Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INCREASED BUOYANCY USING MICROBUBBLES AND NANODROPLETS FOR CELL CLUSTERS AND CIRCULATING TUMOR CELLS RETRIEVAL
Document Type and Number:
WIPO Patent Application WO/2024/081909
Kind Code:
A1
Abstract:
Use of perfluorocarbon (PFC) microbubbles (MB) and nanodroplets (ND) for isolating cells and/or cell clusters from a biological sample is provided.

Inventors:
DE GRACIA LUX CAROLINE (US)
MATTREY ROBERT F (US)
Application Number:
PCT/US2023/076881
Publication Date:
April 18, 2024
Filing Date:
October 13, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV TEXAS (US)
International Classes:
C12M1/00; A61K47/69; A61K49/00; A61B8/08
Attorney, Agent or Firm:
MEIGS, Julie Broadus et al. (US)
Download PDF:
Claims:
CLAIMS We claim: 1. A method for isolating a cell cluster from a biological sample, the method comprising: (a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a cell targeting agent, for a time sufficient for binding of the microbubbles to target cells; (b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the target cells; and (c) isolating the microbubbles bound to a cluster of the target cells from the biological sample. 2. A method for isolating a cell cluster from a biological sample, the method comprising: (a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a cell targeting agent, for a time sufficient for binding of the microbubbles to target cells; (b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the target cells; and (c) isolating the microbubbles bound to a cluster of the target cells from the biological sample, thereby isolating the cluster of the target cells, wherein the isolated target cells are substantially unchanged as compared to prior to isolation. 3. A method for isolating circulating tumor cells from a biological sample, the method comprising: (a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a tumor targeting agent, for a time sufficient for binding of the microbubbles to circulating tumor cells; Page 29 of 32 WBD (US) 59194535v1 (b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the circulating tumor cells; and (c) isolating the microbubbles bound to the circulating tumor cells from the biological sample. 4. The method of claim 3, wherein the isolated microbubbles bound to circulating tumor cells comprise a cell cluster. 5. The method of claims 1, 2, or 4, wherein the cell cluster is 25-300 µm in diameter. 6. The method of any one of claims 1-5, wherein the biological sample is a body fluid sample. 7. The method of claim 6, wherein the body fluid sample is blood. 8. The method of any one of claims 1-7, wherein the biological sample is contacted sequentially with the microbubbles and the nanodroplets. 9. The method of claim 8, wherein the biological sample is contacted with the microbubbles prior to contacting the biological sample with the nanodroplets. 10. The method of any one of claims 1-7, wherein the biological sample is contacted concurrently with the microbubbles and the nanodroplets. 11. The method of any one of claims 1-10, further comprising a step of exposing the isolated microbubbles bound to the cluster of the target cells to air for a time sufficient for release of the cluster of the target cells from the microbubbles, thereby releasing the cluster of the target cells. 12. The method of any one of claims 1-11, wherein the perfluorocarbon gas comprises a 3, 4, or 5 carbon chain. Page 30 of 32 WBD (US) 59194535v1

13. The method of any one of claims 1-12, wherein the perfluorocarbon liquid comprises a 3, 4, or 5 carbon chain. 14. The method of any one of claims 1-13 wherein the microbubbles contacted with the biological sample have an average diameter between 200 nm and 10 μm. 15. The method of any one of claims 1-14, wherein the microbubbles expand by at least 100- fold in average volume. 16. The method of any one of claims 1-15, wherein the microbubbles expand by at least 10- fold in average diameter. 17. The method of any one of claims 1-16, wherein the cell targeting agent or tumor targeting agent targets Epithelial Cell Adhesion Molecule (EpCAM), epithelial growth factor receptor (EGFR), E-cadherin, vimentin, or cytokeratin (CK) on the target cell or the circulating tumor cell. 18. The method of any one of claims 1-17, wherein the microbubbles and nanodroplets are functionalized to bind each other. 19. The method of any one of claims 1-18, wherein the target cells or circulating tumor cells are breast cancer cells, lung cancer cells, bladder cancer cells, colon cancer cells, pancreatic cancer cells, hepatic cancer cells, cholangiocarcinoma cells, gastric cancer cells, or prostate cancer cells. 20. The method of any one of claims 1-19, wherein the contacting of the biological sample and the microbubbles occurs in vivo. 21. The method of any one of claims 1-19, wherein the contacting of the biological sample and the microbubbles occurs ex vivo. 22. The method of any one of claims 1-21, wherein at least 90% of the isolated cells are viable. Page 31 of 32 WBD (US) 59194535v1

Description:
Increased Buoyancy using Microbubbles and Nanodroplets for Cell Clusters and Circulating Tumor Cells Retrieval CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority to U.S. Provisional Application No.63/379,595 filed on October 14, 2022, the entirety of which is incorporated herein by reference. FIELD OF THE INVENTION The present invention generally relates to the field of cell isolation methods, and more particularly the use of perfluorocarbon (PFC) microbubbles (MB) and nanodroplets (ND) for isolating cells and/or cell clusters from a biological sample. BACKGROUND Isolating cells from a subject or a biological sample can require invasive methods, such as biopsy. Isolating circulating cells, such as rare circulating cells, has technical difficulties. For example, circulating tumor cells (CTCs) disseminate in the bloodstream from solid tumors to seed tumor metastases that cause about 90% of solid tumor-associated deaths. Counting CTCs from a simple blood draw provides an indication of tumor invasiveness, as high CTC counts carry worse prognosis. As CTCs maintain primary tumor genetic heterogeneity and phenotype, they bear the promise of serving as clinical biomarkers. Unfortunately, CTCs are apoptotic and rarely survive more than 2.5 h in circulation. Current practices rely on tumor tissue sampling from biopsies for molecular and drug sensitivity profiling to personalize care. It has recently been recognized that multicellular aggregates of CTCs (CTC clusters) that can contain other support cells and stroma from the tumor’s micro-environment become more robust and have a 100-fold greater metastatic potential than single CTCs. However, most of the current research remains focused on the detection of single CTCs that on occasions also captures CTC clusters, likely because CTC clusters are even more rare than single CTCs. There is therefore a need to develop a simple, practical, and cost-efficient technique to isolate cell clusters, e.g., CTC clusters in a sensitive, specific, and fast manner with minimal processing. SUMMARY OF THE INVENTION Disclosed herein are methods of using perfluorocarbon (PFC) microbubbles (MB) and Page 1 of 32 WBD (US) 59194535v1 nanodroplets (ND) to isolate cells and/or cell clusters from a biological sample. In one aspect, the present disclosure provides a method for isolating a cell cluster from a biological sample, the method comprising: a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a cell targeting agent, for a time sufficient for binding of the microbubbles to target cells; b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the target cells; and c) isolating the microbubbles bound to a cluster of the target cells from the biological sample. In another aspect, the present disclosure provides a method for isolating a cell cluster from a biological sample, the method comprising: a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a cell targeting agent, for a time sufficient for binding of the microbubbles to target cells; b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the target cells; and c) isolating the microbubbles bound to a cluster of the target cells from the biological sample, thereby isolating the cluster of the target cells, wherein the isolated target cells are substantially unchanged as compared to prior to isolation. In another aspect, the present disclosure provides a method for isolating circulating tumor cells from a biological sample, the method comprising: a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a tumor targeting agent, for a time sufficient for binding of the microbubbles to circulating tumor cells; b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles to the circulating tumor cells; and c) isolating the microbubbles bound to the circulating tumor cells from the biological sample. In some embodiments of the methods provided herein, the isolated microbubbles bound to circulating tumor cells comprise a cell cluster. In some embodiments, the cell clusters range from 25 to 300 µm in diameter. In some embodiments, the biological sample is a body fluid sample. In some embodiments, the body fluid sample is blood. Page 2 of 32 WBD (US) 59194535v1 In some embodiments of the methods provided herein, the biological sample is contacted sequentially with the microbubbles and the nanodroplets. In some embodiments, the biological sample is contacted with the microbubbles prior to contacting the biological sample with the nanodroplets. In some embodiments, the biological sample is contacted concurrently with the microbubbles and nanodroplets. In some embodiments, the method further comprises a step of exposing the isolated microbubbles bound to the cluster of the target cells to air for a time sufficient for release of the cluster of the target cells from the microbubbles, thereby releasing the cluster of the target cells. In some embodiments of the methods provided herein, the perfluorocarbon gas comprises a 3, 4, or 5 carbon chain. In some embodiments of the methods provided herein, the perfluorocarbon liquid comprises a 3, 4, or 5 carbon chain. In some embodiments, the microbubbles contacted with the biological sample have an average diameter between 200 nm and 10 µm. In some embodiments, the microbubbles expand by at least 100-fold in average volume upon contact with the nanodroplets. In some embodiments, the microbubbles expand by at least 10-fold in average diameter upon contact with the nanodroplets. In some embodiments, microbubble expansion is measured by average microbubble diameter. In some embodiments, the microbubble expansion is measured by average microbubble volume. In some embodiments of the methods provided herein, the cell targeting agent or tumor targeting agent is Epithelial Cell Adhesion Molecule (EpCAM), epithelial growth factor receptor (EGFR), E-cadherin, vimentin, or cytokeratin (CK). In some embodiments, the microbubbles and nanodroplets are functionalized to bind each other. In some embodiments, the target cells or circulating tumor cells are breast cancer cells, lung cancer cells, bladder cancer cells, colon cancer cells, pancreatic cancer cells, hepatic cancer cells, cholangiocarcinoma cells, gastric cancer cells, or prostate cancer cells. In some embodiments, the contacting of the biological sample and the microbubbles occurs in vivo. In some embodiments, the contacting of the biological sample and the microbubbles occurs ex vivo. In some embodiments, at least 90% of the isolated cells are viable. In one aspect, the present disclosure provides a method of cellular profiling, the method comprising: a) isolating cells or cell clusters according to any one of the preceding claims; and b) determining the genetic and/or molecular profile of the isolated cells. Page 3 of 32 WBD (US) 59194535v1 BRIEF DESCRIPTION OF THE DRAWINGS FIG.1 provides an overview of retrieval of CTC clusters using the inflated MB approach described in the present invention. FIGs.2A-F depicts FITC-labeled silica beads (50 µm and/or 12 µm) in microbubble complexes in suspension before and after addition of nanodroplets. FIG.2A demonstrates that only tiny silica beads float (present as impurities in commercially bought silica beads) in a PBS solution when only targeted MBs were added. FIG.2B shows that large beads sink to the bottom of a PBS solution despite being coupled to MBs. The inset in Fig.2B shows a magnified silica bead with attached MBs indicated by arrows. FIG.2C shows that the addition of NDs causes the large beads to float to the surface. FIG.2D shows a mixture of FITC-labeled 12 µm and 50 µm silica beads before addition of any MBs. FIG.2E shows the retrieval of FITC-labeled beads with the addition of MBs only. FIG.2F depicts the retrieval of FITC-labeled beads using the combination of MBs and NDs. FIG.3 shows the impact of ND-driven MB inflation on FITC-labeled RBCs and FITC- labeled K562 cells retrieval as detected by fluorescence microscopy and hemocytometry. FIGs.4A-C demonstrates the ability for anti-EpCAM targeted MBs to bind to H2122 cell clusters and ND-driven improved retrieval in media and blood. Fig.4A shows a representative anti-EPCAM MBs size distribution. Fig.4B provides a representative image of a H2122 cell cluster densely coated with anti-EpCAM MBs. FIG.4C quantifies H2122 cell cluster retrieval from full media using either anti-EpCAM targeted MBs only or the sequential addition of anti-EpCAM targeted MBs and NDs targeted to the MBs. FIG.5A-C presents representative images of H2122 cells isolated using inflated MBs. FIG.5A is a representative dual-fluorescent image of isolated H2122 clusters stained with acridine orange and propidium iodide staining showing that MB inflation did not affect H2122 cell viability. The black and white arrows indicate the same H2122 cluster, with the red arrow indicating an attached MB. FIG.4B-C present representative bright-field and fluorescent images respectively of 100 µm MBs (red arrow) attached to H2122 AO+ cluster (black and white arrows). Page 4 of 32 WBD (US) 59194535v1 FIG.6A-B depicts the retrieval of CTC clusters from blood using inflated MBs. FIG.6A presents representative photographs of whole blood processed with MBs and NDs after 1, 2, and 3 washes. FIG.6B depicts representative microscopy pictures of retrieved CTC clusters from blood (blue = DAPI, green = AF488-anti-EpCAM). FIG.7 shows a representative image of EpCAM-positive naturally occurring CTC clusters isolated from the blood of female Balb/c mice implanted with luciferase-expressing 4T1 (Luc-4T1) metastatic breast cancer cell line in the mammary fat pad. Going clockwise, the panels show the same image for brightfield, DAPI, EpCAM, and CD45 staining. DETAILED DESCRIPTION The present invention provides methods for cell cluster and/or cancer cell isolation using perfluorocarbon (PFC) microbubbles (MBs) and nanodroplets (NDs). The isolated cells may be useful for any purpose, and particular uses in drug sensitivity studies on cultured isolated cells, biomarker discovery for new therapy development, genetic/molecular profiling are envisioned to provide personalized care and prevent metastasis or improve treatment outcome. Microbubbles and Nanodroplets Microbubbles and nanodroplets comprise a perfluorocarbon (PFC) core stabilized by a shell (e.g., phospholipid, polymer, denatured protein). Perfluorocarbon compositions may be a dispersion of a non-continuous perfluorocarbon phase in a continuous non-perfluorocarbon phase (e.g., a liquid-in-liquid emulsion or a gas-in-liquid dispersion). In certain aspects, the continuous phase is aqueous (e.g., water or saline or a mixture of PBS, glycerol and propylene glycol). Perfluorocarbon dispersions may comprise particles encapsulating the perfluorocarbon, which may exist within the particle in a liquid and/or gaseous form. According to certain aspects, the particles may comprise nanodroplets and/or microbubbles. In some implementations, nanodroplets and microbubbles may comprise similar structures with the exception that nanodroplets comprise a liquid perfluorocarbon core and microbubbles comprise a gaseous perfluorocarbon core. Based upon the size of nanodroplets, nanobubbles and microbubbles, different concentrations may be appropriate for a therapeutic composition. As used herein, “nanodroplet” (“ND”) may refer to a particle formed by a surfactant shell encapsulating a liquid core. According to some aspects, the diameter of the nanodroplet or the average diameter of a nanodroplet composition may be no greater than about 50, 75, 100, 125, Page 5 of 32 WBD (US) 59194535v1 150, 175, 200, 225, 250, 275, 300, 350, 400, 500, 600, 700, 800, 900, or 1,000 nm. According to some aspects, the diameter of the nanodroplet or the average diameter of the nanodroplet composition may be at least about 100, 125, 150, 175, 200, 225, 250, 275, or 300 nm in diameter. According to some aspects, the diameter of the nanodroplet or the average diameter of a nanodroplet composition may be about 50 nm – 100 nm, about 50 nm – 200 nm, about 50 nm – 300 nm, about 100 nm – 200 nm, about 100 nm – 300 nm, about 100 nm – 400 nm, about 100 nm – 500 nm, about 100 nm -1 µm, about 200 nm - 1 µm, about 300 nm - 1 µm, or about 500 nm – 1 µm. Nanodroplet compositions may be produced by any method known in the art. Preferably, nanodroplet compositions are produced by methods that result in concentrated and stable nanodroplets at physiological temperatures (e.g., nanodroplets that do not spontaneously evaporate or significantly change size). According to certain aspects, nanodroplets may be formed by any of the methods described in U.S. Pat. App. Pub. No.2018/0272012 to de Gracia Lux et al., published Sep.27, 2018; or de Gracia Lux et al., RSC Adv.2017; 7(77):48561-48568 (doi: 10.1039/C7RA08971F), each of which is herein incorporated by reference in its entirety. According to some aspects, nanodroplets may be formed by a high energy emulsification method, such as sonication at -15 °C, which surprisingly results in more efficient manufacture. In some embodiments, PFC (e.g., PFB, PFP, C 2 BrF 5 , C 3 BrF 7 ) nanoemulsions are prepared by sonication. For example, a first dram vial in dry ice containing an 80:15:5 (v:v:v) PBS/propylene glycol/glycerol excipient solution and a dry phospholipid film (e.g., comprising 1,2-distearoy l-sn-glycero-3-phosphocholine DSPC and 1,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-methoxy(polyethyleneglycol)-2000 (DSPE-PEG2000) in a 9:1 molar ratio) can be heated on a 70 °C heating block for 15 min, followed by sonication in a bath sonicator until the solution turned clear. The phospholipid solution is then allowed to cool to room temperature and then cooled in cold bath that has temperature lower than the boiling point of the PFC [e.g., for PFB, an ice-salt bath (between -10 and -15 °C); for PFP, an ethanol-dry ice bath (about -80 °C)] for 5 minutes. A second dram vial in cold bath that has temperature lower than the boiling point of the PFC [e.g., for PFB, an ice-salt bath (between -10 and -15 °C); for PFP, an ethanol-dry ice bath (about -80 °C)] containing liquid PFC and cold phospholipid mixture can be sonicated with the probe sonicator. Any volume of PFC can be mixed with phospholipid at any proportions. For example, PFB of about 50 μL, 100 μL, 150 μL, 200 μL, 300 μL, 400 μL, 500 μL, or more; about 50-100 μL, 100-150 μL, 150-200 μL, 200-300 μL, 300-400 Page 6 of 32 WBD (US) 59194535v1 μL, 400-500 μL, or more, e.g., 150-200 μL of PFC can be mixed with phospholipid at a concentration of 1-30% v/v (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, or 30% v/v) and sonicated. The PFC-phospholipid mixture is sonicated at a temperature that is lower than the boiling point of the PFC. For instance, the PFC-phospholipid mixture is prepared and sonicated at a temperature that is about 1 °C lower, about 5 °C lower, about 10 °C lower, about 1-2°C lower, about 2-3 °C lower, about 3-4°C lower, about 4-5°C lower, about 5-10°C lower, more than about 10°C lower, at least about 1°C lower, at least about 5°C lower, or at least about 10°C lower than the boiling point of the PFC. Any sonication condition can be used, including power range, operation frequency, amplitude, duration, and pulse. For instance, the PFC-phospholipid mixture can be sonicated with about 10-200 kHz (e.g., 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, or 200 kHz) at about 1-2000 Watt (e.g., 1, 10, 20, 50, 100, 200, 300, 400, 500, 1000, 1500, or 1500 Watt), at about 20-80% amplitude (e.g., 20%, 30%, 40%, 50%, 60%, 70%, or 80%), for about 1-1000 active seconds (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 15, 20, 30, 40, 50, 60 active seconds, or more). In specific embodiments, a 2-mL PFC-phospholipid mixture is sonicated for 10 seconds. “Active seconds” as used herein refer to the total duration of actual sonication, excluding any pauses where sonication is applied in pulse mode. For example, sonication can be applied for 1 second on and 1 second off, 5 seconds on and 5 seconds off, or 10 seconds on and 10 seconds off, for about 1-1000 total active seconds. In a specific embodiment, the PFB/phospholipid mixture is sonicated for 10 seconds at 20% power. The probe tip size can be about 1-10 mm in diameter (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 mm in diameter) when sonicating about 150-200 μL of PFC with phospholipid. If a drop of non-encapsulated PFC remained at the bottom of the vial, the mixture can be further sonicated. Following sonication, the resulting PFC emulsion can be collected by centrifugation, filtration, or both, and stored. Any suitable methods of centrifugation and/or filtration can be used. For instance, centrifugation can be performed at 200-1500 x g (e.g., about 200 x g, 300 x g, 400 x g, 500 x g, 600 x g, 700 x g, 800 x g, 900 x g, 1000 x g, 1100 x g, 1200 x g, 1300 x g, 1400 x g, or 1500 x g). Filtration can be performed using a filter having a pore size that is larger than the size of the nanoemulsions but small enough to remove any debris, for example of a pore size of about 0.1-5 μm (e.g., 0.1 μm, 0.22 μm, 0.45 μm, 1 μm, 2 μm, 3 μm, 4 μm, or 5 μm). According to certain aspects, nanodroplets may be formed by any of the methods described in U.S. Pat. App. Pub. No.2013/0336891 to Dayton et al., published on Dec.19, 2013; Sheeran et al., Ultrasound Med Biol.2011 Sep; Page 7 of 32 WBD (US) 59194535v1 37(9):1518-30 (doi: 10.1016/j.ultrasmedbio.2011.05.021); Sheeran et al., Biomaterials.2012 Apr; 33(11):3262-9 (doi: 10.1016/j.biomaterials.2012.01.021), or Sheeran et al., IEEE Trans Ultrason Ferroelectr Freq Control.2017 Jan; 64(1):252-263 (doi: 10.1109/TUFFC.2016.2619685), each of which is herein incorporated by reference in its entirety. According to certain aspects, nanodroplet emulsifications prepared directly from liquid perfluorocarbons (e.g., via high energy emulsification methods) may be advantageous (e.g., more easily manufactured, higher encapsulation efficiency, no contamination from empty liposomes). Fabrication of nanodroplet emulsifications via microbubble condensation will require much larger volumes of a starting microbubble dispersion to achieve the same amount of liquid perfluorocarbon in an emulsification dose. For example, whereas a 7% (v/v) PFB nanodroplet emulsion, as can be prepared according to the emulsification techniques disclosed herein, can deliver 150 µL of liquid PFB in approximately 2.15 mL total volume, it would require condensation of approximately 22.5 mL of PFB gas to achieve the same volume of liquid PFB. As each 1 µm PFB microbubble should comprise approximately 4.18x10 -12 mL of PFB gas and microbubble compositions prepared according to standard procedures and formulations (e.g., having the same phospholipid content as described herein) should yield approximately 30-150 µL of gas per batch (having broad distributions of microbubble sizes), significantly larger volumes of microbubble dispersions would be needed for condensation to achieve the same volumes of liquid perfluorocarbon as can be directly produced via emulsification. As used herein, “bubbles” or “microbubble” (“MB”) may refer to a particle formed by a surfactant shell encapsulating a gas core. According to some aspects, the microbubble may be no greater than about 10 µm in diameter. Unless otherwise specified, microbubbles, as used herein, may include bubbles less than 1 µm (i.e. nanobubbles), such as microbubbles between, for example, about 50 nm – 100 nm, about 50 nm – 200 nm, about 50 nm – 300 nm, about 100 nm – 200 nm, about 100 nm – 300 nm, about 100 nm – 400 nm, about 100 nm – 500 nm, about 100 nm -1 µm, about 200 nm - 1 µm, about 300 nm - 1 µm, about 500 nm – 1 µm, about 100 nm -10 µm, about 200 nm - 10 µm, about 300 nm - 10 µm, about 500 nm – 10 µm, or about 1 µm – 10 µm. According to some aspects, the average microbubble size within a microbubble composition is at least about 1, 2, 3, 4, or 5 µm. According to some aspects, the average microbubble size is between approximately 1-10, 1-5, 1-4, 1-3, 1-2, 2-5, 2-4, 2-3, 3-5, or 3-4 µm. According to certain aspects, microbubbles may be formed by any of the methods described in U.S. Pat. App. Page 8 of 32 WBD (US) 59194535v1 Pub. No.2013/0336891 to Dayton et al., published on Dec.19, 2013, which is herein incorporated by reference. Both nanodroplets and microbubbles generally comprise a surfactant shell which encapsulates the perfluorocarbon core. The surfactant shell may comprise one or more types of molecules which lower the interfacial tension between the perfluorocarbon core and the continuous phase, such as a physiological aqueous environment. This exterior shell may comprise, for example, lipids (e.g., phospholipids), proteins (e.g., albumin), sugars, and/or polymers. According to some aspects, the surfactant shell may comprise lipids, such as phospholipids, which self-align under certain conditions to form a hydrophilic external surface and a lipophilic or hydrophobic internal surface. The phospholipids may comprise any standard phospholipid used in the art for forming microbubbles, nanodroplets, micelles, liposomes, etc. According to some aspects, the phospholipids may comprise diacylglyceride structures, such as phosphatidic acid (phosphatidate) (PA), phosphatidylethanolamine (cephalin) (PE), phosphatidylcholine (lecithin) (PC), phosphatidylserine (PS), and phosphoinositides (e.g., posphatidylinositol (PI), phosphatidylinositol phosphate (PIP), phosphatidylinositol bisphosphate (PIP2) and phosphatidylinositol trisphosphate (PIP3). In some aspects, the phospholipids may comprise phosphosphingolipids, such as ceramide phosphorylcholine (Sphingomyelin) (SPH), ceramide phosphorylethanolamine (Sphingomyelin) (Cer-PE), and ceramide phosphoryllipid. According to some aspects, the phospholipid comprises 1,2-Distearoyl-sn-Glycero-3- Phosphocholine (DSPC) or derivatives thereof. According to some aspects, the phospholipid comprises 1,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine (DSPE) or derivatives thereof. According to some aspects, the surfactant shell may comprise one or more co-surfactants, including, for example, fluorinated surfactants such as semifluorinated alkanes (e.g., CnF2n+1CmH2m+l and more complex architectures). Semifluorinated alkanes are described, for example, in U.S. Pat. App. Pub. No.2018/0272012 to de Gracia Lux et al. and Bertilla et al., “Semifluorinated Alkanes as Stabilizing Agents of Fluorocarbon Emulsions.” In: Kobayashi K., Tsuchida E., Horinouchi H. (eds) Artificial Oxygen Carrier. Keio University International Symposia for Life Sciences and Medicine, (2005) vol 12. Springer, Tokyo, each of which is herein incorporated by reference in its entirety. According to some aspects, the surfactant molecules may be coupled to polymer chains, such as poly(ethylene glycol) (i.e. the surfactant shell may be PEGylated). For example, the Page 9 of 32 WBD (US) 59194535v1 surfactant molecule may comprise 1,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine-N- [Methoxy(Polyethylene glycol)-2000] (DSPE-PEG2k). PEGylation of the particle may improve anti-flocculation / colloidal stability, . For example, PEGylation may inhibit coalescence and/or Ostwald ripening of nanodroplets and/or microbubbles. PEGylation of the external surface of the particle may also provide favorable conditions (e.g., steric) for performing conjugations which functionalize the particle surface. According to some aspects, the surfactant shell may comprise two or more types of surfactant molecules. According to some aspects, the surfactant shell may comprise three or more types of surfactant molecules. The perfluorocarbon core comprises one or more perfluorocarbons. “Perfluorocarbon” (PFC) as used herein refers to any hydrocarbons of which hydrogen atoms are substituted by fluorine atoms or other halogens. Perfluorocarbon cores help stabilize the perfluorocarbon particles (e.g., nanodroplets) against dissolution, counteracting the effect of outside pressure (blood pressure and Laplace Pressure), since perfluorocarbons are hydrophobic and not prone to escaping the particle. Any perfluorocarbons can be used in the perfluorocarbon core, compositions, and methods of the present disclosure, including but not limited to, three-carbon PFCs [e.g., perfluoropropane (PFP, C 3 F 8 ), 1-bromoheptafluoropropane (C 3 BrF 7 )], four-carbon PFCs [e.g., perfluorobutane (PFB, C 4 F 10 )], and five-carbon PFCs [e.g., perfluoropentane (C5F12)]. The one or more perfluorocarbons may comprise octafluoropropane (OFP) / perfluoropropane (PFP), decafluorobutane (DFB) / perfluorobutane (PFB), dodecafluoropentane (DDFP) / perfluoropentane / perflenapent, tetradecafluorohexane / perfluorohexane, hexadecafluoroheptane / perfluoroheptane, octadecafluorodecalin / perfluorodecalin, or perfluoro(2-methyl-3-pentanone) (PFMP). The perfluorocarbon core may comprise one or more fluorocarbons selected from the following: 1,2-bis(F-alkyl)ethenes; 1,2-bis(F-butyl)ethenes; 1-F- isopropyl,2-F-hexylethenes; 1,2-bis(F-hexyl)ethenes; perfluoromethyldecalins; perfluorodimethyldecalins; perfluoromethyl- and dimethyl- adamantanes; perfluoromethyl-, dimethyl- and trimethyl- bicyclo (3,3,1) nonanes and their homologs; perfluoroperhydrophenanthrene; ethers of formulae: (CF3)2CFO(CF2 CF2)2OCF(CF3)2, (CF3)2CFO(CF2 CF2)3 OCF(CF3)2, (CF3)2CFO(CF2 CF2)2F, (CF3)2CFO(CF2 CF2)3F, (C 3 F 7 ) 3 , N(C 4 F 9 ) 3 , and N(C 5 F 11 ) 3 ; perfluoro-N-methylperhydroquinolines and perfluoro-N- methylperhydroisoquinolines; and perfluoralkyl hydrides, such as C6F13H, C8F17H, C8F16H2 and Page 10 of 32 WBD (US) 59194535v1 the halogenated derivatives C6F13Br, (perflubron), C6F13CBr2CH2Br, 1-bromo 4- perfluoroisopropyl cyclohexane, C8F16Br2, and CF3O(CF2CF2O)uCF2CH2OH with u = 2 or 3. In some specific embodiments, perfluorocarbon used in the perfluorocarbon core, compositions, and methods provided herein has low or very low boiling points (BP), for example PFP (e.g., BP -39 °C), C2BrF5 (BP -21 °C), PFB (e.g., BP -2 °C), and C3BrF7 (e.g., PB +12 °C). One PFC, or a combination of two or more PFCs can be mixed with phospholipids to produce PFC emulsions or PFC compositions provided herein. According to some aspects wherein the dispersion comprises microbubbles, the microbubble core may further comprise non-perfluorocarbon gases such as air, sulfur hexafluoride, and/or nitrogen. The perfluorocarbon composition may comprise a dispersion (e.g., emulsion of nanodroplets) having an surfactant/emulsifier content (e.g., phospholipids and any additional cosurfactants) that results in a stable emulsion. Perfluorocarbon dispersions are well known in the art and the surfactant content may generally be any amount that is known in the art. According to some aspects, the perfluorocarbon composition may comprise a dispersion having a surfactant content of approximately 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, or 10.0 mg/mL. In some aspects, the dispersion is no greater than approximately 3.5 mg/mL surfactant. In some aspects, the dispersion is at least about 1.0 mg/mL surfactant. In some aspects, the dispersion is between about 1.0 and about 3.5 mg/mL surfactant. The perfluorocarbon composition may comprise a dispersion (e.g., emulsion of nanodroplets) having a perfluorocarbon content that results in a stable emulsion. Perfluorocarbon dispersions are well known in the art and the perfluorocarbon content may generally be any amount that is known in the art. According to some aspects, the perfluorocarbon composition may comprise a dispersion that is approximately 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 35%, 40%, 45%, 50% (v/v) of the perfluorocarbon. In some aspects, the dispersion is no greater than about 30% (v/v). In some aspects, the dispersion is at least about 0.5% (v/v). In some aspects, the dispersion is between about 0.5% and 30% (v/v). In some aspects, the dispersion is between about 1-10% (v/v). In some aspects, the dispersion is about 7% (v/v). Generally, larger perfluorocarbon contents can be achieved by increasing the concentration of surfactant. The perfluorocarbon composition may be the end product of a dispersion-forming (e.g., Page 11 of 32 WBD (US) 59194535v1 emulsification) process. In some aspects, the continuous liquid phase of the dispersion may be exchanged with another liquid (e.g., via dialysis), diluted, or concentrated (e.g., via centrifugation). In some aspects, additional components may be added to the dispersion, e.g., by mixing the dispersion with additional fluids (e.g., saline). According to various aspects, the dispersion may be prepared in an excipient solution. According to certain aspects, the excipient solution may comprise one or more of the following components: water, saline, PBS, glycerol, propylene glycol, Ringer’s solution, and dextrose. For example, the excipient solution may comprise a PBS/propylene glycol/glycerol mixture (e.g., 80:15:5 (v:v:v)) or a PBS/propylene glycol mixture (e.g., 4:6 (v:v)). According to some aspects, the excipient solution may be any solution which will not freeze at the working temperatures. The dispersion may be combined with or prepared in any compatible excipient or pharmaceutically acceptable carrier that is suitable for the particular route of administration. Various pharmaceutically acceptable carriers are well known in the pharmaceutical arts. Isolating Cells from Biological Sample Provided herein is a method for isolating cells from a biological sample. In some aspects, the method provided herein comprises a method for isolating a cell cluster or a plurality of clusters from a biological sample, and comprises: a) contacting the body fluid sample with microbubbles, for a time sufficient to allow for binding of the microbubbles to target cells, wherein the microbubbles comprise a cell targeting agent; b) contacting the body fluid sample with nanodroplets to the body fluid to thereby expand the microbubbles and the buoyancy of the microbubbles bound to the target cells; and; c) isolating the microbubbles bound to the target cells from the body fluid sample. In some embodiments, the cell cluster is a tumor cell cluster. In some aspects, the method provided herein is a method for isolating circulating tumor cells from a biological sample, and comprises: a) contacting the biological sample with microbubbles for a time sufficient for allowing binding of the microbubbles to circulating tumor cells, wherein the microbubbles comprise a tumor targeting agent; b) contacting the body fluid sample with nanodroplets to the body fluid, thereby expanding the microbubbles and increasing the buoyancy of the microbubbles bound to the circulating tumor cells; and c) isolating the microbubbles bound to the circulating tumor cells from the body fluid without centrifugation. In some embodiments, the circulating tumor cells comprise one or more circulating tumor cell clusters. A “biological sample” as used herein refers to any biological material, obtained from a Page 12 of 32 WBD (US) 59194535v1 subject, or present within a subject. A biological sample can comprise cells, e.g., a heterologous population of cells, or a homologous population of cells. For example, a biological sample can be a body fluid that is contained in or sampled from the subject, and that contains cells, e.g., liquid suspension of cells, or a heterologous population of cells. A biological sample or a body fluid can include blood, whole blood, urine, cerebrospinal fluid, ascites, and pleural fluid. “Whole blood” as used herein refers to blood within or obtained from a subject, in a crude state, that is not subjected to a process, e.g., centrifugation or filtering. Alternatively or additionally, a biological sample can comprise a tissue sample and/or a liquid suspension of cells. “Isolating” cells or a cell cluster as used herein refers to taking the cells or the cell cluster out of the environment in which they exist, e.g., biological sample, e.g., blood. “Binding” of the microbubbles to cells refers to attaching or attachment of microbubbles to targeted cells by any means, including via a covalent bond, a noncovalent bond, a linker, conjugation, e.g., an antibody-antigen interaction, receptor-ligand interaction. A “target cell” as used herein refers to any cell that the microbubbles, comprising a cell targeting agent, targets and/or binds. A target cell can be a tumor cell or a non-tumor cell. A “cell targeting agent” as used herein refers to an agent that targets and binds any moiety of interest on the cell surface, including a cell surface moiety, a cell surface receptor, or a cell surface ligand. In some embodiments, a “cell targeting agent” is an antibody or antigen-binding domain thereof. “Antibody” as used herein encompasses any type of antibody, including but not limited to monoclonal antibodies, polyclonal antibodies, antigen-binding fragments of intact antibodies (e.g., Fab, Fab’, F(ab’) 2, Fd, Fv, Fc, etc.) that retain the ability to bind to a given antigen, bispecific antibodies, multispecific antibodies, heteroconjugate antibodies, fusion proteins having an antibody or antigen-binding fragment thereof (e.g., a domain antibody), single chain antibodies (scFv), single domain antibodies (sdAbs, also known as nanobodies and VHH antibodies), maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR, humanized antibodies, chimeric antibodies, and any other modified configuration of the immunoglobin molecule that includes an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently-modified antibodies. “Antigen- binding fragment” as used herein refers to a protein or polypeptide sequence that can bind to a specific target or antigen, such as a carbohydrate, polynucleotide, lipid, polypeptide, specific antigenic determinant, epitope, antigen, or protein. A “tumor targeting agent” refers to an agent Page 13 of 32 WBD (US) 59194535v1 that targets and binds any moiety of interest on the tumor cell surface, including a tumor cell surface moiety, a tumor cell surface receptor, or a tumor cell surface ligand. A “time sufficient for binding of the microbubbles” to target cells or circulating tumor cells, as used herein, refers to a time that allows for a certain amount of microbubbles to bind to target cells or circulating tumor cells that is understood by one having ordinary skills in the art. Depending on the amount of microbubbles used to contact the biological sample, a time sufficient for binding of the microbubbles to target cells or circulating tumor cells can be a time that allows for binding of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or more than 95% of the microbubbles to target cells or circulating tumor cells. A time sufficient for binding of the microbubbles to target cells or circulating tumor cells can be any amount of time, for example, 1 second to 10 minutes, 1 second to 1 minute, 1-30 seconds, 30 seconds to 1 minute, 1-5 minutes, 5-10 minutes, or more than 10 minutes. In some embodiments, the microbubbles bind to a target cell or a circulating tumor cell or a circulating tumor cell cluster by a cell targeting agent or a tumor targeting agent. Binding can be in any form or of characteristics, such as via a covalent bond, a noncovalent bond, a linker, or conjugation. The targeted moiety on the cell can be any moiety to which the microbubbles can bind, such as a receptor or other protein moiety. The cell targeting agent or tumor targeting agent can target Epithelial Cell Adhesion Molecule (EpCAM), epithelial growth factor receptor (EGFR), E-cadherin, vimentin, or cytokeratin (CK) on the target cell or the circulating tumor cell. The targeted moiety on the cell can be specific or abundantly expressed in targeted cells or circulating tumor cells. In some embodiments, the microbubbles comprise a plurality of cell targeting agents or tumor targeting agents that targets a plurality of moieties of interest on the cell surface, thereby conducting divalent or multivalent targeting. A microbubble can comprise 50-2,000 copies, 2,000-10,000 copies, or more than 10,000 copies of antibodies on the surface, which can target different moieties on the surface of target cells or circulating tumor cells. For example, a microbubble can comprise 50-1,000, 1,000-2,000, 2,000-3,000, 4,000-5,000, 5,000- 6,000, 6,000-7,000, 7,000-8,000, 9,000-10,000, or more than 10,000 copies of antibodies on the surface, which can target different moieties on the surface of target cells or circulating tumor cells. The plurality of targeted moieties can be specific or abundantly expressed in targeted cells or tumor cells. For example, a microbubble can comprise a plurality of cell targeting agents or tumor targeting agents that each bind Epithelial Cell Adhesion Molecule (EpCAM), epithelial Page 14 of 32 WBD (US) 59194535v1 growth factor receptor (EGFR), E-cadherin, vimentin, or cytokeratin (CK) on the target cell or the circulating tumor cell. “Expanding the microbubbles” refers to increasing the volume and/or diameter of the microbubbles. In some aspects, the microbubbles are expanded, i.e., their average diameter and/or average volume is increased, by the methods provided herein. Microbubbles provided to the biological sample can have an average diameter between about 50 nm – 100 nm, about 50 nm – 200 nm, about 50 nm – 300 nm, about 100 nm – 200 nm, about 100 nm – 300 nm, about 100 nm – 400 nm, about 100 nm – 500 nm, about 100 nm -1 µm, about 200 nm - 1 µm, about 300 nm - 1 µm, about 500 nm – 1 µm, about 100 nm -10 µm, about 200 nm - 10 µm, about 300 nm - 10 µm, about 500 nm – 10 µm, or about 1 µm – 10 µm In specific embodiments, microbubbles provided to the biological sample can have an average diameter between 200 nm and 10 μm, e.g., between 300 nm and 3 μm. The average diameter of the microbubbles can be increased by at least an order of magnitude, at least two orders of magnitude, or more by the methods provided herein. The average volume of the microbubbles can be increased by at least an order of magnitude, two orders of magnitude, three orders of magnitude, four orders of magnitude, five orders of magnitude, six orders or magnitude, at least six orders of magnitude, or more than six orders of magnitude by the methods provided herein. In some embodiments, microbubbles (MBs) inflated by nanodroplets can expand by 10-fold, 50-fold, 100-fold, 150-fold, 200-fold, 300-fold, or 400-fold in diameter as compared to the size of the MBs before inflation. In specific embodiments, MBs are about 300 nm – 3 μm in diameter before inflation, which upon contact with nanodroplets expand to about 50-100 μm in diameter, thereby expanding about 16-133 fold in diameter. “Increasing the buoyancy” of microbubbles, e.g., microbubbles bound to target cells or circulating tumor cells, as used herein refers to increasing the ability for the microbubbles to float to the surface of liquid, e.g., fluid sample. The expansion of the microbubbles, i.e., increase in the average diameter and/or volume of the microbubbles can increase the buoyancy force of the microbubbles while the microbubbles are bound to the targeted cells or tumor cells, and enables retrieval of cells. In some embodiments, the methods provided herein increases the buoyancy of microbubbles by at least an order of magnitude, two orders of magnitude, three orders of magnitude, four orders of magnitude, five orders of magnitude, six orders or magnitude, at least six orders of magnitude, or more than six orders of magnitude. Page 15 of 32 WBD (US) 59194535v1 In some embodiments, the methods provided herein retrieve cells that are circulating cells, e.g., rare circulating cells. “Circulating cells” as used herein refers to any cell that is in a biological sample in vivo, or in a biological sample obtained from the subject. In some embodiments, the circulating cells are circulating tumor cells (CTCs), such as breast cancer cells, lung cancer cells, bladder cancer cells, colon cancer cells, pancreatic cancer cells, hepatic cancer cells, cholangiocarcinoma cells, gastric cancer cells, or prostate cancer cells. CTCs disseminate in the bloodstream from solid tumors to seed tumor metastases. In some embodiments, CTCs is about 10-20 µm in diameter, or about 15-20 µm in diameter. In some embodiments, the cells comprise a cluster. A “cluster” or “cell cluster” as used herein refers to a plurality of cells that are attached with one another or otherwise grouped together to form one region of interest for retrieval. A cluster can comprise any number of plurality of cells, e.g., about 2-10, 2-100, 2-200, 2-300, 2-500, 25-75, 25-100, 2-1000, or more than 1000 cells. In specific embodiments, a cluster comprises about 50 cells. The cluster can comprise a mass that is about 25-500 µm in diameter, such as about 25-10 µm, 25-100 µm, 25- 200 µm, 25-300 µm, 25-400 µm, 50-100 µm, 100-200 µm, 200-300 µm, 300-400 µm, 400-500 µm, or more than 500 µm in diameter. A cluster of cells can have a heterogenous or homogeneous cell population. “Homogeneous” as used herein refers to deriving from the same cell origin or source. “Heterogeneous” as used herein refers to deriving from different cell origin or source. For example, a cluster of cells can be 0-100% homogeneous, e.g., 0%, 1-10%, 10- 20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90%, 90-99%, or 100% homogeneous. When CTCs organize or are shed as multicellular clusters they carry with them support elements such as immune cells, fibroblasts, tumor matrix, etc. As clusters, CTCs are more resilient, can hide from the immune system, are more protected from shear damage, are less apoptotic, were shown to increase metastatic potential by up to 100-fold, and can be shed from even small tumors. Despite convincing evidence that clusters can deliver the promise that CTCs could not, most research from liquid biopsy remains focused on analyzing ctDNA or enumerating CTCs, likely because CTC clusters are very rare in blood and are extremely challenging to retrieve. Microfluidic-based cluster detections are not yet simple enough to be widely adopted. They are challenged by the large size of some clusters as well as their rarity that requires processing a large volume of blood decreasing throughputs. A cluster that can be isolated by the methods provided herein can be a CTC cluster. Page 16 of 32 WBD (US) 59194535v1 In some embodiments of the methods provided herein, the microbubbles and nanodroplets are provided sequentially to the biological sample. The microbubbles can be provided prior to the nanodroplets. In some embodiments, the microbubbles comprise a perfluorocarbon gas, e.g., a perfluorocarbon gas comprising a 3, 4, or 5 carbon chain. In some embodiments, the nanodroplets comprise a perfluorocarbon liquid, e.g., a perfluorocarbon liquid comprising a 3, 4, or 5 carbon chain. In some embodiments, the microbubbles and the nanodroplets are functionalized to bind each other. “Functionalized to bind” as used herein refers to any form of binding, including via covalent bond, noncovalent bond, linker, or conjugation. The methods provided herein can comprise using nanodroplets that do not target, or are not functionalized to bind, to microbubbles. The methods provided herein can comprise using nanodroplets that targets, or functionalized to bind, to microbubbles. In some embodiments of the methods described herein, the addition of NDs that are not functionalized to bind to MBs increases cell or cell cluster retrieval by 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or more, e.g., by 5%, 10%, 15%, 20%, 25%, 30%, 50%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 1000% or more as compared to MBs alone. In some embodiments, the addition of NDs functionalized to bind MBs increases cell or cell cluster retrieval by 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or more, e.g., by 5%, 10%, 15%, 20%, 25%, 30%, 50%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 1000% or more as compared to MBs alone. Functionalizing nanodroplets to bind to microbubbles can increase efficiency of cell retrieval as compared to nanodroplets that are not functionalized. For example, functionalizing nanodroplets to bind to microbubbles can increase efficiency of cell or cell cluster retrieval by 5%, 10%, 15%, 20%, 25%, 30%, or more as compared to non-targeting nanodroplets. In some specific embodiments, functionalizing nanodroplets to bind to microbubbles can increase efficiency of cell retrieval by about 20% as compared to non-targeting nanodroplets. In some embodiments, contacting the biological sample with the microbubbles occurs in vivo. In other embodiments, contacting the biological sample with the microbubbles in vitro, e.g., using a biological sample obtained from the subject. The methods provided herein can retrieve cells of interest, e.g., CTCs, CTC clusters, while they remain viable during or after retrieval. For example, at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or more cells can remain viable during and after retrieval. In specific Page 17 of 32 WBD (US) 59194535v1 embodiments, at least 90% of the isolated cells are viable. The methods provided herein enable the retrieval of cells or cell clusters that are substantially unchanged relative to their condition prior to isolation. For example, the present disclosure provides a method for isolating a cell cluster from a biological sample, comprising: (a) contacting the biological sample with microbubbles, wherein the microbubbles comprise a perfluorocarbon gas and a cell targeting agent, for a time sufficient for binding of the microbubbles to target cells; (b) contacting the biological sample with nanodroplets, wherein the nanodroplets comprise a perfluorocarbon liquid, to thereby expand and increase buoyancy of the microbubbles bound to the target cells; and (c) isolating the microbubbles bound to a cluster of the target cells from the biological sample, thereby isolating the cluster of the target cells, wherein the isolated target cells are substantially unchanged as compared to prior to isolation. “Substantially unchanged” as used herein in the context of cells refers to the absence of significant (e.g., more than 10%, 20%, 30%, 40%, or more) changes in a certain attribute (e.g., size, membrane integrity) of the cells. “Substantially unchanged” can reflect the absence of significant impacts of various stressors on the physiological state related to isolating cells or cell clusters as compared to their physiological state in vivo. For example, cells or cell clusters isolated (e.g., retrieved) using the methods described herein are substantially unchanged as compared to prior to isolation because they are not exposed to mechanical or shear stress, centrifugation, enzymatic digestion, ionizing radiation, alkylating agents, reactive oxygen species, elevated temperatures, reduced temperatures, and/or other physiological stressors. In some embodiments of the methods described herein, retrieved cell clusters remain intact (e.g., undisturbed). In some embodiments, retrieved cell clusters retain 50%, 60%, 70%, 80%, 90%, or 99% of the cell cluster size and/or number prior to isolation. In some embodiments, individual cells or cell clusters increase in size by within less than 1%, 1-10%, or 5-10%. In some embodiments of the methods described herein, the method further comprises a step of exposing the isolated microbubbles bound to the cluster of the target cells to air for a time sufficient for release of the cluster of the target cells from the microbubbles, thereby releasing the cluster of the target cells. For example, microbubbles can be eliminated from isolated cell clusters by exposure of the microbubbles bound to cells or a cell cluster complex to atmospheric air in a time sufficient to enable bursting of microbubbles. In some embodiments, the time sufficient to enable bursting of microbubbles is 30 seconds, 1-10 minutes, 10-20 minutes, 20-30 Page 18 of 32 WBD (US) 59194535v1 minutes, or more than 30 minutes. In some embodiments, exposure of isolated cell clusters to atmospheric air bursts 50%, 60%, 70%, 80%, 90%, 99% or more of attached microbubbles. In some embodiments, exposure of isolated cell clusters to atmospheric air bursts all attached microbubbles. In one aspect, the methods provided herein enable the use of isolated cells or cell clusters for any purpose, e.g., scientific, diagnostic, and therapeutic purposes, including but not limiting to in vitro culturing, molecular profiling and analysis (e.g., genomics, proteomics, glycomics), drug sensitivity analyses, and/or precision medicine development. The method provided herein can offer improvements on the current cell isolation buoyancy method using targeted MBs, the inventors have demonstrated in vitro that the addition of liquid PFC NDs (300 nm) to a MB (0.2- 10 µm) suspension dramatically expands the gas bodies by up to 6 orders of magnitude, and does so without direct contact. NDs act as a PFC and phospholipid reservoirs that transfer to the adjacent NBs or MBs to trigger their growth. According to one aspect of the present disclosure, the number of attached MBs per cell required to induce buoyancy and cell recovery could decrease significantly, as only one attached MB could potentially grow to become 100 µm. This aspect can improve upon the buoyancy technique used to isolate cells. It can also be applied in vivo, by administering the NBs or MBs intravenously targeted to a circulating cell surface receptor of interest, and then adding the liquid PFC to an extracted blood sample to detect the targeted cells. Aspects of the present disclosure can improve upon existing technology in at least two ways: (1) Increasing buoyancy using PFC transfer from superheated PFC NDs will decrease the number of MB per cell needed to cause cells to float; and (2) targeted NBs that are more effective at locating their target both in vitro and particularly in vivo, and allow the accumulation of more NBs at the cell surface, can also induce attached cells to float when they are inflated by the liquid PFC. The current standard for cell isolation is to target magnetic beads to the cell surface receptor of interest that can only be done in vitro and requires special equipment for isolation and then cell handling to remove the magnetic beads. The use of buoyancy to isolate cells improves upon the magnetic bead strategy by not requiring MB removal after isolation, simplifying the isolation technique, and adding the potential of administering the MBs intravenously to search for circulating cells of interest prior to isolation. The current standard for cell isolation that is FDA approved, is the addition of CTC-targeted magnetic beads to the blood sample. This requires specialized equipment not only for isolation, but also to remove the magnetic beads if Page 19 of 32 WBD (US) 59194535v1 downstream analysis is desired. Although isolated CTCs can be further analyzed to investigate their molecular and genetic features, this method decreases their viability, preventing cell expansion and live cell analyses. This method, developed for single circulating tumor cells could accidentally retrieve cell clusters. However, should clusters be retrieved, they could be disrupted during processing, underestimating their count. The use of buoyancy to isolate cells improves upon the magnetic bead strategy because the removal of attached MBs does not require specialized equipment or processing. Similar to cell isolation with MBs, the methods provided herein that adds NDs to inflate MBs merely improves retrieval to lift the much heavier and larger CTC clusters, without adding complexity. The present invention provides sensitive and gentle methods of cell (e.g., CTC cluster) isolation. In the methods provided herein, MB inflation can be used to further increase buoyancy to lift heavier cells and particularly clusters. The present invention does not require specialized equipment, it can interrogate any volume of whole blood, and it retrieves cells and clusters with minimal processing to provide live CTCs clusters in the supernatant within minutes without centrifugation to allow for downstream processing. High viability and unaltered characteristics is attributed to minimal processing and the fact that MBs are easily eliminated because their shell is phospholipid similar to cell membranes, and the highly volatile PFB gas core, is easily lost by simple non-violent manipulation such as minimal increase or decrease of ambient pressure or upon contact with air. Aspects of the present disclosure can provide a solution to when not enough MBs attach to the cell or cell cluster of interest to make it buoyant, or if NBs are used to improve cell interaction but the total gas bodies remain insufficient to cause attached cells to float. When a blood sample or any cell or particle suspension that contains the NBs or MBs attached to cells or particles that need to be isolated is spiked with liquid PFC, preferably as an emulsion of superheated PFC ND such as PFB ND that may be targeted or non-targeted to the attached NBs or MBs, the gas bodies will inflate because of the transfer of PFC molecules from the ND liquid core to the MB gaseous core. This increased buoyancy phenomenon that causes the attached cells/particles to float results directly from the unique physicochemical properties of PFC (e.g., volatility, hydrophobicity, lipophobicity, extreme density between liquid vs gas PFC). Based on our theoretical model of PFC diffusion from NDs to an adjacent gaseous environment, the transfer is because gaseous PFB has less positional restrictions and thus higher entropy than in the liquid state, and liquid PFB is at much higher pressure than its gaseous counterpart due to the Page 20 of 32 WBD (US) 59194535v1 extreme difference in density (1417 for PFB liquid vs 9.9 kg/m 3 for PFB gas).This new strategy should improve the detection limits of cells or particles in any suspension. With conventional buoyancy-based existing techniques, the buoyancy of cells or particles to be detected or isolated depends on the number and size of MBs attached to their surface to overcome the gravitational force exerted on the attached cell or particle. This can be a limitation that severely impacts detection sensitivity when using buoyancy for isolation. Aspects of the present disclosure addresses this problem and improves the sensitivity of isolation by simply adding liquid PFC, preferably as emulsion of superheated PFC ND such as PFB ND, to inflate the attached MBs or NBs, increasing the force to overcome the gravitational force increasing buoyancy and improving cell isolation. Unlike magnetic bead isolation, aspects of the present disclosure can be used in vivo to search for the rare circulating cells and clusters, and do not require cell manipulation to remove the beads after they are isolated. Unlike current buoyancy techniques, aspects of the present disclosure will allow the use of NBs that are more efficient at targeting, and can overcome the gravitational force for large cell or particle masses, by inflating the gas bodies attached to the cell or particle surface. For therapy, there are several schemes that this technology offers that could be exploited. These are merely some potential scenarios out of several. Analyzing Molecular Profile of Isolated Cells Methods are provided herein for evaluating the molecular profile, e.g., the genomic profile, proteomic profile, and/or glycomic profiles of isolated cells from a human subject in need thereof. In some aspects, provided herein are methods for isolating and retrieving intact, live cells of interest from a subject. In some embodiments, the method comprises a) contacting a patient sample with microbubbles, wherein the microbubbles target a specific cell target of interest, and allowing for sufficient binding of the microbubbles to the target cells; b) contacting the patient sample with nanodroplets to expand the microbubbles, increasing their buoyancy; c) retrieving the buoyant target cells coupled to microbubbles; d) identifying the genomic, proteomic, and/or glycomic profiles of the retrieved cells; and e) treating the subject with a treatment regimen predetermined to be effective in a subject with cells having the identified genomic, proteomic, and/or glycomic profiles. In some embodiments, DNA and/or RNA can be extracted from the isolated cells, sequenced, and analyzed for known biomarkers. Methods for genomic profiling are well known Page 21 of 32 WBD (US) 59194535v1 in the art and can be applied to cells isolated using the methods described herein (for example see Malone, E.R et al https://doi.org/10.1186/s13073-019-0703-1 and Nakagawa and Fujita DOI: 10.1111/cas.13505). In some embodiments, the proteomic profile of isolated cells can be evaluated via mass spectrometry or western blotting. In some embodiments, immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH) can be used to detect specific antigen biomarkers. Methods for proteomic profiling are well known in the art and can be applied to cells isolated using the methods described herein (for example, see Gast, MCW et al DOI: 10.1007/s10549-008-0263-3 and Ornstein and Tyson DOI: 10.1016/j.urolonc.2005.11.035 and Duarte and Spencer DOI: 10.3390/proteomes4040029). In some embodiments, the glycomic profile of isolated cells can be determined using mass spectrometry. Methods for glycomic profiling are well described in the art and can be applied to cells isolated using the methods described herein (for example, see Guo et al DOI: 10.3389/fendo.2022.970489 and Krishnamoorthy and Mahal DOI: 10.1021/cb900103n). In some embodiments, functional data regarding drug sensitivity of the isolated cells is evaluated by cellular toxicity assays or identification of specific drug resistance biomarkers as is well described in the art. The genomic, proteomic, glycomic, and/or functional data determined from the above methods can inform and optimize treatment for the subject dependent on the molecular-based therapeutic options available. In one aspect, the methods described herein can be used to determine a precision medicine treatment for a patient in need thereof. In some embodiments, genomic data obtained from isolated cells and subsequent biomarker analysis determines a treatment option for a patient in need thereof. In some embodiments, the proteomic data obtained from isolated cells and subsequent biomarker analysis determines a treatment option for a patient in need thereof. In some embodiments, the glycomic data obtained from isolated cells and subsequent biomarker analysis determines a treatment option for a patient in need thereof. In yet other embodiments, a combination of the genomic, proteomic, and/or glycomic data obtained from isolated cells and subsequent biomarker analyses determines a treatment option for a patient in need thereof. In some embodiments, the treatment is for cancer. In some embodiments, the cancer is selected from the group consisting of breast cancer, lung cancer, bladder cancer, colon cancer, pancreatic cancer, hepatic cancer, cholangiocarcinoma, gastric cancer, and prostate cancer. In some embodiments, the isolated cells are CTCs which can inform a precision oncology approach for treatment as Page 22 of 32 WBD (US) 59194535v1 described in the art (see Labib and Kelley DOI: 10.1002/1878-0261.12901). In some embodiments, the treatment is co-administered with other cancer treatment modalities known in the art. General Considerations It should be noted that whenever a value or range of values of a parameter are recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this disclosure. The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element, e.g., a plurality of elements. The term “including” is used herein to mean, and is used interchangeably with, the phrase “including but not limited to.” The term “including” does not necessarily imply that additional elements beyond those recited must be present. The term “about” or “approximately” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and, thus, the number or numerical range may vary from, for example, between 1% and 20% of the stated number or numerical range. In some aspects, “about” indicates a value within 20% of the stated value. In more preferred aspects, “about” indicates a value within 10% of the stated value. In even more preferred aspects, “about” indicates a value within 1% of the stated value. Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth as used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless otherwise indicated, the numerical properties set forth in the following specification and claims are approximations that may vary depending on the desired properties sought to be obtained in aspects of the present invention. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical values; however, inherently contain certain errors necessarily resulting from error found in their respective measurements. The term “at least” prior to a number or series of numbers is understood to include the Page 23 of 32 WBD (US) 59194535v1 number adjacent to the term “at least”, and all subsequent numbers or integers that could logically be included, as clear from context. When “at least” is present before a series of numbers or a range, it is understood that “at least” can modify each of the numbers in the series or range. Where a range of values is provided, it is understood that each intervening value (e.g., to the tenth of the unit of the lower limit unless the context clearly dictates otherwise) between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention. A “patient” refers to a subject who shows symptoms and/or complications of a disease or condition, is under the treatment of a clinician (e.g., an oncologist), for example, has been diagnosed as having a cancer, and/or is at a risk of developing a cancer. The term “patient” includes human and veterinary subjects, and also including patients of any age (e.g., adults, children, and infants). Any reference to subjects in the present disclosure, should be understood to include the possibility that the subject is a “patient.” As used herein, a “subject” is an animal, such as a mammal, including primate and non- primate animals, and in particular aspects the subject is a human. As used herein, the terms “treating” or “treatment” refer to a beneficial or desired result, such as reducing at least one sign or symptom, e.g., respiratory distress, in a subject. “Treatment” also refers to prevention of a disease or a condition, or prevention of at least one sign or symptoms associated with the disease or the condition (a prophylactic treatment). “Treatment” can also mean prolonging survival as compared to expected survival in the absence of treatment. Further description and explanation of the operating principles can also be found in the discussion of the example and results that follow. Page 24 of 32 WBD (US) 59194535v1 Examples Example 1: Retrieval of Labeled Silica Beads Using Targeted Microbubbles The ability for microbubbles (MB) to retrieve heavy particles was assessed. Approximately 1000 fluorescein isothiocyanate (FITC)-labeled 50-micron silica beads were suspended in 1 mL of PBS. Approximately 10 7 FITC-targeted MBs were then added to the solution, incubated for 30 min (20 RPM) and the entire MBs-beads suspension was placed in a microwell plate (24-well plate). While the tiny beads, present as impurities in the commercially- purchased silica beads, floated (FIG.2A), large beads rapidly settled to the bottom despite the attached MBs (FIG.2B). After the beads settled, 10 10 MB-targeted perfluorobutane (PFB) nanodroplets (ND) were added to the solution. Minutes after addition of the NDs, the large beads floated to the surface (FIG.2C). The ability for MBs was further assessed using a mixture of FITC-labeled 50-micron and 12-micron silica beads. Beads were suspended in 1 mL of PBS and FITC-targeted MBs were added as described above. Without the addition of any MBs, smaller beads floated (FIG.2D). Following the addition of FITC-targeted MBs, silica beads settled to the bottom despite the attached MBs (FIG.2E). Following the addition of MB-targeted NDs as described above, a significant portion of FITC-labeled beads floated to the surface (FIG.2F). The weight of 50- micron silica beads is greater than or equal to that of 130, 15-micron circulating tumor cells (CTCs), suggesting that this approach would work on CTC clusters which average approximately 50 cells. Comparing particle size distribution of FITC-labeled silica beads to the distribution retrieved after adding FITC-targeted MBs or after adding targeted MBs and MB- targeted NDs showed that 50-µm beads could only be retrieved with NDs and the addition of NDs markedly skewed the distribution towards large particle sizes. Example 2: Retrieval of Labeled Cells from Whole Blood Using Targeted Microbubbles The ability for MBs to retrieve cell types of various sizes was assessed. FITC-labeled red blood cells (RBCs), with an average size of 5 microns, or FITC-labeled K562 cells, with an average size of 15 microns were used. FITC-labeled K562 cells are approximately 27 times heavier than that of the FITC-labeled RBCs. Cells were added at various amounts, ranging from 10 3 to 10 6 cells into 7.5 mL of whole blood. FITC-targeted MBs were subsequently added to both and mixed. In one set of samples, non-targeted NDs were added, and retrieval rate of cells was Page 25 of 32 WBD (US) 59194535v1 assessed using fluorescence microscopy and hemocytometry. The addition of non-targeted NDs increased the retrieval rate for RBCs and K562 cells by 2- and 3-fold respectively as compared to that of MBs alone (FIG.3). The addition of targeted NDs increased the overall retrieval rates by an additional ~25%. Retrieval rates for K562 cells were comparable to that of CTC retrieval rates for FDA-approved methods using magnetic beads. Example 3: Retrieval of Live H2122 Cells from Media Using Targeted Microbubbles H2122 lung carcinoma cells that express Epithelial cell adhesion molecule (EpCAM) and grow in grape-like clusters in suspension were cultured. The suspension was divided into wells of 96-well plates and counted using an automated inverted microscope (Cytation 5, Biotek). The entire content of each well containing clusters were combined to produce 3 mL samples of either full media or whole blood containing 1, 2, or 30 clusters of H2122 cells. For a given sample, either 3x10 7 MBs targeted to EpCAM or 3x10 7 MBs conjugated to a non-specific antibody were added, and the sample was mixed head over heel for 30 minutes. After the 30 min mixing step, rosettes of anti-EpCAM-targeted MBs bound to clusters were seen when analyzed via microscopy (FIG.4A). No similar rosettes were seen in the samples with control MBs. Subsequently, 3x10 10 MB-targeted NDs were added to the sample and the supernatant was collected after 10 minutes. Cell retrieval was quantified using microscopy and ImageJ, with the size range of clusters shown in FIG.4B. The control MBs conjugated to a non-specific antibody failed to retrieve any cells or clusters. When only 1 or 2 clusters were present in the sample, they could only be isolated after the addition of MB-targeted NDs (FIG.4C). When 30 clusters were present in the sample, the addition of MB-targeted NDs increased retrieval ~15X as compared to MBs alone (FIG.4C). Isolated H2122 cells were stained with a live/dead contrast stain, acridine orange (AO) and propidium iodide (PI) to determine viability . When assayed promptly after retrieval, approximately 95% of clusters remained alive, for which a representative image is shown in FIG.5A-C. Example 4: Retrieval of H2122 Cells from Whole Blood Using Targeted Microbubbles H2122 lung carcinoma cells were used as described in Example 3.300 clusters of H2122 Page 26 of 32 WBD (US) 59194535v1 cells were spiked into 3 mL of whole blood, to which 100 anti-EpCAM-labeled MBs / cluster were then added and the sample was mixed head over heel for 30 minutes. Following the 30 min mixing, 1000 MB-targeted NDs/MB were added to the sample, and the supernatant was collected after 30 minutes. Supernatant was washed three times with PBS to remove RBCs (FIG.6A) and the supernatant was stained with DAPI and AF488-anti-EpCAM (FIG.6B). Stained samples were then quantified using microscopy, which demonstrated that intact clusters could be isolated from whole blood that showed similar cellular morphology and markers as the unprocessed H2122 cells (FIG.6B). Example 5: Retrieval of Clusters from a Syngeneic Metastatic Triple Negative Breast Cancer Mouse Model A syngeneic metastatic model of triple negative breast cancer in Balb/c mice was used to assess the ability for MB/NDs to retrieve naturally occurring clusters. A triple negative stably transfected luciferase-expressing 4T1 (Luc-4T1) metastatic breast cancer cell line was used to produce CTC clusters in mice.10 6 Luc-4T1 cells were implanted in the mammary fat pad of female Balb/c mice. When lung metastases were detected (~16 days) with bioluminescence imaging, mice were killed and the blood was collected via cardiac puncture. Blood samples were combined for all mice and then split into 2 equal halves. To both samples, 10 7 anti-EpCAM- labeled MBs were added and mixed head over heel for 30 mins. Following the mixing step, 10 10 MB-targeted NDs were added to one sample, with the MB-only sample serving as a control. Supernatant was collected after 30 mins, and stained with APC-antiCD45, AF488-antiEpCAM, and DAPI. Samples were assessed via fluorescence microscopy, which showed that CTC clusters were isolated only with the addition of both EpCAM-targeted MBs and MB-targeted NDs, for which a representative image is shown in FIG.7. Retrieved clusters were bioluminescent which further validated that the retrieved clusters were alive and originated from the Luc-4T1. All publications, patents, and patent applications mentioned in this specification are incorporated herein by reference to the same extent as if each independent publication or patent application was specifically and individually indicated to be incorporated by reference. While the invention has been described in connection with specific embodiments thereof, Page 27 of 32 WBD (US) 59194535v1 it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the invention that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims. Page 28 of 32 WBD (US) 59194535v1