101
|
A multi-scale model for determining the effects of pathophysiology and metabolic disorders on tumor growth. Sci Rep 2020; 10:3025. [PMID: 32080250 PMCID: PMC7033139 DOI: 10.1038/s41598-020-59658-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 01/17/2020] [Indexed: 11/08/2022] Open
Abstract
The search for efficient chemotherapy drugs and other anti-cancer treatments would benefit from a deeper understanding of the tumor microenvironment (TME) and its role in tumor progression. Because in vivo experimental methods are unable to isolate or control individual factors of the TME and in vitro models often do not include all the contributing factors, some questions are best addressed with systems biology mathematical models. In this work, we present a new fully-coupled, agent-based, multi-scale mathematical model of tumor growth, angiogenesis and metabolism that includes important aspects of the TME spanning subcellular-, cellular- and tissue-level scales. The mathematical model is computationally implemented for a three-dimensional TME, and a double hybrid continuous-discrete (DHCD) method is applied to solve the governing equations. The model recapitulates the distinct morphological and metabolic stages of a solid tumor, starting with an avascular tumor and progressing through angiogenesis and vascularized tumor growth. To examine the robustness of the model, we simulated normal and abnormal blood conditions, including hyperglycemia/hypoglycemia, hyperoxemia/hypoxemia, and hypercarbia/hypocarbia - conditions common in cancer patients. The results demonstrate that tumor progression is accelerated by hyperoxemia, hyperglycemia and hypercarbia but inhibited by hypoxemia and hypoglycemia; hypocarbia had no appreciable effect. Because of the importance of interstitial fluid flow in tumor physiology, we also examined the effects of hypo- or hypertension, and the impact of decreased hydraulic conductivity common in desmoplastic tumors. The simulations show that chemotherapy-increased blood pressure, or reduction of interstitial hydraulic conductivity increase tumor growth rate and contribute to tumor malignancy.
Collapse
|
102
|
Stevenson ME, Kay JJM, Atry F, Wickstrom AT, Krueger JR, Pashaie RE, Swain RA. Wheel running for 26 weeks is associated with sustained vascular plasticity in the rat motor cortex. Behav Brain Res 2020; 380:112447. [PMID: 31870777 DOI: 10.1016/j.bbr.2019.112447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 10/25/2022]
Abstract
Vascular pathologies represent the leading causes of mortality worldwide. The nervous system has evolved mechanisms to compensate for the cerebral hypoxia caused by many of these conditions. Vessel dilation and growth of new vessels are two prominent responses to hypoxia, both of which play a critical role in maintaining cerebral homeostasis. One way to facilitate cerebrovascular plasticity, and develop neuroprotection against vascular pathologies, is through aerobic exercise. The present study explored the long-term consequences of aerobic exercise on vascular structure and function in the motor cortex. Rats were assigned to a sedentary condition or were provided access to running wheels for 26 weeks. Rats were then anesthetized, and angiograms were captured using spectral domain optical coherence tomography (SD-OCT) to explore cerebrovascular reactivity in response to altered oxygen and carbon dioxide status. Following this procedure, all rats were euthanized, and unbiased stereological quantification of blood vessel density was collected from sections of the primary motor cortex infused with India ink. Results demonstrated that chronic exercise increased capillary and arteriole surface area densities and enhanced arteriole reactivity in response to hypercapnia-hypoxia, as displayed by increased vasodilation within the motor cortex of exercised animals.
Collapse
Affiliation(s)
- Morgan E Stevenson
- Department of Psychology, University of Wisconsin-Milwaukee, United States
| | - Jacob J M Kay
- Department of Psychology, University of Wisconsin-Milwaukee, United States
| | - Farid Atry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | | | | | - Ramin E Pashaie
- Department of Electrical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Rodney A Swain
- Department of Psychology, University of Wisconsin-Milwaukee, United States.
| |
Collapse
|
103
|
Si P, Honkala A, de la Zerda A, Smith BR. Optical Microscopy and Coherence Tomography of Cancer in Living Subjects. Trends Cancer 2020; 6:205-222. [PMID: 32101724 DOI: 10.1016/j.trecan.2020.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 01/05/2020] [Accepted: 01/07/2020] [Indexed: 12/16/2022]
Abstract
Intravital microscopy (IVM) and optical coherency tomography (OCT) are two powerful optical imaging tools that allow visualization of dynamic biological activities in living subjects with subcellular resolutions. Recent advances in labeling and label-free techniques empower IVM and OCT for a wide range of preclinical and clinical cancer imaging, providing profound insights into the complex physiological, cellular, and molecular behaviors of tumors. Preclinical IVM and OCT have elucidated many otherwise inscrutable aspects of cancer biology, while clinical applications of IVM and OCT are revolutionizing cancer diagnosis and therapies. We review important progress in the fields of IVM and OCT for cancer imaging in living subjects, highlighting key technological developments and their emerging applications in fundamental cancer biology research and clinical oncology investigation.
Collapse
Affiliation(s)
- Peng Si
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Alexander Honkala
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Adam de la Zerda
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA; The Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Bryan Ronain Smith
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
104
|
Li R, Yin H, Hong J, Wang C, He B, Chen Z, Li Q, Xue P, Zhang X. Speckle reducing OCT using optical chopper. OPTICS EXPRESS 2020; 28:4021-4031. [PMID: 32122062 DOI: 10.1364/oe.382369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/23/2019] [Indexed: 06/10/2023]
Abstract
Optical coherence tomography (OCT) has been an important and powerful tool for biological research and clinical applications. However, speckle noise significantly degrades the image quality of OCT and has a negative impact on the clinical diagnosis accuracy. In this paper, we propose a novel speckle noise suppression technique which changes the spatial distribution of sample beam using a special optical chopper. Then a series of OCT images with uncorrelated speckle patterns could be captured and compounded to improve the image quality without degradation of resolution. Typical signal-to-noise ratio improvement of ∼6.4 dB is experimentally achieved in tissue phantom imaging with average number n = 100. Furthermore, compared with conventional OCT, the proposed technique is demonstrated to view finer and clearer biological structures in human skin in vivo, such as sweat glands and blood vessels. The advantages of low cost, simple structure and compact integration will benefit the future design of handheld or endoscopic probe for biomedical imaging in research and clinical applications.
Collapse
|
105
|
Rabiee N, Yaraki MT, Garakani SM, Garakani SM, Ahmadi S, Lajevardi A, Bagherzadeh M, Rabiee M, Tayebi L, Tahriri M, Hamblin MR. Recent advances in porphyrin-based nanocomposites for effective targeted imaging and therapy. Biomaterials 2020; 232:119707. [PMID: 31874428 PMCID: PMC7008091 DOI: 10.1016/j.biomaterials.2019.119707] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 12/05/2019] [Accepted: 12/18/2019] [Indexed: 12/24/2022]
Abstract
Porphyrins are organic compounds that continue to attract much theoretical interest, and have been called the "pigments of life". They have a wide role in photodynamic and sonodynamic therapy, along with uses in magnetic resonance, fluorescence and photoacoustic imaging. There is a vast range of porphyrins that have been isolated or designed, but few of them have real clinical applications. Due to the hydrophobic properties of porphyrins, and their tendency to aggregate by stacking of the planar molecules they are difficult to work with in aqueous media. Therefore encapsulating them in nanoparticles (NPs) or attachment to various delivery vehicles have been used to improve delivery characteristics. Porphyrins can be used in a composite designed material with properties that allow specific targeting, immune tolerance, extended tissue lifetime and improved hydrophilicity. Drug delivery, healing and repairing of damaged organs, and cancer theranostics are some of the medical uses of porphyrin-based nanocomposites covered in this review.
Collapse
Affiliation(s)
- Navid Rabiee
- Department of Chemistry, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Tavakkoli Yaraki
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore; Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, 138634, Singapore
| | | | | | - Sepideh Ahmadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aseman Lajevardi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Lobat Tayebi
- Department of Developmental Sciences, Marquette University, Milwaukee, WI, 53233, USA
| | - Mohammadreza Tahriri
- Department of Developmental Sciences, Marquette University, Milwaukee, WI, 53233, USA.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, USA; Department of Dermatology, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| |
Collapse
|
106
|
Chen X, Du W, Cai Z, Ji S, Dwivedi M, Chen J, Zhao G, Chu J. Uniaxial Stretching of Cell-Laden Microfibers for Promoting C2C12 Myoblasts Alignment and Myofibers Formation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:2162-2170. [PMID: 31856565 DOI: 10.1021/acsami.9b22103] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Fiber-shaped cellular constructs have attracted increasing attention in the regeneration of blood vessels, nerve networks, and skeletal myofibers. Nevertheless, the generation of functional fiber-shaped cellular constructs suffers from limited appropriate microfiber-based fabrication approaches and the maintenance of regenerated tissue functions. Herein, we demonstrate a silicone-tube-based coagulant bath free method to fabricate tens of centimeters long cell-laden microfibers using single UV exposure without pretreatment of nozzles or microchannels. By modulating the exposure time, the gelatin methacrylate microfibers with tissue-like microstructures and mechanical properties are obtained. Then, a culture system integrated with a pillar well-array based stretching device is used to apply uniaxial stretching with various strain ratios in situ to cell-laden microfibers in a 60 mm petri dish. Cells with improved spreading, elongation, and alignment are obtained under uniaxial stretching. Moreover, the promotional effects of uniaxial stretching on the differentiation of C2C12 myoblasts, the formation, and contractility of myofibers become more pronounced with increasing strain ratio and achieve saturation level as strain ratio up to ∼35%.
Collapse
Affiliation(s)
| | - Wenqiang Du
- Nationwide Children's Hospital , Columbus , Ohio 43205 , United States
- The Ohio State University College of Medicine , Columbus , Ohio 43205 , United States
| | | | | | | | - Jianfeng Chen
- Department of Mechanics Engineering , Nanchang University , Nanchang , Jiangxi 330031 , China
| | | | | |
Collapse
|
107
|
Si P, Shevidi S, Yuan E, Yuan K, Lautman Z, Jeffrey SS, Sledge GW, de la Zerda A. Gold Nanobipyramids as Second Near Infrared Optical Coherence Tomography Contrast Agents for in Vivo Multiplexing Studies. NANO LETTERS 2020; 20:101-108. [PMID: 31585502 DOI: 10.1021/acs.nanolett.9b03344] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Developing contrast-enhanced optical coherence tomography (OCT) techniques is important for specific imaging of tissue lesions, molecular imaging, cell-tracking, and highly sensitive microangiography and lymphangiography. Multiplexed OCT imaging in the second near-infrared (NIR-II) window is highly desirable since it allows simultaneous imaging and tracking of multiple biological events in high resolution with deeper tissue penetration in vivo. Here we demonstrate that gold nanobipyramids can function as OCT multiplexing contrast agents, allowing high-resolution imaging of two separate lymphatic flows occurring simultaneously from different drainage basins into the same lymph node in a live mouse. Contrast-enhanced multiplexed lymphangiography of a melanoma tumor in vivo shows that the peritumoral lymph flow upstream of the tumor is unidirectional, and tumor is accessible to such flow. Whereas the lymphatic drainage coming out from the tumor is multidirectional. We also demonstrate real-time tracking of the contrast agents draining from a melanoma tumor specifically to the sentinel lymph node of the tumor and the three-dimensional distribution of the contrast agents in the lymph node.
Collapse
Affiliation(s)
- Peng Si
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford , Stanford , California 94305 , United States
| | - Saba Shevidi
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford , Stanford , California 94305 , United States
| | - Edwin Yuan
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford , Stanford , California 94305 , United States
| | - Ke Yuan
- Department of Medicine , Stanford University , Stanford , California 94305 , United States
| | - Ziv Lautman
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford , Stanford , California 94305 , United States
| | - Stefanie S Jeffrey
- Department of Surgery , Stanford University , Stanford , California 94305 , United States
| | - George W Sledge
- Department of Medicine , Stanford University , Stanford , California 94305 , United States
| | - Adam de la Zerda
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford , Stanford , California 94305 , United States
- Department of Electrical Engineering , Stanford University , 350 Serra Mall , Stanford , California 94305 , United States
- The Chan Zuckerberg Biohub , San Francisco , California 94158 , United States
| |
Collapse
|
108
|
Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer 2020; 20:26-41. [PMID: 31601988 PMCID: PMC8246629 DOI: 10.1038/s41568-019-0205-x] [Citation(s) in RCA: 930] [Impact Index Per Article: 232.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
For a blood-borne cancer therapeutic agent to be effective, it must cross the blood vessel wall to reach cancer cells in adequate quantities, and it must overcome the resistance conferred by the local microenvironment around cancer cells. The brain microenvironment can thwart the effectiveness of drugs against primary brain tumours as well as brain metastases. In this Review, we highlight the cellular and molecular components of the blood-brain barrier (BBB), a specialized neurovascular unit evolved to maintain brain homeostasis. Tumours are known to compromise the integrity of the BBB, resulting in a vasculature known as the blood-tumour barrier (BTB), which is highly heterogeneous and characterized by numerous distinct features, including non-uniform permeability and active efflux of molecules. We discuss the challenges posed by the BBB and BTB for drug delivery, how multiple cell types dictate BBB function and the role of the BTB in disease progression and treatment. Finally, we highlight emerging molecular, cellular and physical strategies to improve drug delivery across the BBB and BTB and discuss their impact on improving conventional as well as emerging treatments, such as immune checkpoint inhibitors and engineered T cells. A deeper understanding of the BBB and BTB through the application of single-cell sequencing and imaging techniques, and the development of biomarkers of BBB integrity along with systems biology approaches, should enable new personalized treatment strategies for primary brain malignancies and brain metastases.
Collapse
Affiliation(s)
- Costas D Arvanitis
- School of Mechanical Engineering, Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Gino B Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
109
|
Abstract
Secondary burn necrosis is the expansion and deepening of the original burn injury several days after injury. Limiting the extent of secondary burn necrosis may improve outcomes. In this study, we examined the ability of the lipid mediator of inflammation-resolution resolvin D2 (RvD2) and chromatin-lysing enzyme (DNase) to reduce secondary burn necrosis. Male Wistar rats were injured using a brass comb with 4 prongs heated in boiling water. This method created 2 parallel rows of 4 rectangular burned areas separated by 3 unburned interspaces. Starting at 2 hours after the burn injury, rats received either 25 ng/kg RvD2 intravenously daily for 7 days or 200 U/kg DNase every 12 hours for 3 days. We documented the necrosis around the initial wounds by digital photography. We used laser Doppler to assess the total blood flux in the burn area. We evaluated the functionality of the capillary network in the interspaces by optical coherence tomography angiography. We performed histological examination of wound skin tissue samples collected at 14 days postburn. We found that the interspace areas were preserved and had higher blood flow in the RvD2-treated group, while the burn areas expanded into the interspace areas, which were confluent by 7 days postburn, in the control-untreated group. We found a larger monocyte-to-neutrophil ratio in the RvD2-treated group compared with the DNase-treated and control groups (P < .05). Overall, RvD2 suppresses secondary necrosis and starts regeneration, highlighting the role of inflammation resolution as a potential therapeutic target in burn care.
Collapse
|
110
|
Li Y, Chen J, Chen Z. Advances in Doppler optical coherence tomography and angiography. TRANSLATIONAL BIOPHOTONICS 2019; 1:e201900005. [PMID: 33005888 PMCID: PMC7523705 DOI: 10.1002/tbio.201900005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/14/2019] [Indexed: 12/22/2022] Open
Abstract
Since the first demonstration of Doppler optical coherence tomography (OCT) in 1997, several functional extensions of Doppler OCT have been developed, including velocimetry, angiogram, and optical coherence elastography. These functional techniques have been widely used in research and clinical applications, particularly in ophthalmology. Here, we review the principles, representative methods, and applications of different Doppler OCT techniques, followed by discussion on the innovations, limitations, and future directions of each of these techniques.
Collapse
Affiliation(s)
- Yan Li
- Beckman Laser Institute, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Jason Chen
- Beckman Laser Institute, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Zhongping Chen
- Beckman Laser Institute, University of California, Irvine, California
- Department of Biomedical Engineering, University of California, Irvine, California
| |
Collapse
|
111
|
Wu M, Small DM, Nishimura N, Adie SG. Computed optical coherence microscopy of mouse brain ex vivo. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-18. [PMID: 31773937 PMCID: PMC6880187 DOI: 10.1117/1.jbo.24.11.116002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/18/2019] [Indexed: 05/25/2023]
Abstract
The compromise between lateral resolution and usable imaging depth range is a bottleneck for optical coherence tomography (OCT). Existing solutions for optical coherence microscopy (OCM) suffer from either large data size and long acquisition time or a nonideal point spread function. We present volumetric OCM of mouse brain ex vivo with a large depth coverage by leveraging computational adaptive optics (CAO) to significantly reduce the number of OCM volumes that need to be acquired with a Gaussian beam focused at different depths. We demonstrate volumetric reconstruction of ex-vivo mouse brain with lateral resolution of 2.2 μm, axial resolution of 4.7 μm, and depth range of ∼1.2 mm optical path length, using only 11 OCT data volumes acquired on a spectral-domain OCM system. Compared to focus scanning with step size equal to the Rayleigh length of the beam, this is a factor of 4 fewer datasets required for volumetric imaging. Coregistered two-photon microscopy confirmed that CAO-OCM reconstructions can visualize various tissue microstructures in the brain. Our results also highlight the limitations of CAO in highly scattering media, particularly when attempting to reconstruct far from the focal plane or when imaging deep within the sample.
Collapse
Affiliation(s)
- Meiqi Wu
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, New York, United States
| | - David M. Small
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, New York, United States
| | - Nozomi Nishimura
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, New York, United States
| | - Steven G. Adie
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, New York, United States
| |
Collapse
|
112
|
Huang L, Fu Y, Chen R, Yang S, Qiu H, Wu X, Zhao S, Gu Y, Li P. SNR-Adaptive OCT Angiography Enabled by Statistical Characterization of Intensity and Decorrelation With Multi-Variate Time Series Model. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:2695-2704. [PMID: 30990423 DOI: 10.1109/tmi.2019.2910871] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In OCT angiography (OCTA), decorrelation computation has been widely used as a local motion index to identify dynamic flow from static tissues, but its dependence on SNR severely degrades the vascular visibility, particularly in low-SNR regions. To mathematically characterize the decorrelation-SNR dependence of OCT signals, we developed a multi-variate time series (MVTS) model. Based on the model, we derived a universal asymptotic linear relation of decorrelation to inverse SNR (iSNR), with the variance in static and noise regions determined by the average kernel size. Accordingly, with the population distribution of static and noise voxels being explicitly calculated in the iSNR and decorrelation (ID) space, a linear classifier is developed by removing static and noise voxels at all SNR, to generate a SNR-adaptive OCTA, termed as ID-OCTA. Then, flow phantom and human skin experiments were performed to validate the proposed ID-OCTA. Both qualitative and quantitative assessments demonstrated that the ID-OCTA offers a superior visibility of blood vessels, particularly in the deep layer. Finally, the implications of this work on both system design and hemodynamic quantification are further discussed.
Collapse
|
113
|
Park J, Choi Y, Chang H, Um W, Ryu JH, Kwon IC. Alliance with EPR Effect: Combined Strategies to Improve the EPR Effect in the Tumor Microenvironment. Theranostics 2019; 9:8073-8090. [PMID: 31754382 PMCID: PMC6857053 DOI: 10.7150/thno.37198] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022] Open
Abstract
The use of nanomedicine for cancer treatment takes advantage of its preferential accumulation in tumors owing to the enhanced permeability and retention (EPR) effect. The development of cancer nanomedicine has promised highly effective treatment options unprecedented by standard therapeutics. However, the therapeutic efficacy of passively targeted nanomedicine is not always satisfactory because it is largely influenced by the heterogeneity of the intensity of the EPR effect exhibited within a tumor, at different stages of a tumor, and among individual tumors. In addition, limited data on EPR effectiveness in human hinders further clinical translation of nanomedicine. This unsatisfactory therapeutic outcome in mice and humans necessitates novel approaches to improve the EPR effect. This review focuses on current attempts at overcoming the limitations of traditional EPR-dependent nanomedicine by incorporating supplementary strategies, such as additional molecular targeting, physical alteration, or physiological remodeling of the tumor microenvironment. This review will provide valuable insight to researchers who seek to overcome the limitations of relying on the EPR effect alone in cancer nanomedicine and go "beyond the EPR effect".
Collapse
Affiliation(s)
- Jooho Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yongwhan Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hyeyoun Chang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Wooram Um
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| |
Collapse
|
114
|
Shin P, Choi W, Joo J, Oh WY. Quantitative hemodynamic analysis of cerebral blood flow and neurovascular coupling using optical coherence tomography angiography. J Cereb Blood Flow Metab 2019; 39:1983-1994. [PMID: 29757059 PMCID: PMC6775585 DOI: 10.1177/0271678x18773432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Functional hyperemia in the rat cortex was investigated using high-speed optical coherence tomography (OCT) angiography and Doppler OCT. OCT angiography (OCTA) was performed to image the hemodynamic stimulus-response over a wide field of view. Temporal changes in vessel diameters in different vessel compartments, which were determined as the diameters of erythrocyte flows in OCT angiograms, were measured in order to monitor localized hemodynamic changes. Our results showed that the dilation of arterioles at the site of activation was accompanied by the dilation of upstream arteries. Relatively negligible dilation was observed in veins. An increase in the OCTA signal was observed during stimulus in multiple capillaries, which may imply that capillary blood flow increases as a result of the expanded arterial blood volume. These results agree with previous observations using two-photon laser scanning microscopy (TPLSM). Doppler OCT was performed to quantitatively measure stimulus-induced blood flow response in pial arteries. The measurement showed small but clear hemodynamic response in upstream arteries with diameters exceeding 100 μm. Our results demonstrate the potential of OCTA and Doppler OCT for the investigation of neurovascular coupling in small animal models.
Collapse
Affiliation(s)
- Paul Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - WooJhon Choi
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - JongYoon Joo
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
115
|
Ahmed S, Galle PR, Neumann H. Molecular endoscopic imaging: the future is bright. Ther Adv Gastrointest Endosc 2019; 12:2631774519867175. [PMID: 31517311 PMCID: PMC6724493 DOI: 10.1177/2631774519867175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
The prediction and final survival rate of gastrointestinal cancers are dependent on the stage of disease. The ideal would be to detect those gastrointestinal lesions at early stage or even premalignant forms which are difficult to detect by conventional endoscopy with white light optical imaging as they show minimum or no changes in morphological characteristics and are thus left untreated. The introduction of molecular imaging has greatly changed the pattern for detecting gastrointestinal lesions from purely macroscopic structural imaging to the molecular level. It allows microscopic examination of the gastrointestinal mucosa with endoscopy after the topical or systemic application of molecular probes. In recent years, major advancements in endoscopic instruments and specific molecular probes have been achieved. This review focuses on the current status of endoscopic imaging and highlights the application of molecular imaging in gastrointestinal and hepatic disease in the context of diagnosis and therapy based on recently published literature in this field. We also discuss the challenges of molecular endoscopic imaging, its future directions and potential that could have a tremendous impact on endoscopic research and clinical practice in future.
Collapse
Affiliation(s)
- Shakil Ahmed
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Helmut Neumann
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
116
|
Zhou HC, Chen N, Zhao H, Yin T, Zhang J, Zheng W, Song L, Liu C, Zheng R. Optical-resolution photoacoustic microscopy for monitoring vascular normalization during anti-angiogenic therapy. PHOTOACOUSTICS 2019; 15:100143. [PMID: 31463195 PMCID: PMC6710376 DOI: 10.1016/j.pacs.2019.100143] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/17/2019] [Accepted: 08/09/2019] [Indexed: 05/03/2023]
Abstract
Monitoring the changes in tumor vascularity is important for anti-angiogenic therapy assessment with therapeutic implications. However, monitoring vascularity is quite challenging due to the lack of appropriate imaging techniques. Here, we describe a non-invasive imaging technique using optical-resolution photoacoustic microscopy (OR-PAM) to track vascular changes in prostate cancer treated with an anti-angiogenic agent, DC101, on a mouse ear xenograft model. Approximately 1-3 days after the initial therapy, OR-PAM imaging detected tumor vascular changes such as reduced vessel tortuosity, decreased vessel diameter and homogenized intratumoral vessel distribution. These observations indicated vessel normalization, which was pathologically validated as increased fractional pericyte coverage, functional perfusion and drug delivery of the vessels. After four DC101 interventions, OR-PAM imaging eventually revealed intratumoral vessel regression. Therefore, OR-PAM imaging of the vasculature offers a promising method to study anti-angiogenic drug mechanisms of action in vivo and holds potential in monitoring and guiding anti-angiogenic therapy.
Collapse
Affiliation(s)
- Hui-Chao Zhou
- Department of Medical Ultrasonic, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ningbo Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Mechanical and Electrical Engineering, Guangzhou University, Guangzhou, China
| | - Huangxuan Zhao
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Tinghui Yin
- Department of Medical Ultrasonic, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianhui Zhang
- School of Mechanical and Electrical Engineering, Guangzhou University, Guangzhou, China
| | - Wei Zheng
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liang Song
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chengbo Liu
- Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Corresponding author at: Research Laboratory for Biomedical Optics and Molecular Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Boulevard, Shenzhen 518055, China.
| | - Rongqin Zheng
- Department of Medical Ultrasonic, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Corresponding author at: Department of Medical Ultrasonic, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital, Sun Yat-sen University, Tian He Road 600#, Guangzhou 510630, China.
| |
Collapse
|
117
|
Xu W, Zhang X, Hu X, Zhiyi C, Huang P. Translational Prospects of ultrasound-mediated tumor immunotherapy: Preclinical advances and safety considerations. Cancer Lett 2019; 460:86-95. [DOI: 10.1016/j.canlet.2019.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022]
|
118
|
Zarco N, Norton E, Quiñones-Hinojosa A, Guerrero-Cázares H. Overlapping migratory mechanisms between neural progenitor cells and brain tumor stem cells. Cell Mol Life Sci 2019; 76:3553-3570. [PMID: 31101934 PMCID: PMC6698208 DOI: 10.1007/s00018-019-03149-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/16/2019] [Accepted: 05/13/2019] [Indexed: 01/18/2023]
Abstract
Neural stem cells present in the subventricular zone (SVZ), the largest neurogenic niche of the mammalian brain, are able to self-renew as well as generate neural progenitor cells (NPCs). NPCs are highly migratory and traverse the rostral migratory stream (RMS) to the olfactory bulb, where they terminally differentiate into mature interneurons. NPCs from the SVZ are some of the few cells in the CNS that migrate long distances during adulthood. The migratory process of NPCs is highly regulated by intracellular pathway activation and signaling from the surrounding microenvironment. It involves modulation of cell volume, cytoskeletal rearrangement, and isolation from compact extracellular matrix. In malignant brain tumors including high-grade gliomas, there are cells called brain tumor stem cells (BTSCs) with similar stem cell characteristics to NPCs but with uncontrolled cell proliferation and contribute to tumor initiation capacity, tumor progression, invasion, and tumor maintenance. These BTSCs are resistant to chemotherapy and radiotherapy, and their presence is believed to lead to tumor recurrence at distal sites from the original tumor location, principally due to their high migratory capacity. BTSCs are able to invade the brain parenchyma by utilizing many of the migratory mechanisms used by NPCs. However, they have an increased ability to infiltrate the tight brain parenchyma and utilize brain structures such as myelin tracts and blood vessels as migratory paths. In this article, we summarize recent findings on the mechanisms of cellular migration that overlap between NPCs and BTSCs. A better understanding of the intersection between NPCs and BTSCs will to provide a better comprehension of the BTSCs' invasive capacity and the molecular mechanisms that govern their migration and eventually lead to the development of new therapies to improve the prognosis of patients with malignant gliomas.
Collapse
Affiliation(s)
- Natanael Zarco
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Emily Norton
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Hugo Guerrero-Cázares
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
119
|
Optical coherence tomography angiography in preclinical neuroimaging. Biomed Eng Lett 2019; 9:311-325. [PMID: 31456891 DOI: 10.1007/s13534-019-00118-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/29/2019] [Accepted: 06/27/2019] [Indexed: 01/22/2023] Open
Abstract
Preclinical neuroimaging allows for the assessment of brain anatomy, connectivity, and function in laboratory animals, such as mice and this imaging field has been a rapidly growing aimed at bridging the translation gap between animal and human research. The progress in the animal research could be accelerated by high-resolution in vivo optical imaging technologies. Optical coherence tomography-based angiography (OCTA) estimates the scattering from moving red blood cells, providing the visualization of functional micro-vessel networks within tissue beds in vivo without a need for exogenous contrast agents. Recent advancement of OCTA methods have expanded its application to neuroimaging of small animal models of brain disorders. In this paper, we overview the recent development of OCTA techniques for blood flow imaging and its preclinical applications in neuroimaging. In specific, a summary of preclinical OCTA studies for traumatic brain injury, cerebral stroke, and aging brain on mice is reviewed.
Collapse
|
120
|
Demidov V, Matveev LA, Demidova O, Matveyev AL, Zaitsev VY, Flueraru C, Vitkin IA. Analysis of low-scattering regions in optical coherence tomography: applications to neurography and lymphangiography. BIOMEDICAL OPTICS EXPRESS 2019; 10:4207-4219. [PMID: 31453005 PMCID: PMC6701530 DOI: 10.1364/boe.10.004207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/24/2019] [Accepted: 07/17/2019] [Indexed: 05/19/2023]
Abstract
Analysis of semi-transparent low scattering biological structures in optical coherence tomography (OCT) has been actively pursued in the context of lymphatic imaging, with most approaches relying on the relative absence of signal as a means of detection. Here we present an alternate methodology based on spatial speckle statistics, utilizing the similarity of a distribution of given voxel intensities to the power distribution function of pure noise, to visualize the low-scattering biological structures of interest. In a human tumor xenograft murine model, we show that these correspond to lymphatic vessels and nerves; extensive histopathologic validation studies are reported to unequivocally establish this correspondence. The emerging possibility of OCT lymphangiography and neurography is novel and potentially impactful (especially the latter), although further methodology refinement is needed to distinguish between the visualized lymphatics and nerves.
Collapse
Affiliation(s)
- Valentin Demidov
- Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada
| | - Lev A. Matveev
- Institute of Applied Physics Russian Academy of Sciences, 46 Ulyanov Street, Nizhniy Novgorod, 603950, Russia
| | - Olga Demidova
- Department of Arts and Science, Seneca College, 1750 Finch Avenue East, Toronto, M2J 2X5, Canada
| | - Alexander L. Matveyev
- Institute of Applied Physics Russian Academy of Sciences, 46 Ulyanov Street, Nizhniy Novgorod, 603950, Russia
| | - Vladimir Y. Zaitsev
- Institute of Applied Physics Russian Academy of Sciences, 46 Ulyanov Street, Nizhniy Novgorod, 603950, Russia
| | - Costel Flueraru
- National Research Council Canada, Information Communication Technology, 1200 Montreal Rd, Ottawa, K1A0R6, Canada
| | - I. Alex Vitkin
- Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada
- University Health Network, Princess Margaret Cancer Centre, 610 University Ave, Toronto, M5G 2C1, Canada
- University of Toronto, Department of Radiation Oncology, 150 College St, Toronto, M5S 3E2, Canada
| |
Collapse
|
121
|
SoRelle ED, Yecies DW, Liba O, Bennett FC, Graef CM, Dutta R, Mitra S, Joubert LM, Cheshier S, Grant GA, de la Zerda A. Spatiotemporal Tracking of Brain-Tumor-Associated Myeloid Cells in Vivo through Optical Coherence Tomography with Plasmonic Labeling and Speckle Modulation. ACS NANO 2019; 13:7985-7995. [PMID: 31259527 PMCID: PMC8144904 DOI: 10.1021/acsnano.9b02656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
By their nature, tumors pose a set of profound challenges to the immune system with respect to cellular recognition and response coordination. Recent research indicates that leukocyte subpopulations, especially tumor-associated macrophages (TAMs), can exert substantial influence on the efficacy of various cancer immunotherapy treatment strategies. To better study and understand the roles of TAMs in determining immunotherapeutic outcomes, significant technical challenges associated with dynamically monitoring single cells of interest in relevant live animal models of solid tumors must be overcome. However, imaging techniques with the requisite combination of spatiotemporal resolution, cell-specific contrast, and sufficient signal-to-noise at increasing depths in tissue are exceedingly limited. Here we describe a method to enable high-resolution, wide-field, longitudinal imaging of TAMs based on speckle-modulating optical coherence tomography (SM-OCT) and spectral scattering from an optimized contrast agent. The approach's improvements to OCT detection sensitivity and noise reduction enabled high-resolution OCT-based observation of individual cells of a specific host lineage in live animals. We found that large gold nanorods (LGNRs) that exhibit a narrow-band, enhanced scattering cross-section can selectively label TAMs and activate microglia in an in vivo orthotopic murine model of glioblastoma multiforme. We demonstrated near real-time tracking of the migration of cells within these myeloid subpopulations. The intrinsic spatiotemporal resolution, imaging depth, and contrast sensitivity reported herein may facilitate detailed studies of the fundamental behaviors of TAMs and other leukocytes at the single-cell level in vivo, including intratumoral distribution heterogeneity and roles in modulating cancer proliferation.
Collapse
Affiliation(s)
- Elliott Daniel SoRelle
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Biophysics Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Derek William Yecies
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Orly Liba
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | | | - Claus Moritz Graef
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Rebecca Dutta
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Siddhartha Mitra
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Lydia-Marie Joubert
- Cell Sciences Imaging Facility, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Samuel Cheshier
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
| | - Adam de la Zerda
- Department of Structural Biology, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Biophysics Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Molecular Imaging Program (MIPS), Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Bio-X Program, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, 299 Campus Dr., Stanford, CA 94305, USA
- The Chan Zuckerberg Biohub, 499 Illinois St., San Francisco, CA 94158, USA
- To whom correspondence should be addressed:
| |
Collapse
|
122
|
A Robust Method for Adjustment of Laser Speckle Contrast Imaging during Transcranial Mouse Brain Visualization. PHOTONICS 2019. [DOI: 10.3390/photonics6030080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Laser speckle imaging (LSI) is a well-known and useful approach for the non-invasive visualization of flows and microcirculation localized in turbid scattering media, including biological tissues (such as brain vasculature, skin capillaries etc.). Despite an extensive use of LSI for brain imaging, the LSI technique has several critical limitations. One of them is associated with inability to resolve a functionality of vessels. This limitation also leads to the systematic error in the quantitative interpretation of values of speckle contrast obtained for different vessel types, such as sagittal sinus, arteries, and veins. Here, utilizing a combined use of LSI and fluorescent intravital microscopy (FIM), we present a simple and robust method to overcome the limitations mentioned above for the LSI approach. The proposed technique provides more relevant, abundant, and valuable information regarding perfusion rate ration between different types of vessels that makes this method highly useful for in vivo brain surgical operations.
Collapse
|
123
|
Andreana M, Sentosa R, Erkkilä MT, Drexler W, Unterhuber A. Depth resolved label-free multimodal optical imaging platform to study morpho-molecular composition of tissue. Photochem Photobiol Sci 2019; 18:997-1008. [PMID: 30882117 DOI: 10.1039/c8pp00410b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multimodal imaging platforms offer a vast array of tissue information in a single image acquisition by combining complementary imaging techniques. By merging different systems, better tissue characterization can be achieved than is possible by the constituent imaging modalities alone. The combination of optical coherence tomography (OCT) with non-linear optical imaging (NLOI) techniques such as two-photon excited fluorescence (TPEF), second harmonic generation (SHG) and coherent anti-Stokes Raman scattering (CARS) provides access to detailed information of tissue structure and molecular composition in a fast, label-free and non-invasive manner. We introduce a multimodal label-free approach for morpho-molecular imaging and spectroscopy and validate the system in mouse skin demonstrating the potential of the system for colocalized acquisition of OCT and NLOI signals.
Collapse
Affiliation(s)
- Marco Andreana
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| | | | | | | | | |
Collapse
|
124
|
Yang S, Liu K, Ding H, Gao H, Zheng X, Ding Z, Xu K, Li P. Longitudinal in vivo intrinsic optical imaging of cortical blood perfusion and tissue damage in focal photothrombosis stroke model. J Cereb Blood Flow Metab 2019; 39. [PMID: 29521548 PMCID: PMC6668510 DOI: 10.1177/0271678x18762636] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A thorough understanding of the spatiotemporal dynamics of blood supply and tissue viability is of great importance in stroke researches. In the current study, vascular and cellular responses to focal ischemia were monitored with optical coherence tomography on chronic rat photothrombotic stroke model. The 3D mapping of blood perfusion and cellular scattering were achieved by analyzing the temporal dynamics and depth attenuation of intrinsic backscattered light respectively. Optical coherence tomography revealed that vessels of different types presented various spatial and temporal dynamics during the photothrombotic occlusion and the later recovery period. The large distal middle cerebral arteries presented a spontaneous recanalization and the small pial microvessels presented a reperfusion along with newly appeared vessels from the peripheral into the core area. The cortical capillary perfusion presented a weak recovery. Compared to the male group, the female rats showed a faster vascular recovery after photothrombotic. Moreover, the dynamic changes of the cellular scattering signal showed a high spatial and temporal correlation with the cortical capillary perfusion. Combined with well-designed photothrombotic stroke model and chronic optical window, optical coherence tomography imaging offers a unique approach to improve the understanding of stroke procedure and evaluate the treatment outcomes.
Collapse
Affiliation(s)
- Shanshan Yang
- 1 State Key Lab of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kezhou Liu
- 2 Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, China.,3 College of Life Information Science and Instruments Engineering, Hangzhou Dianzi University, Hangzhou, China
| | - Huijie Ding
- 3 College of Life Information Science and Instruments Engineering, Hangzhou Dianzi University, Hangzhou, China
| | - Huan Gao
- 4 Qiushi Academy for Advanced Studies (QAAS), Zhejiang University, Hangzhou, China
| | - Xiaoxiang Zheng
- 2 Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, China.,4 Qiushi Academy for Advanced Studies (QAAS), Zhejiang University, Hangzhou, China.,5 Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Zhihua Ding
- 1 State Key Lab of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kedi Xu
- 2 Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, China.,4 Qiushi Academy for Advanced Studies (QAAS), Zhejiang University, Hangzhou, China.,5 Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Peng Li
- 1 State Key Lab of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
125
|
Hu D, Li L, Li S, Wu M, Ge N, Cui Y, Lian Z, Song J, Chen H. Lymphatic system identification, pathophysiology and therapy in the cardiovascular diseases. J Mol Cell Cardiol 2019; 133:99-111. [PMID: 31181226 DOI: 10.1016/j.yjmcc.2019.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
The mammalian circulatory system comprises both the cardiovascular system and the lymphatic system. In contrast to the closed, high-pressure and circular blood vascular circulation, the lymphatic system forms an open, low-pressure and unidirectional transit network from the extracellular space to the venous system. It plays a key role in regulating tissue fluid homeostasis, absorption of gastrointestinal lipids, and immune surveillance throughout the body. Despite the critical physiological functions of the lymphatic system, a complete understanding of the lymphatic vessels lags far behind that of the blood vasculatures due to the challenge of their visualization. During the last 20 years, discoveries of underlying genes responsible for lymphatic vessel biology, combined with state-of-the-art lymphatic function imaging and quantification techniques, have established the importance of the lymphatic vasculature in the pathogenesis of cardiovascular diseases including lymphedema, obesity and metabolic diseases, dyslipidemia, hypertension, inflammation, atherosclerosis and myocardial infraction. In this review, we highlight the most recent advances in the field of lymphatic vessel biology, with an emphasis on the new identification techniques of lymphatic system, pathophysiological mechanisms of atherosclerosis and myocardial infarction, and new therapeutic perspectives of lymphangiogenesis.
Collapse
Affiliation(s)
- Dan Hu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Long Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Sufang Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Manyan Wu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Nana Ge
- Department of Geriatrics, Beijing Renhe Hospital, Beijing, China
| | - Yuxia Cui
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Zheng Lian
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Junxian Song
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
126
|
Todorović J, Dinčić M, Nešović Ostojić J, Zaletel I, Lopičić S, Dundjerović D, Tatić S, Kovačević S, Paunović I, Puškaš N, Marković L. Differences in Chromatin Texture and Nuclear Fractal Dimension Between Hashimoto's and Lymphocytic Thyroiditis Lymphocytes. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2019; 25:762-768. [PMID: 30813976 DOI: 10.1017/s1431927619000163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Previous evidence suggested that lymphocytic thyroiditis (LT) was a variant of Hashimoto's thyroiditis (HT), thus the aim of the current study is to quantify structural changes in histological specimens taken from HT and LT patients. A total of 600 images containing a single lymphocyte nucleus (300 nuclei per group) were obtained from 20 patients with HT and LT. In order to quantify changes in the nuclear architecture of investigated lymphocytes, the fractal dimension (FD) and some gray-level co-occurrence matrix texture parameters (angular second moment, inverse difference moment, contrast, entropy, and correlation) were calculated for each nucleus. A statistically significant difference in the FD of the "binary-outlined" nucleus and that of the corresponding "black-and-white" nucleus was detected between HT and LT lymphocyte nuclei. In addition, there was also a statistically significant difference in contrast and correlation between HT and LT lymphocyte nuclei. In conclusion, the results of this study suggested that there was a difference in structural complexity between investigated lymphocyte nuclei; additionally, LT lymphocytes possessed probably more complex texture and larger variations as well as more asymmetrical nuclei compared with HT lymphocytes. Accordingly, these findings indicate that LT is probably not a variant of HT; however, more complex studies are necessary to estimate differences between these types of thyroiditis.
Collapse
Affiliation(s)
- Jasna Todorović
- Faculty of Medicine,Institute of Pathological Physiology, University of Belgrade,Serbia
| | - Marko Dinčić
- Faculty of Medicine,Institute of Pathological Physiology, University of Belgrade,Serbia
| | | | - Ivan Zaletel
- Faculty of Medicine,Institute of Histology and Embryology, University of Belgrade,Serbia
| | - Srdjan Lopičić
- Faculty of Medicine,Institute of Pathological Physiology, University of Belgrade,Serbia
| | - Duško Dundjerović
- Faculty of Medicine,Institute of Pathology, University of Belgrade,Serbia
| | - Svetislav Tatić
- Faculty of Medicine,Institute of Pathology, University of Belgrade,Serbia
| | - Sanjin Kovačević
- Faculty of Medicine,Institute of Pathological Physiology, University of Belgrade,Serbia
| | - Ivan Paunović
- Faculty of Medicine,Center for Endocrine Surgery, Clinical Center of Serbia, University of Belgrade,Serbia
| | - Nela Puškaš
- Faculty of Medicine,Institute of Histology and Embryology, University of Belgrade,Serbia
| | - Ljiljana Marković
- Faculty of Medicine,Institute of Pathological Physiology, University of Belgrade,Serbia
| |
Collapse
|
127
|
Choi WJ, Li Y, Wang RK. Monitoring Acute Stroke Progression: Multi-Parametric OCT Imaging of Cortical Perfusion, Flow, and Tissue Scattering in a Mouse Model of Permanent Focal Ischemia. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:1427-1437. [PMID: 30714910 PMCID: PMC6660833 DOI: 10.1109/tmi.2019.2895779] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Cerebral ischemic stroke causes injury to brain tissue characterized by a complex cascade of neuronal and vascular events. Imaging during the early stages of its development allows prediction of tissue infarction and penumbra so that optimal intervention can be determined in order to salvage brain function impairment. Therefore, there is a critical need for novel imaging techniques that can characterize brain injury in the earliest phases of the ischemic stroke. This paper examined optical coherence tomography (OCT) for imaging acute injury in experimental ischemic stroke in vivo. Based on endogenous optical scattering signals provided by OCT imaging, we have developed a single, integrated imaging platform enabling the measurement of changes in blood perfusion, blood flow, erythrocyte velocity, and light attenuation within a cortical tissue, during focal cerebral ischemia in a mouse model. During the acute phase (from 5 min to the first few hours following the blood occlusion), the multi-parametric OCT imaging revealed multiple hemodynamic and tissue scattering responses in vivo, including cerebral blood flow deficits, capillary non-perfusion, displacement of penetrating vessels, and increased light attenuation in the cortical tissue at risk that are spatially correlated with the infarct core, as determined by postmortem staining with triphenyltetrazolium chloride. The use of multi-parametric OCT imaging may aid in the comprehensive evaluation of ischemic lesions during the early stages of stroke, thereby providing essential knowledge for guiding treatment decisions.
Collapse
Affiliation(s)
- Woo June Choi
- School of Electrical and Electronics Engineering, College of ICT Engineering, Chung-Ang University, Seoul, 06974, Korea
| | - Yuandong Li
- Department of Bioengineering, University of Washington, Seattle WA 98195, USA
| | - Ruikang K. Wang
- Corresponding author, phone: 206-616-5025; fax: 206-616-5025;
| |
Collapse
|
128
|
Wang CX, Gao ZY, Wang X, Ke C, Zhang Z, Zhang CJ, Fu LM, Wang Y, Zhang JP. Noninvasive and real-time pharmacokinetics imaging of polymeric nanoagents in the thoracoepigastric vein networks of living mice. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-11. [PMID: 31222991 PMCID: PMC6977018 DOI: 10.1117/1.jbo.24.6.066009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/05/2019] [Indexed: 06/09/2023]
Abstract
Noninvasive and real-time visualization of the thoracoepigastric veins (TVs) of living mice was demonstrated by using two-photon excitation (TPE) optical imaging with a Eu-luminescent polymeric nanoagent as the angiographic contrast. The spatiotemporal evolution of the polymeric nanoagent in TVs was monitored for up to 2 h by TPE time-resolved (TPE-TR) bioimaging, which is free from the interference of tissue autofluorescence. A wide field-of-view covering the thoracoabdominal region allowed the visualization of the entire TV network with an imaging depth of 1 to 2 mm and a lateral resolution of 80 μm at submillimeter. Detailed analysis of the uptake, transport, and clearance processes of the polymeric nanoagent revealed a clearance time constant of ∼30 min and an apparent clearance efficiency of 80% to 90% for the nanoagent in both axial and lateral TVs. TPE-TR imaging of the dissected internal organs proved that the liver is mainly responsible for the sequestration of the nanoagent, which is consistent with the apparent retention efficiency of liver, ∼32 % , as determined by the real-time in vivo TV imaging. We demonstrate the potency of TPE-TR modality in the pharmacokinetics imaging of the peripheral vascular systems of animal models, which can be beneficial for related nanotheranostics study.
Collapse
Affiliation(s)
- Chuan-Xi Wang
- Peking University, College of Chemistry and Molecular Engineering, Academy for Advanced Interdisciplinary Studies, Beijing National Laboratory for Molecular Science, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Beijing, China
| | - Zhi-Yue Gao
- Peking University, College of Chemistry and Molecular Engineering, Academy for Advanced Interdisciplinary Studies, Beijing National Laboratory for Molecular Science, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Beijing, China
| | - Xin Wang
- Peking University, College of Chemistry and Molecular Engineering, Academy for Advanced Interdisciplinary Studies, Beijing National Laboratory for Molecular Science, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Beijing, China
| | - Can Ke
- Peking University, College of Chemistry and Molecular Engineering, Academy for Advanced Interdisciplinary Studies, Beijing National Laboratory for Molecular Science, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Beijing, China
| | - Zhuo Zhang
- Renmin University of China, Department of Chemistry, Beijing, China
| | - Chao-Jie Zhang
- Renmin University of China, Department of Chemistry, Beijing, China
| | - Li-Min Fu
- Renmin University of China, Department of Chemistry, Beijing, China
| | - Yuan Wang
- Peking University, College of Chemistry and Molecular Engineering, Academy for Advanced Interdisciplinary Studies, Beijing National Laboratory for Molecular Science, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Beijing, China
| | - Jian-Ping Zhang
- Renmin University of China, Department of Chemistry, Beijing, China
| |
Collapse
|
129
|
Katta N, Estrada AD, McElroy AB, Gruslova A, Oglesby M, Cabe AG, Feldman MD, Fleming RYD, Brenner AJ, Milner TE. Laser brain cancer surgery in a xenograft model guided by optical coherence tomography. Am J Cancer Res 2019; 9:3555-3564. [PMID: 31281497 PMCID: PMC6587169 DOI: 10.7150/thno.31811] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
Higher precision surgical devices are needed for tumor resections near critical brain structures. The goal of this study is to demonstrate feasibility of a system capable of precise and bloodless tumor ablation. An image-guided laser surgical system is presented for excision of brain tumors in vivo in a murine xenograft model. The system combines optical coherence tomography (OCT) guidance with surgical lasers for high-precision tumor ablation (Er:YAG) and microcirculation coagulation (Thulium (Tm) fiber laser). Methods: A fluorescent human glioblastoma cell line was injected into mice and allowed to grow four weeks. Craniotomies were performed and tumors were imaged with confocal fluorescence microscopy. The mice were subsequently OCT imaged prior, during and after laser coagulation and/or ablation. The prior OCT images were used to compute three-dimensional tumor margin and angiography images, which guided the coagulation and ablation steps. Histology of the treated regions was then compared to post-treatment OCT images. Results: Tumor sizing based on OCT margin detection matched histology to within experimental error. Although fluorescence microscopy imaging showed the tumors were collocated with OCT imaging, margin assessment using confocal microscopy failed to see the extent of the tumor beyond ~ 250 µm in depth, as verified by OCT and histology. The two-laser approach to surgery utilizing Tm wavelength for coagulation and Er:YAG for ablation yielded bloodless resection of tumor regions with minimal residual damage as seen in histology. Conclusion: Precise and bloodless tumor resection under OCT image guidance is demonstrated in the murine xenograft brain cancer model. Tumor margins and vasculature are accurately made visible without need for exogenous contrast agents.
Collapse
|
130
|
Shi W, Chen C, Pasarikovski CR, Gao W, Yang VXD. Differential phase standard-deviation-based optical coherence tomographic angiography for human retinal imaging in vivo. APPLIED OPTICS 2019; 58:3401-3409. [PMID: 31044835 DOI: 10.1364/ao.58.003401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/25/2019] [Indexed: 06/09/2023]
Abstract
We present a differential phase standard-deviation (DPSD)-based optical coherence tomographic (OCT) angiography (OCTA) technique to calculate the angiography images of the human retina. The standard deviation was calculated along the depth direction on the differential phase image of two B-scans (from the same position, at different times) to contrast dynamic vascular signals. The performance of a DPSD was verified by both phantom and in vivo experiments. When compared to other OCTA algorithms such as phase variance OCT, speckle variance OCT, and optical microangiography, we showed that a DPSD achieved improved image contrast and higher sensitivity. Furthermore, we also found the improved signal-to-noise ratio and contrast-to-noise ratio of 1.6 dB and 0.5, respectively, in large scanning range images.
Collapse
|
131
|
Demené C, Payen T, Dizeux A, Barrois G, Gennisson JL, Bridal L, Tanter M. 3-D Longitudinal Imaging of Tumor Angiogenesis in Mice in Vivo Using Ultrafast Doppler Tomography. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1284-1296. [PMID: 30799125 DOI: 10.1016/j.ultrasmedbio.2018.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 11/09/2018] [Accepted: 12/17/2018] [Indexed: 06/09/2023]
Abstract
Angiogenesis, the formation of new vessels, is one of the key mechanisms in tumor development and an appealing target for therapy. Non-invasive, high-resolution, high-sensitivity, quantitative 3-D imaging techniques are required to correctly depict tumor heterogeneous vasculature over time. Ultrafast Doppler was recently introduced and provides an unprecedented combination of resolution, penetration depth and sensitivity without requiring any contrast agents. The technique was further extended to three dimensions with ultrafast Doppler tomography (UFD-T). In this work, UFD-T was applied to the monitoring of tumor angiogenesis in vivo, providing structural and functional information at different stages of development. UFD-T volume renderings revealed that our murine model's vasculature stems from pre-existing vessels and sprouts to perfuse the whole volume as the tumor grows until a critical size is reached. Then, as the network becomes insufficient, the tumor core is no longer irrigated because the vasculature is concentrated mainly in the periphery. In addition to spatial distribution and growth patterns, UFD-T allowed a quantitative analysis of vessel size and length, revealing that the diameter distribution of vessels remained relatively constant throughout tumor growth. The network is dominated by small vessels at all stages of tumor development, with more than 74% of the vessels less than 200 µm in diameter. This study also found that cumulative vessel length is more closely related to tumor radius than volume, indicating that the vascularization becomes insufficient when a critical mass is reached. UFD-T was also compared with dynamic contrast-enhanced ultrasound and found to provide complementary information regarding the link between structure and perfusion. In conclusion, UFD-T is capable of in vivo quantitative assessment of the development of tumor vasculature (vessels with blood speed >1 mm/s [sensitivity limit] assessed with a resolution limit of 80 µm) in 3 dimensions. The technique has very interesting potential as a tool for treatment monitoring, response assessment and treatment planning for optimal drug efficiency.
Collapse
Affiliation(s)
- Charlie Demené
- Physics for Medicine Paris, Inserm, ESPCI Paris, CNRS, PSL Research University, Paris, France; Inserm Accélérateur de Recherche Technologique en Ultrasons biomédicaux, Paris, France.
| | - Thomas Payen
- Sorbonnne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Alexandre Dizeux
- Sorbonnne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Guillaume Barrois
- Sorbonnne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Jean-Luc Gennisson
- Physics for Medicine Paris, Inserm, ESPCI Paris, CNRS, PSL Research University, Paris, France
| | - Lori Bridal
- Sorbonnne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Mickael Tanter
- Inserm Accélérateur de Recherche Technologique en Ultrasons biomédicaux, Paris, France
| |
Collapse
|
132
|
Rajabi S, Dehghan MH, Dastmalchi R, Jalali Mashayekhi F, Salami S, Hedayati M. The roles and role-players in thyroid cancer angiogenesis. Endocr J 2019; 66:277-293. [PMID: 30842365 DOI: 10.1507/endocrj.ej18-0537] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Thyroid cancer is the most prevalent endocrine cancer worldwide. Angiogenesis, the formation of new blood vessels, plays a pivotal role in the development and progression of tumors. Over the past years, cancer research has focused on the ability of tumors to induce newly formed blood vessel, because tumor growth and the process of cancer metastasis mainly depends on angiogenesis. Tumor neovascularization occurs following the imbalance between pro-angiogenic and anti-angiogenic factors until the tumor switches to an angiogenic phenotype. A number of signaling factors and receptors that are implicated in the regulation of angiogenesis have been identified and characterized; most notably, the vascular endothelial growth factors (VEGFs) family and their receptors, which are the main pro-angiogenic molecules during early development and in pathological conditions such as cancer. Although thyroid is a highly vascularized organ, angiogenic switch in tumors of this organ leads to the formation of a vast network of blood vessels that favors the dissemination of tumor cells to distant organs and results in deterioration of patient conditions. Accordingly, the identification of key angiogenic biomarkers for thyroid cancer can facilitate diagnosis, prognosis and clinical decision-making and also may help to discover targeting factors for effective cancer therapy as well as monitoring response to therapy. Hence, the main purposes of this review are to summarize the types and mechanisms of angiogenesis emphasizing the prominent factors implicated in thyroid cancer angiogenesis.
Collapse
Affiliation(s)
- Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Romina Dastmalchi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Siamak Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
133
|
Wijeratne PA, Vavourakis V. A quantitative in silico platform for simulating cytotoxic and nanoparticle drug delivery to solid tumours. Interface Focus 2019; 9:20180063. [PMID: 31065337 DOI: 10.1098/rsfs.2018.0063] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
The role of tumour-host mechano-biology and the mechanisms involved in the delivery of anti-cancer drugs have been extensively studied using in vitro and in vivo models. A complementary approach is offered by in silico models, which can also potentially identify the main factors affecting the transport of tumour-targeting molecules. Here, we present a generalized three-dimensional in silico modelling framework of dynamic solid tumour growth, angiogenesis and drug delivery. Crucially, the model allows for drug properties-such as size and binding affinity-to be explicitly defined, hence facilitating investigation into the interaction between the changing tumour-host microenvironment and cytotoxic and nanoparticle drugs. We use the model to qualitatively recapitulate experimental evidence of delivery efficacy of cytotoxic and nanoparticle drugs on matrix density (and hence porosity). Furthermore, we predict a highly heterogeneous distribution of nanoparticles after delivery; that nanoparticles require a high porosity extracellular matrix to cause tumour regression; and that post-injection transvascular fluid velocity depends on matrix porosity, and implicitly on the size of the drug used to treat the tumour. These results highlight the utility of predictive in silico modelling in better understanding the factors governing efficient cytotoxic and nanoparticle drug delivery.
Collapse
Affiliation(s)
- Peter A Wijeratne
- Centre for Medical Imaging Computing, Department of Computer Science, University College London, London, UK
| | - Vasileios Vavourakis
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
134
|
Abstract
Fuelled by innovation, optical microscopy plays a critical role in the life sciences and medicine, from basic discovery to clinical diagnostics. However, optical microscopy is limited by typical penetration depths of a few hundred micrometres for in vivo interrogations in the visible spectrum. Optoacoustic microscopy complements optical microscopy by imaging the absorption of light, but it is similarly limited by penetration depth. In this Review, we summarize progress in the development and applicability of optoacoustic mesoscopy (OPAM); that is, optoacoustic imaging with acoustic resolution and wide-bandwidth ultrasound detection. OPAM extends the capabilities of optical imaging beyond the depths accessible to optical and optoacoustic microscopy, and thus enables new applications. We explain the operational principles of OPAM, its placement as a bridge between optoacoustic microscopy and optoacoustic macroscopy, and its performance in the label-free visualization of tissue pathophysiology, such as inflammation, oxygenation, vascularization and angiogenesis. We also review emerging applications of OPAM in clinical and biological imaging.
Collapse
|
135
|
Tumor Ensemble-Based Modeling and Visualization of Emergent Angiogenic Heterogeneity in Breast Cancer. Sci Rep 2019; 9:5276. [PMID: 30918274 PMCID: PMC6437174 DOI: 10.1038/s41598-019-40888-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/19/2019] [Indexed: 01/29/2023] Open
Abstract
There is a critical need for new tools to investigate the spatio-temporal heterogeneity and phenotypic alterations that arise in the tumor microenvironment. However, computational investigations of emergent inter- and intra-tumor angiogenic heterogeneity necessitate 3D microvascular data from 'whole-tumors' as well as "ensembles" of tumors. Until recently, technical limitations such as 3D imaging capabilities, computational power and cost precluded the incorporation of whole-tumor microvascular data in computational models. Here, we describe a novel computational approach based on multimodality, 3D whole-tumor imaging data acquired from eight orthotopic breast tumor xenografts (i.e. a tumor 'ensemble'). We assessed the heterogeneous angiogenic landscape from the microvascular to tumor ensemble scale in terms of vascular morphology, emergent hemodynamics and intravascular oxygenation. We demonstrate how the abnormal organization and hemodynamics of the tumor microvasculature give rise to unique microvascular niches within the tumor and contribute to inter- and intra-tumor heterogeneity. These tumor ensemble-based simulations together with unique data visualization approaches establish the foundation of a novel 'cancer atlas' for investigators to develop their own in silico systems biology applications. We expect this hybrid image-based modeling framework to be adaptable for the study of other tissues (e.g. brain, heart) and other vasculature-dependent diseases (e.g. stroke, myocardial infarction).
Collapse
|
136
|
Virtual histological staining of unlabelled tissue-autofluorescence images via deep learning. Nat Biomed Eng 2019; 3:466-477. [PMID: 31142829 DOI: 10.1038/s41551-019-0362-y] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/28/2019] [Indexed: 01/16/2023]
Abstract
The histological analysis of tissue samples, widely used for disease diagnosis, involves lengthy and laborious tissue preparation. Here, we show that a convolutional neural network trained using a generative adversarial-network model can transform wide-field autofluorescence images of unlabelled tissue sections into images that are equivalent to the bright-field images of histologically stained versions of the same samples. A blind comparison, by board-certified pathologists, of this virtual staining method and standard histological staining using microscopic images of human tissue sections of the salivary gland, thyroid, kidney, liver and lung, and involving different types of stain, showed no major discordances. The virtual-staining method bypasses the typically labour-intensive and costly histological staining procedures, and could be used as a blueprint for the virtual staining of tissue images acquired with other label-free imaging modalities.
Collapse
|
137
|
Tang J, Erdener SE, Sunil S, Boas DA. Normalized field autocorrelation function-based optical coherence tomography three-dimensional angiography. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-8. [PMID: 30868803 PMCID: PMC6414735 DOI: 10.1117/1.jbo.24.3.036005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/06/2019] [Indexed: 05/02/2023]
Abstract
Optical coherence tomography angiography (OCTA) has been widely used for en face visualization of the microvasculature, but is challenged for real three-dimensional (3-D) topologic imaging due to the "tail" artifacts that appear below large vessels. Further, OCTA is generally incapable of differentiating descending arterioles from ascending venules. We introduce a normalized field autocorrelation function-based OCTA (g1-OCTA), which minimizes the tail artifacts and is capable of distinguishing penetrating arterioles from venules in the 3-D image. g1 ( τ ) is calculated from repeated optical coherence tomography (OCT) acquisitions for each spatial location. The decay amplitude of g1 ( τ ) is retrieved to represent the dynamics for each voxel. To account for the small g1 ( τ ) decay in capillaries where red blood cells are flowing slowly and discontinuously, Intralipid is injected to enhance the OCT signal. We demonstrate that the proposed technique realizes 3-D OCTA with negligible tail projections and the penetrating arteries are readily identified. In addition, compared to regular OCTA, the proposed g1-OCTA largely increased the depth-of-field. This technique provides a more accurate rendering of the vascular 3-D anatomy and has the potential for more quantitative characterization of vascular networks.
Collapse
Affiliation(s)
- Jianbo Tang
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
- Address all correspondence to Jianbo Tang, E-mail:
| | - Sefik Evren Erdener
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Smrithi Sunil
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - David A. Boas
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| |
Collapse
|
138
|
Pijewska E, Gorczynska I, Szkulmowski M. Computationally effective 2D and 3D fast phase unwrapping algorithms and their applications to Doppler optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2019; 10:1365-1382. [PMID: 30891352 PMCID: PMC6420292 DOI: 10.1364/boe.10.001365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 05/07/2023]
Abstract
We propose a simplification for a robust and easy to implement fast phase unwrapping (FPU) algorithm that is used to solve the phase wrapping problem encountered in various fields of optical imaging and metrology. We show that the number of necessary computations using the algorithm can be reduced compared to its original version. FPU can be easily extended from two to three spatial dimensions. We demonstrate the applicability of the two- and three-dimensional FPU algorithm for Doppler optical coherence tomography (DOCT) in numerical simulations, and in the imaging of a flow phantom and blood flow in the human retina in vivo. We introduce an FPU applicability plot for use as a guide in the selection of the most suitable version of the algorithm depending on the phase noise in the acquired data. This plot uses the circular standard deviation of the wrapped phase distribution as a measure of noise and relates it to the root-mean-square error of the recovered, unwrapped phase.
Collapse
|
139
|
Experimental and computational analyses reveal dynamics of tumor vessel cooption and optimal treatment strategies. Proc Natl Acad Sci U S A 2019; 116:2662-2671. [PMID: 30700544 DOI: 10.1073/pnas.1818322116] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cooption of the host vasculature is a strategy that some cancers use to sustain tumor progression without-or before-angiogenesis or in response to antiangiogenic therapy. Facilitated by certain growth factors, cooption can mediate tumor infiltration and confer resistance to antiangiogenic drugs. Unfortunately, this mode of tumor progression is difficult to target because the underlying mechanisms are not fully understood. Here, we analyzed the dynamics of vessel cooption during tumor progression and in response to antiangiogenic treatment in gliomas and brain metastases. We followed tumor evolution during escape from antiangiogenic treatment as cancer cells coopted, and apparently mechanically compressed, host vessels. To gain deeper understanding, we developed a mathematical model, which incorporated compression of coopted vessels, resulting in hypoxia and formation of new vessels by angiogenesis. Even if antiangiogenic therapy can block such secondary angiogenesis, the tumor can sustain itself by coopting existing vessels. Hence, tumor progression can only be stopped by combination therapies that judiciously block both angiogenesis and cooption. Furthermore, the model suggests that sequential blockade is likely to be more beneficial than simultaneous blockade.
Collapse
|
140
|
Liu Y, Broberg MCG, Watanabe M, Rollins AM, Jenkins MW. SLIME: robust, high-speed 3D microvascular mapping. Sci Rep 2019; 9:893. [PMID: 30696870 PMCID: PMC6351571 DOI: 10.1038/s41598-018-37313-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/30/2018] [Indexed: 01/08/2023] Open
Abstract
Three dimensional (3D) microvascular imaging of cubic millimeter to centimeter size volumes often requires much time and expensive instruments. By combining optical clearing with a novel scatter-based optical coherence tomography (OCT) contrast agent, we have greatly extended OCT imaging depth in excised tissues while maintaining a simple and low cost approach that does not require in-depth OCT knowledge. The new method enables fast 3D microvascular mapping in large tissue volumes, providing a promising tool for investigating organ level microvascular abnormalities in large cohorts.
Collapse
Affiliation(s)
- Yehe Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Meredith C G Broberg
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA.,Division of Pediatric Critical Care, UH Rainbow Babies & Children's Hospital, Cleveland, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA. .,Department of Pediatrics, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
141
|
Nishida-Aoki N, Gujral TS. Emerging approaches to study cell-cell interactions in tumor microenvironment. Oncotarget 2019; 10:785-797. [PMID: 30774780 PMCID: PMC6366828 DOI: 10.18632/oncotarget.26585] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/27/2018] [Indexed: 01/19/2023] Open
Abstract
Cell-cell interactions are of crucial importance for tissue formation, homeostasis, regeneration processes, and immune response. Recent studies underlined contribution of cell-cell interaction in tumor microenvironment (TME) for tumor progression and metastasis. Cancer cells modify the host cells to tumor-supportive traits, and the modified host cells contribute to tumor progression by interacting with cancer cells and further modifying other normal cells. However, the complex interaction networks of cancer cells and host cells remained largely unknown. Recent advances in high throughput microscopy and single cells-based molecular analyses have unlocked a new era for studying cell-cell interactions in the complex tissue microenvironment at the resolution of a single cell. Here, we review various model systems and emerging experimental approaches that are used to study cell-cell interactions focusing on the studies of TME. We discuss strengths and weaknesses of each model system and each experimental approach, and how upcoming approaches can solve current fundamental questions of cell-cell interactions in TME.
Collapse
Affiliation(s)
- Nao Nishida-Aoki
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
142
|
Breckwoldt MO, Bode J, Sahm F, Krüwel T, Solecki G, Hahn A, Wirthschaft P, Berghoff AS, Haas M, Venkataramani V, von Deimling A, Wick W, Herold-Mende C, Heiland S, Platten M, Bendszus M, Kurz FT, Winkler F, Tews B. Correlated MRI and Ultramicroscopy (MR-UM) of Brain Tumors Reveals Vast Heterogeneity of Tumor Infiltration and Neoangiogenesis in Preclinical Models and Human Disease. Front Neurosci 2019; 12:1004. [PMID: 30686972 PMCID: PMC6335617 DOI: 10.3389/fnins.2018.01004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022] Open
Abstract
Diffuse tumor infiltration into the adjacent parenchyma is an effective dissemination mechanism of brain tumors. We have previously developed correlated high field magnetic resonance imaging and ultramicroscopy (MR-UM) to study neonangiogenesis in a glioma model. In the present study we used MR-UM to investigate tumor infiltration and neoangiogenesis in a translational approach. We compare infiltration and neoangiogenesis patterns in four brain tumor models and the human disease: whereas the U87MG glioma model resembles brain metastases with an encapsulated growth and extensive neoangiogenesis, S24 experimental gliomas mimic IDH1 wildtype glioblastomas, exhibiting infiltration into the adjacent parenchyma and along white matter tracts to the contralateral hemisphere. MR-UM resolves tumor infiltration and neoangiogenesis longitudinally based on the expression of fluorescent proteins, intravital dyes or endogenous contrasts. Our study demonstrates the huge morphological diversity of brain tumor models regarding their infiltrative and neoangiogenic capacities and further establishes MR-UM as a platform for translational neuroimaging.
Collapse
Affiliation(s)
- Michael O Breckwoldt
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Bode
- Schaller Research Group at the University of Heidelberg and the German Cancer Research Center (DKFZ), Molecular Mechanisms of Tumor Invasion, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Krüwel
- Schaller Research Group at the University of Heidelberg and the German Cancer Research Center (DKFZ), Molecular Mechanisms of Tumor Invasion, Heidelberg, Germany
| | - Gergely Solecki
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) Within the DKFZ, Heidelberg, Germany
| | - Artur Hahn
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Wirthschaft
- Schaller Research Group at the University of Heidelberg and the German Cancer Research Center (DKFZ), Molecular Mechanisms of Tumor Invasion, Heidelberg, Germany
| | - Anna S Berghoff
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) Within the DKFZ, Heidelberg, Germany
| | - Maximilian Haas
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany
| | - Varun Venkataramani
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) Within the DKFZ, Heidelberg, Germany.,Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) Within the DKFZ, Heidelberg, Germany.,Neurology Clinic and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Sabine Heiland
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Bendszus
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix T Kurz
- Neuroradiology Department, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) Within the DKFZ, Heidelberg, Germany.,Neurology Clinic and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Björn Tews
- Schaller Research Group at the University of Heidelberg and the German Cancer Research Center (DKFZ), Molecular Mechanisms of Tumor Invasion, Heidelberg, Germany
| |
Collapse
|
143
|
Solid stress in brain tumours causes neuronal loss and neurological dysfunction and can be reversed by lithium. Nat Biomed Eng 2019; 3:230-245. [PMID: 30948807 PMCID: PMC6452896 DOI: 10.1038/s41551-018-0334-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 11/25/2018] [Indexed: 12/12/2022]
Abstract
The compression of brain tissue by a tumour mass is believed to be a major cause of the clinical symptoms seen in patients with brain cancer. However, the biological consequences of these physical stresses on brain tissue are unknown. Here, via imaging studies in patients and by using mouse models of human brain tumours, we show that a subgroup of primary and metastatic brain tumours, classified as nodular on the basis of their growth pattern, exert solid stress on the surrounding brain tissue, causing a decrease in local vascular perfusion as well as neuronal death and impaired function. We demonstrate a causal link between solid stress and neurological dysfunction by applying and removing cerebral compression, which respectively mimic the mechanics of tumour growth and of surgical resection. We also show that, in mice, treatment with lithium reduces solid-stress-induced neuronal death and improves motor coordination. Our findings indicate that brain-tumour-generated solid stress impairs neurological function in patients, and that lithium as a therapeutic intervention could counter these effects.
Collapse
|
144
|
Melanoma tumour vasculature heterogeneity: from mice models to human. J Cancer Res Clin Oncol 2018; 145:589-597. [PMID: 30547320 DOI: 10.1007/s00432-018-2809-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/30/2018] [Indexed: 01/23/2023]
Abstract
Tumour angiogenesis is defined by an anarchic vasculature and irregularities in alignment of endothelial cells. These structural abnormalities could explain the variability in distribution of nanomedicines in various tumour models. Then, the main goal of this study was to compare and to characterize the tumour vascular structure in different mouse models of melanoma tumours (B16F10 and SK-Mel-28) and in human melanomas from different patients. Tumours were obtained by subcutaneous injection of 106 B16F10 and 3.106 SK-Mel-28 melanoma cells in C57BL/6 and nude mice, respectively. Tumour growth was evaluated weekly, while vasculature was analysed through fluorescent labelling via CD31 and desmin. Significant differences in tumour growth and mice survival were evidenced between the two melanoma models. A fast evolution of tumours was observed for B16F10 melanoma, reaching a tumour size of 100 mm3 in 7 days compared to SK-Mel-28 which needed 21 days to reach the same volumes. Important differences in vascularization were exposed between the melanoma models, characterized by a significant enhancement of vascular density and a significant lumen size for mice melanoma models compared to human. Immunostaining revealed irregularities in endothelium structure for both melanoma models, but structural differences of vasculature were observed, characterized by a stronger expression of desmin in SK-Mel-28 tumours. While human melanoma mainly develops capillaries, structural irregularities are also observed on the samples of this tumour model. Our study revealed an impact of cell type and tumour progression on the structural vasculature of melanoma, which could impact the distribution of drugs in the tumour environment.
Collapse
|
145
|
Nam AS, Ren J, Bouma BE, Vakoc BJ. Demonstration of Triband Multi-Focal Imaging with Optical Coherence Tomography. APPLIED SCIENCES (BASEL, SWITZERLAND) 2018; 8:2395. [PMID: 31308961 PMCID: PMC6628925 DOI: 10.3390/app8122395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We demonstrate an extended depth of focus optical coherence tomography (OCT) system based on the use of chromatic aberration to create displaced focal planes in the sample. The system uses a wavelength-swept source tuning over three spectral bands and three separate interferometers, each of which interfaces to a single illumination/collection fiber. The resulting three imaged volumes are merged in post-processing to generate an image with a larger depth of focus than is obtained from each band individually. The improvements are demonstrated in structural imaging of a porous phantom and a lipid-cleared murine brain, and by angiographic imaging of human skin. By using a coaxial approach with Gaussian beams, this approach enables an extended focus with relatively simple microscope optics and data-merging algorithms.
Collapse
Affiliation(s)
- Ahhyun Stephanie Nam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | - Jian Ren
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | - Brett E. Bouma
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Division of Health Sciences & Technology (HST), Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin J. Vakoc
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Division of Health Sciences & Technology (HST), Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
146
|
Ying Z, Minghui T, Feng B, Ke W. Tanshinone II A improves distribution and anti-tumor efficacy of pegylated liposomal doxorubicin via normalizing the structure and function of tumor vasculature in hepa1-6 hepatoma mice model. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30980-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
147
|
Andleeb F, Ullah H. Optical coherence tomography in early detection of malignancies. MINERVA BIOTECNOL 2018. [DOI: 10.23736/s1120-4826.18.02466-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
148
|
Khoradmehr A, Mazaheri F, Anvari M, Tamadon A. A Simple Technique for Three-Dimensional Imaging and Segmentation of Brain Vasculature U sing Fast Free-of-Acrylamide Clearing Tissue in Murine. CELL JOURNAL 2018; 21:49-56. [PMID: 30507088 PMCID: PMC6275429 DOI: 10.22074/cellj.2019.5684] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/30/2018] [Indexed: 12/26/2022]
Abstract
Objective Fast Free-of-Acrylamide Clearing Tissue (FACT) is a recently developed protocol for the whole tissue
three-dimensional (3D) imaging. The FACT protocol clears lipids using sodium dodecyl sulfate (SDS) to increase the
penetration of light and reflection of fluorescent signals from the depth of cleared tissue. The aim of the present study
was using FACT protocol in combination with imaging of auto-fluorescency of red blood cells in vessels to image the
vasculature of a translucent mouse tissues.
Materials and Methods In this experimental study, brain and other tissues of adult female mice or rats were dissected
out without the perfusion. Mice brains were sliced for vasculature imaging before the clearing. Brain slices and other
whole tissues of rodent were cleared by the FACT protocol and their clearing times were measured. After 1 mm of the
brain slice clearing, the blood vessels containing auto-fluorescent red blood cells were imaged by a z-stack motorized
epifluorescent microscope. The 3D structures of the brain vessels were reconstructed by Imaris software.
Results Auto-fluorescent blood vessels were 3D imaged by the FACT in mouse brain cortex. Clearing tissues of
mice and rats were carried out by the FACT on the brain slices, spinal cord, heart, lung, adrenal gland, pancreas, liver,
esophagus, duodenum, jejunum, ileum, skeletal muscle, bladder, ovary, and uterus.
Conclusion The FACT protocol can be used for the murine whole tissue clearing. We highlighted that the 3D imaging
of cortex vasculature can be done without antibody staining of non-perfused brain tissue, rather by a simple auto-
fluorescence.
Collapse
Affiliation(s)
- Arezoo Khoradmehr
- Research and Clinical Center for Infertility, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fahime Mazaheri
- Research and Clinical Center for Infertility, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Morteza Anvari
- Research and Clinical Center for Infertility, Yazd Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Biology and Anatomical Sciences, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. Electronic Address:
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran. Electronic Address:
| |
Collapse
|
149
|
Matsumoto Y, Asao Y, Sekiguchi H, Yoshikawa A, Ishii T, Nagae KI, Kobayashi S, Tsuge I, Saito S, Takada M, Ishida Y, Kataoka M, Sakurai T, Yagi T, Kabashima K, Suzuki S, Togashi K, Shiina T, Toi M. Visualising peripheral arterioles and venules through high-resolution and large-area photoacoustic imaging. Sci Rep 2018; 8:14930. [PMID: 30297721 PMCID: PMC6175891 DOI: 10.1038/s41598-018-33255-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/06/2018] [Indexed: 01/26/2023] Open
Abstract
Photoacoustic (PA) imaging (PAI) has been shown to be a promising tool for non-invasive blood vessel imaging. A PAI system comprising a hemispherical detector array (HDA) has been reported previously as a method providing high morphological reproducibility. However, further improvements in diagnostic capability will require improving the image quality of PAI and fusing functional and morphological imaging. Our newly developed PAI system prototype not only enhances the PA image resolution but also acquires ultrasonic (US) B-mode images at continuous positions in the same coordinate axes. In addition, the pulse-to-pulse alternating laser irradiation shortens the measurement time difference between two wavelengths. We scanned extremities and breasts in an imaging region 140 mm in diameter and obtained 3D-PA images of fine blood vessels, including arterioles and venules. We could estimate whether a vessel was an artery or a vein by using the S-factor obtained from the PA images at two wavelengths, which corresponds approximately to the haemoglobin oxygen saturation. Furthermore, we observed tumour-related blood vessels around breast tumours with unprecedented resolution. In the future, clinical studies with our new PAI system will help to elucidate various mechanisms of vascular-associated diseases and events.
Collapse
Affiliation(s)
- Yoshiaki Matsumoto
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasufumi Asao
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Japan Science and Technology Agency, ImPACT Program, Cabinet Office, Kyoto, Japan
| | - Hiroyuki Sekiguchi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aya Yoshikawa
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Ishii
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken-Ichi Nagae
- Medical Imaging Development Center, Canon Inc., Kyoto, Japan
| | | | - Itaru Tsuge
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Susumu Saito
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Takada
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Ishida
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masako Kataoka
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takaki Sakurai
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Yagi
- Japan Science and Technology Agency, ImPACT Program, Cabinet Office, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigehiko Suzuki
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Shiina
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
150
|
In-silico dynamic analysis of cytotoxic drug administration to solid tumours: Effect of binding affinity and vessel permeability. PLoS Comput Biol 2018; 14:e1006460. [PMID: 30296260 PMCID: PMC6193741 DOI: 10.1371/journal.pcbi.1006460] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 10/18/2018] [Accepted: 08/25/2018] [Indexed: 12/31/2022] Open
Abstract
The delivery of blood-borne therapeutic agents to solid tumours depends on a broad range of biophysical factors. We present a novel multiscale, multiphysics, in-silico modelling framework that encompasses dynamic tumour growth, angiogenesis and drug delivery, and use this model to simulate the intravenous delivery of cytotoxic drugs. The model accounts for chemo-, hapto- and mechanotactic vessel sprouting, extracellular matrix remodelling, mechano-sensitive vascular remodelling and collapse, intra- and extravascular drug transport, and tumour regression as an effect of a cytotoxic cancer drug. The modelling framework is flexible, allowing the drug properties to be specified, which provides realistic predictions of in-vivo vascular development and structure at different tumour stages. The model also enables the effects of neoadjuvant vascular normalisation to be implicitly tested by decreasing vessel wall pore size. We use the model to test the interplay between time of treatment, drug affinity rate and the size of the vessels’ endothelium pores on the delivery and subsequent tumour regression and vessel remodelling. Model predictions confirm that small-molecule drug delivery is dominated by diffusive transport and further predict that the time of treatment is important for low affinity but not high affinity cytotoxic drugs, the size of the vessel wall pores plays an important role in the effect of low affinity but not high affinity drugs, that high affinity cytotoxic drugs remodel the tumour vasculature providing a large window for the normalisation of the vascular architecture, and that the combination of large pores and high affinity enhances cytotoxic drug delivery efficiency. These results have implications for treatment planning and methods to enhance drug delivery, and highlight the importance of in-silico modelling in investigating the optimisation of cancer therapy on a personalised setting. One of the main challenges in optimising cancer therapy is understanding the in-vivo cancer environment and how it changes over time. The efficacy of chemotherapeutic drugs is known to be strongly dependent on blood vessel wall properties and the architecture of the developing tumour vasculature, which in turn are dependent on biochemical and mechanical interactions between cancer cells and their microenvironment. Here we present a novel in-silico modelling framework of dynamic tumour growth, angiogenesis and drug delivery, and we use it to explore biophysical factors governing the efficient delivery of cytotoxic drugs to solid tumours. We find that the time of treatment and vessel permeability are important factors for the efficacy of chemical agents with low binding affinity, that high affinity drugs can impact the tumour vasculature remodelling and bring vascular structure back to a more normalised state, and that the combination of large-sized vessel wall pores and high affinity enhances cytotoxic drug delivery and efficacy. These results have implications for treatment planning and optimisation, and show how in-silico models can be used to help understand and optimise cancer therapy.
Collapse
|