1
|
Guo X, Liu Z, Wu L, Guo P. Preparation and Evaluation of Hepatoma-Targeting Glycyrrhetinic Acid Composite Micelles Loaded with Curcumin. Pharmaceuticals (Basel) 2025; 18:448. [PMID: 40283886 PMCID: PMC12030034 DOI: 10.3390/ph18040448] [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: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Liver cancer, especially hepatocellular carcinoma, a prevalent malignant tumor of the digestive system, poses significant therapeutic challenges. While traditional chemotherapy can inhibit tumor progression, its clinical application is limited by insufficient efficacy. Hydrophobic therapeutic agents further encounter challenges including low tumor specificity, poor bioavailability, and severe systemic toxicity. This study aimed to develop a liver-targeted, glutathione (GSH)-responsive micellar system to synergistically enhance drug delivery and antitumor efficacy. Methods: A GSH-responsive disulfide bond was chemically synthesized to conjugate glycyrrhetinic acid (GA) with curcumin (Cur) at a molar ratio of 1:1, forming a prodrug Cur-GA (CGA). This prodrug was co-assembled with glycyrrhizic acid (GL) at a 300% w/w loading ratio into micelles. The system was characterized for physicochemical properties, in vitro drug release in PBS (7.4) without GSH and in PBS (5.0) with 0, 5, or 10 mM GSH, cellular uptake in HepG2 cells, and in vivo efficacy in H22 hepatoma-bearing BALB/c mice. Results: The optimized micelles exhibited a hydrodynamic diameter of 157.67 ± 2.14 nm (PDI: 0.20 ± 0.02) and spherical morphology under TEM. The concentration of CUR in micelles can reach 1.04 mg/mL. In vitro release profiles confirmed GSH-dependent drug release, with 67.5% vs. <40% cumulative Cur release observed at 24 h with/without 10 mM GSH. Flow cytometry and high-content imaging revealed 1.8-fold higher cellular uptake of CGA-GL micelles compared to free drug (p < 0.001). In vivo, CGA-GL micelles achieving 3.6-fold higher tumor accumulation than non-targeted controls (p < 0.001), leading to 58.7% tumor volume reduction (p < 0.001). Conclusions: The GA/GL-based micellar system synergistically enhanced efficacy through active targeting and stimuli-responsive release, providing a promising approach to overcome current limitations in hydrophobic drug delivery for hepatocellular carcinoma therapy.
Collapse
Affiliation(s)
- Xueli Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.G.); (Z.L.); (L.W.)
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhongyan Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.G.); (Z.L.); (L.W.)
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lina Wu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.G.); (Z.L.); (L.W.)
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pan Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.G.); (Z.L.); (L.W.)
- Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
2
|
Wang X, Kutschat AP, Aggrey-Fynn J, Hamdan FH, Graham RP, Wixom AQ, Souto Y, Ladigan-Badura S, Yonkus JA, Abdelrahman AM, Alva-Ruiz R, Gaedcke J, Ströbel P, Kosinsky RL, Wegwitz F, Hermann P, Truty MJ, Siveke JT, Hahn SA, Hessmann E, Johnsen SA, Najafova Z. Identification of a ΔNp63-Dependent Basal-Like A Subtype-Specific Transcribed Enhancer Program (B-STEP) in Aggressive Pancreatic Ductal Adenocarcinoma. Mol Cancer Res 2023; 21:881-891. [PMID: 37279184 PMCID: PMC10542885 DOI: 10.1158/1541-7786.mcr-22-0916] [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: 11/14/2022] [Revised: 04/13/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023]
Abstract
A major hurdle to the application of precision oncology in pancreatic cancer is the lack of molecular stratification approaches and targeted therapy for defined molecular subtypes. In this work, we sought to gain further insight and identify molecular and epigenetic signatures of the Basal-like A pancreatic ductal adenocarcinoma (PDAC) subgroup that can be applied to clinical samples for patient stratification and/or therapy monitoring. We generated and integrated global gene expression and epigenome mapping data from patient-derived xenograft models to identify subtype-specific enhancer regions that were validated in patient-derived samples. In addition, complementary nascent transcription and chromatin topology (HiChIP) analyses revealed a Basal-like A subtype-specific transcribed enhancer program in PDAC characterized by enhancer RNA (eRNA) production that is associated with more frequent chromatin interactions and subtype-specific gene activation. Importantly, we successfully confirmed the validity of eRNA detection as a possible histologic approach for PDAC patient stratification by performing RNA-ISH analyses for subtype-specific eRNAs on pathologic tissue samples. Thus, this study provides proof-of-concept that subtype-specific epigenetic changes relevant for PDAC progression can be detected at a single-cell level in complex, heterogeneous, primary tumor material. IMPLICATIONS Subtype-specific enhancer activity analysis via detection of eRNAs on a single-cell level in patient material can be used as a potential tool for treatment stratification.
Collapse
Affiliation(s)
- Xin Wang
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Ana P. Kutschat
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Joana Aggrey-Fynn
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Feda H. Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Alexander Q. Wixom
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yara Souto
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | | | - Jennifer A. Yonkus
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amro M. Abdelrahman
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Roberto Alva-Ruiz
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jochen Gaedcke
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Clinical Research Unit 5002, KFO5002, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Philipp Ströbel
- Clinical Research Unit 5002, KFO5002, University Medical Center Göttingen, 37075 Göttingen, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Robyn Laura Kosinsky
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Florian Wegwitz
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | | | - Mark J. Truty
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jens T. Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), Partner site University Hospital Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan A. Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Elisabeth Hessmann
- Clinical Research Unit 5002, KFO5002, University Medical Center Göttingen, 37075 Göttingen, Germany
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Zeynab Najafova
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| |
Collapse
|
3
|
Toplak Ž, Hendrickx LA, Abdelaziz R, Shi X, Peigneur S, Tomašič T, Tytgat J, Peterlin-Mašič L, Pardo LA. Overcoming challenges of HERG potassium channel liability through rational design: Eag1 inhibitors for cancer treatment. Med Res Rev 2021; 42:183-226. [PMID: 33945158 DOI: 10.1002/med.21808] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/18/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
Two decades of research have proven the relevance of ion channel expression for tumor progression in virtually every indication, and it has become clear that inhibition of specific ion channels will eventually become part of the oncology therapeutic arsenal. However, ion channels play relevant roles in all aspects of physiology, and specificity for the tumor tissue remains a challenge to avoid undesired effects. Eag1 (KV 10.1) is a voltage-gated potassium channel whose expression is very restricted in healthy tissues outside of the brain, while it is overexpressed in 70% of human tumors. Inhibition of Eag1 reduces tumor growth, but the search for potent inhibitors for tumor therapy suffers from the structural similarities with the cardiac HERG channel, a major off-target. Existing inhibitors show low specificity between the two channels, and screenings for Eag1 binders are prone to enrichment in compounds that also bind HERG. Rational drug design requires knowledge of the structure of the target and the understanding of structure-function relationships. Recent studies have shown subtle structural differences between Eag1 and HERG channels with profound functional impact. Thus, although both targets' structure is likely too similar to identify leads that exclusively bind to one of the channels, the structural information combined with the new knowledge of the functional relevance of particular residues or areas suggests the possibility of selective targeting of Eag1 in cancer therapies. Further development of selective Eag1 inhibitors can lead to first-in-class compounds for the treatment of different cancers.
Collapse
Affiliation(s)
- Žan Toplak
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Louise A Hendrickx
- Department of Toxicology and Pharmacology, University of Leuven, Leuven, Belgium
| | - Reham Abdelaziz
- AG Oncophysiology, Max-Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Xiaoyi Shi
- AG Oncophysiology, Max-Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Steve Peigneur
- Department of Toxicology and Pharmacology, University of Leuven, Leuven, Belgium
| | - Tihomir Tomašič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Jan Tytgat
- Department of Toxicology and Pharmacology, University of Leuven, Leuven, Belgium
| | | | - Luis A Pardo
- AG Oncophysiology, Max-Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
4
|
Li Y, Zhou Y, Yue X, Dai Z. Cyanine Conjugate-Based Biomedical Imaging Probes. Adv Healthc Mater 2020; 9:e2001327. [PMID: 33000915 DOI: 10.1002/adhm.202001327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/11/2020] [Indexed: 12/12/2022]
Abstract
Cyanine is a class of fluorescent dye with meritorious fluorescence properties and has motivated numerous researchers to explore its imaging capabilities by miscellaneous structural modification and functionalization strategies. The covalent conjugation with other functional molecules represents a distinctive design strategy and has shown immense potential in both basic and clinical research. This review article summarizes recent achievements in cyanine conjugate-based probes for biomedical imaging. Particular attention is paid to the conjugation with targeting warheads and other contrast agents for targeted fluorescence imaging and multimodal imaging, respectively. Additionally, their clinical potential in cancer diagnostics is highlighted and some concurrent impediments for clinical translation are discussed.
Collapse
Affiliation(s)
- Yang Li
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Yiming Zhou
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Xiuli Yue
- School of Environment Harbin Institute of Technology Harbin 150090 China
| | - Zhifei Dai
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| |
Collapse
|
5
|
Morlandt AB, Moore LS, Johnson AO, Smith CM, Stevens TM, Warram JM, MacDougall M, Rosenthal EL, Amm HM. Fluorescently Labeled Cetuximab-IRDye800 for Guided Surgical Excision of Ameloblastoma: A Proof of Principle Study. J Oral Maxillofac Surg 2020; 78:1736-1747. [PMID: 32554066 PMCID: PMC7541684 DOI: 10.1016/j.joms.2020.05.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE Fluorescently labeled epidermal growth factor receptor (EGFR) antibodies have successfully identified microscopic tumors in multiple in vivo models of human cancers with limited toxicity. The present study sought to demonstrate the ability of fluorescently labeled anti-EGFR, cetuximab-IRDye800, to localize to ameloblastoma (AB) tumor cells in vitro and in vivo. MATERIAL AND METHODS EGFR expression in AB cells was confirmed by quantitative real-time polymerase chain reaction and immunohistochemistry. Primary AB cells were labeled in vitro with cetuximab-IRDye800 or nonspecific IgG-IRDye800. An in vivo patient-derived xenograft (PDX) model of AB was developed. The tumor tissue from 3 patients was implanted subcutaneously into immunocompromised mice. The mice received an intravenous injection of cetuximab-IRDye800 or IgG-IRDye800 and underwent imaging to detect infrared fluorescence using a Pearl imaging system (LI-COR Biosciences, Lincoln, NE). After resection of the overlying skin, the tumor/background ratios (TBRs) were calculated and statistically analyzed using a paired t test. RESULTS EGFR expression was seen in all AB samples. Tumor-specific labeling was achieved, as evidenced by a positive fluorescence signal from cetuximab-IRDye800 binding to AB cells, with little staining seen in the negative controls treated with IgG-IRDye800. In the animal PDX model, imaging revealed that the TBRs produced by cetuximab were significantly greater than those produced by IgG on days 7 to 14 for AB-20 tumors. After skin flap removal to simulate a preresection state, the TBRs increased with cetuximab and were significantly greater than the TBRs with the IgG control for PDX tumors derived from the 3 patients with AB. The excised tissues were embedded in paraffin and examined to confirm the presence of tumor. CONCLUSIONS Fluorescently labeled anti-EGFR demonstrated specificity for AB cells and PDX tumors. The present study is the first report of tumor-specific, antibody-based imaging of odontogenic tumors, of which AB is one of the most clinically aggressive. We expect this technology will ultimately assist surgeons treating AB by helping to accurately assess the tumor margins during surgery, leading to improved long-term local tumor control and less surgical morbidity.
Collapse
Affiliation(s)
- Anthony B Morlandt
- Associate Professor and Section Chief, Division of Oral Oncology, Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Lindsay S Moore
- Resident, Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | - Aubrey O Johnson
- Student, Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Caris M Smith
- Researcher II, Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Todd M Stevens
- Associate Professor, Department of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Jason M Warram
- Associate Professor, Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | - Mary MacDougall
- Dean and Professor, Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada
| | - Eben L Rosenthal
- Professor, Division of Otolaryngology - Head and Neck Surgery, and Associate Director, Department of Clinical Care, Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Hope M Amm
- Assistant Professor, Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
6
|
Ion Channels in Cancer: Orchestrators of Electrical Signaling and Cellular Crosstalk. Rev Physiol Biochem Pharmacol 2020; 183:103-133. [PMID: 32894333 DOI: 10.1007/112_2020_48] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ion channels are pore-forming transmembrane proteins that govern ion flux to regulate a myriad of biological processes in development, physiology, and disease. Across various types of cancer, ion channel expression and activity are often dysregulated. We review the contribution of ion channels to multiple stages of tumorigenesis based on data from in vivo model systems. As intertumoral and intratumoral heterogeneities are major obstacles in developing effective therapies, we provide perspectives on how ion channels in tumor cells and their microenvironment represent targetable vulnerabilities in the areas of tumor-stromal cell interactions, cancer neuroscience, and cancer mechanobiology.
Collapse
|
7
|
Potier-Cartereau M, Raoul W, Weber G, Mahéo K, Rapetti-Mauss R, Gueguinou M, Buscaglia P, Goupille C, Le Goux N, Abdoul-Azize S, Lecomte T, Fromont G, Chantome A, Mignen O, Soriani O, Vandier C. Potassium and Calcium Channel Complexes as Novel Targets for Cancer Research. Rev Physiol Biochem Pharmacol 2020; 183:157-176. [PMID: 32767122 DOI: 10.1007/112_2020_24] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intracellular Ca2+ concentration is mainly controlled by Ca2+ channels. These channels form complexes with K+ channels, which function to amplify Ca2+ flux. In cancer cells, voltage-gated/voltage-dependent Ca2+ channels and non-voltage-gated/voltage-independent Ca2+ channels have been reported to interact with K+ channels such as Ca2+-activated K+ channels and voltage-gated K+ channels. These channels are activated by an increase in cytosolic Ca2+ concentration or by membrane depolarization, which induces membrane hyperpolarization, increasing the driving force for Ca2+ flux. These complexes, composed of K+ and Ca2+ channels, are regulated by several molecules including lipids (ether lipids and cholesterol), proteins (e.g. STIM), receptors (e.g. S1R/SIGMAR1), and peptides (e.g. LL-37) and can be targeted by monoclonal antibodies, making them novel targets for cancer research.
Collapse
Affiliation(s)
| | - William Raoul
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | - Gunther Weber
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | - Karine Mahéo
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | | | | | - Paul Buscaglia
- LBAI UMR 1227, University of Brest, INSERM, Brest, France
| | - Caroline Goupille
- N2C UMR 1069, University of Tours, INSERM, CHRU de Tours, Tours, France
| | - Nelig Le Goux
- LBAI UMR 1227, University of Brest, INSERM, Brest, France
| | | | - Thierry Lecomte
- EA 7501 GICC, University of Tours, CHRU de Tours, Department of Hepato-Gastroenterology and Digestive Oncology, Tours, France
| | - Gaëlle Fromont
- N2C UMR 1069, University of Tours, INSERM, CHRU de Tours, Department of Pathology, Tours, France
| | | | - Olivier Mignen
- LBAI UMR 1227, University of Brest, INSERM, Brest, France
| | - Olivier Soriani
- iBV, INSERM, CNRS, University of the Côte d'Azur, Nice, France
| | | |
Collapse
|
8
|
Near-Infrared Fluorescent Imaging for Monitoring of Treatment Response in Endometrial Carcinoma Patient-Derived Xenograft Models. Cancers (Basel) 2020; 12:cancers12020370. [PMID: 32041116 PMCID: PMC7072497 DOI: 10.3390/cancers12020370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/16/2023] Open
Abstract
Imaging of clinically relevant preclinical animal models is critical to the development of personalized therapeutic strategies for endometrial carcinoma. Although orthotopic patient-derived xenografts (PDXs) reflecting heterogeneous molecular subtypes are considered the most relevant preclinical models, their use in therapeutic development is limited by the lack of appropriate imaging modalities. Here, we describe molecular imaging of a near-infrared fluorescently labeled monoclonal antibody targeting epithelial cell adhesion molecule (EpCAM) as an in vivo imaging modality for visualization of orthotopic endometrial carcinoma PDX. Application of this near-infrared probe (EpCAM-AF680) enabled both spatio-temporal visualization of development and longitudinal therapy monitoring of orthotopic PDX. Notably, EpCAM-AF680 facilitated imaging of multiple PDX models representing different subtypes of the disease. Thus, the combined implementation of EpCAM-AF680 and orthotopic PDX models creates a state-of-the-art preclinical platform for identification and validation of new targeted therapies and corresponding response predicting markers for endometrial carcinoma.
Collapse
|
9
|
Hernandez-Resendiz I, Hartung F, Pardo LA. Antibodies Targeting K V Potassium Channels: A Promising Treatment for Cancer. Bioelectricity 2019; 1:180-187. [PMID: 34471820 DOI: 10.1089/bioe.2019.0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated potassium channels are transmembrane proteins that allow flow of potassium across the membrane to regulate ion homeostasis, cell proliferation, migration, cell volume, and specific processes such as muscular contraction. Aberrant function or expression of potassium channels can underlie pathologies ranging from heart arrhythmia to cancer; the expression of potassium channels is altered in many types of cancer and that alteration correlates with malignancy and poor prognosis. Targeting potassium channels therefore constitutes a promising approach for cancer therapy. In this review, we discuss strategies to target a particular family of potassium channels, the voltage-gated potassium channels (KV) where a reasonable structural understanding is available. We also discuss the possible obstacles and advantages of such a strategy.
Collapse
Affiliation(s)
| | - Franziska Hartung
- AG Oncophysiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Luis A Pardo
- AG Oncophysiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
10
|
Aung T, Asam C, Haerteis S. Ion channels in sarcoma: pathophysiology and treatment options. Pflugers Arch 2019; 471:1163-1171. [PMID: 31377822 DOI: 10.1007/s00424-019-02299-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas are characterized by aggressive growth and a high metastasis potentially leading in most cases to a lethal outcome. These malignant tumors of the connective tissue have a high heterogeneity with numerous genetic mutations resulting in more than 100 types of sarcoma that can be grouped into two main kinds: soft tissue sarcoma and bone sarcoma. Sarcomas are often diagnosed at late disease stage, whereas a guaranteed diagnosis of the sarcoma type is fundamental for successful therapy. However, there is no appropriate therapy available. Therefore, the need for new therapies, which prolong survival and improve quality of life, is high. In the last two decades, the role of ion channels in cancer has emerged. Ion channels seem to be an ideal target for anti-tumor therapies. However, different cancer types have their own altered ion channel pattern, and the knowledge about the tumor-associated ion channel expression is fundamental. Here, we focus on the role of different ion channels in sarcoma, their pathophysiology, and possible treatment options.
Collapse
Affiliation(s)
- Thiha Aung
- Abteilung für Plastische, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Claudia Asam
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany
| | - Silke Haerteis
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
11
|
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, Lehen'kyi V. Monoclonal Antibodies Targeting Ion Channels and Their Therapeutic Potential. Front Pharmacol 2019; 10:606. [PMID: 31231216 PMCID: PMC6561378 DOI: 10.3389/fphar.2019.00606] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) represent a rapidly growing pharmaceutical class of protein drugs that becomes an important part of the precision therapy. mAbs are characterized by their high specificity and affinity for the target antigen, which is mostly present on the cell surface. Ion channels are a large family of transmembrane proteins that control ion transport across the cell membrane. They are involved in almost all biological processes in both health and disease and are widely considered as prospective targets. However, no antibody-based drug targeting ion channel has been developed so far that has progressed to clinical use. Thus, we provide a comprehensive review of the elaborated mAbs against ion channels, describe their mechanisms of action, and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Aline Hantute-Ghesquier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - V'yacheslav Lehen'kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France.,FONDATION ARC, Villejuif, France
| |
Collapse
|
12
|
Debie P, Hernot S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front Pharmacol 2019; 10:510. [PMID: 31139085 PMCID: PMC6527780 DOI: 10.3389/fphar.2019.00510] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Fluorescence imaging is an emerging technology that can provide real-time information about the operating field during cancer surgery. Non-specific fluorescent agents, used for the assessment of blood flow and sentinel lymph node detection, have so far dominated this field. However, over the last decade, several clinical studies have demonstrated the great potential of targeted fluorescent tracers to visualize tumor lesions in a more specific way. This has led to an exponential growth in the development of novel molecular fluorescent contrast agents. In this review, the design of fluorescent molecular tracers will be discussed, with particular attention for agents and approaches that are of interest for clinical translation.
Collapse
Affiliation(s)
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
13
|
Hutchings CJ, Colussi P, Clark TG. Ion channels as therapeutic antibody targets. MAbs 2018; 11:265-296. [PMID: 30526315 PMCID: PMC6380435 DOI: 10.1080/19420862.2018.1548232] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
It is now well established that antibodies have numerous potential benefits when developed as therapeutics. Here, we evaluate the technical challenges of raising antibodies to membrane-spanning proteins together with enabling technologies that may facilitate the discovery of antibody therapeutics to ion channels. Additionally, we discuss the potential targeting opportunities in the anti-ion channel antibody landscape, along with a number of case studies where functional antibodies that target ion channels have been reported. Antibodies currently in development and progressing towards the clinic are highlighted.
Collapse
Affiliation(s)
| | | | - Theodore G. Clark
- TetraGenetics Inc, Arlington Massachusetts, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca New York, USA
| |
Collapse
|
14
|
Lefranc F, Le Rhun E, Kiss R, Weller M. Glioblastoma quo vadis: Will migration and invasiveness reemerge as therapeutic targets? Cancer Treat Rev 2018; 68:145-154. [PMID: 30032756 DOI: 10.1016/j.ctrv.2018.06.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE The purpose of the current review is to highlight, on one hand, the fact that the migratory pattern of glioma cells is the major obstacle to combat them with chemotherapy, and on the other one, the new treatment strategies to overcome this obstacle. METHODS This review surveys several membrane and extracellular molecules involved in glioma cell migration, invasiveness and resistance to apoptosis. RESULTS This review focuses on signaling pathways implicated in the positive regulation of glioblastoma cell migration, including glutamate and ion channel networks, microtubes and membrane-derived extracellular vesicles (EV) containing microRNAs. Glioma cells release glutamate to the extracellular matrix, inducing neuronal cell death, which may facilitate glioma growth and invasion. Glioma cell migration and invasion are further facilitated through ion channels and transporters that modify cellular volume. Microtubes and EV promote connections and communication among glioma cells and with the microenvironment and are associated with progression and resistance to therapy. Potential therapies linked to these pathways for glioblastoma are being developed. CONCLUSION Our view is evolving from an intracellular view of the complex intracellular signaling pathways to one of orchestral machinery, including connections between heterogeneous tumoral and nontumoral cells and with the microenvironment through channels, microtubes, and extracellular miRNA, generating different signals at different times. All of these elements give rise to a new perspective for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Florence Lefranc
- Department of Neurosurgery, Hôpital Erasme; Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium.
| | - Emilie Le Rhun
- University of Lille, U-1192, F-59000 Lille, France; Inserm, U-1192, F-59000 Lille, France; CHU Lille, General and Stereotaxic Neurosurgery Service, F-59000 Lille, France
| | | | - Michael Weller
- Department of Neurology, University Hospital & University of Zurich, Frauenklinikstrasse 26, CH-8091 Zurich, Switzerland
| |
Collapse
|
15
|
Cázares-Ordoñez V, Pardo L. Kv10.1 potassium channel: from the brain to the tumors. Biochem Cell Biol 2017; 95:531-536. [DOI: 10.1139/bcb-2017-0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The KCNH1 gene encodes the Kv10.1 (Eag1) ion channel, a member of the EAG (ether-à-go-go) family of voltage-gated potassium channels. Recent studies have demonstrated that KCHN1 mutations are implicated in Temple–Baraitser and Zimmermann–Laband syndromes and other forms of developmental deficits that all present with mental retardation and epilepsy, suggesting that Kv10.1 might be important for cognitive development in humans. Although the Kv10.1 channel is mainly expressed in the mammalian brain, its ectopic expression occurs in 70% of human cancers. Cancer cells and tumors expressing Kv10.1 acquire selective advantages that favor cancer progression through molecular mechanisms that involve several cellular pathways, indicating that protein–protein interactions may be important for Kv10.1 influence in cell proliferation and tumorigenesis. Several studies on transcriptional and post-transcriptional regulation of Kv10.1 expression have shown interesting mechanistic insights about Kv10.1 role in oncogenesis, increasing the importance of identifying the cellular factors that regulate Kv10.1 expression in tumors.
Collapse
Affiliation(s)
- V. Cázares-Ordoñez
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - L.A. Pardo
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| |
Collapse
|
16
|
Eag1 K + Channel: Endogenous Regulation and Functions in Nervous System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7371010. [PMID: 28367272 PMCID: PMC5358448 DOI: 10.1155/2017/7371010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/27/2016] [Accepted: 01/31/2017] [Indexed: 02/04/2023]
Abstract
Ether-à-go-go1 (Eag1, Kv10.1, KCNH1) K+ channel is a member of the voltage-gated K+ channel family mainly distributed in the central nervous system and cancer cells. Like other types of voltage-gated K+ channels, the EAG1 channels are regulated by a variety of endogenous signals including reactive oxygen species, rendering the EAG1 to be in the redox-regulated ion channel family. The role of EAG1 channels in tumor development and its therapeutic significance have been well established. Meanwhile, the importance of hEAG1 channels in the nervous system is now increasingly appreciated. The present review will focus on the recent progress on the channel regulation by endogenous signals and the potential functions of EAG1 channels in normal neuronal signaling as well as neurological diseases.
Collapse
|