1
|
Chen P, Li S, Nagaoka K, Kakimi K, Kataoka K, Cabral H. Nanoenabled IL-15 Superagonist via Conditionally Stabilized Protein-Protein Interactions Eradicates Solid Tumors by Precise Immunomodulation. J Am Chem Soc 2024. [PMID: 39356776 DOI: 10.1021/jacs.4c08327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Protein complexes are crucial structures that control many biological processes. Harnessing these structures could be valuable for therapeutic therapy. However, their instability and short lifespans need to be addressed for effective use. Here, we propose an innovative approach based on a functional polymeric cloak that coordinately anchors different domains of protein complexes and assembles them into a stabilized nanoformulation. As the polymer-protein association in the cloak is pH sensitive, the nanoformulation also allows targeting the release of the protein complexes to the acidic microenvironment of tumors for aiding their therapeutic performance. Building on this strategy, we developed an IL-15 nanosuperagonist (Nano-SA) by encapsulating the interleukin-15 (IL-15)/IL-15 Receptor α (IL-15Rα) complex (IL-15cx) for fostering synergistic transpresentation in tumors. Upon intravenous administration, Nano-SA stably circulated in the bloodstream, safeguarding the integrity of IL-15cx until reaching the tumor site, where it selectively released the active complex. Thus, Nano-SA significantly amplified the antitumor immune signals while diminishing systemic off-target effects. In murine colon cancer models, Nano-SA achieved potent immunotherapeutic effects, eradicating tumors without adverse side effects. These findings highlight the transformative potential of nanotechnology for advancing protein complex-based therapies.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shangwei Li
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Koji Nagaoka
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
2
|
Kato Y, Mawatari K. Clinical significance of acidic extracellular microenvironment modulated genes. Front Oncol 2024; 14:1380679. [PMID: 39372863 PMCID: PMC11449683 DOI: 10.3389/fonc.2024.1380679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/21/2024] [Indexed: 10/08/2024] Open
Abstract
Background The extracellular pH (pH e ) is known to be acidic. We investigated the effect of mild (pH e 6.8) and severe (pH e 5.9) acidosis on gene expression in mouse B16-BL6 melanoma cells using cDNA microarray analysis and compared them with the acidic pH e dependence of human tumors. Methods B16-BL6 cells were treated with pH e 7.4 (control), pH e 6.8, and pH e 5.9. The mRNA expression was analyzed by using the cDNA microarray. Heat map, volcano plot, and gene ontology enrichment analysis were performed. The data were compared with the gene signatures of published data GSE52031 and GSE8401 and compared with the pathological staging by GEPIA2, and the prognostic signature of proteins was searched by the Human Protein Atlas database. If the acidic pH e -induced and -reduced genes were correlated with shortened and prolonged survival times, respectively, and also correlated with pathological staging, we defined it as "hit" and counted the sum of hit points of eight types of tumors such as breast, colorectal, prostate, gastric, liver, prostate, lung, and head and neck and melanoma. Results Gene expression was differentially and commonly regulated by both pH e s. The number of genes upregulated fourfold or more at pH e 6.8 and 5.9 only for 25 and 131 genes, respectively, and 85 genes were common. The number of genes downregulated fourfold or less at pH e 6.8 and 5.9 only for 63 and 82 genes, respectively, and 118 genes were common. Compared with human mRNA expression data (GSE8401), there is no correlation with the overall pattern of the signature. In seven types of cancer (breast, colorectal, gastric, liver, prostate, lung, and head and neck) and melanoma, the relationship between acidic pH e -modulated gene expression and overall survival was evaluated. As a result, acidic pH e dependency contributing to prognosis was higher in colorectal, lung, and head and neck cancers and lower in prostate cancer. Conclusion Tumor classification based on response to extracellular acidic pH e will provide new insights into chemotherapy strategy for patients with tumors.
Collapse
Affiliation(s)
- Yasumasa Kato
- Department of Oral Function and Molecular Biology, Ohu University School of
Dentistry, Koriyama, Japan
| | | |
Collapse
|
3
|
Mu C, Liu X, Riselli A, Slater J, Escobar E, Dang D, Drapeau S, Delos Santos R, Andosca S, Nguyen H, Larson PEZ, Bok R, Vigneron DB, Kurhanewicz J, Wilson DM, Flavell RR. Protocol for producing hyperpolarized 13C-bicarbonate for clinical MRI of extracellular pH in aggressive tumors. STAR Protoc 2024; 5:103091. [PMID: 38943645 PMCID: PMC11261128 DOI: 10.1016/j.xpro.2024.103091] [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: 02/15/2024] [Revised: 03/29/2024] [Accepted: 05/06/2024] [Indexed: 07/01/2024] Open
Abstract
Tumor acidosis is one of the hallmarks indicating the initiation and progression of various cancers. Here, we present a protocol for preparing a hyperpolarized (HP) 13C-bicarbonate tissue pH MRI imaging contrast agent to detect aggressive tumors. We describe the steps for the formulation and polarization of a precursor molecule 13C-glycerol carbonate (13C-GLC), the post-dissolution reaction, and converting HP 13C-GLC to an injectable HP 13C-bicarbonate solution. We then detail procedures for MRI data acquisition to generate tumor pH maps for assessing tumor aggressiveness. For complete details on the use and execution of this protocol, please refer to Mu et al.1.
Collapse
Affiliation(s)
- Changhua Mu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Xiaoxi Liu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andrew Riselli
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - James Slater
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Evelyn Escobar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Duy Dang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Scott Drapeau
- Makers Lab, Library, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Romelyn Delos Santos
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stacy Andosca
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hao Nguyen
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
4
|
Chen S, Zhang L, Li S, Yuan Y, Jiang B, Jiang Z, Zhang X, Zhou X, Liu M. Detecting biomarkers by dynamic nuclear polarization enhanced magnetic resonance. Natl Sci Rev 2024; 11:nwae228. [PMID: 39144741 PMCID: PMC11321254 DOI: 10.1093/nsr/nwae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/26/2024] [Accepted: 06/16/2024] [Indexed: 08/16/2024] Open
Abstract
Hyperpolarization stands out as a technique capable of significantly enhancing the sensitivity of nuclear magnetic resonance (NMR) and magnetic resonance imaging (MRI). Dynamic nuclear polarization (DNP), among various hyperpolarization methods, has gained prominence for its efficacy in real-time monitoring of metabolism and physiology. By administering a hyperpolarized substrate through dissolution DNP (dDNP), the biodistribution and metabolic changes of the DNP agent can be visualized spatiotemporally. This approach proves to be a distinctive and invaluable tool for non-invasively studying cellular metabolism in vivo, particularly in animal models. Biomarkers play a pivotal role in influencing the growth and metastasis of tumor cells by closely interacting with them, and accordingly detecting pathological alterations of these biomarkers is crucial for disease diagnosis and therapy. In recent years, a range of hyperpolarized DNP molecular bioresponsive agents utilizing various nuclei, such as 13C, 15N, 31P, 89Y, etc., have been developed. In this context, we explore how these magnetic resonance signals of nuclear spins enhanced by DNP respond to biomarkers, including pH, metal ions, enzymes, or redox processes. This review aims to offer insights into the design principles of responsive DNP agents, target selection, and the mechanisms of action for imaging. Such discussions aim to propel the future development and application of DNP-based biomedical imaging agents.
Collapse
Affiliation(s)
- Shizhen Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Lei Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sha Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaping Yuan
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongxing Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Qiu D, He Y, Feng Y, Lin M, Lin Z, Zhang Z, Xiong Y, Hu Z, Ma S, Jin H, Liu J. Tumor perfusion enhancement by microbubbles ultrasonic cavitation reduces tumor glycolysis metabolism and alleviate tumor acidosis. Front Oncol 2024; 14:1424824. [PMID: 39091919 PMCID: PMC11291205 DOI: 10.3389/fonc.2024.1424824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
The tumor microenvironment is increasingly acknowledged as a critical contributor to cancer progression, mediating genetic and epigenetic alterations. Beyond diverse cellular interactions from the microenvironment, physicochemical factors such as tumor acidosis also significantly affect cancer dynamics. Recent research has highlighted that tumor acidosis facilitates invasion, immune escape, metastasis, and resistance to therapies. Thus, noninvasive measurement of tumor acidity and the development of targeted interventions represent promising strategies in oncology. Techniques like contrast-enhanced ultrasound (CEUS) can effectively assess blood perfusion, while ultrasound-stimulated microbubble cavitation (USMC) has proven to enhance tumor blood perfusion. We therefore aimed to determine whether CEUS assesses tumor acidity and whether USMC treatment can modulate tumor acidity. Firstly, we tracked CEUS perfusion parameters in MCF7 tumor models and compared them with in vivo tumor pH recorded by pH microsensors. We found that the peak intensity and area under curve of tumor contrast-enhanced ultrasound correlated well with tumor pH. We further conducted USMC treatment on MCF7 tumor-bearing mice, tracked changes of tumor blood perfusion and tumor pH in different perfusion regions before and after the USMC treatment to assess its impact on tumor acidity and optimize therapeutic ultrasound pressure. We discovered that USMC with 1.0 Mpa significantly improved tumor blood perfusion and tumor pH. Furthermore, tumor vascular pathology and PGI2 assays indicated that improved tumor perfusion was mainly due to vasodilation rather than angiogenesis. More importantly, analysis of glycolysis-related metabolites and enzymes demonstrated USMC treatment can reduce tumor acidity by reducing tumor glycolysis. These findings support that CEUS may serve as a potential biomarker to assess tumor acidity and USMC is a promising therapeutic modality for reducing tumor acidosis.
Collapse
Affiliation(s)
- Danxia Qiu
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yangcheng He
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuyi Feng
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Minhua Lin
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zekai Lin
- Department of Radiology, The Second Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Zhiyi Zhang
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ying Xiong
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhiwen Hu
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Suihong Ma
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hai Jin
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianhua Liu
- Department of Medical Ultrasound, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
6
|
Gammaraccio F, Villano D, Irrera P, Anemone AA, Carella A, Corrado A, Longo DL. Development and Validation of Four Different Methods to Improve MRI-CEST Tumor pH Mapping in Presence of Fat. J Imaging 2024; 10:166. [PMID: 39057737 PMCID: PMC11277679 DOI: 10.3390/jimaging10070166] [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: 05/08/2024] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
CEST-MRI is an emerging imaging technique suitable for various in vivo applications, including the quantification of tumor acidosis. Traditionally, CEST contrast is calculated by asymmetry analysis, but the presence of fat signals leads to wrong contrast quantification and hence to inaccurate pH measurements. In this study, we investigated four post-processing approaches to overcome fat signal influences and enable correct CEST contrast calculations and tumor pH measurements using iopamidol. The proposed methods involve replacing the Z-spectrum region affected by fat peaks by (i) using a linear interpolation of the fat frequencies, (ii) applying water pool Lorentzian fitting, (iii) considering only the positive part of the Z-spectrum, or (iv) calculating a correction factor for the ratiometric value. In vitro and in vivo studies demonstrated the possibility of using these approaches to calculate CEST contrast and then to measure tumor pH, even in the presence of moderate to high fat fraction values. However, only the method based on the water pool Lorentzian fitting produced highly accurate results in terms of pH measurement in tumor-bearing mice with low and high fat contents.
Collapse
Affiliation(s)
- Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Pietro Irrera
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Annasofia A. Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| |
Collapse
|
7
|
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, Del Mercato LL. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening. Adv Healthc Mater 2024:e2401138. [PMID: 38978424 DOI: 10.1002/adhm.202401138] [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: 03/26/2024] [Revised: 06/06/2024] [Indexed: 07/10/2024]
Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
8
|
Ali S, de Gracia Lux C, Brown K, Endsley C, Woodward A, Mattrey R, Lux J. Modulating Nonlinear Acoustic Response of Phospholipid-Coated Microbubbles with pH for Ultrasound Imaging. ACS Sens 2024; 9:2356-2363. [PMID: 38752383 DOI: 10.1021/acssensors.3c02382] [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] [Indexed: 05/25/2024]
Abstract
Activatable microbubble contrast agents for contrast-enhanced ultrasound have a potential role for measuring physiologic and pathologic states in deep tissues, including tumor acidosis. In this study, we describe a novel observation of increased harmonic oscillation of phosphatidylcholine microbubbles (PC-MBs) in response to lower ambient pH using a clinical ultrasound scanner. MB echogenicity and nonlinear echoes were monitored at neutral and acidic pH using B-mode and Cadence contrast pulse sequencing (CPS), a harmonic imaging technique at 7.0 and 1.5 MHz. A 3-fold increase in harmonic signal intensity was observed when the pH of PC-MB suspensions was decreased from 7.4 to 5.5 to mimic normal and pathophysiological levels that can be encountered in vivo. This pH-mediated activation is tunable based on the chemical structure and length of phospholipids composing the MB shell. It is also reliant on the presence of phosphate groups, as the use of lipids without phosphate instead of phospholipids completely abrogated this phenomenon. The increased harmonic signal likely is the result of increased MB oscillation caused by a decrease of the interfacial tension induced at a lower pH, altering the lipid conformation. While relative signal changes are interpreted clinically as mostly related to blood flow, pH effects could be significant contributors, particularly when imaging tumors. While our observation can be used clinically, it requires further research to isolate the effect of pH from other variables. These findings could pave the way toward for the development of new smart ultrasound contrast agents that expand the clinical utility of contrast-enhanced ultrasound.
Collapse
Affiliation(s)
- Shariq Ali
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Caroline de Gracia Lux
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Katherine Brown
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080-3021, United States
| | - Connor Endsley
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Adam Woodward
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Robert Mattrey
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| | - Jacques Lux
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8514, United States
| |
Collapse
|
9
|
Zhao Z, Jin Z, Wu G, Li C, Yu J. TriFNet: A triple-branch feature fusion network for pH determination by surface-enhanced Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 312:124048. [PMID: 38387412 DOI: 10.1016/j.saa.2024.124048] [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: 11/04/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Due to the acidic tumor microenvironment caused by metabolic changes in tumor cells, the accurate pH detection of extracellular fluid is helpful for doctors in precise tumor resection. The combination of Raman spectroscopy and deep learning provides a solution for pH detection. However, most existing studies use one-dimensional convolutional neural networks (1D-CNNs) for spectral analysis, which limits the performance due to insufficient feature extraction. In this work, we propose a 2D triple-branch feature fusion network (TriFNet) for accurate pH determination using surface-enhanced Raman spectra (SERS). Specifically, we design a triple-branch network structure by converting Raman spectra into three types of images to extensively extract complex patterns in spectra. In addition, an attention fusion module, which leverages the complementarity among features in both space and channel, is designed to obtain the valuable information, achieving further accurate pH determination. On our Raman spectral dataset containing 14,137 samples, we achieved mean absolute error (MAE) of 0.059, standard deviation of the absolute error (SD) of 0.07, root mean squared error (RMSE) of 0.092, and coefficient of determination (R2) of 0.991 on the test set. Compared with other published methods, the four metrics showed an average improvement of 47%, 39%, 43%, and 6%, respectively. In addition, visualization validates the diagnostic capability of our model to correlate with biomolecular signatures. Meanwhile, our model has robustness to different SERS chips. These results prove the potential of our method to develop an effective technology based on Raman spectroscopy for accurate pH determination to guide surgery.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Information Science and Technology, Fudan University, Shanghai 200438, China
| | - Ziyi Jin
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Guoqing Wu
- School of Information Science and Technology, Fudan University, Shanghai 200438, China
| | - Cong Li
- School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai 200438, China.
| |
Collapse
|
10
|
Lefevre C, Thibaut MM, Loumaye A, Thissen JP, Neyrinck AM, Navez B, Delzenne NM, Feron O, Bindels LB. Tumoral acidosis promotes adipose tissue depletion by fostering adipocyte lipolysis. Mol Metab 2024; 83:101930. [PMID: 38570069 PMCID: PMC11027574 DOI: 10.1016/j.molmet.2024.101930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE Tumour progression drives profound alterations in host metabolism, such as adipose tissue depletion, an early event of cancer cachexia. As fatty acid consumption by cancer cells increases upon acidosis of the tumour microenvironment, we reasoned that fatty acids derived from distant adipose lipolysis may sustain tumour fatty acid craving, leading to the adipose tissue loss observed in cancer cachexia. METHODS To evaluate the pro-lipolytic capacities of acid-exposed cancer cells, primary mouse adipocytes from subcutaneous and visceral adipose tissue were exposed to pH-matched conditioned medium from human and murine acid-exposed cancer cells (pH 6.5), compared to naive cancer cells (pH 7.4). To further address the role of tumoral acidosis on adipose tissue loss, a pH-low insertion peptide was injected into tumour-bearing mice, and tumoral acidosis was neutralised with a sodium bicarbonate buffer. Prolipolytic mediators were identified by transcriptomic approaches and validated on murine and human adipocytes. RESULTS Here, we reveal that acid-exposed cancer cells promote lipolysis from subcutaneous and visceral adipocytes and that dampening acidosis in vivo inhibits adipose tissue depletion. We further found a set of well-known prolipolytic factors enhanced upon acidosis adaptation and unravelled a role for β-glucuronidase (GUSB) as a promising new actor in adipocyte lipolysis. CONCLUSIONS Tumoral acidosis promotes the mobilization of fatty acids derived from adipocytes via the release of soluble factors by cancer cells. Our work paves the way for therapeutic approaches aimed at tackling cachexia by targeting the tumour acidic compartment.
Collapse
Affiliation(s)
- Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Morgane M Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey Loumaye
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Jean-Paul Thissen
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Benoit Navez
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
11
|
Chaumeil MM, Bankson JA, Brindle KM, Epstein S, Gallagher FA, Grashei M, Guglielmetti C, Kaggie JD, Keshari KR, Knecht S, Laustsen C, Schmidt AB, Vigneron D, Yen YF, Schilling F. New Horizons in Hyperpolarized 13C MRI. Mol Imaging Biol 2024; 26:222-232. [PMID: 38147265 PMCID: PMC10972948 DOI: 10.1007/s11307-023-01888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/27/2023]
Abstract
Hyperpolarization techniques significantly enhance the sensitivity of magnetic resonance (MR) and thus present fascinating new directions for research and applications with in vivo MR imaging and spectroscopy (MRI/S). Hyperpolarized 13C MRI/S, in particular, enables real-time non-invasive assessment of metabolic processes and holds great promise for a diverse range of clinical applications spanning fields like oncology, neurology, and cardiology, with a potential for improving early diagnosis of disease, patient stratification, and therapy response assessment. Despite its potential, technical challenges remain for achieving clinical translation. This paper provides an overview of the discussions that took place at the international workshop "New Horizons in Hyperpolarized 13C MRI," in March 2023 at the Bavarian Academy of Sciences and Humanities, Munich, Germany. The workshop covered new developments, as well as future directions, in topics including polarization techniques (particularly focusing on parahydrogen-based methods), novel probes, considerations related to data acquisition and analysis, and emerging clinical applications in oncology and other fields.
Collapse
Affiliation(s)
- Myriam M Chaumeil
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
| | - James A Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Ferdia A Gallagher
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Martin Grashei
- Department of Nuclear Medicine, TUM School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Guglielmetti
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Joshua D Kaggie
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Weill Cornell Graduate School, New York City, NY, USA
| | | | - Christoffer Laustsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| | - Andreas B Schmidt
- Partner Site Freiburg and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany
- Department of Chemistry, Integrative Biosciences (Ibio), Karmanos Cancer Institute (KCI), Wayne State University, Detroit, MI, 48202, USA
| | - Daniel Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Franz Schilling
- Department of Nuclear Medicine, TUM School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Partner Site Freiburg and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
12
|
Agbana P, Park JE, Rychahou P, Kim KB, Bae Y. Carfilzomib-Loaded Ternary Polypeptide Nanoparticles Stabilized by Polycationic Complexation. J Pharm Sci 2024; 113:711-717. [PMID: 37673172 PMCID: PMC10979393 DOI: 10.1016/j.xphs.2023.08.026] [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: 05/24/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023]
Abstract
Carfilzomib (CFZ) is a second-generation proteasome inhibitor showing great efficacy in multiple myeloma treatment, yet its clinical applications for other diseases such as solid cancers are limited due to low aqueous solubility and poor biostability. Ternary polypeptide nanoparticles (tPNPs) are drug carriers that we previously reported to overcome these pharmaceutical limitations by entrapping CFZ in the core of the nanoparticles and protecting the drugs from degradation in biological media. However, preclinical studies revealed that tPNPs would require further improvement in particle stability to suppress initial burst drug release and thus achieve prolonged inhibition of proteasome activity with CFZ against tumor cells in vivo. In this study, CFZ-loaded tPNPs are stabilized by polycations which have varying pKa values and thus differently modulate nanoparticle stability in response to solution pH. Through polyion complexation, the polycations appeared to stabilize the core of tPNPs entrapping CFZ-cyclodextrin inclusion complexes while allowing for uniform particle size before and after freeze drying. Interestingly, CFZ-loaded tPNPs (CFZ/tPNPs) showed pH-dependent drug release kinetics, which accelerated CFZ release as solution acidity increased (pH < 6) without compromising particle stability at the physiological condition (pH 7.4). In vitro cytotoxicity and proteasome activity assays confirmed that tPNPs stabilized with cationic polymers improved bioactivity of CFZ against CFZ-resistant cancer cells, which would be greatly beneficial in combination with pH-dependent drug release for treatment of solid cancers with drug resistance and tumor microenvironment acidosis by using CFZ and other proteasome inhibitors.
Collapse
Affiliation(s)
- Preye Agbana
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Ji Eun Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Kyung-Bo Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
13
|
Mancini C, Lori G, Pranzini E, Taddei ML. Metabolic challengers selecting tumor-persistent cells. Trends Endocrinol Metab 2024; 35:263-276. [PMID: 38071164 DOI: 10.1016/j.tem.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 03/14/2024]
Abstract
Resistance to anticancer therapy still represents one of the main obstacles to cancer treatment. Numerous components of the tumor microenvironment (TME) contribute significantly to the acquisition of drug resistance. Microenvironmental pressures arising during cancer evolution foster tumor heterogeneity (TH) and facilitate the emergence of drug-resistant clones. In particular, metabolic pressures arising in the TME may favor epigenetic adaptations supporting the acquisition of persistence features in tumor cells. Tumor-persistent cells (TPCs) are characterized by high phenotypic and metabolic plasticity, representing a noticeable advantage in chemo- and radio-resistance. Understanding the crosslink between the evolution of metabolic pressures in the TME, epigenetics, and TPC evolution is significant for developing novel therapeutic strategies specifically targeting TPC vulnerabilities to overcome drug resistance.
Collapse
Affiliation(s)
- Caterina Mancini
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Giulia Lori
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| |
Collapse
|
14
|
Aubert L, Bastien E, Renoult O, Guilbaud C, Özkan K, Brusa D, Bouzin C, Richiardone E, Richard C, Boidot R, Léonard D, Corbet C, Feron O. Tumor acidosis-induced DNA damage response and tetraploidy enhance sensitivity to ATM and ATR inhibitors. EMBO Rep 2024; 25:1469-1489. [PMID: 38366255 PMCID: PMC10933359 DOI: 10.1038/s44319-024-00089-7] [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: 10/08/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/18/2024] Open
Abstract
Tumor acidosis is associated with increased invasiveness and drug resistance. Here, we take an unbiased approach to identify vulnerabilities of acid-exposed cancer cells by combining pH-dependent flow cytometry cell sorting from 3D colorectal tumor spheroids and transcriptomic profiling. Besides metabolic rewiring, we identify an increase in tetraploid cell frequency and DNA damage response as consistent hallmarks of acid-exposed cancer cells, supported by the activation of ATM and ATR signaling pathways. We find that regardless of the cell replication error status, both ATM and ATR inhibitors exert preferential growth inhibitory effects on acid-exposed cancer cells. The efficacy of a combination of these drugs with 5-FU is further documented in 3D spheroids as well as in patient-derived colorectal tumor organoids. These data position tumor acidosis as a revelator of the therapeutic potential of DNA repair blockers and as an attractive clinical biomarker to predict the response to a combination with chemotherapy.
Collapse
Affiliation(s)
- Léo Aubert
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium.
| | - Estelle Bastien
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Ophélie Renoult
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Céline Guilbaud
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Kübra Özkan
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Davide Brusa
- CytoFlux-Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform 2IP, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Corentin Richard
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges‑François Leclerc Cancer Center‑UNICANCER, 21079, Dijon, France
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges‑François Leclerc Cancer Center‑UNICANCER, 21079, Dijon, France
| | - Daniel Léonard
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc, and Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200, Brussels, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, B-1300, Wavre, Belgium.
| |
Collapse
|
15
|
Li S, Wang Q, Ren Y, Zhong P, Bao P, Guan S, Qiu X, Qu X. Oxygen and pH responsive theragnostic liposomes for early-stage diagnosis and photothermal therapy of solid tumours. Biomater Sci 2024; 12:748-762. [PMID: 38131275 DOI: 10.1039/d3bm01514a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The development of cancer treatment is of great importance, especially in the early stage. In this work, we synthesized a pH-sensitive amphiphilic ruthenium complex containing two alkyl chains and two PEG chains, which was utilized as an oxygen sensitive fluorescent probe for co-assembly with lipids to harvest a liposomal delivery system (RuPC) for the encapsulation of a photothermal agent indocyanine green (ICG). The resultant ICG encapsulated liposome (RuPC@ICG) enabled the delivery of ICG into cells via a membrane fusion pathway, by which the ruthenium complex was localized in the cell membrane for better detection of the extracellular oxygen concentration. Such characteristics allowed ratiometric imaging to distinguish the tumour location from normal tissues just 3 days after cancer cells were implanted, by monitoring the hypoxia condition and tracing the metabolism. Moreover, the pH sensitivity of the liposomes favoured cell uptake, and improved the anti-tumour efficiency of the formulation in vivo under NIR irradiation. Assuming liposomal systems have fewer safety issues, our work not only provides a facile method for the construction of a theragnostic system by combining phototherapy with photoluminescence imaging, but hopefully paves the way for clinical translation from bench to bedside.
Collapse
Affiliation(s)
- Siyi Li
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Qinglin Wang
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Yingying Ren
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Pengfei Zhong
- Hebei North University, Hebei 075000, China
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100094, China
| | - Pengtao Bao
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100094, China
| | - Shanyue Guan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xiaochen Qiu
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Xiaozhong Qu
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China.
- Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Shandong 256606, China
| |
Collapse
|
16
|
Schmitt FJ, Mehmood AS, Tüting C, Phan HT, Reisdorf J, Rieder F, Ghane Golmohamadi F, Verma R, Kastritis PL, Laufer J. Effect of Molecular Dynamics and Internal Water Contact on the Photophysical Properties of Red pH-Sensitive Proteins. Biochemistry 2024; 63:82-93. [PMID: 38085825 DOI: 10.1021/acs.biochem.3c00444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The pH dependence of the absorption and (time-resolved) fluorescence of two red-shifted fluorescent proteins, mCardinal and mNeptune, was investigated. Decay-associated spectra were measured following fluorescence excitation at 470 nm in PBS buffer with a pH that ranged from 5.5 to 8.0. The fluorescence of both proteins shows two different decay components. mCardinal exhibits an increase in the long-lived fluorescence component with acidification from 1.34 ns at pH 8.0 to 1.62 ns at pH 5.5. An additional fast decay component with 0.64 ns at pH 8.0 up to 1.1 ns at pH 5.5 was found to be blue-shifted compared to the long-lived component. The fluorescence lifetime of mNeptune is insensitive to pH. DAS of mCardinal were simulated assuming a coupled two-level system to describe the 1S state of the chromophore within two different conformations of the protein. MD simulations were conducted to correlate the experimentally observed pH-induced change in the lifetime in mCardinal with its molecular properties. While the chromophores of both protein variants are stabilized by the same number of hydrogen bonds, it was found that the chromophore in mCardinal exhibits more water contacts compared to mNeptune. In mCardinal, interaction between the chromophore and Glu-145 is reduced as compared to mNeptune, but interaction with Thr-147 which is Ser-147 in mNeptune is stronger in mCardinal. Therefore, the dynamics of the excited-state proton transfer (ESPT) might be different in mCardinal and mNeptune. The pH dependency of ESPT is suggested as a key mechanism for pH sensitivity.
Collapse
Affiliation(s)
- Franz-Josef Schmitt
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| | - Amna Shah Mehmood
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| | - Christian Tüting
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, 06120 Halle, Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle, Saale, Germany
| | - Hoang Trong Phan
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Judith Reisdorf
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| | - Fabian Rieder
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| | - Farzin Ghane Golmohamadi
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| | | | - Panagiotis L Kastritis
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, 06120 Halle, Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, 06120 Halle, Saale, Germany
| | - Jan Laufer
- Institute of Physics, Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 3, 06120 Halle, Saale, Germany
| |
Collapse
|
17
|
Paech D, Weckesser N, Franke VL, Breitling J, Görke S, Deike-Hofmann K, Wick A, Scherer M, Unterberg A, Wick W, Bendszus M, Bachert P, Ladd ME, Schlemmer HP, Korzowski A. Whole-Brain Intracellular pH Mapping of Gliomas Using High-Resolution 31P MR Spectroscopic Imaging at 7.0 T. Radiol Imaging Cancer 2024; 6:e220127. [PMID: 38133553 PMCID: PMC10825708 DOI: 10.1148/rycan.220127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023]
Abstract
Malignant tumors commonly exhibit a reversed pH gradient compared with normal tissue, with a more acidic extracellular pH and an alkaline intracellular pH (pHi). In this prospective study, pHi values in gliomas were quantified using high-resolution phosphorous 31 (31P) spectroscopic MRI at 7.0 T and were used to correlate pHi alterations with histopathologic findings. A total of 12 participants (mean age, 58 years ± 18 [SD]; seven male, five female) with histopathologically proven, newly diagnosed glioma were included between September 2018 and November 2019. The 31P spectroscopic MRI scans were acquired using a double-resonant 31P/1H phased-array head coil together with a three-dimensional (3D) 31P chemical shift imaging sequence (5.7-mL voxel volume) performed with a 7.0-T whole-body system. The 3D volumetric segmentations were performed for the whole-tumor volumes (WTVs); tumor subcompartments of necrosis, gadolinium enhancement, and nonenhancing T2 (NCE T2) hyperintensity; and normal-appearing white matter (NAWM), and pHi values were compared. Spearman correlation was used to assess association between pHi and the proliferation index Ki-67. For all study participants, mean pHi values were higher in the WTV (7.057 ± 0.024) compared with NAWM (7.006 ± 0.012; P < .001). In eight participants with high-grade gliomas, pHi was increased in all tumor subcompartments (necrosis, 7.075 ± 0.033; gadolinium enhancement, 7.075 ± 0.024; NCE T2 hyperintensity, 7.043 ± 0.015) compared with NAWM (7.004 ± 0.014; all P < .01). The pHi values of WTV positively correlated with Ki-67 (R2 = 0.74, r = 0.78, P = .001). In conclusion, 31P spectroscopic MRI at 7.0 T enabled high-resolution quantification of pHi in gliomas, with pHi alteration associated with the Ki-67 proliferation index, and may aid in diagnosis and treatment monitoring. Keywords: 31P MRSI, pH, Glioma, Glioblastoma, Ultra-High-Field MRI, Imaging Biomarker, 7 Tesla Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
| | | | - Vanessa L. Franke
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes Breitling
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Steffen Görke
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Katerina Deike-Hofmann
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Antje Wick
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Moritz Scherer
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Bachert
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Mark E. Ladd
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Heinz-Peter Schlemmer
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Korzowski
- From the Divisions of Radiology (D.P., N.W., K.D.H., H.P.S.) and
Medical Physics in Radiology (V.L.F., J.B., S.G., P.B., M.E.L., A.K.), German
Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,
Germany; Faculties of Medicine (N.W., M.E.L.) and Physics and Astronomy (V.L.F.,
P.B., M.E.L.), University of Heidelberg, Heidelberg, Germany; and Departments of
Neurology (A.W., W.W.), Neurosurgery (M.S., A.U.), and Neuroradiology (M.B.),
Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
18
|
Abstract
Cancers undergo sequential changes to proton (H+) concentration and sensing that are consequences of the disease and facilitate its further progression. The impact of protonation state on protein activity can arise from alterations to amino acids or their titration. Indeed, many cancer-initiating mutations influence pH balance, regulation or sensing in a manner that enables growth and invasion outside normal constraints as part of oncogenic transformation. These cancer-supporting effects become more prominent when tumours develop an acidic microenvironment owing to metabolic reprogramming and disordered perfusion. The ensuing intracellular and extracellular pH disturbances affect multiple aspects of tumour biology, ranging from proliferation to immune surveillance, and can even facilitate further mutagenesis. As a selection pressure, extracellular acidosis accelerates disease progression by favouring acid-resistant cancer cells, which are typically associated with aggressive phenotypes. Although acid-base disturbances in tumours often occur alongside hypoxia and lactate accumulation, there is now ample evidence for a distinct role of H+-operated responses in key events underpinning cancer. The breadth of these actions presents therapeutic opportunities to change the trajectory of disease.
Collapse
Affiliation(s)
- Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Stine Falsig Pedersen
- Department of Biology, University of Copenhagen, University of Copenhagen, Faculty of Science, København, Denmark.
| |
Collapse
|
19
|
Patrucco D, Cutrin JC, Longo DL, Botto E, Cong L, Aime S, Delli Castelli D. In Situ Insonation of Alkaline Buffer Containing Liposomes Leads to a Net Improvement of the Therapeutic Outcome in a Triple Negative Breast Cancer Murine Model. Adv Healthc Mater 2023; 12:e2301480. [PMID: 37709294 DOI: 10.1002/adhm.202301480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Breast cancer is characterized by an acidic micro-environment. Acidic extracellular pH gives cancer cells an evolutionary advantage, hence, neutralization of the extracellular pH has been considered as a potential therapeutic strategy. To address the issue of systemic pH alteration, an approach based on the targeted delivery of the buffering solution to the tumor region is investigated. The method relies on the use of low frequency ultrasound and sono-sensitive liposomes loaded with buffers at alkaline pH (LipHUS). After the i.v. injection of LipHUS, the application of ultrasound (US) at the sites of the pathology induces a local increase of pH that results highly effective in i) inhibiting primary tumor growth, ii) reducing tumor recurrence after surgery, and iii) suppressing metastases' formation. The experiments are carried out on a triple negative breast cancer mouse model. The results obtained demonstrate that localized and triggered release of bicarbonate or PBS buffer from sonosensitive liposomes represents an efficient therapeutic tool for treating triple-negative breast cancer. This approach holds promise for potential clinical translation.
Collapse
Affiliation(s)
- Deyssy Patrucco
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| | - Juan Carlos Cutrin
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| | - Dario Livio Longo
- Istituto di Biostrutture e Bioimmagini (IBB), Consiglio Nazionale delle Ricerche (CNR), Via Tommaso De Amicis, 95, Naples, 80145, Italy
| | - Elena Botto
- Istituto di Biostrutture e Bioimmagini (IBB), Consiglio Nazionale delle Ricerche (CNR), Via Tommaso De Amicis, 95, Naples, 80145, Italy
| | - Li Cong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Silvio Aime
- IRCCS SDN, SYNLAB, Via Gianturco 113, Naples, 80143, Italy
| | - Daniela Delli Castelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| |
Collapse
|
20
|
Mu C, Liu X, Kim Y, Riselli A, Korenchan DE, Bok RA, Delos Santos R, Sriram R, Qin H, Nguyen H, Gordon JW, Slater J, Larson PEZ, Vigneron DB, Kurhanewicz J, Wilson DM, Flavell RR. Clinically Translatable Hyperpolarized 13C Bicarbonate pH Imaging Method for Use in Prostate Cancer. ACS Sens 2023; 8:4042-4054. [PMID: 37878761 PMCID: PMC10683509 DOI: 10.1021/acssensors.3c00851] [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: 05/02/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023]
Abstract
Solid tumors such as prostate cancer (PCa) commonly develop an acidic microenvironment with pH 6.5-7.2, owing to heterogeneous perfusion, high metabolic activity, and rapid cell proliferation. In preclinical prostate cancer models, disease progression is associated with a decrease in tumor extracellular pH, suggesting that pH imaging may reflect an imaging biomarker to detect aggressive and high-risk disease. Therefore, we developed a hyperpolarized carbon-13 MRI method to image the tumor extracellular pH (pHe) and prepared it for clinical translation for detection and risk stratification of PCa. This method relies on the rapid breakdown of hyperpolarized (HP) 1,2-glycerol carbonate (carbonyl-13C) via base-catalyzed hydrolysis to produce HP 13CO32-, which is neutralized and converted to HP H13CO3-. After injection, HP H13CO3- equilibrates with HP 13CO2 in vivo and enables the imaging of pHe. Using insights gleaned from mechanistic studies performed in the hyperpolarized state, we solved issues of polarization loss during preparation in a clinical polarizer system. We successfully customized a reaction apparatus suitable for clinical application, developed clinical standard operating procedures, and validated the radiofrequency pulse sequence and imaging data acquisition with a wide range of animal models. The results demonstrated that we can routinely produce a highly polarized and safe HP H13CO3- contrast agent suitable for human injection. Preclinical imaging studies validated the reliability and accuracy of measuring acidification in healthy kidney and prostate tumor tissue. These methods were used to support an Investigational New Drug application to the U.S. Food and Drug Administration. This methodology is now ready to be implemented in human trials, with the ultimate goal of improving the management of PCa.
Collapse
Affiliation(s)
- Changhua Mu
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Xiaoxi Liu
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Yaewon Kim
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Andrew Riselli
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - David E. Korenchan
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Robert A. Bok
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Romelyn Delos Santos
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Renuka Sriram
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Hecong Qin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Hao Nguyen
- Department
of Urology, University of California, San Francisco, California 94143, United States
| | - Jeremy W. Gordon
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - James Slater
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Peder E. Z. Larson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Daniel B. Vigneron
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - John Kurhanewicz
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - David M. Wilson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
| | - Robert R. Flavell
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94107, United States
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, California 94158, United States
| |
Collapse
|
21
|
Dhakan C, Anemone A, Ventura V, Carella A, Corrado A, Pirotta E, Villano D, Romdhane F, Gammaraccio F, Aime S, Longo DL. Assessing the Therapeutic Efficacy of Proton Transport Inhibitors in a Triple-Negative Breast Cancer Murine Model with Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer Tumor pH Imaging. Metabolites 2023; 13:1161. [PMID: 37999256 PMCID: PMC10673543 DOI: 10.3390/metabo13111161] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Proton transporters play a key role in maintaining the acidic tumor microenvironment; hence, their inhibition has been proposed as a new therapeutic treatment, although few methods can accurately assess their effect in vivo. In this study, we investigated whether MRI-CEST (Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer) tumor pH imaging can be a useful tool to evaluate in vivo the therapeutic efficacy of several Proton Pump Inhibitors (PPIs) in breast cancer. Cell viability and extracellular pH assays were carried out in breast cancer cells cultured at physiological pH (7.4) or acid-adapted (pH of 6.5 and 6.8) following the exposure to inhibitors of V-ATPase (Lansoprazole, Esomeprazole) or NHE1 (Amiloride, Cariporide) at several concentrations. Next, triple-negative breast cancer 4T1 tumor-bearing mice were treated with Lansoprazole or Amiloride and MRI-CEST tumor pH imaging was utilized to assess the in vivo efficacy. Only Lansoprazole induced, in addition to breast cancer cell toxicity, a significant inhibition of proton extrusion. A significant reduction in tumor volume, prolonged survival, and increase in extracellular tumor pH after 1 and 2 weeks were observed after Lansoprazole treatment, whereas no significant changes were detected upon Amiloride treatment. Our results suggested that MRI-CEST tumor pH imaging can monitor the therapeutic efficacy of PPIs in breast cancer murine models.
Collapse
Affiliation(s)
- Chetan Dhakan
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Annasofia Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Vittoria Ventura
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Elisa Pirotta
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Feriel Romdhane
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| | - Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Silvio Aime
- IRCCS SynLAB SDN, Via Gianturco 113, 80143 Naples, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
22
|
Gasymov OK, Bakhishova MJ, Aslanov RB, Melikova LA, Aliyev JA. Membrane Partitioning of TEMPO Discriminates Human Lung Cancer from Neighboring Normal Cells. Acta Naturae 2023; 15:111-120. [PMID: 38234602 PMCID: PMC10790361 DOI: 10.32607/actanaturae.19426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/16/2023] [Indexed: 01/19/2024] Open
Abstract
The plasma membranes of normal and cancer cells of the lung, breast, and colon tissues show considerably different lipid compositions that greatly influence their physicochemical properties. Partitioning of the spin probe 2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPO) into the membranes of human lung normal and carcinoma cells was assessed by EPR spectroscopy to estimate the impact of the lipid compositions. The goal was to reveal potential strategies for cancer therapy attributable to the membrane properties. The study was conducted at pH values of 7.3 and 6.2, relevant to the microenvironments of normal and cancer cells, respectively. The TEMPO partitioning was examined in the temperature interval of 283-317K to reveal the efficacy of local hyperthermia used in chemotherapy. Results indicate that the TEMPO partitioning coefficient for the membranes of human lung carcinoma cells is significantly higher compared with that of neighboring normal cells. Increased partition coefficients were observed at relatively higher temperatures in both normal and cancer cells. However, compared to the normal cells, the cancer cells demonstrated higher partition coefficients in the studied temperature range. The data obtained with C12SL (spin-labeled analog of lauric acid) indicate that increased membrane dynamics of the cancer cells is a possible mechanism for enhanced partitioning of TEMPO. Free energy values for partitioning estimated for pH values of 6.2 and 7.3 show that TEMPO partitioning requires 30% less energy in the cancer cells at pH 7.3. TEMPO and its derivatives have previously been considered as theranostic agents in cancer research. Data suggest that TEMPO derivatives could be used to test if complementary alkalization therapy is effective for cancer patients receiving standard chemotherapy with local hyperthermia.
Collapse
Affiliation(s)
- O. K. Gasymov
- Institute of Biophysics, Ministry of Science and Education Republic of Azerbaijan, Baku, AZ1171 Azerbaijan
| | - M. J. Bakhishova
- Institute of Biophysics, Ministry of Science and Education Republic of Azerbaijan, Baku, AZ1171 Azerbaijan
| | - R. B. Aslanov
- Institute of Biophysics, Ministry of Science and Education Republic of Azerbaijan, Baku, AZ1171 Azerbaijan
| | - L. A. Melikova
- Institute of Biophysics, Ministry of Science and Education Republic of Azerbaijan, Baku, AZ1171 Azerbaijan
- National Center of Oncology, Azerbaijan Republic Ministry of Health, Baku, AZ1012 Azerbaijan
| | - J. A. Aliyev
- National Center of Oncology, Azerbaijan Republic Ministry of Health, Baku, AZ1012 Azerbaijan
| |
Collapse
|
23
|
Hou B, Yi L, Hu D, Luo Z, Gao D, Li C, Xing B, Wang JW, Lee CN, Zhang R, Sheng Z, Zhou B, Liu X. A swallowable X-ray dosimeter for the real-time monitoring of radiotherapy. Nat Biomed Eng 2023; 7:1242-1251. [PMID: 37055542 DOI: 10.1038/s41551-023-01024-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/15/2023] [Indexed: 04/15/2023]
Abstract
Monitoring X-ray radiation in the gastrointestinal tract can enhance the precision of radiotherapy in patients with gastrointestinal cancer. Here we report the design and performance, in the gastrointestinal tract of rabbits, of a swallowable X-ray dosimeter for the simultaneous real-time monitoring of absolute absorbed radiation dose and of changes in pH and temperature. The dosimeter consists of a biocompatible optoelectronic capsule containing an optical fibre, lanthanide-doped persistent nanoscintillators, a pH-sensitive polyaniline film and a miniaturized system for the wireless readout of luminescence. The persistent luminescence of the nanoscintillators after irradiation can be used to continuously monitor pH without the need for external excitation. By using a neural-network-based regression model, we estimated the radiation dose from radioluminescence and afterglow intensity and temperature, and show that the dosimeter was approximately five times more accurate than standard methods for dose determination. Swallowable dosimeters may help to improve radiotherapy and to understand how radiotherapy affects tumour pH and temperature.
Collapse
Affiliation(s)
- Bo Hou
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Luying Yi
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Dehong Hu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Duyang Gao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chao Li
- Department of Spaceborne Microwave Remote Sensing System, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
| | - Bowen Xing
- Department of Precision Instruments, Tsinghua University, Beijing, China
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chuen Neng Lee
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rong Zhang
- Department of Precision Instruments, Tsinghua University, Beijing, China
| | - Zonghai Sheng
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Bin Zhou
- Department of Precision Instruments, Tsinghua University, Beijing, China.
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Materials Research and Engineering, Agency for Science,Technology and Research, Singapore, Singapore.
- Center for Functional Materials, National University of Singapore Suzhou Research Institute, Suzhou, China.
| |
Collapse
|
24
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
25
|
Igarashi T, Kim H, Sun PZ. Detection of tissue pH with quantitative chemical exchange saturation transfer magnetic resonance imaging. NMR IN BIOMEDICINE 2023; 36:e4711. [PMID: 35141979 PMCID: PMC10249910 DOI: 10.1002/nbm.4711] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 05/12/2023]
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) has emerged as a novel means for sensitive detection of dilute labile protons and chemical exchange rates. By sensitizing to pH-dependent chemical exchange, CEST MRI has shown promising results in monitoring tissue statuses such as pH changes in disorders like acute stroke, tumor, and acute kidney injury. This article briefly reviews the basic principles for CEST imaging and quantitative measures, from the simplistic asymmetry analysis to multipool Lorentzian decoupling and quasi-steady-state reconstruction. In particular, the advantages and limitations of commonly used quantitative approaches for CEST applications are discussed.
Collapse
Affiliation(s)
- Takahiro Igarashi
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Hahnsung Kim
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Phillip Zhe Sun
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| |
Collapse
|
26
|
Rizzo R, Onesto V, Morello G, Iuele H, Scalera F, Forciniti S, Gigli G, Polini A, Gervaso F, del Mercato LL. pH-sensing hybrid hydrogels for non-invasive metabolism monitoring in tumor spheroids. Mater Today Bio 2023; 20:100655. [PMID: 37234366 PMCID: PMC10205545 DOI: 10.1016/j.mtbio.2023.100655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
The constant increase in cancer incidence and mortality pushes biomedical research towards the development of in vitro 3D systems able to faithfully reproduce and effectively probe the tumor microenvironment. Cancer cells interact with this complex and dynamic architecture, leading to peculiar tumor-associated phenomena, such as acidic pH conditions, rigid extracellular matrix, altered vasculature, hypoxic condition. Acidification of extracellular pH, in particular, is a well-known feature of solid tumors, correlated to cancer initiation, progression, and resistance to therapies. Monitoring local pH variations, non-invasively, during cancer growth and in response to drug treatment becomes extremely important for understanding cancer mechanisms. Here, we describe a simple and reliable pH-sensing hybrid system, based on a thermoresponsive hydrogel embedding optical pH sensors, that we specifically apply for non-invasive and accurate metabolism monitoring in colorectal cancer (CRC) spheroids. First, the physico-chemical properties of the hybrid sensing platform, in terms of stability, rheological and mechanical properties, morphology and pH sensitivity, were fully characterized. Then, the proton gradient distribution in the spheroids proximity, in the presence or absence of drug treatment, was quantified over time by time lapse confocal light scanning microscopy and automated segmentation pipeline, highlighting the effects of the drug treatment in the extracellular pH. In particular, in the treated CRC spheroids the acidification of the microenvironment resulted faster and more pronounced over time. Moreover, a pH gradient distribution was detected in the untreated spheroids, with more acidic values in proximity of the spheroids, resembling the cell metabolic features observed in vivo in the tumor microenvironment. These findings promise to shed light on mechanisms of regulation of proton exchanges by cellular metabolism being essential for the study of solid tumors in 3D in vitro models and the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Riccardo Rizzo
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Giulia Morello
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
- Department of Mathematics and Physics ‘‘Ennio De Giorgi”, University of Salento, C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Francesca Scalera
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
- Department of Mathematics and Physics ‘‘Ennio De Giorgi”, University of Salento, C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Alessandro Polini
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Francesca Gervaso
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| | - Loretta L. del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), C/o Campus Ecotekne, Via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
27
|
Schwenck J, Sonanini D, Cotton JM, Rammensee HG, la Fougère C, Zender L, Pichler BJ. Advances in PET imaging of cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00576-4. [PMID: 37258875 DOI: 10.1038/s41568-023-00576-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Molecular imaging has experienced enormous advancements in the areas of imaging technology, imaging probe and contrast development, and data quality, as well as machine learning-based data analysis. Positron emission tomography (PET) and its combination with computed tomography (CT) or magnetic resonance imaging (MRI) as a multimodality PET-CT or PET-MRI system offer a wealth of molecular, functional and morphological data with a single patient scan. Despite the recent technical advances and the availability of dozens of disease-specific contrast and imaging probes, only a few parameters, such as tumour size or the mean tracer uptake, are used for the evaluation of images in clinical practice. Multiparametric in vivo imaging data not only are highly quantitative but also can provide invaluable information about pathophysiology, receptor expression, metabolism, or morphological and functional features of tumours, such as pH, oxygenation or tissue density, as well as pharmacodynamic properties of drugs, to measure drug response with a contrast agent. It can further quantitatively map and spatially resolve the intertumoural and intratumoural heterogeneity, providing insights into tumour vulnerabilities for target-specific therapeutic interventions. Failure to exploit and integrate the full potential of such powerful imaging data may lead to a lost opportunity in which patients do not receive the best possible care. With the desire to implement personalized medicine in the cancer clinic, the full comprehensive diagnostic power of multiplexed imaging should be utilized.
Collapse
Affiliation(s)
- Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonathan M Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Department of Immunology, IFIZ Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany.
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
28
|
Chen B, Meng X, Wu W, Zhang Y, Ma L, Chen K, Fang X. A novel CEST-contrast nanoagent for differentiating the malignant degree in breast cancer. RSC Adv 2023; 13:14131-14138. [PMID: 37180024 PMCID: PMC10167945 DOI: 10.1039/d3ra01006f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Different subtypes of breast cancer (BCC) have variable degrees of malignancy, which is closely related to their extracellular pH (pHe). Therefore, it is increasingly significant to monitor the extracellular pH sensitively to further determine the malignancy of different subtypes of BCC. Here, a l-arginine and Eu3+ assembled nanoparticle Eu3+@l-Arg was prepared to detect the pHe of two breast cancer models (TUBO is non-invasive and 4T1 is malignant) using a clinical chemical exchange saturation shift imaging technique. The experiments in vivo showed that Eu3+@l-Arg nanomaterials could respond sensitively to changes of pHe. In 4T1 models, the CEST signal enhanced about 5.42 times after Eu3+@l-Arg nanomaterials were used to detect the pHe. In contrast, few enhancements of the CEST signal were seen in the TUBO models. This significant difference had led to new ideas for identifying subtypes of BCC with different degrees of malignancy.
Collapse
Affiliation(s)
- Bixue Chen
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Xianfu Meng
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai China
- Department of Materials Science, State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai China
| | - Wanlu Wu
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University Shanghai China
| | - Lin Ma
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Kaidong Chen
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| | - Xiangming Fang
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi China
| |
Collapse
|
29
|
Petrunina NA, Shtork AS, Lukina MM, Tsvetkov VB, Khodarovich YM, Feofanov AV, Moysenovich AM, Maksimov EG, Shipunova VO, Zatsepin TS, Bogomazova AN, Shender VO, Aralov AV, Lagarkova MA, Varizhuk AM. Ratiometric i-Motif-Based Sensor for Precise Long-Term Monitoring of pH Micro Alterations in the Nucleoplasm and Interchromatin Granules. ACS Sens 2023; 8:619-629. [PMID: 36662613 DOI: 10.1021/acssensors.2c01813] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
DNA-intercalated motifs (iMs) are facile scaffolds for the design of various pH-responsive nanomachines, including biocompatible pH sensors. First, DNA pH sensors relied on complex intermolecular scaffolds. Here, we used a simple unimolecular dual-labeled iM scaffold and minimized it by replacing the redundant loop nucleosides with abasic or alkyl linkers. These modifications improved the thermal stability of the iM and increased the rates of its pH-induced conformational transitions. The best effects were obtained upon the replacement of all three native loops with short and flexible linkers, such as the propyl one. The resulting sensor showed a pH transition value equal to 6.9 ± 0.1 and responded rapidly to minor acidification (tau1/2 <1 s for 7.2 → 6.6 pH jump). We demonstrated the applicability of this sensor for pH measurements in the nuclei of human lung adenocarcinoma cells (pH = 7.4 ± 0.2) and immortalized embryonic kidney cells (pH = 7.0 ± 0.2). The sensor stained diffusely the nucleoplasm and piled up in interchromatin granules. These findings highlight the prospects of iMs in the studies of normal and pathological pH-dependent processes in the nucleus, including the formation of biomolecular condensates.
Collapse
Affiliation(s)
- Nataliia A Petrunina
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia
| | - Alina S Shtork
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia
| | - Maria M Lukina
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow119435, Russia
| | - Vladimir B Tsvetkov
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Institute of Biodesign and Complex System Modeling, I.M. Sechenov First Moscow State Medical University, Moscow119991, Russia.,A.V. Topchiev Institute of Petrochemical Synthesis RAS, Leninsky Prospect Str. 29, Moscow119991, Russia
| | - Yuri M Khodarovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russia.,The Peoples' Friendship University of Russia, 117198Moscow, Russia
| | - Alexey V Feofanov
- Biological Faculty, Lomonosov Moscow State University, Moscow119992, Russia.,Institute of Gene Biology RAS, Russian Academy of Sciences, Moscow119334, Russia
| | | | - Eugene G Maksimov
- Biological Faculty, Lomonosov Moscow State University, Moscow119992, Russia
| | - Victoria O Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russia
| | - Timofei S Zatsepin
- Department of Chemistry, Lomonosov Moscow State University, Moscow119992, Russia
| | - Alexandra N Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow119435, Russia
| | - Victoria O Shender
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow119435, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russia
| | - Andrey V Aralov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russia
| | - Maria A Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow119435, Russia
| | - Anna M Varizhuk
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow119435, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow119435, Russia.,G4_Interact, USERN, University of Pavia, 27100Pavia, Italy
| |
Collapse
|
30
|
Janasik D, Imielska P, Krawczyk T. Tuning the pH of Activation of Fluorinated Hydrazone-Based Switches─A Pathway to Versatile 19F Magnetic Resonance Imaging Contrast Agents. ACS Sens 2023; 8:721-727. [PMID: 36695323 PMCID: PMC9972467 DOI: 10.1021/acssensors.2c02251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Molecular switches have become an area of great interest in recent years. They are explored as high-density data storage and organic diodes in molecular electronics as well as chemosensors due to their ability to undergo a transition between well-defined structures under the action of external stimuli. One of the types of such switches is hydrazones. They work by changing the configuration from E to Z under the influence of pH or light. The change in configuration is accompanied by a change in the absorption band and changes in the nuclear magnetic resonance (NMR) spectrum. In this publication, the structure-property relationship of fluorinated hydrazone switches was established. A linear relationship between the Hammett substituent constants and the pH where the switching occurs was found. Introduction of strong electron-donating groups allowed obtaining a hydrazone switch of pKa = 6 suitable for application in 19F MRI as contrast agents.
Collapse
Affiliation(s)
- Dawid Janasik
- Department of Chemical Organic Technology and Petrochemistry, Silesian University of Technology, Krzywoustego 4, 44-100Gliwice, Poland
| | - Patrycja Imielska
- Department of Chemical Organic Technology and Petrochemistry, Silesian University of Technology, Krzywoustego 4, 44-100Gliwice, Poland
| | - Tomasz Krawczyk
- Department of Chemical Organic Technology and Petrochemistry, Silesian University of Technology, Krzywoustego 4, 44-100Gliwice, Poland
| |
Collapse
|
31
|
Vladimirov N, Perlman O. Molecular MRI-Based Monitoring of Cancer Immunotherapy Treatment Response. Int J Mol Sci 2023; 24:3151. [PMID: 36834563 PMCID: PMC9959624 DOI: 10.3390/ijms24043151] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapy constitutes a paradigm shift in cancer treatment. Its FDA approval for several indications has yielded improved prognosis for cases where traditional therapy has shown limited efficiency. However, many patients still fail to benefit from this treatment modality, and the exact mechanisms responsible for tumor response are unknown. Noninvasive treatment monitoring is crucial for longitudinal tumor characterization and the early detection of non-responders. While various medical imaging techniques can provide a morphological picture of the lesion and its surrounding tissue, a molecular-oriented imaging approach holds the key to unraveling biological effects that occur much earlier in the immunotherapy timeline. Magnetic resonance imaging (MRI) is a highly versatile imaging modality, where the image contrast can be tailored to emphasize a particular biophysical property of interest using advanced engineering of the imaging pipeline. In this review, recent advances in molecular-MRI based cancer immunotherapy monitoring are described. Next, the presentation of the underlying physics, computational, and biological features are complemented by a critical analysis of the results obtained in preclinical and clinical studies. Finally, emerging artificial intelligence (AI)-based strategies to further distill, quantify, and interpret the image-based molecular MRI information are discussed in terms of perspectives for the future.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
32
|
Liu F, Anton N, Niko Y, Klymchenko AS. Controlled Release and Capture of Aldehydes by Dynamic Imine Chemistry in Nanoemulsions: From Delivery to Detoxification. ACS APPLIED BIO MATERIALS 2023; 6:246-256. [PMID: 36516427 DOI: 10.1021/acsabm.2c00861] [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: 12/15/2022]
Abstract
Current biomedical applications of nanocarriers are focused on drug delivery, where encapsulated cargo is released in the target tissues under the control of external stimuli. Here, we propose a very different approach, where the active toxic molecules are removed from biological tissues by the nanocarrier. It is based on the drug-sponge concept, where specific molecules are captured by the lipid nanoemulsion (NE) droplets due to dynamic covalent chemistry inside their oil core. To this end, we designed a highly lipophilic amine (LipoAmine) capable of reacting with a free cargo-aldehyde (fluorescent dye and 4-hydroxynonenal toxin) directly inside lipid NEs, yielding a lipophilic imine conjugate well encapsulated in the oil core. The formation of imine bonds was first validated using a push-pull pyrene aldehyde dye, which changes its emission color during the reaction. The conjugate formation was independently confirmed by mass spectrometry. As a result, LipoAmine-loaded NEs spontaneously loaded cargo-aldehydes, yielding formulations stable against leakage at pH 7.4, which can further release the cargo in a low pH range (4-6) in solutions and living cells. Using fluorescence microscopy, we showed that LipoAmine NEs can extract pyrene aldehyde dye from cells as well as from an epithelial tissue (chicken skin). Moreover, successful extraction from cells was also achieved for a highly toxic aliphatic aldehyde 4-hydroxynonenal, which allowed obtaining the proof of concept for detoxification of living cells. Taken together, these results show that the dynamic imine chemistry inside NEs can be used to develop detoxification platforms.
Collapse
Affiliation(s)
- Fei Liu
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, Illkirch 67401, France.,INSERM UMR 1260, Regenerative Nanomedicine (RNM), CRBS, Université de Strasbourg, Strasbourg 67000, France
| | - Nicolas Anton
- INSERM UMR 1260, Regenerative Nanomedicine (RNM), CRBS, Université de Strasbourg, Strasbourg 67000, France
| | - Yosuke Niko
- Research and Education Faculty, Multidisciplinary Science Cluster, Interdisciplinary Science Unit, Kochi University, 2-5-1, Akebono-cho, Kochi-shi, Kochi 780-8520, Japan
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, Illkirch 67401, France
| |
Collapse
|
33
|
Jardim-Perassi BV, Irrera P, Lau JYC, Budzevich M, Whelan CJ, Abrahams D, Ruiz E, Ibrahim-Hashim A, Damgaci Erturk S, Longo DL, Pilon-Thomas SA, Gillies RJ. Intraperitoneal Delivery of Iopamidol to Assess Extracellular pH of Orthotopic Pancreatic Tumor Model by CEST-MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2023; 2023:1944970. [PMID: 36704211 PMCID: PMC9836819 DOI: 10.1155/2023/1944970] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/05/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023]
Abstract
The extracellular pH (pHe) of solid tumors is often acidic, as a consequence of the Warburg effect, and an altered metabolic state is often associated with malignancy. It has been shown that acidosis can promote tumor progression; thus, many therapeutic strategies have been adopted against tumor metabolism; one of these involves alkalinization therapies to raise tumor pH to inhibit tumor progression, improve immune surveillance, and overcome resistance to chemotherapies. Chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) is a noninvasive technique that can measure pH in vivo using pH-sensitive contrast agents. Iopamidol, an iodinated contrast agent, clinically used for computed tomography (CT), contains amide group protons with pH-dependent exchange rates that can reveal the pHe of the tumor microenvironment. In this study, we optimized intraperitoneal (IP) delivery of iopamidol to facilitate longitudinal assessments of orthotopic pancreatic tumor pHe by CEST-MRI. Following IV-infusion and IP-bolus injections, we compared the two protocols for assessing tumor pH. Time-resolved CT imaging was used to evaluate the uptake of iopamidol in the tumor, revealing that IP-bolus delivered a high amount of contrast agent 40 min postinjection, which was similar to the amounts reached with the IV-infusion protocol. As expected, both IP and IV injection protocols produced comparable measurements of tumor pHe, showing no statistically significant difference between groups (p=0.16). In addition, we showed the ability to conduct longitudinal monitoring of tumor pHe using CEST-MRI with the IP injection protocol, revealing a statistically significant increase in tumor pHe following bicarbonate administration (p < 0.001). In conclusion, this study shows the capability to measure pHe using an IP delivery of iopamidol into orthotopic pancreatic tumors, which is important to conduct longitudinal studies.
Collapse
Affiliation(s)
| | - Pietro Irrera
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Justin Y. C. Lau
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mikalai Budzevich
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Christopher J. Whelan
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Biological Sciences, University of Illinois, Chicago, IL, USA
| | | | - Epifanio Ruiz
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Arig Ibrahim-Hashim
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sultan Damgaci Erturk
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dario Livio Longo
- Institute of Biostructures and Bioimages (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Shari A. Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert J. Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
34
|
Longo DL, Pirotta E, Gambino R, Romdhane F, Carella A, Corrado A. Tumor pH Imaging Using Chemical Exchange Saturation Transfer (CEST)-MRI. Methods Mol Biol 2023; 2614:287-311. [PMID: 36587132 DOI: 10.1007/978-1-0716-2914-7_18] [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: 01/02/2023]
Abstract
Magnetic resonance imaging (MRI) is a noninvasive imaging technique that allows for physiological and functional studies of the tumor microenvironment. Within MRI, the emerging field of chemical exchange saturation transfer (CEST) has been largely exploited for assessing a salient feature of all solid tumors, extracellular acidosis. Iopamidol-based tumor pH imaging has been demonstrated to provide accurate and high spatial resolution extracellular tumor pH maps to elucidate tumor aggressiveness and for assessing response to therapy, with a high potential for clinical translation. Here, we describe the overall setup and steps for measuring tumor extracellular pH of tumor models in mice by exploiting MRI-CEST pH imaging with a preclinical MRI scanner following the administration of iopamidol. We address issues of pH calibration curve setup, animal handling, pH-responsive contrast agent injection, acquisition protocol, and image processing for accurate quantification and visualization of tumor acidosis.
Collapse
Affiliation(s)
- Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Torino, Italy.
| | - Elisa Pirotta
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Torino, Italy
| | - Riccardo Gambino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Feriel Romdhane
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Torino, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Torino, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Torino, Italy
| |
Collapse
|
35
|
Tang JLY, Moonshi SS, Ta HT. Nanoceria: an innovative strategy for cancer treatment. Cell Mol Life Sci 2023; 80:46. [PMID: 36656411 PMCID: PMC9851121 DOI: 10.1007/s00018-023-04694-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023]
Abstract
Nanoceria or cerium oxide nanoparticles characterised by the co-existing of Ce3+ and Ce4+ that allows self-regenerative, redox-responsive dual-catalytic activities, have attracted interest as an innovative approach to treating cancer. Depending on surface characteristics and immediate environment, nanoceria exerts either anti- or pro-oxidative effects which regulate reactive oxygen species (ROS) levels in biological systems. Nanoceria mimics ROS-related enzymes that protect normal cells at physiological pH from oxidative stress and induce ROS production in the slightly acidic tumour microenvironment to trigger cancer cell death. Nanoceria as nanozymes also generates molecular oxygen that relieves tumour hypoxia, leading to tumour cell sensitisation to improve therapeutic outcomes of photodynamic (PDT), photothermal (PTT) and radiation (RT), targeted and chemotherapies. Nanoceria has been engineered as a nanocarrier to improve drug delivery or in combination with other drugs to produce synergistic anti-cancer effects. Despite reported preclinical successes, there are still knowledge gaps arising from the inadequate number of studies reporting findings based on physiologically relevant disease models that accurately represent the complexities of cancer. This review discusses the dual-catalytic activities of nanoceria responding to pH and oxygen tension gradient in tumour microenvironment, highlights the recent nanoceria-based platforms reported to be feasible direct and indirect anti-cancer agents with protective effects on healthy tissues, and finally addresses the challenges in clinical translation of nanoceria based therapeutics.
Collapse
Affiliation(s)
- Joyce L. Y. Tang
- grid.1022.10000 0004 0437 5432Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111 Australia ,grid.1022.10000 0004 0437 5432Bioscience Discipline Department, School of Environment and Science, Griffith University, Nathan Campus, Brisbane, QLD 4111 Australia
| | - Shehzahdi S. Moonshi
- grid.1022.10000 0004 0437 5432Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111 Australia
| | - Hang T. Ta
- grid.1022.10000 0004 0437 5432Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111 Australia ,grid.1022.10000 0004 0437 5432Bioscience Discipline Department, School of Environment and Science, Griffith University, Nathan Campus, Brisbane, QLD 4111 Australia ,grid.1003.20000 0000 9320 7537Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD 4072 Australia
| |
Collapse
|
36
|
Abdella S, Abid F, Youssef SH, Kim S, Afinjuomo F, Malinga C, Song Y, Garg S. pH and its applications in targeted drug delivery. Drug Discov Today 2023; 28:103414. [PMID: 36273779 DOI: 10.1016/j.drudis.2022.103414] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 02/02/2023]
Abstract
Physiologic pH is vital for the normal functioning of tissues and varies in different parts of the body. The varying pH of the body has been exploited to design pH-sensitive smart oral, transdermal and vaginal drug delivery systems (DDS). The DDS demonstrated promising results in hard-to-treat diseases such as cancer and Helicobacter pylori infection. In some cases, a change in pH of tissues or body fluids has also been employed as a useful diagnostic biomarker. This paper aims to comprehensively review the development and applications of pH-sensitive DDS as well as recent advances in the field.
Collapse
Affiliation(s)
- Sadikalmahdi Abdella
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia; Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Zambia St, Addis Ababa, Ethiopia
| | - Fatima Abid
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Souha H Youssef
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Sangseo Kim
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Franklin Afinjuomo
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Constance Malinga
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Yunmei Song
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation (CPI), University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
37
|
Irrera P, Roberto M, Consolino L, Anemone A, Villano D, Navarro-Tableros V, Carella A, Dastrù W, Aime S, Longo DL. Effect of Esomeprazole Treatment on Extracellular Tumor pH in a Preclinical Model of Prostate Cancer by MRI-CEST Tumor pH Imaging. Metabolites 2022; 13:metabo13010048. [PMID: 36676972 PMCID: PMC9866131 DOI: 10.3390/metabo13010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Novel anticancer treatments target the pH regulating system that plays a major role in tumor progression by creating an acidic microenvironment, although few studies have addressed their effect on tumor acidosis. In this study, we investigated in vivo several proton pump inhibitors (PPIs) targeting NHE-1 (Amiloride and Cariporide) and V-ATPase (Esomeprazole and Lansoprazole) proton transporters in the DU145 androgen-insensitive human prostate cancer model. In cellulo results showed that DU145 are sensitive, with decreasing efficacy, to Amiloride, Esomeprazole and Lansoprazole, with marked cell toxicity both in normoxia and in hypoxia, with almost any change in pH. In vivo studies were performed upon administration of Esomeprazole to assess both the acute and chronic effects, and Iopamidol-based tumor pH imaging was performed to evaluate tumor acidosis. Although statistically significant tumor pH changes were observed a few hours after Esomeprazole administration in both the acute study and up to one week of treatment in the chronic study, longer treatment resulted in a lack of changes in tumor acidosis, which was associated to similar tumor growth curves between treated and control groups in both the subcutaneous and orthotopic models. Overall, this study highlights MRI-CEST tumor pH imaging as a valid approach to monitoring treatment response to PPIs.
Collapse
Affiliation(s)
- Pietro Irrera
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Turin, Italy
| | - Miriam Roberto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Lorena Consolino
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074 Aachen, Germany
| | - Annasofia Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Victor Navarro-Tableros
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Turin, Italy
| | - Walter Dastrù
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | | | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Turin, Italy
- Correspondence:
| |
Collapse
|
38
|
Ryu K, Yoshida A, Funato Y, Yamazaki D, Miki H. PRL stimulates mitotic errors by suppressing kinetochore-localized activation of AMPK during mitosis. Cell Struct Funct 2022; 47:75-87. [PMID: 36336348 PMCID: PMC10511051 DOI: 10.1247/csf.22034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/28/2022] [Indexed: 12/17/2023] Open
Abstract
Phosphatase of regenerating liver (PRL) is frequently overexpressed in various malignant cancers and is known to be a driver of malignancy. Here, we demonstrated that PRL overexpression causes mitotic errors that accompany spindle misorientation and aneuploidy, which are intimately associated with cancer progression. Mechanistic analyses of this phenomenon revealed dysregulation of the energy sensor kinase, AMP-activated protein kinase (AMPK), in PRL-induced mitotic errors. Specifically, immunofluorescence analysis showed that levels of phosphorylated AMPK (P-AMPK), an activated form of AMPK, at the kinetochore were reduced by PRL expression. Moreover, artificial activation of AMPK using chemical activators, such as A769662 and AICAR, in PRL-expressing cells restored P-AMPK signals at the kinetochore and normalized spindle orientation. Collectively, these results indicate the crucial importance of the activation of kinetochore-localized AMPK in the normal progression of mitosis, which is specifically perturbed by PRL overexpression.Key words: cancer, AMPK, PRL, kinetochore, mitotic errors.
Collapse
Affiliation(s)
- Kajung Ryu
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Yoshida
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yosuke Funato
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daisuke Yamazaki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroaki Miki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
39
|
Xiao P, Huang J, Han X, Cheu JWS, Liu Y, Law LH, Lai JHC, Li J, Park SW, Wong CCL, Lam RHW, Chan KWY. Monitor Tumor pHe and Response Longitudinally during Treatment Using CEST MRI-Detectable Alginate Microbeads. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54401-54410. [PMID: 36448714 PMCID: PMC9756293 DOI: 10.1021/acsami.2c10493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/05/2022] [Indexed: 06/17/2023]
Abstract
Imaging pHe of the tumor microenvironment has paramount importance for characterizing aggressive, invasive tumors, as well as therapeutic responses. Here, a robust approach to image pH changes in the tumor microenvironment longitudinally and during sodium bicarbonate treatment was reported. The pH-sensing microbeads were designed and prepared based on materials approved for clinical use, i.e., alginate microbead-containing computed tomography (CT) contrast-agent (iopamidol)-loaded liposomes (Iop-lipobeads). This Iop-lipobead prepared using a customized microfluidic device generated a CEST contrast of 10.6% at 4.2 ppm at pH 7.0, which was stable for 20 days in vitro. The CEST contrast decreased by 11.8% when the pH decreased from 7.0 to 6.5 in vitro. Optimized Iop-lipobeads next to tumors showed a significant increase of 19.7 ± 6.1% (p < 0.01) in CEST contrast at 4.2 ppm during the first 3 days of treatment and decreased to 15.2 ± 4.8% when treatment stopped. Notably, percentage changes in Iop-lipobeads were higher than that of amide CEST (11.7% and 9.1%) in tumors during and after treatment. These findings demonstrated that the Iop-lipobead could provide an independent and sensitive assessment of the pHe changes for a noninvasive and longitudinal monitoring of the treatment effects using multiple CEST contrast.
Collapse
Affiliation(s)
- Peng Xiao
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jianpan Huang
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Xiongqi Han
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jacinth W. S. Cheu
- Department
of Pathology, Li Ka Shing Faculty of Medicine,
The University of Hong Kong, Hong Kong, China
| | - Yang Liu
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Lok Hin Law
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Joseph H. C. Lai
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Jiyu Li
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Se Weon Park
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Carmen C. L. Wong
- Department
of Pathology, Li Ka Shing Faculty of Medicine,
The University of Hong Kong, Hong Kong, China
- State
Key Laboratory of Liver Research, The University
of Hong Kong, Hong Kong, China
| | - Raymond H. W. Lam
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
| | - Kannie W. Y. Chan
- Department
of Biomedical Engineering, City University
of Hong Kong, Hong Kong, China
- City
University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Russell
H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Tung
Biomedical
Sciences Centre, City University of Hong
Kong, Hong Kong, China
- Hong
Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| |
Collapse
|
40
|
Hu X, Ha E, Ai F, Huang X, Yan L, He S, Ruan S, Hu J. Stimulus-responsive inorganic semiconductor nanomaterials for tumor-specific theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Funato Y, Hashizume O, Miki H. Phosphatase-independent role of phosphatase of regenerating liver in cancer progression. Cancer Sci 2022; 114:25-33. [PMID: 36285487 PMCID: PMC9807511 DOI: 10.1111/cas.15625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 01/07/2023] Open
Abstract
Phosphatase of regenerating liver (PRL) is a family of protein tyrosine phosphatases (PTPs) that are anchored to the plasma membrane by prenylation. They are frequently overexpressed in various types of malignant cancers and their roles in cancer progression have received considerable attention. Mutational analyses of PRLs have shown that their intrinsic phosphatase activity is dispensable for tumor formation induced by PRL overexpression in a lung metastasis model using melanoma cells. Instead, PRLs directly bind to cyclin M (CNNM) Mg2+ exporters in the plasma membrane and potently inhibit their Mg2+ export activity, resulting in an increase in intracellular Mg2+ levels. Experiments using mammalian culture cells, mice, and C. elegans have collectively revealed that dysregulation of Mg2+ levels severely affects ATP and reactive oxygen species (ROS) levels as well as the function of Ca2+ -permeable channels. Moreover, PRL overexpression altered the optimal pH for cell proliferation from normal 7.5 to acidic 6.5, which is typically observed in malignant tumors. Here, we review the phosphatase-independent biological functions of PRLs, focusing on their interactions with CNNM Mg2+ exporters in cancer progression.
Collapse
Affiliation(s)
- Yosuke Funato
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Osamu Hashizume
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Hiroaki Miki
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan,Center for Infectious Disease Education and Research (CiDER)Osaka UniversityOsakaJapan
| |
Collapse
|
42
|
Mandal M, Banik D, Karak A, Manna SK, Mahapatra AK. Spiropyran-Merocyanine Based Photochromic Fluorescent Probes: Design, Synthesis, and Applications. ACS OMEGA 2022; 7:36988-37007. [PMID: 36312341 PMCID: PMC9608402 DOI: 10.1021/acsomega.2c04969] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/26/2022] [Indexed: 05/27/2023]
Abstract
Due to ever-increasing insights into their fundamental properties and photochromic behaviors, spiropyran derivatives are still a target of interest for researchers. The interswitching ability of this photochrome between the spiropyran (SP) and merocyanine (MC) isoforms under external stimuli (light, cations, anions, pH etc.) with different spectral properties as well as the protonation-deprotonation of its MC form allows researchers to use it suitably in sensing purposes by developing different colorimetric and fluorometric probes. Selective and sensitive recognition can be achieved by little modification of its SP moiety and functional groups. In this review, we emphasize the recent advancements (from 2019 to 2022) of spiropyran-merocyanine based fluorogenic and chromogenic probes for selective detection of various metal ions, anions, neutral analytes, and pH. We precisely explain their design strategies, sensing mechanisms, and biological and environmental applications. This review may accelerate the improvements in designing more advanced probes with innovative applications in the near future.
Collapse
Affiliation(s)
- Moumi Mandal
- Department
of Chemistry, Indian Institute of Engineering
Science and Technology, Shibpur, Howrah 711103, West Bengal, India
| | - Dipanjan Banik
- Department
of Chemistry, Indian Institute of Engineering
Science and Technology, Shibpur, Howrah 711103, West Bengal, India
| | - Anirban Karak
- Department
of Chemistry, Indian Institute of Engineering
Science and Technology, Shibpur, Howrah 711103, West Bengal, India
| | - Saikat Kumar Manna
- Department
of Chemistry, Haldia Government College, Debhog, Haldia, Purba Medinipur 721657, West Bengal, India
| | - Ajit Kumar Mahapatra
- Department
of Chemistry, Indian Institute of Engineering
Science and Technology, Shibpur, Howrah 711103, West Bengal, India
| |
Collapse
|
43
|
Bogdanov A, Bogdanov A, Chubenko V, Volkov N, Moiseenko F, Moiseyenko V. Tumor acidity: From hallmark of cancer to target of treatment. Front Oncol 2022; 12:979154. [PMID: 36106097 PMCID: PMC9467452 DOI: 10.3389/fonc.2022.979154] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor acidity is one of the cancer hallmarks and is associated with metabolic reprogramming and the use of glycolysis, which results in a high intracellular lactic acid concentration. Cancer cells avoid acid stress major by the activation and expression of proton and lactate transporters and exchangers and have an inverted pH gradient (extracellular and intracellular pHs are acid and alkaline, respectively). The shift in the tumor acid-base balance promotes proliferation, apoptosis avoidance, invasiveness, metastatic potential, aggressiveness, immune evasion, and treatment resistance. For example, weak-base chemotherapeutic agents may have a substantially reduced cellular uptake capacity due to "ion trapping". Lactic acid negatively affects the functions of activated effector T cells, stimulates regulatory T cells, and promotes them to express programmed cell death receptor 1. On the other hand, the inversion of pH gradient could be a cancer weakness that will allow the development of new promising therapies, such as tumor-targeted pH-sensitive antibodies and pH-responsible nanoparticle conjugates with anticancer drugs. The regulation of tumor pH levels by pharmacological inhibition of pH-responsible proteins (monocarboxylate transporters, H+-ATPase, etc.) and lactate dehydrogenase A is also a promising anticancer strategy. Another idea is the oral or parenteral use of buffer systems, such as sodium bicarbonate, to neutralize tumor acidity. Buffering therapy does not counteract standard treatment methods and can be used in combination to increase effectiveness. However, the mechanisms of the anticancer effect of buffering therapy are still unclear, and more research is needed. We have attempted to summarize the basic knowledge about tumor acidity.
Collapse
Affiliation(s)
- Alexey Bogdanov
- Saint Petersburg Clinical Research and Practical Center of Specialized Types of Medical Care (Oncological), Saint Petersburg, Russia
| | | | | | | | | | | |
Collapse
|
44
|
Tumor Biochemical Heterogeneity and Cancer Radiochemotherapy: Network Breakdown Zone-Model. ENTROPY 2022; 24:e24081069. [PMID: 36010733 PMCID: PMC9407148 DOI: 10.3390/e24081069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022]
Abstract
Breakdowns of two-zone random networks of the Erdős–Rényi type are investigated. They are used as mathematical models for understanding the incompleteness of the tumor network breakdown under radiochemotherapy, an incompleteness that may result from a tumor’s physical and/or chemical heterogeneity. Mathematically, having a reduced node removal probability in the network’s inner zone hampers the network’s breakdown. The latter is described quantitatively as a function of reduction in the inner zone’s removal probability, where the network breakdown is described in terms of the largest remaining clusters and their size distributions. The effects on the efficacy of radiochemotherapy due to the tumor micro-environment (TME)’s chemical make-up, and its heterogeneity, are discussed, with the goal of using such TME chemical heterogeneity imaging to inform precision oncology.
Collapse
|
45
|
Li Y, Tang K, Zhang X, Pan W, Li N, Tang B. Tumor microenvironment responsive nanocarriers for gene therapy. Chem Commun (Camb) 2022; 58:8754-8765. [PMID: 35880654 DOI: 10.1039/d2cc02759c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stimuli responsive nanocarriers are important non-viral gene carriers for gene therapy. We discuss the stimulus conditions and then highlight various stimuli responsive nanocarriers in the tumor microenvironment for cancer gene therapy. We hope that this review will inspire readers to develop more effective stimuli responsive nanocarriers for delivering genes.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Kun Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
46
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
47
|
Zhao Y, Zu Z, Xu J, Gore JC, Does MD, Li J, Gochberg DF. Mapping pH using stimulated echoes formed via chemical exchange. Magn Reson Imaging 2022; 92:100-107. [PMID: 35764217 DOI: 10.1016/j.mri.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/10/2022] [Accepted: 06/22/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE RACETE (refocused acquisition of chemical exchange transferred excitations) is a recently developed approach to imaging solute exchange with water. However, it lacks biophysical specificity, as it is sensitive to exchange rates, relaxation rates, solute concentration, and macromolecular content. We modified this sequence and developed a protocol and corresponding metric with specific sensitivity to the solute exchange rate and hence a means for mapping pH. THEORY AND METHODS RACETE splits the two gradients traditionally used in a stimulated-echo sequence into one applied after exciting solutes and one applied after exciting water, hence requiring exchange for echo formation. In this work, we leverage the dependence of the stimulated-echo signal on the exchange process. By preserving the total irradiation power and using a ratio metric, the other signal dependencies cancel, leaving a specific measure of exchange rate. Additionally, artifacts due to off-resonance excitation of water are addressed using a phase cancelling approach; and a gradient-echo imaging sequence with a variable flip angle excitation is tailored for a fast read-out of RECETE prepared signals. This method is validated using numerical simulations and salicylic acid phantom experiments at 9.4 T. RESULTS Numerical simulations and phantom experiments demonstrate that the ratio-metric is a single-variable function of exchange rate with extremely low dependence on confounding factors. Additionally, artifacts due to direct water excitation are removed and robustness to B0 and B1 inhomogeneities is demonstrated. CONCLUSION The proposed method can be used for fast pH mapping with robustness against the confounding effects that widely exist in other methods.
Collapse
Affiliation(s)
- Yu Zhao
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Junzhong Xu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark D Does
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jianqi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Daniel F Gochberg
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
48
|
Evaluation of Syrosingopine, an MCT Inhibitor, as Potential Modulator of Tumor Metabolism and Extracellular Acidification. Metabolites 2022; 12:metabo12060557. [PMID: 35736489 PMCID: PMC9230831 DOI: 10.3390/metabo12060557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 01/14/2023] Open
Abstract
Extracellular acidification has been shown to be an important characteristic of invasive tumors, as it promotes invasion and migration but also resistance to treatments. Targeting transporters involved in the regulation of tumor pH constitutes a promising anti-tumor approach, as it would disrupt cellular pH homeostasis and negatively impact tumor growth. In this study, we evaluated the impact of syrosingopine, an inhibitor of MCT1 and MCT4, as a modulator of tumor metabolism and extracellular acidification in human breast cancer (MDA-MB-231) and pharyngeal squamous cell carcinoma (FaDu) cell models. In both models in vitro, we observed that exposure to syrosingopine led to a decrease in the extracellular acidification rate, intracellular pH, glucose consumption, lactate secretion and tumor cell proliferation with an increase in the number of late apoptotic/necrotic cells. However, in vivo experiments using the MDA-MB-231 model treated with a daily injection of syrosingopine did not reveal any significant change in extracellular pH (pHe) (as measured using CEST-MRI) or primary tumor growth. Overall, our study suggests that targeting MCT could lead to profound changes in tumor cell metabolism and proliferation, and it warrants further research to identify candidates without off-target effects.
Collapse
|
49
|
Dong Z, Milak MS, Mann JJ. Proton magnetic resonance spectroscopy thermometry: Impact of separately acquired full water or partially suppressed water data on quantification and measurement error. NMR IN BIOMEDICINE 2022; 35:e4681. [PMID: 34961997 DOI: 10.1002/nbm.4681] [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: 08/23/2021] [Revised: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
In proton magnetic resonance spectroscopy (1 H MRS) thermometry, separately acquired full water and partially suppressed water are commonly used for measuring temperature. This paper compares these two approaches. Single-voxel 1 H MRS data were collected on a 3-T GE scanner from 26 human subjects. Every subject underwent five continuous MRS sessions, each separated by a 2-min phase. Each MRS session lasted 13 min and consisted of two free induction decays (FIDs) without water suppression (with full water [FW or w]) and 64 FIDs with partial water suppression (with partially suppressed water [PW or w']). Frequency differences between the two FWs, the first two PWs, the second FW and the first PW (FW2 , PW1 ), or between averaged water ( wav' ) and N-acetylaspartate (NAA), were measured. Intrasubject and intersubject variations of the frequency differences were used as a metric for the error in temperature measurement. The intrasubject variations of frequency differences between FW2 and PW1fw2-fw1' , calculated from the five MRS sessions for each subject, were larger than those between the two FWs or between the first two PWs (p = 1.54 x 10-4 and p = 1.72 x 10-4 , respectively). The mean values of intrasubject variations of fw2-fw1' for all subjects were 4.7 and 4.5 times those of fw2-fw1 and fw2'-fw1' , respectively. The intrasubject variations of the temperatures based on frequency differences, fw2-fNAA or ( fw1'-fNAA ), were about 2.5 times greater than those based on averaged water and NAA frequencies (fwav'-fNAA ). The mean temperature measured from (fwav'-fNAA ) (n = 26) was 0.29°C lower than that measured from fw2-fNAA and was 0.83°C higher than that from ( fw1'-fNAA ). It was concluded that the use of separately acquired unsuppressed or partially suppressed water signals may result in large errors in frequency and, consequently, temperature measurement.
Collapse
Affiliation(s)
- Zhengchao Dong
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, New York, USA
- Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA
| | - Matthew S Milak
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, New York, USA
- Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, New York, USA
- Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA
- Department of Radiology, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
50
|
Rizzo R, Onesto V, Forciniti S, Chandra A, Prasad S, Iuele H, Colella F, Gigli G, Del Mercato LL. A pH-sensor scaffold for mapping spatiotemporal gradients in three-dimensional in vitro tumour models. Biosens Bioelectron 2022; 212:114401. [PMID: 35617754 DOI: 10.1016/j.bios.2022.114401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022]
Abstract
The detection of extracellular pH at single cell resolution is challenging and requires advanced sensibility. Sensing pH at high spatial and temporal resolution might provide crucial information in understanding the role of pH and its fluctuations in a wide range of physio-pathological cellular processes, including cancer. Here, a method to embed silica-based fluorescent pH sensors into alginate-based three-dimensional (3D) microgels tumour models, coupled with a computational method for fine data analysis, is presented. By means of confocal laser scanning microscopy, live-cell time-lapse imaging of 3D alginate microgels was performed and the extracellular pH metabolic variations were monitored in both in vitro 3D mono- and 3D co-cultures of tumour and stromal pancreatic cells. The results show that the extracellular pH is cell line-specific and time-dependent. Moreover, differences in pH were also detected between 3D monocultures versus 3D co-cultures, thus suggesting the existence of a metabolic crosstalk between tumour and stromal cells. In conclusion, the system has the potential to image multiple live cell types in a 3D environment and to decipher in real-time their pH metabolic interplay under controlled experimental conditions, thus being also a suitable platform for drug screening and personalized medicine.
Collapse
Affiliation(s)
- Riccardo Rizzo
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy.
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Anil Chandra
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Saumya Prasad
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Francesco Colella
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento, via Arnesano, 73100, Lecce, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy.
| |
Collapse
|