Acoustic and Magnetic Techniques for the Isolation and Analysis of Cells in Microfluidic Platforms by

C. Wyatt Shields IV

Department of Biomedical Engineering Duke University

Date:______Approved:

______W. Monty Reichert, Chair

______Gabriel P. López, Supervisor

______Benjamin B. Yellen

______Stefan Zauscher

______Andrew J. Armstrong

Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biomedical Engineering in the Graduate School of Duke University

2016

ABSTRACT

Acoustic and Magnetic Techniques for the Isolation and Analysis of Cells in Microfluidic Platforms by

C. Wyatt Shields IV

Department of Biomedical Engineering Duke University

Date:______Approved:

______W. Monty Reichert, Chair

______Gabriel P. López, Supervisor

______Benjamin B. Yellen

______Stefan Zauscher

______Andrew J. Armstrong

An abstract of a dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biomedical Engineering in the Graduate School of Duke University

2016

Copyright by C. Wyatt Shields IV 2016

Abstract

Cancer comprises a collection of diseases, all of which begin with abnormal tissue growth from various stimuli, including (but not limited to): heredity, genetic mutation, exposure to harmful substances, radiation as well as poor dieting and lack of exercise. The early detection of cancer is vital to providing life-saving, therapeutic intervention. However, current methods for detection (e.g., tissue biopsy, endoscopy and medical imaging) often suffer from low patient compliance and an elevated risk of complications in elderly patients. As such, many are looking to “liquid biopsies” for clues into presence and status of cancer due to its minimal invasiveness and ability to provide rich information about the native tumor. In such liquid biopsies, peripheral blood is drawn from patients and is screened for key biomarkers, chiefly circulating tumor cells (CTCs). Capturing, enumerating and analyzing the genetic and metabolomic characteristics of these CTCs may hold the key for guiding doctors to better understand the source of cancer at an earlier stage for more efficacious disease management.

The isolation of CTCs from whole blood, however, remains a significant challenge due to their (i) low abundance, (ii) lack of a universal surface marker and (iii) epithelial-mesenchymal transition that down-regulates common surface markers (e.g.,

EpCAM), reducing their likelihood of detection via positive selection assays. These factors potentiate the need for an improved cell isolation strategy that can collect CTCs

iv

via both positive and negative selection modalities as to avoid the reliance on a single marker, or set of markers, for more accurate enumeration and diagnosis.

The technologies proposed herein offer a unique set of strategies to focus, sort and template cells in three independent microfluidic modules. The first module exploits ultrasonic standing waves and a class of elastomeric particles for the rapid and discriminate sequestration of cells. This type of cell handling holds promise not only in sorting, but also in the isolation of soluble markers from biofluids. The second module contains components to focus (i.e., arrange) cells via forces from acoustic standing waves and separate cells in a high throughput fashion via free-flow magnetophoresis. The third module uses a printed array of micromagnets to capture magnetically labeled cells into well-defined compartments, enabling on-chip staining and single cell analysis. These technologies can operate in standalone formats, or can be adapted to operate with established analytical technologies, such as flow cytometry. A key advantage of these innovations is their ability to process erythrocyte-lysed blood in a rapid (and thus high throughput) fashion. They can process fluids at a variety of concentrations and flow rates, target cells with various immunophenotypes and sort cells via positive (and potentially negative) selection. These technologies are chip-based, fabricated using standard clean room equipment, towards a disposable clinical tool. With further optimization in design and performance, these technologies might aid in the early detection, and potentially treatment, of cancer and various other physical ailments.

v

Dedication

To my family, and especially my mother, for teaching me the most important

things in life.

vi

Contents

Abstract ...... iv

List of Tables ...... xiii

List of Figures ...... xiv

Acknowledgements ...... xxvii

Specific Aims ...... xxx

1. Introduction to Microfluidic Cell Sorting ...... 1

1.1 Background and significance of microfluidic cell sorting ...... 1

1.2 Existing approaches for microfluidic cell sorting ...... 4

1.2.1 Fluorescent label-based cell sorting ...... 4

1.2.1.1 Electrokinetics ...... 5

1.2.1.2 Acoustophoresis ...... 10

1.2.1.3 Optical systems ...... 14

1.2.1.4 Mechanical systems ...... 16

1.2.2 Bead-based sorting ...... 18

1.2.2.1 Magnetophoresis ...... 19

1.2.2.2 Acoustophoresis ...... 22

1.2.2.3 Electrokinetics ...... 24

1.2.3 Label-free cell sorting ...... 26

1.2.3.1 Acoustophoresis ...... 27

1.2.3.2 Electrokinetics ...... 29

vii

1.2.3.3 Magnetophoresis ...... 31

1.2.3.4 Optical systems ...... 33

1.2.3.5 Passive cell sorting ...... 34

1.3 Commercialization of existing microfluidic cell sorters ...... 52

1.4 Barriers to commercialization and clinical translation ...... 56

1.4.1 Device-related barriers ...... 56

1.4.2 Commercialization-related barriers ...... 57

1.5 Overcoming the barriers to commercialization ...... 60

1.5.1 Device-related solutions ...... 60

1.5.2 Commercialization-related solutions ...... 62

1.6 Conclusions ...... 64

1.7 Acknowledgements ...... 64

2. Elastomeric Particles for Use in Acoustofluidic Devices ...... 66

2.1 Nucleation and growth synthesis of elastomeric particles ...... 66

2.2 Methods for elastomeric particle preparation ...... 69

2.3 Means of controlling particle size ...... 72

2.4 Means of controlling particle composition ...... 76

2.5 Means of controlling particle surface properties and biofunctionality ...... 78

2.6 Means of measuring and controlling particle acoustic contrast factor ...... 82

2.7 Qualitative verification of particle acoustic contrast factor ...... 88

2.8 Conclusions ...... 90

2.9 Acknowledgements ...... 90

viii

3. Acoustofluidic Devices: Fabrication and Operation ...... 91

3.1 Introduction ...... 91

3.1 Methods of device fabrication ...... 93

3.1.1 Photolithography ...... 94

3.1.2 Deep reactive ion etching ...... 96

3.1.3 Method for cleaning with piranha solution ...... 97

3.1.4 Preparing the borosilicate glass “lid” ...... 98

3.1.5 Anodic bonding ...... 98

3.1.6 Finalizing the acoustofluidic device ...... 100

3.2 Operating the acoustofluidic device ...... 100

3.3 Results and discussion ...... 102

3.4 Conclusions ...... 105

3.5 Acknowledgements ...... 108

4. Acoustofluidic Sequestration of Cells via Elastomeric Particles ...... 109

4.1 Experimental ...... 113

4.1.1 Fabrication of acoustofluidic device ...... 113

4.1.2 Method for synthesis of elastomeric particles ...... 114

4.1.3 Particle binding method ...... 115

4.1.4 Cell preparation ...... 116

4.2 Results and discussion ...... 117

4.2.1 Biofunctional elastomeric particles ...... 117

4.2.2 Acoustic confinement of polystyrene microbeads to the pressure antinodes 118

ix

4.2.3 Acoustic confinement of cells to the pressure antinodes ...... 120

4.3 Conclusions ...... 123

4.4 Acknowledgements ...... 124

5. Magnetic Sorting and Magnetographic Templating ...... 125

5.1 Acoustically enhanced magnetic bead sorting ...... 126

5.1.2 Experimental ...... 127

5.1.3 Performance of acoustically enhanced magnetic bead sorter ...... 129

5.1.4 Conclusions ...... 133

5.2 Magnetographic array for the capture and enumeration of single cells and cell pairs ...... 133

5.2.1 Introduction ...... 133

5.2.2 Experimental ...... 135

5.2.2.1 Magnetographic microchip fabrication ...... 135

5.2.2.2 Sample loading onto the magnetographic microchip ...... 136

5.2.2.3 Positive selection of CD3+ T cells ...... 137

5.2.3 Results and discussion ...... 138

5.2.4 Conclusions ...... 141

5.3 Acknowledgements ...... 142

6. Magnetic Separation of Acoustically Focused Cancer Cells from Blood for Magnetographic Templating and Cellular Analysis ...... 144

6.1 Introduction ...... 145

6.2 Experimental ...... 149

6.2.1 Fabrication and operation of the sorting module ...... 149

x

6.2.2 Fabrication and operation of the templating module ...... 151

6.2.3 Preparation of microparticle samples ...... 153

6.2.4 Preparation of cell samples ...... 154

6.2.5 Model of magnetophoretic cell deflection ...... 155

6.3 Results and discussion ...... 160

6.3.1 Acoustically enhanced magnetic bead separation ...... 160

6.3.2 Magnetic bead capture on the magnetographic array ...... 164

6.3.3 Sorting prostate cancer cells from diluted whole blood ...... 166

6.3.4 Capture of prostate cancer cells for staining and analysis in the magnetographic array ...... 169

6.4 Conclusions ...... 173

6.5 Acknowledgements ...... 175

7. Outlook ...... 176

7.1 Promise of microfluidic cell sorters ...... 176

7.2 Summary ...... 178

7.3 Future directions ...... 183

7.3.1 Integrated modular system for holistic processing of CTCs ...... 183

7.3.2 Isolation of CTCs via negative selection ...... 185

7.3.3 Post-isolation analysis of CTCs: qRT-PCR and CTC culturing ...... 188

7.4 Conclusions and implications of this research ...... 189

Appendix A: Field-Directed Assembly of Patchy Anisotropic Microparticles with Defined Shape ...... 191

A.1 Introduction to field-directed colloidal assembly ...... 191

xi

A.2 Experimental ...... 195

A.2.1 Particle fabrication ...... 195

A.2.2 Particle assembly, modeling and analysis ...... 197

A.3 Results and discussion ...... 198

A.3.1 Microparticle fabrication and characterization ...... 198

A.3.2 Dielectrophoretic assembly of microcubes and microcylinders ...... 200

A.3.3 Magnetic assembly of microcubes and microcylinders ...... 211

A.3.4 Comparison of the results from and magnetic field assembly ...... 217

A.4 Conclusions ...... 222

A.5 Acknowledgements ...... 223

Appendix B: Rapid Self-Assembly of Shaped Microtiles into Large, Close-Packed Crystalline Monolayers on Solid Surfaces ...... 225

B.1 Introduction to passive assembly of shaped colloidal particles ...... 225

B.2 Experimental ...... 226

B.2.1 Fabrication of the microtile particles ...... 226

B.2.2 In situ observation of self-assembly ...... 227

B.2.3 Scanning electron microcopy ...... 227

B.3 Results and discussion ...... 227

B.4 Conclusions ...... 238

B.5 Acknowledgements ...... 239

Biography ...... 276

xii

List of Tables

Table 1: Microfluidic cell sorting technologies that are commercialized, or are approaching commercialization, arranged by type of device and specifying product name, sorting mechanism, company, key metrics of sorting and application notes...... 54

Table 2: Particle size and size dispersity by monomer concentration...... 75

Table 3: Particle size and size dispersity by monomer ratio (8.91 vol.% monomer con.). 75

Table 4: Particle size and size dispersity by spin speed during the growth phase (8.91 vol.% monomer concentration)...... 75

Table 5: Nomenclature of terms used for determining the acoustic contrast factor...... 84

Table 6: Material and acoustic properties of different FMAP particles...... 87

Table 7: Materials for fabrication and basic testing of an acoustofluidic device supporting bulk acoustic standing waves...... 95

Table 8: Model parameters used for the analysis shown in Figure 48C-D...... 157

xiii

List of Figures

Figure 1: Droplet-based microreactor and cell sorter using DEP. Cells expressing and secreting a target antibody (depicted in grey) and cells not expressing a target antibody (depicted in orange) are encapsulated in droplets with a fluorescent detection antibody, incubated off-chip to permit the production of secretion antibodies, and sorted according to the increased fluorescence from the localized packing of detection antibodies on the surface of the bead covered with capture and secretion antibodies. Reprinted with permission from Mazutis et al. [29]. Copyright 2013 Nature Publishing Group...... 8

Figure 2: Direct current (DC) electroosmotic cell sorting. Following laser inspection and cell identification, solvated negative ions in the counterionic layer along the positively charged microchannel floor migrate to the oppositely charged electrode, thereby dragging the surrounding liquid for cell transport[30] to either A) the first outlet (t = Time 0) or B) the second outlet (t = Time 1) [31]...... 10

Figure 3: Acoustofluidic manipulation of cells and particles. In a bulk acoustic standing wave, objects with a A) positive and B) negative φ migrate to the pressure node(s) and antinodes, respectively [49]. C) In a SSAW device, interdigital transducers (IDTs) focus cells along well-defined streamlines according to the driving frequencies of the IDTs (e.g., f1, f2, and f3) for sorting across multiple outlets. D) The cross section of a SSAW device containing four pressure nodes [42]...... 13

Figure 4: Cell sorting by optical force switching. A) When scattering forces (FS) exceed gradient forces (FG) from a focused laser beam, cells are deflected (left); however, when FG exceeds FS, cells are optically trapped (right) [60]. B) A hydrodynamically focused stream of cells is aligned toward the waste outlet whereupon cells of interest detected by laser inspection are captured and displaced by optical tweezers for sorting [67]...... 16

Figure 5: Electromechanical T-switch for cell sorting. A microfabricated cantilever beam reversibly shifts from A) a ‘down’ position (t = Time 0) to B) an ‘up’ position (t = Time 1) when a corresponding pair of electrodes is activated to generate bubbles via isothermal electrolysis, which in turn exerts a mechanical force on the T-switch to redirect fluid flow [74]...... 18

Figure 6: Magnetic isolation of CTCs in microfluidic devices. A) Magnetic beads conjugated with anti-EpCAM are shown to capture and isolate CTCs under free flow in a magnetic field. Reprinted with permission from Hoshino et al. [84]. Copyright 2011

xiv

Royal Society of Chemistry. B) Magnetically labeled CTCs and leukocytes are filtered from blood via deterministic lateral displacement (see Figure 12), focused via inertial focusing (see Figure 10A), and sorted in a magnetic field. Reprinted with permission from Ozkumar et al. [89]. Copyright 2013 Science Translational Medicine...... 21

Figure 7: Acoustic separation of cells using elastomeric particles. A) Elastomeric particles and cells focus to the antinodes and node(s) of an acoustic standing wave, respectively. B) When elastomeric particles bind to target cells, those complexes displace to the pressure antinodes for separation from non-target cells. Adapted from Shields IV et al. [49]. 2014 American Chemical Society...... 24

Figure 8: Multi-target cell sorting using DEP and magnetic trapping. A) Target Cell A is captured with polystyrene beads depicted in green and Target Cell B is captured with superparamagnetic beads depicted in red. B) Target Cell A is removed from non-target cells (depicted in blue) by dielectrophoretic forces and Target Cell B is removed from non-target cells by magnetic trapping. Reprinted with permission from Kim et al. [98]. Copyright 2009 Royal Society of Chemistry...... 26

Figure 9: Sorting by nDEP-assisted field-flow fractionation. A) An array of electrodes displaces cells to equilibrium positions above the floor of a microfluidic channel according to their type (t = Time 0) and B) sorts those cells by propelling them through the channel at rates according to their distance from the wall (t = Time 1) [119]...... 30

Figure 10: Inertial microfluidics for cell sorting in curved microchannels. A) A serpentine microfluidic channel can focus cells into a single streamline. B) A spiral microfluidic channel can sort cells by size (IW and OW indicate the inner wall and outer wall, respectively). C) The cross section of the microfluidic channel showing the balance between lift forces (FL) and Dean drag forces (FD). Reprinted with permission from Kuntaegowdanahalli et al. [160]. Copyright 2009 Royal Society of Chemistry...... 37

Figure 11: Cell sorting by pinched flow fractionation. A) In the pinched segment, cells are first pushed against the wall, and then separated by size upon broadening of the microfluidic channel. B) Cells are aligned in the pinched segment of the channel and follow separate streamlines for sorting by size after exiting the pinched segment. Reprinted with permission from Yamada et al. [166]. Copyright 2004 American Chemical Society...... 39

Figure 12: Cell sorting by deterministic lateral displacement. Large cells (depicted in blue) migrate away from the small cells (depicted in red) in the initial streamline due to the engineered size and spacing of the microposts in the microfluidic channel [180]...... 41

xv

Figure 13: Hydrophoretic cell focusing and sorting. A) A simplified free-body diagram of cells with a low density (shown in blue) separating from cells with a high density (shown in red). The black arrows pointing upwards represent buoyancy forces and the black arrows pointing downwards represent settling forces. B) A top view of the microfluidic channel with herringbone grooves in the ceiling to guide the focusing and separation of cells by density [186]...... 42

Figure 14: Micropatterned ratchets for isolating cells by size and deformability. A) A design of microratchet funnels for fractionating cell populations (dimensions in µm). B) The operation area of the device whereby cells (1, left) enter the chip, (2) reversibly flow through the ratchets for separation by size, and (1, right) exit the chip. C) Schematic of a size exclusion filtration device with various inlets, outlets, and valves (V1-V6). Reprinted with permission from McFaul et al. [191]. Copyright 2012 Royal Society of Chemistry. . 44

Figure 15: Microfluidic filtration mechanisms. Schematic of a A) weir filter, B) pillar filter, and C) cross-flow filter to separate smaller cells (depicted in red) from larger cells (depicted in blue) [193]...... 46

Figure 16: Cell sorting by hydrodynamic filtration. A) Cells are injected into the microfluidic device and are pushed toward the outlets. B) Small cells exit out of the proximal branches whereas C) large cells exit out of the distal branches. Reprinted with permission from Yamada et al. [201]. Copyright 2007 Springer...... 47

Figure 17: Cell sorting by deterministic cell rolling. Target cells (red) interact with the surface, roll across the ridges, and laterally displace toward the gutter side whereas non- target cells (green) flow over the ridges, not interacting with the surface, and exit on the focusing side. Reprinted with permission from Choi et al. [208]. Copyright 2012 Royal Society of Chemistry...... 48

Figure 18: Magnetic self-assembly of biofunctional magnetic beads for isolating rare cells. A) Schematic of a hexagonal array of magnetic ink (left) can guide the self- assembly of magnetic beads conjugated with anti-CD19 mAb in the presence of a vertical magnetic field (right). B) Photograph of the microfluidic device. Optical micrographs of the columns after C) the assembly of magnetic beads, D) the passage of 1,000 Jurkat cells (CD19 negative), and E) the passage of 400 Raji cells (CD19 positive) (scale bar: 80 µm). Reprinted with permission from Saliba et al. [223]. Copyright 2010 Proceedings of the National Academy of Sciences of the USA...... 51

Figure 19: Growth of microfluidic cell sorting technologies in the past 16 years. A) Number of publications versus publication year, organized by articles published in

xvi

engineering, biology and medical journals and B) total number of citations per year that resulted from the query “microfluidic cell sorting” on the Web of Science (Thomson Reuters, Co.)...... 53

Figure 20: Conventional microfluidic cell sorting device setup. Top left: microscope station for inspecting the sorting junction; top right: the field source generator of the microfluidic device (required for most active devices); bottom left: microfluidic device with tubes connected to the inlets and outlets; bottom right: an automatic syringe pump...... 57

Figure 21: Silicone-based particles formed by nucleation and growth synthesis can A) contain various functional groups (e.g., hydroxyl, vinyl and acrylate) for surface modification, B) exhibit narrow size distributions and C) display tunable acoustic properties...... 69

Figure 22: Particles synthesized from Approach I required that Steps 1 and 4 be executed together, followed by Step 5. Particles synthesized from Approach II required Steps 1, 4, and 5 to be executed in sequence. Particles synthesized from Approach III required Steps 1-5 to be executed in sequence...... 70

Figure 23: A) SEM image of particles synthesized from VTMOS. B) Bright-field (left) and fluorescence (right) images of Nile red-impregnated particles synthesized from a 96:1 molar ratio of DMODMS:TMOS. C) Particles from (B) and a mammalian cell (KG-1a, dyed green) focused along the antinodes and node of an acoustic standing wave, respectively (dashed white lines represent the walls of the microchannel). Particles synthesized from different concentrations of (D, E) TMOMS and (F) a 96:1 molar ratio of DMODMS:TMOS, displaying an average size of 230±17 nm, 2.6±0.1 µm and 14.8±0.8 µm, respectively...... 73

Figure 24: Mean particle size as a function of the A) monomer concentration for two DMODMS:TMOS ratios (8:1 and 96:1), B) molar ratio of DMODMS:TMOS and C) spin speed during the growth phase. Error bars represent the standard deviation of the mean...... 74

Figure 25: ATR-FTIR spectra of particles synthesized from A) TMOMS, B) an 8:1 molar ratio of TMOMS:VTMOS and C) an 8:1 molar ratio of TMOMS:AcTMOS...... 77

Figure 26: A) Zeta potential and size of particles made from TMOMS and VTMOS. B) Histograms of the number of adsorbed molecules of SA per particle as measured by a flow cytometer (particles synthesized from a 24:1 molar ratio of VMDMOS:TMOS). From

xvii

top left to bottom right: bare particles, bare particles with adsorbed SA, biotinylated particles with adsorbed biotin-blocked SA, and biotinylated particles with adsorbed, unblocked SA...... 81

Figure 27: Schematic of the acoustic displacement and optical tracking system for measuring the acoustic properties of particles via a traveling wave. Particles travel upward (i.e., against gravity) and are displaced by the incoming traveling wave by an extent proportional to their acoustic properties. Particles are tracked in real time using a water-immersion objective focused on the particle stream coupled to a high-speed camera...... 83

Figure 28: Properties of FMAP particles. A) Fundamental properties of particles by monomer composition: density, pp (solid , left) and compressibility, β (dashed line, right). B) Acoustic properties of particles by monomer composition: acoustic response

parameter, ψp(ρ,c) (solid line, left) and acoustic contrast factor, φp (dashed line, right). .. 88

Figure 29: Evaluation of particles flowing through an acoustic standing wave. A) Fluorescence intensity profiles across the width of an acoustofluidic channel for different particles flowing through the channel as shown in (B) (300 µL/min flow rate; 30 Vpp sinusoid waveform; 2.96 MHz; λ ≈ 500 µm). B) Micrographs of fluorescent particles flowing at the node (top) and antinodes (bottom) of an acoustic standing wave...... 89

Figure 30: Acoustofluidic device supporting bulk acoustic standing waves. Schematic views of the top (A) and bottom (B) of a device comprised of an etched silicon substrate fused to a borosilicate glass “lid”, polydimethylsiloxane (PDMS) blocks connected to silicone tubing and a piezoelectric transducer soldered to wires glued to the bottom of the device. Photographs of the top (C) and bottom (D) of the device are also shown. ... 102

Figure 31: Acoustic focusing of particles with positive and negative acoustic contrast factors. A) Prior to actuation of the PZT transducer, particles with a positive acoustic contrast factor (10 µm, yellow-green polystyrene beads) flowing at 100 µL/min occupied the width of the microchannel. B) After the PZT transducer is actuated (Vpp = 40 V and f = 2.366 MHz), the particles in (A) are shown to focus along the pressure node of the standing wave. C) Particles with a negative acoustic contrast factor focused along the pressure antinodes of the standing wave in the absence of applied flow (Vpp = 40 V and f = 2.366 MHz)...... 103

Figure 32: Focusing performance of an acoustofluidic device. Fluorescence intensity plots of polystyrene beads (shown in Figure 31A-B) are shown for (A) various flow rates (ranging from 0 to 1,000 antL/min) peak- towith-peak a const voltage of 40 V and (B)

xviii

various applied voltages (ranging from 0 to 50 V) with a constant flow rate of 100 µL/min...... 105

Figure 33: Schematic diagram illustrating acoustic confinement of cells in a rectangular acoustofluidic device (not to scale). A) Unbound cells and elastomeric particles in an acoustic standing wave. B) Bound cell-particle immunocomplexes in an acoustic standing wave when the acoustic radiation force acting on the elastomeric particle is greater than that on the cell. C) The region under the solid line represents the locus of properties (effective (net) size and the acoustic contrast factor (φp)) of an elastomeric particle(s) that can displace a typical mammalian cell (13.0 μm diameter) toward a pressure antinode...... 112

Figure 34: Method for binding SA-adsorbed particles to biotinylated anti-CD34 labeled KG-1a cells. Elastomeric particles surface-functionalized with streptavidin specifically bind to cells presenting biotinylated antigens...... 116

Figure 35: Elastomeric particles obtained through nucleation and growth synthesis. A) Optical micrographs of elastomeric particles (. ≈13 µm) used for the confinement of cells. B) Elastomeric particles (red, with adsorbed SA labeled with Alexa-Fluor® 546) transported to the antinodes of an acoustic standing wave within a microfluidic channel (no flow). The locations of the channel walls (dashed lines) and the features of the acoustic standing wave (solid lines) are denoted...... 118

Figure 36: Fluorescence intensity profiles of particles across the microchannel cavity under flow. The distributions of (A) SA-adsorbed elastomeric particles (dia. ≈13 µm) at 15 µL/min, (B) biotinylated polystyrene microbeads (diameter ≈5.2 µm) at 100 µL/min, and (C) complexes of particles in a bound to particles in b at 100 µL/min are shown across the acoustofluidic channel, where W denotes the width of the microchannel (250 µm)...... 120

Figure 37: Acoustophoresis of KG-1a cells and elastomeric particles. A) Bright-field and (B) the corresponding fluorescent images of non-fluorescent, SA-adsorbed elastomeric particles (dia. ≈13 µm) bound to green stained KG-1a cells. C) Complexes of KG-1a cells and elastomeric particles migrating to the pressure antinodes in response to imposition of the acoustic standing wave (≈2 µL/min). D) Fluorescence intensity profiles across the channel width indicating the acoustophoresis of KG-1a cells unbound (left) and bound (right) to SA-adsorbed elastomeric particles in an acoustic standing wave. Fluorescent images within a microfluidic device of: (E) green stained KG-1a cells and Nile red embedded elastomeric particles without SA and focusing to the node and antinodes and node of an acoustic standing wave, respectively, and (F) non-fluorescent SA-adsorbed

xix

elastomeric particles bound to green, anti-CD34-biotin labeled KG-1a cells separated from red stained KG-1a cells without anti-CD34-biotin at the antinodes and nodes of an acoustic standing wave, respectively...... 122

Figure 38: Acoustically enhanced magnetic sorting. A mixture of particles starts at the top and flows down towards the channel bifurcation. The yellow and red particles represent magnetic and nonmagnetic entities, respectively. The black lines and white arrow represent the acoustic standing wave and the magnetic field gradient, respectively...... 127

Figure 39: Control experiments for the acoustically enhanced magnetic sorter. A mixture containing approximately a 1:1 ratio of magnetic to nonmagnetic beads was passed through the device without forces from an acoustic standing wave or magnetic field. Analysis from a paired t test shows no significant difference in the ratio of magnetic to nonmagnetic beads across the inlet or either outlet (p = 0.965; n = 5)...... 129

Figure 40: Acoustic focusing and magnetic sorting of polystyrene particles. Micrographs of polystyrene particles with the lead zirconate titante (PZT) transducer A) turned off and B) turned on. C) Schematic of the acoustically enhanced magnetic particle sorting chip (image not to scale). D) Micrograph of the channel bifurcation, where magnetic beads are shown flowing towards the magnetic outlet and nonmagnetic beads are shown flowing towards the nonmagnetic outlet. Green and red represent magnetic and nonmagnetic beads, respectively, in all images; the sinusoidal lines represent an acoustic standing wave. All scale bars are 250 μm...... 131

Figure 41: Performance of the acoustically enhanced magnetic sorter. The left and right columns represent the outputs from the magnetic and non-magnetic outlets, respectively. A mixture containing a 1:1 ratio of magnetic particles to non-magnetic particles at increasing concentrations was passed through the device at A) 20, B) 200, C) 2,000, D) 20,000 and E) 55,000 particles/sec. Each condition (A-E) indicates the fluorescence intensity of the 533/30 nm laser versus the 585/40 nm laser from a flow cytometer (number of trials for each throughput (A-E), n = 5)...... 132

Figure 42: Magnetographic array for single cell analysis. A) SEM image of the dumbbell- shaped well pairs for capturing magnetically labeled cells. B) Photograph of the finished device. C) An array of well pairs displaying a pitch of 60x120 µm before (top) and 10 min after the deposition of magnetic beads (bottom)...... 135

Figure 43: Sample loading onto the magnetographic microchip. A) Samples were loaded onto the microchip (top) dropwise to the top, (middle) injected underneath a coverslip,

xx

and (bottom) injected into the crevice of a titled coverslip. B) The capture efficiency of the three modes of sample loading. The capture efficiency of sample loading without a coverslip were found to have a significantly higher efficiency (p < 0.001) (n = 5)...... 137

Figure 44: Positive magnetic isolation of lymphocytes against the CD3 antigen yields pure cell populations. A) 14.4% and B) 97.8% of cells in the non-target and target populations were CD3+, respectively...... 137

Figure 45: CD4:CD8 analysis of CD3+ lymphocytes. A) Photograph of the magnetographic device activated by permanent magnets (covered with green tape). The CD4:CD8 ratio determined by the (B) magnetographic microarray and (C-D) flow cytometry was 1.84 and 1.74, respectively...... 139

Figure 46: Programmed pairing of magnetic beads and CD3+ lymphocytes. A) Schematic of the magnetographic cell pair isolations. B) Polarized micromagnets isolate cells of one type to one side in a vertical magnetic field and then cells of a second type to the other side when the field is reversed. C) Fluorescent image of magnetically trapped green stained (top) and red stained (bottom) cell pairs. D) SEM image of magnetically labeled cells in the microwells. E) Capture accuracy of magnetic bead pairs (each color represents the field strength of the reversed field). F) Change in the capture accuracy (loss) of initially captured beads after reversing the magnetic field. The capture accuracy of (G) magnetically labeled cell pairs and (H) the second magnetically labeled cell (for (E-H): n = 5; time starts from the deposition of the second set of cells or beads)...... 141

Figure 47: Schematic of the microfluidic system designed to focus, separate and template cancer cells. Diluted blood (sans erythrocytes) moves through the device, from left to right, whereupon cells are acoustically focused to the center of the microchannel. The black dotted lines represent a half wavelength acoustic standing wave. Magnetically labeled cells are then deflected in the non-resonant portion of the microchannel via a gradient magnetic field, indicated by the fading black arrow. Finally, an array of micromagnets, indicated by the blue rectangles, locally attracts magnetically labeled cells into microwells, indicated by the red boxes, for on-chip staining and analysis. The modules for cell sorting and cell templating were made and operated separately. (N.B., schematic is not to scale)...... 149

Figure 48: Model of magnetophoretic cell deflection assuming a uniform flow velocity profile and a constant magnetic field gradient. A) Scaled illustration of the microfluidic system in which a streamline of magnetically labeled cells (green) separates from a streamline of unlabeled cells (red) in a gradient magnetic field (shaded black arrows). Distances for WR, WNR, CD and LNR are 255, 893, 392 and 10,924 μm, respectively. B)

xxi

Histograms of the numbers of magnetic beads (Nb) bound to LNCaP cells (top; n = 70) and diameters of LNCaP cells (bottom; n = 151). C) Calculated trajectories of LNCaP cells bound to different numbers of magnetic beads, ranging from 0 to 30, at a flow rate of 100 μL/min through the deflection portion of the microchannel (the thick orange line indicates the critical number of beads required for separation (i.e., where D ≥ CD)). D) Calculated trajectories of LNCaP cells at a constant flow rate of 50 μL/min (orange) and 200 μL/min (green) through the deflection portion of the microchannel with Nb = 15±5 beads/cell...... 158

Figure 49: Acoustically enhanced magnetic bead sorting. A) Photograph of the device used to focus and separate particles and cells. Wires leading to a piezoelectric transducer and tubing leading to the microchannel protrude from the back. One of two separation magnets is shown adjacent to the deflection portion microchannel. B) Relative intensity plots display the full width at half maximum (FWHM) of particles across the microchannel at different positions and conditions: B1 and B2 represent the resonant portion of the microchannel with the acoustics off and on, respectively; B3 represents the deflection portion of the microchannel with the acoustics on (f = 2.90 MHz; Vpp = 40 V). C) Corresponding micrographs of fluorescent particles in the device with the acoustics off (top) and on (bottom). D) Performance of the device for separating a 1:1 ratio of magnetic particles to non-magnetic particles at throughputs ranging from 20 to 55,000 particles/sec by altering the volume fraction of particles (displayed along the top of the plots in D), at a flow rate of 200 μL/min (n = 5)...... 162

Figure 50: Magnetographic bead templating. A) Schematics of the magnetographic templating module where magnetic particles were injected (top), captured (middle) and retained for analysis (bottom). Vertical black arrows indicate the direction of the uniform magnetic field, and the horizontal white arrows indicate the direction of polarization of the micromagnets. Parabolic black arrows indicate the direction of fluid flow. B) Capture accuracy of the magnetographic templating module through three conditions: Experiment (i.e., micromagnets are polarized, and the chip is in a vertical magnetic field; n = 2,098 particles), Control 1 (i.e., micromagnets are polarized, but the chip is not in a vertical magnetic field; n = 610 particles) and Control 2 (i.e., micromagnets are not polarized, nor is the chip in a vertical magnetic field; n = 857 particles). Labels on the x- axis correspond to events where one particle is captured per microwell (left), multiple particles are captured per microwell (middle) and no particles were captured, but were within the capture radius (right). C) Photograph of the magnetographic templating module. D) Fluorescent micrographs of the array prior to (left) and after (right) the capture of magnetic particles...... 165

xxii

Figure 51: Separation of human prostate cancer cells from erythrocyte-lysed blood. A) Schematic illustration of the microfluidic device for the acoustically enhanced, magnetic cell separation of cancer cells from erythrocyte-lysed blood. B) Pictures of whole porcine blood first doped with LNCaP cells (6 mL, left), then diluted to 60 mL with erythrocyte lysing buffer (middle) and then concentrated to 6 mL after erythrocyte lysis (right). C) Performance of the device to enrich non-magnetic cells in the non-magnetic outlet (green) and prostate cancer cells in the magnetic outlet (blue) at two different flow rates (left) (initial ratio was 150:1 leukocytes to prostate cancer cells). Separation efficiency of the device to isolate magnetically labeled cells in the magnetic outlet (right) at two different flow rates. D) Example photographs of cultured prostate cancer cells 24-72 hours after separation from blood...... 169

Figure 52: Assembly of human prostate cancer cells in microwells of the magnetographic array for staining and analysis. A) Schematic of the magnetographic array for capturing and organizing magnetically labeled cells (magnetic field gradient indicated by the large, yellow arrow). B) Plot of the pressure and force on a cell caused by a single magnetic bead (MyOneTM Dynabeads®, SA T1), determined using COMSOL software. C) Corresponding magnetic field strength simulations of a single magnetic bead at various distances from the field maxima of a micromagnet using COMSOL software. D) Plot of the net magnetic force on a single cell generated by 15 (blue line) and 30 (red line) magnetic beads. E) Overlaid bright field and fluorescence micrographs of magnetically captured prostate cancer (LNCaP) cells (or clusters of cells), stained on-chip with calcein AM viability stain...... 172

Figure 53: Integrated microfluidic device for the on-chip isolation and analysis of rare cells. The chip contains three modules: (i) a debulking module to remove excess white blood cells via elastomeric particles, (ii) an enriching module to purify rare cells (e.g., CTC candidates), and (iii) a templating module to allow cell immunostaining and automated cellular analysis. Elastomeric particles, white blood cells, rare cells and magnetic particles are indicated by the colors of blue, white, green and black, respectively. The bolded black and shaded black arrows indicate the direction of flow and the direction of the gradient magnetic field, respectively. The dotted lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale...... 184

Figure 54: Debulking leukocytes with elastomeric particles and acoustophoresis. Objects in white, green and blue represent white blood cells, CTC candidates and elastomeric particles, respectively. The bold arrow indicates the direction of flow and the dotted

xxiii

lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale...... 186

Figure 55: Debulking cells with magnetic particles and magnetophoresis. Objects in white, green and black represent white blood cells, CTC candidates and magnetic particles, respectively. The bolded black and shaded black arrows indicate the direction of flow and the direction of the gradient magnetic field, respectively. The dotted lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale...... 187

Figure 56: Schematic depicting steps in the fabrication and assembly of anisotropic, patchy particles. A) Anisotropic microparticles with tunable aspect ratios and vertex angles are generated via photolithography, (B) patterned with various metals, and (C) suspended in liquids and assembled to form distinct structural motifs...... 195

Figure 57: Anisotropic particles. Representative SEM images of SU-8 particles include (A) 10 µm cubes, (B) 10 µm tall cylinders, (C) 10 µm tall hexagonal prisms, (D) 25 µm tall cylinders top-coated with 15 nm Cr and 50 nm Au before removal from silicon wafer substrate, and (E) those cylinders after removal. Image (F) is an optical micrograph of 10 µm parallelepipeds in suspension. Scale bars in all micrographs are 50 μm...... 199

Figure 58: Atomic percentage of detected by EDS along the side of different 25 µm tall cylinders. Smoothed data from the side of an uncoated cylinder (blue), a top-coated cylinder (green), and a side-coated cylinder (red). Metal films coated on surfaces orthogonal to the source were nominally 15 nm of Cr and 50 nm of Au...... 200

Figure 59: Schematic of the inverted DEP assembly configuration. The DEP assembly cell is loaded, inverted, and then excited in an AC electric field with a gradient oriented against the direction of gravity. Only frequencies above 100 Hz were studied, which eliminated particle migration across the cell via direct electrokinetic effects (electrophoresis and electroosmosis) between charged particles and the electrodes. .... 202

Figure 60: Optical micrographs of DEP assemblies of microcubes (10µm) and microcylinders. The direction of the field indicated in (A) is consistent in all images. (A) Contrasting configuration of top-coated cubes when apart and assembled (20 kV m-1 at 10 kHz), (B) chains of top-coated cubes (4 kV m-1 at 1 kHz), (C) chains of top-coated cubes (15 kV m-1 at 1 kHz), (D) chains of top-coated cubes (15 kV m-1 at 10 kHz), (E) chains of uncoated cubes, (F) kinked clusters of angle- (or two-side-) coated cubes (10 kV m-1 at 1 kHz), (G) typical orientations of top-coated cylinders (height and dia. = 10 µm; 10 kV m-1 at 1 kHz), and (H) wires of side-coated cylinders (height = 25 µm, dia. = 10

xxiv

µm; 15 kV m-1 at 10 kHz). Observed assembly motifs are illustrated as an inset of the corresponding image. The scale bars in each image correspond to 50 μm...... 204

Figure 61: A) Simulated electric field intensity landscapes around top-coated cubes in AC electric fields at increasing frequencies (100 Hz – 10 MHz). B) Total electrical energies of the various expected assembly configurations at 20 kHz. The field direction is vertical in all figures and the black bars represent the metallic patches...... 209

Figure 62: Optical micrographs of assemblies of microcubes (10 µm) and microcylinders (dia. = 5 µm, height = 10µm) within magnetic fields. The direction of the field indicated in (A) is consistent in all images. Solutions were in either water or ~1.0 vol.% ferrofluid. A) Zigzagged chains of top-coated cubes in water, (B) elongated rods of top-coated cubes in ferrofluid, (C) zigzagged chains of top-coated cylinders in water, (D) uncoated cylinders in ferrofluid, (E) zigzagged chains of side-coated cylinders in water and (F) edgewise chains of top-coated cylinders in ferrofluid. The assembly motifs observed most frequently in each case are illustrated in the top right-hand corner of each image. Scale bars are 25 μm...... 213

Figure 63: Effect of increasing ferrofluid concentration on particle assembly and local magnetic field strength. A) Relative amount of zigzagged structures formed from top- coated cylinders in increasing concentrations of ferrofluid, and (B) simulations of the magnetic field strength distribution around individual top-coated cubes in increasing concentrations of ferrofluid...... 215

Figure 64: A) Overview of the fabrication and the droplet-assisted self-assembly approach of microtile particles. Monodisperse, shape-anisotropic microtiles were fabricated from SU-8 photoresist and were suspended in water with surfactant. The suspension of microtiles was added to a droplet of chloroform, causing the particles to self-arrange along the liquid-liquid interface. A co-solvent was then added to mix the solutions, to break the assembly into crystallites and to rapidly induce the self-assembly of those crystallites at the liquid-air interface. The solvent mixture was then evaporated such that the polycrystalline monolayer remained stably intact on the surface of the glass. SEM images of unassembled (B) circular, (C) hexagonal and (D) square microtiles with an aspect ratio of 0.2 are displayed as examples of the microtiles made from photolithography...... 229

Figure 65: A) Mechanisms for crystal formation: (i) the particle suspension is deposited on a chloroform droplet; (ii) a stable crystalline monolayer forms at the interface of chloroform and water; (iii) a co-solvent (ethanol) is added to mix the two phases and break the crystal into crystallite fragments; (iv) the crystallite fragments re-emerge

xxv

assemble along the top of the fluid mixture; and (v) the solvent mixture evaporates in minutes, resulting in a polycrystalline monolayer stably supported on the glass substrate. Images (vi-x) show optical micrographs (i.e. square microtiles) of the corresponding events depicted schematically in (i-v). Image x shows a large area of a crystal monolayer. B) A schematic representation of three strategies that did not result in well-defined crystals is shown. C) A schematic of three strategies that resulted in well- defined crystals is shown...... 232

Figure 66: An example of bright field images of crystal monolayers self-assembled using thin circle tiles. 4x image (left) shows the long-range coverage of the self-assembled crystal. 10x images (right) were used for quantification...... 234

Figure 67: A) SEM images (left) and FFTs (right) of assemblies of (i) circular, (ii) hexagonal and (iii) square microtiles with an aspect ratio (height:width) of 0.2 as well as of assemblies of (iv) circular, (v) hexagonal and (vi) square microtiles with an aspect ratio of 0.9 supported on a solid substrate. B) Colorized images of large, polycrystalline monolayers. Analysis of (C) monocrystal size and (D) total surface coverage of the crystals made from the different tiles. The statistically analysis was performed using bright field images...... 236

Figure 68: A) Circular and (B) hexagonal tiles were assembled using different particle concentrations. Particle suspensions of both types, with concentrations of (i) 2x106, (ii) 1x107, (iii) 5x107 and (iv) 2.5x108 particles/mL, were assembled on a chloroform droplet using the methods described in Figure 65C(i) of the manuscript. Scale bars represent 20 µm...... 238

xxvi

Acknowledgements

I would like to express my sincere gratitude to my committee for their time, insight and expertise. By their guidance, I have learned a great deal. I would like to thank Prof. William M. Reichert for demonstrating how to make an impact outside of the walls of a research lab. I thank Prof. Stefan Zauscher for his passionate approach to teaching, mentorship and outreach. I thank Prof. Andrew J. Armstrong for sharing his excellent vision for both fundamental and applied research. And I thank Prof. Benjamin

B. Yellen for his years of mentorship, intellectual input and outstanding creativity.

I thank the many exceptional collaborators I was able to work with, including

Profs. Orlin D. Velev (NC State Univ.), David M. Murdoch, Thomas Laurell (Lund Univ.,

Sweden), Benjamin A. Evans (Elon Univ.), Patrick Charbonneau, Benjamin J. Wiley,

Peter Kingshott (Swinburne Univ. of Tech., Australia), Paul A. Dayton (Univ. of North

Carolina), Kennita A. Johnson (Univ. of North Carolina), Joseph B. Tracy (NC State

Univ.), Steven W. Graves (Univ. of New Mexico), Nick J. Carroll, Ashutosh Chilkoti and

Nan M. Jokerst. I would also like to thank the excellent leadership of (and many great individuals within) the National Science Foundation’s Research Triangle Materials

Research Science and Engineering Center (MRSEC).

I would like to thank everyone in the López lab. Foremost, I would like to thank

Drs. Leah M. Johnson and Lu Gao for laying the groundwork from which I could learn

xxvii

and build my research portfolio. Their mentorship was hugely beneficial to my overall development. I would like to thank the members of “team acoustics” who I have enjoyed working with to the very end, including Korine Ohiri, Daniela Cruz and Dr.

Lizzy Szott. I thank the many other fantastic personalities of the López lab group, including Alice (Linying) Li, Wei Han and Drs. Phanindhar Shivapooja, Vrad Levering,

Ali Ghoorchian, Pearlson Prashanth, Morgan Harris and Qian Yu. I would like to give

special thanks to the talented undergraduate and masters students with whom I

worked, including Stephan Abdo, Ce Bian, Eric Essoyan, Jin Huang, Jonathan Liu,

Carissa Livingston, Oluwatosin Omofoye, Crystal Owens, Christopher Reyes, Aura

Rodriguez, Maggie Smith, Dana Sun, Jeffrey Wang, Jack White, Amy Xiong and Amber

Zhang. My interaction with these students was the most rewarding aspect of my time in graduate school. I would also like to give thanks to the “unsung heroines” for their work, including Elaine Fulton, Carla Sturdivant and especially Katie Krieger.

I would like to thank my core group of friends. In addition to the individuals listed already, all of whom I consider friends, I would like to thank Hrishikesh Rao,

Nicholas Tang, Jeffrey Schaal, Kedar Prabhudesai, Jaydeep Srimani, Rachel Lance, Nick

Bottenus, Tianhe (Cheng) Zhang and Bryan Howell for the many happy memories of running, camping, traveling and enjoying each other’s company. I would like to thank my friends from afar as well, including Matt Miller, Jae Park, Chris Johnson, Bill Donati,

Kenny Erwin and especially Travis Johnston.

xxviii

I would like to heartily thank my loving family. Foremost, I thank my mom and stepdad, Karen Shields and Larry Priest, for their unconditional love and support. I would not be here today if it were not for them, especially my mom, who has been there for me at every step. I thank my dad, Charlie Shields, for his love, encouragement, stimulating conversations and thoughtfulness. I thank my stepmom, Shean Shields, and my four wonderful younger siblings, John, Jeremy, Stephen and Beth Anne, all of whom have been an unending source of love and laughter. I thank my grandmother, Maxine

Harris, and my uncles, aunts and cousins. I especially thank my one-of-a-kind brother,

Lance Shields, for walking through this journey of life with me, being by my side and

always sharing a good laugh.

Most importantly, I would like to give my highest and most sincere thanks to my

outstanding advisor, Prof. Gabriel P. López. Gabriel devoted a tremendous amount of

time and effort in molding me into the scientist and individual I am today. He adapted

his mentorship style to accommodate my strengths; he was careful in providing useful

feedback; and he was quick to connect me to other people and opportunities within and

outside of his network to enhance my research and graduate education. Gabriel taught

me so many things, the most central of which is the importance of fairness, hard work

and integrity, which I will carry with me wherever I go.

xxix

Specific Aims

Circulating tumor cells (CTCs) offer a non-invasive approach to access critical information regarding the native tumors from which they emanate. These rare cells can provide valuable prognostic information for elucidating patient health when enumerated, and they have the potential to provide insightful genetic and biomolecular information to guide doctors toward tailored treatments. In this thesis, I describe a set of approaches to manipulate cells in microfluidic platforms that may be useful for rare cell isolation. This work addresses three specific aims: synthesis of elastomeric particles to capture and acoustically sequester cells in a microfluidic device (Specific Aim 1); development of microfluidic devices to magnetically separate and template magnetic particles (Specific Aim 2); application of tools described previously to isolate cancer cells from whole blood (Specific Aim 3).

The first chapter reviews the prior art used to separate cells in microfluidic platforms. The second, third and fourth chapters, which describe the synthesis and biofunctionalization of elastomeric particles for the capture and acoustic confinement of cells in acoustofluidic devices, address Specific Aim 1. The fifth chapter, which describes the fabrication of two types of devices, addresses Specific Aim 2. The sixth chapter, which describes the application of the tools described in the previous chapter for the isolation of cancer cells from blood, addresses Specific Aim 3. Lastly, Chapter 7 provides an outlook and future directions of this work.

xxx

1. Introduction to Microfluidic Cell Sorting

Accurate and high throughput cell sorting is becoming an increasingly important sample-processing step in molecular and cellular biology, biotechnology and medicine.

While conventional methods to sort cells can provide efficient performance in short timescales, advances in microfluidics have enabled the realization of miniaturized devices offering similar capabilities that exploit a variety of physical principles. I organize these sorting technologies by the type of cell preparation required (i.e., fluorescent label-based sorting, bead-based sorting, and label-free sorting) as well as by the physical principles underlying each sorting mechanism.

1.1 Background and significance of microfluidic cell sorting

Isolating and sorting cells from complex, heterogeneous mixtures represents a critical task in many areas of biology, biotechnology, and medicine. Cell sorting is often used to enrich or purify cell samples into well-defined populations to enhance efficiency in research and development applications. Cell sorting also serves as the first step in many diagnostic and therapeutic practices, such as the enrichment of hematopoietic stem cells for autologous patient treatments [1]. The need to sort cells is rapidly expanding toward the isolation of rarer target cell populations, including the enrichment of circulating tumor cells (CTCs), hematopoietic stem cells (HSCs), and circulating fetal cells (CFCs) from blood [2-5]. Meanwhile, the growing interest in theranostics and

1

personalized medicine, in which treatments are tailored to the prognoses of patients, is further driving the demand for rapid and high performance cell sorting [6].

The first commercial cell sorter, which exploits a technique broadly known as

fluorescence-activated cell sorting (FACS), was invented in 1969 by Herzenberg et al. [7].

Soon after its seminal introduction, FACS was optimized into a translatable technology and later propelled through multiple evolutions that have now firmly established it as the benchmark for modern cell sorting devices [8, 9]. Now, FACS technologies are automated, robust, and capable of exceptional specificity when using multiple morphological and fluorescent cell signatures (e.g., cell surface labels, cell size, and granularity). These systems are also capable of multiplexed detections, analyses, and sorting speeds up to 50,000 cells per second [10, 11]. Similarly, magnetic-activated cell sorting devices, which generally operate by separating cells with magnetic labels from unlabeled cells in a column via a permanent magnet, are widely used due to their rapid, batchwise processing [12].

The immense contributions of current commercial cell sorting platforms are tempered by several significant and persistent limitations, however, including: limited sample throughput and processing speeds that would make processing clinical-scale samples (>500 million cells) unfeasible, high operating pressures that could result in a loss of function or viability, bulky instrumentation that occupy large bench footprints,

2

technical expertise necessary for operating complex machinery, and increased risk of

sample contamination and safety concerns due to the sorting of aerosolized samples.

These limitations, as well as high unit and sample processing costs, must be overcome to enable more efficient clinical application and commercialization. Consequently, the next generation of cell sorting devices must meet higher standards for performance, versatility, and convenience, including: (i) faster sorting rates, (ii) equal or improved accuracies, (iii) ability to process native biological fluids, (iv) ability to process diverse cell types, (v) enhanced capabilities for multiplexed sorting, (vi) simpler operating procedures enabling fully automated systems, (vii) reduced biohazard risk by eliminating aerosols, (viii) reduced cost, and (ix) reduced size for operational convenience and portability.

To address these needs, researchers are actively looking toward microfluidic devices as the platform for the next generation translatable cell sorter. Microchip devices are a proven technology for cellular handling as they can offer precise spatial and temporal control in a greatly miniaturized platform [11, 13]. These devices can be easily made using standard microfabrication tools, which lowers cost and simplifies commercialization efforts. In addition, microfluidics can be used to detect, focus, mix, count, lyse, and analyze individual cells on an integrated platform for complete lab-on-a- chip applications [14-19].

3

1.2 Existing approaches for microfluidic cell sorting

This section will survey recent developments in microchip cell sorting by

organizing each technology into one of three principal categories based on its primary

cell recognition modality: (i) fluorescent label-based, (ii) bead-based, and (iii) label-free

cell sorting. Within each category, several subsections are provided to further categorize

each technology by the physical principles governing the sorting process. I provide

emphasis to the more recent technologies, especially those that integrate multiple

functions on the same device toward a fully integrated point-of-use device.

1.2.1 Fluorescent label-based cell sorting

Fluorescent label-based cell sorting relies on fluorescent probes or stains to identify cells by type. In traditional FACS, fluorescently-labeled cells organized in a laminar flow stream encounter a focused laser beam that scatters into a detector. The fluorescent signal is then analyzed to assign each cell a type for discrete sorting, whereby in the case of FACS, each cell is encapsulated into an aerosol droplet that is charged and electrostatically sorted [9]. To circumvent the need to form of aerosol droplets, many research groups have used fluorescent labels to identify cells in the microfluidic regime for sorting by a variety of mechanisms, as described in detail in this section. Similar to FACS devices, these technologies generally operate by ordering cells in flow streams for: (i) serial interrogation by laser light, (ii) real-time classification, and

4

(iii) rapid, command-driven sorting. Since each cell is processed discretely, fluorescent label-based approaches are often associated with high efficiencies. Further, immunostaining assays are ubiquitous, reliable, and require less preparation time than bead-based labeling, which can help reduce experimental error. These advantages have made fluorescent label-based technologies the mainstays of modern cell sorting technologies and a viable option for many microchip cell sorting devices.

1.2.1.1 Electrokinetics

Electrokinetics describes a family of effects stemming from an applied electric field that results in the migration of particles or cells [20, 21]. In addition to charging aerosol droplets for electrostatic sorting (as in FACS), electrokinetic forces can be used to directly displace cells, or cell-containing droplets, in fluids. For the purposes of this chapter, electrokinetic manipulations are divided into three categories: electrophoresis, dielectrophoresis, and electroosmotic flow. While these mechanisms are phenomenologically distinct, the forces they exert on cells are well suited for sorting within the length scales of microfluidic devices.

1.2.1.1.1 Electrophoresis

Electrophoresis refers to the movement of suspended particles toward an oppositely charged electrode in direct current (DC). Since most cells possess a slight negative charge due to a locus of chemical groups on their surface, they migrate toward

5

the positive electrode during electrophoresis, and the electrophoretic force exerted on that cell is proportional to its charge [20]. Takahashi et al. applied electrophoresis to sort cells in a microchip in which an upstream fluorescence detector identified labeled cells for rapid electrostatic sorting downstream [22]. Yao et al. developed a similar device based on gravity that operated in an upright orientation to process cells without convective flow [23]. A more recent example by Guo et al. showed electrophoretic sorting with much higher throughputs by sorting water-in-oil droplets under continuous flow [24]. In this system, prefocused cells were encapsulated into droplets such that droplets containing single cells were sorted from droplets containing no cells or multiple cells.

1.2.1.1.2 Dielectrophoresis

In contrast to electrophoresis, where cells move in a uniform electric field due to their surface charge, dielectrophoresis (DEP) refers to the movement of cells in a non- uniform electric field due to their polarizability. For movement in response to a dielectrophoretic force, cells do not need to possess a surface charge because, unlike a

DC field, an alternating current (AC) is capable of polarizing the cell (i.e., inducing a dipole moment across the cell) [20]. Once exposed to an AC field, cells migrate either toward or away from the region of strongest field intensity depending on the electrical permeability of the cell and the fluid. Cells with a higher permeability than the fluid are

6

attracted toward the field maxima, which is known as positive DEP (pDEP) [25]. The opposite is true for negative DEP (nDEP), which is often preferable to minimize deleterious effects on cell function or viability that might occur due to high field strengths. The magnitude of the dielectrophoretic force is dependent on the size and properties of the cell, fluid, and the parameters of the electric field, which is useful for sorting cells by size and dielectric properties [20]. Wang et al. developed a system using

lateral nDEP, whereby a set of interdigitated electrodes aligned along both sides of a

microfluidic channel provides repulsive forces to organize cells by precise distances

from the microfluidic channel walls, to enable enhanced on-chip cytometry and sorting across five channels [26].

In contrast to directly sorting cells in a buffered suspension, several groups have developed systems to encapsulate single cells into emulsified droplets for sorting using

DEP, thus enabling continuous genomic and proteomic analyses downstream [27-29].

Unlike FACS, which can generate potentially biohazardous aerosols, water-in-oil droplets provide a safe and rapid way to analyze individual cells post-sorting. Baret et al. applied DEP in a fluorescence-activated droplet sorter to separate up to 2,000 cells/sec

[27]. Agresti et al. used emulsions to generate picoliter-volume reaction vessels for detecting new variants of molecular enzymes and dielectrophoretic sorting [28]. Mazutis et al. showed that cells compartmentalized into emulsions with beads coated with

7

capture antibodies can be used to analyze the secretion of antibodies from cells for downstream sorting using DEP (Figure 1) [29]. These advances hold promise for creating the next generation of cell sorting devices that could also enable clinical detection, analysis, and diagnosis using a single microchip.

Figure 1: Droplet-based microreactor and cell sorter using DEP. Cells expressing and secreting a target antibody (depicted in grey) and cells not expressing a target antibody (depicted in orange) are encapsulated in droplets with a fluorescent detection antibody, incubated off-chip to permit the production of secretion antibodies, and sorted according to the increased fluorescence signal from the localized packing of detection antibodies on the surface of the bead covered with capture and secretion antibodies. Reprinted with permission from Mazutis et al. [29]. Copyright 2013 Nature Publishing Group.

1.2.1.1.3 Electroosmotic flow

Unlike the electrically driven migration of cells within a stationary fluid (such as in electrophoresis and DEP), electroosmotic flow refers to the movement of a fluid due to the electrically induced migration of solvated ions, thereby transporting cells

8

suspended within the fluid (Figure 2) [30]. This principle was applied to sort fluorescent

from non-fluorescent cells in microfabricated FACS devices using DC electroosmosis

[31-33]. While effective, a major disadvantage of DC electric fields for sorting cells is that

Faradaic reactions (e.g., electrolysis of water) generally occur at the anode and cathode

to maintain a constant electric field, which can generate bubbles and harmful

compounds, such as hydrogen peroxide, that may adversely affect cellular viability and

solution pH if not carefully monitored and regulated [34]. As such, Puttaswamy et al.

coupled nDEP with AC electroosmosis to mitigate these effects while retaining the

ability to focus, transport, and sort cells [35].

9

Figure 2: Direct current (DC) electroosmotic cell sorting. Following laser inspection and cell identification, solvated negative ions in the counterionic layer along the positively charged microchannel floor migrate to the oppositely charged electrode, thereby dragging the surrounding liquid for cell transport[30] to either A) the first outlet (t = Time 0) or B) the second outlet (t = Time 1) [31].

1.2.1.2 Acoustophoresis

Acoustophoresis refers to the movement of an object in response to an acoustic pressure wave. Recently, acoustic microfluidic (i.e., acoustofluidic) technologies have provided many new areas of development within analytical flow cytometry, including the sorting of cells [36, 37]. Acoustic forces are amenable to cell handling as they can provide rapid and precise spatial control in microchips without affecting cellular viability [25, 38-40]. In this context, acoustic waves can be divided into three categories: bulk standing waves [41], standing surface acoustic waves (SSAWs) [42], and traveling waves [43].

10

Acoustic standing waves form through the disturbance of a medium (e.g., fluid or a surface) by pressure waves of equal magnitude and frequency traveling in opposite directions that result in a single stationary wave containing fixed regions (i.e., nodes) that exhibit a lack of pressure fluctuation and alternating regions (i.e., antinodes) that exhibit alternating pressure maxima and minima. These contrasting regions can provide precise spatiotemporal manipulation of individual cells, as discussed below.

1.2.1.2.1 Bulk acoustic standing waves

Bulk acoustic standing waves occur when a microfluidic channel is excited by

ultrasound to a resonance mode in which the applied wavelength matches the spatial

dimensions of the microfluidic channel. Here, suspended particles with a radius, a,

experience radiation forces proportional to the square of the pressure amplitude, p0:

2 ( ) = , sin (2 ) (1) 3 2 3 , , 𝛽𝛽𝑓𝑓 𝜋𝜋𝑝𝑝0 𝑎𝑎 𝐹𝐹𝑅𝑅 − � � 𝜙𝜙�𝛽𝛽𝑐𝑐 𝑓𝑓 𝜌𝜌𝑐𝑐 𝑓𝑓� 𝑘𝑘𝑘𝑘 𝜆𝜆 where λ, β, and ρ denote the ultrasonic wavelength, compressibility, density, and the subscripts c and f denote the cell and fluid, respectively [44, 45]. The magnitude of this force is strongly dependent on the volume of the cell, and the direction of this force is determined by the acoustic contrast factor, φ, which depends on the densities and compressibilities of the cell and the fluid. If the φ is positive, cells will travel towards the node(s) of an acoustic standing wave (Figure 3A), whereas, if the φ is negative, cells (or particles) will travel towards the antinodes (Figure 3B). Most cells in physiological

11

buffers display a positive φ due to their relatively higher densities and thus can be precisely and rapidly focused in microfluidic channels without additional diluent buffers (i.e., sheath fluids) [46, 47]. Bulk acoustic waves were first used to sort cells by

Johansson et al., in which fluorescently labeled cells detected by a camera triggered an ultrasound transducer that directed that cell from its initial streamline toward the pressure node, thereby modifying its trajectory to the target outlet [41]. More recently,

Jakobsson et al. demonstrated an acoustofluidic device to focus cells to one side of a microfluidic channel for fluorescence detection, real-time classification, and acoustophoretic sorting [48].

12

Figure 3: Acoustofluidic manipulation of cells and particles. In a bulk acoustic standing wave, objects with a A) positive and B) negative φ migrate to the pressure node(s) and antinodes, respectively [49]. C) In a SSAW device, interdigital transducers (IDTs) focus cells along well-defined streamlines according to the driving frequencies of the IDTs (e.g., f1, f2, and f3) for sorting across multiple outlets. D) The cross section of a SSAW device containing four pressure nodes [42].

1.2.1.2.2 Standing surface acoustic waves

In contrast to bulk acoustic standing waves, SSAW devices form a standing wave

along the floor of the microfluidic channel using interdigital transducers (IDTs),

providing the mechanical perturbations necessary to position cells along well-defined

flow streams in the fluid above (Figure 3C,D) [50]. SSAW devices show particular

promise for fluorescent label-based cell sorting since a single device can provide a large

range of frequencies for dexterous spatial control of single cells and, in turn, multiple

channels for sorting [51, 52]. These devices have efficiently sorted cells in buffer as well

as in water-in-oil droplets across five fluidic channels [53, 54]. Ding et al. further showed

13

that SSAW devices can function as acoustic tweezers to manipulate the spatial orientation and patterning of cells and whole organisms such as C. elegans [55].

1.2.1.2.3 Traveling acoustic waves

While most acoustically actuated microfluidic sorting devices developed thus far

employ bulk standing waves or SSAWs, traveling acoustic waves have also been used to

sort cells in the microfluidic regime. Cho et al. devised a system where a transducer

actuates to deflect target cells into the side outlets at rates above 1,000 cells/sec [43].

Franke et al. exploited acoustic streaming forces generated from non-standing surface

acoustic waves to redirect fluid flow from one outlet to an adjacent outlet [56]. More

recently, Schmid et al. developed a device that produces a non-standing surface acoustic

wave that effectively creates a traveling wave that emanates from the surface to the

microfluidic cavity to continuously deflect cells or droplet-containing cells at rates above

1,000 cells/sec [57].

1.2.1.3 Optical systems

In addition to forces from pressure waves or electric fields, radiation forces produced by a highly focused optical beam have been used for cell manipulation [58].

Optical forces are considered advantageous due to the preservation of cell function, precise spatial control in three dimensions, and manipulation of very small objects (e.g., on the atomic or molecular scale) [59]. A focused laser beam can trap cells due to a

14

mismatch in the refractive index between the cell and its surrounding fluid, producing optical scattering and gradient forces. Scattering forces tend to push objects away from the source of light, whereas gradient forces attract objects to the point of highest intensity produced by the light (i.e., the focusing maxima) (Figure 4A). Similar to pDEP, objects travel toward the beam maxima and become optically trapped when the gradient forces overcome scattering forces, producing so-called optical tweezers. Since its inception, this technique has ushered many new types of analyses for single cells, particles, and molecules [60].

Optics were first used to levitate and transport individual particles in a fluid by

Ashkin in 1970 [61]. Eventually, optical radiation forces were applied to cell sorting by propelling single cells through a microfluidic channel, whereby the long traversal times generally resulted in low throughputs [62]. However, recent innovations in miniaturized optical devices have reinvigorated the use of optics as switches, which have made optics-based methods more feasible for fluorescent label-based cell sorting [63, 64].

Several groups have used focused optical beams to impose switchable radiation forces on a continuous stream of focused cells for on-chip sorting (Figure 4B) [64-67]. Recently,

Wu et al. developed a pulsed laser fluorescence-based cell sorter that rapidly forms a stationary bubble due to localized heating to controllably deflect target cells at 20,000 cells/sec [68, 69].

15

Figure 4: Cell sorting by optical force switching. A) When scattering forces (FS) exceed gradient forces (FG) from a focused laser beam, cells are deflected (left); however, when FG exceeds FS, cells are optically trapped (right) [60]. B) A hydrodynamically focused stream of cells is aligned toward the waste outlet whereupon cells of interest detected by laser inspection are captured and displaced by optical tweezers for sorting [67].

1.2.1.4 Mechanical systems

Instead of using external fields, mechanical sorting systems use discrete mechanical forces to sort cells. Krüger et al. developed an external rotary valve to regulate the throughput within a multi-inlet disposable microfluidic chip by hydrodynamic flow switching for cell counting and sorting [70]. Fu et al. devised a similar device to sort E. coli that also includes a trapping component for enhanced analysis [71]. This design was later simplified to hydrodynamically focus and isolate latex beads from red blood cells [72]. Chen et al. used a similar approach that employed

16

negative pressure gradients to draw fluid from chips and switch the flow of cells to various outlets using gating valves [73].

Some groups have developed on-chip flow switching mechanisms instead of

relying on external valves to direct fluid flow. For example, Ho et al. designed a

micromachined T-switch actuated by electrolytically-produced bubbles to direct the

flow of individual cells for binary sorting (Figure 5) [74]. And Shirasaki et al.

demonstrated cell sorting by using laser light to rapidly heat thermoreversible gelation

polymers that controllably actuate the opening and closing of a microchannel [75].

17

Figure 5: Electromechanical T-switch for cell sorting. A microfabricated cantilever beam reversibly shifts from A) a ‘down’ position (t = Time 0) to B) an ‘up’ position (t = Time 1) when a corresponding pair of electrodes is activated to generate bubbles via isothermal electrolysis, which in turn exerts a mechanical force on the T-switch to redirect fluid flow [74].

1.2.2 Bead-based sorting

While fluorescent label-based cell sorting offers several advantages, the inconvenience of serial detections, discrete sorting, and the inability of the fluorescent labels to directly contribute to the sorting process has led to the development of other effective techniques for rapid cell sorting, including bead-based and label-free cell sorting systems. Bead-based cell sorting systems depend on particles of a specific material, size, and surface-binding capacity to capture target cells, or sometimes non- target cells, for sorting in an external field. The attachment of beads allows bound complexes to experience a force that is different from unbound cells. Consequently,

18

bead-based cell sorting does not require the serial interrogation of cells and can manipulate groups of cells simultaneously. This affords bead-based systems the ability to sort cells at potentially faster rates and without large volumes of diluent.

Additionally, bead-based assays share the immunospecificity of fluorescent label-based sorting, providing the potential for highly accurate targeting and reproducible results.

The first commercial bead-based cell sorting system was a benchtop magnetic cell sorter described by Miltenyi et al. in 1989 [12]. This method of cell isolation was soon miniaturized to the microfluidic domain to sort cells with magnetic and other types of particulate labels.

1.2.2.1 Magnetophoresis

Cell sorting by magnetophoresis (MAP) is facilitated by the use of permanent magnets or electromagnetic coils to exert forces on cells labeled with magnetic particles, magnetically responsive cells, or cells suspended in ferrofluid. In the case of magnetic bead-based sorting, cell surface markers are labeled with particles (typically nanoscopic) containing iron. Cell sorting by MAP is usually performed to either debulk cell populations or to isolate rare cells from native biofluids [76]; and similar to acoustophoretic or dielectrophoretic forces, magnetophoretic forces are highly dependent on size of the magnetic moiety [77].

19

The concept for magnetic cell sorting developed by Miltenyi was quickly miniaturized into a microchip device to sort magnetically labeled cells from unlabeled cells under free-flow MAP [78-80]. Using similar principles, Adams et al. developed a multiplexed cell sorting device using MAP where one cell type labeled with large magnetic beads was separated due to the larger magnetic force acting upon it, and a second cell type labeled with small magnetic beads was then separated from unlabeled cells [81]. Similarly, Carr et al. developed a multiplexed bead sorter that sorts magnetic beads across 25 output fractions based on the magnitude of their magnetic moments

[82].

Creating reliable technologies for isolating rare cells such as CTCs, HSCs and

CFCs remains a major challenge in research and medicine. In particular, CTC isolation is a daunting task due to their low abundance in peripheral blood and their “stealthy” behavior during epithelial-mesenchymal transitions [2, 5, 83]. Recently, however, significant progress has been made in isolating CTCs via MAP. Hoshino et al. devised a system can capture CTCs from blood cells in ratios as low 1:109 (Figure 6A) [84]. In their system, CTCs were labeled with antibodies against epithelial cell adhesion molecule

(EpCAM) that were conjugated to iron oxide nanoparticles, separated, and then stained with anti-cytokeratin, DAPI, and anti-CD45 to confirm the captured cells originated

from tumors. Numerical simulations later showed that an inverted system (i.e., where

20

the magnetic field is oriented opposite to gravity) can help improve the fidelity of rare cell isolation [85]. Darabi et al. developed a similar device with a micropatterned magnetic grid for isolating lymphocytes [86]. Kang et al. developed a microchip device with small parallel chambers perpendicular to the main channel to isolate and retain magnetically labeled CTCs from blood for on-chip cell culture [87]. For more precise control over the deposition of cells, our group has recently shown that the magnetic labels can be used to isolate cells from a population and organize those cells into well- defined compartments across a large magnetographic array for rapid enumeration, cell pairing, and single cell analysis [88].

Figure 6: Magnetic isolation of CTCs in microfluidic devices. A) Magnetic beads conjugated with anti-EpCAM are shown to capture and isolate CTCs under free flow in a magnetic field. Reprinted with permission from Hoshino et al. [84]. Copyright

21

2011 Royal Society of Chemistry. B) Magnetically labeled CTCs and leukocytes are filtered from blood via deterministic lateral displacement (see Figure 12), focused via inertial focusing (see Figure 10A), and sorted in a magnetic field. Reprinted with permission from Ozkumar et al. [89]. Copyright 2013 Science Translational Medicine.

Chen et al. recently developed a device that can capture magnetically-labeled cells for sequential encapsulation into aqueous picoliter vessels containing reagents for single cell analysis [90]. Researchers from the Toner lab recently developed the innovative CTC-iChip that contains three modules on a single microchip to separate

CTCs from peripheral blood for diagnostic analysis: (i) deterministic lateral displacement (a method described below) to debulk nucleated cells (e.g., leukocytes and

CTCs) from erythrocytes and platelets, (ii) inertial focusing to align cells into a focused streamline (a method described in detail below), and (iii) an external magnetic field to separate magnetically labeled CTCs from leukocytes (or magnetically labeled leukocytes from CTCs) (Figure 6B) [89, 91]. These bead-based technologies show great promise for isolating tumor cells from unmodified biological fluids on a single chip with minimal processing steps for rapid analysis and diagnosis.

1.2.2.2 Acoustophoresis

Elastomeric beads (or particles) show promise in acoustofluidic systems due to their unique focusing behaviors. Unlike cells, elastomeric particles focus along the antinodes of an acoustic standing wave due their relatively low density and bulk modulus compared to aqueous fluids (i.e., φ < 0; (1)) [92, 93]. Using silicone elastomeric

22

particles synthesized from nucleation and growth techniques, we have developed a system where biofunctionalized elastomeric particles decorated with streptavidin can capture cells displaying biotinylated antibodies and confine those cells to the antinodes of an acoustic standing wave (Figure 7) [49, 94]. This method of cell confinement can enable rapid, continuous, and discriminate sorting in which unlabeled cells focus at the acoustic node(s) and labeled cells focus at the acoustic antinodes for downstream collection [92]. Also, Lenshof et al. demonstrated that target cells labeled with dense (φ >

0) microbeads could be separated from same-sized non-target cells by virtue of their disparate rate of acoustophoretic migration [95].

23

Figure 7: Acoustic separation of cells using elastomeric particles. A) Elastomeric particles and cells focus to the antinodes and node(s) of an acoustic standing wave, respectively. B) When elastomeric particles bind to target cells, those complexes displace to the pressure antinodes for separation from non-target cells. Adapted from Shields IV et al. [49]. 2014 American Chemical Society.

1.2.2.3 Electrokinetics

Electric forces exerted on cells and particles scale according to their size, thereby enabling the possibility of non-sequential sorting [20]. However, since many cell populations exhibit similar size morphologies, Hu et al. used beads to bind to target cells such that the bound complexes experience greater dielectrophoretic forces in alternating current and thus separate from non-labeled cells [96]. In this system, cells labeled with beads traveled farther than cells without beads toward a separate outlet. This system

24

was modified to sort three types of bacteria in a single pass using polystyrene beads of two distinct sizes [97]. Later, a magnetic trapping component was added to isolate magnetically-labeled cells from the bulk population in an integrated dielectrophoretic-

magnetic activated cell sorter (Figure 8) [98]. In addition, Cheng et al. fabricated a DEP

device that can filter, focus, sort, and trap cells on a single integrated microchip [99].

Their sorting mechanism used angled electrodes and nDEP to deflect bacteria into

individual channels and surface enhanced Raman scattering to characterize the samples.

25

Figure 8: Multi-target cell sorting using DEP and magnetic trapping. A) Target Cell A is captured with polystyrene beads depicted in green and Target Cell B is captured with superparamagnetic beads depicted in red. B) Target Cell A is removed from non- target cells (depicted in blue) by dielectrophoretic forces and Target Cell B is removed from non-target cells by magnetic trapping. Reprinted with permission from Kim et al. [98]. Copyright 2009 Royal Society of Chemistry.

1.2.3 Label-free cell sorting

Label-free cell sorting in microfluidic devices is perhaps the most studied of the three approaches as it encompasses not only active systems (i.e., systems that rely on the use of external fields cells for sorting), but also passive systems, which include mechanisms such as inertial flow, on-chip filtration, and immobilization. In these cases, instead of solely relying on surface markers for labeling cells with fluorophores or beads, label-free sorting relies on the physical differences in the properties of cells such

26

as size, shape, density, elasticity, polarizability, and magnetic susceptibility. The earliest automated cell sorter did not require any labels as it sorted cells by volume using an impedance technique based on the Coulter principle [100]. This technique was developed in 1965 by Fulwyler and is widely regarded as the forerunner of modern flow cytometers and FACS devices [101]. These key concepts have since been expanded to sort cells in the microfluidic regime. Of the sorting assays described, label-free sorting

generally requires the least amount of preparation, making it a highly attractive option

for cell sorting.

1.2.3.1 Acoustophoresis

The majority of acoustofluidic-based approaches for cell sorting are label free, which takes advantage of slight quantitative differences in the size or density (thereby, its acoustic contrast factor, φ) of different cell populations.

1.2.3.1.1 Bulk acoustic standing waves

Unlike the previously described acoustofluidic systems, label-free acoustofluidic

systems generally sort cells based on differences in their size. This method of cell sorting relies on the initial placement of cells in streamlines away from the pressure node(s) for their rapid displacement to the node(s) upon actuation of the device (Figure 3A), thereby separating cells of disparate size or acoustic properties. This concept was first used by

Petersson et al. to purify erythrocytes from a mixture of lipid particles [102, 103]. In a

27

later study, Petersson et al. used hydrodynamic forces to direct particles and cells to the

side walls of an acoustofluidic device (i.e., corresponding to the pressure antinodes), whereupon they migrated toward the node (i.e., the channel center) of the acoustic standing wave at rates proportional to their volume [104]. Similar to DEP and MAP, larger cells experience greater forces in an acoustic standing wave, and thus respond faster to the radiation forces (1), directing larger cells to the center outlet and smaller cells to the side outlets. This concept was later used for debulking platelets from peripheral blood progenitor cells by Dykes et al. [105], for enriching prostate cancer cells from leukocytes by Augustsson et al. [106], and for sorting viable from non-viable cells by Yang et al. [107]. Using a thin wall in an acoustic resonating chip, Fong et al. created an H-filter device that can form bulk acoustic standing waves with the node offset from the center of a microfluidic channel for a new method of acoustic cell sorting [108].

1.2.3.1.1 Standing surface acoustic waves

While the majority of SSAW devices use fluorescent tags to sequentially identify and sort cells, Nam et al. developed a SSAW device to separate platelets from whole blood [109]. In their system, blood was injected into the device and sheath fluid focused the cells into a confined . A separation then followed in which larger cells (e.g., leukocytes and erythrocytes) experienced greater acoustic radiation forces causing them to migrate farther then the surrounding platelets. More recently, researchers from the

28

Huang group have used SSAW devices to sort cells by compressibility [110] and wash cells by separating leukocytes from lysed red blood cells [111].

1.2.3.2 Electrokinetics

Label-free cell sorting devices can employ DEP to separate cells based on their intrinsic dielectric properties (i.e., polarizability). Huang et al. first demonstrated this concept by sorting five cell lines with a microelectronic array [112]. Also, Cummings et al. designed a microchip containing arrays of insulating posts where the resulting nonuniform electric field across the posts exerted different magnitudes of force on cells as they passed [113]. Later, this principle was used to generate nonuniform electric fields near insulating ridges on a microchip to filter and concentrate cells [114]. Vahey et al. introduced an isodielectric separation system for sorting cells without labels [115]. In this system, cells experience dielectrophoretic forces that displace them to their natural equilibrium point across a conductivity gradient to where their net polarization vanishes

(i.e., isodielectric point). With this principle, cells with different conductivities displace by discrete distances along that gradient for efficient on-chip sorting.

An interesting extension of dielectric cell sorting uses field-flow fractionation

(FFF), which includes a family of approaches (e.g., gravitational, centrifugal, thermal, magnetic, and electrical) to position cells by precise distances from the microchannel floor and thus to distinct regions of the parabolic flow profile for separation by flow

29

[116-118]. Vykoukal et al. first developed a DEP FFF system to isolate stem cells from enzyme-digested adipose tissue [119]. This system contained patterned microelectrodes

along the bottom of a microfluidic channel that exerted dielectric forces in the opposite

direction of gravitational forces, which enabled each cell type to reach an equilibrium

height based on its surface charge for rapid sorting under flow (Figure 9) [119]. Cui et al.

demonstrated that a pulsed dielectric cell sorter could utilize nDEP to trap cells

upstream for subsequent elution by size [120]. When the AC field was activated, cells

became indiscriminately trapped to the ceiling of the microchannel via nDEP. Then after

the field was disengaged, larger cells eluted from the microchannel faster than the

smaller cells due to larger fluid forces. Then after reactivating the field, smaller cells

were dielectrically trapped again whereas the larger cells escaped the electrodes.

Figure 9: Sorting by nDEP-assisted field-flow fractionation. A) An array of electrodes displaces cells to equilibrium positions above the floor of a microfluidic channel

30

according to their type (t = Time 0) and B) sorts those cells by propelling them through the channel at rates according to their distance from the wall (t = Time 1) [119].

Some groups have coupled hydrodynamic lift forces, which act on cells exposed to a shear flow [121], with DEP to sort cells by size or electric permeability. Doh and Cho developed a microchip to separate viable from non-viable cells due to their dissimilar polarizabilities, resulting in pDEP and nDEP, respectively [122]. Moon et al. and Shim et al. used hydrodynamic lift coupled with DEP to sort breast cancer cells and CTCs without labels, respectively [123, 124]. Kim et al. sorted cells based on their different sizes during the cell cycle using a DEP fractionation technique [125]. Hydrodynamic forces were used to concentrate cells to one side of the microfluidic channel whereupon cells encountered an AC field that displaced the cells across the channel by distances according to their size and dielectric properties. This technique was later used to separate viable from nonviable yeast cells [126]. An interesting dual function microchip was developed by Sun et al. that could simultaneously sort cells using DEP and size cells using the Coulter Principle via an integrated transistor [127].

1.2.3.3 Magnetophoresis

While the majority of magnetic cell sorting devices require nanoparticles to capture and isolate cells (Figure 6), erythrocytes can be sorted from blood in a similar fashion solely due to their natural iron content in methemoglobin [128, 129]. This label- free method of cell sorting has been used for debulking peripheral blood by removing

31

erythrocytes from leukocytes, platelets, lipids and plasma. Isolating erythrocytes from blood using external magnetic fields was first demonstrated in 1975 by Melville et al.

[130] and was later refined by Zborowski et al. using cell tracking velocimetry [131] and

by Han et al. embedding a ferromagnetic inside of a microdevice [132-134]. This

method of magnetic manipulation from embedded ferromagnetic microwires was more

fully developed later by others [135-137]. Furlani presented a method for separating red

blood cells from white blood cells in plasma by embedding soft-magnetic elements in an

array along a microfluidic channel [138].

Since these magnetic, label-free approaches are only suitable for blood cells with

natural iron content, several groups have employed nontraditional means for

continuously sorting other types of cells in magnetic fields. Roberts et al. created a device

that could sort macrophages from monocytes due to their dissimilar internalization rate

of iron nanoparticles [139]. Since macrophages can endocytose more iron content per cell

than monocytes, macrophages can undergo greater loadings of magnetic material and

thus experience greater MAP forces in an external field.

Some groups have sorted diamagnetic (i.e., unlabeled) cells by suspending them

in ferrofluids, which can separate cells due to differences in their size, shape, or

deformability [140]. Ferrofluids are liquids that are strongly magnetizable (e.g., water

containing iron oxide nanoparticles) and can thus transport diamagnetic particles in a

32

magnetic field. Zeng et al. developed a system where an upstream magnet focused cells suspended in ferrofluid into a single stream, and a downstream magnet displaced those cells according to their size [141]. Similarly, Shen et al. used a paramagnetic solution (i.e.,

of paramagnetic ions) to exert repulsive forces to sort cells by size [142].

1.2.3.4 Optical systems

Similar to the optical switching mechanisms described previously, Hoi et al. created a layered device with perpendicular flow channels on top of one another [143].

In this system, a microscope detects unlabeled cells as they pass through the first channel, which triggers a pulsed laser to rapidly force cells into the second channel at their junction. Lau et al. used laser tweezers for an automated Raman-activated cell sorter that functions without fluorescent labels [144]. While the majority of optical systems employ one focused laser beam, MacDonald et al. used a body-centered tetragonal optical lattice to fractionate a two-component cell mixture in a continuous flow microfluidic device [145].

A major limitation of optical tweezers is their inherently slow processing that stymies the potential for high throughput sorting. Optoelectronic tweezers, however, refer to a class of tools that employ optics and electronics by projecting optical images on a photosensitive substrate. The projected images effectively form transient electrodes on the bottom surface to direct an AC field from a conductive, transparent lid for the

33

precise spatial arrangement of micro- and nanoparticles [146]. Optoelectronic tweezers can thus provide a low-power means to trap, transport, and sort cells without labels

[146]. And while optical tweezers provide high-resolution manipulations, they can also require optical intensities reportedly 100,000 times greater than optoelectronic tweezers while only providing small regions for cellular manipulation [147]. In an early example,

Chiou et al. used optoelectronic tweezers to pattern cells into a large, well-defined array

[147]. Now, optoelectronic tweezers are used to sort cells based on size due to the differing induced velocities in response to applied dielectrophoretic forces [148, 149] as well as viable cells from non-viable cells due to differences in membrane potentials

[150]. These devices have been coupled with electrowetting-on-dielectrics mechanisms to enable multiple delivery rounds of drugs or nutrients to cells followed by electrically- driven waste removal [151]. Optoelectronic tweezers have been integrated into microfluidic devices for single-cell preparation and analysis, enabling the high precision benefits of optics devices with higher throughputs [152].

1.2.3.5 Passive cell sorting

The technologies described thus far involve active methods for cell separation; however, the remaining systems involve passive approaches to separate, isolate, or enrich cell populations [153]. Passive systems consist of a variety of methods that do not rely on fluorescent labels or beads. Instead, these methods rely on the inherent

34

differences in cellular morphology between cell groups (e.g., size, shape, compressibility, and density) and can sort cells using inertial forces, hydrodynamic spreading, deterministic lateral displacement, filtration, transient cellular adhesion, and cellular immobilization [154].

1.2.3.5.1 Inertial focusing in curved channels

Inertial forces can result in the induced migration of cells or particles across

streamlines in laminar flow streams. Typically, inertial forces emanate from boundary

effects of fluid flow adjacent to the walls of a microfluidic channel, causing lift. Inertial

focusing in curved channels refers to a subset of distinct phenomenological techniques

for cell fractionation, which includes the use of serpentine or Archimedean spiral

patterns for cell ordering and sorting [155]. Di Carlo et al. demonstrated that cells could

be differentially focused and sorted based on size under laminar flow using a serpentine

pattern (Figure 10A) [156]. A major benefit of this system is its high throughput (e.g., 1.5 mL/min) without sheath flow or sequential cell manipulation, which is useful for processing native biological fluids and flow cytometry [157, 158]. As previously discussed, an inertial flow microfluidic chip with a deterministic lateral displacement module was incorporated into a magnetic-label based system for CTC isolation (Figure

6B) [89].

35

Similarly, Russom et al. created a system with a spiral channel to apply centrifugal forces to focus and sort cells [159]. As in the case of serpentine patterns, cells can be sorted in a spiral microchannel due to their differential focusing for rapid separation upon branching into multiple channels (e.g., six, Figure 10B,C) [160].

Recently, this system was used by Hou et al. to retrieve CTCs from blood using an approach named Dean flow fractionation [161] and by Nivedita et al. to separate blood cells [162]. Guan et al. and Warkiani et al. demonstrated that a channel with a trapezoidal cross section could further enhance the performance of a spiral microfluidic device to focus and sort cells [163, 164].

36

Figure 10: Inertial microfluidics for cell sorting in curved microchannels. A) A serpentine microfluidic channel can focus cells into a single streamline. B) A spiral microfluidic channel can sort cells by size (IW and OW indicate the inner wall and outer wall, respectively). C) The cross section of the microfluidic channel showing the balance between lift forces (FL) and Dean drag forces (FD). Reprinted with permission from Kuntaegowdanahalli et al. [160]. Copyright 2009 Royal Society of Chemistry.

1.2.3.5.2 Pinched flow and hydrodynamic spreading

In addition to curved channels, inertial forces can play a critical role in straight channels [155]. For example, Parichehreh et al. were able to enrich nucleated cell populations in blood using inertial forces in straight microfluidic channels of large aspect ratios [165]. Similarly, pinched flow fractionation and hydrodynamic spreading are two examples where inertial forces play a role in the ordering of cells for the

37

subsequent sorting. Pinched flow fractionation occurs when a flow stream of cells is pinched by a narrow channel cross section such that cells are constrained and aligned against a side wall and subsequently separate by size once the channel broadens due to the laminar flow profile (Figure 11) [166]. This alignment effect is typically enhanced by sheath fluid, which pushes cells against a wall such that the center of the larger cells are farther from the wall surface than the center of smaller cells, thus giving cells of different sizes slightly different flow trajectory upon broadening of the channel. This method for sorting has been expanded by the addition of multiple asymmetric outlets for better hydrodynamic control [167] as well as by the spatial reorientation of the microfluidic device for gravitationally enhanced separation between cell populations of different size and mass [168].

38

Figure 11: Cell sorting by pinched flow fractionation. A) In the pinched segment, cells are first pushed against the wall, and then separated by size upon broadening of the microfluidic channel. B) Cells are aligned in the pinched segment of the channel and follow separate streamlines for sorting by size after exiting the pinched segment. Reprinted with permission from Yamada et al. [166]. Copyright 2004 American Chemical Society.

Similar to pinched flow fractionation, some groups have used hydrodynamic techniques to direct cells against one wall of a microfluidic channel without a pinched segment for sorting upon broadening of the channel in a method called hydrodynamic spreading [169, 170]. Wu et al. enhanced this effect by coupling hydrodynamic spreading with electroosmosis to sort E. coli and yeast cells [171]. Instead of using hydrodynamics or pinched flow to align cells against one wall of a microfluidic channel, Mach et al. used

39

inertial forces in a straight channel with a large aspect ratio cross section (i.e., height > width) to separate pathogenic bacteria from blood upon hydrodynamic spreading [172].

Microfluidic channels with large aspect ratio cross sections can localize to specific regions due to inertial lift and shear forces, which are readily influenced by channel geometry, particle size, and flow properties changes in the Reynolds number for sorting upon hydrodynamic spreading [173]. This concept was exploited to isolate rare tumor cells spiked in blood [174] and sort cells by their deformability [175]. Instead of using pinched flow or inertial forces, Geislinger et al. devised a hydrodynamic lift system enhanced by hydrodynamic spreading to separate CTC and red blood cells [176, 177].

1.2.3.5.3 Deterministic lateral displacement

Given the standard fabrication methods used to create microfluidic chips, micropatterns within a microfluidic channel (e.g., posts, ridges, groves, ratchets) provide a convenient way to spatially manipulate cells. Deterministic lateral displacement is an important example, where cells navigate through an array of posts for sorting by size. In these systems, control over sorting is given by the design of the array features such that cells smaller than a critical radius (a < Rc) move with the convective flow and cells larger

than a critical radius (a > Rc) move in a direction dictated by the arrays (Figure 12) [178].

Using a periodic array of microposts, in which each row of posts is offset by a certain

periodic distance, Huang et al. showed that smaller cells could more easily traverse

40

between obstacles than larger cells, enabling their separation [179]. The critical sizes of cells and microposts were carefully evaluated by Inglis et al. [180] and have been used to fractionate undiluted whole blood samples as well as isolate cancer cells [181-183]. Later,

Beech et al. showed that deterministic lateral displacement was useful for sorting cells by

not only size, but by shape and deformability [178].

Figure 12: Cell sorting by deterministic lateral displacement. Large cells (depicted in blue) migrate away from the small cells (depicted in red) in the initial streamline due to the engineered size and spacing of the microposts in the microfluidic channel [180].

1.2.3.5.4 Hydrophoretic filtration

Ridge-induced hydrophoretic filtration relies on the formation of a lateral

pressure gradient within a microfluidic channel due to flow-altering micropatterns. A

successive array of slanted obstacles on the microchannel floor and ceiling induces a

pressure gradient across the width of the channel to focus cells to precise locations

within the generated local pressure field according to type and then separates those cells

41

[184]. Using this phenomenon, Choi et al. developed a device that exploits differences in both the size and deformability of cells for passive sorting [185].

Similarly, a device with successive ridges along the channel ceiling has been shown to focus, guide and sort cells (Figure 13) [186]. This device, previously called a microvortex manipulator, contains an array of herringbone shaped slanted grooves in the ceiling of a microfluidic channel that passively focuses cells to defined regions of a microfluidic channel and sorts cells based on their density [187]. Using a different style of microvortex, Sollier et al. sorted CTCs using a sudden expansion (i.e., by introducing sharp corners perpendicular to the flow direction) in a microfluidic channel to create predictable eddy flows to capture CTCs from blood [188]. A similar microfluidic channel design with multiple orifices has been used to sort CTCs from white blood cells [189].

Figure 13: Hydrophoretic cell focusing and sorting. A) A simplified free-body diagram of cells with a low density (shown in blue) separating from cells with a high

42

density (shown in red). The black arrows pointing upwards represent buoyancy forces and the black arrows pointing downwards represent settling forces. B) A top view of the microfluidic channel with herringbone grooves in the ceiling to guide the focusing and separation of cells by density [186].

1.2.3.5.5 Size exclusion filtration

Instead of engineering the size and pattern of uniformly spaced posts to sort cells, size exclusion filtration refers to the use of posts with tiered (i.e., decreasing) spacing as a function of distance for sorting cells in a non-binary fashion. Size exclusion filters consist of a series of linear arrays of pillars that selectively group cells by size and shape. Mohamed et al. developed a size exclusion filter to isolate CFCs from maternal blood [190]. Later, McFaul et al. and Preira et al. showed how structural ratchets can be used to sort cells based on size as well as deformability when used with oscillatory applied pressures (Figure 14) [191, 192].

43

Figure 14: Micropatterned ratchets for isolating cells by size and deformability. A) A design of microratchet funnels for fractionating cell populations (dimensions in µm). B) The operation area of the device whereby cells (1, left) enter the chip, (2) reversibly flow through the ratchets for separation by size, and (1, right) exit the chip. C) Schematic of a size exclusion filtration device with various inlets, outlets, and valves (V1-V6). Reprinted with permission from McFaul et al. [191]. Copyright 2012 Royal Society of Chemistry.

1.2.3.5.6 Cross-flow filtration

The filtration of cell-containing fluids is one of the earliest methods used to fractionate cell populations. Examples of these filters include weir filters, which contain large barriers to trap large cells (Figure 15A); pillar filters, which contain a row of same- sized microposts to trap large cells (Figure 15B); and membrane filters, which contain an array of pores on the floor or ceiling to trap large cells [193]. A major limitation of these filters is that they often clog by trapping larger cells or debris. Recently, however,

44

Earhart et al. devised a membrane filter that could reduce this clogging by using larger pore sizes and incorporating a magnetic component to capture magnetically labeled cells

(e.g., CTCs) at the edges of the pores, thereby allowing unlabeled cells to easily pass through the filter [194].

In contrast, cross-flow filtration, sometimes referred to as tangential flow filtration, uses an array of lateral slits aligned in the direction of flow to fractionate cell populations by size (Figure 15C). This method of filtration is a major advancement over early types of filters such as weir, pillar, and membrane because of their decreased likelihood for clogging because the behave more like a sieve than a dead-end filter [195].

Several cross-flow filter designs have been developed for size-based sorting applications such as the separation of white blood cells from whole blood, plasma from whole blood, and myocytes from non-myocytes [195-199]. Using a cross-flow filter, Chen et al. developed an integrated system for sorting and lysing cells on a single chip with capabilities for DNA purification that can yield amounts comparable to commercial centrifugation methods [200].

45

Figure 15: Microfluidic filtration mechanisms. Schematic of a A) weir filter, B) pillar filter, and C) cross-flow filter to separate smaller cells (depicted in red) from larger cells (depicted in blue) [193].

1.2.3.5.7 Hydrodynamic filtration

The last type of microfluidic filtration is hydrodynamic filtration. Here, aligned

cells are separated by multiple branched outlets, whereby the fluid draining from the

outlets pulls cells from the walls of the main channel at rates that scale according to their

size (Figure 16) [201]. Smaller cells exit the proximal outlets because their center is closer to the wall of the microfluidic channel, enabling their controlled shunting from larger cells (Figure 16B-C). Yamada et al. first showed a hydrodynamic filtration device and its use for the selective enrichment of leukocytes and liver cells [201-203]. Later, Mizuno et

al. combined hydrodynamic filtration to first separate lymphocytes by size, and second

to separate lymphocytes by surface marker expression using MAP for multiplexed

sorting [204]. After cells are filtered, the magnetic field pulls cells with more magnetic

content (due to their higher expression levels) toward the outlet closer to the magnet,

whereas cells with lower expression levels exit the outlet farthest from the magnet.

46

Figure 16: Cell sorting by hydrodynamic filtration. A) Cells are injected into the microfluidic device and are pushed toward the outlets. B) Small cells exit out of the proximal branches whereas C) large cells exit out of the distal branches. Reprinted with permission from Yamada et al. [201]. Copyright 2007 Springer.

1.2.3.5.8 Transient cellular adhesion

Instead of relying in differences in the size, deformability, and membrane polarizability of cells, some groups are examining the impermanent adhesion of cells to a surface to enable a unique class of microfluidic cell sorting methods [205]. Lee et al. showed that a flat surface containing a striated pattern of P-selectin could induce the transient cellular adhesion and lateral displacement of HL60 cells due to high affinity between the PSGL-1 ligand and P-selectin [206]. Bose et al. showed this system could isolate neutrophils from blood with high enrichment performances without significant cellular agitation [207].

47

A similar technique, deterministic cell rolling, is used to sort cells based on their surface interactions with a microfluidic channel. Choi et al. showed that a series of successive ridges along the floor of a microfluidic channel can induce repeated cellular collisions for sorting [208]. In this device, the surface is modified with P-selectin to elicit a brief interaction with target cells and push those cells toward one side of the microfluidic channel (i.e., the gutter side). Non-target cells flow above the ridges toward the other side of the microfluidic channel (i.e., the focusing side) (Figure 17) [208, 209].

Figure 17: Cell sorting by deterministic cell rolling. Target cells (red) interact with the surface, roll across the ridges, and laterally displace toward the gutter side whereas non-target cells (green) flow over the ridges, not interacting with the surface, and exit on the focusing side. Reprinted with permission from Choi et al. [208]. Copyright 2012 Royal Society of Chemistry.

1.2.3.5.9 Cellular immobilization

Instead of relying on transient interactions between cells and a functionalized surface for sorting, which can suffer from limited throughputs due to the reliance of low

48

flow rates, many groups are investigating the non-transient capture, or immobilization, of cells on surfaces or in columns. One of the earliest approaches for fractionating cell populations by immobilization was through affinity chromatography, which sorts by the high affinity interactions between cell-surface receptors and immobilized ligands on the chromatography [210, 211]. This technique is capable of yielding results that do not depend on the size, density, or charge of the cells, and can be performed in various types of matrices [212].

Many of the more recent cell immobilization devices use antibodies for cell- specific isolation, often from whole blood [213-215]. For example, Hyun et al. used a microfluidic device coated with antibodies for the enrichment of CTCs [216]. Instead of using antibodies, Chen et al. developed a nanoroughened surface to selectively capture

CTCs based on their differential adhesion preference compared to non-CTCs [217].

Similarly, Singh et al. developed a shear-based system to isolate human pluripotent stem cells where intact colonies were rapidly isolated based on their adhesion strength to the microfluidic channel floor [218]. These techniques show promise for isolating rare cells such as CTCs due to their ability to process large sample volumes and recover rare cells from liquid biopsies (e.g., less than 1 cell in 1 mL of blood) without relying on surface markers (e.g., EpCAM), which are often downregulated during the mesenchymal- epithelial transition [2, 219].

49

The CTC-chip, which is the predecessor of the previously mentioned CTC-iChip, uses microposts conjugated with anti-EpCAM to immobilize CTCs [220]. In a similar system, antibodies conjugated to a pitched substrate are able to capture specific cells for rapid detection assays [221]. Yu et al. developed a magneto-controllable device where antibody-conjugated iron nanoparticles are magnetically localized around the

circumference of microposts within a microfluidic cavity, whereupon cancer cells

expressing the target antigen are captured for rapid release after disengaging the

magnetic field [222]. Saliba et al. designed a system using a similar concept, which

employs the self-assembly of antibody-labeled magnetic microbeads to form

hexagonally spaced pillars under an applied magnetic field (Figure 18) [223]. The self- assembled columns specifically capture and retain tumor cells that can be recovered for analysis after disengaging the magnetic field. Yoon et al. immobilized CTCs from blood using functionalized graphene oxide nanosheets [224]. Finally, Mittal et al. functionalized a fluid-permeable surface with antibodies for the selective capture of

CTCs [225].

50

Figure 18: Magnetic self-assembly of biofunctional magnetic beads for isolating rare cells. A) Schematic of a hexagonal array of magnetic ink (left) can guide the self- assembly of magnetic beads conjugated with anti-CD19 mAb in the presence of a vertical magnetic field (right). B) Photograph of the microfluidic device. Optical micrographs of the columns after C) the assembly of magnetic beads, D) the passage of 1,000 Jurkat cells (CD19 negative), and E) the passage of 400 Raji cells (CD19 positive) (scale bar: 80 µm). Reprinted with permission from Saliba et al. [223]. Copyright 2010 Proceedings of the National Academy of Sciences of the USA.

While antibodies are traditionally used to capture cells for sorting, aptamers also show promise for the high affinity capture of cells. Xu et al. developed an aptamer-based microfluidic system to passively capture CTCs, thereby enriching rare cells 135-fold in a single pass [226]. Similarly, Sheng et al. used microposts conjugated with aptamers for the isolation of CTCs [227] and Zhao et al. developed a biosensing network containing

51

DNA tentacles functionalized with aptamers along the terminal end for capturing cells

[228].

1.3 Commercialization of existing microfluidic cell sorters

The first cell sorting technology was developed by Mack Fulwyler in 1965 at Los

Alamos Scientific Laboratory [101]. Less than 10 years later, Becton, Dickinson and

Company (BD) in collaboration with Leonard Herzenberg of Stanford University sold the first commercial cell sorter circa 1974 [8]. The first chip-based microfluidic cell sorter, however, which used a spectrophotometer to detect cells in real time for displacement by mechanical pumps, was developed in 1972 by Kamentsky and Melamed [229]. Over

40 years later, of the many microfluidic cell sorters published in the literature (Figure

19), only a small handful of devices are actively progressing towards commercialization

(Table 1). Given the many technological innovations in the past two decades, these technologies are highly diverse, with some offering throughputs that equal leading

FACS devices on the market (i.e., 20,000 to 50,000 cells/sec) [11]. We divide these devices into two principal categories based on their separation modality. Active devices displace cells for sorting using external forces generated by magnetic, electric, acoustic or optical fields, and which usually require external powering equipment (N.B., some magnetic devices use permanent magnets and can thus sort cells without external powering equipment, as categorized separately in Table 1 (see “Active (Type I)” devices)). Passive

52

devices, on the other hand, do not require external fields to redirect cells or fluid flow, but instead sort via internal means such as antibodies for capturing specific cells or geometric features within the device for altering cellular flow (albeit, external power is usually required to move liquid through the device, but this is typical for all cell sorting devices).

Figure 19: Growth of microfluidic cell sorting technologies in the past 16 years. A) Number of publications versus publication year, organized by articles published in engineering, biology and medical journals and B) total number of citations per year that resulted from the query “microfluidic cell sorting” on the Web of Science (Thomson Reuters, Co.).

53

Table 1: Microfluidic cell sorting technologies that are commercialized, or are approaching commercialization, arranged by type of device and specifying product name, sorting mechanism, company, key metrics of sorting and application notes.

Type of Product Mechanism(s) Company Headquarters Key Metrics Application Notes Device (Acquired by) Passive ClearCell® FX Geometric Clearbridge Singapore 8 mL/hr of Automated and label-free System features to trap Biomedics, Pte whole blood, isolation of CTCs certain cells by Ltd. 10,000-fold size [230, 231] enrichment Disposable CTC- Affinity capture GPB Scientific, Richmond, 10 mL/hr of Separation of rare cells by Chip [220] LLC Virginia, USA whole blood immunospecific immobilization within the 54 microfluidic channel

CTC-iChip Deterministic Massachusetts Boston, ~36 mL/hr of Isolation of CTCs from Active (Type 1) lateral General Massachusetts, whole blood whole blood with displacement and Hospital USA (10,000 capabilities for positive and magnetophoresis cells/msec) negative selection (col. with [91] Janssen Diagnostics, LCC) LiquidBiopsy® Magnetophoresis Cynvenio Westlake 5 mL/hr of Automated isolation and [232, 233] Biosystems, Village, whole blood, staining of target cells from Inc. California, 1 cell/billion whole blood for (Distribution: USA sensitivity simultaneous isolation of Thermo Fisher CTCs and cell-free DNA Scientific, Inc.) Active Free Flow Free flow AcouSort AB Lund, Sweden N/A Label-free approach to

(Type 2) Acoustic Chip acoustophoresis acoustically sort cells by [104] size in a continuous flow microchip device

GigaSortTM Pneumatic Cytonome, Inc. Boston, ≤ 48,000 Parallel microchips (24,48 chambers to Massachusetts, cells/sec or 72) for sterile, high hydrodynamically USA throughput cell displace fluids identification and sorting [234] Jet Flow Sorter Bubble-jet flow JSR, Co. Leuven, ~2,000 On-chip imaging and cell generated from Belgium cells/sec identification for multi- Joule heating channel sorting (in [235] collaboration with IMEC vzw) MACSQuant® Fluorescence Owl Bergisch ≤ 30,000 Disposable cartridge with a Tyto detection and Biomedical, Gladbach, cells/sec high frequency switch for electro- Inc. Germany cell sorting with 95% 55 mechanical (Miltenyi viability

switching [236] Biotec GmBH)

Sorting Chip for Focuses and Sony San Jose, ≤ 10,000 Disposable cartridge that Cell Sorter SH800 detects cells for Biotechnology, California, cells/sec aligns automatically in a post-chip sorting Inc. USA permanent workstation for via FACS cell identification technology [237] SSAW Standing surface Ascent Bio- State College, N/A Label-free sorting of cells Acoutophoresis acoustic wave Nano Pennsylvania, using “acoustic tweezers” Chip (SSAW) [52] Technologies, USA Inc. WolfTM Cell Fluorescence NanoCellect San Diego, ~330 Sorts cells while removing Sorter detection and Biomedical, California, cells/sec contaminants such as cell- acoustophoretic Inc. USA free DNA sorting [238, 239]

1.4 Barriers to commercialization and clinical translation

Despite these numerous advances in microfluidic cell sorting, relatively few systems are progressing towards translation (Table 1) [240]. We believe there are a number of factors contributing to this phenomenon. We divide these factors into device- related barriers and commercialization-related barriers, as discussed below.

1.4.1 Device-related barriers

Device-related barriers typically arise from the focus of research labs to produce

new technologies, often at the expense of practicality for the end user. For example,

microfluidic devices usually contain multiple complex components (e.g., fluidic pumps,

valves and sometimes a microscope station and/or a field source generator) that require

control for operation and sometimes coordinated data acquisition (Figure 20). They can

contain several fluidic junctions that must be manually connected to fluidic lines (e.g.,

pneumatic lines [241]), which increases their footprint and renders setup time-

consuming. Not only so, these junctions often require custom tubes and adaptors, thus impeding their integration with standard laboratory tools [242]. These challenges can greatly hinder user adoption and result in the failure of addressing market needs.

Second, the lifetimes of microfluidic devices can be relatively short compared to standard benchtop tools. Due to the compact dimensions of the microchannels, air bubbles and other small obstructions can dramatically diminish their performance in cell sorting applications. As such, the lifespan of microfluidic devices is often curtailed by

56

buildup of debris from cell suspensions, residue from native biofluids (e.g., coagulants

and biofoulants) and aggregates of reagents and synthetic labeling materials. These

aggregates can induce cavitation, which further diminishes performance and

throughput, which necessitates longer operating times and becomes especially

problematic when isolating rare cells [5].

Figure 20: Conventional microfluidic cell sorting device setup. Top left: microscope station for inspecting the sorting junction; top right: the field source generator of the microfluidic device (required for most active devices); bottom left: microfluidic device with tubes connected to the inlets and outlets; bottom right: an automatic syringe pump.

1.4.2 Commercialization-related barriers

Commercialization-related barriers, which relate to intellectual property and market need (among other things), are a direct extension of the former set of barriers and are chiefly responsible for the slowed integration of microfluidic devices into the marketplace. The first major barrier of this type is the saturated landscape for new intellectual property, which was not always a major issue. In the 1960s, many of the

57

patents awarded by Los Alamos Scientific Laboratory (e.g., the patent for the first cell

sorter developed by Mack Fulwyler in 1965) were made available for licensing on a

royalty-free, non-exclusive basis [101, 243]. This freedom to operate resulted in a number

of corporations working to commercialize cell sorting technologies simultaneously, with

the first commercial product appearing less than 10 years later (i.e., by BD in 1974) [8]. In

contrast, a flood of patent applications was filed in the 1990s to protect all potential uses

of microfluidic devices. Now, in the U.S. alone, there are over 2,750 issued patents with the word “microfluidic” in their claims (from an online search on the USPTO patent full- text and image database). These patents, which contain broadly written claims, have sequestered much of the available space for the acquisition and implementation of new intellectual property. Consequently, many promising new technologies may fail to realize their commercial potential due to a lack of freedom to operate (i.e., intellectual security or threat of patent infringement), discouraging inventors from pursuing startup

(or other) ventures and dissuading licensors or investors from financing these endeavors.

The second barrier of this type is the complexity of the target market. An enormous amount of time, money and effort will be required to change the infrastructure of the cell sorting industry from manufacturing benchtop devices to microfluidic tools. Importantly, this change will never take place without a compelling advantage by a new technology fulfilling an unmet need in an efficient and practical

58

manner [244]. As such, it is essential for new technologies to possess a high market potential (i.e., a major competitive advantage in terms of improved overall performance or a significant reduction of cost, or preferably both). Incremental advances in technology are insufficient for driving the shift of the current market infrastructure to microfluidic tools for cell sorting. As shown in Figure 19, the vast majority of literature on microfluidic cell sorting appears in engineering journals, whereas very few articles have advanced into medical journals. In order to better understand the potential market for microfluidic cell sorting, significantly more biological and medical translational research must be conducted.

Thirdly, and as a direct result of the previous two barriers, limited investment opportunities post-invention have greatly hindered the forward progress of microfluidic cell sorters. The entry of these technologies into the marketplace is largely dependent on successful funding, which broadly encompasses early, midterm and late commercial investments (e.g., from bootstrapping, angel investors, corporations and venture capital firms), as well as subsidies to catalyze their intellectual property, development, optimization, standardization, manufacturability, marketability and distribution [245].

Funding is dependent on a variety of factors, including the perception of commercial potential, economic factors and regulatory standards (e.g., especially for devices that involve clinical use). In the U.S., the pathways to regulatory approval are typically by

510(k) clearance (i.e., when the technology is substantially equivalent to a predicate

59

technology) or premarket approval (i.e., a submission to the FDA to show that a new

device is safe and effective), which is significantly more costly and time-intensive [244].

While critical to the translation of a clinical technology, regulatory affairs are extremely

time consuming and costly, thus requiring significant investment, which can be

problematic in a highly competitive market against well-established tools such as FACS machines. As such, to our knowledge, no microfluidic cell sorting technology has been approved for clinical use in the U.S. to date.

1.5 Overcoming the barriers to commercialization

The current market for cell sorting is projected to exceed $5 billion (USD) annually by 2019 [246]. This rapid growth has the potential to fuel the translation of many microfluidic cell sorting technologies to the market, especially if certain strategies are implemented, as we discuss below.

1.5.1 Device-related solutions

We believe the most significant factor contributing to the bottleneck of microfluidic cell sorting devices developed in the lab versus those undergoing translation is the philosophy of innovation. Microfluidic systems are usually developed and then adapted to fit a potential market need. However, this longstanding dogma must be rethought and reversed in order to have any chance of translation towards a significant commercial or clinical impact [247]. Innovators must formulate a hypothesis-

60

driven biological question that fulfills an unmet need, and then work to develop

microfluidics tools that address them.

As such, we believe there are several simple strategies can be implemented to

overcome challenges related to the design of microfluidic devices. First, user interfaces

must be simplified to minimize setup time and maximize ergonomic operation.

Integrating devices with universal tubes and adaptors will facilitate this operation

without specialized training that can easily be integrated into a laboratory or hospital

setting. Devising cartridge-based apparatuses (cf., MACSQuant® Tyto; Table 1) will

enable the facile loading and unloading of microfluidic devices into an external housing

unit to facilitate automation and reliability as well as reduce or obviate long term issues

related to clogging, fouling and the generation of bubbles. Making these cartridges

disposable could eliminate the need for serial cleaning steps and the risk of cross- contamination [248]. These changes will add to the commercial appeal due to perceivably higher user compliances, improved reusability and potential for sustained revenue over the life of the instrument [249]. Further, innovations in manufacturing and materials development may lead to a greater reduction in overall costs (e.g., from advances in 3D printing [250]) [251].

The development of multifunctional, modular microfluidic devices will help to firmly establish their utility in the laboratory and the clinic. Microfluidic devices can independently accomplish a variety of tasks (e.g., mixing [252], trapping [19], lysing [18]

61

and drug screening [14]); however, an easily constructible and reconfigurable device

with connectable and interchangeable parts for cell sorting has not, to our knowledge,

been developed. Such a design could reduce manufacturing costs and enable the

integration of microfluidic cell sorting devices in the clinical workspace without changes in infrastructure, thus allowing these devices to interface with existing laboratory supplies, increasing their analytical and therapeutic value and appeal to users. We note that the C1TM Single-Cell System by Fluidigm is a closely related example of such a device by offering multiple interchangeable integrated fluidic circuits to trap, lyse and sequence single cells (https://www.fluidigm.com/products/c1-system).

1.5.2 Commercialization-related solutions

Building a strong market strategy is critical to overcoming many of the barriers related to commercialization. It is imperative for inventors, entrepreneurs and product development experts to focus on the end market in addition to the product itself at all stages of development, from invention all the way to production. As described in a recent review, the number one reason behind the failure of most startup companies is the lack of a market (or failing to address the needs of a market) [245]. Building a

competent team with knowledge, expertise and vision is vital to increasing one’s

chances for successful commercial venture. However, even with a strong team,

understanding the competitive advantage of the device and building a robust market

62

strategy (i.e., one that addresses the needs, cultural milieu and values of the market) is

essential for success [249].

Aggressively filing patent applications and pursuing strategic licensing can help to reduce risk and add value to companies commercializing microfluidic cell sorting technologies [253]. Intellectual property should be developed in parallel with research, not as an afterthought or in a rush before publication. While it is not required, including strong, validating experimental data in a patent can greatly help reinforce that patent later on during times of intellectual infringement. The obligation to aggressively file patent applications and build a comprehensive patent portfolio can sometimes be perceived as a deterrent to commercialization; however, strategic licensing can mitigate this burden, whereby a technology is licensed to either a startup company or a more established entity with other, complementary patents. Such strategic licensing can allow the inventor and the licensor to forge a partnership to benefit both parties while advancing the technology as a whole. Also, field-of-use licensing enables inventors to restrict the licensee to use the technology only for a particular application, which can provide additional opportunities for licensing. In addition to licensing, forging strong partnerships between academic and industrial entities (e.g., as with the CTC-iChip) can foster an environment conducive for successful commercialization. Navigating the best path forward for securing and managing intellectual property is best decided on a case-

63

by-case basis; however it is essential to the funding, development and forward progress

of microfluidic technologies.

1.6 Conclusions

While the field of microfluidic cell sorting is still relatively new, its outlook is promising. Groundbreaking discoveries in cell sorting have revolutionized the ways in which cells can be separated and analyzed, enabling the realization of high-performance devices that rival the functionality and performance of standard benchtop tools, but with added features for enhanced cellular evaluation. However, several important questions must be answered for these devices to reach their full potential. As these questions are addressed and as innovation continues, we believe microfluidic cell sorters will soon revolutionize the ways in which cell-containing fluids are processed in the laboratory and the hospital.

1.7 Acknowledgements

This chapter contains material from two articles. The full citation for the first article is: Shields IV, CW; Reyes, C.; López, GP. “Microfluidic cell sorting: A review of the advances in the separation of cells from debulking to rare cell isolation,” Lab on a

Chip 2015. 15(5): 1230-1249. DOI: 10.1039/C4LC01246A. The Royal Society of Chemistry granted permission for reuse of this article in this dissertation. The full citation for the second article is: Shields IV, CW; Ohiri, KA; Szott, LM; López, GP. “Translating

64

microfluidics: Cell separation technologies and their barriers to commercialization,”

Cytometry Part B (Submitted).

This work was supported by the National Science Foundation's (NSF's) Research

Triangle Materials Research Science and Engineering Center (MRSEC, DMR-1121107), the National Institutes of Health (R21GM111584), and the NSF Graduate Research

Fellowship Program (GRF-1106401). We gratefully acknowledge Professor Steven W.

Graves at the University of New Mexico as well as James Jett and John Martin (formerly at the Los Alamos National Laboratory) for helpful discussions.

65

2. Elastomeric Particles for Use in Acoustofluidic Devices

Elastomeric particles can provide a unique advantage in acoustofluidic systems due to their negative acoustic contrast behavior, thus separating from most biological moieties. A common elastomer for these applications is polydimethylsiloxane (PDMS), which easily synthesized in bulk. Previously, we have synthesized elastomeric particles using rapid homogenization techniques; however, these methods typically suffer from poor control over particle size and size dispersity [93]. As an alternative, we and other groups have synthesized elastomeric particles using microfluidics, which can provide narrow size distributions, but suffer from low particle production rates [92, 254]. In this chapter, we discuss the synthesis of monodisperse elastomeric particles made in bulk

(via nucleation and growth) that can display negative acoustic contrast factors in water.

2.1 Nucleation and growth synthesis of elastomeric particles

Nucleation and growth methods offer scalable means of synthesizing colloidal particles with precisely specified size for applications in chemical research, industry and medicine. We have used nucleation and growth methods to prepare a class of silicone gel particles that display a range of programmable properties and narrow size distributions. We demonstrate that the acoustic contrast factor of these particles can be tuned such that they undergo acoustophoresis to either the pressure nodes or antinodes of acoustic standing waves in water. We also demonstrate how these particles can be

66

synthesized to display surface functional groups that can be covalently modified for a

range of bioanalytical and acoustophoretic sorting applications.

Several groups have used biofunctional, polymeric particles in suspension to isolate proteins [92, 255], sort cells [25], and deliver drugs, genes or small molecules

[256]. An emerging use of these particles is in acoustofluidic systems, which use standing pressure waves for rapid filtration [38], sorting [106], and flow cytometry [257].

While the majority of laboratory and commercial particles display positive acoustic contrast behavior by focusing along the nodes of an acoustic standing wave, we have shown that elastomeric silicone particles can display negative acoustic contrast behavior and can be used to isolate small bioanalytes and cells to the pressure antinodes for sorting [49, 92, 93]. Previously, we have synthesized elastomeric particles using rapid homogenization techniques; however, these methods typically suffer from poor control over particle size and size dispersity [93]. As an alternative, we and other groups have synthesized elastomeric particles using microfluidics, which can provide narrow size distributions, but suffer from low particle production rates [92, 254].

We have implemented methods for the nucleation and growth of alkoxysilane and silicon alkoxide monomers to develop a class of functional, monodisperse and acoustically programmable (FMAP) particles that can be synthesized in bulk (Figure 21).

These particles (i) can display ample surface groups (e.g., vinyl, acrylate) for high yield conjugations, (ii) exhibit narrow size distributions (e.g., less than 15% CV) and (iii)

67

exhibit tunable acoustic behaviors (i.e., positive or negative acoustic contrast factors, φ)

for versatile manipulations in acoustofluidic systems. FMAP particles can display φ > 0

or φ < 0 behavior by migrating to the node(s) or antinodes of an acoustic standing wave, respectively. The φ is dependent on the density and compressibility of the particle and its suspending fluid; particles with higher densities and bulk moduli relative to the fluid tend to focus along the pressure node(s), whereas particles with the opposite properties

(e.g., elastomeric particles in water) tend to focus along the pressure antinodes [93].

68

Figure 21: Silicone-based particles formed by nucleation and growth synthesis can A) contain various functional groups (e.g., hydroxyl, vinyl and acrylate) for surface modification, B) exhibit narrow size distributions and C) display tunable acoustic properties.

2.2 Methods for elastomeric particle preparation

We used three approaches of a nucleation and growth process to prepare particles from an assortment of alkoxysilane and silicon alkoxide monomers (e.g., trimethoxymethylsilane (TMOMS), dimethoxymethylsilane (DMODMS), vinyltrimethoxysilane (VTMOS), vinylmethyldimethoxysilane (VMDMOS), 3-

(trimethoxysilyl)propyl acrylate (AcTMOS) and tetramethyl orthosilicate (TMOS)). Each approach involves the hydrolytic cleavage of alkoxide groups in the monomers followed by the uniform polycondensation of those monomers in an alkaline catalyst [258]. 69

Figure 22: Particles synthesized from Approach I required that Steps 1 and 4 be executed together, followed by Step 5. Particles synthesized from Approach II required Steps 1, 4, and 5 to be executed in sequence. Particles synthesized from Approach III required Steps 1-5 to be executed in sequence.

Approach I follows the Stöber process in which a small volume of monomers

was added to water and a co-solvent [259]. The monomers underwent both hydrolysis and polycondensation after the addition of a catalyst. To follow this approach, 4.0 mL of ethanol (200-proof, Sigma) were added to 4.6 mL of deionized water (~18 MΩ·cm) as a co-solvent. Then, a given volume of monomers (typically 100 μL) was added to solution with 1.0 mL of NH4OH (14.5 M, Sigma) and was vigorously mixed (i.e., placed in a vortex mixer at a speed of 4-6, Vortex Genie 2, SI-0236, 120 V) for 0.5 hrs.

The second method (i.e., Approach II) is similar to an approach described previously [260, 261]. This approach, which can be used to make relatively large (e.g., 10

70

µm) particles, decouples the hydrolysis and polycondensation steps of Approach I by

hydrolyzing the monomers at a low pH and later polymerizing those monomers at

higher pH. To follow this approach, a given volume of monomers (typically 1.0 mL) was added to 10.0 mL of 0.22 mM HCl and stirred for 2-18 hrs at 500 rpm. Then, 647 μL of

0.21 M NH4OH were added to the solution for an additional 0.5 hrs of stirring.

Approach III introduces a centrifugation step prior to polycondensation, which generated particles with the narrowest size distribution due to the removal of large, non- uniform oligomers (Figure 23A) [262, 263]. To follow this approach, a given volume of monomers (typically 1.0 mL) was added to 2.0 mM HCl and was stirred for 2-18 hrs at

500 rpm. The solution was then centrifuged at 2000xg for 5 min. Next, 7.5 mL of the supernatant was diluted in 7.5 mL of 3.1x10-4 M HCl and stirred at 500 rpm for 5 min.

Then, 15 μL of triethylamine (Sigma, 99.5%) was added to the solution, with continued stirring for 0.5 hrs at 500 rpm.

For each approach, the concentration of monomers, the spin speed used during the growth phase, and the duration of the growth phase were sometimes modified to influence the size of the resulting particles. As necessary, surfactant (0.1-0.3 vol.% F-108,

Sigma) was added to increase colloidal stability. Further, we sometimes incorporated hydrophobic dyes (i.e., Nile red) into the particles (Figure 23B) for their visualization

(e.g., in the presence of calcein dyed mammalian cells) in an acoustofluidic chip (Figure

23C). To illustrate the versatility of the nucleation and growth process, we provide

71

examples of FMAP particles synthesized by all three approaches and highlight the

properties of particles synthesized from TMOMS, VTMOS and varying molar ratios of

DMODMS:TMOS (8:1, 24:1, 48:1, 72:1 and 96:1).

2.3 Means of controlling particle size

By regulating the type of monomers, their concentration and reaction time, we

controlled the size of particles over a large range (e.g., 230 nm to 14.8 µm; Figure 23D-F).

We observed that particles with the narrowest size distribution (i.e., lowest size

dispersity) were synthesized from TMOMS, VTMOS, or high molar ratios (e.g., 96:1) of

DMODMS:TMOS. To measure particle size, we used tunable resistive pulse sensing

(TRPS), which exploits the Coulter principle in a qNano device (IZON Science Ltd.)

[264]. Prior to sizing, particles were stabilized with surfactant (0.1 vol.% F-108, Sigma) and diluted in 1xPBS.

72

Figure 23: A) SEM image of particles synthesized from VTMOS. B) Bright-field (left) and fluorescence (right) images of Nile red-impregnated particles synthesized from a 96:1 molar ratio of DMODMS:TMOS. C) Particles from (B) and a mammalian cell (KG-1a, dyed green) focused along the antinodes and node of an acoustic standing wave, respectively (dashed white lines represent the walls of the microchannel). Particles synthesized from different concentrations of (D, E) TMOMS and (F) a 96:1 molar ratio of DMODMS:TMOS, displaying an average size of 230±17 nm, 2.6±0.1 µm and 14.8±0.8 µm, respectively.

We found that particles synthesized from higher concentrations of monomer in water exhibited larger sizes (Figure 24A); and particles synthesized from larger molar ratios of DMODMS:TMOS exhibited smaller sizes (Figure 24B). We also found that higher spin speeds during the growth phase of particle synthesis resulted in smaller particle sizes (Figure 24C).

73

Figure 24: Mean particle size as a function of the A) monomer concentration for two DMODMS:TMOS ratios (8:1 and 96:1), B) molar ratio of DMODMS:TMOS and C) spin speed during the growth phase. Error bars represent the standard deviation of the mean.

Below we show the sizing results summarized in Figure 24 in tabular form as measured by a qNano device (IZON Science Ltd.). The sizes are averaged from five trials and the coefficient of variation (CV) reflects the average size dispersity by particle for those trials. In Table 2, we show the size of particles synthesized from various monomer concentrations (i.e., 1.92, 5.55, 8.91, 12.05, and 14.98 vol.%) of 8:1 and 96:1 molar ratios of

DMODMS:TMOS in water (spin speed of 500 rpm; Figure 24A). In Table 3, we show the size of particles synthesized from various molar ratios (i.e., TMOMS, 8:1, 24:1, 48:1, 72:1, and 96:1) of DMODMS:TMOS (8.91 vol.% monomer concentration and spin speed of 500 rpm; Figure 24B). In

Table 4, we show the size of particles synthesized from various spin speeds during the growth phase (i.e., 100, 300, 500, 700, and 900 rpm) for 8:1 and 96:1 molar ratios of

DMODMS:TMOS (8.91 vol.% monomer concentration; Figure 24C).

74

Table 2: Particle size and size dispersity by monomer concentration.

Monomer Concentration (vol.%) Size (nm) CV (%) 1.92 (8:1 DMODMS:TMOS) 813 9.1% 5.55 (8:1 DMODMS:TMOS) 1396 12.0% 8.91 (8:1 DMODMS:TMOS) 1889 14.7% 12.05 (8:1 DMODMS:TMOS) 2058 15.9% 14.98 (8:1 DMODMS:TMOS) 2216 14.1% 1.92 (96:1 DMODMS:TMOS) 605 20.7% 5.55 (96:1 DMODMS:TMOS) 1165 10.6% 8.91 (96:1 DMODMS:TMOS) 1505 11.4% 12.05 (96:1 DMODMS:TMOS) 1751 10.5% 14.95 (96:1 DMODMS:TMOS) 1954 11.2%

Table 3: Particle size and size dispersity by monomer ratio (8.91 vol.% monomer con.).

Monomer Ratio Size (nm) CV (%)

TMOMS 2260 19.2% 8:1 DMODMS:TMOS 1889 14.7% 24:1 DMODMS:TMOS 1686 12.9% 48:1 DMODMS:TMOS 1642 12.7% 72:1 DMODMS:TMOS 1581 13.1% 96:1 DMODMS:TMOS 1504 11.4%

Table 4: Particle size and size dispersity by spin speed during the growth phase (8.91 vol.% monomer concentration).

Spin Speed (rpm) Size (nm) CV (%) 100 (8:1 DMODMS:TMOS) 2339 15.3% 300 (8:1 DMODMS:TMOS) 2053 13.0% 500 (8:1 DMODMS:TMOS) 1889 14.7% 700 (8:1 DMODMS:TMOS) 1516 14.0% 900 (8:1 DMODMS:TMOS) 1309 14.6% 100 (96:1 DMODMS:TMOS) 2185 25.8% 300 (96:1 DMODMS:TMOS) 1855 28.7% 500 (96:1 DMODMS:TMOS) 1505 11.4% 700 (96:1 DMODMS:TMOS) 1169 8.3% 900 (96:1 DMODMS:TMOS) 998 8.8%

75

2.4 Means of controlling particle composition

We performed attenuated total reflectance (ATR)-FTIR on concentrated

suspensions of particles synthesized from TMOMS, an 8:1 molar ratio of

TMOMS:VTMOS, and an 8:1 molar ratio of TMOMS:AcTMOS to confirm the presence of

reactive functional groups (i.e., vinyl and acrylate). Following data acquisition, the

absorption spectra were smoothed in MATLAB using an internal smoothing algorithm

and analyzed. Particles prepared from TMOMS monomers displayed the characteristic

absorption peaks of polymethylsiloxane (i.e., absorption peaks near 2960 cm-1 indicating

CH3 bonds, 1,262 and 773 cm-1 indicating Si-CH3 bonds, and 1,115, 1,016, and 930 cm-1 indicating Si-O-Si bonds) (Figure 25A). Particles prepared from an 8:1 molar ratio of

TMOMS:VTMOS and an 8:1 molar ratio of TMOMS:AcTMOS also displayed the characteristic absorption peaks observed in polymethylsiloxane (PMS) at similar wavelengths. In addition to the PMS adsorption peaks, particles formed from an 8:1 molar ratio of TMOMS:VTMOS displayed an absorption peak corresponding to a carbon double bond at 1410 cm-1 (Figure 25B), which is a characteristic adsorption peak for materials containing vinyl groups. Particles formed from an 8:1 molar ratio of

TMOMS:AcTMOS displayed a carbon double bond absorption peak at 1410 cm-1 as well as a carbonyl absorption peak at 1576 cm-1 in addition to the PMS adsorption peaks

(Figure 25C), which are both characteristic adsorption peaks for materials containing

acrylate groups. The PMS particles lacked the absorption peaks at 1410 and 1576 cm-1,

76

indicating vinyl and acrylate groups were not present. The lower intensity of the 1410

and 1576 cm-1 adsorption peaks was due to the higher monomer ratio (i.e., 8:1) between

TMOMS and VTMOS or AcTMOS.

Figure 25: ATR-FTIR spectra of particles synthesized from A) TMOMS, B) an 8:1 molar ratio of TMOMS:VTMOS and C) an 8:1 molar ratio of TMOMS:AcTMOS.

77

2.5 Means of controlling particle surface properties and biofunctionality

We synthesized particles containing different reactive functional groups (i.e., vinyl and acrylate) by polymerizing functional monomers (e.g., VTMOS and AcTMOS).

To examine their colloidal stability, we measured the zeta potential of these functional particles in 1xPBS (without adding surfactant) using the TRPS method (Figure 26A)

[265]. Particles made from 0.96 vol.% VTMOS (average size: 247±51 nm) displayed an average zeta potential -36.2±3.2 mV. By comparison, much larger particles made from

8.91 vol.% TMOMS (1,221±87 nm) displayed an average zeta potential of -29.7±2.4 mV.

The zeta potential of these particles indicates moderate colloidal stability without surfactants [265].

Next, we investigated the covalent modification of the surface of FMAP particles for the attachment of biomolecules. While we have shown that nonspecific physical adsorption can immobilize proteins on silicone particles for biosensing [92] and cellular binding [49], covalent biofunctionalization provides a more stable means of attachment.

To do this, we prepared FMAP particles from a 24:1 molar ratio of VMDMOS:TMOS for a thermally-initiated surface reaction of thiol-polyethylene glycol (PEG)70-biotin (MW

3400, Nanocs, Inc.). A 1.0 mL suspension of particles was washed (i.e., centrifuged at

5,000xg for 5 min and resuspended in deionized water) to remove free alcohols, catalyst and unreacted monomers from solution. A second solution was prepared by adding 16.9 mg of 2,2’-azobis(2-methylpropionamidine) dihydrochloride (V-50, 97%, Sigma) to a 3.4

78

mL solution of a 4:1 volume ratio of ethanol to water and was vigorously mixed for 10 min (i.e., placed in a vortex mixer at a speed of 10, Vortex Genie 2). A third solution was prepared by dissolving 34 mg of thiol-PEG-biotin in 1 mL of deionized water. Finally,

1.0 mL of the washed particles, 1.0 mL of the V-50 solution, and 200 µL of the thiol-PEG- biotin solution were mixed and stirred at 125 rpm for 5 hrs at 70°C. The mixture was then cooled at 4°C for 10 min, centrifuged at 5,000xg for 5 min, and re-suspended in 1.0 mL of 1xPBS. Finally, 100 μL of 33.3 μM Alexa Fluor® 488-labeled streptavidin (SA,

Invitrogen) was added to the suspension and stirred at 125 rpm for 20 hrs at 25°C.

Prior to analysis by flow cytometry, the particles were centrifuged again at

5,000xg for 5 min and resuspended in 1xPBS to remove unbound SA. The particles were then loaded into a flow cytometer (C-6 Accuri, BD Biosciences) and inspected using a blue laser (488 nm, FL1) without gating. A calibration test was performed using a set of calibration beads (Spherotech, 8-Peak Rainbow Calibration Particles) to convert the raw data (i.e., channel counts) to the molecules equivalent of fluorescence (MEFL) of fluorophores per particle. The molecules of SA per particle was then estimated (Figure

26B) by dividing the MEFL by the average number of fluorophores on each SA molecule

(provided by the manufacturer, Invitrogen), which was four in the case of the Alexa

Fluor 488-labeled SA used in this study.

Using the method described above, we also analyzed unmodified (i.e., bare) particles, unmodified particles adsorbed with Alexa 488-SA, and thiol-reacted particles

79

adsorbed with biotin-blocked Alexa 488-SA (Figure 26B). The suspension of bare

particles was prepared by centrifuging 1.0 mL of the unmodified particle suspension at

5,000xg for 5 min, resuspending those particles in 1.0 mL of deionized water,

centrifuging the suspension again at 5,000xg for 5 min, and resuspending those particles

in 1.0 mL of 1xPBS. The 1.0 mL suspension of Alexa 488-SA adsorbed particles was

prepared by adding 100 μL of unblocked 33.3 mM Alexa 488-SA and 1.0 mL of 1xPBS to

the particles then mixing at 125 rpm for 20 hrs at 25°C. Lastly, the 1.0 mL suspension of

thiol-reacted particles adsorbed with biotin-blocked Alexa 488-SA was prepared by

using the same method described above, but the 100 µL of 33.3 mM Alexa 488-SA was

first mixed with 1.0 mL of 4.2x10-2 M NH4OH (Sigma) containing 5.0 mg of free biotin

(Sigma). The solution of blocked-SA was then added to the particles and stirred at 125

rpm for 20 hrs at 25°C. The suspensions of bare particles, unmodified particles adsorbed

with Alexa 488-SA, and thiol-reacted particles adsorbed with biotin-blocked Alexa 488-

SA were each centrifuged at 5,000xg for 5 min and resuspended in an equal volume of

1xPBS immediately prior to inspection by flow cytometry and analyzed using the method described above.

The resulting peaks (i.e., modes) in the number of SA molecules for particles nonspecifically adsorbed with SA, biotinylated particles adsorbed with biotin-blocked

SA, and biotinylated particles adsorbed with unblocked SA were measured to be approximately 5x103, 1x103, and 2.7x104, respectively. The larger mode observed for the

80

unblocked, biotinylated polyvinylsiloxane particles compared to the controls indicated

that the SA adsorption was biospecific, an important hallmark for controlled biomolecular recognition at interfaces [265].

Figure 26: A) Zeta potential and size of particles made from TMOMS and VTMOS. B) Histograms of the number of adsorbed molecules of SA per particle as measured by a flow cytometer (particles synthesized from a 24:1 molar ratio of VMDMOS:TMOS). From top left to bottom right: bare particles, bare particles with adsorbed SA, biotinylated particles with adsorbed biotin-blocked SA, and biotinylated particles with adsorbed, unblocked SA.

81

2.6 Means of measuring and controlling particle acoustic contrast factor

We controlled the acoustic responsiveness of FMAP particles by selecting

different molar ratios of alkoxysilane and silicon alkoxide monomers that could form

different numbers of siloxane bonds. For example, particles synthesized from monomers

that predominately formed three (e.g., TMOMS) or four (e.g., TMOS) siloxane bonds

exhibited positive acoustic contrast behavior, and particles synthesized predominantly

from monomers that form two siloxane bonds (e.g., DMODMS) exhibited negative

acoustic contrast behavior. The latter focusing behavior (i.e., along the pressure

antinodes) is useful since cells generally focus along the pressure node (Figure 23C),

allowing for rapid, continuous sorting in acoustofluidic systems [49, 92, 93].

The acoustic properties of FMAP particles were probed using an acoustic

traveling wave to spatially displace them in a large water bath (Figure 27). This

unconstrained experimental setup avoided boundary effects and acoustic attenuation

that can complicate acoustic radiation force measurements on particles within a tube or

microfluidic chip, while at the same time enabling the observation of individual particles

translating in response to a precisely characterized acoustic field [266]. A 1.0 MHz high- power piston transducer (Valpey Fisher Corp., Hopkinton, MA) was calibrated with a needle hydrophone (Onda Corp., Sunnyvale, CA). The hydrophone was then used to align the focal point of the transducer with the optical focus of a 100x water immersion objective interfaced with a high-speed digital camera (Fastcam SA1, Photron USA, Inc.,

82

San Diego, CA). A blunt 27G needle was placed at the bottom of the water bath to inject

particles such that they would pass through the mutual focal point of the transducer and

camera (Figure 27). The transducer was driven by a waveform generator (Tektronix, Inc.,

Beaverton, OR) connected to an amplifier (ENI, Rochester, NY) to generate sinusoidal

waveforms with pulse lengths of 50,000 cycles at seven discrete pressures (300, 400, 500,

600, 700, 800, and 900 kPa). We analyzed the high speed video of the migration of the

particles to determine their displacement by the acoustic wave.

Figure 27: Schematic of the acoustic displacement and optical tracking system for measuring the acoustic properties of particles via a traveling wave. Particles travel upward (i.e., against gravity) and are displaced by the incoming traveling wave by an extent proportional to their acoustic properties. Particles are tracked in real time using a water-immersion objective focused on the particle stream coupled to a high-speed camera.

We synthesized six batches of particles from TMOMS and different molar ratios of DMODMS:TMOS, measured the displacement of each particle type in response to an acoustic traveling wave (1 MHz), and tracked their migration with a high speed camera

[266]. Using the confocal optical and acoustic system shown in Figure 27, we evaluated

83

the acoustic response parameter, ψp, as well as the density, ρp, compressibility, βp, and

the acoustic contrast factor, φp, for different FMAP particles (Figure 28). Prior to

exposure to the incident wave, particles traveled only in the z-direction, against gravity.

Therefore, once the piston produced acoustic perturbations, any observed displacement

of particles in the x-direction was caused by acoustic radiation forces from the incident

traveling wave (see Table 5 for a list of nomenclature) [267]:

2 | | ( ) = 3 2 + + 2 1 2 6 2 2 2 (2) 𝜋𝜋9𝜌𝜌02𝐴𝐴+ 𝑘𝑘𝑘𝑘 𝜌𝜌0 𝜌𝜌0𝑐𝑐0 𝜌𝜌0 𝐹𝐹𝐴𝐴𝐴𝐴 2 �� − � � 2� � − � � 𝜌𝜌0 𝜌𝜌𝑝𝑝 𝜌𝜌𝑝𝑝𝑐𝑐𝑝𝑝 𝜌𝜌𝑝𝑝 � � Table 5: Nomenclature of𝜌𝜌𝑝𝑝 terms used for determining the acoustic contrast factor.

Symbol Definition Expression

A Potential amplitude of the acoustic particle velocity [266] = P/ρ0ω P Amplitude of the pressure wave

ρ0 and ρp Density of the medium and the particle, respectively c0 and cp Speed of sound in the medium and the particle, respectively f Driving frequency ω Angular frequency = 2πf k Wavenumber of the medium = ω/c0 Velocity of an immersed particle Radius of an immersed particle 𝑣𝑣 η Dynamic viscosity of the fluid and Compressibility of the medium and the particle, respectively Acoustic contrast factor of the particle 𝛽𝛽0 𝛽𝛽𝑝𝑝

𝜑𝜑𝑝𝑝 Using Equation (3) for the acoustic response parameter:

3 2 + + 2 1 2 2 2 0 0 0 0 ( , ) = 𝜌𝜌 𝜌𝜌 𝑐𝑐2 𝜌𝜌 �� − � 𝑝𝑝� � � − 𝑝𝑝� � (3) 𝜌𝜌9 𝜌𝜌2𝑝𝑝+𝑐𝑐𝑝𝑝 𝜌𝜌 𝜓𝜓𝑝𝑝 𝜌𝜌 𝑐𝑐 2 𝜌𝜌0 � � 𝜌𝜌𝑝𝑝 84

and the expression for the amplitude of the acoustic particle velocity potential, A (shown

above in Table 5), we arrived at a simplified expression for the acoustic radiation force

resulting from a traveling wave:

2 = 2 4 6 (4) 𝜋𝜋𝑃𝑃 𝜔𝜔 𝑅𝑅 𝐹𝐹𝐴𝐴𝐴𝐴 6 𝜓𝜓𝑝𝑝 We assumed that friction between the microparticle𝜌𝜌0𝑐𝑐0 and the surrounding medium

results from Stokes’ drag Equation (5) [268]:

= 6 (5)

𝐹𝐹𝐷𝐷 𝜋𝜋𝜋𝜋𝜋𝜋𝜋𝜋 After a brief period of acceleration, the particles reached a constant velocity, v, such that the acoustic radiation force is assumed to be equivalent to the Stokes drag force, we therefore obtained the following expression governing the particle dynamics [269]:

2 6 = (6) 2 4 6 𝜋𝜋𝑃𝑃 𝜔𝜔 𝑅𝑅 6 𝑝𝑝 𝜋𝜋𝜋𝜋𝜋𝜋𝜋𝜋 0 0 𝜓𝜓 The constant terms were collected into a single𝜌𝜌 𝑐𝑐 term, H, for convenience:

= = (7) , where 6 𝑣𝑣 2 3𝜌𝜌0𝑐𝑐0 𝜂𝜂 5 4 𝐻𝐻 𝑅𝑅 𝑃𝑃 𝜓𝜓𝑝𝑝 𝐻𝐻 𝜔𝜔 Linear regression of Equation (7), in which P was the independent variable and v was the dependent variable, yielded the acoustic response parameter, ψp, as its slope

Equation (8):

= (8) 𝑣𝑣𝑣𝑣 𝜓𝜓𝑝𝑝 2 5 Next, to extract the compressibility and acoustic𝑃𝑃 𝑅𝑅 contrast factors of different types of

FMAP particles, we measured their density using a series of centrifugations in salt

85

solutions of known densities as measured by an automated pycnometer (DSA 5000 M,

Anton Paar GmbH; see Table 6 and Figure 28A). We then used the measured acoustic response parameter (Equation (3)) to determine the speed of sound in each type of particle. Using the Newton-Laplace equation (Equation (9)), we calculated the compressibility of each type of particle (see Figure 28A and Table 6 below):

1 = (9)

𝑐𝑐𝑝𝑝 � Lastly, using the values for density and compressibility,𝛽𝛽𝑝𝑝𝜌𝜌𝑝𝑝 we calculated the acoustic

contrast factor for each type of particle (Figure 28B, Table 6) [45]:

1 5 2 = (10) 3 2 𝑝𝑝 + 0 𝑝𝑝 𝑝𝑝 𝜌𝜌 − 𝜌𝜌 𝛽𝛽 𝜑𝜑 � 𝑝𝑝 0 − 0� Due to the assumption of an ideal host fluid𝜌𝜌 in𝜌𝜌 Equation𝛽𝛽 (2) [267], the acoustic response parameters are relative measures of the acoustic properties of the FMAP particles.

Therefore, we measured 2 µm polystyrene beads (mean size of 1.89 µm as measured by a Coulter counter (MS3, Beckman Coulter, Inc.)) as a control in the confocal acoustic traveling wave setup to calibrate the results obtained. Using the known density and compressibility of polystyrene [270], we estimated the theoretical speed of sound (cp ≈

1816) from Equation (9) as a means to determine the theoretical acoustic response parameter of polystyrene (ψp ≈ + 0.045) using Equation (3). We then solved for the ratio between the theoretical acoustic response parameter and the measured acoustic response parameter of polystyrene and used this value to normalize the measured acoustic response parameters of each type of FMAP particle (Figure 28B). We also report 86

the approximate speed of sound of different types of FMAP particles using this method

(see Table 6 below). We found that particles migrated according to their composition, where particles synthesized from larger ratios of DMODMS:TMOS displayed a lower φp.

Table 6: Material and acoustic properties of different FMAP particles.

Density Compressibility Speed of FMAP Particle φ (kg/m3) (GPa-1) Sound (m/s) TMOMS 1040±10 0.40±0.05 1547±200 +0.05±0.01 8:1 DMODMS:TMOS 1030±5.8 0.54±0.10 1339±246 -0.05±0.01 24:1 DMODMS:TMOS 1020±10 0.97±0.19 1004±197 -0.37±0.07 48:1 DMODMS:TMOS 1000±5.8 1.12±0.17 944±139 -0.49±0.07 72:1 DMODMS:TMOS 980±5.8 1.49±0.21 827±114 -0.77±0.11 96:1 DMODMS:TMOS 960±0.0 1.65±0.17 791±82 -0.88±0.09

87

Figure 28: Properties of FMAP particles. A) Fundamental properties of particles by monomer composition: density, pp (solid line, left) and compressibility, β (dashed line, right). B) Acoustic properties of particles by monomer composition: acoustic response

parameter, ψp(ρ,c) (solid line, left) and acoustic contrast factor, φp (dashed line, right).

2.7 Qualitative verification of particle acoustic contrast factor

We then injected the same set of particles into an acoustofluidic microchip and measured their response to a standing wave under high flow conditions (i.e., 300

µL/min). As indicated by their acoustic focusing profiles at a fixed location along the microchannel [8] (Figure 29), particles synthesized from larger ratios of

DMODMS:TMOS (e.g., 96:1) exhibited φ < 0 behavior and focused closer to the pressure antinodes than particles made from smaller monomer ratios (e.g., 8:1). Moreover,

88

particles synthesized from TMOMS exhibited φ > 0 behavior by focusing along the pressure node. The relative slope of their focusing profiles correlated with the calculated acoustic contrast factors, indicating this synthesis method can be used to program the acoustic properties of particles in both traveling and standing acoustic waves by their monomer composition.

Figure 29: Evaluation of particles flowing through an acoustic standing wave. A) Fluorescence intensity profiles across the width of an acoustofluidic channel for different particles flowing through the channel as shown in (B) (300 µL/min flow rate; 30 Vpp sinusoid waveform; 2.96 MHz; λ ≈ 500 µm). B) Micrographs of fluorescent particles flowing at the node (top) and antinodes (bottom) of an acoustic standing wave.

89

2.8 Conclusions

In summary, we show that siloxane-based particles made by nucleation and

growth can provide a precise means for programming particle size, surface functionality

and acoustic properties. These particles are useful for applications in bioanalytical

acoustofluidics as well as other applications, as they are rapidly generated in bulk,

biofunctional and easily programmable.

2.9 Acknowledgements

This chapter contains material from a previously published article: Shields IV,

CW; Sun, D; Johnson, K; Duval, K; Rodriguez, AV; Gao, L; Dayton, PA; López, GP.

“Nucleation and growth synthesis of functional, monodisperse and acoustically programmable particles,” Angewandte Chemie International Edition 2014. 53(31): 8070-

8073. DOI: 10.1002/anie.201402471. John Wiley and Sons granted permission for reuse of this article in this dissertation.

This work was supported by the National Science Foundation’s (NSF’s) Research

Triangle MRSEC (DMR-1121107) and the NSF Graduate Research Fellowship Program

(1106401). We thank Alexander A. Doinikov for his assistance with the theoretical

considerations of particles in an acoustic field.

90

3. Acoustofluidic Devices: Fabrication and Operation

Acoustophoresis refers to the displacement of suspended objects in response to directional forces from sound energy. Given that the suspended objects must be smaller than the incident wavelength of sound and the width of the fluidic channels are typically tens to hundreds of micrometers across, acoustofluidic devices typically use ultrasonic waves generated from a piezoelectric transducer pulsating at high frequencies

(in the megahertz range). At characteristic frequencies that depend on the geometry of the device, it is possible to induce the formation of standing waves that can focus particles along desired fluidic streamlines within a bulk flow. Here, we describe a method for the fabrication of acoustophoretic devices from common materials and clean room equipment. We show representative results for the focusing of particles with positive or negative acoustic contrast factors, which move towards the pressure nodes or antinodes of the standing waves, respectively. These devices offer enormous practical utility for precisely positioning large numbers of microscopic entities (e.g., cells) in stationary or flowing fluids for applications ranging from cytometry to assembly.

3.1 Introduction

Acoustofluidic devices are used to exert directional forces on microscopic entities

(e.g., particles or cells) for their concentration, alignment, assembly, confinement or

separation within quiescent fluids or laminar flow streams [40]. Within this broad class of devices, forces can be generated from bulk acoustic standing waves, surface acoustic

91

standing waves (SSAWs) [42] or acoustic travelling waves [57]. While we focus on the fabrication and operation of devices supporting bulk acoustic standing waves, devices supporting SSAWs have received much attention recently due to their ability to precisely manipulate cells along surfaces [271] and rapidly sort cells in continuous flow channels [272]. Devices supporting bulk acoustic standing waves, however, rearrange particles based on the mechanical vibrations of the walls of the device generated by a piezoelectric transducer, which excites the standing waves in microfluidic cavities at geometrically defined resonant frequencies. This enables the potential for generating higher pressure amplitudes compared to SSAW devices, and thus, faster acoustophoretic transport of microscopic entities [273].

These standing waves consist of a spatially periodic set of pressure nodes and antinodes, which are fixed in position as the pressure oscillates in time. Particles respond to the standing waves by migrating to the pressure nodes or antinodes, depending on the mechanical properties of the particles relative to the fluid, and which are described by the acoustic contrast factor, ɸ (1) [45]. Entities that possess a positive acoustic contrast factor (i.e., ɸ > 0) migrate to the pressure node(s); whereas, entities that possess a negative acoustic contrast factor (i.e., ɸ < 0) migrate to the pressure antinodes

[45]. While the majority of synthetic materials (e.g., polystyrene beads) and cells exhibit positive acoustic contrast, elastomeric particles made from silicone-based materials [94], fatty molecules [102] or other highly elastic constituents exhibit negative acoustic

92

contrast in water. Elastomeric particles in acoustofluidic devices can be used to isolate

small molecules [92] and as means to confine synthetic particles [93] or cells [49] for the purposes of discriminate sorting [274].

Acoustofluidic devices are usually manufactured from standard materials (e.g., silicon and glass) that have sufficient rigidity to support an acoustic standing wave. In many acoustofluidic devices (including the device shown herein), the mechanical waves are designed to resonate at the lowest harmonic mode, which consists of a half- wavelength standing wave spanning the width of the microchannel. This configuration has a pressure node at the center of the channel and pressure antinodes along the peripheries of the channel. It has been shown previously that these systems can be used for chip-based cytometry applications [47, 275, 276] and applications ranging from the trapping of cells to the concentration of cells [38, 277].

3.1 Methods of device fabrication

We describe the process of fabrication, methods for use and representative performance capabilities of an acoustofluidic device that supports bulk acoustic standing waves. This device requires one photolithography step, one etching step and one fusing step to permanently bond a glass “lid” to the etched silicon substrate. We note that other acoustofluidic devices that support bulk acoustic standing waves can be fabricated from glass or quartz capillaries bound to piezoelectric transducers, which is described elsewhere [36, 278]. Silicon-based devices offer the advantages of robustness

93

and control over the flow channel geometry, which together allow for numerous types

of processing for samples containing suspensions of particles and cells. The devices are

reusable provided they are properly cleaned between uses (i.e., by flushing the device

with buffers and detergents).

3.1.1 Photolithography

We designed the photomask using an appropriate software package (AutoCAD,

Autodesk, Inc.) and submitted the design to a qualified photomask printer [279]. Then, in a clean room facility, we rinsed a 6” single-side polished Si wafer with a steady stream of acetone (≥99.5%; see Table 7) followed by a steady stream of methanol (99.8%; see Table 7). We dried the wafer by spraying with N2 gas and placing the wafer on a hot plate at 95°C for 2 min (N.B., the doping profile and crystal orientation of the wafers do not affect the following procedures). We protected the trough outside of the spin coater

(in a standard spin coat hood) by covering with a sheet of Al foil and place the clean Si wafer on the center of the vacuum chuck in the spin coater to secure the wafer.

Next, we deposited positive photoresist directly onto the center of the wafer by carefully pouring until the photoresist covers most of the wafer, taking care to ensure there no bubbles were present in the photoresist. We start the spin cycle by performing the following procedures:

(i) Program a speed of 300 rpm, a ramp of 100 rpm/sec, and a spin time of 5 sec to

begin the spin cycle.

94

(ii) Program to a speed of 1800 rpm, a ramp of 1000 rpm/sec, and a spin time of

60 sec to evenly spread the photoresist.

(iii) Program to a speed of 0 rpm, a ramp of 1000 rpm/sec, and a spin time of 0 sec

to conclude the spin cycle.

We released the vacuum on the chuck and used wafer tweezers to retrieve the wafer from the chuck. We placed the wafer onto a hot plate to bake at 110°C for 165 sec.

Next, we loaded the photomask into the holder of a mask aligner and programed the machine to provide an energy dosage of 1,400 mJ/cm2 (e.g., for an output intensity of

13.5 mW/cm2, use an exposure time of ≈103.7 sec). Then we removed the photopatterned

wafer from the holder and place it in a solution of its corresponding developer (see

Table 7) for 5 min. We removed the wafer from the developer, wash the wafer with a

steady stream of deionized H2O and dry it with N2 gas. N.B., over-developing caused patterns to swell, while under-developing caused incomplete removal of the photoresist along the photo-patterned features. Finally, we inspected the wafer under a microscope to confirm the patterns printed on the photomask were transferred to the photoresist.

Table 7: Materials for fabrication and basic testing of an acoustofluidic device supporting bulk acoustic standing waves.

Name Company Cat. No. Comments Silicon wafers Addison 3P1 6” mechanical grade Engineering, Inc. silicon wafer <111> AZ® 9260 photoresist MicroChemicals AZ9260-Q Positive photoresist GmbH AZ® 400K developer MicroChemicals AZ400K CONC- Dilute 1 part AZ 400k GmbH CS in 4 parts DI H2O 95

H2O2 Sigma Aldrich, 216763 30 wt.% in H2O Co.

H2SO4 Sigma Aldrich, 320501 ACS reagent, 95.0- Co. 98.0% 1165 Photoresist Dow Chemical, DEM-10018073 1-methyl-2- Remover Co. pyrrolidinone based Acetone Sigma Aldrich, 320110 ACS reagent, ≥99.5% Co. Isopropyl alcohol Sigma Aldrich, W292907 ≥99.7%, FCC, FG Co. Methanol Sigma Aldrich, 322415 Anhydrous, 99.8% Co. Borosilicate glass Schott AG 2098576 Size: 120x60±0.1 mm (Nexterion glass B) Thickness: 1±0.005 mm Drill bit for glass and McMaster-Carr, 2954A1 Drill bit size: 1/8” ceramic Inc. Overall length: 1 3/16” Shank diameter: 7/64” Polydimethylsiloxane Sigma Aldrich, 761036 Dow Corning, Co.; (PDMS) kit Co. Sylgard 184®; 10 g clip-pack Biopsy punch Ted Pella, Inc. 15078 Harris uni-core Tip ID: 3.0 mm Tip OD: 3.40 mm Lead zirconate titanate APC Custom order Length: 30.0 mm (PZT) transducer International, (841 WFB) Width: 5.0 mm Ltd. Freq.: 2.46 MHz 2.0 mm wrap for leads Silicone tubing Cole Parmer 07625-22 0.6 mm ID Instrument, Co. Polystyrene beads Thermo Fischer F-8836 10 µm yellow-green Scientific, Inc. fluorescence

3.1.2 Deep reactive ion etching

To perform the deep reactive ion etching, we loaded the photo-patterned Si wafer into the chamber of a deep reactive ion etching instrument and etch the fluidic

96

channels into the Si wafer to the desired depth following standard etching procedures

[280]. Next, we carefully unloaded the sample from the chamber after the etching

process was complete. To remove excess photoresist from the wafer, we prepared a large beaker with a solution of photoresist remover (see Table 7) in a well-ventilated hood dedicated to solvent use and placed it on a hot plate at 65°C. We submerged the wafer in the photoresist removal solution and let it soak for 1 hr. NOTE: Different solutions can be used to remove photoresist (e.g., a solution of acetone (≥99.5%; see Table 7) can remove the photoresist by soaking overnight). Finally, we removed the wafer from the beaker, rinsed it with alternating streams of acetone (≥99.5%; see Table 7) and isopropyl alcohol (≥99.7%; see Table 7), and dried the wafer with N2 gas.

3.1.3 Method for cleaning with piranha solution

In a well-ventilated hood (dedicated to the use of acids), we prepared a piranha

solution by adding H2O2 (30.0 wt.% in water; see Table 7) to H2SO4 (95.0-98.0%; see Table

7) to in a 1:3 ratio in a large, clean beaker. (N.B., Piranha solutions are highly corrosive, are a strong oxidizer and are highly dangerous. Take extreme care in handling piranha solutions and wear the proper safety equipment.) Next, we submerged the ion-etched wafer with the etched-features facing up into the solution and left it for 5 min. We then carefully removed the wafer and fully rinsed it with deionized H2O. We resubmerged

the wafer in the piranha solution for 2 min and repeated the rinse procedure with

deionized H2O. In a separate well-ventilated hood dedicated to solvent use, we washed

97

the wafer with a steady stream of acetone (≥99.5%; see Table 7) followed by a steady

stream of methanol (99.8%; see Table 7) and dried the wafer with N2 gas.

3.1.4 Preparing the borosilicate glass “lid”

Using a scribe tool, we etched straight lines into the borosilicate glass to create

rectangular segments (e.g., 8x4 cm). Then, we carefully snapped the glass to recover the rectangular segments. We took one of these glass segments and place it on top of a

printed copy of the desired design (with actual dimensions) to mark the location of the

inlets and outlets on the glass with a black marker. Using the marks, we drilled the inlet

and outlet holes into the borosilicate glass by fixing a 1/8” drill bit into the mouth of drill

press, placing the rectangular glass segment on top of an Al plate with drilled holes so

that the marks on the glass are above the holes in the Al plate, taping the glass segment

down with tape, and carefully lowering the feed handle to press the drill bit through the

glass. Once finished, we removed the tape and slowly lifted the glass to remove the glass

powder. We rinsed the glass powder from the wafer using warm water and dried it with

a non-lint-producing absorbent cloth and follow the same procedures to drill the other

inlet and outlet holes. Finally, we cleaned the rectangular glass segment with piranha

solution, as described earlier.

3.1.5 Anodic bonding

Using a scribe tool, we etched straight lines into the Si wafer around the

perimeter of the microfluidic chip such that it was slightly smaller than the rectangular

98

glass segment (e.g., 7x3 cm), and we carefully snapped the wafer along the etched lines.

We then rinsed the Si segment with a steady stream of acetone (≥99.5%; see Table 7)

followed by a steady stream of methanol (99.8%; see Table 7). Finally, we placed the

wafer on a hot plate at 95°C for 2 min to dry.

With the etched-features on the Si segment facing up, we carefully added the

clean glass on top of the Si segment and make sure the holes aligned properly. We then

carefully flipped the segments, while ensuring the holes were kept aligned. Since the

glass segment is larger than the Si segment, we secured the two segments with double-

sided tape such that half of the tape secured the vertical edges of the Si segment and the

other half of the tape secured the overhanging glass. We then flipped the segments again

such that the glass segment was on top and placed the segments on top of a metal slab

on a hot plate. We carefully added a second metal slab (e.g., steel) of a sufficiently heavy

weight (i.e., at least 5 kg) directly to the top of the assembled glass and Si segments.

N.B., the metal slab was not in contact with the Si segment or the conductive tape.

Finally, using a high voltage power supply, we connected one lead (power) to

the metallic slab on top of the assembled glass and Si segments and the other lead

(ground) to the bottom metallic slab. We turned the voltage on the underlying hot plate

to 1,000 V. We confirmed the applied voltage was 1,000 V by using multimeter, pressing one probe against the bottom plate and the other probe against the top plate. We left the hot plate at 450°C for 2 hr to allow the glass “lid” to anodically bond to the Si substrate,

99

and we returned after 2 hr to turn off the hot plate, turn off the DC power supply and

remove the device from the metallic slabs.

3.1.6 Finalizing the acoustofluidic device

We scraped the surface of the glass with a razor to remove grime produced by the anodic bonding and clean the surface of the glass with acetone. Next, we prepared a sheet of polydimethylsiloxane (PDMS) approximately 5 mm thick and cut several small, square slabs approximately 10 x 10 mm (see Table 7) [281]. Then, using a 3 mm biopsy

punch, we cut one hole in the center of each PDMS slab so as to insert the silicone tubing

through it, placed the slabs directly on top the holes on the glass substrate, and glued

the slabs with epoxy.

Next, we carefully glue the lead zirconate titanate (PZT) transducer to the Si

segment on the backside of the device, centered-underneath the microchannel. We

soldered two wires to the two conductive areas on the PZT transduce, ensuring that the

wires were securely attached to the PZT transducer. Finally, we inserted the silicone

tubing through the holes in the slabs of PDMS and added additional glue around slabs

and the tubing to secure their attachment.

3.2 Operating the acoustofluidic device

We securely mounted the device onto a microscope stage with the microchannel directly underneath the objective, ensuring the PZT transducer did not make contact with the stage by placing a small insert underneath the device. Using standardized

100

connectors, we connected the silicone tubes from the outlets of the device to syringes secured on syringe pumps. This configuration was intended for “withdrawal mode”; however, the syringe pumps may alternatively be used to inject the sample into the device. We placed the silicone tube leading to the inlet of the device in a vial containing the fluid sample (e.g., a suspension of polystyrene beads or cells). Finally, we placed the vial containing the fluid sample on a stir plate to continuously mix the sample and ensure that a constant concentration of particles or cells is maintained throughout the course of the experiment.

Next, we connected the wires from the PZT transducer to the output from a power amplifier in series with a function generator. We programed the settings on the function generator (e.g., peak-to-peak voltage and frequency) and monitored the output signal from the amplifier using an oscilloscope. We started the function generator and power amplifier to begin actuating the PZT transducer [273]. To estimate the resonant frequency of the device, we followed the simple relation c = λ*f, where c is the speed of sound of the medium (i.e., water), λ is the acoustic wavelength, and f is the frequency of the PZT transducer. In the case of a half-wavelength harmonic (which we show in the following section), the width of the microchannel was half the length of the standing wave.

Finally, we turned on the syringe pump to apply flow and introduce the sample into the device. We monitored the entities flowing through the device with the

101

microscope on fluorescence mode, and we ensured the device focused particles by adjusting the peak-to-peak voltage supplied to the PZT transducer to modify the pressure amplitude and by performing a frequency sweep near the expected resonant frequency to identify the empirical resonant frequency.

3.3 Results and discussion

We designed the acoustofluidic device to contain a trifurcating inlet, a main channel with a width of 300 µm and a trifurcating outlet (Figure 30A-B). We note that we only used one inlet for all experiments in this study (i.e., to achieve sheathless focusing of particles via acoustic radiation forces) by blocking the other inlets with removable plugs. Following the procedures described above, we constructed a chip possessing a channel width of 313 µm, with an error of ≈4% due to imperfections during the microfabrication process (Figure 30C-D). We operated the device at a driving frequency of 2.366 MHz to induce a half-wavelength harmonic standing wave.

Figure 30: Acoustofluidic device supporting bulk acoustic standing waves. Schematic views of the top (A) and bottom (B) of a device comprised of an etched silicon substrate fused to a borosilicate glass “lid”, polydimethylsiloxane (PDMS) blocks connected to silicone tubing and a piezoelectric transducer soldered to wires glued to

102

the bottom of the device. Photographs of the top (C) and bottom (D) of the device are also shown.

We used a signal generator connected to a power amplifier to generate the high frequency sinusoidal waveform to actuate the PZT transducer. We used an oscilloscope to measure the peak-to-peak output voltage (Vpp) generated from the power amplifier to

verify the fidelity of the signal shape and amplitude. Using a syringe pump, we first

injected a suspension of green fluorescent polystyrene beads at a rate of 100 µL/min

without actuation of the PZT transducer as a negative control (Figure 31A). Next, we

actuated the device at 2.366 MHz to form a half-wavelength standing wave across the

width of the microchannel (Vpp = 40 V; Figure 31B). We found that these particles, which have a positive acoustic contrast factor, focused along the pressure node as expected

[273]. We also injected red fluorescent particles with a negative acoustic contrast factor

(i.e., ɸ ≈ −0.88, synthesized from a process described previously) [94] to verify that our device could induce their concentration along the pressure antinodes (Figure 31C).

Figure 31: Acoustic focusing of particles with positive and negative acoustic contrast factors. A) Prior to actuation of the PZT transducer, particles with a positive acoustic contrast factor (10 µm, yellow-green polystyrene beads) flowing at 100 µL/min occupied the width of the microchannel. B) After the PZT transducer is actuated (Vpp = 40 V and f = 2.366 MHz), the particles in (A) are shown to focus along the pressure node of the standing wave. C) Particles with a negative acoustic contrast factor 103

focused along the pressure antinodes of the standing wave in the absence of applied flow (Vpp = 40 V and f = 2.366 MHz).

Finally, we explored the extent of focusing of particles with a positive acoustic contrast factor at a range of flow rates (i.e., 0 to 1,000 µL/min as regulated by a syringe pump) and voltages (i.e., 0 to 50 V). Videos comprised of 15 frames were collected for each condition. ImageJ software was used to sample five of the fluorescence intensity profile across the width of the microchannel. A numerical computing program was used to average the intensity profiles for each condition and to smooth the averaged data using an inline filtering program. As expected, the extent of particle focusing (i.e., as defined by the width of the fluorescence peak, corresponding to the width of the stream of particles) decreased with increasing flow rates (Figure 32A). We also found that the extent of particle focusing increased with increasing applied voltages (Figure 32B).

104

Figure 32: Focusing performance of an acoustofluidic device. Fluorescence intensity plots of polystyrene beads (shown in Figure 31A-B) are shown for (A) various flow rates (ranging from 0 to 1,000 -toL/min)-peak voltage with a constantof 40 V peak and (B) various applied voltages (ranging from 0 to 50 V) with a constant flow rate of 100 µL/min.

3.4 Conclusions

Acoustophoresis offers a simple and rapid approach to precisely arrange microscopic entities within fluidic microchannels without the need of sheath fluids used in hydrodynamic focusing approaches [282]. These devices provide several advantages over other methods of particle or cell manipulation (e.g., magnetophoresis [88, 283], dielectrophoresis [20] or inertial forcing [155]) due to their ability to process entities without high magnetic susceptibilities, electric polarizabilities or a narrow size dispersity. Furthermore, the focusing nodes of an acoustic standing wave can be positioned far from the source of excitation, which is something that is not possible by static magnetic or electric fields as per Earnshaw’s theorem [284]. An additional advantage is that acoustic devices can focus particles across a wide range of applied

105

flow rates and independent of the flow direction, which is not possible in devices that rely on inertial forces for focusing [155], providing the means to efficiently transport particles or cells for enhanced particle inspection for applications such as flow cytometry and particle sizing [11, 285]. The ease of device fabrication and operation can directly allow for the implementation of similar devices for focusing, concentrating, fractionating and sorting objects suspended in fluids [286].

We have shown that the primary radiation forces, which are the strongest forces produced by acoustic standing waves [40], can focus microparticles flowing through a microfluidic channel at flow rates exceeding 10 mL/hr for a single orifice design. For a fixed flow rate of 100 µL/min, we show that our device can focus particles into a narrow streamline (i.e., 50 µm across) without any sheath fluids at voltages as low as 20 Vpp, enabling a low-power method for the batchwise focusing of 10 million particles/min when processing densely concentrated solutions (e.g., 6x108 particles/mL), as an

example. Furthermore, this throughput can be dramatically increased by fabricating

multi-orifice acoustofluidic chips or channels that are actuated with higher harmonics to

produce sets of parallel nodes [257].

While the device shown herein only requires materials and methods used in conventional microfabrication, we emphasize that there are a handful of other techniques that can be used for constructing similar devices [36, 287, 288]. The

106

advantages of this approach include its simplicity as well as the durability of the final device.

The critical steps to the fabrication of these devices include photolithography to define the geometry of the microchannel, reactive ion etching to form the channel in the silicon and anodic bonding to fuse the silicon to a transparent “lid” for observation by fluorescence microscopy. All of these steps require clean room facilities to avoid the collection of dust or debris within the device. Once these steps are complete, however, bonding a PZT transducer and fluidic ports are relatively straightforward and can be performed outside of a clean room.

However, proper treatment of the device is essential for its longevity. This includes (i) incubating the device with passivating reagents (e.g., poly(ethylene glycol) silane) prior to each experiment to protect the channel from residue buildup and (ii) flushing the device with detergents after each experiment. Buildup of debris may compromise the fidelity of the acoustic standing wave and may reduce the ability to efficiently focus particles or cells within the device. We also note that these devices are not well suited for highly polydisperse samples or samples containing entities approaching half of the size of the standing wave.

Acoustofluidic devices provide enormous utility for a variety of applications spanning from colloidal assembly to cell separation and flow cytometry. The ability to process biological samples with precision at high flow rates can allow for the ability of

107

increased throughputs by these microfluidic devices, while reducing costs from superfluous reagents, large sample volumes or bulky equipment for dispensing sheath fluids. The fabrication methods required to make acoustofluidic devices are straightforward and the procedures required for their operation are user-friendly. We hope these procedures will encourage the widespread development of similar devices to catalyze new areas of research for applications across materials science, biotechnology and medicine.

3.5 Acknowledgements

This chapter contains material from a previously published article: Shields IV,

CW; Cruz, DF; Ohiri, KA; Yellen, BB; López, GP. “Fabrication and operation of acoustofluidic devices supporting bulk acoustic standing waves for sheathless focusing of particles,” Journal of Visualized Experiments 2016. e53861 (109): 1-7. DOI: 10.3791/53861.

MyJove Corporation granted permission for reuse of this article in this dissertation.

This work was supported by the National Science Foundation (through grants

DMR-1121107, CMMI-1363483 and Graduate Research Fellowship Program (GRF-

1106401)) and the National Institutes of Health (R21GM111584).

108

4. Acoustofluidic Sequestration of Cells via Elastomeric Particles

The number of miniaturized medical devices for biosensing, cell sequestration, and sorting applications has soared in recent years due to their functional versatility, capacity to analyze small sample volumes, and promise for clinical translation [25, 244].

Acoustofluidics, which relies on the actuation of a piezoelectric transducer to excite a microfluidic device to resonance, has merited considerable attention as a convenient

means for cellular handling without imposing deleterious effects to cellular viability [39,

289, 290]. Through the action of acoustic radiation forces in laminar flows,

acoustofluidics can continuously direct cells into tightly focused streams, which

provides an attractive alternative to centrifugation (e.g., for removing lipids from blood)

[102] and hydrodynamic focusing for flow cytometric analysis [257, 291].

Previous acoustics-based microchip systems have employed resonant pressure waves to selectively focus (i.e., concentrate) and manipulate cell populations based on their morphological characteristics (e.g., size, compressibility, and density) using standing bulk acoustic waves and surface acoustic waves [50, 106]. In these systems, acoustic radiation forces drive cells with differences in such characteristics to the pressure nodes of standing waves at dissimilar rates, enabling continuous sorting at

multi-junction outlets, for example; however, no system has confined cells to the

pressure antinodes of an acoustic standing wave. This study describes the use of

elastomeric particles for recognition (binding), transport, and confinement of cells to the 109

pressure antinodes in a simple acoustofluidic system as an example of a new class of cell sample handling techniques.

Particles will focus to the nodes or antinodes of an acoustic standing wave, depending on their acoustic contrast factor (Equation (10)) [45, 292]. Particles with a positive φp migrate to the pressure node of a standing wave, whereas particles with a

negative φp migrate to the pressure antinodes [25]. Mammalian cells generally exhibit positive φp in aqueous fluids [291]; whereas elastomeric particles generally exhibit

negative φp due to their high compressibility and relatively low densities [92, 293].

For the half-wavelength resonant mode of a fluidic cavity, the pressure node and antinodes correspond to the centerline and the sidewalls of the channel, respectively. As depicted in Figure 33, elastomeric particles bound to cells can cause the cells to transport to the pressure antinodes. To achieve this phenomenon, the acoustic radiation forces acting on the elastomeric particle(s) must be greater those acting on the cell(s) to which they are bound [25]. The magnitude and direction of these forces are determined by the size, density, and compressibility of the components of the assembled complexes as well as the fluid properties [102]. This dependence is represented in Figure 33C, which indicates the maximum φp of an elastomeric particle capable of transporting a typical mammalian cell (13.0 µm in diameter) to a pressure antinode of an acoustic standing wave.

We modeled this relation by imposing the following inequality:

110

> | | (11)

�𝐹𝐹𝑝𝑝� 𝐹𝐹𝑐𝑐 such that,

> (12) 2 2 2 2 𝑉𝑉𝑝𝑝𝜋𝜋𝛽𝛽𝑓𝑓𝑃𝑃0 𝑉𝑉𝑐𝑐𝜋𝜋𝛽𝛽𝑓𝑓𝑃𝑃0 �� � 𝜑𝜑𝑝𝑝� �� � 𝜑𝜑𝑐𝑐� and, 𝜆𝜆 𝜆𝜆

> | | (13)

𝑉𝑉𝑝𝑝�𝜑𝜑𝑝𝑝� 𝑉𝑉𝑐𝑐 𝜑𝜑𝑐𝑐 where Vp and Vc are constrained to values greater than zero. The acoustic contrast factors

φp and φp are less than and greater than zero in this scenario, respectively. The average density of KG-1a cells is assumed from measurements of other mammalian cells [294] and the average size of KG-1a cells was determined by analysis using ImageJ software from bright field microscopy:

= 1.1 × 10 (14) 3 𝑘𝑘𝑘𝑘 𝜌𝜌𝑐𝑐 3 = 13 × 10 𝑚𝑚 (15) −6 𝐷𝐷𝑐𝑐 𝑚𝑚 We also assume:

= 1 (16) 𝛽𝛽𝑐𝑐 𝑓𝑓 𝛽𝛽 (17) = = 1.0 × 10 3 𝑘𝑘𝑘𝑘 𝜌𝜌𝑓𝑓 𝜌𝜌𝑝𝑝 3 that is, where the compressibility of the cell is assumed𝑚𝑚 to match the compressibility of water, and the density of the particle is sufficiently near to the density of water to assume their equality. We thus arrive at the final lower limit expression:

111

1.08 × 10 > (18) −16 𝑝𝑝 𝑉𝑉 𝑝𝑝 indicating the properties necessary of an elastomeric�𝜑𝜑 � particle to transport a typical mammalian cell to a pressure antinode in an acoustic standing wave. In practice, the elastomeric material bound to a cell may be comprised of multiple particles with the same effective volume. Thus, to controllably transport cells to pressure antinodes in acoustic standing waves, it is desirable to synthesize particles with controlled size, sufficiently low density, high compressibility and robust capacity for readily forming complexes with cells.

Figure 33: Schematic diagram illustrating acoustic confinement of cells in a rectangular acoustofluidic device (not to scale). A) Unbound cells and elastomeric

112

particles in an acoustic standing wave. B) Bound cell-particle immunocomplexes in an acoustic standing wave when the acoustic radiation force acting on the elastomeric particle is greater than that on the cell. C) The region under the solid line represents the locus of properties (effective (net) size and the acoustic contrast factor (φp)) of an elastomeric particle(s) that can displace a typical mammalian cell (13.0 μm diameter) toward a pressure antinode.

We previously reported the preparation of elastomeric particles using

homogenization and microfluidic techniques and demonstrated that they can be used to

transport target proteins and positive φp microbeads to the antinodes of an acoustic standing wave [92, 93]. Although convenient, homogenization techniques provide limited control over the size distribution and properties of the resulting particles. In contrast, microfluidic syntheses provide precise control over the dispersity in particle size, but suffer from limited yields. Herein, we present the preparation of a new class of elastomeric silicone particles for the precise spatial control of cells in acoustofluidic devices. We describe how their synthesis by nucleation and growth is easily scalable to generate large quantities of particles that enable immunolabeling for specific binding to cells. As a demonstration of their use, we show that elastomeric particles can confine mammalian cells to the antinodes of an acoustic standing wave, which may provide the basis for several, new, acoustics-based cell handling techniques.

4.1 Experimental

4.1.1 Fabrication of acoustofluidic device

Acoustofluidic chips were made from silicon and glass components using conventional microfabrication techniques: (i) photolithography and (ii) deep reactive-ion

113

etching to generate flow channels (≈250 µm deep), (iii) anodic bonding to adhere the patterned silicon chip to a Pyrex glass lid (Borofloat® 33, Glass B, Schott AG) and (iv) plasma treatment to bond polydimethylsiloxane (PDMS, Sylgard® 184, Dow Corning) ports for the inlet and outlet tubing connections. Inlets and outlets were designed on the backside of the chip to facilitate visual access to the channel by an upright microscope

(Zeiss Axio Imager 2, Carl Zeiss, AG) with a fluorescent lamp (X-cite series 120Q, Lumen

Dynamics). Tubing connections were installed into the bored holes of the PDMS ports and were interfaced with high purity silicone tubing (9628T54, McMaster-Carr Supply

Co.). A lead zirconate titanate (PZT) transducer (841, APC International; 2.93 MHz resonant frequency) was bonded underneath the flow channel using cyanoacrylate glue.

The PZT (0.5, 5 and 30 mm thickness, width and length, respectively) was actuated at ≈2.96 MHz, corresponding to the half-wavelength resonant mode of a channel 250 μm in width via a sinusoid waveform from a function generator (DG1022,

Rigol Technologies, Inc.) connected through a power amplifier (25A250AM6, Amplifier

Research, Corp.). An oscilloscope (TDS2004C, Tektronix, Inc.) measured the peak-to- peak voltage applied to the PZT, which was 31.0 V in all experiments.

4.1.2 Method for synthesis of elastomeric particles

Monomers (totaling 1 mL) (i.e., 1:100 molar ratio of tetramethyl orthosilicate

(TMOS; 98% purity, Sigma) to dimethoxydimethylsilane (DMODMS; ≥99.5% purity,

Sigma)) were added to 10 mL of 2.2x10-3 M HCl and were stirred at 500 rpm for 2-18 hr.

114

A molar excess of NH4OH (Sigma) was then added with continued stirring. Particles could be rendered fluorescent by adding 50 uL of 3.1 mM Nile red in acetone (Sigma) directly prior to the addition of NH4OH. Approximately 1x108 particles were functionalized by resuspending them in 1 mL 1xPBS, adding 25 µL of 33.3 µM fluorescent (Alexa-Fluor® 488 or 546-conjugated) streptavidin (SA), stirring at 100 rpm for 0.5 hr, and storing overnight at 4°C. Prior to use, particles were washed and resuspended in fresh 1xPBS.

4.1.3 Particle binding method

Approximately 1x108 elastomeric particles were washed (i.e., centrifuged at

2,000xg for 120 sec and resuspended in an equal volume of 1xPBS) and incubated with

25 µL of 33.3 µM streptavidin (SA) at 4°C for at least 12 hr to allow for physical adsorption. Prior to incubation with KG-1a cells, particles were centrifuged (2,000xg for

120 sec) and resuspended in fresh 1xPBS. Approximately 106 KG-1a cells were incubated with 25 µL of biotinylated antibodies (CD34 mouse anti-human mAb (clone 581),

Invitrogen) at 4°C for 0.5 hr and were subsequently washed twice (i.e., centrifuged at

400xg for 300 sec and resuspended in an equal volume of 1xPBS). The immunolabeled

KG-1a cells were then mixed with the SA-adsorbed elastomeric particles for 1 hr on an end-over-end rotator at ambient temperature for immunospecific binding (Figure 34).

115

Figure 34: Method for binding SA-adsorbed particles to biotinylated anti-CD34 labeled KG-1a cells. Elastomeric particles surface-functionalized with streptavidin specifically bind to cells presenting biotinylated antigens.

4.1.4 Cell preparation

Acute myelogenous leukemia (KG-1a) cells were cultured in Iscove’s Modified

Dulbecco’s Medium (Invitrogen) with 20% fetal bovine serum supplemented with 50

µg/mL streptomycin, 10 µg/mL penicillin, and 50 µg/mL fungizone (Gibco). Prior to analysis, cells were stained with calcein AM from a Live/Dead® cell viability kit

(Invitrogen). Cells (1x106) were incubated in cold wash buffer with 25 μL of stock biotinylated CD34 antibodies (Mouse Anti-Human mAb (clone 581), Invitrogen) for 0.5 hr. After washing, a concentrated mixture of SA-adsorbed elastomeric particles and biotinylated polystyrene microbeads or biotinylated anti-CD34 labeled KG-1a cells were gently agitated in an end-over-end rotator for 1 hr prior to injection to the acoustofluidic device.

116

4.2 Results and discussion

Below we describe the (i) methods for synthesizing biofunctional elastomeric particles as well as the results for confining (ii) polystyrene beads and (iii) mammalian cells to the pressure antinodes of a standing wave.

4.2.1 Biofunctional elastomeric particles

We prepared silicone gel particles using a simple nucleation and growth process whereby monomers were hydrolyzed in a low pH solution, thus forming nucleation sites for the rapid and uniform growth of particles upon catalysis [261]. Monomers were initially immiscible in water, but became miscible after hydrolysis. Excessive hydrolysis resulted in the formation of nonuniformly sized oligomers, and thus, polydisperse particles [262]. We thus controlled the duration of hydrolysis to achieve a narrow size distribution compared to that of particles prepared by homogenization techniques (18 vs. 51% CVs, respectively; n = 50 particles; Figure 35A) [93].

The reaction mixture contained two types of monomers: one that can form two siloxane linkages (i.e., DMODMS) and one that can form four siloxane linkages (i.e.,

TMOS). The large ratio of DMODMS to TMOS resulted in the formation of particles exhibiting elastic properties and displaying negative φp in water, as evidenced by their transport to the pressure antinodes of an acoustic standing wave (Figure 35B) [25]. A major advantage of this nucleation and growth synthesis is the ability to tune the size of particles by altering concentration of monomers used during the growth process or the

117

duration of the polycondensation reaction [92, 93]. Moreover, the nucleation and growth synthesis enables the facile incorporation of monomers with a variety of functional groups. These features are attractive compared to homogenization and microfluidic- based synthetic approaches, as particles with programmable properties (i.e., size, modulus, and surface chemistry) can be easily manufactured in large quantities for a variety of acoustofluidic applications.

Figure 35: Elastomeric particles obtained through nucleation and growth synthesis. A) Optical micrographs of elastomeric particles (dia. ≈13 µm) used for the confinement of cells. B) Elastomeric particles (red, with adsorbed SA labeled with Alexa-Fluor® 546) transported to the antinodes of an acoustic standing wave within a microfluidic channel (no flow). The locations of the channel walls (dashed lines) and the features of the acoustic standing wave (solid lines) are denoted.

4.2.2 Acoustic confinement of polystyrene microbeads to the pressure antinodes

For initial evaluation of the elastomeric particles, we studied their interaction with biotinylated polystyrene microbeads (Spherotech) as surrogates for cells due to their inherent positive φp behavior in physiological buffers and ample surface binding

sites [93]. We functionalized elastomeric particles by physically adsorbing SA to their

118

surfaces to permit binding to biotin groups on the polystyrene microbeads [92]. As controls, we performed acoustophoresis separately on the elastomeric particles and the polystyrene microbeads. The elastomeric particles exhibited negative φp behavior in the

acoustofluidic chip under flow by migrating to the pressure antinodes (Figure 36A). We

note that high flow rates or multinodal systems [257] with protective hydrodynamic sheath flow can help reduce the aggregation and buildup of negative φp particles as observed in Figure 35B. Polystyrene microbeads exhibited positive φp behavior by migrating to the pressure node (Figure 36B). After binding the SA-adsorbed elastomeric particles to the biotinylated polystyrene microbeads, the bound complexes displaced to the pressure antinodes of the acoustic standing wave (Figure 36C), indicating that elastomeric particles prepared by nucleation and growth and adsorbed with SA are capable of capturing positive φp objects and transporting them to the pressure antinodes.

119

Figure 36: Fluorescence intensity profiles of particles across the microchannel cavity under flow. The distributions of (A) SA-adsorbed elastomeric particles (dia. ≈13 µm) at 15 µL/min, (B) biotinylated polystyrene microbeads (diameter ≈5.2 µm) at 100 µL/min, and (C) complexes of particles in a bound to particles in b at 100 µL/min are shown across the acoustofluidic channel, where W denotes the width of the microchannel (250 µm).

4.2.3 Acoustic confinement of cells to the pressure antinodes

We evaluated the ability of elastomeric particles with adsorbed SA to capture

KG-1a cells presenting biotinylated antibodies against the CD34 cell surface antigen (see

Figure 34) and to transport those cells to the antinodes of an acoustic standing wave.

After co-incubation with gentle agitation, elastomeric particles bound to KG-1a cells

(Figure 37A-B). Once bound, the complexes were injected into an acoustofluidic chip

where they migrated to the pressure antinodes upon excitation of the PZT (Figure 37C-

D).

As a control, KG-1a cells without bound anti-CD34-biotin separated from elastomeric particles and focused along the pressure node (Figure 37D (left), Figure 37E).

Green stained KG-1a cells presenting biotinylated antibodies bound to SA-adsorbed, non-fluorescent elastomeric particles and separated from red stained (PKH26, Sigma)

KG-1a cells without anti-CD34-biotin (Figure 37F). At low flow rates (e.g., less than 50 120

µL/min), isolated complexes at the pressure antinodes generally remained immobilized against the walls of the microchannel, thus enabling the possibility of on-chip washing, staining, and secondary labeling of cells. At high flow rates (e.g., 200 µL/min in Figure

36C), shear forces cause the complexes to flow down the microchannel along the sidewalls, thus allowing their collection through a downstream microchannel trifurcation [102].

121

Figure 37: Acoustophoresis of KG-1a cells and elastomeric particles. A) Bright-field and (B) the corresponding fluorescent images of non-fluorescent, SA-adsorbed elastomeric particles (dia. ≈13 µm) bound to green stained KG-1a cells. C) Complexes of KG-1a cells and elastomeric particles migrating to the pressure antinodes in response to imposition of the acoustic standing wave (≈2 µL/min). D) Fluorescence intensity profiles across the channel width indicating the acoustophoresis of KG-1a cells unbound (left) and bound (right) to SA-adsorbed elastomeric particles in an acoustic standing wave. Fluorescent images within a microfluidic device of: (E) green stained KG-1a cells and Nile red embedded elastomeric particles without SA and focusing to the node and antinodes and node of an acoustic standing wave, respectively, and (F) non-fluorescent SA-adsorbed elastomeric particles bound to green, anti-CD34-biotin labeled KG-1a cells separated from red stained KG-1a cells without anti-CD34-biotin at the antinodes and nodes of an acoustic standing wave, respectively.

122

4.3 Conclusions

We describe the use of elastomeric silicone particles for the capture and transport of cells to the pressure antinodes of an acoustic standing wave. This approach provides:

(i) gentle forces on cells and particles [39], (ii) implementation in continuous laminar flow streams [106], (iii) the ability to tightly focus cells under a range of flow rates, even during flow reversal [291] and (iv) discriminating forces that act on both labeled and unlabeled cell populations. Looking forward, the combination of acoustic standing waves and biofunctional and elastomeric particles holds promise for sorting cells via acoustophoresis. However, in order to achieve a more robust binding between elastomeric particles and cells, we believe other approaches for biofunctionalizing elastomeric particles may be necessary. Isolating cells from complex biological fluids is critical to many diagnostic and therapeutic systems [2, 3]. We envision a miniaturized and continuous flow sorting system in which cells biospecifically immunolabeled with elastomeric particles would displace to, and flow along, pressure antinodes, separating them from unlabeled cells that would be focused to, and flow along, the pressure node.

In this approach, the separated populations of cells could be collected in a continuous fashion at high flow rates at a downstream channel trifurcation [102]. Such a method could provide advantages over systems such as fluorescence-activated cell sorting in that it would not require copious volumes of purified sheath fluid or serial cell-by-cell inspection (e.g., by fluorescence spectrophotometry).

123

4.4 Acknowledgements

This chapter contains material from a previously published article: Shields IV,

CW; Johnson, LM; Gao, L; López, GP. “Elastomeric negative acoustic contrast particles for capture, acoustophoretic transport, and confinement of cells in microfluidic systems,” Langmuir 2014. 30(14): 3923-3927. DOI: 10.1021/la404677w. The American

Chemical Society (ACS) granted permission for reuse of this article in this dissertation.

This work was supported by the National Science Foundation’s (NSF’s) Research

Triangle MRSEC (DMR-1121107), the NSF Graduate Research Fellowship Program

(1106401), the Hartwell Foundation Biomedical Research Fellowship Program and the

NSF EAGER program (CBET-1050176). We thank Dr. Catherine Reyes for her assistance with the writing process and the staff at Duke University’s Shared Materials

Instrumentation Facility for their guidance.

124

5. Magnetic Sorting and Magnetographic Templating

Accurate and high throughput cell sorting is a critical first step in isolating target cells for downstream analysis. The current gold standard for cell sorting, fluorescence- assisted cell sorting (FACS), is capable of multiplexed analyses and can sort up to 50,000

Hz (cells/sec) [11]. However, traditional FACS systems suffer from high costs and large footprints, which make routine clinical use non-ideal, especially for processing complex cell-containing fluids such as blood. Further, these highly advanced systems offer limited means of analyzing cellular, or subcellular, content. In this chapter, we present a miniaturized platform that offers high throughput sorting and detailed analysis of individual cells or particles in a multi-modular microfluidic device.

The first module uses acoustic standing waves to focus cells to the pressure node without the aid of sheath fluids. The second module then sorts magnetically labeled cells

(or magnetic particles) from nonmagnetic entities in a gradient magnetic field. We show this module can sort synthetic beads with purities above 90% (i.e., in both the target and waste outlets) at throughputs of up to 2,000 Hz. The third module contains a spatially periodic array of microwells with aligned micromagnets to capture individual cells for on-chip staining and analysis. We show this array is capable of capturing cells with accuracies exceeding 95%. Following the analysis of cells on-chip, reversing the polarity of the micromagnets allows cells to be ejected and recovered for further analysis.

125

5.1 Acoustically enhanced magnetic bead sorting

Previous magnetic-based microfluidic cell sorting instruments suffer from poor sensitivities and high losses of magnetic content due to the highly non-linear magnetic field distribution from the source dipole [80]. Consequently, magnetic particles and magnetically labeled cells along the distal wall of a microchannel (i.e., furthest from the magnetic field source) experience substantially lower magnetic moments compared to those near the proximal wall (i.e., closest to the magnetic field source). This effect manifests in one of two outcomes: (i) some magnetically labeled cells are not captured

(occurring in situations of weak fields, fast flow rates, etc.) or (ii) nearly all magnetically labeled cells are captured, but the field entrains unlabeled cells, thus reducing purity and exerting large, deleterious forces on isolated species (occurring in situations of strong fields, slow flow rates, etc.).

To overcome this limitation, we have developed a microfluidic instrument that acoustically pre-focuses particles (as surrogates for cells) for rapid, continuous and high throughput magnetic sorting. A key feature of this technology is the acoustic focusing, which enables high precision separations as particles traverse the magnetic field along the same location, thus exposing them to a more uniform magnetic torque (Figure 38)

[80]. In this design, once forces from an acoustic standing wave focus the particles, the channel broadens such that the width of the downstream channel does not resonate with the piezoelectric actuator and constrict particles to the pressure node. This enables

126

magnetic particles to freely migrate across the channel toward the magnetic outlet, and thus should result in higher output purities while maintaining high throughputs.

Figure 38: Acoustically enhanced magnetic sorting. A mixture of particles starts at the top and flows down towards the channel bifurcation. The yellow and red particles represent magnetic and nonmagnetic entities, respectively. The black lines and white arrow represent the acoustic standing wave and the magnetic field gradient, respectively.

5.1.2 Experimental

Using the methods described in Chapter 3, we fabricated an acoustofluidic chip possessing a microchannel with an upstream resonant portion to focus particles and a downstream non-resonant portion. The non-resonant portion was designed to permit the free flow magnetophoresis of magnetic entities toward the magnetic outlet without competing acoustic radiation forces. The resonant and non-resonant portions of the microchannel were 255 μm and 893 μm across, respectively. Consequently, following the relation c = λ*f, we applied a sinusoidal alternating current of ~50 V at a frequency of

2.90 MHz to the piezoelectric transducer with using a signal generator (DG1022, Rigol

127

Technologies, Inc.) in series with a power amplifier (25A250AM6, Amplifier Research,

Corp.).

To generate the gradient magnetic field, we placed a pair of neodymium magnets on either side of the microfluidic chip, directly adjacent to the deflection segment of the microfluidic channel, separated by about 1 mm (½” x ½” x ¼” magnet on the bottom and ½” x ¼” x ¼” on the top; K&J Magnetics, Inc.). Using the magnetic field calculator available from K&J Magnetics, we estimate this produced a magnetic field strength of

0.34 T in the center of the microchannel.

To validate the sorting performance of the instrument, we prepared a mixture of polystyrene microbeads as surrogates for cells. The mixture comprised a 1:1 ratio of yellow magnetic beads (8.43 μm; Spherotech, Inc.) to red non-magnetic beads (10.2 μm;

Spherotech) diluted in deionized water. We prepared five different concentrations of the bead mixture, i.e., 6, 60, 600, 6000 and 16000 beads/μL, corresponding to throughputs of

20, 200, 2000, 20000 and 55000 beads/sec with a constant flow rate of 200 μL/min. We gently agitated the solution throughout the course of the experiment by shaking the vial containing the particles. The sample was infused into the device via two syringe pumps

(NE-300, New Era Pump Systems, Inc.) connected directly to both outlets (80 μL/min withdrawn from the magnetic outlet and 120 μL/min withdrawn from the non-magnetic outlet). Following the completion of each experiment, we analyzed the results on a flow cytometer (Accuri C6; BD Biosciences, Co.).

128

5.1.3 Performance of acoustically enhanced magnetic bead sorter

We examined the performance (i.e., throughput and sensitivity) of the device with polystyrene beads as surrogates for cells (see Chapter 6 for separation results with cells from blood). First, we conducted control tests, i.e., with no acoustic or magnetic stimuli, to determine if any intrinsic bias was present within the instrument. We passed particles through the device at 200 μL/min to match the experimental conditions, and we maintained the concentration at 60 beads/μL. As expected, we found that there was no significant difference between distribution of magnetic beads and non-magnetic beads in the inlet or either outlet (p = 0.965; Figure 39).

Figure 39: Control experiments for the acoustically enhanced magnetic sorter. A mixture containing approximately a 1:1 ratio of magnetic to nonmagnetic beads was passed through the device without forces from an acoustic standing wave or magnetic field. Analysis from a paired t test shows no significant difference in the ratio of magnetic to nonmagnetic beads across the inlet or either outlet (p = 0.965; n = 5).

Next, we investigated the ability of the device to process various throughputs of microbeads by fixing the flow rate and adjusting the sample concentration. In these

129

experiments, we acoustically focused the particles into a single streamline such that each object was positioned the same distance from the field source, allowing for precise control over the force exerted on each magnetic entity. When the lead zirconate titante

(PZT) piezoelectric transducer was off, particles did not focus along a central streamline

(Figure 40A); however, when the PZT transducer was on, the particles tightly focused along the node of the acoustic standing wave and remained focused after entering the non-resonant portion of the channel (Figure 40B). As shown in Figure 40C and Figure

40D, the microchannel bifurcated asymmetrically such that the focused nonmagnetic entities exited out of the nonmagnetic outlet.

130

Figure 40: Acoustic focusing and magnetic sorting of polystyrene particles. Micrographs of polystyrene particles with the lead zirconate titante (PZT) transducer A) turned off and B) turned on. C) Schematic of the acoustically enhanced magnetic particle sorting chip (image not to scale). D) Micrograph of the channel bifurcation, where magnetic beads are shown flowing towards the magnetic outlet and nonmagnetic beads are shown flowing towards the nonmagnetic outlet. Green and red represent magnetic and nonmagnetic beads, respectively, in all images; the sinusoidal lines represent an acoustic standing wave. All scale bars are 250 μm.

Given its non-serial processing of particles and cells, this sorting instrument has the capacity to sort at high throughputs. To measure the sensitivity of the instrument as a function of throughput, we performed a series of experiments where we varied the concentration of beads flowing through the device (i.e., 6 to 16,000 beads/μL) while maintaining a constant flow rate (i.e., 200 μL/min), magnetic field strength (i.e., 0.34 T) and applied voltage to the PZT transducer (i.e., 50 Vpp) (Figure 41).

Using flow cytometry, we found that the instrument was capable of maintaining a sorting efficiency exceeding 90% in both outlets at throughputs up to 2,000 Hz (i.e., particles/sec; Figure 41A-C). The performance of the instrument between throughputs of 131

20 to 2,000 Hz was not substantially different, ranging from 91.4±4.8% to 96.0±0.8% magnetic beads in the magnetic outlet and 90.3±5.5% to 97.5±1.3% nonmagnetic beads in the nonmagnetic outlet. We also tested the throughput of the instrument at high throughputs to match the maximum throughput of commercial FACS devices (i.e.,

20,000 to 50,000 Hz) [11]. However, the purity in either outlet diminished at these high throughputs (average of both outlets was 78.7±3.6% for 20,000 Hz and 64.6±4.1% for

55,000 Hz; Figure 41D-E).

Figure 41: Performance of the acoustically enhanced magnetic sorter. The left and right columns represent the outputs from the magnetic and non-magnetic outlets, respectively. A mixture containing a 1:1 ratio of magnetic particles to non-magnetic 132

particles at increasing concentrations was passed through the device at A) 20, B) 200, C) 2,000, D) 20,000 and E) 55,000 particles/sec. Each condition (A-E) indicates the fluorescence intensity of the 533/30 nm laser versus the 585/40 nm laser from a flow cytometer (number of trials for each throughput (A-E), n = 5).

5.1.4 Conclusions

The results from these studies suggest that the acoustically enhanced magnetic sorting instrument is capable of accurate particle sorting (i.e., maintaining purities above

90% in both outlets) for throughputs up to 2,000 Hz. Further, we established the device functions well at moderately high flow rates, but we note that magnetically labeled cells may require slower flow rates or a stronger magnetic field as the magnetic content in those entities is lower. Overall, this system a new, promising approach for manipulating and separating microscopic entities, and it has direct implications for integration with previously developed techniques (e.g., MACS). Further, magnetic particles or magnetically labeled cells isolated from this approach can be directly infused into the magnetographic array for high precision capture, staining and analysis, as discussed in the following section.

5.2 Magnetographic array for the capture and enumeration of single cells and cell pairs

5.2.1 Introduction

The emergent need for point-of-care devices has spurred development of simplified platforms to organize cells across well-defined templates [295]. These devices employ passive microwells, immunospecific adhesive islands, and electric, optical, and

133

acoustic traps to manipulate cells [19, 20, 49, 94, 296]. In contrast, magnetic templating can control the spatial organization of cells through its ability to readily program ferromagnetic memory states [297]. While it has been applied to control the deposition of magnetic beads [298-303], it has not been used to direct the deposition of heterogeneous cell pairs, which may help provide critical insight into the function of single cells [5, 304]. As such, we developed a simple magnetographic device capable of arraying single cells and pairs of cells with high fidelity. We show this magnetic templating tool can use immunospecific magnetic labels for both the isolation of cells from blood and their organization into spatially defined wells.

We used standard photolithographic techniques to fabricate the microchips.

Briefly, an array of 10x30 µm cobalt micromagnets were patterned by a photolithographic liftoff process and overlaid with a pattern of dumbbell-shaped microwells formed in SU-8 photoresist (Figure 42A). The micromagnets were designed to produce a predominantly vertical field in the microwells by aligning the ends of the micromagnet at the center of each well of the dumbbell. These features were deposited across an area of ≈400 mm2 (>50,000 well pairs per microchip) (Figure 42B). Depending on the programmed magnetization state with respect to the external field, magnetic beads or cells were attracted to one pole and repelled by the other pole of each micromagnet, leading to a biased deposition (Figure 42C) [302].

134

Figure 42: Magnetographic array for single cell analysis. A) SEM image of the dumbbell-shaped well pairs for capturing magnetically labeled cells. B) Photograph of the finished device. C) An array of well pairs displaying a pitch of 60x120 µm before (top) and 10 min after the deposition of magnetic beads (bottom).

5.2.2 Experimental

5.2.2.1 Magnetographic microchip fabrication

The array of 10x30 µm micromagnets was created by spin coating NFR-016-D2

(JSR Micro, Inc.) on a 3” Si wafer (Addison Engineering, Inc.), exposing the wafer to 365

nm UV light through a quartz photomask (Photo Sciences, Inc.), and depositing 10 nm of

Cr, 100 nm of Co, and 100 nm of Au on the patterned side of the wafer using an electron- 135

beam metal evaporator (CHA Industries, Inc.). Wafers were then submersed in microposit remover 1165 (Shipley Co, Inc.) overnight at 25°C. Dumbbell-shaped well pairs were aligned over the micromagnets by spin coating and exposing SU-8 10

(MicroChem, Inc.) with a contact mask aligner (Karl Suss MA6/BA6, Suss MicroTec, AG) to a final thickness of 10 µm.

5.2.2.2 Sample loading onto the magnetographic microchip

Without developed integrated microfluidics, we studied three methods for depositing the fluid sample onto the microchip: (i) by depositing the droplet directly onto the surface, (ii) by following (i) and gently placing a coverslip over the droplet and

(iii) by fixing a coverslip to the surface of the microchip at an angle and depositing the droplet in the wedge (Figure 43A). We deposited 150 µL of a solution containing 2x104

particles/mL onto the microchip in a field of 200 G and collected images after 10 min for

each condition. We found that fluid samples that were deposited directly onto the

surface of the microchip yielded the highest capture efficiency (93.9±1.6%) (Figure 43B).

After each experiment, microchips could be reused by reversing the applied field to eject

beads for washing by a gentle stream of fluid.

136

Figure 43: Sample loading onto the magnetographic microchip. A) Samples were loaded onto the microchip (top) dropwise to the top, (middle) injected underneath a coverslip, and (bottom) injected into the crevice of a titled coverslip. B) The capture efficiency of the three modes of sample loading. The capture efficiency of sample loading without a coverslip were found to have a significantly higher efficiency (p < 0.001) (n = 5).

5.2.2.3 Positive selection of CD3+ T cells

Using a magnetic cell isolation kit (EasySepTM, STEMCELL Technologies), we isolated CD3+ cells from human peripheral blood mononuclear cells (PBMC) (Figure 44).

Figure 44: Positive magnetic isolation of lymphocytes against the CD3 antigen yields pure cell populations. A) 14.4% and B) 97.8% of cells in the non-target and target populations were CD3+, respectively.

137

5.2.3 Results and discussion

To demonstrate the capability of the array to capture cells into a format amenable for rapid image processing, we organized CD3+ lymphocytes using only hand-held permanent magnets. We isolated CD3+ lymphocytes from blood via positive selection using anti-CD3 magnetic nanoparticles (EasySepTM, STEMCELL Technologies) with purities confirmed by flow cytometry (97.8%). We then stained 1x106 CD3+ cells with anti-CD8 Alexa-488 and anti-CD4 Alexa-647 (5 µL of each antibody in 100 µL for 20 min;

BD Bioscience) to determine the CD4:CD8 ratio, a prognostic ratio for assessing the

immune system [305, 306].

Variably spaced neodymium magnets (0.5” x 0.5” x 1”; K&J Magnetics, Inc.) were fixed on either side of the microchip to generate a tunable magnetic field (0-400 G;

Figure 45A). Using this setup, fluorescently labeled cells were deposited, and the populations of CD4+ and CD8+ cells were indiscriminately arrayed, imaged, and enumerated using ImageJ. The resulting CD4:CD8 ratio of 1.84±0.18 (Figure 45B) was confirmed by flow cytometry with a high correlation (5.4% difference; Figure 45C), indicating the magnetographic microarray can pattern cells for the rapid and accurate assessment of critical phenotypical parameters without complex equipment (e.g., function generators or flow cytometers).

138

Figure 45: CD4:CD8 analysis of CD3+ lymphocytes. A) Photograph of the magnetographic device activated by permanent magnets (covered with green tape). The CD4:CD8 ratio determined by the (B) magnetographic microarray and (C-D) flow cytometry was 1.84 and 1.74, respectively.

More complex operations, such as the programmed deposition of cell pairs, can be achieved by leveraging the switchable, bistable magnetization of the micromagnets for the detailed studies of cell-cell interactions (Figure 46A-D) [302]. For these studies, a

200 G horizontal field generated from an electromagnetic coil was used to magnetize the micromagnets [279]. We then captured different concentrations of magnetic beads as surrogates for cells (8.4 µm polystyrene, Spherotech, Inc.) and found that higher bead concentrations did not affect the capture accuracy (>95%).

The opposite side of each micromagnet was then populated with the second

(yellow fluorescent) bead by reversing the direction of the applied magnetic field. We

139

tested several field strengths (i.e. 10, 25, 40, or 55 G) to optimize the conditions for isolating the desired bead in the opposite well without ejecting the first bead. If the field strength was too large, the previously deposited beads could be ejected from their wells due to the repulsive magnetic force overcoming gravity [302]. As shown in Figure 46E, increasing the field strength from 10 to 25 G significantly increased the capture accuracy at 60 min from the deposition of the second bead (p < 0.01), but increases from 25 to 55 G did not affect the capture accuracy (p > 0.10). As shown in Figure 46F, higher field strengths (i.e., 40 and 55 G) resulted in lower capture accuracies compared to lower field strengths (i.e., 10 and 25 G; p < 0.01), which was primarily due to ejection of the initially captured beads when the micromagnets reversed their polarity.

We then arranged pairs of membrane dyed (calcein AM, Invitrogen; PKH26,

Sigma) magnetically labeled CD3+ lymphocytes. First, red stained cells (150 µL of 2x104

cells/mL) were deposited on the microchip in the presence of 250 G vertical magnetic

field. After 20 min, the field was reversed (i.e., to 40, 55 and 70 G) and green stained cells

(150 µL of 2x104 cells/mL) were deposited on the microchip with images taken in 10 min intervals. Fluorescence images were overlaid (Figure 46C) and the capture accuracy of cell pairs was determined (ImageJ).

As seen in Figure 46G, the capture accuracy of pairs of CD3+ lymphocytes was lower than that of magnetic beads (Figure 46E). However, as shown in Figure 46H, the second set of cells (green fluorescent) exhibited an average capture accuracy of

140

91.8±1.9%. This indicates the lower capture accuracy of cell pairs was either due to the ejection of initially captured (red fluorescent) cells or the migration of initially captured cells through the connecting channel, resulting from their relatively high deformability compared to magnetic beads.

Figure 46: Programmed pairing of magnetic beads and CD3+ lymphocytes. A) Schematic of the magnetographic cell pair isolations. B) Polarized micromagnets isolate cells of one type to one side in a vertical magnetic field and then cells of a second type to the other side when the field is reversed. C) Fluorescent image of magnetically trapped green stained (top) and red stained (bottom) cell pairs. D) SEM image of magnetically labeled cells in the microwells. E) Capture accuracy of magnetic bead pairs (each color represents the field strength of the reversed field). F) Change in the capture accuracy (loss) of initially captured beads after reversing the magnetic field. The capture accuracy of (G) magnetically labeled cell pairs and (H) the second magnetically labeled cell (for (E-H): n = 5; time starts from the deposition of the second set of cells or beads).

5.2.4 Conclusions

In summary, we developed a simple device capable of organizing magnetic particles, cells, and pairs of cells into well-defined compartments. A major advantage of

141

this system is the use of specific magnetic labels to both isolate cells and program their deposition. While the design of this device does not enable dynamic control of the spacing between captured cell pairs as does some dielectrophoresis-based devices [307], it can easily capture cells with high fidelity using only permanent magnets and has clinical relevance in the assessment of immune parameters (i.e., the CD4:CD8 ratio).

These demonstrations potentiate a relatively simple and robust device where highly organized spatial arrangement of cells facilitates rapid and accurate analyses towards a functional and low-cost point-of-care device.

5.3 Acknowledgements

This chapter contains material from two articles. The full citation for the first article is: Shields IV, CW; Wang, J; Ohiri, KA; Essoyan, E; Yellen, BB; Armstrong, AJ;

López, GP. “Magnetic separation of acoustically focused cancer cells from blood for magnetographic templating and cellular analysis,” (In preparation). The full citation for the second article is: Shields IV, CW; Livingston, CE; Yellen, BB; López, GP; Murdoch,

DM. “Magnetographic array for the capture and enumeration of single cells and cell pairs,” Biomicrofluidics 2014. 8(4): 041101. DOI: 10.1063/1.4885840. AIP Publishing, LLC granted permission for reuse of this article in this dissertation.

This work was supported by the National Science Foundation’s (NSF’s) Research

Triangle MRSEC (DMR-1121107) and the NSF Graduate Research Fellowship Program

(1106401). We thank Marcia Bentz for her assistance for the preparation of cells,

142

Jonathan Liu for his assistance in the fabrication of the microchips, and the staff at

Duke’s Shared Materials Instrumentation Facility for helpful discussions. We also thank the Duke Center for AIDS Research for assistance in cell isolations and flow cytometry experiments.

143

6. Magnetic Separation of Acoustically Focused Cancer Cells from Blood for Magnetographic Templating and Cellular Analysis

Liquid biopsies hold enormous promise for the next generation of medical diagnoses. At the forefront of this effort, many are seeking to capture, enumerate and analyze circulating tumor cells (CTCs) as a means to prognosticate and develop individualized treatments for cancer. Capturing these rare cells, however, represents a major engineering challenge due to their low abundance, morphology and heterogeneity. A variety of microfluidic tools have been developed to isolate CTCs from drawn blood samples; however, few of these approaches offer a means to separate and analyze cells in an integrated system. We have developed a microfluidic platform comprised of three modules that offers high throughput separation of cancer cells from blood and on-chip organization of those cells for streamlined analyses. The first module uses an acoustic standing wave to rapidly align cells in a contact-free manner. The second module then separates magnetically labeled cells from unlabeled cells, offering purities exceeding 85% for cells and 90% for binary mixtures of synthetic particles.

Finally, the third module contains a spatially periodic array of microwells with underlying micromagnets to capture individual cells for on-chip analyses (e.g., staining, imaging and quantification). This array is capable of capturing with accuracies exceeding 80% for magnetically labeled cells and 95% for magnetic particles. Overall, by virtue of its holistic processing complex biological samples, we believe this system has

144

promise for the isolation and evaluation of rare cancer cells and can be readily extended

to address a variety of applications across single cell biology and immunology.

6.1 Introduction

The isolation and examination of circulating tumor cells (CTCs) represents a critical goal in cancer medicine, as these compartmentalized biomarkers may aid our understanding of how cancer spreads and provide a direct means to profile cancer cells over time during therapy as a marker of response or resistance [308, 309]. As such, capturing CTCs from drawn blood (in so-called liquid biopsies) could eliminate the need for invasive diagnostic procedures, while yielding the same, or similar, information that could shed light onto the regulatory pathways of native tumor sites [310]. At the most basic level, the enumeration of CTCs has been shown to provide direct prognostic value in the metastatic setting for some cancers such as prostate, breast and colorectal [311,

312]. Ensuing studies have shown that post-isolation CTC biomarker analyses (e.g., immunofluorescence, fluorescence in situ hybridization (FISH) and reverse transcription quantitative polymerase chain reaction (RT-qPCR)) can provide useful phenotypic, genetic and regulatory information [313]. More recent studies have demonstrated the ability to create viable cultures from CTCs, thus potentially enabling ex vivo drug testing that could give rise to new medical insights and personalized medicine by guiding doctors towards administering patient-specific therapeutic interventions [314].

145

To these ends, enormous interest has been generated to develop technologies that isolate CTCs from whole blood both rapidly and with high purity. The leading technology for this is CellSearch by Janssen Diagnostics (a Johnson & Johnson

Company), which is the only FDA approved assay for CTC enumeration as a prognostic test for certain types of cancer. CellSearch works by lysing erythrocytes from 7.5 mL of drawn blood, magnetically separating CTC candidates by EpCAM, and staining cells with anti-CD45, anti-cytokeratin, and Hoechst DNA stain. This type of analysis is useful, for example, as recent work suggests that CTC enumeration following therapy may provide useful prognostic information in men with castration-resistant prostate cancer

[315]. However, despite its potential, many doctors are reluctant to order such tests due to their unclear clinical utility and questionable ability to directly inform upon clinical decision-making, which has resulted in a lack of insurance coverage and thus levies cost directly onto patients [316]. However, methods that preserve CTC integrity and viability for predictive biomarker studies in cancer may be able to overcome such issues of clinical utility.

As we highlight in Chapter 1, a variety of microfluidic tools have been developed to overcome these issues [274]. Microfluidic devices can sort cells by active approaches with forces from acoustics [110, 272], magnetics [283] and electrokinetics [20], as examples, as well as by passive approaches like cellular immobilization [220], deterministic lateral displacement [317] and hydrodynamic filtration [201]. While

146

options are diverse, each microfluidic strategy possesses certain advantages and disadvantages. Several persistent challenges have made it difficult for these tools to isolate CTCs from whole blood in a rapid, gentle and precise manner, including [318]:

(i) the low abundance of CTCs (e.g., as low as 1-10 cells per 7.5 mL of whole blood, which comprises tens of billions of other cells) [316], (ii) the highly specific, and sometimes pliable, definition of a CTC (e.g., it expresses cytokeratin 8, 18, and/or 19, does not express CD45, has a nucleus and is larger than 5 μm) [319], (iii) the lack of a universal surface marker [316] and (iv) the epithelial-mesenchymal transition (EMT) of

CTCs during extravasation, which involves the down regulation of common anchoring surface markers (e.g., epithelial cell adhesion molecule (EpCAM)) or loss of an epithelial phenotype (e.g. loss of cytokeratin or tumor specific markers like prostate specific antigen (PSA)) [2].

These challenges necessitate a versatile tool that offers high throughput potential for processing relevant volumes (e.g., 7.5 mL) of blood. While many microfluidic tools have been developed [5], most still suffer from low throughputs (e.g., <1,000 cells/sec),[135] low purities (e.g., <85% purities in the target outlet) [320] and/or the inability to analyze isolated CTC candidates on-chip for post-separation analysis [79, 81].

One microfluidic technology that stands above the others is the CTC-iChip developed by

Toner and coworkers, which offers the ability to separate CTCs via positive or negative selection [89]. This tool has been shown to allow for the isolation of cells in a high

147

throughput fashion, with sensitivities comparable to (or exceeding) CellSearch. While exemplary, this technology is still limited in that it does not organize cells after sorting for post-separation staining, quantification and automated analysis. Certain cell types, such as cellular clusters or small CTCs of similar sizes to red blood cells, may be missed with this approach as well [321]. This shortcoming, which is shared by many other microfluidic technologies, greatly slows the screening process and may lead to more frequent errors, especially as post-separation analyses are typically performed by technicians, and thus reliant upon the accuracy of human examination.

As a potential means to overcome these limitations, we have developed a microfluidic platform designed to separate cancer cells from blood and organize them across a spatially defined array for staining and analysis (Figure 47). This system is comprised of three modules. The first module is designed to align cells using forces from an acoustic standing wave. The second module exploits a magnetic field gradient to separate magnetically labeled cells from unlabeled cells. The combination of these two modules allows the initial positions of the cells to be tightly controlled prior to magnetic sorting, leading to more repeatable sorting conditions [322]. The third module then organizes magnetically labeled cells across a spatially defined magnetographic array for on-chip staining and analysis.

148

Figure 47: Schematic of the microfluidic system designed to focus, separate and template cancer cells. Diluted blood (sans erythrocytes) moves through the device, from left to right, whereupon cells are acoustically focused to the center of the microchannel. The black dotted lines represent a half wavelength acoustic standing wave. Magnetically labeled cells are then deflected in the non-resonant portion of the microchannel via a gradient magnetic field, indicated by the fading black arrow. Finally, an array of micromagnets, indicated by the blue rectangles, locally attracts magnetically labeled cells into microwells, indicated by the red boxes, for on-chip staining and analysis. The modules for cell sorting and cell templating were made and operated separately. (N.B., schematic is not to scale).

6.2 Experimental

6.2.1 Fabrication and operation of the sorting module

To fabricate the sorting module, we used methods described previously [323].

Briefly, a photomask was designed in AutoCAD (Autodesk, Inc.) and sent to a qualified printer (e.g., CAD/Art Services, Inc.). A layer of positive photoresist (AZ 9260;

MicroChemicals, GmbH) was spin coated onto a 6” Si wafer (Addison Engineering,

Inc.), and then exposed to UV irradiation from a mask aligner (365 nm wavelength;

MA6/BA6, Karl Süss KG) to define the microchannel. The wafer was then etched with

149

deep reactive-ion etching (Pegasus deep silicon etcher; SPTS Technologies, Ltd.) and then anodically fused to a precut borosilicate glass “lid” (Borofloat® 33, Glass B; Schott

AG) to form an enclosed microchannel. We bonded a lead zirconate titanate (PZT) transducer (30 x 5 mm, resonant frequency, f0, = 2.93 MHz; APC International, Ltd.) underneath the microchannel via cyanoacrylate glue (Loctite® 495; Loctite Corp.).

Polydimethylsiloxane (PDMS, Sylgard® 184; Dow Corning Corp.) was used to form the inlet and outlet ports, which were bonded to the substrate with epoxy.

The sorting module contained an acoustic focusing module in series with a magnetic separation module. The width of the resonant portion of the microchannel was

255 μm (see Figure 48A for dimensions), corresponding to a half-wavelength resonant frequency of 2.90 MHz (following the relation c = λ * f; where c, λ and f represent the speed of sound, acoustic wavelength and applied frequency, respectively). The width of the microchannel increased by 3.5 times in the magnetic separation region (i.e., to 893

μm) to avoid local resonances that would compete with the magnetophoretic separation of magnetically labeled cells. We used a signal generator (DG1022; Rigol Technologies,

Inc.) to create a sinusoidal signal with peak-to-peak voltages (Vpp) ranging from 2.0 to 3.5

V, which we amplified by a power amplifier (25A250AM6; Amplifier Research, Corp.) to

40 to 70 V, as measured and confirmed by an oscilloscope (TDS2004C; Tektronix, Inc.).

Mixtures of polystyrene particles, and later cells, were prepared by the methods described in Sections 6.2.3 and 6.2.4. These samples were infused into the device using

150

the “withdraw” mode on two syringe pumps (NE-300; New Era Pump Systems, Inc.), connected to each of the outlets. Samples were agitated throughout the course of the experiment to avoid changes in concentration over time. For studies with particles, the syringe pumps withdrew fluid at a constant rate of 200 μL/min (i.e., 80 μL/min through

the magnetic outlet and 120 μL/min through the non-magnetic outlet), with the PZT

transducer driven at 40 Vpp. For studies with cells, a constant flow rate of 25 or 50

μL/min (i.e., 7.5 or 15 μL/min through the magnetic outlet and 17.5 or 35 μL/min

through the non-magnetic outlet) was used with the PZT transducer driven at 70 Vpp.

Particle and cell trajectories through the microchannel were observed with an upright microscope (Axio Imager 2; Carl Zeiss, AG).

6.2.2 Fabrication and operation of the templating module

To fabricate the templating module, we used methods described previously [88].

Briefly, a layer of negative photoresist (NFR-016D2; MicroChemicals, GmbH) was spin coated on a 3” soda lime glass wafer (University Wafer, Inc.) and exposed to UV

irradiation from a mask aligner (365 nm wavelength; MA6/BA6, Karl Süss) to define an

array of 10 x 30 μm micromagnets using UV irradiation from a mask aligner (MA6/BA6;

Karl Süss). Each array had a pitch of 75 μm in the planar directions. Next, 10 nm Cr, 200

nm Co, 10 nm Cr and 50 nm Au were deposited on the photopatterned wafer using an

electron beam metal evaporator (Solution E-Beam; CHA Industries, Inc.). The wafer was

then soaked in 1165 photoresist remover (MicroChem Corp.) overnight at room

151

temperature. Finally, a matching array of 20 μm square “microwells” formed in SU-8 10

(MicroChem) was defined over the micromagnets using a photomask aligner

(MA6/BA6, Karl Süss) such that the center of each microwell was aligned at the same end of each micromagnet (e.g., Figure 47).

After cutting, the glass substrate containing the aligned array of micromagnets and microwells was sealed with a 600 μm tall perfusion chamber (3 x 32 mm

CoverWellTM; Grace Bio-Labs, Inc.). Inlet and outlet ports were created by pressing

adhesive tubing connectors (press fit; Grace Bio-Labs) over top of the inlet and outlet

holes of the perfusion chamber. Prior to injecting the sample into the magnetographic

module, the micromagnets were polarized in the direction of the microwells by exposing

the substrate to a neodymium magnet in plane (1” x ½” x ½”; K&J Magnetics, Inc.), producing a local field strength > 0.3 T. The magnetographic module was then placed in a uniform magnetic field produced from two neodymium magnets (4” x ½” x ¼”; K&J

Magnetics), separated by 3.5”, producing a field strength of 110 G at the center of the two magnets where the device was placed. Depending on the programmed magnetization state with respect to the external field, magnetic particles or magnetically labeled cells were attracted to one pole and repelled by the other pole of each micromagnet, leading to a biased deposition. The initial polarization of the micromagnets was configured such that the field produced near the center of each microwell was attractive.

152

The inlet ports were then connected with silicone tubing (0.6 mm I.D.; Cole

Parmer Instrument, Co.) in series with a syringe pump (NE-300; New Era Pump

Systems). Samples were injected at a low flow rate (e.g., 50 μL/min). Once injected, the flow was arrested for ≈5 min to allow the magnetic particles or magnetically labeled cells to settle and to be captured by the micromagnets. Magnetic particles were analyzed immediately after capture. Magnetically labeled prostate cancer cells were incubated on- chip with green calcein AM (2 μL dye with 158 μL DMSO, diluted 50-fold in PBS;

Thermo Fisher) for 20 min, and were washed by flowing PBS through the perfusion chamber, again at a low flow rate. Cells were analyzed on-chip with an upright optical microscope (Axio Imager 2).

6.2.3 Preparation of microparticle samples

To validate the sorting device, we prepared a mixture of synthetic particles as surrogates for cells comprising a 1:1 ratio of magnetic particles to non-magnetic particles

(8.4 μm yellow polystyrene impregnated with iron oxide and 10.2 μm red polystyrene, respectively; Spherotech, Inc.) diluted in deionized water. We prepared five different concentrations of this mixture, 6, 60, 600, 6,000 and 16,000 particles/μL, which corresponded to throughputs of 20, 200, 2000, 20,000 and approximately 55,000 particles/sec under a constant flow rate of 200 μL/min, as described by the procedures in

Section 6.2.1. Following the completion of each experiment, results were analyzed via a flow cytometer (Accuri C6; BD Biosciences, Co.).

153

6.2.4 Preparation of cell samples

Adherent human prostate cancer (LNCaP) cells were cultured in polystyrene flasks (75 cm2, CellStar®; Greiner Bio-One GmbH) in RPMI culture medium (1640 (1x),

Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum, 5 mL sodium pyruvate (100 mM; Gibco), 5 mL HEPES (1.0 M; Gibco) and 2.8 mL glucose (450 g D-(+)-glucose per liter; Sigma Aldrich, Co.). To release cells, the surface of the flask was rinsed with PBS (1x, without CaCl2 and MgCl2) and incubated in 7 mL of

HyQ®TaseTM cell detachment solution (HyClone; GE Healthcare) for 5 min followed by moderate agitation. Cells were collected and washed twice (i.e., centrifuged 250xg for 5 min and resuspended in an equal volume of PBS).

To prepare the magnetic labels, a 50 μL aliquot of streptavidin (SA)-labeled stock magnetic beads (MyOneTM Dynabeads®, SA T1; Thermo Fisher) was diluted to 30 mL with PBS and was washed twice (i.e., by trapping beads along the side of an Eppendorf tube with a strong, neodymium magnet for 1 min, decanting the supernatant with the magnet still attached and resuspending the beads in PBS). Next, a 20 μL aliquot of

CD326 (EpCAM)-biotin (Miltenyi biotec GmbH) was added directly to the solution under gentle agitation on an end-over-end rotator for 30 min at 4°C. The suspension was then washed using the conditions described above and resuspended in 50 μL PBS.

Cell mixtures were prepared by spiking LNCaP cells, which served as surrogates for CTCs, into whole blood. Erythrocytes were lysed by diluting 6.0 mL of whole

154

porcine blood (BioreclamationIVT, LLC) 10-fold with ACK lysis buffer (Thermo

Scientific) and incubating the solution at room temperature for 5 min. The remaining cells were concentrated by centrifuging at 300xg for 5 min and re-suspending the pellet in 6.0 mL PBS. The solution containing LNCaP cells was added to the lysed blood to yield a ratio of 150:1 leukocytes to LNCaP cells. The magnetic bead mixture was then added to the solution under gentle agitation on an end-over-end rotator for 30 min prior to injection into the microfluidic device.

6.2.5 Model of magnetophoretic cell deflection

We developed a simple model to assess the sensitivity of our device to separate magnetically labeled cells from unlabeled cells in slow flow regimes, i.e., where viscous forces dominate. To determine the conditions necessary to separate cells, we calculated the residence time of particles flowing through the device. Using an optical profilometer

(Zygo NewView 5000 Optical Surface Profilometer; Zygo Corp.), we found the microchannel was 204 µm deep. At the maximum flow rates used in this study, which range from 25 to 200 µL/min, the fluid velocity ranged from ≈2.3 to 18.3 mm/sec, respectively. Given the length of the deflection portion of the microchannel, LNR (10.92 mm), this yields a corresponding range of residence times, τr, of 4.7 to 0.6 sec. Further, we note the microchannel bifurcates at a critical distance, CD, of 392 µm from the centerline of the microchannel in the y-direction (see Figure 48A for directionality); therefore, magnetically labeled cells must travel a distance, D, ≥ CD within a time of τr to

155

separate from unlabeled cells. N.B., in designing the sorting module, we chose a relatively large CD so as to increase the purity of sorted samples by reducing the likelihood of unlabeled cells exiting the magnetic outlet.

Using an online magnetic field calculator from K&J Magnetics, Inc.

(www.kjmagnetics.com/calculator.asp), we estimated the magnetic field strength produced by a pair of neodymium magnets (½ x ¼ x ¼” and ½ x ½ x ¼”) to be ≈0.34 T.

The magnets were placed on the top and bottom of the microfluidic chip, separated by

≈1 mm from the deflection portion of the microchannel in an “end-to-end” configuration such that the north end of one magnet was facing the south end of the other magnet as to cumulate their individual magnetic field strengths. Using a spline fit curve in

MATLAB (MathWorks, Inc.), we estimated that the magnets produced a field gradient of 336 T/m within the microchannel, and we estimated the force imposed on each magnetically labeled cell using the following relation (see Table 8 for a list of model parameters and their corresponding values):

= (19) 𝜕𝜕𝐻𝐻𝑥𝑥 𝐹𝐹𝑚𝑚𝑚𝑚𝑚𝑚 𝑁𝑁𝑏𝑏𝜇𝜇0𝑉𝑉𝑉𝑉 where μ0 is the magnetic permeability of free space𝜕𝜕𝜕𝜕, V is the volume of a magnetic particle and Ms is the saturation magnetization of a single magnetic bead, with Nb being the number of beads attached to a cell. The fields experienced by the beads were genereally greater than 0.01 T, which makes the fixed bead magnetization a realistic assumption [89]. The field gradient was also assumed to be constant, which allows for a

156

straightforward estimation of separation time without requiring a detailed trajectory analysis.

Table 8: Model parameters used for the analysis shown in Figure 48C-D.

Variable Value

μ0 4π x 10-7 N/A2

V 5.315 x 10-19 m3

Ms 18,000 A/m

η 1 x 10-3 (N·s)/m

Reff 9.015 x 10-6 m

2.67 x 108 A/m2 𝝏𝝏𝑯𝑯𝒙𝒙 𝝏𝝏𝝏𝝏

Using an upright microscope (Zeiss Axio Imager 2) and imaging software

(ImageJ), we determined the average cell diameter was 15.6±2.8 μm (Figure 48B (top)), which corroborates with previous reports for LNCaP cells [324]. Next, we found the average number of magnetic beads conjugated with anti-EpCAM bound to an LNCaP cell, Nb, to be 29.9±12.1 beads/cell (excluding cells without magnetic beads, which

represented ≈15% of the population and is proportionate with the number of CTCs in

blood with a reduced expression of EpCAM [325]; Figure 48B (bottom)). Using these

values, we calculated the trajectory of magnetically labeled cells towards the magnetic

outlet by balancing Fmag with the force from Stokes drag, Fdrag. To simplify the expression,

157

the horizontal flow profile of the velocity was assumed to be flat (i.e., Hagen-Poiseuille flow conditions were ignored) [89]:

= = = 6 (20) 𝜕𝜕𝐻𝐻𝑥𝑥 𝐹𝐹𝑚𝑚𝑚𝑚𝑚𝑚 𝑁𝑁𝑏𝑏𝜇𝜇0𝑉𝑉𝑉𝑉 𝐹𝐹𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑 𝜋𝜋𝜋𝜋𝑟𝑟𝑒𝑒𝑒𝑒𝑒𝑒𝑢𝑢𝑥𝑥 where η is the fluid viscosity, reff is the 𝜕𝜕effective𝜕𝜕 radius of the cell complex bound to magnetic beads and ux is the speed of deflection from magnetic exertion (i.e., not due to fluid flow or other effects), which is normal to the direction of flow. We note that this simplified model is used to obtain an approximate measure of the separation time, which ignores edge effects, Brownian motion, non-constant fluid velocity profiles and variation in the magnetic field landscape.

Figure 48: Model of magnetophoretic cell deflection assuming a uniform flow velocity profile and a constant magnetic field gradient. A) Scaled illustration of the 158

microfluidic system in which a streamline of magnetically labeled cells (green) separates from a streamline of unlabeled cells (red) in a gradient magnetic field (shaded black arrows). Distances for WR, WNR, CD and LNR are 255, 893, 392 and 10,924 μm, respectively. B) Histograms of the numbers of magnetic beads (Nb) bound to LNCaP cells (top; n = 70) and diameters of LNCaP cells (bottom; n = 151). C) Calculated trajectories of LNCaP cells bound to different numbers of magnetic beads, ranging from 0 to 30, at a flow rate of 100 μL/min through the deflection portion of the microchannel (the thick orange line indicates the critical number of beads required for separation (i.e., where D ≥ CD)). D) Calculated trajectories of LNCaP cells at a constant flow rate of 50 μL/min (orange) and 200 μL/min (green) through the deflection portion of the microchannel with Nb = 15±5 beads/cell.

At a flow rate of 100 μL/min, our simplified analysis indicates LNCaP cells with

Nb ≥ 15 beads/cell will separate from unlabeled cells and exit the magnetic outlet (Figure

48C). According to the distribution of magnetic beads per LNCaP cell (Nb; Figure 48B

(top)), this would correspond to ≈94% of magnetically labeled cells deflecting to the magnetic outlet. When operating at faster flow rates (e.g., 200 μL/min; see green lines in

Figure 48D), cells with Nb as high as 20 beads/cell would not separate and exit the magnetic outlet, under the assumptions employed. However, at slower flow rates (e.g.,

50 μL/min; see orange lines in Figure 48D), cells with Nb as few as 10 beads/cell would separate and exit the magnetic outlet. Based on this analysis, we operated the device at

25 to 50 μL/min when isolating magnetically labeled cells, but at 200 μL/min when separating magnetic particles from non-magnetic particles. This higher flow rate was selected due to the higher magnetic moment density of magnetic polystyrene particles compared to magnetically labeled cells, where moment scales linearly with the number of magnetic beads on the cell [326].

159

6.3 Results and discussion

6.3.1 Acoustically enhanced magnetic bead separation

Acoustic radiation forces are now routinely used to focus [285], concentrate [327], assemble [328] and sort [104, 272] particles and cells. The advantages of acoustics include its label-free interactions with a broad range of materials (e.g., in contrast to magnetics, which requires magnetically susceptible materials), gentle handling, rapid processing and scalability across multiple harmonics [40]. Primary acoustic radiation forces can be described by:

= , sin(2 ) (21) 22 , , 𝜋𝜋𝑃𝑃0 𝑉𝑉p𝛽𝛽m 𝐹𝐹 � � ∙ 𝜙𝜙�𝛽𝛽𝑚𝑚 𝑝𝑝 𝜌𝜌𝑚𝑚 𝑝𝑝� ∙ 𝑘𝑘𝑘𝑘 where P0, k and Vp represent the𝜆𝜆 acoustic pressure amplitude, wave number and particle volume, respectively [40]. The direction of this force is determined by the acoustic contrast factor:

5 2 = (22) 2 + 𝜌𝜌𝑝𝑝 − 𝜌𝜌𝑚𝑚 𝛽𝛽𝑝𝑝 𝜙𝜙 𝑝𝑝 𝑚𝑚 − 𝑚𝑚 whereby a positive contrast factor indicates𝜌𝜌 𝜌𝜌 a particle𝛽𝛽 (or cell) will migrate towards the

pressure node and a negative factor indicates a particle will migrate towards an

antinode [49, 94]. Importantly, the sign of this acoustic contrast factor depends on the density, ρ, and the compressibility, β, of the particle relative to the fluid medium, which are represented by the subscripts p and m, respectively. Due to their density, cells

160

typically possess a positive acoustic contrast factor in physiological buffers, and thus focus to the pressure node(s).

In this study, we use acoustic radiation forces to set the initial conditions of the

particles in the flow stream at the center of the microfluidic channel (Figure 49A)

without the assistance of sheath buffers or irregularly shaped microchannels [155]. In the

absence of an acoustic standing wave, fluorescent particles (which we initially use as

surrogates for cells) occupied the width of the microchannel (full width at half

maximum (FWHM) = 253 μm; Figure 49B (top); Figure 49C (top)). When the

piezoelectric transducer was excited at a frequency corresponding to a half-wavelength

acoustic standing wave in the resonant portion of the microchannel, particles focused

into a single streamline (FWHM = 22 μm; Figure 49B (middle); Figure 49C (left)) and

remained focused in the deflection portion of the microchannel (FWHM = 55 μm; Figure

49B (bottom); Figure 49C (right)). This ability to retain a focused streamline of particles

is critical for high-purity separation, as non-magnetic entities can thus exit the device

solely through the non-magnetic outlet (i.e., due to the off-center bifurcation of the

microchannel; see scale illustration in Figure 48A).

161

Figure 49: Acoustically enhanced magnetic bead sorting. A) Photograph of the device used to focus and separate particles and cells. Wires leading to a piezoelectric transducer and tubing leading to the microchannel protrude from the back. One of two separation magnets is shown adjacent to the deflection portion microchannel. B) Relative intensity plots display the full width at half maximum (FWHM) of particles across the microchannel at different positions and conditions: B1 and B2 represent the resonant portion of the microchannel with the acoustics off and on, respectively; B3 represents the deflection portion of the microchannel with the acoustics on (f = 2.90 MHz; Vpp = 40 V). C) Corresponding micrographs of fluorescent particles in the device with the acoustics off (top) and on (bottom). D) Performance of the device for separating a 1:1 ratio of magnetic particles to non-magnetic particles at throughputs ranging from 20 to 55,000 particles/sec by altering the volume fraction of particles (displayed along the top of the plots in D), at a flow rate of 200 μL/min (n = 5).

To evaluate the performance of the sorting module, we prepared different concentrations of synthetic particles (i.e., ranging from 6 to 16,588 particles/μL) to flow through the device at a constant flow rate of 200 μL/min, thus corresponding to

162

throughputs of 20 to 55,000 particles/sec (Figure 49D). We found that the device yielded a high purity (i.e., >92%) in both outlets after sorting at throughputs of up to 2,000 particles/sec, which is equivalent to (and sometimes exceeding) the performance of other magnetic microfluidic sorting devices [79, 81, 320, 322].

We then investigated the performance of the device to process higher concentrations of particles to match the throughput of commercial FACS devices, which can sort up to 20,000 to 50,000 cells/sec [11, 274]. At 20,000 particles/sec, the purity decreased to 79±4% magnetic particles and 85±3% non-magnetic particles in the magnetic and non-magnetic outlets, respectively. At 55,000 particles/sec, which exceeds the maximum throughput of most commercial FACS devices, the purity diminished to

66±4% magnetic particles and 63±4% non-magnetic particles in the magnetic and non- magnetic outlets, respectively. We believe the reduction in sorting performance is due to the high volume fraction of particles (e.g., 0.7 vol.%), thus producing significant scattering effects and increasing secondary radiation forces that limit the ability of the standing wave to focus particles [40]. To this end, we believe designing wider fluidic microchannels that operate at multiple harmonic frequencies with parallel microfabricated ferromagnetic strips [81] could alleviate this limitation by creating multiple parallel streams of focused particles to reduce the concentration of particles in any one streamline [257].

163

6.3.2 Magnetic bead capture on the magnetographic array

Next, we investigated the performance of the magnetographic array to template magnetic particles as surrogates for cells. Prior to each experiment, we set the in-plane magnetization of the micromagnets such that when the particles were exposed to a vertical field, they were either attracted to, or repelled by, the open microwell near on the poles of each micromagnet. We injected a sample of ≈2x104 particles/mL magnetic polystyrene particles (18.4 μm dia.; Spherotech) into the device at a low flow rate (e.g.,

50 μL/min) and we arrested the flow for 5 min to allow the particles to assemble in the microwells (Figure 50A). We then imaged the array in sections using an upright microscope.

We quantified the capture accuracy by counting the number of particles in each microwell. N.B., we did not count conditions where microwells neither captured particles nor had particles nearby (i.e., within a ≈40 μm radius from the center of the microwell) as these events were a consequence of a low starting concentration instead of the inability of the micromagnets to capture [88]. We found the array was able to capture one magnetic particle per microwell at an incidence of 95.5±4.4% (Figure 50B). This number decreased to 66.3±4.9% in “Control 1”, where the micromagnets were polarized, but the device was not in a vertical magnetic field. This number further decreased to

17.7±10.4% in “Control 2”, where the micromagnets were not polarized, nor was the device in a vertical magnetic field (which is analogous to randomly depositing particles).

164

We found that only ≈0.05% of microwells failed to capture magnetic particles (i.e., where a magnetic particle was located within the capture radius of a microwell, but was not captured), but this number drastically increased to 24.4±8.2 and 70.2±15.9% in “Control

1” and “Control 2”, respectively. Figure 50C shows an image of the finished device, and

Figure 50D shows micrographs of the magnetographic array before (left) and after

(right) the injection and successful capture of magnetic particles.

Figure 50: Magnetographic bead templating. A) Schematics of the magnetographic templating module where magnetic particles were injected (top), captured (middle) and retained for analysis (bottom). Vertical black arrows indicate the direction of the uniform magnetic field, and the horizontal white arrows indicate the direction of polarization of the micromagnets. Parabolic black arrows indicate the direction of fluid flow. B) Capture accuracy of the magnetographic templating module through

165

three conditions: Experiment (i.e., micromagnets are polarized, and the chip is in a vertical magnetic field; n = 2,098 particles), Control 1 (i.e., micromagnets are polarized, but the chip is not in a vertical magnetic field; n = 610 particles) and Control 2 (i.e., micromagnets are not polarized, nor is the chip in a vertical magnetic field; n = 857 particles). Labels on the x-axis correspond to events where one particle is captured per microwell (left), multiple particles are captured per microwell (middle) and no particles were captured, but were within the capture radius (right). C) Photograph of the magnetographic templating module. D) Fluorescent micrographs of the array prior to (left) and after (right) the capture of magnetic particles.

6.3.3 Sorting prostate cancer cells from diluted whole blood

Using insights from sorting synthetic particles, we used the sorting module to separate prostate cancer cells from processed (i.e., erythrocyte-lysed) whole blood

(Figure 51A). Erythrocyte lysis buffers are used commonly to debulk blood samples, as they have been shown to not affect the viability of nucleated cells for post-isolation analyses [329]. While we note the device is capable of separating cancer cells from unprocessed (albeit diluted) whole blood, we lysed the erythrocytes (comprising about

99.9% of cells in blood) [89] prior to the start of experiments to enhance the overall throughput. For example, separating cancer cells from 7.5 mL of unprocessed whole blood would require 5.2 days of continuous operation at a flow rate of 100 μL/min, as the blood should be diluted ~100-fold to enable adequate acoustic focusing [102].

Instead, separating cancer cells from 7.5 mL of blood sans erythrocytes would reduce the duration of the experiment to 85 minutes, which includes the time to lyse the erythrocytes and re-concentrate the remaining cells. We note, however, that others have

166

used acoustic radiation forces in a microfluidic device to concentrate cells, thus obviating the need to centrifuge cells after lysis [327].

We spiked human prostate cancer cells into 6 mL whole porcine blood at a ratio of ~1:150 LNCaPs to leukocytes. While this ratio yields a concentration of cancer cells that is higher than the typical concentrations of CTCs in the blood of metastatic patients, we selected it as a baseline to allow for more accurate quantification and to demonstrate the efficacy of the sorting device in a proof-of-principle fashion. To prepare the samples, we lysed the erythrocytes, re-concentrated the cell-containing liquid back to its original volume via off-chip centrifugation and performed the labeling assay with anti-EpCAM magnetic beads (MyONETM Dynabeads®, SA T1; Figure 51B). Using insights from our numerical analysis (Figure 48C-D), we reduced the flow rate through the device to 25 or

50 μL/min to increase separation performance. We quantified the efficiency of separation via determining the fold-enrichment of cancer cells in the magnetic outlet and leukocytes in the non-magnetic outlet, respectively, via the relation (Figure 51C (left)):

: = (23) : 𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓𝑓 𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙 𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝 𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐 𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐 𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟 𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒ℎ𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 To perform the enumeration,𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖 we used𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙 𝑙𝑙optical𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙 𝑝𝑝 microscopy𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝𝑝 𝑐𝑐𝑐𝑐𝑐𝑐 𝑐𝑐to𝑐𝑐𝑐𝑐 distinguish𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐 𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟𝑟 cells, whereby

cells with a radius >5 μm that were bound to at least 5 magnetic beads were identified as

LNCaP cells and cells with a radius >5 μm that were not bound to magnetic beads were

identified as leukocytes. We note that LNCaP cells with a low expression of EpCAM

167

may have gone undetected with this methodology, which is a known limitation of positive cell selection.

We found the device was able to enrich cancer cells over 100-fold for both of the flow rates tested and that the performance of the device improved with decreasing flow rate (Figure 51C (left)). In the 50 μL/min case (i.e., 15 and 35 μL/min through the magnetic and non-magnetic outlets, respectively), the device enriched cancer cells 110- fold (n = 5). Reducing the flow rate to 25 μL/min (i.e., 7.5 and 17.5 μL/min through the magnetic and non-magnetic outlets, respectively), improved enrichment of cancer cells to 142-fold (n = 3). These performances correspond to a sorting efficiency of approximately 83% and 89%, respectively (Figure 51C (right)) and a purity of ≈110:150 and 142:150 LNCaPs to leukocytes in the output of the magnetic outlet (up from ~1:150 in the initial mixture). Finally, as a simple proof-of-principle test, we collected the magnetically labeled cells from all 8 trials and suspended the cells in culture media. We found that cells from one of the 8 trials remained viable after 96 hours (Figure 51C), indicating this device may have potential for culturing cancer cells post-isolation.

168

Figure 51: Separation of human prostate cancer cells from erythrocyte-lysed blood. A) Schematic illustration of the microfluidic device for the acoustically enhanced, magnetic cell separation of cancer cells from erythrocyte-lysed blood. B) Pictures of whole porcine blood first doped with LNCaP cells (6 mL, left), then diluted to 60 mL with erythrocyte lysing buffer (middle) and then concentrated to 6 mL after erythrocyte lysis (right). C) Performance of the device to enrich non-magnetic cells in the non-magnetic outlet (green) and prostate cancer cells in the magnetic outlet (blue) at two different flow rates (left) (initial ratio was 150:1 leukocytes to prostate cancer cells). Separation efficiency of the device to isolate magnetically labeled cells in the magnetic outlet (right) at two different flow rates. D) Example photographs of cultured prostate cancer cells 24-72 hours after separation from blood.

6.3.4 Capture of prostate cancer cells for staining and analysis in the magnetographic array

We evaluated the performance of the magnetographic array to capture and retain magnetically labeled cells for on-chip staining and analysis (Figure 52A). We coupled these experiments with simulations from COMSOL to quantify the pressures and forces imposed on single cells by the micromagnets. For the sake of simplicity, we modeled the

169

Co film without the other metallic layers, and we assigned it a relative permeability (μr)

of 250 compared to air. We estimated the magnetic saturation (Ms) using the relation:

= 1.2 = 1,011.65 / (24)

𝑆𝑆 𝐵𝐵 where μb and N represent𝑀𝑀 the Bohr𝜇𝜇 magnetron𝑁𝑁 (i.e., 9.27x10𝑘𝑘𝑘𝑘 𝑚𝑚 -24 A/m2) and the number of atoms (which was calculated via the relation N = ρ*NA/ MW), respectively.

Next, we modeled the magnetic properties of the magnetic beads. Using the specifications provided by the manufacturer, we estimated the beads were magnetized

(M) to 15 emu/g (or 30,600 A/m) by assuming a bead density of 2,040 kg/m3. Next, we calculated the magnetic susceptibility, χv, of the magnetic beads by following the relation:

= (25) 𝑀𝑀 𝜒𝜒𝑉𝑉 whereby H represents the field, which we assumed𝐻𝐻 was 800 G (H = 0.08 T / μ0 = 63,662

A/m). Finally, we calculated that the magnetic permeability of the magnetic beads in our system was 1.481 by the relation μr = 1 + χv.

Using these parameters in COMSOL, we calculated the field strength around a single micromagnet as well as a magnetic bead at various distances from the attractive end of the magnet. We also calculated the magnetic force acting on a magnetic bead at various distances from the attractive end of a magnet, and assuming the contact area of one bead on a cell is roughly proportional to its cross sectional area, we estimated the pressure exerted on a cell by a single bead (P = F/A). Finally, we estimated the force from

170

multiple (i.e., 15 and 30) beads acting on a single cell by the simplifying equivalent volume approximation. In this case, we modeled 15 and 30 beads as a single bead with a radius of 1.23 and 1.55 μm, respectively.

First, we modeled the magnetic force on a single magnetic bead at various distances from the field maxima (i.e., the attractive pole of the micromagnet) (Figure

52B). We found the pressure was approximately 0.3 kPa at maximum, which is several orders of magnitude lower than the approximate force expected to kill a cell (e.g., several MPa) [330]. In Figure 52C, we plot the magnetic field strength distribution around a single micromagnet and a magnetic bead at distances of 5 μm (top), 3 μm

(middle) and 1 μm (bottom) from the field maxima. Additionally, we scaled these calculations to Nb = 15 and 30 beads/cell, corresponding to the labeling conditions observed previously (Figure 48B (top)), by consolidating the volume of multiple beads into a single, high-volume bead and plotted the overall force, which reached maximums of 679 and 997 pN for the cases of Nb = 15 and 30 beads/cell, respectively (Figure 52D).

Finally, to verify the forces imposed by the magnetographic array were insufficient to affect cellular viability, we captured magnetically labeled LNCaP cells against EpCAM in the magnetographic array, achieving a capture accuracy of 80.2±7.8%, and we incubated the cells on-chip with calcein AM for 20 min. We then washed (i.e., by injecting PBS through the device at a low flow rate) and imaged the cells, whereby their

171

green fluorescence indicated viability while captured within the microwells (Figure

52E).

Figure 52: Assembly of human prostate cancer cells in microwells of the magnetographic array for staining and analysis. A) Schematic of the magnetographic array for capturing and organizing magnetically labeled cells (magnetic field gradient indicated by the large, yellow arrow). B) Plot of the pressure and force on a cell caused by a single magnetic bead (MyOneTM Dynabeads®, SA T1), determined using 172

COMSOL software. C) Corresponding magnetic field strength simulations of a single magnetic bead at various distances from the field maxima of a micromagnet using COMSOL software. D) Plot of the net magnetic force on a single cell generated by 15 (blue line) and 30 (red line) magnetic beads. E) Overlaid bright field and fluorescence micrographs of magnetically captured prostate cancer (LNCaP) cells (or clusters of cells), stained on-chip with calcein AM viability stain.

6.4 Conclusions

We have developed a microfluidic instrument capable of isolating rare cells from blood. Once separated, we show this instrument can organize cells across a spatially defined array for staining and on-chip evaluation. This system can quickly process medically relevant volumes of blood (e.g., 6.0 mL of blood in ≈120 min after erythrocyte lysis), which could be expedited with parallelization and further optimization. A key feature of this technology is the acoustic focusing module, which allows magnetic particles or magnetically labeled cells to encounter the magnetic field gradient approximately along the same streamline. We show that the sorting module is capable of isolating magnetic particles and magnetically labeled cells with efficiencies of 92% and

89%, respectively. Further, we show that the templating module is capable of organizing magnetic particles and magnetically labeled cells and permit facile on-chip staining and analysis.

We believe several simple alterations can be made to enhance the operational capabilities of the device. For example, doubling or tripling LNR (Figure 48A) would allow for a corresponding increase in τr and in the throughput or speed of the device.

173

Also, increasing the magnetic field strength with larger neodymium magnets could increase the overall throughput and enhance the sensitivity for isolating magnetically labeled cells with a low Nb (e.g., 5-10 beads/cell). Similarly, increasing the thickness of the cobalt film in the micromagnets of the magnetographic array may enhance the capture accuracy of magnetic particles or magnetically labeled cells. Further, we note that by virtue of the switchable and bistable magnetization of the micromagnets, inverting the uniform, external magnetic field can reverse the polarization of the micromagnets and thus eject magnetically labeled cells from the array on-demand [302].

This feature is may be useful for downstream analyses such as RT-qPCR.

A key limitation of the system is its use of positive selection. While positive selection is the method used in the only FDA approved technology for isolating CTCs

[331], there is a growing consensus of the importance of negative selection assays for isolating CTCs [89]. This is due to the down regulation of EpCAM (the most common surface marker used in the positive selection of CTCs) during the EMT process [2].

Consequently, positive selection assays can miss key cellular biomarkers that could produce misleading diagnoses. To this end, we note that the system shown herein could be adapted to separate cells via negative selection. By magnetically labeling leukocytes against CD45, magnetically labeled cells can be deflected from unlabeled cells (i.e., CTC candidates) after the lysis of erythrocytes. However, we note that these unlabeled cells could not be assembled on the magnetographic array due to the absence of magnetic

174

labels; however, these cells could be collected and stained with anti-cytokeratin, anti-

CD45, anti-EpCAM and Hoechst in a fashion similar to other CTC isolation approaches

[89].

Looking forward, CTCs hold enormous potential to provide a wealth of information (e.g., genetic, proteomic and metabolomic) on remote tumor sites, thus reducing and potentially obviating the need for invasive biopsies and costly imaging procedures. Isolating and analyzing rare tumor cells from blood may forge new pathways in medical practice for the diagnosis and treatment of various ailments towards truly personalized medicine. The tools shown herein are proof-of-principle.

With auxiliary development, this tool can be enhanced and optimized to enable higher throughputs and sensitivities. Furthermore, this instrument may be coupled with other technologies to enable genotyping and molecular characterization of captured CTCs or studies on single CTCs in response to various stimuli and drugs.

6.5 Acknowledgements

This work was supported by the National Science Foundation’s (NSF’s)

Research Triangle Materials Research Science and Engineering Center (MRSEC, DMR-

1121107), the National Institutes of Health (R21GM111584), and NSF Graduate Research

Fellowships (GRF-1106401). The authors would like to thank Lu Gao and David M.

Murdoch for their early contributions, Benjamin A. Evans for helpful discussion and the

staff at Duke University’s Shared Materials Instrumentation Facility.

175

7. Outlook

7.1 Promise of microfluidic cell sorters

Advances in cell biology, cell-based diagnostics and cellular therapies, along with the promise of point-of-care and personalized medicine, have increased the need for cell sorting devices in both basic research and in clinical applications that are safer, more efficient, and easier to use than current technologies. To address this critical need, researchers are looking toward microfluidics due to their accessible fabrication, low reagent consumption, and small footprints which offer significantly scaled-down sizes with design features that are commensurate with single cells [240]. Further, microfluidic devices can offer improved safety over traditional cell sorting technologies by eliminating potentially biohazardous aerosols.

Many of the microfluidic technologies developed to date are still in the prototype or proof-of-concept stage; few have demonstrated the clinical performance of a fully integrated and validated system. In order to reach their full potential, these technologies still require substantial commercial investment for standardization, manufacturability and repeatable performance. As such, microfluidic technologies still suffer from a few key limitations that must be overcome. For example, many of these devices suffer from low throughput due to single-channel or single-orifice designs. Advances in parallelization and other innovative scale-up approaches could help to overcome this barrier. This would prove particularly valuable for diagnostic applications that target

176

rare cell populations from liquid biopsies (e.g., such as CTCs from blood) or multiplexed cell sorting from large sample volumes. Secondly, the limited lifespan of microfluidic chips, which are typically shortened due to blocking or clogging, is another barrier to routine research or clinical use. The commercialization of cell sorting technologies may look to disposable chips to overcome these short lifetimes and enable a sterile, user- friendly operation. Finally, many microfluidic cell sorting systems still require complex sample preparations that reduce ease-of-use and can compromise sorting accuracies due to user variations. Therefore, a major challenge remains to build microfluidic devices that require minimal sample preparation and operational training.

The future of microfluidic cell sorting is promising. With the large variety of cell sorting devices (e.g., from active systems that use acoustic, electric, magnetic or optical forces to passive systems that use inertial effects, filters or immobilization procedures), microfluidic cell sorting technologies can offer speeds and accuracies in a number of ways that rival current commercial devices in a platform that is more efficient, less cumbersome and offers a more straightforward standard operating procedure. Unlike most commercial cell sorting machines, microfluidic devices are highly modular, thereby offering the capacity to perform multiple functions (such as mixing, counting and lysing analyzing single cells) in a single compact device. This ability positions microfluidic technologies at the forefront of the next generation of cell handling devices by providing the capability of performing high-level, post-sorting analyses such as

177

biochemical, secretion, and cell culture assays for true lab-on-a-chip devices for use in the laboratory and the clinic.

7.2 Summary

The results of this work demonstrate a functional microfluidic device (or set of devices) comprised of individual modules that function to sequester, separate and template single cells from heterogeneous mixtures for their direct evaluation on-chip.

We validated the components of the device with commercial beads as surrogates for cells, pure mammalian cell lines and processed blood doped with prostate cancer cells.

Overall, this work demonstrates new and potentially useful methodology to manipulate cells in fluids that may offer advantages over existing commercial methodologies (e.g., fluorescence-assisted cell sorting (FACS) and magnetic-assisted cell sorting (MACS)) and microfluidic methodologies. For example, the treatment of cells bound to elastomeric particles with forces from acoustic standing waves can provide a gentle and discriminate means to direct their movement with a high degree of precision for the purposes of separation and labeling [39, 49]. Further, the organization of cells across a spatially defined template using forces from a magnetic field can allow cells to analyzed in a rapid, automated fashion while leveraging the same magnetic beads used by other methods to sort cells (e.g., MACS) [12].

In Chapter 2, we described a pioneering method for synthesizing a class of elastomeric particles with programmable properties for precise spatial and temporal

178

control in acoustofluidic systems [94]. We synthesized these particles from the nucleation and growth of alkoxysilane and silicon alkoxide monomers in water, and we controlled their density and bulk modulus by reacting monomers with different numbers of siloxane bonds (i.e., two, three or four). We also demonstrated the ability to control the size of the particles over a broad range, from ≈150 nm to 15 μm, while

maintaining a coefficient of variance in size below 15% for most types of particles. Also,

using a newly developed opto-acoustic approach to individually assess the response of

particles to an acoustic traveling wave, we estimated the acoustic contrast factor (ɸ) of

several types of particles in water (i.e., particles comprised of trimethoxymethylsilane

(TMOMS) and a 8:1, 24:1, 48:1, 72:1 and 96:1 molar ratio of dimethoxymethylsilane

(DMODMS) to tetramethyl orthosilicate (TMOS)). We qualitatively verified these

predictions using an acoustofluidic chip to focus particles either to the pressure node or

antinodes of an acoustic standing wave.

In addition to this work, we developed a facile method to render the particles

fluorescent by incorporating hydrophobic dyes (e.g., Nile red) during the growth phase

of particle synthesis. Finally, we developed a method to functionalize the surface of

particles synthesized from vinyltrimethoxysilane (VTMOS) using a thermally-initiated

reaction of thiol-polyethylene glycol (PEG)70-biotin (MW 3400, Nanocs, Inc.) to the vinyl

groups on the particles. Overall, the results from this work has implications in the study

of acoustofluidics, particularly for the rapid and discriminate separation of negative

179

acoustic contrast particles from biological entites, such as cells. This type of discriminate separation may be useful for the isolation of cells and small molecules when they are bound to negative acoustic contrast particles, as discussed more in Chapter 4.

In Chapter 3, we described an approach for the fabrication and operation of

acoustofluidic devices comprised primarily of glass and silicon [273]. Briefly, we used

photolithography to define the microchannel over a silicon substrate, deep reactive ion

etching to form the channel in the substrate and anodic bonding to fuse the silicon to a

glass “lid” with predrilled holes. We finalized the device by gluing tubing adapters over

the predrilled holes and we fixed a piezoelectric transducer to the silicon substrate with

cyanoacrylate glue. We connected silicone tubes to the tubing adapters and soldered

wires to the transducer. Using this device, we acoustically focused particles with

positive and negative acoustic contrast factors, and we measured the extent of focusing

of polystyrene particles as a function of applied voltage and flow rate.

In Chapter 4, we used the acoustofluidic device described in Chapter 3 along

with biofunctionalized silicone particles described in Chapter 2 to capture and confine

mammalian cells to the pressure antinodes of an acoustic standing wave. Given their

relatively high densities compared to aqueous fluids, mammalian cells generally display

positive acoustic contrast behavior by focusing to the node(s) of an acoustic standing

wave. Conversely, given their high elasticity compared to aqueous fluids, silicone

particles generally display negative acoustic contrast behavior by focusing along the

180

antinodes of an acoustic standing wave. By functionalizing the surface of the silicone particles with streptavidin, we bound the particles to mammalian leukemia cells (KG-1a) displaying biotinylated anti-CD34. When injected into an acoustofluidic device, these particle-cell complexes migrated to the pressure antinodes of a standing wave when a

sufficiently high volume and sufficiently negative acoustic contrast factor of silicone

material was bound to a cell. The results from have useful implications in acoustic-based

cell manipulations. For example, binding cells to negative acoustic contrast particles

may permit: (i) rapid and discriminate cell sorting using forces solely from an acoustic

standing wave, (ii) cell immobilization within an acoustofluidic microchannel and

thereby (iii) on-chip staining and cellular analysis. Further, similar types of assays can be

developed and performed to isolate and confine other biological entities of interest such

as proteins, viruses and cell-free DNA [92, 332].

In Chapter 5, we leveraged the ability to acoustically focus particles and cells to generate a high precision (acoustically enhanced) magnetophoretic particle sorter. A major limitation of previous magnetic, microfluidic sorting instruments is the exertion of widely variable magnitudes of force due to the highly nonlinear decay of magnetic fields

[274]. Consequently, entities positioned far away from the field maxima experience substantially less magnetic force than those positioned close, and thus, often are missed in continuous flow separation applications. Therefore, we used forces from an acoustic standing wave to concentrate particles or cells into a single, well-defined streamline of a

181

microchannel such that every particle encounters the magnetic field in the same location, thus generating similar magnitudes of magnetic forces for more precise separations from nonmagnetic particles. We found that we could attain a sorting efficiency above 90% in both the magnetic and nonmagnetic outlets when operating at throughputs as high as

2,000 particles/sec, and that the device can operate with a reduced performance with throughputs as high as 55,000 events/sec.

Also described in Chapter 5, we designed and tested a magnetographic array, comprising an aligned array of micromagnets and microwells, to capture magnetically labeled cells for on-chip cellular analysis. We studied the accuracy of the micromagnets to capture magnetic particles and magnetically labeled cells, and we found that it can capture more than 95% of magnetically labeled cells. Then, as a proof of principle, we captured T cells via magnetic beads coated with anti-CD3, and we labeled those cells against CD4 and CD8 antigens with fluorescent antibodies. We scanned the chip with a fluorescence microscope and quantified the ratio of CD4 to CD8-expressing cells, which is mutually exclusive in most CD3+ T cells, and we compared the results against a flow cytometer. The results between the two methods differed by only 5.4%, indicating the magnetographic chip can be an effective tool for isolating and staining cells for accurate immunological analyses.

In Chapter 6, we combined the systems described in Chapter 5 to create a holistic microfluidic cell processing instrument comprised of three modules to (i) acoustically

182

focus, (ii) magnetically separate and then (iii) magnetically template cells on a magnetographic array. We used this system to process whole blood following the lysis of erythrocytes without dilution as to enable higher throughputs (e.g., processing 1 mL of whole blood per 20 min). We doped human prostate cancer cells into porcine blood as surrogates for CTCs and labeled them with anti-EpCAM coated magnetic beads. In parallel to this work, we developed a numerical model to predict the magnetophoresis and separation of magnetically labeled cells from non-labeled cells. Using insights from this model, we optimized the parameters of this system and found it could enrich cancer cells from blood over 100-fold (attaining purities of prostate cancer cells as high as 89%) and permit their isolation on the magnetographic array for staining and analysis.

7.3 Future directions

7.3.1 Integrated modular system for holistic processing of CTCs

Using the tools we have developed insofar, we envision a single, integrated

microchip instrument for the rapid isolation, immunophenotyping and enumeration of

rare human cancer cells (e.g., circulating tumor cells (CTCs)) on-chip. First, we would lyse erythrocytes using common commercial lysing buffers, as described in Chapter 6.

Next, we would use the device described in Chapter 3 and the elastomeric particles described in Chapters 2 and 4 to capture and debulk leukocytes and granulocytes by using anti-CD45 and/or anti-CD62 as capture reagents [89] on the elastomeric particles

(left hand portion of Figure 53) . Then, the remaining unbound cells would continue

183

flowing through a second resonant portion of the microchannel where cells would be refocused for downstream magnetic separation via forces from a gradient magnetic field, as described in Chapter 5 (middle portion of Figure 53). Finally, the fractionated magnetically labeled cells would be collected and assembled on a magnetographic array for staining and enumeration (right hand portion of Figure 53). Overall, this continuous flow system is designed to process complex, biological mixtures in a single pass, and is particularly well-suited for the isolation of CTCs. The negative selection of leukocytes with elastomeric particles is designed to improve the sensitivity of the positive selection module (operating via magnetophoresis) by removing many of the excess cells. The sensitivity of this device would be compared against various clinical standards such as

MACS, flow cytometry and CellSearch (Janssen Diagnostics, LLC).

Figure 53: Integrated microfluidic device for the on-chip isolation and analysis of rare cells. The chip contains three modules: (i) a debulking module to remove excess white blood cells via elastomeric particles, (ii) an enriching module to purify rare cells (e.g., CTC candidates), and (iii) a templating module to allow cell immunostaining and automated cellular analysis. Elastomeric particles, white blood cells, rare cells and magnetic particles are indicated by the colors of blue, white, green and black, respectively. The bolded black and shaded black arrows indicate the direction of flow

184

and the direction of the gradient magnetic field, respectively. The dotted lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale.

7.3.2 Isolation of CTCs via negative selection

While the positive selection of CTCs is the only FDA approved approach for their

isolation and enumeration [312], and is the approach we propose and describe in the

previous section, many researchers are focusing their efforts on the negative selection fo

CTCs [89, 91]. The motivation for this recent change is two-fold. First, while EpCAM is the predominant marker used for the positive selection of CTCs [312], it is not present on every CTC [216]. Therefore, when selecting these cells via EpCAM, some CTCs are likely to go undetected. Second, and possibly a critical factor in the first, EpCAM is usually downregulated during the epithelial-mesenchymal transition (EMT), thus reducing the likelihood that surface-specific tags on immunospecific beads will capture CTCs [2, 333,

334].

The tools developed in this dissertation can be adapted to isolate CTCs via negative selection. We propose two simplified methods of operating of the microfluidic instrument to capture CTCs via negative selection. The first method involves using forces from an acoustic standing wave and elastomeric particles coated with anti-CD45 and anti-CD62 to debulk leukocytes and granulocytes (Figure 54). In this approach, white blood cells would be continuously removed from solution, and the remaining cells

(considered CTC candidates) would be recovered for staining (e.g., with Hoechst DNA stain, fluorescent anti-EpCAM, fluorescent anti-CD45 and fluorescent anti-cytokeratin) 185

to confirm a tumorous origin via analysis from fluorescence microscopy for enumeration and other downstream analyses.

Figure 54: Debulking leukocytes with elastomeric particles and acoustophoresis. Objects in white, green and blue represent white blood cells, CTC candidates and elastomeric particles, respectively. The bold arrow indicates the direction of flow and the dotted lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale.

One potential roadblock of this approach is the relatively poor binding efficiency between elastomeric particles and cells. Since white blood cells can outnumber CTCs one million to one [312], the inability to remove at least 99.9% of white blood cells may limit the performance of the device and thus lead to more tedious analyses. This difficulty could be overcome by attaching biomimetic polypeptides (e.g., elastin-like polypeptides (ELPs)) to the surfaces of the particles to enhance binding to cells due to the biospecific interactions between the peptides and cell surface antigens. When elastomeric particles are functionalized with ELPs displaying lower critical solution temperature behavior (i.e., where certain stimuli can produce instabilities that drive the

ELPs to precipitate), changes in temperature, salt concentration and pH can trigger a phase transition in the polypeptide to induce the co-aggregation of particles and cells 186

immunospecifically labeled with the same, or similar, temperature-sensitive motifs [335,

336]. In this strategy, we propose the covalent binding of ELPs to the surfaces of the elastomeric particles and antibody-directed binding of ELPs to the surfaces of white blood cells. Modest increases in temperature (e.g., to 37°C) can induce a phase transition and the ELPs will co-aggregate, leading to the capture of cells on the elastomeric particles for acoustofluidic removal.

Alternatively, we can use the device described in Chapter 5 to negatively select

CTCs by debulking white blood cells via magnetic labels and magnetophoresis (Figure

55) [89]. In this approach, instead of using magnetic beads conjugated with anti-EpCAM, as in Chapter 6, we would use magnetic beads conjugated to anti-CD45 and anti-CD62 to magnetically remove leukocytes and granulocytes, respectively.

Figure 55: Debulking cells with magnetic particles and magnetophoresis. Objects in white, green and black represent white blood cells, CTC candidates and magnetic particles, respectively. The bolded black and shaded black arrows indicate the direction of flow and the direction of the gradient magnetic field, respectively. The dotted lines indicate an acoustic standing wave. PZT stands for lead zirconate titanate, the material comprising the piezoelectric transducer. Figure is not to scale.

187

Since CTC candidates obtained from either of these approaches will not have magnetic labels bound to their surface, they cannot magnetically assembled into a magnetographic array. Therefore, we would collect the CTC candidates and incubate them in solution with the same panel of reagents used in commercial CTC tests (i.e.,

Hoechst DNA stain, fluorescent anti-EpCAM, fluorescent anti-CD45 and fluorescent anti-cytokeratin). The samples would be centrifuged and suspended in a small volume of liquid (e.g., 5 µL) for manual inspection under a microscope slide to elucidate the origin of the remaining cells and enumerate cells from a tumorous origin.

7.3.3 Post-isolation analysis of CTCs: qRT-PCR and CTC culturing

In addition to the automated sorting and enumeration of CTCs, our instrument can permit the collection of cells for off-chip analyses such as cell culturing, genotyping, immunophenotyping, mutational analyses and high-throughput gene sequencing [219].

As described previously, CTCs captured into wells of the magnetographic array can be released on demand by virtue of the switchable magnetization of the micromagnets [88].

These cells can be released in bulk for batchwise analytical techniques (e.g., for culturing long-term), or individual cells can be identified and removed for further analysis. To exclusively isolate individual cells, precision micromanipulation devices (e.g., the miBot micromanipulator; Imina Technologies SA) can be used to guide the placement of a thin capillary connected to a pneumatic syringe to extract individual cells from the template, after decoupling the lid of the magnetographic array from the substrate [337].

188

Once recovered, standard tools such as the FLEXsix IFC TacMan standard gene expression workflow qRT-PCR system (Fluidigm Corp.) can be used in series with our cell isolation instrument to assess levels of gene expression. To this end, we would select

various primers of interest for a given type of cancer to amplify the genes corresponding

to common activity in those cells. For example, when screening for prostate cancer, we

would target genes corresponding to prostate-specific antigen, EpCAM, cytokeratins 7/8,

18 and 19, as well as genes not expected in prostate cancer cells such as those encoding

for CD45 and CD62 [338-340].

7.4 Conclusions and implications of this research

The results of this work demonstrate a versatile set of strategies for manipulating particles and cells in microfluidic systems. These methods are particularly suitable for separating and analyzing rare cells, such as CTCs, and may provide useful alternatives to more established techniques (e.g., FACS and MACS), which have scantily changed over the past few decades [341].

Innovation in cell sorting technologies has become critically important, as recent advances in cell therapy and personalized medicine have necessitated advanced cell handling techniques that can provide more detailed insights into the biology of isolated cells, in contrast to previous commercial tools [332]. To this end, there still exists a great need to isolate rare cells from blood in a rapid, gentle and highly selective manner that

189

can permit automated phenotypic analyses and enable extraction for long-term (e.g., cell culture) studies in a compact, microfluidic platform.

The tools described herein may provide a means to address some of these challenges in rare cell isolation and analysis, thus directly imparting implications in oncology, immunology and cellular biology. Additional development and optimization of these tools and methods may create new avenues for developing ‘next-generation’, high performance cell processing devices that could make a significant impact on advanced prognoses and diagnoses for a variety of human ailments and diseases.

190

Appendix A: Field-Directed Assembly of Patchy Anisotropic Microparticles with Defined Shape

A.1 Introduction to field-directed colloidal assembly

Colloidal assembly provides a versatile approach to generate highly ordered superstructures from particulate components by tailoring their intrinsic chemical or physical properties as well as their response to environmental stimuli [342, 343]. Opal gems represent an aesthetic example of a highly ordered structure that can be formed by the self-assembly of monodisperse silica particles into close-packed crystalline lattices

[256, 344, 345]. Drawing inspiration from nature, researchers are striving to develop similar processes that enable the fabrication of highly ordered structures that are otherwise not easily attainable by conventional reductive (top-down) microfabrication

[345]. There exist several approaches to drive the assembly of colloidal structures, including maximization of entropy [346], sedimentation [347, 348], dehydration [349,

350], capillary attraction [351], and Coulombic interactions [352]. While these approaches can provide numerous means to assemble ordered structures, the majority are limited to producing two and three-dimensional close-packed geometries. A few recent studies have employed more complex particulate components to form chains from magnetic

Janus triangles [353] and various cluster assemblies from magnetic Janus spheres [354], amphiphilic Janus spheres [355], and directionally programmable DNA labeled colloids

[356].

191

The use of external fields to drive assembly processes offers the potential to induce tailored, long-range interparticle forces with tunable intensity and directionality that can generate a wide diversity in assembled structures [328, 357, 358]. Another advantage of field-directed assembly is that particle interaction strengths are easily tuned to play a role analogous to changing temperature, which allows assembled structures to be melted, annealed and quenched, or dynamically toggled between different functional states [344]. Dielectrophoresis (DEP) and magnetophoresis (MAP) are two common methods used in directed assembly that can introduce orientation- dependent interactions between multi-component patchy particles. For example, dielectrically susceptible patches positioned at precise locations on the particle surface allow manipulation of the particle in an electric field to be controlled via applied field orientation and frequency [30]. The dynamics of assembly processes can also be controlled by adjusting the wave shape, symmetry, and phase of a multi-dimensional external field [359]. MAP assembly techniques are tuned in a similar manner by adjusting the field magnitude and the type of carrier fluid, leading to a corollary set of structures to those produced by electric field assembly techniques [360-363]. One of the key differences between electric and magnetic field assembly approaches is that the magnetic fields typically used are static, whereas the alternating electric fields typically used can lead to heating and additional electro-hydrodynamic effects that increase the complexity of the particle interactions [360, 364, 365]. Thus, the use of a complementary

192

set of electric and magnetic assembly techniques offers potential for enhanced flexibility in the types of assembled structures that can be achieved.

Within the context of DEP and MAP assembly, selective coating of particles with patches of metal allows for the generation and modulation of orientation-dependent interactions that qualitatively resemble higher order multipolar interactions [366, 367].

Spherical Janus [354, 368-370] and patchy particles [367, 371] have been assembled by

DEP [357] and MAP; however, to date there have been relatively few demonstrations of structures assembled from non-spherical patchy particles [353, 372, 373]. There are several approaches for producing anisotropically shaped microparticles, such as elastic molding [374], emulsion templating [375], rigid molding and micromachining [376], seeded growth [377-379], nonwetting templates [380], and photolithography [353, 381], which, collectively, can generate cubes [378, 382], cylinders and capsules [375], rods [377,

379, 383], tetrahedrons [384] and many other anisotropic shapes [374]. Utilization of particles with anisotropic shape and physical properties can provide additional versatility in controlling the directional interactions between particles to enhance the structural diversity of assemblies to include, for example, different close-packed as well as non-close-packed crystal lattices. These structures have potential applications as metamaterials [385] (e.g., photonic [386-388] and phononic crystals [389]), conductive wires [390], micromirrors [381, 391], biosensors [392] and multiferroic materials [393].

193

Here, we use convenient and widely available methods to fabricate anisotropic,

Janus and patchy particles, such as cubes, cuboids, hexagonal prisms, cylinders, discs, and parallelepipeds (see the overview provided in Figure 56). The diversity of particle shapes is determined not only by photolithographic patterning, but also by the thickness of the photoresist as well as the angle of optical exposure through the photomask. We demonstrate that the shape of particles as well as the placement, type, and thickness of the metal patches on the particles’ surfaces plays an important role in the characteristics of the resulting assemblies, including their structure, orientation, uniformity, and topological complexity. We compare and contrast AC electric and DC magnetic field- based assembly of these anisotropic patchy particles, we model their field-induced interactions through COMSOLTM based numerical simulations, and finally we provide a prospectus for the potential for the use of anisotropic patchy particles as fundamental building blocks in the directed assembly of hierarchical materials such as multi- dimensional wires, lattices, sheets, networks, and branched structures.

194

Figure 56: Schematic depicting steps in the fabrication and assembly of anisotropic, patchy particles. A) Anisotropic microparticles with tunable aspect ratios and vertex angles are generated via photolithography, (B) patterned with various metals, and (C) suspended in liquids and assembled to form distinct structural motifs.

A.2 Experimental

A.2.1 Particle fabrication

Single-side polished 3” (111) Si wafers (Addison Engineering, Inc., San Jose, CA)

were used as substrates. These wafers were rinsed serially with acetone and methanol

and dried at 95°C on a hot plate for 5 min. SU-8 negative photoresist (MicroChem,

Newton, MA) was selected as a base material for microparticle fabrication due to its

ability to generate large aspect ratio structures by altering the concentration of resin in

the prepolymer and spin coating parameters. SU-8 was added drop wise to the wafer

and spin-coated to precise thicknesses following protocols provided by MicroChem

(MicroChem, Inc., Newton, MA). Chrome-printed quartz photomasks (Photo Sciences,

195

Inc., Torrance, CA) contained an array of shapes (circles, squares, or hexagons) of 5 to 10

μm in size with equal spacing in the planar directions were used to pattern approximately 1.1×107 particles per wafer. Spin coated wafers were exposed to UV light

(365nm, 120 - 250 mJ cm-2, Suss MicroTec MA6/BA6, Garching, Germany), post-baked, developed, and rinsed with isopropyl alcohol and dried in nitrogen.

Metal layers were deposited on the structures using an electron beam metal evaporator (EBME) (CHA Industries Solution E-Beam, Fremont, CA) for top, angle, and side-coatings. Top-coatings were achieved by mounting wafers into the EBME in a normal orientation; angle-coatings were achieved by mounting them on an adjustable stage to control the angle of metal deposition. We note that metal coatings covering more than two sides of a cubic, or similar, structure are possible by tilting the stage to angles near 45°. Side-coatings were achieved by removing the particles from wafers with a rubber policeman, suspending particles in water, sonicating for 10 min, depositing the solution on a clean wafer, air drying such that the receding meniscus spontaneously deposited high aspect ratio particles onto their side (where electrostatic forces retained the particles on the surface), and then using an EBME to deposit metal. Patchy particles for DEP assembly were coated with 15 nm Cr followed by 50 nm Au. Patchy particles for

MAP assembly were coated with 15 nm Cr, 25 nm Ni, 15 nm Cr and then 50 nm Au.

196

A.2.2 Particle assembly, modeling and analysis

Fabricated particles were observed under an optical microscope (Zeiss Axio

Imager A2, Thornwood, NY) and a scanning electron microscope (FEI XL30 ESEM,

Hillsboro, OR) with energy-dispersive X-ray spectroscopy (EDS, Bruker XFlash 4010

EDS Detector, Madison, WI). The sides of uncoated, top-coated, and side-coated cylinders were analyzed by EDS. Resultant data was smoothed in MATLAB using two iterations of the built in smoothing function (MathWorks®, Natick, MA) to reduce the noise detected in the signal and highlight the underlying trends. A linear mapping scan was performed across the length of three different particles along three separate locations (totaling n = 9) for each particle type. The mapping analysis was performed to detect the relative presence of various elements across the length of each particle.

The general design of the DEP and MAP assembly cells have been described previously [30, 394]. Particles for DEP assembly were suspended in 1.0 vol.% Tween 20 surfactant (Sigma-Aldrich, St. Louis, MO) to reduce particle aggregation, whereas particles for MAP assembly were suspended in 0 to 4 vol.% ferrofluid (10 nm Fe3O4

nanoparticles suspended in deionized water, EMG 705 from Ferrotec) and 2.0 vol.%

Tween 20. DEP assembly images were taken within seconds to minutes of field actuation

whereas MAP assembly images were taken on the order of minutes to hours.

Modeling of top-coated patchy cubes in electric and magnetic fields was

performed in COMSOLTM using its predefined electrostatics package. The electrical

197

conductance and permittivity of the metal coating, SU-8 polymer, counter-ionic layer

and surrounding fluid were incorporated into the evaluation of electric field distribution, while the magnetic permeability of the metal coating, the SU-8 polymer, and surrounding fluid were incorporated into the evaluation of the magnetic field distribution. The models assumed electric and magnetic field strengths of 20 kV m-1 and

25 kA m-1, respectively. In both models, particles were spatially constrained in two dimensions and the metal-coated face was on the side of the particle, parallel to the applied field. Then, several possible configurations of two DEP assembled top-coated patchy cubes were selected and spatially fixed before performing free energy calculations to predict the most favorable configuration with the lowest energy.

A.3 Results and discussion

A.3.1 Microparticle fabrication and characterization

We fabricated a variety of different types of anisotropic microparticles with precise uniformity in size and shape from SU-8 photoresist (Figure 57) using conventional projection photolithography. The particles used in the experiments had base sizes ranging from 5 to 10 µm and heights ranging from 5 to 25 μm. Scanning electron microscopy (SEM) images revealed that the bases (as defined in Figure 56) of cubes and rectangular prisms were slightly rounded, which was conceivably due to the resolution of the chrome printing on the photomask or the rate sensitivity of the chemical developer.

198

Figure 57: Anisotropic particles. Representative SEM images of SU-8 particles include (A) 10 µm cubes, (B) 10 µm tall cylinders, (C) 10 µm tall hexagonal prisms, (D) 25 µm tall cylinders top-coated with 15 nm Cr and 50 nm Au before removal from silicon wafer substrate, and (E) those cylinders after removal. Image (F) is an optical micrograph of 10 µm parallelepipeds in suspension. Scale bars in all micrographs are 50 μm.

While we focus on the assembly of cubic and cylindrical particles below, we note that other shapes (e.g., pentagonal, star, and washer) are easily attainable using this method. In addition to simple prismatic or prism-like shapes, we were able to generate multifaceted particles with facets slanted in the z-direction by exposing the SU-8 through the photomask at oblique angles. An example of such multifaceted, non- prismatic particles is the parallelepipeds shown in Figure 57F. While the examples shown herein are interesting in the study of directed assembly in their own right, it is expected that feature sizes can be scaled down by orders of magnitude using state-of- the-art fabrication tools of the semiconductor industry.

199

We used several coating strategies, including top-, side-, and “angle”- (or two

adjacent sides) coating, to generate metal patches on shaped particles that allow tunable

dipole-dipole interactions in external fields to modulate particle interactions during

directed assembly, and in turn, to control the resulting morphology of the assembled

structures. Coated particles were characterized by SEM, energy dispersive x-ray

spectroscopy (EDS) and optical microscopy. EDS confirmed that metal deposition can be

generally confined to the top faces of shaped particles on wafers in that minimal

amounts of metal were detected on the sides of top-coated cylinders (Figure 58).

Figure 58: Atomic percentage of gold detected by EDS along the side of different 25 µm tall cylinders. Smoothed data from the side of an uncoated cylinder (blue), a top- coated cylinder (green), and a side-coated cylinder (red). Metal films coated on surfaces orthogonal to the source were nominally 15 nm of Cr and 50 nm of Au.

A.3.2 Dielectrophoretic assembly of microcubes and microcylinders

Prior to AC dielectrophoretic assembly, the metallo-dielectric particles fabricated as described above were stabilized in suspension by non-ionic surfactant (1.0 vol.%

200

Tween 20 in deionized water) to reduce aggregation by hydrophobic and van der Waals interactions. The ionic strength of the fluid suspension, which was on the order 10-6 M according to measured conductance values, was purposefully minimized to not affect the dielectrophoretic assembly process. Particle suspensions were transferred into a DEP assembly chamber consisting of a glass slide, with hydrophobic ring containing the liquid suspension, and covered by a plastic cover slip. The electrodes were two strips of metal film (15 nm Cr and 95 nm Au) deposited by evaporation on a glass slide (Figure

59). Unlike previous experiments with smaller microspheres [357], the cell used here was inverted during DEP actuation [395]. The inverted cell allowed for decreased friction between the large sedimenting particles and the substrate, as the electric field gradient exerted an upward DEP force that partially counters the effect of gravity, making it easier for the particles to move and find their minimum energy orientations under the action of the field. Otherwise, if the electric field gradient was oriented to produce a force on the particles in the same direction as gravity, the sedimentation of particles on the bottom of the cell might constrain their tendency to orient during assembly.

201

Figure 59: Schematic of the inverted DEP assembly configuration. The DEP assembly cell is loaded, inverted, and then excited in an AC electric field with a gradient oriented against the direction of gravity. Only frequencies above 100 Hz were studied, which eliminated particle migration across the cell via direct electrokinetic effects (electrophoresis and electroosmosis) between charged particles and the electrodes.

Effects of electric heating were not observable due to the thin cell, use of deionized water and intermediate frequency. However, AC electrohydrodynamic flows near the edge of the electrodes could induce fluid motion [30], which may disrupt the assemblies. To avoid artifacts from such flows, we chose the site of observation to be in the middle of the 3 mm gap between the coplanar electrodes when performing the dielectric assembly experiments.

The dynamics of DEP-driven assembly of top- and angle-coated cubes as well as top- and side-coated cylinders were observed by optical microscopy (Figure 60). Based on the particle shape and placement of the metallic patches, several distinct assembly motifs were observed. In addition to the particle shape and patchy nature, manipulation of the AC electric field strength and frequency provide the two main parameters to tailor

202

the assembly. The location of the metallic patches on particles is generally discernible in the optical micrographs due to the optical transparency of SU-8. Particles appear transparent when the metallic patch was on one of the sides of the particle (i.e., not in the viewing plane), whereas they appear opaque when the metallic patch was on the top or bottom of the particle (i.e., within the viewing plane). When low concentrations of top-coated cubes (i.e., 5.5×105 particles mL-1) were subjected to field of intermediate intensity and frequency (15 kV m-1 at 10 kHz), single, isolated particles oriented at an angle of 45° within the viewing plane with respect to the field lines (Figure 60A; 77±21% of particles observed had this exact orientation). This 45° diagonal orientation allows for the maximum dipole in the field direction (i.e., the diagonal between opposite corners) to be induced across the square metal patch.

203

Figure 60: Optical micrographs of DEP assemblies of microcubes (10µm) and microcylinders. The direction of the field indicated in (A) is consistent in all images. (A) Contrasting configuration of top-coated cubes when apart and assembled (20 kV m-1 at 10 kHz), (B) chains of top-coated cubes (4 kV m-1 at 1 kHz), (C) chains of top- coated cubes (15 kV m-1 at 1 kHz), (D) chains of top-coated cubes (15 kV m-1 at 10 kHz), (E) chains of uncoated cubes, (F) kinked clusters of angle- (or two-side-) coated cubes (10 kV m-1 at 1 kHz), (G) typical orientations of top-coated cylinders (height and dia. = 10 µm; 10 kV m-1 at 1 kHz), and (H) wires of side-coated cylinders (height = 25 µm, dia. = 10 µm; 15 kV m-1 at 10 kHz). Observed assembly motifs are illustrated as an inset of the corresponding image. The scale bars in each image correspond to 50 μm.

The generalized DEP force acting on dispersed objects during assembly is well understood for spherical particles [30, 396]. However, force calculations become substantially more complex for non-spherical shapes, especially with anisotropic metal coatings and patches. In order to evaluate the DEP interactions and mutual orientation of anisotropic shaped patchy particles, the multipolar interactions must be considered

204

by integrating the electrical field energy over the surface of the particle while accounting for the different polarizabilities of each side of the particle [397]. Consequently, the magnitude of the DEP forces is dependent on the particle design and the field properties, both of which vary in the experiments presented herein; thus evaluate them for a few specific, limiting configurations.

As particles were attracted toward each other dielectrophoretically (4 kV m-1 at 1 kHz), they reoriented and aligned so that their edges were parallel and orthogonal to the field direction such that the particles organized into chains, usually with the metallic patch aligned with the field direction. Interestingly, dimer assemblies of top-coated cubes generally assembled in the direction of the electric field at an angle of approximately 0° (Figure 60A, right). As the chains of top-coated cubes extended in

length, the directional stability of the chains oriented collinearly with the direction of the

applied field increased (Figure 60B; 91±9% of particles observed had this orientation).

These chained, top-coated cubes appeared mostly opaque, indicating that the metallic patches were in the viewing plane and suggesting the metallic face of the chain oriented closest to the electric field maximum (i.e., top of the inverted cell). This is likely due to the higher polarizability of the gold facets on the DEP assembled chain, compared to that of the dielectric SU-8 polymer, which correspondingly drives the particle orientation in the direction of the field gradient [357]. This uniform orientation of metallic patches was observed at both low and high frequencies (i.e., 1 and 10 kHz,

205

respectively). However, when the field strength was increased to 15 kV m-1 at low frequencies (1 kHz), particles tended to assemble without orienting uniformly in the direction of the growing chain. In this case, the cubes within the assembled chains appeared randomly transparent and opaque, indicating the field strength was sufficient to induce chaining prior to reorientation (Figure 60C). In effect, the rapid assembly and strong attraction results in kinetically trapped configurations. Interestingly, at high electric field strengths and high frequencies (15 kV m-1 and 10 kHz), the assembled chains of top-coated cubes rolled onto their sides such that the metallic side of the chain was orthogonal to the viewing plane (Figure 60D), causing the chain became more transparent. This rolling behavior is likely a result of the directional interactions arising from the nonuniform electric field polarization across the particles at high frequencies, as explored below.

Uncoated cubes were also studied and underwent linear chaining (Figure 60E) from dipoles induced across each particle by the AC electric field as previously reported

[398]. Angle (or two-side) coated cubes produced “kinked” chains as they underwent

DEP assembly, where the particles were oriented 45° in-plane from the field direction

(Figure 60F). These structures were less uniform compared to the linear straight chains formed from DEP assembled top-coated cubes as the widths of kinked chains from angle-coated cubes were much broader (36.6±16.4 µm) than the width of chains formed from single top-coated cubes (near 10 µm). However, the cubes contained within these

206

assemblies consistently oriented at 45º to the field direction (81±13% of particles observed had this orientation) with the metallic sides facing in many spatial orientations

(Figure 60F). A preliminary analysis of the polarization energy of these two-side coated cubes (not shown) suggests that they have rather complex angle-dependent polarization and thus can be trapped in a several configurations, which makes these particles more challenging to assemble into desired patterns.

Top-coated cylinders (with an aspect ratio of ~1) assembled into three basic structures in AC electric fields, where a preponderance of a singular architecture was not observed (Figure 60G). The cylinders aligned edgewise (side-by-side) upright against the focal plane, end-to-end flat along the focal plane, and edgewise flat along the focal plane in 34±16%, 32±15%, and 34±8% of observed assemblies, respectively. The lack of a dominant assembly configuration of top-coated cylinders suggests the small aspect ratio and geometry of the circular metallic patches is not conducive for formation of only one free-energy-minimum structure in AC electric fields.

Side-coated cylinders (with an aspect ratio of ~2.5) aligned end-to end and formed elongated wires (Figure 60H) with the metal generally aligning to one side of the chain (91±5% of particles observed had this orientation). In all cases, higher voltages and longer field exposure times resulted in longer chain assemblies in the field direction. It was also observed that all DEP assembled chains immediately disassembled and dispersed on the bottom of the chamber when the field was turned off.

207

To better understand the formation and organization of the patchy particle chains, we performed COMSOLTM based numerical modeling of the two-dimensional electric field distribution of individual top-coated cubes at a range of frequencies (Figure

61A) [367]. By analyzing electric field distributions around the particle, the simulation data were used to interpret the observations of a preferred orientation as well as some of the dynamic features of the assembly process. At low frequencies (<10 kHz), both the metallic coatings and the counterionic layers around the SU-8 facets have greater electrical polarizability than the surrounding aqueous medium, and thus the electric field has features qualitatively similar to a point dipole. At higher frequencies (>>10 kHz), the ionic mobility does not play a role, and SU-8 becomes less polarizable than the surrounding aqueous medium; thus the electric field of the composite structure increasingly resembles a quadrupole (Figure 61A; 10 MHz), displaying four symmetric lobes instead of two, as seen in the case of the dipole (Figure 61A; 100 Hz). Since the metallic coating is more polarizable than the SU-8, this structure can be viewed as a mixture of a point dipole and quadrupole at these higher frequencies. This behavior is in agreement with previously reported results for metal-coated Janus spheres [367].

208

Figure 61: A) Simulated electric field intensity landscapes around top-coated cubes in AC electric fields at increasing frequencies (100 Hz – 10 MHz). B) Total electrical energies of the various expected assembly configurations at 20 kHz. The field direction is vertical in all figures and the black bars represent the metallic patches.

We also used COMSOLTM to calculate the electrical energy of various possible assembly configurations of two top-coated cubes (Figure 61B) in an AC field.

Configurations where both metallic faces were orthogonal to the field produced the highest energy, which is in agreement with experiments as these configurations were not observed. Configurations where one metallic face was orthogonal to the field and the other was parallel to the field produced lower energies. When both metallic faces were in parallel to the field, the total energy was lowest when the two metal faces were 209

on the same side of the cube. This model where the minimum energy structure reveals metal-coated faces aligned with the field (Figure 61B) agrees well with the empirical observations (Figure 60D).

The experimental data and the corresponding electric field calculations show that the rich variety of structural assemblies can be interpreted, and even predicted a priori, by an orientation-dependent energy minimization process for particles having frequency-dependent polarizability. The initial orientation is not necessarily the final one observed in the structure, as the structure evolves through the influence of particle- particle interactions. The conclusions derived during previous work by Velev and coworkers on the assembly of Janus spheres regarding the major driving and angle- orientation role played by the (conductive and hence highly polarized) metal patches remains valid [357], though the specific configuration in the present systems is locked-in by the geometric considerations arising from the shapes of the particles and the locations of their patches as they come together. Increasing the number of metal-coated facets increases the number of kinetically trapped configuration and may complicate the formation of well-organized long-range arrays. However, the results suggest that metal

patches can serve as a good director for assembling a rich variety of metallodielectric

arrays. Similar results using magnetically directed assembly are described below.

210

A.3.3 Magnetic assembly of microcubes and microcylinders

We examined the assembly of anisotropic particles with nickel patches in water

and in aqueous suspensions of ferromagnetic nanoparticles (ferrofluid) subjected to

static, uniform DC magnetic fields created by two electromagnetic coils placed on

opposite sides of the assembly chamber. Prior to assembly in the magnetic field, particles settled on the bottom of the substrate, which constrained their assembly in two dimensions. Particles with patches of higher magnetization than the carrier fluid (e.g., particles coated with ferromagnetic Ni patches in water) had their magnetic moments aligned parallel to the external field [360]. Particles with magnetization lower than the carrier fluid (e.g., uncoated particles in ferrofluid) behaved as non-magnetic cavities inside a magnetizable medium and exhibited an effective diamagnetic response where their magnetic moments aligned antiparallel to the external field [360, 364, 394]. The

MAP force acting on particles during assembly is well understood for spherical isotropic particles [77]. However, similar to the case of DEP, patchy morphologies introduce orientation-dependent interactions that must also be considered to evaluate the MAP forces and torques on the particles.

Figure 62 represents representative images of structures assembled from uncoated and metal-coated anisotropic particles in water and in ferrofluid (~1 vol.% suspensions) within a thin fluid filled chamber formed by two glass surfaces. With the field applied parallel to the plane of the chamber, top-coated cubes in water generally

211

formed staggered alternating, or “zigzagged”, chains (Figure 62A; 73±10% of particles observed had this orientation). However, in ferrofluid, these same particles generally formed edgewise (or side-by-side) chains due to the efficient formation of magnetic quadrupoles resulting from the presence of the diamagnetic SU-8 component with respect to the ferrofluid magnetization (Figure 62B; 91±6% of particles observed had this orientation in 1 vol.% ferrofluid) [360]. The zigzagged assembly morphology observed in

Figure 62A predominated in water or low ferrofluid concentrations over long periods of exposure to the field, during which particle alignment is likely dominated by the interactions between the Ni patches. Top-coated cylinders in water also formed zigzagged chains of cylinders with matching metallic faces aligning along the centerline of the assembly parallel to the magnetic field lines (Figure 62C; 80±10% of particles observed had this orientation), similar to the cubes observed in Figure 62A. Uncoated cylinders in ferrofluid exhibited a diamagnetic response and formed chains parallel to the external field (Figure 62D), as expected [30]. Side-coated cylinders in water usually formed zigzagged chains (Figure 62E; 86±10% of particles observed had this orientation) with the matching metallic faces aligning along the centerline of the assembly parallel to the magnetic field lines, similar to the top-coated cylinders in Figure 62C. However, these same cylinders generally formed edgewise chains in ferrofluid (Figure 62F; 89±4% of particles observed had this orientation in 1.2 vol.% ferrofluid), similar to the top- coated cubes observed in Figure 62B.

212

Figure 62: Optical micrographs of assemblies of microcubes (10 µm) and microcylinders (dia. = 5 µm, height = 10µm) within magnetic fields. The direction of the field indicated in (A) is consistent in all images. Solutions were in either water or ~1.0 vol.% ferrofluid. A) Zigzagged chains of top-coated cubes in water, (B) elongated rods of top-coated cubes in ferrofluid, (C) zigzagged chains of top-coated cylinders in water, (D) uncoated cylinders in ferrofluid, (E) zigzagged chains of side-coated cylinders in water and (F) edgewise chains of top-coated cylinders in ferrofluid. The assembly motifs observed most frequently in each case are illustrated in the top right- hand corner of each image. Scale bars are 25 μm.

This gradual transition from a zigzagged to a close-packed morphology is shown in Figure 63A. When suspended in water or low ferrofluid concentrations (0.1 vol.%), most of the observed structures are in the form of zigzagged chains. However, as the concentration of ferrofluid is increased (to 0.6 to 1.2 vol.%), the chains gradually re- arrange into close-packed structures. Adjusting the concentration of ferrofluid provides an additional level of control in modulating the architectures generated in magnetic fields, which is analogous to the tuning of the frequency of the external field in DEP assembly. In low ferrofluid concentrations, SU-8 exhibits a very weak response to the

213

magnetic field, while the thin Ni coating exhibits a strong ferromagnetic response, which results in formation of alternating chains (Figure 62A, C, E). The alternating chains are similar to the staggered chains observed previously in DEP of metallodielectric Janus particles [357]. At higher concentrations of ferrofluid, the SU-8 exhibits a stronger diamagnetic response, which likely results in the formation of a magnetic quadrupole within the particle, leading to the assembly of more chains that are close-packed in one dimension (Figure 62B, F). An increase in depletion forces at higher ferrofluid concentration may also contribute to the formation of the close-packed structures observed in Figure 62B and F [360].

214

Figure 63: Effect of increasing ferrofluid concentration on particle assembly and local magnetic field strength. A) Relative amount of zigzagged structures formed from top- coated cylinders in increasing concentrations of ferrofluid, and (B) simulations of the magnetic field strength distribution around individual top-coated cubes in increasing concentrations of ferrofluid.

It was thus established that the presence of ferrofluid during MAP is an effective means for controlled formation of close-packed vs. zigzagged structures. This transition is caused by a shift in the response from a predominantly paramagnetic response (due to the Ni patches) to one having a mixture of paramagnetic and diamagnetic responses due to the increasing importance of the SU-8 material in guiding the particle-particle 215

interactions [360]. To illustrate this transition, top-coated cylinders were magnetically assembled in a range of ferrofluid concentrations (0, 0.1, 0.2, 0.4, 0.6 and 1.2 vol.%) and their ordering was measured (Figure 63A) by counting the number of adjacent particles that have zigzag connections within each chain. The percentage of zigzag connections was measured as a function of the ferrofluid concentration and plotted in Figure 63A as a function of the ferrofluid concentration, where a perfect zigzagged chain had a ratio of

1.0 and a perfect close-packed chain had a ratio of 0.0 (Figure 63A). The configurational energies of both close-packed and zigzagged structures shown in Figure 63A were calculated at different ferrofluid concentrations. Similar trends were observed with top coated cubes.

To better understand the tendency to form non-close-packed (zigzagged) vs. close-packed assemblies, we modeled the magnetic field strength distribution around magnetically responsive patchy particles in a range of ferrofluid concentrations in two dimensions using COMSOLTM (Figure 63B). Due to the topographical symmetry of the two-dimensional model, the particles analyzed can be considered cubes, cylinders, or other prismatic shape. Not surprisingly, as the ferrofluid concentration was increased, the degree of magnetization of the SU-8 material also increased, which led to the increasing importance of the quadrupole like behavior, which is qualitatively similar to the effect of AC field frequency shown above. Thus, the ferrofluid concentration is an effective means of controlling the relative polarizability of the metallic patches vs. the

216

rest of the particle, with the resulting consequences for the assembly pattern. We discuss the similarities and the differences between these two types of directed assembly in the following section.

A.3.4 Comparison of the results from electric and magnetic field assembly

Dielectrophoresis and magnetophoresis represent a duality in directed assembly

of particles, in which fields control the energy of interaction between colloidal particles

in a phenomenologically similar way. At their core, both techniques depend on the

interactions driven by the polarizability of the particles relative to a surrounding

medium. For isotropic spherical particles, the higher order moments identically cancel in

a uniform field and are typically negligible in weakly inhomogeneous fields, which

allows the particle to be accurately treated as a point dipole [398]. However, our results reported here show that once the polarization symmetry is broken by anisotropy in the particle’s shape or the presence of an anisotropic surface coating, higher order moments and multipolar interactions can dominate the assembly [397]. The calculation of multipolar interactions for anisotropic particles is nontrivial and is beyond the scope of this work. Moreover, analytically calculated multipolar interactions are typically only accurate at relatively long distances. When two particles are nearly touching, finite element modeling, or other numerical modeling techniques, become necessary for an accurate computation of the local electric and magnetic fields.

217

For linear materials, the particle-particle interaction energy scales with the square of the local fields, = , where F is a general field variable (E or H for 2 𝑈𝑈 −ξ Ϝ electric and magnetic field, respectively) and ξ is the effective contrast factor used to represent the polarization state of the particle relative to the surrounding medium [399].

Particles that are more polarizable than the surrounding medium have positive contrast factor (ξ > 0), and the converse is true for particles less polarizable than the surrounding medium. Thus, minimization of potential energy is achieved by assembly processes in which parts of the particle with a positive contrast factor to move into regions of maximum field, whereas the parts of the particle with a negative contrast factor tend to move towards the regions of minimum field. For non-spherical and patchy particles, the interaction between two particles must be computed through an integral of a Maxwell stress tensor over the particle’s surface [397]. The particle-particle interaction force is proportional to the spatial gradient of the field, while the torques are determined through the rotational derivatives of the interaction energy.

The key differences between electric and magnetic field assembly arise from the disparity of the polarization processes relative to the surrounding media, the polarization timescales, and the diversity of polarization states of different media.

Nearly all materials are strongly affected by electric fields (i.e., most materials have electric permittivities that are substantially different than that of free space), whereas most materials are nearly unresponsive to magnetic fields. Additionally, many materials

218

respond to electric fields at high frequency (e.g., >> 1 kHz), whereas most magnetically responsive materials can respond to low frequencies (e.g., << 1 kHz). As a result of these fundamental differences, the techniques used to modulate the polarization of materials by dielectrophoresis and magnetophoresis can be substantially different. For example, the easiest way to control the electrical polarization of the particle relative to a surrounding medium (e.g., water) is by changing the frequency of an oscillating electric field. Since the relative polarization of such particles can experience a sharp transition over a narrow frequency range, it is possible to modulate a particle’s contrast factor from positive to negative using frequency as the control variable. By contrast, the simplest method for adjusting the magnetization of a particle relative to a surrounding medium is by immersing it inside a magnetic fluid, such as a dispersion of magnetic nanoparticles in an inert carrier solvent, such as water (i.e., ferrofluid). Here, the pertinent control variable is the ferrofluid concentration, which is used to adjust the contrast factor of a magnetic particle from positive to negative in a corollary manner.

Other interesting phenomena that occur in dielectrophoresis, but not in magnetophoresis, are due to the role of the counterions in aqueous media, which can cause electrohydrodynamic flows, AC electrokinetic propulsion or local heating; they can also be involved in electrochemical reactions that occur on metal surfaces [400]. One classical example is the propulsion of nanorods caused by electrochemical reactions on the surfaces of asymmetric particles [401]. Even in cases where electrochemical effects

219

are small, the polarizability of the ion cloud around charged particles leads to an additional time-dependent phenomenon that needs to be considered in dielectrophoretic

assembly processes and allows for an additional frequency dependent control parameter

at frequencies in the kHz range and above [400].

Our results demonstrate how field-driven processes become more complicated when colloidal particles are composed of two different materials that have dramatically different polarizabilities, as is the case for a metallic or magnetic coating on one side of a polymeric particle. For the case of patchy particles in electric fields, the interaction energy must be divided into terms that are related to the surface polarizations from the metallic coatings as well as those related to a volume polarization due to the underlying properties of the base particle material. Here, the diversity among the polarization properties of different materials can yield important differences between electric and magnetic field-directed assembly techniques. In dielectrophoresis, suspended particles typically have dramatically different polarization properties than the immersing fluid

(e.g., water), thereby rendering the particles responsive to DEP forces. In the case of electrolyte solutions, the response of the suspended particles is also strongly dependent on the immersing fluid as the frequency-dependent polarizability of the solution depends on the counterionic layers that can form near particle surfaces [400]. In contrast, in magnetophoresis, most fluids and polymeric materials have nearly the same polarization properties. This intrinsic difference allows for the rich variety of assembly

220

motifs in electric vs. magnetic fields as observed with the structures in Figure 60 and

Figure 62.

A final difference between magnetophoresis and dielectrophoresis of metal- coated Janus-like particles arises from the hysteresis properties that are commonly found in magnetic materials [359], but only rarely in dielectrics. For example, the Ni coating used to induce positive magnetic moments in this work is ferromagnetic, and thus particles can assemble even in the absence of an externally applied magnetic field.

This phenomenon can be exploited in stabilizing the assembled structures once the external field is removed. In our work, we discovered that if the Ni coating was too thick

(e.g., more than about 50 nm), it interfered with our ability to obtain highly reproducible colloidal assemblies. On the other hand, there are relatively few dielectric materials that display hysteresis, though some notable exceptions include perovskite based electric materials, such as lead zirconium titanates, and barium titanates [402].

These considerations suggest that a combination of magnetic and electric field assembly techniques applied in parallel could be used as a tool for making more complex lattices. While the use of these two orthogonal assembly fields was not demonstrated in this work, it is a direction worth pursuing in the context of directed assembly of engineered anisotropic particles with well-defined shape and polarization, as it may allow assembly with even greater levels of control over structural diversity.

221

A.4 Conclusions

We use fabrication techniques capable of producing large quantities of monodisperse anisotropic particles with tunable size, shape, and electromagnetic properties. These methods require only conventional microfabrication equipment, and can fabricate tunable patchy building blocks designed for field-directed assembly of highly ordered structures. As examples, we present a variety of prototype architectures assembled by DEP and MAP. Additionally, we have used computational modeling to describe local fields that lead to assembly as well as to compute potential energies that arise from pairwise interactions between anisotropic particles.

We have also provided an analysis of, and prospectus for, the use of electric and

magnetic fields in providing a direct means for assembling anisotropically shaped and

polarizable colloidal particles into controllable assemblies. The results above show that

metallic facets and patches on anisotropic particles can serve as reliable and well-

controlled directors of their assembly, which drive the particles in specific orientations

that then determine the structure. The induced interactions driven by the metal and the

ones arising from the rest of the particles can be further tuned and re-balanced by changing the frequency of the electric field or the composition of the magnetofluid medium (in magnetically driven structure formation). This allows selecting parameter configurations where only one structure having much lower energy in the field is assembled (e.g., compare the well-assembled motifs in Figure 60B-E vs. the poorly

222

assembled one in Figure 60F). Further, not only is it possible to efficiently use electric and magnetic fields to achieve high levels of control over assembly complexity, but it is also possible to assemble fundamentally different structures by exploiting the means of directing and controlling the particle orientation by the metal and the orthogonalities in the inherent nature of the two field-driven assembly methods. While these particles are relatively large, their ease of fabrication using commonly available microfabrication methods, together with their response and structure formation in the presence of electromagnetic fields, allows them to serve as models for directed assembly of quadrupolar particles and particles with non-spherical shape at a variety of length scales. Future applications for the metallodielectric lattices resulting from such assembly techniques may be found in microelectrodes, micromirrors [381, 391], metamaterials

[385], biosensors [403, 404], phononic [389] and photonic crystals [405], multiferroics, and microactuators [393].

A.5 Acknowledgements

This chapter contains material from a previously published article: Shields IV,

CW; Zhu, S; Yang, Y; Bharti, B; Liu, J; Yellen, BB; Velev, OD; López, GP. “Field- directed assembly of patchy anisotropic microparticles with defined shape,” Soft Matter 2013.

9(38): 9219-9229. DOI: 10.1039/C3SM51119G. The Royal Society of Chemistry granted permission for reuse of this article in this dissertation.

223

This work was supported by the National Science Foundation’s (NSF’s) Research

Triangle MRSEC (DMR-1121107), the NSF Graduate Research Fellowship Program

(1106401) and the Shared Materials Instrumentation Facility (SMIF) Undergraduate User

Program. We thank Dr. Mark Walters and the staff at Duke University’s SMIF and

Henry Taylor at North Carolina State University's Nanofabrication Facility for technical guidance.

224

Appendix B: Rapid Self-Assembly of Shaped Microtiles into Large, Close-Packed Crystalline Monolayers on Solid Surfaces

B.1 Introduction to passive assembly of shaped colloidal particles

A unified approach for the reproducible assembly of microscopic particles with shape anisotropy into monolayered crystals over large areas in a rapid and controllable manner has eluded researchers [406]. In lieu of assembling anisotropic particles, the majority of research in the past several decades has explored the self-assembly of spherical colloids due to their ease of synthesis and abilities to assembly into simple

(e.g., hexagonal close packed (hcp)) structures [256]. A great number of strategies have been developed to assemble these colloids, such as by evaporating solvents [350, 407-

409], trapping particles along various interfaces [410], templating particles in physical wells (or wells of minimum energy) [88] and manipulating (e.g., chaining) particles by external fields [411-413]. Recently, however, the synthesis and assembly of shape- anisotropic particles has generated significant attention because far more complicated structures (i.e., beyond hcp arrangements) are, in principle, attainable by virtue of the different conceivable particle geometries [406, 414, 415]. Assembling shape-anisotropic particles into well-ordered structures is more difficult than assembling spherical particles because both positional (i.e., translational) and orientational (i.e., from axial rotations in three dimensions) ordering is required. One notable approach for assembling microtiles (i.e., a class of anisotropic particles with an aspect ratio,

225

height:width ≠1.0, exploited lateral capillary forces (in the so-called “Cheerios Effect”

[416]) to attract particles across a fluidic boundary layer [417, 418]. This approach used particles that were chemically modified on one side and the assemblies were created by capillary forces at the menisci of the hydrophobic and hydrophilic faces at a liquid- liquid interface. However, a far more desirable outcome would be to generate shape- anisotropic colloidal crystals over large areas on a solid support, where the assembly does not require particle surface modification. These types of new materials would potentially have unique properties that could then be exploited as electronic, MEMS and sensoric devices, particularly if the resultant crystals were fabricated from particles of different shapes, sizes and aspect ratios.

B.2 Experimental

B.2.1 Fabrication of the microtile particles

SU-8 photoresist (MicroChem Corp.) was selected as a base material for generating the microparticles. First, a water-soluble sacrificial layer (Omnicoat, MicroChem) was spin- coated on a polished 3-inch Si wafer (Addison Engineering, Inc.) and baked following the procedures provided by MicroChem. A layer of SU-8 (either SU-8 2002, 3005 or 10)

was then spin-coated on the wafer and baked. Chrome-printed quartz photomasks

(Photo Sciences, Inc.) containing an array of shapes (i.e., 10 µm wide circles, squares or hexagons) with equal spacing in the planar directions were used to pattern ~1.0x107 particles for each 3 inch Si wafer. The wafers were exposed to UV light (365 nm, 120–250

226

mJ cm-2; Suss MicroTec MA6/BA6) through the photomasks, post-baked, developed,

rinsed with isopropyl alcohol and dried with nitrogen gas. Particles were then wetted

with Remover PG (MicroChem) and removed using a pressed-steel razor blade. Particles

were then washed (i.e., centrifuged 8,000xg for 2 min and resuspended in an equal

volume of 0.5 vol.% Tween 20 (Sigma-Aldrich, Co.) in DI water) prior to

experimentation.

B.2.2 In situ observation of self-assembly

The entire assembly process was observed using an inverted microscope (Eclipse Ti-E,

Nikon Instruments, Inc.) and recorded every millisecond using a high-speed shutter

(NIS-D) and charged-coupled device camera (DS-Ri1-U3, cooled colored digital camera)

system.

B.2.3 Scanning electron microcopy

The monolayers were washed with ethanol (Sigma-Aldrich) twice to remove surfactants and air-dried at room temperature. Prior to observation under field emission scanning

electron microcopy (FE-SEM; ZEISS SUPRA 40 VP, Carl Zeiss), samples were coated with 10 nm Au using sputter coating (Emitech International, Ltd.). Images were

captured under a beam strength of 5 keV.

B.3 Results and discussion

Herein, we describe a new facile droplet-based method for the self-assembly of

microtiles (i.e., 10 µm across) that combines two physical mechanisms: (i) the use of an

227

interface formed upon phase separation of two immiscible liquids to trap and uniformly orient particles (a so-called Pickering emulsions [419], and (ii) the mixing of those two liquids with a third, miscible co-solvent that drives the particles to the liquid-air interface for their rapid self-arrangement (similar to the ”Cheerios Effect” at the liquid- air interface [416]). This method of self-assembly overcomes many of the limitations of assembling small (e.g., 10 µm wide) shape-anisotropic particles into large, monolayered crystals by facilitating the organization of particles with various geometries and without surface treatments (Figure 64A). Three geometries of prismatic microtiles (i.e., circles, hexagons and squares) made from a SU-8 negative photoresist, using photolithography described previously [279], with three aspect ratios (i.e., height:width of 0.2, 0.9 and 2.0; see Figure 64B for example images). Photolithography is a highly accessible method to generate a large number of monodisperse microparticles (e.g., 107) in a batch-wise process [279]. Once fabricated and extracted from the surface, the particles were suspended in deionized (DI) water and stabilized with a small volume of surfactant (0.5 vol.% Tween 20) prior to assembly [420].

228

Figure 64: A) Overview of the fabrication and the droplet-assisted self-assembly approach of microtile particles. Monodisperse, shape-anisotropic microtiles were fabricated from SU-8 photoresist and were suspended in water with surfactant. The suspension of microtiles was added to a droplet of chloroform, causing the particles to self-arrange along the liquid-liquid interface. A co-solvent was then added to mix the solutions, to break the assembly into crystallites and to rapidly induce the self- assembly of those crystallites at the liquid-air interface. The solvent mixture was then evaporated such that the polycrystalline monolayer remained stably intact on the surface of the glass. SEM images of unassembled (B) circular, (C) hexagonal and (D) square microtiles with an aspect ratio of 0.2 are displayed as examples of the microtiles made from photolithography.

We assembled the microtile particles in three steps. First, we deposited a 30 µL droplet of dense, hydrophobic chloroform onto a clean glass slide. Next, we added 2 µL of the particle suspension (i.e., with a concentration ranging from 2x106 to 250x106

particles/mL in water) directly to the top of the droplet (Figure 65A(i, vi)). Due to the 229

low miscibility of the two liquids (i.e., ~0.54 mL chloroform in 100 mL water) and their markedly disparate densities (i.e., 1.49 vs. 1.00 g/cm3 for chloroform and water,

respectively), the two liquids remained stably phase separated (there is a mixing at the

liquid-liquid interface). The SU-8 microtiles from the water phase are pinned at the edge

of the two liquids, assemble from the pinning point, and get oriented lengthwise along

the interface of the two liquids to reduce their free energy in a phenomenon called a

Pickering emulsion (Figure 65A(i, vi)) [419, 421]. Finally, all particles are pinned at the interface of the two liquids (Figure 65A(ii, vii)). This mechanism has been observed previously for spherical particles [422]. Emulsion formation was a critical step for forming well-defined crystals because it allowed the particles to achieve the same relative orientation prior to their final arrangement into close-packed structures. When left unaltered, the chloroform evaporated faster due to its higher volatility and the remaining water wetted the surface under the particle layer, thus fragmenting the crystal layer and leading to consistently disorganized arrangements (Figure 65B(i)) [423].

In order to prevent this, a third and final step was added where we introduced a co- solvent (i.e., 30 µL of anhydrous ethanol) to the side of the chloroform phase, prior to evaporation, to rapidly induce the mixing of the two liquids and eliminate the water- chloroform boundary. The mixing process (~tens of seconds) was violent, generating chaotic flows within the mixture, which broke the crystal monolayer into smaller fragment crystallites that still remained fully intact (Figure 65A(iii, viii)). The strong

230

particle-particle interactions (e.g., depletion forces [424]) between microtiles were essential to the stability of the fragment crystallites during the mixing process [425]. At the late stage of this mixing, the fragment crystallites gradually began to emerge at the top of the mixed droplet and reassemble at the liquid-air interface due to lateral capillary forces [418], which eventually produced a polycrystalline monolayer comprised of monocrystal granules, oriented in various directions (Figure 65A(iv, ix))

[4]. The quality and size of crystal monolayers are particle size and shape dependent.

Interestingly, only uniform SU-8 microtiles appeared at the liquid-air interface ultimately forming ordered arrangements, but not the microtiles coated with Au on one side which changed the surface hydrophilicity. After the evaporation of the droplet mixture (~minutes), the polycrystalline monolayer was stably supported on the solid glass surface, allowing it to be inspected by scanning electron microscopy (SEM) and used as a colloidal lithographic mask, as shown below.

231

Figure 65: A) Mechanisms for crystal formation: (i) the particle suspension is deposited on a chloroform droplet; (ii) a stable crystalline monolayer forms at the interface of chloroform and water; (iii) a co-solvent (ethanol) is added to mix the two phases and break the crystal into crystallite fragments; (iv) the crystallite fragments re-emerge assemble along the top of the fluid mixture; and (v) the solvent mixture evaporates in minutes, resulting in a polycrystalline monolayer stably supported on the glass substrate. Images (vi-x) show optical micrographs (i.e. square microtiles) of the corresponding events depicted schematically in (i-v). Image x shows a large area of a crystal monolayer. B) A schematic representation of three strategies that did not result in well-defined crystals is shown. C) A schematic of three strategies that resulted in well-defined crystals is shown.

To further examine the conditions necessary for proper crystal formation, we performed several variations of the droplet-based assembly approach. First, we mixed

232

the particle suspension with chloroform and ethanol prior to dispensing the solution onto the glass substrate (Figure 65B(ii)), which resulted in a disordered array of particles following evaporation of the fluid mixture. Second, we replaced the liquid chloroform with a solid hydrophobic substrate (i.e., polystyrene having a water contact angle ~

95.4°±1.2°); Figure 65B(iii)), which also resulted in a disordered array of particles following the evaporation of the fluid mixture, indicating that the liquid interface serves a critical role in the assembly process. Third, we deposited the co-solvent either immediately after (Figure 65C(i)) or at least 10 seconds after depositing the particle suspension (Figure 65C(ii)). We found that both methods produced well-ordered polycrystalline monolayers, and the quality of the crystals between the two methods was not noticeably different. Finally, we added ethanol to the chloroform drop and added the particle suspension immediately, which also resulted in the formation of well- ordered polycrystalline monolayers (Figure 65C(iii)). We found that this method allowed for higher particle concentrations than the previous methods because the particles remain at the liquid-liquid interface (emulsion step) for only a brief period of time, which avoids the high (and sometimes irreversible) local concentration-induced overlapping and flocculation. This method is also successful without adding surfactant, while small amounts of surfactant generated slightly better crystals.

233

Figure 66: An example of bright field images of crystal monolayers self-assembled using thin circle tiles. 4x image (left) shows the long-range coverage of the self- assembled crystal. 10x images (right) were used for quantification.

It was observed that the shape, aspect ratio and concentration of particles in suspension are all important factors in forming large (mm2), close-packed assemblies.

We analyzed the size of monocrystals (i.e., a singular granular crystallite within a polycrystal) and the surface coverage of polycrystals using both SEM and optical microscopy. Figure 66 shows an example of a long range ordered crystal layer made from circle tiles. Not surprisingly, we found that both hexagonal and circular microtiles assembled into hcp arrangements, whereas square microtiles assembled into cubic close packed (ccp) arrangements, often with an arbitrary shift between rows of particles (see

Figure 67A for representative SEM images and fast Fourier transformations (FFTs) of dried monocrystals (FFTs generated using ImageJ, NIH, USA)). The sizes of the monocrystals were colorized using bright field images and analyzed using Photoshop software (Adobe Systems, Inc.; Figure 67B), and the results showed that the circular

234

microtiles produced the largest monocrystals (mean = 3.34±2.47x104 µm2), which were larger than the monocrystals produced by hexagonal microtiles (mean = 2.59±2.05x104

µm2), while the square microtiles produced the smallest monocrystals (mean =

2.06±1.23x104 µm2; Figure 67C). An apparent reason here is that when the circle tiles re-

assembled at air-liquid interface they have a higher chance to reform close-packed layers

because they do not have angles (one edge), whereas square and hexagon tiles have

multiple edges that place a geometrical restriction reducing the probability of forming

close-packed layers during the re-assembly. We also found that the average surface

coverage of the polycrystals (as measured by the percentage of covered area minus the

percentage of area from voids and crystalline defects) was higher for the assemblies

formed from low aspect ratio microtiles (i.e., 0.2) than the high aspect ratio microtiles

(i.e., 0.9), which was approximately 82±3.95% and 61±6.49%, respectively (Figure 67D).

We found that particles with an aspect ratio close to 1.0 tend to occupy multiple configurations with the same relative expense of energy [426], resulting in crystals displaying more defects and smaller crystallites within the polycrystalline monolayer.

This result corroborates with previous studies as particles with an aspect ratio far from

1.0 tend to occupy less orientational degrees of freedom along a liquid-liquid boundary

(i.e., they tend to orient edgewise along the fluid interface) [427]. Thus, tall tiles with an aspect ratio 0.9 have more faces that can orient at the air at air-liquid interface compared

235

with thin tiles. When thinner tiles have a higher chance to face down a higher crystal size or area is obtained as observed in Figure 67D [426].

Figure 67: A) SEM images (left) and FFTs (right) of assemblies of (i) circular, (ii) hexagonal and (iii) square microtiles with an aspect ratio (height:width) of 0.2 as well as of assemblies of (iv) circular, (v) hexagonal and (vi) square microtiles with an aspect ratio of 0.9 supported on a solid substrate. B) Colorized images of large, polycrystalline monolayers. Analysis of (C) monocrystal size and (D) total surface coverage of the crystals made from the different tiles. The statistically analysis was performed using bright field images.

The difference between different shapes (circle, hexagon, square) is small and could be due to the different meniscus at air-liquid interface. We also found that

236

crystalline monolayers could not form when the initial particle concentration was appreciably low (i.e., < 2x106 particles/mL), as many small crystallites formed separately, or appreciably high (i.e., > 2x108 particles/mL), as particle overlapping and aggregation disrupted the periodicity of the crystal (Figure 68). While we note that the ideal particle concentration for the assembly of large crystalline monolayers is strongly dependent on the size of the particles, we found that the optimal concentration for the 10 µm wide tiles investigated herein is approximately 1x107 to 5x107 particle/mL of water (as measured prior to deposition onto the chloroform droplet). In addition, the volume of the droplets determined the area of the liquid-liquid interface, which directly affected the monolayer size and the number of particles that could be incorporated into the assembly. As such, we note this droplet-based assembly approach can be extended to assemble larger polycrystalline monolayers by dispensing larger single droplets or multiple droplets in parallel within a confined space as long as the ratio of fluid interfacial area (i.e., fluid volume) to particle size is maintained.

237

Figure 68: A) Circular and (B) hexagonal tiles were assembled using different particle concentrations. Particle suspensions of both types, with concentrations of (i) 2x106, (ii) 1x107, (iii) 5x107 and (iv) 2.5x108 particles/mL, were assembled on a chloroform droplet using the methods described in Figure 65C(i) of the manuscript. Scale bars represent 20 µm.

B.4 Conclusions

In summary, we developed a highly translatable approach to rapidly assemble microtiles of various shapes and sizes without special equipment or the use of particle surface treatments. This method of self-assembly overcomes many of the limitations of assembling small tiles (e.g., at least 5 or 10 µm wide) into large monolayer crystals [406,

428, 429]. Although the majority of experimental and theoretical descriptions have been

238

applied to spherical particles similar processes are most likely applicable to anisotropic particles (e.g., particles pinning at interface [429]). There are several advantages of the proposed assembly method over previous techniques, including the: (i) rapid speed of assembly (e.g., occurring in < 120 sec), (ii) large attainable surface coverage areas (mm2 in size that is tunable by droplet size and particle concentration), (iii) facile generation of close-packed monolayers and (iv) potential to assemble a library of particle shapes on solid surfaces for further use (e.g., colloidal lithography) [430]. This demonstration of

“self-assembly on a droplet” is extendable to other systems and types of particles by modifying the solvents and co-solvents used (e.g., methanol or propanol), surfactants

(e.g., with different concentrations or charges) and particle compositions. Furthermore, the use of different particle sizes and shapes opens the possibility of assembling more complex colloidal crystals that are not easily manufactured by standard methods (e.g., microfabrication or other top-down fabrication approaches), which can be easily dehydrated and supported on solid surfaces for a variety of downstream uses, such as in microdevice fabrication.

B.5 Acknowledgements

This chapter contains material from a previously published article: Wang, PY;*

Shields IV, CW;* Zhao, T; Jami, H; López, GP; Kingshott, P. “Rapid self-assembly of shaped microtiles into large, close-packed crystalline monolayers on a solid surface,”

239

Small 2016. 12(2): 1-6. DOI: 10.1002/smll.201503130 (*denotes co-first author). John Wiley and Sons granted permission for reuse of this article in this dissertation.

This work was supported by the National Science Foundation’s (NSF’s) Research

Triangle MRSEC (DMR-1121107), the John Stocker Postdoctoral Research Fellowship

Program through the Scientific Industrial Endowment Fund and the NSF Graduate

Research Fellowship Program (1106401). We gratefully acknowledge Oluwatosin

Omofoye, Jonathan Liu and Crystal E. Owens for their assistance with the fabrication of the particles.

240

References

1. Tomlinson, M.J., et al. (2013) Cell separation: Terminology and practical considerations. Journal of tissue engineering 4, 2041731412472690.

2. Armstrong, A.J., et al. (2011) Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol Cancer Res 9, 997-1007.

3. Wognum, A.W., et al. (2003) Identification and isolation of hematopoietic stem cells. Archives of medical research 34, 461-475.

4. Bischoff, F., et al. (2003) Intact fetal cell isolation from maternal blood: improved isolation using a simple whole blood progenitor cell enrichment approach (RosetteSep). Clin Genet 63, 483-489.

5. Chen, Y., et al. (2014) Rare cell isolation and analysis in microfluidics. Lab on a chip 14, 626-645.

6. Chin, L., et al. (2011) Cancer genomics: from discovery science to personalized medicine. Nature medicine 17, 297-303.

7. Hulett, H.R., et al. (1969) Cell sorting: automated separation of mammalian cells as a function of intracellular fluorescence. Science 166, 747-749.

8. Herzenberg, L.A., et al. (2002) The history and future of the fluorescence activated cell sorter and flow cytometry. Clinical chemistry 48, 1819-1827.

9. Bonner, W.A., et al. (1972) Fluorescence Activated Cell Sorting. Review of Scientific Instruments 43, 404-409.

10. Gossett, D.R., et al. (2010) Label-free cell separation and sorting in microfluidic systems. Anal Bioanal Chem 397, 3249-3267.

11. Piyasena, M.E. and Graves, S.W. (2014) The intersection of flow cytometry with microfluidics and microfabrication. Lab on a Chip 14, 1044-1059.

12. Miltenyi, S., et al. (1990) High gradient magnetic cell separation with MACS. Cytometry 11, 231-238.

241

13. El-Ali, J., et al. (2006) Cells on chips. Nature 442, 403-411.

14. Zhao, M., et al. (2013) An automated high-throughput counting method for screening circulating tumor cells in peripheral blood. Analytical chemistry 85, 2465-2471.

15. Yang, S., et al. (2006) A microfluidic device for continuous, real time blood plasma separation. Lab Chip 6, 871-880.

16. Mernier, G., et al. (2010) Continuous-flow electrical lysis device with integrated control by dielectrophoretic cell sorting. Lab Chip 10, 2077-2082.

17. Sheng, W., et al. (2014) Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip 14, 89- 98.

18. Nan, L., et al. (2014) Emerging microfluidic devices for cell lysis: a review. Lab Chip 14, 1060-1073.

19. Nilsson, J., et al. (2009) Review of cell and particle trapping in microfluidic systems. Analytica chimica acta 649, 141-157.

20. Voldman, J. (2006) Electrical Forces For Microscale Cell Manipulation. Annu Rev Biomed Eng 8, 425-454.

21. Gonzalez, C.F. and Remcho, V.T. (2005) Harnessing dielectric forces for separations of cells, fine particles and macromolecules. Journal of Chromatography A 1079, 59-68.

22. Takahashi, K., et al. (2004) Non-destructive on-chip cell sorting system with real- time microscopic image processing. Journal of nanobiotechnology 2

23. Yao, B., et al. (2004) A microfluidic device based on gravity and electric force driving for flow cytometry and fluorescence activated cell sorting. Lab on a Chip 4, 603-607.

24. Guo, F., et al. (2010) Droplet electric separator microfluidic device for cell sorting. Applied Physics Letters 96, 193701.

242

25. Lenshof, A. and Laurell, T. (2010) Continuous separation of cells and particles in microfluidic systems. Chem. Soc. Rev. 39, 1203-1217.

26. Wang, L., et al. (2007) Dielectrophoresis switching with vertical sidewall electrodes for microfluidic flow cytometry. Lab Chip 7, 1114-1120.

27. Baret, J.C., et al. (2009) Fluorescence-activated droplet sorting (FADS): efficient microfluidic cell sorting based on enzymatic activity. Lab Chip 9, 1850-1858.

28. Agresti, J.J., et al. (2010) Ultrahigh-throughput screening in drop-based microfluidics for directed evolution. Proc Natl Acad Sci U S A 107, 4004-4009.

29. Mazutis, L., et al. (2013) Single-cell analysis and sorting using droplet-based microfluidics. Nature protocols 8, 870-891.

30. Velev, O.D. and Bhatt, K.H. (2006) On-chip micromanipulation and assembly of colloidal particles by electric fields. Soft Matter 2, 738.

31. Fu, A.Y., et al. (1999) A microfabricated fluorescence-activated cell sorter. Nature Biotechnology 17, 1109-1111.

32. Dittrich, P.S. and Schwille, P. (2003) An integrated microfluidic system for reaction, high-sensitivity detection, and sorting of fluorescent cells and particles. Analytical chemistry 75, 5767-5774.

33. Sun, Y., et al. (2007) Design, simulation and experiment of electroosmotic microfluidic chip for cell sorting. Sensors and Actuators A: Physical 133, 340-348.

34. Erlandsson, P.G. and Robinson, N.D. (2011) Electrolysis-reducing electrodes for electrokinetic devices. Electrophoresis 32, 784-790.

35. Valagerahally Puttaswamy, S., et al. (2010) Electrodynamically actuated on-chip flow cytometry with low shear stress for electro-osmosis based sorting using low conductive medium. Microelectronic Engineering 87, 2582-2591.

36. Austin Suthanthiraraj, P.P., et al. (2012) One-dimensional acoustic standing waves in rectangular channels for flow cytometry. Methods 57, 259-271.

37. Kaduchak, G. and Ward, M.D. (2013) Application of acoustic radiation pressure to align cells in a commercial flow cytometer. 045014-045014.

243

38. Lenshof, A., et al. (2012) Acoustofluidics 8: applications of acoustophoresis in continuous flow microsystems. Lab on a chip 12, 1210-1223.

39. Burguillos, M.A., et al. (2013) Microchannel acoustophoresis does not impact survival or function of microglia, leukocytes or tumor cells. PLoS One 8, e64233.

40. Laurell, T., et al. (2007) Chip integrated strategies for acoustic separation and manipulation of cells and particles. Chem Soc Rev 36, 492-506.

41. Johansson, L., et al. (2009) On-chip fluorescence-activated cell sorting by an integrated minaturized ultraonic transducer. Analytical chemistry 81, 5188-5196.

42. Ding, X., et al. (2013) Surface acoustic wave microfluidics. Lab Chip 13, 3626-3649.

43. Cho, S.H., et al. (2010) Human mammalian cell sorting using a highly integrated micro-fabricated fluorescence-activated cell sorter (microFACS). Lab Chip 10, 1567-1573.

44. Gor'kov, L.P. (1962) On the forces acting on a small particle in an acoustical field in an ideal fluid. Soviet Physics Doklady 6, 773-775.

45. Bruus, H. (2012) Acoustofluidics 7: The acoustic radiation force on small particles. Lab on a chip 12, 1014-1021.

46. Augustsson, P. and Laurell, T. (2012) Acoustofluidics 11: Affinity specific extraction and sample decomplexing using continuous flow acoustophoresis. Lab Chip 12, 1742-1752.

47. Goddard, G., et al. (2006) Ultrasonic particle-concentration for sheathless focusing of particles for analysis in a flow cytometer. Cytometry. Part A : the journal of the International Society for Analytical Cytology 69, 66-74.

48. Jakobsson, O., et al. (2014) Acoustic actuated fluorescence activated sorting of microparticles. Lab Chip 14, 1943-1950.

49. Shields IV, C.W., et al. (2014) Elastomeric negative acoustic contrast particles for capture, acoustophoretic transport, and confinement of cells in microfluidic systems. Langmuir 30, 3923-3927.

244

50. Shi, J., et al. (2009) Continuous particle separation in a microfluidic channel via standing surface acoustic waves (SSAW). Lab Chip 9, 3354-3359.

51. Wang, Z. and Zhe, J. (2011) Recent advances in particle and droplet manipulation for lab-on-a-chip devices based on surface acoustic waves. Lab Chip 11, 1280-1285.

52. Lin, S.C., et al. (2012) Surface acoustic wave (SAW) acoustophoresis: now and beyond. Lab on a chip 12, 2766-2770.

53. Ding, X., et al. (2012) Standing surface acoustic wave (SSAW) based multichannel cell sorting. Lab on a chip 12, 4228-4231.

54. Li, S., et al. (2013) An on-chip, multichannel droplet sorter using standing surface acoustic waves. Analytical chemistry 85, 5468-5474.

55. Ding, X., et al. (2012) On-chip manipulation of single microparticles, cells, and organisms using surface acoustic waves. Proceedings of the National Academy of Sciences of the United States of America 109, 11105-11109.

56. Franke, T., et al. (2010) Surface acoustic wave actuated cell sorting (SAWACS). Lab Chip 10, 789-794.

57. Schmid, L., et al. (2014) Sorting drops and cells with acoustics: acoustic microfluidic fluorescence-activated cell sorter. Lab on a chip 14, 3710-3718.

58. Jonas, A. and Zemanek, P. (2008) Light at work: the use of optical forces for particle manipulation, sorting, and analysis. Electrophoresis 29, 4813-4851.

59. Neuman, K.C. and Nagy, A. (2008) Single-molecule force spectroscopy: optical tweezers, magnetic tweezers and atomic force microscopy. Nature methods 5, 491- 505.

60. Moffitt, J.R., et al. (2008) Recent advances in optical tweezers. Annual review of biochemistry 77, 205-228.

61. Ashkin, A. (1970) Acceleration and Trapping of Particles by Radiation Pressure. Physical Review Letters 24, 156-159.

62. Buican, T., et al. (1987) Automated single-cell manipulation and sorting by light trapping. Applied Optics 26, 5311-5316.

245

63. Applegate, R.W., Jr., et al. (2004) Optical trapping manipulation and sorting of cells and colloids in microfluidic systems with diode laser bars. Optics Express 12, 4390-4398.

64. Wang, M.M., et al. (2005) Microfluidic sorting of mammalian cells by optical force switching. Nat Biotechnol 23, 83-87.

65. Applegate, R.W., Jr., et al. (2006) Microfluidic sorting system based on optical waveguide integration and diode laser bar trapping. Lab Chip 6, 422-426.

66. Perroud, T.D., et al. (2008) Microfluidic-based cell sorting of Francisella tularensis infected macrophages using optical forces. Analytical Chemistry 90, 6365-6372.

67. Wang, X., et al. (2011) Enhanced cell sorting and manipulation with combined optical tweezer and microfluidic chip technologies. Lab Chip 11, 3656-3662.

68. Wu, T.H., et al. (2012) Pulsed laser triggered high speed microfluidic fluorescence activated cell sorter. Lab on a chip 12, 1378-1383.

69. Chen, Y., et al. (2014) Pulsed laser activated cell sorting with three dimensional sheathless inertial focusing. Small 10, 1746-1751.

70. Kruger, J., et al. (2002) Development of a microfluidic device for fluorescence activated cell sorting. Journal of Micromechanics and Microengineering 12, 486-494.

71. Fu, A.Y., et al. (2002) An Integrated Microfabricated Cell Sorter. Analytical Chemistry 74, 2451-2457.

72. Wolff, A., et al. (2003) Integrating advanced functionality in a microfabricated high-throughput fluorescent-activated cell sorter. Lab Chip 3, 22-27.

73. Chen, P., et al. (2010) Hydrodynamic gating valve for microfluidic fluorescence- activated cell sorting. Analytica chimica acta 663, 1-6.

74. Ho, C.T., et al. (2005) Micromachined electrochemical T-switches for cell sorting applications. Lab Chip 5, 1248-1258.

246

75. Shirasaki, Y., et al. (2008) On-chip cell sorting system using laser-induced heating of a thermoreversible gelation polymer to control flow Analytical chemistry 78, 695-701.

76. Zborowski, M. and Chalmers, J.J. (2011) Rare cell separation and analysis by magnetic sorting. Analytical chemistry 83, 8050-8056.

77. Gao, Y., et al. (2007) Magnetophoresis of nonmagnetic particles in ferrofluids. J. Phys. Chem. C 111, 10785-10791.

78. Pamme, N. and Wilhelm, C. (2006) Continuous sorting of magnetic cells via on- chip free-flow magnetophoresis. Lab Chip 6, 974-980.

79. Xia, N., et al. (2006) Combined microfluidic-micromagnetic separation of living cells in continuous flow. Biomed Microdevices 8, 299-308.

80. Lai, J.J., et al. (2009) Dynamic bioprocessing and microfluidic transport control with smart magnetic nanoparticles in laminar-flow devices. Lab Chip 9, 1997- 2002.

81. Adams, J.D., et al. (2008) Multitarget magnetic activated cell sorter. Proceedings of the National Academy of Sciences of the United States of America 105, 18165-18170.

82. Carr, C., et al. (2009) Design, fabrication and demonstration of a magnetophoresis chamber with 25 output fractions. J Magn Magn Mater 321, 1440-1445.

83. Yu, M., et al. (2011) Circulating tumor cells: approaches to isolation and characterization. The Journal of cell biology 192, 373-382.

84. Hoshino, K., et al. (2011) Microchip-based immunomagnetic detection of circulating tumor cells. Lab Chip 11, 3449-3457.

85. Hoshino, K., et al. (2012) Computational analysis of microfluidic immunomagnetic rare cell separation from a particulate blood flow. Analytical chemistry 84, 4292-4299.

86. Darabi, J. and Guo, C. (2013) On-chip magnetophoretic isolation of CD4 + T cells from blood. Biomicrofluidics 7, 54106.

247

87. Kang, J.H., et al. (2012) A combined micromagnetic-microfluidic device for rapid capture and culture of rare circulating tumor cells. Lab Chip 12, 2175-2181.

88. Shields IV, C.W., et al. (2014) Magnetographic array for the capture and enumeration of single cells and cell pairs. Biomicrofluidics 8, 041101.

89. Ozkumur, E., et al. (2013) Inertial focusing for tumor antigen-dependent and - independent sorting of rare circulating tumor cells. Sci Transl Med 5, 179ra147.

90. Chen, A., et al. (2013) On-chip magnetic separation and encapsulation of cells in droplets. Lab Chip 13, 1172-1181.

91. Karabacak, N.M., et al. (2014) Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nature protocols 9, 694-710.

92. Cushing, K.W., et al. (2013) Elastomeric negative acoustic contrast particles for affinity capture assays. Anal Chem 85, 2208-2215.

93. Johnson, L.M., et al. (2013) Elastomeric microparticles for acoustic mediated bioseparations. Journal of nanobiotechnology 11, 22.

94. Shields IV, C.W., et al. (2014) Nucleation and growth synthesis of siloxane gels to form functional, monodisperse, and acoustically programmable particles. Angew Chem Int Ed Engl 53, 8070-8073.

95. Lenshof, A., et al. (2014) Efficient Purification of CD41 Lymphocytes from Peripheral Blood Progenitor Cell Products Using Affinity Bead Acoustophoresis. Cytometry. Part A : the journal of the International Society for Analytical Cytology 85, 933-941.

96. Hu, X., et al. (2005) Marker-specific sorting of rare cells using dielectrophoresis. Proceedings of the National Academy of Sciences of the United States of America 102, 15757-15761.

97. Kim, U., et al. (2008) Multitarget dielectrophoresis activated cell sorter. Analytical chemistry 80, 8656-8661.

98. Kim, U. and Soh, H.T. (2009) Simultaneous sorting of multiple bacterial targets using integrated dielectrophoretic-magnetic activated cell sorter. Lab Chip 9, 2313-2318.

248

99. Cheng, I.F., et al. (2007) An integrated dielectrophoretic chip for continuous bioparticle filtering, focusing, sorting, trapping, and detecting. Biomicrofluidics 1, 021503.

100. Coulter, W. (1957) High speed automatic blood cell counter and cell size analyzer. Proc Natl Electron Conf 12, 1034-1040.

101. Fulwyler, M. (1965) Electronic Separation of Biological Cells by Volume. Science 150, 910-911.

102. Petersson, F., et al. (2004) Separation of lipids from blood utilizing ultrasonic standing waves in microfluidic channels. The Analyst 129, 938-943.

103. Petersson, F., et al. (2005) Continuous separation of lipid particles from erythrocytes by means of laminar flow and acoustic standing wave forces. Lab Chip 5, 20-22.

104. Petersson, F., et al. (2007) Free Flow Acoustophoresis: Microfluidic-Based Mode of Particle and Cell Separation. Anal Chem 79, 5117-5123.

105. Dykes, J., et al. (2011) Efficient removal of platelets from peripheral blood progenitor cell products using a novel micro-chip based acoustophoretic platform. PloS one 6, e23074.

106. Augustsson, P., et al. (2012) Microfluidic, label-free enrichment of prostate cancer cells in blood based on acoustophoresis. Anal. Chem. 84, 7954-7962.

107. Yang, A.H. and Soh, H.T. (2012) Acoustophoretic sorting of viable mammalian cells in a microfluidic device. Analytical chemistry 84, 10756-10762.

108. Fong, E.J., et al. (2014) Acoustic focusing with engineered node locations for high- performance microfluidic particle separation. The Analyst 139, 1192-1200.

109. Nam, J., et al. (2011) Separation of platelets from whole blood using standing surface acoustic waves in a microchannel. Lab Chip 11, 3361-3364.

110. Ding, X., et al. (2014) Cell separation using tilted-angle standing surface acoustic waves. Proceedings of the National Academy of Sciences of the United States of America 111, 12992-12997.

249

111. Li, S., et al. (2014) Standing surface acoustic wave (SSAW)-based cell washing. Lab Chip 15, 331-338.

112. Huang, Y., et al. (2002) Dielectrophoretic cell separation and gene expression profiling on microelectronic chip arrays. Analytical chemistry 74, 3362-3371.

113. Cummings, E.B. and Singh, A.K. (2003) Dielectrophoresis in microchips containing arrays of insulating posts- theoretical and experimental results. Analytical Chemistry 75, 4724-4731.

114. Barrett, L.M., et al. (2005) Dielectrophoretic manipulation of particles and cells using insulating ridges in faceted prism microchannels. Analytical chemistry 77, 6798-6804.

115. Vahey, M.D. and Voldman, J. (2008) An equilibrium method for continuous-flow cell sorting using dielectrophoresis. Analytical Chemistry 80, 3135-3143.

116. Roda, B., et al. (2009) Field-flow fractionation in bioanalysis: A review of recent trends. Analytica chimica acta 635, 132-143.

117. Giddings, J.C. (1993) Field-flow fractionation: analysis of macromolecular, colloidal, and particulate materials. Science 260, 1456-1465.

118. Giddings, J.C. (1985) A system based on split-flow lateral-transport thin (SPLITT) separation cells for rapid and continuous particle fractionation. Separation Science and Technology 20, 749-768.

119. Vykoukal, J., et al. (2008) Enrichment of putative stem cells from adipose tissue using dielectrophoretic field-flow fractionation. Lab Chip 8, 1386-1393.

120. Cui, H.H., et al. (2009) Separation of particles by pulsed dielectrophoresis. Lab Chip 9, 2306-2312.

121. Karimi, A., et al. (2013) Hydrodynamic mechanisms of cell and particle trapping in microfluidics. Biomicrofluidics 7, 21501.

122. Doh, I. and Cho, Y.-H. (2005) A continuous cell separation chip using hydrodynamic dielectrophoresis (DEP) process. Sensors and Actuators A: Physical 121, 59-65.

250

123. Moon, H.S., et al. (2011) Continuous separation of breast cancer cells from blood samples using multi-orifice flow fractionation (MOFF) and dielectrophoresis (DEP). Lab Chip 11, 1118-1125.

124. Shim, S., et al. (2013) Antibody-independent isolation of circulating tumor cells by continuous-flow dielectrophoresis. Biomicrofluidics 7, 11807.

125. Kim, U., et al. (2007) Selection of mammalian cells based on their cell-cycle phase using dielectrophoresis. Proceedings of the National Academy of Sciences of the United States of America 104, 20708-20712.

126. Valero, A., et al. (2010) A miniaturized continuous dielectrophoretic cell sorter and its applications. Biomicrofluidics 4

127. Sun, J., et al. (2012) Simultaneous on-chip DC dielectrophoretic cell separation and quantitative separation performance characterization. Analytical chemistry 84, 2017-2024.

128. Toner, M. and Irimia, D. (2005) Blood-on-a-chip. Annual review of biomedical engineering 7, 77-103.

129. Hou, H.W., et al. (2011) Microfluidic Devices for Blood Fractionation. Micromachines 2, 319-343.

130. Melville, D., et al. (1975) Direct magnetic separation of red cells from whole blood. Nature 255, 706.

131. Zborowski, M., et al. (2003) Red blood cell magnetophoresis. Biophysical Journal 84, 2638-2645.

132. Han, K.-H. and Bruno Frazier, A. (2004) Continuous magnetophoretic separation of blood cells in microdevice format. Journal of Applied Physics 96, 5797.

133. Han, K.-H. and Frazier, A.B. (2005) Diamagnetic capture mode magnetophoretic microseparator for blood cells. Journal of Microelectromechanical Systems 14, 1422- 1431.

134. Han, K.H. and Frazier, A.B. (2006) Paramagnetic capture mode magnetophoretic microseparator for high efficiency blood cell separations. Lab Chip 6, 265-273.

251

135. Jung, J. and Han, K.-H. (2008) Lateral-driven continuous magnetophoretic separation of blood cells. Applied Physics Letters 93, 223902.

136. Qu, B.Y., et al. (2008) A glass microfluidic chip for continuous blood cell sorting by a magnetic gradient without labeling. Analytical and bioanalytical chemistry 392, 1317-1324.

137. Seo, H.-K., et al. (2010) Hybrid cell sorters for on-chip cell separation by hydrodynamics and magnetophoresis. Journal of Micromechanics and Microengineering 20, 095019.

138. Furlani, E.P. (2007) Magnetophoretic separation of blood cells at the microscale. Journal of Physics D: Applied Physics 40, 1313-1319.

139. Robert, D., et al. (2011) Cell sorting by endocytotic capacity in a microfluidic magnetophoresis device. Lab Chip 11, 1902-1910.

140. Zhu, T., et al. (2012) Continuous-flow ferrohydrodynamic sorting of particles and cells in microfluidic devices. Microfluidics and Nanofluidics 13, 645-654.

141. Zeng, J., et al. (2013) Magnetic separation of particles and cells in ferrofluid flow through a straight microchannel using two offset magnets. Journal of Magnetism and Magnetic Materials 346, 118-123.

142. Shen, F., et al. (2012) Label-free cell separation using a tunable magnetophoretic repulsion force. Analytical chemistry 84, 3075-3081.

143. Hoi, S.K., et al. (2009) Microfluidic sorting system based on optical force switching. Applied Physics B 97, 859-865.

144. Lau, A.Y., et al. (2008) An integrated optofluidic platform for Raman-activated cell sorting. Lab Chip 8, 1116-1120.

145. MacDonald, M.P., et al. (2003) Microfluidic sorting in an optical lattice. Nature 426, 421-424.

146. Wu, M.C. (2011) Optoelectronic tweezers. Nature Photonics 5, 322-324.

252

147. Chiou, P.Y., et al. (2005) Massively parallel manipulation of single cells and microparticles using optical images. Nature 436, 370-372.

148. Huang, S.B., et al. (2012) A New Optically-Induced Dielectrophoretic (ODEP) Force-Based Scheme for Effective Cell Sorting. International Journal of Electrochemical Science 7, 12656-12667.

149. Huang, S.B., et al. (2013) High-purity and label-free isolation of circulating tumor cells (CTCs) in a microfluidic platform by using optically-induced- dielectrophoretic (ODEP) force. Lab Chip 13, 1371-1383.

150. Ohta, A.T., et al. (2007) Optically controlled cell discrimination and trapping using optoelectronic tweezers. Journal of Selected Topics in Quantum Electronics - IEEE 13, 235-243.

151. Shah, G.J., et al. (2009) EWOD-driven droplet microfluidic device integrated with optoelectronic tweezers as an automated platform for cellular isolation and analysis. Lab on a chip 9, 1732-1739.

152. Huang, K.W., et al. (2013) Microfluidic integrated optoelectronic tweezers for single-cell preparation and analysis. Lab Chip 13, 3721-3727.

153. Sollier, E., et al. (2009) Passive microfluidic devices for plasma extraction from whole human blood. Sensors and Actuators B: Chemical 141, 617-624.

154. Tsutsui, H. and Ho, C.M. (2009) Cell Separation by Non-Inertial Force Fields in Microfluidic Systems. Mechanics research communications 36, 92-103.

155. Di Carlo, D. (2009) Inertial microfluidics. Lab on a chip 9, 3038-3046.

156. Di Carlo, D., et al. (2007) Continuous inertial focusing, ordering, and separation of particles in microchannels. Proceedings of the National Academy of Sciences of the United States of America 104, 18892-18897.

157. Di Carlo, D., et al. (2008) Equilibrium separation and filtration of particles using differential inertial focusing. Analytical Chemistry 80, 2204-2211.

158. Oakey, J., et al. (2010) Particle focusing in staged inertial microfluidic devices for flow cytometry. Analytical chemistry 82, 3862-3867.

253

159. Russom, A., et al. (2009) Differential inertial focusing of particles in curved low- aspect-ratio microchannels. New J Phys 11, 75025.

160. Kuntaegowdanahalli, S.S., et al. (2009) Inertial microfluidics for continuous particle separation in spiral microchannels. Lab Chip 9, 2973-2980.

161. Hou, H.W., et al. (2013) Isolation and retrieval of circulating tumor cells using centrifugal forces. Scientific Reports 3, 1-8.

162. Nivedita, N. and Papautsky, I. (2013) Continuous separation of blood cells in spiral microfluidic devices. Biomicrofluidics 7, 54101.

163. Guan, G., et al. (2013) Spiral microchannel with rectangular and trapezoidal cross-sections for size based particle separation. Sci Rep 3, 1475.

164. Warkiani, M.E., et al. (2014) Slanted spiral microfluidics for the ultra-fast, label- free isolation of circulating tumor cells. Lab Chip 14, 128-137.

165. Parichehreh, V., et al. (2013) Microfluidic inertia enhanced phase partitioning for enriching nucleated cell populations in blood. Lab Chip 13, 892-900.

166. Yamada, M., et al. (2004) Pinched flow fractionation: continuous size separation of particles utilizing a laminar flow profile in a pinched microchannel. Analytical Chemistry 76, 5465-5471.

167. Takagi, J., et al. (2005) Continuous particle separation in a microchannel having asymmetrically arranged multiple branches. Lab Chip 5, 778-784.

168. Huh, D., et al. (2007) Gravity-driven microfluidic particle sorting device with hydrodynamic separation amplification. Analytical Chemistry 79, 1369-1376.

169. Zhang, X., et al. (2006) Continuous flow separation of particles within an asymmetric microfluidic device. Lab Chip 6, 561-566.

170. Wu, Z., et al. (2009) Soft inertial microfluidics for high throughput separation of bacteria from human blood cells. Lab on a chip 9, 1193-1199.

171. Wu, Z., et al. (2007) Microfluidic continuous particle/cell separation via electroosmotic-flow-tuned hydrodynamic spreading. Journal of Micromechanics and Microengineering 17, 1992-1999.

254

172. Mach, A.J. and Di Carlo, D. (2010) Continuous scalable blood filtration device using inertial microfluidics. Biotechnology and bioengineering 107, 302-311.

173. Bhagat, A.A.S., et al. (2008) Enhanced particle filtration in straight microchannels using shear-modulated inertial migration. Physics of Fluids 20, 101702.

174. Zhou, J., et al. (2013) Modulation of aspect ratio for complete separation in an inertial microfluidic channel. Lab Chip 13, 1919-1929.

175. Hur, S.C., et al. (2011) Deformability-based cell classification and enrichment using inertial microfluidics. Lab Chip 11, 912-920.

176. Geislinger, T.M. and Franke, T. (2013) Sorting of circulating tumor cells (MV3- melanoma) and red blood cells using non-inertial lift. Biomicrofluidics 7, 44120.

177. Geislinger, T.M., et al. (2012) Separation of blood cells using hydrodynamic lift. Applied Physics Letters 100, 183701.

178. Beech, J.P., et al. (2012) Sorting cells by size, shape and deformability. Lab Chip 12, 1048-1051.

179. Huang, L.R., et al. (2004) Continuous particle separation through deterministic lateral displacement. Science 304, 987-990.

180. Inglis, D.W., et al. (2006) Critical particle size for fractionation by deterministic lateral displacement. Lab Chip 6, 655-658.

181. Davis, J.A., et al. (2006) Deterministic hydrodynamics: taking blood apart. Proceedings of the National Academy of Sciences of the United States of America 103, 14779-14784.

182. Huang, R., et al. (2008) A microfluidics approach for the isolation of nucleated red blood cells (NRBCs) from the peripheral blood of pregnant women. Prenatal diagnosis 28, 892-899.

183. Liu, Z., et al. (2013) Rapid isolation of cancer cells using microfluidic deterministic lateral displacement structure. Biomicrofluidics 7, 11801.

255

184. Choi, S. and Park, J.K. (2007) Continuous hydrophoretic separation and sizing of microparticles using slanted obstacles in a microchannel. Lab Chip 7, 890-897.

185. Choi, S., et al. (2007) Continuous blood cell separation by hydrophoretic filtration. Lab Chip 7, 1532-1538.

186. Hsu, C.H., et al. (2008) Microvortex for focusing, guiding and sorting of particles. Lab Chip 8, 2128-2134.

187. Stott, S.L., et al. (2010) Isolation of circulating tumor cells using a microvortex- generating herringbone-chip. Proceedings of the National Academy of Sciences of the United States of America 107, 18392-18397.

188. Sollier, E., et al. (2014) Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip 14, 63-77.

189. Hyun, K.A., et al. (2013) Microfluidic flow fractionation device for label-free isolation of circulating tumor cells (CTCs) from breast cancer patients. Biosensors & bioelectronics 40, 206-212.

190. Mohamed, H., et al. (2007) Biochip for separating fetal cells from maternal circulation. Journal of chromatography. A 1162, 187-192.

191. McFaul, S.M., et al. (2012) Cell separation based on size and deformability using microfluidic funnel ratchets. Lab Chip 12, 2369-2376.

192. Preira, P., et al. (2013) Passive circulating cell sorting by deformability using a microfluidic gradual filter. Lab Chip 13, 161-170.

193. Ji, H.M., et al. (2008) Silicon-based microfilters for whole blood cell separation. Biomedical microdevices 10, 251-257.

194. Earhart, C.M., et al. (2014) Isolation and mutational analysis of circulating tumor cells from lung cancer patients with magnetic sifters and biochips. Lab Chip 14, 78-88.

195. Murthy, S.K., et al. (2006) Size-based microfluidic enrichment of neonatal rat cardiac cell populations. Biomedical microdevices 8, 231-237.

256

196. Sethu, P., et al. (2006) Microfluidic diffusive filter for apheresis (leukapheresis). Lab Chip 6, 83-89.

197. VanDelinder, V. and Groisman, A. (2006) Separation of Plasma from Whole Human Blood in a Continuous Cross-Flow in a Molded Microfluidic Device. Analytical Chemistry 78, 3765-3771.

198. VanDelinder, V. and Groisman, A. (2007) Perfusion in microfluidic cross-flow: separation of white blood cells from whole blood and exhange of medium in a continuous flow. Analytical Chemistry 79, 2023-2030.

199. Chen, X., et al. (2008) Microfluidic chip for blood cell separation and collection based on crossflow filtration. Sensors and Actuators B: Chemical 130, 216-221.

200. Chen, X., et al. (2007) Continuous flow microfluidic device for cell separation, cell lysis and DNA purification. Analytica chimica acta 584, 237-243.

201. Yamada, M., et al. (2007) Microfluidic devices for size-dependent separation of liver cells. Biomed Microdevices 9, 637-645.

202. Yamada, M. and Seki, M. (2005) Hydrodynamic filtration for on-chip particle concentration and classification utilizing microfluidics. Lab Chip 5, 1233-1239.

203. Yamada, M. and Seki, M. (2006) Microfluidic particle sorter employing flow splitting and recombining. Analytical chemistry 78, 1357-1362.

204. Mizuno, M., et al. (2013) Magnetophoresis-integrated hydrodynamic filtration system for size- and surface marker-based two-dimensional cell sorting. Analytical chemistry 85, 7666-7673.

205. Didar, T.F. and Tabrizian, M. (2010) Adhesion based detection, sorting and enrichment of cells in microfluidic Lab-on-Chip devices. Lab Chip 10, 3043-3053.

206. Lee, C.H., et al. (2011) Examining the lateral displacement of HL60 cells rolling on asymmetric P-selectin patterns. Langmuir 27, 240-249.

207. Bose, S., et al. (2013) Affinity flow fractionation of cells via transient interactions with asymmetric molecular patterns. Sci Rep 3, 2329.

257

208. Choi, S., et al. (2012) Cell sorting by deterministic cell rolling. Lab Chip 12, 1427- 1430.

209. Choi, S., et al. (2014) A cell rolling cytometer reveals the correlation between mesenchymal stem cell dynamic adhesion and differentiation state. Lab Chip 14, 161-166.

210. Baran, M.M., et al. (1982) Cell sorting using a universally applicable affinity chromatography matrix: Solid-phase anti-fluorescein isothiocyanate antibody. Journal of Immunological Methods 53, 321-334.

211. Hertz, C.M., et al. (1984) Use of cell affinity chromatography for separation of lymphocyte subpopulations. Biotechnology and Bioengineering 27, 603-612.

212. Kumar, A. and Srivastava, A. (2010) Cell separation using cryogel-based affinity chromatography. Nature protocols 5, 1737-1747.

213. Cheng, X., et al. (2007) A microfluidic device for practical label-free CD4(+) T cell counting of HIV-infected subjects. Lab Chip 7, 170-178.

214. Pullagurla, S.R., et al. (2014) Parallel affinity-based isolation of leukocyte subsets using microfluidics: application for stroke diagnosis. Analytical chemistry 86, 4058-4065.

215. Li, P., et al. (2012) Multiparameter cell affinity chromatography: separation and analysis in a single microfluidic channel. Analytical chemistry 84, 8140-8148.

216. Hyun, K.A., et al. (2013) Negative enrichment of circulating tumor cells using a geometrically activated surface interaction chip. Analytical chemistry 85, 4439- 4445.

217. Chen, W., et al. (2013) Nanoroughened surfaces for efficient capture of circulating tumor cells without using capture antibodies. ACS Nano 7, 566-575.

218. Singh, A., et al. (2013) Adhesion strength-based, label-free isolation of human pluripotent stem cells. Nature methods 10, 438-444.

219. Li, P., et al. (2013) Probing circulating tumor cells in microfluidics. Lab Chip 13, 602-609.

258

220. Nagrath, S., et al. (2007) Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 450, 1235-1239.

221. Mahara, A. and Yamaoka, T. (2010) Antibody-immobilized column for quick cell separation based on cell rolling. Biotechnology progress 26, 441-447.

222. Yu, X., et al. (2013) Magneto-controllable capture and release of cancer cells by using a micropillar device decorated with graphite oxide-coated magnetic nanoparticles. Small 9, 3895-3901.

223. Saliba, A.E., et al. (2010) Microfluidic sorting and multimodal typing of cancer cells in self-assembled magnetic arrays. Proceedings of the National Academy of Sciences of the United States of America 107, 14524-14529.

224. Yoon, H.J., et al. (2013) Sensitive capture of circulating tumour cells by functionalized graphene oxide nanosheets. Nature nanotechnology 8, 735-741.

225. Mittal, S., et al. (2012) Antibody-functionalized fluid-permeable surfaces for rolling cell capture at high flow rates. Biophysical journal 102, 721-730.

226. Xu, Y., et al. (2009) Aptamer-based microfluidic device for enrichment, sorrting, and detection of multiple cancer cells. Analytical chemistry 81, 7436-7442.

227. Sheng, W., et al. (2012) Aptamer-enabled efficient isolation of cancer cells from whole blood using a microfluidic device. Analytical chemistry 84, 4199-4206.

228. Zhao, W., et al. (2012) Bioinspired multivalent DNA network for capture and release of cells. Proceedings of the National Academy of Sciences of the United States of America 109, 19626-19631.

229. Kamentsky, L.A. and Melamed, M.R. (1967) Spectrophotometric Cell Sorter. Science 156, 1364-1365.

230. Tan, S.J., et al. (2009) Microdevice for the isolation and enumeration of cancer cells from blood. Biomedical microdevices 11, 883-892.

231. Tan, S.J., et al. (2010) Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients. Biosensors & bioelectronics 26, 1701-1705.

259

232. Winer-Jones, J.P., et al. (2014) Circulating tumor cells: clinically relevant molecular access based on a novel CTC flow cell. PLoS One 9, e86717.

233. Kogot, J.M., et al. (2011) Screening of peptide libraries against protective antigen of Bacillus anthracis in a disposable microfluidic cartridge. PLoS One 6, e26925.

234. Hulspas, R., et al. (2014) Purification of regulatory T cells with the use of a fully enclosed high-speed microfluidic system. Cytotherapy 16, 1384-1389.

235. Vercruysse, D., et al. (2014) A high-speed miniaturized cell sorter with lens-free imaging and thermal bubble based jet flow sorting. International Conference on Miniaturized Systems for Chemistry and Life Sciences, 382-384.

236. Foster, J., et al. Owl Biomedical, Inc., Innovative Micro Technology, Inc. Cartridge for MEMS particle sorting system, US 20120190105 A1.

237. Stibal, M., et al. (2015) Microbial abundance in surface ice on the Greenland Ice Sheet. Frontiers in microbiology 6, 225.

238. Chen, C.H., et al. (2011) Specific sorting of single bacterial cells with microfabricated fluorescence-activated cell sorting and tyramide signal amplification fluorescence in situ hybridization. Analytical chemistry 83, 7269- 7275.

239. Chen, C.H., et al. (2009) Microfluidic cell sorter with integrated piezoelectric actuator. Biomed Microdevices 11, 1223-1231.

240. Sackmann, E.K., et al. (2014) The present and future role of microfluidics in biomedical research. Nature 507, 181-189.

241. Grad, M., et al. (2013) A simple add-on microfluidic appliance for accurately sorting small populations of cells with high fidelity. J Micromech Microeng 23

242. Plouffe, B.D. and Murthy, S.K. (2014) Perspective on microfluidic cell separation: a solved problem? Analytical chemistry 86, 11481-11488.

243. Lester, R. (1981) Secrecy, patents and non-proliferation. In Bulletin of the Atomic Scientists May 1981, pp. 4, Educational Foundation for Nuclear Science, Inc.

260

244. Chin, C.D., et al. (2012) Commercialization of microfluidic point-of-care diagnostic devices. Lab on a chip 12, 2118-2134.

245. Yetisen, A.K., et al. (2015) Entrepreneurship. Lab on a chip 15, 3638-3660.

246. Markets, M.a. (2014) Cell Isolation/ Cell Separation Market by Product (Regent, Media, Bead, Centrifuge), Cell Type (human, stem cell, animal), Technique (Filtration, Surface Market), Application (Research, IVD) & End user (Hospital, Biotechnology)- Forecast to 2019.

247. Caicedo, H.H. and Brady, S.T. (2016) Microfluidics: The Challenge Is to Bridge the Gap Instead of Looking for a 'Killer App'. Trends Biotechnol 34, 1-3.

248. Kuo, J.S. and Chiu, D.T. (2011) Disposable microfluidic substrates: transitioning from the research laboratory into the clinic. Lab on a chip 11, 2656-2665.

249. Becker, H. (2009) Chips, money, industry, education and the "killer application". Lab on a chip 9, 1659-1660.

250. Au, A.K., et al. (2015) 3D-printed microfluidic automation. Lab Chip 15, 1934-1941.

251. Pawell, R.S., et al. (2013) Manufacturing and wetting low-cost microfluidic cell separation devices. Biomicrofluidics 7, 56501.

252. Lee, C.Y., et al. (2011) Microfluidic mixing: a review. International journal of molecular sciences 12, 3263-3287.

253. Shapiro, C. (2001) Navigating the patent thicket: Cross licenses, patent pools, and standard setting. In Innovation Policy and the Economy (Jaffe, A.B., et al., eds), pp. 119-150, MIT Press.

254. Duncanson, W.J., et al. (2012) Microfluidic synthesis of advanced microparticles for encapsulation and controlled release. Lab Chip 12, 2135-2145.

255. Nash, M.A., et al. (2012) Multiplexed enrichment and detection of malarial biomarkers using a stimuli-responsive iron oxide and gold nanoparticle reagent system. ACS Nano 6, 6776-6785.

256. Xia, Y., et al. (2000) Monodisperse colloidal spheres- old materials with new applications. Adv. Mat. 12, 693-713.

261

257. Piyasena, M.E., et al. (2012) Multinode acoustic focusing for parallel flow cytometry. Anal. Chem. 84, 1831-1839.

258. Bogush, G.H. and Zukoski IV, C.F. (1990) Studies of the kinetics of the precipitation of uniform silica particles through the hydrolysis and condensation of silicon alkoxides. J. Colloid Interface Sci. 142, 1-18.

259. Stöber, W., et al. (1968) Controlled growth of monodisperse silica spheres in the micron size range. J. Colloid Interface Sci. 26, 62-69.

260. Obey, T.M. and Vincent, B. (1994) Novel monodisperse 'silicone oil' water emulsions. J. Colloid Interface Sci. 163

261. Goller, M.I., et al. (1997) Inorganic “silicone oil” microgels. Colloids Surf., A 123– 124, 183-193.

262. Miller, C.R., et al. (2005) Functionalized organosilica microspheres via a novel emulsion-based route. Langmuir 21, 9733-9740.

263. Vogel, R., et al. (2007) Fluorescent organosilica micro- and nanoparticles with controllable size. J. Colloid Interface Sci. 310, 144-150.

264. Vogel, R., et al. (2011) Quantitative sizing of nano/microparticles with a tunable elastomeric pore sensor. Anal. Chem. 83, 3499-3506.

265. Kozak, D., et al. (2012) Simultaneous size and zeta potential measurements of individual nanoparticles in dispersion using size-tunable pore sensors. ACS Nano 6, 6990-6997.

266. Dayton, P.A., et al. (2002) The magnitude of radiation force on ultrasound contrast agents. J. Acoust. Soc. Am. 112, 2183.

267. Doinikov, A.A. (2003) Acoustic radiation forces: Classical theory and recent advances. Recent Res Devel 1, 39-67.

268. Woodside, S.M., et al. (1997) Measurement of Ultrasonic Forces for Particle- Liquid Separations. AIChE Journal 43, 1727-1736.

262

269. Barnkob, R., et al. (2010) Measuring the local pressure amplitude in microchannel acoustophoresis. Lab Chip 10, 563-570.

270. Mott, P.H., et al. (2008) The bulk modulus and Poisson's ratio of “incompressible” materials. Journal of Sound and Vibration 312, 572-575.

271. Guo, F., et al. (2015) Controlling cell–cell interactions using surface acoustic waves. Proceedings of the National Academy of Sciences of the United States of America 112, 43-48.

272. Li, P., et al. (2015) Acoustic separation of circulating tumor cells. Proceedings of the National Academy of Sciences of the United States of America 112, 4970-4975.

273. Gao, L., et al. (2015) Two-dimensional spatial manipulation of microparticles in continuous flows in acoustofluidic systems. Biomicrofluidics 9, 014105.

274. Shields IV, C.W., et al. (2015) Microfluidic cell sorting: a review of the advances in the separation of cells from debulking to rare cell isolation. Lab Chip 15, 1230- 1249.

275. Goddard, G.R., et al. (2007) Analytical Performance of an Ultrasonic Particle Focusing Flow Cytometer. Anal Chem 79, 8740-8746.

276. Goddard, G. and Kaduchak, G. (2005) Ultrasonic particle concentration in a line- driven cylindrical tube. The Journal of the Acoustical Society of America 117, 3440.

277. Carugo, D., et al. (2014) A thin-reflector microfluidic resonator for continuous- flow concentration of microorganisms: a new approach to water quality analysis using acoustofluidics. Lab on a chip 14, 3830-3842.

278. Wiklund, M., et al. (2001) Ultrasonic trapping in capillaries for trace-amount biomedical analysis. Journal of Applied Physics 90, 421.

279. Shields IV, C.W., et al. (2013) Field-directed assembly of patchy anisotropic microparticles with defined shape. Soft Matter 9, 9219.

280. Yeom, J., et al. (2005) Maximum achievable aspect ratio in deep reactive ion etching of silicon due to aspect ratio dependent transport and the microloading effect. Journal of Vacuum Science & Technology B: Microelectronics and Nanometer Structures 23, 2319.

263

281. McDonald, J.C. and Whitesides, G.M. (2002) Poly(dimethylsiloxane) as a material for fabricating microfluidic devices. Accounts of Chemical Research 35, 491-499.

282. Golden, J.P., et al. (2012) Hydrodynamic focusing--a versatile tool. Analytical and bioanalytical chemistry 402, 325-335.

283. Hejazian, M., et al. (2015) Lab on a chip for continuous-flow magnetic cell separation. Lab on a chip 15, 959-970.

284. Earnshaw, S. (1842) On the nature of the molecular forces which regulate the constitution of the luminferous ether. Trans Camb Phil Soc 7, 97-112.

285. Grenvall, C., et al. (2014) Two-dimensional acoustic particle focusing enables sheathless chip Coulter counter with planar electrode configuration. Lab on a chip 14, 4629-4637.

286. Au, A.K., et al. (2014) Mail-order microfluidics: evaluation of stereolithography for the production of microfluidic devices. Lab Chip 14, 1294-1301.

287. Lenshof, A., et al. (2012) Acoustofluidics 5: Building microfluidic acoustic resonators. Lab Chip 12, 684-695.

288. Evander, M. and Tenje, M. (2014) Microfluidic PMMA interfaces for rectangular glass capillaries. Journal of Micromechanics and Microengineering 24, 027003.

289. Bruus, H., et al. (2011) Forthcoming Lab on a Chip tutorial series on acoustofluidics: acoustofluidics-exploiting ultrasonic standing wave forces and acoustic streaming in microfluidic systems for cell and particle manipulation. Lab Chip 11, 3579-3580.

290. Wiklund, M. (2012) Acoustofluidics 12: Biocompatibility and cell viability in microfluidic acoustic resonators. Lab Chip 12, 2018-2028.

291. Ward, M., et al. (2009) Fundamentals of Acoustic Cytometry. Current Protocols in Cytometry 49, 1.22.21-21.22.12.

292. Nyborg, W.L. (1978) Ultrasound: Its Applications in Medicine and Biology. Elsevier.

264

293. Leibacher, I., et al. (2013) Acoustophoresis of hollow and core-shell particles in two-dimensional resonance modes. Microfluidics and Nanofluidics

294. Grover, W.H., et al. (2011) Measuring single-cell density. Proceedings of the National Academy of Sciences of the United States of America 108, 10992-10996.

295. Chattopadhyay, P.K., et al. (2014) Single-cell technologies for monitoring immune systems. Nature immunology 15, 128-135.

296. Cheng, X., et al. (2007) A microchip approach for practical label-free CD4+ T-cell counting of HIV-infected subjects in resource-poor settings. J. Acquir. Immune Defic. Syndr. 45, 257-261.

297. Tseng, P., et al. (2012) Magnetic nanoparticle-mediated massively parallel mechanical modulation of single-cell behavior. Nature methods 9, 1113-1119.

298. Yellen, B.B., et al. (2002) Analysis of interactions for magnetic particles assembling on magnetic templates. J. Appl. Phys. 91, 8552.

299. Yellen, B.B., et al. (2003) Printing superparamagnetic colloidal particle arrays on patterned magnetic film. J. Appl. Phys. 93, 7331.

300. Yellen, B.B. and Friedman, G. (2003) Analysis of repulsive interactions in chains of superparamagnetic colloidal particles for magnetic template-based self- assembly. J. Appl. Phys. 93, 8447.

301. Yellen, B.B. and Friedman, G. (2004) Programmable assembly of heterogeneous colloidal particle arrays. Advanced Materials 16, 111-115.

302. Yellen, B.B. and Friedman, G. (2004) Programmable Assembly of Colloidal Particles Using Magnetic Microwell Templates. Langmuir 20, 2553-2559.

303. Yellen, B.B., et al. (2005) Arranging matter by magnetic nanoparticle assemblers. Proc. Natl. Acad. Sci. 102, 8860-8864.

304. Fritzsch, F.S., et al. (2012) Single-cell analysis in biotechnology, systems biology, and biocatalysis. Annual review of chemical and biomolecular engineering 3, 129-155.

265

305. Cooper, D.A., et al. (1988) Characterization of T lymphocyte responses during primary infection with human immunodeficiency virus. J. Infect. Dis. 157, 889- 896.

306. Serrano-Villar, S., et al. (2014) The CD4:CD8 ratio is associated with markers of age-associated disease in virally suppressed HIV-infected patients with immunological recovery. HIV Medicine 15, 40-49.

307. Sen, M., et al. (2013) Cell pairing using a dielectrophoresis-based device with interdigitated array electrodes. Lab Chip 13, 3650-3652.

308. Mateo, J., et al. (2014) The promise of circulating tumor cell analysis in cancer management. Genome Biology 15, 1-10.

309. Zhang, T. and Armstrong, A.J. (2016) Clinical Utility of Circulating Tumor Cells in Advanced Prostate Cancer. Curr Oncol Rep 18, 3.

310. Alix-Panabieres, C. and Pantel, K. (2013) Circulating tumor cells: liquid biopsy of cancer. Clin Chem 59, 110-118.

311. Cristofanilli, M., et al. (2004) Circulating tumor cells, disease progression, and survival in metastatic breast cancer. The New England Journal of Medicine 2004, 8.

312. Miller, M.C., et al. (2010) Significance of Circulating Tumor Cells Detected by the CellSearch System in Patients with Metastatic Breast Colorectal and Prostate Cancer. J Oncol 2010, 617421.

313. Lowes, L.E. and Allan, A.L. (2014) Recent advances in the molecular characterization of circulating tumor cells. Cancers (Basel) 6, 595-624.

314. Yu, M., et al. (2014) Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 345, 216-220.

315. Scher, H.I., et al. (2015) Circulating Tumor Cell Biomarker Panel As an Individual-Level Surrogate for Survival in Metastatic Castration-Resistant Prostate Cancer. Journal of Clinical Oncology 33, 1348-1355.

316. Alix-Panabieres, C. and Pantel, K. (2014) Challenges in circulating tumour cell research. Nat Rev Cancer 14, 623-631.

266

317. McGrath, J., et al. (2014) Deterministic lateral displacement for particle separation: a review. Lab on a chip 14, 4139-4158.

318. Dong, Y., et al. (2013) Microfluidics and circulating tumor cells. J Mol Diagn 15, 149-157.

319. Raimondi, C., et al. (2014) Clinical utility of circulating tumor cell counting through CellSearch((R)): the dilemma of a concept suspended in Limbo. Onco Targets Ther 7, 619-625.

320. Lee, H., et al. (2010) High-speed RNA microextraction technology using magnetic oligo-dT beads and lateral magnetophoresis. Lab on a chip 10, 2764-2770.

321. Sarioglu, A.F., et al. (2015) A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat Methods 12, 685-691.

322. Adams, J.D., et al. (2009) Integrated acoustic and magnetic separation in microfluidic channels. Appl Phys Lett 95, 254103.

323. Shields IV, C.W., et al. (2016) Fabrication and operation of acoustofluidic devices supporting bulk acoustic standing waves for sheathless focusing of particles. Journal of Visualized Experiments e53861, 1-7.

324. Park, S., et al. (2014) Morphological differences between circulating tumor cells from prostate cancer patients and cultured prostate cancer cells. PLoS One 9, e85264.

325. Mikolajczyk, S.D., et al. (2011) Detection of EpCAM-Negative and Cytokeratin- Negative Circulating Tumor Cells in Peripheral Blood. J Oncol 2011, 252361.

326. Muluneh, M., et al. (2014) Track-etched magnetic micropores for immunomagnetic isolation of pathogens. Adv Healthc Mater 3, 1078-1085.

327. Jakobsson, O., et al. (2015) Thousand-fold volumetric concentration of live cells with a recirculating acoustofluidic device. Anal Chem 87, 8497-8502.

328. Owens, C.E., et al. (2016) Highly parallel acoustic assembly of microparticles into well-ordered colloidal crystallites. Soft Matter 12, 717-728.

267

329. Chang, C.L., et al. (2015) Circulating tumor cell detection using a parallel flow micro-aperture chip system. Lab on a chip 15, 1677-1688.

330. Mahara, A., et al. (2014) Complete cell killing by applying high hydrostatic pressure for acellular vascular graft preparation. Biomed Res Int 2014, 379607.

331. Balic, M., et al. (2012) Progress in circulating tumor cell capture and analysis: implications for cancer management. Expert Rev Mol Diagn 12, 303-312.

332. De Mattos-Arruda, L., et al. (2013) Circulating tumour cells and cell-free DNA as tools for managing breast cancer. Nat Rev Clin Oncol 10, 377-389.

333. Armstrong, A.J., et al. (2011) Epithelial-mesenchymal transition in prostate cancer: providing new targets for therapy. Asian journal of andrology 13, 179-180.

334. Armstrong, A.J., et al. (2012) Biomarkers in the management and treatment of men with metastatic castration-resistant prostate cancer. European urology 61, 549- 559.

335. Nash, M.A., et al. (2010) Mixed stimuli-responsive magnetic and gold nanoparticle system for rapid purification, enrichment, and detection of biomarkers. Bioconjugate Chem 21, 2197-2204.

336. MacEwan, S.R. and Chilkoti, A. (2010) Elastin-like polypeptides: biomedical applications of tunable biopolymers. Biopolymers 94, 60-77.

337. Dickinson, A.J., et al. (2013) Automated capillary electrophoresis system for fast single-cell analysis. Analytical chemistry 85, 4797-4804.

338. Ivanov, I., et al. (2013) Chip-based nanostructured sensors enable accurate identification and classification of circulating tumor cells in prostate cancer patient blood samples. Analytical chemistry 85, 398-403.

339. Mao, X., et al. (2008) Detection of TMPRSS2:ERG fusion gene in circulating prostate cancer cells. Asian journal of andrology 10, 467-473.

340. Riethdorf, S., et al. (2008) Review: Biological relevance of disseminated tumor cells in cancer patients. International journal of cancer. Journal international du cancer 123, 1991-2006.

268

341. Robinson, J. and Roederer, M. (2015) Flow cytometry strikes gold. Science 350, 739-740.

342. Mann, S. (2009) Self-assembly and transformation of hybrid nano-objects and nanostructures under equilibrium and non-equilibrium conditions. Nat. Mater. 8, 781-792.

343. Motornov, M., et al. (2012) Field-directed self-assembly with locking nanoparticles. Nano Lett. 12, 3814-3820.

344. Gates, B. and Xia, Y. (2001) Photonic crystals that can be addressed with an external magnetic field. Adv. Mater. 13, 1605-1608.

345. Whitesides, G.M. and Grzybowski, B. (2002) Self-assembly at all scales. Science 295, 2418-2421.

346. Evers, W.H., et al. (2010) Entropy-driven formation of binary semiconductor- nanocrystal superlattices. Nano Lett. 10, 4235-4241.

347. Denkov, N.D., et al. (1992) Mechanism of formation of two-dimensional crystals from latex particles on substrates. Langmuir 8, 3183-3190.

348. Denkov, N.D., et al. (1993) Two-dimensional crystallization. Nature 361, 26.

349. Singh, G., et al. (2011) Multicomponent colloidal crystals that are tunable over large areas. Soft Matter 7, 3290.

350. Singh, G., et al. (2011) Highly Ordered Nanometer-Scale Chemical and Protein Patterns by Binary Colloidal Crystal Lithography Combined with Plasma Polymerization. Adv. Funct. Mater. 21, 540-546.

351. Wang, P.Y., et al. (2016) Rapid Self-Assembly of Shaped Microtiles into Large, Close-Packed Crystalline Monolayers on Solid Surfaces. Small 12, 1-6.

352. Leunissen, M.E., et al. (2005) Ionic colloidal crystals of oppositely charged particles. Nature 437, 235-240.

353. Choi, S.Q., et al. (2011) Synthesis of multifunctional micrometer-sized particles with magnetic, amphiphilic, and anisotropic properties. Adv. Mater. 23, 2348- 2352.

269

354. Yan, J., et al. (2012) Linking synchronization to self-assembly using magnetic Janus colloids. Nature 491, 578-581.

355. Chen, Q., et al. (2011) Supracolloidal Reaction Kinetics of Janus Spheres. Science 331, 199-202.

356. Wang, Y., et al. (2012) Colloids with valence and specific directional bonding. Nature 491, 51-55.

357. Gangwal, S., et al. (2008) Dielectrophoretic assembly of metallodielectric Janus particles in AC electric fields. Langmuir 24, 13312-13320.

358. Velev, O.D. and Gupta, S. (2009) Materials Fabricated by Micro- and Nanoparticle Assembly - The Challenging Path from Science to Engineering. Adv. Mater. 21, 1897-1905.

359. Smoukov, S.K., et al. (2008) Reconfigurable responsive structures assembled from magnetic Janus particles. Soft Matter 5, 1285-1292.

360. Erb, R.M., et al. (2009) Magnetic assembly of colloidal superstructures with multipole symmetry. Nature 457, 999-1002.

361. Ahniyaz, A., et al. (2007) Magnetic field-induced assembly of oriented superlattices from maghemite nanocubes. Proc. Natl. Acad. Sci. U. S. A. 104, 17570-17574.

362. Yang, Y., et al. (2011) Imaginary magnetic tweezers for massively parallel surface adhesion spectroscopy. Nano Lett. 11, 1681-1684.

363. Erb, R.M., et al. (2009) Towards holonomic control of Janus particles in optomagnetic traps. Adv. Mater. 21, 4825-4829.

364. Li, K.H. and Yellen, B.B. (2010) Magnetically tunable self-assembly of colloidal rings. Appl. Phys. Lett. 97, 083105.

365. Erb, R.M., et al. (2008) Magnetic field induced concentration gradients in magnetic nanoparticle suspensions: Theory and experiment. J. Appl. Phys. 103, 063916.

270

366. Pawar, A.B. and Kretzschmar, I. (2010) Fabrication, assembly, and application of patchy particles. Macromol. Rapid Commun. 31, 150-168.

367. Gangwal, S., et al. (2010) Programmed assembly of metallodielectric patchy particles in external AC electric fields. Soft Matter 6, 1413.

368. Loget, G. and Kuhn, A. (2012) Bulk synthesis of Janus objects and asymmetric patchy particles. J. Mat. Chem. 22, 15457.

369. Jiang, S. and Granick, S. (2007) Janus balance of amphiphilic colloidal particles. J. Chem. Phys. 127, 1611021-1611024.

370. Walther, A. and Muller, A.H. (2013) Janus Particles: Synthesis, Self-Assembly, Physical Properties, and Applications. Chem. Rev.

371. Pawar, A.B. and Kretzschmar, I. (2008) Patchy particles by glancing angle deposition. Langmuir 24, 355-358.

372. Yan, J., et al. (2013) Colloidal ribbons and rings from Janus magnetic rods. Nat. Comm. 4, 1516.

373. Glotzer, S.C. and Anderson, J.A. (2010) Nanoparticle assembly: made to order. Nat. Mater. 9, 885-887.

374. Champion, J.A., et al. (2007) Making polymeric micro- and nanoparticles of complex shapes. Proc. Natl. Acad. Sci. U. S. A. 104, 11901-11904.

375. Zoldesi, C.I. and Imhof, A. (2005) Synthesis of monodisperse colloidal spheres, capsules, and microballoons by emulsion templating. Adv. Mater. 17, 924-928.

376. Chen, W., et al. (2012) Photolithographic surface micromachining of polydimethylsiloxane (PDMS). Lab Chip. 12, 391-395.

377. Nikoobakht, B. and El-Sayed, M.A. (2003) Preparation and growth mechanisms of gold nanorods (NRs) using -mediated growth method. Chem. Mater. 15, 1957-1962.

378. Wiley, B.J., et al. (2004) Polyol synthesis of silver nanoparticles- use of chloride and oxygen to promote the formation of single-crystal, truncated cubes and tetrahedrons. Nano Lett. 4, 1733-1739.

271

379. Kuijk, A., et al. (2011) Synthesis of monodisperse, rodlike silica colloids with tunable aspect ratio. J. Am. Chem. Soc. 133, 2346-2349.

380. Herlihy, K.P., et al. (2008) Electrically driven alignment of crystallization of unique anisotropic polymer particles. Langmuir 24, 8421-8426.

381. Bucaro, M.A., et al. (2009) Tunable liquid optics- electrowetting-controlled liquid mirrors based on self-assembled Janus tiles. Langmuir 25, 3876-3879.

382. Zhao, L.-B., et al. (2010) A novel method for generation of amphiphilic PDMS particles by selective modification. Microfluid. Nanofluid. 10, 453-458.

383. Roskov, K.E., et al. (2011) Long-range alignment of gold nanorods in electrospun polymer nano/microfibers. Langmuir 27, 13965-13969.

384. Wiley, B.J., et al. (2006) Right bipyramids of silver- a new shape derived from single twinned seeds. Nano Lett. 6, 765-768.

385. Stebe, K.J., et al. (2009) Oriented Assembly of Metamaterials. Science 325, 159-160.

386. von Blaaderen, A., et al. (1997) Template-Directed Colloidal Crystallization. Nature 385, 321-324.

387. Yethiraj, A., et al. (2004) Large-area electric-field-induced colloidal single crystals for photonic applications. Adv. Mater. 16, 596-600.

388. Zhang, J., et al. (2009) Self-assembly of photonic crystals from polymer colloids. Curr. Opin. Colloid Interface Sci. 14, 103-114.

389. Cheng, W., et al. (2006) Observation and tuning of hypersonic bandgaps in colloidal crystals. Nat. Mater. 5, 830-836.

390. Wang, M.C.P. and Gates, B.D. (2009) Directed Assembly of Nanowires. Mater. Today 12, 34-43.

391. Coyle, S., et al. (2003) Spherical micromirrors from templated self-assembly: Polarization rotation on the micron scale. Appl. Phys. Lett. 83, 767.

272

392. Wink, T., et al. (1997) Self-assembled Monolayers for Biosensors. The Analyst 122, 43R-50R.

393. Levin, I., et al. (2006) Design of Self-Assembled Multiferroic Nanostructures in Epitaxial Films. Adv. Mater. 18, 2044-2047.

394. Khalil, K.S., et al. (2012) Binary colloidal structures assembled through Ising interactions. Nat. Comm. 3, 794.

395. Bharti, B., et al. (2012) Co-assembly of oppositely charged particles into linear clusters and chains of controllable length. Sci. Rep. 2, 1004.

396. Nili, H., et al. (2011) Higher order dielectrophoretic force characterisation of non- spherical particles. J. Phys. Conf. Ser. 301, 012061.

397. Jackson, J.D. (1962) Classical Electrodynamics. John Wiley & Sons Inc.

398. Lumsdon, S.O., et al. (2004) Two-dimensional crystallization of microspheres by a coplanar AC electric field. Langmuir 20, 2108-2116.

399. Yethiraj, A. (2007) Tunable colloids: control of colloidal phase transitions with tunable interactions. Soft Matter 3, 1099.

400. Gangwal, S., et al. (2008) Induced-Charge Electrophoresis of Metallodielectric Particles. Phys. Rev. Lett. 100

401. Paxton, W.F., et al. (2004) Catalytic nanomotors: Autonomous movement of striped nanorods. J. Am. Chem. Soc. 126, 13424-13431.

402. Zhu, X. (2009) Recent patents on perovskite ferroelectric nanostructures. Recent Pat. Nanotechnol. 3, 42-52.

403. Baksh, M.M., et al. (2004) Detection of molecular interactions at membrane surfaces through colloid phase transitions. Nature 427, 139-141.

404. Velev, O.D. and Kaler, E.W. (1999) In situ assembly of colloidal particles into miniaturized biosensors. Langmuir 15, 3693-3698.

405. Wan, Y., et al. (2009) Simulation and fabrication of binary colloidal photonic crystals and their inverse structures. Mater. Lett. 63, 2078-2081.

273

406. Sacanna, S., et al. (2013) Engineering shape: the novel geometries of colloidal self- assembly. Soft Matter 9, 8096.

407. Singh, G., et al. (2011) Highly ordered mixed protein patterns over large areas from self-assembly of binary colloids. Advanced materials 23, 1519-1523.

408. Singh, G., et al. (2011) Large-Area Protein Patterns Generated by Ordered Binary Colloidal Assemblies as Templates. ACS Nano 5, 3542–3551.

409. Wang, P.-Y., et al. (2015) Self-assembled binary colloidal crystal monolayers as cell culture substrates. J. Mater. Chem. B 3, 2545-2552.

410. Dai, Z.F., et al. (2012) Phase Diagram, Design of Monolayer Binary Colloidal Crystals, and Their Fabrication Based on Ethanol-Assisted Self-Assembly at the Air/Water Interface. ACS Nano 6, 6706-6716.

411. Winkleman, A., et al. (2005) Directed Self-Assembly of Spherical Particles on Patterned Electrodes by an Applied Electric Field. Advanced materials 17, 1507- 1511.

412. Ilievski, F., et al. (2011) Templated self-assembly in three dimensions using magnetic levitation. Soft Matter 7, 9113.

413. Bharti, B. and Velev, O.D. (2015) Assembly of Reconfigurable Colloidal Structures by Multidirectional Field-Induced Interactions. Langmuir 31, 7897- 7908.

414. Sacanna, S., et al. (2013) Shaping colloids for self-assembly. Nat Commun 4, 1688.

415. Millan, J.A., et al. (2015) Effect of shape on the self-assembly of faceted patchy nanoplates with irregular shape into tiling patterns. Soft Matter 11, 1386-1396.

416. Vella, D. and Mahadevan, L. (2005) The “Cheerios effect”. American Journal of Physics 73, 817.

417. Bowden, N., et al. (1997) Self-assembly of mesoscale objects into ordered two- dimensional arrays. Science 276, 233-235.

274

418. Bowden, N., et al. (1999) Mesoscale Self-Assembly of Hexagonal Plates Using Lateral Capillary Forces: Synthesis Using the “Capillary Bond”. J Am Chem Soc 121, 5373-5391.

419. Pickering, S.U. (1907) Emulsions. J Chem Soc 91, 2001-2021.

420. Liu, J., et al. (2014) Programmable anisotropic microparticles for self-assembly applications. MRS Proceedings 1622, 1-6.

421. Ramsden, W. (1903) Separation of Solids in the Surface-Layers of Solutions and 'Suspensions'. Proceedings of the Royal Society of London 72, 156-164.

422. Meng, G., et al. (2014) Elastic Instability of a Crystal Growing on a Curved Surface. Science 343, 634-637.

423. Afsar-Siddiqui, A.A., et al. (2004) Dewetting Behavior of Aqueous Cationic Surfactant Solutions on Liquid Films. Langmuir 20, 7575-7582.

424. Sacanna, S. and Pine, D.J. (2011) Shape-anisotropic colloids: Building blocks for complex assemblies. Current Opinion in Colloid & Interface Science 16, 96-105.

425. Denkov, N.D. and Kralchevsky, P.A. (1995) Colloidal structural forces in thin liquid films. Progr Colloid Polym Sci 98, 18-22.

426. Park, B.J., et al. (2013) Geometrically and chemically anisotropic particles at an oil–water interface. Soft Matter 9, 3383.

427. Chatterjee, N. and Flury, M. (2013) Effect of particle shape on capillary forces acting on particles at the air-water interface. Langmuir 29, 7903-7911.

428. Clark, T.D., et al. (2001) Self-Assembly of 10-µm-Sized Objects into Ordered Three-Dimensional Arrays. J Am Chem Soc 123, 7677-7682.

429. Manoharan, V.N. (2015) Colloids at interfaces: Pinned down. Nat Mater 14, 869- 870.

430. Zhang, G. and Wang, D. (2009) Colloidal lithography--the art of nanochemical patterning. Chem Asian J 4, 236-245.

275

Biography

Wyatt Shields was born in Richmond, Virginia on November 23, 1988. He was raised in Powhatan, Virginia, and he graduated from Powhatan High School in 2007.

Wyatt matriculated in the University of Virginia to study Biomedical Engineering and

Materials Science. He attained his first research experiences with Prof. William F. Walker and Prof. Jeffrey J. Saucerman. Wyatt eventually became a member of Tau Beta Pi and the Raven Society, and he served as the founding Associate Editor, and afterwards

Editor-in-Chief, of The Spectra: The Virginia Engineering and Science Research Journal. He graduated in 2011 with a Bachelors of Science degree with high distinction.

Wyatt joined the research group of Prof. Gabriel P. López at Duke University in

2011. Wyatt received an NSF Fellowship and a fellowship to the NSF’s Research Triangle

MRSEC in his first year. Wyatt’s research foci included the development of innovative bioseparation and bioanalytical microsystems as well as the programmed assembly of soft matter. Wyatt was the recipient of the 2013 and 2015 Exceptional Student Award by the International Society for the Advancement of Cytometry, an NSF GROW Award in

2014 to study under Prof. Thomas Laurell at Lund University in Sweden and the 2016

Dean’s Award for Excellence in Mentoring from Duke University. Wyatt has delivered

12 podium presentations at national or international conferences and has mentored 15 students during his time in graduate school. Wyatt’s ultimate goal is to make a lasting impact on his community through research, education and outreach.

276