You are on page 1of 4

TIBTEC 1596 No.

of Pages 4

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

Trends in Biotechnology, Month Year, Vol. xx, No. yy 1


TIBTEC 1596 No. of Pages 4

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

2 Trends in Biotechnology, Month Year, Vol. xx, No. yy


TIBTEC 1596 No. of Pages 4

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

Trends in Biotechnology, Month Year, Vol. xx, No. yy 3


TIBTEC 1596 No. of Pages 4

https://doi.org/10.1016/j.tibtech.2017.12.008
single tumor cell out of millions of cells learning, especially deep learning, will
with unprecedented accuracy. Blood accelerate the paradigm shift from low-
References
specimens can then be sampled in serial to high-content analysis in IFC. Further- 1. Ogle, L.F. et al. (2016) Imagestream detection and char-
fashion (Figure 1, right panel), providing a more, cloud computing can overcome acterisation of circulating tumour cells – a liquid biopsy for
hepatocellular carcinoma? J. Hepatol. 65, 305–313
more comprehensive monitoring of a the computational infrastructure hurdles. 2. Blasi, T. et al. (2016) Label-free cell cycle analysis for high-
patient’s cancer than can be obtained These developments are [10_TD$IF]key for practical throughput imaging flow cytometry. Nat. Commun. 7,
10256
through traditional methods, as was dem- IFC applications to reach the clinic, fueling
3. Betters, D.M. (2015) Use of flow cytometry in clinical prac-
onstrated for hepatocellular carcinoma the applicability of IFC as a diagnostic, tice. J. Adv. Pract. Oncol. 6, 435–440
[1]. Liquid biopsies [97_TD$IF]could also be analyzed prognostic, and therapeutic tool. 4. Giesen, C. et al. (2014) Highly multiplexed imaging of tumor
tissues with subcellular resolution by mass cytometry. Nat.
by imaging technologies other than IFC: Methods 11, 417–422
Recently, multiplex protein detection on [102_TD$IF]Acknowledgments 5. Han, Y. et al. (2016) Review: imaging technologies for flow
circulating tumor cells using imaging This work was supported in part by the US National cytometry. Lab Chip 16, 4639–4647

Science Foundation/UK Biotechnology and Biological 6. Barteneva, N.S. and Vorobjev, I.A. (2016) Imaging Flow
mass cytometry has been demonstrated Cytometry: Methods and Protocols, Springer
[13]. Sciences Research Council under a joint grant NSF
7. Grimwade, L.F. et al. (2017) Applications of imaging flow
DBI 1458626 and BB/N005163 (A.E.C. and P.R.). cytometry in the diagnostic assessment of acute leukae-
mia. Methods 112, 39–45
Despite this promise, IFC is currently pri- 1
Imaging Platform at the Broad Institute of Harvard and 8. Zanella, F. et al. (2010) High content screening: seeing is
marily used in research [98_TD$IF]rather than clinical MIT, 415 Main Street, Cambridge, MA 02142, USA believing. Trends Biotechnol. 28, 237–245
2
School of Science and Technology, Nazarbayev 9. Caicedo, J.C. et al. (2017) Data-analysis strategies for
practice. We see the data analysis as the University, Kababnbay batyr avenue 53, 010000, Astana, image-based cell profiling. Nat. Methods 14, 849–863
primary hurdle: it is often prone to varia- Kazakhstan 10. Hennig, H. et al. (2017) An open-source solution for
3
tion, manual tuning, and interpretation. A.N. Belozersky Institute of Physico-Chemical Biology/ advanced imaging flow cytometry data analysis using
Biological Faculty, M.V. Lomonosov Moscow State machine learning. Methods 112, 201–210
[9_TD$IF]These issues might be overcome with University, 119991, Moscow, Russian Federation 11. Angermueller, C. et al. (2016) Deep learning for computa-
machine learning approaches. As well, 4
College of Engineering, Swansea University, Singleton tional biology. Mol. Syst. Biol. 12, 878
there is a need for standardization of Park, Swansea SA2 8PP, UK 12. Friedrich, M.J. (2017) Going with the flow: the promise and
5
Flow Cytometry Core Facility, Faculty of Medical challenge of liquid biopsies. JAMA 318, 1095–1097
IFC, which should include standard oper- Sciences, Newcastle University, Newcastle upon Tyne 13. Gerdtsson, E. et al. (2018) Multiplex protein detection on
ating procedures and standardized qual- NE1 7RU, UK circulating tumor cells from liquid biopsies using imaging
ity control [10_TD$IF]of hardware performance. 6
Department of Systems Biology and Bioinformatics, mass cytometry. Converg. Sci. Phys. Oncol. 4, 015002
University of Rostock, 18051 Rostock, Germany 14. Chen, C.L. et al. (2016) Deep learning in label-free cell
Although a common practice for conven- 7
Stellenbosch Institute for Advanced Study (STIAS), classification. Sci. Rep. 6, 21471
tional flow cytometry, this has not yet Stellenbosch, [85_TD$IF]7600, South Africa
8
been implemented as such in IFC. Program in Cellular and Molecular Medicine, Boston
Children’s Hospital and Department of Pediatrics, Harvard
Medical School, Boston, MA 02115, USA
User-friendly, robust, and standardized
workflows that can facilitate machine *Correspondence: holgerh@broadinstitute.org (H. Hennig).

4 Trends in Biotechnology, Month Year, Vol. xx, No. yy

You might also like