their metabolites, as well as their effects on endogenous biomolecules including metabolites, lipids, proteins, peptides, and glycans, without the need for labeling. This is enabled by mass spectrometry imaging (MSI) that provides previously inaccessible information in diverse phases of drug discovery and development. We provide a perspective on how MSI technologies and computational tools can be implemented to reveal quantitative spatial drug pharmacokinetics and toxicology, tissue subtyping, and associated biomarkers. We also highlight the emerging potential of comprehensive spatial pharmacology through integration of multimodal MSI data with other spatial technologies. Finally, we describe how to overcome challenges including improving reproducibility and compound annotation to generate robust conclusions that will improve drug discovery and development processes.
Collapse
Affiliation(s)
- Presha Rajbhandari
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | | | - Noreen Hosny
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA; Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Yang E, Kim JH, Tressler CM, Shen XE, Brown DR, Johnson CC, Hahm TH, Barman I, Glunde K. RaMALDI: Enabling simultaneous Raman and MALDI imaging of the same tissue section. Biosens Bioelectron 2023; 239:115597. [PMID: 37597501 PMCID: PMC10544780 DOI: 10.1016/j.bios.2023.115597] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Multimodal tissue imaging techniques that integrate two complementary modalities are powerful discovery tools for unraveling biological processes and identifying biomarkers of disease. Combining Raman spectroscopic imaging (RSI) and matrix-assisted laser-desorption/ionization (MALDI) mass spectrometry imaging (MSI) to obtain fused images with the advantages of both modalities has the potential of providing spatially resolved, sensitive, specific biomolecular information, but has so far involved two separate sample preparations, or even consecutive tissue sections for RSI and MALDI MSI, resulting in images with inherent disparities. We have developed RaMALDI, a streamlined, integrated, multimodal imaging workflow of RSI and MALDI MSI, performed on a single tissue section with one sample preparation protocol. We show that RaMALDI imaging of various tissues effectively integrates molecular information acquired from both RSI and MALDI MSI of the same sample, which will drive discoveries in cell biology, biomedicine, and pathology, and advance tissue diagnostics.
Collapse
Affiliation(s)
- Ethan Yang
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jeong Hee Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Caitlin M Tressler
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Xinyi Elaine Shen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Dalton R Brown
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Cole C Johnson
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Tae-Hun Hahm
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ishan Barman
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Kristine Glunde
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Departments of Oncology and Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
10
|
Miller A, York EM, Stopka SA, Martínez-François JR, Hossain MA, Baquer G, Regan MS, Agar NYR, Yellen G. Spatially resolved metabolomics and isotope tracing reveal dynamic metabolic responses of dentate granule neurons with acute stimulation. Nat Metab 2023; 5:1820-1835. [PMID: 37798473 PMCID: PMC10626993 DOI: 10.1038/s42255-023-00890-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/09/2023] [Indexed: 10/07/2023]
Abstract
Neuronal activity creates an intense energy demand that must be met by rapid metabolic responses. To investigate metabolic adaptations in the neuron-enriched dentate granule cell (DGC) layer within its native tissue environment, we employed murine acute hippocampal brain slices, coupled with fast metabolite preservation and followed by mass spectrometry (MS) imaging, to generate spatially resolved metabolomics and isotope-tracing data. Here we show that membrane depolarization induces broad metabolic changes, including increased glycolytic activity in DGCs. Increased glucose metabolism in response to stimulation is accompanied by mobilization of endogenous inosine into pentose phosphates via the action of purine nucleotide phosphorylase (PNP). The PNP reaction is an integral part of the neuronal response to stimulation, because inhibition of PNP leaves DGCs energetically impaired during recovery from strong activation. Performing MS imaging on brain slices bridges the gap between live-cell physiology and the deep chemical analysis enabled by MS.
Collapse
Affiliation(s)
- Anne Miller
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Elisa M York
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Md Amin Hossain
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Stopka SA, Ruiz D, Baquer G, Bodineau C, Hossain MA, Pellens VT, Regan MS, Pourquié O, Haigis MC, Bi WL, Coy SM, Santagata S, Agar NYR, Basu SS. Chemical QuantArray: A Quantitative Tool for Mass Spectrometry Imaging. Anal Chem 2023; 95:11243-11253. [PMID: 37469028 PMCID: PMC10445330 DOI: 10.1021/acs.analchem.3c00803] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) is a powerful analytical technique that provides spatially preserved detection and quantification of analytes in tissue specimens. However, clinical translation still requires improved throughput, precision, and accuracy. To accomplish this, we created "Chemical QuantArray", a gelatin tissue microarray (TMA) mold filled with serial dilutions of isotopically labeled endogenous metabolite standards. The mold is then cryo-sectioned onto a tissue homogenate to produce calibration curves. To improve precision and accuracy, we automatically remove pixels outside of each TMA well and investigated several intensity normalizations, including the utilization of a second stable isotope internal standard (IS). Chemical QuantArray enables the quantification of several endogenous metabolites over a wide dynamic range and significantly improve over current approaches. The technique reduces the space needed on the MALDI slides for calibration standards by approximately 80%. Furthermore, removal of empty pixels and normalization to an internal standard or matrix peak provided precision (<20% RSD) and accuracy (<20% DEV). Finally, we demonstrate the applicability of Chemical QuantArray by quantifying multiple purine metabolites in 14 clinical tumor specimens using a single MALDI slide. Chemical QuantArray improves the analytical characteristics and practical feasibility of MALDI-MSI metabolite quantification in clinical and translational applications.
Collapse
Affiliation(s)
- Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Daniela Ruiz
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Clément Bodineau
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Md Amin Hossain
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Valentina T Pellens
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Ludwig Center, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Wenya L Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shannon M Coy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, Massachusetts 02115, United States
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, United States
| | - Sankha S Basu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
12
|
Nagano E, Odake K, Shimma S. An Alternative Method for Quantitative Mass Spectrometry Imaging ( q-MSI) of Dopamine Utilizing Fragments of Animal Tissue. Mass Spectrom (Tokyo) 2023; 12:A0128. [PMID: 37538447 PMCID: PMC10394126 DOI: 10.5702/massspectrometry.a0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023] Open
Abstract
Mass spectrometry imaging (MSI) is a well-known method for the ionization of molecules on tissue sections and the visualization of their localization. Recently, different sample preparation methods and new instruments have been used for MSI, and different molecules are becoming visible. On the other hand, although several quantification methods (q-MSI) have been proposed, there is still room for the development of a simplified procedure. Here, we have attempted to develop a reproducible and reliable quantification method using a calibration curve prepared from tissue debris of a frozen section of a sample when we trim the frozen blocks. We discuss the reproducibility of this method across different sample lots and the effect of the biological matrix (ion suppression) on our results. The quantitative performance was evaluated in terms of accuracy and relative standard deviation, and the reliability of the quantitative values obtained by matrix-assisted laser desorption/ionization-MSI was further evaluated by enzyme-linked immunosorbent assay (ELISA). Our q-MSI method for the quantification of dopamine in mouse brain tissue was found to be highly linear, accurate, and precise. The quantitative values obtained by MSI were found to be highly comparable (>85% similarity) to the results obtained by ELISA from the same tissue extracts.
Collapse
Affiliation(s)
- Erika Nagano
- Research and Development Division, Miruion Corporation, Ibaraki, Osaka 567–0085, Japan
| | - Kazuki Odake
- Research and Development Division, Miruion Corporation, Ibaraki, Osaka 567–0085, Japan
| | - Shuichi Shimma
- Research and Development Division, Miruion Corporation, Ibaraki, Osaka 567–0085, Japan
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka 565–0871, Japan
| |
Collapse
|
13
|
Miller A, York E, Stopka S, Martínez-François J, Hossain MA, Baquer G, Regan M, Agar N, Yellen G. Spatially resolved metabolomics and isotope tracing reveal dynamic metabolic responses of dentate granule neurons with acute stimulation. RESEARCH SQUARE 2023:rs.3.rs-2276903. [PMID: 37546759 PMCID: PMC10402263 DOI: 10.21203/rs.3.rs-2276903/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Neuronal activity creates an intense energy demand that must be met by rapid metabolic responses. To investigate metabolic adaptations in the neuron-enriched dentate granule cell (DGC) layer within its native tissue environment, we employed murine acute hippocampal brain slices coupled with fast metabolite preservation, followed by mass spectrometry imaging (MALDI-MSI) to generate spatially resolved metabolomics and isotope tracing data. Here we show that membrane depolarization induces broad metabolic changes, including increased glycolytic activity in DGCs. Increased glucose metabolism in response to stimulation is accompanied by mobilization of endogenous inosine into pentose phosphates, via the action of purine nucleotide phosphorylase (PNP). The PNP reaction is an integral part of the neuronal response to stimulation, as inhibiting PNP leaves DGCs energetically impaired during recovery from strong activation. Performing MSI on brain slices bridges the gap between live cell physiology and the deep chemical analysis enabled by mass spectrometry.
Collapse
|
14
|
Umar U, Ahmed S, Iftikhar A, Iftikhar M, Majeed W, Liaqat A, Shahzad S, Abbas M, Mehmood T, Anwar F. Phenolics Extracted from Jasminum sambac Mitigates Diabetic Cardiomyopathy by Modulating Oxidative Stress, Apoptotic Mediators and the Nfr-2/HO-1 Pathway in Alloxan-Induced Diabetic Rats. Molecules 2023; 28:5453. [PMID: 37513325 PMCID: PMC10383516 DOI: 10.3390/molecules28145453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder defined as hyperglycemia and pancreatic β-cell deterioration, leading to other complications such as cardiomyopathy. The current study assessed the therapeutic effects of phenolic acids extracted from Jasminum sambac phenols of leaves (JSP) against diabetes-induced cardiomyopathy in rats. The rats were divided into four groups, with each group consisting of 20 rats. The rats were given intraperitoneal injections of alloxan monohydrate (150 mg/kg) to induce diabetes. The diabetes-induced groups (III and IV) received treatment for six weeks that included 250 and 500 mg/kg of JSP extract, respectively. In the treated rats, the results demonstrated that JSP extract restored fasting glucose, serum glucose, and hyperlipidemia. Alloxan induced cardiomyopathy, promoted oxidative stress, and altered cardiac function biomarkers, including cardiac troponin I, proBNP, CK-MB, LDH, and IMA. The JSP extract-treated rats showed improved cardiac function indicators, apoptosis, and oxidative stress. In diabetic rats, the mRNA expression of caspase-3, BAX, and Bcl-2 was significantly higher, while Bcl-2, Nrf-2, and HO-,1 was significantly lower. In the treated groups, the expression levels of the BAX, Nrf-2, HO-1, Caspase-3, and Bcl-2 genes were dramatically returned to normal level. According to our findings, the JSP extract prevented cardiomyopathy and heart failure in the hyperglycemic rats by improving cardiac biomarkers and lowering the levels of hyperlipidemia, oxidative stress, apoptosis, hyperglycemia, and hyperlipidemia.
Collapse
Affiliation(s)
- Urooj Umar
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Faisalabad, Faisalabad 38000, Pakistan
| | - Sibtain Ahmed
- Department of Biochemistry, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Asra Iftikhar
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Faisalabad, Faisalabad 38000, Pakistan
| | - Maryam Iftikhar
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Wafa Majeed
- Department of Pharmacy, University of Agriculture, Faisalabad 38000, Pakistan
| | - Atika Liaqat
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Faisalabad, Faisalabad 38000, Pakistan
| | - Sana Shahzad
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Faisalabad, Faisalabad 38000, Pakistan
| | - Mateen Abbas
- Quality Operations Laboratory, Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Tahir Mehmood
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore 53700, Pakistan
| | - Farooq Anwar
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Institute of Chemistry, University of Sargodha, Sargodha 40100, Pakistan
| |
Collapse
|
15
|
Park YM, Meyer MR, Müller R, Herrmann J. Optimization of Mass Spectrometry Imaging for Drug Metabolism and Distribution Studies in the Zebrafish Larvae Model: A Case Study with the Opioid Antagonist Naloxone. Int J Mol Sci 2023; 24:10076. [PMID: 37373226 DOI: 10.3390/ijms241210076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Zebrafish (ZF; Danio rerio) larvae have emerged as a promising in vivo model in drug metabolism studies. Here, we set out to ready this model for integrated mass spectrometry imaging (MSI) to comprehensively study the spatial distribution of drugs and their metabolites inside ZF larvae. In our pilot study with the overall goal to improve MSI protocols for ZF larvae, we investigated the metabolism of the opioid antagonist naloxone. We confirmed that the metabolic modification of naloxone is in high accordance with metabolites detected in HepaRG cells, human biosamples, and other in vivo models. In particular, all three major human metabolites were detected at high abundance in the ZF larvae model. Next, the in vivo distribution of naloxone was investigated in three body sections of ZF larvae using LC-HRMS/MS showing that the opioid antagonist is mainly present in the head and body sections, as suspected from published human pharmacological data. Having optimized sample preparation procedures for MSI (i.e., embedding layer composition, cryosectioning, and matrix composition and spraying), we were able to record MS images of naloxone and its metabolites in ZF larvae, providing highly informative distributional images. In conclusion, we demonstrate that all major ADMET (absorption, distribution, metabolism, excretion, and toxicity) parameters, as part of in vivo pharmacokinetic studies, can be assessed in a simple and cost-effective ZF larvae model. Our established protocols for ZF larvae using naloxone are broadly applicable, particularly for MSI sample preparation, to various types of compounds, and they will help to predict and understand human metabolism and pharmacokinetics.
Collapse
Affiliation(s)
- Yu Mi Park
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Markus R Meyer
- Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Experimental and Clinical Toxicology, Saarland University, 66421 Homburg, Germany
| | - Rolf Müller
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
| |
Collapse
|
16
|
Woodhouse N, Majer J, Marshall P, Hood S, Notingher I. Quantification of Drugs in Brain and Liver Mimetic Tissue Models Using Raman Spectroscopy. APPLIED SPECTROSCOPY 2023; 77:246-260. [PMID: 36320126 PMCID: PMC10034474 DOI: 10.1177/00037028221139494] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Quantitative analysis of drug delivery with in biological systems is an integral challenge in drug development. Analytical techniques are important for assessing both drug target delivery, target action, and drug toxicology. Using mimetic tissue models, we have investigated the efficacy of Raman spectroscopy in quantitative detection of alkyne group and deuterated drugs in rat brain and rat liver tissue models. Lasers with 671 nm and 785 nm wavelengths were assessed for their feasibility in this application due to opposing relative benefits and disadvantages. Thin tissue sections have been tested as a practical means of reducing autofluorescent background by minimizing out-of-focus tissue and therefore maximizing photobleaching rates. Alkyne-tagged drugs were quantitatively measured at 18 ± 5 μg/g drug/tissue mass ratio in rat brain and at 34 ± 6 μg/g in rat liver. Quantification calibration curves were generated for a range of concentrations from 0-500 μg/g. These results show the potential of Raman spectroscopy as a diffraction-limited spatially resolved imaging technique for assessing drug delivery in tissue applications.
Collapse
Affiliation(s)
- Nathan Woodhouse
- School of Physics and Astronomy,
University
of Nottingham, Nottingham, UK
| | | | | | | | - Ioan Notingher
- School of Physics and Astronomy,
University
of Nottingham, Nottingham, UK
| |
Collapse
|
17
|
Quantitative Mass Spectrometry Imaging of Bleomycin in Skin Using a Mimetic Tissue Model for Calibration. Pharmaceuticals (Basel) 2022; 15:ph15121583. [PMID: 36559034 PMCID: PMC9786816 DOI: 10.3390/ph15121583] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The aim of Quantitative mass spectrometry imaging (Q-MSI) is to provide distribution analysis and quantitation from one single mass-spectrometry-based experiment, and several quantitation methods have been devised for Q-MSI. Mimetic tissue models based on spiked tissue homogenates are considered one of the most accurate ways to perform Q-MSI, since the analyte is present in a well-defined concentration in a sample matrix highly similar to the one of the unknown sample to be analyzed. The delivery of drugs in skin is among the most frequent types of pharmaceutical MSI studies. Here, a mimetic tissue model is extended for use on the skin, which, due to its high collagen content, is different from most other tissue as the homogenates become extremely viscous. A protocol is presented which overcomes this by the addition of water and the handling of the homogenate at an elevated temperature where the viscosity is lower. Using a mimetic tissue model, a method was developed for the quantitative imaging of bleomycin in skin. To compensate for the signal drift and the inhomogeneities in the skin, an internal standard was included in the method. The method was tested on skin from a pig which had had an electropneumatic injection of bleomycin into the skin. Quantification was made at several regions in a cross section of the skin at the injection site, and the results were compared to the results of a quantitative LC-MS on a neighboring tissue biopsy from the same animal experiment. The overall tissue concentration determined by the LC-MS was within the range of the different regions quantified by the Q-MSI. As the model provides the results of the same order of magnitude as a LC-MS, it can either be used to replace LC-MS in skin studies where MSI and LC-MS are today carried out in combination, or it can add quantitative information to skin studies which are otherwise carried out by MSI alone.
Collapse
|
18
|
Multimodal imaging distribution assessment of a liposomal antibiotic in an infectious disease model. J Control Release 2022; 352:199-210. [PMID: 36084816 DOI: 10.1016/j.jconrel.2022.08.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/08/2022]
Abstract
Liposomes are promising targeted drug delivery systems with the potential to improve the efficacy and safety profile of certain classes of drugs. Though attractive, there are unique analytical challenges associated with the development of liposomal drugs including human dose prediction given these are multi-component drug delivery systems. In this study, we developed a multimodal imaging approach to provide a comprehensive distribution assessment for an antibacterial drug, GSK2485680, delivered as a liposomal formulation (Lipo680) in a mouse thigh model of bacterial infection to support human dose prediction. Positron emission tomography (PET) imaging was used to track the in vivo biodistribution of Lipo680 over 48 h post-injection providing a clear assessment of the uptake in various tissues and, importantly, the selective accumulation at the site of infection. In addition, a pharmacokinetic model was created to evaluate the kinetics of Lipo680 in different tissues. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was then used to quantify the distribution of GSK2485680 and to qualitatively assess the distribution of a liposomal lipid throughout sections of infected and non-infected hindlimb tissues at high spatial resolution. Through the combination of both PET and MALDI IMS, we observed excellent correlation between the Lipo680-radionuclide signal detected by PET with the GSK2485680 and lipid component signals detected by MALDI IMS. This multimodal translational method can reduce drug attrition by generating comprehensive biodistribution profiles of drug delivery systems to provide mechanistic insight and elucidate safety concerns. Liposomal formulations have potential to deliver therapeutics across a broad array of different indications, and this work serves as a template to aid in delivering future liposomal drugs to the clinic.
Collapse
|
19
|
Hu H, Laskin J. Emerging Computational Methods in Mass Spectrometry Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203339. [PMID: 36253139 PMCID: PMC9731724 DOI: 10.1002/advs.202203339] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/17/2022] [Indexed: 05/10/2023]
Abstract
Mass spectrometry imaging (MSI) is a powerful analytical technique that generates maps of hundreds of molecules in biological samples with high sensitivity and molecular specificity. Advanced MSI platforms with capability of high-spatial resolution and high-throughput acquisition generate vast amount of data, which necessitates the development of computational tools for MSI data analysis. In addition, computation-driven MSI experiments have recently emerged as enabling technologies for further improving the MSI capabilities with little or no hardware modification. This review provides a critical summary of computational methods and resources developed for MSI data analysis and interpretation along with computational approaches for improving throughput and molecular coverage in MSI experiments. This review is focused on the recently developed artificial intelligence methods and provides an outlook for a future paradigm shift in MSI with transformative computational methods.
Collapse
Affiliation(s)
- Hang Hu
- Department of ChemistryPurdue University560 Oval DriveWest LafayetteIN47907USA
| | - Julia Laskin
- Department of ChemistryPurdue University560 Oval DriveWest LafayetteIN47907USA
| |
Collapse
|
20
|
Akakpo JY, Jaeschke MW, Etemadi Y, Artigues A, Toerber S, Olivos H, Shrestha B, Midey A, Jaeschke H, Ramachandran A. Desorption Electrospray Ionization Mass Spectrometry Imaging Allows Spatial Localization of Changes in Acetaminophen Metabolism in the Liver after Intervention with 4-Methylpyrazole. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:2094-2107. [PMID: 36223142 PMCID: PMC9901546 DOI: 10.1021/jasms.2c00202] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Acetaminophen (APAP) overdose is the most common cause of acute liver failure in the US, and hepatotoxicity is initiated by a reactive metabolite which induces characteristic centrilobular necrosis. The only clinically available antidote is N-acetylcysteine, which has limited efficacy, and we have identified 4-methylpyrazole (4MP, Fomepizole) as a strong alternate therapeutic option, protecting against generation and downstream effects of the cytotoxic reactive metabolite in the clinically relevant C57BL/6J mouse model and in humans. However, despite the regionally restricted necrosis after APAP, our earlier studies on APAP metabolites in biofluids or whole tissue homogenate lack the spatial information needed to understand region-specific consequences of reactive metabolite formation after APAP overdose. Thus, to gain insight into the regional variation in APAP metabolism and study the influence of 4MP, we established a desorption electrospray ionization mass spectrometry imaging (DESI-MSI) platform for generation of ion images for APAP and its metabolites under ambient air, without chemical labeling or a prior coating of tissue which reduces chemical interference and perturbation of small molecule tissue localization. The spatial intensity and distribution of both oxidative and nonoxidative APAP metabolites were determined from mouse liver sections after a range of APAP overdoses. Importantly, exclusive differential signal intensities in metabolite abundance were noted in the tissue microenvironment, and 4MP treatment substantially influenced this topographical distribution.
Collapse
Affiliation(s)
- Jephte Yao Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Matthew Wolfgang Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Yasaman Etemadi
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Antonio Artigues
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | | | | | | | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
21
|
Devanathan AS, White NR, Desyaterik Y, De la Cruz G, Nekorchuk M, Terry M, Busman-Sahay K, Adamson L, Luciw P, Fedoriw Y, Estes JD, Rosen EP, Kashuba ADM. Quantitative Imaging Analysis of the Spatial Relationship between Antiretrovirals, Reverse Transcriptase Simian-Human Immunodeficiency Virus RNA, and Fibrosis in the Spleens of Nonhuman Primates. Antimicrob Agents Chemother 2022; 66:e0060922. [PMID: 35856680 PMCID: PMC9380553 DOI: 10.1128/aac.00609-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/07/2022] [Indexed: 01/22/2023] Open
Abstract
Although current antiretroviral therapy (ART) has increased life expectancy, a cure for human immunodeficiency virus (HIV) remains elusive due to the persistence of the virus in tissue reservoirs. In the present study, we sought to elucidate the relationship between antiretrovirals (ARVs) and viral expression in the spleen. We performed mass spectrometry imaging (MSI) of 6 different ARVs, RNAscope in situ hybridization of viral RNA, and immunohistochemistry of three different fibrosis markers in the spleens of 8 uninfected and 10 reverse transcriptase simian-human immunodeficiency virus (RT-SHIV)-infected rhesus macaques (infected for 6 weeks) that had been dosed for 10 days with combination ART. Using MATLAB, computational quantitative imaging analysis was performed to evaluate the spatial and pharmacological relationships between the 6 ARVs, viral RNA, and fibrotic deposition. In these spleens, >50% of the spleen tissue area was not covered by any detectable ARV response (any concentration above the limits of detection for individual ARVs). The median spatial ARV coverage across all tissues was driven by maraviroc followed by efavirenz. Yet >50% of RNA-positive cells were not exposed to any detectable ARV. Quantifiable maraviroc and efavirenz colocalization with RNA-positive cells was usually greater than the in vitro concentration inhibiting 50% replication (IC50). Fibrosis markers covered more than 50% of the spleen tissue area and had negative relationships with cumulative ARV coverages. Our findings suggest that a heterogeneous ARV spatial distribution must be considered when evaluating viral persistence in lymphoid tissue reservoirs.
Collapse
Affiliation(s)
| | - Nicole R. White
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Yury Desyaterik
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Gabriela De la Cruz
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Michael Nekorchuk
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret Terry
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Paul Luciw
- University of California at Davis, Davis, California, USA
| | - Yuri Fedoriw
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Elias P. Rosen
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Angela D. M. Kashuba
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
Kaya F, Zimmerman MD, Antilus-Sainte R, Gengenbacher M, Carter CL, Dartois V. Spatial quantitation of antibiotics in bone tissue compartments by laser-capture microdissection coupled with UHPLC-tandem mass spectrometry. Anal Bioanal Chem 2022; 414:6919-6927. [PMID: 35945288 PMCID: PMC9436889 DOI: 10.1007/s00216-022-04257-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022]
Abstract
Bones are the site of multiple diseases requiring chemotherapy, including cancer, arthritis, osteoporosis and infections. Yet limited methodologies are available to investigate the spatial distribution and quantitation of small molecule drugs in bone compartments, due to the difficulty of sectioning undecalcified bones and the interference of decalcification methods with spatially resolved drug quantitation. To measure drug concentrations in distinct anatomical bone regions, we have developed a workflow that enables spatial quantitation of thin undecalcified bone sections by laser-capture microdissection coupled to HPLC/tandem mass spectrometry, and spatial mapping on adjacent sections by mass spectrometry imaging. The adhesive film and staining methods were optimized to facilitate histology staining on the same sections used for mass spectrometry image acquisition, revealing drug accumulation in the underlying bone tissue architecture, for the first time. Absolute spatial concentrations of rifampicin, bedaquiline, doxycycline, vancomycin and several of their active metabolites are shown for both small rodent bones and larger rabbit bones that more closely resemble human bone density. Overlaid MALDI mass spectrometry images of drugs and histology staining enabled the generation of semi-quantitative data from regions of interest within anatomical bone compartments. These data correlated with absolute drug concentrations determined by HPLC-MS/MS in laser-capture microdissection samples. Collectively, these techniques enable semi- and fully quantitative drug distribution investigations within bone tissue compartments for the first time. Our workflow can be translated to image and quantify not only drugs but also biomarkers of disease to investigate drug penetration as well as mechanisms underlying bone disorders.
Collapse
Affiliation(s)
- Firat Kaya
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Matthew D Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Rosleine Antilus-Sainte
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Martin Gengenbacher
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, 123 Metro Blvd, Nutley, NJ, USA
| | - Claire L Carter
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA.
- Department of Pathology, Hackensack Meridian School of Medicine, 123 Metro Blvd, Nutley, NJ, USA.
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA.
- Department of Medical Sciences, Hackensack Meridian School of Medicine, 123 Metro Blvd, Nutley, NJ, USA.
| |
Collapse
|
23
|
Zang R, Barth A, Wong H, Marik J, Shen J, Lade J, Grove K, Durk MR, Parrott N, Rudewicz PJ, Zhao S, Wang T, Yan Z, Zhang D. Design and Measurement of Drug Tissue Concentration Asymmetry and Tissue Exposure-Effect (Tissue PK-PD) Evaluation. J Med Chem 2022; 65:8713-8734. [PMID: 35790118 DOI: 10.1021/acs.jmedchem.2c00502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The "free drug hypothesis" assumes that, in the absence of transporters, the steady state free plasma concentrations equal to that at the site of action that elicit pharmacologic effects. While it is important to utilize the free drug hypothesis, exceptions exist that the free plasma exposures, either at Cmax, Ctrough, and Caverage, or at other time points, cannot represent the corresponding free tissue concentrations. This "drug concentration asymmetry" in both total and free form can influence drug disposition and pharmacological effects. In this review, we first discuss options to assess total and free drug concentrations in tissues. Then various drug design strategies to achieve concentration asymmetry are presented. Last, the utilities of tissue concentrations in understanding exposure-effect relationships and translational projections to humans are discussed for several therapeutic areas and modalities. A thorough understanding in plasma and tissue exposures correlation with pharmacologic effects can provide insightful guidance to aid drug discovery.
Collapse
Affiliation(s)
- Richard Zang
- IDEAYA Biosciences, South San Francisco, California 94080, United States
| | - Aline Barth
- Global Blood Therapeutics, South San Francisco, California 94080, United States
| | - Harvey Wong
- The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jan Marik
- Genentech, South San Francisco, California 98080, United States
| | - Jie Shen
- AbbVie, Irvine, California 92612, United States
| | - Julie Lade
- Amgen Inc., South San Francisco, California 94080, United States
| | - Kerri Grove
- Novartis, Emeryville, California 94608, United States
| | - Matthew R Durk
- Genentech, South San Francisco, California 98080, United States
| | - Neil Parrott
- Roche Innovation Centre, Basel CH-4070, Switzerland
| | | | | | - Tao Wang
- Coherus BioSciences, Redwood City, California 94605, United States
| | - Zhengyin Yan
- Genentech, South San Francisco, California 98080, United States
| | - Donglu Zhang
- Genentech, South San Francisco, California 98080, United States
| |
Collapse
|
24
|
Tuck M, Grélard F, Blanc L, Desbenoit N. MALDI-MSI Towards Multimodal Imaging: Challenges and Perspectives. Front Chem 2022; 10:904688. [PMID: 35615316 PMCID: PMC9124797 DOI: 10.3389/fchem.2022.904688] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/14/2022] [Indexed: 01/22/2023] Open
Abstract
Multimodal imaging is a powerful strategy for combining information from multiple images. It involves several fields in the acquisition, processing and interpretation of images. As multimodal imaging is a vast subject area with various combinations of imaging techniques, it has been extensively reviewed. Here we focus on Matrix-assisted Laser Desorption Ionization Mass Spectrometry Imaging (MALDI-MSI) coupling other imaging modalities in multimodal approaches. While MALDI-MS images convey a substantial amount of chemical information, they are not readily informative about the morphological nature of the tissue. By providing a supplementary modality, MALDI-MS images can be more informative and better reflect the nature of the tissue. In this mini review, we emphasize the analytical and computational strategies to address multimodal MALDI-MSI.
Collapse
|
25
|
Morosi L, Matteo C, Meroni M, Ceruti T, Fuso Nerini I, Bello E, Frapolli R, D'Incalci M, Zucchetti M, Davoli E. Quantitative measurement of pioglitazone in neoplastic and normal tissues by AP-MALDI mass spectrometry imaging. Talanta 2022; 237:122918. [PMID: 34736656 DOI: 10.1016/j.talanta.2021.122918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022]
Abstract
Pioglitazone is a Peroxisome Proliferator-Activated Receptor (PPAR) agonist of the thiazolidinedione class of compounds with promising anticancer activity. An innovative quantitative mass spectrometry imaging (MSI) method and a HPLC-UV method were developed and validated to investigate its distribution in tumor and liver tissues. The MSI method is based on stable isotope normalization and resulted highly specific and sensitive (0.2 pmol/spot). The correct identification of the drug ion signal is confirmed by MS/MS analysis on tissue. The method shows an optimal lateral resolution (25 μm) relying on the ionization efficiency and fine laser diameter of the atmospheric pressure MALDI source. The HPLC-UV method is simple and straightforward involving quick protein precipitation and shows good sensitivity (50ng/sample) using a small starting volume of biological sample. Thus, it is applicable to samples obtained from both preclinical models and clinical surgical procedures. MSI and HPLC-UV assays were validated assessing linearity, intra- and inter-day precision and accuracy, limit of quantification, selectivity and recovery. These are the first methods developed and validated for the analysis of pioglitazone in tissues, and they were applied successfully to myxoid liposarcoma xenograft-bearing mice, which received clinically relevant drug doses. Pioglitazone was measured by either method in sections of tumor and liver 2, 6 and 24 h post-treatment. Drug distribution was relatively homogeneous.
Collapse
Affiliation(s)
- Lavinia Morosi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Cristina Matteo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Marina Meroni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Tommaso Ceruti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Ilaria Fuso Nerini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Ezia Bello
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Roberta Frapolli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy.
| | - Maurizio D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Massimo Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Enrico Davoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Mass Spectrometry Research Center for Health and Environment and Laboratory of Mass Spectrometry, Via Mario Negri 2, Milan, Italy
| |
Collapse
|
26
|
Schnackenberg LK, Thorn DA, Barnette D, Jones EE. MALDI imaging mass spectrometry: an emerging tool in neurology. Metab Brain Dis 2022; 37:105-121. [PMID: 34347208 DOI: 10.1007/s11011-021-00797-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/11/2021] [Indexed: 12/24/2022]
Abstract
Neurological disease and disorders remain a large public health threat. Thus, research to improve early detection and/or develop more effective treatment approaches are necessary. Although there are many common techniques and imaging modalities utilized to study these diseases, existing approaches often require a label which can be costly and time consuming. Matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) is a label-free, innovative and emerging technique that produces 2D ion density maps representing the distribution of an analyte(s) across a tissue section in relation to tissue histopathology. One main advantage of MALDI IMS over other imaging modalities is its ability to determine the spatial distribution of hundreds of analytes within a single imaging run, without the need for a label or any a priori knowledge. Within the field of neurology and disease there have been several impactful studies in which MALDI IMS has been utilized to better understand the cellular pathology of the disease and or severity. Furthermore, MALDI IMS has made it possible to map specific classes of analytes to regions of the brain that otherwise may have been lost using more traditional methods. This review will highlight key studies that demonstrate the potential of this technology to elucidate previously unknown phenomenon in neurological disease.
Collapse
Affiliation(s)
- Laura K Schnackenberg
- Division of Systems Biology, National Center for Toxicological Research/FDA, 3900 NCTR Rd, Jefferson, AR, USA
| | - David A Thorn
- Division of Systems Biology, National Center for Toxicological Research/FDA, 3900 NCTR Rd, Jefferson, AR, USA
| | - Dustyn Barnette
- Division of Systems Biology, National Center for Toxicological Research/FDA, 3900 NCTR Rd, Jefferson, AR, USA
| | - E Ellen Jones
- Division of Systems Biology, National Center for Toxicological Research/FDA, 3900 NCTR Rd, Jefferson, AR, USA.
| |
Collapse
|
27
|
Pathmasiri KC, Nguyen TTA, Khamidova N, Cologna SM. Mass spectrometry-based lipid analysis and imaging. CURRENT TOPICS IN MEMBRANES 2021; 88:315-357. [PMID: 34862030 DOI: 10.1016/bs.ctm.2021.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mass spectrometry imaging (MSI) is a powerful tool for in situ mapping of analytes across a sample. With growing interest in lipid biochemistry, the ability to perform such mapping without antibodies has opened many opportunities for MSI and lipid analysis. Herein, we discuss the basics of MSI with particular emphasis on MALDI mass spectrometry and lipid analysis. A discussion of critical advancements as well as protocol details are provided to the reader. In addition, strategies for improving the detection of lipids, as well as applications in biomedical research, are presented.
Collapse
Affiliation(s)
- Koralege C Pathmasiri
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Thu T A Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Nigina Khamidova
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States; Laboratory of Integrated Neuroscience, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
28
|
Solon E, Groseclose MR, Ho S, Tanaka K, Nakada N, Linehan S, Nishidate M, Yokoi H, Kaji H, Urasaki Y, Watanabe K, Ishida T, Komatsu R, Yoshida K, Yamazaki H, Saito K, Saito Y, Tanaka Y. Imaging Mass Spectrometry (IMS) for drug discovery and development survey: Results on methods, applications and regulatory compliance. Drug Metab Pharmacokinet 2021; 43:100438. [DOI: 10.1016/j.dmpk.2021.100438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 12/26/2022]
|
29
|
Luo S, Wu Q, Li Y, Lu H. Per-pixel absolute quantitation for mass spectrometry imaging of endogenous lipidomes by model prediction of mass transfer kinetics in single-probe-based ambient liquid extraction. Talanta 2021; 234:122654. [PMID: 34364463 DOI: 10.1016/j.talanta.2021.122654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
With the development of mass spectrometry imaging (MSI), techniques providing quantitative information on the spatial distribution have attracted more attentions recent years. However, for MSI of endogenous compounds in bio-samples, the uncertainty of locally varied sampling efficiencies always hinders accurate absolute quantitation. Here single-probe was used for ambient liquid extraction MSI in rat cerebellum, and standards of phosphatidylcholines (PCs) and cerebrosides (CBs) were doped in extraction solvent. The extraction kinetic curves of endogenous lipids in the ambient liquid extraction during probe parking in single pixel of tissue were investigated. From the results, the extraction kinetic curves were varied between different lipid species in different brain regions, resulting in variations of extraction efficiencies between imaging pixels, and calibration with standards deposited in tissue could not compensate for the variations. In our approach, the theoretical kinetic model of ambient liquid extraction was established, and original concentrations of endogenous lipids in each pixel of tissue were predicted by fitting the experimental extraction kinetic curve in each imaging pixel to the model. The experimental data was demonstrated to be well fitted to the kinetic model with R2 > 0.86, and only with 18-s extraction in each pixel, the original lipid concentrations were predicted accurately with relative errors <23%. With the new method, totally 157 lipids and small metabolites were imaged, and per-pixel quantitation was achieved for 19 PCs and 4 CBs. Compared with conventional quantitative MSI (q-MSI) method, the new q-MSI method had better reproducibility and wider linear range, and produced better contrast in the quantitative images of lipids in brain tissue with less hot spots and noises. The absolute quantitation results by the new method were verified by quantitative LC-MS method with Pearson'r > 0.9 and the slope of the linear fitting line of the correlation plot near 1.
Collapse
Affiliation(s)
- Shifen Luo
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, PR China
| | - Qian Wu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, PR China.
| | - Youmei Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, PR China
| | - Hongmei Lu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, PR China
| |
Collapse
|
30
|
Castellino S, Lareau NM, Groseclose MR. The emergence of imaging mass spectrometry in drug discovery and development: Making a difference by driving decision making. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4717. [PMID: 33724654 PMCID: PMC8365693 DOI: 10.1002/jms.4717] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 05/10/2023]
Abstract
The pharmaceutical industry is a dynamic, science-driven business constantly under pressure to innovate and morph into a higher performing organization. Innovations can include the implementation of new technologies, adopting new scientific methods, changing the decision-making process, compressing timelines, or making changes to the organizational structure. The drivers for the constant focus on performance improvement are the high cost of R&D as well as the lengthy timelines required to deliver new medicines for unmet needs. Successful innovations are measured against both the quality and quantity of potential new medicines in the pipeline and the delivery to patients. In this special feature article, we share our collective experience implementing matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) technology as an innovative approach to better understand the tissue biodistribution of drugs in the early phases of drug discovery to establish pharmacokinetic-pharmacodynamic (PK-PD) relationships, as well as in the development phase to understand pharmacology, toxicology, and disease pathogenesis. In our experience, successful implementation of MALDI IMS in support of therapeutic programs can be measured by the impact IMS studies have on driving decision making in pipeline progression. This provides a direct quantifiable measurement of the return to the organization for the investment in IMS. We have included discussion not only on the technical merits of IMS study conduct but also the key elements of setting study objectives, building collaborations, data integration into the medicine progression milestones, and potential pitfalls when trying to establish IMS in the pharmaceutical arena. We categorized IMS study types into five groups that parallel pipeline progression from the earliest phases of discovery to late stages of preclinical development. We conclude the article with some perspectives on how we see MALDI IMS maintaining relevance and becoming further embedded as an essential tool in the constantly changing environment of the pharmaceutical industry.
Collapse
Affiliation(s)
- Stephen Castellino
- GlaxoSmithKline BioimagingCollegevillePennsylvania19426USA
- Xenovista LLCChapel HillNorth Carolina27516USA
| | | | | |
Collapse
|
31
|
Lan C, Li H, Wang L, Zhang J, Wang X, Zhang R, Yuan X, Wu T, Wu J, Lu M, Ma X. Absolute quantification of 2-hydroxyglutarate on tissue by matrix-assisted laser desorption/ionization mass spectrometry imaging for rapid and precise identification of isocitrate dehydrogenase mutations in human glioma. Int J Cancer 2021; 149:2091-2098. [PMID: 34224582 DOI: 10.1002/ijc.33729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 11/09/2022]
Abstract
Isocitrate dehydrogenase (IDH) gene mutations are important predictive molecular markers to guide surgical strategy in brain cancer therapy. Herein, we presented a method using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) for absolute quantification of 2-hydroxyglutarate (2-HG) on tissues to identify IDH mutations and evaluate tumor residue. This analytical method was tested among 34 glioma patients and validated with gold standard clinical technologies. The cut-off value of 2-HG was set as 0.81 pmol/μg to identify IDH mutant (IDHmt) gliomas with 100% specificity and sensitivity. In addition, 2-HG levels and tumor cell density (TCD) showed positive correlation in IDHmt gliomas by this spatial method. This MALDI MSI-based absolute quantification method has great potentiality for incorporating into surgical workflow in the future.
Collapse
Affiliation(s)
- Chunyan Lan
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China.,Peking Union Medical College Graduate School, Beijing, China
| | - Hainan Li
- Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Lei Wang
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China
| | - Jing Zhang
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China
| | - Xiaodong Wang
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, China
| | - Rumeng Zhang
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China
| | - Xiaoai Yuan
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China
| | - Taihua Wu
- Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Jie Wu
- Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Ming Lu
- Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Xu Ma
- National Centre for Human Genetic Resources, National Research Institute for Health and Family Planning, Beijing, China.,Peking Union Medical College Graduate School, Beijing, China
| |
Collapse
|
32
|
Davoli E, Zucchetti M, Matteo C, Ubezio P, D'Incalci M, Morosi L. THE SPACE DIMENSION AT THE MICRO LEVEL: MASS SPECTROMETRY IMAGING OF DRUGS IN TISSUES. MASS SPECTROMETRY REVIEWS 2021; 40:201-214. [PMID: 32501572 DOI: 10.1002/mas.21633] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 06/11/2023]
Abstract
Mass spectrometry imaging (MSI) has seen remarkable development in recent years. The possibility of getting quantitative or semiquantitative data, while maintaining the spatial component in the tissues has opened up unique study possibilities. Now with a spatial window of few tens of microns, we can characterize the events occurring in tissue subcompartments in physiological and pathological conditions. For example, in oncology-especially in preclinical models-we can quantitatively measure drug distribution within tumors, correlating it with pharmacological treatments intended to modify it. We can also study the local effects of the drug in the tissue, and their effects in relation to histology. This review focuses on the main results in the field of drug MSI in clinical pharmacology, looking at the literature on the distribution of drugs in human tissues, and also the first preclinical evidence of drug intratissue effects. The main instrumental techniques are discussed, looking at the different instrumentation, sample preparation protocols, and raw data management employed to obtain the sensitivity required for these studies. Finally, we review the applications that describe in situ metabolic events and pathways induced by the drug, in animal models, showing that MSI makes it possible to study effects that go beyond the simple concentration of the drug, maintaining the space dimension. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Enrico Davoli
- Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Massimo Zucchetti
- Laboratory of Antitumoral Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Cristina Matteo
- Laboratory of Antitumoral Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Paolo Ubezio
- Laboratory of Antitumoral Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Maurizio D'Incalci
- Laboratory of Antitumoral Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lavinia Morosi
- Laboratory of Antitumoral Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
33
|
Abstract
Mass spectrometry imaging (MSI) is a powerful, label-free technique that provides detailed maps of hundreds of molecules in complex samples with high sensitivity and subcellular spatial resolution. Accurate quantification in MSI relies on a detailed understanding of matrix effects associated with the ionization process along with evaluation of the extraction efficiency and mass-dependent ion losses occurring in the analysis step. We present a critical summary of approaches developed for quantitative MSI of metabolites, lipids, and proteins in biological tissues and discuss their current and future applications.
Collapse
Affiliation(s)
- Daisy Unsihuay
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA; , ,
| | - Daniela Mesa Sanchez
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA; , ,
| | - Julia Laskin
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, USA; , ,
| |
Collapse
|
34
|
Quantitative mass spectrometry imaging of drugs and metabolites: a multiplatform comparison. Anal Bioanal Chem 2021; 413:2779-2791. [PMID: 33770207 PMCID: PMC8007509 DOI: 10.1007/s00216-021-03210-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 01/11/2023]
Abstract
Mass spectrometry imaging (MSI) provides insight into the molecular distribution of a broad range of compounds and, therefore, is frequently applied in the pharmaceutical industry. Pharmacokinetic and toxicological studies deploy MSI to localize potential drugs and their metabolites in biological tissues but currently require other analytical tools to quantify these pharmaceutical compounds in the same tissues. Quantitative mass spectrometry imaging (Q-MSI) is a field with challenges due to the high biological variability in samples combined with the limited sample cleanup and separation strategies available prior to MSI. In consequence, more selectivity in MSI instruments is required. This can be provided by multiple reaction monitoring (MRM) which uses specific precursor ion-product ion transitions. This targeted approach is in particular suitable for pharmaceutical compounds because their molecular identity is known prior to analysis. In this work, we compared different analytical platforms to assess the performance of MRM detection compared to other MS instruments/MS modes used in a Q-MSI workflow for two drug candidates (A and B). Limit of detection (LOD), linearity, and precision and accuracy of high and low quality control (QC) samples were compared between MS instruments/modes. MRM mode on a triple quadrupole mass spectrometer (QqQ) provided the best overall performance with the following results for compounds A and B: LOD 35.5 and 2.5 μg/g tissue, R2 0.97 and 0.98 linearity, relative standard deviation QC <13.6%, and 97-112% accuracy. Other MS modes resulted in LOD 6.7-569.4 and 2.6-119.1 μg/g tissue, R2 0.86-0.98 and 0.86-0.98 linearity, relative standard deviation QC < 19.4 and < 37.5%, and 70-356% and 64-398% accuracy for drug candidates A and B, respectively. In addition, we propose an optimized 3D printed mimetic tissue model to increase the overall analytical throughput of our approach for large animal studies. The MRM imaging platform was applied as proof-of-principle for quantitative detection of drug candidates A and B in four dog livers and compared to LC-MS. The Q-MSI concentrations differed <3.5 times with the concentrations observed by LC-MS. Our presented MRM-based Q-MSI approach provides a more selective and high-throughput analytical platform due to MRM specificity combined with an optimized 3D printed mimetic tissue model.
Collapse
|
35
|
Balluff B, Hopf C, Porta Siegel T, Grabsch HI, Heeren RMA. Batch Effects in MALDI Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:628-635. [PMID: 33523675 PMCID: PMC7944567 DOI: 10.1021/jasms.0c00393] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Mass spectrometry imaging (MSI) has become an indispensible tool for spatially resolved molecular investigation of tissues. One of the key application areas is biomedical research, where matrix-assisted laser desorption/ionization (MALDI) MSI is predominantly used due to its high-throughput capability, flexibility in the molecular class to investigate, and ability to achieve single cell spatial resolution. While many of the initial technical challenges have now been resolved, so-called batch effects, a phenomenon already known from other omics fields, appear to significantly impede reliable comparison of data from particular midsized studies typically performed in translational clinical research. This critical insight will discuss at what levels (pixel, section, slide, time, and location) batch effects can manifest themselves in MALDI-MSI data and what consequences this might have for biomarker discovery or multivariate classification. Finally, measures are presented that could be taken to recognize and/or minimize these potentially detrimental effects, and an outlook is provided on what is still needed to ultimately overcome these effects.
Collapse
Affiliation(s)
- Benjamin Balluff
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, 6229 ER Maastricht, The Netherlands
- Mailing address: Dr. Benjamin Balluff,
Maastricht University, Maastricht MultiModal Molecular Imaging institute
(M4I), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands;
Phone: +31 43 388 1251;
| | - Carsten Hopf
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Tiffany Porta Siegel
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Heike I. Grabsch
- Department
of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC+), 6229 HX Maastricht, The Netherlands
- Pathology
and Data Analytics, Leeds Institute of Medical Research at St. James’s, University of Leeds, LS9 7TF Leeds, U.K.
| | - Ron M. A. Heeren
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
36
|
Wang N, Dartois V, Carter CL. An optimized method for the detection and spatial distribution of aminoglycoside and vancomycin antibiotics in tissue sections by mass spectrometry imaging. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4708. [PMID: 33586279 PMCID: PMC8032321 DOI: 10.1002/jms.4708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 05/08/2023]
Abstract
Suboptimal antibiotic dosing has been identified as one of the key drivers in the development of multidrug-resistant (MDR) bacteria that have become a global health concern. Aminoglycosides and vancomycin are broad-spectrum antibiotics used to treat critically ill patients infected by a variety of MDR bacterial species. Resistance to these antibiotics is becoming more prevalent. In order to design proper antibiotic regimens that maximize efficacy and minimize the development of resistance, it is pivotal to obtain the in situ pharmacokinetic-pharmacodynamic profiles at the sites of infection. Mass spectrometry imaging (MSI) is the ideal technique to achieve this. Aminoglycosides, due to their structure, suffer from poor ionization efficiency. Additionally, ion suppression effects by endogenous molecules greatly inhibit the detection of aminoglycosides and vancomycin at therapeutic levels. In the current study, an optimized method was developed that enabled the detection of these antibiotics by MSI. Tissue spotting experiments demonstrated a 5-, 15-, 35-, and 54-fold increase in detection sensitivity in the washed samples for kanamycin, amikacin, streptomycin, and vancomycin, respectively. Tissue mimetic models were utilized to optimize the washing time and matrix additive concentration. These studies determined the improved limit of detection was 40 to 5 μg/g of tissue for vancomycin and streptomycin, and 40 to 10 μg/g of tissue for kanamycin and amikacin. The optimized protocol was applied to lung sections from mice dosed with therapeutic levels of kanamycin and vancomycin. The washing protocol enabled the first drug distribution investigations of aminoglycosides and vancomycin by MSI, paving the way for site-of-disease antibiotic penetration studies.
Collapse
Affiliation(s)
- Ning Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack School of Medicine, Nutley, New Jersey, USA
| | - Claire L. Carter
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| |
Collapse
|
37
|
Quantitative MALDI mass spectrometry imaging for exploring cutaneous drug delivery of tofacitinib in human skin. Eur J Pharm Biopharm 2020; 159:1-10. [PMID: 33352255 DOI: 10.1016/j.ejpb.2020.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/13/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022]
Abstract
In skin penetration studies, HPLC-MS/MS analysis on extracts of heat-separated epidermis and dermis provides an estimate of the amount of drug penetrated. In this study, MALDI-MSI enabled qualitative skin distribution analysis of endogenous molecules and the drug molecule, tofacitinib and quantitative analysis of the amount of tofacitinib in the epidermis. The delivery of tofacitinib to the skin was investigated in a Franz diffusion cell using three different formulations (two oil-in-water creams, C1 and C2 and an aqueous gel). Further, in vitro release testing (IVRT) was performed and resulted in the fastest release of tofacitinib from the aqueous gel and the lowest from C2. In the ex vivo skin penetration and permeation study, C1 showed the largest skin retention of tofacitinib, whereas, lower retention and higher permeation were observed for the gel and C2. The quantitative MALDI-MSI analysis showed that the content of tofacitinib in the epidermis for the C1 treated samples was comparable to HPLC-MS/MS analysis, whereas, the samples treated with C2 and the aqueous gel were below LOQ. The study demonstrates that MALDI-MSI can be used for the quantitative determination of drug penetration in epidermis, as well as, to provide valuable information on qualitative skin distribution of tofacitinib.
Collapse
|
38
|
Tuck M, Blanc L, Touti R, Patterson NH, Van Nuffel S, Villette S, Taveau JC, Römpp A, Brunelle A, Lecomte S, Desbenoit N. Multimodal Imaging Based on Vibrational Spectroscopies and Mass Spectrometry Imaging Applied to Biological Tissue: A Multiscale and Multiomics Review. Anal Chem 2020; 93:445-477. [PMID: 33253546 DOI: 10.1021/acs.analchem.0c04595] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Michael Tuck
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Landry Blanc
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Rita Touti
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Nathan Heath Patterson
- Mass Spectrometry Research Center, Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232-8575, United States
| | - Sebastiaan Van Nuffel
- Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Sandrine Villette
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Jean-Christophe Taveau
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Andreas Römpp
- Bioanalytical Sciences and Food Analysis, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Alain Brunelle
- Laboratoire d'Archéologie Moléculaire et Structurale, LAMS UMR 8220, CNRS, Sorbonne Université, 4 Place Jussieu, 75005 Paris, France
| | - Sophie Lecomte
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| | - Nicolas Desbenoit
- Institut de Chimie & Biologie des Membranes & des Nano-objets, CBMN UMR 5248, CNRS, Université de Bordeaux, 1 Allée Geoffroy Saint-Hilaire, 33600 Pessac, France
| |
Collapse
|
39
|
Tobias F, Hummon AB. Considerations for MALDI-Based Quantitative Mass Spectrometry Imaging Studies. J Proteome Res 2020; 19:3620-3630. [PMID: 32786684 DOI: 10.1021/acs.jproteome.0c00443] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significant advances in mass spectrometry imaging (MSI) have pushed the boundaries in obtaining spatial information and quantification in biological samples. Quantitative MSI (qMSI) has typically been challenging to achieve because of matrix and tissue heterogeneity, inefficient analyte extraction, and ion suppression effects, but recent studies have demonstrated approaches to obtain highly robust methods and reproducible results. In this perspective, we share our insights into sample preparation, how the choice of matrix influences sensitivity, construction of calibration curves, signal normalization, and visualization of MSI data. We hope that by articulating these guidelines that qMSI can be routinely conducted while retaining the analytical merits of other mass spectrometry modalities.
Collapse
|
40
|
Gachumi G, Purves RW, Hopf C, El-Aneed A. Fast Quantification Without Conventional Chromatography, The Growing Power of Mass Spectrometry. Anal Chem 2020; 92:8628-8637. [PMID: 32510944 DOI: 10.1021/acs.analchem.0c00877] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mass spectrometry (MS) in hyphenated techniques is widely accepted as the gold standard quantitative tool in life sciences. However, MS possesses intrinsic analytical capabilities that allow it to be a stand-alone quantitative technique, particularly with current technological advancements. MS has a great potential for simplifying quantitative analysis without the need for tedious chromatographic separation. Its selectivity relies on multistage MS analysis (MSn), including tandem mass spectrometry (MS/MS), as well as the ever-growing advancements of high-resolution MS instruments. This perspective describes various analytical platforms that utilize MS as a stand-alone quantitative technique, namely, flow injection analysis (FIA), matrix assisted laser desorption ionization (MALDI), including MALDI-MS imaging and ion mobility, particularly high-field asymmetric waveform ion mobility spectrometry (FAIMS). When MS alone is not capable of providing reliable quantitative data, instead of conventional liquid chromatography (LC)-MS, the use of a guard column (i.e., fast chromatography) may be sufficient for quantification. Although the omission of chromatographic separation simplifies the analytical process, extra procedures may be needed during sample preparation and clean-up to address the issue of matrix effects. The discussion of this manuscript focuses on key parameters underlying the uniqueness of each technique for its application in quantitative analysis without the need for a chromatographic separation. In addition, the potential for each analytical strategy and its challenges are discussed as well as improvements needed to render them as mainstream quantitative analytical tools. Overcoming the hurdles for fully validating a quantitative method will allow MS alone to eventually become an indispensable quantitative tool for clinical and toxicological studies.
Collapse
Affiliation(s)
- George Gachumi
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan Canada, S7N 5E5
| | - Randy W Purves
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan Canada, S7N 5E5.,Centre for Veterinary Drug Residues, Canadian Food Inspection Agency, 116 Veterinary Rd, Saskatoon, Saskatchewan Canada, S7N 2R3
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, 68163 Mannheim, Germany
| | - Anas El-Aneed
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan Canada, S7N 5E5
| |
Collapse
|
41
|
Morosi L, Matteo C, Ceruti T, Giordano S, Ponzo M, Frapolli R, Zucchetti M, Davoli E, D'Incalci M, Ubezio P. Quantitative determination of niraparib and olaparib tumor distribution by mass spectrometry imaging. Int J Biol Sci 2020; 16:1363-1375. [PMID: 32210725 PMCID: PMC7085221 DOI: 10.7150/ijbs.41395] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/18/2020] [Indexed: 12/15/2022] Open
Abstract
Rationale: Optimal intratumor distribution of an anticancer drug is fundamental to reach an active concentration in neoplastic cells, ensuring the therapeutic effect. Determination of drug concentration in tumor homogenates by LC-MS/MS gives important information about this issue but the spatial information gets lost. Targeted mass spectrometry imaging (MSI) has great potential to visualize drug distribution in the different areas of tumor sections, with good spatial resolution and superior specificity. MSI is rapidly evolving as a quantitative technique to measure the absolute drug concentration in each single pixel. Methods: Different inorganic nanoparticles were tested as matrices to visualize the PARP inhibitors (PARPi) niraparib and olaparib. Normalization by deuterated internal standard and a custom preprocessing pipeline were applied to achieve a reliable single pixel quantification of the two drugs in human ovarian tumors from treated mice. Results: A quantitative method to visualize niraparib and olaparib in tumor tissue of treated mice was set up and validated regarding precision, accuracy, linearity, repeatability and limit of detection. The different tumor penetration of the two drugs was visualized by MSI and confirmed by LC-MS/MS, indicating the homogeneous distribution and higher tumor exposure reached by niraparib compared to olaparib. On the other hand, niraparib distribution was heterogeneous in an ovarian tumor model overexpressing the multidrug resistance protein P-gp, a possible cause of resistance to PARPi. Conclusions: The current work highlights for the first time quantitative distribution of PAPRi in tumor tissue. The different tumor distribution of niraparib and olaparib could have important clinical implications. These data confirm the validity of MSI for spatial quantitative measurement of drug distribution providing fundamental information for pharmacokinetic studies, drug discovery and the study of resistance mechanisms.
Collapse
Affiliation(s)
- Lavinia Morosi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Cristina Matteo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Tommaso Ceruti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Silvia Giordano
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Laboratory of Mass Spectrometry
| | - Marianna Ponzo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Roberta Frapolli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Massimo Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Enrico Davoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Laboratory of Mass Spectrometry
| | - Maurizio D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Paolo Ubezio
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| |
Collapse
|