1
|
Ding Q, Guo A, Zhang S, Gu C, Wang X, Li X, Gu M, Kim JS. Phototheranostics: An advanced approach for precise diagnosis and treatment of gynecological inflammation and tumors. Biomaterials 2025; 316:123012. [PMID: 39693783 DOI: 10.1016/j.biomaterials.2024.123012] [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: 07/09/2024] [Revised: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Gynecological inflammations have a significant impact on the daily lives of women. Meanwhile, cancers such as ovarian, cervical, and endometrial cancers pose severe threats to their physical and mental well-being. While current options such as conventional pharmacotherapy, surgical interventions, and recent advancements in immunotherapy and targeted therapy provide viable solutions, they possess limitations in effectively addressing the intricacies associated with gynecological diseases. These complexities include post-surgical complications, early cancer detection, and drug resistance. The management of these challenges, however, requires the implementation of innovative treatment modalities. Phototheranostics has emerged as a promising approach to effectively address these challenges. It not only treats inflammation and tumors efficiently but also aids in disease imaging and diagnosis. The distinguishing features of phototheranostics lie in their non-invasive nature, minimal risk of drug resistance, and precise targeting capabilities through the use of photosensitizers or photothermal agents. These distinctive features underscore its potential to revolutionize early diagnosis and treatment of gynecological conditions. This review aims to summarize the application of phototheranostics in managing gynecological inflammation and tumors while highlighting its significant potential for early disease detection and treatment.
Collapse
Affiliation(s)
- Qihang Ding
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China; Department of Chemistry, Korea University, Seoul, 02841, South Korea; Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Aoxue Guo
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China
| | - Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road Nangang District, Harbin, Heilongjiang Province, 150040, China
| | - Chuanqi Gu
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China
| | - Xinyu Wang
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China
| | - Xin Li
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China.
| | - Meijia Gu
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430072, China; Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Hubei International Science and Technology Cooperation Base for Research and Clinical techniques for Brain Glioma Diagnosis and Treatment, Wuhan, 430071, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
2
|
Liao S, Zhou M, Wang Y, Lu C, Yin B, Zhang Y, Liu H, Yin X, Song G. Emerging biomedical imaging-based companion diagnostics for precision medicine. iScience 2023; 26:107277. [PMID: 37520706 PMCID: PMC10371849 DOI: 10.1016/j.isci.2023.107277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
The tumor heterogeneity, which leads to individual variations in tumor microenvironments, causes poor prognoses and limits therapeutic response. Emerging technology such as companion diagnostics (CDx) detects biomarkers and monitors therapeutic responses, allowing identification of patients who would benefit most from treatment. However, currently, most US Food and Drug Administration-approved CDx tests are designed to detect biomarkers in vitro and ex vivo, making it difficult to dynamically report variations of targets in vivo. Various medical imaging techniques offer dynamic measurement of tumor heterogeneity and treatment response, complementing CDx tests. Imaging-based companion diagnostics allow for patient stratification for targeted medicines and identification of patient populations benefiting from alternative therapeutic methods. This review summarizes recent developments in molecular imaging for predicting and assessing responses to cancer therapies, as well as the various biomarkers used in imaging-based CDx tests. We hope this review provides informative insights into imaging-based companion diagnostics and advances precision medicine.
Collapse
Affiliation(s)
- Shiyi Liao
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Mengjie Zhou
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Youjuan Wang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Chang Lu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Baoli Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Ying Zhang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Huiyi Liu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Xia Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Guosheng Song
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
3
|
Roy S, Bag N, Bardhan S, Hasan I, Guo B. Recent Progress in NIR-II Fluorescence Imaging-guided Drug Delivery for Cancer Theranostics. Adv Drug Deliv Rev 2023; 197:114821. [PMID: 37037263 DOI: 10.1016/j.addr.2023.114821] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023]
Abstract
Fluorescence imaging in the second near-infrared window (NIR-II) has become a prevalent choice owing to its appealing advantages like deep penetration depth, low autofluorescence, decent spatiotemporal resolution, and a high signal-to-background ratio. This would expedite the innovation of NIR-II imaging-guided drug delivery (IGDD) paradigms for the improvement of the prognosis of patients with tumors. This work systematically reviews the recent progress of such NIR-II IGDD-mediated cancer therapeutics and collectively brings its essence to the readers. Special care has been taken to assess their performances based on their design approach, such as enhancing their drug loading and triggering release, designing intrinsic and extrinsic fluorophores, and/ or overcoming biological barriers. Besides, the state-of-the-art NIR-II IGDD platforms for different therapies like chemo-, photodynamic, photothermal, chemodynamic, immuno-, ion channel, gas-therapies, and multiple functions such as stimulus-responsive imaging and therapy, and monitoring of drug release and therapeutic response, have been updated. In addition, for boosting theranostic outcomes and clinical translation, the innovation directions of NIR-II IGDD platforms are summarized, including renal-clearable, biodegradable, sub-cellular targeting, and/or afterglow, chemiluminescence, X-ray excitable NIR-IGDD, and even cell therapy. This review will propel new directions for safe and efficient NIR-II fluorescence-mediated anticancer drug delivery.
Collapse
Affiliation(s)
- Shubham Roy
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China
| | - Neelanjana Bag
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Souravi Bardhan
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Ikram Hasan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
4
|
Li B, Chu F, Lu Q, Wang Y, Lane LA. Alternating stealth polymer coatings between administrations minimizes toxic and antibody immune responses towards nanomedicine treatment regimens. Acta Biomater 2021; 121:527-540. [PMID: 33285326 DOI: 10.1016/j.actbio.2020.11.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
In efforts to achieve minimal systemic toxicity and high tumor delivery efficiencies in cancer therapy, various nanomedicine formulations having stealth polymer coatings have been developed for minimizing immune cell uptake and off-target macrophage phagocyte system (MPS) organ accumulation. Despite an initial reduction in immune cell uptake, stealth nanoparticles still initiate an antibody immune response. This response acts on subsequent administrations in treatment regimens resulting in accelerated blood clearance of particles into MPS organs, particularly the liver, where they are retained for prolonged periods. Consequently, doses after the first administration in treatment regimens have diminished tumor accumulation and increased MPS toxicity. Here, we present a strategy reducing antibody responses to each dose in a treatment regimen by alternating between polyethylene-glycol and polymethyloxazoline polymers as the nanoparticle coating between administrations. In a weekly dosing regimen, we find that the first dose of particles having either coating display similar favorable pharmacokinetics and biodistributions, thus allowing the polymers to be used interchangeably. However, when maintaining the same coating in subsequent administrations, we find that particles are in circulation at the height of the antibody immune response resulting in 50-60% decreases of circulation half-lives and tumor accumulation along with 50% increases in liver accumulation. By alternating the polymers used in the nanoparticle coating between administrations, we find each dose maintains favorable in vivo behaviors at the height of the antibody immune response to the previous administration. Furthermore, our strategy increases the clearance of particles uptaken by macrophages and hepatocytes, resulting in marked decreases in hepatotoxicity.
Collapse
|
5
|
de Maar JS, Sofias AM, Porta Siegel T, Vreeken RJ, Moonen C, Bos C, Deckers R. Spatial heterogeneity of nanomedicine investigated by multiscale imaging of the drug, the nanoparticle and the tumour environment. Am J Cancer Res 2020; 10:1884-1909. [PMID: 32042343 PMCID: PMC6993242 DOI: 10.7150/thno.38625] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Genetic and phenotypic tumour heterogeneity is an important cause of therapy resistance. Moreover, non-uniform spatial drug distribution in cancer treatment may cause pseudo-resistance, meaning that a treatment is ineffective because the drug does not reach its target at sufficient concentrations. Together with tumour heterogeneity, non-uniform drug distribution causes “therapy heterogeneity”: a spatially heterogeneous treatment effect. Spatial heterogeneity in drug distribution occurs on all scales ranging from interpatient differences to intratumour differences on tissue or cellular scale. Nanomedicine aims to improve the balance between efficacy and safety of drugs by targeting drug-loaded nanoparticles specifically to tumours. Spatial heterogeneity in nanoparticle and payload distribution could be an important factor that limits their efficacy in patients. Therefore, imaging spatial nanoparticle distribution and imaging the tumour environment giving rise to this distribution could help understand (lack of) clinical success of nanomedicine. Imaging the nanoparticle, drug and tumour environment can lead to improvements of new nanotherapies, increase understanding of underlying mechanisms of heterogeneous distribution, facilitate patient selection for nanotherapies and help assess the effect of treatments that aim to reduce heterogeneity in nanoparticle distribution. In this review, we discuss three groups of imaging modalities applied in nanomedicine research: non-invasive clinical imaging methods (nuclear imaging, MRI, CT, ultrasound), optical imaging and mass spectrometry imaging. Because each imaging modality provides information at a different scale and has its own strengths and weaknesses, choosing wisely and combining modalities will lead to a wealth of information that will help bring nanomedicine forward.
Collapse
|
6
|
Ehlerding EB, Grodzinski P, Cai W, Liu CH. Big Potential from Small Agents: Nanoparticles for Imaging-Based Companion Diagnostics. ACS NANO 2018; 12:2106-2121. [PMID: 29462554 PMCID: PMC5878691 DOI: 10.1021/acsnano.7b07252] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The importance of medical imaging in the diagnosis and monitoring of cancer cannot be overstated. As personalized cancer treatments are gaining popularity, a need for more advanced imaging techniques has grown significantly. Nanoparticles are uniquely suited to fill this void, not only as imaging contrast agents but also as companion diagnostics. This review provides an overview of many ways nanoparticle imaging agents have contributed to cancer imaging, both preclinically and in the clinic, as well as charting future directions in companion diagnostics. We conclude that, while nanoparticle-based imaging agents are not without considerable scientific and developmental challenges, they enable enhanced imaging in nearly every modality, hold potential as in vivo companion diagnostics, and offer precise cancer treatment and maximize intervention efficacy.
Collapse
Affiliation(s)
- Emily B. Ehlerding
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
- Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Piotr Grodzinski
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
- Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
- Carbone Cancer Center, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Christina H. Liu
- Office of Cancer Nanotechnology Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
7
|
Nanoparticle anchoring targets immune agonists to tumors enabling anti-cancer immunity without systemic toxicity. Nat Commun 2018; 9:6. [PMID: 29295974 PMCID: PMC5750237 DOI: 10.1038/s41467-017-02251-3] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/16/2017] [Indexed: 02/01/2023] Open
Abstract
Immunostimulatory agents such as agonistic anti-CD137 and interleukin (IL)−2 generate effective anti-tumor immunity but also elicit serious toxicities, hampering their clinical application. Here we show that combination therapy with anti-CD137 and an IL-2-Fc fusion achieves significant initial anti-tumor activity, but also lethal immunotoxicity deriving from stimulation of circulating leukocytes. To overcome this toxicity, we demonstrate that anchoring IL-2 and anti-CD137 on the surface of liposomes allows these immune agonists to rapidly accumulate in tumors while lowering systemic exposure. In multiple tumor models, immunoliposome delivery achieves anti-tumor activity equivalent to free IL-2/anti-CD137 but with the complete absence of systemic toxicity. Immunoliposomes stimulated tumor infiltration by cytotoxic lymphocytes, cytokine production, and granzyme expression, demonstrating equivalent immunostimulatory effects to the free drugs in the local tumor microenvironment. Thus, surface-anchored particle delivery may provide a general approach to exploit the potent stimulatory activity of immune agonists without debilitating systemic toxicities. Immunostimulatory agents used in cancer treatment often elicit serious toxicities, limiting their clinical application. Here, the authors show that the use of liposomes to intravenously deliver surface-anchored IL-2 and anti-CD137 proteins enables anti-cancer immunity and reduces the toxic side effects.
Collapse
|
8
|
Björnmalm M, Thurecht KJ, Michael M, Scott AM, Caruso F. Bridging Bio-Nano Science and Cancer Nanomedicine. ACS NANO 2017; 11:9594-9613. [PMID: 28926225 DOI: 10.1021/acsnano.7b04855] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The interface of bio-nano science and cancer medicine is an area experiencing much progress but also beset with controversy. Core concepts of the field-e.g., the enhanced permeability and retention (EPR) effect, tumor targeting and accumulation, and even the purpose of "nano" in cancer medicine-are hotly debated. In parallel, considerable advances in neighboring fields are occurring rapidly, including the recent progress of "immuno-oncology" and the fundamental impact it is having on our understanding and the clinical treatment of the group of diseases collectively known as cancer. Herein, we (i) revisit how cancer is commonly treated in the clinic and how this relates to nanomedicine; (ii) examine the ongoing debate on the relevance of the EPR effect and tumor targeting; (iii) highlight ways to improve the next-generation of nanomedicines; and (iv) discuss the emerging concept of working with (and not against) biology. While discussing these controversies, challenges, emerging concepts, and opportunities, we explore new directions for the field of cancer nanomedicine.
Collapse
Affiliation(s)
- Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Kristofer J Thurecht
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The Australian Institute for Bioengineering and Nanotechnology and The Centre for Advanced Imaging, The University of Queensland , Brisbane, Queensland 4072, Australia
| | - Michael Michael
- Division of Cancer Medicine, Peter MacCallum Cancer Centre , Melbourne, Victoria 3000, Australia
- The Peter MacCallum Department of Oncology, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University , Melbourne, Victoria 3084, Australia
- Department of Molecular Imaging and Therapy, Austin Hospital , Heidelberg, Victoria 3084, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| |
Collapse
|
9
|
Ljubimova JY, Sun T, Mashouf L, Ljubimov AV, Israel LL, Ljubimov VA, Falahatian V, Holler E. Covalent nano delivery systems for selective imaging and treatment of brain tumors. Adv Drug Deliv Rev 2017; 113:177-200. [PMID: 28606739 PMCID: PMC5578712 DOI: 10.1016/j.addr.2017.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023]
Abstract
Nanomedicine is a rapidly evolving form of therapy that holds a great promise for superior drug delivery efficiency and therapeutic efficacy than conventional cancer treatment. In this review, we attempt to cover the benefits and the limitations of current nanomedicines with special attention to covalent nano conjugates for imaging and drug delivery in the brain. The improvement in brain tumor treatment remains dismal despite decades of efforts in drug development and patient care. One of the major obstacles in brain cancer treatment is the poor drug delivery efficiency owing to the unique blood-brain barrier (BBB) in the CNS. Although various anti-cancer agents are available to treat tumors outside of the CNS, the majority fails to cross the BBB. In this regard, nanomedicines have increasingly drawn attention due to their multi-functionality and versatility. Nano drugs can penetrate BBB and other biological barriers, and selectively accumulate in tumor cells, while concurrently decreasing systemic toxicity.
Collapse
Affiliation(s)
- Julia Y Ljubimova
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA.
| | - Tao Sun
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA
| | - Leila Mashouf
- Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Alexander V Ljubimov
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Los Angeles, CA 90048, USA
| | - Liron L Israel
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA
| | - Vladimir A Ljubimov
- Department of Neurosurgery and Brain Repair, University of South Florida, 2 Tampa General Circle, Tampa, FL 33606, USA
| | - Vida Falahatian
- Duke University School of Medicine, Department of Biostatistics and Bioinformatics, Clinical Research Training Program (CRTP), 2424 Erwin Road, Suite 1102, Hock Plaza Box 2721, Durham, NC 27710, USA
| | - Eggehard Holler
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA; Institut für Biophysik und Physikalische Biochemie, Universität Regensburg, D-93040 Regensburg, Germany
| |
Collapse
|
10
|
Leitner M, Poturnayova A, Lamprecht C, Weich S, Snejdarkova M, Karpisova I, Hianik T, Ebner A. Characterization of the specific interaction between the DNA aptamer sgc8c and protein tyrosine kinase-7 receptors at the surface of T-cells by biosensing AFM. Anal Bioanal Chem 2017; 409:2767-2776. [PMID: 28229174 PMCID: PMC5366180 DOI: 10.1007/s00216-017-0238-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/18/2017] [Accepted: 01/31/2017] [Indexed: 01/10/2023]
Abstract
We studied the interaction of the specific DNA aptamer sgc8c immobilized at the AFM tip with its corresponding receptor, the protein tyrosine kinase-7 (PTK7) embedded in the membrane of acute lymphoblastic leukemia (ALL) cells (Jurkat T-cells). Performing single molecule force spectroscopy (SMFS) experiments, we showed that the aptamer sgc8c bound with high probability (38.3 ± 7.48%) and high specificity to PTK7, as demonstrated by receptor blocking experiments and through comparison with the binding behavior of a nonspecific aptamer. The determined kinetic off-rate (koff = 5.16 s−1) indicates low dissociation of the sgc8c–PTK7 complex. In addition to the pulling force experiments, simultaneous topography and recognition imaging (TREC) experiments using AFM tips functionalized with sgc8c aptamers were realized on the outer regions surface of surface-immobilized Jurkat cells for the first time. This allowed determination of the distribution of PTK7 without any labeling and at near physiological conditions. As a result, we could show a homogeneous distribution of PTK7 molecules on the outer regions of ALL cells with a surface density of 325 ± 12 PTK7 receptors (or small receptor clusters) per μm2. The specific interaction of the DNA aptamer sgc8c and protein tyrosine kinase-7 (PTK7) on acute lymphoblastic leukemia (ALL) cells was characterized. AFM based single molecule force spectroscopy (SMFS) yielded a kinetic off-rate of 5.16 s−1 of the complex. Simultaneous topography and recognition imaging (TREC) revealed a PTK7 density of 325 ± 12 molecules or clusters per μm2 in the cell membrane ![]()
Collapse
Affiliation(s)
- Michael Leitner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Alexandra Poturnayova
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia.,Institute of Biochemistry and Animal Genetics, Slovak Academy of Sciences, Moyzesova 61, 900 28, Ivanka pri Dunaji, Slovakia
| | - Constanze Lamprecht
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Sabine Weich
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Maja Snejdarkova
- Institute of Biochemistry and Animal Genetics, Slovak Academy of Sciences, Moyzesova 61, 900 28, Ivanka pri Dunaji, Slovakia
| | - Ivana Karpisova
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia
| | - Tibor Hianik
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia
| | - Andreas Ebner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria.
| |
Collapse
|