Open Access
10 November 2016 Label-free high-throughput detection and quantification of circulating melanoma tumor cell clusters by linear-array-based photoacoustic tomography
Pengfei Hai, Yong Zhou, Ruiying Zhang, Jun Ma, Yang Li, Jin-Yu Shao, Lihong V. Wang
Author Affiliations +
Abstract
Circulating tumor cell (CTC) clusters, arising from multicellular groupings in a primary tumor, greatly elevate the metastatic potential of cancer compared with single CTCs. High-throughput detection and quantification of CTC clusters are important for understanding the tumor metastatic process and improving cancer therapy. Here, we applied a linear-array-based photoacoustic tomography (LA-PAT) system and improved the image reconstruction for label-free high-throughput CTC cluster detection and quantification in vivo. The feasibility was first demonstrated by imaging CTC cluster ex vivo. The relationship between the contrast-to-noise ratios (CNRs) and the number of cells in melanoma tumor cell clusters was investigated and verified. Melanoma CTC clusters with a minimum of four cells could be detected, and the number of cells could be computed from the CNR. Finally, we demonstrated imaging of injected melanoma CTC clusters in rats in vivo. Similarly, the number of cells in the melanoma CTC clusters could be quantified. The data showed that larger CTC clusters had faster clearance rates in the bloodstream, which agreed with the literature. The results demonstrated the capability of LA-PAT to detect and quantify melanoma CTC clusters in vivo and showed its potential for tumor metastasis study and cancer therapy.

1.

Introduction

Metastasis, the spread of cancer cells from a primary site to distant organs leading to the growth of new tumors there, accounts for the majority of cancer deaths.1 Blood-borne metastasis comprises multiple steps, including intravazation, survival in the blood circulation, extravazation, and secondary growth in the distant organs, in which the rare circulating tumor cells (CTCs) are the key determinants of metastatic propensity.2 While most CTCs survive in the blood circulation in the form of single cells, CTC clusters also exist and elevate the metastatic potential by 23 to 50 folds.3 Detection and quantification of CTC clusters will provide valuable insights into metastasis as well as cancer therapy. Because the presence of CTCs is positively correlated with metastatic propensity,4 the detection and enumeration of rare CTCs will help to better evaluate the melanoma stage and predict metastatic potential during the diagnosis. In addition, tracking the concentration of CTCs in the circulatory system during treatment will likely be valuable indicators of both tumor responses to therapy and melanoma progression.

Taking advantage of tumor-specific physical and biological properties, such as sizes and surface biomarkers, multiple technologies have been developed to capture and isolate single CTCs and CTC clusters.57 However, most of these existing technologies require blood to be drawn to isolate and capture CTCs ex vivo, reducing the effective CTC detection sensitivity. Optical imaging technologies, such as confocal microscopy, in vivo flow cytometry, and optical coherence tomography, have been applied to detect CTCs in vivo.811 However, suffering from strong optical scattering in biological tissue, these techniques have shallow penetration, limiting them to imaging only small blood vessels, which results in low throughput for CTC detection.

By integrating optical excitation with acoustic detection, photoacoustic tomography (PAT) combines rich optical absorption contrasts with high ultrasonic spatial resolution at depths.12 With 100% relative sensitivity to optical absorption, i.e., a given percentage change in the optical absorption yields the same percentage change in the photoacoustic signal, PAT achieves structural, functional, metabolic, and mechanical imaging of biological tissue.1317 Taking advantage of the strong optical absorption of melanin, photoacoustic techniques have been successfully used for imaging and sensing melanoma CTCs. Photoacoustic flowmetry successfully detected circulating melanoma cells in human blood.18,19 Photoacoustic flow cytometry enables long-term monitoring of melanoma CTCs in vivo. However, these sensing techniques provide inadequate spatial resolution to further characterize melanoma CTCs.20 The microscopic implementation of PAT, optical-resolution photoacoustic microscopy (OR-PAM), images single melanoma CTCs in the blood flows with high spatial-temporal resolution.21 While providing valuable insights into CTCs on a single-cell level, OR-PAM suffers from low throughput.

Here, we apply a linear-array-based photoacoustic tomography (LA-PAT) system for label-free high-throughput melanoma CTC cluster detection and quantification in vivo. Exploiting the strong optical absorption of melanin in the melanoma tumor cells, LA-PAT can achieve label-free detection of melanoma CTC clusters in vivo. By analyzing the contrast-to-noise ratios (CNRs) of the photoacoustic signals, LA-PAT can quantify the number of cells in the CTC clusters and study their circulating kinetics in the bloodstream.

2.

Materials and Methods

2.1.

Linear-Array-Based Photoacoustic Tomography System

To image melanoma CTC clusters, we applied an LA-PAT system based on a handheld probe and improved the image reconstruction.22 A tunable optical parametric oscillator laser (680 to 970 nm, 20-Hz pulse repetition rate) was used for illumination. An excitation wavelength of 680 nm was chosen to achieve the optimal contrast between melanoma tumor cells and blood [Fig. 1(a)]. The laser beam was coupled into a fiber optical bundle that bifurcated into two optical fiber bundles. Laser beams coming out of the two optical fiber bundle strips (20×1.25  mm2) excited the object at an angle of incidence of 30 deg relative to the imaging plane [Figs. 1(b) and 1(c)]. During the experiments, the fluence on the tissue surface was about 10  mJ/cm2, well within the 20  mJ/cm2 safety limit set by the American National Standards Institute. The generated photoacoustic waves were detected by a linear array ultrasonic transducer (Visualsonics, Inc., LZ250, 21-MHz center frequency, 78% one-way bandwidth, 256 elements, 23×3  mm2 array size). The spatial resolutions of the system were 119  μm in the lateral direction, 86  μm in the axial direction, and 1237  μm in the elevational direction.23 Because there were only 64 channels in the data acquisition unit, four-to-one multiplexing was applied during image acquisition. For each laser pulse, the generated photoacoustic signals were captured sequentially by a quarter segment of the linear array (i.e., elements 1 to 64, 65 to 128, 129 to 192, and 193 to 256). Once the data were acquired from all four quarter segments, a two-dimensional photoacoustic image was reconstructed with the universal back-projection algorithm developed by our group.24 The reconstructed photoacoustic images were displayed in an imaging station (Vevo LAZR, Visualsonics, Inc.). Determined by the 20-Hz laser repetition rate and the four-to-one multiplexing in image acquisition, the frame rate was 5  frames/s.

Fig. 1

Absorption spectra of melanin and hemoglobin, and the front and side views of the handheld probe for LA-PAT. (a) Absorption spectra of melanin and hemoglobin. An excitation wavelength of 680 nm was chosen for the optimal CNR. (b) Lateral view of the handheld probe. (c) Elevational view of the handheld probe with a zoomed-in view.

JBO_22_4_041004_f001.png

2.2.

Tumor Cell Culture and Cluster Preparation

B16F10 melanoma tumor cells obtained from ATCC® were cultured in Dulbecco’s modified Eagle’s medium (Invitrogen) supplemented with 10% fetal bovine serum (Invitrogen) at 37°C in air with 5% CO2. Upon reaching 80% confluence, cells growing in a monolayer were incubated with 0.25% trypsin-EDTA solution (Invitrogen) to generate floating cell clusters of various sizes. After adding serum-containing medium to neutralize trypsin, the floating cell clusters were handled in two ways. To mimic melanoma CTC clusters of various sizes, floating cell clusters totaling 1×106 cells were directly suspended in 1 mL of bovine blood for ex vivo experiments or in 1 mL of collected rat blood for tail vein injection. To generate melanoma cell clusters of a certain size for CNR analysis, instead of directly suspending the floating cell clusters in blood, the clusters were mechanically dissociated by pipetting to generate a single-cell suspension. The single cells were then suspended in a blood–agar mixture at a desired concentration and then cooled to become solid. The concentration was controlled so that within each resolution voxel of LA-PAT there was a certain number of melanoma cells, i.e., 5, 10, 20, 30, 40, or 50. The solid mixture was then mechanically dissociated into particles smaller than the resolution voxel of LA-PAT to generate cell clusters of a certain size. The particles were then suspended in 1 mL of bovine blood for CNR analysis.

2.3.

Phantom Preparation

Silicone microtubes with 0.76 mm inner diameter (11-189-15C, Fisher Scientific) were perfused with bovine blood (905, Quad-Five) to mimic blood vessels. The microtubes were embedded 3 mm deep in tissue-mimicking gelatin phantoms. Optical scattering similar to that in biological tissue was achieved by adding 1% intralipid to the gelatin phantoms. Melanoma tumor cell clusters suspended in bovine blood were pumped through the microtubes with a syringe. A syringe pump (BSP-99M, Braintree Scientific) controlled the blood flow speed.

2.4.

Experimental Animals and CTC Cluster Injection

All experimental animal procedures were carried out in conformity with the laboratory animal protocol approved by the Animal Studies Committee at Washington University in St. Louis. Male nude rats (Hsd:RH-Foxn1rnu/Foxn1+, Harlan Co.; 12- to 13-weeks old; 330 to 350 g body weight) were used for all in vivo experiments. Throughout the experiments, the rats were kept under anesthesia with 2% vaporized isoflurane. A homemade animal holder immobilized the rats during the experiments.

3.

Results

3.1.

CNR Analysis of Melanoma Tumor Cell Clusters

To detect and quantify CTC clusters, we first used LA-PAT to image melanoma tumor cell clusters of known sizes and analyzed their CNRs. Clusters with an average of 5, 10, 20, 30, 40, and 50 cells were suspended in blood and imaged by LA-PAT. The CNR for each cluster was calculated [Fig. 2(a)]. The average CNRs were plotted as a function of the number of cells in the clusters, as measured with an optical microscope (OM) (Eclipse TS100, Nikon) [Fig. 2(b)]. The experimental data were fitted to a linear function

Eq. (1)

CNR=0.46×N.
Here, N represents the number of cells in a melanoma tumor cell cluster. Based on a 6-dB CNR threshold, a minimum of four cells can be resolved although this minimum was not directly observed. To verify the relationship between the CNR and the number of cells in the clusters, another independent experiment was performed. Melanoma tumor cell clusters consisting of a uniform but unknown number of cells were equally divided into six groups. Three randomly chosen groups were suspended in blood and imaged by LA-PAT. The numbers of cells in the clusters were calculated based on the CNRs and the relationship above. The other three groups were examined with an OM to determine the numbers of cells in the clusters. The number of cells in the melanoma tumor cell clusters measured by LA-PAT and optical microscopy agreed well with each other [Fig. 2(c)], confirming the relationship between the numbers of cells in the clusters and the CNRs in LA-PAT.

Fig. 2

CNR analysis of melanoma tumor cell clusters. (a) CNR distributions measured with LA-PAT for melanoma tumor cell clusters of different sizes. (b) Average CNRs in LA-PAT as a function of the numbers of cells in melanoma tumor cell clusters. (c) The numbers of cells in melanoma tumor cell clusters measured by LA-PAT agreed well with those measured by an OM.

JBO_22_4_041004_f002.png

3.2.

LA-PAT of CTC Clusters Ex Vivo

We then demonstrated the ability of LA-PAT to detect and quantify CTC clusters ex vivo. A mixture of melanoma tumor cell clusters of various sizes was equally divided into six groups. Three randomly chosen groups were suspended in blood, pumped through microtubes to mimic flowing melanoma CTC clusters, and imaged by LA-PAT [Videos 13]. Each melanoma tumor cell cluster was captured by LA-PAT multiple times, and the average CNR was used to calculate the number of cells in the cluster. Based on the calculated number of cells in the clusters, the melanoma tumor cell clusters were divided into three categories: 10, 11 to 20, and 21. Typical photoacoustic images of the three categories are shown in Figs. 3(a)3(c). The other three groups were measured with an OM to determine the numbers of cells in the clusters and were divided into the same three categories. The size distributions of the clusters measured by LA-PAT and optical microscopy agreed well with each other with a correlation coefficient of 0.96 [Fig. 3(d)], and the results further demonstrated the ability of LA-PAT to detect and quantify melanoma CTC clusters.

Fig. 3

LA-PAT of CTC clusters ex vivo. LA-PAT of a flowing melanoma tumor cell cluster with (a) 7 cells (Video 1), (b) 14 cells (Video 2), and (c) 29 cells (Video 3). The cell numbers are calculated based on the CNR curve in Fig. 2. (d) Distributions of the numbers of cells in melanoma tumor cell clusters, as measured by LA-PAT and OM (Video 1, MP4, 1.91 MB [URL: http://dx.doi.org/10.1117/1.JBO.22.4.041004.1]; Video 2, MP4, 2.01 MB [URL: http://dx.doi.org/10.1117/1.JBO.22.4.041004.2]; Video 3, MP4, 1.84 MB [URL: http://dx.doi.org/10.1117/1.JBO.22.4.041004.3]).

JBO_22_4_041004_f003.png

3.3.

LA-PAT of CTC Clusters In Vivo

To demonstrate the feasibility of LA-PAT in vivo, we imaged melanoma CTC clusters injected into rats. Melanoma tumor cell clusters suspended in 1 mL of rat blood at a concentration of 1×106  cells/mL were injected into the same rat from which the blood was collected a few minutes earlier. The rat tail vein was then monitored by LA-PAT for 45 min to study the circulation kinetics and the clearance rates of CTC clusters. CTC clusters were detected by LA-PAT immediately after injection and during circulating in the rat bloodstreams [Figs. 4(a) and 4(b) and Videos 4 and 5]. The numbers of cells in the CTC clusters were calculated based on the CNRs. The CTC clusters were categorized into three groups based on the calculated number of cells and the clearance curves of the three groups are shown in Fig. 4(c). Larger CTC clusters showed faster clearance rates from the bloodstream than smaller CTC clusters.

Fig. 4

LA-PAT of CTC clusters in rat tail veins in vivo. (a) LA-PAT of CTC clusters in a rat tail vein in vivo immediately after injection (Video 4). (b) LA-PAT of CTC clusters in the rat tail vein in vivo 45 min after injection (Video 5). (c) Clearance curves of small (10 cells), medium (11 to 20 cells), and large (21 cells) CTC clusters in rat bloodstreams (Video 4, MP4, 604 kB [URL: http://dx.doi.org/10.1117/1.JBO.22.4.041004.4]; Video 5, MP4, 613 kB [URL: http://dx.doi.org/10.1117/1.JBO.22.4.041004.5]).

JBO_22_4_041004_f004.png

4.

Discussion and Conclusions

Compared with single CTCs, CTC clusters are rarer, but have a higher metastatic potential. CTC clusters have a faster clearance rate, i.e., a shorter lifetime, in the bloodstream than single CTCs, making them even more difficult to detect. Here, we applied LA-PAT to detect and quantify melanoma CTC clusters in rat in vivo. Since the spatial resolutions of LA-PAT were not enough to resolve single melanoma tumor cells, the numbers of cells in the CTC clusters were quantified based on the CNRs. A linear relationship between the CNRs and the numbers of cells in the CTC clusters was found and verified. In ex vivo experiments, LA-PAT quantified the numbers of cells in the CTC clusters, and the results were validated by an OM. In experiments in vivo, LA-PAT detected and quantified melanoma CTC clusters immediately after injection, as well as when they were circulating in the rat bloodstreams. The results by LA-PAT also showed that larger CTC clusters have faster clearance rates.

The melanoma CTC detection sensitivity of LA-PAT is determined by the CNR. Because the noise is dominated by the fluctuating counts of red blood cells in each resolution voxel, the detection sensitivity is largely dependent on the spatial resolutions of LA-PAT. On one hand, to achieve single cell sensitivity in CTC detection, a higher frequency ultrasonic transducer could be used at the expense of penetration depth. On the other hand, to achieve deeper imaging, a lower frequency ultrasonic transducer can be employed, at the expense of CTC detection sensitivity. The CTC detection sensitivity can be further improved by performing spectral unmixing using multiwavelength illumination. To achieve multiwavelength imaging of flowing CTCs, a second laser with a different wavelength needs to be incorporated into the system.

It is worth pointing out that currently LA-PAT detects only CTCs originating from primary tumors in which cells express melanin. To detect CTCs originating from amelanotic melanoma tumors, other tumor-specific physiological properties, such as sizes and surface biomarkers, can be utilized. The quantification of CTC cluster sizes is indeed based on the assumption that the melanin content is relatively uniform in the tumor cells. This assumption often holds true for tumor cells originating from the same primary tumor. However, if there are several primary tumors, it may affect the accuracy of quantifying CTC cluster sizes.

LA-PAT has the potential to be a powerful tool for both preclinical tumor metastasis study and clinical cancer diagnosis and therapy. As a useful technique for researchers and scientists to better understand the relationship between CTCs and tumor metastasis, LA-PAT can employ ultrasonic transducers of different frequencies for target vessels at various depths and different animal models. Other physiological parameters, including vessel diameter and blood flow speed, can be measured concurrently for further understanding of CTC flow dynamics. LA-PAT can also be used by clinicians to monitor the changes of CTC concentrations in patients’ circulatory systems and to evaluate the outcome of cancer therapy. To further improve LA-PAT for such clinical applications, an automatic CTC detection and counting algorithm should be developed.

Metastasis is a complex biological process that involves multiple steps and many parameters. Photoacoustic imaging, with the ability to quantify numerous parameters at multiple scales based on the same optical absorption contrast, can provide comprehensive information about tumor metastasis. OR-PAM can study CTCs on a single-cell level.21 LA-PAT has been demonstrated to detect and quantify CTC clusters. In addition, photoacoustic imaging can measure other important biological parameters, including the oxygen saturation of hemoglobin, metabolic rate, and tumor stiffness,2527 all of which are closely related to metastasis.2830 With the capability to detect and quantify CTC clusters and the potential to provide multidimensional information on tumor metastasis, LA-PAT is a promising tool for both preclinical cancer metastasis study and clinical tumor therapy.

Acknowledgments

The authors appreciate Professor James Ballard’s close reading of the paper and Eric Zhang’s assistance in cell cluster preparation. This work was sponsored by the National Science Foundation Grant No. 1255930 and the National Institutes of Health (NIH) Grant Nos. R01 CA186567 (NIH Director’s Transformative Research Award) and S10 RR026922 (VSI). L. V. Wang has a financial interest in Microphotoacoustics, Inc., which, however, did not support this work.

References

1. 

D. X. Nguyen, P. D. Bos and J. Massagué, “Metastasis: from dissemination to organ-specific colonization,” Nat. Rev. Cancer, 9 (4), 274 –284 (2009). http://dx.doi.org/10.1038/nrc2622 NRCAC4 1474-175X Google Scholar

2. 

I. J. Fidler, “The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited,” Nat. Rev. Cancer, 3 (6), 453 –458 (2003). http://dx.doi.org/10.1038/nrc1098 NRCAC4 1474-175X Google Scholar

3. 

N. Aceto et al., “Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis,” Cell, 158 (5), 1110 –1122 (2014). http://dx.doi.org/10.1016/j.cell.2014.07.013 CELLB5 0092-8674 Google Scholar

4. 

M. Cristofanilli et al., “Circulating tumor cells, disease progression, and survival in metastatic breast cancer,” N. Engl. J. Med., 351 (8), 781 –791 (2004). http://dx.doi.org/10.1056/NEJMoa040766 NEJMBH Google Scholar

5. 

M. Yu et al., “Circulating tumor cells: approaches to isolation and characterization,” J. Cell Biol., 192 (3), 373 –382 (2011). http://dx.doi.org/10.1083/jcb.201010021 JCLBA3 0021-9525 Google Scholar

6. 

S. Nagrath et al., “Isolation of rare circulating tumour cells in cancer patients by microchip technology,” Nature, 450 (7173), 1235 –1239 (2007). http://dx.doi.org/10.1038/nature06385 Google Scholar

7. 

A. F. Sarioglu et al., “A microfluidic device for label-free, physical capture of circulating tumor cell clusters,” Nat. Methods, 12 (7), 685 –691 (2015). http://dx.doi.org/10.1038/nmeth.3404 1548-7091 Google Scholar

8. 

E. Sahai, “Illuminating the metastatic process,” Nat. Rev. Cancer, 7 (10), 737 –749 (2007). http://dx.doi.org/10.1038/nrc2229 NRCAC4 1474-175X Google Scholar

9. 

H. Seo et al., “In vivo quantitation of injected circulating tumor cells from great saphenous vein based on video-rate confocal microscopy,” Biomed. Opt. Express, 6 (6), 2158 –2167 (2015). http://dx.doi.org/10.1364/BOE.6.002158 BOEICL 2156-7085 Google Scholar

10. 

I. Georgakoudi et al., “In vivo flow cytometry: a new method for enumerating circulating cancer cells,” Cancer Res., 64 (15), 5044 –5047 (2004). http://dx.doi.org/10.1158/0008-5472.CAN-04-1058 Google Scholar

11. 

S. M. Azarin et al., “In vivo capture and label-free detection of early metastatic cells,” Nat. Commun., 6 8094 (2015). http://dx.doi.org/10.1038/ncomms9094 NCAOBW 2041-1723 Google Scholar

12. 

L. V. Wang and S. Hu, “Photoacoustic tomography: in vivo imaging from organelles to organs,” Science, 335 (6075), 1458 –1462 (2012). http://dx.doi.org/10.1126/science.1216210 SCIEAS 0036-8075 Google Scholar

13. 

P. Hai et al., “Near-infrared optical-resolution photoacoustic microscopy,” Opt. Lett., 39 (17), 5192 –5195 (2014). http://dx.doi.org/10.1364/OL.39.005192 OPLEDP 0146-9592 Google Scholar

14. 

J. Ma et al., “Grueneisen relaxation photoacoustic microscopy in vivo,” J. Biomed. Opt., 21 (6), 066005 (2016). http://dx.doi.org/10.1117/1.JBO.21.6.066005 JBOPFO 1083-3668 Google Scholar

15. 

X. Wang et al., “Noninvasive laser-induced photoacoustic tomography for structural and functional in vivo imaging of the brain,” Nat. Biotechnol., 21 (7), 803 –806 (2003). http://dx.doi.org/10.1038/nbt839 NABIF9 1087-0156 Google Scholar

16. 

J. Yao et al., “Label-free oxygen-metabolic photoacoustic microscopy in vivo,” J. Biomed. Opt., 16 (7), 076003 (2011). http://dx.doi.org/10.1117/1.3594786 JBOPFO 1083-3668 Google Scholar

17. 

P. Hai et al., “Photoacoustic elastography,” Opt. Lett., 41 (4), 725 –728 (2016). http://dx.doi.org/10.1364/OL.41.000725 OPLEDP 0146-9592 Google Scholar

18. 

R. M. Weight et al., “Photoacoustic detection of metastatic melanoma cells in the human circulatory system,” Opt. Lett., 31 (20), 2998 –3000 (2006). http://dx.doi.org/10.1364/OL.31.002998 OPLEDP 0146-9592 Google Scholar

19. 

R. M. Weight, P. S. Dale and J. A. Viator, “Detection of circulating melanoma cells in human blood using photoacoustic flowmetry,” in Annual Int. Conf. of the IEEE Engineering in Medicine and Biology Society (EMBC ’09), 106 –109 (2009). http://dx.doi.org/10.1109/IEMBS.2009.5335145 Google Scholar

20. 

E. I. Galanzha and V. P. Zharov, “Photoacoustic flow cytometry,” Methods, 57 (3), 280 –296 (2012). http://dx.doi.org/10.1016/j.ymeth.2012.06.009 MTHDE9 1046-2023 Google Scholar

21. 

L. Wang et al., “Photoacoustic imaging of single circulating melanoma cells in vivo,” Proc. SPIE, 9323 93230A (2015). http://dx.doi.org/10.1117/12.2076987 PSISDG 0277-786X Google Scholar

22. 

A. Needles et al., “Development and initial application of a fully integrated photoacoustic micro-ultrasound system,” IEEE Trans. Ultrason. Ferroelectr. Freq. Control, 60 (5), 888 –897 (2013). http://dx.doi.org/10.1109/TUFFC.2013.2646 ITUCER 0885-3010 Google Scholar

23. 

P. Hai et al., “Photoacoustic tomography of vascular compliance in humans,” J. Biomed. Opt., 20 (12), 126008 (2015). http://dx.doi.org/10.1117/1.JBO.20.12.126008 JBOPFO 1083-3668 Google Scholar

24. 

M. Xu and L. V. Wang, “Universal back-projection algorithm for photoacoustic computed tomography,” Phys. Rev. E, 71 (1), 016706 (2005). http://dx.doi.org/10.1103/PhysRevE.71.016706 Google Scholar

25. 

H. F. Zhang et al., “Functional photoacoustic microscopy for high-resolution and noninvasive in vivo imaging,” Nat. Biotechnol., 24 (7), 848 –851 (2006). http://dx.doi.org/10.1038/nbt1220 NABIF9 1087-0156 Google Scholar

26. 

J. Yao et al., “Noninvasive photoacoustic computed tomography of mouse brain metabolism in vivo,” NeuroImage, 64 (1), 257 –266 (2013). http://dx.doi.org/10.1016/j.neuroimage.2012.08.054 NEIMEF 1053-8119 Google Scholar

27. 

P. Hai et al., “Quantitative photoacoustic elastography in humans,” J. Biomed. Opt., 21 (6), 066011 (2016). http://dx.doi.org/10.1117/1.JBO.21.6.066011 JBOPFO 1083-3668 Google Scholar

28. 

G. P. Luke and S. Y. Emelianov, “Label-free detection of lymph node metastases with US-guided functional photoacoustic imaging,” Radiology, 277 (2), 435 –442 (2015). http://dx.doi.org/10.1148/radiol.2015141909 RADLAX 0033-8419 Google Scholar

29. 

Y. Fong et al., “Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis,” Nat. Cell Biol., 17 (2), 183 –194 (2015). http://dx.doi.org/10.1038/ncb3094 Google Scholar

30. 

J. Fenner et al., “Macroscopic stiffness of breast tumors predicts metastasis,” Sci. Rep., 4 5512 (2014). http://dx.doi.org/10.1038/srep05512 SRCEC3 2045-2322 Google Scholar

Biography

Pengfei Hai is currently a PhD candidate in biomedical engineering at Washington University in St. Louis, under the supervision of Prof. Lihong V. Wang. He received his BS degree in biomedical engineering from Shanghai Jiao Tong University in 2012. His research interests include the technical development and biomedical applications of photoacoustic imaging.

Yong Zhou is currently a graduate student in biomedical engineering at Washington University in St. Louis, under the supervision of Dr. Lihong V. Wang, Gene K. Beare Distinguished Professor. His research focuses on the development of photoacoustic imaging systems.

Ruiying Zhang received her BS degree in optical information science and technology from Wuhan University in Hubei, China. She is currently a PhD student in the Department of Biomedical Engineering at Washington University in St. Louis. Her research interests include optical and photoacoustic/ultrasound imaging with biomedical applications.

Jun Ma received his BSc degree from Huazhong University of Science and Technology and a PhD from the Hong Kong Polytechnic University. Currently, he is working as a postdoctoral research associate at the Biomedical Engineering Department, Washington University in St. Louis. His research interests include optical acoustic and ultrasonic transducer and photoacoustic imaging and its biomedical applications.

Yang Li received his degree of BS summa cum laude in biomedical engineering from the State University of New York at Buffalo in 2013. Then he joined the Optical Imaging Laboratory at Washington University in St. Louis to pursue the degree of PhD in the same field. He is currently working on high-speed, high-throughput detection methods for photoacoustic imaging.

Jin-Yu Shao: Biography is not available.

Lihong V. Wang is a Gene K. Beare Distinguished Professor at Washington University, has published 437 journal articles (h-index=108, citations>47,000) and delivered 433 keynote/plenary/invited talks. His laboratory published the first functional photoacoustic CT and three-dimensional photoacoustic microscopy. He received the Goodman Award for his Biomedical Optics textbook, NIH Director’s Pioneer Award, OSA Mees Medal, IEEE Technical Achievement and Biomedical Engineering Awards, SPIE Britton Chance Biomedical Optics Award, and an honorary doctorate from Lund University, Sweden.

© 2016 Society of Photo-Optical Instrumentation Engineers (SPIE) 1083-3668/2016/$25.00 © 2016 SPIE
Pengfei Hai, Yong Zhou, Ruiying Zhang, Jun Ma, Yang Li, Jin-Yu Shao, and Lihong V. Wang "Label-free high-throughput detection and quantification of circulating melanoma tumor cell clusters by linear-array-based photoacoustic tomography," Journal of Biomedical Optics 22(4), 041004 (10 November 2016). https://doi.org/10.1117/1.JBO.22.4.041004
Published: 10 November 2016
Lens.org Logo
CITATIONS
Cited by 39 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Melanoma

Tumors

In vivo imaging

Blood

Cancer

Photoacoustic tomography

Video


CHORUS Article. This article was made freely available starting 10 November 2017

Back to Top