1
|
Frezzetti D, Caridi V, Marra L, Camerlingo R, D’Alessio A, Russo F, Dotolo S, Rachiglio AM, Esposito Abate R, Gallo M, Maiello MR, Morabito A, Normanno N, De Luca A. The Impact of Inadequate Exposure to Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors on the Development of Resistance in Non-Small-Cell Lung Cancer Cells. Int J Mol Sci 2024; 25:4844. [PMID: 38732063 PMCID: PMC11084975 DOI: 10.3390/ijms25094844] [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: 04/05/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) patients treated with EGFR-tyrosine kinase inhibitors (TKIs) inevitably develop resistance through several biological mechanisms. However, little is known on the molecular mechanisms underlying acquired resistance to suboptimal EGFR-TKI doses, due to pharmacodynamics leading to inadequate drug exposure. To evaluate the effects of suboptimal EGFR-TKI exposure on resistance in NSCLC, we obtained HCC827 and PC9 cell lines resistant to suboptimal fixed and intermittent doses of gefitinib and compared them to cells exposed to higher doses of the drug. We analyzed the differences in terms of EGFR signaling activation and the expression of epithelial-mesenchymal transition (EMT) markers, whole transcriptomes byRNA sequencing, and cell motility. We observed that the exposure to low doses of gefitinib more frequently induced a partial EMT associated with an induced migratory ability, and an enhanced transcription of cancer stem cell markers, particularly in the HCC827 gefitinib-resistant cells. Finally, the HCC827 gefitinib-resistant cells showed increased secretion of the EMT inducer transforming growth factor (TGF)-β1, whose inhibition was able to partially restore gefitinib sensitivity. These data provide evidence that different levels of exposure to EGFR-TKIs in tumor masses might promote different mechanisms of acquired resistance.
Collapse
Affiliation(s)
- Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Vincenza Caridi
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Laura Marra
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Rosa Camerlingo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Amelia D’Alessio
- Laboratory of Toxicology Analysis, Department for the Treatment of Addictions, ASL Salerno, 84124 Salerno, Italy;
| | - Francesco Russo
- Institute of Endocrinology and Experimental Oncology, National Research Council of Italy, 80131 Naples, Italy;
| | - Serena Dotolo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Marianna Gallo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Alessandro Morabito
- Thoracic Department, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.F.); (V.C.); (L.M.); (R.C.); (S.D.); (A.M.R.); (R.E.A.); (M.G.); (M.R.M.); (A.D.L.)
| |
Collapse
|
2
|
Murphy PS, Galette P, van der Aart J, Janiczek RL, Patel N, Brown AP. The role of clinical imaging in oncology drug development: progress and new challenges. Br J Radiol 2023; 96:20211126. [PMID: 37393537 PMCID: PMC10546429 DOI: 10.1259/bjr.20211126] [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: 10/08/2021] [Revised: 02/14/2023] [Accepted: 06/06/2023] [Indexed: 07/03/2023] Open
Abstract
In 2008, the role of clinical imaging in oncology drug development was reviewed. The review outlined where imaging was being applied and considered the diverse demands across the phases of drug development. A limited set of imaging techniques was being used, largely based on structural measures of disease evaluated using established response criteria such as response evaluation criteria in solid tumours. Beyond structure, functional tissue imaging such as dynamic contrast-enhanced MRI and metabolic measures using [18F]flourodeoxyglucose positron emission tomography were being increasingly incorporated. Specific challenges related to the implementation of imaging were outlined including standardisation of scanning across study centres and consistency of analysis and reporting. More than a decade on the needs of modern drug development are reviewed, how imaging has evolved to support new drug development demands, the potential to translate state-of-the-art methods into routine tools and what is needed to enable the effective use of this broadening clinical trial toolset. In this review, we challenge the clinical and scientific imaging community to help refine existing clinical trial methods and innovate to deliver the next generation of techniques. Strong industry-academic partnerships and pre-competitive opportunities to co-ordinate efforts will ensure imaging technologies maintain a crucial role delivering innovative medicines to treat cancer.
Collapse
Affiliation(s)
| | - Paul Galette
- Telix Pharmaceuticals (US) Inc, Fishers, United States
| | | | | | | | - Andrew P. Brown
- Vale Imaging Consultancy Solutions, Harston, Cambridge, United Kingdom
| |
Collapse
|
3
|
Vélez Salazar FM, Patiño Arcila ID. Influence of electric field, blood velocity, and pharmacokinetics on electrochemotherapy efficiency. Biophys J 2023; 122:3268-3298. [PMID: 37421133 PMCID: PMC10465711 DOI: 10.1016/j.bpj.2023.07.004] [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: 12/23/2022] [Revised: 04/13/2023] [Accepted: 07/05/2023] [Indexed: 07/09/2023] Open
Abstract
The convective delivery of chemotherapeutic drugs in cancerous tissues is directly proportional to the blood perfusion rate, which in turns can be transiently reduced by the application of high-voltage and short-duration electric pulses due to vessel vasoconstriction. However, electric pulses can also increase vessel wall and cell membrane permeabilities, boosting the extravasation and cell internalization of drug. These opposite effects, as well as possible adverse impacts on the viability of tissues and endothelial cells, suggest the importance of conducting in silico studies about the influence of physical parameters involved in electric-mediated drug transport. In the present work, the global method of approximate particular solutions for axisymmetric domains, together with two solution schemes (Gauss-Seidel iterative and linearization+successive over-relaxation), is applied for the simulation of drug transport in electroporated cancer tissues, using a continuum tumor cord approach and considering both the electropermeabilization and vasoconstriction phenomena. The developed global method of approximate particular solutions algorithm is validated with numerical and experimental results previously published, obtaining a satisfactory accuracy and convergence. Then, a parametric study about the influence of electric field magnitude and inlet blood velocity on the internalization efficacy, drug distribution uniformity, and cell-kill capacity of the treatment, as expressed by the number of internalized moles into viable cells, homogeneity of exposure to bound intracellular drug, and cell survival fraction, respectively, is analyzed for three pharmacokinetic profiles, namely one-short tri-exponential, mono-exponential, and uniform. According to numerical results, the trade-off between vasoconstriction and electropermeabilization effects and, consequently, the influence of electric field magnitude and inlet blood velocity on the assessment parameters considered here (efficacy, uniformity, and cell-kill capacity) is different for each pharmacokinetic profile deemed.
Collapse
Affiliation(s)
| | - Iván David Patiño Arcila
- Grupo de Investigación e Innovación Ambiental (GIIAM), Institución Universitaria Pascual Bravo, Medellín, Colombia.
| |
Collapse
|
4
|
Vélez Salazar FM, Patiño Arcila ID. Influence of electric pulse characteristics on the cellular internalization of chemotherapeutic drugs and cell survival fraction in electroporated and vasoconstricted cancer tissues using boundary element techniques. J Math Biol 2023; 87:31. [PMID: 37462802 DOI: 10.1007/s00285-023-01963-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/09/2023] [Accepted: 06/30/2023] [Indexed: 07/21/2023]
Abstract
Electroporation has emerged as a suitable technique to induce the pore formation in the cell membrane of cancer tissues, facilitating the cellular internalization of chemotherapeutic drugs. An adequate selection of the electric pulse characteristics is crucial to guarantee the efficiency of this technique, minimizing the adverse effects. In the present work, the dual reciprocity boundary element method (DR-BEM) is applied for the simulation of drug transport in the extracellular and intracellular space of cancer tissues subjected to the application of controlled electric pulses, using a continuum tumour cord approach, and considering both the electro-permeabilization and vasoconstriction phenomena. The developed DR-BEM algorithm is validated with numerical and experimental results previously published, obtaining a satisfactory accuracy and convergence. Using the DR-BEM code, a study about the influence of the magnitude of electric field (E) and pulse spacing (dpulses) on the time behavior and spatial distribution of the internalized drug, as well as on the cell survival fraction, is carried out. In general, the change of drug concentration, drug exposure and cell survival fraction with the parameters E and dpulses is ruled by two important factors: the balance between the electro-permeabilization and vasoconstriction phenomena, and the relative importance of the sources of cell death (electric pulses and drug cytotoxicity); these two factors, in turn, significantly depend on the reversible and irreversible thresholds considered for the electric field.
Collapse
Affiliation(s)
- Fabián Mauricio Vélez Salazar
- Grupo de Investigación e Innovación Ambiental (GIIAM), Institución Universitaria Pascual Bravo, Cl. 73 No 73A-226 (Bloque 7), Medellín, Colombia
- Grupo de Investigación de Ciencias Administrativas, Instituto Tecnológico Metropolitano - ITM, Medellín, Colombia
| | - Iván David Patiño Arcila
- Grupo de Investigación e Innovación Ambiental (GIIAM), Institución Universitaria Pascual Bravo, Cl. 73 No 73A-226 (Bloque 7), Medellín, Colombia.
| |
Collapse
|
5
|
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
|
6
|
Koganemaru S, Kawai T, Fuchigami H, Maeda N, Koyama K, Kuboki Y, Mukohara T, Doi T, Yasunaga M. Quantitative analysis of drug distribution in heterogeneous tissues using dual-stacking capillary electrophoresis-mass spectrometry. Br J Pharmacol 2023; 180:762-774. [PMID: 36377519 DOI: 10.1111/bph.15988] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Intratumour heterogeneity frequently leads to drug resistance, which is a major issue in drug discovery. Drug distribution is one of the key factors for elucidating the resistance mechanism; however, quantitative and regional drug measurement is challenging. Here, we developed a novel ultra-sensitive analytical method and applied it to HER3-targeting antibody-drug conjugate patritumab deruxtecan (HER3-DXd), aiming to explore its payload (DXd) distribution within heterogeneous tissues. EXPERIMENTAL APPROACH The developed analytical method is named LDMS-CE-MS, a capillary electrophoresis-mass spectrometry (CE-MS) coupled with a novel sample preconcentration/separation method called "large-volume dual-sample stacking by micelle collapse and sweeping (LDMS)". First, the analytical performance of LDMS-CE-MS for DXd detection was evaluated. Subsequently, we evaluated the bystander effect of HER3-DXd, where tumour tissues were excised from xenograft models and clinical specimens after administration of HER3-DXd. HER3-high expression, adjacent, and HER3-low expression regions were then sampled by laser microdissection to quantify the released DXd. KEY RESULTS LDMS concentrated DXd by 1000-fold and separated it from the hydrophilic bio-matrix through continuous capture and release by the charged micelles, allowing quantification at sub-attomole-level. DXd concentrations decreased in the order of antigen-high expression > adjacent > antigen-low expression regions in the tumour xenograft model, whereas in clinical specimens, adjacent and antigen-high expression regions had approximately the same concentration. These distributions represent a bystander effect. CONCLUSIONS AND IMPLICATIONS Our LDMS-CE-MS successfully visualized the attomole-level drug distributions in heterogeneous clinical specimens. This new platform opens a new era of quantitative pharmacokinetic analysis, facilitating drug discovery and development.
Collapse
Affiliation(s)
- Shigehiro Koganemaru
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Kawai
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan.,RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Hirobumi Fuchigami
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Naoyuki Maeda
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kumiko Koyama
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yasutoshi Kuboki
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshihiko Doi
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
7
|
Powell NR, Silvola RM, Howard JS, Badve S, Skaar TC, Ipe J. Quantification of spatial pharmacogene expression heterogeneity in breast tumors. Cancer Rep (Hoboken) 2023; 6:e1686. [PMID: 35906899 PMCID: PMC9875649 DOI: 10.1002/cnr2.1686] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/27/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chemotherapeutic drug concentrations vary across different regions of tumors and this is thought to be involved in development of chemotherapy resistance. Insufficient drug delivery to some regions of the tumor may be due to spatial differences in expression of genes involved in the disposition, transport, and detoxification of drugs (pharmacogenes). Therefore, in this study, we analyzed the spatial expression of 286 pharmacogenes in six breast cancer tissues using the recently developed Visium spatial transcriptomics platform to (1) determine if these pharmacogenes are expressed heterogeneously across tumor tissue and (2) to determine which pharmacogenes have the most spatial expression heterogeneity. METHODS AND RESULTS The spatial transcriptomics technology sequences the transcriptome of 55 um diameter barcoded sections (spots) across a tissue sample. We analyzed spatial gene expression profiles of four biobank-sourced breast tumor samples in addition to two breast tumor sample datasets from 10× Genomics. We define heterogeneity as the interquartile range of read counts. Collectively, we identified 8887 spots in tumor regions, 3814 in stroma, 44 in lymphocytes, and 116 in normal regions based on pathologist annotation of the tissues. We showed statistically significant differences in expression of pharmacogenes in tumor regions compared to surrounding non-tumor regions. We also observed that the most heterogeneously expressed genes within tumor regions were involved in reactive oxygen species (ROS) handling and detoxification mechanisms. GPX4, GSTP1, MGST3, SOD1, CYP4Z1, CYB5R3, GSTK1, and NAT1 showed the most heterogeneous expression within tumor regions. CONCLUSIONS The heterogeneous expression of these pharmacogenes may have important implications for cancer therapy due to their ability to impact drug distribution and efficacy throughout the tumor. Our results suggest that chemoresistance caused by expression of GPX4, GSTP1, MGST3, and SOD1 may be intrinsic, not acquired, since the heterogeneity is not specific to chemotherapy-treated samples or cell type. Additionally, we identified candidate chemoresistance pharmacogenes that can be further tested through focused follow-up studies.
Collapse
Affiliation(s)
- Nicholas R. Powell
- Department of Medicine, Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rebecca M. Silvola
- Department of Medicine, Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - John S. Howard
- Department of Medicine, Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sunil Badve
- Department of Pathology and Laboratory MedicineEmory University School of MedicineAtlantaGeorgiaUSA
| | - Todd C. Skaar
- Department of Medicine, Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Joseph Ipe
- Department of Medicine, Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
8
|
Nwabufo CK, Aigbogun OP. The Role of Mass Spectrometry Imaging in Pharmacokinetic Studies. Xenobiotica 2022; 52:811-827. [PMID: 36048000 DOI: 10.1080/00498254.2022.2119900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Although liquid chromatography-tandem mass spectrometry is the gold standard analytical platform for the quantification of drugs, metabolites, and biomarkers in biological samples, it cannot localize them in target tissues.The localization and quantification of drugs and/or their associated metabolites in target tissues is a more direct measure of bioavailability, biodistribution, efficacy, and regional toxicity compared to the traditional substitute studies using plasma.Therefore, combining high spatial resolution imaging functionality with the superior selectivity and sensitivity of mass spectrometry into one analytical technique will be a valuable tool for targeted localization and quantification of drugs, metabolites, and biomarkers.Mass spectrometry imaging (MSI) is a tagless analytical technique that allows for the direct localization and quantification of drugs, metabolites, and biomarkers in biological tissues, and has been used extensively in pharmaceutical research.The overall goal of this current review is to provide a detailed description of the working principle of MSI and its application in pharmacokinetic studies encompassing absorption, distribution, metabolism, excretion, and toxicity processes, followed by a discussion of the strategies for addressing the challenges associated with the functional utility of MSI in pharmacokinetic studies that support drug development.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Omozojie P Aigbogun
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
9
|
Menetrey A, Legouffe R, Haouala A, Bonnel D, Rouits E, Bosq J, Stauber J. Tumor Distribution by Quantitative Mass Spectrometry Imaging of the Inhibitor of Apoptosis Protein Antagonist Xevinapant in Patients with Resectable Squamous Cell Carcinoma of the Head and Neck (EudraCT Number: 2014-004655-31). Anal Chem 2022; 94:12333-12341. [PMID: 36040476 DOI: 10.1021/acs.analchem.2c00943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As tumors are very heterogeneous, investigating the penetration and concentration of an anticancer drug in different histological regions of a tumor is key to evaluate the efficacy, to improve the pharmacokinetics/pharmacodynamics (PK/PD) relationship evaluation, and to confirm the adequacy of the dose regimen. Quantitative mass spectrometry imaging (QMSI) allows for the determination of the tissue distribution of drugs, metabolites, and biomarkers to support quick and precise evaluation of drug efficacy and safety in a single experiment. QMSI was applied in a preoperative window-of-opportunity (WoO) study of the inhibitor of apoptosis protein antagonist xevinapant (Debio 1143) in patients with resectable squamous cell carcinoma of the head and neck (SCCHN). Tumors were isolated, immediately snap-frozen, and sectioned, and then, the molecular distribution of the drug was generated by matrix-assisted laser desorption ionization (MALDI) imaging. Additionally, the different histological regions (tumor, epithelium, salivary glands, muscle, nerve, and blood vessels) were identified on stained sections adjacent to the ones used for QMSI, leading to a specific quantification integrating the biological characterization of the tumor heterogeneity. This innovative approach allowed one to highlight the high affinity of xevinapant for the tumor tissues.
Collapse
Affiliation(s)
- Annick Menetrey
- Debiopharm International SA, Chemin Messidor 5-7, 1002 Lausanne, Switzerland
| | - Raphael Legouffe
- ImaBiotech, Parc Eurasanté, 152 rue du Docteur Yersin, 59120 Loos, France
| | - Amina Haouala
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215 Meyrin, Switzerland
| | - David Bonnel
- ImaBiotech, Parc Eurasanté, 152 rue du Docteur Yersin, 59120 Loos, France
| | - Elisabeth Rouits
- Debiopharm International SA, Chemin Messidor 5-7, 1002 Lausanne, Switzerland
| | - Jacques Bosq
- Sciempath Labo, 7, rue de la Gratiole, 37270 Larcay, France
| | - Jonathan Stauber
- ImaBiotech, Parc Eurasanté, 152 rue du Docteur Yersin, 59120 Loos, France
| |
Collapse
|
10
|
López CL, Brempelis KJ, Matthaei JF, Montgomery KS, Srinivasan S, Roy D, Huang F, Kreuser SA, Gardell JL, Blumenthal I, Chiefari J, Jensen MC, Crane CA, Stayton PS. Arming Immune Cell Therapeutics with Polymeric Prodrugs. Adv Healthc Mater 2022; 11:e2101944. [PMID: 34889072 PMCID: PMC9847575 DOI: 10.1002/adhm.202101944] [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: 09/13/2021] [Revised: 11/16/2021] [Indexed: 01/21/2023]
Abstract
Engineered immune cells are an exciting therapeutic modality, which survey and attack tumors. Backpacking strategies exploit cell targeting capabilities for delivery of drugs to combat tumors and their immune-suppressive environments. Here, a new platform for arming cell therapeutics through dual receptor and polymeric prodrug engineering is developed. Macrophage and T cell therapeutics are engineered to express a bioorthogonal single chain variable fragment receptor. The receptor binds a fluorescein ligand that directs cell loading with ligand-tagged polymeric prodrugs, termed "drugamers." The fluorescein ligand facilitates stable binding of drugamer to engineered macrophages over 10 days with 80% surface retention. Drugamers also incorporate prodrug monomers of the phosphoinositide-3-kinase inhibitor, PI-103. The extended release of PI-103 from the drugamer sustains antiproliferative activity against a glioblastoma cell line compared to the parent drug. The versatility and modularity of this cell arming system is demonstrated by loading T cells with a second fluorescein-drugamer. This drugamer incorporates a small molecule estrogen analog, CMP8, which stabilizes a degron-tagged transgene to provide temporal regulation of protein activity in engineered T cells. These results demonstrate that this bioorthogonal receptor and drugamer system can be used to arm multiple immune cell classes with both antitumor and transgene-activating small molecule prodrugs.
Collapse
Affiliation(s)
- Ciana L López
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Katherine J Brempelis
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - James F Matthaei
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Kate S Montgomery
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Debashish Roy
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Fei Huang
- CSIRO Manufacturing, Bag 10, Bayview Avenue, Clayton, VIC. 3168, Australia
| | - Shannon A Kreuser
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jennifer L Gardell
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Ian Blumenthal
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - John Chiefari
- CSIRO Manufacturing, Bag 10, Bayview Avenue, Clayton, VIC. 3168, Australia
| | - Michael C Jensen
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA,Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, WA 98101, USA,Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Courtney A Crane
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA,Department of Neurological Surgery, University of Washington, Seattle WA 98195, USA
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| |
Collapse
|
11
|
Min X, Zhu J, Shang M, Liu J, Zhang K, Guo L, Li L, Cheng L, Li J. Stiffness Could be a Predictor of AJCC Prognostic Stage Groups in Preoperative Invasive Ductal Carcinoma. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2021; 40:2665-2674. [PMID: 33629753 DOI: 10.1002/jum.15657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVES This study aimed to evaluate the stiffness of 2-dimensional (2D) shear wave elastography (SWE) in preoperatively predicting the prognostic stage groups of invasive ductal carcinoma (IDC). METHODS Eighty-six newly diagnosed lesions on 83 patients with IDCs were analyzed. All parameters from conventional ultrasound and stiffness to virtual touch tissue imaging and quantification were collected, and mean shear wave velocity (SWVmean) was calculated. Data on maximum diameter, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), histologic grading system and Tumor Node Metastasis (TNM) stages were collected. The levels of maximum shear wave velocity (SWVmax), minimum shear wave velocity (SWVmin) and SWVmean were compared. In receiver operating characteristic (ROC) curves analysis, the diagnostic efficacy was found in area under the curve (AUC). Parallel mode was used to improve the predictive value of sensitivity. RESULTS The median stiffness of SWVmax and SWVmean for IDCs were 9.38 and 6.32 m/s for late stage (stages II, III, IV) and 6.39 m/s and 4.72 m/s for early stage (stage I) of the prognostic stage groups, respectively. The median stiffness values in the late stage were significantly higher than those in the early stage (P = .003, P = .005). The optimal cutoff stiffness of SWVmax and SWVmean were 8.62 and 6.13 m/s, respectively. In ROC curves analysis, the AUC for SWVmax was 0.742, and it showed a better diagnostic value than SWVmean (0.725). In predictive diagnosis, the sensitivity for SWVmax and SWVmean were both 62.50%. The parallel mode improved the prediction power of sensitivity to 68.75%. CONCLUSIONS Preoperative SWV level may serve as a promising prognostic imaging indicator for breast IDCs.
Collapse
Affiliation(s)
- Xiang Min
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Health Management Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiang Zhu
- Department of Breast Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jikai Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kai Zhang
- Department of Breast Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Li
- Department of Ultrasound, Medical Section, Jinan Maternal and Child Health Hospital, Jinan, China
| | - Lin Cheng
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
12
|
Serain AF, Morosi L, Ceruti T, Matteo C, Meroni M, Minatel E, Zucchetti M, Salvador MJ. Betulinic acid and its spray dried microparticle formulation: In vitro PDT effect against ovarian carcinoma cell line and in vivo plasma and tumor disposition. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2021; 224:112328. [PMID: 34628206 DOI: 10.1016/j.jphotobiol.2021.112328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/31/2021] [Accepted: 09/28/2021] [Indexed: 01/03/2023]
Abstract
The race against ovarian cancer continue to motivate the research worldwide. It is known that many antitumor drugs have limited penetration into solid tumor tissues due to its microenvironment, thus contributing to their low efficacy. Therapeutic modalities have been exploited to elicit antitumor effects based on microenvironment of tumor, including Photodynamic therapy (PDT). Prospection of natural small molecules and nanotechnology are important tools in the development of new ways of obtaining photoactive compounds that are biocompatible. The Betulinic acid (BA) has shown potential biological effect as bioactive drug, but it has low water solubility. Thus, in the present study, owing to the poor solubility of the BA, its free form (BAF) was compared to a spray dried microparticle betulinic acid/HP-β-CD formulation (BAC) aiming to assess the BAF and BAC efficacy as a photosensitizer in PDT for application in ovarian cancer. BAF and BAC were submitted to assays in the presence of LED (λ = 420 nm) under different conditions (2.75 J/cm2, 5.5 J/cm2, and 11 J/cm2) and in absence of irradiation, after 5 min or 4 h of contact with ovarian carcinoma cells (A2780) or fibroblast murine cells (3T3). Furthermore, HPLC-MS/MS and MALDI-MSI methods were developed and validated in plasma and tumor of mice proving suitable for in vivo studies. The results found a greater photoinduced cytotoxic effect for the BAC at low concentration for A2780 when irradiated with LED with similar results for fluorescence microscopy. The results motivate us to continue the studies with the BA as a potential antitumor bioactive compound.
Collapse
Affiliation(s)
- Alessandra F Serain
- Universidade Estadual de Campinas (UNICAMP), Instituto de Biologia, Departamento de Biologia Vegetal, PPG BTPB, Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil.
| | - Lavinia Morosi
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Tommaso Ceruti
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Cristina Matteo
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Marina Meroni
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Elaine Minatel
- Universidade Estadual de Campinas (UNICAMP), Instituto de Biologia, Departamento de Biologia Estrutural e Funcional, Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Massimo Zucchetti
- Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Marcos J Salvador
- Universidade Estadual de Campinas (UNICAMP), Instituto de Biologia, Departamento de Biologia Vegetal, PPG BTPB, Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil.
| |
Collapse
|
13
|
Kell DB. The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 2021; 26:5629. [PMID: 34577099 PMCID: PMC8470029 DOI: 10.3390/molecules26185629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, my colleagues and I have come to realise that the likelihood of pharmaceutical drugs being able to diffuse through whatever unhindered phospholipid bilayer may exist in intact biological membranes in vivo is vanishingly low. This is because (i) most real biomembranes are mostly protein, not lipid, (ii) unlike purely lipid bilayers that can form transient aqueous channels, the high concentrations of proteins serve to stop such activity, (iii) natural evolution long ago selected against transport methods that just let any undesirable products enter a cell, (iv) transporters have now been identified for all kinds of molecules (even water) that were once thought not to require them, (v) many experiments show a massive variation in the uptake of drugs between different cells, tissues, and organisms, that cannot be explained if lipid bilayer transport is significant or if efflux were the only differentiator, and (vi) many experiments that manipulate the expression level of individual transporters as an independent variable demonstrate their role in drug and nutrient uptake (including in cytotoxicity or adverse drug reactions). This makes such transporters valuable both as a means of targeting drugs (not least anti-infectives) to selected cells or tissues and also as drug targets. The same considerations apply to the exploitation of substrate uptake and product efflux transporters in biotechnology. We are also beginning to recognise that transporters are more promiscuous, and antiporter activity is much more widespread, than had been realised, and that such processes are adaptive (i.e., were selected by natural evolution). The purpose of the present review is to summarise the above, and to rehearse and update readers on recent developments. These developments lead us to retain and indeed to strengthen our contention that for transmembrane pharmaceutical drug transport "phospholipid bilayer transport is negligible".
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK;
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
- Mellizyme Biotechnology Ltd., IC1, Liverpool Science Park, Mount Pleasant, Liverpool L3 5TF, UK
| |
Collapse
|
14
|
Ahn SW, Ferland B, Jonas OH. An Interactive Pipeline for Quantitative Histopathological Analysis of Spatially Defined Drug Effects in Tumors. J Pathol Inform 2021; 12:34. [PMID: 34760331 PMCID: PMC8529341 DOI: 10.4103/jpi.jpi_17_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumor heterogeneity is increasingly being recognized as a major source of variability in the histopathological assessment of drug responses. Quantitative analysis of immunohistochemistry (IHC) and immunofluorescence (IF) images using biomarkers that capture spatialpatterns of distinct tumor biology and drug concentration in tumors is of high interest to the field. METHODS We have developed an image analysis pipeline to measure drug response using IF and IHC images along spatial gradients of local drug release from a tumor-implantable drug delivery microdevice. The pipeline utilizes a series of user-interactive python scripts and CellProfiler pipelines with custom modules to perform image and spatial analysis of regions of interest within whole-slide images. RESULTS Worked examples demonstrate that intratumor measurements such as apoptosis, cell proliferation, and immune cell population density can be quantitated in a spatially and drug concentration-dependent manner, establishing in vivo profiles of pharmacodynamics and pharmacokinetics in tumors. CONCLUSIONS Spatial image analysis of tumor response along gradients of local drug release is achievable in high throughput. The major advantage of this approach is the use of spatially aware annotation tools to correlate drug gradients with drug effects in tumors in vivo.
Collapse
Affiliation(s)
- Sebastian W Ahn
- Department of Radiology, Laboratory for Bio-Micro Devices, Brigham and Women’s Hospital, Boston, MA, USA
| | - Benjamin Ferland
- Department of Radiology, Laboratory for Bio-Micro Devices, Brigham and Women’s Hospital, Boston, MA, USA
| | - Oliver H Jonas
- Department of Radiology, Laboratory for Bio-Micro Devices, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
15
|
Morosi L, Meroni M, Ubezio P, Fuso Nerini I, Minoli L, Porcu L, Panini N, Colombo M, Blouw B, Kang DW, Davoli E, Zucchetti M, D'Incalci M, Frapolli R. PEGylated recombinant human hyaluronidase (PEGPH20) pre-treatment improves intra-tumour distribution and efficacy of paclitaxel in preclinical models. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:286. [PMID: 34507591 PMCID: PMC8434701 DOI: 10.1186/s13046-021-02070-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Scarce drug penetration in solid tumours is one of the possible causes of the limited efficacy of chemotherapy and is related to the altered tumour microenvironment. The abnormal tumour extracellular matrix (ECM) together with abnormal blood and lymphatic vessels, reactive stroma and inflammation all affect the uptake, distribution and efficacy of anticancer drugs. METHODS We investigated the effect of PEGylated recombinant human hyaluronidase PH20 (PEGPH20) pre-treatment in degrading hyaluronan (hyaluronic acid; HA), one of the main components of the ECM, to improve the delivery of antitumor drugs and increase their therapeutic efficacy. The antitumor activity of paclitaxel (PTX) in HA synthase 3-overexpressing and wild-type SKOV3 ovarian cancer model and in the BxPC3 pancreas xenograft tumour model, was evaluated by monitoring tumour growth with or without PEGPH20 pre-treatment. Pharmacokinetics and tumour penetration of PTX were assessed by HPLC and mass spectrometry imaging analysis in the same tumour models. Tumour tissue architecture and HA deposition were analysed by histochemistry. RESULTS Pre-treatment with PEGPH20 modified tumour tissue architecture and improved the antitumor activity of paclitaxel in the SKOV3/HAS3 tumour model, favouring its accumulation and more homogeneous intra-tumour distribution, as assessed by quantitative and qualitative analysis. PEGPH20 also reduced HA content influencing, though less markedly, PTX distribution and antitumor activity in the BxPC3 tumour model. CONCLUSION Remodelling the stroma of HA-rich tumours by depletion of HA with PEGPH20 pre-treatment, is a potentially successful strategy to improve the intra-tumour distribution of anticancer drugs, increasing their therapeutic efficacy, without increasing toxicity.
Collapse
Affiliation(s)
- Lavinia Morosi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy.,Present address: IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marina Meroni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | - Paolo Ubezio
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | - Ilaria Fuso Nerini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy.,Present address: IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Lucia Minoli
- Department of Veterinary Medicine, University of Milan, Lodi, Italy.,Mouse and Animal Pathology Laboratory (MAPLab), Fondazione UniMi, Milan, Italy
| | - Luca Porcu
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | - Nicolò Panini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | - Marika Colombo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | | | - David W Kang
- Halozyme Therapeutics, San Diego, California, USA
| | - Enrico Davoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Laboratory of Mass Spectrometry, Milan, Italy
| | - Massimo Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy
| | - Maurizio D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy.,Present address: IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy.,Present address: Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Roberta Frapolli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, via M. Negri 2, 20156, Milan, Italy.
| |
Collapse
|
16
|
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
|
17
|
He Q, Sun C, Liu J, Pan Y. MALDI-MSI analysis of cancer drugs: Significance, advances, and applications. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
18
|
Prasad M, Postma G, Franceschi P, Morosi L, Giordano S, Falcetta F, Giavazzi R, Davoli E, Buydens LMC, Jansen J. A methodological approach to correlate tumor heterogeneity with drug distribution profile in mass spectrometry imaging data. Gigascience 2020; 9:giaa131. [PMID: 33241286 PMCID: PMC7688471 DOI: 10.1093/gigascience/giaa131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/28/2020] [Accepted: 11/01/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Drug mass spectrometry imaging (MSI) data contain knowledge about drug and several other molecular ions present in a biological sample. However, a proper approach to fully explore the potential of such type of data is still missing. Therefore, a computational pipeline that combines different spatial and non-spatial methods is proposed to link the observed drug distribution profile with tumor heterogeneity in solid tumor. Our data analysis steps include pre-processing of MSI data, cluster analysis, drug local indicators of spatial association (LISA) map, and ions selection. RESULTS The number of clusters identified from different tumor tissues. The spatial homogeneity of the individual cluster was measured using a modified version of our drug homogeneity method. The clustered image and drug LISA map were simultaneously analyzed to link identified clusters with observed drug distribution profile. Finally, ions selection was performed using the spatially aware method. CONCLUSIONS In this paper, we have shown an approach to correlate the drug distribution with spatial heterogeneity in untargeted MSI data. Our approach is freely available in an R package 'CorrDrugTumorMSI'.
Collapse
Affiliation(s)
- Mridula Prasad
- IMM/ Analytical Chemistry, Radboud University, Heyendaalseweg, 6525 AJ Nijmegen, Netherlands
- Unit of Computational Biology, Research and Innovation Center, Fondazione Edmund Mach, 38010 San Michele all’ Adige, Italy
| | - Geert Postma
- IMM/ Analytical Chemistry, Radboud University, Heyendaalseweg, 6525 AJ Nijmegen, Netherlands
| | - Pietro Franceschi
- Unit of Computational Biology, Research and Innovation Center, Fondazione Edmund Mach, 38010 San Michele all’ Adige, Italy
| | - Lavinia Morosi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19-20156 Milan, Italy
| | - Silvia Giordano
- Mass Spectrometry Laboratory, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19-20156 Milan, Italy
| | - Francesca Falcetta
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19-20156 Milan, Italy
| | - Raffaella Giavazzi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19-20156 Milan, Italy
| | - Enrico Davoli
- Mass Spectrometry Laboratory, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa, 19-20156 Milan, Italy
| | - Lutgarde M C Buydens
- IMM/ Analytical Chemistry, Radboud University, Heyendaalseweg, 6525 AJ Nijmegen, Netherlands
| | - Jeroen Jansen
- IMM/ Analytical Chemistry, Radboud University, Heyendaalseweg, 6525 AJ Nijmegen, Netherlands
| |
Collapse
|
19
|
Cartaxo AL, Almeida J, Gualda EJ, Marsal M, Loza-Alvarez P, Brito C, Isidro IA. A computational diffusion model to study antibody transport within reconstructed tumor microenvironments. BMC Bioinformatics 2020; 21:529. [PMID: 33203360 PMCID: PMC7672975 DOI: 10.1186/s12859-020-03854-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background Antibodies revolutionized cancer treatment over the past decades. Despite their successfully application, there are still challenges to overcome to improve efficacy, such as the heterogeneous distribution of antibodies within tumors. Tumor microenvironment features, such as the distribution of tumor and other cell types and the composition of the extracellular matrix may work together to hinder antibodies from reaching the target tumor cells. To understand these interactions, we propose a framework combining in vitro and in silico models. We took advantage of in vitro cancer models previously developed by our group, consisting of tumor cells and fibroblasts co-cultured in 3D within alginate capsules, for reconstruction of tumor microenvironment features.
Results In this work, an experimental-computational framework of antibody transport within alginate capsules was established, assuming a purely diffusive transport, combined with an exponential saturation effect that mimics the saturation of binding sites on the cell surface. Our tumor microenvironment in vitro models were challenged with a fluorescent antibody and its transport recorded using light sheet fluorescence microscopy. Diffusion and saturation parameters of the computational model were adjusted to reproduce the experimental antibody distribution, with root mean square error under 5%. This computational framework is flexible and can simulate different random distributions of tumor microenvironment elements (fibroblasts, cancer cells and collagen fibers) within the capsule. The random distribution algorithm can be tuned to follow the general patterns observed in the experimental models. Conclusions We present a computational and microscopy framework to track and simulate antibody transport within the tumor microenvironment that complements the previously established in vitro models platform. This framework paves the way to the development of a valuable tool to study the influence of different components of the tumor microenvironment on antibody transport.
Collapse
Affiliation(s)
- Ana Luísa Cartaxo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jaime Almeida
- Departamento de Geologia, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal.,Instituto Dom Luiz, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Emilio J Gualda
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Maria Marsal
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Pablo Loza-Alvarez
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A Isidro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal. .,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
20
|
Wang Y, Zhang J, Ren S, Sun D, Huang HY, Wang H, Jin Y, Li F, Zheng C, Yang L, Deng L, Jiang Z, Jiang T, Han X, Hou S, Guo C, Li F, Gao D, Qin J, Gao D, Chen L, Lin SH, Wong KK, Li C, Hu L, Zhou C, Ji H. Branched-Chain Amino Acid Metabolic Reprogramming Orchestrates Drug Resistance to EGFR Tyrosine Kinase Inhibitors. Cell Rep 2020; 28:512-525.e6. [PMID: 31291585 DOI: 10.1016/j.celrep.2019.06.026] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 04/12/2019] [Accepted: 06/05/2019] [Indexed: 02/05/2023] Open
Abstract
Drug resistance is a significant hindrance to effective cancer treatment. Although resistance mechanisms of epidermal growth factor receptor (EGFR) mutant cancer cells to lethal EGFR tyrosine kinase inhibitors (TKI) treatment have been investigated intensively, how cancer cells orchestrate adaptive response under sublethal drug challenge remains largely unknown. Here, we find that 2-h sublethal TKI treatment elicits a transient drug-tolerant state in EGFR mutant lung cancer cells. Continuous sublethal treatment reinforces this tolerance and eventually establishes long-term TKI resistance. This adaptive process involves H3K9 demethylation-mediated upregulation of branched-chain amino acid aminotransferase 1 (BCAT1) and subsequent metabolic reprogramming, which promotes TKI resistance through attenuating reactive oxygen species (ROS) accumulation. Combination treatment with TKI- and ROS-inducing reagents overcomes this drug resistance in preclinical mouse models. Clinical information analyses support the correlation of BCAT1 expression with the EGFR TKI response. Our findings reveal the importance of BCAT1-engaged metabolism reprogramming in TKI resistance in lung cancer.
Collapse
Affiliation(s)
- Yuetong Wang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengxiang Ren
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Dan Sun
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hsin-Yi Huang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Wang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yujuan Jin
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fuming Li
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Zheng
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liu Yang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Deng
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhonglin Jiang
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Jiang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiangkun Han
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shenda Hou
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenchen Guo
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Li
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Qin
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Cheng Li
- School of Life Sciences, Peking University, Beijing 100871, China.
| | - Liang Hu
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; School of Life Science and Technology, Shanghai Tech University, Shanghai 200120, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
21
|
Gonda K, Negishi H, Takano-Kasuya M, Kitamura N, Furusawa N, Nakano Y, Hamada Y, Tokunaga M, Higuchi H, Tada H, Ishida T. Heterogeneous Drug Efficacy of an Antibody-Drug Conjugate Visualized Using Simultaneous Imaging of Its Delivery and Intracellular Damage in Living Tumor Tissues. Transl Oncol 2020; 13:100764. [PMID: 32403030 PMCID: PMC7218300 DOI: 10.1016/j.tranon.2020.100764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/16/2020] [Indexed: 11/30/2022] Open
Abstract
Anticancer drug efficacy varies because the delivery of drugs within tumors and tumor responses are heterogeneous; however, these features are often more homogenous in vitro. This difference makes it difficult to accurately determine drug efficacy. Therefore, it is important to use living tumor tissues in preclinical trials to observe the heterogeneity in drug distribution and cell characteristics in tumors. In the present study, to accurately evaluate the efficacy of an antibody-drug conjugate (ADC) containing a microtubule inhibitor, we established a cell line that expresses a fusion of end-binding protein 1 and enhanced green fluorescent protein that serves as a microtubule plus-end-tracking protein allowing the visualization of microtubule dynamics. This cell line was xenografted into mice to create a model of living tumor tissue. The tumor cells possessed a greater number of microtubules with plus-ends, a greater number of meandering microtubules, and a slower rate of microtubule polymerization than the in vitro cells. In tumor tissues treated with fluorescent dye-labeled ADCs, heterogeneity was observed in the delivery of the drug to tumor cells, and microtubule dynamics were inhibited in a concentration-dependent manner. Moreover, a difference in drug sensitivity was observed between in vitro cells and tumor cells; compared with in vitro cells, tumor cells were more sensitive to changes in the concentration of the ADC. This study is the first to simultaneously evaluate the delivery and intracellular efficacy of ADCs in living tumor tissue. Accurate evaluation of the efficacy of ADCs is important for the development of effective anticancer drugs.
Collapse
Affiliation(s)
- Kohsuke Gonda
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan.
| | - Hiroshi Negishi
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Mayumi Takano-Kasuya
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Narufumi Kitamura
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Naoko Furusawa
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Yasushi Nakano
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Yoh Hamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| | - Masayuki Tokunaga
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Hideo Higuchi
- Department of Physics, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| |
Collapse
|
22
|
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
|
23
|
Li W, Wang HF, Li ZY, Wang T, Zhao CX. Numerical investigation of drug transport from blood vessels to tumour tissue using a Tumour-Vasculature-on-a-Chip. Chem Eng Sci 2019. [DOI: 10.1016/j.ces.2019.115155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
24
|
Besse HC, Barten-van Rijbroek AD, van der Wurff-Jacobs KMG, Bos C, Moonen CTW, Deckers R. Tumor Drug Distribution after Local Drug Delivery by Hyperthermia, In Vivo. Cancers (Basel) 2019; 11:cancers11101512. [PMID: 31600958 PMCID: PMC6826934 DOI: 10.3390/cancers11101512] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023] Open
Abstract
Tumor drug distribution and concentration are important factors for effective tumor treatment. A promising method to enhance the distribution and the concentration of the drug in the tumor is to encapsulate the drug in a temperature sensitive liposome. The aim of this study was to investigate the tumor drug distribution after treatment with various injected doses of different liposomal formulations of doxorubicin, ThermoDox (temperature sensitive liposomes) and DOXIL (non-temperature sensitive liposomes), and free doxorubicin at macroscopic and microscopic levels. Only ThermoDox treatment was combined with hyperthermia. Experiments were performed in mice bearing a human fibrosarcoma. At low and intermediate doses, the largest growth delay was obtained with ThermoDox, and at the largest dose, the largest growth delay was obtained with DOXIL. On histology, tumor areas with increased doxorubicin concentration correlated with decreased cell proliferation, and substantial variations in doxorubicin heterogeneity were observed. ThermoDox treatment resulted in higher tissue drug levels than DOXIL and free doxorubicin for the same dose. A relation with the distance to the vasculature was shown, but vessel perfusion was not always sufficient to determine doxorubicin delivery. Our results indicate that tumor drug distribution is an important factor for effective tumor treatment and that its dependence on delivery formulation merits further systemic investigation.
Collapse
Affiliation(s)
- Helena C Besse
- Center of Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | | | - Kim M G van der Wurff-Jacobs
- Center of Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Clemens Bos
- Center of Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | - Chrit T W Moonen
- Center of Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Roel Deckers
- Center of Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| |
Collapse
|
25
|
Applications of MALDI mass spectrometry imaging for pharmacokinetic studies during drug development. Drug Metab Pharmacokinet 2019; 34:209-216. [DOI: 10.1016/j.dmpk.2019.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/23/2022]
|
26
|
Precision pharmacology: Mass spectrometry imaging and pharmacokinetic drug resistance. Crit Rev Oncol Hematol 2019; 141:153-162. [PMID: 31302407 DOI: 10.1016/j.critrevonc.2019.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/08/2019] [Accepted: 06/13/2019] [Indexed: 12/27/2022] Open
Abstract
Failure of systemic cancer treatment can be, at least in part, due to the drug not being delivered to the tumour at sufficiently high concentration and/or sufficiently homogeneous distribution; this is termed as "pharmacokinetic drug resistance". To understand whether a drug is being adequately delivered to the tumour, "precision pharmacology" techniques are needed. Mass spectrometry imaging (MSI) is a relatively new and complex technique that allows imaging of drug distribution within tissues. In this review we address the applicability of MSI to the study of cancer drug distribution from the bench to the bedside. We address: (i) the role of MSI in pre-clinical studies to characterize anti-cancer drug distribution within the body and the tumour, (ii) the application of MSI in pre-clinical studies to define optimal drug dose or schedule, combinations or new drug delivery systems, and finally (iii) the emerging role of MSI in clinical research.
Collapse
|
27
|
Jove M, Spencer J, Hubbard M, Holden E, O’Dea R, Brook B, Phillips R, Smye S, Loadman P, Twelves C. Cellular Uptake and Efflux of Palbociclib In Vitro in Single Cell and Spheroid Models. J Pharmacol Exp Ther 2019; 370:242-251. [DOI: 10.1124/jpet.119.256693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
|
28
|
Garattini S, Fuso Nerini I, D'Incalci M. Not only tumor but also therapy heterogeneity. Ann Oncol 2019; 29:13-19. [PMID: 29045538 DOI: 10.1093/annonc/mdx646] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- S Garattini
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - I Fuso Nerini
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - M D'Incalci
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
29
|
Falcetta F, Morosi L, Ubezio P, Giordano S, Decio A, Giavazzi R, Frapolli R, Prasad M, Franceschi P, D'Incalci M, Davoli E. Past-in-the-Future. Peak detection improves targeted mass spectrometry imaging. Anal Chim Acta 2018; 1042:1-10. [PMID: 30428975 DOI: 10.1016/j.aca.2018.06.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/19/2018] [Accepted: 06/24/2018] [Indexed: 11/30/2022]
Abstract
Mass spectrometry imaging is a valuable tool for visualizing the localization of drugs in tissues, a critical issue especially in cancer pharmacology where treatment failure may depend on poor drug distribution within the tumours. Proper preprocessing procedures are mandatory to obtain quantitative data of drug distribution in tumours, even at low intensity, through reliable ion peak identification and integration. We propose a simple preprocessing and quantification pipeline. This pipeline was designed starting from classical peak integration methods, developed when "microcomputers" became available for chromatography, now applied to MSI. This pre-processing approach is based on a novel method using the fixed mass difference between the analyte and its 5 d derivatives to set up a mass range gate. We demonstrate the use of this pipeline for the evaluating the distribution of the anticancer drug paclitaxel in tumour sections. The procedure takes advantage of a simple peak analysis and allows to quantify the drug concentration in each pixel with a limit of detection below 0.1 pmol mm-2 or 10 μg g-1. Quantitative images of paclitaxel distribution in different tumour models were obtained and average paclitaxel concentrations were compared with HPLC measures in the same specimens, showing <20% difference. The scripts are developed in Python and available through GitHub, at github.com/FrancescaFalcetta/Imaging_of_drugs_distribution_and_quantifications.git.
Collapse
Affiliation(s)
- Francesca Falcetta
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Lavinia Morosi
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Paolo Ubezio
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Silvia Giordano
- Mass Spectrometry Laboratory, Department of Environmental Health Sciences, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Alessandra Decio
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Raffaella Giavazzi
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Roberta Frapolli
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Mridula Prasad
- IMM/Analytical Chemistry, Radboud University, Heyendaalseweg, 6525 AJ, Nijmegen, Netherlands; Biostatistics and Data Management Group, Fondazione Edmund Mach, 38010, San Michele all' Adige, Italy; Nanotechnology in Medicinal Chemistry, Department of Molecular Biotechnology and Health Sciences, Università di Torino, 10126, Torino, Italy
| | - Pietro Franceschi
- Biostatistics and Data Management Group, Fondazione Edmund Mach, 38010, San Michele all' Adige, Italy
| | - Maurizio D'Incalci
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy
| | - Enrico Davoli
- Mass Spectrometry Laboratory, Department of Environmental Health Sciences, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa, 19-20156, Milan, Italy.
| |
Collapse
|
30
|
Nishimura M, Hayashi M, Mizutani Y, Takenaka K, Imamura Y, Chayahara N, Toyoda M, Kiyota N, Mukohara T, Aikawa H, Fujiwara Y, Hamada A, Minami H. Distribution of erlotinib in rash and normal skin in cancer patients receiving erlotinib visualized by matrix assisted laser desorption/ionization mass spectrometry imaging. Oncotarget 2018; 9:18540-18547. [PMID: 29719624 PMCID: PMC5915091 DOI: 10.18632/oncotarget.24928] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 03/08/2018] [Indexed: 11/25/2022] Open
Abstract
Background The development of skin rashes is the most common adverse event observed in cancer patients treated with epidermal growth factor receptor-tyrosine kinase inhibitors such as erlotinib. However, the pharmacological evidence has not been fully revealed. Results Erlotinib distribution in the rashes was more heterogeneous than that in the normal skin, and the rashes contained statistically higher concentrations of erlotinib than adjacent normal skin in the superficial skin layer (229 ± 192 vs. 120 ± 103 ions/mm2; P = 0.009 in paired t-test). LC-MS/MS confirmed that the concentration of erlotinib in the skin rashes was higher than that in normal skin in the superficial skin layer (1946 ± 1258 vs. 1174 ± 662 ng/cm3; P = 0.028 in paired t-test). The results of MALDI-MSI and LC-MS/MS were well correlated (coefficient of correlation 0.879, P < 0.0001). Conclusions Focal distribution of erlotinib in the skin tissue was visualized using non-labeled MALDI-MSI. Erlotinib concentration in the superficial layer of the skin rashes was higher than that in the adjacent normal skin. Methods We examined patients with advanced pancreatic cancer who developed skin rashes after treatment with erlotinib and gemcitabine. We biopsied both the rash and adjacent normal skin tissues, and visualized and compared the distribution of erlotinib within the skin using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). The tissue concentration of erlotinib was also measured by liquid chromatography-tandem mass spectrometry (LC–MS/MS) with laser microdissection.
Collapse
Affiliation(s)
- Meiko Nishimura
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Mitsuhiro Hayashi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan.,Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Yu Mizutani
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Kei Takenaka
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshinori Imamura
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Naoko Chayahara
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Present Address: Department of Medical Oncology, Kobe Minimally invasive Cancer Center, Kobe, Hyogo, Japan
| | - Masanori Toyoda
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Naomi Kiyota
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Toru Mukohara
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Cancer Center, Kobe University Hospital, Kobe, Hyogo, Japan
| | - Hiroaki Aikawa
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, National Cancer Center, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan.,Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan.,Department of Medical Oncology and Translational Research, Graduate school of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Cancer Center, Kobe University Hospital, Kobe, Hyogo, Japan
| |
Collapse
|
31
|
Niu Y, Zhu J, Li Y, Shi H, Gong Y, Li R, Huo Q, Ma T, Liu Y. Size shrinkable drug delivery nanosystems and priming the tumor microenvironment for deep intratumoral penetration of nanoparticles. J Control Release 2018; 277:35-47. [PMID: 29545106 DOI: 10.1016/j.jconrel.2018.03.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/11/2018] [Indexed: 02/08/2023]
Abstract
The penetration of nanomedicine into solid tumor still constitutes a great challenge for cancer therapy, which lead to the failure of thorough clearance of tumor cells. Aiming at solving this issue, lots of encouraging progress has been made in the development of multistage nanoparticles triggered by various stimuli in the past few years. Besides, the therapeutical effects of nanoagents are also greatly impacted by the complex tumor microenvironment, and remodeling tumor microenvironment has become another important approach for promoting nanoparticles penetration. In this review, we summarize and analyze recent research progress and challenges in promoting nanoparticle penetration based on two kinds of different strategies, which include size shrinkable nanoparticles and priming tumor microenvironments. Especially, many recent reported multi-strategy approaches based on particle size reduction in conjugated with other therapeutic strategies are discussed. And we expect to provide some useful enlightenments and proposals on nanotechnology-based drug delivery systems for more effective therapy of solid tumors.
Collapse
Affiliation(s)
- Yimin Niu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Yang Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huihui Shi
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yaxiang Gong
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Rui Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Huo
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Tao Ma
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Yang Liu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
32
|
Morosi L, Giordano S, Falcetta F, Frapolli R, Licandro SA, Matteo C, Zucchetti M, Ubezio P, Erba E, Visentin S, D'Incalci M, Davoli E. Application of 3D Mass Spectrometry Imaging to TKIs. Clin Pharmacol Ther 2017; 102:748-751. [DOI: 10.1002/cpt.786] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Lavinia Morosi
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Silvia Giordano
- Department of Environmental Health Sciences, Mass Spectrometry Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Francesca Falcetta
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Roberta Frapolli
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Simonetta A. Licandro
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Cristina Matteo
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Massimo Zucchetti
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Paolo Ubezio
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Eugenio Erba
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Sonja Visentin
- Department of Molecular Biotechnology and Health Science; University of Torino; Torino Italy
| | - Maurizio D'Incalci
- Department of Oncology, Cancer Pharmacology Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| | - Enrico Davoli
- Department of Environmental Health Sciences, Mass Spectrometry Laboratory; IRCCS Istituto di Ricerche Farmacologiche Mario Negri; Milano Italy
| |
Collapse
|
33
|
Evaluation of the heterogeneous tissue distribution of erlotinib in lung cancer using matrix-assisted laser desorption ionization mass spectrometry imaging. Sci Rep 2017; 7:12622. [PMID: 28974758 PMCID: PMC5626687 DOI: 10.1038/s41598-017-13025-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/12/2017] [Indexed: 12/22/2022] Open
Abstract
Although drug distribution in tumor tissues has a significant impact on efficacy, conventional pharmacokinetic analysis has some limitations with regard to its ability to provide a comprehensive assessment of drug tissue distribution. Erlotinib is a tyrosine kinase inhibitor that acts on the epidermal growth factor receptor; however, it is unclear how this drug is histologically distributed in lung cancer. We used matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) to analyze erlotinib distribution in the tumor and normal lung tissues of a mouse xenograft model and patient with non-small cell lung cancer. LC-MS/MS showed that the erlotinib tissue concentration in the xenograft tumor tissue was clearly lower than that in the normal tissue at the time of maximum blood concentration. MALDI-MSI showed the heterogeneous distribution of erlotinib at various levels in the murine tissues; interestingly, erlotinib was predominantly localized in the area of viable tumor compared to the necrotic area. In the patient-derived tissue, MALDI-MSI showed that there were different concentrations of erlotinib distributed within the same tissue. For drug development and translational research, the imaging pharmacokinetic study used the combination of MALDI-MSI and LC-MS/MS analyses may be useful in tissues with heterogeneous drug distribution.
Collapse
|
34
|
Galiè M, Boschi F, Scambi I, Merigo F, Marzola P, Altabella L, Lavagnolo U, Sbarbati A, Spinelli AE. Theranostic Role of 32P-ATP as Radiopharmaceutical for the Induction of Massive Cell Death within Avascular Tumor Core. Theranostics 2017; 7:4399-4409. [PMID: 29158835 PMCID: PMC5695139 DOI: 10.7150/thno.21403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 01/04/2023] Open
Abstract
Drug inaccessibility to vast areas of the tumor parenchyma is amongst the major hurdles for conventional therapies. Treatment efficacy rapidly decreases with distance from vessels and most of the tumor cells survive therapy. Also, between subsequent cycles of treatment, spared cancer cells replace those killed near the vessels, improving their access to nutrients, boosting their proliferation rate, and thus enabling tumor repopulation. Because of their property of "acting at a distance," radioisotopes are believed to overcome the physical barrier of vascular inaccessibility. Methods A novel molecular imaging tool called Cerenkov Luminescence Imaging (CLI) was employed for the detection of Cerenkov radiation emitted by beta particles, allowing in vivo tracking of beta-emitters. More precisely we investigated using a xenograft model of colon carcinoma the potential use of 32P-ATP as a novel theranostic radiopharmaceutical for tracing tumor lesions while simultaneously hampering their growth. Results Our analyses demonstrated that 32P-ATP injected into tumor-bearing mice reaches tumor lesions and persists for days and weeks within the tumor parenchyma. Also, the high-penetrating beta particles of 32P-ATP exert a "cross-fire" effect that induces massive cell death throughout the entire tumor parenchyma including core regions. Conclusion Our findings suggest 32P-ATP treatment as a potential approach to complement conventional therapies that fail to reach the tumor core and to prevent tumor repopulation.
Collapse
|
35
|
Villela-Ma LM, Velez-Ayal AK, Lopez-Sanc RDC, Martinez-C JA, Hernandez- JA. Advantages of Drug Selective Distribution in Cancer Treatment: Brentuximab Vedotin. INT J PHARMACOL 2017. [DOI: 10.3923/ijp.2017.785.807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Hubbard ME, Jove M, Loadman PM, Phillips RM, Twelves CJ, Smye SW. Drug delivery in a tumour cord model: a computational simulation. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170014. [PMID: 28573005 PMCID: PMC5451806 DOI: 10.1098/rsos.170014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/25/2017] [Indexed: 05/17/2023]
Abstract
The tumour vasculature and microenvironment is complex and heterogeneous, contributing to reduced delivery of cancer drugs to the tumour. We have developed an in silico model of drug transport in a tumour cord to explore the effect of different drug regimes over a 72 h period and how changes in pharmacokinetic parameters affect tumour exposure to the cytotoxic drug doxorubicin. We used the model to describe the radial and axial distribution of drug in the tumour cord as a function of changes in the transport rate across the cell membrane, blood vessel and intercellular permeability, flow rate, and the binding and unbinding ratio of drug within the cancer cells. We explored how changes in these parameters may affect cellular exposure to drug. The model demonstrates the extent to which distance from the supplying vessel influences drug levels and the effect of dosing schedule in relation to saturation of drug-binding sites. It also shows the likely impact on drug distribution of the aberrant vasculature seen within tumours. The model can be adapted for other drugs and extended to include other parameters. The analysis confirms that computational models can play a role in understanding novel cancer therapies to optimize drug administration and delivery.
Collapse
Affiliation(s)
- M. E. Hubbard
- School of Mathematical Sciences, The University of Nottingham, University Park, Nottingham NG7 2RD, UK
- Author for correspondence: M. E. Hubbard e-mail:
| | - M. Jove
- Department of Medical Oncology, Leeds Teaching Hospitals NHS Trust, University of Leeds, St James’s University Hospital, Leeds LS9 7TF, UK
| | - P. M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| | - R. M. Phillips
- School of Applied Sciences, University of Hudderfield, Queensgate, Huddersfield HD1 3DH, UK
| | - C. J. Twelves
- Leeds Institute of Cancer and Pathology, University of Leeds, St James’s University Hospital, Leeds LS9 7TF, UK
| | - S. W. Smye
- Academic Division of Medical Physics, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
37
|
A Nanostructured Matrices Assessment to Study Drug Distribution in Solid Tumor Tissues by Mass Spectrometry Imaging. NANOMATERIALS 2017; 7:nano7030071. [PMID: 28336905 PMCID: PMC5388173 DOI: 10.3390/nano7030071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 11/17/2022]
Abstract
The imaging of drugs inside tissues is pivotal in oncology to assess whether a drug reaches all cells in an adequate enough concentration to eradicate the tumor. Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging (MALDI-MSI) is one of the most promising imaging techniques that enables the simultaneous visualization of multiple compounds inside tissues. The choice of a suitable matrix constitutes a critical aspect during the development of a MALDI-MSI protocol since the matrix ionization efficiency changes depending on the analyte structure and its physico-chemical properties. The objective of this study is the improvement of the MALDI-MSI technique in the field of pharmacology; developing specifically designed nanostructured surfaces that allow the imaging of different drugs with high sensitivity and reproducibility. Among several nanomaterials, we tested the behavior of gold and titanium nanoparticles, and halloysites and carbon nanotubes as possible matrices. All nanomaterials were firstly screened by co-spotting them with drugs on a MALDI plate, evaluating the drug signal intensity and the signal-to-noise ratio. The best performing matrices were tested on control tumor slices, and were spotted with drugs to check the ion suppression effect of the biological matrix. Finally; the best nanomaterials were employed in a preliminary drug distribution study inside tumors from treated mice.
Collapse
|
38
|
Yan LZ, Dressler EV, Adams VR. Association of hypertension and treatment outcomes in advanced stage non-small cell lung cancer patients treated with bevacizumab or non-bevacizumab containing regimens. J Oncol Pharm Pract 2017; 24:209-217. [DOI: 10.1177/1078155217690921] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Studies suggest that bevacizumab-induced hypertension is prognostic of better outcomes in bevacizumab-treated patients with metastatic colorectal, HER2-negative breast, kidney, and pancreatic cancer. Few have examined this correlation in metastatic non-small cell lung cancer and evaluated whether hypertension independent of bevacizumab can improve the treatment outcomes. Objectives The primary objective was to determine the effect of hypertension on the overall response of advanced non-small cell lung cancer patients from start of the first-line chemotherapy to maintenance therapy. Secondary objectives include the effect of hypertension on the overall survival in all patients and on the overall response in bevacizumab-treated patients. Methods A retrospective chart review for a single institution was conducted from 2008 to 2013 on all patients with advanced non-squamous non-small cell lung cancer who received ≥ 1 cycle of combination chemotherapy. Patients were divided into hypertension versus no hypertension and into bevacizumab versus non-bevacizumab groups. Results Of the 188 advanced non-small cell lung cancer patients evaluated, 62 were treated with bevacizumab-containing regimens. The mean age at diagnosis was 58 years in both the groups. Hypertension independent of bevacizumab did not lead to improved treatment outcomes. However, in the bevacizumab subgroup, hypertensive patients had significantly higher response rates versus non-hypertensive patients (36.7% vs. 12.5%; p = 0.02). There was no significant difference in the overall survival between hypertensive versus non-hypertensive patients. Conclusion While hypertension alone did not significantly improve the treatment outcomes, hypertension in bevacizumab-treated patients with metastatic non-small cell lung cancer led to significantly improved responses. Further prospective studies are needed to confirm the association of hypertension with improved treatment outcomes in metastatic NSCLC.
Collapse
Affiliation(s)
- Lily Z Yan
- Department of Pharmacy, University of Kentucky Medical Center, Lexington, KY, USA
| | - Emily V Dressler
- Division of Cancer Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Val R Adams
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
| |
Collapse
|
39
|
Colmegna B, Morosi L, D'Incalci M. Molecular and Pharmacological Mechanisms of Drug Resistance:An Evolving Paradigm. Handb Exp Pharmacol 2017; 249:1-12. [PMID: 28332049 DOI: 10.1007/164_2017_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The high heterogeneity and genomic instability of malignant tumors explains why even responsive tumors contain cell clones that are resistant for many possible mechanisms involving intracellular drug inactivation, low uptake or high efflux of anticancer drugs from cancer cells, qualitative or quantitative changes in the drug target. Many tumors, however, are resistant because of insufficient exposure to anticancer drugs, due to pharmacokinetic reasons and inefficient and heterogeneous tumor drug distribution, related to a deficient vascularization and high interstitial pressure. Finally, resistance can be related to the activation of anti-apoptotic and cell survival pathways by cancer cells and often enhanced by tumor microenvironment.
Collapse
Affiliation(s)
- Benedetta Colmegna
- Department of Oncology, IRCCS 'Mario Negri', Institute for Pharmacological Research, 20145, Milan, Italy
| | - Lavinia Morosi
- Department of Oncology, IRCCS 'Mario Negri', Institute for Pharmacological Research, 20145, Milan, Italy
| | - Maurizio D'Incalci
- Department of Oncology, IRCCS 'Mario Negri', Institute for Pharmacological Research, 20145, Milan, Italy.
| |
Collapse
|
40
|
Pharmacokinetic/pharmacodynamic modeling of etoposide tumor growth inhibitory effect in Walker-256 tumor-bearing rat model using free intratumoral drug concentrations. Eur J Pharm Sci 2017; 97:70-78. [DOI: 10.1016/j.ejps.2016.10.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/20/2016] [Accepted: 10/30/2016] [Indexed: 11/17/2022]
|
41
|
Heterogeneity of paclitaxel distribution in different tumor models assessed by MALDI mass spectrometry imaging. Sci Rep 2016; 6:39284. [PMID: 28000726 PMCID: PMC5175283 DOI: 10.1038/srep39284] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/22/2016] [Indexed: 11/09/2022] Open
Abstract
The penetration of anticancer drugs in solid tumors is important to ensure the therapeutic effect, so methods are needed to understand drug distribution in different parts of the tumor. Mass spectrometry imaging (MSI) has great potential in this field to visualize drug distribution in organs and tumor tissues with good spatial resolution and superior specificity. We present an accurate and reproducible imaging method to investigate the variation of drug distribution in different parts of solid tumors. The method was applied to study the distribution of paclitaxel in three ovarian cancer models with different histopathological characteristics and in colon cancer (HCT116), breast cancer (MDA-MB-231) and malignant pleural mesothelioma (MPM487). The heterogeneous drug penetration in the tumors is evident from the MALDI imaging results and from the images analysis. The differences between the various models do not always relate to significant changes in drug content in tumor homogenate examined by classical HPLC analysis. The specificity of the method clarifies the heterogeneity of the drug distribution that is analyzed from a quantitative point of view too, highlighting how marked are the variations of paclitaxel amounts in different part of solid tumors.
Collapse
|
42
|
3D Mass Spectrometry Imaging Reveals a Very Heterogeneous Drug Distribution in Tumors. Sci Rep 2016; 6:37027. [PMID: 27841316 PMCID: PMC5107992 DOI: 10.1038/srep37027] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/24/2016] [Indexed: 01/26/2023] Open
Abstract
Mass Spectrometry Imaging (MSI) is a widespread technique used to qualitatively describe in two dimensions the distribution of endogenous or exogenous compounds within tissue sections. Absolute quantification of drugs using MSI is a recent challenge that just in the last years has started to be addressed. Starting from a two dimensional MSI protocol, we developed a three-dimensional pipeline to study drug penetration in tumors and to develop a new drug quantification method by MALDI MSI. Paclitaxel distribution and concentration in different tumors were measured in a 3D model of Malignant Pleural Mesothelioma (MPM), which is known to be a very heterogeneous neoplasm, highly resistant to different drugs. The 3D computational reconstruction allows an accurate description of tumor PTX penetration, adding information about the heterogeneity of tumor drug distribution due to the complex microenvironment. The use of an internal standard, homogenously sprayed on tissue slices, ensures quantitative results that are similar to those obtained using HPLC. The 3D model gives important information about the drug concentration in different tumor sub-volumes and shows that the great part of each tumor is not reached by the drug, suggesting the concept of pseudo-resistance as a further explanation for ineffective therapies and tumors relapse.
Collapse
|
43
|
Pickard AJ, Sohn ASW, Bartenstein TF, He S, Zhang Y, Gallo JM. Intracerebral Distribution of the Oncometabolite d-2-Hydroxyglutarate in Mice Bearing Mutant Isocitrate Dehydrogenase Brain Tumors: Implications for Tumorigenesis. Front Oncol 2016; 6:211. [PMID: 27781195 PMCID: PMC5057413 DOI: 10.3389/fonc.2016.00211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
The prevalence of mutant isocitrate dehydrogenase 1 (IDH1) brain tumors has generated significant efforts to understand the role of the mutated enzyme product d-2-hydroxyglutarate (D2HG), an oncometabolite, in tumorigenesis, as well as means to eliminate it. Glymphatic clearance was proposed as a pathway that could be manipulated to accelerate D2HG clearance and dictated the study design that consisted of two cohorts of mice bearing U87/mutant IDH1 intracerebral tumors that underwent two microdialysis - providing D2HG interstitial fluid concentrations - sampling periods of awake and asleep (activate glymphatic clearance) in a crossover manner. Glymphatic clearance was found not to have a significant effect on D2HG brain tumor interstitial fluid concentrations that were 126.9 ± 74.8 μM awake and 117.6 ± 98.6 μM asleep. These concentrations, although low relative to total brain tumor concentrations of 6.8 ± 3.6 mM, were considered sufficient to be transported by interstitial fluid and taken up into normal cells to cause deleterious effects. A model of D2HG CNS distribution supported this contention and was further supported by in vitro studies that showed D2HG could interfere with immune cell function. The study provides insight into the compartmental distribution of D2HG in the brain, wherein the interstitial fluid serves as a dynamic pathway for D2HG to enter normal cells and contribute to tumorigenesis.
Collapse
Affiliation(s)
- Amanda J Pickard
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Albert S W Sohn
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Thomas F Bartenstein
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Shan He
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA; Department of Microbiology and Immunology, Temple University, Philadelphia, PA, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA; Department of Microbiology and Immunology, Temple University, Philadelphia, PA, USA
| | - James M Gallo
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
44
|
Vázquez R, Licandro SA, Astorgues-Xerri L, Lettera E, Panini N, Romano M, Erba E, Ubezio P, Bello E, Libener R, Orecchia S, Grosso F, Riveiro ME, Cvitkovic E, Bekradda M, D'Incalci M, Frapolli R. Promising in vivo efficacy of the BET bromodomain inhibitor OTX015/MK-8628 in malignant pleural mesothelioma xenografts. Int J Cancer 2016; 140:197-207. [PMID: 27594045 DOI: 10.1002/ijc.30412] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/25/2016] [Accepted: 08/15/2016] [Indexed: 11/06/2022]
Abstract
It has recently been reported that a large proportion of human malignant pleural mesothelioma (MPM) cell lines and patient tissue samples present high expression of the c-MYC oncogene. This gene drives several tumorigenic processes and is overexpressed in many cancers. Although c-MYC is a strategic target to restrain cancer processes, no drugs acting as c-MYC inhibitors are available. The novel thienotriazolodiazepine small-molecule bromodomain inhibitor OTX015/MK-8628 has shown potent antiproliferative activity accompanied by c-MYC downregulation in several tumor types. This study was designed to evaluate the growth inhibitory effect of OTX015 on patient-derived MPM473, MPM487 and MPM60 mesothelioma cell lines and its antitumor activity in three patient-derived xenograft models, MPM473, MPM487 and MPM484, comparing it with cisplatin, gemcitabine and pemetrexed, three agents which are currently used to treat MPM in the clinic. OTX015 caused a significant delay in cell growth both in vitro and in vivo. It was the most effective drug in MPM473 xenografts and showed a similar level of activity as the most efficient treatment in the other two MPM models (gemcitabine in MPM487 and cisplatin in MPM484). In vitro studies showed that OTX015 downregulated c-MYC protein levels in both MPM473 and MPM487 cell lines. Our findings represent the first evidence of promising therapeutic activity of OTX015 in mesothelioma.
Collapse
Affiliation(s)
- Ramiro Vázquez
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Simonetta Andrea Licandro
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | - Emanuele Lettera
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Nicolò Panini
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Michela Romano
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eugenio Erba
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Paolo Ubezio
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Ezia Bello
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Roberta Libener
- Cytogenetic and Molecular Pathology Laboratory, Pathology Unit, Oncology, SS Antonio e Biagio e Cesare Arrigo General Hospital, Alessandria, Italy
| | - Sara Orecchia
- Cytogenetic and Molecular Pathology Laboratory, Pathology Unit, Oncology, SS Antonio e Biagio e Cesare Arrigo General Hospital, Alessandria, Italy
| | - Federica Grosso
- Cytogenetic and Molecular Pathology Laboratory, Pathology Unit, Oncology, SS Antonio e Biagio e Cesare Arrigo General Hospital, Alessandria, Italy
| | | | - Esteban Cvitkovic
- Oncology Therapeutic Development, Clichy, France.,Oncoethix SA (now Oncoethix GmbH, a wholly owned subsidiary of Merck Sharp & Dohme Corp), Lucerne, Switzerland
| | | | - Maurizio D'Incalci
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Roberta Frapolli
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
45
|
Quintela-Fandino M, Lluch A, Manso L, Calvo I, Cortes J, García-Saenz JA, Gil-Gil M, Martinez-Jánez N, Gonzalez-Martin A, Adrover E, de Andres R, Viñas G, Llombart-Cussac A, Alba E, Guerra J, Bermejo B, Zamora E, Moreno-Anton F, Pernas Simon S, Carrato A, Lopez-Alonso A, Escudero MJ, Campo R, Carrasco E, Palacios J, Mulero F, Colomer R. 18F-fluoromisonidazole PET and Activity of Neoadjuvant Nintedanib in Early HER2-Negative Breast Cancer: A Window-of-Opportunity Randomized Trial. Clin Cancer Res 2016; 23:1432-1441. [DOI: 10.1158/1078-0432.ccr-16-0738] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022]
|
46
|
de Morrée E, van Soest R, Aghai A, de Ridder C, de Bruijn P, Ghobadi Moghaddam-Helmantel I, Burger H, Mathijssen R, Wiemer E, de Wit R, van Weerden W. Understanding taxanes in prostate cancer; importance of intratumoral drug accumulation. Prostate 2016; 76:927-36. [PMID: 26997363 DOI: 10.1002/pros.23182] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/04/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Resistance to docetaxel is common in metastatic castration-resistant prostate cancer (mCRPC) and may be caused by sub-therapeutic intratumoral drug concentrations. Cabazitaxel demonstrated survival benefit in docetaxel-pretreated and docetaxel-refractory patients. In this study, we investigated whether the superior antitumor activity of cabazitaxel in mCRPC is explained by higher intratumoral cabazitaxel levels. Since recent studies suggest a reduced efficacy of docetaxel following treatment with novel androgen receptor (AR)-targeted agents, we also investigated taxane efficacy in an enzalutamide-resistant tumor model. METHODS Intratumoral concentrations of docetaxel and cabazitaxel were correlated with antitumor activity in docetaxel-naïve, docetaxel-resistant, and enzalutamide-resistant patient-derived xenografts (PDXs) of prostate cancer. RESULTS Intratumoral drug levels were negatively related to intrinsic and acquired resistance to docetaxel. Also, the observed stronger antitumor activity of cabazitaxel was associated with increased cumulative exposure and higher intratumoral of cabazitaxel concentrations in all PDXs. CONCLUSIONS The superior antitumor activity of cabazitaxel in docetaxel- and enzalutamide-resistant tumors can be partly attributed to higher intratumoral drug concentrations. Especially for patients who are intrinsically resistant to docetaxel resulting from suboptimal intratumoral docetaxel concentrations, cabazitaxel may be the preferred chemotherapeutic agent. Prostate 76:927-936, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ellen de Morrée
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert van Soest
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ashraf Aghai
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Corrina de Ridder
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Herman Burger
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Erik Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wytske van Weerden
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
47
|
Fuso Nerini I, Cesca M, Bizzaro F, Giavazzi R. Combination therapy in cancer: effects of angiogenesis inhibitors on drug pharmacokinetics and pharmacodynamics. CHINESE JOURNAL OF CANCER 2016; 35:61. [PMID: 27357621 PMCID: PMC4928348 DOI: 10.1186/s40880-016-0123-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
Validated preclinical studies have provided evidence that anti-vascular endothelial growth factor (VEGF) compounds enhance the activity of subsequent antitumor therapy, but the mechanism of this potentiation is far from clear. The most widespread explanation is enhanced delivery of therapeutics due to vascular remodeling, lower interstitial pressure, and increased blood flow. While the antiangiogenic effects on vascular morphology have been fairly consistent in both preclinical and clinical settings, the improvement of tumor vessel function is debated. This review focuses on the effect of anti-VEGF therapy on tumor microenvironment morphology and functions, and its therapeutic benefits when combined with other therapies. The uptake and spatial distribution of chemotherapeutic agents into the tumor after anti-VEGF are examined.
Collapse
Affiliation(s)
- Ilaria Fuso Nerini
- Department of Oncology, IRCCS-Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156, Milan, Italy
| | - Marta Cesca
- Department of Oncology, IRCCS-Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156, Milan, Italy
| | - Francesca Bizzaro
- Department of Oncology, IRCCS-Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156, Milan, Italy
| | - Raffaella Giavazzi
- Department of Oncology, IRCCS-Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156, Milan, Italy.
| |
Collapse
|
48
|
Aikawa H, Hayashi M, Ryu S, Yamashita M, Ohtsuka N, Nishidate M, Fujiwara Y, Hamada A. Visualizing spatial distribution of alectinib in murine brain using quantitative mass spectrometry imaging. Sci Rep 2016; 6:23749. [PMID: 27026287 PMCID: PMC4812395 DOI: 10.1038/srep23749] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/14/2016] [Indexed: 01/08/2023] Open
Abstract
In the development of anticancer drugs, drug concentration measurements in the target tissue have been thought to be crucial for predicting drug efficacy and safety. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is commonly used for determination of average drug concentrations; however, complete loss of spatial information in the target tissue occurs. Mass spectrometry imaging (MSI) has been recently applied as an innovative tool for detection of molecular distribution of pharmacological agents in heterogeneous targets. This study examined the intra-brain transitivity of alectinib, a novel anaplastic lymphoma kinase inhibitor, using a combination of matrix-assisted laser desorption ionization–MSI and LC-MS/MS techniques. We first analyzed the pharmacokinetic profiles in FVB mice and then examined the effect of the multidrug resistance protein-1 (MDR1) using Mdr1a/b knockout mice including quantitative distribution of alectinib in the brain. While no differences were observed between the mice for the plasma alectinib concentrations, diffuse alectinib distributions were found in the brain of the Mdr1a/b knockout versus FVB mice. These results indicate the potential for using quantitative MSI for clarifying drug distribution in the brain on a microscopic level, in addition to suggesting a possible use in designing studies for anticancer drug development and translational research.
Collapse
Affiliation(s)
- Hiroaki Aikawa
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Mitsuhiro Hayashi
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Department of Molecular Imaging and Pharmacokinetics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shoraku Ryu
- Department of Molecular Imaging and Pharmacokinetics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Makiko Yamashita
- Department of Molecular Imaging and Pharmacokinetics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Naoto Ohtsuka
- Shimadzu Techno-Research Inc., 3-19-2, Minamirokugo, Ohta-ku, Tokyo 144-0045, Japan
| | - Masanobu Nishidate
- Department of Molecular Imaging and Pharmacokinetics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Translational Clinical Research Science &Strategy Dept., Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan.,Department of Medical Oncology and Translational Research, Graduate school of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yasuhiro Fujiwara
- Strategic Planning Bureau, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Akinobu Hamada
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Department of Molecular Imaging and Pharmacokinetics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Department of Medical Oncology and Translational Research, Graduate school of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| |
Collapse
|
49
|
Chen CW, Wu MH, Chen YF, Yen TY, Lin YW, Chao SH, Tala S, Tsai TH, Su TL, Lee TC. A Potent Derivative of Indolizino[6,7-b]Indole for Treatment of Human Non-Small Cell Lung Cancer Cells. Neoplasia 2016; 18:199-212. [PMID: 27108383 PMCID: PMC4840272 DOI: 10.1016/j.neo.2016.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 12/18/2022] Open
Abstract
The therapeutic effect in non–small cell lung cancer (NSCLC) patients is limited because of intrinsic and acquired resistance. Thus, an unmet need exists for the development of new drugs to improve the therapeutic efficacy in NSCLC patients. In this study, the novel small molecule indolizino[6,7-b]indole derivative BO-1978 was selected to evaluate its therapeutic effects on NSCLC and its preclinical toxicity in animal models. An in vitro cytotoxicity assay revealed that BO-1978 significantly suppressed the growth of various NSCLC cell lines with or without mutations in epidermal growth factor receptor (EGFR). Mechanistically, we demonstrated that BO-1978 exhibited multiple modes of action, including inhibition of topoisomerase I/II and induction of DNA cross-linking. Treatment of NSCLC cells with BO-1978 caused DNA damage, disturbed cell cycle progression, and triggered apoptotic cell death. Furthermore, BO-1978 significantly suppressed the growth of EGFR wild-type and mutant NSCLC tumors in xenograft tumor and orthotopic lung tumor models with negligible body weight loss. The combination of BO-1978 with gefitinib further suppressed EGFR mutant NSCLC cell growth in xenograft tumor and orthotopic lung tumor models. Preclinical toxicity studies showed that BO-1978 administration did not cause apparent toxicity in mice. Based on its significant therapeutic efficacy and low drug toxicity, BO-1978 is a potential therapeutic agent for treatment of NSCLC.
Collapse
Affiliation(s)
- Chi-Wei Chen
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Ming-Hsi Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Fan Chen
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tsai-Yi Yen
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yi-Wen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Shu-Hsin Chao
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Satishkumar Tala
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Tsann-Long Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Te-Chang Lee
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
| |
Collapse
|
50
|
Nakamura H, Abu Lila AS, Nishio M, Tanaka M, Ando H, Kiwada H, Ishida T. Intra-tumor distribution of PEGylated liposome upon repeated injection: No possession by prior dose. J Control Release 2015; 220:406-413. [PMID: 26548975 DOI: 10.1016/j.jconrel.2015.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/12/2015] [Accepted: 11/04/2015] [Indexed: 01/26/2023]
Abstract
Liposomes have proven to be a viable means for the delivery of chemotherapeutic agents to solid tumors. However, significant variability has been detected in their intra-tumor accumulation and distribution, resulting in compromised therapeutic outcomes. We recently examined the intra-tumor accumulation and distribution of weekly sequentially administered oxaliplatin (l-OHP)-containing PEGylated liposomes. In that study, the first and second doses of l-OHP-containing PEGylated liposomes were distributed diversely and broadly within tumor tissues, resulting in a potent anti-tumor efficacy. However, little is known about the mechanism underlying such a diverse and broad liposome distribution. Therefore, in the present study, we investigated the influence of dosage interval on the intra-tumor accumulation and distribution of "empty" PEGylated liposomes. Intra-tumor distribution of sequentially administered "empty" PEGylated liposomes was altered in a dosing interval-dependent manner. In addition, the intra-tumor distribution pattern was closely related to the chronological alteration of tumor blood flow as well as vascular permeability in the growing tumor tissue. These results suggest that the sequential administrations of PEGylated liposomes in well-spaced intervals might allow the distribution to different areas and enhance the total bulk accumulation within tumor tissue, resulting in better therapeutic efficacy of the encapsulated payload. This study may provide useful information for a better design of therapeutic regimens involving multiple administrations of nanocarrier drug delivery systems.
Collapse
Affiliation(s)
- Hiroyuki Nakamura
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Amr S Abu Lila
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Miho Nishio
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Masao Tanaka
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hiroshi Kiwada
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan.
| |
Collapse
|