Вы находитесь на странице: 1из 4

Forum can quantify multiple properties of con- Imaging Flow Cytometry

stituents of interest (including proteins, Equipped with 20, 40, or 60 objec-
Diagnostic Potential of nucleic acids, glycolipids) in multiple sub- tives and up to two charge-coupled
Imaging Flow cellular compartments (nucleus, mito- device cameras, IFC allows thousands
chondria, etc.). The rich information of morphological and spatial properties
Cytometry makes IFC ideal for high-content analysis, to be measured for each individual cell.
Minh Doan,1 Ivan Vorobjev,2,3 as well as machine learning, raising the These include bright [92_TD$IF]field, dark field, and
Paul Rees,1,4 Andrew Filby,5 possibility to profile complex cell pheno- up to ten fluorescent channels (Figure 1)
types, identify rare cells and transition [5]. Similar to its flow cytometry-based
Olaf Wolkenhauer,6,7
states, and, importantly, discover useful siblings, IFC is well-suited to image non-
Anne E. Goldfeld,8
targets for disease diagnosis, personal- adherent or dissociated cells, key for
Judy Lieberman,8
ized medicine, and drug development. many clinical applications such as
Natasha Barteneva,8 Here, we discuss significant recent devel- [93_TD$IF]analyses of bodily fluids like blood, whose
Anne E. Carpenter,1 and opments in the IFC field and a perspective structures can be distorted (smeared) by
Holger Hennig1,6,* on where IFC [89_TD$IF]could be adopted as a placement onto a slide.
diagnostic tool in clinical practice.
Imaging flow cytometry (IFC) cap- Advances in Data Analysis
tures multichannel images of hun- Advances in Instrumentation Low-Content Analysis
dreds of thousands of single cells Conventional Flow Cytometry IFC suffers from a ‘content gap’:
within minutes. IFC is seeing a par- Conventional (zero-spatial-resolution) flow although the images are high in informa-
adigm shift from low- to high-infor- cytometry, that is, without imaging, saw its tion content, containing rich morphologi-
mation-content analysis, driven first commercial instruments in the 1970s. cal and spatial information (even in a
partly by deep learning algorithms. Thanks to continuous standardization and single bright-field channel), data analyses
improvement, flow cytometry is now rou- often have low information content, that
We predict a wealth of applications
tinely used as a diagnostic instrument for is, [94_TD$IF]analyses are based on only very few
with potential translation into clini-
health disorders, especially hematologic selected features, which are often man-
cal practice. diseases [3]. Although conventional flow ually identified by applying binary gates
cytometry is considered high throughput on cell populations of interest. These
Imaging Flow Cytometry because it analyzes up to 100 000 cells approaches are highly subjective, require
Imaging flow cytometry (IFC) combines per second, it is considered to be low in significant user interaction, and only uti-
the high-throughput, multiparameter information content because typically only lize a few morphological features instead
capabilities of conventional flow cytome- a single feature (integrated intensity) is of the hundreds that are inherently pres-
try with morphological and spatial infor- measured per fluorescence marker. The ent in the data, for example, only using
mation, all at [87_TD$IF]single-cell resolution. current trend is to increase the number of cell size to denote cell growth when there
Multichannel digital images of hundreds parameters that can be simultaneously are likely tens or hundreds of other fea-
of thousands of individual cells can be measured by developing instruments with tures that could describe and resolve the
captured within minutes (Figure 1), and more lasers and detectors in combination biological differences in a more reliable
include several fluorescence channels as with new fluorochromes that can be used and powerful way. Such analyses make
well as bright field (transmitted light) and in concert with one another. Mass cytom- use of far less information than is present
dark field (scattered light). The throughput etry can measure in excess of 40 markers in the images. Yet, even with these lim-
of IFC means that it is especially well simultaneously using antibodies tagged itations, some IFC applications are
suited to the analysis of rare cell types with rare earth metals [4]. This platform already heading toward the clinic [6],
such as circulating tumor cells (which significantly increases the number of such as the diagnostic assessment of
are cancer cells that escaped from a pri- parameters measured beyond what is acute leukemia [7].
mary tumor and circulate in the blood- currently achievable with conventional
stream) [1] and transition states[8_TD$IF], such flow cytometry and has [90_TD$IF]driven the adop- High-Content Analysis and Deep
as cell cycle phases (mitosis) [2]. tion of machine learning techniques when Learning
analyzing such multidimensional data, IFC data are now beginning to benefit
By extracting information from these digi- however, it is still limited to intensity-based from significant advancements in
tal images (Figure 1, middle panel), IFC features[91_TD$IF]. high-content analysis from the world of

Trends in Biotechnology, July 2018, Vol. 36, No. 7 649


Image acquisiƟon Feature extracƟon High-content analysis
Hundreds of features
per cell per channel
Area & shape
Intensity
20% leukemic 80% normal
Texture cells cells
Imaging CorrelaƟon
flow Granularity
cytometer
Features

Light Detectors
sources High-content

Low content
single-cell
Cells

mulƟdimensional
data Personalized diagnosis
prognosis
therapy
Up to 12 channels per cell

Figure 1. Imaging Flow Cytometry Acquires Images of Single Cells in High Throughput. Typical throughput is up to 5000 cells/s, but modifications to the
instruments can increase this to 100 000 cells/s [14]. The images of each cell are captured by charge-coupled device detectors as the cell flows past the light sources
(left panel) and morphological features are extracted (middle panel). For instance, patient blood could be analyzed to distinguish leukemic from normal cells or monitor in
vitro or in vivo response to therapeutic intervention (right panel). High-content analysis could be used for personalized diagnosis, prognosis, and therapy.

high-throughput microscopy [8]. High- phenotypes, from pluripotent precursors nature of data obtained using IFC is per-
content analysis might unveil disease to nascent immature cells to terminally fectly suited for deep learning for several
states hidden to the eye of even special- differentiated cells, each with distinct reasons. Deep learning requires a large
ists, leading to entirely new diagnostic morphologies and associated functions. number of examples to train the network
capabilities. In image-based profiling, and IFC can quickly produce millions of
once single cells are identified and seg- However, advanced IFC analysis pipe- single-cell images. In addition, deep
mented, thousands of quantitative met- lines often involve multistep workflows learning operates at the pixel level and
rics can be extracted, including shape, challenging to those who are not compu- does not depend on prerequisite prepro-
intensity, texture, and object relation- tational experts. Momentum to enable a cessing or object segmentation, which is
ships. The general strategy is to ‘measure broader group of biomedical researchers often prone to errors. Deep learning has
everything first, then ask questions later’. and clinicians to carry out complex anal- shown success in identifying colon can-
A morphological profile is created, which yses is growing; for example, a user- cer cells [5] and reconstructing cell cycle
is like a fingerprint of each cell. Then the friendly open-source high-content IFC and disease progression [12]. However,
similarities (correlations) between profiles protocol based on machine learning is major challenges for applying deep learn-
can be compared to define cell subpopu- now available [2,10]. ing to IFC in a clinical setting are the
lations or identify disease-specific pheno- development of user-friendly workflows,
types [9]. This profiling strategy might be Deep learning is revolutionizing computer satisfying heavy computational require-
particularly useful when applying IFC for vision across many domains, including ments, evaluation of diagnostic accuracy,
clinical phenotyping, where the cell computational biology [11]. In particular, and approval for clinical use. The Food
changes present in a chronic disease deep convolutional neural networks have and Drug Administration (FDA) approved
often spread throughout a spectrum of proven to be very powerful (Box 1). The the first clinical deep learning application

650 Trends in Biotechnology, July 2018, Vol. 36, No. 7


Box 1. Image Processing and Deep Learning
Bioimaging research has recently made great strides thanks to deep learning, a subclass of machine learning. A deep learning network consists of an input layer, a
number of hidden layers, and an output layer. The larger the number of hidden layers, the deeper the network. Among several deep learning methods, convolutional
neural networks have shown [78_TD$IF]the most impressive results in object detection [11].

Convolutional neural networks operate on the raw pixels of each image where the network learns to enhance complex abstractions in images, such as eyes, faces, or in
the case of IFC, a cellular phenotype or signature of disease, while suppressing irrelevant information in the images (see [79_TD$IF]Figure I). By rastering a window over the input
image, the feature maps are created (by a mathematical operation called ‘convolution’). The feature maps represent how the input image looks when seen through the
convolutional filter. Pooling refers to downsampling the feature maps to reduce the number of pixels while keeping the relevant information [11]. The output layer is[80_TD$IF], the
classification probability which assigns a class to each input image with a certain probability, for example, Class 1 and 2 could correspond to leukemic and normal cells,
respectively.

In contrast to deep learning, in conventional image analysis, the images are first preprocessed, then cellular objects are identified (segmented) by analysis software,
followed by the extraction of hundreds of human-engineered features per channel and object ([81_TD$IF]middle panel in Figure 1), including shape, intensity, and texture [8,9].
While classical machine learning techniques rely on human-engineered features, deep learning filters can have a much higher level of abstraction and complexity.

Recent open-source frameworks, such as TensorFlow, Keras, [82_TD$IF]PyTorch and CAFFE, provide convenient and effective means to adopt powerful deep learning
architectures into bioimaging research. With a wide range of practical applications emerging in various industries, we expect that such frameworks will become
increasingly stable and robust, and thereby [83_TD$IF]suitable for clinical applications.

Input layer ConvoluƟonal layers


y Output layer
×N
Raw image
Class 1
Class 2

Class n

Learned Feature
conv. filters maps Pooling ClassificaƟon

Figure I. Illustration of a Convolutional Neural Network.

for health care in 2017 (www.forbes.com/ of samples, which are often fragile in resistant neoplastic cells, clotting abnor-
sites/bernardmarr/2017/01/20/first- hematological diseases. Recently, IFC malities of platelet microparticles, or fetal
fda-approval-for-clinical-cloud- was shown to deliver integrated leukemia abnormalities. A major advantage of the
based-deep-learning-in-healthcare). diagnostics in one test [7]. liquid biopsy is that it can be carried out in
a simple, noninvasive way. One reason it
Opportunities Ahead Another potential clinical use of IFC is to took the liquid biopsy so long to develop
Many potential clinical [95_TD$IF]uses of IFC are analyze bodily fluids for rare cells, for is that circulating tumor cells are found in
conceivable, for instance a differential example, the typically small number of low concentrations in the bloodstream
diagnosis of acute and chronic lympho- leukemic cells that remain in the patient [12]. IFC in combination with machine
cytic/myeloid leukemia, possibly using during treatment, known as [96_TD$IF]minimal learning has the potential to identify a
fewer biomarkers, or ideally unstained residual disease (Figure 1, right panel) single tumor cell out of millions of cells
and unmanipulated blood samples. [6]. There is substantial interest in liquid with unprecedented accuracy. Blood
Reducing the number of required, biopsy, the analysis of circulating tumor specimens can then be sampled in serial
descriptive biomarkers would greatly sim- cells, which are extremely rare [12]. Liquid fashion (Figure 1, right panel), providing a
plify the laboratory sample preparation biopsy might detect cancer at an early more comprehensive monitoring of a
and help preserve the intact ‘nativeness’ stage, circulating metastatic or drug- patient’s cancer than can be obtained

Trends in Biotechnology, July 2018, Vol. 36, No. 7 651


*Correspondence: holgerh@broadinstitute.org (H. Hennig).
through traditional methods, as was dem- Furthermore, cloud computing can over-
https://doi.org/10.1016/j.tibtech.2017.12.008
onstrated for hepatocellular carcinoma come the computational infrastructure
[1]. Liquid biopsies [97_TD$IF]could also be analyzed hurdles. These developments are [10_TD$IF]key References
by imaging technologies other than IFC: for practical IFC applications to reach 1. Ogle, L.F. et al. (2016) Imagestream detection and char-
acterisation of circulating tumour cells – a liquid biopsy for
Recently, multiplex protein detection on the clinic, fueling the applicability of IFC hepatocellular carcinoma? J. Hepatol. 65, 305–313
circulating tumor cells using imaging as a diagnostic, prognostic, and thera- 2. Blasi, T. et al. (2016) Label-free cell cycle analysis for high-
mass cytometry has been demonstrated peutic tool. throughput imaging flow cytometry. Nat. Commun. 7,
10256
[13]. 3. Betters, D.M. (2015) Use of flow cytometry in clinical prac-
[102_TD$IF]Acknowledgments tice. J. Adv. Pract. Oncol. 6, 435–440
Despite this promise, IFC is currently pri- This work was supported in part by the US National 4. Giesen, C. et al. (2014) Highly multiplexed imaging of tumor
tissues with subcellular resolution by mass cytometry. Nat.
marily used in research [98_TD$IF]rather than clinical Science Foundation/UK Biotechnology and Biological
Methods 11, 417–422
practice. We see the data analysis as the Sciences Research Council under a joint grant NSF
5. Han, Y. et al. (2016) Review: imaging technologies for flow
DBI 1458626 and BB/N005163 (A.E.C. and P.R.). cytometry. Lab Chip 16, 4639–4647
primary hurdle: it is often prone to varia-
6. Barteneva, N.S. and Vorobjev, I.A. (2016) Imaging Flow
tion, manual tuning, and interpretation. 1
Imaging Platform at the Broad Institute of Harvard and Cytometry: Methods and Protocols, Springer
[9_TD$IF]These issues might be overcome with MIT, 415 Main Street, Cambridge, MA 02142, USA 7. Grimwade, L.F. et al. (2017) Applications of imaging flow
2
School of Science and Technology, Nazarbayev cytometry in the diagnostic assessment of acute leukae-
machine learning approaches. As well, University, Kababnbay batyr avenue 53, 010000, Astana, mia. Methods 112, 39–45
there is a need for standardization of Kazakhstan 8. Zanella, F. et al. (2010) High content screening: seeing is
3
IFC, which should include standard oper- A.N. Belozersky Institute of Physico-Chemical Biology/ believing. Trends Biotechnol. 28, 237–245
Biological Faculty, M.V. Lomonosov Moscow State 9. Caicedo, J.C. et al. (2017) Data-analysis strategies for
ating procedures and standardized qual- University, 119991, Moscow, Russian Federation image-based cell profiling. Nat. Methods 14, 849–863
ity control [10_TD$IF]of hardware performance. 4
College of Engineering, Swansea University, Singleton 10. Hennig, H. et al. (2017) An open-source solution for
Although a common practice for conven- Park, Swansea SA2 8PP, UK advanced imaging flow cytometry data analysis using
5 machine learning. Methods 112, 201–210
Flow Cytometry Core Facility, Faculty of Medical
tional flow cytometry, this has not yet Sciences, Newcastle University, Newcastle upon Tyne 11. Angermueller, C. et al. (2016) Deep learning for computa-
been implemented as such in IFC. NE1 7RU, UK tional biology. Mol. Syst. Biol. 12, 878
6
Department of Systems Biology and Bioinformatics, 12. Friedrich, M.J. (2017) Going with the flow: the promise and
University of Rostock, 18051 Rostock, Germany challenge of liquid biopsies. JAMA 318, 1095–1097
User-friendly, robust, and standardized 7
Stellenbosch Institute for Advanced Study (STIAS), 13. Gerdtsson, E. et al. (2018) Multiplex protein detection on
workflows that can facilitate machine Stellenbosch, [85_TD$IF]7600, South Africa circulating tumor cells from liquid biopsies using imaging
8 mass cytometry. Converg. Sci. Phys. Oncol. 4, 015002
learning, especially deep learning, will Program in Cellular and Molecular Medicine, Boston
Children’s Hospital and Department of Pediatrics, Harvard 14. Chen, C.L. et al. (2016) Deep learning in label-free cell
accelerate the paradigm shift from low- Medical School, Boston, MA 02115, USA classification. Sci. Rep. 6, 21471
to high-content analysis in IFC.

652 Trends in Biotechnology, July 2018, Vol. 36, No. 7

Вам также может понравиться