WO2014004103A1 - Methods for inactivating viruses during a protein purification process - Google Patents

Methods for inactivating viruses during a protein purification process Download PDF

Info

Publication number
WO2014004103A1
WO2014004103A1 PCT/US2013/045677 US2013045677W WO2014004103A1 WO 2014004103 A1 WO2014004103 A1 WO 2014004103A1 US 2013045677 W US2013045677 W US 2013045677W WO 2014004103 A1 WO2014004103 A1 WO 2014004103A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
protein
sample
chromatography
eluate
Prior art date
Application number
PCT/US2013/045677
Other languages
French (fr)
Inventor
Alex Xenopoulos
Original Assignee
Emd Millipore Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=49783749&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014004103(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Emd Millipore Corporation filed Critical Emd Millipore Corporation
Priority to EP13810248.8A priority Critical patent/EP2867359B1/en
Priority to EP21191783.6A priority patent/EP3981873A1/en
Priority to US14/390,924 priority patent/US9809799B2/en
Priority to SG11201406407TA priority patent/SG11201406407TA/en
Priority to ES13810248T priority patent/ES2895173T3/en
Priority to CN201380034780.5A priority patent/CN104411820B/en
Priority to JP2015520259A priority patent/JP6182210B2/en
Publication of WO2014004103A1 publication Critical patent/WO2014004103A1/en
Priority to IN8526DEN2014 priority patent/IN2014DN08526A/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/36Selective adsorption, e.g. chromatography characterised by the separation mechanism involving ionic interaction
    • B01D15/361Ion-exchange
    • B01D15/362Cation-exchange
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/36Selective adsorption, e.g. chromatography characterised by the separation mechanism involving ionic interaction
    • B01D15/361Ion-exchange
    • B01D15/363Anion-exchange
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/38Selective adsorption, e.g. chromatography characterised by the separation mechanism involving specific interaction not covered by one or more of groups B01D15/265 - B01D15/36
    • B01D15/3804Affinity chromatography
    • B01D15/3809Affinity chromatography of the antigen-antibody type, e.g. protein A, G, L chromatography
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F23/00Mixing according to the phases to be mixed, e.g. dispersing or emulsifying
    • B01F23/40Mixing liquids with liquids; Emulsifying
    • B01F23/49Mixing systems, i.e. flow charts or diagrams
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F25/00Flow mixers; Mixers for falling materials, e.g. solid particles
    • B01F25/40Static mixers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F33/00Other mixers; Mixing plants; Combinations of mixers
    • B01F33/80Mixing plants; Combinations of mixers
    • B01F33/81Combinations of similar mixers, e.g. with rotary stirring devices in two or more receptacles
    • B01F33/811Combinations of similar mixers, e.g. with rotary stirring devices in two or more receptacles in two or more consecutive, i.e. successive, mixing receptacles or being consecutively arranged
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F35/00Accessories for mixers; Auxiliary operations or auxiliary devices; Parts or details of general application
    • B01F35/20Measuring; Control or regulation
    • B01F35/21Measuring
    • B01F35/2132Concentration, pH, pOH, p(ION) or oxygen-demand
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F35/00Accessories for mixers; Auxiliary operations or auxiliary devices; Parts or details of general application
    • B01F35/71Feed mechanisms
    • B01F35/715Feeding the components in several steps, e.g. successive steps
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/10061Methods of inactivation or attenuation

Definitions

  • the present invention provides in-line methods tor inactivating viruses during a protein purification process.
  • proteins are produced, by cell coliwre, using either mammalian or bacterial eel! lines engineered to produce the protein of interest such -as a monoclonal antibody.
  • the proteins have to be separated from various impurities such as, host cell proteins (H €Ps) ? endotoxins:, viruses, ON ' A e c.
  • a protein of interest in cell culture, is subjected to a clarification step for removal of cell debris.
  • the clarified cell culture feed containing the protein of interest is then subjected, to one or more chromatography steps, which may sue hide an affinity chromatography step or a catio exchange chromatography step, in order to ensure safety of a protein of interest, especially in case of a therapeutic candidate, it is necessary to inactivate any viruses which may be present in a sample containing the protein of Interest during the purification process.
  • vims maetivation is performed alter a chromatography step (e.g., after affinity chromatography or after cation exchange chromatography).
  • an elution pool containing the protein of Interest Is collected in a large tank or reservoir and subjected to a virus inaetivatlon step/process for an extended period of time with mixing, which may take several hours to a day or longer, in order to achieve complete inactivaiion of any viruses thai may be present in the el uion pool
  • virus inaetivatlon techniques are known in the art including, temperature * H, radiation and exposure to certain chemical agents.
  • the present invention provides methods of virus inaetivation during a protein purification process, which a e several advantages over the methods currently being used in the industry during protein purification. Specifically, the methods described herein obviate the need tor using large tanks or reservoirs for performing the virus inaciivation step during a protein purification process; reduce the overall time required for virus inaetivatlon as well as reduce the overall physical space required to run the virus inaetivatlon operation during a protein purification process, which in turn reduces the overall footprint for the whole purification process.
  • a method for Inactivating one or more viruses, in. a sample in a purification process comprises continuously mixing the sample with one or more virus inactivating agents as the sample flows iroro the first unit operation to the second unit operation.
  • the first unit operation comprises bind and eS.ute chromatography and the second unit operation comprises a flow-through
  • An exemplary bind and eiute chromatography unit operation includes, hut is not limited to, Protein A affinity chromatography,
  • the How-through puri Oeation process comprises two or more matrices selected from the group consisting of activated carbon, anion exchange chromatography media, cation exchange chromatography media and a virus filtration media.
  • the sample comprises a Protein A eluaie
  • target molecules comprising a target molecule.
  • target molecules include, e.g., antibodies.
  • the sample is mixed with one or more virus inactivating agents using one or more in-line static mixers. In other embodiments, the sample is mixed with one or more virus inactivating agents using one or more surge tanks. When static mixers are used, the flow of sample is in the laminar range.
  • one or more virus inactivaiiag a ents are selected from the group consisting of an acid, a salt, a solvent and a detergent
  • a method of inactivating one or more viruses in a Protein A eluate comprises mixing the eluate with one or mors virus inactivating agents using an in-line static mixer, wherein complete virus Inaetivation is achieved in less than 10 minutes or less than 5 minutes or less than 2 minutes or less than 1 minute,
  • a method of inactivating one or more viruses in a Protein A eluate comprises mixing the eiuate with one or more virus Inactivating agents using a surge tank, where complete virus inaetivation is achieved in less than one hour or less than 30 minutes,
  • a method for inactivating one or more viruses comprises: (a) subjecting a sample comprising a target protein (e.g., an antibody) to a Protein A affinity chromatography process, thereby to obtain an eluate; and (b) continuously transferring the eluate t an in-line static mixer to mix one or more virus inacti vating agents with the eluate for a duration of time which is equal to or less than 10 minutes, thereby to inactivate one or more viruses,
  • a target protein e.g., an antibody
  • the Protein A eluate which is subjected to virus inaetivation methods described herein is obtained following a Protein A affinity chromatography process performed in hatch mode.
  • the Protein A affinity chromatography process is performed in a continuous mode, in some embodiments, continuous mode comprises a continuous multi-column chromatography process .
  • one or more virus inactivating agents an acid, used to perform a solution change.
  • the eluate is continuously transferred to a How- through purification, proces step following virus inaetivation, in that the output from the static mixer flows directly into a flow through purification step, which may include use of two or more matrices selected from activated carbon, anion exchange media, cation exchange media and virus filtration media.
  • the eluate i stored following virus inaetivation for an extended period of time e.g., 12 to 24 hours or overnight
  • a flow- through purification, process step or a cation exchange hind and elute e.g., a flow- through purification, process step or a cation exchange hind and elute
  • the virus inactivaiion methods described herein are a part of a larger protein purification process, which may include several steps including, hoi not limited to, e. ., eultnrmg cells expressing protein in a bloreactot; subjecting the cell culture to clarification, which may employ one or more of precipitation, eentriiugation and/or depth filtration;
  • transferring the clarified cell culture io a bind and dute chromatography capture ste e.g.. Protein A affinity chromatography
  • subjecting the output from virus inaetivahon to a flow-through purification process, which employs two or more matrices selected fro activated carbon, anion exchange chromatography media, cation exchange chromatography media and vims filtration media: and formulating the protein in the flow-through from the flow-through purification step using diafiitration coiicentration and sterile filtration. Additional details of such processes can be found, e.g., in co-pending application having reference no, P t 2/ 107, filed concurrently herewith, and the entire contents of which are incorporated by reference herein.
  • a fluid sample continuously flows through the entire process, as described above, from one step to the next.
  • Fi gure I is a schematic depiction of an experimental set up for virus inactivaiion using two in-line static mixers.
  • the set up shown includes; (a) a peristaltic pump or sample feed, (b) two syringe pumps to deliver acid and base, c> two in-line pB probes, and (d) two static mixers.
  • the flow rates are predetermined in batch mode based on the amount of acid/base needed to achieve the desi red pH.
  • the residence time for virus inaetivation is altered by having tubes of appropriate diamete and length after each static mixer and before the pH probe.
  • Biophar aeeutieal maoufaetnrlng requires the mactfvation or removal of viruses (coming from, animal derived components., including mammalian. cells) for drug safety and to meet the standards set forth by the Pood and Drug Adoiioistration (FDA). Typical processes involve a s um er of viral clearance steps thai
  • Some processes used, in the industry involve titration of the solution containing the target protein to a low pH in order to cause destruction of any enveloped viruses and viral components.
  • the sample containing the target protein must be retained at these conditions for an extended period of time, both because time is needed tor virus inaetivation but also - and more importantly - to ensure homogeneous mixing for effective virus inaciivaHon. Therefore, in ease of large scale processes, the sample containing the target protein must be incubated for an extended period of time at a low pH in order to promote efficient virus inaetivstion often with mixing. See. e.g., Shukla et at, . Chromatography B., 848 (200?) 28-39, which describes virus inactivation using incubation of a protein sample for a suitable duration of time at lo w pi t,
  • the pH conditions are established as a balance between a low pH value that is sufficient to cause inactivation and a high enough value to avoid denaturation. of the target protein. Additionally * the sample most be exposed for a certain amount of time to cause a significant reduction, usually 2 to 6 L V in virus activity values (See, e.g., Miesegaes et al, "Analysis of viral clearance unit operations for monoclonal antibodies.” Biotechnology and Bioengineering, Vol. .106, p 238-246 (2010) ⁇ ,
  • the three parameters that are considered important for virus mactivation process are pH value, exposure time and temperature, assuming homogeneous mixing is present. In. ease of large-scale processes, mixing poses a. challenge due to large volumes and additional parameters, such as mix rate and mass transfer, also become important .
  • virus inaetivation is usually performed following elation from: a bind and eiute
  • the chromatography process step e.g., Protein A affinity chromatography or cation exchange chromatography
  • the pH of the elation pool is closer to the desirable pli for virus mactivation.
  • the Protein A chromatography elation pool typically has a pH in the 3.5 to 4.0 range and the cation exchange hind and elute chromatography elution pool typically has a pH about 5.0.
  • the elution pool containing the target protein is adjusted to the pH desired for virus inaetivation and held there for a certain length of time, the combination of pH and time having been shown to result in virus Inaetivation, Longer times are more effective tor virus inacfi vaiion, especially in case of a large-scale process, however, longer times are also known to cause protein damage. Extended exposure to low pH may result in precipitation and formation of aggregates, which i s undesirable and often requi res the use of a depth filter and/or a sterile filter to remove such precipitates and aggregates.
  • agitation in pool tanks can also cause aggregation.
  • Proper mixing is essential in order to homogenize protein pools, and is especially important during mamri3 ⁇ 4eturing when protein solutions need to be treated with acid/base or buffer to adjust phi and/or conductivity (See, e,g Stephen Vazquez-Rev et a!,, "Aggregates in monoclonal antibody manufacturing processes, " Biotechnology and Bioengineering, Vol 108, Issue 7, pages 1494-1 S 8 (201 1 )).
  • the present invention provides novel and improved methods for virus inaetivation (also referred to as i VI" herein) during a protein purification process, which reduces the overall time for virus inaetivation, the cost as well as the overall physical footprint associated with a protein purification process,
  • the methods described herein are able to achieve vims inaetivation in a continuous manner, which significantly reduces the time associated with virus inaetivation relative to most conventional processes, and in tarn, reduces the time for the overall purific tion process, [0032]
  • the methods according to the present invention emplo one or mote in-line static mixers for achieving viru inactivation.
  • the methods according to the present invention employs one or more surge tanks for achieving virus inaetivaiion.
  • the methods described herein facilitate the running of a whole purification process in a continuous manner, i.e., the sample containing the target protein can continuously flo from one process step (or unit operation) to the next process step (or unit operation), without the need to stop the flo w of the sample after a process step.
  • the elation pool from an upstream bind and elate chromatography process -step e.g., Protein A affinity chromatography or cation, exchange chromatography
  • the sample flows continuously into the next process step (e.g., a flow-through purification process step).
  • the eiotioo pool does no have to be mixed or Incubated with a virus inactivating agent for an extended period of time in a pool tank or a vessel before moving on to the next process step lu the purification process.
  • static mixers have been described fo mixing a sample as it is exposed to radiation for inactivating pathogens (see, e.g., U.S. Publication No, 2004013 1497 and PCT Publication No. W02O02O928O6), or mixing a blood sample with a viru inactivation agent using a static mixer (see, e.g., PCT Publication No. WO2004058046); however, there appears to be no teaching or suggestion in the art of the use of static mixers for achieving virus inactivation during a protein purification process, as the sample flows from one unit operation to another.
  • the virus inactivation methods described herein are able to achieve more efficient mixing of the sample containing the target protein with a suitable virus inactivating agent (e.g., low pH) in less time than most conventional processes, f 0036] Due to a shorter processing time, any potential adverse effect on the targe protein quality is minimized. For example, it has been shown that extended exposure to low pM conditions can result in aOectmg the quality of the target protein, e.g.. by causing protein aggregates as well other detrimental changes (See, eg., Wang et ai. "Antibody structure, instability and formulation," J. Pbanm Sci. Vol, 96. pg- 1-26 (2007) ⁇ . By having a shorter exposure t me to conditions that may potentially be detrimental to product quality, any damage to the product quality can be minimized or avoided.
  • a suitable virus inactivating agent e.g., low pH
  • the present invention is based, at least in pari, on a surprisin an
  • the flo w-rate of the sample can be controlled such that to place the flow-rate in the laminar flow range. This allows for a more predictable inactivation compared to higher flow-rates, which contributes to turbulence and leads to a narrower optimum operating window. This result is unexpected as generally, a higher flow-rate is required to achieve efficient mixing, [0038] A process operating in the laminar flow range can be better controlled, as mixing does not depend on the presence of turbulence.
  • the in-line mixing process may tall out of the turbulent regime and lose some of its mixing efficiency. However, if the efficiency is dominated by the laminar flow which exists over the entire flow range, efficienc will not suffer,
  • the methods described herein also offer more control over the process parameters.
  • the methods described herein offer more control over the pH conditions, they offer more control over the entire process and enable a more robust process in general
  • the methods described herein also result In a smaller physical footprint of the process, e.g., by eliminating the need to use a pool tank for virus inactivation.
  • Th methods described herein arc able to reduce the overall footprint of a purification process by replacing large pool tanks that are typically used for virus inactivation with in-line static mixers or surge tanks, which are much smaller than pool tanks, [0041]
  • the methods described herein result in the elimination of .an entire unit operation in a purification process. For example, as discussed above, generally, virus Inactivation is performed in a large pool tank. In most conventional processes,
  • the eluate from the upstream bind and el ate chromatography step is collected m a pool tank, often without any mixing capabilities. Accordingly, the sample (i.e., d e elation pool) has to be transferred to a proper pool tank iih mixin capabilities.
  • the pH is then adjusted to the desirable value, followed by one o two hours of incubation or longer, at the desirable pH value.
  • the pH has to again be adjusted to the pH which 1 ⁇ 4 suitable for the next process step, which Is usually a higher pH than for virus inactivation.
  • One (sterile) or two (depth an sterile) filtration steps may also be used to remove any turbidity from the virus inactivation sample prior to subjecting the sample to the subsequent step. Often, each of these steps may be performed over the course of a day and constitute an. entire separate unit operation,
  • shorter exposure results in less or no turbidity, therefore eliminating the need tor subsequent filtration steps.
  • the methods described herein significantly simplify conventional purification processes by eliminating the entire unit operation which Involves using a po l tank for vims Inactivation,
  • the methods described herein offer easier scalability. Scaling a batch pool tank system, involves increasing the stabilization time as well as the mixing efficiency which is based on the underlying mixing system (e.g., an impeller). For example, if the pool tank volume is increased by a factor of 10, then the mixing efficiency would have to be increased by a factor of 10 In order to retain the same mix time. Again, the mixing time should he minimized to safeguard the protein, while maximized to achieve a certain LEV inactivation. In many eases, a mixer cannot be scaled up to an equal mixing efficiency because of limitations on die impeller ize and available motor PIVL The present invention significantly increases the mixing efficiency and offers scalability based on a diniension!ess.
  • Reynolds number (Re) winch depends on. the dimensions of the pipe or connecting tube which includes the in-line static mixer, the flow velocity, liquid density and viscosity. Reynolds number is defined as the ratio of density X static mixer diameter X flow rate to viscosity. Mixing efficiency can he improved by increasing the number of mixing elements of the static mixer. Better scalability and more predictive performance reduce the process parameters down to only pH and t ime and eliminate the dependency of success on mixin efficiency. [0044] in-line static mixing, as described herein, allows a solution to be modified in a very short time, thereby eliminating much of the stabilization time. This allows the entire -process to be compressed and results in a working volume dial is reasonable for sealing and design purposes.
  • the properties of the sample fluid can be changed by introducing a virus activation, fluid into the ma n fluid stream through a T-valve or a manifold system.
  • the required residence time at the inactivation pH can be guaranteed by increasing the residence time in a tube after static mixer either by increasing the length, or diameter of tube or both.
  • a secondary modification can be done to bring the fluid hack into a condition, tha i desirable for the protein and the next process step.
  • virus inactivation m thods described herein facilitate a purification process to be run in a continuous mode, as described in more detail herein,
  • certai terms are first defined. Additional definitions are set forth throughout the detailed description.
  • the term "'static miser,” as used herein, refers to. a device for mixing two fluid materials, typically liquids (e.g., a sample containing a target protein or an eluate from a bind and elide chromatography process step).
  • the device typically includes mixer elements (also referred to as non-moving elements) contained in cylindrical (tube) housing.
  • mixer elements also referred to as non-moving elements
  • the overall system design incorporates a method for delivering two streams of .fluids into the static mixer. As the streams move through the mixer, the non-moving elements of the static mixer continuously blend the materials. Complete mixing depends on many variables includin properties of the fluids, tube inner diameter, number of mixer elements and their design, In various embodiments described herein, a static mixer is used In-line.
  • 'in-line or 'In-line operation refers to a process of moving a liquid sample through a tube or some other conduit without storage in a essel. Accordingly, in some embodiments according to the present invention, a static mixer is used in a "in-line operation" in a tube through which a liquid sample containing a target protein is moved f om one process step to another.
  • VF virus inaeiivatioi ' or "VF refers to the treatment of a sample containing one or more viruses in a manner such that the one or more viruses are no longer able > replicate or are rendered inactive.
  • Virus inaetivation may be achieved by physical means, e.g., heat, ultraviole light, ultrasonic vibration., or using chemical means, e.g., pH change or addition of a chemical Virus inaetivation is typically a process ste which is used during most protein purification processes, especially in case of purification of therapeutic proteins.
  • VI is performed using one or more in-line static mixers or surge tanks. It i understood that failure to detect one or more viruses in a sample using standard assays known in the art and those described herein, is indicative of complete Inaetivatson of the one or more viruses following treatment of die sample with one or more virus inactivating agents.
  • virus inactivating agent refers to any physical or chemical means capable of rendering one or more viruses inactive or unable to re licate
  • a virus inactivating agent, as used hi th methods described herein may include a solution condition change (e.g., phi, conductivity, temperature, etc) or the addition of a solvent/detergent, a salt, a polymer, a.
  • a virus inaetivation agent is a pfl change, where the virus Inactivating agent is. mixed with a sample containing a target molecule (e.g., an eiua e fro a Protein A. hind and elate chromatography step) using an in-line static mixer or a surge tank.
  • a target molecule e.g., an eiua e fro a Protein A. hind and elate chromatography step
  • virus removal refers to the treatment of virus-containing solution such that viruses are removed from the solution. Virus removal may be done through sieving (e.g. using a nanoSltration membrane with an appropriate pore size) or through adsorption (e.g. using a chromatographic device with media of a charge opposite from thai of the virus).
  • Turbulent flow refers to movement of a fluid in which subeurrents in the fluid display turbulence, movin in irregular patterns, while the overall flow is in one direction. Turbulent flow is common in non-viscous fluids moving at high velocities.
  • laminar flow refers to smooth, orderly movement of a fluid, in which there is no turbulence, and any given sub-current moves more or less in parallel with any other nearb sub-current, Laminar flow is common in viscous fluids,, especially those moving at low velocities, In some embodiments according to the methods described herein, laminar flow is employed.
  • pool tank refers to an container, vessel, reservoir, tank, or bag, which used between process steps and has a ske voJunte to enable collection of the entire volume of output from a process step
  • Pool tanks may he used for holding or storing or manipulating solution conditions of the entire volume of output from a process step.
  • the methods described herein obviate the need to use one or more pool tanks.
  • the methods described herein may use one or more surge tanks,
  • surge tank refers to any container or vessel or bag, which is used between process steps; where the output from a process step flows through the surge tank onto the next process step in a purificatio process. Accordingly, a surge tank is different from a pool tank, in that it is not intended to hold or collect the entire volume of output frorn a process step; but instead enables continuous flow of output from one process step to the next.
  • the -volume -of a surge tank used between two process steps in a methods described herein is no more than 25% of the entire volume of the output f om a process step.
  • the volume of a surge tank is no more than 1 % of the entire volume of the output from a process step. In some other embodiments, the volume of a surge tank, is less than 35%, or less than 30%, or less than 25%, or less than 20%, or less than 15%, or less than 10% of the entire volume of a ceil culture in a Moreaefor, which constitutes the starting material from which a target molecule is to he purified, in some embodiments, vims inacfivaiion, as described herein, is achieved v a a surge tank, where a surge tank is employed for mixing a suitable virus in ctivation agent with a sample containing a target protein (e.g., eluate from a Protein A hind and elate chromatography step),
  • a target protein e.g., eluate from a Protein A hind and elate chromatography step
  • the tern ''connected process refers to a process for purifying a target molecule, where the process comprises two or more process steps (or unit operations), which are in direct fluid communication with, each other, such tha fluid material continuously flows through the process step for unit operations) in the process and is in simultaneous contact with two or more unit operations during the normal operation of the process. It is understood thai at times, at Least one process step (or unit operation) in the process may be temporarily isolated from the other process steps (or unit operations) by a barrier such as a val ve in die closed position. This temporary isolation of individual unit operations may be necessary, for example, during start up or shut down, of the process or during removal/replacement of individual unit operations.
  • Protein A and “ProA” are used interchangeably herein and encompasses Protein A recovered from a native source thereof Protein A produced synthetically (e.g., by peptide synthesis or by recombinant techniques), and variants thereof which retain the ability to bind proteins which have a €% €!3 ⁇ 4 region, such as an Pe region.
  • Protein A cars be purchased commercially from e-p!igen, Pharmacia and Femiatech.
  • Protein A is generally immobilized on a solid phase support material
  • M Pr «A also refers to an affinity chromatography resin or column containing chromatographic- solid-support matrix to which- is covalently attached Protein A. in a particular embodiment.
  • Protein A used in the methods according to the present invention is an alkaline stable form of Protein A.
  • Protein A includes one or more Protein A domains or a functional variants or fragments thereat as described in U.S. Patent Applications Mos. US 12/653 A filed December 18. 2009, and 13/489,999 Filed June 6, 2012, both incorporated by reference herein, which relate to cither wild-type uliimertc forms of B, 2 or C domains or nmltimeric variants of one or more domains of Protein A (e.g.. B, Z or C domain pentamers) with each domain having a truncation of 3 or 4 amino acids from the N-terarinns. where a domain may additionally include a mutation to reduce or eliminate Fab binding.
  • a functional derivative, fragment or variant of Protein A used in the methods according to the present invention may be characterized by a binding constant of at least K- ' i0i M, and preferably ⁇ ⁇ M, for the Pe region of moose igOla or human LgGL An interaction obtained with such value for the binding constant is termed high affinity binding" in the present context.
  • such functional derivative or variant of Protein A comprises at least part of a functional IgG binding domain of wild-type Protein , selected from, the natural, domains E. D, A, B, C or engineered mutants thereof which have retained IgG binding
  • the Protein A is immobilized on a solid support
  • solid support;* "solid pha e; “ ⁇ mat i " and ⁇ chromatography matrix, " as used interchangeably herein, generally refers to any kind of particulate sorbent, resin, or other solid phase (e.g., a membrane, non-woven, monolith, etc.) which in a separation process acts as the adsorbent to separate a target molecule (e.g., an Fe region containing protein such as an immunoglobulin) f om other molecules present in a. mixture.
  • a target molecule e.g., an Fe region containing protein such as an immunoglobulin
  • the target molecule is separated from other molecules as a result of differences in rates at which the individual molecules of the mixture migrate through the matrix under the influence of a moving phase.
  • the matrix consisting of resin particles can be put In columns or cartridges.
  • the matrix examples include polysaccharides (such as agarose and cellulose); and other mechanically stable matrices such as silica (e.g. controlled pore glass), poly(styrenedsvinyi)benEene, polyaerylarmde, ceramic particles and derivatives of any of the above, Typically the matrix carries one or more types of Hgands. However, instances exist where the matrix alone is the chromatographic media (e.g., activated carbon, hydroxy apatite, silica, etc.)
  • chromatographic media e.g., activated carbon, hydroxy apatite, silica, etc.
  • a "ligand” is a functional group that is attached to the chromatography matrix and that determines the binding properties of the matrix.
  • Hgands include, hut are not limited to, ion exchange groups, hydrophobic inte.racii.on groups, hydropMHe interaction groups, thioph!i e interactions groups, metal affinity groups, affinity groups, bioaffinity groups, and mixed mode groups (combinations of the aforementioned).
  • Some ligands that can be used herein include, but are not limited to, strong cation, exchange groups, such as si phopropy!, sulfonic acid: strong anion exchange groups, such as trimeihylammonium chloride; weak cation exchange groups, such as earboxy!ic acid; weak anion exchange groups, such as N$ ' N di.ethylarai.no or DEAE; hydrophobic interaction groups, such as phenyl, butyl, propyl, hexyj; and affinity groups, such as Protein A, Protein G, and Protein L
  • the llgand is Protein A or a variant or fragment thereof.
  • chromatography refers to any ki d of technique which se arates the product of interest (e.g., a therapeutic protein or antibody) from contaminants and/or protein aggregates in a hlopharmaeeutical preparation.
  • affinity chromatography* * refers to a protein separation technique in which a target protein (e.g., a Fc region containing protein of interest or antibody ) -specifically bound to a ligand (e.g.. Protein A), which is typically immobilized onto a solid support (the ligand immobilized on the solid support is referred to herein as a "chromatography matrix" ⁇ .
  • a target protein e.g., a Fc region containing protein of interest or antibody
  • a ligand e.g.. Protein A
  • the target protein generally retains its specific binding affinity for the ligand during the chromatographic steps, while other solutes and/or proteins in the mixture do not hind appreciably or specifically to the ligand.
  • Binding of the target protein to the immobilized ligand allows impurities including contaminating proteins or protein imparities (e.g., HCPs) to be passed through the chromatography matrix while the target protein, remains specifically bound to the immobilized ligand on the solid support material; however, some non-specific binding of the contaminating proteins onto the matrix is typically observed.
  • the chromatography matrix Is typically washed one or more times with a suitable wash buffer, in order to remove the non-speciflcal!y bound proteins (e.g., HCPs) and other impurities before elnting the hound protein from the matrix.
  • the specifically bound protein of interest is subsequently eioted from the matrix using a suitable elution buffer which facilitates the separation, of the protein of interest from the matrix,
  • one or more intermediate wash steps are eliminated from such a process, without decreasing the purity of the e!uted target protein, in other words, in. some embodiments according t the present invention, a protein of interest is allowed to hind to a Protein A containing chromatography matrix and is subsequently elutecf without having the need for one or more intermediate wash steps; however, the purity of the protein of interest in the Protein A elution poo! is not affected.
  • the number of intermediate wash steps are reduced compared to a process which would normally use a certain number of wash steps in order to achieve a certain level of purity of the protein of interest in the Protein A elution pool .
  • the level of host ceil proteins is reduced in the Protein A elution pool, despite the elimination of or reduction in the number of intermediate wash steps.
  • the contaminating solutes in the mixture elute from a column of the ion exchange material taster or slower than the solute of interest or are hound to or excluded from the resin relative to the solute of parallel '3 ⁇ 4n ⁇ exchange chromatography" includes cation exchange, anion e chan e, and mixed mode ion exchange chromatography.
  • cation exchange chromatography can bind the target molecule (e.g., a Fc region containing target protein) followed by elation (cation exchange bind and elate chromatography or i CIEX”) or can predominately bind the impurities while the target molecule "fl ws through” the column ⁇ cation exchange f ow through chromatography or "FT- ClEX' .
  • the solid phase material can bind the target molecule (e.g., an Fc region containing target protein) followed by ei tion or can predominately hind the impurities while the target molecule "Hows through'" the column.
  • the term 'Io exchange matrix” ' refers to a chromatography matrix that is negatively charged (i.e., a cation exchange resin) or positively charged (he,, an anion exchange resin),.
  • the charge may be provided by attaching one or more charged iigands to the matrix, e.g. by eovalent linking.
  • the charge may be an inherent property of the matrix (e.g. as is the ease tor silica, which has an overall negative charge).
  • a "cation exchange matrix”* refers to a chromatography matrix which is negatively charged, and which has tree cations for exchange with cations in an aqueous solution contacted with the matrix,
  • a negativel charged ligaml attached to the solid phase to form the cation exchange matrix may. tor example, he a carhoxylate or sulfonate.
  • cation exchange resins include carhoxy--niethyi ⁇ eellu ose, sulphopropyl (SP) immobilized on agarose (e.g., SP- SEPHARO.SE FAST FLOWTM or SP-SEFHAROSE HIGH PERFORMANCES from GE Healthcare) and sulphonyl immobilized on agarose (e.g. S-SBPHAROSE F AST ' FLOWTM from. GE Healthcare), Additional examples include Fraetogel® EMD SO > , Fractogel ⁇ EMD SE Hi heap, Bshffiuno® S and Fraetoge® EMD COO (EMD Milhpore ,
  • A. "mixed mode ion exchange matrix” or “m xed mode matrix” refers to a chromatography matrix which is covalently modified with, catlonk and/or anionic and hydrophobic moieties.
  • a commercially available mixed mode ion exchange resin is BAKERBOND A 'm (J. T. Baker, Phiiiipsbnrg NJ.) containing weak cation exchange groups, a low concentration of anion exchange groups, and hydrophobic ligands attached to a silica gel solid phase support matrix.
  • Mixed mode cation exchange materials typically have cation exchange and hydrophobic moieties.
  • Suitable mixed mode cation exchange materials are Capto® MC (GE Healthcare) and Eshmuno® HCX (Merck Millipore), Mixed mode anion exchange .materials typically have anion exchange and hydrophobic moieties. Suitable mixed mode anion exchange materials ate Capto3 ⁇ 4 Adhere (GE Healthcare).
  • chromatography matrix which is positively charged., e.g. having one or mo e positively charged hgands, such as quaternary amino groups, attached thereto.
  • anion exchange resin include DEAE cellulose, QAE SEPHADEXTM and FAST Q SEPi!A OSETM (GE Healthcare). Additional examples include Ftaciogeli ; EMD TMAE, Fractogel® EMD TMAE higheap, Eshmunof ) Q and Fractogel® EMD DEAE (Merck Millipore),
  • flow-through process;' ' low-ihiough ode ' and How- through chromatography refer to a product separation technique in which at least one product of interest contained in a hiopharmaceutieal preparation along with one or more impurities is intended to flow through a material, which usually binds the one or more impurities, where the product of interest usually flows-through,
  • binding and elttte process binding and elute m de * and "bind and elute chromatography; 5 as used interchangeably herein, relet to a product separation technique in which, at least one product of interest contained in a biophar aeeutkal preparation along with one or more impurities is contacted with a solid support under conditions which facilitate the binding of the product of interest to the solid support.
  • the product of interest is subsequently elated from the solid support.
  • a s lid support having Protein A attached to the solid support is contacted with a sample containin a product of interest and one or more impurities under suitable conditions which facilitate the binding of the product of interest to the Protein A on the solid support- where the one or more impurities are not expected, to specifically bind to the solid support.
  • the product of interest is subsequently eluted from the Protein A containing solid support, in an attempt to separate the product of interest from the one or more impurities, in the methods described herein, subsequent to elation, the Protein A eluiiors pool is subjected to virus inaetivabon using one or more static mixers or a surge tank, as described herein, where the virus inaetivation can be achieved in a matte of minutes to a out an hour, relative to .convent onal processes where virus inaetivation often takes several hours.
  • any foreign or objectionable molecule including a biological macromoleeule such as a DNA, an RNA, one or more host cell proteins, endotoxins, lipids, aggregates and one or more additives which may be present in a sample containing the product of interest thai is being separated from one or more of the foreign or objectionable molecules. Additional ly, such a contaminant may include any reagent which is used In a step which may occur prior to the separation process..
  • HCP * host cell protein
  • CHOP host cell protein
  • the HCP or CHOP is generally present as an impurity in a cell culture medium or lysatc ( ,g cauliflower a harvested cell culture fluid - CCF'T) comprising a protein of interest such as an antibody or an Fc-contaii ng. protein expressed in a CBO cell).
  • the amount of CHOP present in a mixture comprising a protein of interest provides a measure of the degree of purity for the protein of interest.
  • HCP or CHOP includes, but is not limited to, protei of interest expressed by the host cell such as OHO host ceil.
  • the amount of CHOP in a protein mixture is expressed in parts per million relative to the amount of the protein of interest in the mixture.
  • ft is understood that where the host ceil is another eell type, e.g., a mammalian cell besides CHO, an E. coil, a yeast, an
  • ⁇ -ICP refers io the proteins, other than target protein, found in a lysate of the host cell.
  • ppnt parts per .million or “ppnt” are used interchangeably herein to refer to a measivre of purity of a target protein purified by a method of the invention.
  • the units ppm refer to the amount of HCP or CHOP in nanogranis/milligram per protein of interest in milligrams miililiter (i. ., CHOP ppnHCHOP ng mL)/(p.rote of interest mg mL), where the proteins are in solution ⁇ .
  • ' 'clarify,' * "clarification/ * and 'T ariileati n step/ ' as used herein, refers to a process ste for removing suspended particles and or colloids, thereby to reduce turbidity, of a target molecule containing solution, as measured in NTIJ (nephelometric turbidity ' units). Clarification can be achieved b a variety of .means, including centrifugation or filtration, Centrifugation could be clone in a hatch or continuous mode, while filtration could be done in a normal flow (e.g.
  • depth filtration ⁇ or tangential flow mode I n processes use in the industry today, centrifugation is typically followed b depth filters intended to remove insoluble impurities, which may not have been removed by centrifugation. Furthermore, methods for enhancing clarification efficiency can be used, e.g. precipitation..
  • Precipitation of impurities can be performed by various means such as by iloceu!aiion, pl adjustment (acid precipitation), temperature shifls, phase change due to stimulus-responsive polymers or small molecules, or any combinations of these methods.
  • clarification involves any combinations of two or more of centrifugation. filtration, depth filtration and precipitation, in some emN.Klimen.fs, the processes and systems described herein obviate the need for eentrifugation.
  • purification step may be part of an overall purification process resulting in a "homogeneous" composition or sample, which is used herein to refer to a composition or sample comprising less than 100 ppm MCP in composition comprising the protein of interest, alternatively less than 90 ppm, less than SO ppm, less than 70 pprn, less than 60 ppm, less than 50 ppm, less than 40 ppm, less than 30 ppm, less than 20 ppm, less than 10 ppm. less than S p m, or less than 3 ppm of H P.
  • the product of interest is an immunoglobulin
  • immunoglobulin refers to a protein having a basic fonr-polypeptlde chain structure consisting of two heavy and two light chains, said chains being stabilized, for ex ple, by interchain disulfide bonds, which lias the ability to specifically hind antigen.
  • single-chain immunoglobulin or “single-chain antibody'- (used interchangeably herein)
  • domain refers to a globular region of a heavy or light chain polypeptide comprising peptide loops (e.g., comprising 3 to 4 peptide loops) stabilised, for example, by ⁇ -pieated sheet and/or intraehain disulfide bond.
  • Domains arc further referred to herein as “constant” or “variable , based on the relative lack of sequence variation within the domains of various class members in the case of a “constant” domain, or the significant variation within the domains of various class members in the case of a “ variable” domain.
  • Antibody or polypeptide “domains” are often referred to interchangeably in the art as antibody or polypeptide “regions .
  • the "constant " domains of antibody light chains axe referred to interchangeably as "light chain constant regions”, “light chain constant domains”, “Of ' regions or "CL” domains.
  • the “constant” domains of anti body heavy chains are referred o interchangeably as “heavy chain constant regions " , “heavy chain constant domains", “CH “ regions or ⁇ - ⁇ domains,
  • the 'Variable' * domains of antibody light chains are referred to interchangeably as “light chain variable regions”, “light chain variable domains”, “VL” regions or “VL” domains.
  • the “variable” domains of antibody heavy chains are referred to interchangeably as “heavy chain variable regions”, “heavy chain variable domains”, “Vf ⁇ regions or "VH” domains.
  • Immunoglobulins or antibodies may he monoclonal or polyclonal and may exist in rnonomerie or polymeric form, for example.
  • the t rm fragment refers to a pari or portion of an antibody or antibody chain comprising fewer amino acid residues than an intact or complete antibody or antibod chain. Fragments can be obtained via chemical or enxymatie treatment, of an Intact or com lete antibody or antibody chain. Fragments can also be obtained by recombinant means.
  • fragments include Fab, Fab * , F(ab ' )2, Fc and/or Fv fragments.
  • antigen-binding fragment refers to a polypeptide portion of an immunoglobulin or antibody that binds an antigen or competes with intact antibody (Le, with the intact antibody from which they were derived) for antigen binding (i.e.. specific binding). Binding fragments can be produced by c mbinant DNA techniques, or by enzymatic or chemical cleavage of intact immnnoglobuiiiis.
  • Binding fragments include Fab, Fab 1 . P(ah1 ⁇ 4 . Fv, single chains, and single-chain antibodies.
  • the protein of interest which is purified according to the methods described herein is one which comprises a CH2 C3 ⁇ 43 region and therefore is amenable io purification by Protein A chromatography.
  • T he term '' ⁇ 3 ⁇ 42 € ⁇ 3 e io 1 ' when used herein refers to those amino acid residues in the Fc region of an immunoglobuli molecule which interact, with Protein A.
  • Examples of C»2/CH3 region or Fc region-containing proteins include antibodies, immuneadhesins and fusion proteins comprising a protein of interest fused to, or conjugated with, a C «2/C «3 region or Fc region.
  • methods according to the claimed invention are used for purifying a fragment of an antibody which is an Pc ⁇ regio « containing fragment.
  • Fc region and “It region containing protein' " means that the protein contains heavy and/or light chain constant regions or domains (CH and C . regions as defined previously ) of an immunoglobulin. Proteins containing an "Fc region” can possess the effector functions of an. imnmnog bulin constant domain.
  • An "Fc region” such as CH2/CH3 regions, can bind selectively to affinity ligands such as Protein A or functional variants thereof
  • an F region containing protein specifically binds Protein A. or a functional derivative, variant or fragment thereof.
  • an Pe region containing protein specifically ods Protein 0 or Protein L, or functional derivatives, variants or fragments thereof.
  • an inmio nog!obulin or antibody s directed against an "antigen" of interest Preferably, the antigen Is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can. r sult in a therapeutic benefit in that mamma).
  • the term '"monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, ie the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • each monoclonal, antibody is directed against single determinant on the antigen .
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used i accordance with the present invention may he made by the hybridonm method first, described by lCoh!er et aL Nature 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No, 4,816,567).
  • “Monoclonal antibodies” ma also be isolated f om phage antibody libraries using the techniques described in Clackson et aL, Nature 352:624-628 ⁇ 1991 ) and Marks ct a!., .1. MoL Biol 222:581 -59? (1991 ).
  • Monoclonal antibodies may further include "chimeric' * antibodies
  • immunoglobulins ⁇ in which a portion of the heavy and/or light chain is .identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to correspondin sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well, as fragments of such antibodies, so long as they exhibit the desired biological activity (U,S, Patent Ho. 4,816,567; and Morrison et ah, Proa Nut! Acad. Scl. USA 81 :6851-6855 ( 1984)).
  • the term "liypervariable region" when used herein refers to he amino acid residues of an antibody which are responsible tor antigen-binding.
  • irypervariable region comprises amino acid residues from a
  • vS Humani3 ⁇ 4e3 ⁇ 4 forms of non-human (e.g., murine) antibodies are chimeric antibodies which contain minimal sequence derived from non-human
  • immunoglobulins (recipient antibody ) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a. non-human species ⁇ donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • a. non-human species ⁇ donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • Fv framework region (PR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues which are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanize4 antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially ail of the hypervariable loops correspond to those of a non-human Immunoglobulin and all o substantially ail of she FR regions are hese of a human immunoglobulin sequence.
  • the humanized antibody may comprise at least a portion of an immunoglobulin constant region tFc), typically that of a human immunoglobulin.
  • tFc immunoglobulin constant region
  • a "bidier' is a solution that resists changes in pH by the action of its acid-base conjugate components.
  • Various buffers which can be employed depending, for example, on the desired pH of the buffer are described in Buffers. A Guide for the Preparation and Use of Boilers in Biological Systems, Oaeffroy, D., ed. Caibioehem Corporation ( 1.975).
  • Non- limiting examples of buffers include MBS, MOPS, OPSO, Tris, HBPES. phosphate, acetate, citrate, succinate, and ammonium, buffers, as well as eombi nations of these,
  • sample ⁇ refers to a mixture of a molecule of interest or target protein (e.g.. an Fc region containing protein such as an antibody ) and one or more impurities
  • the sample is subjected to a clarification step and a Protein A affinity chromatography step prior to being, subjected to the virus inactivation methods described herein.
  • the sample comprises cell culture feed., for example, feed from a CHO cell culture, which Is subjected to clarification and a Protein A
  • non-mammalian expression systems refers to all host cells or organisms employed to generate therapeutic proteins, where the host cells o organisms arc of non-mammalian origin. Examples of non-mammalia expression systems are E. colt and Pichia pasioris.
  • eltt fe or "eludon ool” as used herein, refers to a solution, containing a molecule of interest obtained via eiution, tor example, following bind and eiutc chromatography (e.g., using a Protein A affinity chromatography matrix).
  • the e!uate may be subjected to one or more additional purification steps, in some embodiments according to the present invention, an elation pool is obtained, which contains a target protein, e.g., an fc region containing protein, wheiein the elution pool is subjected to virus inactivation, as described herein.
  • conductivity refers to the ability of an a ueous solution to conduct an. electric current between two electrodes. In solution, the current flows by ion transpor Therefore, with an increasing amount of ions present in the aqueous solution, the solution will have a higher conductivity.
  • the unit of measurement for conductivity is nhlllSehens per centimete (m$/cm or mS).
  • the conductivity of a solution may be altered by changing the concentration of ions therein, f or example, the concentration of buffering agent and/or concentration of a salt (e.g., NaCl or CI) in the solution may be altered in order to achieve the desired conductivity.
  • the salt concentration of the various buffers is modified to achieve the desired conductivity
  • the " ⁇ or "Isoelectric point" of a polypeptide refer to the p at which the polypeptide's positive charge balances its negative charge, pi can be calculated from the net charge of the amino acid residues or sialic aci d residues of attached carbohydrates of the polypeptide or can be determined by isoelectric focusing, f 00951
  • process step' or ""unit operation refers to the use of one or more methods or devices to achieve a certain result in a purification process. Examples of proces steps or unit operations which may be employed in a purification process include, but are not limited to, clarification, bind and elute chromatography, virus inactivation. Sow-through purification and formulation, It is understood that each of the process steps or unit operations may employ more than, one ste or method or device to achieve the intended result of that process step or unit operation,
  • continuous process refers to a process for purifying a target molecule, which includes two or more process steps (or unit operations), such that the output from one process step flows directly into the nest process ste in the process, without interruption, and where two or more process steps can bo performed concurrently for at least a portion of their duration.
  • process steps or unit operations
  • a virus inactivation method as described herein, constitutes a process step in a continuous purification process, where a sample flows continuously from a Protein A affinity
  • the virus inactivation process step is performed continuously, he., the e!uate from the previous hind and elute chromatography step tie. Protein A affinity chromatography) flows continuously into the virus inactivation. stop, which employs one or more static mixers and/or surge tanks, after which the virus inactivated eluate may be collected i a storage vessel until the next process step is performed,
  • Vital inactivaiion renders viruses inactive, or unable to replicate or infect which Is important, especially in case of the target molecule being intended for therapeutic use. According, virus mactrvation is typically used during- a protein purification process, especially when the protein is intended for therapeutic use.
  • viruses contain lipid or protein coats that can be inactivated by chemical alteration. Rather than simply rendering the virus inactive, some viral inacti vatiou processes are able denature the virus completely .
  • Some of the more widely used virus inaetivation processes include, e.g., use of one or more of the following: solvent detergent inactivaiion (e.g. with Triton X 100); pasteurization (heating); acidic pH inactivaiion; and ultraviolet (UV) inaetivation. It Is also possible to combine two or more of these processes; e.g., perform acidic pi! inactivaiion at elevated temperature.
  • tnaetivation time used with one target molecule was 60 minutes, whereas for another it was 120 minutes, and further, in ease of one target molecule, the virus XMuLV was not completely inactivated even after 120 minutes. Protein concentration also affected the tnaetivation kinetics. In buffer only, XMuLV was inactivated in. 120 minutes; addition of protein prevented complete inactivaiion with the same pB, temperature, and exposure time. The ionic strength of the Inactivating solution appeared to mitigate the effect of increasing protein concentration. [00103] In.
  • Caprylate has been found to inactivate Hpid-enveloped viruses In MAh production processes.
  • Cell eislture harvest broth containing a Psi d mmas exotoxin A-monocional antibody conjugate and a Psemfamo s monoclonal IgM were each spiked with viruses.
  • Herpes simplex virus- 1 CHSV-l and Vesicular stomatitis virus (VSV) viruses were completely inactivated at 20 ;' C in less than 60 minutes.
  • Solvents/detergents are commonly used for virus inaetivation with plasma proteins, S/D is also used for Inaetivation of enveloped viruses during production of recombinant proteins and MAbs, For example, during the production of B -domain deleted recombinant factor VI if S/D is used for virus inaetivation. Although no viruses may be associated with the C.HO cell line used for production, S/D can be added after a cation exchange step, Concentrations of 0.3% TNBF and 1% Triton X- 100 are targeted for at least 30 minutes.
  • virus macdvation employs exposure of the sample containing the target protein to acidic or knv pit
  • virus inactivation employs exposure of the output or el.uate from the hind and elute chromatography step, which is upstream, of virus inactivation. to acidic pB in-line using a static mixer.
  • the pH used for virus inactivation is typically less than 5.0, or between 3,0 and 4.0. In some embodiments, the pH s 3.6 or lower.
  • the duration of time used for virus inactivation when an in-line static mixer tank, is used, is 10 minutes or less, or 5 minutes or less, or 2 minutes or less, or 1 minute or ess.
  • a surge tank is used between the bind and elute chromatography step and the flow-through process step.
  • the duration of time for virus inactivation, when a surge tank is used is typically 1 hour or less, or 30 minutes or less.
  • the virus inaetivation enables the process to be run continuously, rather than having to collect the sample in a pool tank for virus inaetivati n.
  • Viruses an j . Betermm ng Virus iBaeiivation
  • Virus may be "'cleared ** by two main mechanisms: removal (e.g. by filtration or chromatography) or inaetivation (e.g. low pH, detergent, or irradiation). Regulatory recommendations for biopharmaceutical viral clearance can he found in several documents issued by the FDA and EMEA.
  • the viral clearance capabi lities of a process are assessed using scaled- down versions of individual unit operations, A sample of representative process reed is "' iked' * with a known quantity of virus to simulate a viral contamination, and the amount of virus removed or inactivated by the operation is measured. It is recommended thai the amount of virus used for the spike should be "as high as possible to determine the capacity of the production step to inactivate/remove viruses adequately.” However, the virus spike volume .must not be so great that the composition of the production material is significantly altered; a 10% spike volume is generally regarded as the maximum acceptable spike.
  • Virus is generally measured from sam le collected be.fb.re and after individual unit operations in a purification process using assays that quantify infectious virus particles. Clearance is reported in terms of ihe Logj reduction achieved (log reduction value or LRV).
  • LRV log reduction value
  • the minimal LRV is determined using a calculation dependent upon the virus titer in the starting feed and the amount of final material sampled. Consequently, the LRV that can be claimed for highly effective clearance steps can. be raised, by using high titer virus stocks that allow high levels of spike challenge, and large-volume virus assays that increase assay sensitivity. Conversely, demonstrable LRV can he lowered when, feed materials are cytotoxic or interfere with viral. Infection of the detection cells, as this may require sample dilution, which decreases assay sensitivity.
  • the Tissue Culture Infectious Dose 50% assay is one such method for counting the number of infectious viral particles in a sample.
  • the TCIDjo is the quantity of a pathogenic agent (virus) that will produce a eyt paihie effect in 50% of the cultures inoculated.
  • the TCID50 value is proportional to, but not the same as, the number of infections virions in a sample. Titers determined using this method, are typically reported a T( l3 ⁇ 4i mL.
  • a maximum possible titer for the sample is determined using a Minimal Limit of Detectio (LOO) calculation.
  • LOD Minimal Limit of Detectio
  • This calculation considers the sensitivity of the assay and reports the most virus that could be in. a sample without the assay detecting any.
  • the result of these calculations i a titer reported as " ⁇ X * ', meaning that the actual titer In the sample is X or lower (t a. 95% certainty).
  • This LOD calculation depends solely 00 the quantity of sample tested and any predilytions made to the sample before assaying.
  • LRV Log Reduction. Value
  • Viral clearance validation studies are designed to evaluate the ability of a. MAb purification, process to remove or Inactivate many different kinds of viruses.
  • the FDA recommends use of several, model viruses encompassing large and small particles, DMA and R A genomes, as well as chemically sensitive and resistant lipid enveloped and. non-enveloped strains. The rationale is that demonstration, of robust clearance of an appropriately diverse panel of viruses provides assurance thai an undetected, unknown viral contaminant would also be reduced to minima! levels.
  • Table 1 which, summarizes the properties of a model virus panel appropriate for A process viral clearance validation (adapted from KM Q5A)
  • viral contaminants fall into two broad categories: endogenous viruses known to be present in the source .material, and adventitious agents that may infiltrate the process.
  • Static mixers can combine two functions- in the space of one tube: flow dividing and turbulence.
  • the flow dividing uncti n Is accomplished by 8 series of offset sub-elements whereby the fluid exiting one element impinges on the front blade of the next which is offset by 90 degrees.
  • the turbulence is caused, by the screw-like shape of the sub-elements which causes the fluid to rotate. Turbulence is .induced by ihe inabilit of the fluid to move !aminarly with the flow elements and is exaggerated by increased flow rates.
  • this fiui.dk mixing functionality is given, in the Reynolds number. Re. At lo flow rates and thus, low Re, the static .mixers operate in the laminar flow range.
  • This mixing function allows fresh uureacted activation fluid, such as an acidic buffer, to Interact, wit the virus more efficiently and eliminate occlusion of the virus by old, used acti vation fluid, in ease of a batch mixer, the process is dependent on the efficiency of sweeping away old activation fluid and replacing it with the new activation fluid.
  • Static mixing devices or static mixers are available in a variety of materials (e.g., stainless steel. TeflonTM, copper). In. selecting materials for the static mixers used in the methods described herein, it is desirable to select materials that will not react with or cause reactions In components of the sample flowing through the static mixer. It is also desirable that the material be durable and amenable to sterilization (e.g., by autoclaving or chlorine treatment), especially i the sample Is to be administered to a human or other animal.
  • a static mixer may be opaque or transparent,
  • a static mixer Is a part of a system which includes pH and mass .flow control sensors at the inlet, middle and ' outlet of the static mixer.
  • the relative flow rates of the sample e.g., elnate from the upstream bind and e!ute chromatography step
  • acid and base are adjusted continuously to guarantee the desirable pH value.
  • Control software is used that is based on the previously generated range of data so that the feedback algorithm can be model-based and therefore more efficient than traditional systems.
  • the inlet sensor cluster, pH and flow rate can be used to determine the appropriate acid addition feed rate (F A ) «
  • An additional control algorithm may be used thai predicts the intended final pi!
  • an in-line static mixer is irsed to achieve effective virus inaciivatlon
  • chromatography step so the purification process (e.g., protein A el aie) is mked .inline with acid (usually 1 to 3 M acetic acid) using a 3 -way valve and passed through a static mixer.
  • the static mixer dimensions are chosen so as to enable efficient mixing of the acid and product streams, in order to pro vide sufficient residence time (typically 1 -5 mm.) for robust virus inactivaiion, a tube of sufficient volume may follow after the static mixer.
  • the virus inactivated stream is then, mixed with base (usually 1-2 M iris-base at pH 1 1 ⁇ using a 3 -way valve and passed through a static mixer to enable mixing to increase the pH to a desirable pi for the next purification step in the continuous process, which is typically increased from pf 5 to 8.
  • base usually 1-2 M iris-base at pH 1 1 ⁇ using a 3 -way valve and passed through a static mixer to enable mixing to increase the pH to a desirable pi for the next purification step in the continuous process, which is typically increased from pf 5 to 8.
  • the static mixer diameter and the number of elements can be chosen to enable efficient mixing depending on the total stream flow rate (i.e. feed ⁇ «- acid/base flow rate), density and viscosity.
  • the flow rate is chosen to guarantee adequate residence time in conjunction with the static .mixer (number and diameter) selection.
  • the scale-up is performed by holding constant the Reynolds number. Re.
  • the flow rate is maintained low n ugh so that Re remains in the laminar flow range.
  • inaeiivation is achieved using a surge tank.
  • virus inactivation methods according to the present invention may be used as part of any protein purification process, e.g., a purification process performed, in a batch mode or a purification process performed in a continuous mode.
  • the virus inaeiivation methods are part of a continuous purification process which employs several, process step (or unit operations) that are present bot upstream as well as downstream of the virus inacttvaiion method.
  • the bind and elute chromatography process step which is typically performed prior to the virus inaeiivatlon step in. a protein purification process, is performed in batch mode, typical batch bind and elute chromatography operation employs a large column with multiple runs, usually 1 to 10, to process a hatch of clarified DC l culture feed. Accordingly, in some embodiments, once a hatch process has been run, the e!ution poo! can be collected and pumped into a surge tank to perform virus inacti vaikm. in case of a batch bind and elute chromatography process, multiple surge tanks may be used tor virus inaetivation. Because of the smaller ske of the surge tank, a more efficient mixing and virus inaetivation is achieved,
  • the hind and elute chromatography process step is performed in a continuous mode.
  • the continuous process is a .multiple continuous chromatography process, also referred io as CMC, [00130]
  • CMC .multiple continuous chromatography process
  • typically multiple small columns are used; with each run several (typically 1 -50 ) cycles to process a batch of cell culture feed.
  • a CMC" approach has multiple small elutions which require virus inaetivation.
  • a surge tank is used instead of collecting the small elutions in a pool tank for virus inaetivation.
  • the surge tank used for virus Inacti vation can be used to collect subsequent eltuions, in case of the CMC process, (liven appropriate timing and. surge tank sizing, only one surge tank, might he needed to accomplish virus inacti vation In case of the CMC process.
  • the individual elutions irom the multi-column process are treated, similarly tor virus inacti vation.
  • the sample used is 20 mg mL polyclonal IgG (Seraeare) in. 50mM sodium acetate buffer at pll 5.3.
  • the prediction used to avoid cytotoxicit from the buffer is 1 /50, In order to meet the target of LRV > 4, X-MuLV stock of titer 7.0
  • TClDSO/ml is used to spike feed to 6.0 log TOD50 fmh (-10% spike).
  • Example I Based on the results i Example I, the possibility of inactivating a retrovirus using in-line static mixers is investigated.
  • the pH of the solution is lowered as it flows through a channel b injection of acid at a rate calculated for reducing the pli to 3.4.
  • the pi- ⁇ is adjusted back to neutral for the next step in the process.
  • This experiment determines the exposure lime needed to inactivate X-MuL V using an in-line technique.
  • the experimental setup is shown in Figure L
  • the sample used is 9,9 g/L polyclonal igG (Seracare ⁇ in 20 acetic acid buffet at pli 5.O..
  • the feed spiked with virus is passed through the experimental setup shown in Figure I tor in-line pH inactivation.
  • the set-up consists of: (a) a peristaltic pump to transfer the sample feed; (b) two syringe pumps to deliver acid and base; (c) two in-line pH probes; and (d) two static mixers.
  • the flow r tes are predetermined in batch mode based on the amount of acid/base needed to achieve the pH of interest
  • the 'residence time for virus inaetivatlon is altered by having tubes of appropriate diameter and length used after static mixer and before the pi I probe. The results are summarized in Table IV below.
  • the sample used is 20 mg/mL polyclonal IgO ⁇ Seracare ⁇ in 50.m sodium acetate bailer at pH 5.3.
  • the predilu ion used to avoid cytotoxicity from the buffer is 1/50.
  • TOD50/mL is used to spike teed to 5.6 log TCI DSC) /mL ( -10% spike).
  • Assay medk used is standard X- uLV titration media: McCoys 1 % FBS.
  • virus naclivaii n Is performed In a test tube in order to determine the .minimum time and pH requirements for vims inactivaion when using various additives, such as capryHc acid and detergents such, as Triton X, Tweeu and combinations thereof '
  • the teed used in this study Is 9. g/L Seracare polyclonal IgG in. 20 mM sodium acetate, pH 5,0, Three aliquots of 25 mL each are transferred into three separate 50 nil., centrifuge tubes. To 25 mi, of thi s feed, 1,4 mi, of 3 M. acetic acid, pH 2,5 is added to bring down the pH to pH 3.?, One of the centrifuge tubes is left at room temp at 22 C, The other two centrifuge tubes are then placed in wate baths set a 10°C and 35° Upon equilibration, the temperature and pH of the liquid in the tubes is measured (see.
  • a model is then bu lt of the amounts of acid, and base as a function of eluate volume.
  • a second experiment is performed where the Protein A elaate is continuously -adjusted to the desireti pH based on the model, built.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present application relates to novel and improved methods of achieving virus inactivatlon during a protein purification process.

Description

M ETHODS FOR INACTIVATING VIRUSES URI G A PROTEIN
PURIFICATION PROCESS
Figure imgf000002_0001
[0001 ] The resent application claims the benefit of priority of U,S, Provisional Pate t Application No. 61/666,145, filing date June 29, 2012, die entire content of which is incorporated by reference herein in its entirety,
Έ roiib yesft oi
[0002] The present invention provides in-line methods tor inactivating viruses during a protein purification process.
Background of the Invention
[0003] Large-scale and economic purification of therapeutic proteins and especially monoclonal antibodies Is an increasingly important problem for the biotechnology industry. Generally, proteins are produced, by cell coliwre, using either mammalian or bacterial eel! lines engineered to produce the protein of interest such -as a monoclonal antibody. However, once produced, the proteins have to be separated from various impurities such as, host cell proteins (H€Ps)? endotoxins:, viruses, ON'A e c.
[0004] in a typical purification process, once a protein of interest is expressed, in cell culture, the cell culture feed is subjected to a clarification step for removal of cell debris. The clarified cell culture feed containing the protein of interest is then subjected, to one or more chromatography steps, which may sue hide an affinity chromatography step or a catio exchange chromatography step, in order to ensure safety of a protein of interest, especially in case of a therapeutic candidate, it is necessary to inactivate any viruses which may be present in a sample containing the protein of Interest during the purification process. Generally, vims maetivation is performed alter a chromatography step (e.g., after affinity chromatography or after cation exchange chromatography). Typically, in a large scale process, following a chromatography step, an elution pool containing the protein of Interest Is collected in a large tank or reservoir and subjected to a virus inaetivatlon step/process for an extended period of time with mixing, which may take several hours to a day or longer, in order to achieve complete inactivaiion of any viruses thai may be present in the el uion pool
[0005] Several virus inaetivatlon techniques are known in the art including, temperature* H, radiation and exposure to certain chemical agents.
Summary
[0006] The present invention provides methods of virus inaetivation during a protein purification process, which a e several advantages over the methods currently being used in the industry during protein purification. Specifically, the methods described herein obviate the need tor using large tanks or reservoirs for performing the virus inaciivation step during a protein purification process; reduce the overall time required for virus inaetivatlon as well as reduce the overall physical space required to run the virus inaetivatlon operation during a protein purification process, which in turn reduces the overall footprint for the whole purification process.
[0007] In some embodiments, a method for Inactivating one or more viruses, in. a sample in a purification process is provided, where the method comprises continuously mixing the sample with one or more virus inactivating agents as the sample flows iroro the first unit operation to the second unit operation.
[0008] In some embodiments, the first unit operation comprises bind and eS.ute chromatography and the second unit operation comprises a flow-through
purification process, An exemplary bind and eiute chromatography unit operation includes, hut is not limited to, Protein A affinity chromatography,
[0009] In some embodiments, the How-through puri Oeation process comprises two or more matrices selected from the group consisting of activated carbon, anion exchange chromatography media, cation exchange chromatography media and a virus filtration media..
[001 ] in some embodiments, the sample comprises a Protein A eluaie
comprising a target molecule. Exemplary target molecules include, e.g., antibodies.
[001 11 In some embodiments, the sample is mixed with one or more virus inactivating agents using one or more in-line static mixers. In other embodiments, the sample is mixed with one or more virus inactivating agents using one or more surge tanks. When static mixers are used, the flow of sample is in the laminar range. [0012 j Irs some embodiments, one or more virus inactivaiiag a ents are selected from the group consisting of an acid, a salt, a solvent and a detergent
[0013] In some embodiments, a method of inactivating one or more viruses in a Protein A eluate is provided, where the method comprises mixing the eluate with one or mors virus inactivating agents using an in-line static mixer, wherein complete virus Inaetivation is achieved in less than 10 minutes or less than 5 minutes or less than 2 minutes or less than 1 minute,
[0014] in other em o iments* a method of inactivating one or more viruses in a Protein A eluate is provided, where the method comprises mixing the eiuate with one or more virus Inactivating agents using a surge tank, where complete virus inaetivation is achieved in less than one hour or less than 30 minutes,
[0013] In some embodiments, a method for inactivating one or more viruses comprises: (a) subjecting a sample comprising a target protein (e.g., an antibody) to a Protein A affinity chromatography process, thereby to obtain an eluate; and (b) continuously transferring the eluate t an in-line static mixer to mix one or more virus inacti vating agents with the eluate for a duration of time which is equal to or less than 10 minutes, thereby to inactivate one or more viruses,
[0016] In some embodiments, the Protein A eluate which is subjected to virus inaetivation methods described herein is obtained following a Protein A affinity chromatography process performed in hatch mode. In other embodiments, the Protein A affinity chromatography process is performed in a continuous mode, in some embodiments, continuous mode comprises a continuous multi-column chromatography process .
[0017] In some embodiments, one or more virus inactivating agents an acid, used to perform a solution change.
[0018] In some embodiments, the eluate is continuously transferred to a How- through purification, proces step following virus inaetivation, in that the output from the static mixer flows directly into a flow through purification step, which may include use of two or more matrices selected from activated carbon, anion exchange media, cation exchange media and virus filtration media.
[001 ] in other embodiments, the eluate i stored following virus inaetivation for an extended period of time (e.g., 12 to 24 hours or overnight) in a pool or storage tank, before it is subjected to the next uni operation or process step, e,g., a flow- through purification, process step or a cation exchange hind and elute
chromatography step.
j 0020] In some embodiments described herein, the virus inactivaiion methods described herein are a part of a larger protein purification process, which may include several steps including, hoi not limited to, e. ., eultnrmg cells expressing protein in a bloreactot; subjecting the cell culture to clarification, which may employ one or more of precipitation, eentriiugation and/or depth filtration;
transferring the clarified cell culture io a bind and dute chromatography capture ste (e.g.. Protein A affinity chromatography); subjecting the Protein A dua e to a virus inaetivation method, as described herein; subjecting the output from virus inaetivahon to a flow-through purification process, which employs two or more matrices selected fro activated carbon, anion exchange chromatography media, cation exchange chromatography media and vims filtration media: and formulating the protein in the flow-through from the flow-through purification step using diafiitration coiicentration and sterile filtration. Additional details of such processes can be found, e.g., in co-pending application having reference no, P t 2/ 107, filed concurrently herewith, and the entire contents of which are incorporated by reference herein.
[09211 In some embodiments, a fluid sample continuously flows through the entire process, as described above, from one step to the next.
Brief Descri tion of the Pra ings
[0022] Fi gure I is a schematic depiction of an experimental set up for virus inactivaiion using two in-line static mixers. The set up shown includes; (a) a peristaltic pump or sample feed, (b) two syringe pumps to deliver acid and base, c> two in-line pB probes, and (d) two static mixers. The flow rates are predetermined in batch mode based on the amount of acid/base needed to achieve the desi red pH. The residence time for virus inaetivation is altered by having tubes of appropriate diamete and length after each static mixer and before the pH probe.
Figure imgf000005_0001
[0023] Biophar aeeutieal maoufaetnrlng requires the mactfvation or removal of viruses (coming from, animal derived components., including mammalian. cells) for drug safety and to meet the standards set forth by the Pood and Drug Adoiioistration (FDA). Typical processes involve a s um er of viral clearance steps thai
cumulatively provide the necessary protection.
[0024] Some processes used, in the industry involve titration of the solution containing the target protein to a low pH in order to cause destruction of any enveloped viruses and viral components. Generally, the sample containing the target protein must be retained at these conditions for an extended period of time, both because time is needed tor virus inaetivation but also - and more importantly - to ensure homogeneous mixing for effective virus inaciivaHon. Therefore, in ease of large scale processes, the sample containing the target protein must be incubated for an extended period of time at a low pH in order to promote efficient virus inaetivstion often with mixing. See. e.g., Shukla et at, . Chromatography B., 848 (200?) 28-39, which describes virus inactivation using incubation of a protein sample for a suitable duration of time at lo w pi t,
{'0025] The pH conditions are established as a balance between a low pH value that is sufficient to cause inactivation and a high enough value to avoid denaturation. of the target protein. Additionally* the sample most be exposed for a certain amount of time to cause a significant reduction, usually 2 to 6 L V in virus activity values (See, e.g., Miesegaes et al, "Analysis of viral clearance unit operations for monoclonal antibodies." Biotechnology and Bioengineering, Vol. .106, p 238-246 (2010)},
10026] The three parameters that are considered important for virus mactivation process are pH value, exposure time and temperature, assuming homogeneous mixing is present. In. ease of large-scale processes, mixing poses a. challenge due to large volumes and additional parameters,, such as mix rate and mass transfer, also become important .
[0027] in case of f e region containing proteins (e.g., monoclonal antibodies), virus inaetivation is usually performed following elation from: a bind and eiute
chromatography process step (e.g., Protein A affinity chromatography or cation exchange chromatography) because the pH of the elation pool is closer to the desirable pli for virus mactivation. For example, in processes used in the industry today, the Protein A chromatography elation pool, typically has a pH in the 3.5 to 4.0 range and the cation exchange hind and elute chromatography elution pool typically has a pH about 5.0. [0028] I m st processes used in the industry today, the elution pool containing the target protein is adjusted to the pH desired for virus inaetivation and held there for a certain length of time, the combination of pH and time having been shown to result in virus Inaetivation, Longer times are more effective tor virus inacfi vaiion, especially in case of a large-scale process, however, longer times are also known to cause protein damage. Extended exposure to low pH may result in precipitation and formation of aggregates, which i s undesirable and often requi res the use of a depth filter and/or a sterile filter to remove such precipitates and aggregates.
In addition to iow~pH induced product quality issues, agitation in pool tanks can also cause aggregation. Proper mixing is essential in order to homogenize protein pools, and is especially important during mamri¾eturing when protein solutions need to be treated with acid/base or buffer to adjust phi and/or conductivity (See, e,g„ Vazquez-Rev et a!,, "Aggregates in monoclonal antibody manufacturing processes," Biotechnology and Bioengineering, Vol 108, Issue 7, pages 1494-1 S 8 (201 1 )). j0029] Several studies have shown that shear due to agitation alone may not cause protein aggregation, but may be facilitated by stirring In the presence of a gas-liquid interface (See, e.g., Mahler et ah, "Protein aggregation: Pathways, induction factors and nalysis ' i. Pharm, Sei, 9$(9);2909-·· 2934 (2009), Harrison et aL 'Stability of a single-chain Fv antibody fragment when, exposed to a high shear environment combined with air-liquid interfaces;' Biotechnoh Bioeng, 59:517-519 { 1998)), For example, the aforementioned studies have shown that loss in activity for a single- chain Fv antibody fragment results upon stirring the protein in an incompletely tilled vessel. Further, these studies showed that protein activity was not lost in. the fermentation- roth in the presence of antifoam in the fermentation broth. However, addition of anti oarn may not he the ideal solution, especially since it would add additional purification steps and processing time to a purification process.
| 030| The present invention provides novel and improved methods for virus inaetivation (also referred to as iVI" herein) during a protein purification process, which reduces the overall time for virus inaetivation, the cost as well as the overall physical footprint associated with a protein purification process,
[(KB 1 j The methods described herein are able to achieve vims inaetivation in a continuous manner, which significantly reduces the time associated with virus inaetivation relative to most conventional processes, and in tarn, reduces the time for the overall purific tion process, [0032] In some embodiments described herein the methods according to the present invention emplo one or mote in-line static mixers for achieving viru inactivation. In other embodiments, the methods according to the present invention employs one or more surge tanks for achieving virus inaetivaiion. The methods described herein facilitate the running of a whole purification process in a continuous manner, i.e., the sample containing the target protein can continuously flo from one process step (or unit operation) to the next process step (or unit operation), without the need to stop the flo w of the sample after a process step. Accordingly, in some embodiments according to the present invention, the elation pool from an upstream bind and elate chromatography process -step (e.g., Protein A affinity chromatography or cation, exchange chromatography) can be subjected to virus inactivation in-line, using, a static mixer and the sample flows continuously into the next process step (e.g., a flow-through purification process step).
Accordingly, unlike conventional, processes, the eiotioo pool does no have to be mixed or Incubated with a virus inactivating agent for an extended period of time in a pool tank or a vessel before moving on to the next process step lu the purification process.
[0033] Notably, static mixers have been described fo mixing a sample as it is exposed to radiation for inactivating pathogens (see, e.g., U.S. Publication No, 2004013 1497 and PCT Publication No. W02O02O928O6), or mixing a blood sample with a viru inactivation agent using a static mixer (see, e.g., PCT Publication No. WO2004058046); however, there appears to be no teaching or suggestion in the art of the use of static mixers for achieving virus inactivation during a protein purification process, as the sample flows from one unit operation to another.
[0034] The methods described herein, offer several advantages over the
conventional processes used in the industry today, some of which are described below.
[0035] The virus inactivation methods described herein are able to achieve more efficient mixing of the sample containing the target protein with a suitable virus inactivating agent (e.g., low pH) in less time than most conventional processes, f 0036] Due to a shorter processing time, any potential adverse effect on the targe protein quality is minimized. For example, it has been shown that extended exposure to low pM conditions can result in aOectmg the quality of the target protein, e.g.. by causing protein aggregates as well other detrimental changes (See, eg., Wang et ai. "Antibody structure, instability and formulation," J. Pbanm Sci. Vol, 96. pg- 1-26 (2007)}. By having a shorter exposure t me to conditions that may potentially be detrimental to product quality, any damage to the product quality can be minimized or avoided.
[0037! The present invention is based, at least in pari, on a surprisin an
unexpected observation mat even when the flow of the sample is in the laminar How range (e.g., at slow ilow- ate), efficient mixing as well as efficient virus inactivation can be achieved. Specifically, in case of the some methods described herein, because of use of in-line static mixers, the flo w-rate of the sample can be controlled such that to place the flow-rate in the laminar flow range. This allows for a more predictable inactivation compared to higher flow-rates, which contributes to turbulence and leads to a narrower optimum operating window. This result is unexpected as generally, a higher flow-rate is required to achieve efficient mixing, [0038] A process operating in the laminar flow range can be better controlled, as mixing does not depend on the presence of turbulence. For example, if the upstream How conditions require a reduction in. the flow rate, the in-line mixing process may tall out of the turbulent regime and lose some of its mixing efficiency. However, if the efficiency is dominated by the laminar flow which exists over the entire flow range, efficienc will not suffer,
[0039] The methods described herein, also offer more control over the process parameters. In other words, because the methods described herein offer more control over the pH conditions, they offer more control over the entire process and enable a more robust process in general
0040] In addition to some of the foregoing advantages, the methods described herein also result In a smaller physical footprint of the process, e.g., by eliminating the need to use a pool tank for virus inactivation. In general, there is a growing demand for more flexible manufacturing processes that improve efficiency by reducing the overall physical footprint of the process (he., floor space). Th methods described herein arc able to reduce the overall footprint of a purification process by replacing large pool tanks that are typically used for virus inactivation with in-line static mixers or surge tanks, which are much smaller than pool tanks, [0041] The methods described herein result in the elimination of .an entire unit operation in a purification process. For example, as discussed above, generally, virus Inactivation is performed in a large pool tank. In most conventional processes,
S the eluate from the upstream bind and el ate chromatography step is collected m a pool tank, often without any mixing capabilities. Accordingly, the sample (i.e., d e elation pool) has to be transferred to a proper pool tank iih mixin capabilities. The pH is then adjusted to the desirable value, followed by one o two hours of incubation or longer, at the desirable pH value. Following mixing, the pH has to again be adjusted to the pH which ¼ suitable for the next process step, which Is usually a higher pH than for virus inactivation. One (sterile) or two (depth an sterile) filtration steps may also be used to remove any turbidity from the virus inactivation sample prior to subjecting the sample to the subsequent step. Often, each of these steps may be performed over the course of a day and constitute an. entire separate unit operation,
[0042] In some embodiments, shorter exposure results in less or no turbidity, therefore eliminating the need tor subsequent filtration steps. The methods described herein significantly simplify conventional purification processes by eliminating the entire unit operation which Involves using a po l tank for vims Inactivation,
[0043] The methods described herein offer easier scalability. Scaling a batch pool tank system, involves increasing the stabilization time as well as the mixing efficiency which is based on the underlying mixing system (e.g., an impeller). For example, if the pool tank volume is increased by a factor of 10, then the mixing efficiency would have to be increased by a factor of 10 In order to retain the same mix time. Again, the mixing time should he minimized to safeguard the protein, while maximized to achieve a certain LEV inactivation. In many eases, a mixer cannot be scaled up to an equal mixing efficiency because of limitations on die impeller ize and available motor PIVL The present invention significantly increases the mixing efficiency and offers scalability based on a diniension!ess. number called Reynolds number (Re) winch depends on. the dimensions of the pipe or connecting tube which includes the in-line static mixer, the flow velocity, liquid density and viscosity. Reynolds number is defined as the ratio of density X static mixer diameter X flow rate to viscosity. Mixing efficiency can he improved by increasing the number of mixing elements of the static mixer. Better scalability and more predictive performance reduce the process parameters down to only pH and t ime and eliminate the dependency of success on mixin efficiency. [0044] in-line static mixing, as described herein, allows a solution to be modified in a very short time, thereby eliminating much of the stabilization time. This allows the entire -process to be compressed and results in a working volume dial is reasonable for sealing and design purposes. The properties of the sample fluid can be changed by introducing a virus activation, fluid into the ma n fluid stream through a T-valve or a manifold system. Once the fluid is in the newly desired condition, the required residence time at the inactivation pH can be guaranteed by increasing the residence time in a tube after static mixer either by increasing the length, or diameter of tube or both. At the end of the required residence time, a secondary modification can be done to bring the fluid hack into a condition, tha i desirable for the protein and the next process step.
[0045] The virus inactivation m thods described herein facilitate a purification process to be run in a continuous mode, as described in more detail herein,
0046] in order that the present, invention may be more readily understood, certai terms are first defined. Additional definitions are set forth throughout the detailed description.
[0047] The term "'static miser," as used herein, refers to. a device for mixing two fluid materials, typically liquids (e.g., a sample containing a target protein or an eluate from a bind and elide chromatography process step). The device typically includes mixer elements (also referred to as non-moving elements) contained in cylindrical (tube) housing. The overall system design incorporates a method for delivering two streams of .fluids into the static mixer. As the streams move through the mixer, the non-moving elements of the static mixer continuously blend the materials. Complete mixing depends on many variables includin properties of the fluids, tube inner diameter, number of mixer elements and their design, In various embodiments described herein, a static mixer is used In-line.
[0048] The term 'in-line" or 'In-line operation" refers to a process of moving a liquid sample through a tube or some other conduit without storage in a essel. Accordingly, in some embodiments according to the present invention, a static mixer is used in a "in-line operation" in a tube through which a liquid sample containing a target protein is moved f om one process step to another.
[0049] The term '"virus inaeiivatioi ' or "VF refers to the treatment of a sample containing one or more viruses in a manner such that the one or more viruses are no longer able > replicate or are rendered inactive. Virus inaetivation may be achieved by physical means, e.g., heat, ultraviole light, ultrasonic vibration., or using chemical means, e.g., pH change or addition of a chemical Virus inaetivation is typically a process ste which is used during most protein purification processes, especially in case of purification of therapeutic proteins. In methods described herein, VI is performed using one or more in-line static mixers or surge tanks. It i understood that failure to detect one or more viruses in a sample using standard assays known in the art and those described herein, is indicative of complete Inaetivatson of the one or more viruses following treatment of die sample with one or more virus inactivating agents.
[0050] The term "virus inactivating agent" or "virus inactivatlon agent," refers to any physical or chemical means capable of rendering one or more viruses inactive or unable to re licate, A virus inactivating agent, as used hi th methods described herein may include a solution condition change (e.g., phi, conductivity, temperature, etc) or the addition of a solvent/detergent, a salt, a polymer, a. small molecule, drug molecule or any other suitable entity etc, which interacts with one or more viruses in a sample or physical means (e.g., exposure to !JV light, vibration, etc.), such that exposure to the virus Inactivating agent renders one or more viruses inactive or incapable of replicating, in a particular embodiment, a virus inaetivation agent is a pfl change, where the virus Inactivating agent is. mixed with a sample containing a target molecule (e.g., an eiua e fro a Protein A. hind and elate chromatography step) using an in-line static mixer or a surge tank.
[005 S J The term "virus removal" refers to the treatment of virus-containing solution such that viruses are removed from the solution. Virus removal may be done through sieving (e.g. using a nanoSltration membrane with an appropriate pore size) or through adsorption (e.g. using a chromatographic device with media of a charge opposite from thai of the virus).
[0052] The term "turbulent flow"' refers to movement of a fluid in which subeurrents in the fluid display turbulence, movin in irregular patterns, while the overall flow is in one direction. Turbulent flow is common in non-viscous fluids moving at high velocities.
[(1053] The term "laminar flow" refers to smooth, orderly movement of a fluid, in which there is no turbulence, and any given sub-current moves more or less in parallel with any other nearb sub-current, Laminar flow is common in viscous fluids,, especially those moving at low velocities, In some embodiments according to the methods described herein, laminar flow is employed.
[0054] The term "pool tank"" as used herein refers to an container, vessel, reservoir, tank, or bag, which used between process steps and has a ske voJunte to enable collection of the entire volume of output from a process step, Pool tanks may he used for holding or storing or manipulating solution conditions of the entire volume of output from a process step. In various embodiments according to the present invention, the methods described herein obviate the need to use one or more pool tanks.
[0055] In some embodiments, the methods described herein may use one or more surge tanks,
[0056] The term "su ge tank" as sed herein refers to any container or vessel or bag, which is used between process steps; where the output from a process step flows through the surge tank onto the next process step in a purificatio process. Accordingly, a surge tank is different from a pool tank, in that it is not intended to hold or collect the entire volume of output frorn a process step; but instead enables continuous flow of output from one process step to the next. In some embodiments, the -volume -of a surge tank used between two process steps in a methods described herein, is no more than 25% of the entire volume of the output f om a process step. In another embodiment, the volume of a surge tank is no more than 1 % of the entire volume of the output from a process step. In some other embodiments, the volume of a surge tank, is less than 35%, or less than 30%, or less than 25%, or less than 20%, or less than 15%, or less than 10% of the entire volume of a ceil culture in a Moreaefor, which constitutes the starting material from which a target molecule is to he purified, in some embodiments, vims inacfivaiion, as described herein, is achieved v a a surge tank, where a surge tank is employed for mixing a suitable virus in ctivation agent with a sample containing a target protein (e.g., eluate from a Protein A hind and elate chromatography step),
[0057] The tern ''connected process" refers to a process for purifying a target molecule, where the process comprises two or more process steps (or unit operations), which are in direct fluid communication with, each other, such tha fluid material continuously flows through the process step for unit operations) in the process and is in simultaneous contact with two or more unit operations during the normal operation of the process. It is understood thai at times, at Least one process step (or unit operation) in the process may be temporarily isolated from the other process steps (or unit operations) by a barrier such as a val ve in die closed position. This temporary isolation of individual unit operations may be necessary, for example, during start up or shut down, of the process or during removal/replacement of individual unit operations.
[0058] "Protein A" and "ProA" are used interchangeably herein and encompasses Protein A recovered from a native source thereof Protein A produced synthetically (e.g., by peptide synthesis or by recombinant techniques), and variants thereof which retain the ability to bind proteins which have a€%€!¾ region, such as an Pe region. Protein A cars be purchased commercially from e-p!igen, Pharmacia and Femiatech. Protein A is generally immobilized on a solid phase support material The term MPr«A" also refers to an affinity chromatography resin or column containing chromatographic- solid-support matrix to which- is covalently attached Protein A. in a particular embodiment. Protein A used in the methods according to the present invention is an alkaline stable form of Protein A. In a particular embodiment, Protein A includes one or more Protein A domains or a functional variants or fragments thereat as described in U.S. Patent Applications Mos. US 12/653 A filed December 18. 2009, and 13/489,999 Filed June 6, 2012, both incorporated by reference herein, which relate to cither wild-type uliimertc forms of B, 2 or C domains or nmltimeric variants of one or more domains of Protein A (e.g.. B, Z or C domain pentamers) with each domain having a truncation of 3 or 4 amino acids from the N-terarinns. where a domain may additionally include a mutation to reduce or eliminate Fab binding.
[0059] A functional derivative, fragment or variant of Protein A used in the methods according to the present invention may be characterized by a binding constant of at least K-'i0i M, and preferably Κ ίΓ M, for the Pe region of moose igOla or human LgGL An interaction obtained with such value for the binding constant is termed high affinity binding" in the present context. Preferably, such functional derivative or variant of Protein A comprises at least part of a functional IgG binding domain of wild-type Protein , selected from, the natural, domains E. D, A, B, C or engineered mutants thereof which have retained IgG binding
functionality,
[0060] In various embodiments according to the present invention, the Protein A is immobilized on a solid support,
[0061] The terms "solid support;* "solid pha e;" ^mat i " and ^chromatography matrix," as used interchangeably herein, generally refers to any kind of particulate sorbent, resin, or other solid phase (e.g., a membrane, non-woven, monolith, etc.) which in a separation process acts as the adsorbent to separate a target molecule (e.g., an Fe region containing protein such as an immunoglobulin) f om other molecules present in a. mixture. Usually, the target molecule is separated from other molecules as a result of differences in rates at which the individual molecules of the mixture migrate through the matrix under the influence of a moving phase. The matrix consisting of resin particles can be put In columns or cartridges. Examples of materials for forming the matrix include polysaccharides (such as agarose and cellulose); and other mechanically stable matrices such as silica (e.g. controlled pore glass), poly(styrenedsvinyi)benEene, polyaerylarmde, ceramic particles and derivatives of any of the above, Typically the matrix carries one or more types of Hgands. However, instances exist where the matrix alone is the chromatographic media (e.g., activated carbon, hydroxy apatite, silica, etc.)
[0062] A "ligand" is a functional group that is attached to the chromatography matrix and that determines the binding properties of the matrix. Examples of "Hgands" include, hut are not limited to, ion exchange groups, hydrophobic inte.racii.on groups, hydropMHe interaction groups, thioph!i e interactions groups, metal affinity groups, affinity groups, bioaffinity groups, and mixed mode groups (combinations of the aforementioned). Some ligands that can be used herein include, but are not limited to, strong cation, exchange groups, such as si phopropy!, sulfonic acid: strong anion exchange groups, such as trimeihylammonium chloride; weak cation exchange groups, such as earboxy!ic acid; weak anion exchange groups, such as N$'N di.ethylarai.no or DEAE; hydrophobic interaction groups, such as phenyl, butyl, propyl, hexyj; and affinity groups, such as Protein A, Protein G, and Protein L In various embodiments according to the invention, the llgand is Protein A or a variant or fragment thereof. [00631 The term "chromatography," as used herein, refers to any ki d of technique which se arates the product of interest (e.g., a therapeutic protein or antibody) from contaminants and/or protein aggregates in a hlopharmaeeutical preparation.
[0064] The term "affinity chromatography** refers to a protein separation technique in which a target protein (e.g., a Fc region containing protein of interest or antibody ) -specifically bound to a ligand (e.g.. Protein A), which is typically immobilized onto a solid support (the ligand immobilized on the solid support is referred to herein as a "chromatography matrix"}. The target protein generally retains its specific binding affinity for the ligand during the chromatographic steps, while other solutes and/or proteins in the mixture do not hind appreciably or specifically to the ligand. Binding of the target protein to the immobilized ligand allows impurities including contaminating proteins or protein imparities (e.g., HCPs) to be passed through the chromatography matrix while the target protein, remains specifically bound to the immobilized ligand on the solid support material; however, some non-specific binding of the contaminating proteins onto the matrix is typically observed. The chromatography matrix Is typically washed one or more times with a suitable wash buffer, in order to remove the non-speciflcal!y bound proteins (e.g., HCPs) and other impurities before elnting the hound protein from the matrix. The specifically bound protein of interest is subsequently eioted from the matrix using a suitable elution buffer which facilitates the separation, of the protein of interest from the matrix, In embodiments according to the present invention, one or more intermediate wash steps are eliminated from such a process, without decreasing the purity of the e!uted target protein, in other words, in. some embodiments according t the present invention, a protein of interest is allowed to hind to a Protein A containing chromatography matrix and is subsequently elutecf without having the need for one or more intermediate wash steps; however, the purity of the protein of interest in the Protein A elution poo! is not affected. In other embodiments, the number of intermediate wash steps are reduced compared to a process which would normally use a certain number of wash steps in order to achieve a certain level of purity of the protein of interest in the Protein A elution pool . In various
embodiments according to the present invention, the level of host ceil proteins is reduced in the Protein A elution pool, despite the elimination of or reduction in the number of intermediate wash steps. [0065] The terms "ion-exchange"' and 'Ion-exchange chromatography^ as used interchangeably herein, refer to the chromatographic process in which a solute or anatyte of interest i a mixture, interacts with a charged compound linked (such as by eovalent attachment) to a solid phase ion exchange material such that the sokste or anaiyle of interest interacts non»speetiicaliy with the charged compound more or less as compared to the solute impurities or contaminants in the mixture. The contaminating solutes in the mixture elute from a column of the ion exchange material taster or slower than the solute of interest or are hound to or excluded from the resin relative to the solute of interes '¾n~exchange chromatography" includes cation exchange, anion e chan e, and mixed mode ion exchange chromatography. For example, cation exchange chromatography can bind the target molecule (e.g., a Fc region containing target protein) followed by elation (cation exchange bind and elate chromatography or i CIEX") or can predominately bind the impurities while the target molecule "fl ws through" the column {cation exchange f ow through chromatography or "FT- ClEX' . in case of anion exchange chromatography, the solid phase material can bind the target molecule (e.g., an Fc region containing target protein) followed by ei tion or can predominately hind the impurities while the target molecule "Hows through'" the column.
[0066] The term 'Io exchange matrix"' refers to a chromatography matrix that is negatively charged (i.e., a cation exchange resin) or positively charged (he,, an anion exchange resin),. The charge may be provided by attaching one or more charged iigands to the matrix, e.g. by eovalent linking. Alternatively, or in addition., the charge may be an inherent property of the matrix (e.g. as is the ease tor silica, which has an overall negative charge).
[006?] A "cation exchange matrix"* refers to a chromatography matrix which is negatively charged, and which has tree cations for exchange with cations in an aqueous solution contacted with the matrix, A negativel charged ligaml attached to the solid phase to form the cation exchange matrix may. tor example, he a carhoxylate or sulfonate. Commercially available cation exchange resins include carhoxy--niethyi~eellu ose, sulphopropyl (SP) immobilized on agarose (e.g., SP- SEPHARO.SE FAST FLOW™ or SP-SEFHAROSE HIGH PERFORMANCES from GE Healthcare) and sulphonyl immobilized on agarose (e.g. S-SBPHAROSE F AST' FLOW™ from. GE Healthcare), Additional examples include Fraetogel® EMD SO>, Fractogel^ EMD SE Hi heap, Bshffiuno® S and Fraetoge® EMD COO (EMD Milhpore ,
[0068] A. "mixed mode ion exchange matrix" or "m xed mode matrix" refers to a chromatography matrix which is covalently modified with, catlonk and/or anionic and hydrophobic moieties. A commercially available mixed mode ion exchange resin is BAKERBOND A 'm (J. T. Baker, Phiiiipsbnrg NJ.) containing weak cation exchange groups, a low concentration of anion exchange groups, and hydrophobic ligands attached to a silica gel solid phase support matrix. Mixed mode cation exchange materials typically have cation exchange and hydrophobic moieties. Suitable mixed mode cation exchange materials are Capto® MC (GE Healthcare) and Eshmuno® HCX (Merck Millipore), Mixed mode anion exchange .materials typically have anion exchange and hydrophobic moieties. Suitable mixed mode anion exchange materials ate Capto¾ Adhere (GE Healthcare).
[006 j The term "' nion exchange matrix'* is used herein to refer to a
chromatography matrix which is positively charged., e.g. having one or mo e positively charged hgands, such as quaternary amino groups, attached thereto. Commercially available anion exchange resin include DEAE cellulose, QAE SEPHADEX™ and FAST Q SEPi!A OSE™ (GE Healthcare). Additional examples include Ftaciogeli; EMD TMAE, Fractogel® EMD TMAE higheap, Eshmunof) Q and Fractogel® EMD DEAE (Merck Millipore),
10070] The terms "flow-through process;' ' low-ihiough ode ' and How- through chromatography," as used interchangeably herein, refer to a product separation technique in which at least one product of interest contained in a hiopharmaceutieal preparation along with one or more impurities is intended to flow through a material, which usually binds the one or more impurities, where the product of interest usually flows-through,
[0071 1 The terms "bind and elttte process," "bind and elute m de * and "bind and elute chromatography;5 as used interchangeably herein, relet to a product separation technique in which, at least one product of interest contained in a biophar aeeutkal preparation along with one or more impurities is contacted with a solid support under conditions which facilitate the binding of the product of interest to the solid support. The product of interest is subsequently elated from the solid support. In some embodiments according to the methods described herein, a s lid support having Protein A attached to the solid support is contacted with a sample containin a product of interest and one or more impurities under suitable conditions which facilitate the binding of the product of interest to the Protein A on the solid support- where the one or more impurities are not expected, to specifically bind to the solid support. The product of interest is subsequently eluted from the Protein A containing solid support, in an attempt to separate the product of interest from the one or more impurities, in the methods described herein, subsequent to elation, the Protein A eluiiors pool is subjected to virus inaetivabon using one or more static mixers or a surge tank, as described herein, where the virus inaetivation can be achieved in a matte of minutes to a out an hour, relative to .convent onal processes where virus inaetivation often takes several hours.
[0072] The terms'contaminant "'impurity.;" and
Figure imgf000019_0001
used
interchangeably herein,, refer to any foreign or objectionable molecule, including a biological macromoleeule such as a DNA, an RNA, one or more host cell proteins, endotoxins, lipids, aggregates and one or more additives which may be present in a sample containing the product of interest thai is being separated from one or more of the foreign or objectionable molecules. Additional ly, such a contaminant may include any reagent which is used In a step which may occur prior to the separation process..
[0073] The terms'Chinese hamster ovary ceil protein" and "CHOP" are used interchangeably to refer to a mixture of host cell protein ("HCP*) derived from a. Chinese hamster ovary rCBCF} cell culture. The HCP or CHOP is generally present as an impurity in a cell culture medium or lysatc ( ,g„ a harvested cell culture fluid - CCF'T) comprising a protein of interest such as an antibody or an Fc-contaii ng. protein expressed in a CBO cell). The amount of CHOP present in a mixture comprising a protein of interest provides a measure of the degree of purity for the protein of interest. HCP or CHOP includes, but is not limited to, protei of interest expressed by the host cell such as OHO host ceil. Typically, the amount of CHOP in a protein mixture is expressed in parts per million relative to the amount of the protein of interest in the mixture., ft is understood that where the host ceil is another eell type, e.g., a mammalian cell besides CHO, an E. coil, a yeast, an
I S insect cell or plant cell, ϊ-ICP refers io the proteins, other than target protein, found in a lysate of the host cell.
[0074] The term "parts per .million" or "ppnt" are used interchangeably herein to refer to a measivre of purity of a target protein purified by a method of the invention. The units ppm refer to the amount of HCP or CHOP in nanogranis/milligram per protein of interest in milligrams miililiter (i. ., CHOP ppnHCHOP ng mL)/(p.rote of interest mg mL), where the proteins are in solution}.
[0075] The terms "''clarify,'* "clarification/* and 'T ariileati n step/ ' as used herein, refers to a process ste for removing suspended particles and or colloids, thereby to reduce turbidity, of a target molecule containing solution, as measured in NTIJ (nephelometric turbidity' units). Clarification can be achieved b a variety of .means, including centrifugation or filtration, Centrifugation could be clone in a hatch or continuous mode, while filtration could be done in a normal flow (e.g. depth filtration} or tangential flow mode, I n processes use in the industry today, centrifugation is typically followed b depth filters intended to remove insoluble impurities, which may not have been removed by centrifugation. Furthermore, methods for enhancing clarification efficiency can be used, e.g. precipitation..
Precipitation of impurities can be performed by various means such as by iloceu!aiion, pl adjustment (acid precipitation), temperature shifls, phase change due to stimulus-responsive polymers or small molecules, or any combinations of these methods. In some embodiments described herein, clarification involves any combinations of two or more of centrifugation. filtration, depth filtration and precipitation, in some emN.Klimen.fs, the processes and systems described herein obviate the need for eentrifugation.
[0076] The terms '"purifying/* "separating/' or "isolating," as used
interchangeably herein, refer to increasing the degree of purity of a polypeptide or protein of interest or a target protein from a composition or sample comprising the protein of interest and one or more impurities. Typically, the degree of purity of the protein of interest is increased by removing (completely or partially) at least one impurity from the composition, Λ "purification step" may be part of an overall purification process resulting in a "homogeneous" composition or sample, which is used herein to refer to a composition or sample comprising less than 100 ppm MCP in composition comprising the protein of interest, alternatively less than 90 ppm, less than SO ppm, less than 70 pprn, less than 60 ppm, less than 50 ppm, less than 40 ppm, less than 30 ppm, less than 20 ppm, less than 10 ppm. less than S p m, or less than 3 ppm of H P.
[0077] In some embodiments according to the present invention, the product of interest is an immunoglobulin,
[0078] The term ^immunoglobulin." "Ig" or "antibody" (used interchangeably herein) refers to a protein having a basic fonr-polypeptlde chain structure consisting of two heavy and two light chains, said chains being stabilized, for ex ple, by interchain disulfide bonds, which lias the ability to specifically hind antigen. The term "single-chain immunoglobulin" or "single-chain antibody'- (used
interchangeably herein) refers to a protein having a iwo~polypeptide chain structure consisting of a heavy and a light chain, said chains being stabilized, for example, by interchain peptide linkers, which has the ability to specifically bind antigen. The term "domain" refers to a globular region of a heavy or light chain polypeptide comprising peptide loops (e.g., comprising 3 to 4 peptide loops) stabilised, for example, by β-pieated sheet and/or intraehain disulfide bond. Domains arc further referred to herein as "constant" or "variable , based on the relative lack of sequence variation within the domains of various class members in the case of a "constant" domain, or the significant variation within the domains of various class members in the case of a " variable" domain. Antibody or polypeptide "domains" are often referred to interchangeably in the art as antibody or polypeptide "regions . The "constant" domains of antibody light chains axe referred to interchangeably as "light chain constant regions", "light chain constant domains", "Of ' regions or "CL" domains. The "constant" domains of anti body heavy chains are referred o interchangeably as "heavy chain constant regions", "heavy chain constant domains", "CH" regions or Ό-Γ domains, The 'Variable'* domains of antibody light chains are referred to interchangeably as "light chain variable regions", "light chain variable domains", "VL" regions or "VL" domains. The "variable" domains of antibody heavy chains are referred to interchangeably as "heavy chain variable regions", "heavy chain variable domains", "Vf Π regions or "VH" domains.
[0079] Immunoglobulins or antibodies may he monoclonal or polyclonal and may exist in rnonomerie or polymeric form, for example. Ig antibodies- which exist in pentarneric form and/or gA antibodies which exist in rnonomerie. dimeric or .muiUmerie form. The t rm fragment" .refers to a pari or portion of an antibody or antibody chain comprising fewer amino acid residues than an intact or complete antibody or antibod chain. Fragments can be obtained via chemical or enxymatie treatment, of an Intact or com lete antibody or antibody chain. Fragments can also be obtained by recombinant means. Exemplary fragments include Fab, Fab*, F(ab')2, Fc and/or Fv fragments. OOBO] The term "antigen-binding fragment" refers to a polypeptide portion of an immunoglobulin or antibody that binds an antigen or competes with intact antibody (Le,, with the intact antibody from which they were derived) for antigen binding (i.e.. specific binding). Binding fragments can be produced by c mbinant DNA techniques, or by enzymatic or chemical cleavage of intact immnnoglobuiiiis.
Binding fragments include Fab, Fab1. P(ah¼. Fv, single chains, and single-chain antibodies.
[0081) The protein of interest which is purified according to the methods described herein is one which comprises a CH2 C¾3 region and therefore is amenable io purification by Protein A chromatography. T he term ''<¾2€Η3 e io 1' when used herein refers to those amino acid residues in the Fc region of an immunoglobuli molecule which interact, with Protein A. Examples of C»2/CH3 region or Fc region-containing proteins include antibodies, immuneadhesins and fusion proteins comprising a protein of interest fused to, or conjugated with, a C«2/C«3 region or Fc region.
[0082] In a particular embodiment, methods according to the claimed invention are used for purifying a fragment of an antibody which is an Pc~regio« containing fragment.
[0083] The term "Fc region" and "It region containing protein'" means that the protein contains heavy and/or light chain constant regions or domains (CH and C . regions as defined previously ) of an immunoglobulin. Proteins containing an "Fc region" can possess the effector functions of an. imnmnog bulin constant domain. An "Fc region" such as CH2/CH3 regions, can bind selectively to affinity ligands such as Protein A or functional variants thereof In some embodiments, an F region containing protein specifically binds Protein A. or a functional derivative, variant or fragment thereof. In other embodiments, an Pe region containing protein specifically ods Protein 0 or Protein L, or functional derivatives, variants or fragments thereof.
[0084] Generally, an inmio nog!obulin or antibody s directed against an "antigen" of interest Preferably, the antigen Is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can. r sult in a therapeutic benefit in that mamma).
[0085] The term '"monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, ie the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.. Monoclonal antibodies; are highly specific, being directed against a single antigenic site.
Furthermore, in. contrast to conventional {polyclonal} antibody preparations which typically include different antibodies directed against different determinants
(epitopes), each monoclonal, antibody is directed against single determinant on the antigen . The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used i accordance with the present invention may he made by the hybridonm method first, described by lCoh!er et aL Nature 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No, 4,816,567). "Monoclonal antibodies" ma also be isolated f om phage antibody libraries using the techniques described in Clackson et aL, Nature 352:624-628 { 1991 ) and Marks ct a!., .1. MoL Biol 222:581 -59? (1991 ).
[0086] Monoclonal antibodies may further include "chimeric'* antibodies
(immunoglobulins} in which a portion of the heavy and/or light chain is .identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to correspondin sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well, as fragments of such antibodies, so long as they exhibit the desired biological activity (U,S, Patent Ho. 4,816,567; and Morrison et ah, Proa Nut! Acad. Scl. USA 81 :6851-6855 ( 1984)). [0087] The term "liypervariable region" when used herein refers to he amino acid residues of an antibody which are responsible tor antigen-binding. The
irypervariable region comprises amino acid residues from a
Figure imgf000024_0001
determining region*' or *€DiT (i.e.. residues 24-34 (LI ), 50-56 ( 12) and 89-9? (1,3) in the light chain variable domain and 31 -35 (H I ), 50-65 (B2) and 95- 102 (H3) in the heavy chain variable domain: ahat et aL, Sequences of Proteins of
immunological interest, 5,B Ed. Public Health Service, National institutes of Health, Bethesda, Md, (1991 )} and/or those residues from a hypervariable loop" (i.e. residues 26-32 (Li), 50-52 (L2) and 91 -96 (1,3) n the light chain variable domain and 26-32 (HI ), 53-55 (02) and 96-101 (I B) in the heav -chain variable domain; Cfrofhia and Lesk j. oL Biol. 196:901-91 7 ( 1987)). "F m w rk" or "FR" residues are those variable domain res dues other than the hyperv¾rsable re ion residues as herein defined.
[0088] vSHumani¾e¾. forms of non-human (e.g., murine) antibodies are chimeric antibodies which contain minimal sequence derived from non-human
itnmun.ogbbul.in. For the most part, humanized antibodies are human
immunoglobulins (recipient antibody ) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a. non-human species {donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, Fv framework region (PR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues which are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanize4 antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially ail of the hypervariable loops correspond to those of a non-human Immunoglobulin and all o substantially ail of she FR regions are hese of a human immunoglobulin sequence. The humanized antibody may comprise at least a portion of an immunoglobulin constant region tFc), typically that of a human immunoglobulin. For further details, see Jones et aL Nature 321 :522-525 (1986); Riechmann et aL, Nature 332:323-329 (1988}; and Presta, Curr. Op. Struct. Biol. 2:593-596 ( 1992). |0089] A "bidier' is a solution that resists changes in pH by the action of its acid-base conjugate components. Various buffers which can be employed depending, for example, on the desired pH of the buffer are described in Buffers. A Guide for the Preparation and Use of Boilers in Biological Systems, Oaeffroy, D., ed. Caibioehem Corporation ( 1.975). Non- limiting examples of buffers include MBS, MOPS, OPSO, Tris, HBPES. phosphate, acetate, citrate, succinate, and ammonium, buffers, as well as eombi nations of these,
[0090] The term "solution," ^composition'' or "sample as used herein., refers to a mixture of a molecule of interest or target protein (e.g.. an Fc region containing protein such as an antibody ) and one or more impurities, in some embodiments, the sample is subjected to a clarification step and a Protein A affinity chromatography step prior to being, subjected to the virus inactivation methods described herein. In some embodiments, the sample comprises cell culture feed., for example, feed from a CHO cell culture, which Is subjected to clarification and a Protein A
chromatography step prior to virus inactivation.
(0091 ] The term "non-mammalian expression systems" as used herein refers to all host cells or organisms employed to generate therapeutic proteins, where the host cells o organisms arc of non-mammalian origin. Examples of non-mammalia expression systems are E. colt and Pichia pasioris.
[0092] The term "eltt fe" or "eludon ool" as used herein, refers to a solution, containing a molecule of interest obtained via eiution, tor example, following bind and eiutc chromatography (e.g., using a Protein A affinity chromatography matrix). The e!uate may be subjected to one or more additional purification steps, in some embodiments according to the present invention, an elation pool is obtained, which contains a target protein, e.g., an fc region containing protein, wheiein the elution pool is subjected to virus inactivation, as described herein.
[0093] The term "conductivity" refers to the ability of an a ueous solution to conduct an. electric current between two electrodes. In solution, the current flows by ion transpor Therefore, with an increasing amount of ions present in the aqueous solution, the solution will have a higher conductivity. The unit of measurement for conductivity is nhlllSehnens per centimete (m$/cm or mS). and can be measured using a commercially available conductivity meter (eg., sold by Orion), The conductivity of a solution may be altered by changing the concentration of ions therein, f or example, the concentration of buffering agent and/or concentration of a salt (e.g., NaCl or CI) in the solution may be altered in order to achieve the desired conductivity. in some embodiments, the salt concentration of the various buffers is modified to achieve the desired conductivity,
[0094] The "ρΓ or "Isoelectric point" of a polypeptide refer to the p at which the polypeptide's positive charge balances its negative charge, pi can be calculated from the net charge of the amino acid residues or sialic aci d residues of attached carbohydrates of the polypeptide or can be determined by isoelectric focusing, f 00951 The term "process step'" or ""unit operation," as used interchangeably herein, refers to the use of one or more methods or devices to achieve a certain result in a purification process. Examples of proces steps or unit operations which may be employed in a purification process include, but are not limited to, clarification, bind and elute chromatography, virus inactivation. Sow-through purification and formulation, It is understood that each of the process steps or unit operations may employ more than, one ste or method or device to achieve the intended result of that process step or unit operation,
[0096] The term "continuous process," as used herein, refers to a process for purifying a target molecule, which includes two or more process steps (or unit operations), such that the output from one process step flows directly into the nest process ste in the process, without interruption, and where two or more process steps can bo performed concurrently for at least a portion of their duration. In other words, in case of a continuous process, it is not necessary to complete a process step before the next process step is started, but. a portion, of the sample is always moving through the process steps.
[0097] In some embodiments, the different process steps are connected to be operated in a continuous manner, in some embodiments, a virus inactivation method, as described herein, constitutes a process step in a continuous purification process, where a sample flows continuously from a Protein A affinity
chromatography step to th virus inactivation step to the next step in the process, which is typically a flow-through purification process step.
[0098] In some embodiments, the virus inactivation process step is performed continuously, he., the e!uate from the previous hind and elute chromatography step tie. Protein A affinity chromatography) flows continuously into the virus inactivation. stop, which employs one or more static mixers and/or surge tanks, after which the virus inactivated eluate may be collected i a storage vessel until the next process step is performed,
H- Exemplary Viras ¾c^^
[0099] Vital inactivaiion renders viruses inactive, or unable to replicate or infect which Is important, especially in case of the target molecule being intended for therapeutic use. According,, virus mactrvation is typically used during- a protein purification process, especially when the protein is intended for therapeutic use.
[00100] Many viruses contain lipid or protein coats that can be inactivated by chemical alteration. Rather than simply rendering the virus inactive, some viral inacti vatiou processes are able denature the virus completely . Some of the more widely used virus inaetivation processes Include, e.g., use of one or more of the following: solvent detergent inactivaiion (e.g. with Triton X 100); pasteurization (heating); acidic pH inactivaiion; and ultraviolet (UV) inaetivation. It Is also possible to combine two or more of these processes; e.g., perform acidic pi! inactivaiion at elevated temperature.
[001 Ol'j Several virus Inactivating agents for biotechnology products are known in the art. See, e.g., Gail Sofer, '''Virus Inaetivation in the 1990s and into the 21st
Century, Pari 4, Culture Media. Biotechnology Products, and Vaccines ' Biopharm International, January 2003, pp. 50-57), some of which are described belo w.
[00102 J Low pli has been shown to inactivate the xenotropfc murine leukemia vims {XMuLV}. In one study, pi I 3.5-4.0 was found to he effective at 18-26 °C and very little difference was seen in inaetivation kinetics for pB 3.7 u io pH 4.1 . At 2-8 °C, however. pB 4, 1 Inactivaiion was slower and required up to one hour, compared, to abou 30 minutes for pH 3.7. Further, variability was observed for different target molecules being purified. For example, tnaetivation time used with one target molecule was 60 minutes, whereas for another it was 120 minutes, and further, in ease of one target molecule, the virus XMuLV was not completely inactivated even after 120 minutes. Protein concentration also affected the tnaetivation kinetics. In buffer only, XMuLV was inactivated in. 120 minutes; addition of protein prevented complete inactivaiion with the same pB, temperature, and exposure time. The ionic strength of the Inactivating solution appeared to mitigate the effect of increasing protein concentration. [00103] In. another stud , low pH was investigated for mactivaiion of XMuLV nd pseudorabies virus (FRY'} viruses m ease of eight different monoclonal antibodies (MS) produced in either Sp2/0 or NSO mouse ceil lines. The pi I values used ranged between 3.14 and 3.62. .resulting in LRVs ranging from around 3 to above 6 (i . virus cannot be measured in the sample). These data are being used to support generic virus inaetiva ion approaches. A different study of 13 products, mostly but not all MAbs. illustrated the ability of ptl 3,6 -4,0 to inactivate several different viruses in five to 60 minutes.
00104] Caprylate has been found to inactivate Hpid-enveloped viruses In MAh production processes. Cell eislture harvest broth containing a Psi d mmas exotoxin A-monocional antibody conjugate and a Psemfamo s monoclonal IgM were each spiked with viruses. Herpes simplex virus- 1 CHSV-l ) and Vesicular stomatitis virus (VSV) viruses were completely inactivated at 20;'C in less than 60 minutes.
However, at 5C'C only partial inaetivation of VSV as shown after 120 minutes. A non-ionteed form of the caprylate is maintained aver a broad pH range, and the non- ionized form Is effective in viral inacttvatkm at concentrations between 0,001 and 0,07 weight %, VSV and vaccini virus were inactivated slower than HSY-1 at pH 6.3.
(001 5] Detergents have also been, used as a virus inaetivation agent, Triton X- 100 (0.5%, 4 C'C) completely inactivated respiratory syncytial virus (RSV} and friend murine leukemia virus (FrMuLV) viruses within four hours without influencing the binding capacity of a number of MAbs, LoglO reduction values were >3.8 for FrMuLV and >5.4 for RSV. Other data have shown that MuLV was not inactivated, by 0, 1 to I H Tween,
[00106] Solvents/detergents (S/D) are commonly used for virus inaetivation with plasma proteins, S/D is also used for Inaetivation of enveloped viruses during production of recombinant proteins and MAbs, For example, during the production of B -domain deleted recombinant factor VI if S/D is used for virus inaetivation. Although no viruses may be associated with the C.HO cell line used for production, S/D can be added after a cation exchange step, Concentrations of 0.3% TNBF and 1% Triton X- 100 are targeted for at least 30 minutes. S/D treatment has been shown to completely and rapidly inactivated all the enveloped viruses tested, which include parainf uenza-3 virus (Pl~3), XMnLV, infectious bovine rbi otracheitis viru (IBRj. and malignant catarrhal fever virus (MCF), (See, Sorer et a!.. Id.) [00107} Beta-p'ropioiaeione has been proposed for viral inactivation in n ked NA vaccines. It was found thai for a 16-hour treatment at 4 °C. the initial concentration of β-propio!aetone should not exceed 0,25% in order to prevent loss of gene expression.
[00108] In some embodiments according to the present invention, virus macdvation employs exposure of the sample containing the target protein to acidic or knv pit Accordingly, in some embodiments described herein, virus inactivation employs exposure of the output or el.uate from the hind and elute chromatography step, which is upstream, of virus inactivation. to acidic pB in-line using a static mixer. The pH used for virus inactivation is typically less than 5.0, or between 3,0 and 4.0. In some embodiments,, the pH s 3.6 or lower. The duration of time used for virus inactivation, when an in-line static mixer tank, is used, is 10 minutes or less, or 5 minutes or less, or 2 minutes or less, or 1 minute or ess. In. other embodiments, a surge tank is used between the bind and elute chromatography step and the flow-through process step. The duration of time for virus inactivation, when a surge tank is used, is typically 1 hour or less, or 30 minutes or less. In case of both use of in-line static mixers or surge tanks, the virus inaetivation enables the process to be run continuously, rather than having to collect the sample in a pool tank for virus inaetivati n.
HI. Exemplary Viruses an j .Betermm ng Virus iBaeiivation
{00109] Virus may be "'cleared** by two main mechanisms: removal (e.g. by filtration or chromatography) or inaetivation (e.g. low pH, detergent, or irradiation). Regulatory recommendations for biopharmaceutical viral clearance can he found in several documents issued by the FDA and EMEA.
001 10} 'The viral clearance capabi lities of a process are assessed using scaled- down versions of individual unit operations, A sample of representative process reed is "' iked'* with a known quantity of virus to simulate a viral contamination, and the amount of virus removed or inactivated by the operation is measured. It is recommended thai the amount of virus used for the spike should be "as high as possible to determine the capacity of the production step to inactivate/remove viruses adequately." However, the virus spike volume .must not be so great that the composition of the production material is significantly altered; a 10% spike volume is generally regarded as the maximum acceptable spike. [001 1 11 Virus is generally measured from sam le collected be.fb.re and after individual unit operations in a purification process using assays that quantify infectious virus particles. Clearance is reported in terms of ihe Logj reduction achieved (log reduction value or LRV). When a unit operation achieves clearance to such a degree that no virus is detected in the processed material, the minimal LRV is determined using a calculation dependent upon the virus titer in the starting feed and the amount of final material sampled. Consequently, the LRV that can be claimed for highly effective clearance steps can. be raised, by using high titer virus stocks that allow high levels of spike challenge, and large-volume virus assays that increase assay sensitivity. Conversely, demonstrable LRV can he lowered when, feed materials are cytotoxic or interfere with viral. Infection of the detection cells, as this may require sample dilution, which decreases assay sensitivity.
[00.1 .12J Several methods are known in the art to assay for virus inieetlvity. The Tissue Culture Infectious Dose 50% assay is one such method for counting the number of infectious viral particles in a sample. The TCIDjo is the quantity of a pathogenic agent (virus) that will produce a eyt paihie effect in 50% of the cultures inoculated. The TCID50 value is proportional to, but not the same as, the number of infections virions in a sample. Titers determined using this method, are typically reported a T( l¾i mL.
[00! 13] When no virus at ail is detected, by the assay, a maximum possible titer for the sample is determined using a Minimal Limit of Detectio (LOO) calculation. This calculation considers the sensitivity of the assay and reports the most virus that could be in. a sample without the assay detecting any. The result of these calculations i a titer reported as "< X*', meaning that the actual titer In the sample is X or lower (t a. 95% certainty). This LOD calculation, depends solely 00 the quantity of sample tested and any predilytions made to the sample before assaying. When the titer Is reported as "< X'\ the corresponding Log Reduction. Value (LRV is reported as meaning that the resulting .LRV is Y of higher,
[001 14] Viral clearance validation studies are designed to evaluate the ability of a. MAb purification, process to remove or Inactivate many different kinds of viruses. The FDA recommends use of several, model viruses encompassing large and small particles, DMA and R A genomes, as well as chemically sensitive and resistant lipid enveloped and. non-enveloped strains. The rationale is that demonstration, of robust clearance of an appropriately diverse panel of viruses provides assurance thai an undetected, unknown viral contaminant would also be reduced to minima! levels. Four viruses that fit these guidelines are shown in Table 1 , which, summarizes the properties of a model virus panel appropriate for A process viral clearance validation (adapted from KM Q5A)
Table I
Virus Family Natura Genom Envelop Diaroeie Shape esistan i Host e Type e r (n j ce to
physio- cnemica! treatment s
Minute virus Parvo Mouse DNA no 18-24 lcosohedr Very high of ice a!
Xenotropic Retro Mouse RNA yes 80-110 Spherical ¾ Low murine
virus
Pseudorabse Herpe Swine DNA yes 120-200 Spherical " Medium s virus s
Reovims 3 eo Variou DNA no 60-80 Spherical Medium
s
[001 15 ] Generally, viral contaminants fall into two broad categories: endogenous viruses known to be present in the source .material, and adventitious agents that may infiltrate the process.
[00 ! 16) The methods described herein can be used for inactivating both of these categories of viruses.
I . Static MM
[001 17) Although static mixers are used in other industries for in-line mixing, they have not been widely used in the pharmaceutical industry because processes generally operate in hatch mode. Further, although in-line buffer mixing ami dilution techniques have been described for protein purification processes (e.g. in commercial systems offered by Techni rom, BioRad and GE Healthcare), it has not been used for virus inactivation, especially as virus motivation is generally performed in pool tanks for large scale processes,
[001 1 %} One skilled in the art will recognize that numerous static mixing devices can be used in the present invention, so long that the mixer disrupts the liquid flow in order to enable full mixing. [00! 19] Static mixers can combine two functions- in the space of one tube: flow dividing and turbulence. The flow dividing uncti n Is accomplished by 8 series of offset sub-elements whereby the fluid exiting one element impinges on the front blade of the next which is offset by 90 degrees. The turbulence is caused, by the screw-like shape of the sub-elements which causes the fluid to rotate. Turbulence is .induced by ihe inabilit of the fluid to move !aminarly with the flow elements and is exaggerated by increased flow rates. The measurement of this fiui.dk mixing functionality is given, in the Reynolds number. Re. At lo flow rates and thus, low Re, the static .mixers operate in the laminar flow range. This mixing function allows fresh uureacted activation fluid, such as an acidic buffer, to Interact, wit the virus more efficiently and eliminate occlusion of the virus by old, used acti vation fluid, in ease of a batch mixer, the process is dependent on the efficiency of sweeping away old activation fluid and replacing it with the new activation fluid.
[00 ! 20] Static mixing devices or static mixers are available in a variety of materials (e.g., stainless steel. Teflon™, copper). In. selecting materials for the static mixers used in the methods described herein, it is desirable to select materials that will not react with or cause reactions In components of the sample flowing through the static mixer. It is also desirable that the material be durable and amenable to sterilization (e.g., by autoclaving or chlorine treatment), especially i the sample Is to be administered to a human or other animal. A static mixer may be opaque or transparent,
[00121 ) In some embodiments according to the methods described herein, a static mixer Is a part of a system which includes pH and mass .flow control sensors at the inlet, middle and 'outlet of the static mixer. The relative flow rates of the sample (e.g., elnate from the upstream bind and e!ute chromatography step), acid and base are adjusted continuously to guarantee the desirable pH value. Control software is used that is based on the previously generated range of data so that the feedback algorithm can be model-based and therefore more efficient than traditional systems. For example, the inlet sensor cluster, pH and flow rate, can be used to determine the appropriate acid addition feed rate (FA)« An additional control algorithm may be used thai predicts the intended final pi! value at pH 2.0, If the value is not achieved correctly, the pump FA is changed to bring the fluid to the correct value. This could occur for a number of reasons, e.g., there being additional buffering capacity within the feed front the chromatographic step or the titration value may not be linear. V. Process for Virus inactivation t in an in-U Static Mh«r
[00.122] As described above, in some embodiments according to the methods described herein, an in-line static mixer is irsed to achieve effective virus inaciivatlon,
[00123] In order to enable low pH continuous vims inacfivation tor a continuous process, as described herein, the e!uate from the previous bind and elute
chromatography step so the purification process (e.g., protein A el aie) is mked .inline with acid (usually 1 to 3 M acetic acid) using a 3 -way valve and passed through a static mixer. The static mixer dimensions are chosen so as to enable efficient mixing of the acid and product streams, in order to pro vide sufficient residence time (typically 1 -5 mm.) for robust virus inactivaiion, a tube of sufficient volume may follow after the static mixer. The virus inactivated stream is then, mixed with base (usually 1-2 M iris-base at pH 1 1 } using a 3 -way valve and passed through a static mixer to enable mixing to increase the pH to a desirable pi for the next purification step in the continuous process, which is typically increased from pf 5 to 8.
[ 00124] The static mixer diameter and the number of elements can be chosen to enable efficient mixing depending on the total stream flow rate (i.e. feed«- acid/base flow rate), density and viscosity. The flow rate is chosen to guarantee adequate residence time in conjunction with the static .mixer (number and diameter) selection. The scale-up is performed by holding constant the Reynolds number. Re. Notably, in methods according to the present invention* the flow rate is maintained low n ugh so that Re remains in the laminar flow range.
VI. P ocess for Virus l!ttacttyattiea. Us ng a Surge Tank
[00125] In some embodiments according to the present invention, virus
inaeiivation is achieved using a surge tank.
[00126] The virus inactivation methods according to the present invention may be used as part of any protein purification process, e.g., a purification process performed, in a batch mode or a purification process performed in a continuous mode.
[00127] In some embodiments described herein, the virus inaeiivation methods are part of a continuous purification process which employs several, process step (or unit operations) that are present bot upstream as well as downstream of the virus inacttvaiion method.
[00128] in some embodiments, the bind and elute chromatography process step, which is typically performed prior to the virus inaeiivatlon step in. a protein purification process, is performed in batch mode, typical batch bind and elute chromatography operation employs a large column with multiple runs, usually 1 to 10, to process a hatch of clarified cei l culture feed. Accordingly, in some embodiments, once a hatch process has been run, the e!ution poo! can be collected and pumped into a surge tank to perform virus inacti vaikm. in case of a batch bind and elute chromatography process, multiple surge tanks may be used tor virus inaetivation. Because of the smaller ske of the surge tank, a more efficient mixing and virus inaetivation is achieved,
[00129] In some other embodiments, the hind and elute chromatography process step is performed in a continuous mode. In a particular embodiment, the continuous process is a .multiple continuous chromatography process, also referred io as CMC, [00130] In case of a CMC operation, typically multiple small columns are used; with each run several (typically 1 -50 ) cycles to process a batch of cell culture feed. Because of the small column si¾e and a large number of cycles, a CMC" approach has multiple small elutions which require virus inaetivation. Instead of collecting the small elutions in a pool tank for virus inaetivation, in some embodiments, a surge tank is used instead.
[0 13.1 J Use of a surge tank, as described herein, provides a better solution homogeneity more efficiently because of its smaller size and better mixing capabilities, relative to a pool tank used in conventional processes. The solution i then held for a time sufficient enough for robust virus inaeti vation, usually 1 - 30 minutes or under 1 hour, i.e. significantly shorter than what is required when using a larger pool tank. After the hold step* the pH and conductivity are adjusted in the surge tank to the desired value set for the next unit operation. The virus inactivated solution, is then pumped to feed the next unit operation. Once emptied, the surge tank used for virus Inacti vation can be used to collect subsequent eltuions, in case of the CMC process, (liven appropriate timing and. surge tank sizing, only one surge tank, might he needed to accomplish virus inacti vation In case of the CMC process. The individual elutions irom the multi-column process are treated, similarly tor virus inacti vation. [00132] This invention is further illustrated by the following examples which should not be construed as limiting, The contents of all referen s, patents and published patent applications cited throughout this application, as well as the Figures, are incorporated herein by reference.
EExxaampllee
Figure imgf000035_0001
tubes
[00133] in this representative experiment, a retrovirus spiked into -a -solution containing a Ab is inactivated by short-time incubation at low p -L The goal of the experiment is to understand the pi ! and exposure time required for complete retrovirus inaciivation in a solutio containing high, concentrations of protein (antibody), I'h minimal time required to inactivate X-MuLV is determined at pH's ranging from 3, 1 -3,5. The experiment is performed in test tabes. The maximum experimental time tested for inaetivation is five minutes using a static miser.
[00 ! 34] The sample used is 20 mg mL polyclonal IgG (Seraeare) in. 50mM sodium acetate buffer at pll 5.3. The prediction used to avoid cytotoxicit from the buffer is 1 /50, In order to meet the target of LRV > 4, X-MuLV stock of titer 7.0
TClDSO/ml, is used to spike feed to 6.0 log TOD50 fmh (-10% spike).
Accounting lor the 1/50 prediction and the -1/5 dilution, of the material in the process of acidifying and neutralising the sample (1/250 altogether), this results in an observed. LRV of > 4,44 (assuming target spike level is achieved). Assay media used is standard X-MuLV titration media: McCoys * 1 % FBS, I X
penicillin/streptomycin, I L-glutamine, I X NEAA. The results are summarized in Table 11 below. Table II
X-MuLV log reductio values CLRV) target pB -7 3.10 3.50 actual pH ~? 2.85 336
1 mm not tested >4.36 >4;?5
3 min not tested 436 4.75
5 mm 0.1 ¾4 36 ¾4 75
[0 135] The results clearly indic te thai X~M«I. V is rapidly inactivated to (he point of non-detection within one minute at both pH 2.85 and 3.36, No inaettvation is seen at pH ?, The high protein conc n ration (20 g/mL'Seracare polyclonal IgG) of these buffers is not an impediment to achieving rapid maeisvahou (See, e.g., Kurt Brorson et al. "Bracketed Generic Inaetivation of Rodent Retroviruses by to p! l Treatment for Monoclonal Antibodies and Recombinant Proteins,** Biotech, Bioeng. Vol. 82, No. 3 (2003» xamp e 2. Time dependence of X-M»LV low-pH vtrtts Inact ation n jcst lubes for both polyclonal and nionocl nal lfeg jeg
(001361 In this representative experiment,, the same protocol as described in Example .1 is followed, in order to better understand at what pH. there is no virus inaettvation, or alternatively, the minimum pH where there s mactivation,
[00.137] Both polyclonal IgG (Seraeare) and two monoclonal antibodies (MAbOS and MAb04) produced in QIC) ceils are used. The results are summarized in Table III below, where the results with ** >" indicate that no virus is detected in the sample. Table Ϊ
Figure imgf000037_0001
[0 138] As result's in 1 able 1 indicate thai half a minute is ade ua e for virus maetivation at pH 3.3. Increasing the pH to 3,6 extends the time needed to 1.1 minutes. Ten minutes are required at pi above pB 3,6, while at pH 4 and above, no virus mactivation is observed even with one hour exposure.
'Exam le 3, Tim le >entle«cf of X-Mu 1. V to ~pH yirm n Jnaefiyat aoti wiife static
roxcrs
[0 139] In this representative experiment, the pH and exposure time required for complete retrovirus inactivation. in a high protein ( A'b) feed solution using static mixers is investigated.
[00140] Based on the results i Example I, the possibility of inactivating a retrovirus using in-line static mixers is investigated. The pH of the solution is lowered as it flows through a channel b injection of acid at a rate calculated for reducing the pli to 3.4. At the downstream end of the channel, the pi-ί is adjusted back to neutral for the next step in the process. This experiment determines the exposure lime needed to inactivate X-MuL V using an in-line technique.. The experimental setup is shown in Figure L
[00141 ] The sample used is 9,9 g/L polyclonal igG (Seracare} in 20 acetic acid buffet at pli 5.O.. The feed spiked with virus is passed through the experimental setup shown in Figure I tor in-line pH inactivation. The set-up consists of: (a) a peristaltic pump to transfer the sample feed; (b) two syringe pumps to deliver acid and base; (c) two in-line pH probes; and (d) two static mixers. The flow r tes are predetermined in batch mode based on the amount of acid/base needed to achieve the pH of interest The 'residence time for virus inaetivatlon is altered by having tubes of appropriate diameter and length used after static mixer and before the pi I probe. The results are summarized in Table IV below.
Table IV
Seraeare Seraeare time (mm) LRV (&) pH 3,3 tim* (mitt) LEV («) pH. 3,6
0.5 >3.6 «.6 2.80
0.9 >3,b LI >3.6
I A >3.6 I.? >3.,6
2.3* >3.6 2. 3,6
3,5 >3,6 4.4 >3,6
'3 fractions, in succession, for 90 'seconds each are collected to ensure consistent virus inacttvation in the 4.5 mi.n ti.me frame
[00142] As summarized in Table IV, complete maetivation is observed at pli 3.3 for ail the time points. For pM 3,6, time > 1 J mm gives complete inacdvation. In addition, the fractions collected at residence times 2,3 and 2,8 mm show complete virus inaeiivatioo over the 4.5 iron time frame of data collection, thereby indicating that in-line low pH virus maetivation is consistent over time,
Exa.rn.pie 4. ime dependence
ηι| Γ*
[00143] In this representative experiment, the pH and exposure time required for complete retrovirus inaeuvation in a high protein ( MAb) feed solution when using static misers is investigated. The experimental setup is shown in. Figure I ,
[00144] The sample used is 20 mg/mL polyclonal IgO {Seracare} in 50.m sodium acetate bailer at pH 5.3. The predilu ion used to avoid cytotoxicity from the buffer is 1/50. In order to meet the rget of LR.V > 4, X- uLV stock of titer 6,9
TOD50/mL is used to spike teed to 5.6 log TCI DSC) /mL ( -10% spike). Assay medk used is standard X- uLV titration media: McCoys 1 % FBS. IX
penieillin/stieptomydm IX L~$utamine, 1 ' EAA.
[00145] For the acidification and neutralization. 3 M acetic acid and 2 M iris buiier are used. Control samples are also generated using only tubes .no static mixer) n order to directly cheek the effect of the static mixer. The results are summarized in Table V below.
Table V
Sample Time 1 "iter ( g TOP* LEV
(mm)
pH 7 Hold •"60 3.8 n/a pli 3.4 with static mixer 0.5 ·: 1.5 >2.2. -4.3
1 2.9 (19
1 <-0.5 >4 3
3 ··().5 >4.3 pH 3.4 without static mixer 0.25 3.3 0.4
0.5 3.3 0,5 s '5 1 t Π " /?
J 3.0 0.7 p!i 7 with static mixer 4, 1 -03
TFF/UC 2.4 Prep (positive con roi I) n/a 6.5 n/a
{00146] As summarized in Table , no virus inactivatlon is observed for the positive control and the two samples exposed to pH 7, either when held in a lest, tube or when passed through a static mixer at the maximum exposure time, 3 mm.. The samples taken through the static mixer show complete virus inaciivation at times longer than one minute, Static mixer is critical for virus inaciivation as very little ioaeiivatlon occurs in its absence. At least two minutes at target pi l 3,4 is required to achieve >4 FRY of X-MuLV, The static mixer itself does not cause any inaciivation at neutral pH. Example 5. Time dependence of ° nL¥ m mmtwniim using eafirvlfc add and .defem nts in test tubes
['001.47) .n this experiment, virus naclivaii n Is performed In a test tube in order to determine the .minimum time and pH requirements for vims inactivaion when using various additives, such as capryHc acid and detergents such, as Triton X, Tweeu and combinations thereof'
[00148 [ Both pure Mab in buffer as well m Mab in clarified ceil culture are used in this experiment. While the foregoing additives are known- to effect virus inaeiivation, this example demonstrates that bv usina static mixers, virus inaeti vaiioo can e obtained in shorter time frame. The results of this representative experiment are shown in Table VI,
Table VI
Pure MAbft? MAbfl'5 cel culture
Tins* CaprykU Trite wee T PTw«en2 C'apryia CaprylsU Trite
Us iw « 2ίϊ m nX it 0 e 2W « 29 mM B X mM ii p.H S,5 {8,5 <t%> mM pH pH 5.5 (»,5
%) %)
0 0 0 0 O 0 0 0 0
1 3.l >3-1 >1! 0.5 >3J 2.91 2.63 >V1
5 3.1 >3.) 3J i 5. 3.8 .S 3.1
10 >3,1 >3.i 3J VS. 3,t 23.8 >3,S >3.i
20 >3. j 3J 3J 3.8 S3,S >3,t
[0 140] As shown in T able V), in t e se of purified λ l-\b. one v. rj.i u.te appi :ars to ί.'ί,. til e virus H taetivatson WHO. au auus t.sves used except t ;
Twe en. In a is of the larified e ell culture, onlv Triton ! X at 0.5% : results in complete virus inacti vation after one minute, while eaprylate at both pH and salinities requires a minimum of f ve minutes. Example 6. Effect of salt a id MAb eon mtratfon on X-Ma t ¥ v ms
Inaet iyaf aoa at; , Η 3.6 asi¾ atk mixers
[00150] in. this representative experiment, the effect of MAb and salt concentration on the minimum time and pH require for vi us tnactivation. is investigated. MAb is prepared to 21 g/L -using 20 mM acetic acid; pH 3,2 and then titrated to pH 5.0 using 1 M NaOH, The conductivity of that solution is 1 .4 mS/cm. The lower concentrations are made by diluting -with 20 mM acetic acid, pH 3.2 and then titrated to pH 5 using 10 M NaOH. The conductivity of these solutions is 1 .4 mS/cm, The high salt solution s adjusted by NaCl addition to a final molarity of 250.mM. All solutions are adjusted to pH 3.6 for virus ί motivation using the inline static mixer setup, The results are summarized in Table VIL
Table VH
[IgCi] g/L fNaC!J aiM time (sec) LRV
4.4 0 20 2.8
4.4 0 40 3,5
4,4 0 60 3.5
4.4 250 20 3.9
4,4 250 40 4
4.4 250 60 4
21.8 0 20 .1.5
21.8 0 40 >4.3
21.8 0 60 >4.3
[00.151 J As summarized in Table VH, the most relevant solution is the one with a high concentration of MAb and the low salt concentration, which mimics a Protein. A elation pool. For such ¾ sample, 40 seconds are found to be sufficient for complete virus inactivatioo at a pi! of 3.6. Example 7.
Figure imgf000042_0001
[00152] In this representative: experiment, the effect of H on virus hiactivaii n. using static mixers is investigated. IgG is prepared to 9 g/L using 20 mM acetic acid, pH 3.2 nd then titrated to pB 5,0 using 10 M. NaO , All solutions are thea adjusted to the desired pH for virus inaetivaison using the in-line static raixer setup. The results are summarized in Table VI 11 below.
Table VIII
tiiae (sec) LRV (§ pH 3.4 LRV . pli 3.5 LRY (a), 3.6
20 3,2 3.1 3.1
40 >4 >4 3,2
60 4 > 4
[00153] As shown in Table VII, about one minute is required for complete virus inactivaiion at pH 3.6, whereas even shorter times are adequate at tower pH.
Figure imgf000042_0002
[00154] !n this representative experiment,, the effect of temperature on virus inaetivafion is investigated. Generally, in-line virus Inaetlvation is particularly conducive to exposure of the solution to higher temperatures; therefore, it is important to determine the effect of temperature o virus inactivaiion.
[0 155] The teed used in this study Is 9. g/L Seracare polyclonal IgG in. 20 mM sodium acetate, pH 5,0, Three aliquots of 25 mL each are transferred into three separate 50 nil., centrifuge tubes. To 25 mi, of thi s feed, 1,4 mi, of 3 M. acetic acid, pH 2,5 is added to bring down the pH to pH 3.?, One of the centrifuge tubes is left at room temp at 22 C, The other two centrifuge tubes are then placed in wate baths set a 10°C and 35° Upon equilibration, the temperature and pH of the liquid in the tubes is measured (see. Table IX below), A 10% virus spike i subsequently added to each of the tubes. 5 ml, samples, are collected at the stipulated time points and 0.4 nil, of 2 rn tris-base, pB 1 1 . is added to increase the phi to pfl 7,0, Table IX
ime H 3.6 (¾ 12¾C pH 3.67 (¾ 22°C ≠i 3.78 (§. 33¾€ (mm)
4. J
>4.1 > .U >4. i >4.G
>4. 4J ,0
[00156] The results smmn&rized in. Table IX indicate that, high temperature is preferred for virus Inactiva ion. It is surprising that even with a higher measured pit, the higher temperature achieves complete maeiivatiors at the I minute time point.
Example 9, Il¾ ,jf s tat ic mixers to ¾ect;i<? ¾ ie pfj gt¾t hi ¾fee¾tie»
fOOI 57] in this representative experiment, the use of static mi ers to accelerate pH stabilization was investigated. In general, it is desirable that the time needed for the phi to reach Its desired value is mmimized,
[00158] In this example, experiments ate done at various flow rates arid tube lengths and diameters, all selected so that only laminar flow is achieved, i.e. with Reynolds number below 100, The starting and target desired pH values are 5.0 ·ι· 0.1. a d 3.3 ±· 0.1, respectively.
[001 59] The time seeded to reach the desired target pf-l value is recorded as a function of time or solution volume processed. The volume is expressed in ter ms of tube dead volumes and data, is collected at a flow rate of 10 mL/rnm. The results are summarized in Table X below.
Table X
Reynold* # # of dead volumes fdimeusinssless units)o static mixer 40-50
static mixer elements 0-100
static mixer elements 0 [00160] T he results shown in Table X. indicate thai pH stabilization occurs .faster when usin more static mixer elements, while still 'remaining well into he laminar flow.
Example
Figure imgf000044_0001
[00161 J In this epresentative experiment, a clarified cell culture of a MAb is subjected to Protein A chromatography. The Protein A eluafc is collected in ten separate tractions, spanning about three column volumes. Each fraction is separately adjusted to two pH values, 33 and 3.6, The amounts of acid needed m reduce the pH to the desired value and of base needed to then increase the pH to the desired value are determined, as described herein, These amounts are different for each fraction because the different amounts of MAb result ia different buffering capacity for each sample,
[00162] A model is then bu lt of the amounts of acid, and base as a function of eluate volume. A second experiment is performed where the Protein A elaate is continuously -adjusted to the desireti pH based on the model, built. Virus
concentration is measured following virus inaetivatiou to confirm that vims motivation is achieved.
Exam le I Ϊ. ISffec M in-line vims in activate on .M Ab j¾maJhy
[00163] In. this representative experiment, the beneficial effect of shorter exposure time on MAb product quality is investigated. Two monoclonal antibodies are purified using Protein A chromatography, samples are collected in test tabes and immediately incubated at various pH (i.e., pH 3. 3.3. 3.6 and 4) and times (he., L 2, 5 and. 15 and 90 minutes),
[00164] The samples arc then neutralized and tested for presence of aggregates by Size Exclusion Chromatography (SEC) and SDS gels and lor changes in charge variants by weak cation exc hange chromatography (WCX-10). The identi ty of the resulting protein is also analyzed by Liquid Chromatography / Mass Spectrometry CLC/MS), .3 [00165J The specification is most thoroughl understood in light of the teachings of the references cited within the specification which are hereby incorporated by reference-. The embodiments within the specification provide m illustration of embodiments In this invention and should not be construed to limit its scope. The skilled artisan readily recognises that many other embodiments are encompassed by this invention. Ail publications and Inventions are incorporated by reference in their entirety. To the extent that the material incorporated by reference contradicts or is inconsistent with the present specification, the present specification will supercede any such materia!. The citation of any references herein is not an admission, that such references arc prior art to the present invention.
[00166] Unless otherwise indicated, all numbers expressing quantities of ingredients, cell culture, treatment conditions, and so forth used in the specification, including claims, arc to be understood as being modified in ail instances by the term "about." Accordingly,, unless otherwise .indicated to the contrary., the numerical parameters are approximations and may vary depending upon the desired properties sought to be obtained by the present invention. Unless otherwise indicated, the term " t !eas * preceding a series of elements is to be understood to refer to every element in the series. Those skilled in. the art will, recognize, or be able to ascertain, using no more than routine experimentation, man equivalents to the specific embodiments of the invention described herein. Such equivalents are Intended to be encompassed b the following claims.
("001.67] Many modifications and variations of this invention can. be made without departing from its spirit and scope, as will be apparent to those skilled In the art. The specific embodiments described herein are offered by way of example only and are not meant to be limiting in any way. It Is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being Indicated by the following claims.

Claims

Claims
What is ektaed is:
1 , A method for inactivating one or more i uses in a sam le, wherein the method comprises continuously mixing the sam le with one or more virus inactivating agents as the sample flows from the first unit operation to the second unit operation during a process for purifying a target molecule.
2, 'The method of claim 1 , wherein the first unit operation comprises bind and eiute chromatography and the second unit operation comprises a flow-through purification process.
3, The method of claim 2, wherein the hind and eiute chromatography comprises Protein A affinity chromatography,
4, The method of claim 2, wherein the flow-through purification process comprises two or more matrices selected from the group consisting of activated carbon, anion exchange chromatography media, cation exchange chromatography media and a virus filtration media,
5, The method of claim L wherein the sample comprises a Protein A eluate.
6, The method of claim .1 , wherein the target molecule Is a antibody or an F e region containing protein,
7, 'The method of claim L wherein the sample is mixed with one or more virus inactivating agents using one or more in-line static mixers.
8, The method of claim 1 , wherein the sample is mixed with one or more vi us inactivating agents using one or more surge tanks.
9, The method of claim 1 , wherein the one or more virus inactivating agents are selected from the group consisting (if an acid, a salt, a solvent and a detergent.
10. The method of claim. ?, wherein the flow of sample is in the laminar flow range.
1 1. A method of inactivating one or more viruses in a Protein Λ efuatc the method comprising mixing the eluate with one or more virus inactivating agent?; using one or more in-line static mixers, wherein complete virus inaetivatioo is achieved n less than 10 minutes or less than 5 minutes or less than 3 minutes or less than 1 minute.
12. A method of inactivating one or more viruses in a Protei A eluate, the method comprising mixing the eluate with one or more virus inactivating agents using a surge tank* wherein complete virus Inactivation is achieved in less than 1 hour or less than 30 minutes,
13. A method for inactivating one or more viruses, the method comprising:
(a) subjecting a. sample comprising a target protein to a Protein A affinity chromatography process, thereby to obtain an eluate; and
(b) continuously transferring the eluate to an in-line static mixer to mix one or more virus inactivating agents with the eluate for a duration of time which is equal to or less than 10 minutes,
thereby to inactivate one or more viruses.
14. The method of claim 13, wherein the Protein A affinity chromatography process is performed in batch mode.
15. The method of claim 13, wherein the Protein A affinity chromatography process is performed in a continuous mode.
16. The method of claim 1 , wherein the process comprises continuous multi- column chromatography.
17. The method of claim 13, wherein the one or more virus inactivating agents is an acid. 18, The method of claim 13 , wherein the target protein is an antibody.
19, The method of claim 13, further comprising the step of continuously transferring output from step (b) into a flow-through purification process step.
20, The method of claim 19, wherein the flow-through purification step composes two or .more matrices selected from, activated carbon, anion exchange media, cation exchange media and virus fil er.
PCT/US2013/045677 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process WO2014004103A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP13810248.8A EP2867359B1 (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process
EP21191783.6A EP3981873A1 (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process
US14/390,924 US9809799B2 (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process
SG11201406407TA SG11201406407TA (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process
ES13810248T ES2895173T3 (en) 2012-06-29 2013-06-13 Methods to inactivate viruses during a protein purification process
CN201380034780.5A CN104411820B (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process
JP2015520259A JP6182210B2 (en) 2012-06-29 2013-06-13 Method for virus inactivation during protein purification process
IN8526DEN2014 IN2014DN08526A (en) 2012-06-29 2014-10-13

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261666145P 2012-06-29 2012-06-29
US61/666,145 2012-06-29

Publications (1)

Publication Number Publication Date
WO2014004103A1 true WO2014004103A1 (en) 2014-01-03

Family

ID=49783749

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/045677 WO2014004103A1 (en) 2012-06-29 2013-06-13 Methods for inactivating viruses during a protein purification process

Country Status (8)

Country Link
US (1) US9809799B2 (en)
EP (2) EP3981873A1 (en)
JP (2) JP6182210B2 (en)
CN (2) CN107188926B (en)
ES (1) ES2895173T3 (en)
IN (1) IN2014DN08526A (en)
SG (1) SG11201406407TA (en)
WO (1) WO2014004103A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014137903A3 (en) * 2013-03-08 2014-10-30 Genzyme Corporation Continuous purification of therapeutic proteins
WO2015158776A1 (en) 2014-04-15 2015-10-22 Boehringer Ingelheim International Gmbh Methods, apparatuses, and systems for continuously inactivating a virus during manufacture of a biological product
WO2016059925A1 (en) * 2014-10-14 2016-04-21 東洋エンジニアリング株式会社 Virus inactivation and sampling device
CN105841984A (en) * 2016-03-18 2016-08-10 中国石油天然气股份有限公司 Polymer injection allocation technology equipment detection evaluation apparatus
WO2016207328A1 (en) * 2015-06-24 2016-12-29 Glycotope Gmbh PROCESS FOR THE PURIFICATION OF γ-CARBOXYLATED POLYPEPTIDES
US9630165B2 (en) 2014-01-17 2017-04-25 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US10087214B2 (en) 2014-01-17 2018-10-02 Genzyme Corporation Sterile chromatography and manufacturing processes
WO2020153390A1 (en) * 2019-01-23 2020-07-30 第一三共株式会社 Method for antibody purification including step where activated carbon material is used
US10865224B2 (en) 2012-06-29 2020-12-15 Emd Millipore Corporation Purification of biological molecules
WO2021257387A1 (en) 2020-06-16 2021-12-23 Boehringer Ingelheim International Gmbh A method of providing a homogeneous feed stream within a plug flow reactor
US11369703B2 (en) 2018-08-31 2022-06-28 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US11547957B2 (en) 2017-05-11 2023-01-10 Emd Millipore Corporation Method of maintaining narrow residence time distributions in continuous flow systems
WO2023059800A3 (en) * 2021-10-07 2023-06-01 Regeneron Pharmaceuticals, Inc. Systems and methods of ph modeling and control
EP4001291A4 (en) * 2019-07-18 2023-07-26 Abion Inc. Method for purifying diglycosylated interferon-beta protein
US11732003B2 (en) 2017-05-11 2023-08-22 Emd Millipore Corporation Mechanical method of maintaining narrow residence time distributions in continuous flow systems

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3277397A1 (en) * 2015-03-31 2018-02-07 GE Healthcare Bio-Sciences AB A liquid chromatograhy system, a device, and a method
JP7032046B2 (en) * 2016-01-22 2022-03-08 旭化成メディカル株式会社 Continuous constant flow rate purification method for bioactive substances
EP3426683A1 (en) * 2016-03-11 2019-01-16 Boehringer Ingelheim International GmbH Methods for continuously inactivating a virus during manufacture of a protein
US11155794B2 (en) * 2016-08-23 2021-10-26 Alexion Pharmaceuticals, Inc. Method of purifying a heterologous protein from an egg white
JP6596043B2 (en) 2017-07-31 2019-10-23 株式会社Subaru Vehicle travel control system
GB201802593D0 (en) * 2018-02-16 2018-04-04 Fujifilm Diosynth Biotechnologies Uk Ltd Rig
SG11202103790WA (en) * 2018-11-15 2021-05-28 Bayer Healthcare Llc Viral inactivation methods for continuous manufacturing of antibodies
WO2020117760A1 (en) * 2018-12-03 2020-06-11 Duke University Purification method for recombinant proteins and nanoparticles
FR3099066B1 (en) * 2019-07-26 2021-08-13 Novasep Process Purification process of a target substance with viral inactivation
WO2021197249A1 (en) * 2020-03-30 2021-10-07 上海复宏汉霖生物技术股份有限公司 System and method for continuous purification of target molecules
EP4357009A1 (en) 2022-10-17 2024-04-24 Sartorius Stedim Biotech GmbH Method and apparatus for operating a continuous downstream process

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6358534B1 (en) * 1997-04-08 2002-03-19 Baxter Aktiengesellschaft Immunotolerant prothrombin complex preparation
US6468778B1 (en) * 1999-05-19 2002-10-22 Aventis Behring Gmbh Process for the inactivation of viruses
WO2002092806A1 (en) 2001-05-15 2002-11-21 V.I. Technologies, Inc. Apparatus for the inactivation of pathogens in protein-containingfluids
US20020177693A1 (en) * 1997-06-20 2002-11-28 Wytold Lebing Chromatographic method for high yield purification and viral inactivation of antibodies
US20040131497A1 (en) 2002-09-19 2004-07-08 Thomas Lengsfeld Process for sterilization of biological compositions containg protein
WO2004058046A2 (en) 2002-12-20 2004-07-15 Baxter International Inc. Fluid treatment method, apparatus and system
WO2009138484A2 (en) * 2008-05-15 2009-11-19 Novo Nordisk A/S Antibody purification process
US20100135987A1 (en) * 2008-10-20 2010-06-03 Hickman Robert K Isolation and purification of antibodies using protein a affinity chromatography
US20100221844A1 (en) 2008-12-24 2010-09-02 Millipore Corporation Caustic stable chromatography ligands
WO2012051147A1 (en) * 2010-10-11 2012-04-19 Abbott Laboratories Processes for purification of proteins
US20130046056A1 (en) 2011-06-08 2013-02-21 Emd Millipore Corporation Chromatography matrices including novel staphylococcus aureus protein a based ligands

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4639513A (en) * 1984-02-02 1987-01-27 Cuno Inc. Intravenously injectable immunoglobulin G (IGG) and method for producing same
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0629135B1 (en) 1992-03-02 2000-07-26 Bioeng, Inc. Viral inactivation method
JPH09500015A (en) 1993-06-23 1997-01-07 ニューヨーク ブラッド センター,インコーポレイティド System for virus inactivation of blood
WO1996015231A2 (en) 1994-11-10 1996-05-23 Immuno Aktiengesellschaft Method for producing biologicals in protein-free culture
US6686191B1 (en) 1995-09-22 2004-02-03 Bayer Healthcare Llc Preparation of virally inactivated intravenously injectable immune serum globulin
GB9611698D0 (en) 1996-06-05 1996-08-07 Iatros Ltd Fluid processing
US20040256329A1 (en) 2003-06-13 2004-12-23 Peter Meserol Photodynamic biological fluid pathogen inactivation/filtering apparatus and method
US8271139B2 (en) 2003-10-17 2012-09-18 Asahi Kasei Bioprocess, Inc. Multi-stage accurate blending system and method
WO2005090403A2 (en) 2004-03-12 2005-09-29 Biovest International, Inc. Method and apparatus for antibody purification
CA2604877A1 (en) 2005-04-11 2006-10-19 J.P. Sercel Associates Inc. Protein purification
WO2009045897A1 (en) 2007-10-03 2009-04-09 Dyax Corp. Systems and methods for purifying proteins
AU2009307737B2 (en) * 2008-10-20 2015-07-23 Abbvie Inc. Viral inactivation during purification of antibodies
WO2011028753A1 (en) 2009-09-01 2011-03-10 Genentech, Inc. Enhanced protein purification through a modified protein a elution
US9527010B2 (en) * 2009-09-25 2016-12-27 Ge Healthcare Bio-Sciences Corp. Separation system and method
WO2012014183A1 (en) * 2010-07-30 2012-02-02 Pfizer Inc. Tandem purification of proteins
CN103562145B (en) * 2010-12-06 2016-09-21 颇尔公司 The continuous process of biological product
KR102157227B1 (en) * 2011-07-08 2020-09-17 이엠디 밀리포어 코포레이션 Improved depth filters for disposable biotechnological processes
US9096648B2 (en) 2011-08-19 2015-08-04 Emd Millipore Corporation Methods of reducing level of one or more impurities in a sample during protein purification
CN102492040B (en) * 2011-12-29 2014-06-25 嘉和生物药业有限公司 Method for purifying anti-HER2 or/and anti-HER3 antibody proteins
EP2682168A1 (en) 2012-07-02 2014-01-08 Millipore Corporation Purification of biological molecules
MX2015012114A (en) * 2013-03-08 2016-01-12 Genzyme Corp Continuous purification of therapeutic proteins.

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6358534B1 (en) * 1997-04-08 2002-03-19 Baxter Aktiengesellschaft Immunotolerant prothrombin complex preparation
US20020177693A1 (en) * 1997-06-20 2002-11-28 Wytold Lebing Chromatographic method for high yield purification and viral inactivation of antibodies
US6468778B1 (en) * 1999-05-19 2002-10-22 Aventis Behring Gmbh Process for the inactivation of viruses
WO2002092806A1 (en) 2001-05-15 2002-11-21 V.I. Technologies, Inc. Apparatus for the inactivation of pathogens in protein-containingfluids
US20040131497A1 (en) 2002-09-19 2004-07-08 Thomas Lengsfeld Process for sterilization of biological compositions containg protein
WO2004058046A2 (en) 2002-12-20 2004-07-15 Baxter International Inc. Fluid treatment method, apparatus and system
WO2009138484A2 (en) * 2008-05-15 2009-11-19 Novo Nordisk A/S Antibody purification process
US20100135987A1 (en) * 2008-10-20 2010-06-03 Hickman Robert K Isolation and purification of antibodies using protein a affinity chromatography
US20100221844A1 (en) 2008-12-24 2010-09-02 Millipore Corporation Caustic stable chromatography ligands
WO2012051147A1 (en) * 2010-10-11 2012-04-19 Abbott Laboratories Processes for purification of proteins
US20130046056A1 (en) 2011-06-08 2013-02-21 Emd Millipore Corporation Chromatography matrices including novel staphylococcus aureus protein a based ligands

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
BUFFERS: "A Guide for the Preparation and Use of Buffers in Biological Systems", 1975, CALBIOCHEM CORPORATION
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
GAIL SOFER: "Virus Inactivation in the 1990s — and into the 21st Century, Part 4, Culture Media, Biotechnology Products, and Vaccines", BIOPHARM INTERNATIONAL, January 2003 (2003-01-01), pages 50 - 57, XP009171415
HARRISON ET AL.: "Stability of a single-chain Fv antibody fragment when exposed to a high shear environment combined with air-liquid interfaces", BIOTECHNOL. BIOENG., vol. 59, 1998, pages 517 - 519
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "equences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KURT BRORSON ET AL.: "Bracketed Generic Inactivation of Rodent Retroviruses by Low pH Treatment for Monoclonal Antibodies and Recombinant Proteins", BIOTECH.BIOENG, vol. 82, no. 3, 2003, XP002576168, DOI: doi:10.1002/bit.10574
MAHLER ET AL.: "Protein aggregation: Pathways, induction factors and analysis", J. PHARM. SCI, vol. 98, no. 9, 2009, pages 2909 - 2934, XP055321457, DOI: doi:10.1002/jps.21566
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MIESEGAES ET AL.: "Analysis of viral clearance unit operations for monoclonal antibodies", BIOTECHNOLOGY AND BIOENGINEERING, vol. 106, 2010, pages 238 - 246, XP055146691, DOI: doi:10.1002/bit.22662
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
SHUKLA ET AL., J. CHROMATOGRAPHY B., vol. 848, 2007, pages 28 - 39
VAZQUEZ-REY ET AL.: "Aggregates in monoclonal antibody manufacturing processes", BIOTECHNOLOGY AND BIOENGINEERING, vol. 108, no. 7, 2011, pages 1494 - 1508, XP055365984, DOI: doi:10.1002/bit.23155
WANG ET AL.: "Antibody structure, instability and formulation", J. PHARM. SCI., vol. 96, 2007, pages 1 - 26, XP009084505, DOI: doi:10.1002/jps.20727

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10865224B2 (en) 2012-06-29 2020-12-15 Emd Millipore Corporation Purification of biological molecules
AU2014226126B2 (en) * 2013-03-08 2019-02-14 Genzyme Corporation Continuous purification of therapeutic proteins
WO2014137903A3 (en) * 2013-03-08 2014-10-30 Genzyme Corporation Continuous purification of therapeutic proteins
US10711034B2 (en) 2013-03-08 2020-07-14 Genzyme Corporation Integrated continuous manufacturing of therapeutic protein drug substances
US9650412B2 (en) 2013-03-08 2017-05-16 Genzyme Corporation Integrated continuous manufacturing of therapeutic protein drug substances
US9650413B2 (en) 2013-03-08 2017-05-16 Genzyme Corporation Integrated continuous manufacturing of therapeutic protein drug substances
US9657056B2 (en) 2013-03-08 2017-05-23 Genzyme Corporation Integrated continuous manufacturing of therapeutic protein drug substances
US11912739B2 (en) 2014-01-17 2024-02-27 Genzyme Corporation Sterile chromatography and manufacturing processes
US11839861B2 (en) 2014-01-17 2023-12-12 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US9630165B2 (en) 2014-01-17 2017-04-25 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US10071364B2 (en) 2014-01-17 2018-09-11 Genzyme Corporation Sterile affinity chromatography resin
US10087214B2 (en) 2014-01-17 2018-10-02 Genzyme Corporation Sterile chromatography and manufacturing processes
US10919021B2 (en) 2014-01-17 2021-02-16 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
CN114225072B (en) * 2014-04-15 2024-01-02 勃林格殷格翰国际公司 Method, apparatus and system for continuous inactivation of viruses during manufacture of biological products
CN114225072A (en) * 2014-04-15 2022-03-25 勃林格殷格翰国际公司 Method, apparatus and system for continuous inactivation of viruses during manufacture of biological products
EP4234569A3 (en) * 2014-04-15 2023-10-18 Boehringer Ingelheim International GmbH Method and use for continuously inactivating a virus during manufacture of a biological product
EP4234569A2 (en) 2014-04-15 2023-08-30 Boehringer Ingelheim International GmbH Method and use for continuously inactivating a virus during manufacture of a biological product
CN111632167A (en) * 2014-04-15 2020-09-08 勃林格殷格翰国际公司 Method, apparatus and system for continuous inactivation of viruses during manufacture of biological products
WO2015158776A1 (en) 2014-04-15 2015-10-22 Boehringer Ingelheim International Gmbh Methods, apparatuses, and systems for continuously inactivating a virus during manufacture of a biological product
US11725191B2 (en) 2014-04-15 2023-08-15 Boehringer Ingelheim International Gmbh Methods, apparatuses, and systems for continuously inactivating a virus during manufacture of a biological product
US10435670B2 (en) 2014-04-15 2019-10-08 Boehringer Ingelheim International Gmbh Methods, apparatuses, and systems for continuously inactivating a virus during manufacture of a biological product
CN111632167B (en) * 2014-04-15 2021-12-28 勃林格殷格翰国际公司 Method, apparatus and system for continuous inactivation of viruses during manufacture of biological products
WO2016059925A1 (en) * 2014-10-14 2016-04-21 東洋エンジニアリング株式会社 Virus inactivation and sampling device
JP2016077190A (en) * 2014-10-14 2016-05-16 東洋エンジニアリング株式会社 Virus inactivating and sampling device
US10391187B2 (en) 2014-10-14 2019-08-27 Tec Project Services Corporation Apparatus for virus inactivation and sampling
WO2016207328A1 (en) * 2015-06-24 2016-12-29 Glycotope Gmbh PROCESS FOR THE PURIFICATION OF γ-CARBOXYLATED POLYPEPTIDES
CN105841984A (en) * 2016-03-18 2016-08-10 中国石油天然气股份有限公司 Polymer injection allocation technology equipment detection evaluation apparatus
US11547957B2 (en) 2017-05-11 2023-01-10 Emd Millipore Corporation Method of maintaining narrow residence time distributions in continuous flow systems
US11732003B2 (en) 2017-05-11 2023-08-22 Emd Millipore Corporation Mechanical method of maintaining narrow residence time distributions in continuous flow systems
US11369703B2 (en) 2018-08-31 2022-06-28 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
WO2020153390A1 (en) * 2019-01-23 2020-07-30 第一三共株式会社 Method for antibody purification including step where activated carbon material is used
EP4001291A4 (en) * 2019-07-18 2023-07-26 Abion Inc. Method for purifying diglycosylated interferon-beta protein
WO2021257387A1 (en) 2020-06-16 2021-12-23 Boehringer Ingelheim International Gmbh A method of providing a homogeneous feed stream within a plug flow reactor
WO2023059800A3 (en) * 2021-10-07 2023-06-01 Regeneron Pharmaceuticals, Inc. Systems and methods of ph modeling and control

Also Published As

Publication number Publication date
JP6182210B2 (en) 2017-08-16
EP2867359A4 (en) 2015-11-11
EP3981873A1 (en) 2022-04-13
JP2017148051A (en) 2017-08-31
JP6457575B2 (en) 2019-01-23
CN107188926A (en) 2017-09-22
IN2014DN08526A (en) 2015-05-15
CN104411820B (en) 2017-05-03
CN104411820A (en) 2015-03-11
EP2867359B1 (en) 2021-09-15
US9809799B2 (en) 2017-11-07
CN107188926B (en) 2021-02-09
ES2895173T3 (en) 2022-02-17
US20150064769A1 (en) 2015-03-05
SG11201406407TA (en) 2014-11-27
JP2015522017A (en) 2015-08-03
EP2867359A1 (en) 2015-05-06

Similar Documents

Publication Publication Date Title
EP2867359A1 (en) Methods for inactivating viruses during a protein purification process
Marichal‐Gallardo et al. State‐of‐the‐art in downstream processing of monoclonal antibodies: Process trends in design and validation
JP6471183B2 (en) Purification of biomolecules
TWI675041B (en) Continuous multistep process for purifying antibodies
EP3041857B1 (en) Protein a chromatography
CA2830640A1 (en) Novel protein purification methods
JP2019525925A (en) Affinity chromatography wash buffer
JP2018524146A (en) A method for continuous elution of products from a chromatography column
TW201444863A (en) A method for increasing pyro-glutamic acid formation of a protein
WO2015118149A1 (en) System, apparatus and method for anti-rsv antibodies and formulations
WO2013063298A1 (en) Methods of reducing or eliminating protein modification and degradation arising from exposure to uv light
JP2021528957A (en) Method of producing anti-C5 antibody
JP2014527528A (en) Use of small molecules for purification methods of biomolecules
JP2022522966A (en) How to move a batch production process to a continuous production process
TW201831889A (en) Method for sampling fluid streams for monitoring contaminants in a continuous flow
WO2022203044A1 (en) Viral clearance test method
JP2011041475A (en) Antibody production method
Saab Investigation the Plugging Behavior of Virus filters
TW201945385A (en) Full flow-through process for purifying recombinant proteins
WO2021072210A1 (en) Methods of purifying ranibizumab or a ranibizumab variant

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13810248

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14390924

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2013810248

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015520259

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE