Epi-detected Hyperspectral Stimulated Raman ... - ACS Publications

Publication Date (Web): August 2, 2018 ... SRS images at representative Raman shifts (e.g., 2845, 2885 and 2935 cm-1) delineate the biochemical ... SR...
1 downloads 0 Views 2MB Size
Subscriber access provided by UNIV OF DURHAM

Article

Epi-detected Hyperspectral Stimulated Raman Scattering Microscopy for Label-free Molecular Subtyping of Glioblastomas Bae Kideog, Wei Zheng, Kan Lin, See Wee Lim, Yuk Kien Chong, Carol Tang, Nicolas King, Christopher Beng Ti Ang, and Zhiwei Huang Anal. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.analchem.8b01677 • Publication Date (Web): 02 Aug 2018 Downloaded from http://pubs.acs.org on August 2, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

Epi-detected Hyperspectral Stimulated Raman Scattering Microscopy for Label-

2

free Molecular Subtyping of Glioblastomas

3 4

Bae Kideog1, Wei Zheng1,2, Kan Lin1,2, See Wee Lim3, Yuk Kien Chong3, Carol

5

Tang3,4,5, Nicolas K King3,4, Christopher Beng Ti Ang3,4,6,7, and Zhiwei Huang1*

6 7

1

8

Engineering, National University of Singapore, Singapore 117576

9

2

Optical Bioimaging Laboratory, Department of Biomedical Engineering, Faculty of

Department of Medicine, Yong Loo Lin School of Medicine, National University of

10

Singapore, Singapore 119260

11

3

National Neuroscience Institute, Singapore 308433

12

4

Duke-National University of Singapore Medical School, Singapore 169857

13

5

Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer

14

Research, National Cancer Centre, Singapore 169610

15

6

16

Singapore, Singapore 117597

17

7

18

Research (A*STAR), Singapore 117609

Department of Physiology, Yong Loo Lin School of Medicine, National University of

Singapore Institute for Clinical Sciences, Agency for Science, Technology and

19 20

*Correspondence to:

21

Dr. Zhiwei Huang, Optical Bioimaging Laboratory, Department of Biomedical

22

Engineering, Faculty of Engineering, National University of Singapore, 9 Engineering

23

Drive 1, Singapore 117576

24

Tel: +65- 6516-8856

25

Fax: +65- 6872-3069

26

E-mail: [email protected]

27 28

Running title: SRS imaging for label-free molecular subtyping of GBMs

29 30

Key words: glioblastoma; stimulated Raman scattering microscopy; multivariate

31

curve resolution, principal component analysis, linear discriminant analysis

32

1

ACS Paragon Plus Environment

Analytical Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

ABSTRACT

2

We report the development and implementation of an epi-detected spectral-focusing

3

hyperspectral stimulated Raman scattering (SRS) imaging technique for label-free

4

biomolecular subtyping of glioblastomas (GBMs). The hyperspectral SRS imaging

5

technique developed generates SRS image stacks (from 2800 to 3020 cm-1 at 7 cm-1

6

intervals) within 30 s through controlling the time delay between the chirped pump

7

and Stokes beams. SRS images at representative Raman shifts (e.g., 2845, 2885 and

8

2935 cm-1) delineate the biochemical variations and morphological differences

9

between proneural and mesenchymal subtypes of GBMs. Multivariate curve

10

resolution (MCR) analysis on hyperspectral SRS images enables the quantification of

11

major biomolecule distributions in mesenchymal and proneural GBMs. Further

12

principal component analysis (PCA) and linear discriminant analysis (LDA) together

13

with leave-one SRS spectrum-out, cross-validation (LOOCV) yields a diagnostic

14

sensitivity of 96.7% (29/30) and specificity of 88.9% (28/36) for differentiation

15

between mesenchymal and proneural subtypes of GBMs. This study shows great

16

potential of applying hyperspectral SRS imaging technique developed for rapid, label-

17

free molecular subtyping of GBMs in neurosurgery.

18 19

2

ACS Paragon Plus Environment

Page 2 of 33

Page 3 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

1. Introduction

2

Glioblastoma (GBM) is the most aggressive brain tumor with fatality rate of

3

13,000 deaths each year in the United States1. Despite the implementation of surgical

4

resection and the postsurgical therapies (radiotherapy, chemotherapy) together, the

5

overall median survival remains at ~12 to 15 months2–5. Inter-patient variability to

6

treatment response is also commonly observed. In recent years, with the development

7

of The Cancer Genome Atlas (TCGA) efforts to deep-profile GBM tumors, evidence

8

has suggested that the poor prognosis could be attributed to the molecular complexity

9

and heterogeneity of GBM, accounting for significant differences in response to the

10

treatments6,7. These findings underscore the limitations of traditional pathologic

11

analyses to diagnose and subsequently influence treatment decisions. The current

12

histological system for brain tissue classification is unable to distinguish the

13

molecular subtypes of GBMs due to the lack of their cellular differences by

14

histopathology8. Recent research on the genomic and proteomic profiling can be used

15

to differentiate the subtypes with features that are described as proneural, proliferative

16

and mesenchymal9. The genetic approaches involve multiple steps for sample

17

preparation (e.g., homogenisation of tissue specimens, extraction and amplification of

18

DNA strains, etc) which make the overall process very labor-intensive and time-

19

consuming, unsuited for intraoperative diagnostic uses. It is highly desirable to

20

develop a rapid screening imaging technique with molecular specificity for GBM

21

molecular subtyping in neurosugery.

22

Stimulated Raman scattering (SRS) microscopy is a nonresonant background-free

23

vibrational spectroscopic imaging technique with high biochemical specificity10–12. In

24

SRS imaging, a pump wave (ωp) and a Stokes wave (ωs < ωp) are tightly focused onto

25

the sample; if the frequency difference (ωp − ωs) matches the vibrational frequency of

3

ACS Paragon Plus Environment

Analytical Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

the specific chemical bond, the SRS signal (translated as a loss of the pump beam

2

intensity or an increase of the Stokes beam intensity) can be resonantly enhanced up

3

to 104-fold with phase-sensitive detection. While the fixed wavelengths of the two

4

beams only allow resonance with a single Raman band, a broader spectral information

5

can be obtained by tuning the frequency of either the pump or Stokes beams for

6

acquisition of hyperspectral SRS images. With the high biochemical selectivity and

7

sensitivity, SRS microscopy has emerged as an appealing tool for label-free,

8

quantitative imaging in biological and biomedical systems13–19. SRS technique has

9

been explored for diagnosis and tumor margin delineation of GBMs in brain tissue

10

samples20–23. But to date, SRS microscopy has not been investigated for

11

differentiation between different molecular subtypes of GBMs in brain tissue. In this

12

study, we report the development and implementation of an epi-detected hyperspectral

13

stimulated Raman scattering (SRS) imaging technique based on spectral-focusing24

14

for label-free biomolecular subtyping of GBMs without pretreatments of tissue

15

samples. The hyperspectral SRS images of GBMs acquired delineate the

16

morphological and spectroscopic differences between the mesenchymal and proneural

17

GBMs for histopathological and biomolecular assessments without labeling.

18

Multivariate curve resolution (MCR) analysis on hyperspectral SRS images and

19

multivariate statistical analysis (e.g., principal components analysis (PCA), linear

20

discriminant analysis (LDA)) on SRS spectra are conducted to establish diagnostic

21

model for differentiation between the molecular subtypes of GBMs.

22 23

2. Materials and Methods

24

2.1 Epi-detected spectral-focusing hyperspectral SRS microscopy

25

Figure 1 shows the schematic of an epi-detected spectral-focusing hyperspectral

4

ACS Paragon Plus Environment

Page 4 of 33

Page 5 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

SRS microscopy system developed for label-free tissue imaging. The excitation light

2

source consists of a femtosecond (fs) laser (Insight DS dual, Spectra-Physics Inc.)

3

with two laser outputs: The fixed laser output generates 1041 nm fs laser pulses

4

(pulsewidth of 120 fs) with a repetition rate of 80 MHz, serving as the Stokes beam

5

for SRS imaging. The tunable output ranging from 680 to 1300 nm (pulsewidth of

6

100 fs) is used as the pump beam for SRS imaging. For SRS imaging, the intensity of

7

1041 nm Stokes beam is modulated at 20 MHz by an electro-optic modulator (EOM)

8

(APE-Berlin). The 1041 nm Stokes beam are collinearly combined with the pump

9

beam through a dichroic mirror and delivered into a multiphoton scanning microscope

10

(MPM-4R, Thorlabs Inc.). For epi-SRS detection, a home-built polarization control

11

module consisting of a quarter-wave plate (AQWP10M-980, Thorlabs) and a

12

polarizing beam-splitter (PBS252, Thorlabs), whose optical axis are set at 450 with

13

respect to each other, is placed before the objective to circularly polarize the Stokes

14

and pump beams. The circularly polarized excitation laser beams are scanned with a

15

pair of galvo mirrors and focused onto the sample by the objective (XLUMPLFLN

16

20X, NA=1.0, Olympus Inc.) for tissue imaging. A portion of the scattered beams is

17

epi-collected by the objective, and directed towards the same polarization control

18

module. Another round of transmission through the quarter-wave plate in the module

19

vertically polarizes the collected beam, which then allows them to be reflected

20

towards a large area photodiode (FDS1010, Thorlabs Inc.) by the subsequent

21

polarizing beam-splitter. Before the photodiode, a bandpass filter is used to spectrally

22

isolate the Stokes beam so that only the pump beam alone will be detected. The

23

modulation of the pump beam intensity due to the stimulated Raman loss (SRL)

24

process is demodulated by a lock-in amplifier to obtain the SRL signal. The time

25

constant of the lock-in amplifier is set to be 2 µs to match to the dwell time set for

5

ACS Paragon Plus Environment

Analytical Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

SRS imaging. For hyperspectral SRS imaging, the fs pump and Stokes laser beams

2

are chirped by using SF-57 glass rods with the lengths of 48 and 50 cm to generate the

3

1.7 ps pump beam and 2 ps Stokes beam, respectively, before combined onto a

4

dichroic mirror. The Raman shift differences between the pump and Stokes is scanned

5

by controlling the time delay between the pump and Stokes pulses. The movement of

6

the motorized delay stage is synchronized to the frame trigger of the microscope to

7

achieve automatic spectral scanning of SRS images at different Raman shifts. The

8

SRS intensity changes due to differences in the pulse overlap of the pump and Stokes

9

beam during the scanning are calibrated by measuring the intensity changes of

10

Rhodamine 6G over the same scanning range24. We have evaluated the relationship of

11

epi-detected SRS signal versus the thickness of fresh pork brain tissue (Figure S1). It

12

shows that epi-detected SRS signal increases with the increasing of brain tissue

13

thickness, and reaches a maximum signal level when tissue thickness goes up to 1

14

mm, probably due to the strong back-scattering effect in brain tissue25. The above

15

results indicate that the epi-detected spectral-focusing hyperspectral SRS microscopy

16

system developed in this work has an advantage of rapid characterization of bulky

17

brain tissues without tissue pretreatment in clinical settings.

18 19

2.2 Tissue Samples

20

Graded brain tumor specimens were obtained with written informed consent, as part

21

of a National Neuroscience Institute study protocol approved by the SingHealth

22

Centralized Institutional Review Board. The brain tissue samples are obtained from

23

National Neuroscience Institute (NNI), Singapore. The tissue samples are snap-frozen

24

in liquid nitrogen and stored at -80°C before sending for SRS imaging. The tissue

25

specimens (~3x2x1 mm3 in size) are thawed first, and then sandwiched between a

6

ACS Paragon Plus Environment

Page 6 of 33

Page 7 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

microscope slide and coverslip for epi-detected hyperspectral SRS imaging. In this

2

preliminary study, 9 brain tissue samples are used for SRS imaging, including 5

3

mesenchymal and 6 proneural GBM subtypes based on The Cancer Genome Atlas

4

(TCGA) Research Network.

5 6

2.3 Multivariate statistical analysis

7

In this study, multivariate curve resolution (MCR) analysis is used to decompose

8

the hyperspectral SRS image stack into the MCR spectrum of each major component

9

as well as the chemical distributions in the tissue. The underlying principle of MCR is

10

based on the bilinear model of spectrum of an unknown mixture where the measured

11

data matrix (D) may be decomposed into pure concentration maps (C) and pure

12

spectra (ST) of the k species of the sample as follows:

13

 = ∑    +



(1)

14

where E is the error matrix containing the residual variation of the data. The number

15

of components (k) contributing to the data matrix can be determined by principal

16

component analysis (PCA). With an initial estimation for spectra matrix S, the C and

17

ST are calculated and then optimized iteratively using alternative least squares (ALS)

18

algorithm. In each iteration, the fitting result with raw data is compared with the

19

percentage of variance explained. The process continues until the fitting error reaches

20

the convergence set (1%). In the optimization process, non-negative concentrations

21

and spectra are imposed as constraint. All MCR analysis is performed using the

22

MCR-ALS toolbox in MATLAB (MathWorks, Natick, MA).

23

For tissue classification, the multivariate statistical analysis technique (e.g.,

24

principal components analysis and linear discriminant analysis (PCA-LDA)) is used

25

as a statistical model. First, PCA reduces the dimensionality of the SRS spectrum

7

ACS Paragon Plus Environment

Analytical Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

ranging from 2800 to 3020 cm−1 with a subset of 30 data points. The SRS spectra are

2

first standardized to ensure that the mean of the spectra is zero and the standard

3

deviation (SD) is one, accounting for the influence of inter- and/or intra-subject

4

spectral variability on PCA. Mean centering ensure that the principal components

5

(PCs) form an orthogonal basis. Thus, PCA extracts a set of orthogonal PCs

6

comprising loadings and scores that account for most of the total variance in original

7

spectra. Each loading vector is associated with the original spectrum via PC score,

8

which represents the weight of that particular component against the basis spectrum.

9

The most diagnostically significant PCs are determined with unpaired two-sided

10

Student’s t-test (p < 0.001) and then fed into LDA modeling for tissue classification.

11

The model determines the discriminant function that maximizes the variances in the

12

dataset between different groups while minimizing the variances between members of

13

the same group. The performance of the diagnostic algorithms rendered by the PCA–

14

LDA models for correctly predicting the subtypes of GBMs is estimated in an

15

unbiased manner using the leave one spectrum-out, cross-validation method on all the

16

SRS spectra. In this approach, the spectra from each same tissue are withdrawn from

17

the data set and the PCA–LDA modeling is redeveloped based on the remaining SRS

18

spectra. The redeveloped PCA–LDA diagnostic algorithm is then used to classify the

19

withdrawn spectra. This process is repeated until all the spectra are tested. Receiver

20

operating characteristic (ROC) curves are generated by successively changing the

21

thresholds to determine the correct and incorrect classifications for all tissue

22

examined. Multivariate statistical analysis is performed using the in-house written

23

scripts in the Matlab programming environment.

24

8

ACS Paragon Plus Environment

Page 8 of 33

Page 9 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

3. Results

2

The epi-detected spectral-focusing hyperspectral SRS imaging technique enables

3

rapid acquisition of hyperspectral SRS image stacks (ranging from 2800 to 3020 cm-1

4

at 7 cm-1 intervals) of GBMs without tissue pretreatments. The entire hyperspectral

5

SRS stack can be obtained within 30 s with 2.4 µs of pixel dwell time for 352 x 352

6

pixels (160 x 160 µm). Figures 2(a-h) show the representative epi-detected

7

hyperspectral SRS images of mesenchymal and proneural GBMs at the Raman shifts

8

of 2845, 2885 and 2935 cm-1, respectively. Each image provides distinct biomolecular

9

information on the two subtypes of GBMs. At the Raman shift of 2845 cm−1, lipid

10

distributions in GBMs are visualized due to the symmetric stretching of CH2 bonds of

11

the lipid molecules (Figures 2a and 2e). The hyperspectral SRS image at 2885 cm−1

12

shows a large overlap with the SRS image at 2845 cm−1, as the SRS signals are

13

generated due to the asymmetric stretching of the CH2 bonds in the same lipid

14

molecules (Figures 2b and 2f). The relatively higher intensity at 2885 cm−1 over 2845

15

cm−1 suggests that the lipids tend to be in a solid phase instead of liquid, and thus,

16

they are possibly in the form of spherical micelles26. At the Raman shift of 2935 cm−1,

17

CH3 stretch band of both lipids and proteins are strongly resonated, giving rise to the

18

uniform brightness of the tissues (Figures 2c and 2g). To increase the specific contrast

19

of the proteins in the hyperspectral SRS images, we subtract the SRS image at 2935

20

cm−1 from the SRS image at 2845 cm−1, providing a better visualization of protein

21

distributions in GBMs (Figures 2d and 2h).

22

To assess the cellular morphology of GBM tissues, the SRS at 2845 cm−1 (Figures

23

2a and 2e) and the subtracted images (Figures 2d and 2h) are overlaid together (Figure

24

3: SRS at 2845 cm−1 in green channel and the subtracted image in red channel). In

25

Figure 3a, the mesenchymal GBM is characterised by lipid droplets (white arrow) and

9

ACS Paragon Plus Environment

Analytical Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

amorphous tissue background (orange arrow). On the other hand, the proneural GBM

2

shows greater extent of biological features which include myelins and cells (white and

3

orange arrows in Figure 3b). Figure 3c shows the average SRS spectra of the two

4

subtypes of GBM covering the high wavenumber region from 2800 to 3020 cm-1. The

5

corresponding SRS image stacks are available as movies in the supporting document

6

(Videos S1 and S2). The unique spectroscopic patterns suggest their remarkable

7

differences in terms of biomolecular makeups. The mesenchymal GBM spectrum

8

shows higher intensities at 2845 and 2885 cm-1, possibly due to the dense lipid

9

droplets in tissue; whereas a more intense peak (2935 cm-1) in proneural SRS

10

spectrum could be contributed by the abundance of cellular proteins in proneural

11

GBMs.

12

We further fed the hyperspectral SRS image stack into the MCR algorithm for

13

decomposition of SRS spectra obtained from the hyperspectral SRS images. The

14

entire hyperspectral SRS image stack is divided into 4 to 6 segments (~ square ROI of

15

120 x 120 pixels) from which average SRS spectra are obtained. First, we determine

16

the number of major chemical components in the tissues based on the eigenvalues

17

computed using principal component analysis (PCA). The first two eigenvalues make

18

up of >88% of the total variance and thus, the number of components in our MCR

19

analysis is set as two. Figures 4a and 4b show the two MCR spectra obtained for each

20

subtype. Based on the reference to the Raman spectra of the relevant chemicals27, the

21

first MCR spectra in Figure 4a are most likely to represent cellular proteins. However,

22

there are no observable spectral differences between the two for further classification.

23

In contrast, the second MCR spectra of the mesenchymal and proneural GBM in

24

Figure 4b belong to a family of fatty acids, oleic acid and arachidonic acid,

25

respectively, based on the relevant Raman spectra measured (Figure S2). In addition

10

ACS Paragon Plus Environment

Page 10 of 33

Page 11 of 33 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Analytical Chemistry

1

to the spectral analysis, the concentration maps retrieved by MCR analysis on Figures

2

4c and 4d give a clearer separation between the lipid (green color) and protein (red

3

color) distributions with no cross-talks between the two channels, thereby enabling

4

better visualization of the key cellular features such as myelins, cells and lipid

5

droplets in GBMs. In the concentration maps retrieved by MCR analysis, the pixel

6

intensities are contributed by specific biomolecules. Thus, we can quantify the relative

7

lipid content in the two subtypes of GBM tissues by comparing the pixel intensities of

8

the concentration maps (Figures 4c and 4d) with those of pure oleic acid (100% v/v).

9

Figure 5 shows that the average lipid content in the mesenchymal GBM is ~3-fold

10

higher compared to the proneural GBM (unpaired two-sided Student’s t-test, p