Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AN AUTOMATED DEVICE AND METHOD TO PURIFY BIOMATERIALS FROM A MIXTURE BY USING MAGNETIC PARTICLES AND DISPOSABLE PRODUCT-CONTACT MATERIALS
Document Type and Number:
WIPO Patent Application WO/2022/165310
Kind Code:
A1
Abstract:
This invention relates to a device and method of using the device for purification that separates material of interest from contaminating materials using non-porous magnetic particles and single-use or disposable materials that come in contact with the material of interest. The process encompasses multiple cycles in a single batch to reduce the cost of magnetic particles. This method can be executed in a fully automated manner by a controller that manages different inputs and outputs of system hardware.

Inventors:
MEHTA SUNIL (US)
Application Number:
PCT/US2022/014511
Publication Date:
August 04, 2022
Filing Date:
January 31, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ATHEMBIO (US)
International Classes:
B03C1/28; B01L3/00; G01N33/543
Foreign References:
US20160184737A12016-06-30
US20190339261A12019-11-07
US20120270331A12012-10-25
US20210260600A12021-08-26
Other References:
SHAIKH YONAS S., PERCY KAMPEIS: "Development of a novel disposable filter bag for separation of biomolecules with functionalized magnetic particles", ENGINEERING IN LIFE SCIENCES, vol. 17, no. 7, 7 March 2017 (2017-03-07), pages 817 - 828, XP055954382, DOI: 10.1002/elsc.201600190
Attorney, Agent or Firm:
SCHWARTZMAN, Robert A. (US)
Download PDF:
Claims:
We claim: 1. A device to separate material(s) of interest from a mixture containing plurality of materials, the device comprising: a disposable separation chamber with an inlet and an outlet; a disposable bag connected by tubing to the inlet and the outlet; a pump configured to move fluid in a tubing system including between the bag and the separation chamber; at least one valve configured to direct fluid in the tubing system; at least one pH, conductivity, pressure, occlusion, bubble, and/or optical density sensor; a magnetic field generation unit configured to generate a magnetic field adjacent the separation chamber; a controller in communication with: (i) the pump to control its speed and direction, (ii) the magnetic field generation unit to control a strength of the magnetic field (iii) the valve(s) to control its/their open or closed state (iii) the sensor(s)s to receive inputs from the sensor(s) (iv) a human machine interface in which a user inputs recipe parameters for a process and a user can input to control the pump, the valve(s), and the magnetic field; wherein in operation, the controller changes the state of valve(s) and creates a fluid path through which the pump transfers a fluid to the separation chamber, wherein in operation, the controller changes the state of valve(s) and creates a fluid path through which the pump recirculates the fluid to provide mixing of particles with the fluid, wherein in operation, the controller increases the strength of the magnetic field to immobilize particles in the separation chamber, wherein in operation, the controller changes the state of valve(s) and creates a fluid path through which the pump transfers a fluid to the bag, wherein in operation, the controller lowers the strength of the magnetic field to suspend particles in the separation chamber. 2. The device of claim 1, wherein the separation chamber has a conical bottom with apex at its bottommost point.  

3. The device of claim 1 or 2, further comprising particles that: (i) are mostly non-porous; (ii) are attracted to a magnetic field; and (iii) have an outer surface with affinity for a material of interest. 4. The device of claim 3, wherein the average size of the particles is between 10-5000 nanometers. 5. The device of claim 3 wherein the particles have cubic or tetrahedron shape with an average size between 10-5000 nanometers. 6. The device of any one of claims 3-5, wherein the particles have a surface to area ratio per unit volume greater than 4.8. 7. The device of any one of claims 1-6, wherein at least 10 percent of product-contact materials are disposable, e.g., at least 20, 30, 40, or 50 percent. 8. The device of any one of claims 1-7, wherein the magnetic field generation unit is configured to create a high gradient magnetic field with magnetizable or ferromagnetic material in the separation chamber. 9. The device of any one of claims 1-8, further comprising an impeller in the separation chamber configured to mix particles with fluid. 10. The device of any one of claims 1-9, wherein the device is configured for more than one cycle of process to be run without changing product-contact material. 11. The device of any one of claims 1-10, wherein all product contact surfaces are sterilized prior to use.  

12. The device of any one of claims 1-11, wherein the separation chamber comprises a body and a cavity defined therein configured to receive a magnet, optionally with the magnet held in the cavity. 13. The device of claim 12 wherein the cavity comprises a material that has increased flexibility relative to a material of the body. 14. A particle that: (i) is mostly non-porous; (ii) is attracted to a magnetic field; and (iii) has an outer surface with affinity for a material of interest. 15. The particle of claim 14, wherein the size of the particle is between 10-5000 nanometers. 16. The particle of claim 14, wherein the particle has a cubic or tetrahedron shape with an average size between 10-5000 nanometers. 17. The particle of any one of claims 14-16, having a surface to area ratio per unit volume greater than 4.8. 18. A method of separating a material of interest from a mixture containing a plurality of materials, the method comprising: mixing a starting material containing a mixture of materials with particles that are attracted to a magnetic field and have specific affinity for the material of interest, placing the mixed starting material and particles in the separation chamber of the device of any one of claims 1-13, capturing particles in the separation chamber by changing the strength of a magnetic field and discarding or collecting the flow-through material, washing particles with a buffer in the presence of the magnetic field, washing particles by recirculating a buffer in the absence of the magnetic field and then in the presence of the magnetic field to retain particles in the separation chamber, and discarding wash buffer, introducing elution buffer in the separation chamber, and mixing via pump recirculation in the absence of the magnetic field followed by in the presence of the   magnetic field to elute the material specifically bound to the beads, and repeating this step to improve recoveries and to clean the beads, thereby separating the material of interest. 19. The method of claim 18, wherein the particles comprise the particle of any one of claims 14-17. 20. The method of claim 18 or 19, wherein the particles are regenerated by exposing the particles to regeneration buffer in the absence of the magnetic field followed by in the presence of the magnetic field to remove any non-specific impurities bound to the particles. 21. The method of any one of claims 18-20, wherein the starting material is unpurified or partially purified cell culture or microbial culture. 22. The method of any one of claims 18-20, wherein the starting material is unpurified or partially purified bodily fluid from animals or humans. 23. The method of any one of claims 18-20, wherein the material of interest is organic or inorganic material. 24. The method of any one of claims 18-20, wherein the material of interest is a virus, bacteria, cell, or organelle. 25. The method of any one of claims 18-20, wherein the material of interest is a protein, carbohydrate, lipid, DNA, or RNA. 26. The method of any one of claims 18-25, wherein the volume of starting material is greater than 50 milliliters. 27. The method of any one of claims 18-26, wherein the volume of starting material is greater than 1 liter.  

Description:
An automated device and method to purify biomaterials from a mixture by using magnetic particles and disposable product-contact materials Statement of Priority [0001] The present application claims priority from U.S. Application No.17/163,472, filed January 31, 2021, the disclosure of which is incorporated herein by reference in its entirety. Field of the Invention [0002] The present invention is related to methods and systems that are used for separating macromolecules, including biomaterials, such as cells, cellular components, proteins, lipids, carbohydrates and tissues from a mixture. Background [0003] Biotherapeutics have revolutionized healthcare globally. As compared to traditional chemical-based simple molecule drugs, biotherapeutics are generally complex molecules or particles. Many of the biotherapeutics are recombinant proteins, like antibodies, as they can have a very high affinity for therapeutic targets, fewer side effects, lower immunogenic rejection, and long half-life. Some other biotherapeutics include viruses, bacteria, whole cells or parts of cells (for example, exosomes), carbohydrates, lipids, DNA, or RNA. [0004] Biotherapeutics typically have higher manufacturing costs as compared to chemical- based drugs. Much of their manufacturing cost is attributed to complex downstream purification processes (MAbs.2009 Sep-Oct; 1(5): 443–452). [0005] Recombinant proteins can be produced using a variety of host expression systems, including bacteria, yeast, and mammalian cells. Recombinant antibodies are generally produced in mammalian cells as some other host systems lack sophisticated post-translational machinery that is required to produce functional antibodies. [0006] Due to the complexity of proteins, they are recombinantly produced in host organisms either intracellularly (e.g., bacterial host expression systems) or secreted extracellularly (e.g., yeast and mammalian expression systems). When mammalian expression systems are used to produce proteins, the cell culture harvest is clarified by separating cells from the supernatant using centrifugation and/or filtration technologies. Harvest clarification requires expensive   equipment and/or single-use filter modules, large manufacturing floor space, a large volume of buffers, and expensive utilities. Also, harvest clarification adds at least one additional day to the processing time. An increase in cell density in cultures improves productivity, which can reduce the overall cost of therapeutics. At the same time, the increase in cell density poses several challenges for traditional harvest clarification technologies. Disc-stack centrifuges are commonly used for harvest clarification to clarify cell culture and discharge unwanted cells in the form of a slurry. During this process, some of the product is lost with the discharged slurry; however, due to a limited amount of discharged cycles, it has an insignificant impact on the overall product yield if the cell density is low. If the cell concentration increases, product loss also increases as the number of discharge cycles increases with increasing cell density. Additionally, disc-stack centrifuges exert high shear force on cells, causing the release of host cell proteins and proteolytic enzymes in the clarified harvest (Subramanian, G. (2014). Continuous Processing in Pharmaceutical Manufacturing.: Wiley‐VCH Verlag GmbH & Co. KGaA). This not only creates downstream purification challenges but also negatively impacts product quality. Filtration based technologies also become impractical for high cell density cultures as they require very large systems and surface areas while causing significant loss of product and high levels of host cell protein contamination (Subramanian, G. (2014). Continuous Processing in Pharmaceutical Manufacturing.: Wiley‐VCH Verlag GmbH & Co. KGaA). [0007] High cell density not only poses challenges for upstream processes but also downstream processes as much larger chromatography columns are required for purification and host cell protein removal becomes challenging. Current chromatography technologies rely on porous beads to create large chromatography surface area with a relatively smaller volume. Due to the very small size of pores in beads, the penetration of the molecules to be purified requires significant diffusion time leading to long processing time. The chromatography columns packed with these beads also require high-pressure feeding of liquids due to small pore channels in the beads that increase flow resistance. In addition, each cycle requires a cleaning and regeneration step as the smaller pore size retains some of the non-specific materials after retrieval of the product of interest. Not only does this increase processing times, it also requires large amounts of additional buffers for washing and regenerating the chromatography resin. If the affinity ligands coupled to the resin are sensitive to cleaning agents, the chromatography material can only be used for one cycle. Also, therapeutics or vaccine macromolecules that have a larger size (e.g.,   viruses, bacteria, whole cells, or exosomes) than the pores in the chromatography resin cannot be purified using current chromatography processes. [0008] Regarding the materials used for the chromatography system, most systems use stainless-steel equipment that requires cleaning between batches to reduce cross-contamination. The cleaning validation of systems is expensive and adds significant delays between batches. In addition, the cleaning process requires significant infrastructure and once installed, the equipment becomes immobile as it is hard-piped. Single-use systems have gained traction in recent years due to their lower capital requirements, flexibility, reduced turn-around times, no- risk of cross-contamination between batches, and environmental friendliness. Although most of the single-use chromatography systems use disposable product contact surfaces, the column is still reused for multiple batches due to its expensive cost. Simulated moving bed technology has tried to make the columns disposable by increasing the number of cycles at the cost of adding additional processing time. However, as all these systems use porous chromatography media/resin, the challenges related to porous chromatography resin remain. [0009] Purified and unpurified biomaterials from bodily fluids are also valuable and used for many different purposes. For example, immunoglobulins purified from donated human plasma (intravenous immunoglobulin or IVIG) are used to treat several disorders including life- threatening sepsis. An anti-hemophilic factor, Factor VIII, purified from donated human plasma is used to treat hemophilia in factor VIII deficient patients. Immunoglobulins purified from animals immunized with toxins are used to treat poisoning from venomous snake bites. In the absence of a vaccine or therapeutic during a pathogenic outbreak, convalescent sera from a recovered healthy donor can be used as a therapeutic to treat critical patients or as a prophylactic treatment for the population at risk (Casadevall A, Scharff MD. Return to the past: the case for antibody-based therapies in infectious diseases. Clin Infect Dis.1995;21(1):150–161; Nat Rev Microbiol.2004;2(9):695–703). [0010] There are some drawbacks to using the convalescent serum. Generally, newly recovered donors are not able to donate large amounts of serum and can easily go into hypovolemic shock. In addition, donated unpurified sera can potentially harm the receiving critical patients as it contains many biomaterials (including infectious agents) other than therapeutic immunoglobulins (Gajic O, et al. Transfusion-related acute lung injury in the critically ill: prospective nested case- control study. Am J Respir Crit Care Med.2007;176(9):886–891). For example, viruses can   unintentionally be transmitted from donor’s serum to recipients. Also, immunological reactions from serum such as serum sickness are common (medlineplus.gov/ency/article/000820.htm ). [0011] During serum donations, a limited amount of the biomolecule of interest is collected from each donor as they cannot donate more than a certain amount (unless they are sacrificed). In many applications of plasma (including factor VIII, IVIG, and anti-venom), the plasma from several donors is shipped to a processing facility, pooled, and biomaterials of interest are isolated. This process usually takes months to complete, and if the goal is to combat a novel pathogen during an epidemic or a pandemic, the pathogen may mutate during that time and the purified therapeutic may not be effective. Summary of the Invention [0012] The devices, products, and methods described in this invention solve many problems associated with the purification processes of biotherapeutics. This method significantly reduces the total processing time and simplifies downstream processing by eliminating or reducing the need for clarification before chromatography. All product contact surfaces are suitable for biotherapeutics manufacturing (e.g., meet USP class VI requirements) and are replaced after each batch but can support multiple cycles of processing within the same batch. The method described here uses particles that are attracted to magnets (ferromagnetic, paramagnetic, or superparamagnetic), have a large surface area to volume ratio (per unit volume) due to their smaller size, are preferably non-porous, and have specific binding properties. These particles can be various forms of iron or cobalt oxides (e.g., Fe 3 O 4 ) or a mixture of them. The surface to area ratio per unit volume is greater than 2, e.g., greater than 3, greater than 4, greater than 4.8, or greater than 5, 10, 15, 20, or more. The magnetic core of the particles is rigid and non-porous, but the tethered ligands or linkers may create porous structure around the particles. This porous structure volume is less than the volume of the rigid core of the particles. In some embodiments, porous structure volume is less than 90 percent volume of the rigid core of the particles. In some embodiments, porous structure volume is less than 80, 70, 60, or 50 percent volume of the rigid core of the particles. Particles having a porous structure volume that is less than 90 percent volume of the rigid core of the particles are considered to be “mostly non-porous”. These particles exhibit very short residence time for binding as most of the binding sites are on the surface. The diameter or one of the sides of these particles is from 10-10,000 nm, which is up to   20,000 times smaller than the standard porous chromatography resin beads. In some embodiments, the size is less than 1000 nm, e.g., less than 900, 800, 700, 600, 500, or 400 nm. In some embodiments, the size is in the range of 10-5,000 nm, e.g., 50-1,000, 50-800, 100-500, or 200-400 nm. In some embodiments, the particles are non-spherical. In some embodiments, the particles are cubic or tetrahedral in shape. The surface of the particles is modified to have properties such as affinity for a specific type of molecules (e.g., modification with Protein A ligand for antibody purification or modification with antibodies that specifically bind a biomaterial), positive or negative charge, hydrophobic interactions, or a combination of two or more of these properties. The particles may be referred to as magnetic particles or beads in this invention. The biomolecules or biomaterials to be purified from the crude mixture can be antibodies, proteins, nucleic acids (e.g., DNA and RNA), organelles, viruses, viral vectors (e.g., adeno-associated viruses, lentivirus, poxviruses etc.), cells, exosomes, bacteria, yeast, pathogens, or other organic entities. [0013] The particles of the invention are mixed in with starting material which may be cell culture, partially clarified broth, blood or other bodily fluids, or other biomolecule-containing mixtures. An external magnetic field is applied to recover these particles from the mixture. If the magnetic susceptibility is low due to the smaller size of the magnetic particles, a high gradient magnetic field is used to isolate particles mixed in with the mixture. A high-gradient magnetic field is generated when a magnetic field is applied across an array of magnetically soft metal (e.g., steel wool). This results in a localized high magnetic field around the magnetically soft metal and allows separation of particles that are otherwise weakly attracted to magnets. Under challenging test conditions with a mixture of Fe 3 O 4 particles (300 nm; cubic shaped) and a high concentration of Saccharomyces cerevisiae in media, >99% of the particles were captured after application of a high gradient magnetic field generated by a magnetic field ranging from 0.1-1.7 Tesla. Small-scale batch chromatography methods to purify proteins (in microgram or nanogram scale) using magnetic beads are currently available but those mostly use porous magnetic beads that are not recycled and are operated manually. Commercially available magnetic particles are spherical with a diameter that is equal to or greater than 1 micron and used only for a single cycle at a very small scale (e.g., Sera-Mag from GE Healthcare Life Sciences and Dynabeads from ThermoFisher Scientific). These particles are not well suited for the current invention due to their porosity, large size, and recycling capabilities.   [0014] The magnetic particles of the invention have a high surface area to volumetric ratio. For example, the surface area to volumetric ratio of tetrahedron and cubic particles is at least 25% greater than the ratio of spherical particles. The attachment of ligands to the surface of magnetic particles can be achieved directly with the ligand or by a linker that can bind to the ligand and the inorganic surface of magnetic particles. In one embodiment, linkers with silane groups are used to chemically bond with magnetic particles. The other end of the linker is chosen such that it can readily bind to the ligand that is used for purification. For example, the epoxide group at the other end of the linker can react with primary amine, sulfhydryl, or carboxyl groups that are commonly found on different amino acids of proteins.3-glycidoxypropyl trimethoxysilane or 3- glycidoxypropyl triethoxysilane are two examples of such linkers. In studies by the inventors, 0.5-3% w/w treatment with 3-glycidoxypropyl trimethoxysilane provided excellent conjugation to Fe3O4 cubic particles of 300 nm average size. [0015] Ligands can be proteins (e.g., antibodies, single chain antibodies, affinity proteins, poly- lysine, poly-arginine), carbohydrates (e.g., heparin), matrices (e.g., naturally secreted polymers), polynucleotides (e.g., oligo dT), nucleic acids (e.g., cDNA, primers), or molecules (e.g., primary, secondary, and tertiary amines, sulfonic acid group) that have strong affinity for specific macromolecules or particles. [0016] During affinity chromatography, macromolecules of interest bind to the magnetic particles that have a surface affinity for those macromolecules. The recovered magnetic particles can be washed multiple times to remove impurities and then elution is performed by introducing an elution buffer that dissociates the binding of the macromolecules of interest from the magnetic particles. In one embodiment, a low pH buffer is used to dissociate antibodies that are captured by protein A coupled magnetic particles. Similarly, imidazole is used to elute histidine-tagged protein bound to Ni-NTA particles and high salt buffers are used to remove charged proteins bound to ionic exchange particles. The eluted macromolecules flow through while the magnetic particles are retained due to the external magnetic field. If the affinity performance of the particles is reduced after a certain number of cycles, due to nonspecific occlusion of specific binding sites by impurities, the particles can be regenerated using regeneration solutions (e.g. NaOH or chaotropic agents such as urea or guanidine). [0017] Flow-through chromatography reduces impurities by binding the impurities to magnetic particles and letting the macromolecules of interest flow through. The surface of magnetic   particles is modified such that they have an affinity for impurities but not for the macromolecules of interest. These impurities could be DNA, host cell proteins, or other unwanted macromolecules. In one embodiment, positive-charged quaternary ammonium magnetic particles can be used to remove contaminating negative-charged DNA from the protein of interest. The contaminating impurities bind to magnetic particles while macromolecules of interest flow through and are collected. After each cycle, magnetic particles are regenerated to remove the bound contaminants so that magnetic particles can be reused for subsequent cycles. [0018] These and other aspects of the invention are set forth in more detail in the description of the invention below. Brief Description of the Drawings [0019] Fig.1 depicts an example of an affinity purification method. [0020] Fig.2 depicts an overview of the process to purify immunoglobulins from human blood. [0021] Fig.3 depicts an example to purify biomaterials from bodily fluids. [0022] Figs.4A-4B depict dynamic binding capacities of protein A coupled magnetic particles with (A) purified polyclonal IgG and (B) polyclonal IgG from whole blood. [0023] Fig.5 depicts an example of a chamber with rigid and flexible parts. [0024] Figs.6A-6B depict (A) binding kinetics and (B) reusability of protein A coupled magnetic particles. [0025] Fig.7 depicts an example of a chamber with multiple magnet cavities. Detailed Description of the Invention [0026] The binding of macromolecules or impurities (for flow-through mode) is performed in a mixing container where the magnetic particles are mixed in with a starting solution containing a mixed population of macromolecules and impurities (e.g., cell culture or partially clarified cell culture). Mixing is performed to improve the binding kinetics of the target molecule to the magnetic particles. The mixing can be performed by employing standard mixing technologies or by recirculation of fluid via a single pump or multiple pumps (e.g., P1 in Fig.1). The material moves through the separation chamber if the pumps are used for mixing (Fig.1). The mixing generated by a pump can be enhanced by reversing the flow intermittently, using very high flow rates, and starting mixing before the removal of the magnetic field. After the binding of   macromolecules to the magnetic particles is complete, the magnetic field is introduced via either switching on electromagnets or physically moving permanent magnets closer to the separation chamber. To capture smaller particles with low magnetic susceptibility, a high gradient magnetic field can be generated by employing ferromagnetic stainless-steel wool or other ferromagnetic support material inside the separation chamber. Multiple permanent magnets can also be used to improve retention efficiency of the magnetic particles. In one embodiment, a series of magnets is placed (or magnetic field is applied) near the inlet to capture incoming magnetic particles and another series of magnets is placed (or magnetic field is applied) near the outlet to capture any remaining particles that are not captured by the magnets placed near the inlet. The magnetic particles with the bound target molecule are retained while the remaining material is pumped out of the system into waste. The retention efficiency of the magnetic particles can again be increased by employing standard mixing technologies (e.g., rotating impeller) or by simple recirculation of fluid via a single pump or multiple pumps. Mixing not only increases the retention efficiency but also provides homogeneous capture of magnetic particles on a ferromagnetic support material. In the absence of mixing the magnetic particles can easily clog the ferromagnetic support material and can cause overpressure. The mixing can be gradually reduced for complete capture of magnetic particles as increased mixing will prevent complete capture of particles. If overpressure is detected by the pressure sensor, a fluid path is created (e.g., V3 opens with remaining valves closed) and a pump (e.g., P1) recirculates the liquid within the separation chamber without a magnetic field. Then, a gradually increasing or constant magnetic field can be applied so that particles are uniformly captured in the separation chamber. [0027] Magnetic particles may require resuspension during the initial binding and washing. The resuspension of the particles is performed by turning off the magnetic field and recirculating liquid (between inlet and outlet of the separation chamber) at high flow rates by pumps or mixing within the separation chamber using standard rotary, reciprocating, or vibrating impellers. The chamber shape and material have an impact on resuspension of the particles. [0028] During affinity purification, the bound particles are washed either in the presence or absence of a magnetic force to remove any nonspecifically bound impurities. The bound macromolecules are eluted from the magnetic particles by the introduction of an elution buffer to the separation chamber in the presence or the absence of an external magnetic force. If the binding performance of particles reduces after multiple cycles, the particles can be cleaned using   a sanitization buffer (e.g., sodium hydroxide, urea, or guanidine hydrochloride). The whole cycle can be repeated multiple times until a complete batch is purified. [0029] In case of flow-through purification, macromolecules of interest (e.g., recombinant proteins) do not bind to the magnetic particles and flow through while the impurities (e.g., DNA) are captured by the magnetic particles. Magnetic particles can be regenerated using a sanitization buffer for additional cycles. [0030] One of the advantages of using nonporous magnetic beads is that unlike porous chromatography resins, the impurities do not get trapped in the pores and require significantly less sanitization. Porous chromatography media requires sanitization with high sodium hydroxide concentration (e.g., 0.5 M) after every cycle. Fifty purification cycles were performed to isolate immunoglobulins from whole human blood using protein A coupled magnetic particles. Sanitization with a low sodium hydroxide concentration of 0.1 N was performed after every 5 cycles, Results (Fig.6B) show that binding capacity of the magnetic particles did not change significantly throughout 50 cycles. In each cycle, a specific volume of starting material (out of a batch) is pumped into the system. This is called the cycle volume and is calculated based on the amount of magnetic particles that are loaded into the system, the estimated amount of material to be purified (product or impurity amount), the desired number of cycles or the total processing time, and recycling capability of magnetic particles. For example, if protein A magnetic particles with a capacity of 50 g/L (gram of antibody captured per liter of magnetic particles) are used to affinity purify a batch of 2000 L with 5 g/L antibody concentration, a single cycle will require 200 L of magnetic particles. On the other hand, if the magnetic particles are recycled for 50 cycles, only 4 L of magnetic particles are required. The time required to run 50 cycles will be around 50 times more than to run a single cycle, but the savings realized from using a lesser amount of magnetic particles and smaller size of the equipment greatly outweighs the longer processing time. If a single cycle takes 10 minutes, 50 cycles will take just over 8 hours, which is a reasonable processing time for a 2000 L batch as traditional methods require 2- 4 days of processing. [0031] Depending on the binding affinity of the ligand and the concentration of molecule to be purified, the chromatography media may have maximum binding capacity when the concentration of molecule to be purified is much higher than the binding capacity of the time media. In this case, the dynamic binding capacity (when most of the molecules are bound and   some are unbound) will be lower than the maximum binding capacity. The inventors’ results with purified polyclonal human IgG show that there was no difference between the maximum binding capacity and dynamic binding capacity of magnetic particles coupled with protein A (Fig.4A). However, results from male human blood show that maximum binding capacity was over 50 g/L (grams of purified antibody per liter of magnetic particles) whereas the dynamic binding capacity was about 40 g/L (Fig.4B). [0032] Referring to Fig.5, in regard to the separation chamber 10, its shape is designed with an inlet 12 and an outlet 14 such that they are away or spaced apart from each other (e.g., positioned on opposite sides of the chamber). Spaced apart inlet and outlet allow maximal residence time within the chamber for the material entering the chamber. The internal diameter of tubing is large enough (e.g., larger than 0.125 inch diameter) to prevent clogging from magnetic particles before they are completely suspended. In one embodiment, a cylindrical separation chamber with a conical bottom (with the apex at the bottom) is used to isolate magnetic particles during different parts of the process. In another embodiment, the top of the separation chamber is conical (with the apex at the top). A fully-cylindrical shaped chamber can also be used. Another alternative is a cuboid shaped separation chamber that can provide better resuspension of particles by improving mixing. The separation chamber is fabricated out of materials that are poorly attracted by a magnetic force (e.g., LDPE, high durometer silicone, polypropylene, polystyrene, EVA, PVC, etc.). The chamber can be made out of rigid material or flexible material or a combination of both (Fig.5). If the complete separation chamber is constructed from rigid material and the magnetic field is removed from the separation chamber, the magnetic particles tend to compact and stick to the walls even after strong mixing. This is problematic as it leads to incomplete washing resulting in increased impurities in the elution, and it reduces the capacity of the media. When the outer shell or body 16 of the separation chamber 10 is rigid (e.g., polycarbonate or polypropylene) and the shell holding the magnets (magnet cavity) 18 is flexible (e.g., silicone), experimental results show that concentrated magnetic particles mix more rapidly after the magnetic field is removed. In addition, the results show that separation of magnetic particles from fluid is much faster. The increased separation dynamics is due to the flexible material’s ability to protrude into the chamber, resulting in an increase of the exposed magnetic surface area for the particles to attract to and the flexible material in between the particles and magnet adapts to the shape of magnet to provide higher magnetic field. Magnet cavity is shaped as a sleeve or   cover to the magnet (temporary or permanent) (see, e.g., M in Figs.1 and 3). The chamber may contain steel wool or wire mesh 20 to enhance the magnetic field via high gradient magnetic field generation, to capture magnetic particles that are small and have weak magnetic susceptibility. In one embodiment, the material of steel wool or wire mesh (1 micron-1000 micron) is ferromagnetic stainless-steel (e.g., 430 or 410 stainless steel) to provide corrosion resistance. The ferromagnetic steel wool can also be coated with metals (e.g., nickel or chromium) or plastics to substantially reduce or eliminate corrosion. Steel wool or wire mesh can be situated anywhere within the chamber preferably around the magnet cavity or cavities. When the process is scaled-up, either the chamber can be scaled up appropriately or multiple chambers can be used in parallel or multiple magnet cavities can be added to an appropriately scaled-up chamber (Fig.7). For example, if 50 mL of magnetic chromatography media in one chamber with single magnet cavity is used for purification from 10 L of starting material, the scaled-up process to purify 40 L of material may include a four times scaled up chamber in volume with one large magnet cavity or four chambers that are connected in parallel, or four times scaled up chamber in volume with four magnet cavities (Fig.7). The separation chamber of Figs.5 or 7 may be used in the systems of Figs.1-3 described below. [0033] An external magnetic field, generally in the range of 0.1-1.7 Tesla, is applied to the separation chamber by either moving permanent magnets to the proximity or turning on an electromagnet surrounding the chamber or by other means. As required in the purification process, the controller controls the magnetic force applied to the separation chamber by turning it on or off or by reducing or increasing its strength. [0034] In one embodiment, separation efficiency and throughput of the process is increased by placing multiple separation chambers in parallel and/or in series. Multiple separation chambers increase processing flow rates significantly due to parallel processing. When the separation chambers are placed in series, they improve retention of particles significantly at higher flow rates. Placing the separation chamber in series has another potential advantage of using a lesser amount of chromatography magnetic particles for separation if the particles have much higher maximal binding capacity than their dynamic binding capacity. An excess amount of starting material is loaded in the first chamber with an amount of chromatography media that is calculated based on its maximal binding capacity. This allows some macromolecules to remain in the unbound state which will move to the second chamber in series. The second chamber   contains an amount of chromatography media that is calculated based on its dynamic binding capacity, so that almost all of the remaining unbound macromolecules will bind. This will lead to a larger amount of purified material per unit of chromatography media without any significant addition to the purification time. [0035] The complete disposable manifold including tubing, separation chamber (with or without steel wool), mixing bag, pH/conductivity flow cell, optical density and absorbance measurement flow cell, and filter is pre-assembled and is generally sterilized before use and is capable of withstanding multiple cycles in a batch. Tubing or pipes that have single-use product contact surfaces are used to circulate different fluids through the system. For example, flexible silicone, C-flex, or PVC tubing can be used to connect different parts of the system to channel fluids. A disposable manifold is connected to bags or vessels containing magnetic particle slurry, various buffers, starting material (e.g., bioreactor), and final purified material via connectors or welding of thermoplastic tubes. The connectors can be aseptic or non-aseptic. [0036] Product contact surfaces that come in contact with the material of interest include the inner surfaces of tubes, connectors, all flow-cells (e.g., pH/conductivity, OD etc.), pressure sensor, and the separation chamber. The product contact surfaces also include the outer surface of the particles. [0037] A controller with HMI (human machine interface) is used by the user to automatically or manually control and receive information from parts of the system, which include pumps (speed, direction, start/stop, feedback etc.), valves (open, close, feedback), occlusion/pressure sensors, optical density and absorbance detector, bubble sensors, in-line pH/conductivity meter, temperature sensor, and magnetic-field control system. This information is provided as analog or digital signals to and from the controller. Commercial automation systems from various vendors such as Siemens, Backhoff, or Allen-Bradley are available as complete solutions that include controller, input/output digital or analog cards, and HMI. Examples are Siemens SIMATIC S7- 1200 kit, Beckhoff TwinCAT 3 automation platform, and Delta V. [0038] The user inputs various process parameters into a process screen that is part of the HMI. Process parameters include cycle volume, incubation, mixing, wash, and recirculation times, pH, conductivity, and optical density and absorbance values to start or stop a step. Once the user starts the process by providing input to the HMI, the controller can follow the process screen and automatically run the process using entered process parameters. Multiple valves are controlled   (Fig.1; V1-V10) to form different fluid paths during different phases of the purification process. In one embodiment, pinch valves are used to open and close the fluid path within a flexible tube. [0039] The present invention now is described more fully hereinafter with reference to the accompanying drawings, in which some embodiments of the invention are shown. This invention may, however, be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art. [0040] Like numbers refer to like elements throughout. In the figures, the thickness of certain lines, layers, components, elements or features may be exaggerated for clarity. [0041] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “includes”, “include”, “including”, “comprises”, and/or “comprising,” when used in this specification, specify the presence of stated features, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, steps, operations, elements, components, and/or groups thereof. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items. [0042] Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skills in the art to which this invention belongs. It will be further understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the specification and relevant art and should not be interpreted in an idealized or overly formal sense unless expressly so defined herein. Well-known functions or constructions may not be described in detail for brevity and/or clarity. [0043] It will be understood that when an element is referred to as being “on”, “attached” to, “connected” to, “coupled” with, “contacting”, etc., another element, it can be directly on, attached to, connected to, coupled with or contacting the other element or intervening elements may also be present. In contrast, when an element is referred to as being, for example, “directly on”, “directly attached” to, “directly connected” to, “directly coupled” with or “directly   contacting” another element, there are no intervening elements present. It will also be appreciated by those of skill in the art that references to a structure or feature that is disposed “adjacent” another feature may have portions that overlap or underlie the adjacent feature. [0044] Fluids can be liquid or air. Embodiments with liquids may also be applicable to air. [0045] Having described the present invention, the same will be explained in greater detail in the following examples, which are included herein for illustration purposes only, and which are not intended to be limiting to the invention. [0046] The scheme shown in Fig.1 is an example to introduce various fluids at different process steps during the purification process. Two examples of automated methods to purify biotherapeutics and an example of an automated method to purify from bodily fluids are described below. In the affinity purification method, the material of interest binds to the particles while in the flow-through purification method, the material of interest flows-through and the material bound to the particles is generally discarded. Example 1: Affinity purification method [0047] Various macromolecules or particles can be purified from a mixture using the affinity purification method. The magnetic particles can successfully separate viruses, viral vectors (e.g., lentivirus, AAV, poxvirus). For this process, magnetic particles coupled with negatively charged matrix or molecules (e.g., quaternary or tertiary ammonium, primary amines, polyamines, poly- lysine, poly-arginine) or specific affinity ligands (e.g., proteins, antibodies, antibody fragments, single chain antibodies) are used to isolate viruses or viral vectors from the cell culture or other starting material. This process does not require removal of cells from the culture prior to purification. Protein A coupled magnetic particles were used to purify a monoclonal antibody (mAb) from CHO cell cultures. The results show 100% recovery of mAb with similar profile as a commercially purified human antibody on SDS-PAGE. [0048] Affinity purification method is also used to purify nucleic acids. In one embodiment, mRNA with Poly A tail is purified from the reaction mixture or crude extracts using magnetic particles that are coupled with oligo dT. Specific mRNAs can be purified by magnetic particles that are coupled with either complementary sequence of the mRNA or having 1 or more complementary nucleotides along with oligo dT. This method can be used to purify mRNA for vaccines in a single step.   [0049] In Fig.1, V1-V10 are the valves, P1 indicates a bi-directional pump, F/R indicates forward/reverse directions of the pump, OD/Abs indicates Optical Density/absorbance measurement or sensor(s), pH/cond indicates pH and conductivity sensor, BS1-4 indicates bubble sensors, N or S indicates poles of a magnet M, and B1-B2 indicates bags or containers. [0050] Different steps for the affinity purification process are detailed below. The pump is turned off and all valves are closed at the end of each step. All flexible bags or containers containing different liquids are connected via paths such as tubing as per Fig.1. A bag or container holding the magnetic particles is attached to the tube connected to valves V10. Starting material is not connected to the system at this stage. Step 1. Loading of magnetic particles: [0051] After the disposable has been loaded into the system, valves V1 and V10 are opened while the remaining valves remain closed. Pump P1 is turned on to move magnetic particles in liquid suspension from the container or flexible bag B1 to the separation chamber. Once the bubble sensor BS2 detects air (due to emptying of container B1), the pump P1 is stopped after the remaining material in the tubing between the container B1 and the pump P1 has entered the separation chamber. Thereafter valve V3 is opened, valves V1 and V10 are closed and pump P1 is turned on to recirculate material in the separation chamber. At the same time, the magnetic force is turned on to capture magnetic particles as they recirculate through the loop. The capture of the magnetic particles by the separation chamber can be monitored by measuring optical density and absorbance that is specific for the particles, by the OD sensor (e.g., at 370 nm). The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the magnetic particles are captured, the pump P1 is reversed, the valve V3 is closed, and the valves V1 and V10 are opened to fill the container B1 with liquid in which magnetic particles were suspended. The bag B1 can be manually or mechanically shaken to suspend magnetic particles that may be left in the bag. Once the bubble sensor BS2 detects bubbles, the pump P1 direction is switched to forward to move the liquid back into the separation chamber. Once the bubble sensor BS2 detects bubbles, the valve V10 is closed and the valve V3 is opened to recirculate again in the loop until optical density or absorbance drops substantially. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. At this stage, the supernatant is   discarded by reversing the pump P1 direction and opening valve V9 and closing valve V3. The pump P1 is stopped after bubble sensor BS3 detects air. Step 2. Washing of magnetic particles with magnetic field: [0052] After valve V9 is closed, the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until the bubble sensor BS1 detects liquid. Equilibration buffer is used to wash the particles and to reduce non-specific binding to the particles. Phosphate Buffered Saline (PBS) and Tris based buffers are examples of an equilibration buffer. At that stage, valve V3 is opened and valve V5 is closed. Washing progress can be assessed by monitoring data from inline pH/conductivity sensors pH/cond. Generally, after less than a minute of washing the magnetic particles in this manner, pump P1 direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. This process of washing can be repeated 1-2 times. Cleaning / Regeneration step: [0053] Whenever magnetic particles require regeneration, sanitization, or robust cleaning, sanitization or cleaning buffer connected to valve V8 can be used. The magnetic field is turned off and the equilibration buffer is introduced by opening valves V8 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V8 is closed. After recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump can be reduced to allow better capture of magnetic particles. Once the optical density is reduced substantially, the pump P1 direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow emptying of the remaining liquid in the loop. At last, the pump P1 is stopped, and all valves are closed. The regeneration step is generally followed by Step 5 or Step 2 to remove traces of regeneration buffer from the system. Step 3. Binding of magnetic particles to macromolecules:   [0054] The bag B1 is removed and the starting material source is attached to the tube connected to valve V10. The starting material source could be a bioreactor or a bag or vessel containing the starting material. Starting material is introduced into the system by opening valves V1 and V10, turning off the magnetic field, and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At this stage, valve V1 is closed, valve V2 is opened, and the pump P1 continues to run in the forward direction for a set time until the total volume of starting material loaded into the system is equal to the specified cycle volume. If the mixing tank B2 is a rigid container, an air vent with 0.2 micron filter is attached to the top of the container to displace the air by liquid coming in. Once the set amount of cycle volume has been pumped in, valve V4 is opened and valves V10 and V1 are closed to mix magnetic beads with starting material. The flow direction can be switched back and forth to improve mixing. Once the incubation time required for the binding of macromolecules to the beads is complete, the magnetic field is turned on to capture the magnetic beads in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Most of the beads can be captured by this recirculation process and capture efficiency can be measured by optical density sensor OD. The pump direction is then reversed to capture the remaining magnetic particles, and valve V4 is closed and valve V9 is opened to discard the macromolecule- depleted starting material to waste. When bubble sensor BS3 detects air, valve V2 is closed and valves V3 and V4 are opened for only a couple of seconds to allow emptying of the remaining liquid in the loop. Step 4. Washing of magnetic particles with magnetic field: [0055] Step 2 from above is followed. Step 5. Washing of magnetic particles without magnetic field: [0056] Valve V9 is closed, the magnetic field is turned off, and the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. After recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the OD370 or an optical density that specifically   detects the particles is reduced substantially, the pump P1 direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. This process of washing is repeated 1-2 times. Step 6. Elution: [0057] Step 6.1: Valve V9 is closed, valves V6 and V1 are opened, and the pump P1 is operated for a short time in forward direction to fill the tube up to the pump P1. Then valve V6 is closed, valve V9 is opened, and the pump P1 is reversed. This step cleans tubes with the elution buffer. [0058] Step 6.2: To fill the separation chamber with the elution buffer, the magnetic field is turned off and the elution buffer is introduced by opening valve V6 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V6 is closed. After recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the OD to detect magnetic particles is reduced substantially and OD to detect eluted material (e.g., OD at 280 nm for proteins) is maximal, valve V7 is opened, valve V3 is closed, and the pump direction is reversed to run until bubble sensor BS4 detects air. To empty the remaining liquid in the loop, valve V3 is opened momentarily while the pump P1 is running and then valves V3 and V7 are closed. This elution process is repeated from step 6.2 for 1-2 times to collect eluted molecules. When this process is repeated, the step to clean the tubes with the elution buffer (Step 6.1) is not required. [0059] After step 6, steps 2-6 are repeated until the end of the bioreactor is detected by air in the BS2 sensor. At that point the last cycle is completed by running steps 2-6 and the disposable assembly can be discarded. Example 2: Flow-through purification method [0060] In Fig.1, V1-V10 are the valves, P1 indicates a bi-directional pump, F/R indicates forward/reverse directions of the pump, OD/Abs indicates Optical Density/absorbance   measurement or sensor(s), pH/cond indicates pH and conductivity sensor, BS1-4 indicates bubble sensors, N or S indicates poles of a magnet M, and B1-B2 indicates bags or containers. [0061] Different steps for the flow-through purification process are detailed below. The pump and all valves are closed at the end of each step. All flexible bags or containers containing different liquids are connected via paths such as tubing as per Fig.1. A bag or container holding the magnetic particles is attached to the tube connected to valve V10. Starting material is not connected to the system at this stage. Step 1. Loading of magnetic particles: [0062] After the disposable has been loaded into the system, valves V1 and V10 are opened while the remaining valves remain closed. The pump P1 is turned on to move magnetic particles in liquid suspension from the container or flexible bag B1 to the separation chamber. Once the bubble sensor BS2 detects air (due to emptying of the container B1), the pump P1 is stopped after the remaining material in the tubing between the container B1 and the pump P1 has entered the separation chamber. Thereafter valve V3 is opened, valves V1 and V10 are closed and pump P1 is turned on to recirculate material in the separation chamber. At the same time, the magnetic force is turned on to capture magnetic particles as they recirculate through the loop. The capture of the magnetic particles by the separation chamber can be monitored by measuring optical density and absorbance that is specific for the particles, by the OD sensor (e.g., at 370 nm). Once the magnetic particles are captured, the pump P1 is reversed, valve V3 is closed, and valves V1 and V10 are opened to fill container B1 with liquid in which magnetic particles were suspended. The bag B1 can be manually shaken to suspend magnetic particles that may be left in the bag B1. Once the bubble sensor BS2 detects bubbles, the pump P1 direction is switched to forward direction to move the liquid back into the separation chamber. Once the bubble sensor BS2 detects bubbles, valve V10 is closed and valve V3 is opened to recirculate again in the loop until optical density or absorbance drops substantially. At this stage, the supernatant is discarded by reversing the pump direction and opening valve V9 and closing valve V3. The pump P1 is stopped after bubble sensor BS3 detects air. Step 2. Washing of magnetic particles with magnetic field:   [0063] After valve V9 is closed, the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. Washing progress can be assessed by monitoring data from inline pH/conductivity sensors pH/cond. After less than a minute of washing the magnetic particles in this manner, the pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. This process of washing can be repeated 1-2 times. Cleaning / Regeneration step: [0064] Whenever magnetic particles require regeneration, sanitization, or robust cleaning, sanitizing or cleaning buffer connected to valve V8 can be used. The magnetic field is turned off and the equilibration buffer is introduced by opening valves V8 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V8 is closed. After recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the optical density specific to the particles is reduced substantially, the pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. At last, the pump P1 is stopped, and all valves are closed. The regeneration step is generally followed by Step 6 or Step 2 to remove regeneration buffer traces from the system. Step 3. Binding of magnetic particles to impurities: [0065] Starting material is introduced into the system by opening valve V10, turning off the magnetic field, and running the pump P1 in the forward direction for a set time while valve V2 is opened. If the mixing tank B2 is a rigid container, an air vent with a 0.2 micron filter is attached to the top of the container to displace the air by liquid coming in. Once the set amount of liquid equal to the cycle volume has been pumped in, valve V4 is opened and valve V10 is closed to mix magnetic beads with starting material. Once the incubation time required for the binding of impurities to the beads is complete, the magnetic field is turned on to capture the magnetic beads   in the separation chamber. Lowering the flow rate of the pump P1 allows better capture of magnetic particles. The majority of the beads can be captured by this recirculation process. The pump direction is then reversed, valve V4 closed and valve V7 is opened to capture the remaining magnetic particles and to collect the purified material. When bubble sensor BS4 detects air, valve V2 is closed and valve V3 is opened momentarily to allow emptying of remaining liquid in the loop. Step 4. Washing of magnetic particles with magnetic field: [0066] Washing with the equilibrium buffer can be used to improve yield by recovering the leftover starting material from the fluid path. This step can be eliminated to shorten the total processing time. The equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. After less than a minute of washing the magnetic particles in this manner, the pump direction is reversed, valve V7 is opened, and valve V3 is closed. When bubble sensor BS4 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. Step 5. Cleaning / Regeneration: [0067] Magnetic particles are regenerated to remove the bound impurities by washing with regeneration, sanitization, or cleaning buffer that is connected to valve V8. The magnetic field is turned off and the regeneration buffer is introduced by opening valves V8 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V8 is closed. After the recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the optical density specific to the particles (e.g., OD370) is reduced substantially, pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily to allow the emptying of the remaining liquid in the loop. Step 6. Washing of magnetic particles without magnetic field:   [0068] The magnetic field is turned off and the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. After recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the optical density specific to the particles (e.g., OD370) is reduced substantially, pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily and closed to allow the emptying of the remaining liquid in the loop. At last, the pump P1 is stopped, and all valves are closed. Washing progress can be assessed by monitoring data from inline pH/conductivity sensors pH/cond. This process of washing is repeated 1-2 times. [0069] After step 6, steps 3-6 are repeated until the end of bioreactor is detected by air in the bubble sensor BS2. At that point the last cycle is completed by running steps 3-6 and the disposable assembly can be discarded. Example 3: Method to purify biomaterials from bodily fluids [0070] The device and method of using the device described in this invention are not only useful for biomanufacturing but also enable the purification of biomaterials from crude bodily liquids of animals or humans. In the device and method described here, biomaterials of interest can be purified at the donation site and the remaining material can be returned to the donor after capturing the biomaterial of interest. With the return of biomaterial-depleted bodily fluid, this process can be repeated multiple times to purify much larger amounts of biomaterials of interest in a single donation while eliminating the risk of hypovolemic shock for the donor. Also, this significantly reduces processing, shipping, storage, and logistics costs over the existing methods. Another advantage of the method is that blood and other bodily fluids can directly be used without requiring any pre-processing (e.g., removal of cells etc.). [0071] Multiple cycles of blood collection allow purification of significantly higher amounts of immunoglobulins that can treat multiple recipients without causing hypovolemic shock to the donor. Another advantage of this new method of purification is that recipients of the purified material have a significantly lower risk of serum sickness as compared to the convalescent serum and removes blood type matching. This device and process have the potential to save millions of   human lives in an epidemic or a pandemic. This process can be used to efficiently manage current and emerging strains of SARS-CoV-2 as antibodies purified from donations will be against the most recent propagating strains of the virus. These purified antibodies can then be injected in a short timeframe to the recipients who are exposed to the same strain of the virus. The isolated immunoglobulins from convalescent sera can be screened against pathogens to identify neutralizing antibodies and this information can be used in developing recombinant antibodies and vaccines. [0072] In several disorders, harmful or toxic biomaterials accumulate in patient’s blood. This method can be used to target those harmful or toxic biomaterials and reduce their circulating concentration. For example, in rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, plaque psoriasis, Crohn’s disease, and ulcerative colitis, the tumor necrosis factor (TNF) concentration is elevated. Several antibody-drug treatments are available that target TNF but after repeated dosing, most patients develop neutralizing antibodies against those drugs requiring them to switch to a different drug. This method can also be used to reduce circulating TNF by specifically capturing and removing TNF from blood to ameliorate the condition. Since no external drug is infused into the patients, patients do not suffer from the drug side effects and this method of treatment can effectively be used lifelong by them. This process is similar to the flow- through purification process. [0073] Similar to the effects of repeated dosing of anti-TNF antibody therapeutics, repeated dosing of other protein therapeutics also leads to the generation of neutralizing antibodies against the administered protein therapeutic that reduces their effectiveness over time. In contrast, this method of treatment can effectively be used lifelong by patients. [0074] In another embodiment, this method is used to remove excess drugs from blood to reduce their side effects. For example, a high dose of a drug for the treatment of cancer or other indications is needed to have a maximal effect. After a drug has specifically bound to the target, any free drug may remain circulating in the blood, causing side effects. This technology can be used to specifically remove unbound or free drugs remaining in the blood to reduce its side- effects. [0075] The device uses a single-use or disposable kit and particles with specific properties. All product contact surfaces in a single-use kit are suitable for the medical device (e.g., meet USP class VI requirements) and can be replaced after each donation but can support multiple cycles of   processing within the same donation. For donations from animals, the disposable kit can potentially be re-used for more than one donation after a wash cycle with sanitization and equilibration buffer to sanitize and wash single-use surfaces that come in contact with bodily fluids. The method described here uses particles that are attracted to magnets (e.g., ferromagnetic, paramagnetic, or superparamagnetic), have large surface area due to their smaller size, are preferably non-porous, and have specific binding properties. These particles exhibit very short residence time for binding as most of the binding sites are on the surface. The size of these particles is from 10-10,000 nm, which is up to 20,000 times smaller than the standard porous chromatography resin beads. The surface of these particles is modified with ligands that have specific binding properties (e.g., surface attachment of protein A for antibody purification, or attachment of antibodies that have an affinity for a specific biomaterial, or attachment of positive or negative charged ligands, or attachment of hydrophobic ligands, or ligands with mixed properties). [0076] The bodily fluids containing target biomaterial are drawn either directly or indirectly in a bag containing these particles that have an affinity for the targeted biomaterial and are mixed. Other buffers or reagents can be added to the collection bag for conditioning. For example, citrate or heparin can be added to the collection bag to manage blood clotting. An external magnetic field is applied to recover these particles from bodily fluids. If the magnetic susceptibility is low due to the smaller size of magnetic particles, a high gradient magnetic field is used to isolate particles mixed in with bodily fluids. Small-scale batch chromatography methods to purify proteins (in microgram scale) using magnetic beads are currently available but those mostly use porous magnetic beads that are not recycled and are operated manually. Commercially available magnetic particles are spherical with a diameter that is equal to or greater than 1 micron and used only for a single cycle at a very small scale (e.g., Sera-Mag from GE Healthcare Life Sciences and Dynabeads from ThermoFisher Scientific). These particles are not well suited for the current invention due to their porosity, large size, and recycling capabilities. [0077] As narrow pore channels lead to poor mass transfer of macromolecules, porous affinity chromatography media has slow binding kinetics resulting in long residence time. The inventors’ results show that even with high viscosity whole human blood, the nonporous magnetic particles   have very fast binding kinetics with most of the immunoglobulins binding in 30 seconds (Fig. 6A). [0078] The magnetic particles described in this invention have a high surface area to volumetric ratio. For example, the surface area to the volumetric ratio of tetrahedron and cubic particles is at least 25% greater than the ratio of spherical particles. The attachment of a ligand to the surface of magnetic particles is achieved by a linker that can bind to the ligand and the inorganic surface of magnetic particles. In one embodiment, linkers with silane groups are used to chemically bond with magnetic particles. The other end of the linker is chosen such that it can directly or indirectly bind to the ligand that is used for purification. For example, the epoxide group at the other end of the linker can react with primary amine, sulfhydryl, or carboxyl groups that are commonly found on different amino acids of ligand proteins.3-glycidoxypropyl trimethoxysilane or 3-glycidoxypropyl triethoxysilane are two examples of such linkers. In the inventors’ studies, 0.5-3% w/w treatment with 3-glycidoxypropyl trimethoxysilane provided excellent conjugation to Fe3O4 cubic particles of 300 nm average size. [0079] When the donor’s bodily fluids are mixed in with magnetic particles that have a surface affinity for biomaterials of interest, those biomaterials bind to the magnetic particles. Magnetic particles are captured in a separation chamber by applying an external magnetic force. Flow- through material is collected and can be infused into the donor. The captured magnetic particles can then be washed multiple times to remove impurities and then elution is performed by introducing an elution buffer that dissociates the binding of the macromolecules of interest from the magnetic particles. In one embodiment, low pH buffer (generally below pH 4.0) is used to dissociate antibodies or immunoglobulins that are captured by protein A-coupled magnetic particles. In another embodiment, low or high pH buffer (below 6.5 or higher than 8.4) is used to dissociate macromolecules bound to antibody-coupled magnetic particles. Similarly, change in pH and/or salt concentration can be used to elute factor VIII bound to quaternary ammonium particles with the anion-exchange property. If specific cells are the targeted material, the elution buffer may contain peptides or molecules that compete with interaction (between affinity magnetic particles and receptor or molecule on cell’s surface) or enzymes that destabilize the interaction. The eluted biomaterials flow through and are collected or discarded (depending on the application) while the magnetic particles are retained due to the external magnetic field.   [0080] During affinity purification, the bound particles are washed either in the presence or absence of a magnetic force to remove any nonspecifically bound impurities. The bound macromolecules are eluted from the magnetic particles by introducing an elution buffer to the separation chamber in the presence or absence of an external magnetic force. The whole cycle can be repeated multiple times until the desired amount of target biomaterial is purified or removed. [0081] If the affinity performance of particles reduces after a certain number of cycles, due to nonspecific occlusion of specific binding sites by impurities, the particles can be regenerated using regeneration solutions (e.g., solution with NaOH or chaotropic agents such as urea, guanidine, etc.). [0082] In each cycle, a specific volume of bodily fluid is collected in a bag that is attached to the system. This volume is based on guidelines for the amount of bodily fluid that can be drawn from a subject at a time. This is called the cycle volume. For example, the typical blood donation range for humans is 250-500 mL. If multiple cycles are to be performed, 250 mL of blood can be drawn per cycle (cycle volume) so that as soon as the bodily fluid donation bag is empty, another 250 mL of blood draw is initiated. An overview of the process to purify immunoglobulins from human blood is shown in Fig.2. [0083] Magnetic beads with affinity against target molecule and conditioning buffer or reagents (e.g., anti-clotting agent) can either be pre-loaded in the bag, or added later via another bag that can be aseptically attached, or aseptically injected via a syringe. The mixture in the bag is mixed by recirculation, impeller, or other mechanical means. The magnetic beads are recycled and their replenishment after the first cycle is generally not required. However, an anti-clotting agent or another appropriate buffer may need to be injected before every cycle of bodily fluid collection. [0084] A cylindrical separation chamber with a conical bottom (with an apex at the bottom) is used to isolate magnetic particles during different parts of the process. In another embodiment, the top of the separation chamber is conical (with an apex at the top). The separation chamber is fabricated out of materials that have poor attraction for magnetic force (e.g., LDPE, high durometer silicone, polypropylene, polystyrene, EVA, PVC, etc.). The chamber may contain steel wool or wire mesh to enhance the magnetic field via high gradient magnetic field generation and capture magnetic particles that are small and have weak magnetic susceptibility.   In one embodiment, the material of steel wool or wire mesh (1 micron-1000 micron) is ferromagnetic stainless-steel (e.g., 430 or 410 stainless steel) to provide corrosion resistance. The ferromagnetic steel wool can also be coated with metals (e.g., nickel or chromium) or plastics to substantially reduce or eliminate corrosion. [0085] The external magnetic field, generally in the range of 0.1-1.7 Tesla, is applied to the separation chamber by either moving permanent magnets to the proximity or turning on an electromagnet surrounding the chamber or by other means. As required in the purification process, the controller controls the magnetic force applied to the separation chamber by turning it on or off or by reducing or increasing its strength. [0086] The complete disposable manifold comprising tubing, connectors, separation chamber (with or without steel wool), mixing bag, pH/conductivity flow cell, optical density measurement flow cell, pressure sensors, absorbance measurement flow cell, and the filter is pre-assembled and pre-sterilized prior to use. The complete assembly is designed to withstand multiple cycles in a batch. Tubing or pipes that have single-use product contact surfaces are used to circulate different fluids through the system. For example, flexible silicone, C-flex, or PVC tubing is used to connect different parts of the system to direct fluids. A disposable manifold is connected to bags or vessels containing magnetic particles, different buffers, donated bodily fluids, and final purified material via aseptic connectors or welding of a thermoplastic tube. [0087] A controller with HMI (human machine interface) is used by the user to automatically or manually control and receive information from parts of the system, which comprise pumps (speed, direction, start/stop, feedback etc.), valves (open, close, feedback), occlusion/pressure sensors, optical density and absorbance detector, bubble sensors, in-line pH/conductivity meter, temperature sensor, weight load cells, and magnetic-field control system (Fig.2). This information is provided as analog or digital signals to and from the controller. Commercial automation systems from various vendors such as Siemens, Beckhoff, or Allen-Bradley are available as complete solutions that include controller, input/output digital or analog cards, and HMI. Examples are Siemens SIMATIC S7-1200 kit, Beckhoff TwinCAT 3, and Delta V automation platforms. During the process, process data can be electronically stored or exported out (to an external data management system) by these platforms to meet CFR Part 11 compliance.   [0088] The user inputs various process parameters into a process screen that is part of the HMI. This can be turned into a recipe that is executed each time a process is run. Process parameters include cycle volume, incubation, mixing, wash, and recirculation times, pH, conductivity, and optical density and absorbance values to start or stop a step. Once the user starts the process by providing input to the HMI, the controller can follow the process screen and automatically run the process using entered process parameters. Multiple valves are controlled (Fig.1; V1-V13) to form different fluid paths during different phases of the purification process. In one embodiment, pinch valves are used to open and close the fluid path within a flexible tube. The scheme shown in Fig.2 is an example to introduce various fluids at different process steps during the purification process. An automated method to process bodily fluids is described below. Process steps [0089] In Fig.3, V1-V13 are the valves, P1 indicates a bi-directional pump, F/R indicates forward/reverse directions of the pump, OD/Abs indicates Optical Density/absorbance measurement, pH/cond indicates pH and conductivity sensor(s), BS1-2 indicates bubble sensors, M indicates a magnet, N or S indicates poles of a magnet, and B1-B2 indicates bags or containers. [0090] The pump is turned off and all valves are closed at the end of each step. All flexible bags or containers containing different liquids are connected as per Fig.3. A set amount of bodily fluid (cycle volume) is collected into the bodily fluid donation bag (e.g., 250 mL of blood). Sterile magnetic particles and appropriate modifiers (e.g., citrate or heparin for blood) are either injected directly in the bodily fluid donation bag or transferred through a bag containing magnetic particles. The donation and return bags are placed or hung on load cells so that accurate weight can be measured. The real-time weight information is continuously communicated to the controller. The bodily fluids start flowing into the bodily fluid donation bag once the source is connected and valve V12 is opened. For example, a needle for an IV tube is inserted in one arm of the donor for blood collection and the donation bag is hung at a lower height than the body. Once the cycle volume (corresponding to weight detected by the load cell) is collected, valve V12 is closed and Step 1 is initiated. Step 1. Binding of magnetic particles to targeted macromolecules:   [0091] Bodily fluids are introduced into the remaining system by opening valves V1 and V10, turning off the magnetic field, and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At this stage, valve V1 is closed, valve V2 is opened, and the pump P1 continues to run in the forward direction until bubble sensor BS2 senses air as the bodily fluid donation bag runs empty. If the mixing bag B2 is a rigid container (instead of a flexible bag), an air vent with a 0.2 micron filter is attached to the top of the container to displace the air by liquid coming in. Once the bubble sensor BS2 detects air, valve V4 is opened and valve V10 is closed to mix magnetic beads with starting material. [0092] If another cycle of bodily fluid donation is required, a set amount of anticoagulant or other required buffers is injected in the donation bag at this time and then valve V12 is opened to allow another collection of bodily fluid for the second cycle. Once the cycle volume (corresponding to weight detected by the load cell) is collected, valve V12 is closed. [0093] The flow direction of the pump P1 can be changed multiple times to improve mixing. Once the incubation time required for the binding of macromolecules to the beads is complete (typically 0.5-10 minutes), a magnetic field is turned on to capture the magnetic beads in the separation chamber. The flow rate of the pump P1 is reduced to allow better capture of magnetic particles. The majority of the beads can be captured by this recirculation process and capture efficiency can be monitored by an optical density/absorbance sensor (e.g., monitoring OD370 for Fe 3 O 4 particles). The pump direction is then reversed, valve V4 is closed, and valve V11 is opened to capture the remaining magnetic particles and to collect the bodily fluids (without the macromolecule of interest) in the return bag. When bubble sensor BS2 detects air, valves V3 and V4 are opened and closed once to remove remnant liquid in the loop. Thereafter, valves V2 and V11 are closed, and the pump P1 is stopped. After this step, valve V13 is opened to return the bodily fluid (without the target macromolecule) to the donor. To ensure that no air can go into the return tube to the donor, valve V13 closes immediately once a set weight of liquid (generally 10% below the cycle volume) is infused back into the patient. To avoid hypovolemic shock, the controller is programmed such that at least 50% of the collected volume is infused back to the donor before collecting bodily fluids for the second cycle. Step 2. Washing of magnetic particles with the magnetic field:   [0094] In this step, the magnetic field stays on and the magnetic particles are washed with an equilibration buffer to remove impurities. The equilibration buffer is chosen such that it does not disrupt the binding between the targeted macromolecule and magnetic particle (e.g., PBS or PlasmaLyte solution). After valve V11 is closed, the equilibration buffer is introduced into the system by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. Washing progress can be assessed by monitoring data from inline pH/conductivity sensors pH/cond. After less than a minute of washing the magnetic particles in this manner, the pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS2 detects air, valve V3 is opened momentarily and then closed to allow emptying of remaining liquid in the loop. This process of washing can be repeated 1-2 times. Step 3. Washing of magnetic particles without the magnetic field: [0095] After valve V9 is closed, the magnetic field is turned off and the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed to generate mixing by recirculation for 0.5-2 minutes. Once the recirculation is complete, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the optical density specific to the particles (e.g., OD370) is reduced substantially, the pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS2 detects air, valve V3 is opened and closed to allow the emptying of the remaining liquid in the loop. This process of washing is repeated 1-2 times. Step 4. Elution: The elution buffer is used to dissociate targeted macromolecules from magnetic particles. For example, low pH buffer (100 mM sodium citrate buffer with pH<4) can be used to dissociate both antibody-antigen interactions as well as protein A-IgG interactions. [0096] Step 4.1: After valve V9 is closed, valves V6 and V1 are opened, and the pump P1 is operated momentarily in the forward direction to fill the tube up to the pump P1. Then valve V6 is closed, valve V9 is opened, and the pump direction is reversed. This step cleans tubes with the elution buffer.   [0097] Step 4.2: To fill the separation chamber with the elution buffer, the magnetic field is turned off and the elution buffer is introduced by opening valve V6 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V6 is closed. After the recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of magnetic particles. Once the OD to detect particles (e.g., OD370) is reduced substantially and the OD to detect eluted molecules (e.g., OD at 280 nm for protein-based macromolecules) is maximal, valve V7 is opened, valve V3 is closed, and pump direction is reversed to run until bubble sensor BS2 detects air. To empty the remaining liquid in the loop, valve V3 is opened and closed while the pump P1 is running and then valves V3 and V7 are closed. This elution process is repeated from step 4.2 for 1-2 times to collect eluted macromolecules or biomaterials. When this process is repeated, the step to clean the tubes with the elution buffer (Step 4.1) is not required. Step 5. Washing of magnetic particles without the magnetic field: [0098] At first, the magnetic field is turned off, the equilibration buffer is introduced by opening valves V5 and V1 and running the pump P1 in the forward direction until bubble sensor BS1 detects liquid. At that stage, valve V3 is opened and valve V5 is closed. After the recirculation of the magnetic particles to create mixing, the magnetic field is turned on to collect the magnetic particles in the separation chamber. The flow rate of the pump P1 can be reduced to allow better capture of the magnetic particles. Once the optical density specific to the particles (e.g., OD370) is reduced substantially, the pump direction is reversed, valve V9 is opened, and valve V3 is closed. When bubble sensor BS3 detects air, valve V3 is opened momentarily and closed to allow emptying of remaining liquid in the loop. At last, the pump P1 is stopped, and all valves are closed. Washing progress can be assessed by monitoring data from inline pH/conductivity sensors pH/cond. This process of washing is repeated 1-2 times. [0099] After step 5, steps 1-4 are repeated until sufficient targeted macromolecules have been purified. At that point, the last cycle is completed, and the disposable assembly can be discarded. [0100] If the purified material is not usable (to remove autoantibodies or overproduction of cytokines, etc.), it is discarded. Otherwise, the purified material is saved. If immunoglobulins are the product, the purified immunoglobulins are kept at low pH for viral inactivation and then a   set amount of neutralizing buffer is added (via a syringe or aseptically connected external bag) to increase the pH to physiological range (about pH 6-8) and make the final solution isotonic. The purified macromolecules can be exposed to UV or are nanofiltered to remove or reduce contaminating virus particles. Depending on the amount of the collected macromolecules, the final product can be aliquoted into several bags or vials and stored at a lower temperature. An appropriate dosage can be administered to patients in need. The purified macromolecules can also be lyophilized to increase their shelf life and to reduce transportation and logistics costs. [0101] In addition to the applications in medical and biomanufacturing areas, there are many other industrial applications of this platform. For example, in the food and beverage industry this platform can be used to remove unwanted impurities to reduce toxicity or to enhance flavors. The device and method can be used to extract specific components that may have value. This method can also isolate components that are at very low concentration in a mixture. [0102] The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein.