WO2023177904A1 - Sterile filtration of lipid nanoparticles and filtration analysis thereof for biological applications - Google Patents

Sterile filtration of lipid nanoparticles and filtration analysis thereof for biological applications Download PDF

Info

Publication number
WO2023177904A1
WO2023177904A1 PCT/US2023/015564 US2023015564W WO2023177904A1 WO 2023177904 A1 WO2023177904 A1 WO 2023177904A1 US 2023015564 W US2023015564 W US 2023015564W WO 2023177904 A1 WO2023177904 A1 WO 2023177904A1
Authority
WO
WIPO (PCT)
Prior art keywords
filtration
alkyl
approximately
lipid
lnps
Prior art date
Application number
PCT/US2023/015564
Other languages
French (fr)
Inventor
Jack F. Kramarczyk
Anton ZVEREV
Andrew Lawrence ZYDNEY
Original Assignee
Modernatx, Inc.
The Penn State Research Foundation
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
Application filed by Modernatx, Inc., The Penn State Research Foundation filed Critical Modernatx, Inc.
Publication of WO2023177904A1 publication Critical patent/WO2023177904A1/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D65/00Accessories or auxiliary operations, in general, for separation processes or apparatus using semi-permeable membranes
    • B01D65/10Testing of membranes or membrane apparatus; Detecting or repairing leaks
    • B01D65/109Testing of membrane fouling or clogging, e.g. amount or affinity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle

Definitions

  • LNPs Lipid nanoparticles
  • mRNA-based vaccines and therapeutics e.g., mRNA-encoded immunogens, e.g., after intramuscular injection.
  • mRNA is bound by an ionizable lipid that occupies part of the LNPs.
  • SUMMARY [0004] The present disclosure addresses filtration of solutions containing LNPs, and in particular, presents improved methods of filtration of LNPs.
  • Filtration e.g., sterile filtration
  • the present disclosure further relates to filter performance during filtering of LNPs.
  • the present disclosure relates to a method of evaluating sterile filtration of LNPs. The method includes filtering a first solution lacking LNPs through at least one membrane while controlling a filtration control parameter to satisfy a control parameter threshold; and determining a first hydraulic resistance of the membrane following filtration of the first solution.
  • the method further includes filtering a second solution containing LNPs through the at least one membrane while controlling the filtration control parameter to satisfy the control parameter threshold; and determining a second hydraulic resistance of the membrane following filtration of the second solution. Additionally, the method further includes computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; imaging the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling. [0006] In at least one aspect, the present disclosure relates to a method of filtering a solution containing lipid nanoparticles.
  • the method includes setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold.
  • FIG.1 is a schematic illustration of a lipid nanoparticle, according to an aspect of the present disclosure.
  • FIGS.2A and 2B are images of a first layer of filter media taken by a scanning electron microscope (SEM), according to an aspect of the present disclosure.
  • FIG.3 is an SEM image of a second layer of filter media, according to an aspect of the present disclosure.
  • FIG.4 is a plot of filtrate flux at different transmembrane pressures (TMPs) for exemplary filter media, of which FIGS.2A-2B depict a first layer thereof and FIG.3 depicts a second layer thereof, according to an aspect of the present disclosure.
  • TMPs transmembrane pressures
  • FIG.5 is a plot of filtration resistance at different TMPs for the filter media of FIGS.2A-2B and 3, according to an aspect of the present disclosure.
  • FIG.6 is an SEM image of a first layer of filter media in an unused condition, according to an aspect of the present disclosure.
  • FIG.7 is an SEM image of the first layer of the filter media of FIG.6 in a used condition, according to an aspect of the present disclosure.
  • FIG.8 is an SEM image of a second layer of filter media in an unused condition, according to an aspect of the present disclosure.
  • FIG.9 is an SEM image of the second layer of the filter media of FIG.8 in a used condition, according to an aspect of the present disclosure.
  • FIG.10 is a plot of filtrate flux for different filter media formulations, according to an aspect of the present disclosure.
  • FIG.11 is a plot relating to a derivative analysis based on the test data from FIG. 4-5, according to an aspect of the present disclosure.
  • FIG.12 is a plot of filtrate flux as observed through multiple filters of a two-stage filtering operation, according to an aspect of the present disclosure.
  • FIG.13 is a plot of filtrate flux data from a pressure stepping experiment, according to an aspect of the present disclosure.
  • FIG.14 is a plot of filtrate flux data from a pressure stepping experiment, according to an aspect of the present disclosure.
  • FIG.15 is a plot of filtrate flux data from a pressure stepping experiment using two different feed solutions, according to an aspect of the present disclosure.
  • FIG.16 is a plot of resistance over filtration time from a first pressure stepping experiment, according to an aspect of the present disclosure.
  • FIG.17 is a plot of resistance over filtration time from a second pressure stepping experiment, according to an aspect of the present disclosure.
  • FIG.18 is a plot of resistance over filtration time from a third pressure stepping experiment, according to an aspect of the present disclosure.
  • FIG.19 is an exemplary depiction of a filtration system according to various aspects of the present disclosure.
  • FIG.20A and FIG.20B are plots of resistance versus filtration time during pressure stepping, according to aspects of the present disclosure.
  • FIG.21 is a plot of resistance versus transmembrane pressure for LNPs as deposited on a membrane, according to an aspect of the present disclosure.
  • FIGS.22A-22D are plots of resistance versus transmembrane pressure for various membranes, according to certain aspects of the present disclosure.
  • FIG.23 is a plot of operating pressure versus reciprocal pore diameter, according to an aspect of the present disclosure.
  • FIGS.24A-24F are scanning electron microscopy and environmental scanning electron microscopy images of clean and fouled membranes, according to aspects of the present disclosure.
  • FIG.25A is an atomic force microscopy image of a partially fouled membrane
  • FIG.25B depicts deposit height of the deposited LNPs in FIG.25A as a function of surface distance between two points shown in FIG.25A, according to aspects of the present disclosure.
  • FIG.26 is a high resolution SEM image of deposited LNPs on a membrane, according to an aspect of the present disclosure.
  • FIGS.27A-27F are schematic diagrams of physical mechanisms affecting behavior of deposited LNPs.
  • LNPs The large size and unique behavior of LNPs affects filtration performance.
  • a relatively higher lipid content may cause some solutions containing LNPs to exhibit particular behavior during filtering, such as patterns of aggregation or deposition on filter media, which differ from solutions without LNPs.
  • filter capacities and LNP yields are generally both smaller than those for certain existing biotherapeutics. Lower capacity and significant yield loss can occur during sterile filtration of vaccines, viruses, and gene therapies due to the very similar size of these biotherapeutics and the pore size of sterilizing grade filters through which they are passed to ensure sterility of the final product.
  • the observed capacity and yield may be attributable to both the relatively large size of the LNPs and to unexpected behavior as discussed herein.
  • a method of determining the pressure-dependent behavior in sterile filtration of an LNP solution includes (i) imaging clean or un-fouled filter media in addition to used or fouled filter media, (ii) evaluating filtration performance of individual filter media layers, and (iii) analyzing performance at different transmembrane pressures (TMPs).
  • the filter media may be implemented as a membrane or a membrane assembly, for example.
  • the analysis may be conducted using experimental data obtained where control parameters, including but not limited to TMP, are adjusted.
  • the foregoing method further can include performing re-filtration to assess an extent to which foulants are retained and/or removed by the filter media.
  • the method further can include performing pressure stepping to assess the influence of LNP deposition at different pressures and with different feed solution compositions.
  • At least one aspect of the present disclosure provides for the observation and characterization of the sterile filtration performance of LNP filtration as a function of TMP for a dual layer media assembly. In contrast to typical behavior associated with compressible fouling deposits, experimental data obtained during testing indicates an increase in filtrate flux and filter media capacity with increasing TMP.
  • kits including LNPs in solution as described herein, a membrane, and a controller according to various examples below.
  • LNPs in solution as described herein, a membrane, and a controller according to various examples below.
  • FIG.1 shows a schematic illustration of a lipid nanoparticle, shown as LNP 10, for use in a biological application such as a pharmaceutical drug delivery system.
  • the LNP 10 includes four lipid components including distearolyphosphatidycholine (DSPC) 12, a cholesterol 14, a polyethylene glycol (PEG)-lipid 16, and an ionizable lipid (shown as charged ionizable lipid 18 and neutral ionizable lipid 20).
  • DSPC 12 is configured to provide a general support structure (e.g., shield, support, etc.) for the LNP 10.
  • the cholesterol 14 is configured as a stabilizing element to aid in transfection of the mRNA into cells (e.g., introduction of the mRNA into cells by non-viral methods).
  • the PEG-lipid 16 is configured to provide a stealth effect (e.g., reduce nonspecific binding) and/or as a stabilizing element in the LNP 10.
  • the ionizable lipids e.g., the charged ionizable lipid 18, the neutral ionizable lipid 20
  • the LNP 10 can be substantially spherical in shape.
  • the LNP 10 has an average particle size (e.g., diameter, etc.) greater than approximately 100 nm. In some embodiments, the average particle size is between approximately 50 nm and approximately 200 nm.
  • the LNP 10 can be (i) substantially spherical or generally ellipsoidal in shape and (ii) have a diameter of approximately 50 nm to approximately 200 nm. In general, the LNP 10 can have diverse structural configurations. [0049] In the process of creating a vaccine or therapeutic product relying on an LNP delivery system, a solution containing LNP 10 is first filtered to remove or reduce the quantity of microorganisms (e.g., bacteria) and/or other contaminants present in the final product. Sterile filtration is a process of filtering a solution to remove such contaminants without adversely affecting the final product.
  • microorganisms e.g., bacteria
  • sterile filtration of LNP 10 may include passing a solution containing LNP 10 (e.g., LNP 10 and buffer, etc.) through filter media.
  • the filter media may include a single or multi-layer filter media (e.g., multi- membrane filter media, an assembly constructed of multiple filters, etc.).
  • the LNP 10 may also be referred to herein as mRNA-LNP 10.
  • the production of mRNA vaccines begins with the in vitro enzymatic synthesis of the mRNA from a DNA template.
  • the downstream purification process involves a series of chromatographic separations and tangential flow filtration.
  • the purified mRNA is encapsulated in LNPs by mixing the mRNA (in an acidic aqueous buffer) with the lipids (in ethanol).
  • the resulting LNP suspension can be buffer exchanged and concentrated by ultrafiltration or diafiltration followed by sterile filtration as part of the final fill-finish operation.
  • Sterile filtration is commonly performed using normal-flow (dead-end) filtration through membranes containing a 0.2 ⁇ m filter, which can retain microbes of approximately 107 colony forming units / cm 2 of filter area, for example.
  • LNPs Sterile filtration of LNPs, as well as other large viral and non-viral delivery vehicles, can be challenging because these products can be similar in size to the 0.2 ⁇ m size of certain sterilizing-grade filters, among other reasons.
  • pressure is applied to the feed solution upstream of the filter media to force the feed solution through the filter media.
  • the filter media captures contaminants along the surface or within pores of the filter media while allowing smaller particles to pass therethrough.
  • filtration continues until the filter media becomes clogged (e.g., fouled) and the filtrate flux across the filter media drops below a threshold value.
  • Typical biotherapeutics may exhibit any of the classical fouling models (complete pore blockage, intermediate pore blockage, pore constriction, or cake formation). Under each of these behavioral models, foulant is deposited onto the membrane. For typical biotherapeutics, the deposited foulant is typically either incompressible or compressible. “Incompressible” foulant is foulant for which a filtration resistance is always constant (that is, the flux varies linearly with TMP). “Compressible” foulant is foulant for which filtration resistance increases with higher TMP (as an example, the flux may vary less-than-linearly with TMP).
  • the fouling behavior in sterile filtration of an LNP solution may be evaluated using imaging and data analysis.
  • At least one exemplary method of performing filtration of an LNP-containing solution includes (i) imaging clean or un-fouled filter media in addition to used or fouled filter media, (ii) analyzing filtration performance (e.g., of individual filter media layers and/or of the overall system at different transmembrane pressures (TMPs)) and (iii) adjusting one or more filtration control parameters in responsive to at least one of the individual filter media layer performance or the TMP performance.
  • material may be accumulated for a content analysis to determine the correlation between the accumulated material and the feed stream.
  • the content analysis may be performed to assess the extent to which components and/or product-related impurities, such as cholesterol crystals, are present in the accumulated material.
  • one or more filtration control parameters may be adjusted utilizing a feedback loop based on at least one measured value (e.g., a measured pressure, a measured flow rate, a measured product quality, a measured yield, or any combination of any of the foregoing) and/or based on a calculation obtained from a fouling model that uses one or more of the measured values as an input.
  • the filtration control parameters may include one or more of a pressure, a filter load (e.g., in L/m 2 or g/m 2 ), a flow rate, a temperature, a pH, a salt type or concentration, and a filtration operational mode, and the concentration of additives.
  • an exemplary method for filtering LNP-containing solution may include controlling filtration based on either a specific value associated with a given parameter or a gradient of values.
  • one or more of the pH, the salt concentration, and the concentration of additives may be adjusted during filtering.
  • the filter media is selected based on an associated pressure or an associated range of pressures.
  • pressure may vary between approximately 0.5 psi to approximately 50 psi, or greater than approximately 0 psi to approximately 70 psi. In some embodiments, the pressure is approximately 10 psi, approximately 20 psi, approximately 30 psi, approximately 40 psi, approximately 50 psi, approximately 60 psi, approximately 70 psi or approximately 80 psi.
  • filter load may vary from approximately 10 L/m 2 to approximately 5,000 L/m 2 . In some embodiments, flow rate (flux) may vary from approximately 20 L/m 2 /hr to approximately 2,000 L/m 2 /hr.
  • the temperature may be between approximately 2 °C to approximately 40 °C.
  • the filtration may take place under refrigeration, e.g., between approximately 2 °C to approximately 8 °C.
  • the filtration may take place in ambient conditions of between approximately 15 °C to approximately 25 °C (e.g., room temperature).
  • the filtration may take place at elevated temperatures, e.g., between approximately 30 °C and approximately 40 °C or between approximately 40 °C and 50 °C.
  • the temperature may affect inter-particle attractive forces and may influence viscosity and filter performance.
  • elevated temperatures may be used to induce microbial cell death, thereby increasing efficiency and effectiveness of filtration.
  • the pH may vary between approximately 4 to approximately 8.
  • LNPs may experience greater charging when the pH is approximately 1 pH unit below the isoelectric point (the pH is approximately 6 to approximately 7 for various exemplary lipids). When charged, LNPs may be expected to repel each other, thereby contributing to decreased fouling.
  • a filtration process of filtering LNPs may include filtering the LNPs at a pH of approximately 5, where insignificant fouling is observed. In contrast, higher pH (e.g., above 7.5) may be associated with increased fouling.
  • a salt concentration may vary between approximately 10 M to approximately 1000 M. In some embodiments, a salt concentration may be increased so as to lower the repulsion between LNPs. A relatively lower salt concentration is expected to enhance filtration at low pH.
  • an additive may be one selected from the group consisting of salts, buffers, kosmotropes, chaotropes, detergents, surfactants, PEG, amino acids, organic solvents, acids or bases.
  • a filter operational mode may vary between a first mode and a second mode. For example, a filter operational mode may vary between a constant pressure mode and a constant flux mode.
  • each of the constant pressure mode and a constant flux mode may assume a constant temperature.
  • an exemplary filtration process may begin in a first mode and switch to a second mode, or vice versa, or may alternate between modes.
  • an exemplary filtration process may employ variable pressure and/or variable flux (e.g., stepping alternately up and down, for example from approximately 15 psi to approximately 5 psi, and from approximately 5 psi to approximately 15 psi or vice versa), stepping continuously up or down (for example, from approximately 5 psi to approximately 10 psi to approximately 15 psi or vice versa).
  • an exemplary filtration process may employ continuous changes in response to certain parameters (for example, a controller may continuously adjust the pressure based on the filtrate flow rate, or based on the filtrate volume, or based on another parameter, or any combination of any of the foregoing).
  • an exemplary filtration process may be carried out intermittently rather than continuously, where the filtration is paused one or more times during a batch (i.e., during a “run”).
  • temperature may be maintained at an approximately constant temperature throughout a batch (e.g., at any setpoint between approximately 2 °C and approximately 40 °C).
  • the temperature may be varied, in any of the ways described above (e.g., stepwise or continuously monitoring and adjusting the temperature).
  • other filtration control parameters may be utilized, including but not limited to one or more product quality attributes as control parameters.
  • a sensor may be provided that measures particle size and/or concentration (which are indicative of the yield and/or the product retention). Information from the sensor may be utilized to determine the extent to which the one or more product quality attributes change during filtration. In response to a change being detected – e.g., an indication that the one or more product quality attributes has varied with respect to an initially determined one or more product quality attributes – then at least one aspect of the filtration process may be varied.
  • a method of filtering LNP-containing solution comprises controlling a flow rate of the feed solution.
  • the flow rate is controlled based on the properties of the solution to be filtered.
  • an operator or user may input various parameters or other information relating to the solution into a computer or microcomputer of a controller via a user interface.
  • the controller is configured set a target flow rate based on the properties of the solution or other information provided by the operator or user.
  • the flow rate may be in a range from approximately 1000 L/m 2 /hr to approximately 2000 L/m 2 /hr for solutions that have a water content that exceeds a threshold water content.
  • the flow rate may be approximately 1500 L/m 2 /hr.
  • the flow rate may be in a range from approximately 45 L/m 2 /hr to approximately 550 L/m 2 /hr for solutions such as biologics that have a lower aqueous content (that is, aqueous content that does not exceed the above- mentioned threshold).
  • the flow rate may be approximately 500 L/m 2 /hr.
  • the flow rate may be in a range of approximately 10 L/m 2 /hr to approximately 50 L/m 2 /hr, approximately 50 L/m 2 /hr to approximately 100 L/m 2 /hr, approximately 100 L/m 2 /hr to approximately 200 L/m 2 /hr, or approximately 200 L/m 2 /hr to approximately 500 L/m 2 /hr.
  • a method of filtering LNP-containing solution comprises controlling a temperature of the LNP-containing solution and/or controlling a temperature at which filtration occurs (i.e., an environmental temperature).
  • a method of filtering LNP-containing solution comprises controlling a pH of the LNP-containing solution.
  • a method of filtering LNP-containing solution includes controlling (i) a salt concentration of the LNP-containing solution and/or (ii) a type of salt in the LNP-containing solution.
  • a method of filtering LNP-containing solution includes setting one or more filtration control parameters to one or more respective control target values; determining, after filtration has begun, a deviation between one or more actual control values and the one or more control target values; and, in response to determining that the deviation exceeds a threshold value, adjusting the one or more filtration control parameters.
  • one or more filtration control parameters such as flow rate or pressure are adjustable to achieve a target control flow rate and a target control pressure, respectively.
  • a method of filtering LNP-containing solution includes controlling an operational mode of filtration.
  • a pulse-width-modulation (PWM) control may be used, as implemented via a controller or microcontroller.
  • the operational mode may be selected from and/or switched between a continuous filtration and an intermittent filtration. In intermittent filtration, the filtration is periodically stopped and restarted (e.g., “pulsed”).
  • the period between operational time for the filtering – that is, the “down” time when the filtering is stopped — may range from several minutes to several hours, e.g., (i) from more than approximately 2 minutes to less than approximately 5 hours, (ii) between approximately 1 second to approximately 6 hours, (iii) approximately 30 seconds, (iv) between approximately 4 minutes to approximately 8 minutes, (v) approximately 10 minutes, or (vi) approximately two hours, for example.
  • a pause (of fixed or variable duration) may be imposed between a first filtering period and a second filtering period. Such a pause may relieve the accumulated foulant from the filter surface. In particular, increased yield and/or lower resistance may be realized following such a pause.
  • the length or imposition of a pause may be determined based at least in part on a feedback loop used for controlling one or more filtration control parameters. Accordingly, in some embodiments, one or more filtration control parameters may be adjusted to promote creation and/or maintaining of such pauses. [0077] In some embodiments, during one or more stops (pauses) between filtration, one or more filtration control parameters may be evaluated. That is, after a first filtration period and before a second filtration, a determination may be made as to whether one or more filtration control parameters should be adjusted. For example, actual values for the one or more filtration control parameters may be compared to target and/or predicted values and adjusted accordingly.
  • a method of filtering LNP-containing solution includes introducing one or more additives to the feed-containing solution.
  • the additives may enhance filtration by promoting disassociation between LNPs and/or between LNPs and the filter media.
  • the additives may enhance filtration by altering one or more biophysical properties to reduce or prevent aggregation of the LNPs.
  • the biophysical properties include, but are not limited to, a pH value, a salt concentration, and a salt type.
  • the additive may include, but is not limited to, polyethylene glycol (PEG).
  • a method of filtering LNP-containing solution includes assessing an extent to which foulants are retained and/or removed by the filter media, and optionally, performing refiltration.
  • the methods include performing pressure stepping, or gradient pressure changes.
  • a method of filtering LNP-containing solution may be performed for a dual layer filter media assembly.
  • the filter media may be implemented as a membrane or a membrane assembly.
  • a non-transitory computer readable medium is configured to store instructions which, when executed by a processor, cause a controller to adjust one or more filtration control parameters.
  • one or more filtration control parameters are adjusted to account for actual and/or predicted behavior of LNPs in solution.
  • experimental data obtained during testing of filtration of LNP-containing solutions indicates an increase in filtrate flux and filter media capacity with increasing TMP.
  • Experimental data indicates that high TMP filtration of LNPs may reduce filtration resistance and increase filter media capacity.
  • Sterile filtration is a process of filtering a solution to remove such contaminants without adversely affecting the final product.
  • sterile filtration of LNP may include passing a solution containing LNP (e.g., LNP and buffer, etc.) through filter media.
  • the filter media may include a single or multi-layer filter media (e.g., multi-membrane filter media, an assembly constructed of multiple filters, etc.).
  • pressure is applied to the feed solution upstream of the filter media to force the feed solution through the filter media. That is, the solution is pressurized to a given fluid pressure.
  • the fluid pressure may be set as a filtration control parameter and, in some embodiments, can be adjusted during filtration.
  • the filtration may be performed under substantially constant pressure, which may be referred to as “constant pressure filtration.”
  • the filter media can capture contaminants along the surface or within pores of the filter media while allowing smaller particles to pass therethrough.
  • “constant flux filtration” may be carried out instead of or in addition to constant pressure filtration.
  • a substantially constant flowrate is applied to the feed solution upstream of the filter media. As the filter media becomes clogged, the pressure across the filter media increases above a threshold value.
  • a filtration system may be controlled based on a filtration control parameter.
  • the filtration control parameter may in turn be based on or correspond to a threshold value.
  • the threshold value is a maximum resistance value which is observed with increasing TMP until a given pressure value or range of pressures is reached, before resistance declines thereafter, as discussed in greater detail below.
  • the threshold value is a pressure value associated with the maximum resistance value.
  • the filtration control parameter may be based on an estimated value, a sensed value or a combination thereof.
  • a tangential flow device may be used.
  • a method of filtering LNP-containing solution may include filtering the solution through a tangential flow device and adjusting one or more filtration control parameters to enhance LNP recovery.
  • Tangential flow filtration involves passing a feed solution parallel to the filter media, where the permeate is collected and the retentate is re-cycled back to the feed solution. By alternating a tangential flow of fluid through the filter media, continuous filtration may be achieved.
  • the selection of filter media may be made depending on the properties of the solution and the desired filter performance. In particular, filter media may be selected based on the size of contaminants (e.g., bacteria) that need to be removed. However, the size of the LNP particles (in comparison to the average media pore size required to retain bacteria) may require large filter sizes to ensure sufficient filter capacity at the required filtration rates for production. Accordingly, the techniques of some embodiments include decreasing the extent to which small particles constrict passages in a filter.
  • the filters are sterilizing-grade filter media.
  • Some embodiments comprise filtration methods in which a filter media includes a first layer having a first pore size and a second layer having a second pore size smaller than the first pore size.
  • the filter media includes a coarse filtration layer (e.g., pre-filtration layer) and a fine filtration layer coupled to the coarse filtration layer.
  • the coarse filtration layer is layered on top of the fine filtration layer and positioned upstream from the fine filtration layer during the sterile filtration operation to remove the largest contaminants from the feed solution.
  • the coarse filtration layer includes a media layer (e.g., membrane layer) having an 0.8 ⁇ m rating that is structured to remove particles as small as approximately 0.8 ⁇ m in diameter.
  • the fine filtration layer includes a media layer (e.g., membrane layer) having an 0.2 ⁇ m rating that is structured to remove particles as small as approximately 0.2 ⁇ m in diameter.
  • the fine filtration layer may have a smaller rating, such as approximately 0.1 ⁇ m.
  • At least one of the coarse filtration layer or the fine filtration layer may have a larger rating, such as approximately 0.3 ⁇ m, approximately 0.4 ⁇ m, approximately 0.45 ⁇ m, approximately 0.65 ⁇ m, approximately 0.8 ⁇ m, approximately 1.0 ⁇ m, approximately 2 ⁇ m, approximately 3 um, approximately 5 ⁇ m, or approximately 10 ⁇ m.
  • the filter media is considered an absolute pore size rating, whereas in some embodiments, the pore size is considered nominal.
  • the filter media contains symmetrical pores. In some embodiments, the filter media contains asymmetrical pores.
  • the filter media are implemented as a single layer, whereas in other embodiments, the filter media may be implemented as a dual-layer assembly, or a filter assembly having a different number of layers.
  • one or more of the coarse filtration layer or the fine filtration layer may have differing filter ratings from the foregoing.
  • the coarse filtration layer may be configured to remove particles as small as approximately 0.45 ⁇ m in diameter.
  • the coarse filtration layer (the upstream filter layer) and the fine filtration layer (the downstream filter layer) are selected from the filter media listed in Table 1 below.
  • a method of filtering LNPs includes filtering through a two-layer filter wherein the first layer and/or the second layer of the multi- layer filter is selected from the filters listed in Table 1.
  • exemplary filter media may be selected from one or more of the following materials: polyethersulfone (PES), polyvinylidene fluoride or polyvinylidene difluoride (PVDF), nylon or cellulose acetate (CA), for example.
  • exemplary filtration systems may vary with respect to one or more of the following: a pore size, a pore distribution, a pore geometry (e.g., symmetrical versus asymmetrical), a filter layer arrangement (e.g., a single layer filter or a multi-layer filter), filter assembly (e.g., stacking), etc.
  • a “symmetrical” pore geometry refers to a pore configuration having a substantially cylindrical profile
  • an “asymmetrical” pore geometry refers to a pore configuration having narrowing at one end of the pore, for example.
  • a method of filtering a solution containing lipid nanoparticles includes setting at least one filtration control parameter to at least one target filtration control parameter. Some embodiments comprise performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size. Some embodiments comprise determining, during or after the first period, a value of the at least one filtration control parameter; and adjusting the at least one filtration control parameter in response to determining that the value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold.
  • the actual value may be an actual value or estimated value.
  • a filtration control parameter may be maintained or adjusted in consideration of the filter age. For example, a flow rate may be maintained within a range of flow rates or increased above a threshold rate to increase flow until a filter requires replacement. Controlling the flow rate in this manner, among other filtration control parameters, may contribute to higher yields and increased capacity.
  • the filtration control parameter is a threshold value that corresponds to a characteristic of the membrane itself and/or the feed solution (e.g., a solution containing LNPs, a buffer solution, etc.).
  • Some embodiments include directing, through filter media, a feed solution containing LNPs.
  • the feed solution may be pressurized to a set pressure.
  • the method may include sensing a filtrate flux and/or the throughput of the filter media over time at different TMPs.
  • high TMP may reduce filtration resistance. As noted above, contrary to behaviors typically associated with compressible fouling deposits, the capacity of the filter media for sterile filtration of LNPs is shown to increase with increasing TMP.
  • some embodiments comprise filtering a LNP solution at a TMP of approximately 2 psi, approximately 8 psi, approximately 14 psi, approximately 15 psi, approximately 16 psi, approximately 17 psi, approximately 18 psi, approximately 19 psi, or approximately 20 psi, or more.
  • TMP is monitored continuously during filtration via one or more sensors.
  • the sensors may include pressure sensors placed upstream of the filter.
  • the outlet of the filter may be at atmospheric pressure, but a sensor may nonetheless be positioned at the outlet in case atmospheric pressure is not maintained.
  • the TMP corresponds to the upstream pressure less the downstream pressure.
  • the sensors may be any suitable pressure sensor, including a digital sensor such as PendoTECH pressure sensors made by PendoTECH Corp. of Princeton, NJ or the SciLog® pressure sensor (e.g., the SciPres® sensor) made by Parker Hannifin Corp. of Oxnard, CA.
  • the methods include adjusting different filtration control parameters.
  • one or more filtration control parameters may be adjusted during a filtration process (e.g., after a filtration “run” has started but before it has finished).
  • one or more filtration control parameters may be adjusted via a programmable controller or manually (e.g., by a manual operator).
  • a programmable controller or a plurality of programmable controllers may be used to control one or more components of an exemplary filtration system as shown in FIG.19.
  • FIG.19 depicts an exemplary filtration system including a feed vessel (e.g., a fluid reservoir), a liquid transfer mechanism (e.g., a pump, a motor-driven pump, a micro-pump, a fluid supply source, a pressure source), a filter assembly (e.g., a stacked filter arrangement, a filter membrane assembly, a plurality of filter layers), and a filtrate vessel (e.g., a filtrate reservoir).
  • a feed vessel e.g., a fluid reservoir
  • a liquid transfer mechanism e.g., a pump, a motor-driven pump, a micro-pump, a fluid supply source, a pressure source
  • a filter assembly e.g., a stacked filter arrangement, a filter membrane assembly, a plurality of filter layers
  • a filtrate vessel
  • the liquid transfer mechanism is configured to transfer liquid throughout the system, e.g., a pressurized gas overlay utilizing compressed air or compressed nitrogen, among other substances.
  • a pressurized gas overlay utilizing compressed air or compressed nitrogen, among other substances.
  • Each circle in FIG.19 denotes an exemplary sensor.
  • integrated sensors may be used and the number of sensors may differ from what is shown.
  • Various sensors that are utilized may include, but are not limited to, a pressure sensor, a flow rate sensor, a temperature sensor, etc.
  • the sensors may be used upstream of the filter, downstream of the filter, or upstream and downstream of the filter, as well as in the feed vessel and/or in the filtrate vessel.
  • the filter assembly may include multiple media and/or multiple separate filters in series or in parallel.
  • a complex assembly having multiple filters may, in some embodiments, utilize sensors at multiple locations within the filter assembly.
  • information from any one of the sensors may be provided (e.g., in the form of a digital signal) to a controller which may be accessible to a user.
  • the controller may automatically adjust one or more filtration control parameters and/or allow a user to adjust one or more filtration control parameters.
  • one or more filtration control parameters may be adjusted in response to determining that a calculated parameter exceeds a threshold value.
  • the calculated parameter may be a calculated resistance to flow across the filter media.
  • the calculated parameter may be any parameter associated with a fouling model such as the fouling models discussed above.
  • the one or more filtration control parameters may be calibrated to influence resistance.
  • resistance at the start of filtration was found to be independent of applied TMP, but resistance transitions to a high resistance regime earlier for lower values of TMP. This can lead to reduced filter media capacity relative to filtrations performed at higher TMP.
  • larger values of TMP prolong operating times in the low-resistance regime (e.g., below 100 psi-m 2 -s/L), delaying the transition to high resistance, which results in greater overall filter media capacity.
  • a method of filtering LNP-containing solution comprises (i) measuring the resistance, (ii) comparing the measured resistance to a threshold value, and (iii) in response to determining that the measured resistance exceeds the threshold value, adjusting one or more filtration control parameters.
  • a controller may utilize resistance values, so as to increase a feed pressure with an increase in resistance (e.g., linearly proportional to resistance), so as to maintain a substantially constant resistance.
  • at least one exemplary method of controlling filtering includes adjusting one or more filtration control parameters in response to imaging data.
  • the imaging data is obtained by imaging at least one layer of the filter media to visually assess one or more properties of particulate contamination (e.g., the foulant).
  • the one or more properties of particular contamination include, but are not limited to, a quantity thereof, a distribution thereof, and/or a geometrical profile of the particulate contamination along the surface of the layer(s).
  • the method may include taking one or more SEM images of an outer surface of each media layer at a resolution that is approximately equal to a filter rating and/or a pore size of the media layer.
  • imaging may be used in characterizing filtration results to determine which operational modes may correlate to various degrees of fouling.
  • the method may include characterizing the solution containing LNPs based on the imaging and a determined extent of fouling.
  • the solution may be characterized by categorizing it in terms of a fouling model (e.g., complete pore blockage, intermediate pore blockage, pore constriction, or cake formation) and/or based on the nature of the foulant (e.g., incompressible or compressible).
  • the method further includes (i) evaluating individual layers of filter media, (ii) comparing the filtration performance between the filter layers, and (iii) controlling one or more filtration control parameters based on a differential in performance between the individual layers. [0107] In some embodiments, the method further includes determining an extent to which contaminants are retained and/or removed by the filter media. Among other benefits, such a process may yield information regarding whether additional stages are required to achieve the desired performance, and/or whether the type and/or geometry of the filter media needs to be adjusted.
  • the method includes performing re-filtration processing in which a feed sample containing LNPs is passed through redundant stages of filter media (e.g., passing the feed solution through a first filter media to generate a first filtered solution, and then passing the first filtered solution through a second filter media that matches the type and geometry of the first filter media).
  • Some embodiments comprise using a re-filtration process involving two sequential filters (e.g., each filter being two layers).
  • approximately 85% to approximately 100% of LNPs in the LNP-containing solution are recovered.
  • approximately 95% to approximately 98% of LNPs are recovered.
  • approximately 90% to 96% of LNPs are recovered.
  • the method further includes adjusting one or more filtration control parameters based on data from a re-filtration operation. In some embodiments, a derivative analysis of re-filtration data (data relating to the LNP-containing solution) may be performed. [0111] In at least one embodiment, the method further includes conducting additional adjustments to one or more filtration control parameters based on temporal changes in test conditions. For instance, in at least one embodiment, the method additionally includes performing pressure stepping to account for the influence of (i) membrane fouling at different pressures; and (ii) changes in the composition of the feed solution, where the pressure stepping data may be used to determine whether to adjust one or more filtration control parameters.
  • the method additionally includes performing pressure stepping in reverse. Such approaches may result in a TMP-dependent change in filtration performance. In turn, one or more filtration control parameters may be adjusted to account for the TMP-dependent change. [0113] In some embodiments, the method additionally includes adjusting one or more filtration control parameters based on expected temporal changes in the composition of the feed solution which may impact filtration performance. [0114] Techniques for characterizing the underlying mechanisms controlling sterile filtration of LNPs according to an aspect of the present disclosure will now be described. In some aspects, the method may include additional, fewer, and/or different operations.
  • a method includes evaluating the filtration performance of filter media using a feed solution containing LNPs (e.g., a solution containing LNPs, a buffer solution, etc.).
  • the method may include performing evaluations of sterile filter media to determine how filtrate flux and/or the throughput of the filter media vary with time at different TMPs.
  • FIGS.4-5 show scatter plots of experimental data from sterile filtration of an example feed solution at different TMPs. Each dataset shows the change in the filtrate flux and volumetric throughput of the filtrate throughout the duration of the test.
  • TMP refers to the pressure drop across the filter media (e.g., in units of psi)
  • filtrate flux refers to a rate of transfer of the feed solution through the filter media (e.g., in units of L/m 2 /hr)
  • volumemetric throughput refers to a total amount of filtrate (e.g., permeate, filtered solution, etc.) processed by the filter media at different times throughout the test per unit membrane area (e.g., in units of L/m 2 ).
  • FIG.4 shows a scatter plot of filtrate flux as a function of the volumetric throughput during constant pressure filtration of LNPs through a Sartopore® 2 XLG capsule at the foregoing pressures.
  • the tests indicate that the rate of decrease in filtrate flux increased over the duration of the test regardless of TMP, with the filtrate flux decreasing by as much as approximately 99% for some of the tests.
  • a method according to the present disclosure includes evaluating the data using different filtration performance parameters and/or derived metrics.
  • the method may include plotting the test data in terms of measured resistance to flow across the filter media during each test, where “resistance” refers to the value of TMP divided by the filtrate flux at each data point during the test (e.g., the TMP normalized by the measured filtrate flux at each data point, in units of psi-m 2 -s/L).
  • resistance refers to the value of TMP divided by the filtrate flux at each data point during the test (e.g., the TMP normalized by the measured filtrate flux at each data point, in units of psi-m 2 -s/L).
  • FIG.5 shows a scatter plot of the measured resistance at each data point from FIG.4.
  • FIG.5 shows a scatter plot of resistance as a function of the volumetric throughput during constant pressure filtration of LNPs through the Sartopore® 2 XLG capsules at pressures of approximately 2 psi, approximately 8 psi, approximately 14 psi and approximately 20 psi.
  • FIG.5 shows data from FIG.4 re-plotted according to the following equation: [0121]
  • the resistance at the start of filtration is similar for each data set, regardless of the applied TMP.
  • the resistance transitions to a high resistance regime much earlier for lower values of TMP. This leads to reduced filter media capacity relative to tests performed at higher TMP.
  • the filtrate flux (J) is plotted as a function of the volumetric throughput (v), defined as the ratio of the cumulative filtrate volume (V) to the membrane area (A). In each case, the curves are concave down on the semi-log plot, with the absolute value of the slope increasing with increasing throughput.
  • Vmax filter capacity
  • FIGS.20A-20B show the total resistance of the membrane with the deposited LNPs as a function of filtration time during a pressure-stepping experiment, using both Isopore and Sartopore membranes.
  • the TMP was varied in a stepwise fashion in 30-minute intervals, starting at 140 kPa (20 psi), decreasing to 56 kPa (8 psi), and then returning to 20 psi for deposited LNPs on a 0.2 ⁇ m pore size Isopore track etch membrane (FIG.20A) and on the 0.2 ⁇ m layer of the Sartopore 2 XLG (FIG.20B).
  • the hydraulic resistance of clean Isopore and Sartopore membranes were 3.0 ⁇ 0.1 x 10-3 and 2.1 ⁇ 0.1 x 10-3 (psi/(L/m 2 /h)) (7.5 ⁇ 0.3 x 107 and 5.2 ⁇ 0.2 x 107 Pa/m/s), respectively, which are more than 100-fold smaller than the values measured in FIGS.20A-20B. Because the total resistance (i.e., membrane plus deposit) is controlled predominantly by the contribution from the deposited LNPs, and the membrane is an incompressible polymer at the pressures studied, the measured total resistance was taken as representative of the resistance of the deposited LNP.
  • the resistance for the fouling deposit on the Isopore membrane (FIG.20A) returned to its original value upon increasing the pressure back to 20 psi, indicating a reversible behavior, i.e., there were no permanent changes in the fouling deposit.
  • the resistance of the fouling deposit on the Sartopore 2 XLG membrane (FIG.20B) during the second 20 psi step was below the initial resistance at this TMP. This may be attributable to the transmission of some of the deposited LNP through a Sartopore XLG, as permeate samples obtained during the second filtration at 20 psi showed a low but non-zero level of mRNA based on the concentration.
  • the method further includes conducting tests on individual layers of filter media and comparing differences in the filtration performance between the filter layers.
  • FIG.10 shows a scatter plot of sterile filtration performance using individual layers of the filter media from FIGS.2A, 2B and 3 discussed below. More particularly, FIG.10 depicts filtrate flux as a function of the volumetric throughput for filtration through the individual layers of the Sartopore® 2 XLG dual-layer filter.
  • FIG.10 data were obtained for the coarse filtration layer 102, the fine filtration layer 104, and the combined filter media including both the coarse filtration layer 102 and the fine filtration layer 104 at a fixed TMP (e.g., approximately 14 psi).
  • TMP e.g., approximately 14 psi
  • the capacity of the coarse filtration layer 102 is significantly larger than either the fine filtration layer 104 or the combined filter media 100 (e.g., approximately 7 times greater than the combined filter media 100 for the same TMP).
  • the fine filtration layer 104 tested on its own showed the lowest capacity of all three data sets and provided the lowest measured LNP recovery.
  • the 0.2 ⁇ m layer alone showed a rapid flux decline and a small filter capacity V max of approximately 16 L/m 2 .
  • the results indicate a modest increase in the overall filter capacity when compared with the fine filtration layer 104 on its own, which can be attributed to the use of the coarse filtration layer 102 as a pre-filter that protects the fine filtration layer 104 from key contaminants (e.g., foulants, etc.).
  • the dual-layer filter 100 showed an intermediate filter capacity Vmax of approximately 39 L/m 2 , substantially smaller than the 0.8 ⁇ m layer alone yet almost two and a half-fold higher than the 0.2 ⁇ m layer.
  • the smaller value of ⁇ for the dual layer filter reflects the removal of foulants by the pre-filter.
  • the complete pore blockage model poorly fits the flux data for the 0.8 ⁇ m layer alone, suggesting that the fouling observed in this layer is not completely described by a pore blockage mechanism.
  • the method of characterizing the factors governing and/or contributing to filter media fouling may further include performing a derivative analysis to determine the physical mechanism with the greatest contribution to overall filter fouling (e.g., complete pore blockage, pore constriction, intermediate blockage, or cake formation).
  • a derivative analysis may be performed for the derivative of TMP (P) with respect to filtrate volume (V), as shown below in Equation (4), where n is defined as noted above for Equation (3).
  • FIG.11 shows a scatter plot of a derivative analysis performed on the experimental data from FIGS.4-5.
  • FIG.11 depicts a derivative plot where the y-axis corresponds to the second derivative and the x-axis corresponds to the first derivative. As shown, the data are plotted on log-log axis scaling.
  • FIG.12 shows test data from an example re-filtration operation.
  • FIG.12 shows filtrate flux as a function of volumetric throughput during filtration of the fresh LNP and previously filtered LNP through the Sartopore® 2 XLG according to the working examples.
  • the test data were obtained from a re-filtration process involving two sequential filters (each being two layers).
  • a first filtration operation labeled as “1 st filtration” in FIG.12
  • a feed sample containing fresh (e.g., unfiltered) LNPs is passed through a first stage sterile Sartopore® 2 XLG filter media at a constant TMP of 14 psi to produce a first filtration permeate.
  • the first filtration permeate is directed through a redundant sterile Sartopore® 2 XLG filter having the same geometry as the first stage filter media (labeled as “refiltration” in FIG.12).
  • the data in FIG.12 were obtained from filtration experiments performed on the same day, with the collected permeate used within 1 hour of the first filtration.
  • the redundant sterile filter media still shows significant fouling at the conclusion of the test, the data show an approximately 2-fold increase in the filter capacity relative to filter media in the first filtration stage.
  • an approximately 2-fold increase in the filter capacity was observed as TMP was increased from approximately 2 psi to approximately 20 psi.
  • the method further includes conducting a derivative analysis of the data from the re-filtration operation.
  • a derivative analysis of re-filtration data may be performed. Such an analysis may show, for example, that the re-filtration data indicates filter fouling behavior that is consistent with the complete pore blockage model, similar to test data from FIGS.2A-2B discussed below.
  • Pressure Stepping [0139]
  • the method further includes conducting additional evaluation to determine how the performance of the sterile filtration operation varies with temporal changes in test conditions. For instance, in the example filtration operation described herein, the method additionally includes performing pressure stepping experiments to determine the influence of (i) LNP deposition at different pressures; and (ii) changes in the composition of the feed solution.
  • FIG.13 shows a scatter plot showing test data from a pressure stepping experiment using the Sartopore® 2 XLG filter media.
  • a feed solution containing LNPs is directed through sterile filter media at constant TMP.
  • the TMP is fixed at approximately 14 psi for a first time period (e.g., within a range from the start of the filtration up to approximately 150 seconds or approximately 200 seconds, or another suitable period).
  • a pressure upstream of the filter media is adjusted to cause a first stepwise change in TMP during the test.
  • the TMP is adjusted after the first time period from a first TMP of approximately 14 psi to a second TMP of approximately 1 psi. Testing continues at constant TMP for a second time period until a trend in filtration performance is established.
  • a pressure upstream of the filter media is adjusted to cause a second stepwise change in TMP back to the first TMP value.
  • Testing continues at the first TMP for a third time period until a trend in filtration performance is established. It should be appreciated that test parameters in the pressure testing experiment may be adjusted as desired to determine the effects of changes in test conditions.
  • the results from the pressure stepping experiment indicate non-linear behavior with respect to the filtrate flux.
  • Filtration of LNP solutions may exhibit non-linear behavior with respect to any of filtrate flux, resistance or transmembrane pressure (TMP), for example.
  • TMP transmembrane pressure
  • non-linear changes in filtrate flux were observed in response to stepwise changes in TMP.
  • the data in FIG.13 show an approximately 70-fold increase in filtrate flux in response to an approximately 14- fold increase in TMP.
  • the test data at approximately 14 psi after the stepwise change in TMP is continuous with the data trends observed at the start of the test, indicating no irreversible changes in filter fouling due to operation at 1 psi.
  • the data in FIG. 13 show that performing sterile filtration of LNPs at high TMP also reduces the rate of change in filtrate flux (e.g., the rate of decline of the filtrate flux).
  • the method additionally includes performing pressure stepping experiment in reverse, which may help identify or yield further insight into the physical mechanism or mechanisms affecting TMP-dependent changes in filtration performance.
  • FIG.14 shows a scatter plot of test data of a reverse pressure stepping experiment starting at an initial TMP of approximately 1 psi.
  • Such an experiment in addition to any of the experiments described herein, may be carried out utilizing a kit including a filter (e.g., filter media) and a solution containing LNPs.
  • a filter e.g., filter media
  • a solution containing LNPs e.g., water
  • a stepwise change in TMP from 14 psi to 1 psi causes an approximately 150-fold reduction in filtrate flux, which is even greater than the changes observed during the initial pressure stepping experiments.
  • the resistance to sterile filtration of LNPs is shown to decrease at large TMP.
  • the method additionally includes performing tests to determine how temporal changes in the composition of the feed solution impact filtration performance (e.g., to determine whether the TMP-dependent changes in filtration performance are also observed with the buffer solution).
  • FIG.15 shows test data from a pressure stepping experiment in which the composition of the feed solution is changed during the test.
  • the sterile filtration test is performed using a Sartopore® 2 XLG dual layer media.
  • the test begins by introducing a feed solution containing LNPs at constant pressure (e.g., approximately 14 psi). After a first time period of constant pressure filtration, the LNP feed solution is replaced with a buffer solution that does not include LNPs. Testing continues for a second test period after which the TMP is rapidly decreased from approximately 14 psi to approximately 2 psi.
  • changes in filtrate flux of the buffer solution are similar to those observed at the end of filtration with the feed solution containing LNP.
  • a similar pressure dependence as the LNP feed solution is also observed in response to changing TMP. As shown in FIG.15, an approximately 7-fold decrease in TMP results in an approximately 30-fold decrease in filtrate flux.
  • results from testing of the Sartopore® 2 XLG dual layer filter media show high levels of LNP recovery despite observed fouling of the filter media.
  • LNP recovery of between approximately 90%, approximately 91%, approximately 92%, approximately 93%, approximately 94%, approximately 95%, approximately 96%, approximately 97% or approximately 98% was exhibited in some embodiments.
  • SEM imaging discussed below, confirms fouling primarily on the upper surface of the fine filtration layer of the filter media.
  • FIGS.2A, 2B and 3 show SEM images of an example sterile filter media, shown as filter media 100. The SEM images depicted herein were taken as described in the working examples discussed below.
  • the filter media 100 includes a polyethersulfone, dual-layer sterile filter media such as the Sartopore® XLG dual-layer filter (e.g., Sartopore® 2 XLG filter made by Sartorius Stedim Biotech GmBH of Göttingen, Germany) or other suitable sterilizing-grade filter media, as described in the working examples.
  • the SEM images in FIGS.2A, 2B, 3 and 6-9 are SEM images of the Sartopore® 2 XLG filter media according to the working examples.
  • FIGS.4-5 and 10-18 depict data obtained using the same batch of LNPs made according to the working examples.
  • the filter media 100 includes a coarse filtration layer 102 (e.g., pre-filtration layer) and a fine filtration layer 104 coupled to the coarse filtration layer 102.
  • the coarse filtration layer 102 is layered on top of the fine filtration layer 104 and positioned upstream from the fine filtration layer 104 during the sterile filtration operation to remove the largest contaminants from the feed solution.
  • FIGS.2A-2B show SEM images of an upper surface of the coarse filtration layer 102 of the filter media 100 at 5 ⁇ m resolution.
  • FIG.2A and FIG.2B are SEM images of the 0.8 ⁇ m layer of the Sartopore® 2 XLG, after filtration of buffer (FIG.2A) and after filtration of the LNPs (FIG.2B) through the dual-layer filter at a pressure of approximately 14 psi until a flux decline of more than 99% occurred, with scale bars of 5 ⁇ m.
  • FIG.3 shows an SEM image of an upper surface of the fine filtration layer 104 of the filter media 100 at 5 ⁇ m resolution, where the fine filtration layer 104 is in an unused condition.
  • a method according to at least one aspect includes characterizing the factors governing and/or contributing to filter media fouling.
  • the method includes imaging at least one layer of the filter media to visually assess one or more properties of particulate contamination (e.g., the foulant).
  • the one or more properties of particular contamination include, but are not limited to, a quantity thereof, a distribution thereof, and/or a geometrical profile of the particulate contamination along the surface of the layer(s).
  • the method may include taking one or more images using microscopy of an outer surface of each media layer at a resolution that is approximately equal to a filter rating and/or a pore size of the media layer. A variety of microscopy imaging tools may be used, as discussed further below.
  • the method includes taking “before” and “after” images of each individual media layer in a clean (e.g., unused) condition and an at least partially fouled (e.g., used) condition.
  • FIGS.6–7 are SEM images of an outer surface of a new and used coarse filtration layer of the filter media 100 of FIGS.2A-2B, shown as new coarse layer 108 and used coarse layer 110, respectively.
  • the overall structure of the coarse filtration layer 102 and pores within the used coarse layer 110 remain visible in the SEM image.
  • the SEM images indicate only minimal fouling on the coarse filtration layer 102.
  • FIGS.8-9 are SEM images of an outer surface of a new and used fine filtration layer of the filter media 100 of FIG.3, shown as new fine layer 112 and used fine layer 114, respectively.
  • FIGS.8-9 are SEM images of the 0.2 ⁇ m layer of the Sartopore® 2 XLG filter, before and after filtration of the LNPs through the dual-layer filter at a pressure of approximately 14 psi until a flux decline of more than 99% occurred, with a scale bar of 5 ⁇ m.
  • the SEM images in FIGS. 8-9 show extensive fouling on the used fine layer 114 with contaminants completely blocking the entrance to the pores across at least one section of the used fine layer 114.
  • the 0.8 ⁇ m layer has a rough, asymmetric surface with relatively low pore density.
  • the 0.2 ⁇ m layer appears substantially smoother with a higher density of relatively circular pore openings.
  • There is no visible difference in the 0.8 ⁇ m pre-filter after fouling suggesting most of the pores remain fully open, which is consistent with the very high filtration capacity when the 0.8 ⁇ m layer was used alone, as indicated in FIG.10.
  • much of the surface of the 0.2 ⁇ m layer is covered with LNPs 10, appearing as an aggregated layer on top of the membrane.
  • mRNA-LNPs may also be referred to as “mRNA-LNPs” or “deposited LNPs.”
  • the structure and organization of the LNPs seen in the SEM images in FIGS.2A, 2B, 3 and 6-9 may be affected by the sample preparation (drying) and may appear different in the native (wetted) environment, although the high degree of surface fouling is consistent with the pore blockage mechanism identified by the derivative analysis in FIG.11.
  • a method of evaluating sterile filtration of LNPs is provided.
  • the method includes obtaining LNPs, e.g., an mRNA-LNP solution.
  • the method includes depositing the LNPs on membranes respectively varying in pore size.
  • the membranes may be a plurality of membranes wherein each membrane has a pore size that is larger or smaller by at least 0.1 um than another of the membranes. For example, membranes of 0.1 um, 0.2 um and 0.4 um pore size may be evaluated.
  • the membranes were then housed in a filtration cell on top of a mesh.
  • the method may further include determining the hydraulic resistance of the membranes when buffer was filtered through the membranes. In particular, the buffer flux was measured at various transmembrane pressures to determine the hydraulic resistance.
  • the method further includes supplying the filtration cell with the mRNA-LNPs, supplying buffer through a feed reservoir, and performing filtering of the buffer and the mRNA-LNP solution through the membranes at a constant TMP to deposit LNPs on the membrane.
  • the method further involves comparing the buffer flux of the membrane with the buffer to that with the deposited LNPs, and determining the hydraulic resistance of the deposited LNPs.
  • a method of evaluating sterile filtration of liquid nanoparticles (LNPs) is provided.
  • the method includes filtering a first solution lacking LNPs (e.g., buffer) through at least one membrane.
  • the method may include controlling a filtration control parameter during filtering to satisfy a control parameter threshold.
  • the filtration control parameter may include a TMP during filtering and/or a flux of the first solution through the membrane.
  • the control parameter threshold may be a target TMP and/or target flux of the first solution.
  • controlling the filtration control parameter to satisfy the control parameter threshold may include adjusting the TMP and/or flux of the first solution to approximately equal the target TMP and/or the target flux through the membrane.
  • the method additionally includes determining a first hydraulic resistance of the membrane following filtration of the first solution.
  • the method further includes filtering a second solution containing LNPs through the at least one membrane; and determining a second hydraulic resistance of the membrane following filtration of the second solution. During filtering, the filtration control parameter may be controlled to satisfy the control parameter threshold.
  • the method further includes computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; and imaging the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling (and/or the computed difference between the first hydraulic resistance and the second hydraulic resistance).
  • evaluating an SEM image, for example, as well as experimental filtration data allows for characterization of both membrane performance and the behavior of LNPs themselves. This information can be used in determining whether or not to alter membrane selection or adjust one or more filtration control properties as discussed further herein.
  • the one or more filtration control properties is a pressure value, P CRITICAL , discussed herein.
  • WORKING EXAMPLES The following working examples are illustrative and non-limiting.
  • Example 1- Materials Experiments were performed with an mRNA-LNP solution (LNP) provided by Moderna Inc. of Cambridge, MA. Frozen LNP solution was thawed in a room temperature water bath immediately before use. [0164] Sterile filtration data were obtained using track-etch polycarbonate Isopore membranes with 0.1 ⁇ m, 0.2 ⁇ m, and 0.4 ⁇ m pore sizes (MilliporeSigma, Bedford, MA), as discussed below.
  • LNPs were deposited on the polycarbonate Isopore membranes with 0.1 ⁇ m, 0.2 ⁇ m, and 0.4 ⁇ m pore sizes, which have smooth and uniform surfaces with a parallel array of straight-through cylindrical pores, facilitating microscopic analysis.
  • sterile filtration data were obtained using Sartopore® 2 XLG dual-layer asymmetric polyethersulfone (PES) filters having a 0.8 ⁇ m pore size layer on top of a 0.2 ⁇ m layer.
  • PES polyethersulfone
  • the filter capsule was fed by a 1-liter stainless-steel feed reservoir made by Alloy Products Corporation, of Waukesha, WI.
  • the reservoir was pressurized with compressed air and controlled using a pressure regulator.
  • TMP was monitored using a digital pressure gauge made by Ashcroft Inc. of Stratford, CT. The pressure gauge was placed immediately upstream of the filter, with the permeate outlet maintained at atmospheric pressure.
  • the membranes were initially flushed with a minimum of 100 L/m 2 of deionized water followed by Tris-sucrose formulation buffer with the latter prepared by mixing appropriate quantities of Tris-base (MilliporeSigma, Catalog Number 9210), Tris–HCl (Invitrogen, 15567-027), and sucrose (MilliporeSigma, 8550).
  • Tris-base MilliporeSigma, Catalog Number 9210
  • Tris–HCl Invitrogen, 15567-027
  • sucrose MeilliporeSigma, 8550.
  • the membrane hydraulic permeability was evaluated by measuring the filtrate flux at several TMP (up to approximately 70 kPa) using the Tris-sucrose buffer. All experiments were performed at room temperature (21 ⁇ 1 o C).
  • the feed reservoir and the filter capsule were then emptied, and refilled with the LNP solution (typically ⁇ 5 mL for a 4.1 cm 2 membrane area).
  • the feed reservoir was filled with buffer, and the system was then re-pressurized.
  • the buffer was filtered at a constant TMP for at least 1 hour to stably deposit LNPs on the membrane. Filtrate flow was evaluated by continuous mass readings on an OHAUS Ranger TM 3000 scale, with the data logged every second using the OHAUS Serial Port Data Collection Software, both made by OHAUS Corp. of Parsippany, NJ.
  • Permeate concentrations were evaluated based on UV absorbance at 230 nm using a microplate reader made by Tecan of Mannedorf, Switzerland. [0169] The buffer flux through the fouled membrane (with the deposited LNPs) was less than 10% of the buffer flux evaluated through the clean membrane at the same TMP. The hydraulic resistance of the deposited LNPs was then determined by measuring the buffer flux at several specific TMP values obtained with both increasing and decreasing TMP, often using more than one cycle to examine the stability (or hysteresis) of the deposited LNPs. The extent of fouling may be determined based on the difference in hydraulic resistance as compared to the buffer as being, for example, light, moderate or extensive.
  • Example 3 - LNP and Filter Characterization LNP size distribution was determined by Dynamic Light Scattering (DLS) using a Zetasizer Nano ZS90 made by Malvern Panalytical, United Kingdom. Samples were analyzed at approximately 20oC after equilibration for approximately 120 s. The light scattering intensity was evaluated for a total of 10 runs.
  • Example 3a – SEM Characterization LNPs captured on the external surface of the filter were examined by SEM and ESEM.
  • the fouled membrane was removed from the filtration cell, dried inside an Isotemp® 200 Series oven made by ThermoFisher Scientific of Waltham, MA at 30 oC for one hour, and cut into pieces before being mounted on conductive pins with a double-sided carbon tape on circular pins.
  • a thin layer of conductive gold-platinum alloy was applied using an SCD 050 sputter coater made by Bal-Tec AG of Balzers, Lichtenstein to reduce sample charging. Surface images were obtained with a SIGMA VP Field Emission SEM at 3.0 kV made by Zeiss of Jena, Germany.
  • ESEM samples were imaged in the hydrated state at 75% relative humidity, which was maintained by applying a pressure of 6 Torr at 7 o C with the samples mounted on a Peltier cooling stage. SEM and ESEM images were obtained with a Quanta 250 ESEM (Thermo-Scientific, Hillsboro, OR) at 3.0 and 20 keV, respectively. [0172] Microscopic characterization was performed according to some embodiments as shown in FIGS.24A-24F.
  • FIGS.24A-24F depict SEM and ESEM images of the 0.2 ⁇ m Isopore (FIGS.24A-24C) and 0.2 ⁇ m layer of the Sartopore 2 XLG (FIGS.24D-24F) showing the clean membranes (FIG.24A and FIG.24D), SEM images of the heavily fouled membranes (FIG.24B and FIG.24E), and ESEM images of the heavily fouled membranes (FIG.24C and FIG.24F). Scale bars are 10 ⁇ m on each of FIGS.24A-24F. [0173] More particularly, deposited LNPs were explored by both SEM and ESEM as shown in FIGS.24A-24F.
  • FIGS.24A and 24D ESEM images of the clean Isopore and Sartopore membranes (not shown) looked nearly identical to the SEM images in FIGS.24A and 24D.
  • the SEM and ESEM images of the fouled Isopore membrane show that the pores are almost completely covered by an amorphous deposit. This deposit is thought to primarily be comprised of the various lipids present in the original LNP.
  • the SEM image of the upper surface of the fouled Sartopore 2 XLG (FIG.24E) shows a partial amorphous deposit in addition to a small number of individual LNPs.
  • the fouled Sartopore 2 XLG membrane still shows a significant number of open pores, more than would be expected given the high degree of fouling during the LNP filtration.
  • the open pores may be artifacts arising from the drying of the fouled membrane prior to SEM analysis, as such open pores were not visible in the ESEM image (FIG.24F).
  • the similar structures of the deposits on the Isopore (polycarbonate) and Sartopore (polyethersulfone) membranes are qualitatively consistent with the resistance data in FIG.20 and indicate that membrane surface chemistry has a limited effect on the properties of the deposited LNPs.
  • the SEM and ESEM images both suggest that fouling occurs primarily on the upper surface of the filter.
  • Example 3b Atomic Force Microscopy Characterization
  • the deposited LNPs were analyzed by Atomic Force Microscopy (AFM) in a hydrated state.
  • the fouled membrane was removed from the filtration cell and immediately mounted in a petri dish with carbon tape. Sufficient buffer was added to fully cover the membrane.
  • the sample was then imaged using the PeakForce Tapping mode on a BioScope Resolve AFM (Bruker Corp., Billerica, MA) with a scan rate of 0.3 Hz and a peak force setpoint of approximately 500 pN.
  • a 6 x 6 ⁇ m 2 scan was produced with 512 x 512 pixels.
  • a ScanAsyst-Fluid+ cantilever (Bruker Corp., Billerica, MA) with a nominal spring constant of 0.7 N/m and a tip radius of 2 nm was used to generate images, with image processing performed using Gwyddion 2.61 software (Czech Metrology Institute, Jihlava, Czech Republic).
  • Gwyddion 2.61 software Zech Metrology Institute, Jihlava, Czech Republic.
  • the deposited LNPs were formed on the 0.4 ⁇ m Isopore membrane by filtering the LNP until a flux decline of only 50%.
  • the fouled membrane was examined in the hydrated state by AFM.
  • the surface of the Isopore membrane is only partially covered by an amorphous deposit (FIG. 25A), with a small number of individual LNPs (or small LNP aggregates) visible near the bottom of the figure.
  • the height scans across two locations of the fouled membrane (FIG. 25B) show that the amorphous deposit and the individual LNP are approximately 100 ⁇ 15 nm thick, corresponding to the expected size of LNPs.
  • the open pores in FIG.25B are clearly visible as sharp triangular depressions, reflecting the conical shape of the AFM tip.
  • Example 3c – Bubble Point Measurements [0177] The bubble points for the Isopore membranes and the 0.2 ⁇ m layer of the Sartopore 2 XLG were evaluated using the approach described in ASTM F316. The membranes were placed in a 25 mm stainless steel holder and flushed with at least 100 L/m 2 of deionized water. The filter exit was connected to a dip tube that was submerged in deionized water. The upstream surface of the membrane was then pressurized using a compressed nitrogen tank, with the pressure increased at a rate of approximately 1 psi per minute. The bubble point was determined as the nitrogen pressure at which the first bubble was observed rising in the beaker. The bubble point for each membrane was measured three times, with the results presented as the mean ⁇ standard deviation.
  • the 0.1 ⁇ m Isopore membrane was estimated to have a maximum pore diameter of 0.3 ⁇ m based on SEM images showing large pores formed by clusters of three pores that measured approximately 0.3 ⁇ m across.
  • the maximum pore diameters estimated using Equation 4 for the 0.2 and 0.4 ⁇ m Isopore membranes and the 0.2 ⁇ m layer of the Sartopore 2 XLG were consistent with values determined from the SEM images of the clean membranes (FIGS.22A-22D).
  • the estimated diameters of the largest pores (Table 2) are larger than the nominal pore sizes.
  • Example 4 – Pressure Stepping [0181] As described above, pressure stepping experiments were carried out in some embodiments.
  • FIG.16 depicts results in which the filtration was started at approximately 14 psi for a first period of approximately 150 seconds. After the first period, the pressure was abruptly decreased to approximately 1 psi for approximately 300 s before being returned to approximately 14 psi. In both cases, the data are plotted as the resistance (evaluated directly from the flux and TMP data using Equation 1), as a function of filtration time.
  • FIG.17 depicts results from a reverse pressure stepping experiment involving a single continuous filtration run. In the reverse pressure stepping experiment, filtration was started at approximately 1 psi and increased to approximately 14 psi. Following the increase to approximately 14 psi, the pressure was returned to approximately 1 psi.
  • Example 6 Buffer Permeability
  • the direct effect of pressure on resistance was examined by first fouling the Sartopore® 2 XLG filter with LNPs 10 and then measuring the buffer flux through the fouled membrane at different TMPs.
  • FIG.18 shows the total resistance during buffer permeability measurements performed with the Sartopore® 2 XLG membrane.
  • the filter was fed by two pressure reservoirs, one containing the LNPs and one containing buffer, using a 3-way stopcock valve.
  • the initial LNP filtration was conducted at a constant TMP of approximately 14 psi until a flux decline of approximately 90%.
  • the feed was then rapidly switched to buffer at approximately the same TMP. That is, filtration was started with LNPs 10 at approximately 14 psi followed by buffer at approximately 14 psi, buffer at approximately 1 psi, and then buffer at approximately 14 psi.
  • the calculated values of the resistance during this experiment are summarized in FIG.18.
  • the decrease in resistance with increasing TMP is contrary to the expectation for a compressible fouling deposit and is also different from the pressure- independent resistance expected for an incompressible fouling deposit.
  • biphasic behavior was observed wherein the resistance of the deposited LNPs 10 increases at relatively lower pressure (consistent with a compressible filtration medium) and decreasing at higher pressure.
  • the expected behavior for biologics is that they tend to form compressible fouling deposits characterized by an increase in the hydraulic resistance with increasing TMP, although lipid-based dispersions may exhibit different behavior.
  • the pressure-dependent behavior is consistent across the multiple experiments shown in FIGS.4, 15, 16, 17 and 18, suggesting that this phenomenon is intrinsic to LNPs.
  • a threshold value is a maximum resistance value which is observed with increasing TMP until a given pressure value or range of pressures is reached, before resistance declines thereafter.
  • the hydraulic resistance of the deposited LNPs on the 0.2 ⁇ m Isopore membrane was obtained by measuring the buffer flux over a range of TMP, as shown in FIG.21.
  • the LNPs were deposited and maintained in equilibrium at 20 psi for one hour, after which the TMP was reduced to 14, 8, and then 2 psi in a step wise fashion before being returned to 20 psi.
  • Pressure stepping was repeated for two complete cycles for a single deposit formed on the 0.2 ⁇ m Isopore membrane (labeled as the first and second decrease and increase in FIG.21).
  • FIG.21 plots the steady-state values of the hydraulic resistance, as evaluated from the average filtrate flux determined over approximately 15 min of filtration at each TMP, as a function of TMP.
  • the error bars represent the range of the 3-4 data points obtained at each pressure.
  • the results for the multiple pressure cycles appear substantially identical, indicating that there are no irreversible changes in the deposited LNPs over the range of TMP.
  • the total time for this experiment was 6 h, indicating that the deposited LNPs also remain stable for extended periods of time.
  • the resistance of the deposit increased with increasing TMP up to a pressure of approximately 14 psi, reaching a maximum value of approximately 8.0 ⁇ 0.3 psi/(L/m 2 /h), before decreasing to approximately 2.0 ⁇ 0.2 psi/(L/m 2 /h) at 20 psi.
  • the increase in resistance with increasing TMP at pressures below approximately 14 psi is typical of a compressible fouling deposit.
  • the sharp reduction in resistance when the TMP was increased from 14 to 20 psi is in the opposite direction, with the resistance decreasing with increasing TMP (as in FIGS.20A-20B). As appreciated from FIG.21, the presence of a maximum in the hydraulic resistance of a fouling deposit at intermediate TMP was observed.
  • Example 8 Filtration with Varying Pore Size
  • the effect of the pore size of the underlying membrane on the hydraulic resistance of the deposited LNPs was evaluated by depositing LNPs on 0.2 and 0.4 ⁇ m Isopore membranes as well as on the 0.2 ⁇ m layer of the Sartopore 2 XLG.
  • the LNP was deposited at 20 psi, with the buffer flux then evaluated by step-wise decreasing the TMP; the data for the 0.2 ⁇ m Isopore are taken from the first cycle in FIG.21.
  • a modified procedure was also performed for a 0.1 ⁇ m Isopore membrane, in which the deposit was formed at 2 psi with the TMP then increased step-wise to 40 psi.
  • FIGS.22A-D The resistance versus TMP for all four membranes is shown in FIGS.22A-D; the first cycle alone is shown for ease of illustration. In all instances, the resistance of the clean membrane was much less than that determined for the membrane with the fouling deposit; thus, the resistance data in FIGS.22A-D is representative of the resistance of the deposited LNPs.
  • FIGS.22A-22D show resistance as a function of TMP for deposited LNPs formed on the 0.1 ⁇ m Isopore membrane (FIG.22A), the 0.2 ⁇ m Isopore membrane (FIG.22B), the 0.4 ⁇ m Isopore membranes (FIG.22C) and the 0.2 ⁇ m layer of the Sartopore 2 XLG membrane (FIG.22D).
  • the resistance data for the deposits formed on the 0.1 ⁇ m and 0.2 ⁇ m membranes showed no measurable hysteresis up to 20 psi, while the resistance for the deposits on the 0.4 ⁇ m Isopore and 0.2 ⁇ m Sartopore 2 XLG showed a lower resistance on the second cycle (as in FIG.20B).
  • This behavior was consistent with the lack of measurable LNP concentration in the permeate samples obtained through the 0.1 and 0.2 ⁇ m pore size Isopore membranes, while there was a detectable concentration for the first few permeate samples obtained at high TMP with the larger pore size membranes.
  • the resistance for the deposited LNPs on the 0.1 ⁇ m Isopore and both of the 0.2 ⁇ m membranes exhibits a distinct maximum (Rmax) over the pressure range examined in these experiments, with the pressure value corresponding to Rmax being greatest on the 0.1 ⁇ m Isopore (approximately 32 psi) and smallest (approximately 8 psi) on the Sartopore 2 XLG membrane.
  • Rmax the resistance for the deposited LNPs on the 0.1 ⁇ m Isopore and both of the 0.2 ⁇ m membranes exhibits a distinct maximum (Rmax) over the pressure range examined in these experiments, with the pressure value corresponding to Rmax being greatest on the 0.1 ⁇ m Isopore (approximately 32 psi) and smallest (approximately 8 psi) on the Sartopore 2 XLG membrane.
  • the deposited LNPs exhibit compressible behavior when TMP was less than the pressure value corresponding to Rmax.
  • the resistance of the deposited LNPs formed on the 0.4 ⁇ m Isopore membrane decreases with increasing pressure and shows no regions of compressible behavior at the tested TMPs (i.e., where the pressure corresponding to Rmax ⁇ approximately 2 psi), with the resistance at approximately 20 psi being 6-fold smaller than that at approximately 2 psi.
  • the pressure value corresponding to Rmax represents a threshold value (PCRITICAL) which can be utilized for pressure control, e.g., as an input to a controller as discussed below.
  • FIG.23 depicts the pressure P CRITICAL for the fouled membranes plotted as a function of the inverse of pore size (pore diameter).
  • FIG.23 shows TMP at the maximum resistance evaluated for deposited LNPs formed on the surface of membranes having different pore sizes.
  • the error bars represent the range between the neighboring pressures evaluated experimentally; thus, the PCRITICAL range for the 0.2 ⁇ m Isopore was from approximately 8 to 20 psi based on the results in FIGS.22A-22D.
  • the error bars on the reciprocal pore diameter are determined using the values in Table 1.
  • the threshold pressure corresponding to the maximum in the resistance, P CRITICAL increased with decreasing pore diameter, with the linear dependence on 1/dp in general accordance with the Young-Laplace Equation (Equation (4)).
  • the shaded region below and to the right of the line define the space where the deposited LNPs behave as a compressible foulant layer, with the resistance increasing with increasing TMP.
  • the region above and to the left of the line describes conditions in which the combination of higher TMP and larger pore size allow the amorphous deposit to intrude into and through the pores, effectively opening (unblocking) these pores and reducing the overall resistance to flow.
  • FIG.26 depicts a high-resolution SEM image of deposited LNPs on the 0.2 ⁇ m layer of the Sartopore 2 XLG membrane, according to an aspect of the present disclosure.
  • FIG.26 indicates possible intrusion of the amorphous fouling deposit through some of the pores.
  • regions in the amorphous deposit formed on the 0.2 ⁇ m Isopore membrane appear annularly shaped (in particular, “donut”- shaped) in the SEM image, suggesting that intrusion had occurred through those pores (FIG. 24B).
  • the annular structure is shown in FIG.26.
  • the “donut” shape may represent regions in which the amorphous deposit intruded through larger membrane pores, causing a defect (“donut hole”) in the fouling layer so as to unblock those pores.
  • FIGS.20-26 suggest that the reduction in the resistance of the fouling deposit at high TMP is due at least in part to intrusion of an amorphous deposit into and through the pores in the underlying membrane when the pressure exceeds the intrusion pressure, P C .
  • Intrusion was also observed experimentally by a detectable non-zero concentration in the permeate samples obtained at high TMP, particularly for the larger pore size membranes, and may contribute to the hysteresis observed for the deposited LNPs formed on the larger pore size membranes.
  • a portion of pores that have been intruded into i.e., penetrated by the LNPs 10) may remain open when the TMP is reduced.
  • FIGS.27A-27F are schematic depictions of the pressure dependence of the hydraulic resistance of the deposited LNPs based on the intrusion observation described above.
  • the LNPs deposit and coalesce during filtration forming an amorphous deposit (FIG.27A ⁇ FIG.27B).
  • the deposit reversibly spreads (FIG. 27C FIG.27D), with the hydraulic resistance increasing with increasing TMP.
  • TMP > P CRITICAL the deposit is pushed into and through the membrane pores (FIG.27E ⁇ 27F).
  • a subset of LNPs 10 begin to accumulate on the upper surface of the membrane due to the size-based retention of individual LNPs 10 (FIG. 27A), which may also be affected by surface chemistry-driven adsorption.
  • filtration may be performed to control one or more filtration control parameters, including (i) an operating pressure, (ii) an operating temperature, (iii) a formulation buffer, or (iv) concentration.
  • one or more of these parameters or other parameters may be controlled in view of PCRITICAL, e.g., to increase filter capacity.
  • the foregoing parameters (i)-(iv) may be selected for a given pore size and/or a particle size distribution for the LNPs 10.
  • a controller as discussed below, is configured to receive inputs relating to a plurality of filtration control parameters.
  • the filtration control parameters may be adjusted to increase capacity for a given membrane for filtering of LNPs 10, thereby enhancing efficiency of filtration.
  • the control parameter may be derived or determined based on calculations such as a derivative of a measured value (e.g., any of the measured values discussed herein) or an adjustment to a measured value.
  • filtration control parameters may be maintained as constants or altered, e.g., at different points during a filtration cycle.
  • filtration may be performed to initiate filtering of LNP-containing solution at a pressure approximately equal to PCRITICAL or within a predetermined deviation from PCRITICAL (e.g., 5%, 10%, or 20%).
  • filtration may be performed where P CRITICAL is obtained within a first period before subsequently being lowered.
  • filtration may be controlled at PCRITICAL from the outset of filtration and maintained for at least part or all of a filtration cycle.
  • a controller may be utilized to control one or more filtration control parameters.
  • the controller may include a processor and an input/output interface or terminal.
  • the processor may include one or more processors or one or more processors that include multiple processing cores.
  • the processor may be implemented as a single- or multi-chip processor, a digital signal processor (DSP), an application specific integrated circuit (ASIC), a field programmable gate array (FPGA), or other programmable logic device, discrete gate or transistor logic, discrete hardware components, or any combination thereof designed to perform the functions described herein.
  • DSP digital signal processor
  • ASIC application specific integrated circuit
  • FPGA field programmable gate array
  • a processor may be a microprocessor or any conventional processor, or state machine.
  • a processor may be implemented as a combination of computing devices, such as a combination of a DSP and a microprocessor, a plurality of microprocessors, one or more microprocessors in conjunction with a DSP core, or any other such configuration.
  • the one or more processors may be shared by multiple circuits (e.g., one or more circuits may share the same processor which, in some example embodiments, may execute instructions stored, or otherwise accessed, via different areas of memory).
  • the one or more processors may be structured to perform or otherwise execute certain operations independent of one or more co- processors.
  • two or more processors may be coupled via a bus to enable independent, parallel, pipelined, or multi-threaded instruction execution.
  • the controller may include a memory device that is configured to store machine- readable media.
  • the machine-readable media being readable by the processor in order to execute the programs stored therein.
  • the memory device e.g., memory, memory unit, storage device
  • the memory device may include one or more devices (e.g., RAM, ROM, Flash memory, hard disk storage) for storing data and/or machine-readable media for completing or facilitating the various processes, layers and modules described in the present disclosure.
  • the memory device may be communicatively coupled to the processor to provide computer code, machine-readable media, or instructions to the processor for executing at least some of the processes described herein.
  • the memory device may be or include tangible, non- transient volatile memory or non-volatile memory. Accordingly, the memory device may include database components, object code components, script components, or any other type of information structure for supporting the various activities and information structures described herein.
  • the memory device may include or store a database of target values (e.g., a target pressure, a threshold pressure such as PCRITICAL, or target temperature) or other control parameters.
  • target values e.g., a target pressure, a threshold pressure such as PCRITICAL, or target temperature
  • the controller may be communicably coupled to at least one component of the filtration apparatus to control filtration control parameters including, for example, TMP, a static pressure upstream of the membrane, fluid flux through the membrane, test duration, fluid flows, and others.
  • the controller may form part of a control circuit that includes an input/output (I/O) interface (e.g., a network interface card, a communications interface, etc.) that electrically couples the controller with various actuators and control equipment of the filtration apparatus, such as a pump, linear actuator (e.g., for a syringe), a flow control valve, or another actuator and/or piece of flow control equipment.
  • I/O input/output
  • the control system also includes at least one sensor that is communicably coupled to the controller through the I/O interface and that provide an indication of operating conditions of the filtration apparatus to the controller.
  • the operating conditions may include a static pressure upstream of the membrane, a fluid flux through the membrane, a fluid temperature, or another fluid condition.
  • the controller may be configured to power or otherwise control operation of the actuators and/or control equipment of the filtration apparatus based on sensor data from the at least one sensor.
  • the controller may be configured to send a control signal, via the I/O interface, to a variable speed pump so that a flux through the membrane satisfies (e.g., is approximately equal to, is within a threshold range of, etc.) a target flux.
  • the controller may also be configured to automatically determine characteristics of the fluid, membrane, or fluid/membrane combination.
  • the controller may be configured to iteratively modify the pressure across and/or fluid flux through the membrane to determine hydraulic resistance or other characteristics across a range of fluid control parameters, and to determine fluid control parameters that improve sterile filtration performance.
  • the controller may cause the filtration apparatus to filter the LNPs 10 according to one or more inputs to a program allowing for setting and control of filtration parameters.
  • a programmable pressure limit may be set to a desired static pressure.
  • a control algorithm implemented by the program may utilize the pressure value for P CRITICAL (corresponding to Rmax) as follows.
  • pressure is controlled to reach PCRITICAL and to maintain PCRITICAL for a given duration before declining.
  • pressure is controlled to increase to within a set limit from PCRITICAL, e.g., 10% of P CRITICAL , and then lowered before P CRITICAL is reached.
  • multiple successive filtration cycles may be performed in which PCRITICAL or a set limit from PCRITICAL is reached once per cycle.
  • notifications are provided to an operator or user including whether a given pressure, such as P CRITICAL , is attained.
  • the controller can be configured to allow selection of parameters for notification purposes (e.g., in the form of an alarm, such as an audio, visual or audiovisual notification), where the parameters include but not limited to a retentate volume, a total run time, a back pressure, low and high pressures, or a filtrate or permeate weight alarm, for example.
  • a lipid nanoparticle refers to a nanoscale construct comprising lipid molecules and that comprises in a spherical (e.g., spheroid) geometry.
  • the LNP contains a bleb region, e.g., as described in Brader et al., Biophysical Journal 120: 1- 5 (2021), which is incorporated herein by reference in its entirety.
  • LNPs comprise one or more lipids and a nucleic acid cargo (i.e., polynucleotide) of interest.
  • the LNPs do not comprise a nucleic acid cargo.
  • the lipid is an ionizable lipid (e.g., an ionizable amino lipid), sometimes referred to in the art as an “ionizable cationic lipid”.
  • lipid nanoparticles further comprise other components, including one or more of a phospholipid, a structural lipid, and a molecule capable of reducing particle aggregation, for example a PEG or PEG-modified lipid.
  • the lipid nanoparticle comprises at least one ionizable cationic lipid, at least one non-cationic lipid, at least one sterol, and/or at least one polyethylene glycol (PEG)-modified lipid.
  • the lipid nanoparticles can be generated using components, compositions, and methods as are generally known in the art, see for example PCT/US2016/052352; PCT/US2016/068300; PCT/US2017/037551; PCT/US2015/027400; PCT/US2016/047406; PCT/US2016000129; PCT/US2016/014280; PCT/US2016/014280; PCT/US2017/038426; PCT/US2014/027077; PCT/US2014/055394; PCT/US2016/52117; PCT/US2012/069610; PCT/US2017/027492; PCT/US2016/059575 and PCT/US2016/069491 all of which are incorporated by reference herein in their entireties.
  • the lipid nanoparticle is a lipid nanoparticle described in Intl. Pub. Nos. WO 2013/123523, WO 2012/170930, WO 2011/127255, WO 2008/103276; or U.S. Pub. No.2013/0171646, each of which is herein incorporated by reference in its entirety.
  • the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid, based on the lipid components, e.g., not including the nucleotide cargo.
  • the lipid nanoparticle may comprise a molar ratio of 20-50%, 20-40%, 20-30%, 30-60%, 30-50%, 30-40%, 40-60%, 40-50%, or 50-60% ionizable cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 20%, 30%, 40%, 50, or 60% ionizable cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 5-25% non-cationic lipid, based on the lipid components, e.g., not including the nucleotide cargo.
  • the lipid nanoparticle may comprise a molar ratio of 5-20%, 5-15%, 5-10%, 10- 25%, 10-20%, 10-25%, 15-25%, 15-20%, or 20-25% non-cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 5%, 10%, 15%, 20%, or 25% non-cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 25-55% sterol, based on the lipid components, e.g., not including the nucleotide cargo.
  • the lipid nanoparticle may comprise a molar ratio of 25-50%, 25-45%, 25-40%, 25-35%, 25- 30%, 30-55%, 30-50%, 30-45%, 30-40%, 30-35%, 35-55%, 35-50%, 35-45%, 35-40%, 40- 55%, 40-50%, 40-45%, 45-55%, 45-50%, or 50-55% sterol.
  • the lipid nanoparticle comprises a molar ratio of 25%, 30%, 35%, 40%, 45%, 50%, or 55% sterol.
  • the lipid nanoparticle comprises a molar ratio of 0.5-15% PEG-modified lipid, based on the lipid components, e.g., not including the nucleotide cargo.
  • the lipid nanoparticle may comprise a molar ratio of 0.5-10%, 0.5-5%, 1-15%, 1-10%, 1-5%, 2-15%, 2-10%, 2-5%, 5-15%, 5-10%, or 10-15%.
  • the lipid nanoparticle comprises a molar ratio of 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15% PEG-modified lipid.
  • the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid, 5-25% non-cationic lipid, 25-55% sterol, and 0.5-15% PEG-modified lipid, based on the lipid components, e.g., not including the nucleotide cargo.
  • the lipid nanoparticles described herein have a diameter from about 1 nm to about 100 nm such as, but not limited to, from about 1 nm to about 20 nm, from about 1 nm to about 30 nm, from about 1 nm to about 40 nm, from about 1 nm to about 50 nm, from about 1 nm to about 60 nm, from about 1 nm to about 70 nm, from about 1 nm to about 80 nm, from about 1 nm to about 90 nm, from about 5 nm to about from 100 nm, from about 5 nm to about 10 nm, about 5 nm to about 20 nm, from about 5 nm to about 30 nm, from about 5 nm to about 40 nm, from about 5 nm to about 50 nm, from about 5 nm to about 60 nm, from about 5 nm to about 70 nm, from about 5 nm to about 100 nm
  • the lipid nanoparticles described herein have a diameter from about 10 to 500 nm.
  • the lipid nanoparticle can have a diameter greater than 100 nm, greater than 150 nm, greater than 200 nm, greater than 250 nm, greater than 300 nm, greater than 350 nm, greater than 400 nm, greater than 450 nm, greater than 500 nm, greater than 550 nm, greater than 600 nm, greater than 650 nm, greater than 700 nm, greater than 750 nm, greater than 800 nm, greater than 850 nm, greater than 900 nm, greater than 950 nm or greater than 1000 nm.
  • the ratio between the lipid components of the LNP composition and the polynucleotide cargo can be from about 10:1 to about 60:1 (wt/wt).
  • the ratio between the lipid components and the polynucleotide e.g., mRNA
  • the ratio between the lipid components and the polynucleotide can be about 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, 30:1, 31:1, 32:1, 33:1, 34:1, 35:1, 36:1, 37:1, 38:1, 39:1, 40:1, 41:1, 42:1, 43:1, 44:1, 45:1, 46:1, 47:1, 48:1, 49:1, 50:1, 51:1, 52:1, 53:1, 54:1, 55:1, 56:1, 57:1, 58:1, 59:1 or 60:1 (wt/wt).
  • the wt/wt ratio of the lipid components of the LNP composition to the polynucleotide is about 20:1 or about 15:1.
  • the lipid nanoparticles described herein can comprise polynucleotides (e.g., mRNA) in a lipid:polynucleotide weight ratio of 5:1, 10:1, 15:1, 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, 55:1, 60:1 or 70:1, or a range or any of these ratios such as, but not limited to, 5:1 to about 10:1, from about 5:1 to about 15:1, from about 5:1 to about 20:1, from about 5:1 to about 25:1, from about 5:1 to about 30:1, from about 5:1 to about 35:1, from about 5:1 to about 40:1, from about 5:1 to about 45:1, from about
  • the lipid nanoparticles described herein can comprise the polynucleotide in a concentration from approximately 0.1 mg/ml to 2 mg/ml such as, but not limited to, 0.1 mg/ml, 0.2 mg/ml, 0.3 mg/ml, 0.4 mg/ml, 0.5 mg/ml, 0.6 mg/ml, 0.7 mg/ml, 0.8 mg/ml, 0.9 mg/ml, 1.0 mg/ml, 1.1 mg/ml, 1.2 mg/ml, 1.3 mg/ml, 1.4 mg/ml, 1.5 mg/ml, 1.6 mg/ml, 1.7 mg/ml, 1.8 mg/ml, 1.9 mg/ml, 2.0 mg/ml or greater than 2.0 mg/ml.
  • the composition or pharmaceutical composition disclosed herein can contain more than one polynucleotide.
  • a composition or pharmaceutical composition disclosed herein can contain two or more polynucleotides (e.g., RNA, e.g., mRNA) formulated in the same lipid nanoparticle.
  • a single LNP can contain two or more polynucleotides.
  • a composition or pharmaceutical composition disclosed herein can comprise a mixture of LNPs, each containing one or more polynucleotides, which may be the same or different.
  • LNP1 can contain polynucleotide A; LNP2 can contain polynucleotides B and C; LNP3 can contain polynucleotides D, E, and F; LNP4 can contain polynucleotides E, F, and G, etc.
  • the lipid nanoparticles described herein can be geometrically engineered to modulate macrophage and/or the immune response.
  • the geometrically engineered particles can have varied shapes, sizes and/or surface charges to incorporate the polynucleotides described herein for targeted delivery such as, but not limited to, pulmonary delivery (see, e.g., Intl. Pub. No. WO 2013/082111, herein incorporated by reference in its entirety).
  • the geometrically engineering particles can include are, but are not limited to, fenestrations, angled arms, asymmetry and surface roughness, and charge that can alter the interactions with cells and tissues.
  • the lipid nanoparticles described herein are stealth nanoparticles or target-specific stealth nanoparticles such as, but not limited to, those described in U.S. Pub. No.2013/0172406, herein incorporated by reference in its entirety.
  • the stealth or target-specific stealth nanoparticles can comprise a polymeric matrix, which can comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polyesters, polyanhydrides, polyethers, polyurethanes, polymethacrylates, polyacrylates, polycyanoacrylates, or combinations thereof.
  • polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyester
  • the LNPs can be prepared using microfluidic mixers or micromixers.
  • Exemplary microfluidic mixers can include, but are not limited to, a slit interdigital micromixer including, but not limited to those manufactured by Microinnova (Allerheiligen bei Wildon, Austria) and/or a staggered herringbone micromixer (SHM) (see Zhigaltsevet al., “Bottom-up design and synthesis of limit size lipid nanoparticle systems with aqueous and triglyceride cores using millisecond microfluidic mixing,” Langmuir 28:3633-40 (2012); Belliveau et al., “Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA,” Molecular Therapy-Nucleic Acids.1:e37 (2012); Chen et al., “Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation,” J.
  • methods of making LNP using SHM further comprise mixing at least two input streams wherein mixing occurs by microstructure-induced chaotic advection (MICA).
  • MICA microstructure-induced chaotic advection
  • Methods of generating LNPs using SHM include those disclosed in U.S. Pub. Nos.2004/0262223 and 2012/0276209, each of which is incorporated herein by reference in their entirety.
  • the polynucleotides described herein can be formulated in lipid nanoparticles using microfluidic technology (see Whitesides, George M., “The Origins and the Future of Microfluidics,” Nature 442: 368-373 (2006); and Abraham et al., “Chaotic Mixer for Microchannels,” Science 295: 647-651 (2002)).
  • the polynucleotides can be formulated in lipid nanoparticles using a micromixer chip such as, but not limited to, those from Harvard Apparatus (Holliston, MA) or Dolomite Microfluidics (Royston, UK).
  • a micromixer chip can be used for rapid mixing of two or more fluid streams with a split and recombine mechanism.
  • Ionizable Lipids [0225]
  • the lipid nanoparticles described herein comprise ionizable lipids.
  • the term “ionizable lipid” has its ordinary meaning in the art and may refer to a lipid comprising one or more charged moieties.
  • an ionizable lipid may be positively charged or negatively charged.
  • An ionizable lipid may be positively charged, in which case it can be referred to as “cationic lipid”.
  • an ionizable lipid molecule may comprise an amine group, and can be referred to as an ionizable amino lipid.
  • a “charged moiety” is a chemical moiety that carries a formal electronic charge, e.g., monovalent (+1, or -1), divalent (+2, or -2), trivalent (+3, or -3), etc. The charged moiety may be anionic (i.e., negatively charged) or cationic (i.e., positively charged).
  • Examples of positively-charged moieties include amine groups (e.g., primary, secondary, and/or tertiary amines), ammonium groups, pyridinium group, guanidine groups, and imidizolium groups.
  • the charged moieties comprise amine groups.
  • Examples of negatively charged groups or precursors thereof include carboxylate groups, sulfonate groups, sulfate groups, phosphonate groups, phosphate groups, hydroxyl groups, and the like.
  • the charge of the charged moiety may vary, in some cases, with the environmental conditions, for example, changes in pH may alter the charge of the moiety, and/or cause the moiety to become charged or uncharged. In general, the charge density of the molecule may be selected as desired.
  • the terms “charged” or “charged moiety” does not refer to a “partial negative charge” or “partial positive charge” on a molecule.
  • the terms “partial negative charge” and “partial positive charge” are given its ordinary meaning in the art.
  • a “partial negative charge” may result when a functional group comprises a bond that becomes polarized such that electron density is pulled toward one atom of the bond, creating a partial negative charge on the atom.
  • the ionizable lipid is an ionizable amino lipid, sometimes referred to in the art as an “ionizable cationic lipid”.
  • the ionizable amino lipid may have a positively charged hydrophilic head and a hydrophobic tail that are connected via a linker structure.
  • an ionizable lipid may also be a lipid including a cyclic amine group.
  • the ionizable lipid may be selected from, but not limited to, an ionizable lipid described in International Publication Nos. WO 2013/086354 and WO 2013/116126; each of which is herein incorporated by reference in its entirety.
  • the ionizable lipid may be selected from, but not limited to, formula CLI-CLXXXII of US Patent No.7,404,969; which is herein incorporated by reference in its entirety.
  • the lipid may be a cleavable lipid such as those described in International Publication No. WO 2012/170889, herein incorporated by reference in its entirety.
  • the lipid may be synthesized by methods known in the art and/or as described in International Publication No. WO 2013/086354; each of which is herein incorporated by reference in its entirety.
  • the lipid comprises at least one tertiary amino group, wherein at least one of the three groups of the tertiary amino group comprises a C 6-30 saturated or unsaturated carbon chain optionally interrupted by an –C(O)O– ester group.
  • the disclosure relates to a compound of Formula (I): or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched ; wherein R’ branched is: wherein denotes a point of attachment; wherein R a ⁇ , R a ⁇ , R a ⁇ , and R a ⁇ are each independently selected from the group consisting of H, C 2-12 alkyl, and C 2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C 2-14 alkenyl; R 4 is selected from the group consisting of -(CH 2 ) n OH, wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and wherein denotes a point of attachment; wherein R 10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the
  • R’ a is R’ branched ; R’ branched is ; a ⁇ a ⁇ denotes a point of attachment; R , R , R a ⁇ , and R a ⁇ are each H; R 2 and R 3 are each C1-14 alkyl; R 4 is -(CH2)nOH; n is 2; each R 5 is H; each R 6 is H; M and M’ are each -C(O)O-; R’ is a C 1-12 alkyl; l is 5; and m is 7.
  • R’ a is R’ branched ;
  • R’ branched is denote a ⁇ a ⁇ s a point of attachment;
  • R , R , R a ⁇ , and R a ⁇ are each H;
  • R 2 and R 3 are each C 1-14 alkyl;
  • R 4 is -(CH 2 ) n OH; n is 2;
  • each R 5 is H;
  • each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C1-12 alkyl; l is 3; and m is 7.
  • R’ a is R’ branched ;
  • R’ branched is denotes a point of attachment;
  • R a ⁇ is C2-12 alkyl;
  • R a ⁇ , R a ⁇ , and R a ⁇ are each H;
  • R 2 and R 3 are each C1-14 alkyl;
  • R 4 is R 10 NH(C 1-6 alkyl);
  • n2 is 2;
  • R 5 is H; each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C1-12 alkyl; l is 5; and m is 7.
  • R’ a is R’ branched ;
  • R’ branched is denotes a poi a ⁇ a ⁇ nt of attachment;
  • R , R , and R a ⁇ are each H;
  • R a ⁇ is C2-12 alkyl;
  • R 2 and R 3 are each C1-14 alkyl;
  • R 4 is - (CH2)nOH;
  • n is 2;
  • each R 5 is H; each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C 1-12 alkyl; l is 5; and m is 7.
  • the compound of Formula (I) is selected from: [0239] In some embodiments, the compound of Formula (I) is: [0240] In some embodiments, the compound of Formula (I) is: . [0241] In some embodiments, the compound of Formula (I) is: . [0242] In some embodiments, the compound of Formula (I) is: .
  • the disclosure relates to a compound of Formula (Ia): (Ia) or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched ; wherein R’ branched is: wherein denotes a point of attachment; wherein R a ⁇ , R a ⁇ , and R a ⁇ are each independently selected from the group consisting of H, C 2- 12 alkyl, and C2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C 2-14 alkenyl; R 4 is selected from the group consisting of -(CH 2 ) n OH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and wherein denotes a point of attachment; wherein R 10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consist
  • the disclosure relates to a compound of Formula (Ib): or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched ; wherein R’ branched is: wherein denotes a point of attachment; wherein R a ⁇ , R a ⁇ , R a ⁇ , and R a ⁇ are each independently selected from the group consisting of H, C 2-12 alkyl, and C 2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C 1-14 alkyl and C2-14 alkenyl; R 4 is -(CH2)nOH, wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; each R 5 is independently selected from the group consisting of C 1-3 alkyl, C2-3 alkenyl, and H; each R 6 is independently selected from the group consisting of C1-3 alkyl, C 2-3 alkenyl, and H; M and M’ are each independently selected from the
  • R’ a is R’ branched ;
  • R’ branched is denotes a point of attachment;
  • R a ⁇ , R a ⁇ , and R a ⁇ are each H;
  • R 2 and R 3 are each C1-14 alkyl;
  • R 4 is -(CH 2 ) n OH; n is 2;
  • each R 5 is H;
  • each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C 1-12 alkyl; l is 5; and
  • m is 7.
  • R’ a is R’ branched ;
  • R’ branched is denotes a point of attachment;
  • R a ⁇ , R a ⁇ , a ⁇ and R are each H;
  • R 2 and R 3 are each C1-14 alkyl;
  • R 4 is -(CH2)nOH;
  • n is 2;
  • each R 5 is H;
  • each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C1-12 alkyl; l is 3; and m is 7.
  • R’ a is R’ branched ;
  • R’ branched is denotes a point of attachment;
  • R a ⁇ and R a ⁇ are each H;
  • R a ⁇ is C 2-12 alkyl;
  • R 2 and R 3 are each C 1-14 alkyl;
  • R 4 is -(CH 2 ) n OH;
  • n is 2;
  • each R 5 is H;
  • each R 6 is H;
  • M and M’ are each -C(O)O-;
  • R’ is a C1-12 alkyl; l is 5; and m is 7.
  • the disclosure relates to a compound of Formula (Ic): or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched ; wherein R’ branched is: ; wherein denotes a point of attachment; wherein R a ⁇ , R a ⁇ , R a ⁇ , and R a ⁇ are each independently selected from the group consisting of H, C2-12 alkyl, and C2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C 2-14 alkenyl; R 4 is wherein denotes a point of attachment; wherein R 10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R 5 is independently selected from the group consisting of C 1-3 alkyl, C
  • R’ a is R’ branched ;
  • R’ branched is denotes a point of attachment;
  • R a ⁇ , R a ⁇ , and R a ⁇ are each H;
  • R a ⁇ is C2-12 alkyl;
  • R 2 and R 3 are each C1-14 alkyl;
  • R 4 is denotes a point of attachment;
  • R 10 is NH(C 1-6 alkyl); n2 is 2; each R 5 is H; each R 6 is H; M and M’ are each -C(O)O-;
  • R’ is a C 1-12 alkyl; l is 5; and m is 7.
  • the compound of Formula (Ic) is: [0251]
  • the disclosure relates to a compound of Formula (II): or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein R’ branched is: and R’ cyclic is: ; and wherein denotes a point of attachment; R a ⁇ and R a ⁇ are each independently selected from the group consisting of H, C1-12 alkyl, and C 2-12 alkenyl, wherein at least one of R a ⁇ and R a ⁇ is selected from the group consisting of C 1-12 alkyl and C2-12 alkenyl; R b ⁇ and R b ⁇ are each independently selected from the group consisting of H, C1-12 alkyl, and C 2-12 alkenyl, wherein at least one of R b ⁇ and R b ⁇ is selected from the group consisting of C 1-12 alkyl and C 2-12
  • the disclosure relates to a compound of Formula (II-a): (II-a) or its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein R’ branched is: and R’ b is: wherein denotes a point of attachment; R a ⁇ and R a ⁇ are each independently selected from the group consisting of H, C 1-12 alkyl, and C 2-12 alkenyl, wherein at least one of R a ⁇ and R a ⁇ is selected from the group consisting of C 1-12 alkyl and C2-12 alkenyl; R b ⁇ and R b ⁇ are each independently selected from the group consisting of H, C 1-12 alkyl, and C 2-12 alkenyl, wherein at least one of R b ⁇ and R b ⁇ is selected from the group consisting of C 1-12 alkyl and C2-12 alkenyl; R 2 and R 3 are each independently selected from
  • the disclosure relates to a compound of Formula (II-b): r its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein wherein denotes a point of attachment; R a ⁇ and R b ⁇ are each independently selected from the group consisting of C 1-12 alkyl and C 2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R 4 is selected from the group consisting of -(CH 2 ) n OH wherein n is selected from the group consisting wherein denotes a point of attachment; wherein R 10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1
  • the disclosure relates to a compound of Formula (II-c): r its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein wherein denotes a point of attachment; wherein R a ⁇ is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C 2-14 alkenyl; R 4 is selected from the group consisting of -(CH 2 ) n OH wherein n is selected from the group consisting wherein denotes a point of attachment; wherein R 10 is N(R) 2 ; each R is independently selected from the group consisting of C 1-6 alkyl, C 2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; R’ is a C1-12 alkyl or C
  • the disclosure relates to a compound of Formula (II-d): r its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein wherein denotes a point of attachment; wherein R a ⁇ and R b ⁇ are each independently selected from the group consisting of C 1-12 alkyl and C 2-12 alkenyl; R 4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting wherein denotes a point of attachment; wherein R 10 is N(R) 2 ; each R is independently selected from the group consisting of C 1-6 alkyl, C 2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l
  • the disclosure relates to a compound of Formula (II-e): wherein R’ a is R’ branched or R’ cyclic ; wherein wherein denotes a point of attachment; wherein R a ⁇ is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R 2 and R 3 are each independently selected from the group consisting of C1-14 alkyl and C 2-14 alkenyl; R 4 is -(CH 2 ) n OH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; R’ is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9.
  • each R’ independently is a C1-12 alkyl.
  • each R’ independently is a C 2-5 alkyl.
  • R’ b is: and R 2 and R 3 are each independently a C 1-14 alkyl.
  • R’ b is: and R 2 and R 3 are each independently a C 6-10 alkyl.
  • R’ b is: are each a C8 alkyl.
  • R’ branched is: R 3 are each independently a C6-10 alkyl.
  • R 3 are each independently a C 6-10 alkyl.
  • R’ branched is: is: , R a ⁇ is a C 2-6 alkyl, and R 2 and R 3 are each a C8 alkyl.
  • R’ branched is: b ⁇ is: R are each a C1-12 alkyl.
  • R’ branched is: is: are each a C2-6 alkyl.
  • m and l are each independently selected from 4, 5, and 6 and each R’ independently is a C1-12 alkyl.
  • m and l are each 5 and each R’ independently is a C 2-5 alkyl.
  • R’ branched is: independently selected from 4, 5, and 6, each R’ independently is a C 1-12 alkyl, and R a ⁇ and R b ⁇ are each a C1-12 alkyl.
  • R’ branched is: is: are each 5, each R’ independently is a C2-5 alkyl, and R a ⁇ and R b ⁇ are each a C2-6 alkyl.
  • R’ branched is: , m and l are each independently selected from 4, 5, and 6, R’ is a C 1-12 alkyl, R a ⁇ is a C 1-12 alkyl and R 2 and R 3 are each independently a C6-10 alkyl.
  • R’ branched is: is: are each 5, R’ is a C2-5 alkyl, R a ⁇ is a C 2-6 alkyl, and R 2 and R 3 are each a C 8 alkyl.
  • R’ branched is: are each independently selected from 4, 5, and 6, each R’ independently is a C1-12 alkyl, R a ⁇ and R b ⁇ are each a C 1-12 alkyl, 1 wherein R 0 is NH(C 1-6 alkyl), and n2 is 2.
  • R’ branched is: are each 5, each R’ independently is a C2-5 alkyl, R a ⁇ and R b ⁇ are each a C2-6 alkyl, and R 4 is , wherein R 10 is NH(CH 3 ) and n2 is 2.
  • R’ branched is: and R’ b is: m and l are each independently selected from 4, 5, and 6, R’ is a C 1-12 alkyl, R 2 and R 3 are each independently a C 6-10 alkyl, R a ⁇ is a C1-12 alkyl, wherein R 10 is NH(C1-6 alkyl) and n2 is 2.
  • R’ branched is: alkyl, R a ⁇ is a C 2-6 alkyl, R 2 and R 3 are each a C 8 alkyl, wherein R 10 is NH(CH3) and n2 is 2.
  • R 4 is -(CH 2 ) n OH and n is 2, 3, or 4.
  • R 4 is -(CH2)nOH and n is 2.
  • R’ branched is: is: are each independently selected from 4, 5, and 6, each R’ independently is a C1-12 alkyl, R a ⁇ and R b ⁇ are each a C 1-12 alkyl, R 4 is -(CH 2 ) n OH, and n is 2, 3, or 4.
  • R’ branched is: each 5, each R’ independently is a C 2-5 alkyl, R a ⁇ and R b ⁇ are each a C 2-6 alkyl, R 4 is -(CH 2 ) n OH, and n is 2.
  • the disclosure relates to a compound of Formula (II-f): r its N-oxide, or a salt or isomer thereof, wherein R’ a is R’ branched or R’ cyclic ; wherein R’ branched is: and R’ b is: wherein denotes a point of attachment; R a ⁇ is a C 1-12 alkyl; R 2 and R 3 are each independently a C 1-14 alkyl; R 4 is - (CH 2 ) n OH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; R’ is a C 1-12 alkyl; m is selected from 4, 5, and 6; and l is selected from 4, 5, and 6.
  • m and l are each 5, and n is 2, 3, or 4.
  • R’ is a C2-5 alkyl, R a ⁇ is a C2-6 alkyl, and R 2 and R 3 are each a C6-10 alkyl.
  • m and l are each 5, n is 2, 3, or 4, R’ is a C2-5 alkyl, R a ⁇ is a C2-6 alkyl, and R 2 and R 3 are each a C6-10 alkyl.
  • the disclosure relates to a compound of Formula (II-g): wherein R a ⁇ is a C 2-6 alkyl; R’ is a C 2-5 alkyl; and R 4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group wherein denotes a point of attachment, R 10 is NH(C 1-6 alkyl), and n2 is selected from the group consisting of 1, 2, and 3.
  • the disclosure relates to a compound of Formula (II-h): wherein R a ⁇ and R b ⁇ are each independently a C2-6 alkyl; each R’ independently is a C 2-5 alkyl; and R 4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group wherein denotes a point of attachment, R 10 is NH(C1-6 alkyl), and n2 is selected from the group consisting of 1, 2, and 3. [0288] In some embodiments of the compound of Formula (II-g) or (II-h), R 4 is , wherein R 10 is NH(CH 3 ) and n2 is 2.
  • R 4 is - (CH 2 ) 2 OH.
  • the disclosure relates to a compound having the Formula (III): or a salt or isomer thereof, wherein R1, R2, R3, R4, and R5 are independently selected from the group consisting of C5-20 alkyl, C5- 20 alkenyl, -R”MR’, -R*YR”, -YR”, and -R*OR”; each M is independently selected from the group consisting of -C(O)O-, -OC(O)-, -OC(O)O-, -C(O)N(R’)-, -N(R’)C(O)-, -C(O)-, -C(S)-, -C(S)S-, -SC(S) -, -CH(OH)-, -P(O)(OR’)O-
  • R1, R2, R3, R4, and R5 are each C5-20 alkyl; X 1 is -CH2-; and X 2 and X 3 are each -C(O)-.
  • the compound of Formula (III) is: .
  • the ionizable lipids are one or more of the compounds described in WO 2020/146805; WO 2020/081938; WO 2020/214946; WO 2019/036030; WO 2019/036000; WO 2019/036028; WO 2019/036008; WO 2018/200943; WO 2018/191657; WO 2017/117528; WO 2017/075531; WO 2017/004143; WO 2015/199952; and WO 2015/074085; each of which is incorporated herein in its entirety.
  • the ionizable lipids are one or more of the compounds described in WO 2020/214946 having structure: or a pharmaceutically acceptable salt thereof, wherein each R la is independently hydrogen, R 1c , or R 3d ; each R lb is independently R lc or R ld ; each R lc is Independently –(CH)2C(O)X 1 R 3 ; each R ld is independently -C(O)R 4 ; each R 2 is independently -[C(R 2a ) 2 ] c ;R 2b ; each R 2a is independently hydrogen or lower alkyl (e.g., C1-C6alkyl); R 2b is -N(L I -B) 2 , -(OCH 2 CH 2 ) 6 OH; or -(OCH 2 CH 2 ) 6 OCH 3 ; each R 3 and R 4 is independently aliphatic (e.g., C 6 -C 30 aliphatic);
  • the ionizable lipids are one or more of the compounds described in WO 2015/074085 having the structure: wherein R 1 and R 2 are the same or different, each a linear or branched alkyl with 1-9 carbons, or an alkenyl or alkynyl with 2 to 11 carbon atoms, L1 and L2 are the same or different, each a linear alkyl having 5 to 18 carbon atoms, or form a heterocycle with N, X1 is a bond, or is -CO-O- whereby L2-CO-O-R2 is formed X 2 is S or O, L 3 is a bond or a lower alkyl, or form a heterocycle with N, R3 is a lower alkyl, and R4 and R5 are the same or different, each a lower alkyl; or a pharmaceutically acceptable salt thereof.
  • the ionizable lipids are one or more of the compounds described in Buschmann, M. D. et al., Vaccines, 2021, 9, 65, which incorporated herein in its entirety (the structures provided below include their theoretical pKas):
  • the ionizable lipid is selected from Compound II, Compound III, Compound VI, Compound A1, and Compound A2:
  • Polynucleotides for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, RNA including messenger RNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix formation, aptamers, vectors, etc., described in detail herein.
  • the polynucleotide is RNA.
  • the polynucleotide is mRNA.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises from about 900 to about 100,000 nucleotides (e.g., from 900 to 1,000, from 900 to 1,100, from 900 to 1,200, from 900 to 1,300, from 900 to 1,400, from 900 to 1,500, from 1,000 to 1,100, from 1,000 to 1,100, from 1,000 to 1,200, from 1,000 to 1,300, from 1,000 to 1,400, from 1,000 to 1,500, from 1,187 to 1,200, from 1,187 to 1,400, from 1,187 to 1,600, from 1,187 to 1,800, from 1,187 to 2,000, from 1,187 to 3,000, from 1,187 to 5,000, from 1,187 to 7,000, from 1,187 to 10,000, from 1,187 to 25,000, from 1,187 to 50,000, from 1,187 to 70,000, or from 1,187 to 100,000 nucleotides).
  • nucleotides e.g., from 900 to 1,000, from 900 to 1,100,
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a nucleotide sequence (e.g., an open reading frame (ORF)) encoding a polypeptide, wherein the length of the nucleotide sequence (e.g., an ORF) is at least 500 nucleotides in length, e.g., at least about 500, 600, 700, 80, 900, 1,000, 1,050, 1,100, 1,187, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,100, 2,200, 2,300, 2,400, 2,500, 2,600, 2,700, 2,800, 2,900, 3,000, 3,100, 3,200, 3,300, 3,400, 3,500, 3,600, 3,700, 3,800, 3,900, 4,000, 4,100, 4,200, 4,300, 4,400, 4,500, 4,600, 4,700, 4,800, 4,800
  • the length is up to and including 100,000 nucleotides.
  • the polynucleotide of the composition that comprises a nucleotide sequence (e.g., an ORF) encoding a polypeptide is DNA.
  • the polynucleotide of the composition is RNA.
  • the polynucleotide is, or functions as, an mRNA.
  • the mRNA comprises a nucleotide sequence (e.g., an ORF) that encodes at least one polypeptide, and is capable of being translated to produce the encoded polypeptide in vitro, in vivo, in situ or ex vivo.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a nucleotide sequence (e.g., an ORF) encoding a polypeptide and further comprises at least one nucleic acid sequence that is noncoding, e.g., a microRNA binding site, e.g., a miRNA binding site that binds to miR-142 and/or a miRNA binding site that binds to miR-126.
  • a nucleotide sequence e.g., an ORF
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a 5′-UTR and a 3′UTR.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a 5′ terminal cap.
  • Nonlimiting examples of 5′ terminal caps include Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2′-fluoro-guanosine, 7-deaza- guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, 2-azidoguanosine, Cap2, Cap4, 5′ methylG cap, or an analog thereof.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a poly-A-tail.
  • the polyA tail is about 100 nucleotides in length.
  • the poly A tail is 100 nucleotides in length. In some instances, the poly A tail is 50-150, 75-150, 85-150, 90-150, 90-120, 90-130, or 90-150 nucleotides in length.
  • the polynucleotides e.g., a RNA, e.g., an mRNA
  • One such feature that aids in protein trafficking is the signal sequence, or targeting sequence.
  • the peptides encoded by these signal sequences are known by a variety of names, including targeting peptides, transit peptides, and signal peptides.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) comprises a nucleotide sequence (e.g., an ORF) that encodes a signal peptide operably linked to a nucleotide sequence that encodes a polypeptide of interest, such as a therapeutic polypeptide,.
  • a nucleotide sequence e.g., an ORF
  • the “signal sequence” or “signal peptide” is a polynucleotide or polypeptide, respectively, which is from about 30-210, e.g., about 45-80 or 15-60 nucleotides (e.g., about 20, 30, 40, 50, 60, or 70 amino acids) in length that, optionally, is incorporated at the 5′ (or N-terminus) of the coding region or the polypeptide, respectively. Addition of these sequences results in trafficking the encoded polypeptide to a desired site, such as the endoplasmic reticulum or the mitochondria through one or more targeting pathways.
  • a desired site such as the endoplasmic reticulum or the mitochondria through one or more targeting pathways.
  • the polynucleotide e.g., a RNA, e.g., an mRNA
  • a RNA e.g., an mRNA
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) comprises a 5′ cap, a 5′-UTR, a nucleotide sequence (e.g., an ORF, e.g., a sequence optimized ORF) encoding a polypeptide, a 3′-UTR, and a polyA tail, or any combination thereof, the 5′ UTR or 3′ UTR optionally comprising at least one microRNA binding site.
  • a nucleotide sequence e.g., an ORF, e.g., a sequence optimized ORF
  • a sequence-optimized nucleotide sequence e.g., a codon-optimized mRNA sequence encoding a polypeptide, is a sequence comprising at least one synonymous nucleobase substitution with respect to a reference sequence (e.g., a wild type nucleotide sequence encoding the polypeptide).
  • a sequence-optimized nucleotide sequence can be partially or completely different in sequence from the reference sequence.
  • a reference sequence encoding polyserine uniformly encoded by UCU codons can be sequence-optimized by having 100% of its nucleobases substituted (for each codon, U in position 1 replaced by A, C in position 2 replaced by G, and U in position 3 replaced by C) to yield a sequence encoding polyserine which would be uniformly encoded by AGC codons.
  • the percentage of sequence identity obtained from a global pairwise alignment between the reference polyserine nucleic acid sequence and the sequence-optimized polyserine nucleic acid sequence would be 0%.
  • the protein products from both sequences would be 100% identical.
  • sequence optimization also sometimes referred to codon optimization
  • results can include, e.g., matching codon frequencies in certain tissue targets and/or host organisms to ensure proper folding; biasing G/C content to increase mRNA stability or reduce secondary structures; minimizing tandem repeat codons or base runs that can impair gene construction or expression; customizing transcriptional and translational control regions; inserting or removing protein trafficking sequences; removing/adding post translation modification sites in an encoded protein (e.g., glycosylation sites); adding, removing or shuffling protein domains; inserting or deleting restriction sites; modifying ribosome binding sites and mRNA degradation sites; adjusting translational rates to allow the various domains of the protein to fold properly; and/or reducing or eliminating problem secondary structures within the polynucleotide.
  • sequence optimization tools, algorithms and services are known in the art, non-limiting examples include services from GeneArt (Life Technologies), DNA2.0 (Menlo Park CA) and/or are proprietary methods. [0326] In some embodiments, the sequence optimization method is multiparametric and comprises one, two, three, four, or more methods disclosed herein and/or other optimization methods known in the art.
  • a polynucleotide (e.g., a RNA, e.g., an mRNA) of the composition comprises a sequence-optimized nucleotide sequence (e.g., an ORF) encoding a polypeptide, wherein the polypeptide encoded by the sequence-optimized nucleotide sequence has improved properties (e.g., compared to the polypeptide as encoded by a reference nucleotide sequence that is not sequence optimized), e.g., improved properties related to expression efficacy after administration in vivo.
  • a sequence-optimized nucleotide sequence e.g., an ORF
  • Such properties include, but are not limited to, improving nucleic acid stability (e.g., mRNA stability), increasing translation efficacy in the target tissue, reducing the number of truncated proteins expressed, improving the folding or preventing misfolding of the expressed proteins, reducing toxicity of the expressed products, reducing cell death caused by the expressed products, increasing and/or decreasing protein aggregation.
  • nucleic acid stability e.g., mRNA stability
  • increasing translation efficacy in the target tissue reducing the number of truncated proteins expressed, improving the folding or preventing misfolding of the expressed proteins, reducing toxicity of the expressed products, reducing cell death caused by the expressed products, increasing and/or decreasing protein aggregation.
  • the sequence-optimized nucleotide sequence (e.g., an ORF) is codon optimized for expression in human subjects, having structural and/or chemical features that avoid one or more of the problems in the art, for example, features which are useful for optimizing formulation and delivery of nucleic acid-based therapeutics while retaining structural and functional integrity; overcoming a threshold of expression; improving expression rates; half-life and/or protein concentrations; optimizing protein localization; and avoiding deleterious bio-responses such as the immune response and/or degradation pathways.
  • Methods for optimizing codon usage are known in the art.
  • an ORF of any one or more of the sequences provided herein may be codon optimized.
  • Codon optimization may be used to match codon frequencies in target and host organisms to ensure proper folding; bias GC content to increase mRNA stability or reduce secondary structures; minimize tandem repeat codons or base runs that may impair gene construction or expression; customize transcriptional and translational control regions; insert or remove protein trafficking sequences; remove/add post translation modification sites in encoded protein (e.g., glycosylation sites); add, remove or shuffle protein domains; insert or delete restriction sites; modify ribosome binding sites and mRNA degradation sites; adjust translational rates to allow the various domains of the protein to fold properly; or reduce or eliminate problem secondary structures within the polynucleotide.
  • encoded protein e.g., glycosylation sites
  • add, remove or shuffle protein domains add or delete restriction sites
  • modify ribosome binding sites and mRNA degradation sites adjust translational rates to allow the various domains of the protein to fold properly; or reduce or eliminate problem secondary structures within the polynucleotide.
  • Codon optimization tools, algorithms and services are known in the art - non-limiting examples include services from GeneArt (Life Technologies), DNA2.0 (Menlo Park CA) and/or are proprietary methods.
  • the open reading frame (ORF) sequence is optimized using optimization algorithms.
  • Features, which can be considered beneficial in some embodiments, can be encoded by or within regions of the polynucleotide and such regions can be upstream (5′) to, downstream (3′) to, or within the region that encodes the polypeptide. These regions can be incorporated into the polynucleotide before and/or after sequence-optimization of the protein encoding region or open reading frame (ORF).
  • the polynucleotide comprises a 5′ UTR, a 3′ UTR and/or a microRNA binding site. In some embodiments, the polynucleotide comprises two or more 5′ UTRs and/or 3′ UTRs, which can be the same or different sequences. In some embodiments, the polynucleotide comprises two or more microRNA binding sites, which can be the same or different sequences.
  • any portion of the 5′ UTR, 3′ UTR, and/or microRNA binding site, including none, can be sequence-optimized and can independently contain one or more different structural or chemical modifications, before and/or after sequence optimization.
  • the polynucleotides of the compositions are modified.
  • the modified polynucleotides can be chemically modified and/or structurally modified.
  • modified polynucleotides When the polynucleotides of the compositions are chemically and/or structurally modified the polynucleotides can be referred to as “modified polynucleotides.”
  • modified nucleosides and modified nucleotides of a polynucleotide e.g., RNA polynucleotides, such as mRNA polynucleotides.
  • nucleoside refers to a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as “nucleobase”).
  • organic base e.g., a purine or pyrimidine
  • nucleobase also referred to herein as “nucleobase”.
  • nucleotide refers to a nucleoside including a phosphate group. Modified nucleotides can be synthesized by any useful method, such as, for example, chemically, enzymatically, or recombinantly, to include one or more modified or non-natural nucleosides. Polynucleotides can comprise a region or regions of linked nucleosides.
  • modified polynucleotides disclosed herein can comprise various distinct modifications.
  • the modified polynucleotides contain one, two, or more (optionally different) nucleoside or nucleotide modifications.
  • a modified polynucleotide, introduced to a cell can exhibit one or more desirable properties, e.g., improved protein expression, reduced immunogenicity, or reduced degradation in the cell, as compared to an unmodified polynucleotide.
  • a polynucleotide of the lipid nanoparticles are structurally modified.
  • a “structural” modification is one in which two or more linked nucleosides are inserted, deleted, duplicated, inverted or randomized in a polynucleotide without significant chemical modification to the nucleotides themselves. Because chemical bonds will necessarily be broken and reformed to effect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides. For example, the polynucleotide “ATCG” can be chemically modified to “AT-5meC-G”.
  • Therapeutic lipid nanoparticles comprise, in some embodiments, at least one nucleic acid (e.g., RNA), wherein the nucleic acid comprises nucleotides and/or nucleosides that can be standard (unmodified) or modified as is known in the art.
  • nucleotides and nucleosides comprise modified nucleotides or nucleosides.
  • modified nucleotides and nucleosides can be naturally-occurring modified nucleotides and nucleosides or non-naturally occurring modified nucleotides and nucleosides. Such modifications can include those at the sugar, backbone, or nucleobase portion of the nucleotide and/or nucleoside as are recognized in the art. [0337] In some embodiments, a naturally-occurring modified nucleotide or nucleoside is one as is generally known or recognized in the art. Non-limiting examples of such naturally occurring modified nucleotides and nucleosides can be found, inter alia, in the MODOMICS database.
  • a non-naturally occurring modified nucleotide or nucleoside is one as is generally known or recognized in the art.
  • Non-limiting examples of such non-naturally occurring modified nucleotides and nucleosides can be found, inter alia, in published International Patent Application Nos. PCT/US2012/058519; PCT/US2013/075177; PCT/US2014/058897; PCT/US2014/058891; PCT/US2014/070413; PCT/US2015/36773; PCT/US2015/36759; PCT/US2015/36771; or PCT/IB2017/051367, each of which is incorporated by reference herein in its entirety.
  • RNA e.g., mRNA
  • nucleotides and nucleosides comprise standard nucleoside residues such as those present in transcribed RNA (e.g. A, G, C, or U).
  • nucleotides and nucleosides comprise standard deoxyribonucleosides such as those present in DNA (e.g. dA, dG, dC, or dT).
  • nucleic acids e.g., DNA nucleic acids and RNA nucleic acids, such as mRNA nucleic acids
  • nucleic acids can comprise standard nucleotides and nucleosides, naturally-occurring nucleotides and nucleosides, non-naturally-occurring nucleotides and nucleosides, or any combination thereof.
  • Nucleic acids e.g., DNA nucleic acids and RNA nucleic acids, such as mRNA nucleic acids
  • a particular region of a nucleic acid contains one, two or more (optionally different) types of standard and/or modified nucleotides and nucleosides.
  • a modified RNA nucleic acid e.g., a modified mRNA nucleic acid
  • introduced to a cell or organism exhibits reduced degradation in the cell or organism, respectively, relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides.
  • a modified RNA nucleic acid (e.g., a modified mRNA nucleic acid), introduced into a cell or organism, may exhibit reduced immunogenicity in the cell or organism, respectively (e.g., a reduced innate response) relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides.
  • Nucleic acids e.g., RNA nucleic acids, such as mRNA nucleic acids
  • Nucleic acids in some embodiments, comprise non-natural modified nucleotides that are introduced during synthesis or post-synthesis of the nucleic acids to achieve desired functions or properties. The modifications may be present on internucleotide linkages, purine or pyrimidine bases, or sugars.
  • Modified nucleotide base pairing encompasses not only the standard adenosine- thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures, such as in those nucleic acids having at least one chemical modification.
  • modified nucleobases in nucleic acids comprise N1-methyl-pseudouridine (m1 ⁇ ), 1-ethyl- pseudouridine (e1 ⁇ ), 5-methoxy-uridine (mo5U), 5-methyl-cytidine (m5C), and/or pseudouridine ( ⁇ ).
  • modified nucleobases in nucleic acids comprise 5-methoxymethyl uridine, 5-methylthio uridine, 1-methoxymethyl pseudouridine, 5-methyl cytidine, and/or 5-methoxy cytidine.
  • the polyribonucleotide includes a combination of at least two (e.g., 2, 3, 4 or more) of any of the aforementioned modified nucleobases, including but not limited to chemical modifications.
  • nucleic acids e.g., RNA nucleic acids, such as mRNA nucleic acids
  • RNA nucleic acids are uniformly modified (e.g., fully modified, modified throughout the entire sequence) for a particular modification.
  • a nucleic acid can be uniformly modified with N1-methyl-pseudouridine, meaning that all uridine residues in the mRNA sequence are replaced with N1-methyl-pseudouridine.
  • a nucleic acid can be uniformly modified for any type of nucleoside residue present in the sequence by replacement with a modified residue such as those set forth above.
  • the nucleic acids may be partially or fully modified along the entire length of the molecule.
  • one or more or all or a given type of nucleotide may be uniformly modified in a nucleic acid, or in a predetermined sequence region thereof (e.g., in the mRNA including or excluding the polyA tail).
  • all nucleotides X in a nucleic acid are modified nucleotides, wherein X may be any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
  • the nucleic acid may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e., any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 50% to 100%
  • the nucleic acids may contain at a minimum 1% and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50% modified nucleotides, at least 80% modified nucleotides, or at least 90% modified nucleotides.
  • the nucleic acids may contain a modified pyrimidine such as a modified uracil or cytosine.
  • At least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid is replaced with a modified uracil (e.g., a 5-substituted uracil).
  • the modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • cytosine in the nucleic acid is replaced with a modified cytosine (e.g., a 5-substituted cytosine).
  • the modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • the polynucleotides of the lipid nanoparticles can be generated using components, compositions, and methods as are generally known in the art, see, e.g., International Patent Application Publication Nos.
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • IVT in vitro transcription
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • a polynucleotide disclosed herein can be constructed by chemical synthesis using an oligonucleotide synthesizer.
  • a polynucleotide (e.g., a RNA, e.g., an mRNA) disclosed herein is made by using a host cell.
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • a polynucleotide is made by one or more combination of the IVT, chemical synthesis, host cell expression, or any other methods known in the art.
  • Naturally occurring nucleosides, non-naturally occurring nucleosides, or combinations thereof, can totally or partially naturally replace occurring nucleosides present in the candidate nucleotide sequence and can be incorporated into a sequence-optimized nucleotide sequence (e.g., a RNA, e.g., an mRNA).
  • the resultant polynucleotides e.g., mRNAs, can then be examined for their ability to produce protein and/or produce a therapeutic outcome.
  • the polynucleotides can be purified prior to their inclusion in the lipid nanoparticles. Purification of the polynucleotides described herein can include, but is not limited to, polynucleotide clean-up, quality assurance and quality control.
  • Clean-up can be performed by methods known in the arts such as, but not limited to, AGENCOURT® beads (Beckman Coulter Genomics, Danvers, MA), poly-T beads, LNATM oligo-T capture probes (EXIQON® Inc., Vedbaek, Denmark) or HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC).
  • purification of a polynucleotide removes impurities that can reduce or remove an unwanted immune response, e.g., reducing cytokine activity.
  • the polynucleotide is purified prior to inclusion in a lipid nanoparticle using column chromatography (e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC- HPLC), or (LCMS)).
  • column chromatography e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC- HPLC), or (LCMS)
  • the purified polynucleotide is at least about 80% pure, at least about 85% pure, at least about 90% pure, at least about 95% pure, at least about 96% pure, at least about 97% pure, at least about 98% pure, at least about 99% pure, or about 100% pure prior to inclusion in a lipid nanoparticle.
  • a quality assurance and/or quality control check can be conducted using methods such as, but not limited to, gel electrophoresis, UV absorbance, or analytical HPLC.
  • the polynucleotide can be sequenced by methods including, but not limited to reverse-transcriptase-PCR. 2) Quantification of Polynucleotides [0360] In some embodiments, the polynucleotides, their expression products, as well as degradation products and metabolites can be quantified according to methods known in the art. [0361] In some embodiments, the polynucleotide can be quantified using methods such as, but not limited to, ultraviolet visible spectroscopy (UV/Vis).
  • UV/Vis ultraviolet visible spectroscopy
  • a non-limiting example of a UV/Vis spectrometer is a NANODROP® spectrometer (ThermoFisher, Waltham, MA).
  • the quantified polynucleotide can be analyzed in order to determine if the polynucleotide can be of proper size, or to check that no degradation of the polynucleotide has occurred.
  • Degradation of the polynucleotide can be checked by methods (such as, but not limited to, agarose gel electrophoresis, HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP- HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE), capillary gel electrophoresis (CGE)); and UPLC (e.g., RP-UPLC).
  • HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP- HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE), capillary gel electrophoresis (CGE)); and UPLC (e.g., RP-UP
  • the lipid composition of a lipid nanoparticle disclosed herein can include one or more components in addition to the lipid components described above (e.g., in addition to the ionizable cationic lipid, non-cationic lipid, sterol, and PEG-modified lipid).
  • the lipid composition can include one or more permeability enhancer molecules, carbohydrates, polymers, surface altering agents (e.g., surfactants), or other components.
  • a permeability enhancer molecule can be a molecule described by U.S. Patent Application Publication No.2005/0222064.
  • Carbohydrates can include simple sugars (e.g., glucose) and polysaccharides (e.g., glycogen and derivatives and analogs thereof).
  • a polymer can be included in and/or used to encapsulate or partially encapsulate a composition disclosed herein (e.g., an LNP composition).
  • a polymer can be biodegradable and/or biocompatible.
  • a polymer can be selected from, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, polystyrenes, polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyleneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates.
  • the LNP can further contain a phosphate conjugate.
  • the phosphate conjugate can increase in vivo circulation times and/or increase the targeted delivery of the nanoparticle.
  • Phosphate conjugates can be made by the methods described in, e.g., Intl. Pub. No.
  • the LNP can also contain a polymer conjugate (e.g., a water soluble conjugate) as described in, e.g., U.S. Pub. Nos.2013/0059360, 2013/0196948, and 2013/0072709. Each of the references is herein incorporated by reference in its entirety.
  • the LNPs can comprise a conjugate to enhance the delivery of nanoparticles in a subject. Further, the conjugate can inhibit phagocytic clearance of the nanoparticles in a subject.
  • the conjugate can be a “self” peptide designed from the human membrane protein CD47 (e.g., the “self” particles described by Rodriguez et al, Science 2013339, 971-975, herein incorporated by reference in its entirety). As shown by Rodriguez et al. the self peptides delayed macrophage-mediated clearance of nanoparticles which enhanced delivery of the nanoparticles.
  • the LNPs can comprise a carbohydrate carrier.
  • the carbohydrate carrier can include, but is not limited to, an anhydride-modified phytoglycogen or glycogen-type material, phytoglycogen octenyl succinate, phytoglycogen beta-dextrin, anhydride-modified phytoglycogen beta-dextrin (e.g., Intl. Pub. No. WO 2012/109121, herein incorporated by reference in its entirety).
  • the LNPs can be coated with a surfactant or polymer to improve the delivery of the particle.
  • the LNP can be coated with a hydrophilic coating such as, but not limited to, PEG coatings and/or coatings that have a neutral surface charge as described in U.S.
  • the LNPs can be engineered to alter the surface properties of particles so that the lipid nanoparticles can penetrate the mucosal barrier as described in U.S. Pat. No.8,241,670 or Intl. Pub. No. WO 2013/110028, each of which is herein incorporated by reference in its entirety.
  • the LNPs engineered to penetrate mucus can comprise a polymeric material (e.g., a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer.
  • the polymeric material can include, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates.
  • LNPs engineered to penetrate mucus can also include surface altering agents such as, but not limited to, polynucleotides, anionic proteins (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin
  • the mucus penetrating LNP can be a hypotonic formulation comprising a mucosal penetration enhancing coating.
  • the formulation can be hypotonic for the epithelium to which it is being delivered.
  • hypotonic formulations can be found in, e.g., Intl. Pub. No. WO 2013/110028, herein incorporated by reference in its entirety.
  • Other Lipids a) Phospholipids
  • the lipid composition of a lipid nanoparticle disclosed herein can comprise one or more phospholipids, for example, one or more saturated or (poly)unsaturated phospholipids or a combination thereof.
  • phospholipids comprise a phospholipid moiety and one or more fatty acid moieties.
  • a phospholipid moiety can be selected, for example, from the non-limiting group consisting of phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol, phosphatidyl serine, phosphatidic acid, 2-lysophosphatidyl choline, and a sphingomyelin.
  • a fatty acid moiety can be selected, for example, from the non-limiting group consisting of lauric acid, myristic acid, myristoleic acid, palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, alpha-linolenic acid, erucic acid, phytanoic acid, arachidic acid, arachidonic acid, eicosapentaenoic acid, behenic acid, docosapentaenoic acid, and docosahexaenoic acid.
  • Particular phospholipids can facilitate fusion to a membrane.
  • a cationic phospholipid can interact with one or more negatively charged phospholipids of a membrane (e.g., a cellular or intracellular membrane). Fusion of a phospholipid to a membrane can allow one or more elements (e.g., a therapeutic agent) of a lipid-containing composition (e.g., LNPs) to pass through the membrane permitting, e.g., delivery of the one or more elements to a target tissue.
  • elements e.g., a therapeutic agent
  • a lipid-containing composition e.g., LNPs
  • Non-natural phospholipid species including natural species with modifications and substitutions including branching, oxidation, cyclization, and alkynes are also contemplated.
  • a phospholipid can be functionalized with or cross-linked to one or more alkynes (e.g., an alkenyl group in which one or more double bonds is replaced with a triple bond).
  • alkynes e.g., an alkenyl group in which one or more double bonds is replaced with a triple bond.
  • an alkyne group can undergo a copper- catalyzed cycloaddition upon exposure to an azide.
  • Such reactions can be useful in functionalizing a lipid bilayer of a nanoparticle composition to facilitate membrane permeation or cellular recognition or in conjugating a nanoparticle composition to a useful component such as a targeting or imaging moiety (e.g., a dye).
  • Phospholipids include, but are not limited to, glycerophospholipids such as phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines, phosphatidylinositols, phosphatidy glycerols, and phosphatidic acids. Phospholipids also include phosphosphingolipid, such as sphingomyelin.
  • a phospholipid comprises 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2- dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-gly cero- phosphocholine (DMPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), l,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-di- O-octadecenyl-sn-glycero
  • a phospholipid useful or potentially useful in the present disclosure is an analog or variant of DSPC.
  • a phospholipid useful or potentially useful in the present disclosure is a compound of Formula (IV): or a salt thereof, wherein: each R 1 is independently optionally substituted alkyl; or optionally two R 1 are joined together with the intervening atoms to form optionally substituted monocyclic carbocyclyl or optionally substituted monocyclic heterocyclyl; or optionally three R 1 are joined together with the intervening atoms to form optionally substituted bicyclic carbocyclyl or optionally substitute bicyclic heterocyclyl; n is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; m is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; A is of the formula: each instance of L 2 is independently a bond or optionally substituted C1-6 alkylene, wherein one methylene unit of the optionally substituted C 1-6 alkylene is optionally replaced with O, N(R N)
  • the phospholipids may be one or more of the phospholipids described in PCT/US2018/037922 (published as WO 2018232357).
  • A) Phospholipid Head Modifications [0380]
  • a phospholipid useful or potentially useful in the present disclosure comprises a modified phospholipid head (e.g., a modified choline group).
  • a phospholipid with a modified head is DSPC, or analog thereof, with a modified quaternary amine.
  • at least one of R 1 is not methyl. In certain embodiments, at least one of R 1 is not hydrogen or methyl.
  • the compound of Formula (IV) is of one of the following formulae: , or a salt thereof, wherein: each t is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; each u is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and each v is independently 1, 2, or 3.
  • a compound of Formula (IV) is of Formula (IV-a): salt thereof.
  • a phospholipid useful or potentially useful in the present disclosure comprises a cyclic moiety in place of the glyceride moiety.
  • a phospholipid useful in the present disclosure is DSPC, or analog thereof, with a cyclic moiety in place of the glyceride moiety.
  • a phospholipid useful or potentially useful in the present disclosure comprises a modified tail.
  • a phospholipid useful or potentially useful in the present disclosure is DSPC, or analog thereof, with a modified tail.
  • a “modified tail” may be a tail with shorter or longer aliphatic chains, aliphatic chains with branching introduced, aliphatic chains with substituents introduced, aliphatic chains wherein one or more methylenes are replaced by cyclic or heteroatom groups, or any combination thereof.
  • a phospholipid useful or potentially useful in the present disclosure comprises a modified phosphocholine moiety, wherein the alkyl chain linking the quaternary amine to the phosphoryl group is not ethylene (e.g., n is not 2). Therefore, in certain embodiments, a phospholipid useful or potentially useful in the present disclosure is a compound of Formula (IV), wherein n is 1, 3, 4, 5, 6, 7, 8, 9, or 10. For example, in certain embodiments, a compound of Formula (IV) is of one of the following formulae: salt thereof.
  • a phospholipid useful or potentially useful in the present disclosure comprises a modified phosphocholine moiety, wherein the alkyl chain linking the quaternary amine to the phosphoryl group is not ethylene (e.g., n is not 2). Therefore, in certain embodiments, a phospholipid is useful.
  • an alternative lipid is used in place of a phospholipid.
  • an alternative lipid is oleic acid.
  • the alternative lipid is one of the following: , , , ,
  • the lipid composition of a lipid nanoparticle disclosed herein can comprise one or more structural lipids.
  • structural lipid refers to sterols and also to lipids containing sterol moieties.
  • Incorporation of structural lipids in the lipid nanoparticle may help mitigate aggregation of other lipids in the particle.
  • Structural lipids can be selected from the group including but not limited to, cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, hopanoids, phytosterols, steroids, and mixtures thereof.
  • the structural lipid is a sterol.
  • “sterols” are a subgroup of steroids consisting of steroid alcohols.
  • the structural lipid is a steroid.
  • the structural lipid is cholesterol.
  • the structural lipid is an analog of cholesterol.
  • the structural lipid is alpha-tocopherol.
  • the structural lipids may be one or more of the structural lipids described in PCT/US2018/037922 (published as WO 2018/232357).
  • the structural lipid is cholesterol.
  • the amount of the structural lipids (e.g., cholesterol) in the lipid composition ranges from about 20 mol% to about 60 mol%.
  • PEG Polyethylene Glycol
  • the lipid composition of a lipid nanoparticle disclosed herein can comprise one or more polyethylene glycol (PEG) lipids.
  • PEG-lipid refers to polyethylene glycol (PEG)- modified lipids.
  • PEG-lipids include PEG-modified phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG- CerC14 or PEG-CerC20), PEG-modified dialkylamines and PEG-modified 1,2- diacyloxypropan-3-amines.
  • PEGylated lipids are also referred to as PEGylated lipids.
  • a PEG lipid can be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid.
  • the PEG-lipid includes, but is not limited to 1,2- dimyristoyl-sn-glycerol methoxypolyethylene glycol (PEG-DMG), 1,2-distearoyl-sn-glycero- 3-phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEG- DAG), PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), or PEG-l,2- dimyristyloxlpropyl-3
  • the PEG-lipid is selected from the group consisting of a PEG-modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG- modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG- modified dialkylglycerol, and mixtures thereof.
  • the lipid moiety of the PEG-lipids includes those having lengths of from about C14 to about C22, preferably from about C14 to about C16.
  • a PEG moiety for example an mPEG-NH 2 , has a size of about 1000, 2000, 5000, 10,000, 15,000 or 20,000 daltons.
  • the PEG-lipid is PEG 2k - DMG.
  • the lipid nanoparticles described herein can comprise a PEG lipid which is a non-diffusible PEG.
  • Non-limiting examples of non-diffusible PEGs include PEG-DSG and PEG-DSPE.
  • PEG-lipids are known in the art, such as those described in U.S. Patent No.8158601 and International Publ. No. WO 2015/130584 A2, which are incorporated herein by reference in their entirety.
  • the lipid component of a lipid nanoparticle composition may include one or more molecules comprising polyethylene glycol, such as PEG or PEG-modified lipids. Such species may be alternately referred to as PEGylated lipids.
  • a PEG lipid is a lipid modified with polyethylene glycol.
  • a PEG lipid may be selected from the non-limiting group including PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, and mixtures thereof.
  • a PEG lipid may be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid.
  • the PEG-modified lipids are a modified form of PEG DMG.
  • PEG-DMG has the following structure: [0403]
  • PEG lipids useful in the present disclosure can be PEGylated lipids described in International Publication No. WO 2012/099755, which is herein incorporated by reference in its entirety. Any of these exemplary PEG lipids described herein may be modified to comprise a hydroxyl group on the PEG chain.
  • the PEG lipid is a PEG-OH lipid.
  • a “PEG-OH lipid” (also referred to herein as “hydroxy-PEGylated lipid”) is a PEGylated lipid having one or more hydroxyl (–OH) groups on the lipid.
  • the PEG-OH lipid includes one or more hydroxyl groups on the PEG chain.
  • a PEG-OH or hydroxy-PEGylated lipid comprises an –OH group at the terminus of the PEG chain.
  • a PEG lipid useful in the present disclosure is a compound of Formula (V).
  • R 3 is –OR O ;
  • R O is hydrogen, optionally substituted alkyl, or an oxygen protecting group;
  • r is an integer between 1 and 100, inclusive;
  • L 1 is optionally substituted C 1-10 alkylene, wherein at least one methylene of the optionally substituted C 1-10 alkylene is independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, O, N(R N ), S, C(O), C(O)N(R N ), NR N C(O), C(O)O, - OC(O), OC(O)O, -OC(O)N(R N ), NR N C(O)O, or NR N C(O)N(R N );
  • D is a moiety obtained by click chemistry or a moiety cleavable under physiological conditions;
  • m is 0,
  • the compound of Formula (V) is a PEG-OH lipid (i.e., R 3 is –OR O , and R O is hydrogen).
  • the compound of Formula (V) is of Formula (V-OH): (V-OH), or a salt thereof.
  • a PEG lipid useful in the present disclosure is a PEGylated fatty acid.
  • a PEG lipid useful in the present disclosure is a compound of Formula (VI).
  • R 3 is–OR O ;
  • R O is hydrogen, optionally substituted alkyl or an oxygen protecting group;
  • r is an integer between 1 and 100, inclusive;
  • the compound of Formula (VI) is of Formula (VI-OH): (VI-OH), or a salt thereof. In some embodiments, r is 45. [0409] In yet other embodiments the compound of Formula (VI) is: salt thereof. [0410] In some embodiments, the compound of Formula (VI) is (Compound I). [0411] In some aspects, the lipid composition of the pharmaceutical compositions disclosed herein does not comprise a PEG-lipid. [0412] In some embodiments, the PEG-lipids may be one or more of the PEG lipids described in PCT/US2018/037922 (published as WO 2018232357).
  • a PEG lipid comprises a PEG-modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG-modified dialkylglycerol, and/or mixtures thereof.
  • the PEG-modified lipid is PEG-DMG, PEG-c-DOMG (also referred to as PEG-DOMG), PEG-DSG and/or PEG-DPG.
  • a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising DSPC, a structural lipid, and a PEG lipid comprising PEG-DMG.
  • a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising DSPC, a structural lipid, and a PEG lipid comprising a compound having Formula VI.
  • a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and the PEG lipid comprising a compound having Formula V.
  • a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and the PEG lipid comprising a compound having Formula VI.
  • a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and a PEG lipid comprising a compound having Formula V or VI.
  • a LNP comprises an ionizable cationic lipid of , and a PEG lipid comprising Formula VI.
  • a LNP comprises an ionizable cationic lipid of , and an alternative lipid comprising oleic acid.
  • a LNP comprises an ionizable cationic lipid of , an alternative lipid comprising oleic acid, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VI.
  • a LNP comprises an ionizable cationic lipid of a phospholipid comprising DOPE, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VI.
  • a LNP comprises an ionizable cationic lipid of , a phospholipid comprising DOPE, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VII.
  • an ionizable lipid is one or more compounds such as Compound A1 shown below.
  • a LNP comprises an N:P ratio of from about 2:1 to about 30:1.
  • a LNP comprises an N:P ratio of about 6:1.
  • a LNP comprises an N:P ratio of about 3:1.
  • a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of from about 10:1 to about 100:1.
  • a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of about 20:1.
  • a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of about 10:1.
  • the disclosure has a mean diameter from about 50nm to about 150nm.
  • a LNP has a mean diameter from about 70nm to about 120nm.
  • a method of filtering a solution containing lipid nanoparticles comprising setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold.
  • the method includes stopping the filtration after the first period has elapsed, wherein the determination of the actual value occurs after the first period.
  • the method includes following adjusting of the at least one filtration control parameter, resuming filtration for a second period.
  • the filtration control parameter is a flow rate of the solution, an environmental temperature, a solution temperature, a pH of the solution or a salt concentration of the solution.
  • the filtration control parameter is a fluid pressure of the solution; optionally, the filtration control parameter is P CRITICAL .
  • the method includes introducing one or more additives to the solution.
  • the determination of the actual value occurs after the first period and prior to the second period.
  • the method further comprises monitoring a transmembrane pressure during the filtration.
  • the first layer is upstream of the second layer.
  • the first pore size is 0.8 ⁇ m and the second pore size is 0.2 ⁇ m.
  • a non-transitory computer readable medium which is configured to store instructions which, when executed by a processor, cause a controller to adjust the at least one filtration control parameter according to the techniques herein.
  • a method of evaluating sterile filtration of liquid nanoparticles is provided, the method comprising filtering a first solution lacking LNPs through at least one membrane while controlling at least one filtration control parameter to satisfy a control parameter threshold; determining a first hydraulic resistance of the membrane following filtration of the first solution; filtering a second solution containing LNPs through the at least one membrane while controlling the at least one filtration control parameter to satisfy the threshold; determining a second hydraulic resistance of the membrane following filtration of the second solution; computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; and performing microscopic imaging of the at least one membrane following filtration with the second solution to produce
  • the at least one filtration control parameter is a fluid pressure of the solution.
  • the method includes monitoring a transmembrane pressure during filtering of at least the second solution.
  • each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc.
  • all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above.
  • a range includes each individual member.
  • Such joining may be stationary (e.g., permanent) or moveable (e.g., removable or releasable).
  • FIG. 4 Although the figures and description may illustrate a specific order of method steps, the order of such steps may differ from what is depicted and described, unless specified differently above. Also, two or more steps may be performed concurrently or with partial concurrence, unless specified differently above. All such variations are within the scope of the disclosure. [0451] It is important to note that the construction and arrangement of the various exemplary embodiments are illustrative only. Those skilled in the art who review this disclosure will readily appreciate that modifications are possible (e.g., variations in sizes or dimensions, the values of parameters, etc.) without materially departing from the teachings of the subject matter described herein.

Abstract

Methods of filtering a solution containing lipid nanoparticles are provided. The methods include setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold.

Description

STERILE FILTRATION OF LIPID NANOPARTICLES AND FILTRATION ANALYSIS THEREOF FOR BIOLOGICAL APPLICATIONS CROSS-REFERENCE TO RELATED APPLICATIONS [0001] The present application claims priority to U.S. Provisional Application No. 63/321,196, filed on March 18, 2022, U.S. Provisional Application No.63/322,562, filed on March 22, 2023, and U.S. Provisional Application No.63/335,816, filed on April 28, 2022, the entire contents of each of which are incorporated herein by reference. TECHNICAL FIELD [0002] The present disclosure relates generally to sterile filtration of lipid nanoparticles (LNPs) for biological applications. More specifically, the present disclosure relates to sterile filtration of lipid nanoparticles used in biological applications such as mRNA-based products, including vaccines and therapeutics. BACKGROUND [0003] Lipid nanoparticles (LNPs) have been developed as delivery systems for mRNA- based vaccines and therapeutics. In the vaccine context, LNP delivery systems express mRNA-encoded immunogens, e.g., after intramuscular injection. In some delivery systems, mRNA is bound by an ionizable lipid that occupies part of the LNPs. SUMMARY [0004] The present disclosure addresses filtration of solutions containing LNPs, and in particular, presents improved methods of filtration of LNPs. Filtration, e.g., sterile filtration, may be performed to reduce or eliminate the presence of microorganisms and/or other contaminants. The present disclosure further relates to filter performance during filtering of LNPs. [0005] In at least one aspect, the present disclosure relates to a method of evaluating sterile filtration of LNPs. The method includes filtering a first solution lacking LNPs through at least one membrane while controlling a filtration control parameter to satisfy a control parameter threshold; and determining a first hydraulic resistance of the membrane following filtration of the first solution. The method further includes filtering a second solution containing LNPs through the at least one membrane while controlling the filtration control parameter to satisfy the control parameter threshold; and determining a second hydraulic resistance of the membrane following filtration of the second solution. Additionally, the method further includes computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; imaging the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling. [0006] In at least one aspect, the present disclosure relates to a method of filtering a solution containing lipid nanoparticles. The method includes setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold. [0007] It should be appreciated that all combinations of the foregoing concepts and additional concepts discussed in greater detail below (provided such concepts are not mutually inconsistent) are contemplated as being part of the subject matter disclosed herein. In particular, all combinations of claimed subject matter appended at the end of this disclosure are contemplated as being part of the subject matter disclosed herein. BRIEF DESCRIPTION OF THE DRAWINGS [0008] The foregoing and other features of the present disclosure will become more fully apparent from the following description and appended claims, taken in conjunction with the accompanying drawings. Understanding that these drawings depict only several implementations in accordance with the disclosure and are therefore, not to be considered limiting of its scope, the present subject matter is described with additional specificity and detail through use of the accompanying drawings. [0009] FIG.1 is a schematic illustration of a lipid nanoparticle, according to an aspect of the present disclosure. [0010] FIGS.2A and 2B are images of a first layer of filter media taken by a scanning electron microscope (SEM), according to an aspect of the present disclosure. [0011] FIG.3 is an SEM image of a second layer of filter media, according to an aspect of the present disclosure. [0012] FIG.4 is a plot of filtrate flux at different transmembrane pressures (TMPs) for exemplary filter media, of which FIGS.2A-2B depict a first layer thereof and FIG.3 depicts a second layer thereof, according to an aspect of the present disclosure. [0013] FIG.5 is a plot of filtration resistance at different TMPs for the filter media of FIGS.2A-2B and 3, according to an aspect of the present disclosure. [0014] FIG.6 is an SEM image of a first layer of filter media in an unused condition, according to an aspect of the present disclosure. [0015] FIG.7 is an SEM image of the first layer of the filter media of FIG.6 in a used condition, according to an aspect of the present disclosure. [0016] FIG.8 is an SEM image of a second layer of filter media in an unused condition, according to an aspect of the present disclosure. [0017] FIG.9 is an SEM image of the second layer of the filter media of FIG.8 in a used condition, according to an aspect of the present disclosure. [0018] FIG.10 is a plot of filtrate flux for different filter media formulations, according to an aspect of the present disclosure. [0019] FIG.11 is a plot relating to a derivative analysis based on the test data from FIG. 4-5, according to an aspect of the present disclosure. [0020] FIG.12 is a plot of filtrate flux as observed through multiple filters of a two-stage filtering operation, according to an aspect of the present disclosure. [0021] FIG.13 is a plot of filtrate flux data from a pressure stepping experiment, according to an aspect of the present disclosure. [0022] FIG.14 is a plot of filtrate flux data from a pressure stepping experiment, according to an aspect of the present disclosure. [0023] FIG.15 is a plot of filtrate flux data from a pressure stepping experiment using two different feed solutions, according to an aspect of the present disclosure. [0024] FIG.16 is a plot of resistance over filtration time from a first pressure stepping experiment, according to an aspect of the present disclosure. [0025] FIG.17 is a plot of resistance over filtration time from a second pressure stepping experiment, according to an aspect of the present disclosure. [0026] FIG.18 is a plot of resistance over filtration time from a third pressure stepping experiment, according to an aspect of the present disclosure. [0027] FIG.19 is an exemplary depiction of a filtration system according to various aspects of the present disclosure. [0028] FIG.20A and FIG.20B are plots of resistance versus filtration time during pressure stepping, according to aspects of the present disclosure. [0029] FIG.21 is a plot of resistance versus transmembrane pressure for LNPs as deposited on a membrane, according to an aspect of the present disclosure. [0030] FIGS.22A-22D are plots of resistance versus transmembrane pressure for various membranes, according to certain aspects of the present disclosure. [0031] FIG.23 is a plot of operating pressure versus reciprocal pore diameter, according to an aspect of the present disclosure. [0032] FIGS.24A-24F are scanning electron microscopy and environmental scanning electron microscopy images of clean and fouled membranes, according to aspects of the present disclosure. [0033] FIG.25A is an atomic force microscopy image of a partially fouled membrane, and FIG.25B depicts deposit height of the deposited LNPs in FIG.25A as a function of surface distance between two points shown in FIG.25A, according to aspects of the present disclosure. [0034] FIG.26 is a high resolution SEM image of deposited LNPs on a membrane, according to an aspect of the present disclosure. [0035] FIGS.27A-27F are schematic diagrams of physical mechanisms affecting behavior of deposited LNPs. [0036] Reference is made to the accompanying drawings throughout the following detailed description. The illustrative implementations described in the detailed description, drawings, and claims are not meant to be limiting. Other implementations may be utilized, and other changes may be made, without departing from the spirit or scope of the subject matter presented here. It will be readily understood that the aspects of the present disclosure, as generally described herein, and illustrated in the figures, can be arranged, substituted, combined, and designed in a variety of different configurations, all of which are explicitly contemplated and made part of this disclosure. DETAILED DESCRIPTION [0037] In general, mRNA-based vaccines and therapeutics are of growing interest because of their high efficacy, rapid development timelines, and excellent safety profiles. Encapsulation of mRNA in LNPs promotes effective delivery of the mRNA to ensure protein translation. Solutions containing LNPs undergo filtration during production of mRNA-based vaccines and therapeutics. [0038] The large size and unique behavior of LNPs affects filtration performance. A relatively higher lipid content may cause some solutions containing LNPs to exhibit particular behavior during filtering, such as patterns of aggregation or deposition on filter media, which differ from solutions without LNPs. Further, filter capacities and LNP yields are generally both smaller than those for certain existing biotherapeutics. Lower capacity and significant yield loss can occur during sterile filtration of vaccines, viruses, and gene therapies due to the very similar size of these biotherapeutics and the pore size of sterilizing grade filters through which they are passed to ensure sterility of the final product. The observed capacity and yield may be attributable to both the relatively large size of the LNPs and to unexpected behavior as discussed herein. [0039] The underlying mechanisms controlling sterile filtration of LNPs (e.g., the factors controlling performance of sterile filtration of LNP) are described herein. [0040] According to an aspect of the present disclosure, a method of determining the pressure-dependent behavior in sterile filtration of an LNP solution (a solution containing LNPs) is provided. The method includes (i) imaging clean or un-fouled filter media in addition to used or fouled filter media, (ii) evaluating filtration performance of individual filter media layers, and (iii) analyzing performance at different transmembrane pressures (TMPs). The filter media may be implemented as a membrane or a membrane assembly, for example. The analysis may be conducted using experimental data obtained where control parameters, including but not limited to TMP, are adjusted. [0041] According to an aspect of the present disclosure, the foregoing method further can include performing re-filtration to assess an extent to which foulants are retained and/or removed by the filter media. According to an aspect of the present disclosure, the method further can include performing pressure stepping to assess the influence of LNP deposition at different pressures and with different feed solution compositions. [0042] At least one aspect of the present disclosure provides for the observation and characterization of the sterile filtration performance of LNP filtration as a function of TMP for a dual layer media assembly. In contrast to typical behavior associated with compressible fouling deposits, experimental data obtained during testing indicates an increase in filtrate flux and filter media capacity with increasing TMP. In particular, experimental data indicates that high TMP filtration of LNPs may reduce filtration resistance and increase filter media capacity. [0043] According to at least one aspect of the present disclosure, a kit is provided including LNPs in solution as described herein, a membrane, and a controller according to various examples below. [0044] The various concepts introduced above and discussed in greater detail below may be implemented in any of numerous ways, as the described concepts are not limited to any particular manner of implementation. Examples of specific implementations and applications are provided primarily for illustrative purposes. [0045] Various numerical values herein are provided for reference purposes only. Unless otherwise indicated, all numbers expressing quantities of properties, parameters, conditions, and so forth, used in the specification and claims are to be understood as being modified in all instances by the term “about” or “approximately.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification are approximations. Any numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. The term “about” or “approximately” when used before a numerical designation, e.g., a quantity and/or an amount including ranges, indicates approximations which may vary by 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value). In some embodiments, the term “about” or “approximately” in the context of a logarithmic scale may indicate approximations varying by ^ 0.1 log, ^ 0.2 log, ^ 0.3 log, ^ 0.4 log or ^ 0.5 log from a stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value). Lipid Nanoparticle and Filtration Summary [0046] FIG.1 shows a schematic illustration of a lipid nanoparticle, shown as LNP 10, for use in a biological application such as a pharmaceutical drug delivery system. In at least one embodiment, the LNP 10 includes four lipid components including distearolyphosphatidycholine (DSPC) 12, a cholesterol 14, a polyethylene glycol (PEG)-lipid 16, and an ionizable lipid (shown as charged ionizable lipid 18 and neutral ionizable lipid 20). [0047] The DSPC 12 is configured to provide a general support structure (e.g., shield, support, etc.) for the LNP 10. The cholesterol 14 is configured as a stabilizing element to aid in transfection of the mRNA into cells (e.g., introduction of the mRNA into cells by non-viral methods). The PEG-lipid 16 is configured to provide a stealth effect (e.g., reduce nonspecific binding) and/or as a stabilizing element in the LNP 10. Lastly, the ionizable lipids (e.g., the charged ionizable lipid 18, the neutral ionizable lipid 20) are configured to improve intracellular delivery and/or promote encapsulation of the RNA in the LNP 10. [0048] As shown in FIG.1, the LNP 10 can be substantially spherical in shape. In some embodiments, the LNP 10 has an average particle size (e.g., diameter, etc.) greater than approximately 100 nm. In some embodiments, the average particle size is between approximately 50 nm and approximately 200 nm. In some embodiments, the LNP 10 can be (i) substantially spherical or generally ellipsoidal in shape and (ii) have a diameter of approximately 50 nm to approximately 200 nm. In general, the LNP 10 can have diverse structural configurations. [0049] In the process of creating a vaccine or therapeutic product relying on an LNP delivery system, a solution containing LNP 10 is first filtered to remove or reduce the quantity of microorganisms (e.g., bacteria) and/or other contaminants present in the final product. Sterile filtration is a process of filtering a solution to remove such contaminants without adversely affecting the final product. For example, sterile filtration of LNP 10 may include passing a solution containing LNP 10 (e.g., LNP 10 and buffer, etc.) through filter media. The filter media may include a single or multi-layer filter media (e.g., multi- membrane filter media, an assembly constructed of multiple filters, etc.). The LNP 10 may also be referred to herein as mRNA-LNP 10. [0050] The production of mRNA vaccines begins with the in vitro enzymatic synthesis of the mRNA from a DNA template. The downstream purification process involves a series of chromatographic separations and tangential flow filtration. The purified mRNA is encapsulated in LNPs by mixing the mRNA (in an acidic aqueous buffer) with the lipids (in ethanol). The resulting LNP suspension can be buffer exchanged and concentrated by ultrafiltration or diafiltration followed by sterile filtration as part of the final fill-finish operation. [0051] Sterile filtration is commonly performed using normal-flow (dead-end) filtration through membranes containing a 0.2 µm filter, which can retain microbes of approximately 107 colony forming units / cm2 of filter area, for example. Sterile filtration of LNPs, as well as other large viral and non-viral delivery vehicles, can be challenging because these products can be similar in size to the 0.2 µm size of certain sterilizing-grade filters, among other reasons. [0052] During filtration, pressure is applied to the feed solution upstream of the filter media to force the feed solution through the filter media. The filter media captures contaminants along the surface or within pores of the filter media while allowing smaller particles to pass therethrough. Speaking generally, filtration continues until the filter media becomes clogged (e.g., fouled) and the filtrate flux across the filter media drops below a threshold value. Filter Properties and Operation [0053] Typical biotherapeutics may exhibit any of the classical fouling models (complete pore blockage, intermediate pore blockage, pore constriction, or cake formation). Under each of these behavioral models, foulant is deposited onto the membrane. For typical biotherapeutics, the deposited foulant is typically either incompressible or compressible. “Incompressible” foulant is foulant for which a filtration resistance is always constant (that is, the flux varies linearly with TMP). “Compressible” foulant is foulant for which filtration resistance increases with higher TMP (as an example, the flux may vary less-than-linearly with TMP). [0054] In contrast, according to some aspects of the present disclosure, behavior may be exhibited in which filtration resistance decreases with higher TMP (that is, the flux varies more-than-linearly with TMP). Such behavior indicates that the foulant in LNP-containing solution is neither a typical compressible fouling deposit nor a typical incompressible fouling deposit. [0055] In particular, LNP-containing solution under constant flux can exhibit unique filtration behavior. Pressure observations (as a function of different fluxes and as a function of throughout) may differ from expected behavior. Unexpected fouling behavior, observed as rates of and dependencies of pressure increase and or flux decline, and or observed capacity, has been exhibited. [0056] The fouling behavior in sterile filtration of an LNP solution (a solution containing LNPs) may be evaluated using imaging and data analysis. At least one exemplary method of performing filtration of an LNP-containing solution includes (i) imaging clean or un-fouled filter media in addition to used or fouled filter media, (ii) analyzing filtration performance (e.g., of individual filter media layers and/or of the overall system at different transmembrane pressures (TMPs)) and (iii) adjusting one or more filtration control parameters in responsive to at least one of the individual filter media layer performance or the TMP performance. [0057] In some embodiments, material may be accumulated for a content analysis to determine the correlation between the accumulated material and the feed stream. In some embodiments, the content analysis may be performed to assess the extent to which components and/or product-related impurities, such as cholesterol crystals, are present in the accumulated material. [0058] In some embodiments, one or more filtration control parameters may be adjusted utilizing a feedback loop based on at least one measured value (e.g., a measured pressure, a measured flow rate, a measured product quality, a measured yield, or any combination of any of the foregoing) and/or based on a calculation obtained from a fouling model that uses one or more of the measured values as an input. [0059] In some embodiments, the filtration control parameters may include one or more of a pressure, a filter load (e.g., in L/m2 or g/m2), a flow rate, a temperature, a pH, a salt type or concentration, and a filtration operational mode, and the concentration of additives. For one or more of the foregoing filtration control parameters, an exemplary method for filtering LNP-containing solution may include controlling filtration based on either a specific value associated with a given parameter or a gradient of values. In some embodiments, one or more of the pH, the salt concentration, and the concentration of additives may be adjusted during filtering. [0060] In some embodiments, the filter media is selected based on an associated pressure or an associated range of pressures. In some embodiments, pressure may vary between approximately 0.5 psi to approximately 50 psi, or greater than approximately 0 psi to approximately 70 psi. In some embodiments, the pressure is approximately 10 psi, approximately 20 psi, approximately 30 psi, approximately 40 psi, approximately 50 psi, approximately 60 psi, approximately 70 psi or approximately 80 psi. [0061] In some embodiments, filter load may vary from approximately 10 L/m2 to approximately 5,000 L/m2. In some embodiments, flow rate (flux) may vary from approximately 20 L/m2/hr to approximately 2,000 L/m2/hr. [0062] In some embodiments, the temperature may be between approximately 2 °C to approximately 40 °C. For example, in some embodiments, the filtration may take place under refrigeration, e.g., between approximately 2 °C to approximately 8 °C. In some embodiments, the filtration may take place in ambient conditions of between approximately 15 °C to approximately 25 °C (e.g., room temperature). In some embodiments, the filtration may take place at elevated temperatures, e.g., between approximately 30 °C and approximately 40 °C or between approximately 40 °C and 50 °C. The temperature may affect inter-particle attractive forces and may influence viscosity and filter performance. In some embodiments, elevated temperatures may be used to induce microbial cell death, thereby increasing efficiency and effectiveness of filtration. [0063] In some embodiments, the pH may vary between approximately 4 to approximately 8. In particular, LNPs may experience greater charging when the pH is approximately 1 pH unit below the isoelectric point (the pH is approximately 6 to approximately 7 for various exemplary lipids). When charged, LNPs may be expected to repel each other, thereby contributing to decreased fouling. In some embodiments, a filtration process of filtering LNPs may include filtering the LNPs at a pH of approximately 5, where insignificant fouling is observed. In contrast, higher pH (e.g., above 7.5) may be associated with increased fouling. [0064] In some embodiments, a salt concentration may vary between approximately 10 M to approximately 1000 M. In some embodiments, a salt concentration may be increased so as to lower the repulsion between LNPs. A relatively lower salt concentration is expected to enhance filtration at low pH. [0065] In some embodiments, an additive may be one selected from the group consisting of salts, buffers, kosmotropes, chaotropes, detergents, surfactants, PEG, amino acids, organic solvents, acids or bases. [0066] In some embodiments, a filter operational mode may vary between a first mode and a second mode. For example, a filter operational mode may vary between a constant pressure mode and a constant flux mode. In some embodiments, each of the constant pressure mode and a constant flux mode may assume a constant temperature. In some embodiments, an exemplary filtration process may begin in a first mode and switch to a second mode, or vice versa, or may alternate between modes. [0067] In some embodiments, an exemplary filtration process may employ variable pressure and/or variable flux (e.g., stepping alternately up and down, for example from approximately 15 psi to approximately 5 psi, and from approximately 5 psi to approximately 15 psi or vice versa), stepping continuously up or down (for example, from approximately 5 psi to approximately 10 psi to approximately 15 psi or vice versa). In some embodiments, an exemplary filtration process may employ continuous changes in response to certain parameters (for example, a controller may continuously adjust the pressure based on the filtrate flow rate, or based on the filtrate volume, or based on another parameter, or any combination of any of the foregoing). In some embodiments, an exemplary filtration process may be carried out intermittently rather than continuously, where the filtration is paused one or more times during a batch (i.e., during a “run”). In some embodiments, temperature may be maintained at an approximately constant temperature throughout a batch (e.g., at any setpoint between approximately 2 °C and approximately 40 °C). In some embodiments, the temperature may be varied, in any of the ways described above (e.g., stepwise or continuously monitoring and adjusting the temperature). [0068] Further, in some embodiments, other filtration control parameters may be utilized, including but not limited to one or more product quality attributes as control parameters. For example, in some embodiments, a sensor may be provided that measures particle size and/or concentration (which are indicative of the yield and/or the product retention). Information from the sensor may be utilized to determine the extent to which the one or more product quality attributes change during filtration. In response to a change being detected – e.g., an indication that the one or more product quality attributes has varied with respect to an initially determined one or more product quality attributes – then at least one aspect of the filtration process may be varied. For example, the flowrate may be lowered, or a pause (as discussed in further detail below) may be instituted, among other things. [0069] In some embodiments, a method of filtering LNP-containing solution comprises controlling a flow rate of the feed solution. For example, in some embodiments, the flow rate is controlled based on the properties of the solution to be filtered. In some embodiments, an operator or user may input various parameters or other information relating to the solution into a computer or microcomputer of a controller via a user interface. The controller is configured set a target flow rate based on the properties of the solution or other information provided by the operator or user. [0070] In some embodiments, the flow rate may be in a range from approximately 1000 L/m2/hr to approximately 2000 L/m2/hr for solutions that have a water content that exceeds a threshold water content. For example, in some embodiments, the flow rate may be approximately 1500 L/m2/hr. [0071] Furthermore, in some embodiments, the flow rate may be in a range from approximately 45 L/m2/hr to approximately 550 L/m2/hr for solutions such as biologics that have a lower aqueous content (that is, aqueous content that does not exceed the above- mentioned threshold). For example, in some embodiments, the flow rate may be approximately 500 L/m2/hr. In some embodiments, the flow rate may be in a range of approximately 10 L/m2/hr to approximately 50 L/m2/hr, approximately 50 L/m2/hr to approximately 100 L/m2/hr, approximately 100 L/m2/hr to approximately 200 L/m2/hr, or approximately 200 L/m2/hr to approximately 500 L/m2/hr. [0072] In some embodiments, a method of filtering LNP-containing solution comprises controlling a temperature of the LNP-containing solution and/or controlling a temperature at which filtration occurs (i.e., an environmental temperature). [0073] In some embodiments, a method of filtering LNP-containing solution comprises controlling a pH of the LNP-containing solution. In some embodiments, a method of filtering LNP-containing solution includes controlling (i) a salt concentration of the LNP-containing solution and/or (ii) a type of salt in the LNP-containing solution. [0074] In some embodiments, a method of filtering LNP-containing solution includes setting one or more filtration control parameters to one or more respective control target values; determining, after filtration has begun, a deviation between one or more actual control values and the one or more control target values; and, in response to determining that the deviation exceeds a threshold value, adjusting the one or more filtration control parameters. For example, one or more filtration control parameters such as flow rate or pressure are adjustable to achieve a target control flow rate and a target control pressure, respectively. [0075] In some embodiments, a method of filtering LNP-containing solution includes controlling an operational mode of filtration. For example, a pulse-width-modulation (PWM) control may be used, as implemented via a controller or microcontroller. The operational mode may be selected from and/or switched between a continuous filtration and an intermittent filtration. In intermittent filtration, the filtration is periodically stopped and restarted (e.g., “pulsed”). The period between operational time for the filtering – that is, the “down” time when the filtering is stopped – may range from several minutes to several hours, e.g., (i) from more than approximately 2 minutes to less than approximately 5 hours, (ii) between approximately 1 second to approximately 6 hours, (iii) approximately 30 seconds, (iv) between approximately 4 minutes to approximately 8 minutes, (v) approximately 10 minutes, or (vi) approximately two hours, for example. [0076] In some embodiments, a pause (of fixed or variable duration) may be imposed between a first filtering period and a second filtering period. Such a pause may relieve the accumulated foulant from the filter surface. In particular, increased yield and/or lower resistance may be realized following such a pause. In some embodiments, the length or imposition of a pause may be determined based at least in part on a feedback loop used for controlling one or more filtration control parameters. Accordingly, in some embodiments, one or more filtration control parameters may be adjusted to promote creation and/or maintaining of such pauses. [0077] In some embodiments, during one or more stops (pauses) between filtration, one or more filtration control parameters may be evaluated. That is, after a first filtration period and before a second filtration, a determination may be made as to whether one or more filtration control parameters should be adjusted. For example, actual values for the one or more filtration control parameters may be compared to target and/or predicted values and adjusted accordingly. [0078] In some embodiments, a method of filtering LNP-containing solution includes introducing one or more additives to the feed-containing solution. In some embodiments, the additives may enhance filtration by promoting disassociation between LNPs and/or between LNPs and the filter media. In some embodiments, the additives may enhance filtration by altering one or more biophysical properties to reduce or prevent aggregation of the LNPs. The biophysical properties include, but are not limited to, a pH value, a salt concentration, and a salt type. In some embodiments, the additive may include, but is not limited to, polyethylene glycol (PEG). [0079] According to some aspects, a method of filtering LNP-containing solution includes assessing an extent to which foulants are retained and/or removed by the filter media, and optionally, performing refiltration. [0080] According to some aspects, the methods include performing pressure stepping, or gradient pressure changes. [0081] In some embodiments, a method of filtering LNP-containing solution may be performed for a dual layer filter media assembly. The filter media may be implemented as a membrane or a membrane assembly. [0082] In some embodiments, a non-transitory computer readable medium is configured to store instructions which, when executed by a processor, cause a controller to adjust one or more filtration control parameters. In some embodiments, one or more filtration control parameters are adjusted to account for actual and/or predicted behavior of LNPs in solution. In contrast to typical behavior associated with compressible fouling deposits, experimental data obtained during testing of filtration of LNP-containing solutions indicates an increase in filtrate flux and filter media capacity with increasing TMP. Experimental data indicates that high TMP filtration of LNPs may reduce filtration resistance and increase filter media capacity. Filtration Systems [0083] In the process of creating a vaccine or therapeutic product relying on an LNP delivery system, a solution containing LNP is filtered to remove or reduce the quantity of microorganisms (e.g., bacteria) and/or other contaminants present in the final product. Sterile filtration is a process of filtering a solution to remove such contaminants without adversely affecting the final product. For example, sterile filtration of LNP may include passing a solution containing LNP (e.g., LNP and buffer, etc.) through filter media. The filter media may include a single or multi-layer filter media (e.g., multi-membrane filter media, an assembly constructed of multiple filters, etc.). [0084] During filtration, pressure is applied to the feed solution upstream of the filter media to force the feed solution through the filter media. That is, the solution is pressurized to a given fluid pressure. The fluid pressure may be set as a filtration control parameter and, in some embodiments, can be adjusted during filtration. In some embodiments, the filtration may be performed under substantially constant pressure, which may be referred to as “constant pressure filtration.” The filter media can capture contaminants along the surface or within pores of the filter media while allowing smaller particles to pass therethrough. [0085] In some embodiments, “constant flux filtration” may be carried out instead of or in addition to constant pressure filtration. In “constant flux” filtration, a substantially constant flowrate is applied to the feed solution upstream of the filter media. As the filter media becomes clogged, the pressure across the filter media increases above a threshold value. [0086] In some embodiments, a filtration system may be controlled based on a filtration control parameter. In some embodiments, the filtration control parameter may in turn be based on or correspond to a threshold value. In some embodiments, the threshold value is a maximum resistance value which is observed with increasing TMP until a given pressure value or range of pressures is reached, before resistance declines thereafter, as discussed in greater detail below. In some embodiments, the threshold value is a pressure value associated with the maximum resistance value. In some embodiments, the filtration control parameter may be based on an estimated value, a sensed value or a combination thereof. [0087] In some embodiments, a tangential flow device may be used. For example, a method of filtering LNP-containing solution may include filtering the solution through a tangential flow device and adjusting one or more filtration control parameters to enhance LNP recovery. Tangential flow filtration involves passing a feed solution parallel to the filter media, where the permeate is collected and the retentate is re-cycled back to the feed solution. By alternating a tangential flow of fluid through the filter media, continuous filtration may be achieved. [0088] The selection of filter media may be made depending on the properties of the solution and the desired filter performance. In particular, filter media may be selected based on the size of contaminants (e.g., bacteria) that need to be removed. However, the size of the LNP particles (in comparison to the average media pore size required to retain bacteria) may require large filter sizes to ensure sufficient filter capacity at the required filtration rates for production. Accordingly, the techniques of some embodiments include decreasing the extent to which small particles constrict passages in a filter. In some embodiments, the filters are sterilizing-grade filter media. [0089] Some embodiments comprise filtration methods in which a filter media includes a first layer having a first pore size and a second layer having a second pore size smaller than the first pore size. In some embodiments the filter media includes a coarse filtration layer (e.g., pre-filtration layer) and a fine filtration layer coupled to the coarse filtration layer. In some embodiments, the coarse filtration layer is layered on top of the fine filtration layer and positioned upstream from the fine filtration layer during the sterile filtration operation to remove the largest contaminants from the feed solution. [0090] In some embodiments, the coarse filtration layer includes a media layer (e.g., membrane layer) having an 0.8 µm rating that is structured to remove particles as small as approximately 0.8 µm in diameter. In some embodiments, the fine filtration layer includes a media layer (e.g., membrane layer) having an 0.2 µm rating that is structured to remove particles as small as approximately 0.2 µm in diameter. [0091] In some embodiments, the fine filtration layer may have a smaller rating, such as approximately 0.1 µm. In some embodiments, at least one of the coarse filtration layer or the fine filtration layer may have a larger rating, such as approximately 0.3 µm, approximately 0.4 µm, approximately 0.45 µm, approximately 0.65 µm, approximately 0.8 µm, approximately 1.0 µm, approximately 2 µm, approximately 3 um, approximately 5 µm, or approximately 10 µm. In some embodiments, the filter media is considered an absolute pore size rating, whereas in some embodiments, the pore size is considered nominal. In some embodiments, the filter media contains symmetrical pores. In some embodiments, the filter media contains asymmetrical pores. In some embodiments, the filter media are implemented as a single layer, whereas in other embodiments, the filter media may be implemented as a dual-layer assembly, or a filter assembly having a different number of layers. [0092] In some embodiments, one or more of the coarse filtration layer or the fine filtration layer may have differing filter ratings from the foregoing. In some embodiments, the coarse filtration layer may be configured to remove particles as small as approximately 0.45 µm in diameter. In some embodiments, the coarse filtration layer (the upstream filter layer) and the fine filtration layer (the downstream filter layer) are selected from the filter media listed in Table 1 below. In some embodiments, a method of filtering LNPs includes filtering through a two-layer filter wherein the first layer and/or the second layer of the multi- layer filter is selected from the filters listed in Table 1. [0093] As appreciated from Table 1, exemplary filter media may be selected from one or more of the following materials: polyethersulfone (PES), polyvinylidene fluoride or polyvinylidene difluoride (PVDF), nylon or cellulose acetate (CA), for example. Further, in addition to the material composition of the filter media, exemplary filtration systems may vary with respect to one or more of the following: a pore size, a pore distribution, a pore geometry (e.g., symmetrical versus asymmetrical), a filter layer arrangement (e.g., a single layer filter or a multi-layer filter), filter assembly (e.g., stacking), etc. A “symmetrical” pore geometry refers to a pore configuration having a substantially cylindrical profile, whereas an “asymmetrical” pore geometry refers to a pore configuration having narrowing at one end of the pore, for example.
Figure imgf000021_0001
Figure imgf000022_0001
Table 1 Methods of Filtering LNPs [0094] In some embodiments, a method of filtering a solution containing lipid nanoparticles includes setting at least one filtration control parameter to at least one target filtration control parameter. Some embodiments comprise performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size. Some embodiments comprise determining, during or after the first period, a value of the at least one filtration control parameter; and adjusting the at least one filtration control parameter in response to determining that the value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold. The actual value may be an actual value or estimated value. [0095] In some embodiments, a filtration control parameter may be maintained or adjusted in consideration of the filter age. For example, a flow rate may be maintained within a range of flow rates or increased above a threshold rate to increase flow until a filter requires replacement. Controlling the flow rate in this manner, among other filtration control parameters, may contribute to higher yields and increased capacity. In some embodiments, the filtration control parameter is a threshold value that corresponds to a characteristic of the membrane itself and/or the feed solution (e.g., a solution containing LNPs, a buffer solution, etc.). [0096] Some embodiments include directing, through filter media, a feed solution containing LNPs. In some embodiments, the feed solution may be pressurized to a set pressure. [0097] In some embodiments, the method may include sensing a filtrate flux and/or the throughput of the filter media over time at different TMPs. [0098] In some embodiments, high TMP may reduce filtration resistance. As noted above, contrary to behaviors typically associated with compressible fouling deposits, the capacity of the filter media for sterile filtration of LNPs is shown to increase with increasing TMP. Thus, some embodiments comprise filtering a LNP solution at a TMP of approximately 2 psi, approximately 8 psi, approximately 14 psi, approximately 15 psi, approximately 16 psi, approximately 17 psi, approximately 18 psi, approximately 19 psi, or approximately 20 psi, or more. [0099] In some embodiments, TMP is monitored continuously during filtration via one or more sensors. In some embodiments, the sensors may include pressure sensors placed upstream of the filter. For example, the outlet of the filter may be at atmospheric pressure, but a sensor may nonetheless be positioned at the outlet in case atmospheric pressure is not maintained. The TMP corresponds to the upstream pressure less the downstream pressure. The sensors may be any suitable pressure sensor, including a digital sensor such as PendoTECH pressure sensors made by PendoTECH Corp. of Princeton, NJ or the SciLog® pressure sensor (e.g., the SciPres® sensor) made by Parker Hannifin Corp. of Oxnard, CA. [0100] In some aspects, the methods include adjusting different filtration control parameters. In some aspects, one or more filtration control parameters may be adjusted during a filtration process (e.g., after a filtration “run” has started but before it has finished). In some aspects, one or more filtration control parameters may be adjusted via a programmable controller or manually (e.g., by a manual operator). [0101] In some embodiments, a programmable controller or a plurality of programmable controllers may be used to control one or more components of an exemplary filtration system as shown in FIG.19. In particular, FIG.19 depicts an exemplary filtration system including a feed vessel (e.g., a fluid reservoir), a liquid transfer mechanism (e.g., a pump, a motor-driven pump, a micro-pump, a fluid supply source, a pressure source), a filter assembly (e.g., a stacked filter arrangement, a filter membrane assembly, a plurality of filter layers), and a filtrate vessel (e.g., a filtrate reservoir). The liquid transfer mechanism is configured to transfer liquid throughout the system, e.g., a pressurized gas overlay utilizing compressed air or compressed nitrogen, among other substances. Each circle in FIG.19 denotes an exemplary sensor. [0102] In some embodiments, integrated sensors may be used and the number of sensors may differ from what is shown. Various sensors that are utilized may include, but are not limited to, a pressure sensor, a flow rate sensor, a temperature sensor, etc. The sensors may be used upstream of the filter, downstream of the filter, or upstream and downstream of the filter, as well as in the feed vessel and/or in the filtrate vessel. In some embodiments, the filter assembly may include multiple media and/or multiple separate filters in series or in parallel. A complex assembly having multiple filters may, in some embodiments, utilize sensors at multiple locations within the filter assembly. In some embodiments, information from any one of the sensors may be provided (e.g., in the form of a digital signal) to a controller which may be accessible to a user. In some embodiments, the controller may automatically adjust one or more filtration control parameters and/or allow a user to adjust one or more filtration control parameters. [0103] In particular, one or more filtration control parameters may be adjusted in response to determining that a calculated parameter exceeds a threshold value. For example, the calculated parameter may be a calculated resistance to flow across the filter media. The calculated parameter may be any parameter associated with a fouling model such as the fouling models discussed above. [0104] According to an aspect of the present disclosure, the one or more filtration control parameters may be calibrated to influence resistance. As noted herein, resistance at the start of filtration was found to be independent of applied TMP, but resistance transitions to a high resistance regime earlier for lower values of TMP. This can lead to reduced filter media capacity relative to filtrations performed at higher TMP. In contrast, larger values of TMP prolong operating times in the low-resistance regime (e.g., below 100 psi-m2-s/L), delaying the transition to high resistance, which results in greater overall filter media capacity. Accordingly, in some embodiments, a method of filtering LNP-containing solution comprises (i) measuring the resistance, (ii) comparing the measured resistance to a threshold value, and (iii) in response to determining that the measured resistance exceeds the threshold value, adjusting one or more filtration control parameters. In some embodiments, a controller may utilize resistance values, so as to increase a feed pressure with an increase in resistance (e.g., linearly proportional to resistance), so as to maintain a substantially constant resistance. [0105] In some aspects, at least one exemplary method of controlling filtering includes adjusting one or more filtration control parameters in response to imaging data. In at least one aspect, the imaging data is obtained by imaging at least one layer of the filter media to visually assess one or more properties of particulate contamination (e.g., the foulant). In particular, the one or more properties of particular contamination include, but are not limited to, a quantity thereof, a distribution thereof, and/or a geometrical profile of the particulate contamination along the surface of the layer(s). The method may include taking one or more SEM images of an outer surface of each media layer at a resolution that is approximately equal to a filter rating and/or a pore size of the media layer. In some embodiments, imaging may be used in characterizing filtration results to determine which operational modes may correlate to various degrees of fouling. For example, SEM images indicating that high pressure is associated with fewer fouling deposits may inform a decision to operate at higher TMPs. Further, the method may include characterizing the solution containing LNPs based on the imaging and a determined extent of fouling. The solution may be characterized by categorizing it in terms of a fouling model (e.g., complete pore blockage, intermediate pore blockage, pore constriction, or cake formation) and/or based on the nature of the foulant (e.g., incompressible or compressible). [0106] In some embodiments, the method further includes (i) evaluating individual layers of filter media, (ii) comparing the filtration performance between the filter layers, and (iii) controlling one or more filtration control parameters based on a differential in performance between the individual layers. [0107] In some embodiments, the method further includes determining an extent to which contaminants are retained and/or removed by the filter media. Among other benefits, such a process may yield information regarding whether additional stages are required to achieve the desired performance, and/or whether the type and/or geometry of the filter media needs to be adjusted. In some embodiments, the method includes performing re-filtration processing in which a feed sample containing LNPs is passed through redundant stages of filter media (e.g., passing the feed solution through a first filter media to generate a first filtered solution, and then passing the first filtered solution through a second filter media that matches the type and geometry of the first filter media). [0108] Some embodiments comprise using a re-filtration process involving two sequential filters (e.g., each filter being two layers). [0109] In some embodiments, approximately 85% to approximately 100% of LNPs in the LNP-containing solution are recovered. In some embodiments, approximately 95% to approximately 98% of LNPs are recovered. In some embodiments, approximately 90% to 96% of LNPs are recovered. In some embodiments, approximately 65% to approximately 99% of LNPs are recovered. [0110] In some embodiments, the method further includes adjusting one or more filtration control parameters based on data from a re-filtration operation. In some embodiments, a derivative analysis of re-filtration data (data relating to the LNP-containing solution) may be performed. [0111] In at least one embodiment, the method further includes conducting additional adjustments to one or more filtration control parameters based on temporal changes in test conditions. For instance, in at least one embodiment, the method additionally includes performing pressure stepping to account for the influence of (i) membrane fouling at different pressures; and (ii) changes in the composition of the feed solution, where the pressure stepping data may be used to determine whether to adjust one or more filtration control parameters. [0112] In some embodiments, the method additionally includes performing pressure stepping in reverse. Such approaches may result in a TMP-dependent change in filtration performance. In turn, one or more filtration control parameters may be adjusted to account for the TMP-dependent change. [0113] In some embodiments, the method additionally includes adjusting one or more filtration control parameters based on expected temporal changes in the composition of the feed solution which may impact filtration performance. [0114] Techniques for characterizing the underlying mechanisms controlling sterile filtration of LNPs according to an aspect of the present disclosure will now be described. In some aspects, the method may include additional, fewer, and/or different operations.
Evaluation of Filtration Performance and Characteristics [0115] In at least one aspect, a method according to the present disclosure includes evaluating the filtration performance of filter media using a feed solution containing LNPs (e.g., a solution containing LNPs, a buffer solution, etc.). For example, the method may include performing evaluations of sterile filter media to determine how filtrate flux and/or the throughput of the filter media vary with time at different TMPs. FIGS.4-5 show scatter plots of experimental data from sterile filtration of an example feed solution at different TMPs. Each dataset shows the change in the filtrate flux and volumetric throughput of the filtrate throughout the duration of the test. [0116] As used herein, TMP refers to the pressure drop across the filter media (e.g., in units of psi), “filtrate flux” refers to a rate of transfer of the feed solution through the filter media (e.g., in units of L/m2/hr), and “volumetric throughput” refers to a total amount of filtrate (e.g., permeate, filtered solution, etc.) processed by the filter media at different times throughout the test per unit membrane area (e.g., in units of L/m2). [0117] As shown in FIG.4, tests were conducted at four different values of TMP, including approximately 2 psi, approximately 8 psi, approximately 14 psi, and approximately 20 psi, using a two-layer Sartopore® 2 XLG filter having a coarse layer and a fine layer. In particular, FIG.4 shows a scatter plot of filtrate flux as a function of the volumetric throughput during constant pressure filtration of LNPs through a Sartopore® 2 XLG capsule at the foregoing pressures. The tests indicate that the rate of decrease in filtrate flux increased over the duration of the test regardless of TMP, with the filtrate flux decreasing by as much as approximately 99% for some of the tests. However, the test data indicate filter capacity increased with TMP for the same filter media type and geometry. [0118] Table 2 below summarizes values of total volumetric throughput at each test condition shown in FIG.4. Notably, the LNP recovery (e.g., the concentration of LNPs in the permeate relative to the concentration of LNPs in the feed solution) was similar at each test condition, regardless of the amount of filter fouling (e.g., restriction across the filter media). The values shown in Table 2 are approximate values.
Figure imgf000029_0002
Table 2 [0119] In some aspects, a method according to the present disclosure includes evaluating the data using different filtration performance parameters and/or derived metrics. For example, the method may include plotting the test data in terms of measured resistance to flow across the filter media during each test, where “resistance” refers to the value of TMP divided by the filtrate flux at each data point during the test (e.g., the TMP normalized by the measured filtrate flux at each data point, in units of psi-m2-s/L). [0120] For example, FIG.5 shows a scatter plot of the measured resistance at each data point from FIG.4. In particular, FIG.5 shows a scatter plot of resistance as a function of the volumetric throughput during constant pressure filtration of LNPs through the Sartopore® 2 XLG capsules at pressures of approximately 2 psi, approximately 8 psi, approximately 14 psi and approximately 20 psi. FIG.5 shows data from FIG.4 re-plotted according to the following equation:
Figure imgf000029_0001
[0121] As shown, the resistance at the start of filtration is similar for each data set, regardless of the applied TMP. However, the resistance transitions to a high resistance regime much earlier for lower values of TMP. This leads to reduced filter media capacity relative to tests performed at higher TMP. In contrast, larger values of TMP prolong operating times in the low-resistance regime (e.g., below 100 psi-m2-s/L), delaying the transition to high resistance, which results in greater overall filter media capacity. [0122] Dynamic light scattering (DLS) analysis of the permeate and feed samples used to obtain the data depicted in FIGS.4-5 showed no substantial change in the LNP size distribution, with the Z-average diameter for the permeate samples within 2% of that for the feed. The initial filtrate flux (Jo) was approximately equal to the buffer flux evaluated immediately prior to the filtration experiment (at approximately the same TMP), with the Jo values increasing linearly with increasing TMP, consistent with a constant membrane permeability. The filtrate flux (J) is plotted as a function of the volumetric throughput (v), defined as the ratio of the cumulative filtrate volume (V) to the membrane area (A). In each case, the curves are concave down on the semi-log plot, with the absolute value of the slope increasing with increasing throughput. [0123] The initial rate of flux decline, -dJ/dv, decreases with increasing transmembrane pressure, while the filter capacity (Vmax), evaluated as the volumetric throughput at which the filtrate flux declines to 10% of its initial value, increases from 32 L/m2 at 2 psi to 69 L/m2 at 20 psi. For most biologics, increasing transmembrane pressure during sterile filtration results in either decreased capacity or constant capacity. The observed large increase in capacity for filtration of LNPs, and decrease in rate of flux decline, is unusual for most sterile filtration processes. The flux declined by more than an order of magnitude due to the deposition of LNP, primarily on the upper surface of the 0.2 µm layer of the Sartopore 2 XLG membrane. More strikingly, the capacity increased with increasing TMP, which is the opposite of what would be expected for a compressible fouling deposit. [0124] The increase in capacity with increasing TMP was assessed by directly evaluating hydraulic resistance of the deposited LNPs (defined as the ratio of the TMP to the filtrate flux) as a function of TMP. A layer of LNP was first deposited on the surface of the filter, buffer was filtered through this layer for approximately 60 minutes to obtain a stable flux, after which the buffer flux was measured at multiple TMP. FIGS.20A-20B show the total resistance of the membrane with the deposited LNPs as a function of filtration time during a pressure-stepping experiment, using both Isopore and Sartopore membranes. [0125] In particular, the TMP was varied in a stepwise fashion in 30-minute intervals, starting at 140 kPa (20 psi), decreasing to 56 kPa (8 psi), and then returning to 20 psi for deposited LNPs on a 0.2 µm pore size Isopore track etch membrane (FIG.20A) and on the 0.2 µm layer of the Sartopore 2 XLG (FIG.20B). The hydraulic resistance of clean Isopore and Sartopore membranes were 3.0 ± 0.1 x 10-3 and 2.1 ± 0.1 x 10-3 (psi/(L/m2/h)) (7.5 ± 0.3 x 107 and 5.2 ± 0.2 x 107 Pa/m/s), respectively, which are more than 100-fold smaller than the values measured in FIGS.20A-20B. Because the total resistance (i.e., membrane plus deposit) is controlled predominantly by the contribution from the deposited LNPs, and the membrane is an incompressible polymer at the pressures studied, the measured total resistance was taken as representative of the resistance of the deposited LNP. [0126] The initial resistance at 20 psi was constant for both membranes with values of approximately 0.23 ± 0.02 and 0.42 ± 0.2 (psi/(L/m2/h)) for the Isopore and Sartopore 2 XLG membranes, respectively, confirming that the deposited LNP had formed a stable fouling layer. An incompressible fouling layer is expected to exhibit a constant resistance over the full range of TMP, the behavior observed with the clean membranes. In contrast, the resistance of a compressible system is expected to be higher at high pressure. Surprisingly, when the TMP was reduced to 8 psi, the hydraulic resistance increased by approximately two-fold, which is opposite of what would be expected for a fouling deposit that is compressible. The resistance for the fouling deposit on the Isopore membrane (FIG.20A) returned to its original value upon increasing the pressure back to 20 psi, indicating a reversible behavior, i.e., there were no permanent changes in the fouling deposit. In contrast, the resistance of the fouling deposit on the Sartopore 2 XLG membrane (FIG.20B) during the second 20 psi step was below the initial resistance at this TMP. This may be attributable to the transmission of some of the deposited LNP through a Sartopore XLG, as permeate samples obtained during the second filtration at 20 psi showed a low but non-zero level of mRNA based on the concentration. [0127] In some embodiments, the method further includes conducting tests on individual layers of filter media and comparing differences in the filtration performance between the filter layers. For example, FIG.10 shows a scatter plot of sterile filtration performance using individual layers of the filter media from FIGS.2A, 2B and 3 discussed below. More particularly, FIG.10 depicts filtrate flux as a function of the volumetric throughput for filtration through the individual layers of the Sartopore® 2 XLG dual-layer filter. [0128] In particular, for FIG.10, data were obtained for the coarse filtration layer 102, the fine filtration layer 104, and the combined filter media including both the coarse filtration layer 102 and the fine filtration layer 104 at a fixed TMP (e.g., approximately 14 psi). FIG. 10 shows the flux profiles as a function of volumetric throughput at a constant pressure of approximately 14 psi for the Sartopore® 2 XLG dual-layer filter housed in an ultrafiltration cell. The lower capacity of the dual-layer filter compared to that seen in FIGS.4-5 reflects batch-to-batch variability in the results. Both the 0.2 and 0.8 µm layers showed no measurable change in LNP size in the filtrate samples as determined by DLS. The 0.8 µm layer alone had the highest filtrate flux and lowest rate of flux decline, with a filter capacity Vmax of approximately 230 L/m2 (defined as the throughput corresponding to 90% flux decline). [0129] As shown in FIG.10, the capacity of the coarse filtration layer 102 is significantly larger than either the fine filtration layer 104 or the combined filter media 100 (e.g., approximately 7 times greater than the combined filter media 100 for the same TMP). In contrast, the fine filtration layer 104 tested on its own showed the lowest capacity of all three data sets and provided the lowest measured LNP recovery. In particular, the 0.2 µm layer alone showed a rapid flux decline and a small filter capacity Vmax of approximately 16 L/m2. With respect to the combined filter media (the dual-layer filter) 100, the results indicate a modest increase in the overall filter capacity when compared with the fine filtration layer 104 on its own, which can be attributed to the use of the coarse filtration layer 102 as a pre-filter that protects the fine filtration layer 104 from key contaminants (e.g., foulants, etc.). The dual-layer filter 100 showed an intermediate filter capacity Vmax of approximately 39 L/m2, substantially smaller than the 0.8 µm layer alone yet almost two and a half-fold higher than the 0.2 µm layer. These results demonstrate that the bulk of the fouling occurs in the 0.2 µm layer of the Sartopore® 2 XLG, with the 0.8 µm layer providing protection to the downstream layer (which may be by retaining foulants that are present in the feed). [0130] The solid curves in FIG.10 were obtained using the classical form of the complete pore blockage model:
Figure imgf000032_0001
[0131] The best-fit value of ^^, a parameter describing the rate of blockage, was determined by non-linear regression using Mathematica. The model closely fits the flux data for both the 0.2 µm layer alone and for the dual layer (α = 0.053 and 0.022 m2/L, respectively, with R2 values > 0.98), consistent with LNP fouling being predominantly due to pore blockage in the 0.2 µm layer. The smaller value of ^^ for the dual layer filter reflects the removal of foulants by the pre-filter. In contrast, the complete pore blockage model poorly fits the flux data for the 0.8 µm layer alone, suggesting that the fouling observed in this layer is not completely described by a pore blockage mechanism. The substantial difference in capacity between the individual membrane layers, as well as the difference in fouling mechanisms, suggests that the major foulants are likely smaller than 0.8 µm in size, indicating that the LNPs themselves can be the primary foulants rather than higher order, large-scale aggregates. [0132] In some embodiments, the method of characterizing the factors governing and/or contributing to filter media fouling may further include performing a derivative analysis to determine the physical mechanism with the greatest contribution to overall filter fouling (e.g., complete pore blockage, pore constriction, intermediate blockage, or cake formation). In at least one embodiment, the derivative analysis includes comparing the first and second derivatives of filtration time (t) with respect to the cumulative filtrate volume (V) as provided in Equation (3) below:
Figure imgf000033_0001
where the value of n corresponds to the mode of filter media fouling (e.g., n = 0 corresponds to cake formation (in which foulants accumulate on the membrane surface in a permeable cake), n = 1.5 corresponds to pore constriction (in which the pore radius decreases as foulants deposit along the pore walls), n = 2 corresponds to complete pore blocking (in which foulants block pore entrances), and n = 1 (in which foulants either block pore entrances or deposit on previously blocked pores) corresponds to intermediate pore blocking). [0133] In some embodiments, a derivative analysis may be performed for the derivative of TMP (P) with respect to filtrate volume (V), as shown below in Equation (4), where n is defined as noted above for Equation (3).
Figure imgf000033_0002
[0134] For example, FIG.11 shows a scatter plot of a derivative analysis performed on the experimental data from FIGS.4-5. In particular, FIG.11 depicts a derivative plot where the y-axis corresponds to the second derivative and the x-axis corresponds to the first derivative. As shown, the data are plotted on log-log axis scaling. All derivatives were evaluated numerically using a finite difference analysis accounting for the non-constant intervals for the volumetric throughput (accurate to second order in ∆v), with the derivative averaged over approximately 20 second intervals to minimize numerical noise. [0135] The data at various values of TMP overlap along a linear trend line with a slope corresponding to the value of n=2, indicating a filter fouling mode consistent with complete pore blockage in the filter media. In particular, the TMP values were approximately 2 psi, approximately 8 psi, approximately 14 psi and approximately 20 psi for LNP filtration through a Sartopore® 2 XLG filter. The solid lines represent the slope (n=2, as noted above). [0136] FIG.12 shows test data from an example re-filtration operation. In particular, FIG.12 shows filtrate flux as a function of volumetric throughput during filtration of the fresh LNP and previously filtered LNP through the Sartopore® 2 XLG according to the working examples. The test data were obtained from a re-filtration process involving two sequential filters (each being two layers). In a first filtration operation (labeled as “1st filtration” in FIG.12), a feed sample containing fresh (e.g., unfiltered) LNPs is passed through a first stage sterile Sartopore® 2 XLG filter media at a constant TMP of 14 psi to produce a first filtration permeate. Next, the first filtration permeate is directed through a redundant sterile Sartopore® 2 XLG filter having the same geometry as the first stage filter media (labeled as “refiltration” in FIG.12). The data in FIG.12 were obtained from filtration experiments performed on the same day, with the collected permeate used within 1 hour of the first filtration. [0137] As shown in FIG.12, although the redundant sterile filter media still shows significant fouling at the conclusion of the test, the data show an approximately 2-fold increase in the filter capacity relative to filter media in the first filtration stage. In particular, an approximately 2-fold increase in the filter capacity was observed as TMP was increased from approximately 2 psi to approximately 20 psi. The data suggest that the first filtration stage reduces the concentration of contaminants even at high LNP recovery. In addition, although foulants were removed during the first filtration, they were not removed completely. The general fouling behavior during re-filtration was unchanged, with pore blockage still the dominant mechanism. This suggests that fouling might occur by a stochastic mechanism, with a small fraction of the LNPs being captured by the sterilizing-grade layer of the Sartopore® 2 XLG membrane. This fouling behavior can depend on the individual particle morphology and surface characteristics, flow velocity, and/or membrane pore structure at specific locations on the filter. [0138] In some embodiments, the method further includes conducting a derivative analysis of the data from the re-filtration operation. In some embodiments, a derivative analysis of re-filtration data (data relating to the LNP-containing solution) may be performed. Such an analysis may show, for example, that the re-filtration data indicates filter fouling behavior that is consistent with the complete pore blockage model, similar to test data from FIGS.2A-2B discussed below. Pressure Stepping [0139] In at least one embodiment, the method further includes conducting additional evaluation to determine how the performance of the sterile filtration operation varies with temporal changes in test conditions. For instance, in the example filtration operation described herein, the method additionally includes performing pressure stepping experiments to determine the influence of (i) LNP deposition at different pressures; and (ii) changes in the composition of the feed solution. [0140] FIG.13 shows a scatter plot showing test data from a pressure stepping experiment using the Sartopore® 2 XLG filter media. During a first part of the test, a feed solution containing LNPs is directed through sterile filter media at constant TMP. As shown in FIG.13, the TMP is fixed at approximately 14 psi for a first time period (e.g., within a range from the start of the filtration up to approximately 150 seconds or approximately 200 seconds, or another suitable period). Next, a pressure upstream of the filter media is adjusted to cause a first stepwise change in TMP during the test. In the example of FIG.13, the TMP is adjusted after the first time period from a first TMP of approximately 14 psi to a second TMP of approximately 1 psi. Testing continues at constant TMP for a second time period until a trend in filtration performance is established. [0141] As shown in FIG.13, at the end of the second time period, a pressure upstream of the filter media is adjusted to cause a second stepwise change in TMP back to the first TMP value. Testing continues at the first TMP for a third time period until a trend in filtration performance is established. It should be appreciated that test parameters in the pressure testing experiment may be adjusted as desired to determine the effects of changes in test conditions. [0142] Contrary to the expected behavior associated with existing biotherapeutics, such as compressible cake fouling of the filter media, the results from the pressure stepping experiment indicate non-linear behavior with respect to the filtrate flux. Filtration of LNP solutions may exhibit non-linear behavior with respect to any of filtrate flux, resistance or transmembrane pressure (TMP), for example. In particular, non-linear changes in filtrate flux were observed in response to stepwise changes in TMP. In particular, the data in FIG.13 show an approximately 70-fold increase in filtrate flux in response to an approximately 14- fold increase in TMP. Additionally, the test data at approximately 14 psi after the stepwise change in TMP is continuous with the data trends observed at the start of the test, indicating no irreversible changes in filter fouling due to operation at 1 psi. Moreover, the data in FIG. 13 show that performing sterile filtration of LNPs at high TMP also reduces the rate of change in filtrate flux (e.g., the rate of decline of the filtrate flux). [0143] In some embodiments, the method additionally includes performing pressure stepping experiment in reverse, which may help identify or yield further insight into the physical mechanism or mechanisms affecting TMP-dependent changes in filtration performance. [0144] For example, FIG.14 shows a scatter plot of test data of a reverse pressure stepping experiment starting at an initial TMP of approximately 1 psi. Such an experiment, in addition to any of the experiments described herein, may be carried out utilizing a kit including a filter (e.g., filter media) and a solution containing LNPs. As shown in FIG.14, a stepwise change in TMP from 14 psi to 1 psi causes an approximately 150-fold reduction in filtrate flux, which is even greater than the changes observed during the initial pressure stepping experiments. As with previous testing, and contrary to behavior expected with compressible cake fouling, the resistance to sterile filtration of LNPs is shown to decrease at large TMP. The data indicates that high TMPs may reduce filtration resistance. In some embodiments, at high TMPs, the LNPs 10 can foul the dual-layer filter by blocking the pores in the downstream sterilizing-grade membrane layer. [0145] In some embodiments, the method additionally includes performing tests to determine how temporal changes in the composition of the feed solution impact filtration performance (e.g., to determine whether the TMP-dependent changes in filtration performance are also observed with the buffer solution). For example, FIG.15 shows test data from a pressure stepping experiment in which the composition of the feed solution is changed during the test. As with other tests described herein, the sterile filtration test is performed using a Sartopore® 2 XLG dual layer media. The test begins by introducing a feed solution containing LNPs at constant pressure (e.g., approximately 14 psi). After a first time period of constant pressure filtration, the LNP feed solution is replaced with a buffer solution that does not include LNPs. Testing continues for a second test period after which the TMP is rapidly decreased from approximately 14 psi to approximately 2 psi. [0146] As shown in FIG.15, changes in filtrate flux of the buffer solution are similar to those observed at the end of filtration with the feed solution containing LNP. A similar pressure dependence as the LNP feed solution is also observed in response to changing TMP. As shown in FIG.15, an approximately 7-fold decrease in TMP results in an approximately 30-fold decrease in filtrate flux. The results indicate that the TMP-dependent filtration performance may not be attributable to changes in LNP deposition in the filter media but rather to how the existing deposited LNPs respond to changes in TMP. [0147] Additional pressure stepping experiments were carried out according to the working examples. The dependence of resistance on pressure was observed, and the results (e.g., in FIGS.16-17) were contrary to the expectation that an incompressible fouling deposit would have a resistance that is independent of pressure, while a compressible fouling deposit would show a resistance that is greater at high pressure because compression increases the packing density of the deposit, making it less permeable. Rather, the data obtained with LNPs 10 indicate that the unique characteristics of the LNPs lead to a sharp reduction in resistance at high transmembrane pressures. [0148] Furthermore, results from testing of the Sartopore® 2 XLG dual layer filter media show high levels of LNP recovery despite observed fouling of the filter media. In particular, LNP recovery of between approximately 90%, approximately 91%, approximately 92%, approximately 93%, approximately 94%, approximately 95%, approximately 96%, approximately 97% or approximately 98% was exhibited in some embodiments. Contrary to behaviors typically associated with compressible fouling deposits, the capacity of the filter media for sterile filtration of LNPs is shown to increase with increasing TMP. [0149] SEM imaging, discussed below, confirms fouling primarily on the upper surface of the fine filtration layer of the filter media. Re-filtering of LNP solution contributes to less fouling but similar behavior with respect to changes in filtrate flux over time, suggesting a reduction in concentration of key foulants in the LNP solution. As appreciated from FIG.11, derivative plotting shows a slope of n = 2, consistent with complete pore blockage of the filter media. However, pressure stepping experiments show behavior that is opposite from that expected with compressible fouling deposits. Imaging of Fouling Phenomena [0150] In some embodiments, deposits on filters of the LNP 10 (also called the mRNA- LNP 10) can be characterized using imaging, microscopy, or hydrodynamic metrological techniques or any combination of the foregoing. For example, one or more of scanning electron microscopy (SEM), environmental scanning electron microscopy (ESEM), atomic force microscopy (AFM), or hydraulic resistance measurements can be used to characterize deposition on filter media. [0151] FIGS.2A, 2B and 3 show SEM images of an example sterile filter media, shown as filter media 100. The SEM images depicted herein were taken as described in the working examples discussed below. In at least one embodiment, the filter media 100 includes a polyethersulfone, dual-layer sterile filter media such as the Sartopore® XLG dual-layer filter (e.g., Sartopore® 2 XLG filter made by Sartorius Stedim Biotech GmBH of Göttingen, Germany) or other suitable sterilizing-grade filter media, as described in the working examples. Among FIGS.2A, 2B and 3-18, the SEM images in FIGS.2A, 2B, 3 and 6-9 are SEM images of the Sartopore® 2 XLG filter media according to the working examples. FIGS.4-5 and 10-18 depict data obtained using the same batch of LNPs made according to the working examples. [0152] As shown in FIGS.2A, 2B and 3, the filter media 100 includes a coarse filtration layer 102 (e.g., pre-filtration layer) and a fine filtration layer 104 coupled to the coarse filtration layer 102. In at least one embodiment, the coarse filtration layer 102 is layered on top of the fine filtration layer 104 and positioned upstream from the fine filtration layer 104 during the sterile filtration operation to remove the largest contaminants from the feed solution. [0153] FIGS.2A-2B show SEM images of an upper surface of the coarse filtration layer 102 of the filter media 100 at 5 µm resolution. Specifically, FIG.2A and FIG.2B are SEM images of the 0.8 µm layer of the Sartopore® 2 XLG, after filtration of buffer (FIG.2A) and after filtration of the LNPs (FIG.2B) through the dual-layer filter at a pressure of approximately 14 psi until a flux decline of more than 99% occurred, with scale bars of 5 µm. [0154] FIG.3 shows an SEM image of an upper surface of the fine filtration layer 104 of the filter media 100 at 5 µm resolution, where the fine filtration layer 104 is in an unused condition. [0155] In some aspects, a method according to at least one aspect includes characterizing the factors governing and/or contributing to filter media fouling. In at least one aspect, the method includes imaging at least one layer of the filter media to visually assess one or more properties of particulate contamination (e.g., the foulant). In particular, the one or more properties of particular contamination include, but are not limited to, a quantity thereof, a distribution thereof, and/or a geometrical profile of the particulate contamination along the surface of the layer(s). The method may include taking one or more images using microscopy of an outer surface of each media layer at a resolution that is approximately equal to a filter rating and/or a pore size of the media layer. A variety of microscopy imaging tools may be used, as discussed further below. [0156] In at least one embodiment, the method includes taking “before” and “after” images of each individual media layer in a clean (e.g., unused) condition and an at least partially fouled (e.g., used) condition. For example, FIGS.6–7 are SEM images of an outer surface of a new and used coarse filtration layer of the filter media 100 of FIGS.2A-2B, shown as new coarse layer 108 and used coarse layer 110, respectively. The overall structure of the coarse filtration layer 102 and pores within the used coarse layer 110 remain visible in the SEM image. Thus, the SEM images indicate only minimal fouling on the coarse filtration layer 102. [0157] FIGS.8-9 are SEM images of an outer surface of a new and used fine filtration layer of the filter media 100 of FIG.3, shown as new fine layer 112 and used fine layer 114, respectively. Specifically, FIGS.8-9 are SEM images of the 0.2 µm layer of the Sartopore® 2 XLG filter, before and after filtration of the LNPs through the dual-layer filter at a pressure of approximately 14 psi until a flux decline of more than 99% occurred, with a scale bar of 5 µm. In contrast to SEM images of the coarse filtration layer 102, the SEM images in FIGS. 8-9 show extensive fouling on the used fine layer 114 with contaminants completely blocking the entrance to the pores across at least one section of the used fine layer 114. [0158] Returning to FIGS.6-7, as seen in these figures, the 0.8 µm layer has a rough, asymmetric surface with relatively low pore density. In contrast, as seen in FIGS.8-9, the 0.2 µm layer appears substantially smoother with a higher density of relatively circular pore openings. There is no visible difference in the 0.8 µm pre-filter after fouling, suggesting most of the pores remain fully open, which is consistent with the very high filtration capacity when the 0.8 µm layer was used alone, as indicated in FIG.10. In contrast, as seen in FIG.9, much of the surface of the 0.2 µm layer is covered with LNPs 10, appearing as an aggregated layer on top of the membrane. These may also be referred to as “mRNA-LNPs” or “deposited LNPs.” The structure and organization of the LNPs seen in the SEM images in FIGS.2A, 2B, 3 and 6-9 may be affected by the sample preparation (drying) and may appear different in the native (wetted) environment, although the high degree of surface fouling is consistent with the pore blockage mechanism identified by the derivative analysis in FIG.11. [0159] In at least one aspect, a method of evaluating sterile filtration of LNPs is provided. In particular, performance of a filter during LNP filtering can be evaluated. The method includes obtaining LNPs, e.g., an mRNA-LNP solution. The method includes depositing the LNPs on membranes respectively varying in pore size. For example, the membranes may be a plurality of membranes wherein each membrane has a pore size that is larger or smaller by at least 0.1 um than another of the membranes. For example, membranes of 0.1 um, 0.2 um and 0.4 um pore size may be evaluated. The membranes were then housed in a filtration cell on top of a mesh. The method may further include determining the hydraulic resistance of the membranes when buffer was filtered through the membranes. In particular, the buffer flux was measured at various transmembrane pressures to determine the hydraulic resistance. The method further includes supplying the filtration cell with the mRNA-LNPs, supplying buffer through a feed reservoir, and performing filtering of the buffer and the mRNA-LNP solution through the membranes at a constant TMP to deposit LNPs on the membrane. The method further involves comparing the buffer flux of the membrane with the buffer to that with the deposited LNPs, and determining the hydraulic resistance of the deposited LNPs. [0160] In at least one embodiment, a method of evaluating sterile filtration of liquid nanoparticles (LNPs) is provided. The method includes filtering a first solution lacking LNPs (e.g., buffer) through at least one membrane. The method may include controlling a filtration control parameter during filtering to satisfy a control parameter threshold. The filtration control parameter may include a TMP during filtering and/or a flux of the first solution through the membrane. The control parameter threshold may be a target TMP and/or target flux of the first solution. As such, controlling the filtration control parameter to satisfy the control parameter threshold may include adjusting the TMP and/or flux of the first solution to approximately equal the target TMP and/or the target flux through the membrane. The method additionally includes determining a first hydraulic resistance of the membrane following filtration of the first solution. The method further includes filtering a second solution containing LNPs through the at least one membrane; and determining a second hydraulic resistance of the membrane following filtration of the second solution. During filtering, the filtration control parameter may be controlled to satisfy the control parameter threshold. [0161] Additionally, the method further includes computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; and imaging the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling (and/or the computed difference between the first hydraulic resistance and the second hydraulic resistance). In particular, evaluating an SEM image, for example, as well as experimental filtration data, allows for characterization of both membrane performance and the behavior of LNPs themselves. This information can be used in determining whether or not to alter membrane selection or adjust one or more filtration control properties as discussed further herein. In some embodiments, the one or more filtration control properties is a pressure value, PCRITICAL, discussed herein. WORKING EXAMPLES [0162] The following working examples are illustrative and non-limiting. Example 1- Materials [0163] Experiments were performed with an mRNA-LNP solution (LNP) provided by Moderna Inc. of Cambridge, MA. Frozen LNP solution was thawed in a room temperature water bath immediately before use. [0164] Sterile filtration data were obtained using track-etch polycarbonate Isopore membranes with 0.1 µm, 0.2 µm, and 0.4 µm pore sizes (MilliporeSigma, Bedford, MA), as discussed below. More specifically, LNPs were deposited on the polycarbonate Isopore membranes with 0.1 µm, 0.2 µm, and 0.4 µm pore sizes, which have smooth and uniform surfaces with a parallel array of straight-through cylindrical pores, facilitating microscopic analysis. In addition, sterile filtration data were obtained using Sartopore® 2 XLG dual-layer asymmetric polyethersulfone (PES) filters having a 0.8 µm pore size layer on top of a 0.2 µm layer. [0165] Membranes were sealed in 25-mm diameter capsules provided by Sartorius Stedim Biotech GmBH of Göttingen, Germany. Data were obtained for the individual layers of the filter housed at the base of a 25-mm Amicon 8010 filtration cell made by MilliporeSigma, of Bedford, MA. The filtration cell was operated without a magnetic stirrer, and with a 5 µm pore size nylon mesh placed beneath the filter membrane to promote flow distribution.
Example 2 – Filtration [0166] Filtration experiments were performed at substantially constant TMP. The filter capsule was fed by a 1-liter stainless-steel feed reservoir made by Alloy Products Corporation, of Waukesha, WI. The reservoir was pressurized with compressed air and controlled using a pressure regulator. TMP was monitored using a digital pressure gauge made by Ashcroft Inc. of Stratford, CT. The pressure gauge was placed immediately upstream of the filter, with the permeate outlet maintained at atmospheric pressure. [0167] The membranes were initially flushed with a minimum of 100 L/m2 of deionized water followed by Tris-sucrose formulation buffer with the latter prepared by mixing appropriate quantities of Tris-base (MilliporeSigma, Catalog Number 9210), Tris–HCl (Invitrogen, 15567-027), and sucrose (MilliporeSigma, 8550). The membrane hydraulic permeability was evaluated by measuring the filtrate flux at several TMP (up to approximately 70 kPa) using the Tris-sucrose buffer. All experiments were performed at room temperature (21 ± 1 oC). [0168] The feed reservoir and the filter capsule were then emptied, and refilled with the LNP solution (typically < 5 mL for a 4.1 cm2 membrane area). The feed reservoir was filled with buffer, and the system was then re-pressurized. The buffer was filtered at a constant TMP for at least 1 hour to stably deposit LNPs on the membrane. Filtrate flow was evaluated by continuous mass readings on an OHAUS RangerTM 3000 scale, with the data logged every second using the OHAUS Serial Port Data Collection Software, both made by OHAUS Corp. of Parsippany, NJ. Permeate concentrations (LNP concentrations) were evaluated based on UV absorbance at 230 nm using a microplate reader made by Tecan of Mannedorf, Switzerland. [0169] The buffer flux through the fouled membrane (with the deposited LNPs) was less than 10% of the buffer flux evaluated through the clean membrane at the same TMP. The hydraulic resistance of the deposited LNPs was then determined by measuring the buffer flux at several specific TMP values obtained with both increasing and decreasing TMP, often using more than one cycle to examine the stability (or hysteresis) of the deposited LNPs. The extent of fouling may be determined based on the difference in hydraulic resistance as compared to the buffer as being, for example, light, moderate or extensive. Example 3 - LNP and Filter Characterization [0170] LNP size distribution was determined by Dynamic Light Scattering (DLS) using a Zetasizer Nano ZS90 made by Malvern Panalytical, United Kingdom. Samples were analyzed at approximately 20ºC after equilibration for approximately 120 s. The light scattering intensity was evaluated for a total of 10 runs. Example 3a – SEM Characterization [0171] LNPs captured on the external surface of the filter were examined by SEM and ESEM. The fouled membrane was removed from the filtration cell, dried inside an Isotemp® 200 Series oven made by ThermoFisher Scientific of Waltham, MA at 30 ºC for one hour, and cut into pieces before being mounted on conductive pins with a double-sided carbon tape on circular pins. A thin layer of conductive gold-platinum alloy was applied using an SCD 050 sputter coater made by Bal-Tec AG of Balzers, Lichtenstein to reduce sample charging. Surface images were obtained with a SIGMA VP Field Emission SEM at 3.0 kV made by Zeiss of Jena, Germany. ESEM samples were imaged in the hydrated state at 75% relative humidity, which was maintained by applying a pressure of 6 Torr at 7 ºC with the samples mounted on a Peltier cooling stage. SEM and ESEM images were obtained with a Quanta 250 ESEM (Thermo-Scientific, Hillsboro, OR) at 3.0 and 20 keV, respectively. [0172] Microscopic characterization was performed according to some embodiments as shown in FIGS.24A-24F. FIGS.24A-24F depict SEM and ESEM images of the 0.2 µm Isopore (FIGS.24A-24C) and 0.2 µm layer of the Sartopore 2 XLG (FIGS.24D-24F) showing the clean membranes (FIG.24A and FIG.24D), SEM images of the heavily fouled membranes (FIG.24B and FIG.24E), and ESEM images of the heavily fouled membranes (FIG.24C and FIG.24F). Scale bars are 10 µm on each of FIGS.24A-24F. [0173] More particularly, deposited LNPs were explored by both SEM and ESEM as shown in FIGS.24A-24F. Images of the upper surface of the 0.2 µm Isopore and Sartopore 2 XLG membranes were obtained after flushing with buffer (clean membrane) or after filtration of the LNP at a constant pressure of 2 psi until the flux declined by more than 99% compared to the buffer flux through the clean (unfouled membrane). This corresponded to filtration of 2 L/m2 for the Isopore and 18 L/m2 for the Sartopore 2 XLG; the much higher capacity of the Sartopore filter is consistent with the slightly greater porosity (pore density) and larger average pore size of this membrane (Table 3, discussed below, and FIGS.24A and 24D). ESEM images of the clean Isopore and Sartopore membranes (not shown) looked nearly identical to the SEM images in FIGS.24A and 24D. [0174] The SEM and ESEM images of the fouled Isopore membrane (FIGS.24B and 24C) show that the pores are almost completely covered by an amorphous deposit. This deposit is thought to primarily be comprised of the various lipids present in the original LNP. The SEM image of the upper surface of the fouled Sartopore 2 XLG (FIG.24E) shows a partial amorphous deposit in addition to a small number of individual LNPs. The fouled Sartopore 2 XLG membrane still shows a significant number of open pores, more than would be expected given the high degree of fouling during the LNP filtration. The open pores may be artifacts arising from the drying of the fouled membrane prior to SEM analysis, as such open pores were not visible in the ESEM image (FIG.24F). The similar structures of the deposits on the Isopore (polycarbonate) and Sartopore (polyethersulfone) membranes are qualitatively consistent with the resistance data in FIG.20 and indicate that membrane surface chemistry has a limited effect on the properties of the deposited LNPs. The SEM and ESEM images both suggest that fouling occurs primarily on the upper surface of the filter. Example 3b – Atomic Force Microscopy Characterization [0175] The deposited LNPs were analyzed by Atomic Force Microscopy (AFM) in a hydrated state. The fouled membrane was removed from the filtration cell and immediately mounted in a petri dish with carbon tape. Sufficient buffer was added to fully cover the membrane. The sample was then imaged using the PeakForce Tapping mode on a BioScope Resolve AFM (Bruker Corp., Billerica, MA) with a scan rate of 0.3 Hz and a peak force setpoint of approximately 500 pN. A 6 x 6 µm2 scan was produced with 512 x 512 pixels. A ScanAsyst-Fluid+ cantilever (Bruker Corp., Billerica, MA) with a nominal spring constant of 0.7 N/m and a tip radius of 2 nm was used to generate images, with image processing performed using Gwyddion 2.61 software (Czech Metrology Institute, Jihlava, Czech Republic). [0176] To determine thickness of a fouling deposit, AFM characterization of a partially fouled 0.4 µm Isopore membrane was carried out, as shown in FIGS.25A-25B. The traces in FIG.25B show the height of the deposit as a function of the surface distance at the two locations indicated on FIG.25A by the two horizontal lines. The deposited LNPs were formed on the 0.4 µm Isopore membrane by filtering the LNP until a flux decline of only 50%. The fouled membrane was examined in the hydrated state by AFM. In this case, the surface of the Isopore membrane is only partially covered by an amorphous deposit (FIG. 25A), with a small number of individual LNPs (or small LNP aggregates) visible near the bottom of the figure. The height scans across two locations of the fouled membrane (FIG. 25B) show that the amorphous deposit and the individual LNP are approximately 100 ± 15 nm thick, corresponding to the expected size of LNPs. The open pores in FIG.25B are clearly visible as sharp triangular depressions, reflecting the conical shape of the AFM tip. Example 3c – Bubble Point Measurements [0177] The bubble points for the Isopore membranes and the 0.2 µm layer of the Sartopore 2 XLG were evaluated using the approach described in ASTM F316. The membranes were placed in a 25 mm stainless steel holder and flushed with at least 100 L/m2 of deionized water. The filter exit was connected to a dip tube that was submerged in deionized water. The upstream surface of the membrane was then pressurized using a compressed nitrogen tank, with the pressure increased at a rate of approximately 1 psi per minute. The bubble point was determined as the nitrogen pressure at which the first bubble was observed rising in the beaker. The bubble point for each membrane was measured three times, with the results presented as the mean ± standard deviation. [0178] The largest pore diameters for the different membranes were evaluated from bubble point pressure measurements using the Young-Laplace equation, Equation 4:
Figure imgf000047_0001
where γ is the interfacial surface tension, θ is the contact angle between the fluid and the pore wall, and dp is the effective diameter of the largest membrane pores. [0179] Table 3 depicts the measured bubble point pressures using deionized water and calculated pore diameters for the Isopore membranes and the 0.2 µm layer of the Sartopore 2 XLG filter.
Figure imgf000048_0001
a The pore diameter for 0.1 µm Isopore membrane was determined from SEM images as discussed above. Table 3 [0180] The 0.1 µm Isopore membrane was estimated to have a maximum pore diameter of 0.3 µm based on SEM images showing large pores formed by clusters of three pores that measured approximately 0.3 µm across. The maximum pore diameters estimated using Equation 4 for the 0.2 and 0.4 µm Isopore membranes and the 0.2 µm layer of the Sartopore 2 XLG were consistent with values determined from the SEM images of the clean membranes (FIGS.22A-22D). For each membrane, the estimated diameters of the largest pores (Table 2) are larger than the nominal pore sizes. Example 4 – Pressure Stepping [0181] As described above, pressure stepping experiments were carried out in some embodiments. Specifically, pressure stepping experiments were carried out to examine the effects of pressure on the filtration behavior. TMP was varied in a step-wise fashion during a single, continuous filtration run, without any disruption in the LNP feed. FIG.16 depicts results in which the filtration was started at approximately 14 psi for a first period of approximately 150 seconds. After the first period, the pressure was abruptly decreased to approximately 1 psi for approximately 300 s before being returned to approximately 14 psi. In both cases, the data are plotted as the resistance (evaluated directly from the flux and TMP data using Equation 1), as a function of filtration time. [0182] The initial step-down in pressure resulted in an approximately 7-fold increase in resistance, while the final step-up in pressure resulted in an approximately 6-fold reduction in resistance. This variation in resistance with TMP is generally qualitatively consistent with the higher capacity at higher TMP seen in FIGS.4-5. Example 5 – Reverse Pressure Stepping [0183] FIG.17 depicts results from a reverse pressure stepping experiment involving a single continuous filtration run. In the reverse pressure stepping experiment, filtration was started at approximately 1 psi and increased to approximately 14 psi. Following the increase to approximately 14 psi, the pressure was returned to approximately 1 psi. The initial step-up in pressure caused a small (approximately 1.5-fold) increase in resistance, likely due to the relatively low level of fouling after the 150 seconds of filtration at low TMP. However, the final reduction in TMP caused an approximately 13-fold increase in resistance, similar to the behavior seen in the experiment that was started at 14 psi (FIG.16). Example 6 –Buffer Permeability [0184] The direct effect of pressure on resistance was examined by first fouling the Sartopore® 2 XLG filter with LNPs 10 and then measuring the buffer flux through the fouled membrane at different TMPs. FIG.18 shows the total resistance during buffer permeability measurements performed with the Sartopore® 2 XLG membrane. To avoid de-pressurizing the system, the filter was fed by two pressure reservoirs, one containing the LNPs and one containing buffer, using a 3-way stopcock valve. The initial LNP filtration was conducted at a constant TMP of approximately 14 psi until a flux decline of approximately 90%. The feed was then rapidly switched to buffer at approximately the same TMP. That is, filtration was started with LNPs 10 at approximately 14 psi followed by buffer at approximately 14 psi, buffer at approximately 1 psi, and then buffer at approximately 14 psi. The calculated values of the resistance during this experiment are summarized in FIG.18. [0185] The resistance of the fouled membrane while filtering buffer remained stable at a value approximately equal to that obtained at the end of the LNP filtration, indicating that the deposited foulants were relatively stable (at least in response to a short buffer flush). [0186] The reduction in TMP from 14 to 1 psi caused an approximately 3-fold increase in the resistance, as seen in FIG.18, corresponding to an approximately 40-fold decrease in flux. The resistance returned to its previous value when the TMP was increased back to approximately 14 psi, indicating that the behavior of the fouling deposit was fully reversible. Thus, an unexpected reduction in fouling and increase in capacity were observed at high TMP. Similarly to FIGS.16-17, the decrease in resistance with increasing TMP is contrary to the expectation for a compressible fouling deposit and is also different from the pressure- independent resistance expected for an incompressible fouling deposit. In particular, biphasic behavior was observed wherein the resistance of the deposited LNPs 10 increases at relatively lower pressure (consistent with a compressible filtration medium) and decreasing at higher pressure. In contrast, the expected behavior for biologics is that they tend to form compressible fouling deposits characterized by an increase in the hydraulic resistance with increasing TMP, although lipid-based dispersions may exhibit different behavior. [0187] The pressure-dependent behavior is consistent across the multiple experiments shown in FIGS.4, 15, 16, 17 and 18, suggesting that this phenomenon is intrinsic to LNPs. The increased pressure may deform the fouling deposit, e.g., at the entrance to a blocked pore, potentially due to a distortion of the LNPs 10 when subjected to greater TMP. This behavior appears at least partially or fully reversible, with the LNPs 10 resuming their previous state when the TMP is reduced, restoring the high resistance of the fouling deposit. Example 7 – Filtration with Pressure Stepping [0188] As noted above, in some embodiments, a threshold value is a maximum resistance value which is observed with increasing TMP until a given pressure value or range of pressures is reached, before resistance declines thereafter. In some embodiments, the hydraulic resistance of the deposited LNPs on the 0.2 µm Isopore membrane was obtained by measuring the buffer flux over a range of TMP, as shown in FIG.21. The LNPs were deposited and maintained in equilibrium at 20 psi for one hour, after which the TMP was reduced to 14, 8, and then 2 psi in a step wise fashion before being returned to 20 psi. [0189] Pressure stepping was repeated for two complete cycles for a single deposit formed on the 0.2 µm Isopore membrane (labeled as the first and second decrease and increase in FIG.21). FIG.21 plots the steady-state values of the hydraulic resistance, as evaluated from the average filtrate flux determined over approximately 15 min of filtration at each TMP, as a function of TMP. The error bars represent the range of the 3-4 data points obtained at each pressure. The results for the multiple pressure cycles appear substantially identical, indicating that there are no irreversible changes in the deposited LNPs over the range of TMP. The total time for this experiment was 6 h, indicating that the deposited LNPs also remain stable for extended periods of time. The resistance of the deposit increased with increasing TMP up to a pressure of approximately 14 psi, reaching a maximum value of approximately 8.0 ± 0.3 psi/(L/m2/h), before decreasing to approximately 2.0 ± 0.2 psi/(L/m2/h) at 20 psi. The increase in resistance with increasing TMP at pressures below approximately 14 psi is typical of a compressible fouling deposit. The sharp reduction in resistance when the TMP was increased from 14 to 20 psi is in the opposite direction, with the resistance decreasing with increasing TMP (as in FIGS.20A-20B). As appreciated from FIG.21, the presence of a maximum in the hydraulic resistance of a fouling deposit at intermediate TMP was observed. Example 8 – Filtration with Varying Pore Size [0190] The effect of the pore size of the underlying membrane on the hydraulic resistance of the deposited LNPs was evaluated by depositing LNPs on 0.2 and 0.4 µm Isopore membranes as well as on the 0.2 µm layer of the Sartopore 2 XLG. The LNP was deposited at 20 psi, with the buffer flux then evaluated by step-wise decreasing the TMP; the data for the 0.2 µm Isopore are taken from the first cycle in FIG.21. A modified procedure was also performed for a 0.1 µm Isopore membrane, in which the deposit was formed at 2 psi with the TMP then increased step-wise to 40 psi. The resistance versus TMP for all four membranes is shown in FIGS.22A-D; the first cycle alone is shown for ease of illustration. In all instances, the resistance of the clean membrane was much less than that determined for the membrane with the fouling deposit; thus, the resistance data in FIGS.22A-D is representative of the resistance of the deposited LNPs. [0191] In particular, FIGS.22A-22D show resistance as a function of TMP for deposited LNPs formed on the 0.1 µm Isopore membrane (FIG.22A), the 0.2 µm Isopore membrane (FIG.22B), the 0.4 µm Isopore membranes (FIG.22C) and the 0.2 µm layer of the Sartopore 2 XLG membrane (FIG.22D). The resistance data for the deposits formed on the 0.1 µm and 0.2 µm membranes showed no measurable hysteresis up to 20 psi, while the resistance for the deposits on the 0.4 µm Isopore and 0.2 µm Sartopore 2 XLG showed a lower resistance on the second cycle (as in FIG.20B). This behavior was consistent with the lack of measurable LNP concentration in the permeate samples obtained through the 0.1 and 0.2 µm pore size Isopore membranes, while there was a detectable concentration for the first few permeate samples obtained at high TMP with the larger pore size membranes. [0192] The resistance for the deposited LNPs on the 0.1 µm Isopore and both of the 0.2 µm membranes exhibits a distinct maximum (Rmax) over the pressure range examined in these experiments, with the pressure value corresponding to Rmax being greatest on the 0.1 µm Isopore (approximately 32 psi) and smallest (approximately 8 psi) on the Sartopore 2 XLG membrane. For each membrane, the deposited LNPs exhibit compressible behavior when TMP was less than the pressure value corresponding to Rmax. The opposite effect was observed when TMP was greater than the pressure value corresponding to Rmax. In contrast, the resistance of the deposited LNPs formed on the 0.4 µm Isopore membrane decreases with increasing pressure and shows no regions of compressible behavior at the tested TMPs (i.e., where the pressure corresponding to Rmax ≤ approximately 2 psi), with the resistance at approximately 20 psi being 6-fold smaller than that at approximately 2 psi. Thus, the pressure value corresponding to Rmax represents a threshold value (PCRITICAL) which can be utilized for pressure control, e.g., as an input to a controller as discussed below. [0193] The increase in the value of PCRITICAL with decreasing pore size resembles the behavior observed in bubble point testing described above, where the pressure to overcome the surface tension of a wetted pore is inversely proportional to the pore diameter. A related phenomenon may be at least partially attributable to the reduction in the resistance of the fouling deposit at high TMP. In particular, assuming the fouling deposit are an amorphous and fluid-like layer, higher TMP may lead to intrusion of a portion of the deposit into and through some of the larger membrane pores, effectively unblocking the pores. [0194] FIG.23 depicts the pressure PCRITICAL for the fouled membranes plotted as a function of the inverse of pore size (pore diameter). In particular, FIG.23 shows TMP at the maximum resistance evaluated for deposited LNPs formed on the surface of membranes having different pore sizes. The error bars represent the range between the neighboring pressures evaluated experimentally; thus, the PCRITICAL range for the 0.2 µm Isopore was from approximately 8 to 20 psi based on the results in FIGS.22A-22D. The error bars on the reciprocal pore diameter are determined using the values in Table 1. The threshold pressure corresponding to the maximum in the resistance, PCRITICAL, increased with decreasing pore diameter, with the linear dependence on 1/dp in general accordance with the Young-Laplace Equation (Equation (4)). As shown in FIG.23, a linear regression fit model was suitable with R2 = 0.98; the solid line represents a best fit, with the intercept fixed at zero. The shaded region below and to the right of the line define the space where the deposited LNPs behave as a compressible foulant layer, with the resistance increasing with increasing TMP. The region above and to the left of the line describes conditions in which the combination of higher TMP and larger pore size allow the amorphous deposit to intrude into and through the pores, effectively opening (unblocking) these pores and reducing the overall resistance to flow. [0195] The interfacial surface tension γ of the fluid-like deposit present on the membrane was estimated directly from the slope of the linear regression fit in FIG.23 based on Equation (4) using PCRITICAL as the pressure, yielding γdeposit = approximately 2.5 ± 0.3 mN/m (assuming a contact angle near 0°). Example 9 – Intrusion Control [0196] In some embodiments, filtration may be controlled to limit intrusion of an amorphous deposit into and through the pores of a membrane when filtering LNPs. [0197] FIG.26 depicts a high-resolution SEM image of deposited LNPs on the 0.2 µm layer of the Sartopore 2 XLG membrane, according to an aspect of the present disclosure. High-resolution SEM was obtained to assess the shape and characteristics of the amorphous deposited LNPs on the membrane. FIG.26 indicates possible intrusion of the amorphous fouling deposit through some of the pores. In particular, regions in the amorphous deposit formed on the 0.2 µm Isopore membrane appear annularly shaped (in particular, “donut”- shaped) in the SEM image, suggesting that intrusion had occurred through those pores (FIG. 24B). The annular structure is shown in FIG.26. The “donut” shape may represent regions in which the amorphous deposit intruded through larger membrane pores, causing a defect (“donut hole”) in the fouling layer so as to unblock those pores. [0198] FIGS.20-26 suggest that the reduction in the resistance of the fouling deposit at high TMP is due at least in part to intrusion of an amorphous deposit into and through the pores in the underlying membrane when the pressure exceeds the intrusion pressure, PC. Intrusion was also observed experimentally by a detectable non-zero concentration in the permeate samples obtained at high TMP, particularly for the larger pore size membranes, and may contribute to the hysteresis observed for the deposited LNPs formed on the larger pore size membranes. In particular, a portion of pores that have been intruded into (i.e., penetrated by the LNPs 10) may remain open when the TMP is reduced. The presence of a plurality of pores opened in this manner lowers the overall hydraulic resistance in subsequent pressure cycles. In contrast, the lack of hysteresis on the smaller pore size membranes indicates that the fluid-like amorphous layer of deposited LNPs re-forms more easily than for the large pore size membranes when the TMP is reduced when operating below or close to PC. Accordingly, the behavior of the deposit appears reversible when applying low to moderate pressures and using smaller pore size membranes (e.g., 0.1 μm and 0.2 μm). [0199] FIGS.27A-27F are schematic depictions of the pressure dependence of the hydraulic resistance of the deposited LNPs based on the intrusion observation described above. In particular, the LNPs deposit and coalesce during filtration forming an amorphous deposit (FIG.27A ^
Figure imgf000054_0001
FIG.27B). At TMP < PCRITICAL, the deposit reversibly spreads (FIG. 27C FIG.27D), with the hydraulic resistance increasing with increasing TMP. At TMP > PCRITICAL, the deposit is pushed into and through the membrane pores (FIG.27E ^
Figure imgf000054_0002
27F). [0200] In particular, during filtration, a subset of LNPs 10 begin to accumulate on the upper surface of the membrane due to the size-based retention of individual LNPs 10 (FIG. 27A), which may also be affected by surface chemistry-driven adsorption. It is thought that the LNPs 10 coalesce, thereby creating an amorphous deposit that spans the pores of the underlying membrane (FIG.27B) with interfacial surface tension γdeposit. When the pressure is less than PCRITICAL, it is theorized that an increase in TMP causes the amorphous deposit to spread in a layer of about 100 nm thickness ^ 15 nm, similar to the diameter of a single LNP in the feed sample (FIGS.27C-27D). Spreading of the layer thus covers a greater fraction of the membrane surface, thereby increasing resistance. When the pressure is greater than PCRITICAL = (4γdeposit)/dp , the high TMP causes the amorphous deposit to intrude into and through some of the membrane pores, effectively unblocking these pores and reducing the overall hydraulic resistance to flow (FIG.27E and FIG.27F). This results in unique behavior where the resistance decreases with increasing TMP, opposite to that observed for compressible foulants. The maximum in resistance thus occurs at the maximum pressure for which the amorphous deposit remains intact (with no intrusion through the pores of the underlying membrane). As mentioned, this behavior appears to be reversible on smaller pore size membranes where the transmission of LNP is low or negligible (at least for TMP that is, for example, no larger than 10% of PCRITICAL). The transmission of LNPs 10 causes hysteresis is at least on the larger pore size membranes. FIG.27F depicts a portion of the amorphous deposit intruding into and penetrating through the previously blocked pores, which is consistent with the SEM images showing “donut”-shaped regions in the fouling deposit (FIG.26). [0201] In particular, in at least one embodiment, filtration may be performed to control one or more filtration control parameters, including (i) an operating pressure, (ii) an operating temperature, (iii) a formulation buffer, or (iv) concentration. In particular, one or more of these parameters or other parameters may be controlled in view of PCRITICAL, e.g., to increase filter capacity. Further, in some embodiments, the foregoing parameters (i)-(iv) may be selected for a given pore size and/or a particle size distribution for the LNPs 10. For example, in at least one embodiment, a controller, as discussed below, is configured to receive inputs relating to a plurality of filtration control parameters. The filtration control parameters may be adjusted to increase capacity for a given membrane for filtering of LNPs 10, thereby enhancing efficiency of filtration. In some embodiments, the control parameter may be derived or determined based on calculations such as a derivative of a measured value (e.g., any of the measured values discussed herein) or an adjustment to a measured value. Further, various filtration control parameters may be maintained as constants or altered, e.g., at different points during a filtration cycle. [0202] For example, in some embodiments, filtration may be performed to initiate filtering of LNP-containing solution at a pressure approximately equal to PCRITICAL or within a predetermined deviation from PCRITICAL (e.g., 5%, 10%, or 20%). In some embodiments, filtration may be performed where PCRITICAL is obtained within a first period before subsequently being lowered. In some embodiments, filtration may be controlled at PCRITICAL from the outset of filtration and maintained for at least part or all of a filtration cycle. Exemplary Controllers [0203] As noted above a controller according to one or more exemplary embodiments may be utilized to control one or more filtration control parameters. In some embodiments, the controller may include a processor and an input/output interface or terminal. The processor may include one or more processors or one or more processors that include multiple processing cores. The processor may be implemented as a single- or multi-chip processor, a digital signal processor (DSP), an application specific integrated circuit (ASIC), a field programmable gate array (FPGA), or other programmable logic device, discrete gate or transistor logic, discrete hardware components, or any combination thereof designed to perform the functions described herein. A processor may be a microprocessor or any conventional processor, or state machine. A processor may be implemented as a combination of computing devices, such as a combination of a DSP and a microprocessor, a plurality of microprocessors, one or more microprocessors in conjunction with a DSP core, or any other such configuration. In some embodiments, the one or more processors may be shared by multiple circuits (e.g., one or more circuits may share the same processor which, in some example embodiments, may execute instructions stored, or otherwise accessed, via different areas of memory). Alternatively or additionally, the one or more processors may be structured to perform or otherwise execute certain operations independent of one or more co- processors. In other example embodiments, two or more processors may be coupled via a bus to enable independent, parallel, pipelined, or multi-threaded instruction execution. All such variations are intended to fall within the scope of the present disclosure. [0204] The controller may include a memory device that is configured to store machine- readable media. The machine-readable media being readable by the processor in order to execute the programs stored therein. The memory device (e.g., memory, memory unit, storage device) may include one or more devices (e.g., RAM, ROM, Flash memory, hard disk storage) for storing data and/or machine-readable media for completing or facilitating the various processes, layers and modules described in the present disclosure. The memory device may be communicatively coupled to the processor to provide computer code, machine-readable media, or instructions to the processor for executing at least some of the processes described herein. Moreover, the memory device may be or include tangible, non- transient volatile memory or non-volatile memory. Accordingly, the memory device may include database components, object code components, script components, or any other type of information structure for supporting the various activities and information structures described herein. The memory device may include or store a database of target values (e.g., a target pressure, a threshold pressure such as PCRITICAL, or target temperature) or other control parameters. [0205] The controller may be communicably coupled to at least one component of the filtration apparatus to control filtration control parameters including, for example, TMP, a static pressure upstream of the membrane, fluid flux through the membrane, test duration, fluid flows, and others. For example, the controller may form part of a control circuit that includes an input/output (I/O) interface (e.g., a network interface card, a communications interface, etc.) that electrically couples the controller with various actuators and control equipment of the filtration apparatus, such as a pump, linear actuator (e.g., for a syringe), a flow control valve, or another actuator and/or piece of flow control equipment. [0206] In some embodiments, the control system also includes at least one sensor that is communicably coupled to the controller through the I/O interface and that provide an indication of operating conditions of the filtration apparatus to the controller. The operating conditions may include a static pressure upstream of the membrane, a fluid flux through the membrane, a fluid temperature, or another fluid condition. The controller may be configured to power or otherwise control operation of the actuators and/or control equipment of the filtration apparatus based on sensor data from the at least one sensor. For example, the controller may be configured to send a control signal, via the I/O interface, to a variable speed pump so that a flux through the membrane satisfies (e.g., is approximately equal to, is within a threshold range of, etc.) a target flux. The controller may also be configured to automatically determine characteristics of the fluid, membrane, or fluid/membrane combination. For example, the controller may be configured to iteratively modify the pressure across and/or fluid flux through the membrane to determine hydraulic resistance or other characteristics across a range of fluid control parameters, and to determine fluid control parameters that improve sterile filtration performance. [0207] In some embodiments, the controller may cause the filtration apparatus to filter the LNPs 10 according to one or more inputs to a program allowing for setting and control of filtration parameters. For example, a programmable pressure limit may be set to a desired static pressure. A control algorithm implemented by the program may utilize the pressure value for PCRITICAL (corresponding to Rmax) as follows. In some embodiments, pressure is controlled to reach PCRITICAL and to maintain PCRITICAL for a given duration before declining. In some embodiments, pressure is controlled to increase to within a set limit from PCRITICAL, e.g., 10% of PCRITICAL, and then lowered before PCRITICAL is reached. In some embodiments, multiple successive filtration cycles may be performed in which PCRITICAL or a set limit from PCRITICAL is reached once per cycle. In some embodiments, notifications are provided to an operator or user including whether a given pressure, such as PCRITICAL, is attained. In particular, the controller can be configured to allow selection of parameters for notification purposes (e.g., in the form of an alarm, such as an audio, visual or audiovisual notification), where the parameters include but not limited to a retentate volume, a total run time, a back pressure, low and high pressures, or a filtrate or permeate weight alarm, for example. Lipid Nanoparticles (LNPs) [0208] A lipid nanoparticle (LNP) refers to a nanoscale construct comprising lipid molecules and that comprises in a spherical (e.g., spheroid) geometry. In some embodiments, the LNP contains a bleb region, e.g., as described in Brader et al., Biophysical Journal 120: 1- 5 (2021), which is incorporated herein by reference in its entirety. In some embodiments, LNPs comprise one or more lipids and a nucleic acid cargo (i.e., polynucleotide) of interest. In some embodiments, the LNPs do not comprise a nucleic acid cargo. In some embodiments, the lipid is an ionizable lipid (e.g., an ionizable amino lipid), sometimes referred to in the art as an “ionizable cationic lipid”. In some embodiments, lipid nanoparticles further comprise other components, including one or more of a phospholipid, a structural lipid, and a molecule capable of reducing particle aggregation, for example a PEG or PEG-modified lipid. In some embodiments, the lipid nanoparticle comprises at least one ionizable cationic lipid, at least one non-cationic lipid, at least one sterol, and/or at least one polyethylene glycol (PEG)-modified lipid. The lipid nanoparticles can be generated using components, compositions, and methods as are generally known in the art, see for example PCT/US2016/052352; PCT/US2016/068300; PCT/US2017/037551; PCT/US2015/027400; PCT/US2016/047406; PCT/US2016000129; PCT/US2016/014280; PCT/US2016/014280; PCT/US2017/038426; PCT/US2014/027077; PCT/US2014/055394; PCT/US2016/52117; PCT/US2012/069610; PCT/US2017/027492; PCT/US2016/059575 and PCT/US2016/069491 all of which are incorporated by reference herein in their entireties. [0209] In some embodiments, the lipid nanoparticle is a lipid nanoparticle described in Intl. Pub. Nos. WO 2013/123523, WO 2012/170930, WO 2011/127255, WO 2008/103276; or U.S. Pub. No.2013/0171646, each of which is herein incorporated by reference in its entirety. [0210] In some embodiments, the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid, based on the lipid components, e.g., not including the nucleotide cargo. For example, the lipid nanoparticle may comprise a molar ratio of 20-50%, 20-40%, 20-30%, 30-60%, 30-50%, 30-40%, 40-60%, 40-50%, or 50-60% ionizable cationic lipid. In some embodiments, the lipid nanoparticle comprises a molar ratio of 20%, 30%, 40%, 50, or 60% ionizable cationic lipid. [0211] In some embodiments, the lipid nanoparticle comprises a molar ratio of 5-25% non-cationic lipid, based on the lipid components, e.g., not including the nucleotide cargo. For example, the lipid nanoparticle may comprise a molar ratio of 5-20%, 5-15%, 5-10%, 10- 25%, 10-20%, 10-25%, 15-25%, 15-20%, or 20-25% non-cationic lipid. In some embodiments, the lipid nanoparticle comprises a molar ratio of 5%, 10%, 15%, 20%, or 25% non-cationic lipid. [0212] In some embodiments, the lipid nanoparticle comprises a molar ratio of 25-55% sterol, based on the lipid components, e.g., not including the nucleotide cargo. For example, the lipid nanoparticle may comprise a molar ratio of 25-50%, 25-45%, 25-40%, 25-35%, 25- 30%, 30-55%, 30-50%, 30-45%, 30-40%, 30-35%, 35-55%, 35-50%, 35-45%, 35-40%, 40- 55%, 40-50%, 40-45%, 45-55%, 45-50%, or 50-55% sterol. In some embodiments, the lipid nanoparticle comprises a molar ratio of 25%, 30%, 35%, 40%, 45%, 50%, or 55% sterol. [0213] In some embodiments, the lipid nanoparticle comprises a molar ratio of 0.5-15% PEG-modified lipid, based on the lipid components, e.g., not including the nucleotide cargo. For example, the lipid nanoparticle may comprise a molar ratio of 0.5-10%, 0.5-5%, 1-15%, 1-10%, 1-5%, 2-15%, 2-10%, 2-5%, 5-15%, 5-10%, or 10-15%. In some embodiments, the lipid nanoparticle comprises a molar ratio of 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15% PEG-modified lipid. [0214] In some embodiments, the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid, 5-25% non-cationic lipid, 25-55% sterol, and 0.5-15% PEG-modified lipid, based on the lipid components, e.g., not including the nucleotide cargo. [0215] In some embodiments, the lipid nanoparticles described herein have a diameter from about 1 nm to about 100 nm such as, but not limited to, from about 1 nm to about 20 nm, from about 1 nm to about 30 nm, from about 1 nm to about 40 nm, from about 1 nm to about 50 nm, from about 1 nm to about 60 nm, from about 1 nm to about 70 nm, from about 1 nm to about 80 nm, from about 1 nm to about 90 nm, from about 5 nm to about from 100 nm, from about 5 nm to about 10 nm, about 5 nm to about 20 nm, from about 5 nm to about 30 nm, from about 5 nm to about 40 nm, from about 5 nm to about 50 nm, from about 5 nm to about 60 nm, from about 5 nm to about 70 nm, from about 5 nm to about 80 nm, from about 5 nm to about 90 nm, about 10 to about 20 nm, about 10 to about 30 nm, about 10 to about 40 nm, about 10 to about 50 nm, about 10 to about 60 nm, about 10 to about 70 nm, about 10 to about 80 nm, about 10 to about 90 nm, about 20 to about 30 nm, about 20 to about 40 nm, about 20 to about 50 nm, about 20 to about 60 nm, about 20 to about 70 nm, about 20 to about 80 nm, about 20 to about 90 nm, about 20 to about 100 nm, about 30 to about 40 nm, about 30 to about 50 nm, about 30 to about 60 nm, about 30 to about 70 nm, about 30 to about 80 nm, about 30 to about 90 nm, about 30 to about 100 nm, about 40 to about 50 nm, about 40 to about 60 nm, about 40 to about 70 nm, about 40 to about 80 nm, about 40 to about 90 nm, about 40 to about 100 nm, about 50 to about 60 nm, about 50 to about 70 nm about 50 to about 80 nm, about 50 to about 90 nm, about 50 to about 100 nm, about 60 to about 70 nm, about 60 to about 80 nm, about 60 to about 90 nm, about 60 to about 100 nm, about 70 to about 80 nm, about 70 to about 90 nm, about 70 to about 100 nm, about 80 to about 90 nm, about 80 to about 100 nm and/or about 90 to about 100 nm. [0216] In some embodiments, the lipid nanoparticles described herein have a diameter from about 10 to 500 nm. In some embodiments, the lipid nanoparticle can have a diameter greater than 100 nm, greater than 150 nm, greater than 200 nm, greater than 250 nm, greater than 300 nm, greater than 350 nm, greater than 400 nm, greater than 450 nm, greater than 500 nm, greater than 550 nm, greater than 600 nm, greater than 650 nm, greater than 700 nm, greater than 750 nm, greater than 800 nm, greater than 850 nm, greater than 900 nm, greater than 950 nm or greater than 1000 nm. [0217] The ratio between the lipid components of the LNP composition and the polynucleotide cargo can be from about 10:1 to about 60:1 (wt/wt). In some embodiments, the ratio between the lipid components and the polynucleotide (e.g., mRNA) can be about 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 21:1, 22:1, 23:1, 24:1, 25:1, 26:1, 27:1, 28:1, 29:1, 30:1, 31:1, 32:1, 33:1, 34:1, 35:1, 36:1, 37:1, 38:1, 39:1, 40:1, 41:1, 42:1, 43:1, 44:1, 45:1, 46:1, 47:1, 48:1, 49:1, 50:1, 51:1, 52:1, 53:1, 54:1, 55:1, 56:1, 57:1, 58:1, 59:1 or 60:1 (wt/wt). In some embodiments, the wt/wt ratio of the lipid components of the LNP composition to the polynucleotide (such as a polynucleotide encoding a therapeutic agent) is about 20:1 or about 15:1. [0218] In some embodiments, the lipid nanoparticles described herein can comprise polynucleotides (e.g., mRNA) in a lipid:polynucleotide weight ratio of 5:1, 10:1, 15:1, 20:1, 25:1, 30:1, 35:1, 40:1, 45:1, 50:1, 55:1, 60:1 or 70:1, or a range or any of these ratios such as, but not limited to, 5:1 to about 10:1, from about 5:1 to about 15:1, from about 5:1 to about 20:1, from about 5:1 to about 25:1, from about 5:1 to about 30:1, from about 5:1 to about 35:1, from about 5:1 to about 40:1, from about 5:1 to about 45:1, from about 5:1 to about 50:1, from about 5:1 to about 55:1, from about 5:1 to about 60:1, from about 5:1 to about 70:1, from about 10:1 to about 15:1, from about 10:1 to about 20:1, from about 10:1 to about 25:1, from about 10:1 to about 30:1, from about 10:1 to about 35:1, from about 10:1 to about 40:1, from about 10:1 to about 45:1, from about 10:1 to about 50:1, from about 10:1 to about 55:1, from about 10:1 to about 60:1, from about 10:1 to about 70:1, from about 15:1 to about 20:1, from about 15:1 to about 25:1, from about 15:1 to about 30:1, from about 15:1 to about 35:1, from about 15:1 to about 40:1, from about 15:1 to about 45:1, from about 15:1 to about 50:1, from about 15:1 to about 55:1, from about 15:1 to about 60:1 or from about 15:1 to about 70:1. [0219] In some embodiments, the lipid nanoparticles described herein can comprise the polynucleotide in a concentration from approximately 0.1 mg/ml to 2 mg/ml such as, but not limited to, 0.1 mg/ml, 0.2 mg/ml, 0.3 mg/ml, 0.4 mg/ml, 0.5 mg/ml, 0.6 mg/ml, 0.7 mg/ml, 0.8 mg/ml, 0.9 mg/ml, 1.0 mg/ml, 1.1 mg/ml, 1.2 mg/ml, 1.3 mg/ml, 1.4 mg/ml, 1.5 mg/ml, 1.6 mg/ml, 1.7 mg/ml, 1.8 mg/ml, 1.9 mg/ml, 2.0 mg/ml or greater than 2.0 mg/ml. [0220] In some embodiments, the composition or pharmaceutical composition disclosed herein can contain more than one polynucleotide. For example, a composition or pharmaceutical composition disclosed herein can contain two or more polynucleotides (e.g., RNA, e.g., mRNA) formulated in the same lipid nanoparticle. A single LNP can contain two or more polynucleotides. Additionally or alternatively, a composition or pharmaceutical composition disclosed herein can comprise a mixture of LNPs, each containing one or more polynucleotides, which may be the same or different. For example LNP1 can contain polynucleotide A; LNP2 can contain polynucleotides B and C; LNP3 can contain polynucleotides D, E, and F; LNP4 can contain polynucleotides E, F, and G, etc. [0221] The lipid nanoparticles described herein can be geometrically engineered to modulate macrophage and/or the immune response. The geometrically engineered particles can have varied shapes, sizes and/or surface charges to incorporate the polynucleotides described herein for targeted delivery such as, but not limited to, pulmonary delivery (see, e.g., Intl. Pub. No. WO 2013/082111, herein incorporated by reference in its entirety). Other physical features the geometrically engineering particles can include are, but are not limited to, fenestrations, angled arms, asymmetry and surface roughness, and charge that can alter the interactions with cells and tissues. [0222] In some embodiments, the lipid nanoparticles described herein are stealth nanoparticles or target-specific stealth nanoparticles such as, but not limited to, those described in U.S. Pub. No.2013/0172406, herein incorporated by reference in its entirety. The stealth or target-specific stealth nanoparticles can comprise a polymeric matrix, which can comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polyesters, polyanhydrides, polyethers, polyurethanes, polymethacrylates, polyacrylates, polycyanoacrylates, or combinations thereof. [0223] The LNPs can be prepared using microfluidic mixers or micromixers. Exemplary microfluidic mixers can include, but are not limited to, a slit interdigital micromixer including, but not limited to those manufactured by Microinnova (Allerheiligen bei Wildon, Austria) and/or a staggered herringbone micromixer (SHM) (see Zhigaltsevet al., “Bottom-up design and synthesis of limit size lipid nanoparticle systems with aqueous and triglyceride cores using millisecond microfluidic mixing,” Langmuir 28:3633-40 (2012); Belliveau et al., “Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA,” Molecular Therapy-Nucleic Acids.1:e37 (2012); Chen et al., “Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation,” J. Am. Chem. Soc.134(16):6948-51 (2012)). Exemplary micromixers include Slit Interdigital Microstructured Mixer (SIMM-V2) or a Standard Slit Interdigital Micro Mixer (SSIMM) or Caterpillar (CPMM) or Impinging-jet (IJMM,) from the Institut für Mikrotechnik Mainz GmbH, Mainz Germany. In some embodiments, methods of making LNP using SHM further comprise mixing at least two input streams wherein mixing occurs by microstructure-induced chaotic advection (MICA). According to this method, fluid streams flow through channels present in a herringbone pattern causing rotational flow and folding the fluids around each other. This method can also comprise a surface for fluid mixing wherein the surface changes orientations during fluid cycling. Methods of generating LNPs using SHM include those disclosed in U.S. Pub. Nos.2004/0262223 and 2012/0276209, each of which is incorporated herein by reference in their entirety. [0224] In some embodiments, the polynucleotides described herein can be formulated in lipid nanoparticles using microfluidic technology (see Whitesides, George M., “The Origins and the Future of Microfluidics,” Nature 442: 368-373 (2006); and Abraham et al., “Chaotic Mixer for Microchannels,” Science 295: 647-651 (2002)). In some embodiments, the polynucleotides can be formulated in lipid nanoparticles using a micromixer chip such as, but not limited to, those from Harvard Apparatus (Holliston, MA) or Dolomite Microfluidics (Royston, UK). A micromixer chip can be used for rapid mixing of two or more fluid streams with a split and recombine mechanism. • i. Ionizable Lipids [0225] The lipid nanoparticles described herein comprise ionizable lipids. As used herein, the term “ionizable lipid” has its ordinary meaning in the art and may refer to a lipid comprising one or more charged moieties. In some embodiments, an ionizable lipid may be positively charged or negatively charged. An ionizable lipid may be positively charged, in which case it can be referred to as “cationic lipid”. In certain embodiments, an ionizable lipid molecule may comprise an amine group, and can be referred to as an ionizable amino lipid. As used herein, a “charged moiety” is a chemical moiety that carries a formal electronic charge, e.g., monovalent (+1, or -1), divalent (+2, or -2), trivalent (+3, or -3), etc. The charged moiety may be anionic (i.e., negatively charged) or cationic (i.e., positively charged). Examples of positively-charged moieties include amine groups (e.g., primary, secondary, and/or tertiary amines), ammonium groups, pyridinium group, guanidine groups, and imidizolium groups. In some embodiments, the charged moieties comprise amine groups. Examples of negatively charged groups or precursors thereof include carboxylate groups, sulfonate groups, sulfate groups, phosphonate groups, phosphate groups, hydroxyl groups, and the like. The charge of the charged moiety may vary, in some cases, with the environmental conditions, for example, changes in pH may alter the charge of the moiety, and/or cause the moiety to become charged or uncharged. In general, the charge density of the molecule may be selected as desired. [0226] It should be understood that the terms “charged” or “charged moiety” does not refer to a “partial negative charge” or “partial positive charge” on a molecule. The terms “partial negative charge” and “partial positive charge” are given its ordinary meaning in the art. A “partial negative charge” may result when a functional group comprises a bond that becomes polarized such that electron density is pulled toward one atom of the bond, creating a partial negative charge on the atom. Those of ordinary skill in the art will, in general, recognize bonds that can become polarized in this way. [0227] In some embodiments, the ionizable lipid is an ionizable amino lipid, sometimes referred to in the art as an “ionizable cationic lipid”. In some embodiments, the ionizable amino lipid may have a positively charged hydrophilic head and a hydrophobic tail that are connected via a linker structure. [0228] In addition to these, an ionizable lipid may also be a lipid including a cyclic amine group. [0229] In some embodiments, the ionizable lipid may be selected from, but not limited to, an ionizable lipid described in International Publication Nos. WO 2013/086354 and WO 2013/116126; each of which is herein incorporated by reference in its entirety. [0230] In some embodiments, the ionizable lipid may be selected from, but not limited to, formula CLI-CLXXXXII of US Patent No.7,404,969; which is herein incorporated by reference in its entirety. [0231] In some embodiments, the lipid may be a cleavable lipid such as those described in International Publication No. WO 2012/170889, herein incorporated by reference in its entirety. In some embodiments, the lipid may be synthesized by methods known in the art and/or as described in International Publication No. WO 2013/086354; each of which is herein incorporated by reference in its entirety. [0232] In some aspects, the lipid comprises at least one tertiary amino group, wherein at least one of the three groups of the tertiary amino group comprises a C6-30 saturated or unsaturated carbon chain optionally interrupted by an –C(O)O– ester group. [0233] In some aspects, the disclosure relates to a compound of Formula (I): or its N-oxide, or a salt or isomer thereof,
Figure imgf000066_0001
wherein R’a is R’branched; wherein R’branched is: wherein denotes a point of attachment;
Figure imgf000066_0002
Figure imgf000066_0003
wherein R, R, R, and R are each independently selected from the group consisting of H, C2-12 alkyl, and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH, wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and
Figure imgf000066_0004
wherein denotes a point of attachment; wherein
Figure imgf000066_0005
R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R5 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; M and M’ are each independently selected from the group consisting of -C(O)O- and -OC(O)-; R’ is a C1-12 alkyl or C2-12 alkenyl; l is selected from the group consisting of 1, 2, 3, 4, and 5; and m is selected from the group consisting of 5, 6, 7, 8, 9, 10, 11, 12, and 13. [0234] In some embodiments of the compounds of Formula (I), R’a is R’branched; R’branched is ; aα aβ
Figure imgf000067_0001
denotes a point of attachment; R , R , R, and R are each H; R2 and R3 are each C1-14 alkyl; R4 is -(CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0235] In some embodiments of the compounds of Formula (I), R’a is R’branched; R’branched is denote aα aβ
Figure imgf000067_0002
s a point of attachment; R , R , R, and R are each H; R2 and R3 are each C1-14 alkyl; R4 is -(CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 3; and m is 7. [0236] In some embodiments of the compounds of Formula (I), R’a is R’branched; R’branched is denotes a point of attachment; R is C2-12 alkyl; R, R
Figure imgf000067_0003
, and R are each H; R2 and R3 are each C1-14 alkyl; R4 is
Figure imgf000067_0004
R10 NH(C1-6 alkyl); n2 is 2; R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0237] In some embodiments of the compounds of Formula (I), R’a is R’branched; R’branched is denotes a poi aα aβ
Figure imgf000068_0003
nt of attachment; R , R , and R are each H; R is C2-12 alkyl; R2 and R3 are each C1-14 alkyl; R4 is - (CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0238] In some embodiments, the compound of Formula (I) is selected from:
Figure imgf000068_0001
[0239] In some embodiments, the compound of Formula (I) is:
Figure imgf000068_0002
[0240] In some embodiments, the compound of Formula (I) is:
Figure imgf000069_0001
. [0241] In some embodiments, the compound of Formula (I) is:
Figure imgf000069_0002
. [0242] In some embodiments, the compound of Formula (I) is:
Figure imgf000069_0003
. [0243] In some aspects, the disclosure relates to a compound of Formula (Ia):
Figure imgf000069_0004
(Ia) or its N-oxide, or a salt or isomer thereof, wherein R’a is R’branched; wherein R’branched is: wherein denotes a point of attachment;
Figure imgf000069_0006
Figure imgf000069_0005
wherein R, R, and R are each independently selected from the group consisting of H, C2- 12 alkyl, and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and
Figure imgf000070_0001
wherein denotes a point of attachment; wherein
Figure imgf000070_0002
R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R5 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; M and M’ are each independently selected from the group consisting of -C(O)O- and -OC(O)-; R’ is a C1-12 alkyl or C2-12 alkenyl; l is selected from the group consisting of 1, 2, 3, 4, and 5; and m is selected from the group consisting of 5, 6, 7, 8, 9, 10, 11, 12, and 13. [0244] In some aspects, the disclosure relates to a compound of Formula (Ib): or its N-oxide, or a salt or isomer thereof,
Figure imgf000070_0003
wherein R’a is R’branched; wherein R’branched is: wherein denotes a point of attachment;
Figure imgf000071_0001
Figure imgf000071_0002
wherein R, R, R, and R are each independently selected from the group consisting of H, C2-12 alkyl, and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is -(CH2)nOH, wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; each R5 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; M and M’ are each independently selected from the group consisting of -C(O)O- and -OC(O)-; R’ is a C1-12 alkyl or C2-12 alkenyl; l is selected from the group consisting of 1, 2, 3, 4, and 5; and m is selected from the group consisting of 5, 6, 7, 8, 9, 10, 11, 12, and 13. [0245] In some embodiments of Formula (I) or (Ib), R’a is R’branched; R’branched is denotes a point of attachment; R, R, and R are each
Figure imgf000071_0003
H; R2 and R3 are each C1-14 alkyl; R4 is -(CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0246] In some embodiments of Formula (I) or (Ib), R’a is R’branched; R’branched is denotes a point of attachment; R, R,
Figure imgf000072_0001
and R are each H; R2 and R3 are each C1-14 alkyl; R4 is -(CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 3; and m is 7. [0247] In some embodiments of Formula (I) or (Ib), R’a is R’branched; R’branched is denotes a point of attachment; R and R are each H; R
Figure imgf000072_0002
is C2-12 alkyl; R2 and R3 are each C1-14 alkyl; R4 is -(CH2)nOH; n is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0248] In some aspects, the disclosure relates to a compound of Formula (Ic): or its N-oxide, or a salt or isomer thereof,
Figure imgf000072_0003
wherein R’a is R’branched; wherein R’branched is: ; wherein denotes a point of attachment;
Figure imgf000072_0004
Figure imgf000072_0005
wherein R, R, R, and R are each independently selected from the group consisting of H, C2-12 alkyl, and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is
Figure imgf000073_0001
wherein denotes a point of attachment; wherein
Figure imgf000073_0002
R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R5 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; each R6 is independently selected from the group consisting of C1-3 alkyl, C2-3 alkenyl, and H; M and M’ are each independently selected from the group consisting of -C(O)O- and -OC(O)-; R’ is a C1-12 alkyl or C2-12 alkenyl; l is selected from the group consisting of 1, 2, 3, 4, and 5; and m is selected from the group consisting of 5, 6, 7, 8, 9, 10, 11, 12, and 13. [0249] In some embodiments, R’a is R’branched; R’branched is denotes a
Figure imgf000073_0003
point of attachment; R, R, and R are each H; R is C2-12 alkyl; R2 and R3 are each C1-14 alkyl; R4 is denotes a point of attachment; R10 is NH(C1-6 alkyl);
Figure imgf000073_0004
n2 is 2; each R5 is H; each R6 is H; M and M’ are each -C(O)O-; R’ is a C1-12 alkyl; l is 5; and m is 7. [0250] In some embodiments, the compound of Formula (Ic) is:
Figure imgf000074_0001
[0251] In some aspects, the disclosure relates to a compound of Formula (II): or its N-oxide, or a salt or isomer thereof,
Figure imgf000074_0002
wherein R’a is R’branched or R’cyclic; wherein R’branched is: and R’cyclic is: ; and
Figure imgf000074_0003
wherein
Figure imgf000074_0004
denotes a point of attachment; R and R are each independently selected from the group consisting of H, C1-12 alkyl, and C2-12 alkenyl, wherein at least one of R and R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R and R are each independently selected from the group consisting of H, C1-12 alkyl, and C2-12 alkenyl, wherein at least one of R and R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and
Figure imgf000075_0001
wherein
Figure imgf000075_0002
denotes a point of attachment; wherein R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1-12 alkyl or C2-12 alkenyl; Ya is a C3-6 carbocycle; R*”a is selected from the group consisting of C1-15 alkyl and C2-15 alkenyl; and s is 2 or 3; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0252] In some aspects, the disclosure relates to a compound of Formula (II-a): (II-a) or its N-oxide, or a salt or isomer thereof,
Figure imgf000075_0003
wherein R’a is R’branched or R’cyclic; wherein R’branched is: and R’b is:
Figure imgf000076_0001
Figure imgf000076_0002
wherein denotes a point of attachment;
Figure imgf000076_0003
R and R are each independently selected from the group consisting of H, C1-12 alkyl, and C2-12 alkenyl, wherein at least one of R and R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R and R are each independently selected from the group consisting of H, C1-12 alkyl, and C2-12 alkenyl, wherein at least one of R and R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5, and
Figure imgf000076_0004
wherein
Figure imgf000076_0005
denotes a point of attachment; wherein R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0253] In some aspects, the disclosure relates to a compound of Formula (II-b):
Figure imgf000077_0001
r its N-oxide, or a salt or isomer thereof, wherein R’a is R’branched or R’cyclic; wherein
Figure imgf000077_0002
wherein denotes a point of attachment; R and R are each independently selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting
Figure imgf000077_0003
wherein denotes a point of attachment; wherein R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0254] In some aspects, the disclosure relates to a compound of Formula (II-c):
Figure imgf000078_0001
r its N-oxide, or a salt or isomer thereof, wherein R’a is R’branched or R’cyclic; wherein
Figure imgf000078_0002
wherein denotes a point of attachment; wherein R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting
Figure imgf000078_0003
wherein denotes a point of attachment; wherein
Figure imgf000078_0004
R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; R’ is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0255] In some aspects, the disclosure relates to a compound of Formula (II-d):
Figure imgf000079_0001
r its N-oxide, or a salt or isomer thereof, wherein R’a is R’branched or R’cyclic; wherein
Figure imgf000079_0002
wherein denotes a point of attachment; wherein R and R are each independently selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group consisting
Figure imgf000079_0003
wherein denotes a point of attachment; wherein R10 is N(R)2; each R is independently selected from the group consisting of C1-6 alkyl, C2-3 alkenyl, and H; and n2 is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; each R’ independently is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0256] In some aspects, the disclosure relates to a compound of Formula (II-e):
Figure imgf000080_0001
wherein R’a is R’branched or R’cyclic; wherein
Figure imgf000080_0002
wherein denotes a point of attachment; wherein R is selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; R2 and R3 are each independently selected from the group consisting of C1-14 alkyl and C2-14 alkenyl; R4 is -(CH2)nOH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; R’ is a C1-12 alkyl or C2-12 alkenyl; m is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9; l is selected from 1, 2, 3, 4, 5, 6, 7, 8, and 9. [0257] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), m and l are each independently selected from 4, 5, and 6. In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), m and l are each 5. [0258] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), each R’ independently is a C1-12 alkyl. In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), each R’ independently is a C2-5 alkyl. [0259] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’b is: and R2 and R3 are each independently a C1-14 alkyl. [0260] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’b is: and R2 and R3 are each independently a C6-10 alkyl. [0261] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’b is:
Figure imgf000081_0001
are each a C8 alkyl. [0262] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000081_0002
R3 are each independently a C6-10 alkyl. [0263] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000081_0003
R3 are each independently a C6-10 alkyl. [0264] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000081_0004
is: , R is a C2-6 alkyl, and R2 and R3 are each a C8 alkyl. [0265] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000081_0006
is:
Figure imgf000081_0005
R are each a C1-12 alkyl. [0266] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000082_0002
is:
Figure imgf000082_0001
are each a C2-6 alkyl. [0267] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), m and l are each independently selected from 4, 5, and 6 and each R’ independently is a C1-12 alkyl. [0268] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), m and l are each 5 and each R’ independently is a C2-5 alkyl. [0269] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000082_0003
independently selected from 4, 5, and 6, each R’ independently is a C1-12 alkyl, and R and R are each a C1-12 alkyl. [0270] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000082_0005
is:
Figure imgf000082_0004
are each 5, each R’ independently is a C2-5 alkyl, and R and R are each a C2-6 alkyl. [0271] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000082_0006
, m and l are each independently selected from 4, 5, and 6, R’ is a C1-12 alkyl, R is a C1-12 alkyl and R2 and R3 are each independently a C6-10 alkyl. [0272] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000083_0001
is:
Figure imgf000083_0002
are each 5, R’ is a C2-5 alkyl, R is a C2-6 alkyl, and R2 and R3 are each a C8 alkyl. [0273] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d),
Figure imgf000083_0009
[0275] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000083_0004
is:
Figure imgf000083_0003
are each independently selected from 4, 5, and 6, each R’ independently is a C1-12 alkyl, R and R are each a C1-12 alkyl, 1
Figure imgf000083_0005
wherein R 0 is NH(C1-6 alkyl), and n2 is 2. [0276] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000083_0007
is:
Figure imgf000083_0006
are each 5, each R’ independently is a C2-5 alkyl, R and R are each a C2-6 alkyl, and R4 is
Figure imgf000083_0008
, wherein R10 is NH(CH3) and n2 is 2. [0277] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000084_0002
and R’b is:
Figure imgf000084_0001
m and l are each independently selected from 4, 5, and 6, R’ is a C1-12 alkyl, R2 and R3 are each independently a C6-10 alkyl, R is a C1-12 alkyl,
Figure imgf000084_0003
wherein R10 is NH(C1-6 alkyl) and n2 is 2. [0278] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000084_0004
alkyl, R is a C2-6 alkyl, R2 and R3 are each a C8 alkyl,
Figure imgf000084_0005
wherein R10 is NH(CH3) and n2 is 2. [0279] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R4 is -(CH2)nOH and n is 2, 3, or 4. In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R4 is -(CH2)nOH and n is 2. [0280] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000084_0007
is:
Figure imgf000084_0006
are each independently selected from 4, 5, and 6, each R’ independently is a C1-12 alkyl, R and R are each a C1-12 alkyl, R4 is -(CH2)nOH, and n is 2, 3, or 4. [0281] In some embodiments of the compound of Formula (II), (II-a), (II-b), (II-c), (II-d), or (II-e), R’branched is:
Figure imgf000084_0008
each 5, each R’ independently is a C2-5 alkyl, R and R are each a C2-6 alkyl, R4 is -(CH2)nOH, and n is 2. [0282] In some aspects, the disclosure relates to a compound of Formula (II-f):
Figure imgf000085_0001
r its N-oxide, or a salt or isomer thereof, wherein R’a is R’branched or R’cyclic; wherein R’branched is:
Figure imgf000085_0003
and R’b is:
Figure imgf000085_0002
wherein
Figure imgf000085_0004
denotes a point of attachment; R is a C1-12 alkyl; R2 and R3 are each independently a C1-14 alkyl; R4 is - (CH2)nOH wherein n is selected from the group consisting of 1, 2, 3, 4, and 5; R’ is a C1-12 alkyl; m is selected from 4, 5, and 6; and l is selected from 4, 5, and 6. [0283] In some embodiments of the compound of Formula (II-f), m and l are each 5, and n is 2, 3, or 4. [0284] In some embodiments of the compound of Formula (II-f) R’ is a C2-5 alkyl, R is a C2-6 alkyl, and R2 and R3 are each a C6-10 alkyl. [0285] In some embodiments of the compound of Formula (II-f), m and l are each 5, n is 2, 3, or 4, R’ is a C2-5 alkyl, R is a C2-6 alkyl, and R2 and R3 are each a C6-10 alkyl. [0286] In some aspects, the disclosure relates to a compound of Formula (II-g):
Figure imgf000085_0005
wherein R is a C2-6 alkyl; R’ is a C2-5 alkyl; and R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group
Figure imgf000086_0001
wherein denotes a point of attachment, R10 is NH(C1-6 alkyl), and n2 is selected from the group consisting of 1, 2, and 3. [0287] In some aspects, the disclosure relates to a compound of Formula (II-h):
Figure imgf000086_0002
wherein R and R are each independently a C2-6 alkyl; each R’ independently is a C2-5 alkyl; and R4 is selected from the group consisting of -(CH2)nOH wherein n is selected from the group
Figure imgf000086_0003
wherein denotes a point of attachment, R10 is NH(C1-6 alkyl), and n2 is selected from the group consisting of 1, 2, and 3. [0288] In some embodiments of the compound of Formula (II-g) or (II-h), R4 is
Figure imgf000086_0004
, wherein R10 is NH(CH3) and n2 is 2. [0289] In some embodiments of the compound of Formula (II-g) or (II-h), R4 is - (CH2)2OH. [0290] In some aspects, the disclosure relates to a compound having the Formula (III):
Figure imgf000087_0001
or a salt or isomer thereof, wherein R1, R2, R3, R4, and R5 are independently selected from the group consisting of C5-20 alkyl, C5- 20 alkenyl, -R”MR’, -R*YR”, -YR”, and -R*OR”; each M is independently selected from the group consisting of -C(O)O-, -OC(O)-, -OC(O)O-, -C(O)N(R’)-, -N(R’)C(O)-, -C(O)-, -C(S)-, -C(S)S-, -SC(S) -, -CH(OH)-, -P(O)(OR’)O-, -S(O)2-, an aryl group, and a heteroaryl group; X1, X2, and X3 are independently selected from the group consisting of a bond, -CH2-, -(CH2)2-, -CHR-, -CHY-, -C(O)-, -C(O)O-, -OC(O)-, -C(O)-CH2-, -CH2-C(O)-, -C(O)O-CH2-, -OC(O)-CH2-, -CH2-C(O)O-, -CH2-OC(O)-, -CH(OH)-, -C(S)-, and -CH(SH)-; each Y is independently a C3-6 carbocycle; each R* is independently selected from the group consisting of C1-12 alkyl and C2-12 alkenyl; each R is independently selected from the group consisting of C1-3 alkyl and a C3-6 carbocycle; each R’ is independently selected from the group consisting of C1-12 alkyl, C2-12 alkenyl, and H; and each R” is independently selected from the group consisting of C3-12 alkyl and C3-12 alkenyl, and wherein: i) at least one of X1, X2, and X3 is not -CH2-; and/or ii) at least one of R1, R2, R3, R4, and R5 is -R”MR’. [0291] In some embodiments, R1, R2, R3, R4, and R5 are each C5-20 alkyl; X1 is -CH2-; and X2 and X3 are each -C(O)-. [0292] In some embodiments, the compound of Formula (III) is:
Figure imgf000088_0001
. [0293] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2020/146805; WO 2020/081938; WO 2020/214946; WO 2019/036030; WO 2019/036000; WO 2019/036028; WO 2019/036008; WO 2018/200943; WO 2018/191657; WO 2017/117528; WO 2017/075531; WO 2017/004143; WO 2015/199952; and WO 2015/074085; each of which is incorporated herein in its entirety. [0294] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2020/146805 having structure:
Figure imgf000088_0002
or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R1 is optionally substituted C1-C24 alkyl or optionally substituted C2-C24 alkenyl; R2 and R3 are each independently optionally substituted C1-C36 alkyl; R4 and R5 are each independently optionally substituted C1-C6 alkyl, or R4 and R5 join, along with the N to which they are attached, to form a heterocyclyl or heteroaryl; L1, L2, and L3 are each independently optionally substituted C1-C18 alkylene; G1 is a direct bond, -(CH2)nO(C=O)-, -(CH2)n(C=O)O-, or -(C=)-; G2 and G3 are each independently -(C=O)O- or -O(C=O)-; and n is an integer greater than 0. [0295] In some embodiments, the ionizable lipids are one or more of the compounds described in WO/2020/081938 having structure:
Figure imgf000089_0001
or a pharmaceutically acceptable salt, tautomer or stereoisomer thereof, wherein: G1 is - N(R3)R4 or -OR5; R1 is optionally substituted branched, saturated or unsaturated C12-C36 alkyl; R2 is optionally substituted branched or unbranched, saturated or unsaturated C12-C36 alkyl when L is -C(=0)-; or R2 is optionally substituted branched or unbranched, saturated or unsaturated C4-C36 alkyl when L is C6-C12 alkylene, C6-C12 alkenylene, or C2-C6 alkynylene; R3 and R4 are each independently H, optionally substituted branched or unbranched, saturated or unsaturated C1-C6 alkyl; or R3and R4 are each independently optionally substituted branched or unbranched, saturated or unsaturated C1-C6 alkyl when L is C6-C12 alkylene, C6- C12 alkenylene, or C2-C6 alkynylene; or R3 and R4, together with the nitrogen to which they are attached, join to form a heterocyclyl; R5 is H or optionally substituted C1-C6 alkyl; L is - C(=O)-, C6-C12 alkylene, C6-C12 alkenylene, or C2-C12alkynylene (e.g., C2-C6 alkynylene); and n is an integer from 1 to 12. [0296] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2020/214946 having structure:
Figure imgf000090_0001
or a pharmaceutically acceptable salt thereof, wherein each Rla is independently hydrogen, R1c, or R3d; each Rlb is independently Rlc or Rld; each Rlc is Independently –(CH)2C(O)X1R3; each Rld is independently -C(O)R4; each R2 is independently -[C(R2a)2]c;R2b; each R2a is independently hydrogen or lower alkyl (e.g., C1-C6alkyl); R2b is -N(LI-B)2, -(OCH2CH2)6OH; or -(OCH2CH2)6OCH3; each R3and R4 is independently aliphatic (e.g., C6-C30 aliphatic); each L1 is independently alkylene (e.g., C1-C10 alkylene); each B is independently hydrogen or an ionizable nitrogen-containing group; each X1 is independently a covalent bond or O; each a is independently an integer (e.g., 1-10); each b is independently an integer (e.g., 1-10); and each c is independently an integer (e.g., 1-10). [0297] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2019/036030 having the structure:
Figure imgf000091_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: X is N, and Y is absent; or X is CR, and Y is NR; L1 is -O(C=O)R1, -(C=O)OR1, -C(=O)R1, -OR1, S(O)xR1, -S-SR1, -C(=O)SR1, -SC(=O)R1, -NRaC(=O)R1, -C(=O)NRbRc, -NRaC(=O)NRbRc, -OC(=O)NRbRc or -NRaC(=O)OR1; L2 is -O(C=O)R2, -(C=O)OR2, -C(=O)R2, -OR2, -S(O)xR2, -S-SR2, -C(=O)SR2, -SC(=O)R2, -NRdC(=O)R2, -C(=O)NReRf, -NRdC(=O)NReRf, -OC(=O)NReRf; -NRdC(=O)OR2 or a direct bond to R2; L3 is -O(C=O)R3 or -(C=O)OR3; G1 and G2 are each independently C2-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C1-C24 heteroalkylene or C2-C24 heteroalkenylene when X is CR, and Y is NR; and G3 is C1-C24 heteroalkylene or C2-C24 heteroalkenylene when X is N, and Y is absent; Ra, Rb, Rd and Re are each independently H or C1-C12 alkyl or C1-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; each R is independently H or C1-C12 alkyl; R1, R2 and R3 are each independently C1-C24 alkyl or C2-C24 alkenyl; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, heteroalkylene and heteroalkenylene is independently substituted or unsubstituted unless otherwise specified. [0298] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2019/036000 having the structure:
Figure imgf000092_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, - C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, -NRaC(=O)NRa-, -OC(=O)NRa-, -NRaC(=O)O- or a direct bond; G1 is C1-C2 alkylene, -(C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is -C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa- or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon- carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon- carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon- carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon- carbon double bond; R5 and R6 are each independently H or methyl; R7 is H or C1-C20 alkyl; R8 is OH, -N(R9)(C=O)R10, -(C=O)NR9R10, -NR9R10, -(C=O)OR11 or -O(C=O)R11, provided that G3 is C4-C6 alkylene when R8 is -NR9R10, R9 and R10 are each independently H or C1-C12 alkyl; R11 is aralkyl; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2, wherein each alkyl, alkylene and aralkyl is optionally substituted. [0299] In some embodiments, the ionizable lipids are one or more of the compounds described in WO/2019/036028 having the structure:
Figure imgf000093_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: X and X’ are each independently N or CR; Y and Y’ are each independently absent, -O(C=O)-, -(C=O)O- or NR, provided that: a) Y is absent when X is N; b) Y’ is absent when X’ is N; c) Y is -O(C=O)-, -(C=O)O- or NR when X is CR; and d) Y’ is -O(C=O)-, -(C=O)O- or NR when X’ is CR, L1 and L1’ are each independently -O(C=O)R1, -(C=O)OR1 , -C(=O)R1, -OR1, -S(O)zR1, -S- SR1, -C(=O)SR1, -SC(=O)R1, -NRaC(=O)R1, -C(=O)NRbRc, -NRaC(=O)NRbRc, - OC(=O)NRbRc or -NRaC(=O)OR1; L2 and L2 are each independently -O(C=O)R2, -(C=O)OR2, -C(=O)R2, -OR2, -S(O)zR2, -S- SR2, -C(=O)SR2, -SC(=O)R2, -NRdC(=O)R2, -C(=O)NReRf, -NRdC(=O)NReRf, -OC(=O)NReRf; -NRdC(=O)OR2 or a direct bond to R2; G1, G1’, G2 and G2’ are each independently C2-C12 alkylene or C2-C12 alkenylene; G3 is C2-C24 heteroalkylene or C2-C24 heteroalkenylene; Ra, Rb, Rd and Re are, at each occurrence, independently H, C1-C12 alkyl or C2-C12 alkenyl; Rc and Rf are, at each occurrence, independently C1-C12 alkyl or C2-C12 alkenyl; R is, at each occurrence, independently H or C1-C12 alkyl; R1 and R2 are, at each occurrence, independently branched C6-C24 alkyl or branched C6-C24 alkenyl; z is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, heteroalkylene and heteroalkenylene is independently substituted or unsubstituted unless otherwise specified. [0300] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2019/036008 have the structure:
Figure imgf000095_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L1 is -O(C=O)R1, -(C=O)OR1, -C(=O)R1, -OR1, -S(O)xR1, -S-SR1, - C(=O)SR1, -SC (=O)R1, -NRaC(=O)R1, -C(=O)NRbRc , -NRaC(=O)NRbRc, -OC(=O)NRbRc or -NRaC(=O)OR1; L2 is -O(C=O)R2, -(C=O)OR2, -C(=O)R2, -OR2, -S(O)xR2, -S-SR2, - C(=O)SR2, -SC (=O)R2, -NRdC(=O)R2, -C(=O)NReRf , -NRdC(=O)NReRf, -OC(=O)NReRf or –NRdC(=O)OR2 or a direct bond to R2; G1 and G2 are each independently C2-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C3-C8 cycloalkylene or C3-C8 cycloalkenylene; Ra, Rb, Rd and Re are each independently H or C1-C12 alkyl or C1-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; R1 and R2 are each independently branched C6-C24 alkyl or branched C6-C24 alkenyl; R3 is -N(R4)R5; R4 is C1-C12 alkyl; R5 is substituted C1-C12 alkyl; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, cycloalkylene, cycloalkenylene, aryl and aralkyl is independently substituted or unsubstituted unless otherwise specified. [0301] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2018/200943 having the structure:
Figure imgf000096_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: L1 is -O(C=O)R1, -(C=O)OR1, -C(=O)R1, -OR1, -S(O)xR1, -S-SR1, - C(=O)SR1, -SC(=O)R1, -NRaC(=O)R1, -C(=O)NRbRc , -NRaC(=O)NRbRc, -OC(=O)NRbRc or -NRaC(=O)OR1; L2 is -O(C=O)R2, -(C=O)OR2, -C(=O)R2, -OR2, -S(O)xR2, -S-SR2, -C(=O)SR2, -SC (=O)R2, -NRdC(=O)R2, -C(=O)NReRf , -NRdC(=O)NReRf, -OC(=O)NReRf; -NRdC(=O)OR2 or a direct bond to R2; G1a and G2a are each independently C2-C12 alkylene or C2-C12 alkenylene; G1b and G2b are each independently C1-C12 alkylene or C2-C12 alkenylene; G3 is C1-C24 alkylene, C2-C24 alkenylene, C3-C8 cycloalkylene or C3-C8 cycloalkenylene; Ra, Rb, Rd and Re are each independently H or C1-C12 alkyl or C2-C12 alkenyl; Rc and Rf are each independently C1-C12 alkyl or C2-C12 alkenyl; R1 and R2 are each independently branched C6-C24 alkyl or branched C6-C24 alkenyl; R3a is -C(=O)N(R4a)R5a or -C(=O)OR6; R3b is -NR4b C(=O)R5b; R4a is C1-C12 alkyl; R4b is H, C1-C12 alkyl or C2-C12 alkenyl; R5a is H, C1-C8 alkyl or C2-C8 alkenyl; R5b is C2-C12 alkyl or C2-C12 alkenyl when R4b is H; or R5b is C1-C12 alkyl or C2-C12 alkenyl when R4b is C1-C12 alkyl or C2-C12 alkenyl; R6 is H, aryl or aralkyl; and x is 0, 1 or 2, and wherein each alkyl, alkenyl, alkylene, alkenylene, cycloalkylene, cycloalkenylene, aryl, and aralkyl is independently substituted or unsubstituted. [0302] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2018/191657 having the structure
Figure imgf000097_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: G1 is -OH, -NR3R4, -(C=O)NR5 or -NR3(C=O)R5; G2 is -CH2- or -(C=O)-; R is, at each occurrence, independently H or OH; R1 and R2 are each independently optionally substituted branched, saturated or unsaturated C12-C36 alkyl; R3 and R4 are each independently H or optionally substituted straight or branched, saturated or unsaturated C1-C6 alkyl; R5 is optionally substituted straight or branched, saturated or unsaturated C1-C6 alkyl; and n is an integer from 2 to 6. [0303] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2017/117528 having the structure:
Figure imgf000098_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)y, -S-S-, -C(=O)S-, SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, -N(Ra)C(=O)N(Ra)-, -OC(=O)N(Ra)- or -N(Ra)C(=O)O-, and the other of G1 or G2 is, at each occurrence, -O(C=O)-, -(C=O)O-, - C(=O)-, -O-, -S(O)y, -S-S-, -C(=O)S-, -SC(=O)-, -N(Ra)C(=O)-, -C(=O)N(Ra)-, - N(Ra)C(=O)N(Ra)-, -OC(=O)N(Ra)- or -N(Ra)C(=O)O- or a direct bond; L is, at each occurrence, ~O(C=O)-, wherein ~ represents a covalent bond to X; X is CRa; Z is alkyl, cycloalkyl or a monovalent moiety comprising at least one polar functional group when n is 1; or Z is alkylene, cycloalkylene or a polyvalent moiety comprising at least one polar functional group when n is greater than 1; Ra is, at each occurrence, independently H, C1-C12 alkyl, C1-C12 hydroxylalkyl, C1-C12 aminoalkyl, C1-C12 alkylaminylalkyl, C1-C12 alkoxyalkyl, C1-C12 alkoxycarbonyl, C1-C12 alkylcarbonyloxy, C1-C12 alkylcarbonyloxyalkyl or C1-C12 alkylcarbonyl; R is, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R together with the carbon atom to which it is bound is taken together with an adjacent R and the carbon atom to which it is bound to form a carbon-carbon double bond; R1 and R2 have, at each occurrence, the following structure, respectively:
Figure imgf000098_0002
a1 and a2 are, at each occurrence, independently an integer from 3 to 12; b1 and b2 are, at each occurrence, independently 0 or 1; c1 and c2 are, at each occurrence, independently an integer from 5 to 10; d1 and d2 are, at each occurrence, independently an integer from 5 to 10; y is, at each occurrence, independently an integer from 0 to 2; and n is an integer from 1 to 6, wherein each alkyl, alkylene, hydroxylalkyl, aminoalkyl, alkylaminylalkyl, alkoxyalkyl, alkoxycarbonyl, alkylcarbonyloxy, alkylcarbonyloxyalkyl and alkylcarbonyl is optionally substituted with one or more substituent. [0304] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2017/075531 having the structure:
Figure imgf000099_0001
or a pharmaceutically acceptable salt, prodrug or stereoisomer thereof, wherein: one of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O-, and the other of L1 or L2 is -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, -C(=O)S-, SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, NRaC(=O)NRa-, -OC(=O)NRa- or -NRaC(=O)O- or a direct bond; G1 and G2 are each independently unsubstituted C1-C12 alkylene or C1-C12 alkenylene; G3 is C1-C24 alkylene, C1-C24 alkenylene, C3-C8 cycloalkylene, C3-C8 cycloalkenylene; Ra a is H or C1-C12 alkyl; R1 and R2 are each independently C6-C24 alkyl or C6-C24 alkenyl; R3 is H, OR5, CN, -C(=O)OR4, -OC(=O)R4 or -NR5C(=O)R4; R4 is C1-C12 alkyl; R5 is H or C1-C6 alkyl; and x is 0, 1 or 2. [0305] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2017/004143 having the structure:
Figure imgf000100_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O-, -C(=O)-, -O-, -S(O)x-, -S-S-, - C(=O)S-, -SC(=O)-, -NRaC(=O)-, -C(=O)NRa-, -NRaC(=O)NRa-, -OC(=O)NRa-, -NRaC(=O)O- or a direct bond; G1 is C1-C2 alkylene, -(C=O)-, -O(C=O)-, -SC(=O)-, -NRaC(=O)- or a direct bond; G2 is -C(=O)-, -(C=O)O-, -C(=O)S-, -C(=O)NRa- or a direct bond; G3 is C1-C6 alkylene; Ra is H or C1-C12 alkyl; R1a and R1b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R1a is H or C1-C12 alkyl, and R1b together with the carbon atom to which it is bound is taken together with an adjacent R1b and the carbon atom to which it is bound to form a carbon- carbon double bond; R2a and R2b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon- carbon double bond; R3a and R3b are, at each occurrence, independently either (a): H or C1-C12 alkyl; or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon- carbon double bond; R4a and R4b are, at each occurrence, independently either: (a) H or C1-C12 alkyl; or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon- carbon double bond; R5 and R6 are each independently H or methyl; R7 is C4-C20 alkyl; R8 and R9 are each independently C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7-membered heterocyclic ring; a, b, c and d are each independently an integer from 1 to 24; and x is 0, 1 or 2. [0306] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2015/199952 having the structure:
Figure imgf000101_0001
or a pharmaceutically acceptable salt, tautomer, prodrug or stereoisomer thereof, wherein: L1 and L2 are each independently -O(C=O)-, -(C=O)O- or a carbon-carbon double bond; Rla and Rlb are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) Rla is H or C1-C12 alkyl, and Rlb together with the carbon atom to which it is bound is taken together with an adjacent Rlb and the carbon atom to which it is bound to form a carbon-carbon double bond; R2a and R2b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R2a is H or C1-C12 alkyl, and R2b together with the carbon atom to which it is bound is taken together with an adjacent R2b and the carbon atom to which it is bound to form a carbon-carbon double bond; R3a and R3b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R3a is H or C1-C12 alkyl, and R3b together with the carbon atom to which it is bound is taken together with an adjacent R3b and the carbon atom to which it is bound to form a carbon-carbon double bond; R4a and R4b are, at each occurrence, independently either (a) H or C1-C12 alkyl, or (b) R4a is H or C1-C12 alkyl, and R4b together with the carbon atom to which it is bound is taken together with an adjacent R4b and the carbon atom to which it is bound to form a carbon-carbon double bond; R5 and R6 are each independently methyl or cycloalkyl; R7 is, at each occurrence, independently H or C1-C12 alkyl; R8 and R9 are each independently unsubstituted C1-C12 alkyl; or R8 and R9, together with the nitrogen atom to which they are attached, form a 5, 6 or 7- membered heterocyclic ring comprising one nitrogen atom; a and d are each independently an integer from 0 to 24; b and c are each independently an integer from 1 to 24; and e is 1 or 2, provided that: at least one of Rla, R2a, R3a or R4a is C1-C12 alkyl, or at least one of L1 or L2 is - O(C=O)- or -(C=O)O-; and Rla and Rlb are not isopropyl when a is 6 or n-butyl when a is 8. [0307] In some embodiments, the ionizable lipids are one or more of the compounds described in WO 2015/074085 having the structure:
Figure imgf000102_0001
wherein R1 and R2 are the same or different, each a linear or branched alkyl with 1-9 carbons, or an alkenyl or alkynyl with 2 to 11 carbon atoms, L1 and L2 are the same or different, each a linear alkyl having 5 to 18 carbon atoms, or form a heterocycle with N, X1 is a bond, or is -CO-O- whereby L2-CO-O-R2 is formed X2 is S or O, L3 is a bond or a lower alkyl, or form a heterocycle with N, R3 is a lower alkyl, and R4 and R5 are the same or different, each a lower alkyl; or a pharmaceutically acceptable salt thereof. [0308] In some embodiments, the ionizable lipids are one or more of the compounds described in Buschmann, M. D. et al., Vaccines, 2021, 9, 65, which incorporated herein in its entirety (the structures provided below include their theoretical pKas):
Figure imgf000103_0001
Figure imgf000104_0001
[0309] In some embodiments, the ionizable lipid is selected from Compound II, Compound III, Compound VI, Compound A1, and Compound A2:
Figure imgf000104_0002
Figure imgf000105_0001
• ii. Polynucleotides [0310] Polynucleotides for use in accordance with the present disclosure include, but are not limited to, one or more of DNA, RNA including messenger RNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix formation, aptamers, vectors, etc., described in detail herein. [0311] In some embodiments, the polynucleotide is RNA. In some embodiments, the polynucleotide is mRNA. [0312] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises from about 900 to about 100,000 nucleotides (e.g., from 900 to 1,000, from 900 to 1,100, from 900 to 1,200, from 900 to 1,300, from 900 to 1,400, from 900 to 1,500, from 1,000 to 1,100, from 1,000 to 1,100, from 1,000 to 1,200, from 1,000 to 1,300, from 1,000 to 1,400, from 1,000 to 1,500, from 1,187 to 1,200, from 1,187 to 1,400, from 1,187 to 1,600, from 1,187 to 1,800, from 1,187 to 2,000, from 1,187 to 3,000, from 1,187 to 5,000, from 1,187 to 7,000, from 1,187 to 10,000, from 1,187 to 25,000, from 1,187 to 50,000, from 1,187 to 70,000, or from 1,187 to 100,000 nucleotides). [0313] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a nucleotide sequence (e.g., an open reading frame (ORF)) encoding a polypeptide, wherein the length of the nucleotide sequence (e.g., an ORF) is at least 500 nucleotides in length, e.g., at least about 500, 600, 700, 80, 900, 1,000, 1,050, 1,100, 1,187, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,100, 2,200, 2,300, 2,400, 2,500, 2,600, 2,700, 2,800, 2,900, 3,000, 3,100, 3,200, 3,300, 3,400, 3,500, 3,600, 3,700, 3,800, 3,900, 4,000, 4,100, 4,200, 4,300, 4,400, 4,500, 4,600, 4,700, 4,800, 4,900, 5,000, 5,100, 5,200, 5,300, 5,400, 5,500, 5,600, 5,700, 5,800, 5,900, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, or 90,000 nucleotides. In some embodiments, the length is up to and including 100,000 nucleotides. [0314] In some embodiments, the polynucleotide of the composition that comprises a nucleotide sequence (e.g., an ORF) encoding a polypeptide is DNA. [0315] In some embodiments, the polynucleotide of the composition is RNA. In some embodiments, the polynucleotide is, or functions as, an mRNA. In some embodiments, the mRNA comprises a nucleotide sequence (e.g., an ORF) that encodes at least one polypeptide, and is capable of being translated to produce the encoded polypeptide in vitro, in vivo, in situ or ex vivo. [0316] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a nucleotide sequence (e.g., an ORF) encoding a polypeptide and further comprises at least one nucleic acid sequence that is noncoding, e.g., a microRNA binding site, e.g., a miRNA binding site that binds to miR-142 and/or a miRNA binding site that binds to miR-126. [0317] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a 5′-UTR and a 3′UTR. [0318] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a 5′ terminal cap. Nonlimiting examples of 5′ terminal caps include Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2′-fluoro-guanosine, 7-deaza- guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, 2-azidoguanosine, Cap2, Cap4, 5′ methylG cap, or an analog thereof. [0319] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) of the lipid nanoparticle comprises a poly-A-tail. In some embodiments, the polyA tail is about 100 nucleotides in length. In some instances, the poly A tail is 100 nucleotides in length. In some instances, the poly A tail is 50-150, 75-150, 85-150, 90-150, 90-120, 90-130, or 90-150 nucleotides in length. [0320] The polynucleotides (e.g., a RNA, e.g., an mRNA) can also comprise nucleotide sequences that encode additional features that facilitate trafficking of the encoded polypeptides to therapeutically relevant sites. One such feature that aids in protein trafficking is the signal sequence, or targeting sequence. The peptides encoded by these signal sequences are known by a variety of names, including targeting peptides, transit peptides, and signal peptides. In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) comprises a nucleotide sequence (e.g., an ORF) that encodes a signal peptide operably linked to a nucleotide sequence that encodes a polypeptide of interest, such as a therapeutic polypeptide,. [0321] In some embodiments, the “signal sequence” or “signal peptide” is a polynucleotide or polypeptide, respectively, which is from about 30-210, e.g., about 45-80 or 15-60 nucleotides (e.g., about 20, 30, 40, 50, 60, or 70 amino acids) in length that, optionally, is incorporated at the 5′ (or N-terminus) of the coding region or the polypeptide, respectively. Addition of these sequences results in trafficking the encoded polypeptide to a desired site, such as the endoplasmic reticulum or the mitochondria through one or more targeting pathways. Some signal peptides are cleaved from the protein, for example by a signal peptidase after the proteins are transported to the desired site. [0322] In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) is sequence optimized. In some embodiments, the polynucleotide (e.g., a RNA, e.g., an mRNA) comprises a 5′ cap, a 5′-UTR, a nucleotide sequence (e.g., an ORF, e.g., a sequence optimized ORF) encoding a polypeptide, a 3′-UTR, and a polyA tail, or any combination thereof, the 5′ UTR or 3′ UTR optionally comprising at least one microRNA binding site. [0323] A sequence-optimized nucleotide sequence, e.g., a codon-optimized mRNA sequence encoding a polypeptide, is a sequence comprising at least one synonymous nucleobase substitution with respect to a reference sequence (e.g., a wild type nucleotide sequence encoding the polypeptide). [0324] A sequence-optimized nucleotide sequence can be partially or completely different in sequence from the reference sequence. For example, a reference sequence encoding polyserine uniformly encoded by UCU codons can be sequence-optimized by having 100% of its nucleobases substituted (for each codon, U in position 1 replaced by A, C in position 2 replaced by G, and U in position 3 replaced by C) to yield a sequence encoding polyserine which would be uniformly encoded by AGC codons. The percentage of sequence identity obtained from a global pairwise alignment between the reference polyserine nucleic acid sequence and the sequence-optimized polyserine nucleic acid sequence would be 0%. However, the protein products from both sequences would be 100% identical. [0325] Some sequence optimization (also sometimes referred to codon optimization) methods are known in the art (and discussed in more detail below) and can be useful to achieve one or more desired results. These results can include, e.g., matching codon frequencies in certain tissue targets and/or host organisms to ensure proper folding; biasing G/C content to increase mRNA stability or reduce secondary structures; minimizing tandem repeat codons or base runs that can impair gene construction or expression; customizing transcriptional and translational control regions; inserting or removing protein trafficking sequences; removing/adding post translation modification sites in an encoded protein (e.g., glycosylation sites); adding, removing or shuffling protein domains; inserting or deleting restriction sites; modifying ribosome binding sites and mRNA degradation sites; adjusting translational rates to allow the various domains of the protein to fold properly; and/or reducing or eliminating problem secondary structures within the polynucleotide. Sequence optimization tools, algorithms and services are known in the art, non-limiting examples include services from GeneArt (Life Technologies), DNA2.0 (Menlo Park CA) and/or are proprietary methods. [0326] In some embodiments, the sequence optimization method is multiparametric and comprises one, two, three, four, or more methods disclosed herein and/or other optimization methods known in the art. [0327] In some embodiments, a polynucleotide (e.g., a RNA, e.g., an mRNA) of the composition comprises a sequence-optimized nucleotide sequence (e.g., an ORF) encoding a polypeptide, wherein the polypeptide encoded by the sequence-optimized nucleotide sequence has improved properties (e.g., compared to the polypeptide as encoded by a reference nucleotide sequence that is not sequence optimized), e.g., improved properties related to expression efficacy after administration in vivo. Such properties include, but are not limited to, improving nucleic acid stability (e.g., mRNA stability), increasing translation efficacy in the target tissue, reducing the number of truncated proteins expressed, improving the folding or preventing misfolding of the expressed proteins, reducing toxicity of the expressed products, reducing cell death caused by the expressed products, increasing and/or decreasing protein aggregation. [0328] In some embodiments, the sequence-optimized nucleotide sequence (e.g., an ORF) is codon optimized for expression in human subjects, having structural and/or chemical features that avoid one or more of the problems in the art, for example, features which are useful for optimizing formulation and delivery of nucleic acid-based therapeutics while retaining structural and functional integrity; overcoming a threshold of expression; improving expression rates; half-life and/or protein concentrations; optimizing protein localization; and avoiding deleterious bio-responses such as the immune response and/or degradation pathways. [0329] Methods for optimizing codon usage are known in the art. For example, an ORF of any one or more of the sequences provided herein may be codon optimized. Codon optimization, in some embodiments, may be used to match codon frequencies in target and host organisms to ensure proper folding; bias GC content to increase mRNA stability or reduce secondary structures; minimize tandem repeat codons or base runs that may impair gene construction or expression; customize transcriptional and translational control regions; insert or remove protein trafficking sequences; remove/add post translation modification sites in encoded protein (e.g., glycosylation sites); add, remove or shuffle protein domains; insert or delete restriction sites; modify ribosome binding sites and mRNA degradation sites; adjust translational rates to allow the various domains of the protein to fold properly; or reduce or eliminate problem secondary structures within the polynucleotide. Codon optimization tools, algorithms and services are known in the art - non-limiting examples include services from GeneArt (Life Technologies), DNA2.0 (Menlo Park CA) and/or are proprietary methods. In some embodiments, the open reading frame (ORF) sequence is optimized using optimization algorithms. [0330] Features, which can be considered beneficial in some embodiments, can be encoded by or within regions of the polynucleotide and such regions can be upstream (5′) to, downstream (3′) to, or within the region that encodes the polypeptide. These regions can be incorporated into the polynucleotide before and/or after sequence-optimization of the protein encoding region or open reading frame (ORF). Examples of such features include, but are not limited to, untranslated regions (UTRs), microRNA sequences, Kozak sequences, oligo(dT) sequences, poly-A tail, and detectable tags and can include multiple cloning sites that can have XbaI recognition. [0331] In some embodiments, the polynucleotide comprises a 5′ UTR, a 3′ UTR and/or a microRNA binding site. In some embodiments, the polynucleotide comprises two or more 5′ UTRs and/or 3′ UTRs, which can be the same or different sequences. In some embodiments, the polynucleotide comprises two or more microRNA binding sites, which can be the same or different sequences. Any portion of the 5′ UTR, 3′ UTR, and/or microRNA binding site, including none, can be sequence-optimized and can independently contain one or more different structural or chemical modifications, before and/or after sequence optimization. [0332] In some embodiments, the polynucleotides of the compositions are modified. The modified polynucleotides can be chemically modified and/or structurally modified. When the polynucleotides of the compositions are chemically and/or structurally modified the polynucleotides can be referred to as “modified polynucleotides.” [0333] The present disclosure provides for modified nucleosides and modified nucleotides of a polynucleotide (e.g., RNA polynucleotides, such as mRNA polynucleotides). A “nucleoside” refers to a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as “nucleobase”). A “nucleotide” refers to a nucleoside including a phosphate group. Modified nucleotides can be synthesized by any useful method, such as, for example, chemically, enzymatically, or recombinantly, to include one or more modified or non-natural nucleosides. Polynucleotides can comprise a region or regions of linked nucleosides. Such regions can have variable backbone linkages. The linkages can be standard phosphodiester linkages, in which case the polynucleotides would comprise regions of nucleotides. [0334] The modified polynucleotides disclosed herein can comprise various distinct modifications. In some embodiments, the modified polynucleotides contain one, two, or more (optionally different) nucleoside or nucleotide modifications. In some embodiments, a modified polynucleotide, introduced to a cell, can exhibit one or more desirable properties, e.g., improved protein expression, reduced immunogenicity, or reduced degradation in the cell, as compared to an unmodified polynucleotide. [0335] In some embodiments, a polynucleotide of the lipid nanoparticles are structurally modified. As used herein, a “structural” modification is one in which two or more linked nucleosides are inserted, deleted, duplicated, inverted or randomized in a polynucleotide without significant chemical modification to the nucleotides themselves. Because chemical bonds will necessarily be broken and reformed to effect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides. For example, the polynucleotide “ATCG” can be chemically modified to “AT-5meC-G”. The same polynucleotide can be structurally modified from “ATCG” to “ATCCCG”. Here, the dinucleotide “CC” has been inserted, resulting in a structural modification to the polynucleotide. [0336] Therapeutic lipid nanoparticles comprise, in some embodiments, at least one nucleic acid (e.g., RNA), wherein the nucleic acid comprises nucleotides and/or nucleosides that can be standard (unmodified) or modified as is known in the art. In some embodiments, nucleotides and nucleosides comprise modified nucleotides or nucleosides. Such modified nucleotides and nucleosides can be naturally-occurring modified nucleotides and nucleosides or non-naturally occurring modified nucleotides and nucleosides. Such modifications can include those at the sugar, backbone, or nucleobase portion of the nucleotide and/or nucleoside as are recognized in the art. [0337] In some embodiments, a naturally-occurring modified nucleotide or nucleoside is one as is generally known or recognized in the art. Non-limiting examples of such naturally occurring modified nucleotides and nucleosides can be found, inter alia, in the MODOMICS database. [0338] In some embodiments, a non-naturally occurring modified nucleotide or nucleoside is one as is generally known or recognized in the art. Non-limiting examples of such non-naturally occurring modified nucleotides and nucleosides can be found, inter alia, in published International Patent Application Nos. PCT/US2012/058519; PCT/US2013/075177; PCT/US2014/058897; PCT/US2014/058891; PCT/US2014/070413; PCT/US2015/36773; PCT/US2015/36759; PCT/US2015/36771; or PCT/IB2017/051367, each of which is incorporated by reference herein in its entirety. [0339] In some embodiments, at least one RNA (e.g., mRNA) is not chemically modified and comprises the standard ribonucleotides consisting of adenosine, guanosine, cytosine and uridine. In some embodiments, nucleotides and nucleosides comprise standard nucleoside residues such as those present in transcribed RNA (e.g. A, G, C, or U). In some embodiments, nucleotides and nucleosides comprise standard deoxyribonucleosides such as those present in DNA (e.g. dA, dG, dC, or dT). [0340] Hence, nucleic acids (e.g., DNA nucleic acids and RNA nucleic acids, such as mRNA nucleic acids) can comprise standard nucleotides and nucleosides, naturally-occurring nucleotides and nucleosides, non-naturally-occurring nucleotides and nucleosides, or any combination thereof. [0341] Nucleic acids (e.g., DNA nucleic acids and RNA nucleic acids, such as mRNA nucleic acids), in some embodiments, comprise various (more than one) different types of standard and/or modified nucleotides and nucleosides. In some embodiments, a particular region of a nucleic acid contains one, two or more (optionally different) types of standard and/or modified nucleotides and nucleosides. [0342] In some embodiments, a modified RNA nucleic acid (e.g., a modified mRNA nucleic acid), introduced to a cell or organism, exhibits reduced degradation in the cell or organism, respectively, relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides. [0343] In some embodiments, a modified RNA nucleic acid (e.g., a modified mRNA nucleic acid), introduced into a cell or organism, may exhibit reduced immunogenicity in the cell or organism, respectively (e.g., a reduced innate response) relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides. [0344] Nucleic acids (e.g., RNA nucleic acids, such as mRNA nucleic acids), in some embodiments, comprise non-natural modified nucleotides that are introduced during synthesis or post-synthesis of the nucleic acids to achieve desired functions or properties. The modifications may be present on internucleotide linkages, purine or pyrimidine bases, or sugars. The modification may be introduced with chemical synthesis or with a polymerase enzyme at the terminus of a chain or anywhere else in the chain. Any of the regions of a nucleic acid may be chemically modified. [0345] Modified nucleotide base pairing encompasses not only the standard adenosine- thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures, such as in those nucleic acids having at least one chemical modification. One example of such non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil. Any combination of base/sugar or linker may be incorporated into nucleic acids. [0346] In some embodiments, modified nucleobases in nucleic acids (e.g., RNA nucleic acids, such as mRNA nucleic acids) comprise N1-methyl-pseudouridine (m1ψ), 1-ethyl- pseudouridine (e1ψ), 5-methoxy-uridine (mo5U), 5-methyl-cytidine (m5C), and/or pseudouridine (ψ). In some embodiments, modified nucleobases in nucleic acids (e.g., RNA nucleic acids, such as mRNA nucleic acids) comprise 5-methoxymethyl uridine, 5-methylthio uridine, 1-methoxymethyl pseudouridine, 5-methyl cytidine, and/or 5-methoxy cytidine. In some embodiments, the polyribonucleotide includes a combination of at least two (e.g., 2, 3, 4 or more) of any of the aforementioned modified nucleobases, including but not limited to chemical modifications. [0347] In some embodiments, nucleic acids (e.g., RNA nucleic acids, such as mRNA nucleic acids) are uniformly modified (e.g., fully modified, modified throughout the entire sequence) for a particular modification. For example, a nucleic acid can be uniformly modified with N1-methyl-pseudouridine, meaning that all uridine residues in the mRNA sequence are replaced with N1-methyl-pseudouridine. Similarly, a nucleic acid can be uniformly modified for any type of nucleoside residue present in the sequence by replacement with a modified residue such as those set forth above. [0348] The nucleic acids may be partially or fully modified along the entire length of the molecule. For example, one or more or all or a given type of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may be uniformly modified in a nucleic acid, or in a predetermined sequence region thereof (e.g., in the mRNA including or excluding the polyA tail). In some embodiments, all nucleotides X in a nucleic acid (or in a sequence region thereof) are modified nucleotides, wherein X may be any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C. [0349] The nucleic acid may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e., any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%). It will be understood that any remaining percentage is accounted for by the presence of unmodified A, G, U, or C. [0350] The nucleic acids may contain at a minimum 1% and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50% modified nucleotides, at least 80% modified nucleotides, or at least 90% modified nucleotides. For example, the nucleic acids may contain a modified pyrimidine such as a modified uracil or cytosine. In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid is replaced with a modified uracil (e.g., a 5-substituted uracil). The modified uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the nucleic acid is replaced with a modified cytosine (e.g., a 5-substituted cytosine). The modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). [0351] The polynucleotides of the lipid nanoparticles can be generated using components, compositions, and methods as are generally known in the art, see, e.g., International Patent Application Publication Nos. WO 2015/051173, WO 2017/049286, WO 2016/100812, WO 2016/014846, WO 2016/011226, WO 2016/011222, WO 2016/011306, WO 2015/196128, WO 2013/151736, WO 2013/151672, WO 2013/151671, WO 2013/151670, WO 2013/151669, WO 2013/151668, WO 2013/151666, WO 2013/151667, WO 2013/151665, WO 2013/151664, WO 2013/151663, WO 2013/151736, WO 2013/151668, WO 2013/151666, WO 2013/151665, WO 2013/151670, WO 2013/151672, WO 2015/089511, WO 2015/051173, WO 2015/051169, each of which is incorporated by reference herein in its entirety. [0352] In some aspects, a polynucleotide (e.g., a RNA, e.g., an mRNA) disclosed herein can be constructed using in vitro transcription (IVT). In other aspects, a polynucleotide (e.g., a RNA, e.g., an mRNA) disclosed herein can be constructed by chemical synthesis using an oligonucleotide synthesizer. [0353] In other aspects, a polynucleotide (e.g., a RNA, e.g., an mRNA) disclosed herein is made by using a host cell. In certain aspects, a polynucleotide (e.g., a RNA, e.g., an mRNA) disclosed herein is made by one or more combination of the IVT, chemical synthesis, host cell expression, or any other methods known in the art. [0354] Naturally occurring nucleosides, non-naturally occurring nucleosides, or combinations thereof, can totally or partially naturally replace occurring nucleosides present in the candidate nucleotide sequence and can be incorporated into a sequence-optimized nucleotide sequence (e.g., a RNA, e.g., an mRNA). The resultant polynucleotides, e.g., mRNAs, can then be examined for their ability to produce protein and/or produce a therapeutic outcome. 1) Purification of Polynucleotides [0355] The polynucleotides can be purified prior to their inclusion in the lipid nanoparticles. Purification of the polynucleotides described herein can include, but is not limited to, polynucleotide clean-up, quality assurance and quality control. Clean-up can be performed by methods known in the arts such as, but not limited to, AGENCOURT® beads (Beckman Coulter Genomics, Danvers, MA), poly-T beads, LNATM oligo-T capture probes (EXIQON® Inc., Vedbaek, Denmark) or HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC). [0356] In some embodiments, purification of a polynucleotide removes impurities that can reduce or remove an unwanted immune response, e.g., reducing cytokine activity. [0357] In some embodiments, the polynucleotide is purified prior to inclusion in a lipid nanoparticle using column chromatography (e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC- HPLC), or (LCMS)). [0358] In some embodiments, the purified polynucleotide is at least about 80% pure, at least about 85% pure, at least about 90% pure, at least about 95% pure, at least about 96% pure, at least about 97% pure, at least about 98% pure, at least about 99% pure, or about 100% pure prior to inclusion in a lipid nanoparticle. [0359] A quality assurance and/or quality control check can be conducted using methods such as, but not limited to, gel electrophoresis, UV absorbance, or analytical HPLC. In another embodiment, the polynucleotide can be sequenced by methods including, but not limited to reverse-transcriptase-PCR. 2) Quantification of Polynucleotides [0360] In some embodiments, the polynucleotides, their expression products, as well as degradation products and metabolites can be quantified according to methods known in the art. [0361] In some embodiments, the polynucleotide can be quantified using methods such as, but not limited to, ultraviolet visible spectroscopy (UV/Vis). A non-limiting example of a UV/Vis spectrometer is a NANODROP® spectrometer (ThermoFisher, Waltham, MA). The quantified polynucleotide can be analyzed in order to determine if the polynucleotide can be of proper size, or to check that no degradation of the polynucleotide has occurred. Degradation of the polynucleotide can be checked by methods (such as, but not limited to, agarose gel electrophoresis, HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP- HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE), capillary gel electrophoresis (CGE)); and UPLC (e.g., RP-UPLC). Other Lipid Nanoparticle Components [0362] The lipid composition of a lipid nanoparticle disclosed herein can include one or more components in addition to the lipid components described above (e.g., in addition to the ionizable cationic lipid, non-cationic lipid, sterol, and PEG-modified lipid). For example, the lipid composition can include one or more permeability enhancer molecules, carbohydrates, polymers, surface altering agents (e.g., surfactants), or other components. For example, a permeability enhancer molecule can be a molecule described by U.S. Patent Application Publication No.2005/0222064. Carbohydrates can include simple sugars (e.g., glucose) and polysaccharides (e.g., glycogen and derivatives and analogs thereof). A polymer can be included in and/or used to encapsulate or partially encapsulate a composition disclosed herein (e.g., an LNP composition). A polymer can be biodegradable and/or biocompatible. A polymer can be selected from, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, polystyrenes, polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyleneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates. [0363] The LNP can further contain a phosphate conjugate. The phosphate conjugate can increase in vivo circulation times and/or increase the targeted delivery of the nanoparticle. Phosphate conjugates can be made by the methods described in, e.g., Intl. Pub. No. WO 2013033438 or U.S. Pub. No.2013/0196948. The LNP can also contain a polymer conjugate (e.g., a water soluble conjugate) as described in, e.g., U.S. Pub. Nos.2013/0059360, 2013/0196948, and 2013/0072709. Each of the references is herein incorporated by reference in its entirety. [0364] The LNPs can comprise a conjugate to enhance the delivery of nanoparticles in a subject. Further, the conjugate can inhibit phagocytic clearance of the nanoparticles in a subject. In some embodiments, the conjugate can be a “self” peptide designed from the human membrane protein CD47 (e.g., the “self” particles described by Rodriguez et al, Science 2013339, 971-975, herein incorporated by reference in its entirety). As shown by Rodriguez et al. the self peptides delayed macrophage-mediated clearance of nanoparticles which enhanced delivery of the nanoparticles. [0365] The LNPs can comprise a carbohydrate carrier. As a non-limiting example, the carbohydrate carrier can include, but is not limited to, an anhydride-modified phytoglycogen or glycogen-type material, phytoglycogen octenyl succinate, phytoglycogen beta-dextrin, anhydride-modified phytoglycogen beta-dextrin (e.g., Intl. Pub. No. WO 2012/109121, herein incorporated by reference in its entirety). [0366] The LNPs can be coated with a surfactant or polymer to improve the delivery of the particle. In some embodiments, the LNP can be coated with a hydrophilic coating such as, but not limited to, PEG coatings and/or coatings that have a neutral surface charge as described in U.S. Pub. No.2013/0183244, herein incorporated by reference in its entirety. [0367] The LNPs can be engineered to alter the surface properties of particles so that the lipid nanoparticles can penetrate the mucosal barrier as described in U.S. Pat. No.8,241,670 or Intl. Pub. No. WO 2013/110028, each of which is herein incorporated by reference in its entirety. [0368] The LNPs engineered to penetrate mucus can comprise a polymeric material (e.g., a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer. The polymeric material can include, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates. [0369] LNPs engineered to penetrate mucus can also include surface altering agents such as, but not limited to, polynucleotides, anionic proteins (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin β4 dornase alfa, neltenexine, erdosteine) and various DNases including rhDNase. [0370] In some embodiments, the mucus penetrating LNP can be a hypotonic formulation comprising a mucosal penetration enhancing coating. The formulation can be hypotonic for the epithelium to which it is being delivered. Non-limiting examples of hypotonic formulations can be found in, e.g., Intl. Pub. No. WO 2013/110028, herein incorporated by reference in its entirety. 1) Other Lipids a) Phospholipids [0371] The lipid composition of a lipid nanoparticle disclosed herein can comprise one or more phospholipids, for example, one or more saturated or (poly)unsaturated phospholipids or a combination thereof. In general, phospholipids comprise a phospholipid moiety and one or more fatty acid moieties. [0372] A phospholipid moiety can be selected, for example, from the non-limiting group consisting of phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol, phosphatidyl serine, phosphatidic acid, 2-lysophosphatidyl choline, and a sphingomyelin. [0373] A fatty acid moiety can be selected, for example, from the non-limiting group consisting of lauric acid, myristic acid, myristoleic acid, palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, alpha-linolenic acid, erucic acid, phytanoic acid, arachidic acid, arachidonic acid, eicosapentaenoic acid, behenic acid, docosapentaenoic acid, and docosahexaenoic acid. [0374] Particular phospholipids can facilitate fusion to a membrane. For example, a cationic phospholipid can interact with one or more negatively charged phospholipids of a membrane (e.g., a cellular or intracellular membrane). Fusion of a phospholipid to a membrane can allow one or more elements (e.g., a therapeutic agent) of a lipid-containing composition (e.g., LNPs) to pass through the membrane permitting, e.g., delivery of the one or more elements to a target tissue. [0375] Non-natural phospholipid species including natural species with modifications and substitutions including branching, oxidation, cyclization, and alkynes are also contemplated. For example, a phospholipid can be functionalized with or cross-linked to one or more alkynes (e.g., an alkenyl group in which one or more double bonds is replaced with a triple bond). Under appropriate reaction conditions, an alkyne group can undergo a copper- catalyzed cycloaddition upon exposure to an azide. Such reactions can be useful in functionalizing a lipid bilayer of a nanoparticle composition to facilitate membrane permeation or cellular recognition or in conjugating a nanoparticle composition to a useful component such as a targeting or imaging moiety (e.g., a dye). [0376] Phospholipids include, but are not limited to, glycerophospholipids such as phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines, phosphatidylinositols, phosphatidy glycerols, and phosphatidic acids. Phospholipids also include phosphosphingolipid, such as sphingomyelin. [0377] In some embodiments, a phospholipid comprises 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2- dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-gly cero- phosphocholine (DMPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), l,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-di- O-octadecenyl-sn-glycero-3-phosphocholine (18:0 Diether PC), 1-oleoyl-2 cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1-hexadecyl-sn- glycero-3-phosphocholine (C16 Lyso PC), 1,2-dilinolenoyl-sn-glycero-3-phosphocholine,1,2- diarachidonoyl-sn-glycero-3-phosphocholine, 1,2-didocosahexaenoyl-sn-glycero-3- phosphocholine, 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (ME 16.0 PE), 1,2- distearoyl-sn-glycero-3-phosphoethanolamine, 1,2-dilinoleoyl-sn-glycero-3- phosphoethanolamine, 1,2-dilinolenoyl-sn-glycero-3-phosphoethanolamine, 1,2- diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1,2-didocosahexaenoyl-sn-glycero-3- phosphoethanolamine, 1,2-dioleoyl-sn-glycero-3-phospho-rac-(1-glycerol) sodium salt (DOPG), sphingomyelin, or mixtures thereof. [0378] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure is an analog or variant of DSPC. In certain embodiments, a phospholipid useful or potentially useful in the present disclosure is a compound of Formula (IV):
Figure imgf000121_0001
or a salt thereof, wherein: each R1 is independently optionally substituted alkyl; or optionally two R1 are joined together with the intervening atoms to form optionally substituted monocyclic carbocyclyl or optionally substituted monocyclic heterocyclyl; or optionally three R1 are joined together with the intervening atoms to form optionally substituted bicyclic carbocyclyl or optionally substitute bicyclic heterocyclyl; n is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; m is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; A is of the formula:
Figure imgf000122_0001
each instance of L2 is independently a bond or optionally substituted C1-6 alkylene, wherein one methylene unit of the optionally substituted C1-6 alkylene is optionally replaced with O, N(RN), S, C(O), C(O)N(RN), NRNC(O), C(O)O, OC(O), OC(O)O, OC(O)N(RN), NRNC(O)O, or -NRNC(O)N(RN); each instance of R2 is independently optionally substituted C1-30 alkyl, optionally substituted C1-30 alkenyl, or optionally substituted C1-30 alkynyl; optionally wherein one or more methylene units of R2 are independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(RN), O, S, C(O), C(O)N(RN), NRNC(O), - NRNC(O)N(RN), C(O)O, OC(O), OC(O)O, -OC(O)N(RN), NRNC(O)O, C(O)S, SC(O), C(=NRN), C(=NRN)N(RN), - NRNC(=NRN), NRNC(=NRN)N(RN), C(S), C(S)N(RN), NRNC(S), NRNC(S)N(RN), S(O), - OS(O), S(O)O, -OS(O)O, OS(O)2, S(O)2O, OS(O)2O, N(RN)S(O), S(O)N(RN), N(RN)S(O)N(RN), - OS(O)N(RN), N(RN)S(O)O, S(O)2, N(RN)S(O)2, S(O)2N(RN), N(RN)S(O)2N(RN), - OS(O)2N(RN), or -N(RN)S(O)2O; each instance of RN is independently hydrogen, optionally substituted alkyl, or a nitrogen protecting group; Ring B is optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; and p is 1 or 2; provided that the compound is not of the formula:
Figure imgf000123_0001
, wherein each instance of R2 is independently unsubstituted alkyl, unsubstituted alkenyl, or unsubstituted alkynyl. [0379] In some embodiments, the phospholipids may be one or more of the phospholipids described in PCT/US2018/037922 (published as WO 2018232357). A) Phospholipid Head Modifications [0380] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure comprises a modified phospholipid head (e.g., a modified choline group). In certain embodiments, a phospholipid with a modified head is DSPC, or analog thereof, with a modified quaternary amine. For example, in embodiments of Formula (IV), at least one of R1 is not methyl. In certain embodiments, at least one of R1 is not hydrogen or methyl. In certain embodiments, the compound of Formula (IV) is of one of the following formulae: ,
Figure imgf000123_0002
or a salt thereof, wherein: each t is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; each u is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and each v is independently 1, 2, or 3. [0381] In certain embodiments, a compound of Formula (IV) is of Formula (IV-a):
Figure imgf000124_0001
salt thereof. [0382] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure comprises a cyclic moiety in place of the glyceride moiety. In certain embodiments, a phospholipid useful in the present disclosure is DSPC, or analog thereof, with a cyclic moiety in place of the glyceride moiety. In certain embodiments, the compound of Formula (IV) is of Formula (IV-b):
Figure imgf000124_0002
salt thereof. B) Phospholipid Tail Modifications [0383] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure comprises a modified tail. In certain embodiments, a phospholipid useful or potentially useful in the present disclosure is DSPC, or analog thereof, with a modified tail. As described herein, a “modified tail” may be a tail with shorter or longer aliphatic chains, aliphatic chains with branching introduced, aliphatic chains with substituents introduced, aliphatic chains wherein one or more methylenes are replaced by cyclic or heteroatom groups, or any combination thereof. For example, in certain embodiments, the compound of (IV) is of Formula (IV-a), or a salt thereof, wherein at least one instance of R2 is each instance of R2 is optionally substituted C1-30 alkyl, wherein one or more methylene units of R2 are independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(RN), O, S, C(O), C(O)N(RN), NRNC(O), NRNC(O)N(RN), C(O)O, OC(O), OC(O)O, OC(O)N(RN), NRNC(O)O, C(O)S, SC(O), C(=NRN), C(=NRN)N(RN), NRNC(=NRN), NRNC(=NRN)N(RN), C(S), C(S)N(RN), NRNC(S), NRNC(S)N(RN), S(O), OS(O), S(O)O, OS(O)O, OS(O)2, - S(O)2O, OS(O)2O, N(RN)S(O), S(O)N(RN), N(RN)S(O)N(RN), -OS(O)N(RN), N(RN)S(O)O, S(O)2, N(RN)S(O)2, S(O)2N(RN), N(RN)S(O)2N(RN), OS(O)2N(RN), or N(RN)S(O)2O. [0384] In certain embodiments, the compound of Formula (IV) is of Formula (IV-c):
Figure imgf000125_0001
(IV-c), or a salt thereof, wherein: each x is independently an integer between 0-30, inclusive; and each instance is G is independently selected from the group consisting of optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(RN), O, S, C(O), C(O)N(RN), NRNC(O), - NRNC(O)N(RN), C(O)O, OC(O), OC(O)O, OC(O)N(RN), NRNC(O)O, C(O)S, SC(O), - C(=NRN), C(=NRN)N(RN), NRNC(=NRN), NRNC(=NRN)N(RN), C(S), C(S)N(RN), NRNC(S), NRNC(S)N(RN), S(O), OS(O), S(O)O, OS(O)O, OS(O)2, S(O)2O, OS(O)2O, N(RN)S(O), - S(O)N(RN), N(RN)S(O)N(RN), OS(O)N(RN), N(RN)S(O)O, S(O)2, N(RN)S(O)2, S(O)2N(RN), N(RN)S(O)2N(RN), OS(O)2N(RN), or N(RN)S(O)2O. Each possibility represents separate embodiments. [0385] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure comprises a modified phosphocholine moiety, wherein the alkyl chain linking the quaternary amine to the phosphoryl group is not ethylene (e.g., n is not 2). Therefore, in certain embodiments, a phospholipid useful or potentially useful in the present disclosure is a compound of Formula (IV), wherein n is 1, 3, 4, 5, 6, 7, 8, 9, or 10. For example, in certain embodiments, a compound of Formula (IV) is of one of the following formulae:
Figure imgf000125_0002
salt thereof. • b) Alternative Lipids [0386] In certain embodiments, a phospholipid useful or potentially useful in the present disclosure comprises a modified phosphocholine moiety, wherein the alkyl chain linking the quaternary amine to the phosphoryl group is not ethylene (e.g., n is not 2). Therefore, in certain embodiments, a phospholipid is useful. [0387] In certain embodiments, an alternative lipid is used in place of a phospholipid. [0388] In certain embodiments, an alternative lipid is oleic acid. [0389] In certain embodiments, the alternative lipid is one of the following: , ,
Figure imgf000126_0001
,
Figure imgf000127_0001
. • c) Structural Lipids [0390] The lipid composition of a lipid nanoparticle disclosed herein can comprise one or more structural lipids. As used herein, the term “structural lipid” refers to sterols and also to lipids containing sterol moieties. [0391] Incorporation of structural lipids in the lipid nanoparticle may help mitigate aggregation of other lipids in the particle. Structural lipids can be selected from the group including but not limited to, cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, tomatine, ursolic acid, alpha-tocopherol, hopanoids, phytosterols, steroids, and mixtures thereof. In some embodiments, the structural lipid is a sterol. As defined herein, “sterols” are a subgroup of steroids consisting of steroid alcohols. In certain embodiments, the structural lipid is a steroid. In certain embodiments, the structural lipid is cholesterol. In certain embodiments, the structural lipid is an analog of cholesterol. In certain embodiments, the structural lipid is alpha-tocopherol. [0392] In some embodiments, the structural lipids may be one or more of the structural lipids described in PCT/US2018/037922 (published as WO 2018/232357). [0393] In some embodiments, the structural lipid is cholesterol. In some embodiments, the amount of the structural lipids (e.g., cholesterol) in the lipid composition ranges from about 20 mol% to about 60 mol%. d) Polyethylene Glycol (PEG)-Lipids [0394] The lipid composition of a lipid nanoparticle disclosed herein can comprise one or more polyethylene glycol (PEG) lipids. [0395] As used herein, the term “PEG-lipid” refers to polyethylene glycol (PEG)- modified lipids. Non-limiting examples of PEG-lipids include PEG-modified phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG- CerC14 or PEG-CerC20), PEG-modified dialkylamines and PEG-modified 1,2- diacyloxypropan-3-amines. Such lipids are also referred to as PEGylated lipids. For example, a PEG lipid can be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid. [0396] In some embodiments, the PEG-lipid includes, but is not limited to 1,2- dimyristoyl-sn-glycerol methoxypolyethylene glycol (PEG-DMG), 1,2-distearoyl-sn-glycero- 3-phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEG- DAG), PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), or PEG-l,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA). [0397] In some embodiments, the PEG-lipid is selected from the group consisting of a PEG-modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG- modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG- modified dialkylglycerol, and mixtures thereof. [0398] In some embodiments, the lipid moiety of the PEG-lipids includes those having lengths of from about C14 to about C22, preferably from about C14 to about C16. In some embodiments, a PEG moiety, for example an mPEG-NH2, has a size of about 1000, 2000, 5000, 10,000, 15,000 or 20,000 daltons. In some embodiments, the PEG-lipid is PEG2k- DMG. [0399] In some embodiments, the lipid nanoparticles described herein can comprise a PEG lipid which is a non-diffusible PEG. Non-limiting examples of non-diffusible PEGs include PEG-DSG and PEG-DSPE. PEG-lipids are known in the art, such as those described in U.S. Patent No.8158601 and International Publ. No. WO 2015/130584 A2, which are incorporated herein by reference in their entirety. [0400] In general, some of the other lipid components (e.g., PEG lipids) of various formulae, described herein may be synthesized as described International Patent Application No. PCT/US2016/000129, filed December 10, 2016, entitled “Compositions and Methods for Delivery of Therapeutic Agents,” which is incorporated by reference in its entirety. [0401] Thus, the lipid component of a lipid nanoparticle composition may include one or more molecules comprising polyethylene glycol, such as PEG or PEG-modified lipids. Such species may be alternately referred to as PEGylated lipids. A PEG lipid is a lipid modified with polyethylene glycol. A PEG lipid may be selected from the non-limiting group including PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, and mixtures thereof. For example, a PEG lipid may be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, or a PEG-DSPE lipid. [0402] As noted above, in some embodiments the PEG-modified lipids are a modified form of PEG DMG. PEG-DMG has the following structure:
Figure imgf000129_0001
[0403] In some embodiments, PEG lipids useful in the present disclosure can be PEGylated lipids described in International Publication No. WO 2012/099755, which is herein incorporated by reference in its entirety. Any of these exemplary PEG lipids described herein may be modified to comprise a hydroxyl group on the PEG chain. In certain embodiments, the PEG lipid is a PEG-OH lipid. As generally defined herein, a “PEG-OH lipid” (also referred to herein as “hydroxy-PEGylated lipid”) is a PEGylated lipid having one or more hydroxyl (–OH) groups on the lipid. In certain embodiments, the PEG-OH lipid includes one or more hydroxyl groups on the PEG chain. In certain embodiments, a PEG-OH or hydroxy-PEGylated lipid comprises an –OH group at the terminus of the PEG chain. Each possibility represents separate embodiments. [0404] In certain embodiments, a PEG lipid useful in the present disclosure is a compound of Formula (V). Provided herein are compounds of Formula (V): (V), or salts thereof, wherein: R3 is –ORO; RO is hydrogen, optionally substituted alkyl, or an oxygen protecting group; r is an integer between 1 and 100, inclusive; L1 is optionally substituted C1-10 alkylene, wherein at least one methylene of the optionally substituted C1-10 alkylene is independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, O, N(RN), S, C(O), C(O)N(RN), NRNC(O), C(O)O, - OC(O), OC(O)O, -OC(O)N(RN), NRNC(O)O, or NRNC(O)N(RN); [0405] D is a moiety obtained by click chemistry or a moiety cleavable under physiological conditions; m is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; A is of the formula:
Figure imgf000130_0001
each instance of L2 is independently a bond or optionally substituted C1-6 alkylene, wherein one methylene unit of the optionally substituted C1-6 alkylene is optionally replaced with O, N(RN), S, C(O), C(O)N(RN), NRNC(O), C(O)O, OC(O), OC(O)O, OC(O)N(RN), NRNC(O)O, or -NRNC(O)N(RN); each instance of R2 is independently optionally substituted C1-30 alkyl, optionally substituted C1-30 alkenyl, or optionally substituted C1-30 alkynyl; optionally wherein one or more methylene units of R2 are independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(RN), O, S, C(O), C(O)N(RN), NRNC(O), - NRNC(O)N(RN), C(O)O, OC(O), OC(O)O, -OC(O)N(RN), NRNC(O)O, C(O)S, SC(O), - C(=NRN), C(=NRN)N(RN), NRNC(=NRN), -NRNC(=NRN)N(RN), C(S), C(S)N(RN), NRNC(S), NRNC(S)N(RN), S(O) , OS(O), S(O)O, -OS(O)O, OS(O)2, S(O)2O, OS(O)2O, N(RN)S(O), - S(O)N(RN), N(RN)S(O)N(RN), OS(O)N(RN), N(RN)S(O)O, S(O)2, N(RN)S(O)2, S(O)2N(RN), N(RN)S(O)2N(RN), OS(O)2N(RN), or -N(RN)S(O)2O; each instance of RN is independently hydrogen, optionally substituted alkyl, or a nitrogen protecting group; [0406] Ring B is optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl; and p is 1 or 2. [0407] In certain embodiments, the compound of Formula (V) is a PEG-OH lipid (i.e., R3 is –ORO, and RO is hydrogen). In certain embodiments, the compound of Formula (V) is of Formula (V-OH):
Figure imgf000131_0001
(V-OH), or a salt thereof. In certain embodiments, a PEG lipid useful in the present disclosure is a PEGylated fatty acid. In certain embodiments, a PEG lipid useful in the present disclosure is a compound of Formula (VI). Provided herein are compounds of Formula (VI):
Figure imgf000131_0002
or a salt thereof, wherein: R3 is–ORO; RO is hydrogen, optionally substituted alkyl or an oxygen protecting group; r is an integer between 1 and 100, inclusive; R5 is optionally substituted C10-40 alkyl, optionally substituted C10-40 alkenyl, or optionally substituted C10-40 alkynyl; and optionally one or more methylene groups of R5 are replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(RN), O, S, C(O), C(O)N(RN), - NRNC(O), NRNC(O)N(RN), C(O)O, OC(O), OC(O)O, OC(O)N(RN), NRNC(O)O, C(O)S, -SC(O), C(=NRN), C(=NRN)N(RN), NRNC(=NRN), NRNC(=NRN)N(RN), C(S), C(S)N(RN), -NRNC(S), NRNC(S)N(RN), S(O), OS(O), S(O)O, OS(O)O, OS(O)2, S(O)2O, OS(O)2O, -N(RN)S(O), S(O)N(RN), N(RN)S(O)N(RN), OS(O)N(RN), N(RN)S(O)O, S(O)2, N(RN)S(O)2, S(O)2N(RN), N(RN)S(O)2N(RN), OS(O)2N(RN), or N(RN)S(O)2O; and each instance of RN is independently hydrogen, optionally substituted alkyl, or a nitrogen protecting group. [0408] In certain embodiments, the compound of Formula (VI) is of Formula (VI-OH):
Figure imgf000132_0001
(VI-OH), or a salt thereof. In some embodiments, r is 45. [0409] In yet other embodiments the compound of Formula (VI) is:
Figure imgf000132_0002
salt thereof. [0410] In some embodiments, the compound of Formula (VI) is
Figure imgf000132_0003
(Compound I). [0411] In some aspects, the lipid composition of the pharmaceutical compositions disclosed herein does not comprise a PEG-lipid. [0412] In some embodiments, the PEG-lipids may be one or more of the PEG lipids described in PCT/US2018/037922 (published as WO 2018232357). [0413] In some embodiments, a PEG lipid comprises a PEG-modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG-modified dialkylglycerol, and/or mixtures thereof. In some embodiments, the PEG-modified lipid is PEG-DMG, PEG-c-DOMG (also referred to as PEG-DOMG), PEG-DSG and/or PEG-DPG. [0414] In some embodiments, a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising DSPC, a structural lipid, and a PEG lipid comprising PEG-DMG. [0415] In some embodiments, a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising DSPC, a structural lipid, and a PEG lipid comprising a compound having Formula VI. [0416] In some embodiments, a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and the PEG lipid comprising a compound having Formula V. [0417] In some embodiments, a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and the PEG lipid comprising a compound having Formula VI. [0418] In some embodiments, a LNP comprises an ionizable cationic lipid of any of Formula I, II or III, a phospholipid comprising a compound having Formula IV, a structural lipid, and a PEG lipid comprising a compound having Formula V or VI. [0419] In some embodiments, a LNP comprises an ionizable cationic lipid of
Figure imgf000133_0001
, and a PEG lipid comprising Formula VI. [0420] In some embodiments, a LNP comprises an ionizable cationic lipid of
Figure imgf000133_0002
, and an alternative lipid comprising oleic acid. [0421] In some embodiments, a LNP comprises an ionizable cationic lipid of
Figure imgf000134_0001
, an alternative lipid comprising oleic acid, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VI. [0422] In some embodiments, a LNP comprises an ionizable cationic lipid of
Figure imgf000134_0002
a phospholipid comprising DOPE, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VI. [0423] In some embodiments, a LNP comprises an ionizable cationic lipid of
Figure imgf000134_0003
, a phospholipid comprising DOPE, a structural lipid comprising cholesterol, and a PEG lipid comprising a compound having Formula VII. [0424] In some embodiments, an ionizable lipid is one or more compounds such as Compound A1 shown below.
Figure imgf000135_0001
Compound A1. [0425] In some embodiments, a LNP comprises an N:P ratio of from about 2:1 to about 30:1. [0426] In some embodiments, a LNP comprises an N:P ratio of about 6:1. [0427] In some embodiments, a LNP comprises an N:P ratio of about 3:1. [0428] In some embodiments, a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of from about 10:1 to about 100:1. [0429] In some embodiments, a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of about 20:1. [0430] In some embodiments, a LNP comprises a wt/wt ratio of the ionizable cationic lipid component to the RNA of about 10:1. the disclosure has a mean diameter from about 50nm to about 150nm. [0431] In some embodiments, a LNP has a mean diameter from about 70nm to about 120nm. Exemplary Methods [0432] According to at least one aspect of the disclosure, a method of filtering a solution containing lipid nanoparticles is provided, the method comprising setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold. [0433] According to at least one aspect of the disclosure, the method includes stopping the filtration after the first period has elapsed, wherein the determination of the actual value occurs after the first period. [0434] According to at least one aspect of the disclosure, the method includes following adjusting of the at least one filtration control parameter, resuming filtration for a second period. [0435] According to at least one aspect of the disclosure, the filtration control parameter is a flow rate of the solution, an environmental temperature, a solution temperature, a pH of the solution or a salt concentration of the solution. [0436] According to at least one aspect of the disclosure, the filtration control parameter is a fluid pressure of the solution; optionally, the filtration control parameter is PCRITICAL. [0437] According to at least one aspect of the disclosure, the method includes introducing one or more additives to the solution. [0438] According to at least one aspect of the disclosure, in the foregoing method, the determination of the actual value occurs after the first period and prior to the second period. [0439] According to at least one aspect of the disclosure, the method further comprises monitoring a transmembrane pressure during the filtration. [0440] According to at least one aspect of the disclosure, the first layer is upstream of the second layer. [0441] According to at least one aspect of the disclosure, the first pore size is 0.8 µm and the second pore size is 0.2 µm. [0442] According to at least one aspect of the disclosure, a non-transitory computer readable medium is provided which is configured to store instructions which, when executed by a processor, cause a controller to adjust the at least one filtration control parameter according to the techniques herein. [0443] According to at least one aspect of the disclosure, a method of evaluating sterile filtration of liquid nanoparticles (LNPs) is provided, the method comprising filtering a first solution lacking LNPs through at least one membrane while controlling at least one filtration control parameter to satisfy a control parameter threshold; determining a first hydraulic resistance of the membrane following filtration of the first solution; filtering a second solution containing LNPs through the at least one membrane while controlling the at least one filtration control parameter to satisfy the threshold; determining a second hydraulic resistance of the membrane following filtration of the second solution; computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; and performing microscopic imaging of the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling of the at least one membrane. [0444] According to at least one aspect of the disclosure, the at least one filtration control parameter is a fluid pressure of the solution. [0445] According to at least one aspect of the disclosure, the method includes monitoring a transmembrane pressure during filtering of at least the second solution. Definitions [0446] As will be understood by one of skill in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. [0447] It should be noted that the term “example” as used herein to describe various embodiments is intended to indicate that such embodiments are possible examples, representations, and/or illustrations of possible embodiments. [0448] As utilized herein, the term “substantially” and similar terms are intended to have a broad meaning in harmony with the common and accepted usage by those of ordinary skill in the art to which the subject matter of this disclosure pertains. It should be understood by those of skill in the art who review this disclosure that these terms are intended to allow a description of certain features described and claimed without restricting the scope of these features to the precise numerical ranges provided. Accordingly, these terms should be interpreted as indicating that insubstantial or inconsequential modifications or alterations of the subject matter described and claimed are considered to be within the scope of the invention as recited in the appended claims. [0449] The terms “coupled,” “connected,” and the like as used herein mean the joining of two members directly or indirectly to one another. Such joining may be stationary (e.g., permanent) or moveable (e.g., removable or releasable). [0450] Although the figures and description may illustrate a specific order of method steps, the order of such steps may differ from what is depicted and described, unless specified differently above. Also, two or more steps may be performed concurrently or with partial concurrence, unless specified differently above. All such variations are within the scope of the disclosure. [0451] It is important to note that the construction and arrangement of the various exemplary embodiments are illustrative only. Those skilled in the art who review this disclosure will readily appreciate that modifications are possible (e.g., variations in sizes or dimensions, the values of parameters, etc.) without materially departing from the teachings of the subject matter described herein. Other substitutions, modifications, changes and omissions may also be made to the various exemplary embodiments without departing from the scope of the embodiments described herein. [0452] While this specification contains implementation details, these should not be construed as limitations on the scope of any embodiment or of what may be claimed, but rather as descriptions of features specific to particular implementations of particular embodiments. Certain features described in this specification in the context of separate implementations can also be implemented in combination in a single implementation. Conversely, various features described in the context of a single implementation can also be implemented in multiple implementations separately or in any suitable subcombination. [0453] Moreover, although features may be described above as acting in certain combinations and even initially claimed as such, one or more features from a claimed combination can in some cases be excised from the combination, and the claimed combination may be directed to a subcombination or variation of a subcombination.

Claims

WHAT IS CLAIMED IS: 1. A method of evaluating sterile filtration of liquid nanoparticles (LNPs), the method comprising: filtering a first solution lacking LNPs through at least one membrane while controlling at least one filtration control parameter to satisfy a control parameter threshold; determining a first hydraulic resistance of the membrane following filtration of the first solution; filtering a second solution containing LNPs through the at least one membrane while controlling the at least one filtration control parameter to satisfy the threshold; determining a second hydraulic resistance of the membrane following filtration of the second solution; computing a difference between the first hydraulic resistance and the second hydraulic resistance to determine an extent of fouling of the at least one membrane; and performing microscopic imaging of the at least one membrane following filtration with the second solution to produce at least one image thereof, and characterizing the second solution based on the at least one image and the extent of fouling of the at least one membrane. 2. The method according to claim 1, wherein the at least one filtration control parameter is a fluid pressure of the solution. 3. The method according to claim 1, further comprising monitoring a transmembrane pressure during filtering of at least the second solution. 4. A non-transitory computer readable medium configured to store instructions which, when executed by a processor, cause a controller to adjust the at least one filtration control parameter according to claim 1. 5. The method of claim 1, wherein performing microscopic imaging includes performing at least one of scanning electron microscopy or atomic force microscopy. 6. A method of filtering a solution containing lipid nanoparticles, the method comprising: setting at least one filtration control parameter to at least one target filtration control parameter; performing filtration of the solution for a first period in a multi-layer filter comprising at least a first layer having a first pore size and at least a second layer having a second pore size smaller than the first pore size; optionally determining, during or after the first period, an actual value of the at least one filtration control parameter; and optionally adjusting the at least one filtration control parameter in response to determining that the actual value of the at least one control parameter differs from the at least one target filtration control parameter by at least a predetermined threshold. 7. The method of filtering the solution according to claim 6 further comprising stopping the filtration after the first period has elapsed, wherein the determination of the actual value occurs after the first period. 8. The method according to claim 6, further comprising, following adjusting of the at least one filtration control parameter, resuming filtration for a second period. 9. The method according to claim 8, wherein the determination occurs after the first period and prior to the second period. 10. The method according to any of claims 6-9, wherein the at least one filtration control parameter is a pH of the solution. 11. The method according to any of claims 6-9, wherein the at least one filtration control parameter is a salt concentration of the solution. 12. The method according to any of claims 6-9, further comprising introducing one or more additives to the solution. 13. The method according to claim 6, wherein the first layer is upstream of the second layer. 14. The method according to claim 6 or claim 13, wherein the first pore size is 0.8 µm and the second pore size is 0.2 µm. 15. The method according to claim 6, wherein the at least one filtration control parameter is an environmental temperature.
PCT/US2023/015564 2022-03-18 2023-03-17 Sterile filtration of lipid nanoparticles and filtration analysis thereof for biological applications WO2023177904A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263321196P 2022-03-18 2022-03-18
US63/321,196 2022-03-18
US202263322562P 2022-03-22 2022-03-22
US63/322,562 2022-03-22
US202263335816P 2022-04-28 2022-04-28
US63/335,816 2022-04-28

Publications (1)

Publication Number Publication Date
WO2023177904A1 true WO2023177904A1 (en) 2023-09-21

Family

ID=85979546

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/015564 WO2023177904A1 (en) 2022-03-18 2023-03-17 Sterile filtration of lipid nanoparticles and filtration analysis thereof for biological applications

Country Status (1)

Country Link
WO (1) WO2023177904A1 (en)

Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040262223A1 (en) 2001-07-27 2004-12-30 President And Fellows Of Harvard College Laminar mixing apparatus and methods
US20050222064A1 (en) 2002-02-20 2005-10-06 Sirna Therapeutics, Inc. Polycationic compositions for cellular delivery of polynucleotides
US7404969B2 (en) 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
WO2008103276A2 (en) 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
WO2011127255A1 (en) 2010-04-08 2011-10-13 Merck Sharp & Dohme Corp. Preparation of lipid nanoparticles
US8158601B2 (en) 2009-06-10 2012-04-17 Alnylam Pharmaceuticals, Inc. Lipid formulation
WO2012099755A1 (en) 2011-01-11 2012-07-26 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US8241670B2 (en) 2004-04-15 2012-08-14 Chiasma Inc. Compositions capable of facilitating penetration across a biological barrier
WO2012109121A1 (en) 2011-02-07 2012-08-16 Purdue Research Foundation Carbohydrate nanoparticles for prolonged efficacy of antimicrobial peptide
US20120276209A1 (en) 2009-11-04 2012-11-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
WO2012170889A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Cleavable lipids
WO2012170930A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc Lipid nanoparticle compositions and methods for mrna delivery
US20130059360A1 (en) 2005-04-12 2013-03-07 Nektar Therapeutics Polymer-based compositions and conjugates of antimicrobial agents
WO2013033438A2 (en) 2011-08-31 2013-03-07 Mallinckrodt Llc Nanoparticle peg modification with h-phosphonates
US20130072709A1 (en) 2006-02-21 2013-03-21 Nektar Therapeutics Segmented Degradable Polymers and Conjugates Made Therefrom
WO2013082111A2 (en) 2011-11-29 2013-06-06 The University Of North Carolina At Chapel Hill Geometrically engineered particles and methods for modulating macrophage or immune responses
WO2013086354A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US20130171646A1 (en) 2010-08-09 2013-07-04 So Jung PARK Nanop article-oligonucleotide hybrid structures and methods of use thereof
US20130172406A1 (en) 2007-09-28 2013-07-04 Bind Biosciences, Inc. Cancer Cell Targeting Using Nanoparticles
US20130183244A1 (en) 2010-09-10 2013-07-18 The Johns Hopkins University Rapid Diffusion of Large Polymeric Nanoparticles in the Mammalian Brain
WO2013110028A1 (en) 2012-01-19 2013-07-25 The Johns Hopkins University Nanoparticle formulations with enhanced mucosal penetration
US20130196948A1 (en) 2010-06-25 2013-08-01 Massachusetts Insitute Of Technology Polymers for biomaterials and therapeutics
WO2013116126A1 (en) 2012-02-01 2013-08-08 Merck Sharp & Dohme Corp. Novel low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
WO2013123523A1 (en) 2012-02-19 2013-08-22 Nvigen, Inc. Uses of porous nanostructure in delivery
WO2013151668A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of secreted proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
US20150036773A1 (en) 2010-04-01 2015-02-05 Ismail Lakkis Cancellation System for Millimeter-Wave Radar
WO2015051173A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc Polynucleotide molecules and uses thereof
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
WO2015074085A1 (en) 2013-11-18 2015-05-21 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
WO2015089511A2 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2015130584A2 (en) 2014-02-25 2015-09-03 Merck Sharp & Dohme Corp. Lipid nanoparticle vaccine adjuvants and antigen delivery systems
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016011306A2 (en) 2014-07-17 2016-01-21 Moderna Therapeutics, Inc. Terminal modifications of polynucleotides
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2016011222A2 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Circular polynucleotides
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2016100812A1 (en) 2014-12-19 2016-06-23 Moderna Therapeutics, Inc. Terminal modifications of polynucleotides
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049286A1 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a morpholino linker
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018191657A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
WO2018200943A1 (en) 2017-04-28 2018-11-01 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018232357A1 (en) 2017-06-15 2018-12-20 Modernatx, Inc. Rna formulations
WO2019036030A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036028A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036008A1 (en) 2017-08-16 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036000A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
CN110433667A (en) * 2019-09-02 2019-11-12 天津大学 Antipollution ant-scaling seperation film and preparation method thereof
WO2020081938A1 (en) 2018-10-18 2020-04-23 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020146805A1 (en) 2019-01-11 2020-07-16 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020214946A1 (en) 2019-04-18 2020-10-22 Translate Bio, Inc. Cystine cationic lipids
US20210378980A1 (en) * 2018-09-20 2021-12-09 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof

Patent Citations (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040262223A1 (en) 2001-07-27 2004-12-30 President And Fellows Of Harvard College Laminar mixing apparatus and methods
US20050222064A1 (en) 2002-02-20 2005-10-06 Sirna Therapeutics, Inc. Polycationic compositions for cellular delivery of polynucleotides
US8241670B2 (en) 2004-04-15 2012-08-14 Chiasma Inc. Compositions capable of facilitating penetration across a biological barrier
US7404969B2 (en) 2005-02-14 2008-07-29 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20130059360A1 (en) 2005-04-12 2013-03-07 Nektar Therapeutics Polymer-based compositions and conjugates of antimicrobial agents
US20130072709A1 (en) 2006-02-21 2013-03-21 Nektar Therapeutics Segmented Degradable Polymers and Conjugates Made Therefrom
WO2008103276A2 (en) 2007-02-16 2008-08-28 Merck & Co., Inc. Compositions and methods for potentiated activity of biologicaly active molecules
US20130172406A1 (en) 2007-09-28 2013-07-04 Bind Biosciences, Inc. Cancer Cell Targeting Using Nanoparticles
US8158601B2 (en) 2009-06-10 2012-04-17 Alnylam Pharmaceuticals, Inc. Lipid formulation
US20120276209A1 (en) 2009-11-04 2012-11-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
US20150036773A1 (en) 2010-04-01 2015-02-05 Ismail Lakkis Cancellation System for Millimeter-Wave Radar
WO2011127255A1 (en) 2010-04-08 2011-10-13 Merck Sharp & Dohme Corp. Preparation of lipid nanoparticles
US20130196948A1 (en) 2010-06-25 2013-08-01 Massachusetts Insitute Of Technology Polymers for biomaterials and therapeutics
US20130171646A1 (en) 2010-08-09 2013-07-04 So Jung PARK Nanop article-oligonucleotide hybrid structures and methods of use thereof
US20130183244A1 (en) 2010-09-10 2013-07-18 The Johns Hopkins University Rapid Diffusion of Large Polymeric Nanoparticles in the Mammalian Brain
WO2012099755A1 (en) 2011-01-11 2012-07-26 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
WO2012109121A1 (en) 2011-02-07 2012-08-16 Purdue Research Foundation Carbohydrate nanoparticles for prolonged efficacy of antimicrobial peptide
WO2012170930A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc Lipid nanoparticle compositions and methods for mrna delivery
WO2012170889A1 (en) 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Cleavable lipids
WO2013033438A2 (en) 2011-08-31 2013-03-07 Mallinckrodt Llc Nanoparticle peg modification with h-phosphonates
WO2013082111A2 (en) 2011-11-29 2013-06-06 The University Of North Carolina At Chapel Hill Geometrically engineered particles and methods for modulating macrophage or immune responses
WO2013086354A1 (en) 2011-12-07 2013-06-13 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2013110028A1 (en) 2012-01-19 2013-07-25 The Johns Hopkins University Nanoparticle formulations with enhanced mucosal penetration
WO2013116126A1 (en) 2012-02-01 2013-08-08 Merck Sharp & Dohme Corp. Novel low molecular weight, biodegradable cationic lipids for oligonucleotide delivery
WO2013123523A1 (en) 2012-02-19 2013-08-22 Nvigen, Inc. Uses of porous nanostructure in delivery
WO2013151665A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of proteins associated with human disease
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151664A1 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of proteins
WO2013151669A1 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
WO2013151667A1 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides
WO2013151671A1 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of cosmetic proteins and peptides
WO2013151663A1 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of membrane proteins
WO2013151670A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of nuclear proteins
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151672A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of oncology-related proteins and peptides
WO2013151668A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of secreted proteins
WO2015051173A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc Polynucleotide molecules and uses thereof
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
WO2015074085A1 (en) 2013-11-18 2015-05-21 Arcturus Therapeutics, Inc. Ionizable cationic lipid for rna delivery
WO2015089511A2 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2015130584A2 (en) 2014-02-25 2015-09-03 Merck Sharp & Dohme Corp. Lipid nanoparticle vaccine adjuvants and antigen delivery systems
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015199952A1 (en) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2016011222A2 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Circular polynucleotides
WO2016011306A2 (en) 2014-07-17 2016-01-21 Moderna Therapeutics, Inc. Terminal modifications of polynucleotides
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2016100812A1 (en) 2014-12-19 2016-06-23 Moderna Therapeutics, Inc. Terminal modifications of polynucleotides
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017049286A1 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a morpholino linker
WO2017075531A1 (en) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2017117528A1 (en) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018191657A1 (en) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
WO2018200943A1 (en) 2017-04-28 2018-11-01 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
WO2018232357A1 (en) 2017-06-15 2018-12-20 Modernatx, Inc. Rna formulations
WO2019036008A1 (en) 2017-08-16 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036030A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036028A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
WO2019036000A1 (en) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipids for use in lipid nanoparticle formulations
US20210378980A1 (en) * 2018-09-20 2021-12-09 Modernatx, Inc. Preparation of lipid nanoparticles and methods of administration thereof
WO2020081938A1 (en) 2018-10-18 2020-04-23 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020146805A1 (en) 2019-01-11 2020-07-16 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
WO2020214946A1 (en) 2019-04-18 2020-10-22 Translate Bio, Inc. Cystine cationic lipids
CN110433667A (en) * 2019-09-02 2019-11-12 天津大学 Antipollution ant-scaling seperation film and preparation method thereof

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
ABRAHAM ET AL.: "Chaotic Mixer for Microchannels", SCIENCE, vol. 295, 2002, pages 647 - 651
BELLIVEAU ET AL.: "Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA", MOLECULAR THERAPY-NUCLEIC ACIDS, vol. 1, 2012, pages e37, XP002715253, DOI: 10.1038/mtna.2012.28
BRADER ET AL., BIOPHYSICAL JOURNAL, vol. 120, 2021, pages 1 - 5
BUSCHMANN, M. D. ET AL., VACCINES, vol. 9, 2021, pages 65
CHEN ET AL.: "Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation", J. AM. CHEM. SOC., vol. 134, no. 16, 2012, pages 6948 - 51, XP002715254, DOI: 10.1021/ja301621z
JOHNSON THOMAS F. ET AL: "Liposome Sterile Filtration Characterization via X-ray Computed Tomography and Confocal Microscopy", MEMBRANES, vol. 11, no. 11, 22 November 2021 (2021-11-22), pages 905, XP093050417, DOI: 10.3390/membranes11110905 *
MENDRET J ET AL: "In situ dynamic characterisation of fouling under different pressure conditions during dead-end filtration: Compressibility properties of particle cakes", JOURNAL OF MEMBRANE SCIENCE, ELSEVIER BV, NL, vol. 333, no. 1-2, 5 February 2009 (2009-02-05), pages 20 - 29, XP026031525, ISSN: 0376-7388, [retrieved on 20090205], DOI: 10.1016/J.MEMSCI.2009.01.035 *
MESSERIAN KEVORK OLIVER ET AL: "Pressure-dependent fouling behavior during sterile filtration of mRNA-containing lipid nanoparticles", BIOTECHNOLOGY AND BIOENGINEERING, vol. 119, no. 11, 8 August 2022 (2022-08-08), Hoboken, USA, pages 3221 - 3229, XP093050365, ISSN: 0006-3592, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/bit.28200> [retrieved on 20230531], DOI: 10.1002/bit.28200 *
MESSERIAN KEVORK OLIVER ET AL: "Sterile filtration of lipid nanoparticles for mRNA vaccines and therapeutics", 21 March 2022 (2022-03-21), pages 1 - 2, XP093050566, Retrieved from the Internet <URL:https://acs.digitellinc.com/sessions/414716/view> [retrieved on 20230523] *
RODRIGUEZ ET AL., SCIENCE, vol. 339, 2013, pages 971 - 975
WHITESIDES, GEORGE M.: "The Origins and the Future of Microfluidics", NATURE, vol. 442, 2006, pages 368 - 373, XP055123139, DOI: 10.1038/nature05058
ZHIGALTSEVET: "Bottom-up design and synthesis of limit size lipid nanoparticle systems with aqueous and triglyceride cores using millisecond microfluidic mixing", LANGMUIR, vol. 28, 2012, pages 3633 - 40, XP055150435, DOI: 10.1021/la204833h
ZOURNA KALLIOPI ET AL: "Optimizing the Filtration of Liposomes Using Sterilizing-Grade Filters", PDA JOURNAL OF PHARMACEUTICAL SCIENCE AND TECHNOLOGY, vol. 75, no. 2, 30 September 2020 (2020-09-30), US, pages 128 - 140, XP093050501, ISSN: 1079-7440, Retrieved from the Internet <URL:http://dx.doi.org/10.5731/pdajpst.2020.011866> [retrieved on 20230523], DOI: 10.5731/pdajpst.2020.011866 *

Similar Documents

Publication Publication Date Title
JP7091393B2 (en) Method of Encapsulating Nucleic Acid in Lipid Nanoparticle Host
US20230235359A1 (en) Nucleic acid-containing lipid particles and related methods
JP7086870B2 (en) Compositions and Methods for Delivering Messenger RNA
JP5080779B2 (en) Method for producing liposome preparation
CN116549671A (en) Improved ICE-based lipid nanoparticle formulations for delivery of MRNA
JP5639338B2 (en) Liposome production system and production method
JP2014534232A (en) Single use system for aseptically producing lipid-nucleic acid particles
JP2010184947A (en) Method and apparatus for producing liposome
JP2016515815A (en) Lipid nanoparticles and related methods for transfection
ES2832330T3 (en) Vector for pulmonary delivery, inducing agent and uses
CN103585107A (en) Composition for the manufacture of a therapeutic agent for cancer or inflammatory disease
DK2774991T3 (en) Drug administration system for use in the treatment or diagnosis of neurological disorders
EP2774991B1 (en) Drug delivery system for use in the treatment or diagnosis of neurological disorders
WO2023177904A1 (en) Sterile filtration of lipid nanoparticles and filtration analysis thereof for biological applications
RU2297827C2 (en) Method and the device for carrying out vesicle extrusion under high pressure
US20140221461A1 (en) Nucleic acid molecules and uses thereof
EP1970078A1 (en) Composition inhibiting the expression of target gene in eyeball and remedy for disease in eyeball
CN111565833B (en) Development of method and apparatus for producing lipid particles having desired particle diameter
WO2010110318A1 (en) Therapeutic agent for arteriosclerotic diseases comprising nucleic acid
WO2023193002A1 (en) Cross mixers for lipid nanoparticle production, and methods of operating the same
Kennison et al. Coupling of Leaflet Phase Behavior in Asymmetric Giant Unilamellar Vesicles Prepared by Hemifusion: Investigating the Influence of the Low-Metling Lipid
Gilbert et al. Lipid sponge phase as a matrix for enzyme encapsulation: structure and dynamics
Enoki et al. Partition Coefficient of a Transmembrane Peptide, between Lo and Ld Phases: Does the Peptide Distinguish Macro from Nano Domains?

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: 23716052

Country of ref document: EP

Kind code of ref document: A1