Capture, purification, and release of biological substances using a surface coating

Information

  • Patent Grant
  • 11674958
  • Patent Number
    11,674,958
  • Date Filed
    Friday, March 27, 2020
    4 years ago
  • Date Issued
    Tuesday, June 13, 2023
    11 months ago
Abstract
This invention relates to a surface coating for capture circulating rare cells, comprising a nonfouling composition to prevent the binding of non-specific cells and adsorption of serum components; a bioactive composition for binding the biological substance, such as circulating tumor cells; with or without a linker composition that binds the nonfouling and bioactive compositions. The invention also provide a surface coating for capture and purification of a biological substance, comprising a releasable composition to release the non-specific cells and other serum components; a bioactive composition for binding the biological substance, such as circulating tumor cells; with or without a linker composition that binds the releasable and bioactive compositions. The present invention also discloses a novel microfluidic chip, with specific patterned microstructures to create a flow disturbance and increase the capture rate of the biological substance.
Description
CROSS-REFERENCES TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application No. 15/378,938, filed Dec. 14, 2016, which is a continuation of U.S. patent application No. 14/128,354, filed May 20, 2014, now U.S. Pat. No. 9,541,480, which is a 371 national stage entry of PCT Application No. PCT/US2012/044701, filed Jun. 28, 2012, which claims the benefit of U.S. Provisional Application No. 61/502,844, filed Jun. 29, 2011, and claims the benefit of U.S. Provisional Application No. 61/606,220, filed Mar. 2, 2012, the entirety of which are incorporated by reference.


STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT

Not Applicable


REFERENCE TO A “SEQUENCE LISTING,” A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK

Table 1 is the amino acid sequence of EpAb4-1 antibody.


BACKGROUND OF THE INVENTION

The shedding of cells into the circulation is an intrinsic property of the malignant tumor, and this feature provides important information with regard to the diagnosis, staging, treatment response and survival of cancer patients. For example, Pantel et al found the number of circulating tumor cells (CTCs) in the blood is correlated with the aggressiveness of the cancer as well as the efficacy of the therapy. (Pantel, K. et. al., “Detection, clinical relevance and specific biological properties of disseminating tumor cells”, Nat Rev Cancer. 2008, 8(5):329-40).


However, CTCs, as few as one per 109 blood cells in patients with metastatic cancer, are rare cells. This makes the detection and isolation of CTCs technically challenging (see Kahn et al. Breast Cancer Res Treat 2004, 86:237-47). An enrichment process is therefore necessary to effectively detect and isolate CTCs.


An example of such enrichment process is the use of a highly overexpressed cell surface biomarker with high specificity and sensitivity for CTCs, such as the epithelial cell adhesion molecule (EpCAM). The Cellsearch Systet™ (Veridex), the only FDA-approved platform for CTC detection, utilizes anti-EpCAM antibody-coated magnetic nanoparticles to capture and enrich CTCs, followed by cytokeratin immunostaining. The AdnaTest (AdnaGen AG, Germany), another commercially available system for CTC detection, adopts similar immunomagnetic approach by using anti-EpCAM and Mucin 1 (MUC) conjugated magnetic beads. More recently, “CTC chips” based on anti-EpCAM antibody-coated microfluidics chip were developed for CTC detection and enrichment (Nagrath et al, Nature 2007, 450:1235-9). However, the disadvantage of the above techniques is the low detection rate of pure CTCs, due to the non-specific binding of blood cells with anti-EpCAM antibody.


In order to maximize the detection and isolation of CTCs, it is necessary to reduce the nonspecific binding of other circulating blood cells. This can be achieved by surface modification with bioinert materials. For example. Kaladhar et al. observed a significant fewer circulating blood cells (e.g. platelets, leukocytes, and erythrocytes) binding onto the solid substrate modified with supported monolayer of various lipid compositions containing phosphatidyl-choline, cholesterol, and glycolipid (Kaladhar et al, Langmuir 2004, 20: 11115-22 and Kaladhar et al, J Biomed Mater Res A 2006, 79A:23-35).


Despite the advance in the detection and isolation CTCs technology, there is still a need for a more specific and effective method for detecting, purification and releasing CTCs and other biological substances for further cultivation and characterization.


BRIEF SUMMARY OF THE INVENTION

In one aspect, the present invention is directed to a surface coating to capture a circulating rare cell (CRC). This surface coating increases the capture efficiency of a CRC, such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and reduces the binding of non-specific cells or protein adsorption.


The surface coating comprises 1) a nonfouling composition that reduces the binding of nonspecific blood cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures a CRC. The surface coating further comprises a linker composition that attaches to the nonfouling composition and the bioactive composition, as illustrated in FIG. 1A.


In another aspect, the present invention is directed to a surface coating to capture and release a biological substance. This surface coating increases the capture efficiency of a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and enhances the removal or release of the non-specific cells or protein from the surface coating.


The surface coating comprises 1) a releasable composition for releasing or removing nonspecific blood cells and other blood components, such as protein, from the surface coating; and 2) a bioactive composition that captures a biological substance. The surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.


The present invention is also directed to a microfluidic device, with specific microstructure designs to create a disturbed flow of blood, body fluid or biological samples to increase the capture rate of the biological substance.


The present invention is also directed to a method of manufacturing the surface coating, comprising a) forming the nonfouling or the releasable composition; and b) attaching the the linker composition with the nonfouling/releasable composition from step a) and the bioactive composition, or c) attaching the nonfouling/releasable composition from step a) with the bioactive composition.


The present invention is also directed to methods to capture and release the biological substance from the surface coating. The biological substance on the surface coating can be purified by removing the non-specific cells or protein. The captured biological substance can be released by air bubbles, ultraviolet irradiation and the like.


The present invention is also directed to uses of a biotinylated anti-EpCam antibody, EpAb4-1 antibody, to capture a CTC.





BRIEF DESCRIPTION OF THE DRAWINGS

Embodiments of the present invention may be described with reference to the accompanying drawings.



FIG. 1A illustrates schematically an embodiment of the surface coating comprising a nonfouling composition, a linker composition and a bioactive composition.



FIG. 1B illustrates schematically the binding of a circulating tumor cell with the surface coating from FIG. 1A



FIG. 2A to FIG. 2F illustrate the chemical structures of examples of nonfouling materials.



FIG. 3 illustrates the chemical reactions of conjugation between the functional groups on the nonfouling composition and the bioactive composition.



FIG. 4A illustrates schematically the attachment of the surface coating and the solid substrate without a surface linker.



FIG. 4B and FIG. 4C illustrate schematically a linker composition with a cleavable functional group.



FIG. 4D illustrates schematically the attachment of the surface coating and the solid substrate using a surface linker.



FIG. 5A and FIG. 5B illustrates schematically the formation of the surface coating on a solid substrate.



FIGS. 6A and 6B illustrate schematically the components of a microfluidic chip.



FIG. 6C illustrates schematically the microfluidic chip assembly to capture CTCs from a biological sample.



FIG. 7A to FIG. 7H illustrate schematically the designs of the microstructures on the solid substrate.



FIGS. 7I and 7J illustrate the capture efficiency of various microstructure designs in DMEM solution and blood respectively.



FIG. 8 illustrates the shear stresses of a buffer solution to release the non-specific cells and purify the captured biological substance.



FIG. 9. illustrates schematically the release of biological substance by the air bubble method.



FIG. 10A illustrates schematically the surface coating with a cleavable linker composition on a solid substrate.



FIG. 10B illustrates schematically the release of the biologic substance from the surface coating in FIG. 10A.



FIG. 11 illustrates QCM-D response of the surface coating construction.



FIG. 12 illustrates the QCM-D response of the addition of bovine serum albumin to the surface coating.



FIG. 13 are the photographs of the non-specific cells (top images) and the CTCs (bottom images) on the surface coating before and after the buffer rinse.



FIG. 14A illustrates the capture efficiency and non-specific blood cell binding of various surface coatings.



FIG. 14B are photo images which illustrate the non-specific blood cell binding of various surface coatings before and after the buffer rinse.



FIG. 15A to FIG. 15C are the photographs of the non-specific cells and the biological substance on the surface coating before and after the buffer rinse purification.



FIG. 16 illustrates the various shear stress and flushing time for the removal of HCT116 and NIH-3T3 cell populations from the surface coating.



FIG. 17 are the photographs of the CTCs released by the air bubbles.



FIG. 18 illustrates the cell cultures of the released CTCs on day 1, day 10 and day 14.



FIG. 19 illustrates schematically a CTC filtration device.



FIG. 20 illustrates the CTC binding specificity of biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgG antibody.





DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to a surface coating to effectively capture a circulating rare cell (CRC), such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like.


In one embodiment, the surface coating for the capture of a CRC comprises 1) a nonfouling composition that prevents the binding of non-specific cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures the circulating rare cells. The nonfouling composition and the bioactive composition are joined by discrete functional groups or moieties present in the nonfouling and bioactive compositions. Generally, a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.


In one group of embodiments, complementary DNA fragments are used for binding the nonfouling composition and the bioactive composition. The fragments are attached to each of the compositions and can be partially or completely complementary over their lengths. A suitable length of DNA will generally be at least 15, 20, 25, 35, 50, 100 or more bases in length. An example of the DNA used in the present invention is an DNA tweezer. (See, B Yurke et al., A DNA-fueled molecular machine made of DNA. Nature 2000, 406:605-608.)


In another group of embodiments, the surface coating comprises 1) a nonfouling composition; 2) a bioactive composition; and 3) a linker composition, which joins the nonfouling composition to the bioactive composition. Sec FIG. 1A.


The present invention is also directed to a surface coating to effectively capture a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like, purify the biological substance on the surface of the surface coating by releasing or removing the non-specific cells and other serum components (e.g. protein) through a buffer rinse, and release the captured biological substance from the surface coating.


The surface coating for the capture and purification of a biological substance comprises 1) a releasable composition for releasing nonspecific blood cells and other blood components, such as protein, through a buffer rinse; and 2) a bioactive composition that captures a biological substance. The releasable composition and the bioactive composition are joined by discrete functional groups or moieties present in the releasable and bioactive compositions. Generally, a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.


In one embodiment, the surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.


As will be explained in more detail below, the surface coating can be incorporated into the following configurations: cell cultural dishes, microfluidic channels, microfluidic chips, filtration filter, capillaries, tubes, beads, nanoparticles, or the like, with an inner diameter ranging from about 50 to about 1000 um.


Nonfouling and Releasable Composition


The “nonfouling” composition (see FIG. 1A) reduces the binding of non-specific cells and adsorption of the serum protein.


The “releasable” composition comprises a nonfouling composition which also acts as a “lubricating” surface such that only low flow shear stress is required to remove or release the non-specific cells or blood components from the surface coating, while the biological substance remains intact.


The nonfouling composition is selected from the group consisting of: a supported lipid layer such as liposomes, supported lipid bilayers (SLBs) or lipid multilayer, polypeptides, polyelectrolyte multilayers (PEMs), polyvinyl alcohol, polyethylene glycol (PEG) as illustrated in FIG. 2A, hydrogel polymers, extracellular matrix proteins, carbohydrate, polymer brushes, zwitterionic materials such as poly(carboxybetaine) (pCB)) as illustrated in FIG. 2D, poly(sulfobetaine)(pSB) as illustrated in FIG. 2E and pDMAEMA as illustrated in FIG. 2F, small organic compounds, and the combination of above materials forming a single or a multi-layer.


For those embodiments in which the nonfouling composition comprises supported lipid bilayers (SLBs), the SLBs typically comprise lipids such as, for example, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(cap biotinyl) (sodium salt) (b-PE) as illustrated in FIG. 2B and 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC). The protein resistant property of a SLB can be explained by the presence of neutral and zwitterionic phosphatidylcholine headgroups in a wide pH range, as well as an aqueous thin film formed between the hydrophilic lipid head groups and the bulk solution (see. Johnson et al., Biophys J 1991, 59:289-94).


In another group of embodiments, the nonfouling composition comprises PEG, preferably PEG with a molecular weight from about 100 to about 100,000 and exhibits a nonfouling property.


In yet another group of embodiments, the nonfouling composition comprises polyelectrolyte multilayers (PEMs) or a polymer brush. Examples of suitable PEMs useful in the present invention include, but are not limited to, poly-L-lysine/poly-L-glutamic acid (PLL/PLGA), poly-L-lysine/poly-L-aspartic acid or similar counter ionic polyelectrolytes. The polymer brush comprises ([2-(acryloyloxy)ethyl] trimethyl ammonium chloride, TMA)/(2-carboxy ethyl acrylate, CAA) copolymer as illustrated in FIG. 2C. Generally, the nonfouling layer has a thickness from a few nanometers up to hundreds microns.


The nonfouling composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment, either directly to a functional group present in the bioactive composition, or directly to a functional group that is part of the linkage composition.


In some embodiments, the functional groups of the nonfouling composition (prior to covalent attachment) are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the linker or bioactive composition. In other embodiments, the functional groups of the nonfouling composition (prior to non-covalent attachment) which are first members of a binding pair, are selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present in either the linker or bioactive composition.


The Linker Composition


The linker composition joins the nonfouling/releasable composition and the bioactive composition and comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the bioactive composition.


In some embodiments, the linker composition comprises functional groups (prior to covalent attachment) selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the nonfouling or bioactive composition.


In other embodiments, the linker composition comprises functional groups (prior to non-covalent attachment) which are first members of a binding pair, selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the bioactive composition.


The functional groups on the linker composition can also be a cleavable functional group, selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro pulse mechanism, a magnetic material cleavable by the absence of the magnetic force, a polyelectrolyte material cleavable by breaking the electrostatic interaction, a DNA cleavable by hybridization, and the like.


Bioactive Composition


The bioactive composition joins to either the linker composition or the nonfouling composition, and comprises a binding moiety selective for the detection of the biological substance or CRC.


The bioactive composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling layer or to a functional group that is part of the linker composition.


In some embodiments, the functional groups of the bioactive composition (prior to covalent attachment) are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups which are selected to be reactive with functional groups present in either the nonfouling or linker composition. In other embodiments, the functional groups of the bioactive composition (prior to non-covalent attachment) are selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen-antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the linker composition.


The binding moiety of the bioactive composition has specific affinity with the biological substance through molecular recognition, chemical affinity, or geometrical/shape recognition. Examples of the binding moiety for the detection of the biological substance include, but are not limited to: synthetic polymers, molecular imprinted polymers, extracellular matrix proteins, binding receptors, antibodies, DNA, RNA, antigens or any other surface markers which present high affinity to the biological substance. A preferred antibody is the anti-EpCAM membrane protein antibody (commercially available from many sources, including R&D Systems, Minn., USA), which provides high specificity for CTCs because EpCAM is frequently overexpressed in the lung, colorectal, breast, prostate, head and neck and hepatic malignancies, but is absent from haematologic cells. Another preferred antibody is Anti-HER2, which has high specificity for CTCs but absent in haematologic cells.


In one embodiment, the anti-EpCAM membrane protein antibody is EpAb4-1 antibody, comprising a heavy chain sequence with SEQ ID No:1 and alight chain sequence with SEQ ID NO: 2 shown in Table 1.









TABLE 1





Amino Acid Sequence of VH and VL domains of EpAb4-1 antibody




















FW1
CDR1
FW2
CDR2





SEQ ID
QIQLVQSGPELKKPGETV

GYTFTNYG

WVKQAPGKGLK

INTYTGEP



NO: 1
KISCKAS

MN

WMGW



(VH)









SEQ ID
DIVMTQAAFSNPVTLGTS

RSSKSLLH

WYLQKPGQSPQ

HMSNLAS



NO: 2
ASISC

SNGITYLY

LLIY



(VL)






FW3
CDR3
FW4
Family





SEQ ID
TYGDDFKGRFAFSLETSA

FGRSVDF

WGQGTSVTVSS
VH9


NO: 1
STAYLQINNLKNEDTATY





(VH)
FCAR








SEQ ID
GVPDRFSSSGSGTDFTLRI

AQNLENPR

FGGGTKLEIK
VK24/25


NO: 2
SRVEAEDVGIYYC

T





(VL)










Complementary-determining regions 1-3 (CDR1-3), framework regions 1-4 (FW1-4) for both the VH and VL domains are shown. The V domain families were aligned by VBASE2 database (www.vbase2.org).


The bioactive composition can have a variety of thicknesses, selected so that it does not affect the function or the performance of the surface coating.


In one embodiment, the conjugation linkers or catalysts for the nonfouling composition and the bioactive compositions are biotin/avidin or their derivatives. In another embodiment, the conjugation linkers or catalysts for the nonfouling composition and the bioactive composition are EDC/NHS. In yet another preferred embodiment, the conjugation linker or catalysts for the nonfouling composition and the bioactive compositions are sulfo-SMCC. FIG. 3 schematically illustrates the chemical reactions of these embodiments.


Solid Substrate


In some embodiments, the surface coating is attached to the solid substrate without a surface linker, as illustrated in FIG. 4A. The nonfouling/releasable composition is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.


In other embodiments, the surface coating is attached to the solid substrate with a surface linker, as illustrated in FIG. 4D. Examples of the solid substrate used in the present invention include, but are not limited to: metals, plastics, glass, silicon wafers, hydroxylated poly(methyl methacrylate) (PMMA), and a combination thereof. The shape of the solid substrate include, but are not limited to: planar, circular and irregular shapes with micro, or nano-structures such as nanoparticles, nanowires, and a combination thereof.


The surface linker composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the solid substrate. Examples of the surface linker for binding the surface coating to a glass substrate include, but are not limited to, silane, aminopropyltriethoxy silane, aminopropyltrimethoxy silane, silane-PEG-NH2, silane-PEG-N3 (PEG molecular weight is about 1,000 to about 30,000 daltons) and silane-PEG biotin.


In one group of embodiments, the surface linker comprises a cleavable functional group selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro-pulse mechanism, an iron or magnetic material in which the absence of the magnetic force will release the nonfouling composition, a polyelectrolyte material cleavable by breaking the electrostatic interaction, an DNA cleavable by hybridization, and the like.


In one embodiment, the nonfouling composition comprises silane-functionalized PEG and the solid substrate is preferably selected from the group consisting of silicon, glass, hydroxylated poly(methyl methacrylate) (PMMA), aluminum oxide, TiO2 and the like. In another embodiment, the nonfouling composition comprises thiol-functionalized compounds and the solid substrate is preferably selected from the group consisting of Au, Ag, Pt, and the like.


The Method of Manufacturing the Surface Coating



FIGS. 5A and 5B show the steps of forming the surface coating:


1. Formation of the nonfouling/releasable composition (e.g. SLB or PEG) with appropriate functional group (biotin);


2. Attaching the functional group (streptavidin) on the linker composition to the functional group (biotin) on the nonfouling/releasable composition;


3. Formation of the bioactive composition and attaching the functional group (biotin) on the bioactive composition to the functional group (streptavidin) on the linker composition.


The surface coating without a linker composition can be formed by:


1. Formation of the nonfouling/releasable composition with appropriate functional group (e.g. carboxyl group of N-glutaryl phosphatidylethanolamine or NGPE);


2. Formation and attaching the functional group (primary amine) on the bioactive composition to the functional group (carboxyl group of NOPE) on the nonfouling/releasable composition in step 1.


The steps in forming the surface coating as described above can be reversed, and the steps for forming the surface coating without a link composition as described above can be reversed.


Microfluidic Chip


As illustrated in FIG. 6A, the microfluidic chip comprises a first solid substrate 1 (e.g. PMMA) and a second solid substrate 2 (e.g. glass), wherein the first and second solid substrates are adhered together using an adhesive means 3 or other means.


Referring to FIG. 6B, the surface of one or both solid substrates can be engraved with microstructures 4. In one group of embodiments, the microstructures 4 are arranged in a linear fashion. In another group of embodiments, the microstructures 4 are arranged in herringbone fashion. The shaded region on the adhesive 3 in FIG. 6B is carved out to accommodate the microstructures 4 on the surface of the solid substrate 1. A sealed channel 5 is created by adhering the first solid substrate 1 and the second solid substrate 2 together with an adhesive 3. The height of the channel 5 is determined by the thickness of the adhesive 3.


Once the microfluidic chip is formed, the surface coating can be attached to one or both solid substrates. In one group of embodiments, the surface coating is attached to the solid substrate with a surface linker. In another group of embodiments, the surface coating is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.


Referring to FIG. 6C, the microstructures 4 on the solid substrate are perpendicular to the flow direction and create a chaotic or disturbed flow of the blood, body fluid or biologic sample as it passes through the sealed channel 5 of the microfluidic chip. The disturbed flow enhances the biological substance-surface coating contact.


Two factors govern the capture efficiency of the microfluidic chip:


(1) The linear speed of the blood, body fluid or biological sample, which determines the contact time of the biological substance and the surface coating. In a preferred embodiment, the linear speed is about 0.1 mm/s to 1 mm/s. In a more preferred embodiment, the linear speed is about 0.42 mm/s or 0.5 ml/h for Design E in FIG. 7F.


(2) The flow disturbance of the blood, body fluid or biological sample, created by the microstructures 4 on the solid substrate(s). The flow disturbance increases contact between the biological substance and the surface coating.



FIG. 7A shows various designs of the microstructures 4 on the solid substrate. The microstructures in Design F are arranged in a herringbone pattern whereas the microstructures in Designs A-E and H are arranged in a linear pattern. The dimensions of the microstructures 4 are as follows: the length is about 50 mm for O-D and G and about 120 mm for E-F, the height is about 30 μm, the width is about 1.5 mm for O and A about 3.0 mm for B, and about 5.5 mm for C-G. The height of the sealed channel 5 varies with the thickness of the adhesive 3, preferably about 30-90 μm, more preferably about 60 μm.



FIG. 7B-7H show the details of Designs A-G in FIG. 7A. Design G in FIG. 7H is the preferred pattern, with the following dimensions: the width of Microstructure (W) is about 150 μm, the length of microstructure (L) is about 1000 μm, the distance between two rows of microstructures (Sr) is about 250 μm, the distance between two adjacent microstructures (Sc) is about 350 μm, the height of the microstructure (D) is about 30 μm and the height of the sealed channel 5 (H) is about 60 μm.


The biological substance capture efficiency of the various designs are shown in FIG. 7I and FIG. 7J. Capture rate is defined as (captured biological substance/original biological substance in the testing sample)×100%. Channel O has no microstructure and has the lowest biological substance capture rate, at 27% and 1% for DMEM sample and blood sample, respectively. Design E has a 80% capture rate for HCT116 cancer cells spiked in DMEM, and a 30% capture rate for HCT116 cancer cells spiked in blood sample. Design F has the best capture rate, on average over 70% of HCT116 cancer cells spiked in blood sample were captured (see FIG. 7J).


Flow Purification


The biological substance on the surface coating can be further purified by removing the non-specific cells and other blood components on the surface of the nonfouling/releasable composition. The nonfouling/releasable composition has low affinity for non-specific cells and other blood components. Therefore, rinsing the surface coating with a low flow buffer solution of about 0.8 dyne/cm2 to about 50 dyne/cm2 is sufficient to remove non-specific cells and other blood components on the nonfouling/releasable composition while the biological substance remains on the surface coating.


In a preferred embodiment, the shear force of the buffer rinse is about 2.5 to about 10 dyne/cm2. FIG. 8 shows that when the shear stress of the buffer flow is about 3.3 dyne/cm2, 80% of the non-specific cells (i.e. white blood cells) were removed while none of the biological substance (i.e. HCT 116 cancer cells) were removed from the surface coating. When the shear stress of the buffer flow was increased to 8 dyne/cm2, almost all of the non-specific cells were removed while none of the biological substance was removed from the surface coating.


Release of the Biological Substance


After removing the majority of the non-specific cells and blood components by flow purification, the biological substance can be released from the surface coating.


If the nonfouling/releasable composition comprises a lipid or a mixture of lipid, the captured biological substance can be released by introducing an air bubble solution or oil phase. As shown in FIG. 9, the surface coating comprises a nonfouling composition A (lipid bilayer) and a bioactive composition B (antibody) and is bound to a solid substrate S. The biological substance, CTC, is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A. As the air bubble approaches the lipid bilayer, the hydrophobic tails of the lipid bilayer are turned upside down due to its high affinity with the air inside the air bubble, which is also hydrophobic. This breaks up the hydrophilic-hydrophilic interaction at the surface of the lipid bilayer and allows the air bubble to “lift off” the top layer of the lipid bilayer, together with the CTC bound on the bioactive composition.


If the nonfouling composition comprises a composition other than a lipid or a mixture of lipid, the captured biological substance can be released by breaking the cleavable functional group on the linker composition or on the surface linker. This release mechanism is illustrated in FIGS. 10A and 10B. FIG. 10A shows a surface coating on a solid substrate, wherein the surface coating comprises a bioactive composition B, a linker composition with a cleavable functional group C, and a nonfouling composition A. The surface coating is attached to a solid substrate S (e.g. glass) by a surface linker 1. FIG. 10B shows the release of the biologic substance (e.g. CTC) from the surface coating in FIG. 10A. The biologic substance is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A. The surface coating is irradiated with 365 nm ultraviolet light, which breaks the cleavable functional group on the linker composition C and the biologic substance is released for subsequent analysis but maintaining the viability.


The biological substance can also be released by other mechanisms. In one group of embodiments, the linker composition or the surface linker comprises an electrosensitive cleavable functional group, and the biological substance is released by electro pulse mechanism. In another group of embodiments, the linker composition or the surface linker comprises a magnetic material as the cleavable functional group, and the absence of the magnetic field or force releases the biological substance. In yet another group of embodiments, the linker composition or the surface linker comprises a PEM as the cleavable functional group, and the biological substance is released by changing the electrostatic interaction between the layers. In yet another group of embodiments, the linker composition or the surface linker comprises an DNA piece as the cleavable functional group, and the biological substance is released by DNA hybridization.


EXAMPLES

The following examples further illustrate the present invention. These examples are intended merely to be illustrative of the present invention and are not to be construed as being limiting.


Example 1: Preparation of the Two-Layer Surface Coating

Preparation of the Nonfouling Composition:


Supported lipid bilayer (SLB) was prepared by the following steps:


(1) POPC and b-PE (commercially available from Avanti Polar Lipids, USA) were dissolved in chloroform and the final lipid concentration was 5 mg/mL. The POPC/b-PE solution was vortex dried under a slow stream of nitrogen to form a thin, uniform POPC/b-PE film. The POPC/b-PE film was further dried in a vacuum chamber overnight to remove residual chloroform.


(2) The POPC/biotin-PE film in step (1) was dispersed in and mixed with a phosphate buffer containing 10 mM of phosphate buffered saline, 150 mM of sodium chloride aqueous solution, and 0.02% (w/v) of sodium azide (NaN3, commercially available from Sigma-Aldrich, USA), with the pH adjusted to 7.2. The mixed solution was filtered through the 100-nm, followed by the 50-nm Nuclepore® track-etched polycarbonate membranes (Whatman Schleicher & Schuell, Germany) at least 10 times under 150 psi at room temp.


(3) The filtered solution in step (2) was passed through the LIPEX™ Extruder (Northern Lipids, Inc. Canada) to generate a homogenous population of unilamillar vesicles. The size of the POPC/biotin-PE vesicles was about 65±3 nm, determined by the dynamic laser light scattering detector (Zetasizer Nano ZS, Malvern Instruments, Germany).


Preparation of the Bioactive Composition


Biotinylated EpCAM Antibody was prepared by the following steps:


(1) The anti-EpCAM monoclonal antibody (OC98-1 or EpAb4-1) was generated by method described by Chen et al (Clin Vaccine Immunol 2007; 14:404-11).


(2) The antibody in step (1) was dissolved in a buffer solution containing 10 mM of PBS and 150 mM of NaCl, with a pH about 7.2. The concentration of the antibody buffer solution was about 0.65 mg/mL, determined by Nanodrop 1000 spectrophotometer (Thermo Scientific, USA).


(3) The antibody solution in step (2) was mixed with 10 mM of Sulfo NHS-LC-Biotin (with a molar ratio of 1 to 10) and dissolved in Milli-Q water (Milli-Q RO system, USA) at room temperature for 30 min. Excess biotin was removed by dialysis in phosphate buffered saline at 4° C. for 24 h, with a buffer change every 12 h.


(4) The ratio of biotin and antibody in the biotinylated anti-EpCAM antibody (bOC98-1 or bEpAb4-1) was 1.5 to 1, determined by the HABA assay using a biotin quantitation kit (Pierce, USA).


Alternatively, commercially available biotinylated goat anti-human anti-EpCAM antibody from R and D Systems (Minneapolis, Minn.) could be used.


Preparation of Solid Substrates of the Present Invention


Glass substrate (such as microscope coverslips from Deckglaser, Germany) were cleaned with 10% DECON 90 (Decon Laboratories Limited, England), rinsed with Milli-Q water, dried under nitrogen gas, and exposed to oxygen plasma in a plasma cleaner (Harrick Plasma, Ithaca, N.Y., U.S.A.) at 100 mtorr for 10 min. Prior to each use, the glass substrate was rinsed with ethanol and dried under nitrogen gas.


Silicon oxide based solid substrates (e.g. silicon wafer or glass coverslips) were cleaned with piranha solution (70% sulfuric acid and 30% hydrogen peroxide (v/v)) at 120° C. for 40 min, subsequently washed with distilled water and rinsed with acetone. The solid substrates were dried under a stream of nitrogen and treated with a plasma cleaner.


For the vapor phase silanization reaction, clean silicon oxide substrates and a Petri-dish containing 150 uL of 3-(aminopropyl)-triethoxysilane (Sigma, USA) were placed in a desiccator (Wheaton dry-seal desiccator, 100 nm) under reduced pressure at ˜0.3 Torr for 16 h. The substrates were cleaned by acetone and dried under nitrogen stream.


Construction of the SLB Surface Coating on a Solid Substrate


0.25 mg/ml of POPC/b-PE vesicle solution described above was added to the cleaned solid substrate to form a SLB coated solid substrate. This was followed by an extensive rinse with a phosphate buffer containing 10 mM PBS and 150 mM NaCl (pH=7.2) to remove excess POPC/b-PE vesicles. Biotin was the functional group in the SLB which binds with the functional group (streptavidin) in the linker composition.


0.1 mg/mL of streptavidin (SA) solution (commercially available from Pierce Biotechnology. Rockford, Ill., USA) was added to the SLB coated solid substrate and incubated for 1 hour, followed with a PBS buffer rinse to remove excess SA.


About 0.05 mg/mL of b-Anti-EpCAM solution was added to the SA-SLB coated solid substrate to form the surface coating of the present invention.


Construction of the PEG Surface Coating on a Solid Substrate


The biotinylated PEG silane solution (Si-bPEGs) was added to the clean glass substrate and incubated for 1 hour to form a Si-bPEG nonfouling composition on the glass substrate, followed by an ethanol rinse to remove excess Si-bPEGs. Silane was the surface linker and the biotin was the functional group that bind with the functional group (SA) in the linker composition.


0.1 mg/mL of SA solution was added to the Si-bPEGs coated solid substrate and incubated for 1 hour, followed by a PBS buffer rinse to remove excess SA.


0.05 mg/mL of b-Anti-EpCAM solution was added and bound with SA-Si-bPEGs surface coating, followed by PBS buffer rinse to remove excess b-Anti-EpCAM.


Construction of the PEM Surface Coating on a Solid Substrate


Physical deposition of PEM films was performed by batch and static conditions as follows: initially, all polypeptides were dissolved in 10 mM Tris-HCl buffer with 0.15 M NaCl, pH 7.4. Solid substrates were then immersed in PLL (MW 15000-30000; Sigma, St Louis, Mo.) solution (1 mg/mL) for 10 min at room temperature, followed by rinsing with 1 mL of Tris-HCl buffer for 1 min. To couple PLGA, the PLL-coated slide was subsequently immersed in the PLGA solution (MW 3000-15000, Sigma, St Louis, Mo., 1 mg/mL) for 10 min, followed by rinsing with 1 mL of Tris-HCl buffer for 1 min. Lastly, substrates were cleaned with fresh PBS to remove uncoupled polypeptides. The resulting c-(PLL/PLGA)i, where i was denoted as the number of polyelectrolyte pairs generated by repeating the above steps: i) 0.5 was referred to c-PLL only, i) 1 was referred to c-(PLL/PLGA)1, and the like.


QCM-D Characterization of the SLB Surface Coating


The construction of the surface coating was monitored by quartz crystal microbalance with dissipation (QCM-D). The QCM-D response in FIG. 11 shows the construction of the surface coating on a SiO2-pretreated quartz crystal. First, 0.25 mg/mL of POPC/b-PE vesicle mixture (in phosphate buffer) was dispensed into the QCM chamber at point (I). The normalized frequency change F and dissipation shift D were 26.0±0.7 Hz and 0.19±0.03×10-6 respectively, which are the characteristics of a highly uniformed lipid bilayer. After two buffer washes (denoted as *), 0.1 mg/mL of SA solution was dispensed at point II. • SA binding was saturated at F=52.8±5.4 Hz and D=3.84±0.54×10-6. At point (III), 0.025 mg/mL of OC98-1 antibody solution was dispensed into the QCM chamber and there was no frequency or dissipation change. This shows there was no interaction between the OC98-1 antibody and the SA-lipid bilayer surface. In contrast, adding biotinylated antibody solution (bOC98-1 or bEpAb4-1) at point (IV) resulted in frequency and dissipation change, with equilibrated shifts of F=39.4±6.8 Hz and D=1.63±0.28×10-6. This demonstrates the binding of biotinylated antibody to SA-lipid bilayer surface.


The characteristics of the SLB nonfouling composition on the surface coating were examined using QCM-D (FIG. 12). Bovine serum albumin (BSA, commercially available from Sigma-Aldrich. USA) was added to the surface coating and there was a sudden change in frequency and dissipation, with equilibrated shifts of F=6.9 Hz and D=3.35×10-6. This indicates an immediate BSA adsorption. Three buffer rinses (*) caused an increase in frequency and a decrease in dissipation, with saturated shifts of F=6.1 Hz and D=3.16×10-6. This indicates the adsorbed BSA can be easily removed from the surface coating and thus, a very weak interaction between BSA and SLB.


Example 2: Preparation of the Microfluidic Chip

The microfluidic chip can be prepared by the following steps:


1. A commercial CO2 laser scriber (Helix 24, Epilog, USA) was used to engrave the microtrenches to form microstructures on the PMMA substrate.


2. The PMMA substrate, glass substrate and nuts were cleaned with MeOH, detergent and water, followed by 10 min sonication. The nuts and the solid substrates were dried by nitrogen gas and baked for 10 min at 60° C.


3. The PMMA substrate was bonded with nuts by chloroform treatment.


4. PMMA substrate and the glass slide were joined together using an adhesive (e.g. 3M doubled sided tape from 3M, USA).


Example 3: CTCs Binding to the Anti-EnCAM Functionalized SLB Surface Coating

Eight blood samples were used to determine the CTC capture rate of the Anti-EpCAM functionalized SLB surface coating in a microfluidic chip in Example 2. Each blood sample contained 2 ml of blood from a stage IV colon cancer patient and the sample was introduced to the sealed channel of the microfluidic chip at 0.5 ml/hr, controlled by a syringe pump. Subsequently, the sealed channel in the microfluidic chip was rinsed with 0.5 ml of PBS buffer at the flow rate of 1 ml/h, followed by in situ immunostaining.


The number of CTCs captured per ml of blood for these 8 samples were 26, 34, 36, 39, 47, 67 79, and 99, 25% of the blood samples had 79 or higher CTC count per ml of testing sample and the median CTC count was 43 per ml of testing sample. There was minimal binding of the non-specific cells and proteins after the buffer rinse.


As a comparison, the CTC count for the FDA approved Veridex CellSearch is as follows: 25% of the samples had 3 or more CTCs per 7.5 ml of testing sample and the median CTC counts was 0.


The anti-EpCAM functionalized SLB surface was incubated with 150 uL of HCT116 cancer cell spiked human blood (with HCT116 cancer cell density of approximately 10 to 100 per 100 μL of blood), followed by a buffer rinse to remove non-specific cells. FIG. 13 shows the surface coating before and after the buffer rinse. Prior to the buffer rinse, the surface coating was covered with non-specific cells (upper left) and four HCT116 cancer cells (lower left). After the buffer rinse, almost all of the non-specific cells were removed (upper right) but the four HCT116 cancer cell (lower right) remained on the surface coating.


The results show the surface coating of the present invention is effective in capturing CTCs and releasing the non-specific cells.


Example 4: Comparison of Capture Efficiency and Nonfouling Property of Various Surface Conditions

The capture rate of HCT116 cancer cells (biological substance) and the nonfouling property of six different surface conditions are illustrated in FIG. 14A.


The results show that the surface coatings of the present invention (lipid/SA/b-anti-EpCAM and PEG (15 mM)/SA/b-anti-EpCAM) are more effective in capturing the biological substance. There is less binding of the non-specific cells (white blood cells or WBC) on the surface coatings of the present invention compare to a surface coating without a nonfouling composition (glass only).



FIG. 14B shows the non-specific blood cell binding of the following surfaces: (A) Glass only; (B) biotinylated SLB (b-SLB), (C) Streptavidin conjugated-bSLB, and (D) OC98-1-conjugated bSLB. These surfaces were incubated with diluted human blood from healthy donor (1 uL of blood in 100 uL PBS buffer) for 4 hours, followed by a PBS buffer rinse. Images (E) to (H) are the after rinse images which correspond to the surface coatings in (A) to (D). The results show that after a buffer rinse, there is less non-specific blood cell on the surface coatings with a releasable composition (i.e. SLB) compare to the surface coating without a releasable composition (i.e. glass only).


Example 5: Purification by Flow

The differentiated flow shear could selectively “flush” out the non-specific cells based on the affinity of these cells to the nonfouling composition, while the biological substance remains on the surface coating.


In this study, the surface coating comprised a SLB, a linker composition and fibronectin as the bioactive composition. FIG. 15A shows fibroblast 3T3 (green) and colon cancer cell line HCT116 (red) were incubated on the surface coating for 4 h. The surface coating was rinsed with a buffer solution, which has a shear stress of 3 dyne/cm2.


The HCT 116 cells (red) were flushed away from the surface coating within 5 min of the buffer rinse, as shown in FIG. 15B. The fibroblast 3T3 cells (green) remained on the surface coating after 30 min of buffer rinse, as shown in FIG. 15C, due to its high affinity to fibronectin.


The result shows a shear stress about 3 dyne/cm2 is sufficient to remove the non-specific cells from the releasable composition.



FIG. 16 summarizes the respective shear stress and flushing time for the HCT116 and NIH-3T3 cell populations (non-specific cells). To remove HCT116 cells from the releasable compostion of the surface coating, the shear stress is about 3 to about 4.5 dyne/cm2. To remove NIH-3T3 cells from the releasable composition of the surface coating, the shear stress is about 8.5 to about 12 dyne/cm2 (N/N0 is the percentage of the cells remains attached to the surface coating using various shear stresses. N is the final cell number and NO is the initial cell number.)


Example 6: Release of CTCs from the Surface Coating

The captured HCT116 cancer cells on the surface coating in Example 3 were released by introducing air bubbles. FIG. 17 shows HCT116 cells in the red circle were removed from the surface coating within 3 seconds of introducing air bubbles.


Example 7: Culture of Released CTCs from the Surface Coating

The captured CTCs were incubated with 5 mM of EDTA at 37° C. for 5 to 10 min and released by flowing a culture medium into the sealed channel of the microfluidic chip. A total of 18 colo205 cells were released from this procedure. The released colo205 cells, together with a serum-containing culture medium and antibiotics (penicillin+streptomycin+gentamicin), were placed into a 48-well tissue cultured polystyrene plate for cultivation.



FIGS. 18 A-18C show a portion of 18 colo205 cells on day 1, on day 10 and day 14 respectively. This study demonstrates the released colo205 cells retained their viability for subsequent cell culture.


Example 8: Capture CTCs Through a CTC Filtration Device

Any membranes, tubes, capillaries, beads, nanoparticles or channels can be coated with the surface coating of the present invention. FIG. 19 illustrates schematically a filtration device, wherein the filtration filter is coated with the surface coating of the present invention. The filter could accommodate high volume blood flow and capture a biological substance for a diagnostic or therapeutic purpose. To access the patient's blood or body fluid, a catheter can be inserted into the patient's vein or fistula and the patient's blood flows through the CTC filtration device, wherein the surface coating on the filters captures the CTCs. The filtered blood flows back to the patient.


Example 9: Capture CTCs Through a Biotinylated EPAb4-1 Antibody

The binding specificity of biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody and biotinylated EpCam antibody (commercially available from R&D system, USA) were examined using the HCT116 (colorectal) CTCs and SAS (tongue) CTCs.


The CTCs were spiked in a buffer solution (about 105 CTCs/ml). The CTC-spiked buffer solution was introduced to the surface coatings with the following bioactive composition: biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgG antibody.


The CTC binding specificy of the antibodies was determined by colorimetric method, by measuring the absorption optical density at 490 nm. FIG. 20 shows biotinylated EpAb 4-1 is effective in capturing HCT116 CTCs and SAS CTCs.

Claims
  • 1. A microfluidic chip for selectively enriching rare cells, comprising: a first solid substrate and a second solid substrate, wherein the first solid substrate or the second solid substrate comprises a series of microstructures configured to interact with cells, wherein the series of microstructures are perpendicular to a flow direction of the microfluidic chip, and wherein the first solid substrate and the second solid substrate are configured to be bound parallel to one another; anda surface coating for capturing the rare cells, wherein the surface coating comprises (i) a non-fouling composition that reduces binding of non-specific cells or adsorption of serum proteins compared to a surface coating lacking the non-fouling composition and (ii) a bioactive composition which selectively binds to the rare cells, wherein the bioactive composition is an antibody, wherein the non-fouling composition of the surface coating is non-covalently associated with the bioactive composition and wherein the surface coating is attached to the first or second solid substrate by a non-covalent interaction,wherein the first solid substrate or the second solid substrate comprises the surface coating, and wherein said non-fouling composition comprises a supported lipid bilayer.
  • 2. The microfluidic chip of claim 1, wherein the series of microstructures comprises microstructures arranged in a linear pattern such that microstructures in a first pair of adjacent rows have a distance separating the adjacent rows, thereby forming a first gap, and wherein said first gap is not in line with a second gap formed by a second pair of adjacent rows, wherein said first gap and second gap are in adjacent columns.
  • 3. The microfluidic chip of claim 1, wherein the antibody is a biotinylated EpCAM antibody.
  • 4. The microfluidic chip of claim 1, wherein the first solid substrate comprises a plastic substrate and the second solid substrate comprises a glass substrate.
  • 5. The microfluidic chip of claim 4, wherein the glass substrate is located below the plastic substrate in a working configuration.
  • 6. The microfluidic chip of claim 1, wherein the first solid substrate comprises the series of microstructures, and wherein the first solid substrate is located above the second solid substrate in a working configuration.
  • 7. The microfluidic chip of claim 1, further comprising an adhesive for bonding the first solid substrate to the second solid substrate.
  • 8. The microfluidic chip of claim 7, wherein the adhesive comprises an inner hollow opening in a form of a channel.
  • 9. The microfluidic chip of claim 8, wherein the inner hollow opening in a form of a channel is configured to encompass the series of microstructures.
  • 10. The microfluidic chip of claim 8, wherein the inner hollow opening in a form of a channel determines a path for the rare cells to travel through the microfluidic chip.
  • 11. The microfluidic chip of claim 7, wherein a thickness of the adhesive determines a height of a channel of the microfluidic chip.
  • 12. The microfluidic chip of claim 1, wherein the antibody comprises a heavy chain and a light chain that binds EpCAM, wherein (a) the heavy chain comprises CDR1, CDR2, and CDR3 of SEQ ID NO: 1, and (b) the light chain comprises CDR1, CDR2, and CDR3 of SEQ ID NO: 2.
  • 13. The microfluidic chip of claim 1, further comprising a syringe pump, wherein the syringe pump is configured to apply buffer at a flow rate configured to release non-specific cells from the non-fouling layer without releasing cells selectively bound to the bioactive composition.
  • 14. The microfluidic chip of claim 1, further comprising a syringe pump configured to aid rinsing the microfluidic chip with a buffer at a shear force of about 2.5 to about 10 dyne/cm2.
  • 15. The microfluidic chip of claim 1, wherein the non-fouling composition is coupled to the first solid substrate by a linker, and wherein the non-fouling composition is coupled to the second solid substrate by a linker.
  • 16. The microfluidic chip of claim 1, wherein the non-covalent interaction is selected from the group consisting of hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, magnetic force, and combinations thereof.
  • 17. The microfluidic chip of claim 1, wherein the first solid substrate is completely coated by the non-fouling composition, and wherein the second solid substrate is completely coated by the non-fouling composition.
  • 18. The microfluidic chip of claim 1, further comprising (i) a rare cell bound to the bioactive composition, and (ii) a liquid which comprises an air bubble, wherein the air bubble disrupts an interaction of the non-fouling composition with the first solid substrate or the second solid substrate.
US Referenced Citations (424)
Number Name Date Kind
3784015 Kasten Jan 1974 A
5147606 Charlton et al. Sep 1992 A
5554686 Frisch, Jr. et al. Sep 1996 A
5646001 Terstappen et al. Jul 1997 A
5652148 Doshi et al. Jul 1997 A
5707799 Hansmann et al. Jan 1998 A
5837115 Austin et al. Nov 1998 A
5842787 Kopf-Sill et al. Dec 1998 A
5885470 Parce et al. Mar 1999 A
5952173 Hansmann et al. Sep 1999 A
6039897 Lochhead et al. Mar 2000 A
6046295 Frisch, Jr. et al. Apr 2000 A
6153113 Goodrich et al. Nov 2000 A
6271309 Roberts et al. Aug 2001 B1
6280622 Goodrich et al. Aug 2001 B1
6322683 Wolk et al. Nov 2001 B1
6361749 Terstappen et al. Mar 2002 B1
6365362 Terstappen et al. Apr 2002 B1
6372542 Martin et al. Apr 2002 B1
6562616 Toner et al. May 2003 B1
6613525 Nelson et al. Sep 2003 B2
6620627 Liberti et al. Sep 2003 B1
6623982 Liberti et al. Sep 2003 B1
6632652 Austin et al. Oct 2003 B1
6645731 Terstappen et al. Nov 2003 B2
6685841 Lopez et al. Feb 2004 B2
6699952 Chaikof et al. Mar 2004 B2
6790366 Terstappen et al. Sep 2004 B2
6790599 Madou Sep 2004 B1
6844028 Mao et al. Jan 2005 B2
6887578 Gleason et al. May 2005 B2
6890426 Terstappen et al. May 2005 B2
6955738 Derand et al. Oct 2005 B2
6960449 Wang et al. Nov 2005 B2
7005493 Huang et al. Feb 2006 B2
7056657 Terstappen et al. Jun 2006 B2
7067194 Mao et al. Jun 2006 B2
7117807 Bohn et al. Oct 2006 B2
7150812 Huang et al. Dec 2006 B2
7190818 Ellis et al. Mar 2007 B2
7229760 Zohlnhofer et al. Jun 2007 B2
7276170 Oakey et al. Oct 2007 B2
7282350 Rao et al. Oct 2007 B2
7318902 Oakey et al. Jan 2008 B2
7332288 Terstappen et al. Feb 2008 B2
7368163 Huang et al. May 2008 B2
7374944 Thompson et al. May 2008 B2
7428325 Douglass et al. Sep 2008 B2
7431969 Gleason et al. Oct 2008 B2
7442515 Ratner et al. Oct 2008 B2
7472794 Oakey et al. Jan 2009 B2
7485343 Branson et al. Feb 2009 B1
7501157 Mao et al. Mar 2009 B2
7531120 Van Rijn et al. May 2009 B2
7579077 Dubrow et al. Aug 2009 B2
7588550 Leonard et al. Sep 2009 B2
7629029 Mao et al. Dec 2009 B2
7687241 Chen et al. Mar 2010 B2
7695775 Kobrin et al. Apr 2010 B2
7713689 Chilkoti May 2010 B2
7723112 Clarke et al. May 2010 B2
7727399 Leonard et al. Jun 2010 B2
7735652 Inglis et al. Jun 2010 B2
7736891 Nelson et al. Jun 2010 B2
7777010 Logtenberg Aug 2010 B2
7783098 Douglass et al. Aug 2010 B2
7785810 Chen et al. Aug 2010 B2
RE41762 Lopez et al. Sep 2010 E
7815922 Chaney et al. Oct 2010 B2
7846393 Tai et al. Dec 2010 B2
7846445 Schellenberger et al. Dec 2010 B2
7846743 Tai et al. Dec 2010 B2
7850633 Leonard et al. Dec 2010 B2
7855068 Cao Dec 2010 B2
7855279 Schellenberger et al. Dec 2010 B2
7863012 Rao et al. Jan 2011 B2
7879444 Jiang et al. Feb 2011 B2
RE42249 Lopez et al. Mar 2011 E
7901950 Connelly et al. Mar 2011 B2
RE42315 Lopez et al. May 2011 E
7955704 Lowery et al. Jun 2011 B2
7960166 Vacanti et al. Jun 2011 B2
7973136 Lazar et al. Jul 2011 B2
7981688 Stayton et al. Jul 2011 B2
7985475 Dubrow Jul 2011 B2
7988840 Huang et al. Aug 2011 B2
7993821 Chiu et al. Aug 2011 B2
8008032 Forsyth et al. Aug 2011 B2
8012480 Lorence Sep 2011 B2
8021318 Leonard et al. Sep 2011 B2
8021614 Huang et al. Sep 2011 B2
8025854 Ohman et al. Sep 2011 B2
8057418 Korbling et al. Nov 2011 B2
8063187 Chu et al. Nov 2011 B2
D650091 Odeh Dec 2011 S
8069782 Fragala et al. Dec 2011 B2
8083706 Leonard et al. Dec 2011 B2
8092684 Leonard et al. Jan 2012 B2
8093365 Wisniewski et al. Jan 2012 B2
8097153 Leonard et al. Jan 2012 B2
8097162 Leonard et al. Jan 2012 B2
8101720 Lazar et al. Jan 2012 B2
8158410 Tang et al. Apr 2012 B2
8158728 Desimone et al. Apr 2012 B2
8178602 Mao et al. May 2012 B2
8186913 Toner et al. May 2012 B2
8282799 Huang et al. Oct 2012 B2
8288116 Chen et al. Oct 2012 B2
8288170 Tai et al. Oct 2012 B2
8304230 Toner et al. Nov 2012 B2
8308699 Zhang et al. Nov 2012 B2
8333934 Cao et al. Dec 2012 B2
8343440 Yoshioka Jan 2013 B2
8357528 Vacanti et al. Jan 2013 B2
8367314 Chilkoti Feb 2013 B2
8372579 Toner et al. Feb 2013 B2
8414806 Sun et al. Apr 2013 B2
8445225 Kuhn et al. May 2013 B2
8481336 Earhart et al. Jul 2013 B2
8491516 Leonard et al. Jul 2013 B2
8507283 Stayton et al. Aug 2013 B2
8545983 Jiang et al. Oct 2013 B2
8557528 Hauch et al. Oct 2013 B2
8557577 Hauch et al. Oct 2013 B2
8574660 Weaver et al. Nov 2013 B2
8579117 Sturm et al. Nov 2013 B2
8632838 Roth et al. Jan 2014 B2
8663625 Stroock et al. Mar 2014 B2
8669044 Chiu et al. Mar 2014 B2
8796184 Chilkoti et al. Aug 2014 B2
8821812 Ohman et al. Sep 2014 B2
8822231 Melin et al. Sep 2014 B2
8835144 Jiang et al. Sep 2014 B2
8895298 Toner et al. Nov 2014 B2
8911957 Irimia et al. Dec 2014 B2
8921102 Fuchs et al. Dec 2014 B2
8980568 Lin et al. Mar 2015 B2
8986966 Toner et al. Mar 2015 B2
8986988 Karnik et al. Mar 2015 B2
9016221 Brennan et al. Apr 2015 B2
9056318 Bergman et al. Jun 2015 B2
9140697 Tseng et al. Sep 2015 B2
9174222 Huang et al. Nov 2015 B2
9494500 Chang et al. Nov 2016 B2
9541480 Chang et al. Jan 2017 B2
10107726 Shao et al. Oct 2018 B2
10112198 Chang et al. Oct 2018 B2
10495644 Chang et al. Dec 2019 B2
10605708 Shao Mar 2020 B2
20010031309 Lee et al. Oct 2001 A1
20010036556 Jen Nov 2001 A1
20020009759 Terstappen et al. Jan 2002 A1
20020055093 Abbott et al. May 2002 A1
20020098535 Wang et al. Jul 2002 A1
20020119482 Nelson et al. Aug 2002 A1
20020125192 Lopez et al. Sep 2002 A1
20020141913 Terstappen et al. Oct 2002 A1
20020160139 Huang et al. Oct 2002 A1
20020182633 Chen et al. Dec 2002 A1
20030022216 Mao et al. Jan 2003 A1
20030071525 Tong et al. Apr 2003 A1
20030087338 Messersmith et al. May 2003 A1
20030096226 Logtenberg May 2003 A1
20030129676 Terstappen et al. Jul 2003 A1
20030134416 Yamanishi et al. Jul 2003 A1
20030138645 Gleason et al. Jul 2003 A1
20030157054 Gillies Aug 2003 A1
20030159999 Oakey et al. Aug 2003 A1
20030163084 Griffiths et al. Aug 2003 A1
20030206901 Chen Nov 2003 A1
20030213551 Derand et al. Nov 2003 A1
20030216534 Chaikof et al. Nov 2003 A1
20040004043 Terstappen et al. Jan 2004 A1
20040009471 Cao Jan 2004 A1
20040028875 Van Rijn et al. Feb 2004 A1
20040038339 Kufer et al. Feb 2004 A1
20040053334 Ratner et al. Mar 2004 A1
20040072269 Rao et al. Apr 2004 A1
20040109853 McDaniel Jun 2004 A1
20040115721 Mao et al. Jun 2004 A1
20040118757 Terstappen et al. Jun 2004 A1
20040175407 McDaniel Sep 2004 A1
20040225249 Leonard et al. Nov 2004 A1
20040254419 Wang et al. Dec 2004 A1
20050025797 Wang et al. Feb 2005 A1
20050042766 Ohman et al. Feb 2005 A1
20050058576 Pranis et al. Mar 2005 A1
20050079132 Wang et al. Apr 2005 A1
20050100675 Mao et al. May 2005 A1
20050107870 Wang et al. May 2005 A1
20050147758 Mao et al. Jul 2005 A1
20050153342 Chen Jul 2005 A1
20050175501 Thompson et al. Aug 2005 A1
20050178286 Bohn et al. Aug 2005 A1
20050181195 Dubrow Aug 2005 A1
20050181463 Rao et al. Aug 2005 A1
20050186685 Kange et al. Aug 2005 A1
20050215764 Tuszynski et al. Sep 2005 A1
20050230272 Lee et al. Oct 2005 A1
20050255327 Chaney et al. Nov 2005 A1
20050265980 Chen et al. Dec 2005 A1
20050267440 Herman et al. Dec 2005 A1
20050288398 Messersmith et al. Dec 2005 A1
20060002825 Derand et al. Jan 2006 A1
20060009550 Messersmith et al. Jan 2006 A1
20060014013 Saavedra et al. Jan 2006 A1
20060057180 Chilkoti et al. Mar 2006 A1
20060076295 Leonard et al. Apr 2006 A1
20060079740 Silver et al. Apr 2006 A1
20060088666 Kobrin et al. Apr 2006 A1
20060093836 Huang et al. May 2006 A1
20060134599 Toner et al. Jun 2006 A1
20060137438 Lenzing et al. Jun 2006 A1
20060159916 Dubrow et al. Jul 2006 A1
20060160066 Bhatia et al. Jul 2006 A1
20060166183 Short et al. Jul 2006 A1
20060169642 Oakey et al. Aug 2006 A1
20060173394 Stroock et al. Aug 2006 A1
20060194192 Rao et al. Aug 2006 A1
20060237390 King et al. Oct 2006 A1
20060251795 Kobrin et al. Nov 2006 A1
20060252046 Short et al. Nov 2006 A1
20060252054 Lin et al. Nov 2006 A1
20060254972 Tai et al. Nov 2006 A1
20060285996 Ohman et al. Dec 2006 A1
20070003549 Ignatovich et al. Jan 2007 A1
20070010702 Wang et al. Jan 2007 A1
20070025883 Tai et al. Feb 2007 A1
20070026381 Huang et al. Feb 2007 A1
20070026416 Fuchs Feb 2007 A1
20070026469 Fuchs et al. Feb 2007 A1
20070032620 Gleason et al. Feb 2007 A1
20070037173 Allard et al. Feb 2007 A1
20070048859 Sears Mar 2007 A1
20070059716 Balis et al. Mar 2007 A1
20070071762 Ts'o et al. Mar 2007 A1
20070072220 Chilkoti Mar 2007 A1
20070077276 Haynie Apr 2007 A1
20070122406 Chamberlain et al. May 2007 A1
20070131622 Oakey et al. Jun 2007 A1
20070154960 Connelly et al. Jul 2007 A1
20070160502 Hwang Jul 2007 A1
20070172903 Toner et al. Jul 2007 A1
20070178133 Rolland Aug 2007 A1
20070187250 Huang et al. Aug 2007 A1
20070202536 Yamanishi et al. Aug 2007 A1
20070231851 Toner et al. Oct 2007 A1
20070259424 Toner et al. Nov 2007 A1
20070264675 Toner et al. Nov 2007 A1
20070266777 Bergman et al. Nov 2007 A1
20070281353 Vacanti et al. Dec 2007 A1
20080009780 Leonard et al. Jan 2008 A1
20080023399 Inglis et al. Jan 2008 A1
20080026486 Cooper et al. Jan 2008 A1
20080090239 Shoemaker et al. Apr 2008 A1
20080113350 Terstappen May 2008 A1
20080114096 Qu et al. May 2008 A1
20080131425 Garcia et al. Jun 2008 A1
20080147178 Pacetti et al. Jun 2008 A1
20080149566 Messersmith et al. Jun 2008 A1
20080176271 Silver et al. Jul 2008 A1
20080181861 Jiang et al. Jul 2008 A1
20080188638 Breitenkamp et al. Aug 2008 A1
20080206757 Lin et al. Aug 2008 A1
20080207913 Breitenkamp et al. Aug 2008 A1
20080213853 Garcia et al. Sep 2008 A1
20080220531 Stayton et al. Sep 2008 A1
20080241892 Roitman et al. Oct 2008 A1
20080248499 Chiu et al. Oct 2008 A1
20080255305 Brook et al. Oct 2008 A1
20080274335 Bowman et al. Nov 2008 A1
20080311182 Ferrari et al. Dec 2008 A1
20080312356 Kobrin et al. Dec 2008 A1
20090020431 Voccia et al. Jan 2009 A1
20090029043 Rong et al. Jan 2009 A1
20090036982 Aharoni et al. Feb 2009 A1
20090060791 Hagiwara et al. Mar 2009 A1
20090068760 Nelson et al. Mar 2009 A1
20090093610 Textor et al. Apr 2009 A1
20090098017 Celik-Butler et al. Apr 2009 A1
20090105463 Berry et al. Apr 2009 A1
20090114344 Barinov et al. May 2009 A1
20090117574 Labgold et al. May 2009 A1
20090136982 Tang et al. May 2009 A1
20090139931 Leonard et al. Jun 2009 A1
20090142772 Lau et al. Jun 2009 A1
20090156460 Jiang et al. Jun 2009 A1
20090181441 Jin et al. Jul 2009 A1
20090203536 Vermette et al. Aug 2009 A1
20090215088 Forsyth et al. Aug 2009 A1
20090226499 Wisniewski et al. Sep 2009 A1
20090247424 Chilkoti et al. Oct 2009 A1
20090259015 Jiang et al. Oct 2009 A1
20090259302 Trollsas et al. Oct 2009 A1
20090263457 Trollsas et al. Oct 2009 A1
20090264317 Ofir et al. Oct 2009 A1
20090269323 Luk et al. Oct 2009 A1
20090281250 Desimone et al. Nov 2009 A1
20090285873 Lim et al. Nov 2009 A1
20090292234 Leonard et al. Nov 2009 A1
20090298067 Irimia et al. Dec 2009 A1
20090311734 Greve et al. Dec 2009 A1
20090317836 Kuhn et al. Dec 2009 A1
20100004578 Leonard et al. Jan 2010 A1
20100028526 Martin et al. Feb 2010 A1
20100055733 Lutolf et al. Mar 2010 A1
20100059414 Sturm et al. Mar 2010 A1
20100061892 Flaim et al. Mar 2010 A1
20100062156 Kurth et al. Mar 2010 A1
20100063570 Pacetti et al. Mar 2010 A1
20100081735 Mao et al. Apr 2010 A1
20100092393 Haghgooie et al. Apr 2010 A1
20100092491 Anastasi Apr 2010 A1
20100096327 Gin et al. Apr 2010 A1
20100099160 Jiang et al. Apr 2010 A1
20100099579 Chilkoti Apr 2010 A1
20100112026 Karp May 2010 A1
20100118642 Ho et al. May 2010 A1
20100137984 Lowery et al. Jun 2010 A1
20100140160 Dubrow et al. Jun 2010 A1
20100143438 Todd et al. Jun 2010 A1
20100143741 Bell et al. Jun 2010 A1
20100145286 Zhang et al. Jun 2010 A1
20100151491 Himmelhaus et al. Jun 2010 A1
20100152708 Li et al. Jun 2010 A1
20100159462 Takayama et al. Jun 2010 A1
20100160645 Breitenkamp et al. Jun 2010 A1
20100169990 Clarke et al. Jul 2010 A1
20100173402 Chen Jul 2010 A1
20100198131 Leonard et al. Aug 2010 A1
20100209612 Rong et al. Aug 2010 A1
20100210745 Mcdaniel et al. Aug 2010 A1
20100226943 Brennan et al. Sep 2010 A1
20100233146 Mcdaniel Sep 2010 A1
20100233693 Kopf-Sill et al. Sep 2010 A1
20100233694 Kopf-Sill et al. Sep 2010 A1
20100233812 Sun et al. Sep 2010 A1
20100247492 Kuhn et al. Sep 2010 A1
20100247760 Houben et al. Sep 2010 A1
20100248334 Mcdaniel Sep 2010 A1
20100248358 Yoshioka Sep 2010 A1
20100273991 Luk et al. Oct 2010 A1
20100278892 Krauland et al. Nov 2010 A1
20100279321 Chiu et al. Nov 2010 A1
20100280252 Breitenkamp et al. Nov 2010 A1
20100285581 Hauch et al. Nov 2010 A1
20100285972 Dubrow et al. Nov 2010 A1
20100294146 Fragala et al. Nov 2010 A1
20100304485 Karnik et al. Dec 2010 A1
20100311599 Wheeler et al. Dec 2010 A1
20100316842 Tuteja et al. Dec 2010 A1
20100323918 Huang et al. Dec 2010 A1
20100330025 Messersmith et al. Dec 2010 A1
20100331965 Dugas et al. Dec 2010 A1
20110005997 Kurth et al. Jan 2011 A1
20110008404 Lyon et al. Jan 2011 A1
20110027803 Moussavi et al. Feb 2011 A1
20110048947 Petronis et al. Mar 2011 A1
20110054347 Goss et al. Mar 2011 A1
20110056884 Leonard et al. Mar 2011 A1
20110059468 Earhart et al. Mar 2011 A1
20110062083 Leonard et al. Mar 2011 A1
20110066097 Leonard et al. Mar 2011 A1
20110091864 Karlsson et al. Apr 2011 A1
20110097277 Jiang et al. Apr 2011 A1
20110105712 Jiang et al. May 2011 A1
20110105982 Leonard et al. May 2011 A1
20110117674 Melin et al. May 2011 A1
20110143119 Bell et al. Jun 2011 A1
20110165161 Lin et al. Jul 2011 A1
20110165415 Ma et al. Jul 2011 A1
20110171663 Smith et al. Jul 2011 A1
20110192233 Aizenberg et al. Aug 2011 A1
20110195104 Jiang et al. Aug 2011 A1
20110212085 Joseloff et al. Sep 2011 A1
20110212297 Dhinojwala et al. Sep 2011 A1
20110212440 Viovy et al. Sep 2011 A1
20110217449 Lowery et al. Sep 2011 A1
20110224383 Sill et al. Sep 2011 A1
20110236904 Hauch et al. Sep 2011 A1
20110240064 Wales et al. Oct 2011 A1
20110240595 Dubrow Oct 2011 A1
20110250626 Williams et al. Oct 2011 A1
20110250679 Chang Oct 2011 A1
20110256619 Vacanti et al. Oct 2011 A1
20110266492 Stayton et al. Nov 2011 A1
20110275530 Walfish et al. Nov 2011 A1
20110282005 Jiang et al. Nov 2011 A1
20110294186 Fuchs et al. Dec 2011 A1
20110300551 Rao et al. Dec 2011 A1
20110300603 Forsyth et al. Dec 2011 A1
20110301442 Luecke et al. Dec 2011 A1
20110305660 Stayton et al. Dec 2011 A1
20110305872 Li et al. Dec 2011 A1
20110305881 Schultz et al. Dec 2011 A1
20110305895 Roth et al. Dec 2011 A1
20110305898 Zhang et al. Dec 2011 A1
20110305909 Weaver et al. Dec 2011 A1
20120003711 Tseng et al. Jan 2012 A1
20120006728 Huang et al. Jan 2012 A1
20120015146 Advincula et al. Jan 2012 A1
20120015835 Fuchs et al. Jan 2012 A1
20120021200 Koberstein et al. Jan 2012 A1
20120028342 Ismagilov et al. Feb 2012 A1
20120037544 Lane et al. Feb 2012 A1
20120045828 Davis et al. Feb 2012 A1
20120052415 Fragala et al. Mar 2012 A1
20120058302 Eggenspieler et al. Mar 2012 A1
20120058500 Mitchell et al. Mar 2012 A1
20120061304 Leonard et al. Mar 2012 A1
20120064150 Wisniewski et al. Mar 2012 A1
20120077246 Hong et al. Mar 2012 A1
20120094327 Young et al. Apr 2012 A1
20120114742 Martinez et al. May 2012 A1
20120178094 Kuhn Jul 2012 A1
20120196273 Huang et al. Aug 2012 A1
20120252022 Walfish et al. Oct 2012 A1
20120270209 Shah et al. Oct 2012 A1
20120301900 Kang et al. Nov 2012 A1
20130121895 Tang et al. May 2013 A1
20130143197 Heyneker Jun 2013 A1
20140017776 Kopf-Sill Jan 2014 A1
20140296095 Lin et al. Oct 2014 A1
20170199184 Chang et al. Jul 2017 A1
Foreign Referenced Citations (35)
Number Date Country
1646912 Jul 2005 CN
1731901 Feb 2006 CN
101701039 May 2010 CN
101765762 Jun 2010 CN
102011193 Apr 2011 CN
103261436 Aug 2013 CN
103998932 Aug 2014 CN
0783694 Nov 2003 EP
2359689 Aug 2011 EP
1569510 Nov 2011 EP
2359689 Aug 2015 EP
2427468 Mar 2011 GB
2472927 May 2011 GB
WO-9823948 Jun 1998 WO
WO-9920649 Apr 1999 WO
WO-2007048459 May 2007 WO
WO-2007079229 Jul 2007 WO
WO-2007079250 Jul 2007 WO
WO-2008157257 Dec 2008 WO
WO-2007079250 Mar 2009 WO
WO-2009051734 Apr 2009 WO
WO-2009088933 Jul 2009 WO
WO-2009140326 Nov 2009 WO
WO-2010123608 Oct 2010 WO
WO-2010124227 Oct 2010 WO
WO-2010132795 Nov 2010 WO
WO-2012016136 Feb 2012 WO
WO-2012094642 Jul 2012 WO
WO-2012103025 Aug 2012 WO
WO-2012116073 Aug 2012 WO
WO-2013003624 Jan 2013 WO
WO-2013006828 Jan 2013 WO
WO-2013036620 Mar 2013 WO
WO-2013131001 Sep 2013 WO
WO-2015153816 Oct 2015 WO
Non-Patent Literature Citations (114)
Entry
Sharma, H. et al. Apr. 2011, published online Dec. 9, 2010. Unconventional low-cost fabrication and patterning techniques for point of care diagnostics. Annals of Biomedical Engineering 39(4): 1313-1327; specif, pp. 1313, 1316, 1317.
Lawrence, M.B. et al. 1991. Leukocytes roll on a selectin at physiologic flow rates: distinction from and prerequisite for adhesion through integrins. Cell 65: 859-873.
Martinez, A.W. et al. 2008. Three-dimensional microfluidic devices fabricated in layered paper and tape. Proceedings of the National Academy of Sciences (PNAS) 105(50): 19606-19611.
Kaladhar, K. et al. 2004. Supported cell mimetic monolayers and their interaction with blood. Langmuir 20: 11115-11122; specif, pp. 11115, 11116.
Pappas, D. et al. 2007. Cellular separations: a review of new challenges in analytical chemistry. Analytics Chimica Acts 601: 26-35; specif, pp. 27, 29, 32.
Krasowska, M. et al. Wetting films in attachment of the colliding bubble. Advances in Colloid and Interface Sciences 134-135: 138-150; specif, pp. 138, 144, 145.
Johnson, S.J. et al. 1991. Structure ofsn adsorbed dimyristoylphosphstidylcholine bilayer measured with specular reflection of neutrons. Biophysics Journal 59: 289-294; specif, pp. 289, 293.
Co-pending U.S. Appl. No. 16/698,699, inventors Chang; Ying-Chih et al., filed Nov. 27, 2019.
Notice of allowance dated Sep. 1, 2016 for U.S. Appl. No. 14/128,354.
Notice of allowance dated Nov. 3, 2016 for U.S. Appl. No. 14/128,354.
Office action dated Mar. 23, 2016 for U.S. Appl. No. 14/128,354.
Park, et al. Continuous focusing of microparticles using inertial lift force and vorticity via multi-orifice microfluidic channels. Lab on a Chip 9.7 (2009): 939-948.
U.S. Appl. No. 15/378,938 Office Action dated Feb. 14, 2019.
U.S. Appl. No. 15/378,938 Office Action dated Oct. 29, 2019.
Adams, et al. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. J Am Chem Soc. Jul. 9, 2008;130(27):8633-41. doi: 10.1021/ja8015022. Epub Jun. 17, 2008.
Adams, et al. Integrated acoustic and magnetic separation in microfluidic channels. Appl Phys Lett. Dec. 21, 2009;95(25):254103.
Allard, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. Oct. 15, 2004;10(20):6897-904.
Balasubramanian, et al. Confocal images of circulating tumor cells obtained using a methodology and technology that removes normal cells. Mol Pharm. Sep.-Oct. 2009;6(5):1402-8. doi: 10.1021/mp9000519.
Balic, et al. Micrometastasis: detection methods and clinical importance. Cancer Biomarkers 9.1-6 (2011): 397-419.
Barkley, et al. Bubble-induced detachment of affinity-adsorbed erythrocytes. Biotechnol Appl Biochem. Oct. 2004;40(Pt 2):145-9.
Barradas, et al. Towards the biological understanding of CTC: capture technologies, definitions and potential to create metastasis. Cancers 5.4 (2013): 1619-1642.
Bhagat, et al. Continuous particle separation in spiral microchannels using Dean flows and differential migration. Lab Chip. Nov. 2008;8(11):1906-14. doi: 10.1039/b807107a. Epub Sep. 24, 2008.
Burgoyne. A method for rapid fabrication of microfluidic devices. Chips and Tips. Royal Society of Chemistry. (Jun. 30, 2009). Retrieved on Feb. 6, 2019, Retrieved from the internet: URL: https://blogs.rsc.org/chipsandtips/2009/06/30a-method-for-rapid-fabrication-of-microfluidic-devices/ pp. 1-6.
Cao, et al. Detachment strategies for affinity-adsorbed cells. Enzyme and microbial technology. 2002; 31: 153-160.
Cavalli, et al. Micro- and nanobubbles: a versatile non-viral platform for gene delivery. Int J Pharm. Nov. 18, 2013;456(2):437-45. doi: 10.1016/j.ijpharm.2013.08.041. Epub Sep. 2, 2013.
Cima, et al. Label-free isolation of circulating tumor cells in microfluidic devices: Current research and perspectives. Biomicrofluidics. Jan. 24, 2013;7(1):11810. doi: 10.1063/1.4780062. eCollection 2013.
Cohen, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. Jul. 1, 2008;26(19):3213-21. doi: 10.1200/JCO.2007.15.8923.
Cremer, et al. Writing and erasing barriers to lateral mobility into fluid phospholipid bilayers. Langmuir 15.11 (1999): 3893-3896.
Dainiak, et al. Cell chromatography: separation of different microbial cells using IMAC supermacroporous monolithic columns Biotechnol Prog. Mar.-Apr. 2005;21(2):644-9.
De Giorgi, et al. Application of a filtration- and isolation-by-size technique for the detection of circulating tumor cells in cutaneous melanoma. J Invest Dermatol. Oct. 2010;130(10):2440-7. doi: 10.1038/jid.2010.141. Epub Jun. 10, 2010.
Dharmasiri, et al. High-throughput selection, enumeration, electrokinetic manipulation, and molecular profiling of low-abundance circulating tumor cells using a microfluidic system. Anal Chem. Mar. 15, 2011;83(6):2301-9. doi: 10.1021/ac103172y. Epub Feb. 14, 2011.
Dickson, et al. Efficient capture of circulating tumor cells with a novel immunocytochemical microfluidic device. Biomicrofluidics. Sep. 2011;5(3):34119-3411915. doi: 10.1063/1.3623748. Epub Aug. 22, 2011.
Extended European Search Report and Search Opinion dated Feb. 28, 2017 for European Patent Application No. EP15773744.6.
Fehm, et al. Cytogenetic evidence that circulating epithelial cells in patients with carcinoma are malignant. Clin Cancer Res. Jul. 2002;8(7):2073-84.
Fehm, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective, multicenter trial. Breast Cancer Res Treat. Nov. 2010;124(2):403-12. doi: 10.1007/s10549-010-1163-x. Epub Sep. 22, 2010.
Garstecki, et al. Formation of droplets and bubbles in a microfluidic T-junction-scaling and mechanism of break-up. Lab Chip. Mar. 2006;6(3):437-46. Epub Jan. 25, 2006.
Geers, et al. Targeted liposome-loaded microbubbles for cell-specific ultrasound-triggered drug delivery. Small. Dec. 9, 2013;9(23):4027-35. doi: 10.1002/smll.201300161. Epub Jun. 5, 2013.
Glasmastar, et al. Protein adsorption on supported phospholipid bilayers. J Colloid Interface Sci. Feb. 1, 2002;246(1):40-7.
Gomez-Suarez, et al. Analysis of bacterial detachment from substratum surfaces by the passage of air-liquid interfaces. Appl Environ Microbiol. Jun. 2001;67(6):2531-7.
Hong, et al. Detecting circulating tumor cells: current challenges and new trends. Theranostics 3.6 (2013): 377-394.
“Hsiung, et al. A planar interdigitated ring electrode array via dielectrophoresis for uniform patterning of cells. Biosens Bioelectron. Dec. 1, 2008;24(4):869-875.”
Hsu, et al. Microvortex for focusing, guiding and sorting of particles. Lab Chip. Dec. 2008;8(12):2128-34. doi: 10.1039/b813434k. Epub Oct. 30, 2008.
Kaizuka, et al. Structure and dynamics of supported intermembrane junctions. Biophys J. Feb. 2004;86(2):905-12.
Kang, et al. A combined micromagnetic-microfluidic device for rapid capture and culture of rare circulating tumor cells. Lab Chip. Jun. 21, 2012;12(12):2175-81. doi: 10.1039/c2lc40072c. Epub Mar. 28, 2012.
Kang, et al. Isomagnetophoresis to discriminate subtle difference in magnetic susceptibility. Journal of the American Chemical Society 130.2 (2008): 396-397.
Kim, et al. Rapid prototyping of microfluidic systems using a PDMS/polymer tape composite. Lab Chip. May 7, 2009;9(9):1290-3. doi: 10.1039/b818389a. Epub Feb. 10, 2009.
Krivacic, et al. A rare-cell detector for cancer. Proc Natl Acad Sci USA. Jul. 20, 2004;101(29):10501-4. Epub Jul. 12, 2004.
Kuo, et al. Deformability considerations in filtration of biological cells. Lab Chip. Apr. 7, 2010;10(7):837-42. doi: 10.1039/b922301k. Epub Jan. 19, 2010.
Li, et al. Negative enrichment of target cells by microfluidic affinity chromatography. Anal Chem. Oct. 15, 2011;83(20):7863-9. doi: 10.1021/ac201752s. Epub Sep. 22, 2011.
Mahalingam, et al. Formation, stability, and mechanical properties of bovine serum albumin stabilized air bubbles produced using coaxial electrohydrodynamic atomization. Langmuir. Jun. 17, 2014;30(23):6694-703. doi: 10.1021/la5011715. Epub Jun. 4, 2014.
Office action dated Jul. 26, 2017 for U.S. Appl. No. 15/072,287.
Office action dated Aug. 2, 2017 for U.S. Appl. No. 14/836,390.
Olmos, et al. Circulating tumour cell (CTC) counts as intermediate end points in castration-resistant prostate cancer (CRPC): a single-centre experience. Ann Oncol. Jan. 2009;20(1):27-33. doi: 10.1093/annonc/mdn544. Epub Aug. 11, 2008.
Ozkumur, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci Transl Med. Apr. 3, 2013;5(179):179ra47. doi: 10.1126/scitranslmed.3005616.
Panchision, et al. Optimized flow cytometric analysis of central nervous system tissue reveals novel functional relationships among cells expressing CD133, CD15, and CD24. Stem Cells. Jun. 2007;25(6):1560-70. Epub Mar. 1, 2007.
PCT/US2015/023956 International Search Report dated Sep. 30, 2015.
Schiro, et al. Sensitive and high-throughput isolation of rare cells from peripheral blood with ensemble-decision aliquot ranking. Angew Chem Int Ed Engl. May 7, 2012;51(19):4618-22. doi: 10.1002/anie.201108695. Epub Feb. 22, 2012.
Shah, et al. Biopolymer system for cell recovery from microfluidic cell capture devices. Anal Chem. Apr. 17, 2012;84(8):3682-8. doi: 10.1021/ac300190j. Epub Apr. 3, 2012.
Shih, et al. Flow-focusing regimes for accelerated production of monodisperse drug-loadable microbubbles toward clinical-scale applications. Lab Chip. Dec. 21, 2013;13(24):4816-26. doi: 10.1039/c3lc51016f.
Singer, et al. The fluid mosaic model of the structure of cell membranes. Science. Feb. 18, 1972;175(4023):720-31.
Stott, et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci U S A. Oct. 26, 2010;107(43):18392-7. doi: 10.1073/pnas.1012539107. Epub Oct. 7, 2010.
Stroock, et al. Chaotic mixer for microchannels. Science. Jan. 25, 2002;295(5555):647-51.
Sun, et al. High-performance size-based microdevice for the detection of circulating tumor cells from peripheral blood in rectal cancer patients. PLoS One. Sep. 16, 2013;8(9):e75865. doi: 10.1371/journal.pone.0075865. eCollection 2013.
Tan, et al. Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients. Biosens Bioelectron. Dec. 15, 2010;26(4):1701-5. doi: 10.1016/j.bios.2010.07.054. Epub Jul. 22, 2010.
Thorsteinsson, et al. The clinical significance of circulating tumor cells in non-metastatic colorectal cancer-a review. European Journal of Surgical Oncology (EJSO) 37.6 (2011): 459-465.
Triffo, et al. Monitoring lipid anchor organization in cell membranes by PIE-FCCS. J Am Chem Soc. Jul. 4, 2012;134(26):10833-42. doi: 10.1021/ja300374c. Epub Jun. 14, 2012.
Tseng, et al. Tethered fibronectin liposomes on supported lipid bilayers as a prepackaged controlled-release platform for cell-based assays. Biomacromolecules. Aug. 13, 2012;13(8):2254-62. doi: 10.1021/bm300426u. Epub Jul. 11, 2012.
Vona, et al. Isolation by size of epithelial tumor cells : a new method for the immunomorphological and molecular characterization of circulating tumor cells. Am J Pathol. Jan. 2000;156(1):57-63.
Wang, et al. Highly efficient capture of circulating tumor cells by using nanostructured silicon substrates with integrated chaotic micromixers. Angew Chem Int Ed Engl. Mar. 21, 2011;50(13):3084-8.doi: 10.1002/anie.201005853. Epub Mar. 4, 2011.
Wang, et al. Open-tubular capillary cell affinity chromatography: single and tandem blood cell separation. Anal Chem. Mar. 15, 2008;80(6):2118-24. doi: 10.1021/ac702553w. Epub Feb. 21, 2008.
Wang, et al. Shear stress induces endothelial differentiation from a murine embryonic mesenchymal progenitor cell line. Arterioscler Thromb Vase Biol. Sep. 2005;25(9):1817-23. Epub Jun. 30, 2005.
Wu, et al. Antibody conjugated supported lipid bilayer for capturing and purification of viable tumor cells in blood for subsequent cell culture. Biomaterials. Jul. 2013;34(21):5191-9. doi: 10.1016/j.biomaterials.2013.03.096. Epub Apr. 21, 2013.
Xu, et al. A cancer detection platform which measures telomerase activity from live circulating tumor cells captured on a microfilter. Cancer Res. Aug. 15, 2010;70(16):6420-6. doi: 10.1158/0008-5472.CAN-10-0686. Epub Jul. 27, 2010.
Yang et al. Fabrication of Phospholipid Bilayer-Coated Microchannels for On-Chip Immunoassays. Anal Chem 73(2):165-169 (Jan. 15, 2001).
Alix-Panabieres, et al. Challenges in circulating tumour cell research. Nat Rev Cancer. Sep. 2014;14(9):623-31. doi: 10.1038/nrc3820. Epub Jul. 31, 2014.
Ananthanarayanan, et al. Neural stem cell adhesion and proliferation on phospholipid bilayers functionalized with RGD peptides. Biomaterials, Elsevier Science Publishers BV., Barking GB, vol. 31, No. 33, Nov. 1, 2010, pp. 8706-8715.
Antolovic, et al. Heterogeneous detection of circulating tumor cells in patients with colorectal cancer by immunomagnetic enrichment using different EpCAM-specific antibodies. BMC Biotechnol. Apr. 28, 2010;10:35. doi: 10.1186/1472-6750-10-35.
Baeuerle, et al. EpCAM (CD326) finding its role in cancer. Br J Cancer. Feb. 12, 2007;96(3):417-23. Epub Jan. 9, 2007.
Balzar, et al. Epidermal growth factor-like repeats mediate lateral and reciprocal interactions of Ep-CAM molecules in homophilic adhesions. Mol Cell Biol. Apr. 2001;21(7):2570-80.
Chaudry, et al. EpCAM an immunotherapeutic target for gastrointestinal malignancy: current experience and future challenges. Br J Cancer. Apr. 10, 2007;96(7):1013-9. Epub Feb. 27, 2007.
Chen, et al. Generation and characterization of monoclonal antibodies against dengue virus type 1 for epitope mapping and serological detection by epitope-based peptide antigens. Clin Vaccine Immunol. Apr. 2007;14(4):404-11. Epub Feb. 7, 2007.
Cornell, et al. A biosensor that uses ion-channel switches. Letters to Nauture. Jun. 5, 1997. vol. 387. p. 580-583.
European search report and written opinion dated May 2, 2015 for EP Application No. 12805303.0.
“European search report dated Jan. 29, 2016 for EP 15182577.5”.
Gervais, Luc. Capillary Microfluidic Chips for Point-of-Care Testing: from Research Tools to Decentralized Medical Diagnostics. InfoScience. 2011. Thesis 5047. Available at http://infoscience.epfl.ch/record/165376/files/EPFL_TH5047.pdf.
Holmen, et al. Heterogeneity of human nasal vascular and sinusoidal endothelial cells from the inferior turbinate. Am J Respir Cell Mol Biol. Jan. 2005;32(1):18-27. Epub Oct. 21, 2004.
Huang, et al. Type I Collagen-Functionalized Supported Lipid Bilayer as a Cell Culture Platform. Biomacromolecules, vol. 11, No. 5, May 10, 2010, pp. 1231-1240.
International search report and written opinion dated May 30, 2013 for PCT Application No. PCT/US2013/028667 with publication.
International search report and written opinion dated Dec. 10, 2012 for PCT/US2012/044701.
Ishihara, et al. Photoinduced graft polymerization of 2-methacryloyloxyethyl phosphorylcholine on polyethylene membrane surface for obtaining blood cell adhesion resistance. Colloids and Surfaces B: Biointerfaces, vol. 18, No. 3-4, Oct. 1, 2000, pp. 325-355.
Johnson, et al. Structure of an adsorbed dimyristoylphosphatidylcholine bilayer measured with specular reflection of neutrons. Biophys J. Feb. 1991;59(2):289-94.
Kahn, et al. Enumeration of circulating tumor cells in the blood of breast cancer patients after filtration enrichment: correlation with disease stage. Breast Cancer Res Treat. Aug. 2004;86(3):237-47.
Kaladhar, et al. Cell mimetic lateral stabilization of outer cell mimetic bilayer on polymer surfaces by peptide bonding and their blood compatibility. J Biomed Mater Res A. Oct. 2006;79(1):23-35.
Kaladhar, et al. Supported cell mimetic monolayers and their interaction with blood. Langmuir. Dec. 7, 2004;20(25):11115-22.
Karabacak, et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat Protoc. Mar. 2014;9(3):694-710. doi: 10.1038/nprot.2014.044. Epub Feb. 27, 2014.
Lawrence, et al. Leukocytes roll on a selectin at physiologic flow rates: distinction from and prerequisite for adhesion through integrins.Cell. May 31, 1991;65(5):859-73.
Lin, et al. Adhesion of antibody-functionalized polymersomes. Langmuir. Apr. 25, 2006;22(9):3975-9.
Lin, J.J et al. 2006. Adhesion of antibody-functionalized polymersomes. Langmuir 22: 3975-3979. specif, pp. 3975, 3979.
Nagrath, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. Dec. 20, 2007;450(7173):1235-9.
NCBI Direct Submission. NM_002354.2. Homo sapiens epithelial cell adhesion molecule (EPCAM), mRNA. Feb. 5, 2012. [Retrieved from the Internet: http://www.ncbi.nlm.nih.gov/nuccore/218505669?sat=15&satkey=5763417.
Notice of allowance dated Jul. 7, 2016 for U.S. Appl. No. 14/065,265.
“Office action dated Jan. 21, 2015 for U.S. Appl. No. 14/065,265.”
Office action dated Mar. 9, 2016 for U.S. Appl. No. 14/065,265.
“Office action dated May 29, 2015 for U.S. Appl. No. 14/065,265.”
“Office action dated Mar. 23, 16 for U.S. Appl. No. 14/128,345”.
“Office action dated Mar. 9, 16 for U.S. Appl. No. 14/065,265”.
Pantel, et al. Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer. May 2008;8(5):329-40. doi: 10.1038/nrc2375.
Patriarca, et al. Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev. Feb. 2012;38(1):68-75. doi: 10.1016/j.ctrv.2011.04.002. Epub May 14, 2011.
Phillips, et al. Enrichment of cancer cells using aptamers immobilized on a microfluidic channel. Anal Chem. Feb. 1, 2009;81(3):1033-9. doi: 10.1021/ac802092j.
Phillips, J.A. et al. 2009. Enrichment of cancer cells using aptamers immobilized on a microfluidic channel. Analytical Chemistry81 : 1 033-1 039. specif, pp. 1 034, 1 035, 1 036, 1 037, 1 038.
Ruf, et al. Characterisation of the new EpCAM-specific antibody HO-3: implications for trifunctional antibody immunotherapy of cancer. Br J Cancer. Aug. 6, 2007;97(3):315-21. Epub Jul. 10, 2007.
Xu, et al. Aptamer-based microfluidic device for enrichment, sorting, and detection of multiple cancer cells. Anal Chem. Sep. 1, 2009;81(17):7436-42. doi: 10.1021/ac9012072.
Xu, Y. et al. 2009. Aptamer-based microfluidic device for enrichment, sorting, and detection of multiple cancer cells. AnalyticalChemistry 81: 7436-7442. specif. pp. 7436, 7437, 7439, 7440.
Yurke, et al. A DNA-fuelled molecular machine made of DNA. Nature. Aug. 10, 2000;406(6796):605-8.
Related Publications (1)
Number Date Country
20210088514 A1 Mar 2021 US
Provisional Applications (2)
Number Date Country
61606220 Mar 2012 US
61502844 Jun 2011 US
Continuations (2)
Number Date Country
Parent 15378938 Dec 2016 US
Child 16832396 US
Parent 14128354 US
Child 15378938 US