159
A Miniaturized Microfluidic Cytometer Platform for Point-of- Care Blood Testing Applications by James Jiahua Dou A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Electrical and Computer Engineering © Copyright by James Dou 2017

A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

A Miniaturized Microfluidic Cytometer Platform for Point-of-Care Blood Testing Applications

by

James Jiahua Dou

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Department of Electrical and Computer Engineering

© Copyright by James Dou 2017

Page 2: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

ii

A Miniaturized Microfluidic Cytometer Platform for Point-of-Care

Blood Testing Applications

James Jiahua Dou

Doctor of Philosophy

Department of Electrical and Computer Engineering

University of Toronto

2017

Abstract

In recent years, microfluidic lab-on-a-chip devices have made tremendous progress. Many new

features, new processes and fabrication methodologies have been proposed and realized in the

last two decades. In the field of clinical diagnostics, however, there still lack devices and

products that truly utilize microfluidic lab-on-a-chip technologies. My thesis is focused on

developing a novel blood testing platform that can be commercialized as a point-of-care

diagnostic tool using microfluidics and optical detection technologies. Access to laboratory

quality blood testing has been difficult and sometimes prohibitive for many populations around

the world, which lead to increased mortality, morbidity and healthcare cost. In this work, I

research and developed a particle imaging and detection system using microfluidics to control

sample motion. The system used fluorescence based detection methodology. A novel image

analysis algorithm is proposed and implemented to detect and track particles captured by the

optical detector. My thesis investigates the fluidic design, on-chip fluidic control, optical

detection system and particle/cell counting algorithm. The overall system is designed to be

capable of commercialized as a point-of-care portable blood analysis system deployed in remote

health settings. In addition, CD4 T cell counting, which is the current gold standard in

HIV/AIDS disease monitoring, is used as an example assay developed on the platform. A

Page 3: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

iii

microfluidic chip, that can complete the cell counting analysis within 15 minutes directly from a

single finger prick blood sample is designed, fabricated and characterized. This work also studies

the sample preparation steps required for the cell counting assay and the impact of various

system parameters. The system described in this thesis highlights a portable blood testing

platform that can be further developed and commercialized as a clinical tool for cell, protein and

nucleic acid based laboratory tests conducted at point-of-care. The outcome of this work

demonstrates the feasibility and utility of such system or product in medical diagnostics in

remote health settings.

Page 4: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

iv

Acknowledgments

I would like to express my gratitude to my supervisor, Prof. Stewart Aitchison, whose expertise,

understanding, and patience, added considerably to my graduate experience. I appreciate his vast

knowledge and skill in many areas (e.g., vision, aging, ethics, interaction with participants), and

his assistance in writing reports (i.e., grant proposals, scholarship applications and this thesis).

His passion, energy, relentless curiosity, and clear vision have defined research to me. This thesis

would not have been possible without the mentorship and tremendous support of Prof. Aitchison.

I would like to thank the other members of my committee, Dr. Axel Gunther, and Dr. Yu Sun for

the assistance they provided at all levels of the research project.

I have been blessed to work with friendly colleagues. Dr. Lu Chen and Rakesh Nayyar were a

pleasure to work with in the lab. I am grateful to Dr. Aju Jugessur for great discussion and lab

work on silicon photonics and nanofabrication. I would also like to thank Dr. Francis Mandy,

without whose motivation and encouragement I would not have considered developing novel

point-of-care technologies for global health. Appreciation also goes out to Jason Grenier, Luis

Fernandez, Stephen Ho, Dr. Jianzhao Li of the Department of Electrical and Computer

Engineering for all the wonderful support and fruitful discussions.

I would also like to thank my family for the support they provided me through my entire life and

in particular, I owe my parents Jinglie and Xinxuan, among a multitude of other things, for

giving me the curiosity to explore the exciting research of combining medicine with technology.

I must acknowledge my wife and best friend, Jenny, without whose love, encouragement and

patience, I would not have finished this thesis.

Finally, I recognize that this research would not have been possible without the financial

assistance of NSERC, the University of Toronto Graduate Studies, the Department of Electrical

and Computer Engineering at the University of Toronto (Teaching Assistantships, Graduate

Research Scholarships) and the Ministry of Research and Innovation, Government of Ontario,

and express my gratitude to those agencies.

Page 5: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

v

Table of Contents

Acknowledgments .......................................................................................................................... iv

Table of Contents ............................................................................................................................ v

List of Tables ............................................................................................................................... viii

List of Figures ................................................................................................................................ ix

Chapter 1 Introduction ............................................................................................................ 1

1.1 Introduction ......................................................................................................................... 1

1.2 Flow Cytometry .................................................................................................................. 2

1.2.1 Working Principle ................................................................................................... 3

1.2.2 Data Analysis .......................................................................................................... 7

1.2.3 Labels ...................................................................................................................... 8

1.2.4 State of the Art ........................................................................................................ 9

1.2.5 Modern Cytometry Applications ............................................................................ 9

1.3 Aims of this Work ............................................................................................................. 10

1.4 Organization of Thesis ...................................................................................................... 12

Chapter 2 Background .......................................................................................................... 13

2.1 Microfluidic Lab-on-a-Chip .............................................................................................. 13

2.1.1 History of Microfluidics ....................................................................................... 13

2.1.2 Recent Advances ................................................................................................... 14

2.1.3 Microfabrication Technologies ............................................................................. 16

2.2 Point-of-Care Testing ........................................................................................................ 21

2.2.1 Microfluidics and POCT ....................................................................................... 23

2.2.2 Lab on a chip and Global Health .......................................................................... 25

2.2.3 Motivation of this Thesis ...................................................................................... 29

Page 6: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

vi

Chapter 3 Passive Microfluidic Systems .............................................................................. 33

3.1 Introduction ....................................................................................................................... 33

3.2 Flow Control Methodology in Microfabricated Flow Cytometers ................................... 34

3.2.1 Acoustic ................................................................................................................ 34

3.2.2 Electrical ............................................................................................................... 35

3.2.3 Micro Structure ..................................................................................................... 36

3.2.4 Micro Flow Cytometers without Focusing ........................................................... 36

3.3 Microfluidics Design Concept .......................................................................................... 37

3.4 Capillary Microfluidic Systems ........................................................................................ 37

3.4.1 Governing equations of Fluid Mechanics ............................................................. 38

3.4.2 Numerical Modeling ............................................................................................. 40

3.4.3 Fluidic Resistance Calculation .............................................................................. 46

3.4.4 Microchannel Design ............................................................................................ 48

3.4.5 Capillary Microfluidic Device Fabrication ........................................................... 49

3.4.6 Capillary Microfluidic Device Characterization ................................................... 50

3.5 Conclusion ........................................................................................................................ 54

Chapter 4 Active Microfluidic System ................................................................................ 55

4.1 Introduction ....................................................................................................................... 55

4.2 Bellows Actuation System ................................................................................................ 56

4.3 Bellows Transport System Design .................................................................................... 57

4.3.1 Bellows Actuation Volume Calculation ............................................................... 58

4.4 Bellows Actuation System and Microfluidics Design ...................................................... 61

4.4.1 Material Characterization ...................................................................................... 62

4.4.2 Microfluidic Channel Design ................................................................................ 66

4.4.3 Bellows Slide Fabrication ..................................................................................... 66

4.4.4 Characterization .................................................................................................... 68

Page 7: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

vii

4.5 On-Chip Sample Preparation ............................................................................................ 78

4.5.1 Reagents ................................................................................................................ 79

4.5.2 Reagent Handling and Incorporation .................................................................... 79

4.5.3 Reagent Drying ..................................................................................................... 81

4.5.4 Re-suspension ....................................................................................................... 82

4.5.5 Microfluidic Mixer ................................................................................................ 83

4.6 Conclusion ........................................................................................................................ 93

Chapter 5 Optics and Detection .......................................................................................... 95

5.1 Introduction ....................................................................................................................... 95

5.2 Particle Detection using Optics ......................................................................................... 95

5.2.1 Optical Detection Methodology Literature Review .............................................. 95

5.2.2 Dynamic Particle Detection and Counting ........................................................... 97

5.2.3 Cell Detection and Enumeration using Capillary Microfluidic Devices ............ 105

5.3 Beadarray Multiplexed Detection ................................................................................... 117

5.3.1 Underlying Strategy ............................................................................................ 118

5.3.2 Proof of Concept Demonstration ........................................................................ 121

5.4 Conclusion ...................................................................................................................... 128

Chapter 6 Conclusion and Future Work ......................................................................... 129

6.1 Thesis Work Summary ................................................................................................... 129

6.2 Impact ............................................................................................................................. 131

6.3 Future Outlook ................................................................................................................ 132

References ................................................................................................................................... 133

Page 8: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

viii

List of Tables

Table 1 - Comparison between polymer and glass as substrate of microfluidic systems for bio

and chemical applications. ............................................................................................................ 20

Table 2 - Prevalent diseases in developing countries. DALY is a measure of the disease burden

on healthcare system. Type of test is the conventional testing methodology.102 .......................... 28

Table 3 - Materials used in autofluorescence characterization. .................................................... 65

Table 5 - Performance of passive micromixers in recent development141. ................................... 83

Table 6 - Performance summary of active micromixers published in literature141. ...................... 85

Table 7 - Mixing time characterization result. Relative mean fluorescence intensity was

calculated as the ratio between liquid control mean fluorescence intensity and measured mean

fluorescence intensity for each scenario. This characterization was completed using design 2 of

the resuspension prototype slide shown in Figure 4-21. ............................................................... 92

Table 8 - Mixing time characterization result. This characterization was completed using design

4 of the resuspension prototype slide shown in Figure 4-21. With the optimized reagent chamber

dimension, the mixing time can be further reduced. ..................................................................... 93

Table 9 - Mixing time characterization result. This characterization was completed using design

4 of the resuspension prototype slide shown in Figure 4-21 and surface treatment of the reagent

plug. .............................................................................................................................................. 93

Table 10 - Camera comparison summary. .................................................................................. 100

Table 11 – Comparison of results using wide field dynamic counting system and flow cytometer.

..................................................................................................................................................... 113

Table 12 - Concentration levels of IL-6 cytokine tested as proof of concept demonstration on

prototype. .................................................................................................................................... 125

Page 9: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

ix

List of Figures

Figure 1-1 - Schematic of a flow cell in a conventional flow cytometer.1 ..................................... 4

Figure 1-2 - Optical setup in a typical modern flow cytometer.1 .................................................... 6

Figure 1-3 – A sample single parameter histogram of particles labeled with FITC dye. FITC

(fluorescein isothiocyanate) is a synthetic organic compound that is widely used as a fluorescent

dye in many applications. Its absorption peak is at 494 nm and emission maximum is at 512 nm.1

......................................................................................................................................................... 7

Figure 1-4 – Sample of a two-parameter (dual color fluorescence) histogram. Particles in this

examples are labeled with PE and FITC dyes. PE (Phycoerythrin) is another organic

fluorochrome whose absorption peak is at 488 nm and 532 nm, and emission maximum is at 585

nm. 1 ................................................................................................................................................ 8

Figure 3-1 - Schematic of liquid plug in a rectangular microfluidic channel. 117 ......................... 39

Figure 3-2 – Schematic illustration of two reservoirs and a straight microchannel connecting the

two reservoirs. ............................................................................................................................... 40

Figure 3-3 – Schematic of microfluidic setup in the numerical model. ........................................ 42

Figure 3-4 – Snapshots of flow in the microchannel at different times. The contact angle for this

simulation is set at 70 degrees. The length of the channel shown in this figure is 1 mm. ............ 43

Figure 3-5 – Axial velocity of liquid in the microchannel with respect to time. .......................... 44

Figure 3-6 – Axial velocity of liquid in the microchannel with respect to time. The cell size is

1.5µm (10 cells per height of channel). The contact angle is 30 degrees. All other conditions are

the same as those in Figure 3-5. .................................................................................................... 45

Figure 3-7 – An example of a capillary microfluidic device. (a) Top view of a capillary

microfluidic device with a circular sample inlet, serpentine microchannel and a tapering structure

to enhance capillary force. (b) Side view of the rectangular microfluidic channel. ..................... 47

Page 10: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

x

Figure 3-8 - Graphical illustration of photolithography process used by Epigem to produce

capillary microfluidic devices. ...................................................................................................... 50

Figure 3-9 – Serpentine microfluidic structure for fluidic flow characterization. The channels

were designed with the same cross-sectional dimensions as the detection channels in the

cell/particle counting device. Channel length of each pass is 20 mm. .......................................... 51

Figure 3-10 – Experimental results on filling characterization of the microfluidic device

corresponding to the serpentine microfluidic structure shown in Fig. 4. (a) Observed filling time

of each linear section of the serpentine microfluidic device. (b) Fluid flow speed and channel

filling distance characterization results based on the filling time and the distance for each pass in

the serpentine microfluidic device. An inversely proportional relationship between flow speed

and the channel length can be seen. .............................................................................................. 52

Figure 3-11 - Capillary microfluidic chip design layout. This design relies on capillary forces to

manipulate sample flow. The device has a volumetric design to allow a sample volume of 2 µL to

be processed. ................................................................................................................................. 53

Figure 4-1 – Bellows transport concept. The soft elastomer is depressed under external force F.

The deflection of the elastomer induces a pressure change inside the chamber and actuates the

fluidic motion inside the microchannel that is connected to the chamber. ................................... 56

Figure 4-2 - Bellows slide concept. The entire device consists of a soft elastomer semi-sphere,

bonded to a plastic fluidic chip. The fluidic channels are connected to the bellows. When bellows

is depressed, the reduction in volume inside the bellows increases the pressure inside the

microchannel which subsequently pushes the liquid sample forward. ......................................... 57

Figure 4-3 – A graphical illustration of the coordinate system and variables used in bellows

volume change calculation. ........................................................................................................... 58

Figure 4-4 – Volume change induced by bellows deflection. Bellows deflection is actuated from

the top of the semi-sphere in this diagram and the amount of deflection is denoted by d. ........... 59

Figure 4-5 - Volume change of the bellows as a function of bellows deflection. ........................ 60

Figure 4-6 - Linear stepper motor from Haydon Kerk. (www.haydonkerkexpress.com) ............ 61

Page 11: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

xi

Figure 4-7 - Microfluidic chip layout of the bellows slide. The bellows slide was fabricated using

injection molding and it was used to test the fluidic actuation and on-chip flow control. ........... 62

Figure 4-8 - Autofluoscence levels of different PMMA and COC material under different

excitation and emission optical setup. .......................................................................................... 64

Figure 4-9 - A photograph of the injection molded microfluidic bellows chip using the PMMA

material selected in previous section. The bellows chip is 1 inch wide and 3 inches long. ......... 67

Figure 4-10 - Schematics of the stepper motor setup used to test and characterize on-chip fluidic

actuation ........................................................................................................................................ 69

Figure 4-11 - Fluidic control eletronics setup block diagram. This setup was used to

experimentally characterize and test the fluidic actuation using bellows concept. ...................... 70

Figure 4-12 - LabView program interface. This tool was used to control the motion of the stepper

motor for on-chip fluidic actuation. .............................................................................................. 71

Figure 4-13 - A picture showing the mechanical setup of the bellows actuation. ........................ 73

Figure 4-14 - Schematics of microfluidic connections used in on-chip fluidic actuation testing

and characterization. ..................................................................................................................... 74

Figure 4-15 - Pictures of the bellows actuation experimental setup. (a) Side view of the motion

control board, stepper motor, bellows slide and re-suspension slide. (b) Top view of the

experimental setup. The engineered blood sample was introduced into the re-suspension slide

and actuated back and forth inside the device. ............................................................................. 75

Figure 4-16 - Fluidic linear flow speed measured as a function of time. Graph (a) is obtained

with a bead sample only while (b-d) were obtained with a sample of beads mixed with

Immunotrols. Three different pumping conditions were tested in this experiment: the stepper

motor pumping distance (2 µm, 10 µm, and 30 µm at a speed of 50). .......................................... 76

Figure 4-17 – Fluidic linear speed plotted as a function of time in the detection microchannel

using bellows actuation. The measurement was made on the resuspension slide described earlier

in this work. .................................................................................................................................. 77

Page 12: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

xii

Figure 4-19 – 3D drawing of reagent plug used to handle and incorporate reagents into the

microfluidic cartridge. ................................................................................................................... 81

Figure 4-20 - Reagent plug coated with dried fluorescently labelled CD4 antibodies. The pink

color indicates the high concentration of antibodies. .................................................................... 82

Figure 4-21 - Microfluidic mixer design. The “wiggly” channels are mixing structures which

utilizes Dean’s flow to enhance mixing efficiency. ...................................................................... 86

Figure 4-22 - Dean flow. In curved channels, when inertia is important, faster moving fluid near

the channel center tends to continue outward, and to conserve mass, more stagnant fluid near the

walls re-circulates inward. This creates two counter-rotating vortices perpendicular to the

primary flow direction164 .............................................................................................................. 87

Figure 4-23 - Resuspension slide design layout. In this design, four different mixer were

proposed to optimize the mixing and fluidic motion in the microchannel. The circular holes are

reagent plug chambers where plugs are inserted. ......................................................................... 90

Figure 4-24 - Fully assembled microfluidic re-suspension prototype slides with reagent plugs

inserted. The re-suspension prototype slides were used to charaterize on-chip mixing and re-

suspension of dried reagents. ........................................................................................................ 91

Figure 5-1 – Optical imaging system of the cell/particle detection and analysis platform. ........ 103

Figure 5-2 – Illustration of the multiplexed detection system described in this paper. The optical

detector continually takes images as particles/cells move into the detection window.

Particles/cells labelled with different fluorophores can only be detected in the corresponding sub-

regions within the detection window depending on the filter setup. (a) a graphical illustration of

the underline principle of the technique; (b) transmission spectrum of the left sub-region of the

detection window; (c) transmission spectrum of the right sub-region of the detection window. 105

Figure 5-3 – CD3 antibody concentration titration curve for signal to background ratio. ......... 108

Figure 5-4 – CD4 antibody concentration titration curve for signal to background ratio. ......... 108

Page 13: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

xiii

Figure 5-5 – Positive events intensity histogram for PE channel at 50ng CD4 antibody per 100µL

blood. .......................................................................................................................................... 109

Figure 5-7 – Linearity test result. Cell concentrations range from 150 to 720 per μL were tested

for the comparison with flow cytometer. (Each data point is an average value for 3 measurements

with standard deviation bar.) ....................................................................................................... 114

Figure 5-8 – Two color fluorescence image captured through a custom setup with two half-moon

shaped optical filters placed together side by side. The CD4 cells labelled with PE

(phycoerythrin) were shown in the left panel while the CD3 cells labelled with PE-Cy5.5 were

shown in the right panel. ............................................................................................................. 116

Figure 5-9 – Combined images is showing four detected MESF beads. .................................... 117

Figure 5-10 – Multiplexed beadarray detection process illustration.192 ..................................... 119

Figure 5-11 – Bead complexes and reagents explanation. .......................................................... 120

Figure 5-12 – Two color multiplexed beadarray detection concept illustration. The optical

imaging area is divided into two sections: left is the identification channel and the right is the

quantification channel. ................................................................................................................ 121

Figure 5-13 - Dilution steps performed to obtain different IL-6 concentration levels to be

measured. .................................................................................................................................... 124

Figure 5-14 – Histogram of multiplexed beads detected by the optical detection system described

and developed in this chapter. The beads fluorescence intensities are evenly distributed on a log

scale. ............................................................................................................................................ 126

Figure 5-15 - Correlation between fluorescence intensity of reporter or detection antibody and

target analyte concentration. In this experiment, bead type number 4 in the BD kit was used. The

target analyte was IL-6 cytokine as described in Section 5.3.2 on Page 79. .............................. 127

Page 14: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

1

Chapter 1 Introduction

1.1 Introduction

The enumeration of cells, bacteria and viruses in human body fluids is of primary importance in

medicine and immunology. The human immune system is a network of white blood cells,

proteins, tissues and organs. It is a critical component of human body that prevents infection and

the growth of bacteria, viruses and parasites. It also prevents the unwanted growth of cancerous

cells. When compromised, the immune system does not perform its functions and infections

occur. On the other hand, the immune system is overactive and attacks the normal cells of the

body, leading to autoimmune disorder. Maintaining the proper functions of human immune

system is critical in keeping people healthy and preventing infections.

Examples of the clinical importance of the immune system include: counting of CD4 T-cells in

HIV positive subjects and of granulocytes/platelets in patients receiving chemotherapy1.

Currently, flow cytometry is the tool of choice for rapid blood cell analysis2. In this technique,

cells are suspended in a fluid stream and passed through the detection region where there are

illuminated by a laser. Light scattered in different directions can be used to distinguish

differences in size and internal complexity of the cells, whereas light emitted from fluorescently

labeled antibodies can identify a wide array of cell surface and cytoplasmic antigens. This

technique is widely used in cell counting, sorting, biomarker detection and protein engineering.

Moreover, multi-parametric analysis of the physical and/or chemical characteristics of up to

thousands of particles per second can be completed.

The objective of this work is to research and develop a miniaturized, compact imaging cytometer

system that can bring facility bound diagnostic work to the field. More importantly, it can help

doctors and physicians to develop personalized therapy for patients based on customized

biomarker testing for each individual. The model application is a CD4 cell counting assay for

HIV disease monitoring.

Page 15: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

2

Figure 1-1 - Concept drawing of the portable blood testing workflow from a droplet of sample from a finger

prick.

Conceptually, the portable analysis instrument will consist of a disposable cartridge and a

handheld/portable electronic device as illustrated in Figure 1-1. The plastic cartridge contains

slow-dried reagents coated on the surface of a disposable microfluidic device during fabrication

and packaging. Disposability reduces the cost of cleaning and eliminates cross contamination

between tests. This point-of-care analysis system offers rapid, effective, accurate and low-cost

HIV monitoring that is suitable for remote areas and resource poor settings. In addition, the

proposed handheld cytometer will have built-in software and hardware quality control

mechanisms to ensure the proper operation of the system, which is vital for such portable

instruments deployed in the field that lacks well established infrastructure.

This chapter lays the background information and motivation of this thesis. It explains the

working principles of a flow cytometer and overviews the common applications of flow

cytometry in clinical setting. The second half of this chapter reviews current efforts to develop

portable flow cytometry solutions that can be deployed at the point of care.

1.2 Flow Cytometry

Flow cytometry is the method of choice for rapid analysis of multiple characteristics of single

cells. The information obtained is both qualitative and quantitative. Whereas in the past flow

cytometers were found only in larger academic centers, advances in technology now make it

possible for community hospitals to use this methodology. Contemporary flow cytometers are

Page 16: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

3

much smaller, less expensive, more user-friendly, and well suited for high-volume operation.

Flow cytometry is used for immunophenotyping of a variety of specimens, including whole

blood, bone marrow, serous cavity fluids, cerebrospinal fluid, urine, and solid tissues.

Characteristics that can be measured include cell size, cytoplasmic complexity, DNA or RNA

content, and a wide range of membrane-bound and intracellular proteins1.

1.2.1 Working Principle

The working principles of a flow cytometer are shown schematically in Figure 1-2. The flow

cytometer measures the optical and fluorescence characteristics of single cells (or any other

particle, including nuclei, microorganisms, chromosome preparations, and latex beads). Physical

properties, such as size (represented by forward angle light scatter) and internal complexity

(represented by right-angle or side scatter) can be used to differentiate certain cell populations. A

beam of light of a single wavelength is directed onto a hydro dynamically focused stream of

liquid. A number of detectors are aimed at the detection point to image the subject. By using the

principles of light scattering, and the emission of light from fluorochrome molecules conjugated

to the subject, it is possible to generate specific multi-parameter data from microscopic particles

in the size range of 0.5 µm to 40 µm in diameter. Hydro-dynamic focusing of particles is

accomplished through the introduction of sheath flow surrounding the sample stream; ensuring

only one particle is interrogated at a time.

Page 17: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

4

Figure 1-2 - Schematic of a flow cell in a conventional flow cytometer.1

Figure 1-2 also illustrates the principle of hydrodynamic focusing in a flow cytometer. The

sample is injected into the center of a sheath flow. The combined flow is reduced in diameter,

forcing the cells or particles into the center of the fluid stream. This allows the laser beam to

interrogate cells/particles one at a time. As the sheath fluid moves, it creates a drag force on the

narrowing central chamber, which alters the velocity of the central fluid stream whose flow front

becomes parabolic with greatest velocity at its center while effectively having zero velocity at

the wall. The net effect produces a single file of particles, known as hydrodynamic focusing.

Under optimal conditions, the fluid in the center will not mix with the surrounding sheath fluid.

The flow characteristic of the central column can be modeled using the Reynolds number (Re):

𝑅𝑒 =𝜌𝑉𝐷

𝜇 (1 – 1)

Where D is the tube diameter, v is the mean fluid speed, 𝜌 is the density of the liquid and 𝜇 being

the viscosity of the fluid. Laminar flow usually occurs when either the fluid is moving slowly or

the fluid is viscous, under which conditions Reynolds number (Re) is low. As Reynolds number

increases, the flow will transition from laminar to turbulent flow at a specific range of Reynolds

Page 18: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

5

numbers. The laminar-turbulent transition range depends on the disturbance levels in the fluidic

system. Typically, laminar flow occurs when the Reynolds number is below a threshold value of

approximately 2000, while the transition range is usually between 1,800 and 2,100.3

After forming single file, particles or cells, are interrogated by a beam of light. Scattering and

fluorescence emission provides information about particle/cell properties. Light that is scattered

in the forward direction, up to 20◦ offset from the laser beam axis, is known as the forward

scatter channel signal (FSC). The FSC signal depends on the particle/cell size and can be used to

distinguish between debris and living cells. On the other hand, light measured at 90◦ angle to the

excitation is termed side scatter signal (SSC). The SSC collects information about the granular

content within a particle/cell. Combing FSC and SSC, a flow cytometer is able to differentiate

different cell types in a heterogeneous population.

Another dimension to the flow cytometer is the ability of carry out fluorescence measurements,

which can provide quantitative and qualitative data about fluorochrome-labeled cell surface

receptors, or intracellular molecules such as DNA and cytokines. Typically, in flow cytometry

different fluorescence channels are used to detect emission of different wavelength.

As cells or particles pass through the detection volume, fluorochromes are excited to a higher

energy state. When electrons of the fluorochrome relax to the ground state, photons of light are

emitted with specific spectral properties unique to different fluorochromes. One unique aspect of

flow cytometry is that it can measure fluorescence per cell or particle. This contrasts with

spectrophotometry, in which the percent absorption and transmission of specific wavelengths of

light is measured for a bulk sample.

Typically, the scattered optical signal and the emitted fluorescence light from cells and particles

are converted to electrical pulses by optical detectors such as silicon photodiodes or

photomultiplier (PMT) devices. Because of its higher sensitivity and lower noise, PMTs are

widely used in conventional flow cytometry.

Figure 1-3 is a graphical illustration of the optical components in a flow cytometer. Collimated

laser beam is focused at the interrogation region. Emitted light is directed to different optical

detectors with filters and mirrors.

Page 19: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

6

Figure 1-3 - Optical setup in a typical modern flow cytometer.1

The electrical signals are then processed by a series of amplifiers, with both linear and log

amplification. Logarithmic amplification is commonly used in fluorescence imaging of

biological samples, since this technique expands the scale of weak signals and compresses the

scale for strong signals, resulting in a distribution that is easy to display on a histogram.

The measurement from each detector shown in Figure 1-3 is referred to as a “parameter”. The

data acquired in each parameter represent the number of occurrence of such parameter, which

usually means the number of particles or cells displaying the physical feature or surface marker

of interest. Finally, the amplified signals are processed using a standard analog-to-digital

converter (ADC) which in turn allows further data processing and analysis, and the generation of

histograms and scatter plots.

Page 20: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

7

1.2.2 Data Analysis

An important principle of flow cytometry data analysis is to selectively visualize the

particles/cells of interest while eliminating results from unwanted subjects such as debris and

dead cells. This process is called gating.

Gating can be done based on the physical characteristics of particles/cells, surface markers or

intracellular contents. In addition to FSC /SSC gating, newer strategies utilizes fluorescence

parameters with scatter signals to distinguish monocytes, lymphocytes and granulocytes.1

In addition to gating, single parameter histograms are also often used to display a single

measurement with light intensity on the x-axis and the number of events on the y-axis. The

sample histogram shown in Figure 1-4 is useful for evaluating the total number of particles/cells

in the sample that possess the physical properties selected for or which express the surface

marker of interest.

Figure 1-4 – A sample single parameter histogram of particles labeled with FITC dye. FITC (fluorescein

isothiocyanate) is a synthetic organic compound that is widely used as a fluorescent dye in many applications.

Its absorption peak is at 494 nm and emission maximum is at 512 nm.1

Page 21: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

8

A more complex and informative process is the two-parameter histogram shown in Figure 1-5,

one on the x-axis and the other on the y-axis. The cell counts of the measurements are shown as a

density plot, or contour map.

Figure 1-5 – Sample of a two-parameter (dual color fluorescence) histogram. Particles in this example are

labeled with PE and FITC dyes. PE (Phycoerythrin) is another organic fluorochrome whose absorption peak

is at 488 nm and 532 nm, and emission maximum is at 585 nm. 1

In this example, R2 encompasses the Phycoerythrin (PE)-labelled B cells whereas R5 contains

the fluorescein isothiocyanate (FITC)-labelled T cells. Both PE and FITC are organic fluorescent

dyes commonly used in microscopy and flow cytometry in biological applications. PE has an

absorption maximum at 488 nm or 532 nm, and an emission peak at 585 nm while FITC has an

absorption maximum at 494 nm and an emission peak at 512 nm.1,4 The top right quadrant

contains a few “activated T cells” that possess some HLA-DR (Human Leukocyte Antigen –

antigen D related) expression. HLA-DR is a type of cell surface receptor that mediates

acceptance or rejection of tissue or organ transplants.5,6 As the T cells with HLA-DR expression

are labeled with both antibody markers (PE and FITC), they are grouped in their own region

(R3). R4 contains cells negative for both FITC and PE, as shown in Figure 1-5.

1.2.3 Labels

To achieve specific detection and analysis of biological samples, target particles or analytes are

typically conjugated with labels. Most common labels used in flow cytometry are fluorescent

labels and isotope labels.1

Page 22: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

9

1.2.3.1 Fluorescent Labels

A wide range of fluorophores can be used as labels in flow cytometry. Fluorophores are typically

conjugated to an antibody that recognizes a target feature on, or in the cell, they may also be

attached to a chemical entity with affinity for the cell membrane or another cellular structure.

Each fluorophore has a characteristic peak excitation and emission wavelength, and the emission

spectra often overlap. Consequently, the combination of fluorescence labels which can be used

depends on the wavelength of the excitation sources used and on the detectors available.

1.2.3.2 Isotope Labelling

Currently, due to the limited available bandwidth in fluorescence detection, the maximum

number of labels that can be used in a flow cytometer is 17. In another approach intended to

overcome the fluorescent labelling limit, lanthanide isotopes are attached to antibodies. This

method could theoretically allow the use of 40 to 60 distinguishable labels and has been

introduced into a plasma, ionizing them and allowing time-of-flight mass spectrometry to

identify the associated isotopes. Although this method permits the use of a large number of

labels, it currently has lower throughput capacity than traditional flow cytometers. In addition,

this labelling technique destroys the cells, precluding their recovery by sorting.

1.2.4 State of the Art

The state of the art flow cytometers can perform measurements on samples labelled with up to 17

fluorescence markers simultaneously. Adding the forward scattering and side scattering, that

implies a total of 19 parameters that can be detected simultaneously, making flow cytometry a

powerful tool in clinical and research tool in biology and medical applications. The measurable

parameters of today’s flow cytometers include total DNA content, nuclear antigens, chromosome

analysis and sorting, cell detection and enumeration, enzymatic activity and characterizing

multidrug resistance in cancer cells.

1.2.5 Modern Cytometry Applications

Modern flow cytometry has application in a number of fields, including molecular biology,

pathology, immunology, plant biology and marine biology. In medicine the flow cytometer is

the tool of choice for measuring or monitoring a wide range of conditions such as

transplantation, hematology, tumor immunology and chemotherapy, prenatal diagnosis, genetics

Page 23: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

10

and sperm sorting for sex pre-selection. For example, in marine biology, the auto-fluorescence

properties of photosynthetic plankton can be exploited by flow cytometry in order to characterize

abundance and community structure. Similarly, in protein engineering, flow cytometry is used in

conjunction with yeast display and bacterial display to identify cell surface-displayed protein

variants with desired properties.

1.3 Aims of this Work

The flow cytometer has become an important tool in today’s clinical and research settings. The

introduction of novel labels such as quantum dots and nanoparticles enabled development of

novel probes and techniques to detect and characterize microscopic particles. On the other hand,

the development of new optics and electronics has opened doors for miniaturization and

advancement of flow cytometry instrumentation. The aim of this thesis is to develop a portable

particle detection and analysis system, which can be integrated with a handheld device, enabling

a more mobile and effective cell cytometry analysis.

Prior to this work, attempts at realizing portable/handheld cytometer were made. A

electrochemical based particle/cell detection system was developed by Li et al.7 where detection

was accomplished by measuring the impedance of the sample. Morgan et al.8 also developed a

microfabricated flow cytometer system where integrated photonic circuits and fibers were used

to excite and collect optical signals of target particles and molecules. However, these attempts

lacked a design approach that can be practically packaged in a commercial instrument for use in

clinical or low resource settings. In this thesis, microfluidic, lab-on-a-chip devices were

developed which allow various clinical and biological assays to be developed in a cartridge

format. Its size and physical properties brings significant advantages to developing new tools for

biological, chemical and medical analysis. It presents an ideal platform for the potable

cytometer/cell analysis.

Page 24: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

11

Figure 1-6 – Rendering of the proposed handheld cytometer. The instrument is intended to be a multi-test

platform system that can be used for a range of applications.

The ultimate aim of this work is to develop a portable, cartridge based cytometer system, as

illustrated in the 3D rendering shown in Figure 1-6, that can be integrated with a commercial

grade instrument to produce a portable system which can be taken into a rural or remote setting.

The key challenges in this work include integration of optics, lyophilization, fluorescence

imaging and microfluidic lab-on-a-chip devices.

The target requirements for this portable cytometer are summarized as follows:

Sample collection: finger prick

Sample volume required: 10 µL

Requirement for metering: Yes

On-chip reagent storage: Yes

Format of on-chip reagent: Dried

Mixing and actuation: Active and/or passive

Sample analysis throughput: > 0.28 µL/min

Overall test completion time (for CD4 cell counting assay): < 20 min

Instrument hardware must be portable, less than 2 kilograms in weight

Optical detection methodology: fluorescence

Light source: LED or Laser with emission wavelength at 532 nm

Detection optics: must have a magnification of 10x and a field of view of 1mm by 1mm

The proposed system will advance HIV monitoring, treatment, patient care, and point-of-care

analytical tools in general. The process of prognosis will be extremely efficient by this platform’s

ability to deliver a patient’s current condition in a matter of minutes. By integrating the smart

Page 25: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

12

electronic device functions including position tracking, communication, biometric identification

with advanced optical sensing technologies, this system can provide a complete and total

solution from patient identification, disease monitoring to patient care, tracking and treatment.

1.4 Organization of Thesis

The tremendous promise of microfluidic lab-on-a-chip devices, particularly in the application of

point-of-care testing, is the main motivation for this work.

Chapter 1 introduces the flow cytometry and its application in modern biology and medicine.

Chapter 2 focuses on the background of the work, including topics in point-of-care testing,

microfluidics, and current state-of-the-art in microfabricated and miniaturized flow cytometry

instruments and devices. Starting from Chapter 3, the thesis describes the main contribution of

this work: microfluidics of the disposable device, detection methodology, and image acquisition

and analysis. Chapter 3 and 4 presents on-chip fluidics developed for the miniaturized flow

cytometry application. In Chapter 5, a unique and novel imaging approach is proposed and

characterized while Chapter 6 is a summary of the thesis with a brief look ahead on potential

future work.

Page 26: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

13

Chapter 2 Background

2.1 Microfluidic Lab-on-a-Chip

Microfluidics is the study of systems that analyzes small amount of fluids (10-9 to 10-18 liters),

using channels with dimensions of tens to hundreds of micrometers. Some of the first research

and scientific projects involving microfluidics began with the development of high pressure

liquid chromatography9 and capillary electrophoresis10,11.

2.1.1 History of Microfluidics

Towards the end of 1970s, the field of microfluidics emerged as a valuable tool for analytical

chemistry as well as pharmaceutical research. A silicon micro-machined gas chromatograph was

first developed in 1975 by Terry12,13. This remarkable gas chromatograph was capable of

separating simple mixture of compounds in a span of a few seconds. The device included a valve

to control sample injection, a column that is 1.5 meters long for separation, with all functional

components integrated in a single silicon wafer. A thermal conductivity detector was fabricated

on a separate silicon wafer and mechanically clamped to the wafer containing the column.

However, this research was not very well received by the academia community. Related research

were primarily focused on the fabrication of component such as micropumps14-18,

microvalves19,20, and chemical sensors21,22. The need to precisely control liquid flow led to the

development of micropumps and valves in the integrated systems.23-25 Furthermore, the concept

of miniaturized total chemical analysis system (µTAS), also known as Lab-on- Chip (LOC), was

first proposed by Manz et al. in 199026, in which silicon chip analyzers incorporating sample

pretreatment, separation, and detection played a fundamental role. At the time, µTAS was

envisioned as a new concept for chemical sensing, given the sensor technologies available were

not providing the necessary requirements in terms of selectivity and lifetime. In the beginning,

the main purpose and motivation for miniaturization was to enhance the analytical performance

of the device. However, it was realized that smaller overall sensor architecture also meant

smaller consumption of sample and reagent. The concept of µTAS enabled the integration of

separation techniques that could provide the monitoring of many components within a single

Page 27: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

14

device. It was envisioned such devices were capable of performing multiple functions in the

same device20,21, including sample handling, transport, analysis, detection and measurement,

hence the name total analysis system.

Early theoretical considerations of the miniaturization concept demonstrated that pumping using

electro-osmosis was an effective and possible method to transport samples inside the on-chip,

interconnected channel systems, especially those where separation was required26. On the other

hand, conventional pumps utilizing high pressure systems showed problems in such systems

during fluid and sample transport when interconnecting channels were on the order less than 20

µm wide or deep. Research efforts continued to develop novel pumping methodologies16,27 and

sample introduction17,20 in such microsystems.

Electrophoresis in planar chips was successfully integrated and demonstrated for the first time in

1992 using silicon and glass substrates28,29. These results showed the feasibility of using

electroosmotic pumping for flow control in interconnected microchannels without the use of

valves and the use of glass and silicon as a potential material of choice for µTAS. The concept of

µTAS that integrates sample introduction, separation and detection within the same device was

first demonstrated. Subsequent efforts were made towards increasing the separation efficiency.29-

32, where amino acids and dyes were separated in less than 30 seconds with plate height of 0.3

µm. In addition, automated repetitive sample introduction and separation on a time scale of

seconds were also achieved. The most common detection method was laser induced

fluorescence, which was used to detect mixtures of fluorescein derivatives and fluorescein

isothiocyanate-labeled amino acids separated on-chip.33 In addition to the separation of

biological samples, applications related to the reaction and handling of biomolecules and cells

began to develop. These include the use of microfabricated chambers to carry out Polymerase

Chain Reaction (PCR) to amplify DNA strands,34 the measurement of cellular metabolism in

micromachined channels,35 and the use of microfabricated devices for flow cytometry

application.36

2.1.2 Recent Advances

Recent advances in microfluidics science and technology are enabling more functions and

features that can be integrated in a lab-on-a-chip device to complete more complex analysis.

Page 28: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

15

Progress has been made on a multitude of fronts, from materials to fabrication techniques, from

fluidic manipulation to detection, and from integration to application. Iliescu et al.37 reviewed

silicon and glass microfluidic systems. A polymer/glass-hybrid approach was proposed and

presented 38,39 which led to a renaissance in silicon based detectors for microfluidic systems.

Droplet based PCR40, cellular culture41 and nanowire for label-free cardiac enzyme detection42

were also developed in recent research activities.

A great deal of research has also focused on the development of on-chip fluidic manipulation and

functionalities, including sample separation, mixing, fluidic management and detection.

Although microfluidic electrophoretic systems received more initial attention than

chromatographic ones, important progress has been made in both areas, as covered in greater

detail in recent review articles on microfluidic chromatography43,44 and electrophoretic

approaches.45,46

Optical detection methods have several advantages over other electrical based methods.

Typically, optical detection techniques have good detection limits, they can be isolated from the

fluid inside the fluidic channels, and can be used to monitor a wide variety of compounds.47

Mogensen et al. provided a very in-depth investigation on this topic in a recent review.48 There

are several approaches to optical detection that are currently being implemented in microfluidic

systems; these can be classified as label-based, such as fluorescence and chemiluminescence, or

label-free.49 More specifically, Laser-induced fluorescence (LIF) is the most frequently used

optical method in microfluidic systems because of its low detection limits and availability of off-

the-shelf components and devices to design such systems.50 In many instances, the detection

optics are not integrated in the fluidic chip itself. For LIF, a light source such as a laser is used

for excitation and a CCD or photodiode detector is used for detection.51,52 Integration of an LED

excitation source and a photodiode detector in a micro-system has been shown, producing

relatively high limits of detection of 100 nM for rhodamine 6G and 10 μM for fluorescein.53

Integration of pumps into microfluidic devices can significantly decrease external equipment

needs and reduces the dead volumes from interfacing with pumps. However, it could increase

complexity and difficulty in manufacturing of the microfluidic devices. A detailed review on

micropumps and microvalves has also recently been published by Au et al.54 Because flow inside

microfluidic channels is strictly laminar due to the microscopic channel size, the main driving

Page 29: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

16

force for mixing in a microfluidic system is by diffusion. Several micromixer designs have been

developed in the pursuit of thorough and rapid mixing of multiple samples.55 Detailed reviews of

mixing systems for microfluidics have been written by Jeong et al.56 and Lee et al. 50

These are only few examples highlighting the recent advances in the field of microfluidic lab-on-

a-chip research. Nge et al. has presented a more thorough and complete in-depth summary of

microfluidics research in a recent review article.57

2.1.3 Microfabrication Technologies

The development of microfabrication technology for microfluidic lab-on-a-chip systems

stemmed from the readily available and recent advances in microelectronics manufacturing

processes. It is clear from the history of the development of microfluidics, the material (silicon

and glass) and fabrication process (photolithography) were directly taken from the

microelectronics industry.20,21 Gradually plastics became the dominant material selection for

microfluidics devices based on its excellent optical quality, biocompatibility and ease of

manufacturing at both prototyping stage and for large scale production. The following sections

outline some of the most common fabrications technologies currently used.

2.1.3.1 Laser Processing

Laser ablation was used by Grzybowski et al. for rapid fabrication of elastomeric masters

(poly(dimethylsiloxane) or PDMS) for micro-contact printing (µCP) and a technique named

controlled sagging micro-contact printing (CSµCP) was developed. This approach is capable of

patterning structures with dimensions of 1 µm in width without the need of a cleanroom

environment, or specialized photolithographic tools.58 With the advent of ultrafast photonics, Ho

et al.59,60 demonstrated the feasibility of creating 3D structures inside a fuse silica bulk. In this

approach, the fused silica was directly exposed to ultrafast laser pulses followed by chemical

etching with diluted hydrofluoric acid. Both woodpile structures and 3D microfluidic channel

networks were fabricated with a resolution of 5 µm.

Page 30: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

17

2.1.3.2 Lithography

2.1.3.2.1 Photolithography

Photolithography is the process of transferring geometric shapes on a mask to the surface of a

silicon wafer.61-63 It uses light to transfer a geometric pattern from a photomask to a light-

sensitive chemical material known as “photoresist”. A series of chemical treatments then either

etches the exposure pattern into, or enables the deposition of new material in the desired pattern.

The steps involved in the photolithographic process are wafer cleaning; barrier layer formation;

photoresist application; soft baking; mask alignment; exposure and development; and hard-

baking.

Many microfluidic lab-on-a-chip devices have been fabricated using photolithography

techniques, particularly using SU-8 photoresist. For example, the preparation and

characterization of optical and mechanical properties of SU-8 negative photoresist for the

fabrication of high aspect-ratio structures were reported by Lorenz and co-workers.64 Using

alternating spin-coating of SU-8 photoresist and exposure steps followed by a single

development step to remove the unpolymerized resist, Guérin et al. have fabricated monolithic

SU-8 channels.65 Microchannels of 80-nm width on carbon-based resist were fabricated by

Johnson et al. on Si, SiO2, and gold substrates by exposing them to a metastable argon atom

beam in the presence of dilute vapors of trimethylpentaphenyltrisiloxane.66

2.1.3.2.2 Soft Lithography

Pioneered by Whitesides and Quake, soft lithography is a unique technique to fabricate structures

using elastomeric stamps, molds and confirmable photomasks.67 It is generally used to build

features measured typically on the order of nanometer to micrometer scale.68 The key advantage

of soft lithography include lower cost than traditional photolithography, more suitable for

biotechnology applications and processes,69 and ability to rapidly generate prototype device.70

Since its first publication in 199867, soft lithography has become the most widely used rapid

prototyping technique in the research and development of microfluidic systems. The work

summarized in Section 2.1.2 was predominantly completed using soft lithography fabrication

process.

Page 31: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

18

2.1.3.3 Injection Molding

As research on microfluidic lab-on-a-chip devices grows and matures, many research activities

are focused on the manufacturing of integrated microfluidic devices on a mass-production scale

with relative low costs.71 This is especially critical for applications where disposable devices are

used such as medical diagnostics and analysis. For mass production, polymer material presents a

very attractive alternative material choice compared to glass and silicon given its cost and ease of

manufacturing.

Micro-injection molding is the process of transferring a thermoplastic material in the form of

granules from a hopper into a heated barrel so that it becomes molten and soft. The material is

then forced under pressure inside a mold cavity where it is subjected to holding pressure for a

specific time to compensate for material shrinkage. The material solidifies as the mold

temperature is decreased below the glass-transition temperature of the polymer. After sufficient

time, the material freezes into the mold shape and gets ejected, and the cycle is repeated. A

typical cycle lasts between few seconds to few minutes. The process has a set of advantages that

makes it commercially applicable with potential for further developments in the future.

Advantages include the wide range of thermoplastics available and the potential for full-

automation with short cycle times,72,73 cost-effectiveness for mass-production process, especially

for disposable products44,45,74-76, very accurate shape replication and good dimension control45,46,

low maintenance costs of capital equipment, when compared to lithographic methods77, and

applicability of the large amount of industrial information and know-how available from

conventional injection molding. Within certain limitations, this may be scaled down to micro-

injection molding.

A comparison between micro-injection molding and other techniques for microfluidic device

manufacturing, such as hot-embossing and PDMS casting, is also available in the literature76.

Other micromachining techniques have been presented using deep reactive ion etching (DRIE)

and surface fusion bonding (SFB)78 and an electron cyclotron resonance (ECR) source79,80 to

produce high-aspect-ratio narrow-gap silicon devices.

On the other hand, polymers have some limitations related to their properties or processing

techniques relative to glass. These include limited operation-temperature range, higher auto-

fluorescence and limited well-established surface modification techniques. Table 1 presents a

Page 32: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

19

comparison between polymers and glass for manufacturing microfluidic devices, where

information is compiled from the literature.77,81-88 When it comes to processing, mass-production

processing techniques impose limitations on the moldable geometry of the microfluidic device.

These geometrical limitations restrict the flexibility of integrating external functional elements

within a mass-manufacturing technique.

Page 33: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

20

Table 1 - Comparison between polymer and glass as substrate of microfluidic systems for bio and chemical

applications.

Polymers Glass

Manufacturing costs Lower costs than glass, especially

in mass volume

More expensive to

manufacture as the fabrication

process are more complex

Fabrication complexity Simpler fabrication process Time consuming and

expensive, and usually wet

chemistry is used.

Clean room facilities Cleanroom environment is

necessary

Clean-room facilities are

required.

Properties A wide range of polymer material

to choose from to tailor the

desired requirements

(mechanical, optical and

chemical)

Less variability in available

properties compared to

polymer.

Operation

temperature

Narrower range due to the low

glass transition temperature

Wider range of operation

temperature than polymer.

Optical properties and

fluorescence detection

Higher auto fluorescence in the

UV end of the spectrum and

lower transparency than glass

Superior optical property than

glass.

Bonding Different bonding options are

available, including adhesives,

thermal fusion and mechanical

welding

More time consuming than

polymer. Possible bonding

processes include thermal,

adhesive and anodic bonding.

Page 34: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

21

Surface treatment Direct treatment available

including oxygen plasma

treatment

Established chemical

modification procedures for

glass are available.

Compatibility with

organic solvents or

strong acids

Generally not compatible with

most organic solvents and in

some cases, strong bases or acids

Excellent resistance to

solvents and acids.

Joule heating Significant Joule heating due to

low thermal conductivity

More resistant to Joule

heating.

Electro osmotic flow

(EOF)

Smaller EOF because of lack of

ionisable functional groups

Higher EOF compared to

polymers.

Geometrical flexibility More flexibility for geometrical

designs with a wide selection of

different cross-sections (curved,

vertical or V-groove); high aspect

ratio and arbitrary wall angle

Limited to 2D designs due to

the isotropic nature of the

etching process. Less flexibility

in cross-sections than

polymer.

Permeability to gasses Higher gas permeability relative

to glass

Does not meet the gas

permeability requirements for

some biological applications

such as living mammalian

cells.

2.2 Point-of-Care Testing

Point-of-care testing (POCT), also known as bedside testing, is defined as medical testing

conducted at, or near the site of patient care.89 Typically, these are simple blood or urine tests

which can be performed by nurses, healthcare workers or patients. Examples of such POCT

Page 35: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

22

systems include rapid diagnostic tests for HIV, malaria, pregnancy test, blood glucose monitor,

blood chemistry and electrolytes analysis, and portable ultrasound just to name a few. The main

driving force in the development of POCT is to improve the accessibility of medical tests to the

patients. This increases the likelihood that the patient, physician and healthcare professionals to

receive the results faster, which leads to immediate clinical and patient care decisions.

Generally, POCT consists of a portable, or handheld instrument and a disposable test kit or

cartridge.90 The ultimate goal is to collect patient sample and obtain the test results in a very

short period of time at, or near the location of the patient so that clinical decisions can be made

faster. Cheaper, faster and smarter POCT devices have increased the use in a wide range of

disease diagnosis and testing, such as diabetes, acute coronary syndrome, malaria and

HIV/AIDS.91

A major class of POC diagnostic tests is the lateral flow test, which uses a membrane or paper

strip to indicate the presence of protein markers such as pathogen antigens or host antibodies. On

a membrane, addition of sample induces capillary action without user intervention. As the

sample flows across the membrane, it reacts with embedded labeling reagents in the membrane,

and flows over an area that contains capture molecules. The labeled, captured analytes are

usually read and interpreted by human eye to form a visible band. In the U.S., lateral flow tests

are used for diagnosis in a small number of indications, most notably pregnancy as well as

infections with streptococcus or flu. On the other hand, in developing countries or remote areas,

the lateral flow test is widely used to diagnose infectious diseases such as HIV and malaria.

Although the test is simple to perform, the single-flow action does not resemble the multi-step

procedures of laboratory-based assays that are crucial for producing highly reproducible,

quantitative, and sensitive results. As the lateral flow test comprises a multibillion dollar market,

with many of the technologies now consolidated at the company Alere (formerly Inverness

Medical Innovations), there has been significant industry interest in trying to improve their

performance over the last few decades, but so far without significant progress.

The other major class of successful POC tests is the blood glucose test, which can be considered

as a classic example for a high impact POC diagnostics product that has improved millions of

diabetic patients’ lives, and now acts as a pillar of the entire diagnostics industry. The glucose

test is also performed on membranes, but is typically classified differently from lateral flow

Page 36: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

23

immunoassays as the rest of the analytical method is altogether different. The glucose test uses

signal amplification by a redox enzyme, which generally generates an electrochemical signal to

be detected by a reader.92 The glucose test, however, is by nature is somewhat unique. On the

technology side, the concentration of the analyte is in the mM range, which far exceeds the

concentration of most diagnostics markers. On the market side, the frequency of testing, typically

multiple times a day, outnumbers that of most other tests, which creates a billion-dollar market

world-wide. Both factors contribute to the rapid development and tremendous success of the

POC glucose testing.

POC testing has seen significant and rapid advances in the last decade.92 A number of

technologies and products were successfully developed and commercialized. The research and

development of POC tests often require interdisciplinary expertise, ranging from biology,

chemistry, to fluidics, electromechanical or optical detection. The availability of POC tests for

infectious and non-communicable diseases enabled faster and more affordable medical

diagnostics to be conducted at or near patient, significantly reducing the burden on healthcare

systems world-wide.

2.2.1 Microfluidics and POCT

With the emergence of new POC diagnostic technologies in the market, there has recently been a

resurgence in interest to develop novel and clever methods to re-invent both lateral flow

immunoassays and the glucose test, to significantly improve their detection limit, quality control

and readout systems, as well as expanding their range of targets. Examples include marrying

modern LOC concepts, such as sophisticated flow control, to diagnostic tests performed on paper

and membranes.93-95 In the current concept of LOC-based devices, the iSTAT handheld system

(now part of Abbott) was among the first commercially successful products. The iSTAT system

marries miniature fluidics and electrochemical detection to conduct clinical chemistry

measurements, such as electrolyte levels and limited immunoassays using a disposable test kit.

Another interesting hybrid of LOC technologies with lateral flow is the A1cNow® test for

diabetic patients formerly from Metrika (now Bayer Healthcare), which uses multiple strips

integrated with detection optics in a single package.

Page 37: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

24

The clinical need for new POC diagnostic tests remains high, especially for tests that can detect

low concentrations of the target and with an ability to quantify the result. Currently, tests such as

lateral flow immunoassays can detect analytes present in a sample solution at high native

concentrations ranging from µM to mM. For targets present at low native concentrations that are

beyond the detection limit of the current detection methodologies, the assay systems require

amplification either of the signal or the target, which typically increases the complexity of the

testing device, and is not available in current POC tests.96 In addition, current POC can mostly

produce qualitative results. The challenges in designing and manufacturing quantitative tests

remain prohibitive. Finally, multiplexing is another desirable feature that current POC tests strive

to incorporate.

POC testing is beginning to benefit from the potential and recent advances in microfluidics

research. The degree of integration of a microfluidic technology can vary from having a

disposable microfluidic chip used with peripheral equipment (pumps, reader, etc.) to having all

functions needed for processing and analyzing a sample and reporting the results on a chip. The

main selection criteria are portability, time to result and cost per test. The time to result is

between seconds and minutes as devices are often used at the patient side and timely results are

key requirements. Multiplexing is usually done for a few analytes. The size and weight of the

device are minimal and affect the portability and energy consumption of the reader peripheral.

The cost per test must be low in order for the test to be performed routinely and fit into pricing

and reimbursement policies that are relevant for the geography where the tests are performed. As

can be seen from above, there is a large number of requirements that POC diagnostics must meet.

Generally, technologies for research and central laboratories meet these requirements by using a

variety of peripheral equipment, several sampling methods, flexible protocols, and a number of

signal detection formats. In contrast, a POC microfluidic device is optimized during

manufacturing for a particular application.

Central laboratory testing is done mostly on clinical analyzers. The main selection criteria are

throughput and cost per test. Samples are sent from the patient to the central laboratory and

placed in a queue with an option for high priority. The time to result can be from several minutes

to hours and is usually not critical. Clinical analyzers have a large variety of analysis capabilities

and can detect hundreds of analytes. Machines can be meters in size and weigh more than a ton.

Page 38: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

25

Operators of clinical analyzers are usually trained technicians. In clinical laboratories that

perform a large number of tests, the instrument is often provided and the instrument cost may be

small compared to the running costs. The cost per test should be low enough to be done

routinely, but can be higher for less common analytes.

Microfluidics is, at its heart, a technology, with a primary goal of improving the performance of

end products. Lab-on-a-chip (LOC) technologies utilizes the manipulation of fluids and particles

in a microfluidic system, which can be fabricated in a glass or polymer substrate. LOC enables

the significant reduction in sample and reagent volumes compared with conventional bench

based, macroscopic analysis techniques. Secondly, chemical separation procedures are much

faster and more efficient at these small dimensions.11 Arrays of similar structures on one chip

allow for a large group of measurements to be made under the same conditions and at the same

time. Most importantly, since sample handling, reactions, separation, transport and detection all

take place on the same substrate, supplementary connecting interfaces between these different

functions are eliminated. For building POC diagnostic devices, there have been dazzling progress

in the development of individual LOC components, but unfortunately only very few microfluidic

technologies have made the leap to fully functioning integrated devices that provide real clinical

value.96

2.2.2 Lab on a chip and Global Health

Lab-on-a-chip (LOC) technologies have a tremendous potential to improve the efficiency of

healthcare system in Low and Middle Income countries (LMIC). Ever since the modern

inception of LOC and microfluidic technologies around 1990, use in remote settings has been

perceived as potentially one of the most powerful applications of the technology. Indeed,

portable LOC devices are now beginning to be used in remote settings, as a result of

developments in integrating fluid actuation, sample preparation, sample separation, signal

amplification, and signal detection into a single device.

There is an urgent need in developing countries for new and innovative health-related

technologies, and specifically, new technologies for health diagnostics, to improve patient care.

For example, in one survey of international scientists familiar with the public health programs of

Page 39: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

26

developing countries, Singer et al. discovered that the overall top priority in new technology

development for global health was “modified molecular technologies for affordable, simple

diagnosis of infectious diseases”.97 Similarly, in a study by the Bill and Melinda Gates

Foundation and the NIH to identify “Grand Challenges for Global Health”, two of the 14

priorities involved diagnosis and measurement of patients’ health statuses.98 Microfluidic LOC

technology holds substantial potential for fulfilling these challenges by automating complex

diagnostic procedures that are normally performed in a centralized laboratory on a microfluidic

chip. This outcome could empower healthcare workers and patients with simple-to-use POC tests

to generate important health-related information in even the most remote settings. To this effect,

funding by philanthropic foundations (such as those from Doris Duke, Soros, and Gates) are

leading the development of microfluidics technologies for diagnostics in LMICs. The broad aim

of these scientific initiatives is to combine new diagnostic and prevention methods with

treatment to improve global health.99,100

In both developed and developing countries, early and accurate diagnosis for every disease is

critical for the well-being of patients: it permits prompt and proper treatment of patients, and

minimizes the waste of public resources on ineffective treatments.97 In developing countries, the

value of diagnosis for certain diseases is sometimes mitigated by the lack of available treatment.

Early diagnosis can allow patients to receive required therapy and medication on time, reducing

morbidity and mortality, and investments in diagnostics and prevention can be more cost-

effective than treatment.101 Moreover, point-of-care devices can enable epidemiological

surveillance of diseases,102 which is an especially challenging problem in developing countries.

For scientists and engineers who aim to design new diagnostic technologies, a crucial question

for achieving real world impact is which health conditions in developing countries are most in

need of diagnostic devices. In a study led by Murray and Lopez, the World Health Organization

conducted an unprecedented and comprehensive initiative to compile statistics for comparing the

relative burden of diseases, conditions, injuries, and risk factors on a global scale.103,104 Table 2

summarizes the most common diseases by disability adjusted life years (DALYs) in developing

countries, which is a metric that accounts for years of life lost due to premature death and

disability. Infectious diseases constitute a large burden of disease in developing countries

(32.1%; by comparison, they represent only 3.7% of total DALYs in developed countries). The

combination of HIV/AIDS, malaria, and tuberculosis (TB), constitutes an important 12% of

Page 40: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

27

DALYs in developing countries. The social impact of these diseases stretches beyond the DALY

statistics, however, since HIV/AIDS (along with common co-infections of TB) targets healthy

adults, thereby leaving behind villages of orphans which destroy the underlying functioning of

entire communities.105

In addition to infectious diseases, the burden of non-communicable diseases in developing

countries is often underappreciated.106 The list of important non-communicable diseases include

cardiovascular disease (such as ischemic heart disease and stroke), cancer, neuropsychiatric

conditions (such as unipolar depressive disorder), and respiratory diseases (such as chronic

obstructive pulmonary disorder and asthma). As the standard of living in developing countries

improves and average life span increases, the burden of disease will gradually shift to the non-

communicable diseases. Already, obesity and diabetes are increasingly prevalent in developing

countries.107 As these trends develop, accessibility of the corresponding diagnostic technologies

in developing countries can be difficult in developing countries due to the lack of infrastructure

and inadequacy in the healthcare systems.

Page 41: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

28

Table 2 - Prevalent diseases in developing countries. DALY is a measure of the disease burden on healthcare

system. Type of test is the conventional testing methodology.105

Disease % DALY

Type of Test

Communicable diseases 32.1

Respiratory infection 6.8 Immunoassay

HIV/AIDS 6.1 Immunoassay

Diarrheal disease 4.5 ELISA

Malaria 3.4 Microscopy; immunochromatography

Tuberculosis 2.5 Microscopy; PCR

Non-communicable diseases 43.5

Neuropsychiatric conditions 11.7 Hormone levels

Cardiovascular disease 9.5 ELISA

Sense order disease 4.6 clinical diagnosis

Cancer 4.2 Immunoassay

Respiratory diseases (asthma) 3.5 Spirometry

Digestive diseases 3.0 Complete blood count and blood chemistry

Maternal perinatal and nutritional conditions

11.8 No tests available

Perinatal conditions 7.0 Clinical diagnosis

Nutritional deficiencies 2.5 Immunoassay; cell count

Maternal condition 2.4 Hematology

Intentional injuries 3.3 Cell culture; immunoassay

Unintentional injuries 9.2 Analytical toxicology

Page 42: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

29

To diagnose this wide array of diseases and conditions, assays with a variety of methodologies

will be needed. The types of assays that are currently used to diagnose them are listed in Table 1;

some assays are in great need of new diagnostic methods.

The use of point-of-care testing systems in the resource limited settings pose a series of design

challenges, including affordability, time to analysis result, ease of use and rugged system design.

In addition, the system must be operable without access to stable electricity and clean water.

These constraints hold direct pertinence to the design of the diagnostic technology. It is

necessary to take them into consideration at the earliest design stages to ensure the performance

of the product is satisfied.

2.2.3 Motivation of this Thesis

A key problem with the current healthcare system lies in the disease diagnostics. Most of the

medical devices and instrumentations are designed for use in the central laboratories and

hospitals where established infrastructure must be available to operate the equipment effectively.

As a result, for people living in resource limited settings or remote areas around the world,

access to medical diagnosis is very limited and sometimes non-existent. The aim of this thesis is

to develop novel platforms that enable easy access to state-of-the-art in vitro diagnostics by

utilizing recent advances in microfluidic lab-on-a-chip technologies. The target assay chosen in

this work is CD4 T cell count test, which is a measure of a human’s immune system strength.

2.2.3.1 CD4 T Cell Count and HIV/AIDS

The CD4 T-cell count (CD4 count) serves as the major laboratory indicator of immune function

in patients who have HIV infection. It is one of the key factors in determining both the urgency

of antiretroviral therapy (ART) initiation and the need for prophylaxis for opportunistic

infections.108 It is also the strongest predictor of subsequent disease progression and survival

according to findings from clinical trials and cohort studies109-111.

CD4 cells or T-cells are a type of white blood cells that play a major role in protecting your body

from infection. They send signals to activate your body’s immune response when they detect

“intruders,” like viruses or bacteria. Once a person is infected with HIV, the virus begins to

attack and destroy the CD4 cells of the person’s immune system. HIV uses the machinery of the

Page 43: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

30

CD4 cells to multiply (make copies of itself) and spread throughout the body. This process is

called the HIV life cycle.

A CD4 count is a lab test that measures the number of CD4 cells in a blood sample. It is an

important indicator of how well the immune system is working. The CD4 count of a healthy

adult/adolescent ranges from 500 cells/mm3 to 1,200 cells/mm3. A very low CD4 count (less than

200 cells/mm3) is one of the ways to determine whether a person living with HIV has progressed

to stage 3 infection (AIDS). ART involves taking a combination of HIV medicines every day. It

prevents HIV from multiplying and destroying patient’s infection-fighting CD4 cells. ART

cannot cure HIV, but it can help patient live a longer, healthier life and reduce the risk of HIV

transmission by suppressing the virus.

When the amount of HIV in a patient blood is lowered by ART, it allows the CD4 cells to

reproduce and increase in number. The higher the CD4 count, the more capable patient is to fight

HIV and other infections. ART is recommended for everyone with HIV, but the urgency to start

ART is greater in people with low or rapidly falling CD4 counts. A falling CD4 count indicates

that HIV is advancing and damaging patient’s immune system.

Traditionally, a CD4 T cell count is completed using flow cytometry, a well-established

methodology that requires sophisticated infrastructure and technical expertise to operate. In

developing countries, the lack of healthcare infrastructure and resources make it impossible to

monitor the CD4 T cells in all HIV patients. There is an urgent need to develop simple to use,

affordable POC CD4 testing solutions with vigorous quality control protocols.

2.2.3.2 Current Gap and Proposed Concept

Flow cytometry is an important blood analysis technology that remains largely inaccessible for

clinical use in low and middle-income countries (LMICs) due to size, cost, and the lack of

infrastructure and skilled personnel. The result is a highly inefficient health system where a

patient’s blood must be sent to a central laboratory (when available). The patient must return

weeks later to get test results. This delay blocks clinician decision making; test results often do

not make it back to the clinician; and patients are often “lost to follow up” leading to increased

morbidity and death.

Page 44: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

31

This thesis aims to develop a novel laboratory-quality diagnostic platform, which can be

described as a mini flow cytometer. The platform is intended to be mobile, simple to use, and

inexpensive. By leveraging advances in microfluidic and biomarker technologies, we plan to

research and develop a microfluidic based point-of-care system that enables the detection of cell-

surface and blood serum protein biomarkers. The ultimate objective is from a single drop of

blood, health workers in remote locations will rapidly and accurately perform tests to diagnose or

monitor a range of infectious and non-communicable diseases. The portable, or handheld

platform will allow for testing at community level facilities, mobile clinics and on hospital

wards. Simplicity of use will allow front-line nurses, laboratory technicians and advanced

community health workers to reliably perform accurate tests. Disposable microfluidic cartridges

do not require cold chain and have a minimum 12-month shelf life. Quantitative results will be

available within tens of minutes (depending on test). A rechargeable battery will allow for day-

long continuous use. The low cost of platform and tests will allow for the cost-effective uptake

of the platform in low-test throughput sites (5-20 tests per day). Cloud connectivity enables the

review of results from a central location for quality control, clinical decision-making support,

and facilitates electronic medical record (EMR) data aggregation. Diagnostic testing available at

the point-of-care, especially in remote or rural settings, will improve patient care and morbidity

and mortality outcomes, improve health worker motivation, increase health system efficiencies,

and significantly reduce health system costs.

Starting from Chapter 3, this thesis will present the design and development of a novel

microfluidic based imaging and detection system. This portable system forms the core of a

handheld cytometer that can be further integrated into a point-of-care tool for clinical testing and

diagnosis. As a first demonstration of its clinical application, a CD4 T cell counting test is part of

the objective of this thesis and described in the subsequent chapters. Microfluidic chips were

designed, fabricated and tested against the current state-of-the-art flow cytometers to validate the

performance of the detection methodology. Engineering challenges in designing on-chip

functions, such as reagent incorporation, blood cell labeling and mixing, on-chip fluidic actuation

and fluorescence detection were described in detail in Chapter 3, 4 and 5.

The same detection platform can be further expanded to image and analyze blood serum proteins

using beadarray technology. Chapter 5 also included an introduction the concept of the beadarray

Page 45: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

32

system and provided a proof-of-concept demonstration and analysis towards building a portable

or handheld ELISA-like beadarray instrument.

Page 46: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

33

Chapter 3 Passive Microfluidic Systems

In flow cytometry, precise control of fluid flow is critical to ensure the proper functioning of the

instrument. For microflow, or microfluidic-based cytometers, this is of utmost importance to the

overall system. Typically, conventional flow cytometers employ a sheath flow surrounding the

sample stream to form the particles/cells into a single file stream. This stream must be stable

without significant pulsation and well aligned with the detection optics for optimal sensing of

each particle/cell. The shape, width, and height of the detected optical signal of the particle/cell

passing through the interrogation point corresponds to its position, speed in the flow stream.

Hence, a slight variation in flow will result in variation of the detected signal.

3.1 Introduction

Capillary driven flow is ideal for disposable, on-site analytical systems, such as point-of-care

devices because of no external energy such as electricity or mechanical forces are required. The

complexity of the system is also reduced compared to other active fluidic systems. However, the

flow characteristics is heavily dependent on the channel geometry and material properties. In a

passive actuation system, system design and power requirements are significantly reduced as no

active components are needed. The interfacial energy, or surface tension, is the dominant driving

force that moves the liquid sample in the microfluidic channel.

This chapter discusses concept and strategies of control fluidic flow on-chip. The first sections

introduce various methodologies used in microfluidic based flow cytometry applications and

reviews the microfluidic system design concept for the proposed cell/particle imaging platform.

This chapter then describes the concept, theory and experimental results on capillary microfluidic

systems, one of two fluidic transport strategies, developed during this thesis.

Page 47: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

34

3.2 Flow Control Methodology in Microfabricated Flow Cytometers

The fluidic system in a flow cytometer is used to control the motion of the particles suspended in

a liquid medium, transporting target particles to the interrogation point for optical

characterization. For optimal performance, the particles must be positioned at the center of the

optical beam and only one particle is being illuminated by the laser at any time during operation.

In conventional flow cytometry, hydrodynamic focusing is used to organize the particles and

cells into single file formation. A sheath flow of distilled water is pumped through the instrument

into the flow cell to create the hydrodynamic focusing effect. In portable systems aimed for

applications at point-of-care where microfluidic technologies are typically used, generation of

precise flow control to produce sheath flow is a challenge. In micro fabricated flow cytometer

devices and systems, other approaches were developed.

There are two distinct approaches in creating sheathless particle focusing in a microfluidic based

flow cell in cytometry: field based and flow assisted method. Field based approaches include the

application of external physical forces such as electric, acoustic and optical forces, whereas flow

assisted methods entails micro-channel or micro structures that form physical barriers to move

particles out of their streamlines and into desired focusing pattern.

3.2.1 Acoustic

Similar to electric fields, acoustic waves can generate pressure gradients in a fluid transporting

suspended particles either to the pressure nodes (minimum pressure amplitude) or the antinodes

(maximum pressure amplitude). Particles can be trapped or focused with resonating transducers

generating the acoustic wave field (confocal or planar fields). When a standing wave is generated

in a medium, the acoustic pressure at position x can be described by the following relationship:

∆𝑝(𝑥) = 𝑝𝑜 sin(𝑘𝑥) cos(𝑤𝑡) (3 – 2)

where 𝑝𝑜 is the acoustic pressure amplitude, 𝑘 is the wave number of ultrasonic radiation (𝑘 =

2𝜋/𝜆, λ is the wavelength), x is the distance from the nodal position in the medium, 𝑤 is the

angular frequency, and t is time. An acoustic radiation force on a particle can be expressed as the

following:

Page 48: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

35

𝐹𝑎𝑐 = −4

3𝜋𝑅3𝑘𝐸𝑎𝑐𝐴 sin(2𝑘𝑥) (3 – 3)

where 𝑅 is the particle radius, 𝐸𝑎𝑐 is the averaged acoustic energy density, and A is the constant

given by material density, compressibility and the sound velocity in the medium and particle.

When A is positive, the particles move to the nodal position of the acoustic standing wave.112

3.2.2 Electrical

Dielectrophoresis can be used to focus micrometer and even nanometer sized particles in a flow

stream. A non-uniform electric field drives motion of dielectric particles in a certain direction.

The dielectrophoretic force acting on a particle in a non-uniform electric field is represented by

the following expression:

𝐹𝐷𝐸𝑃 = 𝜋𝑎3𝜖𝑚𝑅𝑒[𝑓𝐶𝑀]∇|𝐸|2 (3 – 1)

where 𝑎 is particle radius, 𝜖𝑚 is the permittivity of the suspending medium, 𝑅𝑒[𝑓𝐶𝑀] is the real

part of the Clausius-Mossotti factor, 𝑓𝐶𝑀, and E is the applied non-uniform electric field.113

The direction of the particle movement is determined by the sign of the real part of the Clausius-

Mossotti factor which depends on the permittivity and conductivity of the particle and the

suspending medium, and the frequency of the applied electric field. Particles move either toward

the region of high-electric field strength (positive DEP) or to the minimum field gradient

(negative DEP). 113

An example of using DEP to focus micro-particles is demonstrated by Yu et al.114 The elliptical

channel was fabricated by bonding of two soda-lime glass wafers after chemical etching and

electrode deposition. The electric field gradient was generated in the radial direction from the

electrode pattern and was minimal at the center of the channel. Therefore, particles were directed

towards the center from all directions by way of negative DEP. The etched channel of 50 µm in

depth, 250 µm in width and 100 µm center-to-center distance between two adjacent electrodes

focused micro-beads and human leukemia HL-60 cells to regions 10-15 µm in diameter at 15 V

peak to peak at a frequency of 10 kHz.

Page 49: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

36

3.2.3 Micro Structure

Microfluidic channels can be fabricated with accurate dimensions and shapes. Recent advances

in micro/nanofluidics enable detailed understanding of fluid/particle transport which led to the

development of new techniques for self-ordering of biological particles without external forces.

Many researchers investigated micro-structures and their impact on fluid flow in a microfluidic

device to achieve the desired fluidic profile.

Choi et al. described a unique particle ordering principle called hydrophoresis that refers to the

movement of suspended particles under the influence of a microstructure induced pressure

field.115,116 The hydrophoretic ordering principle is governed by anisotropic obstacles, a kind of

the physical barrier. Upon application of a fluid flow into the channel, the anisotropic fluidic

resistance of the V-shaped obstacles generates rotational fluid streams117. Flow streams force

particles to migrate laterally and into the center of the microchannel. The streamlines starting at

the center move upward or downward in the z-direction along with the particles, and their

motions are determined by steric hindrance mechanism. The steric hindrance occurs when the

obstacles prevent rotational flows of large particles. A particle with a diameter that is similar to

the obstacle gap will steer its position toward the center of the z-axis due to the particle-wall

interaction. Therefore, the particle can be focused to the channel center and remain in its focused

position. The authors reported the sheathless focusing of 10 µm and 15 µm polystyrene particles

within the standard deviation of 22 µm and 18 µm in 1 mm wide channels. In addition, the

authors confirmed that the focusing effect was not affected by the flow rate in a range from 2 to 9

µL/min. It was discovered that the hydrophoretic focusing is dependent on particle size. The

smaller particles were found to have large focusing variation.

3.2.4 Micro Flow Cytometers without Focusing

In microfluidics based cytometry system, another approach is non-focusing flow in the detection

region. The target particles/cells will be moving inside a microfluidic device. Instead of being

interrogated one at a time, the particles/cells are probed by laser in a group. This approach

eliminates the complex fluidic controls that a conventional flow cytometer entails, and makes the

optical alignment less critical for the target fluorescent excitation and detection. However, the

optical signal to background ratio is compromised and the forward and side scattering signals are

lost in the measurement, yielding effectively an imaging flow cytometer system. Given the goal

Page 50: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

37

of this work being development of a portable and affordable cell analyzer, designing a functional

cell analyzer with simplest system architecture and specification is desired.

3.3 Microfluidics Design Concept

Both passive and active fluidic actuation approaches were investigated in this thesis. In passive

microfluidics, sample transport was driven by the capillary forces generated between the liquid

and micro-channel structures. This approach further simplifies the system design and power

requirements on the overall system. Since there is no particle focusing or sheath flow required,

the main objective of microfluidics system is to transport fluid sample through the detection

region by capillary forces only. The key design considerations of the microfluidic structures in

this approach include uniform fluid flow, metering of sample processed and channel filling

pattern and rate.

In addition, active fluid transport was also studied. A pressure based fluidic actuation system was

developed to control the fluidic motion on-chip. In this approach, a pneumatic interface between

the actuation hardware and microfluidic device was designed. By varying the volume of the

microfluidic device, a pressure difference was created inside the microfluidic channels which

was the dominating force in driving the motion of liquid sample.

Note in a fluidic system, both of these mechanisms, capillary and pressure based, are present and

affecting the flow characteristics. Depending on the application and the design requirements, one

phenomena can be suppressed by tweaking the design parameters to achieve the desired

outcomes. The following sections describe the capillary and pressure based microfluidic

transport systems developed in this thesis. Strategies on how to manipulate, actuate fluid flow, as

well as parameters that can effectively change fluidic flow properties are discussed in this

chapter.

3.4 Capillary Microfluidic Systems

Page 51: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

38

The physical laws describing, for example, the forces produced by inertia and interfacial tensions

are valid at any scale, but the relative magnitude of these forces reverses as the volume of the

liquid decreases and the surface/volume ratio increases. Solid-liquid-air interfacial phenomena

can be exploited and harnessed for controlling, guiding, and affecting the transport of liquids.

3.4.1 Governing equations of Fluid Mechanics

The flow of fluid through a control volume can be described by the complete Navier-Stokes

equations. These equations can be derived from the principles of conservation of mass,

momentum and energy. The conservation of mass equation states that at all times (for

incompressible, steady flow) the mass entering the control volume is equal to the mass leaving

the control volume,

𝜕𝑚

𝜕𝑡+ ∇ ∙ (𝜌𝑉) = 0 (3 – 4)

where m denotes the mass and ρ, v denote density and volume respectively. When liquid

penetrates into the capillary tube by capillary action at the gas-air interface, the fluidic motion is

driven completely by the capillary action. This is known as the capillary driven flow. In the last

two decades, extensive studies have addressed capillary force as the primary driving force

instead of electrical or mechanical means.118-121

Page 52: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

39

Figure 3-1 - Schematic of liquid plug in a rectangular microfluidic channel. 120

To accurately calculate the capillary force in a microfluidic channel, it is necessary to obtain the

shape of the liquid/air interface. Although there is some work on calculating the interface shape

in rectangular microchannels,122-124 it is difficult to estimate the exact interface shape and

capillary force for microchannel with general cross sectional shape. To simplify the analysis, the

pressure difference at the interface in a rectangular microchannel can be described as the

following by assuming a constant interface curvature and constant contact angle on the channel

inner surfaces, as shown in Figure 3-1.120

∆𝑃 = 𝜎 (1

𝑅𝑤+

1

𝑅ℎ) (3 – 5)

Where 𝜎 is the surface tension, 𝑅𝑤, and 𝑅ℎ are radii of curvature in y- (width) and z- (height)

directions, respectively. Applying the following relations between curvature, channel size, and

contact angle, where

𝑅𝑤 =𝑤

2 cos 𝜃, 𝑅ℎ =

2 cos 𝜃 (3 – 6)

The total capillary force, Fc, can be expressed as120

Page 53: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

40

𝐹𝑐 = ∆𝑃𝑤ℎ = 2𝜎 cos 𝜃 (1

𝑤+

1

ℎ) 𝑤ℎ = 2𝜎 cos 𝜃 (𝑤 + ℎ) (3 – 7)

This approximation provides an analytical calculation on capillary forces experienced by the

liquid sample in a rectangular microchannel. It can be used to balance the capillary and fluidic

resistance when designing a capillary microfluidic system.

3.4.2 Numerical Modeling

To assist with the microfluidic chip design, a numerical model was constructed to predict the

fluid flow in the capillary system. A computational model for free surface flow that can

accommodate the presence of obstacles in the flow was developed by Simulent and used in this

work.125 This model was developed to provide an analytical model and to aid the design of the

capillary microfluidic devices for the cytometry application.

3.4.2.1 Objective

The objective of this phase of the project is to model the flow of liquid in a simple microchannel

geometry, as shown in Figure 3-2, to determine the feasibility of using the numerical modeling.

The first reservoir will be filled with liquid and due to the capillary forces, the liquid will move

through the microchannel to reach the second reservoir. The simulation can be used to determine

the effect of parameters such as the geometry of the channel (size, shape) and the properties of

the liquid (contact angle, surface tension) on the time required for the liquid to reach from one

side to the other side.

Figure 3-2 – Schematic illustration of two reservoirs and a straight microchannel connecting the two

reservoirs.

Page 54: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

41

The reservoirs are 1.58 mm diameter by 1 mm height cylinders and are connected together

through a micro-channel of 15 microns high, 200 microns wide, and 15 mm long.

A single-phase algorithm was used to calculate the time required for liquid to flow from one

reservoir to another. One feature of the micro-channel geometry is that the maximum aspect ratio

is 1,000 to 1. This means for a good resolution, the number of nodes may be excessive. A rough

estimate shows at least that more that 15 million cells are needed to capture the simulation

domain. For this reason, some simplifications must be considered to lower the simulation time at

the same time to capture the capillary effect of the blood movement.

Simulations were run using a software tool developed by Dr. Hamideh Parizi at Simulent. Three

simulations with different mesh cell numbers were performed to find the effect of the cell sizes

on the accuracy of the simulations. The purpose of this simulation was to confirm and validate

the experimental results and observations. Hence only three mesh sizes were chosen in this

study. The results would also demonstrate how efficient the Simulent code was for such

problems and, if needed, what type, or types, of modifications it would require to make it faster

and more efficient.

Since surface tension is the dominant parameter in microfluidics, modelling surface tension and

contact angle play a very important role in all of the applications. A computational model for free

surface flows that can accommodate the presence of obstacles in the flow is developed by

Simulent and adapted for this work. The specific attributes of this numerical model are listed as

follows:

The numerical model uses a software tool for free surface flows and interfaces with no-slip

boundary conditions. Because of the dominance and effectiveness of surface tension force

applied in the code, it is very accurate and efficient for microfluidic analysis and it would be a

powerful tool for designing and prototyping new biochips and components. This mathematical

model has been extensively validated against experiments and has been already tested to model a

moving droplet in a microchannel, as a result of applying electro-capillary forces 125.

Page 55: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

42

3.4.2.2 Simulation Results

In order to capture the capillary flow in the microchannel, we need to have enough number of

cells in the smallest dimension of the channel, which is 15 microns. On the other hand, the height

of liquid column in the reservoir is at least 500 microns and the length of the channel is 15 mm.

To reduce to total number of cells in the whole calculation domain, it was decided that only part

of the reservoir is considered in the calculation domain and it is assumed that the height of liquid

remains constant. (Please refer to Figure 3-2 for details). Also, since the width of the channel (w)

is much larger that its height (h) the two boundaries at the “y” direction were considered to be

symmetric, with minimum number of cells. In addition since the flow velocity is very low and in

order to reduce the number of cells further, the simulations were performed only for one

millimeter of the microchannel,( L = 1 mm).

Figure 3-3 – Schematic of microfluidic setup in the numerical model.

Page 56: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

43

Figure 3-4 – Snapshots of flow in the microchannel at different times. The contact angle for this simulation is

set at 70 degrees. The length of the channel shown in this figure is 1 mm.

These simulations were used to confirm and validate the experimental observations and results.

As a result, only three different simulations were performed. To see the effect of the mesh size

on the simulation results, two different cell sizes, i.e., 3.5 and 2.5 microns were used. The total

number of mesh cells in each case was 264,552 and 681,952, respectively.

To characterize the effect of the contact angle, the simulations were performed at two contact

angles of 44.3 degrees and 70 degrees, respectively. These two contact angles were chosen to

model the material property of acrylic substrate such as PMMA. In Figure 3-4, snapshots of

simulation results at different time for a contact angle of 70 degrees are shown. In the last frame,

Page 57: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

44

the maximum distance that the liquid has been able to travel is shown, which is less than one

millimeter.

Due to the limitation of computer memory, the length of channel must be kept short. In Figure

3-5, the simulation results for the above three case are shown. This Figure shows the variation of

the flow velocity with time. As expected the velocity rises to its maximum at the very beginning

of the liquid movement and then drops as the liquid flows more into the channel. Different

curves represent various contact angles and simulation conditions (small or large mesh size). The

solid lines were plotted from discrete outputs from numerical simulation while the dashed lines

were logarithmic fit of the simulated discrete data points.

Figure 3-5 – Calculated axial velocity of liquid in the microchannel with respect to time.

Page 58: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

45

Clearly, the effect of mesh cell size in this range is not very significant. However, the effect of

the contact angle is more remarkable. At contact angle of 70 degrees, the flow has stopped in the

channel after about 0.04 second. This has been also shown from analytical investigation.

Figure 3-6 – Axial velocity of liquid in the microchannel with respect to time. The cell size is 1.5µm (10 cells

per height of channel). The contact angle is 30 degrees. All other conditions are the same as those in Figure

3-5.

One important observation is the oscillation of the flow velocity at the very beginning of the

capillary flow. To investigate the effect of the cell size and to eliminate the numerical error,

another simulation was performed with cell size of 1.5 microns (10 cells within the height of

channel). The preliminary results for a contact angle of 30 degrees are shown in Figure 3-6.

There are several important issues reported in the literatures that will affect the flow of liquid

in the microchannel:

Page 59: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

46

The aspect ratio of the channel, height to width ratio, has great effect on the behaviour

of the liquid. In general, the maximum center line velocity decreases with decreasing

channel aspect ratio 126.

Contact angle has a great effect on the velocity of the liquid in the microchannel. The

greater the contact angle (less wettability), the lower the velocity. In present case in

which there is only 500-1,000 microns of liquid column in the reservoir, the capillary

forces are more remarkable than the pressure force. This makes the effect of contact

angle more important.

For the above reason, the effect of gravity may also be neglected. Due to very low liquid

velocity, the use of the equilibrium contact angle instead of the dynamic contact angle

must be investigated in more details.

3.4.3 Fluidic Resistance Calculation

In a capillary system, the fluidic resistance is dependent on two factors: cross sectional area of

the microchannel and the fluid viscosity. Extensive research efforts have been put into providing

analytical models to predict fluidic resistance of various types of microfluidic channels.

The flow rate Q of a liquid plug in a capillary fluidic system is determined by the wettability of

the microchannels, the viscosity of the liquid, the total flow resistance and the capillary pressure

in the capillary system127:

𝑄 =1

𝜂

Δ𝑃

𝑅𝑓=

Δ𝑃

𝑅 (3 – 8)

Where 𝜂 is the viscosity of the liquid, Δ𝑃 is the difference in pressure inside and in front of the

liquid, and 𝑅𝑓 is the friction factor and 𝑅 is the hydraulic fluidic resistance of the system.

Page 60: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

47

Figure 3-7 – An example of a capillary microfluidic device. (a) Top view of a capillary microfluidic device

with a circular sample inlet, serpentine microchannel and a tapering structure to enhance capillary force. (b)

Side view of the rectangular microfluidic channel.

In a capillary microfluidic system shown in Figure 3-7, the capillary pressure Pc of a liquid-air

meniscus in a channel with rectangular cross-sectional profile can be expressed as the

following127:

𝑃𝑐 = −𝛾 (cos 𝛼𝑏+cos 𝛼𝑡

𝑎+

cos 𝛼𝑙+cos 𝛼𝑟

𝑏) (3 – 9)

Where 𝛾 is the surface tension of the liquid, 𝛼𝑡,𝑏,𝑙,𝑟 are the contact angles of the liquid on the top,

bottom, left and right side wall, respectively, a and b are the depth and width of the

microchannel.

The flow resistance of the rectangular microchannel shown in Figure 3-7 can be expressed with a

Fourier Series and can be approximated by a linear term:3

Page 61: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

48

𝑅𝐹 = [1

12(1 +

5

6

𝑎

𝑏)

𝑎𝑏𝑅𝐻2

𝐿]

−1

(3 – 10)

When the condition of a < b is met. In the above equation, L is the length of the microchannel,

and 𝑅𝐻 denotes the hydraulic radius of the microchannel, which can be further expressed as the

following:

𝑅𝐻 =2𝐴

𝑃=

𝑎𝑏

𝑎+𝑏 (3 – 11)

Where P being the perimeter and A is the area of the cross section of the microfluidic channel.

Similar to fluid dynamics in macroscopic application, the flow in a microchannel can be

approximated by capillary pressure divided by the fluidic resistance, which continually increases

as the channel is being filled under the influence of capillary force. The filling of microchannels

is determined by the surface tension of the liquid, and the chemistry and geometry of the micro-

structures in a microfluidic system.

3.4.4 Microchannel Design

A typical lymphocyte has a size of 6 to 8 µm in diameter. To improve sensitivity, a mono layer

of cells or particles distribution is desired during detection. On the other hand, according to the

equations described in Section 3.4.1, the fluidic resistance of the microfluidic system is strongly

dependent on the channel cross section. A shallow channel, on the order of 10 µm, with a width

of 600 µm and 1 mm in length, has fluidic resistance of 2.04x1015 m-3. If the channel is 100 µm

deep while all other parameters remain the same, the fluidic resistance becomes 2.39x1013 m-3. A

10 fold increase in channel depth resulted 100 fold decrease in fluidic resistance. Small channel

depth of less than 10 µm also increases the likelihood of microchannel clogging during

flow2,4,128,129 as blood samples often contain lipids and other molecules in addition to the

cells.6,130,131

For the dynamic imaging approach proposed and developed in this work, the microfluidic

channels must match with the active area of the imaging sensor in order to capture all the

cells/particles in a sample. To ensure the entire cross-section of the microchannel at detection

completely lies within the image sensor field of view, the microchannels must have a width of

500 – 900 µm with a depth of ~ 20 – 50 µm at detection region.

Page 62: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

49

To characterize and experimentally investigate flow properties of the capillary microfluidic

systems, testing structure shown in Figure 3-3 was designed and fabricated. This prototype

enables further understanding of the impact of fluidic channel geometry on the capillary flow.

3.4.5 Capillary Microfluidic Device Fabrication

The microfluidic chips were fabricated using standard photolithographic technique and the

wafers were packaged using a laminar press process, shown graphically in Figure 3-8. The base

layer, made of plastic acrylic, has fluidic structures defined in SU-8 negative photoresist

(Microchem Corp.) The microfluidic channels were patterned using a standard photolithography

technique – first an underlayer is deposited (spin coated then dried) and fully cured, then a

second layer is deposited in the same way and patterned by exposing through a photo mask.

Exposed samples were baked and developed to form the desired features. The lid or capping

layer, also made of plastic acrylic, has a partially cured SU-8 photoresist layer deposited with

mechanically drilled through holes to form the inlets and outlets. The base and the capping layers

are then assembled in registration in bonding jogs and heated under mechanical load in a

laminating press and held for a period of time to form the bond. The fabrication and packaging

were completed by Epigem, UK (http://www.epigem.co.uk).

Page 63: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

50

Figure 3-8 - Graphical illustration of photolithography process used by Epigem to produce capillary

microfluidic devices.

3.4.6 Capillary Microfluidic Device Characterization

The microfluidic device was characterized in two approaches: fluidic flow characterization and

cell/particle counting characterization.

3.4.6.1 Fluid Transport

The microfluidic devices were designed based on capillary fluidic properties described

previously. Once fabricated using the process described in Section 3.4.5, the chips were first

characterized to determine their fluidic flow properties. The fluidic speed characterization chip

had a serpentine design shown in Figure 3-9.

To test the fluidic flow in the capillary system, colored food dye samples mixed with distilled

water were used to enhance the visualization. A blue dye was used in this case to characterize the

fluidic motion inside the microfluidic channels.

Page 64: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

51

Figure 3-9 – Serpentine microfluidic structure for fluidic flow characterization. The channels were designed

with the same cross-sectional dimensions as the detection channels in the cell/particle counting device.

Channel length of each pass is 20 mm.

In a typical capillary microfluidic device, the flow resistance is dependent on the cross-sectional

area, the viscosity of the fluid moving inside the channel and the length of the channel. The

larger the cross-sectional area, the smaller the fluidic resistance seen by the liquid. The fluidic

channel has a cross sectional area of 800 x 20 µm and each pass is 20 mm long. The width of the

channel confines with the width of the optical detector, whereas the depth of the channel is

limited by the target cell dimensions. Ideally during detection a uniform, a single layer

distribution of cells inside the microfluidic device is desired. This eliminates potential errors that

may result in cell detection and tracking due to cells stacking and also minimizes the background

noise of blood plasma and other cell populations. On the other hand, if the channel depth is too

narrow, the cross-sectional area is reduced and fluidic resistance increases accordingly, thus

reducing the fluidic flow rate of the entire system.

Page 65: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

52

Figure 3-10 – Experimental results on filling characterization of the microfluidic device corresponding to the

serpentine microfluidic structure shown in Fig. 4. (a) Observed filling time of each linear section of the

serpentine microfluidic device. (b) Fluid flow speed and channel filling distance characterization results based

on the filling time and the distance for each pass in the serpentine microfluidic device. An inversely

proportional relationship between flow speed and the channel length can be seen.

Figure 3-10 demonstrates the flow properties of the colored dye when the sample is introduced in

the PMMA device. As expected in the fluidic simulation, the flow speed decreases as the liquid

propagates inside the microchannel when only exposed to capillary forces.

From the microfluidic channel dimensions and filling time of each linear section of the

serpentine channel, the fluidic velocity can be calculated along the entire microfluidic device. It

is known that the flow rate of a liquid in a capillary fluidic system is determined by the viscosity

of the liquid, the total flow resistance and the capillary pressure of the capillary pump127,132.

Since the serpentine channel has the same capillary pressure throughout the whole channel due to

constant surface wettability and cross sectional area, the flow rate of a liquid in the channel will

Page 66: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

53

be inversely proportional to the distance between entrance and the liquid filling front namely the

total flow resistance. The experimental results on filing characterization of the device are shown

in Figure 3-10. Because the particles/cells are imaged dynamically inside the microchannel and

fluidic motion is driven purely by capillary force, this investigation enables the validation of

capillary fluid system at experimental conditions. It also guides the design of the cell

enumeration device and the selection of detection window along the channel for the final design

so that a desired particle moving speed can be achieved during image capture; hence achieving

the required signal to background ratio of the captured images.

The result of this fluid flow characterization enabled the refinement of the capillary microfluidic

devices for cell counting. From this exercise, the length of the leading microfluidic channel can

be determined to yield the optimal fluid flow speed at the interrogation region. The PMMA

microfluidic chip was then used to characterize the fluidic flow.

3.4.6.2 Volume Metering

For many applications such as cell or particle enumeration, sample volume that has been

analyzed must be measured in order to produce an accurate volumetric concentration result. As a

result, the microfluidic cartridge must be designed to be able to calculate the exact volume of

sample that was imaged or processed.

Figure 3-11 - Capillary microfluidic chip design layout. This design relies on capillary forces to manipulate

sample flow. The device has a volumetric design to allow a sample volume of 2 µL to be processed.

In the passive microfluidic device designed in this thesis, the sample chamber is designed to have

a pre-determined overall volume. Figure 3-11 illustrates this design principle. The liquid sample

flow past the detection point will be imaged and analyzed. Hence, the volume downstream of the

detection point must be precisely set to a known value. Using this information, the cell or particle

concentration can be calculated based on the total number of positive events captured or

Page 67: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

54

detected. In the case of CD4 T cell analysis, to produce a statistically accurate result, a minimum

of 1000 events must be acquired133,134. Since the CD4 T cell concentration varies from 500 to

1500 per microliter, a sample volume of 2 µL must be analyzed in order to yield an accurate cell

counting result. The microfluidic device shown in Figure 3-11 has a chamber size of exactly 2

µL so that when the chamber is completely filled, the system has processed 2 µL of sample. A

capillary stop valve135 was designed at the end of exiting microchannel (shown in Figure 3-11).

Once the chamber if filled and the liquid front reaches the capillary valve, the flow will stop, due

to capillary force and surface tension, which signals the end of the entire analysis.

3.5 Conclusion

In this chapter, we investigated capillary-driven fluidic transport mechanism. Capillary driven

flow is a passive technique where no external forces are required to manipulate the sample fluids

on-chip. This approach is strongly dependent on material property, more specifically surface

contact angle with the liquid. The smaller the contact angle, the higher wettability of the material

which translate to larger capillary forces. In order to optimize or enhance the fluidic flow on-

chip, surface treatment such as oxygen plasma is often required.

A detailed analysis on capillary-driven fluid flow was conducted in this work. The flow

characteristics were investigated and strategies to control the flow speed were presented in this

chapter. The on-chip fluidic management is a key component in the disposable cartridge that

completes the cell enumeration analysis. The knowledge gained in this part will be utilized in

developing the point-of-care cell analysis device as described in the subsequent chapters.

Page 68: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

55

Chapter 4 Active Microfluidic System

In this chapter, the active microfluidic system is studied. A volumetric pressure based pneumatic

actuation concept is designed and developed in this part of the thesis. This approach is more

suitable for complex on-chip operations such as mixing, incubation and other similar chemical

reactions which require precise control of fluid flow.

4.1 Introduction

Capillary microfluidic devices offer the advantage of simple design since the fluidic actuation is

passive and no active components are required. The elimination of external energy sources

allows for reduction in the overall system complexity. However, the disadvantage of capillary

pumping in a microfluidic system is the strong dependency on surface tension between the liquid

and the channel walls. The surface tension is a function of the contact angle between the liquid

and material. Hence the hydrophobicity, also known as wettability, of the material is critical in

determining the fluidic transport inside the microchannel. For most applications, a hydrophilic

surface is desired to enhance the fluidic flow. To achieve that, it often requires surface treatment

of the material. This surface treatment could be a plasma oxygen treatment to increase the water

contact angle and improve the wettability of the surface. However, for mass production of

microfluidic devices and cartridges, this process is expensive and it is difficult to achieve

consistent fluidic transport. In addition, more complex operations such as mixing and incubation

require more consistent fluidic transport properties. Capillary force alone may not be sufficient

for these types of functions.

To overcome this drawback with capillary microfluidic systems, an alternate fluidic transport

approach was also investigated in this work. The alternative uses a volumetric pressure based

actuation approach. In this thesis, a soft elastomer – bellows – based pneumatic interface was

designed to generate the pressure change for fluidic transport.

Page 69: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

56

The first section of this chapter describes the design principle of the active microfluidic system.

Microfluidic devices were fabricated and tested. Additional operations and functions for sample

preparation was also developed and characterized in this chapter, followed by a conclusion.

4.2 Bellows Actuation System

In collaboration with thinXXS Microtechnology AG (www.thinxxs.com), an alternative fluidic

actuation mechanism was developed to mechanically control volume change in the fluidic

system to actuate sample flow. Figure 4-1 illustrates the basic concept of the pumping

mechanism. In this approach, an elastic film, or bellows that contains certain amount of pumping

volume integrated with the cartridge. During actuation, the elastic film of the bellows deflects

under external mechanical force, leading to a volume change which subsequently changes the

pressure in the microfluidic channel for fluidic transport.

Figure 4-1 – Bellows transport concept. The soft elastomer is depressed under external force F. The deflection

of the elastomer induces a pressure change inside the chamber and actuates the fluidic motion inside the

microchannel that is connected to the chamber.

Page 70: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

57

4.3 Bellows Transport System Design

The bellows transport system has two modes of operation: forward pumping which pushes the

liquid move the sample forward and reverse suction which retracts the sample backward.

To design the appropriate bellows transport system, a key parameter must be specified to ensure

proper control of fluidic motion in the microfluidic channel, namely: the total sample volume

that the bellows can transport.

Figure 4-2 - Bellows slide concept. The entire device consists of a soft elastomer semi-sphere, bonded to a

plastic fluidic chip. The fluidic channels are connected to the bellows. When bellows is depressed, the

reduction in volume inside the bellows increases the pressure inside the microchannel which subsequently

pushes the liquid sample forward.

In order to estimate the internal bellows volume needed, the following calculation steps are

applied:

1. Calculate the back-end gas spring pressure required to transport the sample trough the mixing

and detection zone depending on the size of the gas spring.

2. Calculate the front-end volume change of the air in the bellows vs. the internal bellows

volume for the pressure range selected.

Page 71: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

58

3. Calculate the motion of a stepper motor actuation deflecting the pneumatic bellows depending

on the bellows volume and the diameter of the actuator in order to get an indication for the

pulsation of the sample flow through the detection zone.

4.3.1 Bellows Actuation Volume Calculation

The first design parameter is the bellows volume. The deflection of bellows membrane

compresses the air inside the bellows chamber, hence increasing the pressure inside. The

increased pressure will subsequently push the liquid plug to move forward in the direction shown

in Figure 4-1. As a result, the relationship between bellows membrane deflection and pressure

change inside the bellows chamber must be established. The following calculation establishes the

change in bellows chamber volume as a function of bellows deflection.

Figure 4-3 – A graphical illustration of the coordinate system and variables used in bellows volume change

calculation.

The volume of the semi sphere shown in Figure 4-3 can be calculated from the following

equation:

Page 72: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

59

𝑉 = ∫ 𝜋𝑥2𝑟

0𝑑𝑧 (4 – 1)

Figure 4-4 – Volume change induced by bellows deflection. Bellows deflection is actuated from the top of the

semi-sphere in this diagram and the amount of deflection is denoted by d.

When the bellows is deflected, the integral is changed to the following equation to represent the

volume inside the bellows after deflection. (Also illustrated in Figure 4-4).

∫ 𝜋𝜌2𝑟 cos 𝜃

0𝑑𝑧 (4 – 2)

Applying the Pythagorean Theorem,

𝜌2 + 𝑧2 = 𝑟2

Or 𝜌2 = 𝑟2 − 𝑧2 (4 – 3)

Substitute Equation (4 – 3) in Equation (4 – 2), the volume of the deflected bellows can be

calculated using the following:

𝑉 =1

2∫ 𝜋(𝑟2 − 𝑧2)𝑑𝑧

𝑟 cos 𝜃

−𝑟 cos 𝜃 (4 – 4)

Page 73: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

60

Integrating gives:

𝑉 =1

2𝜋 [𝑟2𝑧 −

𝑧

3

3

]−𝑟 cos 𝜃

𝑟 cos 𝜃

Resulting in the following expression for the volume:

𝑉 = 𝜋𝑟3 (cos 𝜃 −1

3cos3 𝜃) (4 – 5)

Where:

𝜃 = cos−1 (𝑟−𝑑

𝑟) (4 – 6)

Where V is the volume of air inside the bellows, r is the radius of the bellows and d is the amount

of deflection applied to the bellows. Results of this analysis are illustrated in Figure 4-5 below

where the volume change incurred as a result of bellows deflection is plotted.

Figure 4-5 - Volume change of the bellows as a function of bellows deflection.

0

200

400

600

800

1000

1200

0 1 2 3 4 5 6

Vo

lum

e ch

ange

L)

Bellows deflection d (mm)

Volume Change as a function of Bellows Deflection

r = 5 mm

r = 10 mm

r = 15 mm

Page 74: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

61

From this analysis, it is clear under the same bellows deflection displacement, the larger bellows

will result in a larger volume change inside the microfluidic system, hence more samples

transported through the microchannel. On the other hand, since it produces smaller volume

change, a smaller bellows would have more precise control of fluidic motion inside the

microchannel per unit deflection of the bellows.

4.4 Bellows Actuation System and Microfluidics Design

The bellows slide, fabricated using injection molding process by thinXXS Microtechnology AG,

comprised two parts: a soft elastomer membrane and a hard-plastic substrate. The schematic of

this mechanical structure is shown in Figure 4-2.

The bellows deflection is driven by linear stepper motor shown in Figure 4-6. Linear stepper

motors have a compact foot print with low power consumption. They also offer very fine and

well controlled displacement, as small as 2 µm per step, which makes it ideal for microfluidic

application. An actuation pin can be installed at the tip of the shaft shown in the Figure 4-13 on

Page 73 to deflect the bellows membrane to drive the fluidic motion inside the microchannel.

Figure 4-6 - Linear stepper motor from Haydon Kerk. (www.haydonkerkexpress.com)

According to the analysis completed, to generate the same pressure change, a smaller bellows

size requires a smaller bellows deflection. In addition, a smaller actuation pin diameter requires a

bigger displacement to deflect the spherical bellows. The size and height of the deflection defines

the motor motion and therefore the number of motor steps.

Page 75: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

62

Figure 4-7 - Microfluidic chip layout of the bellows slide. The bellows slide was fabricated using injection

molding and it was used to test the fluidic actuation and on-chip flow control.

For the selected range of bellows and actuation pins, the number of 2 µm motor steps per µl

ranges from 15 to 30 mm (this value would double in case of 1 µm motor resolution). Assuming

the flow rate through the microfluidic system is set to a target of 0.2 µl/min, this results into 5

steps/min (10 steps in case of 1 µm displacement per step), which is assessed to be critical in

terms of pulsation. In general, the design has to reflect a compromise between the tendency to

keep all elements as small as possible (due to cost) and allowing enough motor steps to transport

the sample through the detection zone at the required flow rate. Figure 4-7 is a picture of the

microfluidic chip layout of the bellows slide that was used to test and evaluate the on-chip fluidic

management and actuation.

4.4.1 Material Characterization

The active microfluidic devices were fabricated using injection molding process, as it is the

preferred method in mass production for plastic medical consumables. Since the fluidic

component of the cartridge is comprised of two parts: microfluidic channels fabricated in a

plastic substrate and a thin covering film. Both material must be characterized. Two most

commonly used thermo plastic material that can be used include polymethyl methacrylate

(PMMA) and cyclic olefin copolymer (COC).

Page 76: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

63

PMMA is a transparent thermoplastic often used in sheet form. It is a clear, colorless polymer

with high mechanical strength, high Young’s modulus and low elongation at break. It has

excellent optical quality and is shatter-resistant. It is one of the hardest thermoplastics and is also

highly scratch resistant. It exhibits low moisture and water absorbing capacity. COC is an

amorphous polymer material that is widely used in medical device industry. It has excellent

optical transmission, high purity, and resistant to moisture. It is also resistant to chemicals as it is

becoming more and more important in microfluidics manufacturing.136,137

Since optical detection of target cells is used by measuring their fluorescence intensities, the

optical properties of the material used to fabricate the chip, specifically auto fluorescence,

becomes a critical evaluation parameter. A range of different PMMA and COC polymer material

were characterized by measuring their auto fluorescence in the 400 nm to 750 nm range to

determine the best material choice.

Page 77: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

64

Figure 4-8 - Autofluoscence levels of different PMMA and COC material under different excitation and

emission optical setup.

The material label represents different common injection molding plastic material that was

supplied by the injection molding foundry thinXXS Microtechnology (http://www.thinxxs.com/)

in Germany. Table 3 summarizes the name and physical characteristics of each material

investigated in this work. For each substrate material, a 3 inch by 1 inch slide was provided.

Auto fluorescence measurement was made on a standard fluorescence microscope (Olympus

BX51). Excitation filters were chosen to match potential fluorescence dye that may be used in

0

500

1000

1500

2000

2500

3000

3500

dark field COC BlackPMMA

COP PC PP OpaquePC

PMMA Black COC Black COP OpaquePC

Au

tofl

uo

resc

ence

inte

nsi

ty

Material type

Plastic material autofluorescence measured under different excitation wavelength

ex:387/11, em:440/40 ex:485/20, em:525/30

ex:560/25, ex:607/36 ex:650/13, em:684/24

Page 78: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

65

the detection system. Each data point was captured under 20 ms exposure time using a

Photometrics CoolSNAP HQ2 monochrome CCD camera (http://www.photometrics.com/) under

non-cooled operation mode. Dark field was a reference data point measured when no material

was present while all other excitation and emission setup were the same.

Table 3 - Materials used in autofluorescence characterization.

Label name Material full name Physical characteristics

COC Cyclic olefin copolymer Transparent polymer

PMMA Polymethyl methacrylate Transparent polymer

COP Cyclic olefin polymer Transparent polymer

PC Polycarbonate Transparent polymer

PP Polypropylene Transparent polymer

Opaque PC Polycarbonate White polycarbonate

Black PMMA Polymethyl methacrylate Black PMMA, non-transparent

Black COC Cyclic olefin copolymer Black COC, non-transparent

Black COP Cyclic olefin polymer Black COP, non-transparent

Opaque PP Polypropylene White PP, non-transparent

As expected, the shorter the excitation wavelength, the stronger the auto fluorescence. When

excited with longer wavelengths, the auto fluorescence of all these plastic material reduced

significantly. Of the material tested, black PMMA (see Figure 4-8) exhibited the least auto

fluorescence levels across all excitation wavelengths. Another transparent variant of PMMA,

also has minimal auto fluorescence level even when excited by 387 nm violet light source. The

experimental results suggest the 3 black materials have the lowest level of auto fluorescence (the

Page 79: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

66

level of auto fluorescence are COP < PMMA < PC as shown in Figure 4-8). Based on the

current results and considering material property compatibility, black PMMA would be the ideal

material choice for the microfluidic device.

4.4.2 Microfluidic Channel Design

The microchannels used for fluid transport have a width of 800 microns and a depth of 400

microns. The width of the microchannel at detection zone was designed to be 200 microns to

match the size of the optical sensor.

Based on the analysis completed, a microfluidic device using bellows actuation was designed

that can meet the sample transport requirement for CD4 T cell enumeration. Since typical healthy

individual’s CD4 T cell concentration ranges from 700 to 1000 per microliter, according to

Poisson statistics133,138, the counting outcome is accurate only if 1000 events are encountered and

detected. That means a minimum of 2 µL sample must be analyzed, or processed. Typically, in

conventional flow cytometry, 100 µL of sample is prepared and analyzed. The larger volume can

make the sample preparation process less prone to statistical variations. On the other hand,

microfluidic systems are not well-suited for applications that require processing of large sample

volumes. To reduce the statistical uncertainty on the cell enumeration result, a larger volume (10

µL) is used for sample preparation while only a sub-set of the prepared sample (2 µL) is used for

image analysis to produce the final cell counting result.

4.4.3 Bellows Slide Fabrication

Microfluidic prototypes were fabricated using injection molding by thinXXS. Figure 4-9 is a

picture of the fabricated bellows slide using PMMA material as characterized in Section 4.4.1.

Page 80: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

67

Figure 4-9 - A photograph of the injection molded microfluidic bellows chip using the PMMA material

selected in previous section. The bellows chip is 1 inch wide and 3 inches long.

Injection molding is the most commonly used manufacturing process for producing parts by

injecting liquid material into a mold and allowing it to set. A wide variety of products are

manufactured using injection molding, which vary greatly in their size, complexity, and

application. Material for the part is fed into a heated barrel, mixed and forced into a molding

cavity. Once cooled, the material will harden and conforms to the shape of the cavity. 139-141

Injection molding is used to produce thin-walled plastic parts for a wide variety of applications,

including electronics, biomedical, automotive and industrial. Injection molding process is widely

used to manufacture parts and components with a variety of sizes from microscopic devices to

entire body panels of cars.

Parts to be injection molded must be very carefully designed to facilitate the molding process;

the material used for the part, the desired shape and features of the part, the material of the mold,

and the properties of the molding machine must all be taken into account. The versatility of

injection molding is facilitated by this breadth of design considerations and possibilities. It is the

most common modern method of manufacturing parts; it is ideal for producing high volumes of

the same object.142

Page 81: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

68

Adding to the capabilities of micro-molding is the ability to mold two different materials on the

same part at the same time. The two different thermoplastic resins are injected in synchronization

into the molding cavity so that it requires only one mold cycle.

4.4.3.1 Micro injection molding for microfluidics

To produce very small components, such as microfluidic devices where the critical features are

on the order of tens of micrometers, the injection molding process requires maximum possible

accuracy and precision. From the material and machine to the mold, everything must be

streamlined to this objective.

Micro-molding is defined as a very unique Injection Molding process requiring specialized

molding machine capable of delivering high injection speed, high injection pressure, precise shot

control, uniform melt temperature and ultra-fine resolution using servo-electric drives and

sophisticated controls.

4.4.4 Characterization

A bellows slide was designed according to the design principles described in previous sections.

The microfluidic device was fabricated using injection molding in PMMA to minimize auto

fluorescence. A thin PMMA film was used to cover the injection molded microfluidic channel

network. The bellows had a size of 15 mm diameter, which is sufficient to transport 50 µL of

sample on-chip.

4.4.4.1 Experimental Setup

A linear stepper motor was used to deflect the bellows membrane. A metal pin was placed at the

end of stepper motor shaft to push the bellows membrane. The experiment was executed using

the setup according to schematics shown in Figure 4-10.

Page 82: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

69

Figure 4-10 - Schematics of the stepper motor setup used to test and characterize on-chip fluidic actuation

4.4.4.1.1 Electronics

A universal motion controller (TMC1110) from Trinamic (www.trinamic.com) was used to

control the motion of the stepper motor. Figure 4-11 on Page 70 is a block diagram of the

electronics setup.

Page 83: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

70

Figure 4-11 - Fluidic control eletronics setup block diagram. This setup was used to experimentally

characterize and test the fluidic actuation using bellows concept.

A LabVIEW program was developed to interface the motion controller via USB. All hardware

was integrated in a mechanical structure to obtain a compact format as shown in Figure 4-13. A

screenshot of the LabVIEW program is shown in Figure 4-12. The motion controller can drive

the stepper motor in 256 microsteps per full step. This enabled precise control of bellows

deflection. The stepper motor can drive the shaft to move at a wide range of speeds from 0.16

µm/second to 0.341 mm/second. The wide range of speeds allows the fluidic actuation for

different application requirements. The motor can also move in both forward and backward

direction to drive the liquid sample back and forth inside the microfluidic channel for mixing and

incubation purposes, as described in Section 4.2 and Section 4.3.

Page 84: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

71

Figure 4-12 - LabView program interface. This tool was used to control the motion of the stepper motor for

on-chip fluidic actuation.

The following list is a detailed description of the functionality of the LabVIEW program on how

to control the stepper motor.

1. Communication port interfacing the motion controller circuit board

2. Electronic pulse initialization where the program will establish the initial configuration of

the electronic and stepper motor system.

3. Stopping the stepper motor.

4. This function drives the stepper motor forward until it touches the bellows. Then the

motor steps. The stepper motor moves at a pre-defined speed.

Page 85: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

72

5. This function drives the stepper motor backward until it reaches the original or home

position. The motion speed is pre-defined.

6. Drives the stepper motor forward

7. Drives the stepper motor backward.

8. Specify a given amount of stepper motor travel distance. The distance is defined in

micrometers.

9. Speed of the stepper motor movement. Default value is 200, which corresponds to a

linear speed of 2mm/min.

10. This function specifies direction of motion: forward or backward.

11. Motion starts once this button is pressed. The button returns to the original unpressed

state once the pre-defined displacement is reached.

4.4.4.1.2 Mechanics

Figure 4-15 is a picture that illustrates the mechanical setup of the bellows actuation and

controller. The linear actuator had a travel distance of 9 mm. According to calculation carried out

in Section 4.3.1 on Page 58, for bellows with a diameter of 15 mm, 3 mm deflection of bellows

membrane would induce approximately 50 µL volume change, while at full 9 mm motor travel it

can transport up to 200 µL of sample inside the microfluidic channel.

.

Page 86: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

73

Figure 4-13 - A picture showing the mechanical setup of the bellows actuation.

4.4.4.1.3 Testing Slide

To test the fluidic actuation and detection, a separate testing slide was designed and fabricated.

The testing slide included a sample inlet, an outlet, and a detection zone for optical interrogation.

The design of the device is shown in Figure 4-7 on page 62.

Page 87: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

74

Figure 4-14 - Schematics of microfluidic connections used in on-chip fluidic actuation testing and

characterization.

During fluidic transport testing, samples were introduced at the inlet. A flexible, plastic tube was

used to connect the bellows slide with the testing slide via fluidic connector. Figure 4-14 is a

schematic of the experimental setup of the microfluidic connections while Figure 4-15 are

pictures of the experiment setup.

Page 88: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

75

Figure 4-15 - Pictures of the bellows actuation experimental setup. (a) Side view of the motion control board,

stepper motor, bellows slide and re-suspension slide. (b) Top view of the experimental setup. The engineered

blood sample was introduced into the re-suspension slide and actuated back and forth inside the device.

4.4.4.1.4 Fluidic Transport Observations

Using experimental setup described, the fluidic transport properties of the bellows actuated

microfluidic system were characterized. The liquid test sample used was colored food dye mixed

with water. The following experimental observations were made:

The bellows slide was initially actuated at a linear speed of 16.7 µm/second. After the liquid

sample breaks into the microchannel, the speed was reduced to a linear speed of 0.167

µm/second. The narrow taper in the testing slide means a large increase in the fluidic resistance

in the system, as resistance is proportional to the cross-sectional dimension of the microchannel

as described in Section 3.4.3. Once the steady state is reached, the flow rate of liquid sample was

very stable at 0.615 (±0.016) μL/min. Figure 4-16 is a plot of the speed characterization on

fluidic transport measurement.

Page 89: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

76

The narrow detection region means large fluidic resistance. To overcome this resistance, there is

a pressure built-up inside the channel before liquid breaks into the detection channel. This means

there will be wasted or dead volume of sample during analysis. However, the high resistance is

desirable as it results more precise fluidic speed control. The large fluidic resistance makes the

system less responsive to the stepper motor actuation, as illustrated by the 200 seconds settling

time shown in Figure 4-16, resulting more precise fluidic actuation during analysis.

In the second part of the chip characterization, Immuno Trol, a standard flow cytometry control

sample, was used to test the fluidic actuation. The Immuno Trol sample has the same physical

property, such as particle density, viscosity, cell distribution as a real blood sample. The results

from this analysis will reveal how blood sample would behave inside the microchannels during

analysis.

Figure 4-16 on Page 76 is a graph on speed measurements completed with beads in immunotrol

blood when pumping the blood through the detection channel and then stop once the sample has

passed the detection zone, the fluid linear speed stabilized around the range of 0.2-0.8 mm/s after

the first 200 seconds.

Figure 4-16 - Fluidic linear flow speed measured as a function of time. Graph (a) is obtained with a bead

sample only while (b-d) were obtained with a sample of beads mixed with Immunotrols. Three different

Page 90: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

77

pumping conditions were tested in this experiment: the stepper motor pumping distance (2 µm, 10 µm, and 30

µm at a speed of 50).

Figure 4-16 showed a graph on speed measurements done with fluorescently labeled beads in an

Immunotrol blood sample. In this experiment, the fluorescent beads were mixed with the blood

sample and their individual speeds were measured using image analysis techniques. The x, y

positions of the particles were recorded in each frame. Since the frame rate is fixed, by

calculating the spatial distance between two subsequent frames, the linear speed of particle in the

sample plug can be deduced. After the sample breaks into the detection channel, the sample was

continuously pumped at a very slow speed. (Stepper motor was moving at a speed of 0.167

µm/s.) By actively control the blood, the settling time for fluidics to stabilize can be shortened to

less than 100 seconds.

Figure 4-17 – Fluidic linear speed plotted as a function of time in the detection microchannel using bellows

actuation. The measurement was made on the resuspension slide described earlier in this work.

0.00

2.00

4.00

6.00

8.00

10.00

12.00

0.0 100.0 200.0 300.0 400.0 500.0 600.0 700.0 800.0 900.0 1000.0

Flu

idic

lin

ear

spee

d (

mm

/s)

Time lapsed from sample entrance (seconds)

Fluidic flow speed measured in the microfluidic device using bellows actuation methodology

0415 bead blood flow rate20um@2000 followed by 4um@2 permin

0417 bead blood flow rate

Page 91: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

78

The same fluidic actuation experiment was also performed on wider microchannels with width

of 700 µm). Wider channels had less pressure build-up before the liquid entered the detection

channel area as shown in Figure 4-18. And the flow was easier to control and more responsive to

the operation of the bellows deflection. By repeatedly pushing the bellows by 2 µm at a pumping

speed of 0.167 µm/second, it is feasible to control the flow speed to be within 0.3-0.5 mm/second

range. The particle speed within the flow slowed down to the 0.3-0.5 mm/second range within

100 seconds. In addition, there was large fluctuation in particle speed due to the fact that

individual particles travel at different speeds even under the same actuation conditions.

Figure 4-18 – Fluidic speed plotted as a function of time for the wider 700 µm channel using bellows

actuation.

4.5 On-Chip Sample Preparation

To develop practical point-of-care devices, sample preparation is a key function that must be

designed and integrated in the microfluidic chip. For CD4 T cell enumeration, a single step

sample preparation of staining the blood sample is required. Other blood testing that involves

Page 92: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

79

probing of intracellular protein, or DNA molecules often require more complex and cumbersome

sample preparation procedure such as washing, cell lyse and amplification. This section

describes the design and development of the on-chip sample preparation sub-system that includes

reagent and reagent incorporation, reagent re-constitution and mixing functions for the cell

enumeration assay.

4.5.1 Reagents

The reagents required to complete a CD4 T cell absolute count are anti-human CD3+ antibody

and anti-human CD4+ antibody. Each antibody is labeled with a specific fluorescent dye to

enable optical detection. The dye PE-Cy5 is used to conjugate with CD3 antibody while PE is

used to conjugate with CD4 antibody. This arrangement enables the use of single wavelength to

excite both fluorescent molecules so that both the CD3 and CD4 cell populations can be counted

simultaneously. The fluorescently labeled antibodies were purchased from BioLegend

(www.biolegend.com), catalogue number 344605/344606 (PE anti-human CD4), and catalogue

number 300410 PE/Cy5 anti-human CD3.

4.5.2 Reagent Handling and Incorporation

A number of different approaches for the incorporation of the dry reagents into the microfluidic

cartridge have been investigated:

a) The application of the reagent directly in the microfluidic channel prior to bonding the chip

via liquid spotting; and drying of reagent after bonding

b) Drying of the reagent on a plastic plug, followed by the application of the plug on bonded chip

by a heat staking process (allows separate batch manufacturing of functionalization process and

cartridge manufacturing process)

c) Drying of reagent on a plastic disk, and followed by insertion of the disks in to the chip prior

to bonding and packaging of the chip

d) Drying of reagent as a pellet, and deposit the pellet inside the fluidic chip prior to bonding and

packaging

Page 93: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

80

A qualitative analysis was conducted to evaluate the different strategies of incorporating reagents

into the disposable cartridge. The following table summarizes the advantages and disadvantages

of each approach.

Table 4 - Qualitative analysis of five, reagent drying approaches investigated in this thesis.

Reagent

Drying

Direct in Channel Plug Carrier Disk Carrier Pellet Comments

Coated

material

Must be microfluidic

chip material;

potential surface

treatment may be

required and its

impact on bonding is

unknown.

Plug material

could be

different from

microfluidic

chip material

Disk material

could be

different

from

microfluidic

chip material

No media

required

Must be

compatible with

future assays

and

requirementsw

Drying

process

Tested and proven Tested and

proven with

sugar sample

Tested and

proven

Tested and

proven

Either freeze

drying or slow

drying

Integrati

on

Prior to microfluidic

chip bonding and

assembly

After bonding

of the

microfluidic

chip

Prior to

microfluidic

chip bonding

and assembly

Prior to

microfluidic

chip

bonding

and

assembly

Preferably

integration

completed

before bonding

Re-

suspensi

on

Need to verify Tested for

selected

reagents; need

verification on

Need

verification

Need

verification

Page 94: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

81

target

reagents

Handling No handling

required

Pick and place

handling

Difficult to

handle

Delicate

handling,

difficult to

automate

Since during bonding and assembly, the microfluidic chips are exposed to high temperatures ( >

65 degree Celsius), the reagent integration is best to be completed afterwards to prevent loss of

protein activity. Given the analysis conducted, it is clear that the reagent plug is the best

approach to introduce dried reagents to the microfluidic chip in terms of both handling and

preserving function.

4.5.3 Reagent Drying

Reagent drying was completed at Reametrix using their proprietary slow drying process, which

has received US Food and Drug Administration (FDA) approval. Both CD3 and CD4 antibody

reagents were dried on a plastic transfer plug

Figure 4-19 – 3D drawing of reagent plug used to handle and incorporate reagents into the microfluidic

cartridge.

A stock solution primarily comprised of sugar additives was used to protect the protein

molecular structure and chemical activity during the drying process. Five microliters of the

Page 95: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

82

antibody reagents were first dispensed on a plastic reagent plug, shown in Figure 4-19. The

reagent plug was then slow dried under room condition in a vacuum. Stock sugar solution was

also added to the reagent plug to preserve the chemical activity of the antibody. Figure 4-20 is a

picture of reagent plug after drying. The pink color exhibited by the dried pellet indicates a high

concentration of antibody.

Figure 4-20 - Reagent plug coated with dried fluorescently labelled CD4 antibodies. The pink color indicates

the high concentration of antibodies.

4.5.4 Re-suspension

The sample preparation component allows the on-chip dried reagents to be mixed with the blood

sample. This part of the microfluidic system must accomplish two main objectives: re-suspend

the dried powder reagents and conjugate of the target biomolecule with appropriate fluorescent

Page 96: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

83

tags. Several design concepts were proposed and the based on the functionality, one concept was

chosen and a prototype microfluidic device was fabricated and characterized.

4.5.5 Microfluidic Mixer

The mixing channel must first suspend the dried powder reagents in the blood sample. The re-

suspended reagents will then be mixed with the sample. After incubation, the stained sample will

be ready for detection. Typically, the same sample preparation process takes 30 minutes to

complete in a traditional lab setting under the standard flow cytometry protocol. Using a

microfluidic device, the reaction time is significantly reduced. The entire sample preparation: re-

suspension, mixing and incubation can be completed in less than 10 minutes.

The aim of a microfluidic mixer is to thoroughly mix multiple samples, or material in a

microchannel. In these devices, mixing is essentially achieved by enhancing the diffusion effect

between different samples. Typical microfluidic mixer can be divided into two categories:

passive mixer and active mixer. In passive mixer, capillary force driven diffusion is the primary

driving force for mixing. Geometries and structures are often used to introduce turbulence in

otherwise laminar microfluidic flow. Common passive microfluidic mixers include split and

combine, T or Y shaped microchannels, and barriers or obstacles structure embedded in the

channels. Table 5 is a summary of passive microfluidic mixer that were studied in recent years in

literature143.

Table 5 - Performance of passive micromixers in recent development143.

Categories Mixing Technique Mixing Time (ms) Mixing Length (µm)

Lamination Wedged shaped

inlets144

1 1

90 rotation145 - -

Zigzag channels Elliptic-shape

barriers146

- 10,000

Folding structure147 489 -

Page 97: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

84

3D serpentine

structure

Creeping structure148 - -

Stacked shim

structure149

- -

Multiple splitting,

stretching and

recombining flows150

- -

Unbalanced driving

force151

- -

Embedded barriers SMX152 - -

Multidirectional

vortices153

- 4,255

Twisted channels Split-and-

recombine154

730 96,000

Surface chemistry Obstacle shape155 - 1,000

T-/Y- mixer156 - 1,000

On the other hand, an external energy source is applied to perturb the sample flow and enhance

mixing in the active mixers. In active mixing, an external force is used to enhance mixing, such

as mechanical, acoustic or thermal. Table 6 outlines the recent development in passive

microfluidic mixer143.

Page 98: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

85

Table 6 - Performance summary of active micromixers published in literature143.

Categories Mixing Technique Mixing Time

(ms)

Mixing Length

(µm)

Acoustic/Ultrasonic Acoustically driven sidewall-trapped

microbubbles157

120 650

Acoustic streaming induced by

surface acoustic wave158

600 10,000

Dielectrophoretic Chaotic advection based on Linked

Twisted Map159

- 1,000

Electrokinetic time-

pulsed

Chaotic electric fields 100 Width*5.0

Periodic electro-osmosis flow160 - 200

Electrohydrodynamic

force

Staggered herringbone structure161 - 825

Staggered herringbone structure162 - 2300

Thermal actuation Thermal163 - 6,000

Magneto-

hydrodynamic flow

High operating frequency164 1,100 500

Electrokinetic

instability

Low Reynolds number139 - 1,200

Low Reynolds number165 - 1,200

For a point-of-care device, the key design considerations are low power consumption and ease of

integration with the detection and sample introduction protocols. Robustness and reliability are

two other major design inputs as well. As a result, an active microfluidic mixer, shown in Figure

4-21 was designed and developed in this thesis to meet the desired requirements.

Page 99: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

86

Figure 4-21 - Microfluidic mixer design. The “wiggly” channels are mixing structures which utilizes Dean’s

flow to enhance mixing efficiency.

This mixer design relies on Dean’s flow to enhance the turbulence in the microchannels. This

improves the mixing efficiency to reduce the reaction time and device geometry. The sample

plug will be actuated and moves back and forth around the center point similar to a pendulum,

where dried reagent is encapsulated via a plastic reagent plug. An external pneumatic pumping

system, as described in Section 4.2 actuates the sample plug movement in the microfluidic

channel. As the blood samples moves back and forth inside the microchannel, it re-suspends and

mixes with the dried reagent. The sample, along with reagent, moves through the serpentine

structure. This design utilizes Dean’s flow to introduce turbulence into the flow, hence

improving the mixing efficiency.160

4.5.5.1 Physics of Dean Flow

In microfluidic channels, since the dimensions of the channels are on the order of hundreds of

micrometers, the flow is typically laminar. This is beneficial for certain applications but is a

significant limitation for applications such as mixing. To have improved mixing results, turbulent

flow is required to allow thorough mixing of different liquid samples. Typically, diffusion is the

phenomena that was exploited in on-chip mixing in microfluidic systems. To further enhance the

Page 100: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

87

rate of mixing and ensure a thorough re-constitution of the dried reagent on-chip, other physical

phenomena were often utilized as well such as Dean’s flow166.

Figure 4-22 - Dean flow. In curved channels, when inertia is important, faster moving fluid near the channel

center tends to continue outward, and to conserve mass, more stagnant fluid near the walls re-circulates

inward. This creates two counter-rotating vortices perpendicular to the primary flow direction166

Secondary flow arises when a fluid flows through a curved channel because of a mismatch of the

velocity in the downstream direction between fluid in the center and near-wall regions of a

channel. Therefore, fluid elements near the channel centerline have larger inertia than fluid near

the channel walls, and would tend to flow outward around a curve, creating a pressure gradient in

the radial direction of the channel. Because the channel is enclosed, relatively stagnant fluid near

the walls re-circulates inward due to this centrifugal pressure gradient, creating two symmetric

vortices as shown in Figure 4-22. A dimensionless number that describes the magnitude of this

flow was first established by W. R. Dean167, and a more generally accepted form of this Dean

number is described by Berger et al168 as 𝜅 = (𝐻

2𝑅)

1

2𝑅𝑒, where H is the width of the curved

channel, R is the radius of the channel curvature and Re is the Reynolds number. Berger et al.

noted that the ratio of the channel dimension to the radius of curvature, defined by the parameter

𝛿 =𝐻

2𝑅, also has important effects on the shape of the secondary flow168. Following Squires and

Quake119 the secondary flow velocity scales as 𝑈𝐷~𝜅2 𝜇

𝜌𝐻. Besides giving a measure of the Dean

Page 101: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

88

flow velocity, increases in Dean number are associated with changes in shape of the secondary

flow, with the centers of the symmetric vortices moving towards the outer wall and development

of boundary layers with increasing κ.168

4.5.5.2 Applications of secondary flow in microfluidic systems

Secondary flows have been employed in microfluidic systems primarily for applications in fluid

mixing. Mixing in microfluidic systems has been extensively explored because of the difficulty

in quickly mixing fluid streams without the aid of turbulence.169,170

Most techniques are based on the concept of increasing the interfacial area for diffusive mixing

to occur. Often the concept of chaotic advection is used, whereby fluid interfaces are stretched

and folded to increase the interfacial area to an extreme level171. Because of the exponential

growth in stretching of fluid interfaces in these systems, the positions of individual fluid

elements cannot be confidently assigned, recapitulating an aspect of turbulent flow that leads to

good mixing.

Secondary flows in curved microfluidic channels have been used to increase the interfacial area

for diffusive mixing. One of the first examples is the use of three-dimensional ‘‘twisted’’

channels analogous to macro-scale systems that take advantage of chaotic advection172.

Effective mixing in microfluidic channels requires that fluids be manipulated to increase the

interfacial surface area between initially distinct fluid regions so that diffusion can complete the

mixing process in a reasonable time. Unfortunately, the rapid mixing produced by turbulent

flows is usually not available at the micro-scale because the Reynolds number (Re)173 is typically

below the critical value for transition to turbulence. Thus, some other mechanism must be

employed to enhance mixing.

It has been shown that a “twisted pipe” has the potential to enhance mixing even at low Reynolds

numbers174. This mixing enhancement is possible because of the phenomenon known as chaotic

advection175,176, in which simple regular velocity fields produce chaotic particle trajectories.

Dynamical systems theory shows that chaotic particle motion can occur when a velocity field is

either two-dimensional and time-dependent or three-dimensional (with or without time

dependence)170. The occurrence of chaotic advection typically indicates rapid distortion and

Page 102: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

89

elongation of material interfaces. This process significantly increases the area across which

diffusion occurs, which leads to rapid mixing.

Advantages of using secondary flow in curved channels for mixing include: (i) the relatively

simple design and operation and ease of fabrication, (ii) enhancement of mixing with increasing

flow rate, and (iii) applicability to a range of different fluids of varying viscosities, densities, and

conductivities. However, it should be noted that these techniques are often not appropriate for

many microfluidic lab-on-a-chip applications dealing with small volumes of fluid, since mixing

enhancement becomes negligible for lower flow rates where κ < 1.

In the mixer design shown in Figure 4-21, secondary flow in serpentine channels was utilized to

enhance mixing. Active fluidic control was also implemented to reduce the mixing time and

increase mixing rate. This approach enables rapid and effective, thorough mixing of blood

sample with dried reagents on-chip, which is a critical requirement for the point-of-care CD4 T

cell counting test.

4.5.5.3 Mixing Testing

A re-suspension microfluidic chip was designed to test and characterize the on-chip dried reagent

re-constitution and mixing. Figure 4-23 is a picture of the re-suspension slide designed and

fabricated using injection molding process. The microfluidic device was made from PMMA. The

channels are 800 µm wide and 400 µm deep. The circular hole in the middle of the reagent

chamber is where reagent plugs were inserted after drying.

Page 103: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

90

Figure 4-23 - Resuspension slide design layout. In this design, four different mixers were proposed to optimize

the mixing and fluidic motion in the microchannel. The circular holes are reagent plug chambers where plugs

are inserted.

The plastic COC plugs, coated with dried antibody reagents, are inserted and assembled with the

re-suspension microfluidic chips via a snap fit. Figure 4-24 illustrates an assembled chip. The

testing and characterization of re-suspension, mixing and incubation was completed using the

bellows actuation setup shown in Figure 4-15.

Page 104: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

91

Figure 4-24 - Fully assembled microfluidic re-suspension prototype slides with reagent plugs inserted. The re-

suspension prototype slides were used to characterize on-chip mixing and re-suspension of dried reagents.

The same experimental setup that was used in bellows actuation characterization was used in

mixing and re-suspension testing. Figure 4-14 on Page 74 illustrates the block diagram of the

fluidic setup and connections used in mixing testing. The re-suspension slide was connected to

the bellows actuation slide via a soft, tygon tube. Plastic microfluidic connectors were used to

connect the tube and the plastic microfluidic device with an air tight seal. The bellows membrane

deflection induced a volume change in the microfluidic system, which results in a pressure

change inside the microchannels to drive the liquid sample forward. When the bellows returned

to the original state, the volume inside the microfluidic system expanded, thus liquid sample

retracts and moves backward inside the microchannel. Figure 4-15 on Page 75 is a photograph of

the experimental setup used in this part of the work.

4.5.5.4 Testing Procedure

Fluorescently labeled antibodies were purchased from BioLegend as described in Section 4.5.1.

The liquid reagent was used as the control sample on the flow cytometer for re-suspension

testing. Ten micro liters of Immunotrol blood control sample, purchased from Becton Dickinson,

was injected into the microfluidic slide via the sample port. After connecting and sealing the

Reagent plug

Page 105: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

92

bellows slide with the re-suspension slide, the stabilized blood sample was then pushed forward

to the reagent chamber. The blood sample was then actuated back and forth around the reagent

chamber to re-constitute and mix with the dried reagent. After mixing is complete, the stabilized

blood sample was then pushed forward to the sample outlet, which was transferred to a flow

cytometer for cell counting using a pipette.

4.5.5.5 Experimental Observations and Results

4.5.5.5.1 Mixing Time

The stabilized blood sample was actuated back and forth inside the microfluidic mixer for 10, 15,

20, and 30 passes using the experimental setup shown in Figure 4-15 on Page 75. Table 7 below

summaries the mean fluorescence intensity measured by flow cytometer as a function of varying

mixing time. From this investigation, it was clear that 15 passes of back and forth actuation, (~4-

5 minutes) was required to re-suspend the dried reagents thoroughly.

Table 7 - Mixing time characterization result. Relative mean fluorescence intensity was

calculated as the ratio between liquid control mean fluorescence intensity and measured

mean fluorescence intensity for each scenario. This characterization was completed using

design 2 of the resuspension prototype slide shown in Figure 4-23.

Number of Passes Mixing Time (sec) Relative Mean Fluorescence Intensity

5 passes 50 0.2

10 passes 100 0.5

15 passes 150 0.7

20 passes 200 0.98

25 passes 250 1

30 passes 300 1

4.5.5.5.2 Reagent Chamber Design

In Figure 4-23 on Page 90, there were three different micro-mixer designs in the re-suspension

slide. As shown in the figure, design 2 had a reagent chamber that was wider than the reagent

plug while in design 4, the reagent chamber has the same width as the reagent plug. It was

observed design 4 worked much better than design 2, since liquid tend to flow by reagent plug on

the side instead of underneath the plug with reagent if the chamber was wider than the reagent

plug. This significantly reduced the interaction time between dried reagent and liquid sample

during re-suspension, hence leads to longer re-suspension time.

Page 106: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

93

Table 8 - Mixing time characterization result. This characterization was completed using

design 4 of the resuspension prototype slide shown in Figure 4-23. With the optimized

reagent chamber dimension, the mixing time can be further reduced.

Number of Passes Mixing Time (sec) Relative Mean Fluorescence Intensity

5 passes 50 0.3

10 passes 100 0.7

15 passes 150 0.98

20 passes 200 1

25 passes 250 1

30 passes 300 1

4.5.5.5.3 Reagent Plug

Some of the reagent plugs underwent oxygen plasma treatment prior to reagent drying. The

oxygen plasma treatment improved the adhesion of the dried reagents: no reagent came off the

plug surface but re-suspension took longer (up to 30 passes to dissolve the dried reagents). The

oxygen plasma treatment made the plastic plug hydrophilic, especially along the circular walls of

the tip. During mixing, the liquid tends to flow through the reagent chamber around the plug,

hence reducing contact with the dried reagent and increasing the required re-suspension time.

Table 9 - Mixing time characterization result. This characterization was completed using

design 4 of the resuspension prototype slide shown in Figure 4-23 and surface treatment of

the reagent plug.

Number of Passes Mixing Time (sec) Relative Mean Fluorescence Intensity

5 passes 50 0.25

10 passes 100 0.65

15 passes 150 0.78

20 passes 200 94

25 passes 250 1

30 passes 300 1

4.6 Conclusion

In this chapter, we investigated the two fluidic transport mechanisms: capillary driven flow and

volumetric pressure driven flow. Capillary driven flow is a passive technique where no external

forces or energies are required to manipulate the sample fluids on-chip, whereas volumetric

pressure driven flow requires an external energy source and components to generate the pressure

Page 107: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

94

needed to achieve the desired flow control. Capillary driven fluid flow is strongly dependent on

material property, more specifically surface contact angle with the liquid. Hence to optimize or

enhance the fluidic flow on-chip, surface treatment such as oxygen plasma is often required. The

active fluidic manipulation method is less dependent on the material of the microfluidic device

and it produces more consistent and precise fluidic transport outcome, although more energy

consumption is necessary and the overall system is more complex.

A detailed analysis on each fluidic actuation approach: capillary driven and volumetric pressure

driven fluid flow was conducted in this work. The flow characteristics were investigated and

strategies to control the flow speed were presented in this chapter. The on-chip fluidic

management is a key component in the disposable cartridge that completes the cell enumeration

analysis, whereas volumetric pressure driven flow requires external energy sources and

components to generate the pressure needed to achieve the desired flow control.

Page 108: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

95

Chapter 5 Optics and Detection

After the fluidic system of the proposed portable cell/particle detection system is finalized, the

optical detection sub-system can be designed for cell/particle detection and counting. Since a

dynamic counting approach is used in this work, maintaining a uniform flow speed is critical in

ensuring accurate cell counting results. The detailed analysis and results of particle detection and

enumeration are shown in this chapter.

5.1 Introduction

This chapter focuses on the development of an optical based imaging module to detect and

enumerate microparticle and cells populations. A brief literature review explains the current

common approaches and research results, followed by a detailed description of the dynamic cell

counting method and experimental results of the proposed detection concept. The second part of

this chapter discusses the utility of using this imaging technique to develop a beadarray detection

platform for the analysis of proteins and DNA/RNA molecules.

5.2 Particle Detection using Optics

As described in the introduction chapter, there are a number of detection approaches. Optical and

fluorescent detection is the most commonly used approach and widely adopted in laboratory-

based technologies such as flow cytometry and microscopy. It offers better sensitivity, accuracy

and precision, but suffers on weight, size and instrumentation cost.

5.2.1 Optical Detection Methodology Literature Review

Optical detection of particles and cells can be classified in two main categories: far field and near

field imaging. Far field imaging is a direct imaging approach where lenses are used to capture

Page 109: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

96

directly the particles/cells whereas in near field imaging, the diffraction pattern of the subject is

captured and original image is re-constructed from the diffraction pattern.

5.2.1.1 Static Imaging

The most common optics based approach is far field static imaging of cells or microparticles.

The working principle is the same as a modern microscope. Typically if fluorescence is utilized

to specifically target cells of interest, expensive and bulky optical components, such as filters,

dichroic mirrors and condensers, are required177.

With the advances in microfluidic lab-on-a-chip technologies, researchers began to investigate

mobile platforms that can detect and analyze cells at point of care. Ozcan et al.178 engineered an

imaging module that can be attached to a mobile phone to analyze and image cells. The system

comprises affordable off-the-shelf optical components that are readily obtainable. The imaging

system has a field of view of ~81 mm2 and a depth of field of 2 mm, permitting analyzing

volumes > 0.1 mL for high throughput applications.

Static imaging offers the best optical quality and flexibility in detecting target particles/cells. The

optical detector also has the freedom to vary the exposure time to optimize the acquired signal

intensity level. The main disadvantage related to this type of imaging technique is the ability to

multiplex multiple target particle/cell population detection. The overall system will get

significantly more complex mechanically and becomes impractical to be miniaturized to a

portable, point-of-care analytical or diagnostic tool.

5.2.1.2 Microfabricated Flow Cytometry

Microfabricated flow cytometry is another common approach179-181. Recent development in

micro/nano fabrication in the semiconductor industry has accelerated research efforts and

progress in this area. In these approaches, the underlying system design follows that of a

conventional flow cytometer where particle focusing is utilized to form a single file of

cell/particle before being interrogated by a laser beam. Different detectors have been used to

capture individual emission wavelength of the target fluorescence. Morgan et al.8 developed a

device where all the optical devices were monolithically integrated in a Si wafer while the optical

routing was accomplished via integrated waveguides.

Page 110: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

97

A microfabricated flow cytometry, in essence, employs the same technology and process as

conventional flow cytometry. Without the necessary fluidic system, it is not feasible to achieve

the required flow rate and analysis throughput for practical portable and point-of-care

applications.

5.2.1.3 Near Field Imaging

Far field imaging offers excellent image quality. However, it requires use of filters and lenses,

which makes the instrument larger and more expensive. Another approach is that of near field or

contact imaging, where the subject is placed in close proximity of the detector (tens of microns).

A diffraction pattern is generated and captured by the optical sensor. Upon numerical

reconstruction, the original subject image can be re-constructed182-186.

Coskun et al.187 developed a lens-free imaging system that can detect fluorescently labeled cells.

In this work, the optical sensor was placed directly on top of the microfluidic channel with target

cells, hence the name lens free imaging. A similar method was described in a patent

application188, where homogeneous particles in a solution can be detected in a lens-less setup

using the holography technique.

The main problem associated with near field imaging is the requirement of placing the target

cells/particles close to optical sensor. This is also detrimental to any microfluidic based portable

analytical systems that can be useful and effective in clinical and commercial applications.

5.2.2 Dynamic Particle Detection and Counting

A dynamic counting/detection approach was developed in this work to improve the mechanical

stability and reduce the active components in the system. When combined with capillary

microfluidic devices described earlier, this forms a simple and flexible multiple fluorescence

detection system. This could be miniaturized on a handheld platform as a portable cytometer or

cell analysis device. The following sections describe the dynamic imaging, its principle and

implementation.

Page 111: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

98

5.2.2.1 Design Concept

The static imaging requires stitching of multiple images to cover the entire imaging area due to

the limitation on the size of field of view. To image multiple biomarkers with difference

fluorescence antibodies, filter wheels must be used to choose the appropriate fluorescence filter

just as in a fluorescence microscope. On the other hand, in conventional flow cytometry,

individual particles/cells are interrogated one by one. Although different fluorescent dyes can be

distinguished through complex optical systems involving dichroic mirror and filters, the entire

setup is bulky and prone to mechanical instability. The fluidic system is also very complex to

produce single column particles/cells so that only one particle/cell is being examined at a time.

To improve the detection, a dynamic imaging approach is designed and developed in this work.

This dynamic imaging approach uses wide microfluidic microfluidic channel structures to

regulate and control the fluid flow during analysis. The particles are moving in multiple streams

in parallel passing through the optical detector. The use of microfluidic device eliminates

complex fluidic control system present in a typical flow cytometer. The multi-stream imaging

also improves the throughput of this approach so that the final particle/cell analysis platform is

practical and feasible to become a commercial cell analyzing device. The dynamic imaging,

while particles are moving inside the microchannel, further eliminates the need of filter wheels in

multi wavelength fluorescence detection. The reduction of the required mechanical components

makes this an ideal platform for a portable or handheld cell analysis device.

5.2.2.2 Optical Detector

There is a wide range of optical detectors that are used in biomedical and scientific imaging

applications, including one dimensional avalanche photodiode, photomultiplier, and two

dimensional CCD (charge coupled device) and CMOS (complementary metal-oxide

semiconductor) sensors. For the imaging methodology described in the previous section

(5.2.2.1), a two dimensional image sensor is required.

Page 112: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

99

5.2.2.2.1 CCD vs CMOS

Much has been written about the relative advantages of CMOS versus CCD image sensors. Both

technologies and markets have been evolving, affecting not only what is technically feasible, but

also what is commercially available. Some applications are best served by CMOS sensors while

for others a CCD sensor might be the superior choice. In most visible imaging application,

CMOS outperforms CCD imagers, while in Near Infrared (NIR) application, CCD may be a

preferred.189,190 In scientific applications where sensitivity is critical to capture weak

fluorescence signal, CCD is also the preferred device. It also has hardware binning features to

enable combination of adjacent pixels to act as one giant pixel, hence significantly reducing

readout noise and improving signal to background ratio.

5.2.2.2.2 Optical Sensor Selection and Characterization

Several CCD and CMOS cameras that are designed for scientific imaging applications were

identified and tested. For the target application, sensitivity and dynamic range are the two most

important parameters. Recent advances in electronics and semiconductor manufacturing process

have enabled the realization of wide dynamic range in all the optical image sensors. The real

limiting factor in this characterization becomes the sensitivity. The other consideration is form

factor, since the eventual goal is to develop a portable, handheld cell analyzer platform for global

health. Affordable devices with small foot print, low power consumption are also important

considerations in choosing the image detector.

Four different Lumenera cameras have been tested. These low cost, scientific grade Lumenera

modules are compared with a more expensive Pixelfly USB camera which was used as the bench

mark. An inverted fluorescence microscope was used for the experiments. The microscope

attached halogen lamp was used for the transmitting light. The testing parameter was chosen to

be close to the condition for CD4 cell detection.

The testing conditions are described as follows: 10X objective lens (NA = 0.25) was focused on

a cover glass slide, the lamp was adjusted so that the average intensity reading on Pixelfly USB

is about 2000 when using 20 ms exposure time and 1x1 binning. The lamp light setting was kept

the same throughout the tests.

Page 113: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

100

Within the four Lumenera cameras, only LW110 is a CMOS camera. Other three cameras are all

CCD cameras with hardware binning capabilities. LW160 camera uses the same SONY

ICX285AL sensor (type 2/3) as Pixelfly USB camera. LW130 camera uses a smaller SONY

ICX205AL sensor (type 1/2). Images were always acquired using 16 bits bit depth. The gamma

is set to 1 to keep the original data untouched. Whenever available, the smallest gain that can still

cover the whole bit depth of the camera at strong light condition is used. The actual gain used are

(LW160, LW110 gain=1), (LW130, Lt365 gain=0.7). The background was acquired using the

same 20 ms condition except the camera was fully covered. The following table listed the testing

results.

Table 10 - Camera comparison summary.

Camera model binning exposure I average stdev var(I) SNR*

Pixelfly USB 1x1 20ms 2076.81 130.2 8476.02 27.1

2x2 20ms 8422.04 268.24 35976.35 32.9

Lumenera LW160RM-sci 1x1 20ms 934.36 80.45 3236.10 24.3

2x2 20ms 3739.44 155.1 12028.01 30.7

4x4 20ms 15039.43 316.56 50105.12 36.5

Lumenera LW130RM-sci 1x1 20ms 268.18 54.3 1474.25 16.9

2x2 20ms 1075.1 97.49 4752.15 23.9

4x4 20ms 4341.97 183.39 16815.95 30.5

lumenera LW110 1x1 20ms 344.77 52.33 1369.21 19.4

Lumenera lt365 1x1 20ms 684.85 73.27 2684.25 22.4

2x2 20ms 2738.76 140.85 9919.36 28.8

3x3 20ms 6173.01 208.62 21761.15 32.4

4x4 20ms 10994.44 275.6 37977.68 35.0

8x8 20ms 44235.2 554.81 153907.07 41.0

Page 114: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

101

* The signal to noise ratio (SNR) here is based on the conditions used above, which will be

different for the actual CD4 cell signal detection.

5.2.2.2.3 Camera Characterization Results

The commonly used setting on the Pxelfly USB camera for CD4 cell detection are 2x2 binning

with 20 ms exposure time (blue color). Other camera settings that can achieve comparable SNR

have been identified (green color). LW110 has a rather low SNR number. Combined with its

small pixel size at 3.6 μm, it is unlikely it can meet the requirements for CD4 cell detection.

Lt365 is a high-end camera. Its great performance has a high price tag. Since LW160 is

comparable to Pixelfly USB that can detect CD4 cells. It is unlikely that we will choose Lt365.

However, its superior performance will be useful for an application requiring better sensitivity

such as beads assay.

Both LW160 and LW130 have settings that comparable with the Pixelfly USB. The LW130

camera is especially interesting. At 4x4 binning, its SNR is at 30.5. Although the ICX205 sensor

has a smaller pixel size of 4.65 μm comparing to the 6.45 μm pixel size for ICX285 sensor, its

4x4 binning has an effective pixel size of 18.6 μm while the 2x2 binning of ICX285 is only 12.9

μm. It is reasonable to say that the LW130 can detect CD4 cells at the same condition as Pixelfly

USB even considering that the ICX205 sensor has a smaller quantum yield comparing to ICX285

sensor. In fact, a test using CD4 PE antibody stained Immunotrol sample has been conducted.

The results demonstrated comparable detection sensitivity to the Pixelfly USB camera.

5.2.2.3 Imaging Lens

At high numerical apertures of the microscope, depth of field is determined primarily by wave

optics, while at lower numerical apertures, the geometrical optics dominates the phenomenon.

Using a variety of different criteria for determining when the image becomes unacceptably sharp,

several authors have proposed different formulas to describe the depth of field in a microscope.

The total depth of field is given by the sum of the wave and geometrical optical depths of fields

as:191

𝑑𝑡𝑜𝑡 =𝜆𝑜𝑛

𝑁𝐴2 +𝑛

𝑀⋅𝑁𝐴𝑒 (5 – 1)

Page 115: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

102

Where 𝑑𝑡𝑜𝑡 represents the depth of field, 𝝀 is the wavelength of illuminating light, 𝑛 is the

refractive index of the medium (usually air (1.000) or immersion oil (1.515)) between the

coverslip and the objective front lens element, and 𝑁𝐴 equals the objective numerical aperture.

The variable 𝑒 is the smallest distance that can be resolved by a detector that is placed in the

image plane of the microscope objective, whose lateral magnification is 𝑀. Using this equation,

the depth of field 𝑑𝑡𝑜𝑡 and wavelength 𝜆 must be expressed in similar units. For example, if

𝑑𝑡𝑜𝑡 is to be calculated in micrometers, 𝜆 must also be formulated in micrometers (700

nanometer red light is entered into the equation as 0.7 micrometers). Notice that the diffraction-

limited depth of field (the first term in the equation) shrinks inversely with the square of the

numerical aperture, while the lateral limit of resolution is reduced in a manner that is inversely

proportional to the first power of the numerical aperture. Thus, the axial resolution and thickness

of optical sections that can be attained are affected by the system numerical aperture much more

so than is the lateral resolution of the microscope.192

5.2.2.4 Optics Setup

A handheld green laser pointer, emission wavelength 532 nm with power of 18 mW was used to

excite the fluorescent dyes. Emission filters used in the fluorescence detection system were

purchased from Semrock (585/40 product number FF01-585-40 and 708/75 product number

FF01-708-75). An Olympus microscope objective lens (10x, NA 0.30) for emission light

collection was purchased from Spectra Physics. The CCD camera (Pixelfly USB) was purchased

from PCO-TECH Inc.(Kelheim, Germany). The fluorescence signals were acquired in a 2x2

binning configuration to enhance the signal to noise ratio. The optics was assembled in a lens

tube (SM1L, SM1V and SM1ZM, Thorlabs) configuration.

5.2.2.4.1 Measurement Setup

This imaging system is based on a modified fluorescence microscopy imaging approach, where

the excitation filter and dichroic mirrors are not required. Figure 5-1 is a schematic diagram of

the optical detection system. Upon illumination, fluorescently labelled particles/cells of interest

will be excited and emit light. The imaging system, a microscope objective lens, magnifies the

Page 116: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

103

target particles/cells, collects emitted light and projects to the optical detector. The light then

passes through emission filter and is captured by the optical detector. The samples under testing

are loaded onto a custom designed disposable plastic microfluidic chip. A thin blood smear is

formed once the sample fills inside the microfluidic channel, whereby particles are imaged and

characterized based on their optical properties. Furthermore, an image analysis program is used

to analyze and process the acquired optical images for particle and cell detection and

enumeration. The only optical elements in this system are the imaging lens and emission filters.

Unlike a conventional fluorescence microscope setup, the dichroic mirrors and excitation filters

are eliminated, which reduces the complexity of the platform and makes the optical system easier

to miniaturize.

Figure 5-1 – Optical imaging system of the cell/particle detection and analysis platform.

For multi-color detection, a custom designed emission filter was used in the optical detection

system. The filter set, (optical property shown in Figure 5-2) incorporates two half-moon shaped

fluorescence filter into a single round cell. This specific custom filter allows for two-color

fluorescence detection side by side simultaneously.

The imaging system uses wide field dynamic imaging approach that does not require any moving

components, such as filter wheels and rotation stages, needed in standard fluorescent detection

for multi-color (or multiplexed) analysis, a key feature and advantage of this analytical platform.

Page 117: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

104

During analysis, a time-series images of particles moving inside the microfluidic device was

captured. An optical detection area, defined by the size of the CCD detector and the

magnification of objective lens, is used to examine and measure the fluorescence signal. The

CCD detector starts data acquisition when the sample enters the analysis chamber. As the fluidic

sample moves into and fills the analysis chamber, the detector continues capturing optical images

over time until the chamber is full and fluidic flow is stopped. By analyzing the acquired images,

the number of fluorescent particles of interest which pass under the detection window can be

counted, generating the final cell count. The captured images are also analyzed to render the

intensity of particles and to generate counting statistics. The microfluidic chamber is designed

and fabricated with a precise volume; hence the final results are well calibrated. An optical filter

with a single filter or an array of filters is placed in front of the optical detector. When an array of

filters is used, the system has the ability to do multi-color (or multiplexed) fluorescent detection

without any additional (moving) components, a key feature of this analytical platform. Figure 5-2

is a graphical illustration that further describes the underlying principle of this technique.

Page 118: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

105

Figure 5-2 – Illustration of the multiplexed detection system described in this paper. The optical detector

continually takes images as particles/cells move into the detection window. Particles/cells labelled with

different fluorophores can only be detected in the corresponding sub-regions within the detection window

depending on the filter setup. (a) a graphical illustration of the underline principle of the technique; (b)

transmission spectrum of the left sub-region of the detection window; (c) transmission spectrum of the right

sub-region of the detection window.

5.2.3 Cell Detection and Enumeration using Capillary Microfluidic Devices

In order to demonstrate the proof of concept of this cell analysis platform, CD4 T cell counting

was chosen as the initial test, in direct comparison with clinical flow cytometry. CD4 T cells are

a specific type of white blood cell and its concentration can be a measure of human immune

system strength. This assay is one of the most predictive tests for HIV/AIDS monitoring in

infected individuals and is used to determine the timing for the initiation of anti-retroviral drug

therapy. The test of our cell analysis system is conducted using Immuno-Trol control cell

samples for single population CD4 T cell detection.

Page 119: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

106

5.2.3.1 Sample Preparation

Samples were stained with 40 μL PE (excitation 532 nm, emission 585 nm) labeled CD4 antibody

and 6 μL of PE-Cy5.5 (excitation 532 nm, emission 670 nm) labelled CD3 antibody. A single

droplet of stained Immuno-Trol, approximately 5 μL, was transported to the inlet reservoir of the

microfluidic device shown in Figure 3-11. The fluid inside the microfluidic chip was then

completely driven by capillary force. The microfluidic channel can be divided up into three

zones: flow restrictor, detection window and capillary pump. The flow restrictor is a linear

microchannel, with a cross sectional area of 15 μm x 100 μm. This zone is to reduce the fluid

flow to a reasonable rate so that particle/cell motion can be captured by the optical detector. The

detection window has an area of 500 μm x 800 μm, which matches the size of the 1M pixel CCD

sensor through magnification of the objective. The acquired fluorescence images will be

processed and analyzed to produce the final cell/particle counting statistics. The final section of

the microchannel serves a dual purpose: a volumetric reservoir and a capillary pump. As the fluid

sample fills the microfluidic device, the fluidic resistance increases and the flow speed reduces.

To make the whole instrument functional and practical, the sample flow needs to maintain a

uniform rate so that the acquired images can be analyzed to produce an accurate cell

concentration measurement within a reasonable time frame. The widening of the fluidic channel

reduces the fluidic resistance seen by the sample fluid inside the microchannel and effectively

acts as a capillary pump. This capillary actuation counteracts the decrease in the fluidic flow in

the previous sections along the microfluidic device, maintaining a uniform flow profile.

5.2.3.2 Antibody Concentration

In flow cytometry, detailed sample preparation procedures were involved in each analysis,

including CD4 T cell counting. Blood samples are typically lysed, then diluted with Phosphate

buffered saline (PBS) buffer solution. During analysis, distilled water was added to the fluidics

to create hydrodynamic focusing which further dilutes the blood sample. These steps

significantly reduce the unbound antibody concentration remaining in the blood sample. Hence,

in flow cytometry, there is no significant impact on the recorded mean fluorescence intensity.

In the proposed microfluidic based point-of-care system, the only sample preparation involved is

staining with fluorescent dyes. As a result, the background fluorescent signal of the blood sample

became the dominant source of noise.

Page 120: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

107

The background fluorescence primarily came from two sources: auto-fluorescence from the

blood and unbound antibodies in the solution. To avoid any complex on-chip sample washing

processes, the auto-fluorescence of blood is difficult to be further reduced. The unbound

antibody in the solution, however, can be optimized. A range of antibody concentration was used

to study its impact on background and signal to background ratio. 100 microliter of stabilized

blood control sample was separated into two vials: one to be run on flow cytometer while the

other on the microfluidic platform. The flow cytometry sample preparation followed the standard

protocols, including lysing, adding buffer solution and staining while the microfluidic sample

only included staining of fluorescently labeled CD3 and or CD4 antibodies.

CD3 and CD4 antibody concentration optimization was conducted. The optimization was based

on the concentration at which the CD3 and CD4 cell intensity plateau was reached. The deciding

factor here is the average intensity of CD3 and CD4 cells in their respective fluorescent channel,

since the background intensity is low in flow cytometry. However, the same is not necessarily

true for the dynamic wide-field imaging detection approach developed in this thesis. The correct

identification of CD3 and CD4 cell event in the wide-field imaging detection is based on whether

the signal can be well separated from the rest of the background. As a result, the optimization

will need to be based on the signal to background ratio. Experiments using a series of different

concentration of CD3 and CD4 antibody were conducted. The results of their respective average

signal to background ratio was used to identify ideal antibody concentration range.

The concentration range used on the flow cytometer was used as a starting point. The

concentration we chose for CD3 antibody testing were 10, 25, 50, 100, 250, 500 and 2500 ng per

100 µL blood. The concentration range used for CD4 antibody testing were 2.5, 5, 10, 25, 50 and

250 ng per 100 µL blood. Two optical channels, PE and PE-Cy5, were used to observe CD4 and

CD3 respectively. The results are shown in Figure 5-3 and Figure 5-4 where the average signal to

background ratio is plotted against the logarithm of each antibody concentration. Based on the

results, the best average signal to background was achieved at 100 ng and 50 ng per 100 µL

blood for CD3 and CD4 antibody respectively.

Page 121: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

108

Figure 5-3 – CD3 antibody concentration titration curve for signal to background ratio.

Figure 5-4 – CD4 antibody concentration titration curve for signal to background ratio.

1.00

1.50

2.00

2.50

3.00

3.50

1.00 1.50 2.00 2.50 3.00 3.50 4.00

Sig

/BG

(A

U)

log of antibody quantity (ng)

CD3 Antibody titration curve

1

1.5

2

2.5

3

3.5

0.50 1.00 1.50 2.00 2.50

Sig

/BG

(A

U)

log of antibody quantity (ng)

CD4 Antibody titration curve

Page 122: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

109

Figure 5-3 and Figure 5-4 are titration curves for both CD3 and CD4 antibody concentrations.

From the range of reagents tested in this part of the thesis project, there is an optical level of

antibody concentration for each cell population as shown. Each data point in these two figures

represents average values of three separate measurements. The error bars indicate the variation in

the experimental measurements.

When the antibody concentration is low, there are not enough antibody molecules in the solution

to bind with antigens on cell surface, resulting a weaker fluorescence signal. On the other hand,

if the antibody concentration is too high, all the cell surface binding sites are occupied and there

remains a large number of antibody molecules in the solution, resulting a high background signal

and a low signal to background ratio.

Figure 5-5 – Positive events intensity histogram for PE channel at 50ng CD4 antibody per 100µL blood.

In addition, it was also observed that the population of positive event separation in the CD4

antibody concentration tests that are at or close to optimal concentration. Figure 5-5 shows the

positive events intensity histogram in PE channel when 50ng PE CD4 antibody per 100 µL was

used for the test. There is a clear separation between two intensity populations. It is most likely

that the two populations are CD4 cells and monocytes since the CD4 antigen is also weakly

Page 123: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

110

expressed on monocytes. This capability to differentiate cell populations is not crucial for the

CD4 assay. However, it will be critical for other cell counting assays or even beadarray assays.

5.2.3.3 Image Analysis

An image analysis program was developed in collaboration with an undergraduate project

student James Durst, to produce the counting results. The program takes the fluorescence images

acquired by the CCD camera, and based on the intensity level of pixels it detects and tracks the

cells of interest. The entrance region of the detection window is scanned in every captured frame

to search for fluorescent events. Fluorescent objects are detected if groups of pixels display

intensity levels exceeding a pre-determined threshold. Since the fluidic flow inside the

microchannels are strictly laminar, the program would look for small regions where a fluorescent

particle is expected in subsequent frames. Since only a small portion of the entire images are

processed, it significantly improves the processing speed and reduces computing power

requirements. This is very important for real time analysis especially at a high frame rate. To

track fluorescently labelled subjects’ location within each frame, a virtual bounding box is placed

on each particle/cell, recording its maximum and minimum x and y coordinates. The center

position of each box is then computed and marked as the current position of the cell. This

process will be repeated for each frame captured and analyzed to produce a final enumeration

result. In addition to the entrance scanning of target population, particles/cells of interest are also

detected and counted upon exiting the detection window. This secondary analysis is compared

with the entrance scanning to ensure the accuracy of counting. Finally, the detection of

fluorescent subjects by continuously tracking them through multiple image frames increases the

reliability of the counting results, which makes the technique readily applicable for health-related

measurements such as CD4 T cell counts in human blood.

5.2.3.4 Detection Optimization

Since dynamic counting is used in determining the cell/particle statistics, the exposure time of

the optical detector is one of the most important parameter in optimizing the signal to

background ratio of the acquired images and such counting accuracy. The ultimate goal is to

maximize the signal to background ratio, but the ability to this maximization is affected by the

Page 124: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

111

exposure time and flow speed. If the CCD integration time is too short, the fluorescence signal

captured by the detector would be low and compromising signal to background ratio. On the

other hand, if the integration time is too long, the particles/cells might be travelling too fast for

the detectors such that it can create motion blur in images or even been missed by detector

completely. Motion blur will compromise the signal to background ratio, while cells flowing

through the detection window will be and missed registering on the CCD and will also lead to

errors in counting and analysis results. The optimal integration time should allow the

fluorescently labelled particles/cells to produce bright signal comparing with background, and at

the same time the electronic circuitry that drives the CCD is fast enough to capture all the

cells/particles of interest with proper sampling rate.

Page 125: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

112

Figure 5-6 – Snapshots illustrating the effect of optical detector exposure time. (a-d) shows a steady decrease

in detector exposure time from 50ms to 10ms. The particles/cells shape appears more circular at shorter

exposure, while the signal to background ratio is lower.

5.2.3.5 Comparison with Flow Cytometry

Using our microfluidic approach, enumeration measurements were first conducted using 10-um

Flow-CountTM polystyrene microspheres in phosphate buffered saline (PBS) solutions. The

initial experiments were performed on an Olympus BX50 upright fluorescence microscope. Band

pass filter sets were used for fluorescence excitation and emission measurements. An average

count of 970 ±70 particles/μL was recorded with our microfluidic chips. Using the same testing

material the flow cytometer recorded a count of 1,007 ± 100 particles/μL.

(a) 50 ms exposure, S/B: 4200/2000 (b) 25 ms exposure, S/B: 1700/912

(c) 15 ms exposure, S/B: 858/540 (d) 10 ms exposure, S/B: 695/500

Direction of fluid flow

Ch

an

ne

l wid

th

100 μm

Page 126: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

113

This dynamic approach was then tested using Immuno-Trol Control Cells, commonly used

stabilized blood samples for ensuring clinical labs can count CD4 cells at two concentrations.

Immuno-Trol High CD4 counts are in the range of a normal blood sample, and Immuno-Trol

Low is at the level of an immune-compromised individual. A clinically validated commercial

CD4 antibody with Phycoerythrin (PE) dye was used in testing these stabilized blood samples.

The testing produced an average count of 620 ± 15 cells/μL and 184 ± 17 cells/μL for high and

low concentrations whereas the flow cytometer measured 670 ± 70 cells/μL and 158 ± 28

cells/μL respectively. For each sample, the counting test was repeated 10 times.

Table 11 – Comparison of results using wide field dynamic counting system and flow

cytometer.

FlowCount Fluorospheres

(#of particles/μL)

Immuno-Trol High

(# of cells/μL)

Immuno-Trol Low

(# of cells/μL)

Flow Cytometer 1007 100 670 70 158 28

Prototype 970 70 620 15 184 17

5.2.3.5.1 Linearity

To demonstrate the performance of this platform in real world applications, a range of cell

concentrations of interest to CD4 enumeration assay was tested on this platform, from 180 per μL

to 720 per μL as plotted in Figure 5-7. Two types of Immuno-Trol samples were directly used in

this analysis: low (180/ μL) and high (700/ μL) concentration controls while the intermediate cell

concentrations were obtained through dilution from the high concentration Immuno-Trol sample.

In total, there were four concentrations tested in this part of the characterization: 180/μL, 380

/μL, 500 /μL and 700 /μL. The same samples were tested through our portable system and clinical

flow cytometer in parallel and cell statistics were plotted in Figure 5-7. The direct comparison

showed great agreement between the two methods, proving the accuracy of the portable cell

analysis platform under a wide range of cell concentration that is of clinical importance.

Page 127: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

114

Figure 5-7 – Linearity test result. Cell concentrations range from 150 to 720 per μL were tested for the

comparison with flow cytometer. (Each data point is an average value for 3 measurements with standard

deviation bar.)

5.2.3.5.2 Mixed Population

In clinical relevant assays, the ability of detecting multiple biological targets is of primary

importance. The multiplexing provides additional information for each sample being studied and

allows identification and characterization of multiple cell populations or cell subpopulations. In

the particular case of CD4 analysis, both CD3 and CD4 cells need to be enumerated to eliminate

possible inclusion of monocytes which also weakly expresses the CD4 biomarker on its cell

surface. To develop a portable cell analyzer that can produce both statistics, a two-color

detection system must be implemented. As a demonstration of the multiplexing capability of this

platform, fresh whole blood sample was used to count cells with both CD3+ and CD4+

biomarkers for absolute CD4 T cell counts. Similar to the schematics shown in Figure 5-1 and

Figure 5-2 where the optical detection window was divided into two sub-regions, two half-moon

shaped fluorescence filters were combined and placed in front of the optical detector. Particles

y = 0.9611xR² = 0.9825

0

100

200

300

400

500

600

700

800

0 200 400 600 800 1000

pro

toty

pe

CD

4 c

ou

nts

flowcytometer CD4 cell counts

Series1

Linear (Series1)

Page 128: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

115

labelled with specific fluorophores will be present in the corresponding panel/region of the

optical detector as shown in Figure 5-2. Fresh whole blood sample was prepared as described in

the material section. In the mixed population experiment, cells with the CD3+ antigen were

bound with antibody conjugated PE-Cy5.5 (excitation 532nm/emission 705nm) molecules,

corresponding to the right panel of the detection window shown in Figure 5-2, while CD4+ cells

were bound with antibody conjugated PE (excitation 532nm/emission 585nm) molecules, the left

panel of the detection window in Figure 5-2. In a single run, statistics of cells expressing CD3

and CD4 surface biomarkers could be obtained. Cells that express both CD3 and CD4

biomarkers on the cell surfaces are leukocytes that we are interested in counting in absolute CD4

counting analysis since monocytes do not express CD3+ antigens on their surface.

Similar to previous experiments, the stained samples were run through both the clinical flow

cytometer and our portable cell analyzer system in parallel. This direct comparison showed

excellent agreement between these two approaches and demonstrated the functionality and

accuracy of this dynamic cell counting technique. The imaging system reported in this work can

be further miniaturized and integrated into a handheld electronic device, such as a smart phone,

to be deployed in the field as a point-of-care testing instrument.

Page 129: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

116

Figure 5-8 – Two color fluorescence image captured through a custom setup with two half-moon shaped

optical filters placed together side by side. The CD4 cells labelled with PE (phycoerythrin) were shown in the

left panel while the CD3 cells labelled with PE-Cy5.5 were shown in the right panel.

5.2.3.5.3 Limit of Detection

The sensitivity (or limit of detection) of the system is very important for the detection especially

when the signal is weak. Experiments have been conducted using beads that have known

fluorescence intensity based on their Molecules of Equivalent Soluble Fluorochrome (MESF)

value. The beads with PE dyes used here have MESF of 14975. The magnified images of

detected MESF beads during our acquisition were shown in Figure 5-9.

585 nm +/- 20 nm emission channel 705 nm +/- 70 nm emission channel

Page 130: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

117

Figure 5-9 – Combined images is showing four detected MESF beads.

Using the signal and noise values acquired on these images, the limit of detection of the system

was determined to be 6400 MESF unit based on that the minimum detectable signal need to be 3

times of noise. The sensitivity of a typical flow cytometer is in the 900 MESF unit range for

fluorescein. Currently, the sensitivity of our system is not as good as flow cytometer, but

additional optimization could help to improve the limit of detection further. Both a more

powerful light source and a better sensor such as backside illuminated sensor can be used to

increase the sensitivity of the system. In addition, our detection is based on image recognition.

The signal can be reliably detected even before the signal reaches the level of 3 times of the

noise. This can further increase our sensitivity. By proper optimization, it is reasonable to expect

this detection system can reach sensitivity in 1,000 MESF unit range.

5.3 Beadarray Multiplexed Detection

A further development of the detection system described in this work is multiplexed beadarray

platform. Similar to the multiplexed beadarray technology from Luminex Corporation, it can

simultaneously detect multiple protein, or enzyme molecules. This approach uses the sensitivity

of amplified fluorescence detection to measure soluble analytes in a particle-based immunoassay,

Page 131: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

118

which will significantly widen the detection capacity of the detection platform. Target analytes

can range from blood serum proteins, enzymes to DNA/RNA molecules.

5.3.1 Underlying Strategy

The method is based on two-dimensional fluorescence detection system to identify the bead array

populations in solution. Each bead population can be functionalized with an antibody, or antigen

for a specific test. This detection methodology is similar to the Luminex xMAP® technology

where multiplexed discrete assays are performed on the surface of color coded beads or

microspheres. These beads are read in a desktop analyzer in which lasers or LEDs, optical

detectors and high speed digital signal processors combine to generate fluorescence intensity

based analytical data. The fluorescence intensities of each color of beads are differentiated and

can be correlated to analyte concentration.

In this detection approach, the beads are mixed with blood or serum sample, with a reporter

fluorochrome label in a single or multiple mixing step, followed by necessary washing step. We

may be able to mix the beads, blood or serum, with a reporter fluorochrome tag in a single

mixing step, followed by one washing step. One or more bead populations can be included to

measure unspecific binding. An additional bead can be deployed with a specific fluorescent

intensity, the same as our reporter fluorochrome, to establish a threshold range, positive for all

the analytes measured, as a control signal to the analysis.

The multiplexed beadarray detection concept that is proposed is further illustrated in Figure 5-10.

Page 132: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

119

Figure 5-10 – Multiplexed beadarray detection process illustration.193

The fluorescent beads are conjugated with antibodies on their surfaces. Several types of beads,

each with its unique fluorescence intensity levels, are mixed together. The bead solution is then

mixed with a sample solution where target analyte would react and bind to the antibodies on the

surface of fluorescent beads. The binding is specific and would only occur if the target analyte is

present in the solution. Otherwise no binding will take place and a negative signal will be

detected. A second reporting reagent with detector antibody is then added to the mixture. Similar

to the previous reaction, if the target analyte is present in the sample solution, specific binding

would occur and the detector antibodies would bind to the target analyte and form a bead

complex as shown in Figure 5-11.

Page 133: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

120

Figure 5-11 – Bead complexes and reagents explanation.

Figure 5-11 illustrates an example of the bead complex after staining is complete. In the case

shown, the fluorescent bead emits in the red channel, at 670 nm. This fluorescent bead is

functionalized with the antibodies, which will capture target antigen floating in the sample

solution. If the antigen is present in the solution, it will conjugate to the surface of the bead via

antibody-antigen binding. A second reagent containing detection or reporter antibody, will then

be added to the mixed solution. In this illustration, the reporter antibody is also pre-labeled with

PE dyes that emits in 585 nm. The reporter antibody will bind to the bead complex, forming the

final bead complex as shown in Figure 5-11. From the graphic illustration, it is clear that the

more antigen molecules in the solution, the more reporter antibody will bound to the surface of

the bead complex, yielding a stronger fluorescence signal in 585 nm channel. The presence of the

target antigen will be decided by the fluorescence signal in 670 nm channel and 585 nm channel,

while the concentration of the target antigen can be determined by the fluorescence intensity

level in 585 nm channel.

Figure 5-12 graphically shows how the bead complex are detected and the target antigen

concentration can be measured and calculated. In a two channel detection scheme a, the first

channel can be used to detect the presence of fluorescent beads as described in Figure 5-11. The

presence of a specific type of bead can be determined by its unique fluorescence intensity. The

fluorescence intensity captured in the second channel is used to quantify the analyte

concentration in the solution. As shown in the illustration, the higher the analyte concentration,

Page 134: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

121

the more bound bead complex will be formed and hence yielding a stronger fluorescence signal.

Using this approach, it is possible to use the first channel to identify the presence of a specific

analyte in a solution and using the second channel to quantify its concentration in the solution.

Figure 5-12 – Two color multiplexed beadarray detection concept illustration. The optical imaging area is

divided into two sections: left is the identification channel and the right is the quantification channel.

5.3.2 Proof of Concept Demonstration

To further demonstrate the multiplexed beadarray concept proposed in previous sections, a

proof-of-principle experiment was designed and implemented. The experiment was designed

based on a commercially available assay kit, BC Cytometric Bead Array (CBA) Mouse

Inflammation Kit (Category number 552364), from Becton Dickinson. This kit can be used to

quantitatively measure Interleukin-6 (IL-6), Interleukin-10 (IL-10), Monocyte Chemoattractant

Protein-1 (MCP-1), Interferon-γ (IFN-γ), Tumor Necrosis Factor (TNF) and Interleikin-12p70

(IL-12p70) protein levels in a single sample. This kit has been optimized for analysis of specific

proteins in tissue culture, EDTA plasma and serum samples.

Page 135: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

122

5.3.2.1 Experimental Setup

As shown in Figure 5-12, the optical detection area is divided into two sub regions. The one on

the left is coated with a band pass filter with center wavelength of 670 nm. This sub region is

used to identify the presence of the target analyte. The sub region on the right-hand side of

Figure 5-12 has a band pass filter with center wavelength of 585 nm. This region is used to

measure target analyte concentration. The detection optics were the same as that used for

CD3/CD4 cell counting, as illustrated in Figure 5-1.

5.3.2.2 Interleukin 6 Protein

Interleukin-6 (IL-6) protein, a blood serum protein, is used in this demonstration. It is an

inflammation marker that includes information on human health. It is an interleukin194-196 that

acts as both a pro-inflammatory cytokine5,197,198 and an anti-inflammatory myokine199. In

humans, it is encoded by the IL6 gene.200

IL-6 is secreted by T cells and macrophages201-204 to stimulate immune response, for example,

during infection and after trauma, especially burns or other tissue damage leading to

inflammation. IL-6 also plays a role in fighting infection, as IL-6 has been shown in mice to be

required for resistance against bacterium Streptococcus pneumoniae.205

A sample with IL-6 proteins is mixed with the fluorescent beads first. The beads are also

conjugated with IL-6 antibodies on their surfaces. IL-6 proteins in suspended in the sample

solution will then bind to the functionalized fluorescent beads. In a second mixing step, detection

antibodies are added to the solution. The detection antibodies are also fluorescently labeled as

shown in Figure 5-10. Upon mixing and incubation, the detection antibodies (anti-IL-6) will bind

to the IL-6 – bead complex as shown in Figure 5-10, forming a complex in Figure 5-11. This

complex has two fluorescence signatures: one is emitting at 585 nm from fluorescent beads while

the other is emitting at 670 nm from the functionalized detection antibodies. The higher the

concentration of IL-6 in the original sample solution, the more binding of IL-6 with antibodies

will take place and hence the fluorescence intensity in the 670 nm channel will be stronger. If

there are no IL-6 present in the solution, then no antibody-antigen binding will occur during

incubation and reaction and hence no fluorescence signal will be observed in the 670 nm

channel. The 585 nm channel is used to identify and confirm the presence of the IL-6 protein

Page 136: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

123

molecules in the solution as shown in Figure 5-12. When a bead complex travels from the first

detection region to the second detection region, if an IL-6 protein is captured by the bead, the

fluorescent signal will be present in both sub panels. Otherwise, only the first sub region, or the

identification sub panel, will show fluorescence signal while the quantification channel will

remain dark since there are no binding between detection antibodies with the target analyte.

5.3.2.3 Sample Preparation

The IL-6 cytokine standards, purchased from BD (http://bd.com/), catalogue number 552364, has

a known concentration of 5,000 pg/mL. The solution was diluted a number of times to obtain

different concentration levels, and incubated for over 3 hours to ensure all antibodies are

conjugated with the fluorescent beads. The dilution procedure that was used to obtain various

concentrations were outlined as follows and also illustrated graphically in Figure 5-13:

Opened one vial of lyophilized Human Inflammatory Cytokine Standards. Transferred the

standard to a 15-mL polypropylene tube. Labeled the tube “Top Standard.”

Reconstituted the standards with 2 mL of Assay Diluent.

Allowed the reconstituted standard to equilibrate for at least 15 minutes at room

temperature.

Gently mixed the reconstituted protein by pipette only. Did not vortex or mix vigorously.

Labeled eight 12 × 75-mm tubes and arranged them in the following order: 1:2, 1:4, 1:8,

1:16, 1:32, 1:64, 1:128, and 1:256.

Pipetted 300 μL of Assay Diluent in each of the 12 ×75-mm tubes.

Performed a serial dilution:

Transferred 300 μL from the Top Standard to the 1:2 dilution tube and mixed thoroughly

by pipette only. Did not vortex.

Continued making serial dilutions by transferring 300 μL from the 1:2 tube to the 1:4 tube

and so on to the 1:256 tube.

Prepared one 12 × 75-mm tube containing only Assay Diluent to serve as the 0 pg/mL

negative control.

Page 137: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

124

Figure 5-13 - Dilution steps performed to obtain different IL-6 concentration levels to be measured.

5.3.2.4 Assay

The prepared samples with different analyte concentrations were then divided into two groups:

one group was processed on a clinical flow cytometer while the other was analyzed using the

optical detection setup shown in Figure 5-1. Each concentration was processed individually after

which data were collected and processed together. The experiment was performed based on the

procedure described below:

Vortexed the mixed Capture Beads and add 50 µL to all assay tubes.

Added 50 µL of the Human Inflammatory Cytokine Standard dilutions to the control

tubes as listed in the following table.

Added 50 µL of each unknown sample to the appropriately labeled sample tubes.

Incubated the assay tubes for 1.5 hours at room temperature, protected from light.

Added 1 mL of Wash Buffer to each assay tube and centrifuge at 200g for 5 minutes.

Carefully and consistently aspirated and discarded the supernatant, leaving approximately

100 µL of liquid in each assay tube.

Page 138: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

125

Added 50 µL of the Human Inflammatory Cytokine PE Detection Reagent to all assay

tubes. Gently agitated the tubes to re-suspend the pellet.

Incubated the assay tubes for 1.5 hours at room temperature, protected from light.

Added 1 mL of Wash Buffer to each assay tube and centrifuge at 200g for 5 minutes.

Acquire standards from lowest (0 pg/mL) to highest (Top Standard) concentration,

followed by the test samples

CBA samples must be acquired on the same day they are prepared. Prolonged storage of

samples, once the assay is complete, can lead to increased background and reduced sensitivity.

Table 11 summarizes the concentration levels that were tested after a series of dilution

described in Section 5.3.2.3.

Table 12 - Concentration levels of IL-6 cytokine tested as

proof of concept demonstration on prototype.Concentration

Tube label (pg/mL) Standard dilution

1 0 (negative control) no standard dilution(Assay Diluent only) 2 20 1:256 3 40 1:128 4 80 1:64 5 156 1:32 6 312.5 1:16 7 625 1:8 8 1,250 1:4 9 2,500 1:2

10 5,000 Top Standard

Samples were run on BD FACSCanto II and a benchtop prototype using the dynamic

imaging approach. Concentration curves were generated from both instruments and compared.

5.3.2.5 Results and Discussions

The BD Cytometric Bead Array Kit included a 6-peak bead populations and a negative control

sample. The 6 peak beads were injected onto the microfluidic devices described in Section 4.5.4

on Page 82. These beads are fluorescent polystyrene microspheres with different intensities.

Their excitation wavelength is 532 nm and emission is centered at 650 nm. The beads were

processed one by one using the optical detection system shown in Figure 5-1, with bellows

Page 139: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

126

actuation described in Section 4.3 on Page 57. The fluorescence intensity of each bead was

detected and recorded. Figure 5-14 shows the histogram of the captured bead population.

Figure 5-14 – Histogram of multiplexed beads detected by the optical detection system described and

developed in this chapter. The beads fluorescence intensities are evenly distributed on a log scale.

Each column in Figure 5-14 represents a bead population that is differentiated by its fluorescence

intensities. From this experimental demonstration, it is clear that the detection system designed

and developed in this thesis can differentiate up to 6 different types of bead population based on

their intensity levels. In a multiplexed bead array implementation with a two wavelength

excitation configuration, this system can be used to detect up to 36 markers simultaneously.

A second set of experiments related to multiplexed beadarray platform was performed to

correlate the detected fluorescence intensity with analyte concentration. The same beadarray

0

2

4

6

8

10

12

4.00 4.20 4.40 4.60 4.80 5.00 5.20 5.40 5.60 5.80 6.00

FREQ

UEN

CY

LOG OF RELATIVE INTENSITY

Beads population identification

Beads 1

Beads 2

Beads 3

Beads 4

Beads 5

Beads 6

Page 140: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

127

standard kit was used in these experiments. Bead number 4 in Figure 5-14 was chosen for this

experiment.

Figure 5-15 - Correlation between fluorescence intensity of reporter or detection antibody and target analyte

concentration. In this experiment, bead type number 4 in the BD kit was used. The target analyte was IL-6

cytokine as described in Section 5.3.2 on Page 121.

From the experiments performed in this work, it is clear that a mulplexed beadarray detection

platform can be implemented to detect and measure protein concentrations in a liquid sample.

With a two laser excitation configuration, this platform can detect up to 36 biomarkers or

analytes simultaneously. The detection capacity can be further expanded by introducing

additional laser sources and optical components. A second excitation wavelength at 405 nm can

be added to the optical system to detect and measure additional markers and target analytes. The

current detection platform has a detection limit of <50 pg/mL. Further optimization can be done

to the detection system to improve the detection limit. The laser source used was a green DPSS

laser pointer emitting at 532 nm with 10 mW output power. A more powerful laser source can

significantly improve the fluorescence signal of the beads. Additional optical components can be

y = 0.0015xR² = 0.9957

0

1

2

3

4

5

6

7

8

10 100 1000 10000

Rel

ativ

e Fl

uo

resc

ent

Inte

nsi

ty

IL-6 concentration (pg/mL)

IL-6 calibration curve

Page 141: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

128

introduced to improve the signal to background ratio as well. The optical beam of the laser

pointer has a Gaussian profile. For multiplex beadarray application, a uniform illumination is

preferred for data processing. Beam shaping components such as microlens arrays or diffractive

optical element should be used to homogenize the beam intensity to achieve a top-hat profile that

is desired for beadarray detection and fluorescence intensity differentiation.

5.4 Conclusion

An optical imaging methodology for particle detection and analysis that functions much like a

handheld cytometer was proposed and demonstrated. By utilizing a unique wide field dynamic

imaging technique, we have eliminated all moving components in a conventional multi-color

fluorescence detection system, such as filter wheels and rotating stages. These mechanical

components are prone to wear and failure. Extreme mobility in a point of care device is a key

technological advantage. An arrayed set of filters placed in front of an optical detector that

enables multi-color fluorescence detection without excess mechanical components. This multi-

color capability makes our platform versatile and simple to combine/implement new tests.

The imaging and detection platform can not only capture cells, but also smaller biomolecules

such as proteins and DNA/RNA molecules. By using beadarray technology, an ELISA like assay

can be developed on this platform. A proof-of-concept demonstration of this expansion was

presented in the last part of this thesis. IL-6, a blood serum protein biomarker, was measured and

quantified in this experiment. This work would enable the development of a handheld or portable

ELISA system that can conduct much wide range of blood testing at point-of-care, such as

cardiovascular diseases, sepsis and diarrhea diseases just to name a few.

Page 142: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

129

Chapter 6 Conclusion and Future Work

A portable imaging and detection system was designed and developed in this work. This system

could be used as the foundation of a handheld point-of-care testing platform for cell, protein and

DNA/RNA analysis. As a demonstration, a CD4 T cell counting test was developed using this

imaging and detection platform. Microfluidic devices were designed, fabricated and

characterized. A direct comparison was also made against the state-of-the-art flow cytometry to

demonstrate the performance of the system.

6.1 Thesis Work Summary

The following list summarizes the key findings and outcomes made in this work:

A capillary microfluidic cell detection and counting device was designed and fabricated. The

microfluidic device relies on surface tension and capillary pumps to actuate sample flow inside

the microchannel. The channel cross section was designed in a way such that uniform flow rate is

maintained throughout the detection and analysis.

Passive and active fluidic actuation mechanisms were investigated in this work and an active

volumetric based actuation approach was designed and developed. A soft elastomer membrane

was used to act as the pneumatic interface between the microfluidic cartridge and the instrument.

The underlying principle was the same as bellows where external force was applied to compress

the air inside the microfluidic device. The reduction in volume resulted in increase in pressure

inside which in turn drove the fluidic sample forward. Similarly, an increase in volume would

lower the pressure inside the microchannel system and hence retract the fluidic sample. With this

approach, it was feasible to actuate fluidic samples inside the microfluidic device backward and

forward as required. Using this approach, an active mixer was designed and fabricated to label

the blood cells with fluorescent dyes on-chip by utilizing Dean’s flow. The active actuation

combined with curved microchannel structures greatly enhanced mixing efficiency. For CD4 T

cell counting test, a conventional labeling process which takes more than 30 minutes in a

benchtop setting was successfully reduced to less than 5 minutes.

Page 143: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

130

A common and critical challenge in POC devices and technologies is about functionalization and

reagent incorporation. Usually this step requires cumbersome and complex protocols which

makes the overall microfluidic device design more difficult to manufacture, hence increasing the

cost of production and reduce the reliability. In this work, a novel reagent handling and

integration process was proposed and demonstrated. A plastic reagent plug had a concave cone

shape end where a droplet of reagent solution with a volume of 5 uL was dispensed on. The

reagent was slow-dried to a pellet form, and the plug was then inserted into the microfluidic

cartridge by snap fit or pressure. This entire process did not require any wet chemistry or surface

treatment on microchannels. The reagent plugs can be transferred and inserted into the device

after the entire microfluidic device is fabricated and packaged, which reduced the risks of

damage of antibody affinity during the packing and bonding process. Finally, the entire process

could be automated to reduce the cost improve ease of integration at high volume.

Antibody concentration is another important parameter in the detection of the target

biomolecules. In the CD4 counting test designed in this work, there was no washing steps

involved in order to reduce the complexity of the fluidic system. Hence the signal-to-background

ratio was higher than for a sample with a washing step. To improve the signal-to-background, a

very effective method was to reduce or optimize the background signal. Since the background

was dominated by the blood sample, where unbound antibody was floating freely, the antibody

concentration had a large impact on the assay performance. Larger antibody concentration would

ensure signal from target cells were bright, but at the same time, it would increase the

background fluorescence as excess antibody would result in a higher background signal. An

optimized level of antibody concentration was determined based on the measured signal-to-

background ratio for a number of concentration levels.

An optical imaging methodology for particle detection and analysis that functions much like a

handheld cytometer was proposed and demonstrated. By utilizing a unique wide field dynamic

imaging technique, we have eliminated all moving components in a conventional multi-color

fluorescence detection system, such as filter wheels and rotating stages. These mechanical

components are prone to wear and failure. Extreme mobility in a point of care device is a key

technological advantage. An arrayed set of filters placed in front of an optical detector that

enables multi-color fluorescence detection without excess mechanical components. This multi-

color capability makes our platform versatile and simple to combine/implement new tests.

Page 144: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

131

The imaging and detection platform can not only capture cells, but also smaller biomolecules

such as proteins and DNA/RNA molecules. By using beadarray technology, an ELISA like assay

can be developed on this platform. A proof-of-concept demonstration of this expansion was

presented in the last part of this thesis. IL-6, a blood serum protein biomarker, was measured and

quantified in this experiment. This work would enable the development of a handheld or portable

ELISA system that can conduct much wide range of blood testing at point-of-care, such as

cardiovascular diseases, sepsis and diarrhea diseases just to name a few.

6.2 Impact

Access to decentralized diagnostics can save millions of lives globally, yet the lack of

supervision and training opportunities for overburdened nurses and community health workers in

remote settings can negatively affect quality of testing and care. This thesis has built the

foundation of portable or handheld point-of-care testing and diagnostic system that will bring lab

quality clinical diagnostic testing to patients at point of care.

Insufficient access to simple, accurate and affordable diagnostic testing in remote health settings

makes it difficult to provide timely, evidence-based clinical care. The result is millions of

preventable deaths from infectious and non-communicable diseases worldwide, reduced

economic growth and limited human development. Remote health is defined as settings that do

not have access to 24-hour laboratory facilities including low and middle-income countries, rural

Ontario hospitals, aboriginal health settings, intensive care units, ambulances, point of entry

diagnostics and military applications.

The system designed and characterized in this thesis can be further developed into a mobile,

rugged, inexpensive and easy-to-use diagnostic platform that produces laboratory quality results.

From single drops of blood, healthcare workers in remote locations can rapidly and accurately

perform tests to diagnose or monitor a range of infectious and non-communicable diseases. The

platform does not require infrastructure such as refrigeration, stable electricity or highly trained

technicians. Simplicity of use allows community health workers with limited training to test

effectively. Affordability makes the platform cost effective in remote health facilities anywhere

in the world. Its small size and weight facilitate patient testing in remote locations, use in mobile

clinics and deployment in hospitals where a platform can be shared between hospital wards and

outpatient clinics.

Page 145: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

132

Currently, in many countries and communities, people do not have access to lifesaving

diagnostic testing because often times there is none: labs that do exist do not operate 24/7 while

sophisticated diagnostic platforms cannot be operated because of lack of trained personnel and

stable electricity and refrigeration, or people live too far from health settings that do offer testing.

The result is millions of preventable deaths from infectious and non-communicable diseases

globally, reduced economic growth, and limited human development. By integrating smart

electronic device functions, including position tracking, bar code scanning and two-way

communication, the envisioned system has the potential to provide a complete and total solution

from patient identification and disease monitoring to patient care, tracking and treatment. This

tool could change the challenges in global health by providing rugged, mobile and simple-to-use

diagnostics in remote health settings that reduce morbidity, decrease healthcare costs, and save

lives.

6.3 Future Outlook

The handheld cytometer developed in this thesis is a very powerful tool. Novel and unique point-

of-care assays can be developed using this platform as demonstrated in the experiments

performed. Additional work is required to further expand the detection capability of the

instrument, as well as development of new assays and tests to target more diseases and

applications.

Further optimization to the optical system can improve the detection limit, sensitivity and

dynamic range, which are all important parameters for many clinical tests. Additional modalities

may also be incorporated into the system such as Raman detection and holographic imaging.

These functionalities could allow more parameters be characterized, reduction of optical

system’s physical size and detection limit.

To develop new and innovative assays, more on-chip fluidic functionalities must also be

researched and developed to satisfy more complex sample preparation processes. On-chip

washing, filtration, dilution, temperature cycling,fluidic valves and more involved fluidic

systems must be incorporated into the current microfluidic architecture to complete the required

steps. In addition, by adding additional sample processing features or capabilities, such as

isothermal amplification, this detection methodology can be useful in developing a portable

nucleic acid test that can be engineered as a molecular diagnostic tool at point-of-care.

Page 146: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

133

References

1 Brown, M. & Wittwer, C. Flow cytometry: principles and clinical applications in

hematology. Clinical chemistry 46, 1221-1229 (2000).

2 Marti, G. E., Stetler-Stevenson, M., Bleesing, J. J. & Fleisher, T. A. in Seminars in

hematology. 93-99 (WB Saunders).

3 Aksel, J. H. S. N. Fluid Mechanics. 2 edn, 164-167 (Springer-Belin, 2004).

4 Van Dilla, M. A. Flow cytometry: instrumentation and data analysis. (Academic Pr,

1985).

5 Schall, T. J. Biology of the RANTES/SIS cytokine family. Cytokine 3, 165-183 (1991).

6 Lodish, H. F. et al. Molecular cell biology. Vol. 4 (WH Freeman New York, 2000).

7 Wang, Y.-N. et al. On-chip counting the number and the percentage of CD4+ T

lymphocytes. Lab on a Chip 8, 309-315, doi:10.1039/B713932B (2008).

8 Barat, D., Benazzi, G., Mowlem, M. C., Ruano, J. M. & Morgan, H. Design, simulation

and characterisation of integrated optics for a microfabricated flow cytometer. Optics

Communications 283, 1987-1992 (2010).

9 Martin, A. J. P. & Synge, R. L. M. A new form of chromatogram employing two liquid

phases: A theory of chromatography. 2. Application to the micro-determination of the

higher monoamino-acids in proteins. Biochemical Journal 35, 1358-1368 (1941).

10 Kunkel, H. G. & Tiselius, A. ELECTROPHORESIS OF PROTEINS ON FILTER

PAPER. The Journal of General Physiology 35, 89-118 (1951).

11 Kuschel, M., Buhlmann, C. & Preckel, T. High-throughput protein and DNA analysis

based on microfluidic on-chip electrophoresis. Journal of the Association for Laboratory

Automation 10, 319-326 (2005).

12 Terry, S. C. A gas chromatography system fabricated on a silicon wafer using integrated

circuit technology. PhD thesis, Stanford, (1975).

13 Terry, S. C., Jerman, J. H. & Angell, J. B. A gas chromatographic air analyzer fabricated

on a silicon wafer. Electron Devices, IEEE Transactions on 26, 1880-1886,

doi:10.1109/T-ED.1979.19791 (1979).

14 van Lintel, H. T. G., van De Pol, F. C. M. & Bouwstra, S. A piezoelectric micropump

based on micromachining of silicon. Sensors and Actuators 15, 153-167,

doi:http://dx.doi.org/10.1016/0250-6874(88)87005-7 (1988).

15 Pol, F. C. M. v. d., Wonnink, D. G. J., Elwenspoek, M. & Fluitman, J. H. J. A thermo-

pneumatic actuation principle for a microminiature pump and other micromechanical

devices. Sensors and Actuators 17, 139-143 (1989).

Page 147: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

134

16 Smits, J. G., Dalke, S. I. & Cooney, T. K. The constituent equations of piezoelectric

bimorphs. Sensors and Actuators A: Physical 28, 41-61 (1991).

17 Shoji, S., Nakagawa, S. & Esashi, M. Micropump and sample-injector for integrated

chemical analyzing systems. Sensors and Actuators A: Physical 21, 189-192 (1990).

18 Van de Pol, F., Van Lintel, H., Elwenspoek, M. & Fluitman, J. A thermopneumatic

micropump based on micro-engineering techniques. Sensors and Actuators A: Physical

21, 198-202 (1990).

19 Shoji, S., Esashi, M. & Matsuo, T. Prototype miniature blood gas analyser fabricated on a

silicon wafer. Sensors and Actuators 14, 101-107 (1988).

20 Esashi, M. Integrated micro flow control systems. Sensors and Actuators A: Physical 21,

161-167 (1990).

21 van der Schoot, B. H. & Bergveld, P. ISFET based enzyme sensors. Biosensors 3, 161-

186 (1988).

22 Verheggen, T. P., Beckers, J. & Everaerts, F. Simple sampling device for capillary

isotachophoresis and capillary zone electrophoresis. Journal of Chromatography A 452,

615-622 (1988).

23 Thomas, L. J. J. & Bessman, S. P. PROTOTYPE FOR AN IMPLANTABLE

MICROPUMP POWERED BY PIEZOELECTRIC DISK BENDERS. ASAIO Journal

21, 516-522 (1975).

24 Spencer, W. J., Corbett, W. T., Dominguez, L. R. & Shafer, B. D. An electronically

controlled piezoelectric insulin pump and valves. Sonics and Ultrasonics, IEEE

Transactions on 25, 153-156, doi:10.1109/T-SU.1978.31006 (1978).

25 Woias, P. Micropumps—past, progress and future prospects. Sensors and Actuators B:

Chemical 105, 28-38 (2005).

26 Manz, A., Graber, N. & Widmer, H. á. Miniaturized total chemical analysis systems: a

novel concept for chemical sensing. Sensors and actuators B: Chemical 1, 244-248

(1990).

27 Van de Pol, F. C. M., Van Lintel, H. T. G., Elwenspoek, M. & Fluitman, J. H. J. A

thermopneumatic micropump based on micro-engineering techniques. Sensors and

Actuators A: Physical 21, 198-202, doi:http://dx.doi.org/10.1016/0924-4247(90)85038-6

(1990).

28 Manz, A. et al. Planar chips technology for miniaturization and integration of separation

techniques into monitoring systems:: Capillary electrophoresis on a chip. Journal of

Chromatography A 593, 253-258 (1992).

Page 148: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

135

29 Harrison, D. J., Manz, A., Fan, Z., Luedi, H. & Widmer, H. M. Capillary electrophoresis

and sample injection systems integrated on a planar glass chip. Analytical chemistry 64,

1926-1932 (1992).

30 Effenhauser, C. S., Manz, A. & Widmer, H. M. Glass chips for high-speed capillary

electrophoresis separations with submicrometer plate heights. Analytical Chemistry 65,

2637-2642 (1993).

31 Harrison, D. J. et al. Micromachining a miniaturized capillary electrophoresis-based

chemical analysis system on a chip. Science 261, 895-897 (1993).

32 Harrison, D. J., Glavina, P. & Manz, A. Towards miniaturized electrophoresis and

chemical analysis systems on silicon: an alternative to chemical sensors. Sensors and

Actuators B: Chemical 10, 107-116 (1993).

33 Seiler, K., Harrison, D. J. & Manz, A. Planar glass chips for capillary electrophoresis:

repetitive sample injection, quantitation, and separation efficiency. Analytical Chemistry

65, 1481-1488 (1993).

34 Northrup, M. A., Ching, M.T., White, R.M., Watson, R.T. in Transducers '93. 924-926.

35 Bousse, L. M., R.J.; Kirk, G.; Dawes, T.; Lam, P.; Bemiss, W.R.; P. J. in Transducers.

916-919.

36 Sobek, D. Y., A.M.; Gray, M.L.; Senturia, S.D. in Micro Electro Mechanical Systems

Proceedings. 219-224.

37 Iliescu, C., Taylor, H., Avram, M., Miao, J. & Franssila, S. Biomicrofluidics 6, 016505

(2012).

38 Washburn, A. L., Luchansky, M. S., Bowman, A. L. & Bailey, R. C. Anal. Chem. 82, 69

(2009).

39 Anderson, R. R. et al. Lab Chip 11, 2088 (2011).

40 Wang, F. & Burns, M. A. Biomed. Microdevices 11, 1071 (2009).

41 Sung, J. H. & Shuler, M. L. Lab Chip 9, 1385 (2009).

42 Chua, J. H., Chee, R. E., Agarwal, A., Wong, S. M. & Zhang, G. J. Anal. Chem. 81, 6266

(2009).

43 Lavrik, N. V., Taylor, L. T. & Sepaniak, M. J. Anal. Chim. Acta 694, 6 (2011).

44 Kutter, J. P. J. Chromatogr., A 1221, 72 (2012).

45 Breadmore, M. C. J. Chromatogr., A 1221, 42 (2012).

46 Kenyon, S. M., Meighan, M. M. & Hayes, M. A. Electrophoresis 32, 482 (2011).

Page 149: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

136

47 Angelescu, D. E. Highly Integrated Microfluidics Design. (2011).

48 Mogensen, K. B. & Kutter, J. P. Optical detection in microfluidic systems.

Electrophoresis 30, S92-100, doi:10.1002/elps.200900101 (2009).

49 Baker, C. A., Duong, C. T., Grimley, A. & Roper, M. G. Bioanalysis 1, 967 (2009).

50 Myers, F. B. & Lee, L. P. Lab Chip 8, 2015 (2008).

51 Mitra, I. et al. Anal. Chem. 84, 3621 (2012).

52 Mainz, E. R. et al. Anal. Methods 4, 414 (2012).

53 Pais, A., Banerjee, A., Klotzkin, D. & Papautsky, I. Lab Chip 8, 794 (2008).

54 Au, A. K., Lai, H., Utela, B. R. & Folch, A. Micromachines 2, 179 (2011).

55 Johnson, T. J., Ross, D. & Locascio, L. E. Anal. Chem. 74, 45 (2002).

56 Jeong, G. S., Chung, S., Kim, C. B. & Lee, S. H. Analyst 135, 460 (2010).

57 Nge, P. N., Rogers, C. I. & Woolley, A. T. Advances in microfluidic materials, functions,

integration, and applications. Chemical reviews 113, 2550-2583 (2013).

58 Grzybowski, B. A., Haag, R., Bowden, N. & Whitesides, G. M. Generation of

Micrometer-Sized Patterns for Microanalytical Applications Using a Laser Direct-Write

Method and Microcontact Printing. Analytical Chemistry 70, 4645-4652,

doi:10.1021/ac9807621 (1998).

59 Ho, S., Haque, M., Herman, P. R. & Aitchison, J. S. Femtosecond laser-assisted etching

of three-dimensional inverted-woodpile structures in fused silica. Optics Letters 37,

1682-1684, doi:10.1364/OL.37.001682 (2012).

60 Ho, S., Herman, P. & Aitchison, J. S. Single- and multi-scan femtosecond laser writing

for selective chemical etching of cross section patternable glass micro-channels. Appl.

Phys. A 106, 5-13, doi:10.1007/s00339-011-6675-7 (2012).

61 Berkowski, K. L., Plunkett, K. N., Yu, Q. & Moore, J. S. Introduction to

Photolithography: Preparation of Microscale Polymer Silhouettes. Journal of Chemical

Education 82, 1365, doi:10.1021/ed082p1365 (2005).

62 Smith, B. & Suzuki, K. Microlithography: Science and Technology, 2nd ed. Vol. 126

(CRC Press, 2007, 2007).

63 Sheats, J. R. & Smith, B. W. Microlithography: Science and Technology. (Marcel

Dekker, 1998).

64 Lorenz, H. et al. SU-8: a low-cost negative resist for MEMS. Journal of Micromechanics

and Microengineering 7, 121 (1997).

Page 150: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

137

65 Guerin L.J.; Bossel M.; Demierre M., C. S. R. P. in Transducers '97. 1419-1421.

66 Johnson, K. et al. Using neutral metastable argon atoms and contamination lithography to

form nanostructures in silicon, silicon dioxide, and gold. Applied physics letters 69, 2773-

2775 (1996).

67 Xia, Y. & Whitesides, G. M. Soft Lithography. Angewandte Chemie International

Edition 37, 550-575, doi:10.1002/(SICI)1521-3773(19980316)37:5<550::AID-

ANIE550>3.0.CO;2-G (1998).

68 Xia, Y. & Whitesides, G. M. SOFT LITHOGRAPHY. Annual Review of Materials

Science 28, 153-184, doi:doi:10.1146/annurev.matsci.28.1.153 (1998).

69 Quake, S. R. & Scherer, A. From Micro- to Nanofabrication with Soft Materials. Science

290, 1536-1540, doi:10.1126/science.290.5496.1536 (2000).

70 Rogers, J. A. & Nuzzo, R. G. Recent progress in soft lithography. Materials Today 8, 50-

56, doi:http://dx.doi.org/10.1016/S1369-7021(05)00702-9 (2005).

71 Attia, U., Marson, S. & Alcock, J. Micro-injection moulding of polymer microfluidic

devices. Microfluidics and Nanofluidics 7, 1-28, doi:10.1007/s10404-009-0421-x (2009).

72 L., N. M. E. W. W. in Proceedings of SPIE - The International Society for Optical

Engineering.

73 Donggang, Y. & Byung, K. Simulation of the filling process in micro channels for

polymeric materials. Journal of Micromechanics and Microengineering 12, 604 (2002).

74 Wimberger-Friedl, R. Injection Molding of Sub-jam Grating Optical Elements.

Specialized Molding Techniques: Application, Design, Materials and Processing, 149

(2008).

75 Pirskanen, J. et al. Replication of sub-micrometre features using microsystems

technology. Plastics, rubber and composites 34, 222-226 (2005).

76 Mazzeo, A., Dirckx, M. & Hardt, D. in ANTEC-CONFERENCE PROCEEDINGS-.

2931.

77 De Mello, A. Focus: Plastic fantastic? Lab on a Chip 2, 31N-36N (2002).

78 Klaassen, E. H. et al. Silicon fusion bonding and deep reactive ion etching: a new

technology for microstructures. Sensors and Actuators A: Physical 52, 132-139 (1996).

79 Juan, W. & Pang, S. High‐aspect‐ratio Si etching for microsensor fabrication. Journal

of Vacuum Science & Technology A 13, 834-838 (1995).

80 Snow, E., Juan, W., Pang, S. & Campbell, P. Si nanostructures fabricated by anodic

oxidation with an atomic force microscope and etching with an electron cyclotron

resonance source. Applied physics letters 66, 1729-1731 (1995).

Page 151: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

138

81 Becker, H. & Gärtner, C. Polymer microfabrication technologies for microfluidic

systems. Analytical and bioanalytical chemistry 390, 89-111 (2008).

82 Becker, H. & Locascio, L. E. Polymer microfluidic devices. Talanta 56, 267-287 (2002).

83 Inc., M. < http://www.micralyne.com> (2012).

84 Gerlach, A. et al. Microfabrication of single-use plastic microfluidic devices for high-

throughput screening and DNA analysis. Microsystem Technologies 7, 265-268 (2002).

85 Erickson, D. & Li, D. Integrated microfluidic devices. Analytica Chimica Acta 507, 11-

26 (2004).

86 Soper, S. A. et al. Surface modification of polymer-based microfluidic devices. Analytica

Chimica Acta 470, 87-99 (2002).

87 Whitesides, G. M. The origins and the future of microfluidics. Nature 442, 368-373

(2006).

88 Soper, S. A. et al. Peer Reviewed: Polymeric Microelectromechanical Systems.

Analytical chemistry 72, 642 A-651 A (2000).

89 Kost, G. J. Guidelines for point-of-care testing. Improving patient outcomes. American

journal of clinical pathology 104, S111-127 (1995).

90 College of American Pathologists POCT toolkit,

<http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportle

ts%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.

contentReference%7D=committees%2Fpointofcare%2Fpoct_toolkit.html&_state=maxim

ized&_pageLabel=cntvwr> (

91 Publications, T. Point of Care Diagnostic Testing World Markets. 493 (2015).

92 Yager, P., Domingo, G. J. & Gerdes, J. Point-of-care diagnostics for global health. Annu.

Rev. Biomed. Eng. 10, 107-144 (2008).

93 Fu, E., Ramsey, S. A., Kauffman, P., Lutz, B. & Yager, P. Transport in two-dimensional

paper networks. Microfluidics and nanofluidics 10, 29-35 (2011).

94 Martinez, A. W., Phillips, S. T., Butte, M. J. & Whitesides, G. M. Patterned paper as a

platform for inexpensive, low‐volume, portable bioassays. Angewandte Chemie

International Edition 46, 1318-1320 (2007).

95 Martinez, A. W. et al. Programmable diagnostic devices made from paper and tape. Lab

on a Chip 10, 2499-2504 (2010).

96 Chin, C. D., Linder, V. & Sia, S. K. Lab Chip 12, 2118 (2012).

Page 152: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

139

97 Daar, A. S. et al. Top ten biotechnologies for improving health in developing countries.

Nature genetics 32, 229-232 (2002).

98 Varmus, H., Klausner, R., Zerhouni, E. & Acharya, T. Grand challenges in global health.

Science 302, 398 (2003).

99 Christianson, A., Streetly, A. & Darr, A. Lessons from thalassaemia screening in Iran:

Screening programmes must consider societal values. BMJ: British Medical Journal 329,

1115 (2004).

100 Sachs, J. & Malaney, P. The economic and social burden of malaria. Nature 415, 680-685

(2002).

101 Laxminarayan, R. et al. Advancement of global health: key messages from the Disease

Control Priorities Project. The Lancet 367, 1193-1208 (2006).

102 Robertson, B. H. & Nicholson, J. K. New microbiology tools for public health and their

implications 1. Annu. Rev. Public Health. 26, 281-302 (2005).

103 Murray, C. J. & Lopez, A. D. Alternative projections of mortality and disability by cause

1990–2020: Global Burden of Disease Study. The Lancet 349, 1498-1504 (1997).

104 Hunter, J. World Bank, World Development Report 1993: Investing in Health. ANNALS-

ASSOCIATION OF AMERICAN GEOGRAPHERS 85, 774-775 (1995).

105 Chin, C. D., Linder, V. & Sia, S. K. Lab-on-a-chip devices for global health: Past studies

and future opportunities. Lab on a Chip 7, 41-57 (2007).

106 Beaglehole, R. & Yach, D. Globalisation and the prevention and control of non-

communicable disease: the neglected chronic diseases of adults. The Lancet 362, 903-908

(2003).

107 Browning, J. D. et al. Prevalence of hepatic steatosis in an urban population in the United

States: impact of ethnicity. Hepatology 40, 1387-1395 (2004).

108 Mellors, J. W. et al. Plasma viral load and CD4+ lymphocytes as prognostic markers of

HIV-1 infection. Annals of internal medicine 126, 946-954 (1997).

109 Badri, M., Lawn, S. D. & Wood, R. Utility of CD4 cell counts for early prediction of

virological failure during antiretroviral therapy in a resource-limited setting. BMC

Infectious Diseases 8, 89, doi:10.1186/1471-2334-8-89 (2008).

110 Peter, T. et al. Challenges in implementing CD4 testing in resource-limited settings.

Cytometry. Part B, Clinical cytometry 74 Suppl 1, S123-130, doi:10.1002/cyto.b.20416

(2008).

111 Thairu, L., Katzenstein, D. & Israelski, D. Operational challenges in delivering CD4

diagnostics in sub-Saharan Africa. AIDS care 23, 814-821,

doi:10.1080/09540121.2010.541416 (2011).

Page 153: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

140

112 MASUDO, T. & Okada, T. Ultrasonic radiation-novel principle for microparticle

separation. Analytical Sciences/Supplements 17, i1341-i1344 (2002).

113 Morgan, H., Holmes, D. & Green, N. G. in Nanobiotechnology, IEE Proceedings-. 76-81

(IET).

114 Yu, C. et al. A three-dimensional dielectrophoretic particle focusing channel for

microcytometry applications. Microelectromechanical Systems, Journal of 14, 480-487

(2005).

115 Choi, S. & Park, J.-K. Continuous hydrophoretic separation and sizing of microparticles

using slanted obstacles in a microchannel. Lab on a Chip 7, 890-897,

doi:10.1039/B701227F (2007).

116 Choi, S., Song, S., Choi, C. & Park, J.-K. Continuous blood cell separation by

hydrophoretic filtration. Lab on a Chip 7, 1532-1538, doi:10.1039/B705203K (2007).

117 Choi, S., Song, S., Choi, C. & Park, J.-K. Sheathless Focusing of Microbeads and Blood

Cells Based on Hydrophoresis. Small 4, 634-641, doi:10.1002/smll.200700308 (2008).

118 Haeberle, S. & Zengerle, R. Microfluidic platforms for lab-on-a-chip applications. Lab on

a Chip 7, 1094-1110, doi:10.1039/B706364B (2007).

119 Squires, T. M. & Quake, S. R. Microfluidics: Fluid physics at the nanoliter scale. Reviews

of Modern Physics 77, 977-1026 (2005).

120 Ichikawa, N., Hosokawa, K. & Maeda, R. Interface motion of capillary-driven flow in

rectangular microchannel. Journal of Colloid and Interface Science 280, 155-164,

doi:http://dx.doi.org/10.1016/j.jcis.2004.07.017 (2004).

121 de Gennes, P.-G. B.-W., Francoise; Quere, David. Capillarity and Wetting Phenomena.

(Springer-Verlag New York, 2004).

122 Ajaev, V. S. & Homsy, G. M. Steady Vapor Bubbles in Rectangular Microchannels.

Journal of Colloid and Interface Science 240, 259-271,

doi:http://dx.doi.org/10.1006/jcis.2001.7562 (2001).

123 Ajaev, V. S. in Interfacial Fluid Mechanics 125-140 (Springer US, 2012).

124 Ajaev, V., Gatapova, E. & Kabov, O. A. Two-phase viscous flows in channels with

chemically patterned walls. Bulletin of the American Physical Society 58 (2013).

125 Mohseni, K., Arzpeyma, A. & Dolatabadi, A. Behaviour of a Moving Droplet under

Electrowetting Actuation: Numerical Simulation. The Canadian Journal of Chemical

Engineering 84, 17-21, doi:10.1002/cjce.5450840104 (2006).

126 Mukhopadhyay, S., Roy, S. S., Mathur, A., Tweedie, M. & McLaughlin, J. A.

Experimental study on capillary flow through polymer microchannel bends for

Page 154: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

141

microfluidic applications. Journal of Micromechanics and Microengineering 20, 055018

(2010).

127 Zimmermann, M., Schmid, H., Hunziker, P. & Delamarche, E. Capillary pumps for

autonomous capillary systems. Lab on a Chip 7, 119-125 (2007).

128 Watson, J. V. Introduction to flow cytometry. (Cambridge University Press, 2004).

129 Raveche, E. et al. Introduction to Flow Cytometry. Flow Cytometry in Drug Discovery

and Development, 3-21 (2011).

130 Alberts, B. et al. Molecular Biology of the Cell (3rd edn). Trends in Biochemical

Sciences 20, 210-210 (1995).

131 Cooper, G. M. & Hausman, R. E. The cell. (Sinauer Associates Sunderland, 2000).

132 Juncker, D. et al. Autonomous microfluidic capillary system. Analytical chemistry 74,

6139-6144 (2002).

133 Dalgaard, P. Introductory Statistics with R. 2nd Edition edn, (Springer, 2008).

134 Mandel, J. The Statistical Analysis of Experimental Data (Dover Publications, 1984).

135 Zimmermann, M., Hunziker, P. & Delamarche, E. Valves for autonomous capillary

systems. Microfluidics and Nanofluidics 5, 395-402 (2008).

136 Klein, R. Material Properties of Plastics. 1st Edition edn, (Wiley-VCH Verlag GmbH &

Co. KGaA, 2011).

137 Malloy, R. A. Plastic Part Design for Injection Molding. 2nd edn, (Hanser Gardner

Publisher, 2010).

138 Pishro-Nik, H. Introduction to Probability, Statistics and Random Processes. (Kappa

Research, LLC, 2014).

139 Lam, Y. C., Gan, H. Y., Nguyen, N. T. & Lie, H. Micromixer based on viscoelastic flow

instability at low Reynolds number. Biomicrofluidics 3, 014106, doi:10.1063/1.3108462

(2009).

140 Bryce, D. M. Plastic Injection Molding: Manufacturing Process Fundamentals. (Society

of Manufacturing Engineers, 1996).

141 Rosato, D. V., Rosato, D. V. & Rosato, M. G. Injection Molding Handbook. (Kluwer

Academic Publishers, 2000).

142 Wikipedia. Injection Molding,

<http://en.wikipedia.org/wiki/Injection_moulding#cite_note-2> (2015).

Page 155: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

142

143 Lee, C.-Y., Chang, C.-L., Wang, Y.-N. & Fu, L.-M. Microfluidic Mixing: A Review.

International Journal of Molecular Sciences 12, 3263-3287, doi:10.3390/ijms12053263

(2011).

144 Buchegger, W., Wagner, C., Lendl, B., Kraft, M. & Vellekoop, M. J. A highly uniform

lamination micromixer with wedge shaped inlet channels for time resolved infrared

spectroscopy. Microfluidics and Nanofluidics 10, 889-897 (2011).

145 Tofteberg, T., Skolimowski, M., Andreassen, E. & Geschke, O. A novel passive

micromixer: lamination in a planar channel system. Microfluidics and Nanofluidics 8,

209-215 (2010).

146 CY Lee, C. L., MF Hung, T Chin, LM Fu. Experimental and numerical investigation into

mixing efficiency of micromixers with different geometric barriers. Material Science

Forum, 391-396 (2006).

147 Chen, Z. et al. Performance analysis of a folding flow micromixer. Microfluidics and

nanofluidics 6, 763-774 (2009).

148 Kang, T. G., Singh, M. K., Anderson, P. D. & Meijer, H. E. A chaotic serpentine mixer

efficient in the creeping flow regime: from design concept to optimization. Microfluidics

and nanofluidics 7, 783-794 (2009).

149 Moon, D. & Migler, K. B. Forced assembly and mixing of melts via planar polymer

micro-mixing. Polymer 51, 3147-3155, doi:10.1016/j.polymer.2010.04.070 (2010).

150 Neerincx, P. E., Denteneer, R. P. J., Peelen, S. & Meijer, H. E. H. Compact Mixing Using

Multiple Splitting, Stretching, and Recombining Flows. Macromolecular Materials and

Engineering 296, 349-361, doi:10.1002/mame.201000338 (2011).

151 C.H. Lin, C. H. T., L.M. Fu. A rapid three-dimensional vortex micromixer utilizing self-

rotation effects under low Reynolds number conditions. Journal of Micromechanics and

Microengineering 15, 935-943 (2005).

152 Singh, M. K., Anderson, P. D. & Meijer, H. E. Understanding and Optimizing the SMX

Static Mixer. Macromolecular rapid communications 30, 362-376,

doi:10.1002/marc.200800710 (2009).

153 Tsai, R.-T. & Wu, C.-Y. An efficient micromixer based on multidirectional vortices due

to baffles and channel curvature. Biomicrofluidics 5, 014103, doi:10.1063/1.3552992

(2011).

154 Hardt, S., Pennemann, H. & Schönfeld, F. Theoretical and experimental characterization

of a low-Reynolds number split-and-recombine mixer. Microfluidics and Nanofluidics 2,

237-248 (2006).

155 Jain, M., Yeung, A. & Nandakumar, K. Induced charge electro osmotic mixer: Obstacle

shape optimization. Biomicrofluidics 3, 022413, doi:10.1063/1.3167279 (2009).

Page 156: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

143

156 Jain, M. & Nandakumar, K. Novel index for micromixing characterization and

comparative analysis. Biomicrofluidics 4, 031101, doi:10.1063/1.3457121 (2010).

157 Daniel Ahmed, X. M., Bala Krishna Juluri, Tony Jun Huang. A fast microfluidic mixer

based on acoustically driven sidewall-trapped microbubbles. Microfluidics and

Nanofluidics, 727-731, doi:10.1007/s10404-009-0444-3 (2009).

158 T-D. Luong, V.-N. P., N-T. Nguyen. High-throughput micromixers based on acoustic

streaming induced by surface acoustic wave. Microfluidics and Nanofluidics 10, 619-625,

doi:10.1007/s10404-010-0694-0 (2010).

159 Michele Campisi, D. A., Francesco Damiani, Paolo Dario. A soft-lithographed chaotic

electrokinetic micromixer for efficient chemical reactions in lab-on-a-chips. Journal of

Micro-Nano Mechatronics 5, 69-76, doi:10.1007/s12213-010-0024-3 (2010).

160 Lim, C. Y., Lam, Y. C. & Yang, C. Mixing enhancement in microfluidic channel with a

constriction under periodic electro-osmotic flow. Biomicrofluidics 4, 014101,

doi:10.1063/1.3279790 (2010).

161 Du, Y., Zhang, Z., Yim, C., Lin, M. & Cao, X. A simplified design of the staggered

herringbone micromixer for practical applications. Biomicrofluidics 4, 024105,

doi:10.1063/1.3427240 (2010).

162 Zhang, Z., Yim, C., Lin, M. & Cao, X. Quantitative characterization of micromixing

simulation. Biomicrofluidics 2, 034104, doi:10.1063/1.2966454 (2008).

163 Xu, B., Wong, T. N., Nguyen, N.-T., Che, Z. & Chai, J. C. K. Thermal mixing of two

miscible fluids in a T-shaped microchannel. Biomicrofluidics 4, 044102,

doi:10.1063/1.3496359 (2010).

164 Y. Wang, J. Z., B. T. F. Chung, P. Dutta. A rapid magnetic particle driven micromixer.

Microfluidics and Nanofluidics, 375-389, doi:10.1007/s10404-007-0188-x (2008).

165 Huang, M.-Z., Yang, R.-J., Tai, C.-H., Tsai, C.-H. & Fu, L.-M. Application of

electrokinetic instability flow for enhanced micromixing in cross-shaped microchannel.

Biomed Microdevices 8, 309-315, doi:10.1007/s10544-006-0034-z (2006).

166 Di Carlo, D. Inertial microfluidics. Lab on a Chip 9, 3038-3046 (2009).

167 Dean, W. R. Note on the motion of fluid in a curved pipe. Philosophical magazine series

7 4, 208-223 (1927).

168 S.A. Berger, L. T., L.S. Yao. Flow in curved pipes. Annual Review of Fluid Mechanics

15, 461-512 (1983).

169 Bringer, M. R., Gerdts, C. J., Song, H., Tice, J. D. & Ismagilov, R. F. Microfluidic

systems for chemical kinetics that rely on chaotic mixing in droplets. Philosophical

transactions. Series A, Mathematical, physical, and engineering sciences 362, 1087-

1104, doi:10.1098/rsta.2003.1364 (2004).

Page 157: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

144

170 Dinh, T. X. & Ogami, Y. Mixing Enhancement by Microrotor in Step Channel. Journal

of Fluids Engineering 133, 021101-021101, doi:10.1115/1.4003420 (2011).

171 Ottino, J. M. Mixing, Chaotic Advection, And Turbulence. Annual Review of Fluid

Mechanics 22, 207-253 (1990).

172 Liu, R. H. et al. Passive mixing in a three-dimensional serpentine microchannel.

Microelectromechanical Systems, Journal of 9, 190-197 (2000).

173 R.C. Anderson, G. J. B., A. Puski, X. Su. in Proceedings Solid-State Sensors and

Actuator Workshop. 7-10.

174 Jones, S. W., Thomas, O. M. & Aref, H. Chaotic advection by laminar flow in a twisted

pipe. Journal of Fluid Mechanics 209, 335-357, doi:doi:10.1017/S0022112089003137

(1989).

175 Aref, H. Stirring by chaotic advection. Journal of Fluid Mechanics 143, 1-21,

doi:doi:10.1017/S0022112084001233 (1984).

176 Aref, H. Chaotic Advection of Fluid Particles. Philosophical Transactions of the Royal

Society of London A: Mathematical, Physical and Engineering Sciences 333, 273-288

(1990).

177 Lin, B. et al. (Google Patents, 2010).

178 Zhu, H., Mavandadi, S., Coskun, A. F., Yaglidere, O. & Ozcan, A. Optofluidic

Fluorescent Imaging Cytometry on a Cell Phone. Analytical Chemistry 83, 6641-6647,

doi:10.1021/ac201587a (2011).

179 Yang, S.-Y. et al. A cell counting/sorting system incorporated with a microfabricated

flow cytometer chip. Measurement Science and Technology 17, 2001 (2006).

180 Fu, A. Y., Spence, C., Scherer, A., Arnold, F. H. & Quake, S. R. A microfabricated

fluorescence-activated cell sorter. Nature biotechnology 17, 1109-1111 (1999).

181 Godin, J. et al. Microfluidics and photonics for Bio‐System‐on‐a‐Chip: A review

of advancements in technology towards a microfluidic flow cytometry chip. Journal of

biophotonics 1, 355-376 (2008).

182 Benton, S. A. & Bove Jr, V. M. Holographic imaging. (John Wiley & Sons, 2008).

183 Collier, R. Optical holography. (Elsevier, 2013).

184 Hariharan, P. Optical Holography: Principles, techniques and applications. Vol. 20

(Cambridge University Press, 1996).

185 Marquet, P. et al. Digital holographic microscopy: a noninvasive contrast imaging

technique allowing quantitative visualization of living cells with subwavelength axial

accuracy. Optics letters 30, 468-470 (2005).

Page 158: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

145

186 Sung, Y. et al. Optical diffraction tomography for high resolution live cell imaging.

Optics express 17, 266-277 (2009).

187 Coskun, A. F., Su, T.-W. & Ozcan, A. Wide field-of-view lens-free fluorescent imaging

on a chip. Lab on a Chip 10, 824-827, doi:10.1039/B926561A (2010).

188 Vykoukal, J., Vykoukal, D. M., Stone, G. P. & Alt, E. U. (Google Patents, 2010).

189 Dalsa. CCD vs CMOS, <https://www.teledynedalsa.com/imaging/knowledge-

center/appnotes/ccd-vs-cmos/> (2015).

190 Access, I. CCD or CIS: The Technology Decision. 6 (2012).

191 Chartier, G. Introduction to optics. (Springer Science & Business Media, 2005).

192 Pedrotti, F. L. & Pedrotti, L. S. Introduction to Optics 2nd Edition. Introduction to Optics

2nd Edition by Frank L. Pedrotti, SJ, Leno S. Pedrotti New Jersey: Prentice Hall, 1993 1

(1993).

193 Luminex xMAP technology, <https://www.luminexcorp.com/clinical/our-

technology/xmap-technology/> (2015).

194 Wikipedia. Interleukin, <http://en.wikipedia.org/wiki/Interleukin> (2015).

195 Schindler, R. & Dinarello, C. A. in Growth Factors, Differentiation Factors, and

Cytokines (ed Andreas Habenicht) Ch. 7, 85-102 (Springer Berlin Heidelberg, 1990).

196 di Giovine, F. S. & Duff, G. W. Interleukin 1: the first interleukin. Immunology Today 11,

13-20, doi:http://dx.doi.org/10.1016/0167-5699(90)90005-T (1990).

197 Lackie, J. A dictionary of biomedicine. (Oxford University Press, 2010).

198 McCoy, C. E. 1. Abstract 2. Introduction 3. Cytokine regulation of miRNA 3.1.

Interleukin-1 3.2. Tumor necrosis factor 3.3. Interleukin-6. Frontiers in Bioscience 16,

2161-2171 (2011).

199 Pedersen, B. K., Åkerström, T. C., Nielsen, A. R. & Fischer, C. P. Role of myokines in

exercise and metabolism. Journal of applied physiology 103, 1093-1098 (2007).

200 Ferguson-Smith, A. C. et al. Regional localization of the interferon-β2B-cell stimulatory

factor 2/hepatocyte stimulating factor gene to human chromosome 7p15-p21. Genomics

2, 203-208 (1988).

201 North, R. An introduction to macrophage activation. Lymphokines 3, 1-10 (1981).

202 Zwilling, B. S. & Eisenstein, T. K. Macrophage-pathogen interactions. (M. Dekker,

1994).

203 Natsuka, S. et al. Macrophage differentiation-specific expression of NF-IL6, a

transcription factor for interleukin-6. Blood 79, 460-466 (1992).

Page 159: A Miniaturized Microfluidic Cytometer Platform for Point ...tspace.library.utoronto.ca/bitstream/1807/80810/1/Dou_James_J... · detection system and particle/cell counting algorithm

146

204 Cromwell, O. et al. Expression and generation of interleukin-8, IL-6 and granulocyte-

macrophage colony-stimulating factor by bronchial epithelial cells and enhancement by

IL-1 beta and tumour necrosis factor-alpha. Immunology 77, 330 (1992).

205 van der Poll, T. et al. Interleukin-6 gene-deficient mice show impaired defense against

pneumococcal pneumonia. Journal of Infectious Diseases 176, 439-444 (1997).