WO2022051247A1 - Methods for treating or preventing inflammatory events - Google Patents

Methods for treating or preventing inflammatory events Download PDF

Info

Publication number
WO2022051247A1
WO2022051247A1 PCT/US2021/048352 US2021048352W WO2022051247A1 WO 2022051247 A1 WO2022051247 A1 WO 2022051247A1 US 2021048352 W US2021048352 W US 2021048352W WO 2022051247 A1 WO2022051247 A1 WO 2022051247A1
Authority
WO
WIPO (PCT)
Prior art keywords
inflammation
subject
mcm
formulation
mineral
Prior art date
Application number
PCT/US2021/048352
Other languages
French (fr)
Inventor
Anna B. CLEMENTS
Jae Sung Lee
William Murphy
Martin Ostrowski
Original Assignee
Dianomi Therapeutics, Inc.
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 Dianomi Therapeutics, Inc. filed Critical Dianomi Therapeutics, Inc.
Publication of WO2022051247A1 publication Critical patent/WO2022051247A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2006IL-1
    • 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
    • A61K9/5005Wall or coating material
    • A61K9/501Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • ARDS acute respiratory distress syndrome
  • CRS cytokine release syndrome
  • hypercytokinemia cytokine storm
  • CRS cytokine release syndrome
  • hypercytokinemia cytokine storm
  • ARDS is one of the leading causes of death in patients infected with COVID-19, and the number of ARDS deaths and hospitalizations in the US has been increasing significantly.
  • COVID-19 Prior to the emergence of COVID-19, ARDS accounted for 75,000 deaths, 3.6 million hospital days, and $5 billion in healthcare costs in the U.S. annually.
  • inflammation can be triggered by transplant (e.g., graft versus host disease (GvHD)) or CAR-T therapy.
  • transplant e.g., graft versus host disease (GvHD)
  • CAR-T therapy e.g., CAR-T therapy
  • GvHD graft versus host disease
  • CARs chimeric antigen receptors
  • clinical manifestations of CRS can develop within the first days from CAR-T cell infusion and can include high fever, increased levels of acute phase proteins, respiratory and cardiovascular insufficiency, which if severe and left untreated may lead to death.
  • gout an inflammatory arthritic disease affecting millions of people in the U.S with significant economic burden.
  • gout sufferers can have frequent, excruciating, and protracted attacks as well as permanent joint damage.
  • Gout develops when excessive levels of uric acid accumulate in tissue and form crystals which deposit in various joints causing excruciating pain and inflammation. These crystals stimulate neutrophil ingress into the joint and activate synovial tissue macrophages to produce mediators of inflammation, including, interleukin (IL)- 1 P, tumor necrosis factor (TNF)-a, and IL-6.
  • IL interleukin
  • TNF tumor necrosis factor
  • IL-6 tumor necrosis factor
  • ARDS acute respiratory distress syndrome
  • CRS cytokine release syndrome
  • GVHD graft versus host disease
  • sepsis inflammation caused by chimeric antibody receptor T-cell (CAR-T) therapy or inflammation associated with gout.
  • CAR-T chimeric antibody receptor T-cell
  • the treatment or prevention involves administering an admixture of interleukin- 1 receptor agonist (ILl-Ra) or its equivalents, including anakinra along with a mineral coated microparticle (MCM) to a subject in need thereof.
  • ILl-Ra interleukin- 1 receptor agonist
  • MCM mineral coated microparticle
  • MCM mineral-coated microparticle
  • An MCM is a biomimetic, tailorable, mineral coated microparticle.
  • MCMs can bind (e.g., adsorb), stabilize, and release proteins, peptides and nucleic acid molecules.
  • MCMs can be used in combination with IL-IRa for prevention and/or treatment of inflammation.
  • MCMs are able to maintain activity, stabilize structure, and provide superior loading capacity compared to current therapeutic protein delivery platforms.
  • intraarticular delivery of anakinra using MCMs can sustain biologically active protein release, thereby extending the time between treatments.
  • MCMs can utilize calcium phosphate mineral (CaP) coatings to bind and release therapeutic proteins in a sustained and controllable manner.
  • CaP mineral coatings can be precipitated onto core materials (such as, for example, beta-tri calcium phosphate (P-TCP) particles, hydroxyapatite particles, or polymer microspheres) by incubating (e.g., for a period of 5-10 days) in modified simulated body fluid (mSBF), a calcium phosphate solution similar to human blood plasma.
  • P-TCP beta-tri calcium phosphate
  • mSBF modified simulated body fluid
  • Proteins can bind to the CaP mineral surface through electrostatic interactions between the polar and charged groups of the proteins and the positively (Ca2+) and negatively (PO4 3- ) charged ions in the CaP mineral.
  • the mineral surface can serve as a platform for binding and stabilizing proteins to the coating by maintaining the protein’s conformational structure. Moreover, because of the plate-like structure of the mineral coatings, significantly greater loading capacity of protein is possible compared to other delivery platforms. Release and delivery of therapeutic proteins from the MCMs can occur via dissolution of the CaP mineral coatings. As MCMs are inorganic components, they also do not elicit unwanted immunogenic responses.
  • MCMs can be constructed of generally regarded as safe (GRAS) materials. They can also be added to an existing formulation and optionally lyophilized to create a product that is stable at room temperature, can be stockpiled, and can be distributed without need for refrigeration. The lyophilized composition can be reconstituted and used at the point of administration.
  • GRAS safe
  • a method for treating inflammation can include (a) providing a formulation comprising a biologic; and (b) admixing the formulation with a mineral coated microparticle (MCM) to provide an admixed formulation, wherein the MCM adsorbs the biologic and provides a sustained delivery of the biologic when administered in vivo, wherein the inflammation is exemplified by an increase in one or more members of the IL-1 family of cytokines or IL-6 production, and wherein the admixed formulation is administered to a subject in need thereof.
  • MCM mineral coated microparticle
  • the inflammation is acute respiratory distress syndrome (ARDS).
  • ARDS acute respiratory distress syndrome
  • the inflammation is associated with graft versus host disease (GVHD).
  • the inflammation is associated with infection by the SARS-CoV-2 virus.
  • the inflammation is associated with cytokine storm syndrome.
  • the inflammation is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
  • CAR-T chimeric antigen receptor T-cell
  • the inflammation is associated with gout.
  • the inflammation is associated with urate crystal formation and/or presence.
  • the inflammation is acute inflammation.
  • the biologic is interleukin-1 receptor agonist (IL-IRa), or an analog thereof.
  • IL-IRa interleukin-1 receptor agonist
  • the biologic is anakinra or a derivative thereof.
  • an amount of biologic administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
  • the admixed formulation is provided prophylactically to a subject in need thereof.
  • a method for reducing the expression of IL-1 or IL-6 can include administering a single dose of a formulation to a patient exhibiting elevated serum concentrations of IL-1 or IL-6 associated with an acute inflammatory event, wherein the formulation comprises a sustained release admixture, wherein the sustained release admixture comprises a drug delivery vehicle and IL-lRa.
  • the elevated serum concentration of IL-1 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 in vivo.
  • the elevated serum concentration of IL-6 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-6 in vivo.
  • the elevated serum concentrations of IL-1 and IL-6 are elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 and IL-6 in vivo.
  • no more than one administration of the formulation is performed in a 24 hour, 48 hour, or 72 hour period.
  • the method further comprises administering a subsequent dose of the formulation after the single administration of the formulation.
  • the elevated serum concentration of IL-1 or IL-6 is at least about 2, at least about 5, at least about 10, at least about 20, or at least about 50-times higher than a patient not having the acute inflammatory event.
  • the expression of IL-1 or IL-6 resulting from the single administration is reduced compared to a single administration of IL-lRa alone.
  • the expression of IL-1 or IL-6 is reduced by at least about 2, 5, 10, 20, or 50-fold.
  • the expression of IL-1 or IL-6 lasts over a period of 3, 5, or 7 days in vivo.
  • the acute inflammatory event is acute respiratory distress syndrome (ARDS).
  • ARDS acute respiratory distress syndrome
  • the acute inflammatory event is a gout flare.
  • the acute inflammatory event is graft versus host disease (GVHD).
  • the acute inflammatory event is associated with infection by the SARS-CoV-2 virus or influenza virus. [0038] In some embodiments, the acute inflammatory event is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
  • CAR-T chimeric antigen receptor T-cell
  • the drug delivery vehicle is a mineral coated microparticle (MCM).
  • an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
  • a method for preventing inflammation can comprise administering an admixture of IL-IRa and a mineral coated microparticle (MCM) to a subject having an elevated risk of inflammation.
  • MCM mineral coated microparticle
  • the subject exhibits or is at risk of a pulmonary edema.
  • the subject requires a high fraction of inspired oxygen (FiCh).
  • the subject has been infected with the SARS-Cov-2 virus or influenza virus.
  • the subject has received a transplant.
  • the subject has undergone chimeric antigen receptor T-cell (CAR- T) therapy.
  • CAR- T chimeric antigen receptor T-cell
  • the subject is at risk of inflammation of immune cells, lymphocytes, or mast cells.
  • the subject is at risk of respiratory or cardiovascular inflammation.
  • the subject is showing symptoms of inflammation.
  • the subject is at an onset of inflammation.
  • the subject is exhibiting hyperoxia.
  • the subject has elevated serum concentrations of IL-1 or IL-6.
  • no more than one administration of the admixture is performed in a 24 hour, 48 hour, or 72 hour period.
  • the method further comprises administering a subsequent dose of the admixture after the single administration of the admixture.
  • the MCM provides for sustained release of the IL-IRa.
  • an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
  • FIG. 1 shows a scanning electron micrograph (SEM) image of an example MCM
  • FIG. 2 shows an example method for fabricating an MCM and binding protein to the surface of the MCM
  • FIG. 3A shows a conceptual graphic of example repeated bolus delivery of proteins not delivered by MCMs
  • FIG. 3B shows a conceptual graphic of a single sustained release delivery of proteins from an example MCM
  • FIG. 4 shows an example benefit of MCM binding on the effect of anakinra in vivo.
  • a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in some embodiments, to A only (optionally including elements other than B); in some embodiments, to B only (optionally including elements other than A); in some embodiments, to both A and B (optionally including other elements); etc.
  • the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in some embodiments, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in some embodiments, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in some embodiments, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
  • the term "about” or “approximately” as used herein means within an acceptable error range for the particular value as determined, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system.
  • "about” can mean within 3 or more than 3 standard deviations, per the practice in the art. In certain embodiments, such as with respect to biological systems or processes, the term can mean within an order of magnitude, including within 5 -fold, and within 2-fold of a value. In certain embodiments, when the term “about” or “approximately” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below those numerical values. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20%, 10%, 5%, or 1%. In certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 10%.
  • the term “about” is used to modify a numerical value above and below the stated value by a variance of 5%. In certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 1%.
  • administering refers to the placement of the dose as disclosed herein into a subject by a method or route which results in at least partial delivery of the composition at an appropriate extracellular location of a target tissue.
  • the dose adsorbed to the MCM component can be, for example, injected into a subject in need thereof by either intradermal, intra-muscular, subcutaneous, intra-articular, periarticular or intravenous administration.
  • the protein dose adsorbed to the MCM component administered parenterally, e.g., by intravenous, intra-arterial, intracardiac, intraspinal, intraosseous, intra-articular, intra-synovial, subcutaneous, intradermal, intra- tendinous, intraligamentous or intramuscular administration.
  • the bioactive compound captured within the inorganic precipitate is administered by implantation, infiltration or infusion.
  • the therapeutically effective amount can vary depending upon the intended application or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined such as by a board-certified physician.
  • the terms “treat,” “treatment,” “treating” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a disorder.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced.
  • treatment is “effective” if the progression of a disorder is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms in the absence of treatment.
  • Beneficial clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (e.g., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
  • tissue refers to an aggregation of morphologically similar cells and associated intercellular matter, e.g., extracellular matrix, acting together to perform one or more specific functions in the body.
  • tissues fall into one of four basic types: muscle, nerve, epidermal, and connective.
  • a tissue is substantially solid, e.g., cells within the tissue are strongly associated with one another to form a multicellular solid tissue.
  • a tissue is substantially non-solid, e.g., cells within the tissue are loosely associated with one another, or not at all physically associated with one another, but may be found in the same space, bodily fluid, etc.
  • extracellular means being situated or taking place outside a cell or cells.
  • a “subject” refers to a vertebrate, such as a mammal (e.g., a non-human mammal), such as a primate or a human. Mammals include, but are not limited to, primates, humans, farm animals, rodents, sport animals, and pets. [0079] As used herein, "bioavailability" is a fraction (%) of an administered drug that reaches the systemic circulation. By definition, when a medication is administered intravenously, its bioavailability is 100%. However, when a medication is administered via routes other than intravenous, its bioavailability can be lower than that of intravenous.
  • bioavailability equals the ratio of comparing the area under the plasma drug concentration curve versus time (AUC) for the extravascular formulation to the AUC for the intravascular formulation.
  • AUC area under the plasma drug concentration curve versus time
  • the bottom value of the deviation range is employed to represent real bioavailability to calculate drug dose for the drug taker to achieve systemic drug concentrations similar to the intravenous formulation.
  • the bottom value of the deviation range can be used in order to ensure the anticipated efficacy will be met unless the drug is associated with narrow therapeutic window. Bioavailability can be measured over any suitable period of time.
  • formulation generically indicates the beneficial agents and mineral coated microparticles are formulated, mixed, added, dissolved, suspended, solubilized, formulated into a solution, carried and/or the like in or by the fluid, gas, or solid in a physical-chemical form acceptable for patient administration.
  • Effective amount or “therapeutically effective amount” means a dosage sufficient to alleviate one or more symptoms of the condition being treated, or to otherwise provide a pharmacological and/or physiologic effect, as may be determined by an objective measure or a patient derived subjective measure.
  • an “effective amount” refers to the optimal amount of a dose adsorbed to the MCM needed to elicit a clinically significant improvement in the symptoms and/or pathological state associated with a disease state, infection, or disorder to be treated.
  • the disease state, infection, or disorder to be treated is a viral pathogen.
  • the dose adsorbed to the MCM is administered as a treatment.
  • the dose adsorbed to the MCM is administered prophylactically as a preventative measure.
  • an “effective amount”, a “therapeutically effective amount”, a “prophylactically effective amount” and a “diagnostically effective amount” is the amount of the unbound active agent and the active agent adsorbed to the mineral coated microparticle needed to elicit a biological response following administration.
  • a subject in need thereof' also used interchangeably herein with "a patient in need thereof' refers to a subject susceptible to or at risk of a specified disease, disorder, or condition.
  • the methods disclosed herein can be used with a subset of subjects who are susceptible to or at elevated risk of infection by a condition for which the treatment is provided. Because some of the method embodiments of the present disclosure are directed to specific subsets or subclasses of identified subjects (that is, the subset or subclass of subjects “in need” of assistance in addressing or vaccinating against one or more specific conditions noted herein), not all subjects will fall within the subset or subclass of subjects as described herein for certain diseases, disorders or conditions. "Elevated risk” can include any statistically relevant threshold when comparing one population of subjects to another.
  • IL-1 refers to the inflammatory members of the Interleukin- 1 family of cytokines. Such members include interleukin- la, interleukin- ip, interleukin- 18, interleukin- 36a, interleukin-36p, interleukin-36y, and interleukin-33.
  • Acute inflammatory events represent a significant unmet medical need. Examples include acute respiratory distress syndrome (ARDS), such as induced by SARS-CoV-2 infection (COVID-19) or influenza, cytokine release syndrome, complications arising from CAR-T therapy, sepsis, or graft-versus-host disease.
  • ARDS acute respiratory distress syndrome
  • COVID-19 SARS-CoV-2 infection
  • influenza cytokine release syndrome
  • CAR-T therapy sepsis
  • sepsis sepsis
  • graft-versus-host disease graft-versus-host disease
  • Inflammation and oxidative stress can play a role in the pathogenesis of ARDS and related conditions. Such conditions can be caused by or associated with many factors including viral infections, bacterial infections, sepsis, exposure to toxic substances, lung injury, mechanical ventilation, or high-oxygen environments.
  • coronavirus infections e.g., coronavirus infections
  • coronavirus infections can be characterized by elevated pro-inflammatory markers in the serum, evidence of monocyte/macrophage activation, activated coagulation and pro-inflammatory cytokine and chemokine profiles, implying that the host response is an important factor in this disease.
  • lung inflammation can intensify after viral clearance, peaking 1-2 weeks after infection in animal models and in human SARS-1 (severe acute respiratory syndrome-1) patients. This suggests that clinical deterioration later in the disease course can be due to damage from uncontrolled immune responses rather than uncontrolled viral replication. It also defines a window for therapy after presentation of most patients but before refractory ARDS is established.
  • the materials and methods described herein can address both lung inflammation and hyperoxic lung injury.
  • Interleukin-1 IL-1
  • IL-1R IL-1 receptor
  • the ILl-Ra as delivered from MCM binds to IL-1R without activating inflammation.
  • Hyperoxia often used to treat ARDS, but also worsens lung injury.
  • the methods described herein alleviates the need for hyperoxia treatment.
  • GvHD Graft-versus-host disease
  • GvHD can cause acute inflammation that can be treated or prevented using the methods described herein.
  • GvHD is a syndrome that can be characterized by inflammation in different organs, in some cases with the specificity of epithelial cell apoptosis and crypt drop out.
  • GvHD can be associated with stem cell transplants such as those that occur with bone marrow transplants.
  • GvHD can also apply to other forms of transplanted tissues such as solid organ transplants.
  • White blood cells of the donor's immune system which remain within the donated tissue (the graft) can recognize the recipient (the host) as foreign (non-self).
  • the white blood cells present within the transplanted tissue can then attack the recipient's body's cells, which can lead to GvHD.
  • Transplant rejection can occur when the immune system of the transplant recipient rejects the transplanted tissue
  • GvHD can occur when the donor's immune system's white blood cells reject the recipient.
  • the underlying principle is similar, but the details and course may differ.
  • GvHD can also occur after a blood transfusion if the blood products used have not been irradiated or treated with an approved pathogen reduction system.
  • CAR-T therapy can induce acute inflammatory events that are suitable for treatment or prevention using the materials and methods described herein.
  • CARs chimeric antigen receptors
  • the basic structure of CARs comprises a tumor-targeting domain, usually from the single-chain fragment variables (scFvs) of a monoclonal antibody (mAb), fused to at least one immune tyrosine activatory motif (ITAM), which can be the CD3 zeta chain, and one or more costimulatory endodomains.
  • scFvs single-chain fragment variables
  • mAb monoclonal antibody
  • ITAM immune tyrosine activatory motif
  • CD3 zeta chain CD3 zeta chain
  • Gout develops when excessive levels of uric acid accumulate in tissue and form crystals which deposit in various joints causing excruciating pain and inflammation. These crystals stimulate neutrophil ingress into the joint and activate synovial tissue macrophages to produce mediators of inflammation, including, interleukin (IL)-lp, tumor necrosis factor (TNF)- a, and IL-6. Without treatment, more frequent/protracted polyarthritis attacks occur, eventually resulting in permanent joint damage. IL-ip released from leukocytes may be a trigger for a cascade of inflammatory mediators and cytokines which are responsible for gout flares and subsequent damage.
  • IL interleukin
  • TNF tumor necrosis factor
  • Pro-inflammatory cytokines including IL- 1 p, TNF-a, IL-6 and IL-8, can be significantly elevated in bronchoalveolar lavage fluid and plasma of ARDS patients. Furthermore, plasma levels of IL-1 P, one of the most active pro-inflammatory cytokines, can be predictive of clinical outcomes in patients with severe ARDS. In some cases, large increases in the expressions of IL- ip, IL-6, TNF-a and integrin ligand ICAM-1 can be found in lung tissue exposed to hyperoxia.
  • Protection can be concomitant with a smaller increase in lung tissue expressions of IL-1 p, IL-6, TNF-a and ICAM-1 and better preservation of arterial blood pCh, consistent with a key role for inflammation in the pathogenesis of hyperoxia-induced ARDS.
  • Interleukin- la (IL-la) and interleukin- ip (IL-1 p) are prototypic proteins of the IL-1 superfamily that exert pleiotropic pro-inflammatory effects on many cell types throughout the body. Binding of IL-la or IL-ip to the IL-1 receptor type I (IL-1RI) can allow for the recruitment of the interleukin- 1 receptor accessory protein (IL-lRAcP), which can induce a biological response that can involve activation of the nuclear factor-KB (NF-KB) and mitogen- activated protein kinase (MAPK) pathways, both of which are potent inflammatory pathways. Additionally, IL-1 signaling has been shown to produce a positive feedback loop, which amplifies IL-1 expression.
  • IL-1RI interleukin- 1 receptor accessory protein
  • NF-KB nuclear factor-KB
  • MAPK mitogen- activated protein kinase
  • Anakinra the recombinant form of naturally occurring Interleukin- 1 receptor antagonist (IL-IRa), is a 17 kD anti-inflammatory protein which binds to the Interleukin-1 receptor (IL- 1RI) with an affinity similar to IL-1, but does not produce an inflammatory signal.
  • IL-IRa or equivalents (including anakinra) with MCMs can improve the anti-inflammatory activity and duration of effect of anakinra to develop a therapeutic for the prevention and treatment of ARDS.
  • Anakinra possesses properties that allow it to inhibit detrimental IL-1 activity, including its high binding affinity and target specificity with the IL-1 receptor, low toxicity, and low molecular weight (17kD) when compared to other potential antiinflammatory therapeutic proteins.
  • Anakinra can be used for treating inflammatory conditions including cytokine release syndrome (CRS), sepsis with features of macrophage activation syndrome (MAS), and acute respiratory distress syndrome (ARDS).
  • CRS cytokine release syndrome
  • MAS macrophage activation syndrome
  • ARDS acute respiratory distress syndrome
  • effective dosing for inflammatory events including COVID-19, can require twice daily intravenous (i.v.) administration of 5 mg/kg of anakinra, which can limit its practical use without improved delivery outside of, and even within, a hospital setting.
  • Anakinra has a very short in vivo half-life (20 minutes-4 hours) and the high dose required to inhibit IL-1 activity (10- 100-fold molar excess of IL-1).
  • anakinra can be quickly cleared from circulation because of enzymatic degradation, renal filtration, rapid distribution into organs, hepatic metabolism, and initial scavenging by receptors. Rapid clearance can reduce its efficacy for preventing inflammation, even when administered at frequent dosing intervals.
  • Some patients can require twice daily, hour-long intravenous (i.v.) infusions of gram quantities of anakinra for effective treatment for ARDS.
  • Anakinra can also be dosed as a daily subcutaneous injection for the treatment of some inflammatory conditions.
  • the need for daily administration of large quantities of protein can present a burdensome dosing regimen for patients who are unaccustomed to self-administered injections. Additionally, daily injections can cause injection site reactions.
  • MCMs are calcium phosphate based, micron-sized particles which bind, stabilize, and release proteins upon coating dissolution.
  • a SEM (scanning electron microscope) image of an example MCM can be seen in FIG. 1.
  • the nano structured, plate-like surface of the micron sized MCMs can provide a large surface for protein binding that also stabilizes the protein structure. These mineral coatings can stabilize protein structures and release biologically active proteins as they dissolve.
  • Proteins can bind to the surface of the MCM through electrostatic interactions between the polar and charged group of the protein and the calcium and phosphate groups of the mineral coating, similar to hydroxyapatite chromatography. Binding to the mineral coating can preserve the protein’s tertiary structure while simultaneously protecting the protein from degradation, even in harsh environments. Proteins can then be released from the MCM upon coating dissolution, providing sustained release and improved half-life. Because proteins are loaded onto the surface of MCMs after the MCMs are fabricated and sterilized, MCMs can be added to an already formulated therapeutic to improve its half-life, much like an excipient ingredient. This unique characteristic greatly simplifies the manufacturing process when compared to other sustained delivery technologies, such as polymer encapsulation or PEGylation, and is a key advantage of the MCM.
  • MCMs are simple and scalable.
  • P- tricalcium phosphate cores can be incubated in modified simulated body fluid (mSBF), a solution which contains the same ionic constituents of human blood plasma, and the mineral coating can precipitate onto the core material.
  • mSBF modified simulated body fluid
  • the MCMs can be freeze dried and sterilized using dry heat sterilization and depyrogenation techniques.
  • the resulting MCMs can have acceptable bioburden and endotoxin levels for injectable materials and are stable at room temperature.
  • a number of approaches can be taken when combining the MCM with formulated anakinra.
  • One example approach is to combine the MCMs with the formulated anakinra at the point of care, much like the approach that is taken with lyophilized therapeutics which are reconstituted prior to administration. This approach can take advantage of the stability of anakinra in the excipient buffer used for Kineret (an FDA approved anakinra product) and the stability of MCMs when they are freeze dried.
  • a second example approach is to add the MCMs to anakinra during formulation and packaging. A formulation with the components combined can be easier to administer in the field and in the clinic.
  • the MCM can stabilize macromolecules.
  • the diameter of the MCM can also be tailored for any suitable purpose. In some embodiments, the diameter of the MCM is about 1 micrometer (um), about 3 um, about 5 um, about 10 um, about 30 um, about 50 um, about 80 um, about 100 um, about 120 um, about 150 um, about 200 um, about 300 um, or about 500 um.
  • the diameter of the MCM is at least about 1 micrometer (um), at least about 3 um, at least about 5 um, at least about 10 um, at least about 30 um, at least about 50 um, at least about 80 um, at least about 100 um, at least about 120 um, at least about 150 um, at least about 200 um, at least about 300 um, or at least about 500 um.
  • the diameter of the MCM is at most about 1 micrometer (um), at most about 3 um, at most about 5 um, at most about 10 um, at most about 30 um, at most about 50 um, at most about 80 um, at most about 100 um, at most about 120 um, at most about 150 um, at most about 200 um, at most about 300 um, or at most about 500 um.
  • the diameter of the MCM can be tailored by using a larger or smaller core material or by the conditions of deposition of the mineral coating on the core material, which conditions can include time of reaction or concentration of components in the simulated body fluid solution.
  • the diameter of the core and MCM can be measured using laser diffraction. Laser diffraction can provide the Dv (10), Dv (50), and Dv (90) of the particles. An increase in the particle diameter after the coating process can be an indicator of the level of coating. Particle diameter can also have a large impact on particle performance, including dissolution kinetics, loading of active pharmaceutical ingredients, release rate of active pharmaceutical ingredients, the immune response to the particle after administration.
  • the starting core materials have a Dv (10), Dv (50) and Dv (90) of approximately 5 um, 15 um, and 30 um respectively.
  • suitable ranges for the Dv (10), Dv (50), and Dv(90) of uncoated materials can span from 0.01-10 um, 0.01-30 um, and 0.01 to 50 um respectively.
  • the coated MCMs have a Dv (10), Dv (50) and Dv (90) of approximately 15 um, 25 um, and 30 um respectively.
  • suitable ranges for the Dv (10), Dv (50), and Dv(90) of the coated microparticles can span from 0.1-20 um, 0.1-40 um, and 0.1 to 70 um respectively.
  • the coated MCMs have a Dv(10) that is approximately 3X larger than the Dv(10) of the starting core material. In some embodiments, the coated MCMs have a Dv(50) that is approximately 1.5X larger than the Dv(50) of the starting core material.
  • the diameter of the core material and the MCMs can also be measured using staining and visual assessment using imaging software.
  • the mean particle diameter of the core material is approximately 5 um. In some embodiments, the mean particle diameter of the coated MCM is 8 um to 10 um. In some embodiments, the mean particle diameter of the coated MCM is 5 um-8 um. In other embodiments, the mean particle diameter of the coated MCM is 1 um-5 um. In some embodiments, the mean particle diameter of the coated MCM is 8 um-20 um. In some embodiments, the mean diameter of the coated MCM is 2X the diameter of the starting core material. In some embodiments, the mean diameter of the coated MCM is greater than 2X the diameter of the starting core material.
  • the surface area of the microparticle can directly impact the loading capacity, the dissolution rate, and the release rate of the mineral coated microparticle. Additionally, coating of the starting core materials significantly increases the surface area of the particle. In some embodiments the surface area of the starting core material is approximately 1 to 3 m 2 /g. In some embodiments, the surface area of the starting core material is between 0.1 to 1 m 2 /g. In some embodiments, the surface area of the coated MCM is approximately 25 m 2 /g. In some embodiments the surface area of the coated MCM is between 10 m 2 /g to 50 m 2 /g. In some embodiments, the surface area of the coated MCM is approximately 10X greater than that of the starting uncoated core material.
  • the MCM can also be an excipient.
  • an "excipient” is a substance formulated alongside the active ingredient of a medication, included for the purpose of long-term stabilization, bulking up solid formulations that contain potent active ingredients in small amounts (thus often referred to as “bulking agents", “fillers”, or “diluents”), or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption, reducing viscosity, or enhancing solubility.
  • Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerns such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation or aggregation over the expected shelf life.
  • the selection of appropriate excipients also depends upon the route of administration and the dosage form, as well as the active ingredient and other factors.
  • the MCM can be a stabilizer to increase the halflife of a therapeutic, either alone or in combination with other excipients.
  • FIG. 1 shows a micrograph of an example MCM having a nano- structured calcium phosphate mineral coating. These can provide a platform for sustained delivery of biological macromolecules.
  • the mineral coated microparticles offer an injectable and systemic or localized delivery system that can lower the dose and off-target side-effects when compared to bolus injection.
  • Mineral coated microparticles offer a delivery system that can sustainably release proteins while maintaining their activity. In some cases, these microparticles can remain localized when injected in vivo and offer a localized delivery system which can allow for lower therapeutic dosages when compared to systemic subcutaneous or intravenous delivery. Further, release of protein from mineral coated microparticles can be tailored by altering the coating composition. In addition, mineral coated microparticles have a high binding capacity for biological macromolecules which allows them to sustainably deliver a suitable dose of protein with little delivery system material. This may widen the applicability of sustained delivery systems.
  • FIG. 3A delivery of the protein without the MCM can result in rapid clearing of the protein (and the diminution of therapeutic effect) from the body. This can require frequent dosing, as is the case with anakinra not delivered by MCM.
  • FIG. 3B shows that sustained release of anakinra from an MCM can sustain the dose in the therapeutic window for an extended period of time, ultimately requiring less protein and less need for attention from doctors.
  • the formulation includes a mineral coated microparticle, wherein the mineral coated microparticle comprises a core; a mineral coating on the core; and an anti-inflammatory protein.
  • the core is a nucleation site for coating precipitation.
  • the protein is adsorbed to the mineral coating.
  • the protein is incorporated throughout the mineral coating.
  • the protein is adsorbed to multiple layers of mineral coating on the core.
  • multiple, different active agents are adsorbed to the mineral coating along with a protein.
  • multiple proteins are adsorbed to the mineral coating.
  • the formulation includes a carrier, wherein the carrier is for a mineral coated microparticle, wherein the mineral coated microparticle comprises a core; a mineral coating on the core; and an antiinflammatory protein adsorbed to the mineral coating.
  • another active agent is adsorbed to the mineral coating along with the protein.
  • the carrier is a liquid.
  • the carrier is a solution or a liquid.
  • the carrier is a gel.
  • the carrier is a gas.
  • the carrier is a solid.
  • the carrier contains an active agent.
  • the active agent is an IL-IRa or an equivalent thereof such as anakinra.
  • the active agent in the carrier contains the same anti-inflammatory protein adsorbed on or incorporated within the mineral coating.
  • Suitable liquid carriers include water, saline, isotonic saline, phosphate buffered saline, Ringer's lactate, and the like.
  • Suitable gel carriers include collagen, hydrogels, polymer gels, polyethylene glycol, and the like.
  • Formulations can also include other components such as surfactants, preservatives, and excipients.
  • Surfactants can reduce or prevent surface-induced aggregation of the active agent and the mineral coated microparticles.
  • Various surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbitol fatty acid esters. Amounts can range from about 0.001 and about 4% by weight of the formulation.
  • preservatives include, for example, phenol, o-cresol, m-cresol, p- cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p- hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p- hydroxybenzoate, benzethonium chloride, chlorphenesine (3p- chlorphenoxypropane-l,2-diol) and mixtures thereof.
  • the preservative can be present in concentrations ranging from about 0.1 mg/ml to about 20 mg/ml, including from about 0.1 mg/ml to about 10 mg/ml.
  • a preservative can be used in pharmaceutical compositions such as, but not limited to those described in “Remington: The Science and Practice of Pharmacy, 19th edition, 1995,” which is incorporated herein by reference in its entirety for all purposes.
  • Formulations can include suitable buffers such as sodium acetate, glycylglycine, HEPES (4-(2-hydroxyethyl)-l- piperazineethanesulfonic acid) and sodium phosphate.
  • Excipients include components for tonicity adjustment, antioxidants, and stabilizers used in the preparation of pharmaceutical formulations.
  • Other inactive ingredients include, for example, L-histidine, L-histidine monohydrochloride monohydrate, sorbitol, polysorbate 80, sodium citrate, sodium chloride, and EDTA disodium.
  • Suitable core materials on which the mineral coating is formed include polymers, ceramics, metals, glass and combinations thereof in the form of particles.
  • Suitable particles can be, for example, agarose beads, latex beads, magnetic beads, polymer beads, ceramic beads, metal beads (including magnetic metal beads), glass beads and combinations thereof.
  • the microparticle can include ceramics (e.g., hydroxyapatite, beta-tricalcium phosphate (beta-TCP, P-TCP), magnetite, neodymium), plastics (e.g., polystyrene, poly-caprolactone), hydrogels (e.g., polyethylene glycol; poly(lactic-co-glycolic acid), and the like, and combinations thereof.
  • Suitable core materials can be those that dissolve in vivo such as, for example, beta-tricalcium phosphate (beta-TCP, P-TCP).
  • Suitable microparticle sizes can range from about 1 pm to about 100 pm in diameter. In some cases, the diameter is about 1 pm, about 5 pm, about 10 pm, about 20 pm, about 30 pm, about 40 pm, about 50 pm, about 60 pm, about 70 pm, about 80 pm, about 90 pm, about 100 pm, about 120 pm, about 150 pm, or about 200 pm. Microparticle diameter can be measured by, for example, measurements taken from microscopic images (including light and electron microscopic images), filtration through a size-selection substrate, and the like.
  • Suitable core materials include those materials non-toxic to humans and animals. Suitable core materials also include those materials that degrade and/or dissolve in humans and animals. Suitable core materials include P-tri calcium phosphate (P-TCP), hydroxyapatite (HA), poly(lactic-co-glycolic acid) (PLGA), and combinations thereof.
  • P-TCP P-tri calcium phosphate
  • HA hydroxyapatite
  • PLGA poly(lactic-co-glycolic acid)
  • P-tricalcium phosphate cores are can be suitable as the P-tricalcium phosphate degrades rapidly after mineral coating dissolution. Both P-tricalcium phosphate and hydroxyapatite are can also be suitable cores because they dissolve into calcium and phosphate ions which are easily metabolized by the body.
  • the core material can be dissolved following mineral coating formation.
  • the core material is non-degradable.
  • the mineral coating can include calcium, phosphate, carbonate, and combinations thereof.
  • a core material is incubated in a modified simulated body fluid. Simulated body fluid contains the same ion constituents at the same concentrations as human blood plasma. Modified simulated body fluid contains similar, but altered ion constituents as human blood plasma. In some embodiments, the modified simulated body fluid contains twice the concentration of calcium and phosphate as human blood plasma along with the other ionic components of human blood plasma at physiological concentrations.
  • the modified simulated body fluid can include calcium and phosphate, which form the mineral coating on the surface of the core, which results in the mineral coated microparticle. Because the modified simulated body fluid contains a supersaturation of calcium and phosphate, a mineral coating precipitates from solution onto the core material to form the mineral coating. Different mineral coating morphologies can be achieved by varying the amounts and ratios of calcium, phosphate, and carbonate in the modified simulated body solution during coating precipitation. Other ions, or dopants, can also be added to the modified simulated body fluid during coating formation to change the coating composition and/or morphology. Different mineral coating morphologies include, for example, plate-like structure, spherulite-like structure.
  • High carbonate concentration can result in a mineral coating having a plate-like structure.
  • Low carbonate concentration can result in a mineral coating having a spherulite-like structure.
  • the mineral coating morphology can also affect adsorption of the active agent.
  • the mineral coating morphology can also affect the preservation of activity of the active agent release from the mineral coating.
  • the modified simulated body fluid (mSBF) for use in the methods of the present disclosure can include from about 5 mM to about 12.5 mM calcium ions, including from about 7 mM to about 10 mM calcium ions, and including about 8.75 mM calcium ions; from about 2 mM to about 12.5 mM phosphate ions, including from about 2.5 mM to about 7 mM phosphate ions, and including from about 3.5 mM to about 5 mM phosphate ions; and from about 4 mM to about 100 mM carbonate ions.
  • the mSBF can further include about 145 mM sodium ions, from about 6 mM to about 9 mM potassium ions, about 1.5 mM magnesium ions, from about 150 mM to about 175 mM chloride ions, about 4 mM HCO3", and about 0.5 mM SO4 ⁇ " ions.
  • the pH of the mSBF can range from about 4 to about 7.5, including from about 5.3 to about 6.8, including from about 5.7 to about 6.2, and including from about 5.8 to about 6.1.
  • Suitable mSBF can include, for example: about 145 mM sodium ions, about 6 mM to about 9 mM potassium ions, about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 150 mM to about 175 mM chloride ions, about 4.2 mM HCO3', about 2 mM to about 5 mM HPCM ⁇ " ions, and about 0.5 mM SO4 ⁇ - ions.
  • the pH of the simulated body fluid may be from about 5.3 to about 7.5, including from about 6 to about 6.8.
  • the mSBF may include, for example: about 145 mM sodium ions, about 6 mM to about 17 mM potassium ions, about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 150 mM to about 175 mM chloride ions, about 4.2 mM to about 100 mM HCO3', about 2 mM to about 12.5 mM phosphate ions, and about 0.5 mM 804 ⁇ " ions.
  • the pH of the simulated body fluid may be from about 5.3 to about 7.5, including from about 5.3 to about 6.8.
  • the mSBF includes: about 145 mM sodium ions, about 6 mM to about 9 mM potassium ions, from about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 60 mM to about 175 mM chloride ions, about 4.2 mM to about 100 mM HCO3', about 2 mM to about 5 phosphate ions, about 0.5 mM 804 ⁇ " ions, and a pH of from about 5.8 to about 6.8, including from about 6.2 to about 6.8.
  • the mSBF includes: about 145 mM sodium ions, about 9 mM potassium ions, about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 172 mM chloride ions, about 4.2 mM HCO3', about 5 mM to about 12.5 mM phosphate ions, about 0.5 mM SO4 ⁇ ‘ ions, from about 4 mM to about 100 mM CO3 ⁇ ‘, and a pH of from about 5.3 to about 6.0.
  • a core incubated in a formulation of modified simulated body fluid.
  • the layer of mineral coating forms on the core during the incubation period of minutes to days.
  • the mineral coated microparticle can be removed from the modified simulated body fluid and washed.
  • a mineral coated microparticle can be incubated in a second, third, fourth, etc. modified simulated body fluid until an appropriate number of layers of mineral coating is achieved.
  • a new layer of mineral coating forms on the previous layer.
  • active agents such as proteins can be included in the modified simulated body fluid to incorporate active agents within the layer of mineral coating during mineral formation.
  • the active agent can be a protein or can be a different active agent.
  • the mineral coated microparticle can then be incubated in a carrier comprising at least one active agent to adsorb the agent to the layer of mineral coating.
  • another layer of mineral coating can be formed by incubating the microparticle in another formulation of modified simulated body fluid.
  • layers of mineral coating can incorporate an active agent in the mineral, layers can have an active agent adsorbed to the layer of mineral, the layer of mineral coating can be formed without incorporating an active agent or adsorbing an active agent, and combinations thereof.
  • Mineral coated microparticles having different layers of mineral coating can be prepared by forming a layer of mineral using one formulation of modified simulated body fluid, then incubating the mineral coated microparticle in a different formulation of modified simulated body fluid.
  • mineral coated microparticles can be prepared to have a plurality of layers of mineral coating wherein each layer is different.
  • Embodiments are also contemplated that include two or more layers of mineral coating that are the same combined with one or more layers of mineral coating that are the different.
  • One of the active agents can be a protein such as an antigen or an mRNA that expresses an antigen when administered to a subject in need of vaccination.
  • the composition of the mineral coating in the different layers advantageously allows for tailored release kinetics of the active agent or active agents from each layer of the mineral coating.
  • the active agent can be included in the mSBF. As mineral formation occurs, active agents become incorporated with the mineral coating.
  • magnetic material can be incorporated into mineral coatings.
  • superparamagnetic iron oxide linked to bovine serum albumin can be incorporated into mineral coatings.
  • Linked proteins e.g., bovine serum albumin
  • the mineral coating further includes a dopant.
  • Suitable dopants include halogen ions, for example, fluoride ions, chloride ions, bromide ions, and iodide ions.
  • the dopant(s) can be added with the other components of the mSBF prior to incubating the substrate in the mSBF to form the mineral coating.
  • the dopant ions can alter the dissolution kinetics of the mineral and can thus alter the release kinetics of protein or other active agent from the mineral coating.
  • halogen ions including fluoride ions can be used.
  • Suitable fluoride ions can be provided by fluoride ion-containing agents such as water-soluble fluoride salts, including, for example, alkali and ammonium fluoride salts. Incorporation of fluoride alters the stability of the mineral coating.
  • the fluoride ion-containing agent can be included in the mSBF to provide an amount of up to 100 mM fluoride ions, including from about 0.001 mM to 100 mM, including about 0.01 mM to about 50 mM, including from about 0.1 mM to about 15 mM, and including about 1 mM fluoride ions.
  • Inclusion of one or more dopants in the mSBF can result in the formation of a halogen-doped mineral coating that can have significantly different morphologies and/or dissolution and release kinetics.
  • the different morphology may be beneficial for preserving the activity of the active agent release from the mineral coating.
  • the control of mineral coating dissolution can be beneficial when tailoring the coating to have sufficient release kinetics for the active agent to enhance efficacy.
  • magnetic materials including magnetite, magnetite-doped plastics, and neodymium, are used for the microparticle core material. Including magnetic materials results in the formation of MCM for which location and/or movement/positioning of the MCM by application of a magnetic force is enabled.
  • the alternate use of magnetic microparticle core materials can allow for spatial control of where the active agent and/or the protein is delivered.
  • the mineral coatings may be formed by incubating the substrate with the mSBF at a temperature of about 37°C for a period of time ranging from about 3 days to about 10 days.
  • the mineral coated microparticles can be contacted with a solution containing the protein. This contact can form a protein loaded mineral coated microparticle.
  • Other active agent(s) can also be adsorbed to the mineral coating along with the protein by including them in the solution with the protein.
  • the microparticles can be contacted with a second solution containing other active agent(s) after loading with the protein. Addition of other active agents can make the delivery of protein more efficient or effective. In some embodiments, only a protein is incorporated, adsorbed, or loaded onto or into the mineral coating.
  • active agent refers to a biologically active molecule.
  • protein loaded mineral coated microparticle refers to a mineral coated microparticle which has protein adsorbed to the mineral coating and/or has protein incorporated throughout the coating.
  • the protein and/or other active agent(s) can be contacted with the mineral coated microparticle using any suitable method. For example, a solution of the protein and/or other active agent(s) can be pipetted, poured, or sprayed onto the mineral coated microparticle. Alternatively, the mineral coated microparticle can be dipped in a solution including protein and/or other active agent(s) along with the protein. Alternatively, the mineral coated microparticle can be bathed or incubated in a solution containing protein and/or other active agent(s).
  • the protein, and/or other active agent(s) can adsorb to the mineral coating by an electrostatic interaction between the protein or active agent and the mineral coating of the mineral coated microparticle.
  • Suitable active agents include biological molecules. Suitable active agents include proteins, small molecules, hormones, steroids, NSAIDs, cytokines, therapeutic proteins, antibodies, receptor antagonists, or the like. Adsorption of the protein, or other active agents along with the protein, to the mineral coated microparticles can be tailored by changing the mineral constituents (e.g., high carbonate and low carbonate microspheres), by changing the amount of mineral coated microparticles incubated with the protein, or other active agents along, by changing the concentration of protein, or other active agents in the incubation solution, and combinations thereof.
  • the mineral coatings can be analyzed to determine the morphology and composition of the mineral coatings.
  • the composition of the mineral coatings can be analyzed by energy dispersive X-ray spectroscopy, Fourier transform infrared spectrometry, X-ray diffractometry, and combinations thereof.
  • Suitable X-ray diffractometry peaks can be, for example, at 26° and 31°, which correspond to the (0 0 2) plane, the (2 1 1) plane, the (1 1 2) plane, and the (2 0 2) plane for the hydroxyapatite mineral phase.
  • Suitable X-ray diffractometry peaks can be, for example, at 26° and 31°, which correspond to the (0 0 2) plane, the (1 1 2) plane, and the (3 0 0) plane for carb onate- substituted hydroxyapatite.
  • Other suitable X-ray diffractometry peaks can be, for example, at 16°, 24°, and 33°, which correspond to the octacalcium phosphate mineral phase.
  • Suitable spectra obtained by Fourier transform infrared spectrometry analysis can be, for example, a peak at 450-600 cm‘l, which corresponds to O-P-O bending, and a peak at 900-1200 cm' ⁇ , which corresponds to asymmetric P-0 stretch of the PO4 ⁇ ' group of hydroxyapatite.
  • Suitable spectra peaks obtained by Fourier transform infrared spectrometry analysis can be, for example, peaks at 876 cm' ⁇ , 1427 cm' ⁇ , and 1483 cm' ⁇ , which correspond to the carbonate (CO3 ⁇ ‘) group.
  • the peak for HPOd ⁇ " can be influenced by adjusting the calcium and phosphate ion concentrations of the mSBF used to prepare the mineral coating.
  • the HPOd ⁇ " peak can be increased by increasing the calcium and phosphate concentrations of the mSBF.
  • the HPCM ⁇ " peak can be decreased by decreasing the calcium and phosphate concentrations of the mSBF.
  • Another suitable peak obtained by Fourier transform infrared spectrometry analysis can be, for example, a peak obtained for the octacalcium phosphate mineral phase at 1075 cm" ' , which can be influenced by adjusting the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating.
  • the 1075 cm'l peak can be made more distinct by increasing the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating.
  • the 1075 cm'l peak can be made less distinct by decreasing the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating.
  • Energy dispersive X-ray spectroscopy analysis can also be used to determine the calcium/phosphorus ratio of the mineral coating.
  • the calcium/phosphorus ratio can be increased by decreasing the calcium and phosphate ion concentrations in the mSBF.
  • the calcium/phosphorus ratio may be decreased by increasing the calcium and phosphate ion concentrations in the mSBF.
  • the mSBF can include calcium and phosphate ions in a ratio ranging from about 10: 1 to about 0.2: 1, including from about 2.5: 1 to about 1 : 1.
  • the morphology of the mineral coatings can be analyzed by scanning electron microscopy, for example. Scanning electron microscopy can be used to visualize the morphology of the resulting mineral coatings.
  • the morphology of the resulting mineral coatings can be, for example, a spherulitic microstructure, plate-like microstructure, and/or a net-like microstructure. Suitable average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2 pm to about 42 pm. Suitable average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2 pm to about 4 pm.
  • average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2.5 pm to about 4.5 pm. In some embodiments, average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 16 pm to about 42 pm.
  • Mineral coated microparticles can be stored for later use, washed and stored for later use, washed and immediately used for adsorption, or immediately used for adsorption without washing. Storage of mineral coated microparticles can include lyophilization.
  • mice were given a single subcutaneous injection of anakinra or anakinra bound to MCM (DTX020). Serum anakinra concentration was measured after treatment. Intraperitoneal injections of IL-ip were administered 1, 5, and 7 days after treatment and the resulting inhibition of IL-linduced upregulation of IL-6 was examined in serum. DTX020 elevated anakinra concentration 5 and 7 days after administration (left), and inhibited IL-1 induced levels of IL-6 (right) at all timepoints examined (p ⁇ 0.05), suggesting improved anti-inflammatory activity, mean ⁇ SE.
  • MCM-bound anakinra can increase the serum concentration of anakinra for 5 and 7 days after administration when compared to an equivalent dose of anakinra without the addition of MCMs.
  • MCMs can both significantly lower the required dose and decrease the required frequency of dosing of anakinra to inhibit IL-1 induced inflammation and prevent and treat ARDS and other inflammatory conditions.
  • a single dose that successfully inhibits inflammation for 7 days can prevent the development of ARDS in high-risk patients.
  • a single dose that successfully inhibits inflammation for 1, 2, 3, 4 5, 6 7, 8, 9, 10 or more days can significantly reduce the duration and/or magnitude of a gout flare.
  • a combination of anakinra with MCMs can be delivered subcutaneously, not requiring an intravenous (i.v.) line.
  • the subcutaneous injection comprises a single dose injection of anakinra with MCMs.
  • the subcutaneous single dose injection of anakinra with MCMs can be administered to ARDS patients in conjunction with other concurrent treatments.
  • the subcutaneous single dose injection of anakinra with MCMs can be administer to gout patients in conjunction with other concurrent treatments.
  • a method for treating inflammation can include providing a formulation comprising a biologic, admixing the formulation with a mineral coated microparticle (MCM), wherein the MCM adsorbs to the biologic and provides a sustained delivery of biologic when administered in vivo, wherein the inflammation is exemplified by an increase in IL-1 or IL-6 production, and wherein admixed formulation is administered to a subject in need thereof.
  • MCM mineral coated microparticle
  • the method for treating inflammation includes providing a formulation subcutaneously. In some embodiments, the method for treating inflammation includes providing a formulation through localized injection. In some embodiments, the localized injection is into a joint. In some embodiments, the formulation is injected into the synovial cavity of a joint.
  • the inflammation is acute respiratory distress syndrome (ARDS).
  • the inflammation is associated with graft versus host disease (GVHD).
  • the inflammation is associated with infection by the SARS- CoV-2 virus.
  • the inflammation is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
  • the inflammation is chronic or acute inflammation.
  • the inflammation is associate with a gout flare.
  • the method of treatment reduces the inflammation associated with a gout flare.
  • the method of treatment reduces the duration of a gout flare.
  • the method of treatment prevents the formation of urate crystals that induce of gout flare.
  • the method of treatment prevents inflammation occurring that is caused by urate crystals.
  • the inflammation is systemic. In some embodiments the inflammation is localized in the lungs. In some embodiments the inflammation is localized within a joint.
  • the biologic is interleukin-1 receptor agonist (IL-IRa), or an analog thereof, such as anakinra.
  • IL-IRa interleukin-1 receptor agonist
  • the concentration of the MCM in the formulation is the same as the concentration of anakinra. In some embodiments, the concentration of the anakinra is 10X greater than the concentration of the MCMs in the formulation. Suitable ratios of MCM concentration to anakinra concentration can range from 100: 1 to 10: 1, from 1 : 1 to 1 : 10, and from 1 : 10 to 1 : 1000. In some embodiments, suitable concentrations of MCMs can range from 0.1 mg/ml to 10 mg/ml. In some embodiments, suitable concentrations of MCMs can range from 10 ug/ml to 100 ug/ml. In some embodiments, approximately 0.7 mg of MCM is administered subcutaneously per kg of body weight.
  • approximately 0.1 mg of MCM is administered subcutaneously per kg of body weight.
  • Suitable dosing ranges for MCMs may include 10 ug/kg to 100 ug/ml, 0.1 mg/kg to 1 mg/kg, or 1 mg/kg to 5 mg/kg.
  • approximately 7 mg of anakinra per kg of body weight is administered subcutaneously.
  • approximately 1 mg of anakinra per kg of body weight is administered.
  • Suitable dosing ranges of anakinra may include 0.1 mg/kg to 1 mg/kg or 1 mg/kg to 10 mg/kg.
  • the amount of MCMs or anakinra administered is depends on the volume or mass of tissue where the treatment is administered. For example, the amount of anakinra and MCM in a localized injection used to treat or prevent a gout flare is dependent on the size of the joint.
  • approximately 1-5 mg of MCMs per ml of synovial fluid within a joint is administered. In some embodiments, approximately 2.5 mg of MCMs per ml of synovial fluid is administered. In some embodiments, approximately 5-50 mg of MCMs per ml of synovial fluid is administered.
  • approximately 25 mg of MCMs per ml of synovial fluid is administered. In some embodiments, less than 1 mg of MCMs per ml of synovial fluid is administered. In some embodiments, approximately 0.35 mg of MCMs per ml of synovial fluid is administered. In some embodiments for local administration, the ratio of MCMs to anakinra can be 1 : 1.
  • an amount of biologic administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM (e.g., by at least about 50%, at least about 75%, at least about 85%, at least about 90%, or at least about 95%).
  • the admixed formulation is provided prophylactically to a subject in need thereof.
  • a method for reducing the expression of IL-1 and/or IL-6 in vivo comprising a single administration of a formulation to a patient exhibiting elevated serum concentrations of IL-1 and/or IL-6, the formulation comprising a sustained release admixture, wherein the admixture comprises a drug delivery vehicle and IL- IRa, wherein the elevated serum concentration of IL-1 and/or IL-6 is associated with an acute inflammatory event.
  • the serum concentration of IL-1 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 in vivo.
  • the serum concentration of IL-6 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-6 in vivo.
  • the serum concentrations of IL-1 and IL-6 are elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 and IL-6 in vivo.
  • no more than one administration of the formulation is performed in a 24 hour, 48 hour, or 72 hour period.
  • the method further comprises administering a subsequent dose of the formulation after the single administration of the formulation.
  • the serum concentration of IL-1 and/or IL-6 is at least about 2, at least about 5, at least about 10, at least about 20, or at least about 50-times higher than a patient not having the acute inflammatory event.
  • the expression IL-1 and/or IL-6 resulting from the single administration can be reduced compared to a single administration of IL-IRa alone (e.g., by at least about 2, 5, 10, 20, or 50-fold). In some cases, the reduction of IL-1 and/or IL-6 serum concentration lasts over a period of 3, 5, or 7 days in vivo.
  • the drug delivery vehicle can be a mineral coated microparticle (MCM).
  • an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
  • a method for preventing inflammation in vivo comprising administering an admixture of IL-IRa and a mineral coated microparticle (MCM) to a subject having an elevated risk of inflammation.
  • MCM mineral coated microparticle
  • the subject is at risk of inflammation of immune cells, lymphocytes, or mast cells. In some embodiments, the subject is at risk of respiratory or cardiovascular inflammation.
  • the subject is showing signs of inflammation. In some embodiments, the subject is at an onset of inflammation. In some embodiments, the subject is exhibiting hyperoxia.
  • the subject is experiencing a gout flare.
  • Indications of a gout flare include painful joint swelling and inflammation and/or the presence of urate crystals in the synovial fluid of a joint.
  • the subject is at risk of developing a gout flare.
  • Increased risk can be indicated by high uric acid levels, formation of urate crystals in a joint, or a history of gout flares.
  • the subject has elevated serum concentrations of IL-1 and/or IL-6.
  • the MCM can provide for sustained release of the IL-IRa.
  • the subject has elevated synovial concentrations of IL-1 and/or IL-6 within a joint.
  • the methods described herein can exhibit a number of advantages over previous therapies for treating ARDS such as associated with COVID-19, including: (1) decreased need for recombinant anakinra, mitigating supply, storage, and stockpiling limitations; (2) does not require i.v. line for delivery, (3) decreased fluid (volume) load relative to anakinra alone, (4) a single dose can be effective for many patients, (5) stability is much greater than altematives, increasing value for field use, and (6) does not require significant medical training for subcutaneous administration. Additionally, the methods described herein can exhibit a number of advantages for treating and preventing gout flares.
  • the methods described herein can 1) significantly shorten the duration and magnitude of a gout flare, 2) reduce joint damage caused by the gout flare, 3) reduce the number of injections required for treatment, and 4) reduce the amount of biologic required for treatment.
  • the methods described herein can 1) dramatically reduce the amount of protein required to prevent flares, 2) allow for biologies with short half-lives to be used to prevent gout flares, 3) and lengthen the duration between treatment administration.

Abstract

In an aspect, provided herein is a method for treating or preventing inflammation such as acute respiratory distress syndrome (ARDS), graft versus host disease (GVHD) or inflammation caused by chimeric antibody receptor T-cell (CAR-T) therapy. In some embodiments, the treatment or prevention involves administering an admixture of interleukin-1 receptor agonist (IL1-Ra) and a mineral coated microparticle (MCM) to a subject in need thereof. This formulation can substantially reduce the amount of IL1-Ra needed and/or the frequency of administration.

Description

METHODS FOR TREATING OR PREVENTING INFLAMMATORY EVENTS
CROSS-REFERENCE
[0001] This application claims priority to U.S. Provisional Patent Application No. 63/073,135, filed September 1, 2020, which application is herein incorporated by reference in its entirety for all purposes.
BACKGROUND
[0002] Inflammatory events are significant. For example, acute respiratory distress syndrome (ARDS) cytokine release syndrome (CRS), hypercytokinemia, and cytokine storm can be devastating disorders with high morbidity and mortality rates characterized by rapidly progressive hypoxic lung failure. It can be triggered by factors including sepsis, pneumonia, overwhelming infections, trauma, CO VID-19 infection and other conditions. ARDS is one of the leading causes of death in patients infected with COVID-19, and the number of ARDS deaths and hospitalizations in the US has been increasing significantly. Prior to the emergence of COVID-19, ARDS accounted for 75,000 deaths, 3.6 million hospital days, and $5 billion in healthcare costs in the U.S. annually.
[0003] Furthermore, inflammation can be triggered by transplant (e.g., graft versus host disease (GvHD)) or CAR-T therapy. With respect to the latter, genetically engineering T cells with chimeric antigen receptors (CARs) represents a highly sophisticated and radically innovative way of treating cancer. Unfortunately, clinical manifestations of CRS can develop within the first days from CAR-T cell infusion and can include high fever, increased levels of acute phase proteins, respiratory and cardiovascular insufficiency, which if severe and left untreated may lead to death.
[0004] Another example of an inflammatory event is gout, an inflammatory arthritic disease affecting millions of people in the U.S with significant economic burden. Without treatment, gout sufferers can have frequent, excruciating, and protracted attacks as well as permanent joint damage. Gout develops when excessive levels of uric acid accumulate in tissue and form crystals which deposit in various joints causing excruciating pain and inflammation. These crystals stimulate neutrophil ingress into the joint and activate synovial tissue macrophages to produce mediators of inflammation, including, interleukin (IL)- 1 P, tumor necrosis factor (TNF)-a, and IL-6. Without treatment, more frequent/protracted polyarthritis attacks occur, eventually resulting in permanent joint damage. Chronic inflammation often remains present after gout symptoms are under control. SUMMARY
[0005] Recognized herein is an urgent need for novel therapies for treating and preventing inflammatory events, including ARDS, CRS, hypercytokinemia and cytokine storm. [0006] In an aspect, provided herein is a method for treating or preventing inflammation such as acute respiratory distress syndrome (ARDS), cytokine release syndrome (CRS), cytokine storm, graft versus host disease (GVHD), sepsis, or inflammation caused by chimeric antibody receptor T-cell (CAR-T) therapy or inflammation associated with gout. In some embodiments, the treatment or prevention involves administering an admixture of interleukin- 1 receptor agonist (ILl-Ra) or its equivalents, including anakinra along with a mineral coated microparticle (MCM) to a subject in need thereof. Anakinra (trade name Kineret) is a recombinant IL-IRa protein but can differ in that it is not-glycosylated, can be manufactured in Escherichia coh. and can differ in sequence by one methionine added to its N-terminus. An admixture of MCMs and anakinra can substantially reduce the amount of anakinra needed and/or the frequency of administration.
[0007] As described herein, the need for improved methods for treatment or prevention of inflammation is addressed by using a mineral-coated microparticle (MCM). An MCM is a biomimetic, tailorable, mineral coated microparticle. MCMs can bind (e.g., adsorb), stabilize, and release proteins, peptides and nucleic acid molecules. In some cases, MCMs can be used in combination with IL-IRa for prevention and/or treatment of inflammation. When used as an excipient for therapeutic proteins, MCMs are able to maintain activity, stabilize structure, and provide superior loading capacity compared to current therapeutic protein delivery platforms. [0008] For example, intraarticular delivery of anakinra using MCMs can sustain biologically active protein release, thereby extending the time between treatments.
[0009] MCMs can utilize calcium phosphate mineral (CaP) coatings to bind and release therapeutic proteins in a sustained and controllable manner. CaP mineral coatings can be precipitated onto core materials (such as, for example, beta-tri calcium phosphate (P-TCP) particles, hydroxyapatite particles, or polymer microspheres) by incubating (e.g., for a period of 5-10 days) in modified simulated body fluid (mSBF), a calcium phosphate solution similar to human blood plasma. Proteins can bind to the CaP mineral surface through electrostatic interactions between the polar and charged groups of the proteins and the positively (Ca2+) and negatively (PO4 3- ) charged ions in the CaP mineral. The mineral surface can serve as a platform for binding and stabilizing proteins to the coating by maintaining the protein’s conformational structure. Moreover, because of the plate-like structure of the mineral coatings, significantly greater loading capacity of protein is possible compared to other delivery platforms. Release and delivery of therapeutic proteins from the MCMs can occur via dissolution of the CaP mineral coatings. As MCMs are inorganic components, they also do not elicit unwanted immunogenic responses.
[0010] MCMs can be constructed of generally regarded as safe (GRAS) materials. They can also be added to an existing formulation and optionally lyophilized to create a product that is stable at room temperature, can be stockpiled, and can be distributed without need for refrigeration. The lyophilized composition can be reconstituted and used at the point of administration.
[0011] In an aspect, provided herein is a method for treating inflammation. The method can include (a) providing a formulation comprising a biologic; and (b) admixing the formulation with a mineral coated microparticle (MCM) to provide an admixed formulation, wherein the MCM adsorbs the biologic and provides a sustained delivery of the biologic when administered in vivo, wherein the inflammation is exemplified by an increase in one or more members of the IL-1 family of cytokines or IL-6 production, and wherein the admixed formulation is administered to a subject in need thereof.
[0012] In some embodiments, the inflammation is acute respiratory distress syndrome (ARDS). [0013] In some embodiments, the inflammation is associated with graft versus host disease (GVHD).
[0014] In some embodiments, the inflammation is associated with infection by the SARS-CoV-2 virus.
[0015] In some embodiments, the inflammation is associated with cytokine storm syndrome.
[0016] In some embodiments, the inflammation is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
[0017] In some embodiments, the inflammation is associated with gout.
[0018] In some embodiments the inflammation is associated with urate crystal formation and/or presence.
[0019] In some embodiments, the inflammation is acute inflammation.
[0020] In some embodiments, the biologic is interleukin-1 receptor agonist (IL-IRa), or an analog thereof.
[0021] In some embodiments, the biologic is anakinra or a derivative thereof.
[0022] In some embodiments, an amount of biologic administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
[0023] In some embodiments, the admixed formulation is provided prophylactically to a subject in need thereof. [0024] In an aspect, provided herein is a method for reducing the expression of IL-1 or IL-6. The method can include administering a single dose of a formulation to a patient exhibiting elevated serum concentrations of IL-1 or IL-6 associated with an acute inflammatory event, wherein the formulation comprises a sustained release admixture, wherein the sustained release admixture comprises a drug delivery vehicle and IL-lRa.
[0025] In some embodiments, the elevated serum concentration of IL-1 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 in vivo.
[0026] In some embodiments, the elevated serum concentration of IL-6 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-6 in vivo.
[0027] In some embodiments, the elevated serum concentrations of IL-1 and IL-6 are elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 and IL-6 in vivo.
[0028] In some embodiments, no more than one administration of the formulation is performed in a 24 hour, 48 hour, or 72 hour period.
[0029] In some embodiments, the method further comprises administering a subsequent dose of the formulation after the single administration of the formulation.
[0030] In some embodiments, the elevated serum concentration of IL-1 or IL-6 is at least about 2, at least about 5, at least about 10, at least about 20, or at least about 50-times higher than a patient not having the acute inflammatory event.
[0031] In some embodiments, the expression of IL-1 or IL-6 resulting from the single administration is reduced compared to a single administration of IL-lRa alone.
[0032] In some embodiments, the expression of IL-1 or IL-6 is reduced by at least about 2, 5, 10, 20, or 50-fold.
[0033] In some embodiments, the expression of IL-1 or IL-6 lasts over a period of 3, 5, or 7 days in vivo.
[0034] In some embodiments, the acute inflammatory event is acute respiratory distress syndrome (ARDS).
[0035] In some embodiments, the acute inflammatory event is a gout flare.
[0036] In some embodiments, the acute inflammatory event is graft versus host disease (GVHD).
[0037] In some embodiments, the acute inflammatory event is associated with infection by the SARS-CoV-2 virus or influenza virus. [0038] In some embodiments, the acute inflammatory event is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
[0039] In some embodiments, the drug delivery vehicle is a mineral coated microparticle (MCM).
[0040] In some embodiments, an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
[0041] In an aspect, provided herein is a method for preventing inflammation. The method can comprise administering an admixture of IL-IRa and a mineral coated microparticle (MCM) to a subject having an elevated risk of inflammation.
[0042] In some embodiments, the subject exhibits or is at risk of a pulmonary edema.
[0043] In some embodiments, the subject requires a high fraction of inspired oxygen (FiCh).
[0044] In some embodiments, the subject has been infected with the SARS-Cov-2 virus or influenza virus.
[0045] In some embodiments, the subject has received a transplant.
[0046] In some embodiments, the subject has undergone chimeric antigen receptor T-cell (CAR- T) therapy.
[0047] In some embodiments, the subject is at risk of inflammation of immune cells, lymphocytes, or mast cells.
[0048] In some embodiments, the subject is at risk of respiratory or cardiovascular inflammation.
[0049] In some embodiments, the subject is showing symptoms of inflammation.
[0050] In some embodiments, the subject is at an onset of inflammation.
[0051] In some embodiments, the subject is exhibiting hyperoxia.
[0052] In some embodiments, the subject has elevated serum concentrations of IL-1 or IL-6.
[0053] In some embodiments, no more than one administration of the admixture is performed in a 24 hour, 48 hour, or 72 hour period.
[0054] In some embodiments, the method further comprises administering a subsequent dose of the admixture after the single administration of the admixture.
[0055] In some embodiments, the MCM provides for sustained release of the IL-IRa.
[0056] In some embodiments, an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
[0057] Additional aspects and advantages of the present disclosure will become readily apparent to those skilled in this art from the following detailed description, wherein only illustrative embodiments of the present disclosure are shown and described. As will be realized, the present disclosure is capable of other and different embodiments, and its several details are capable of modifications in various obvious respects, all without departing from the disclosure. Accordingly, the drawings and description are to be regarded as illustrative in nature, and not as restrictive.
INCORPORATION BY REFERENCE
[0058] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.
BRIEF DESCRIPTION OF THE DRAWINGS
[0059] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings (also “figure” and “FIG.” herein), of which:
[0060] FIG. 1 shows a scanning electron micrograph (SEM) image of an example MCM;
[0061] FIG. 2 shows an example method for fabricating an MCM and binding protein to the surface of the MCM;
[0062] FIG. 3A shows a conceptual graphic of example repeated bolus delivery of proteins not delivered by MCMs;
[0063] FIG. 3B shows a conceptual graphic of a single sustained release delivery of proteins from an example MCM; and
[0064] FIG. 4 shows an example benefit of MCM binding on the effect of anakinra in vivo.
DETAILED DESCRIPTION
[0065] While various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.
Definitions [0066] Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosure herein belongs. As used herein, the singular forms “a,” “an,” and “the,” are intended to include the plural forms as well, unless the context clearly indicates otherwise. The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, e.g., elements that are conjunctively present in some cases and disjunctively present in other cases. Thus, as a nonlimiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in some embodiments, to A only (optionally including elements other than B); in some embodiments, to B only (optionally including elements other than A); in some embodiments, to both A and B (optionally including other elements); etc.
[0067] As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in some embodiments, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in some embodiments, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in some embodiments, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc. In certain embodiments, the term "about" or "approximately" as used herein means within an acceptable error range for the particular value as determined, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system.
[0068] Whenever the term “at least,” “greater than,” or “greater than or equal to” precedes the first numerical value in a series of two or more numerical values, the term “at least,” “greater than” or “greater than or equal to” applies to each of the numerical values in that series of numerical values. For example, greater than or equal to 1, 2, or 3 is equivalent to greater than or equal to 1, greater than or equal to 2, or greater than or equal to 3. [0069] Whenever the term “no more than,” “less than,” or “less than or equal to” precedes the first numerical value in a series of two or more numerical values, the term “no more than,” “less than,” or “less than or equal to” applies to each of the numerical values in that series of numerical values. For example, less than or equal to 3, 2, or 1 is equivalent to less than or equal to 3, less than or equal to 2, or less than or equal to 1.
[0070] In certain embodiments, "about" can mean within 3 or more than 3 standard deviations, per the practice in the art. In certain embodiments, such as with respect to biological systems or processes, the term can mean within an order of magnitude, including within 5 -fold, and within 2-fold of a value. In certain embodiments, when the term “about” or “approximately” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below those numerical values. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20%, 10%, 5%, or 1%. In certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 10%. In certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 5%. In certain embodiments, the term “about” is used to modify a numerical value above and below the stated value by a variance of 1%.
[0071] When a range of values is listed herein, it is intended to encompass each value and subrange within that range. For example, “1-5 ng” or “from about 1 ng to about 5 ng” is intended to encompass 1 ng, 2 ng, 3 ng, 4 ng, 5 ng, 1-2 ng, 1-3 ng, 1-4 ng, 1-5 ng, 2-3 ng, 2-4 ng, 2-5 ng, 3-4 ng, 3-5 ng, and 4-5 ng.
[0072] It will be further understood that the terms “comprises,” “comprising,” “includes,” and/or “including,” when used herein, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. [0073] As used herein, the term “administering,” refers to the placement of the dose as disclosed herein into a subject by a method or route which results in at least partial delivery of the composition at an appropriate extracellular location of a target tissue. In certain embodiments, the dose adsorbed to the MCM component can be, for example, injected into a subject in need thereof by either intradermal, intra-muscular, subcutaneous, intra-articular, periarticular or intravenous administration. In certain embodiments, the protein dose adsorbed to the MCM component administered parenterally, e.g., by intravenous, intra-arterial, intracardiac, intraspinal, intraosseous, intra-articular, intra-synovial, subcutaneous, intradermal, intra- tendinous, intraligamentous or intramuscular administration. In certain embodiments, the bioactive compound captured within the inorganic precipitate is administered by implantation, infiltration or infusion.
[0074] The therapeutically effective amount can vary depending upon the intended application or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined such as by a board-certified physician.
[0075] As used herein, the terms “treat,” “treatment,” “treating” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a disorder. The term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disorder is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms in the absence of treatment. Beneficial clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (e.g., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable. The term “treatment” of a disease also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
[0076] As used herein, the term “tissue” or “target tissue” refers to an aggregation of morphologically similar cells and associated intercellular matter, e.g., extracellular matrix, acting together to perform one or more specific functions in the body. In some embodiments, tissues fall into one of four basic types: muscle, nerve, epidermal, and connective. In some embodiments, a tissue is substantially solid, e.g., cells within the tissue are strongly associated with one another to form a multicellular solid tissue. In some embodiments, a tissue is substantially non-solid, e.g., cells within the tissue are loosely associated with one another, or not at all physically associated with one another, but may be found in the same space, bodily fluid, etc.
[0077] As used herein, the term “extracellular” means being situated or taking place outside a cell or cells.
[0078] A “subject” refers to a vertebrate, such as a mammal (e.g., a non-human mammal), such as a primate or a human. Mammals include, but are not limited to, primates, humans, farm animals, rodents, sport animals, and pets. [0079] As used herein, "bioavailability" is a fraction (%) of an administered drug that reaches the systemic circulation. By definition, when a medication is administered intravenously, its bioavailability is 100%. However, when a medication is administered via routes other than intravenous, its bioavailability can be lower than that of intravenous. In some cases, bioavailability equals the ratio of comparing the area under the plasma drug concentration curve versus time (AUC) for the extravascular formulation to the AUC for the intravascular formulation. In some cases, to ensure that the drug taker who has poor absorption is dosed appropriately, the bottom value of the deviation range is employed to represent real bioavailability to calculate drug dose for the drug taker to achieve systemic drug concentrations similar to the intravenous formulation. To dose without the prerequisite of drug taker's absorption state, the bottom value of the deviation range can be used in order to ensure the anticipated efficacy will be met unless the drug is associated with narrow therapeutic window. Bioavailability can be measured over any suitable period of time.
[0080] As used herein, the term “formulation”, generically indicates the beneficial agents and mineral coated microparticles are formulated, mixed, added, dissolved, suspended, solubilized, formulated into a solution, carried and/or the like in or by the fluid, gas, or solid in a physical-chemical form acceptable for patient administration.
[0081] "Effective amount" or "therapeutically effective amount" means a dosage sufficient to alleviate one or more symptoms of the condition being treated, or to otherwise provide a pharmacological and/or physiologic effect, as may be determined by an objective measure or a patient derived subjective measure. In certain embodiments, an “effective amount” refers to the optimal amount of a dose adsorbed to the MCM needed to elicit a clinically significant improvement in the symptoms and/or pathological state associated with a disease state, infection, or disorder to be treated. In certain embodiments the disease state, infection, or disorder to be treated is a viral pathogen. In certain embodiments, the dose adsorbed to the MCM is administered as a treatment. In certain embodiments, the dose adsorbed to the MCM is administered prophylactically as a preventative measure. As used herein, an “effective amount”, a “therapeutically effective amount”, a “prophylactically effective amount” and a “diagnostically effective amount” is the amount of the unbound active agent and the active agent adsorbed to the mineral coated microparticle needed to elicit a biological response following administration.
[0082] As used herein, "a subject in need thereof' (also used interchangeably herein with "a patient in need thereof') refers to a subject susceptible to or at risk of a specified disease, disorder, or condition. The methods disclosed herein can be used with a subset of subjects who are susceptible to or at elevated risk of infection by a condition for which the treatment is provided. Because some of the method embodiments of the present disclosure are directed to specific subsets or subclasses of identified subjects (that is, the subset or subclass of subjects “in need” of assistance in addressing or vaccinating against one or more specific conditions noted herein), not all subjects will fall within the subset or subclass of subjects as described herein for certain diseases, disorders or conditions. "Elevated risk" can include any statistically relevant threshold when comparing one population of subjects to another.
[0083] As used herein, IL-1 refers to the inflammatory members of the Interleukin- 1 family of cytokines. Such members include interleukin- la, interleukin- ip, interleukin- 18, interleukin- 36a, interleukin-36p, interleukin-36y, and interleukin-33.
Inflammation and ARDS
[0084] Acute inflammatory events represent a significant unmet medical need. Examples include acute respiratory distress syndrome (ARDS), such as induced by SARS-CoV-2 infection (COVID-19) or influenza, cytokine release syndrome, complications arising from CAR-T therapy, sepsis, or graft-versus-host disease.
[0085] Inflammation and oxidative stress can play a role in the pathogenesis of ARDS and related conditions. Such conditions can be caused by or associated with many factors including viral infections, bacterial infections, sepsis, exposure to toxic substances, lung injury, mechanical ventilation, or high-oxygen environments. The present disclosure and discussion in the context of one condition (e.g., coronavirus infections) does not preclude use of the materials and methods disclosed herein with other conditions. For example, coronavirus infections, including COVID-19, can be characterized by elevated pro-inflammatory markers in the serum, evidence of monocyte/macrophage activation, activated coagulation and pro-inflammatory cytokine and chemokine profiles, implying that the host response is an important factor in this disease. Furthermore, lung inflammation can intensify after viral clearance, peaking 1-2 weeks after infection in animal models and in human SARS-1 (severe acute respiratory syndrome-1) patients. This suggests that clinical deterioration later in the disease course can be due to damage from uncontrolled immune responses rather than uncontrolled viral replication. It also defines a window for therapy after presentation of most patients but before refractory ARDS is established.
[0086] The materials and methods described herein can address both lung inflammation and hyperoxic lung injury. Interleukin-1 (IL-1) is increased with both COVID-19 and hyperoxia. IL- 1 binding to IL-1 receptor (IL-1R) amplifies inflammation and results in ARDS. The ILl-Ra as delivered from MCM binds to IL-1R without activating inflammation. Hyperoxia often used to treat ARDS, but also worsens lung injury. The methods described herein alleviates the need for hyperoxia treatment.
Inflammation and Graft- Versus-Host Disease or CAR-T Therapy
[0087] Graft-versus-host disease (GvHD) can cause acute inflammation that can be treated or prevented using the methods described herein. GvHD is a syndrome that can be characterized by inflammation in different organs, in some cases with the specificity of epithelial cell apoptosis and crypt drop out. GvHD can be associated with stem cell transplants such as those that occur with bone marrow transplants. GvHD can also apply to other forms of transplanted tissues such as solid organ transplants.
[0088] White blood cells of the donor's immune system which remain within the donated tissue (the graft) can recognize the recipient (the host) as foreign (non-self). The white blood cells present within the transplanted tissue can then attack the recipient's body's cells, which can lead to GvHD. Transplant rejection can occur when the immune system of the transplant recipient rejects the transplanted tissue, while GvHD can occur when the donor's immune system's white blood cells reject the recipient. The underlying principle (alloimmunity) is similar, but the details and course may differ. GvHD can also occur after a blood transfusion if the blood products used have not been irradiated or treated with an approved pathogen reduction system.
[0089] Furthermore, CAR-T therapy can induce acute inflammatory events that are suitable for treatment or prevention using the materials and methods described herein.
[0090] Genetically engineering T cells with chimeric antigen receptors (CARs) represents a highly sophisticated and radically innovative way of treating cancer. The basic structure of CARs comprises a tumor-targeting domain, usually from the single-chain fragment variables (scFvs) of a monoclonal antibody (mAb), fused to at least one immune tyrosine activatory motif (ITAM), which can be the CD3 zeta chain, and one or more costimulatory endodomains. The FDA has thus far approved two distinct CD 19 CAR-T cell products in pediatric/young adult has paved the way to their availability outside clinical trials. Unfortunately, patients often develop ARDS. Clinical manifestations of ARDS can develop within the first days from CD19 CAR-T cell infusion and include high fever, increased levels of acute phase proteins, respiratory and cardiovascular insufficiency, which if severe and left untreated may lead to death. Therefore, there is a need for the methods of treatment and/or prevention of ARDS as described herein. Inflammation and Gout
[0091] Gout develops when excessive levels of uric acid accumulate in tissue and form crystals which deposit in various joints causing excruciating pain and inflammation. These crystals stimulate neutrophil ingress into the joint and activate synovial tissue macrophages to produce mediators of inflammation, including, interleukin (IL)-lp, tumor necrosis factor (TNF)- a, and IL-6. Without treatment, more frequent/protracted polyarthritis attacks occur, eventually resulting in permanent joint damage. IL-ip released from leukocytes may be a trigger for a cascade of inflammatory mediators and cytokines which are responsible for gout flares and subsequent damage.
Anti-inflammatories and IL-IRa
[0092] Pro-inflammatory cytokines, including IL- 1 p, TNF-a, IL-6 and IL-8, can be significantly elevated in bronchoalveolar lavage fluid and plasma of ARDS patients. Furthermore, plasma levels of IL-1 P, one of the most active pro-inflammatory cytokines, can be predictive of clinical outcomes in patients with severe ARDS. In some cases, large increases in the expressions of IL- ip, IL-6, TNF-a and integrin ligand ICAM-1 can be found in lung tissue exposed to hyperoxia. Protection can be concomitant with a smaller increase in lung tissue expressions of IL-1 p, IL-6, TNF-a and ICAM-1 and better preservation of arterial blood pCh, consistent with a key role for inflammation in the pathogenesis of hyperoxia-induced ARDS.
[0093] Interleukin- la (IL-la) and interleukin- ip (IL-1 p) are prototypic proteins of the IL-1 superfamily that exert pleiotropic pro-inflammatory effects on many cell types throughout the body. Binding of IL-la or IL-ip to the IL-1 receptor type I (IL-1RI) can allow for the recruitment of the interleukin- 1 receptor accessory protein (IL-lRAcP), which can induce a biological response that can involve activation of the nuclear factor-KB (NF-KB) and mitogen- activated protein kinase (MAPK) pathways, both of which are potent inflammatory pathways. Additionally, IL-1 signaling has been shown to produce a positive feedback loop, which amplifies IL-1 expression.
[0094] Anakinra, the recombinant form of naturally occurring Interleukin- 1 receptor antagonist (IL-IRa), is a 17 kD anti-inflammatory protein which binds to the Interleukin-1 receptor (IL- 1RI) with an affinity similar to IL-1, but does not produce an inflammatory signal. As described herein, combination of IL-IRa or equivalents (including anakinra) with MCMs can improve the anti-inflammatory activity and duration of effect of anakinra to develop a therapeutic for the prevention and treatment of ARDS. Anakinra possesses properties that allow it to inhibit detrimental IL-1 activity, including its high binding affinity and target specificity with the IL-1 receptor, low toxicity, and low molecular weight (17kD) when compared to other potential antiinflammatory therapeutic proteins. Anakinra can be used for treating inflammatory conditions including cytokine release syndrome (CRS), sepsis with features of macrophage activation syndrome (MAS), and acute respiratory distress syndrome (ARDS). However, effective dosing for inflammatory events, including COVID-19, can require twice daily intravenous (i.v.) administration of 5 mg/kg of anakinra, which can limit its practical use without improved delivery outside of, and even within, a hospital setting.
[0095] Anakinra has a very short in vivo half-life (20 minutes-4 hours) and the high dose required to inhibit IL-1 activity (10- 100-fold molar excess of IL-1). As with other low molecular weight proteins, anakinra can be quickly cleared from circulation because of enzymatic degradation, renal filtration, rapid distribution into organs, hepatic metabolism, and initial scavenging by receptors. Rapid clearance can reduce its efficacy for preventing inflammation, even when administered at frequent dosing intervals. Some patients can require twice daily, hour-long intravenous (i.v.) infusions of gram quantities of anakinra for effective treatment for ARDS. The need for gram quantities of anakinra on a per week basis to treat each patient represents a major supply and storage challenge for large numbers of patients. Local administration of anakinra by injection to prevent or treat inflammation is often not feasible, as local injections are often performed by physicians and daily administration is therefore not possible for outpatient treatments.
[0096] Anakinra can also be dosed as a daily subcutaneous injection for the treatment of some inflammatory conditions. The need for daily administration of large quantities of protein can present a burdensome dosing regimen for patients who are unaccustomed to self-administered injections. Additionally, daily injections can cause injection site reactions.
Mineral Coated Microparticles
[0097] Addition of MCMs to formulated anakinra prior to subcutaneous administration can significantly improve its anti-inflammatory activity and can reduce the amount of protein required to prevent and treat inflammatory conditions, such as ARDS or gout. MCMs are calcium phosphate based, micron-sized particles which bind, stabilize, and release proteins upon coating dissolution. A SEM (scanning electron microscope) image of an example MCM can be seen in FIG. 1. The nano structured, plate-like surface of the micron sized MCMs can provide a large surface for protein binding that also stabilizes the protein structure. These mineral coatings can stabilize protein structures and release biologically active proteins as they dissolve. Proteins can bind to the surface of the MCM through electrostatic interactions between the polar and charged group of the protein and the calcium and phosphate groups of the mineral coating, similar to hydroxyapatite chromatography. Binding to the mineral coating can preserve the protein’s tertiary structure while simultaneously protecting the protein from degradation, even in harsh environments. Proteins can then be released from the MCM upon coating dissolution, providing sustained release and improved half-life. Because proteins are loaded onto the surface of MCMs after the MCMs are fabricated and sterilized, MCMs can be added to an already formulated therapeutic to improve its half-life, much like an excipient ingredient. This unique characteristic greatly simplifies the manufacturing process when compared to other sustained delivery technologies, such as polymer encapsulation or PEGylation, and is a key advantage of the MCM.
[0098] As shown in FIG. 2, MCMs are simple and scalable. To manufacture MCMs, P- tricalcium phosphate cores can be incubated in modified simulated body fluid (mSBF), a solution which contains the same ionic constituents of human blood plasma, and the mineral coating can precipitate onto the core material. After formation, the MCMs can be freeze dried and sterilized using dry heat sterilization and depyrogenation techniques. The resulting MCMs can have acceptable bioburden and endotoxin levels for injectable materials and are stable at room temperature. A number of approaches can be taken when combining the MCM with formulated anakinra. One example approach is to combine the MCMs with the formulated anakinra at the point of care, much like the approach that is taken with lyophilized therapeutics which are reconstituted prior to administration. This approach can take advantage of the stability of anakinra in the excipient buffer used for Kineret (an FDA approved anakinra product) and the stability of MCMs when they are freeze dried. A second example approach is to add the MCMs to anakinra during formulation and packaging. A formulation with the components combined can be easier to administer in the field and in the clinic.
[0099] As described herein, the MCM can stabilize macromolecules. The diameter of the MCM can also be tailored for any suitable purpose. In some embodiments, the diameter of the MCM is about 1 micrometer (um), about 3 um, about 5 um, about 10 um, about 30 um, about 50 um, about 80 um, about 100 um, about 120 um, about 150 um, about 200 um, about 300 um, or about 500 um. In some embodiments, the diameter of the MCM is at least about 1 micrometer (um), at least about 3 um, at least about 5 um, at least about 10 um, at least about 30 um, at least about 50 um, at least about 80 um, at least about 100 um, at least about 120 um, at least about 150 um, at least about 200 um, at least about 300 um, or at least about 500 um. In some embodiments, the diameter of the MCM is at most about 1 micrometer (um), at most about 3 um, at most about 5 um, at most about 10 um, at most about 30 um, at most about 50 um, at most about 80 um, at most about 100 um, at most about 120 um, at most about 150 um, at most about 200 um, at most about 300 um, or at most about 500 um. The diameter of the MCM can be tailored by using a larger or smaller core material or by the conditions of deposition of the mineral coating on the core material, which conditions can include time of reaction or concentration of components in the simulated body fluid solution. [00100] The diameter of the core and MCM can be measured using laser diffraction. Laser diffraction can provide the Dv (10), Dv (50), and Dv (90) of the particles. An increase in the particle diameter after the coating process can be an indicator of the level of coating. Particle diameter can also have a large impact on particle performance, including dissolution kinetics, loading of active pharmaceutical ingredients, release rate of active pharmaceutical ingredients, the immune response to the particle after administration. In some embodiments, the starting core materials have a Dv (10), Dv (50) and Dv (90) of approximately 5 um, 15 um, and 30 um respectively. In some embodiments, suitable ranges for the Dv (10), Dv (50), and Dv(90) of uncoated materials can span from 0.01-10 um, 0.01-30 um, and 0.01 to 50 um respectively. In some embodiments, the coated MCMs have a Dv (10), Dv (50) and Dv (90) of approximately 15 um, 25 um, and 30 um respectively. In some embodiments, suitable ranges for the Dv (10), Dv (50), and Dv(90) of the coated microparticles can span from 0.1-20 um, 0.1-40 um, and 0.1 to 70 um respectively. In some embodiments, the coated MCMs have a Dv(10) that is approximately 3X larger than the Dv(10) of the starting core material. In some embodiments, the coated MCMs have a Dv(50) that is approximately 1.5X larger than the Dv(50) of the starting core material.
[00101] The diameter of the core material and the MCMs can also be measured using staining and visual assessment using imaging software. In some embodiments, the mean particle diameter of the core material is approximately 5 um. In some embodiments, the mean particle diameter of the coated MCM is 8 um to 10 um. In some embodiments, the mean particle diameter of the coated MCM is 5 um-8 um. In other embodiments, the mean particle diameter of the coated MCM is 1 um-5 um. In some embodiments, the mean particle diameter of the coated MCM is 8 um-20 um. In some embodiments, the mean diameter of the coated MCM is 2X the diameter of the starting core material. In some embodiments, the mean diameter of the coated MCM is greater than 2X the diameter of the starting core material.
[00102] The surface area of the microparticle can directly impact the loading capacity, the dissolution rate, and the release rate of the mineral coated microparticle. Additionally, coating of the starting core materials significantly increases the surface area of the particle. In some embodiments the surface area of the starting core material is approximately 1 to 3 m2/g. In some embodiments, the surface area of the starting core material is between 0.1 to 1 m2/g. In some embodiments, the surface area of the coated MCM is approximately 25 m2/g. In some embodiments the surface area of the coated MCM is between 10 m2/g to 50 m2/g. In some embodiments, the surface area of the coated MCM is approximately 10X greater than that of the starting uncoated core material. [00103] The MCM can also be an excipient. As used herein, an "excipient" is a substance formulated alongside the active ingredient of a medication, included for the purpose of long-term stabilization, bulking up solid formulations that contain potent active ingredients in small amounts (thus often referred to as "bulking agents", "fillers", or "diluents"), or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption, reducing viscosity, or enhancing solubility. Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerns such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation or aggregation over the expected shelf life. The selection of appropriate excipients also depends upon the route of administration and the dosage form, as well as the active ingredient and other factors. The MCM can be a stabilizer to increase the halflife of a therapeutic, either alone or in combination with other excipients.
[00104] FIG. 1 shows a micrograph of an example MCM having a nano- structured calcium phosphate mineral coating. These can provide a platform for sustained delivery of biological macromolecules. The mineral coated microparticles offer an injectable and systemic or localized delivery system that can lower the dose and off-target side-effects when compared to bolus injection.
[00105] Mineral coated microparticles offer a delivery system that can sustainably release proteins while maintaining their activity. In some cases, these microparticles can remain localized when injected in vivo and offer a localized delivery system which can allow for lower therapeutic dosages when compared to systemic subcutaneous or intravenous delivery. Further, release of protein from mineral coated microparticles can be tailored by altering the coating composition. In addition, mineral coated microparticles have a high binding capacity for biological macromolecules which allows them to sustainably deliver a suitable dose of protein with little delivery system material. This may widen the applicability of sustained delivery systems.
[00106] As shown in FIG. 3A, delivery of the protein without the MCM can result in rapid clearing of the protein (and the diminution of therapeutic effect) from the body. This can require frequent dosing, as is the case with anakinra not delivered by MCM. In contrast, FIG. 3B shows that sustained release of anakinra from an MCM can sustain the dose in the therapeutic window for an extended period of time, ultimately requiring less protein and less need for attention from doctors.
[00107] In some embodiments, the formulation includes a mineral coated microparticle, wherein the mineral coated microparticle comprises a core; a mineral coating on the core; and an anti-inflammatory protein. In some embodiments, the core is a nucleation site for coating precipitation. In some embodiments, the protein is adsorbed to the mineral coating. In some embodiments the protein is incorporated throughout the mineral coating. In some embodiments, there are layers of mineral coating on the core. In some embodiments, the protein is adsorbed to multiple layers of mineral coating on the core. In some embodiments, multiple, different active agents are adsorbed to the mineral coating along with a protein. In some embodiments, multiple proteins are adsorbed to the mineral coating.
[00108] There can be additional active agents in the fluid in which the MCMs are suspended ("carrier") during their manufacture and/or upon administration. In some embodiments, the formulation includes a carrier, wherein the carrier is for a mineral coated microparticle, wherein the mineral coated microparticle comprises a core; a mineral coating on the core; and an antiinflammatory protein adsorbed to the mineral coating. In some embodiments, another active agent is adsorbed to the mineral coating along with the protein. In some embodiments, the carrier is a liquid. In some embodiments, the carrier is a solution or a liquid. In some embodiments, the carrier is a gel. In some embodiments the carrier is a gas. In some embodiments, the carrier is a solid. In some embodiments, the carrier contains an active agent. In some embodiments, the active agent is an IL-IRa or an equivalent thereof such as anakinra. In some embodiments, the active agent in the carrier contains the same anti-inflammatory protein adsorbed on or incorporated within the mineral coating.
[00109] Suitable liquid carriers include water, saline, isotonic saline, phosphate buffered saline, Ringer's lactate, and the like. Suitable gel carriers include collagen, hydrogels, polymer gels, polyethylene glycol, and the like.
[00110] Formulations can also include other components such as surfactants, preservatives, and excipients. Surfactants can reduce or prevent surface-induced aggregation of the active agent and the mineral coated microparticles. Various surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbitol fatty acid esters. Amounts can range from about 0.001 and about 4% by weight of the formulation. Pharmaceutically acceptable preservatives include, for example, phenol, o-cresol, m-cresol, p- cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p- hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p- hydroxybenzoate, benzethonium chloride, chlorphenesine (3p- chlorphenoxypropane-l,2-diol) and mixtures thereof. The preservative can be present in concentrations ranging from about 0.1 mg/ml to about 20 mg/ml, including from about 0.1 mg/ml to about 10 mg/ml. A preservative can be used in pharmaceutical compositions such as, but not limited to those described in “Remington: The Science and Practice of Pharmacy, 19th edition, 1995,” which is incorporated herein by reference in its entirety for all purposes. Formulations can include suitable buffers such as sodium acetate, glycylglycine, HEPES (4-(2-hydroxyethyl)-l- piperazineethanesulfonic acid) and sodium phosphate. Excipients include components for tonicity adjustment, antioxidants, and stabilizers used in the preparation of pharmaceutical formulations. Other inactive ingredients include, for example, L-histidine, L-histidine monohydrochloride monohydrate, sorbitol, polysorbate 80, sodium citrate, sodium chloride, and EDTA disodium.
[00111] Suitable core materials on which the mineral coating is formed include polymers, ceramics, metals, glass and combinations thereof in the form of particles. Suitable particles can be, for example, agarose beads, latex beads, magnetic beads, polymer beads, ceramic beads, metal beads (including magnetic metal beads), glass beads and combinations thereof. The microparticle can include ceramics (e.g., hydroxyapatite, beta-tricalcium phosphate (beta-TCP, P-TCP), magnetite, neodymium), plastics (e.g., polystyrene, poly-caprolactone), hydrogels (e.g., polyethylene glycol; poly(lactic-co-glycolic acid), and the like, and combinations thereof. Suitable core materials can be those that dissolve in vivo such as, for example, beta-tricalcium phosphate (beta-TCP, P-TCP).
[00112] Suitable microparticle sizes can range from about 1 pm to about 100 pm in diameter. In some cases, the diameter is about 1 pm, about 5 pm, about 10 pm, about 20 pm, about 30 pm, about 40 pm, about 50 pm, about 60 pm, about 70 pm, about 80 pm, about 90 pm, about 100 pm, about 120 pm, about 150 pm, or about 200 pm. Microparticle diameter can be measured by, for example, measurements taken from microscopic images (including light and electron microscopic images), filtration through a size-selection substrate, and the like.
[00113] Any suitable material can be used as the core upon which the mineral coating is formed. Suitable core materials include those materials non-toxic to humans and animals. Suitable core materials also include those materials that degrade and/or dissolve in humans and animals. Suitable core materials include P-tri calcium phosphate (P-TCP), hydroxyapatite (HA), poly(lactic-co-glycolic acid) (PLGA), and combinations thereof. P-tricalcium phosphate cores are can be suitable as the P-tricalcium phosphate degrades rapidly after mineral coating dissolution. Both P-tricalcium phosphate and hydroxyapatite are can also be suitable cores because they dissolve into calcium and phosphate ions which are easily metabolized by the body. In other embodiments, the core material can be dissolved following mineral coating formation. In other embodiments, the core material is non-degradable. [00114] The mineral coating can include calcium, phosphate, carbonate, and combinations thereof. To prepare a mineral coated microparticle a core material is incubated in a modified simulated body fluid. Simulated body fluid contains the same ion constituents at the same concentrations as human blood plasma. Modified simulated body fluid contains similar, but altered ion constituents as human blood plasma. In some embodiments, the modified simulated body fluid contains twice the concentration of calcium and phosphate as human blood plasma along with the other ionic components of human blood plasma at physiological concentrations. The modified simulated body fluid can include calcium and phosphate, which form the mineral coating on the surface of the core, which results in the mineral coated microparticle. Because the modified simulated body fluid contains a supersaturation of calcium and phosphate, a mineral coating precipitates from solution onto the core material to form the mineral coating. Different mineral coating morphologies can be achieved by varying the amounts and ratios of calcium, phosphate, and carbonate in the modified simulated body solution during coating precipitation. Other ions, or dopants, can also be added to the modified simulated body fluid during coating formation to change the coating composition and/or morphology. Different mineral coating morphologies include, for example, plate-like structure, spherulite-like structure. High carbonate concentration can result in a mineral coating having a plate-like structure. Low carbonate concentration can result in a mineral coating having a spherulite-like structure. The mineral coating morphology can also affect adsorption of the active agent. The mineral coating morphology can also affect the preservation of activity of the active agent release from the mineral coating.
[00115] The modified simulated body fluid (mSBF) for use in the methods of the present disclosure can include from about 5 mM to about 12.5 mM calcium ions, including from about 7 mM to about 10 mM calcium ions, and including about 8.75 mM calcium ions; from about 2 mM to about 12.5 mM phosphate ions, including from about 2.5 mM to about 7 mM phosphate ions, and including from about 3.5 mM to about 5 mM phosphate ions; and from about 4 mM to about 100 mM carbonate ions.
[00116] In some embodiments, the mSBF can further include about 145 mM sodium ions, from about 6 mM to about 9 mM potassium ions, about 1.5 mM magnesium ions, from about 150 mM to about 175 mM chloride ions, about 4 mM HCO3", and about 0.5 mM SO4^" ions. [00117] The pH of the mSBF can range from about 4 to about 7.5, including from about 5.3 to about 6.8, including from about 5.7 to about 6.2, and including from about 5.8 to about 6.1. [00118] Suitable mSBF can include, for example: about 145 mM sodium ions, about 6 mM to about 9 mM potassium ions, about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 150 mM to about 175 mM chloride ions, about 4.2 mM HCO3', about 2 mM to about 5 mM HPCM^" ions, and about 0.5 mM SO4^- ions. The pH of the simulated body fluid may be from about 5.3 to about 7.5, including from about 6 to about 6.8.
[00119] In some embodiments, the mSBF may include, for example: about 145 mM sodium ions, about 6 mM to about 17 mM potassium ions, about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 150 mM to about 175 mM chloride ions, about 4.2 mM to about 100 mM HCO3', about 2 mM to about 12.5 mM phosphate ions, and about 0.5 mM 804^" ions. The pH of the simulated body fluid may be from about 5.3 to about 7.5, including from about 5.3 to about 6.8.
[00120] In some embodiments, the mSBF includes: about 145 mM sodium ions, about 6 mM to about 9 mM potassium ions, from about 5 mM to about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 60 mM to about 175 mM chloride ions, about 4.2 mM to about 100 mM HCO3', about 2 mM to about 5 phosphate ions, about 0.5 mM 804^" ions, and a pH of from about 5.8 to about 6.8, including from about 6.2 to about 6.8.
[00121] In some embodiments, the mSBF includes: about 145 mM sodium ions, about 9 mM potassium ions, about 12.5 mM calcium ions, about 1.5 mM magnesium ions, about 172 mM chloride ions, about 4.2 mM HCO3', about 5 mM to about 12.5 mM phosphate ions, about 0.5 mM SO4^‘ ions, from about 4 mM to about 100 mM CO3^‘, and a pH of from about 5.3 to about 6.0.
[00122] In embodiments that include a layered mineral coating, a core can be incubated in a formulation of modified simulated body fluid. The layer of mineral coating forms on the core during the incubation period of minutes to days. After the initial layer of mineral coating is formed on the core, the mineral coated microparticle can be removed from the modified simulated body fluid and washed. To form a plurality of layers of mineral coating a mineral coated microparticle can be incubated in a second, third, fourth, etc. modified simulated body fluid until an appropriate number of layers of mineral coating is achieved. During each incubation period a new layer of mineral coating forms on the previous layer. These operations are repeated until the appropriate number of layers of mineral coating is achieved.
[00123] During mineral formation, active agents such as proteins can be included in the modified simulated body fluid to incorporate active agents within the layer of mineral coating during mineral formation. The active agent can be a protein or can be a different active agent. Following formation of each layer of mineral, the mineral coated microparticle can then be incubated in a carrier comprising at least one active agent to adsorb the agent to the layer of mineral coating. After incorporating an active agent within a layer of mineral coating and/or adsorbing an active agent to a layer of mineral coating, another layer of mineral coating can be formed by incubating the microparticle in another formulation of modified simulated body fluid. In some cases, layers of mineral coating can incorporate an active agent in the mineral, layers can have an active agent adsorbed to the layer of mineral, the layer of mineral coating can be formed without incorporating an active agent or adsorbing an active agent, and combinations thereof. Mineral coated microparticles having different layers of mineral coating can be prepared by forming a layer of mineral using one formulation of modified simulated body fluid, then incubating the mineral coated microparticle in a different formulation of modified simulated body fluid. Thus, mineral coated microparticles can be prepared to have a plurality of layers of mineral coating wherein each layer is different. Embodiments are also contemplated that include two or more layers of mineral coating that are the same combined with one or more layers of mineral coating that are the different. One of the active agents can be a protein such as an antigen or an mRNA that expresses an antigen when administered to a subject in need of vaccination.
[00124] Tailoring the composition of the mineral coating in the different layers advantageously allows for tailored release kinetics of the active agent or active agents from each layer of the mineral coating. In embodiments where one or more active agents is incorporated within the mineral coating, the active agent can be included in the mSBF. As mineral formation occurs, active agents become incorporated with the mineral coating. In other embodiments, magnetic material can be incorporated into mineral coatings. For example, superparamagnetic iron oxide linked to bovine serum albumin can be incorporated into mineral coatings. Linked proteins (e.g., bovine serum albumin) can adsorb onto the mineral coating to incorporate the magnetic material with the mineral coating. In some embodiments, the mineral coating further includes a dopant. Suitable dopants include halogen ions, for example, fluoride ions, chloride ions, bromide ions, and iodide ions. The dopant(s) can be added with the other components of the mSBF prior to incubating the substrate in the mSBF to form the mineral coating. The dopant ions can alter the dissolution kinetics of the mineral and can thus alter the release kinetics of protein or other active agent from the mineral coating.
[00125] In some embodiments, halogen ions including fluoride ions can be used. Suitable fluoride ions can be provided by fluoride ion-containing agents such as water-soluble fluoride salts, including, for example, alkali and ammonium fluoride salts. Incorporation of fluoride alters the stability of the mineral coating. The fluoride ion-containing agent can be included in the mSBF to provide an amount of up to 100 mM fluoride ions, including from about 0.001 mM to 100 mM, including about 0.01 mM to about 50 mM, including from about 0.1 mM to about 15 mM, and including about 1 mM fluoride ions. Inclusion of one or more dopants in the mSBF can result in the formation of a halogen-doped mineral coating that can have significantly different morphologies and/or dissolution and release kinetics. The different morphology may be beneficial for preserving the activity of the active agent release from the mineral coating. The control of mineral coating dissolution can be beneficial when tailoring the coating to have sufficient release kinetics for the active agent to enhance efficacy. In some embodiments, magnetic materials, including magnetite, magnetite-doped plastics, and neodymium, are used for the microparticle core material. Including magnetic materials results in the formation of MCM for which location and/or movement/positioning of the MCM by application of a magnetic force is enabled. The alternate use of magnetic microparticle core materials can allow for spatial control of where the active agent and/or the protein is delivered. The mineral coatings may be formed by incubating the substrate with the mSBF at a temperature of about 37°C for a period of time ranging from about 3 days to about 10 days.
[00126] To adsorb the protein to the mineral coated microparticle, the mineral coated microparticles can be contacted with a solution containing the protein. This contact can form a protein loaded mineral coated microparticle. Other active agent(s) can also be adsorbed to the mineral coating along with the protein by including them in the solution with the protein. Alternatively, the microparticles can be contacted with a second solution containing other active agent(s) after loading with the protein. Addition of other active agents can make the delivery of protein more efficient or effective. In some embodiments, only a protein is incorporated, adsorbed, or loaded onto or into the mineral coating. As used herein, "active agent" refers to a biologically active molecule. As used herein, “protein loaded mineral coated microparticle” refers to a mineral coated microparticle which has protein adsorbed to the mineral coating and/or has protein incorporated throughout the coating. The protein and/or other active agent(s) can be contacted with the mineral coated microparticle using any suitable method. For example, a solution of the protein and/or other active agent(s) can be pipetted, poured, or sprayed onto the mineral coated microparticle. Alternatively, the mineral coated microparticle can be dipped in a solution including protein and/or other active agent(s) along with the protein. Alternatively, the mineral coated microparticle can be bathed or incubated in a solution containing protein and/or other active agent(s). The protein, and/or other active agent(s) can adsorb to the mineral coating by an electrostatic interaction between the protein or active agent and the mineral coating of the mineral coated microparticle. Suitable active agents include biological molecules. Suitable active agents include proteins, small molecules, hormones, steroids, NSAIDs, cytokines, therapeutic proteins, antibodies, receptor antagonists, or the like. Adsorption of the protein, or other active agents along with the protein, to the mineral coated microparticles can be tailored by changing the mineral constituents (e.g., high carbonate and low carbonate microspheres), by changing the amount of mineral coated microparticles incubated with the protein, or other active agents along, by changing the concentration of protein, or other active agents in the incubation solution, and combinations thereof.
[00127] Additional details regarding methods for producing the modified simulated body fluid (mSBF) and/or for forming or binding molecules to the MCM can be found in "Addition of Mineral-Coated microparticles to soluble interleukin-1 receptor antagonist injected subcutaneously improves and extends systematic interleukin- 1 inhibition" A.E.B. Clements, et. al., Advanced Therapeutics, vol. 1, issue 7, 1800048, November 2018; "Single-dose mRNA therapy via biomaterial-mediated sequestration of overexpressed proteins", Khalil et al., Sci. Adv. 2020; 6; or "Nanostructured mineral coatings stabilize proteins for therapeutic delivery", X. Yu, et al., Adv. Mater. 2017 September, 29(33), each of which is incorporated herein by reference in its entirety for all purposes.
[00128] After completing the mineral coating preparation, the mineral coatings can be analyzed to determine the morphology and composition of the mineral coatings. The composition of the mineral coatings can be analyzed by energy dispersive X-ray spectroscopy, Fourier transform infrared spectrometry, X-ray diffractometry, and combinations thereof.
Suitable X-ray diffractometry peaks can be, for example, at 26° and 31°, which correspond to the (0 0 2) plane, the (2 1 1) plane, the (1 1 2) plane, and the (2 0 2) plane for the hydroxyapatite mineral phase. Suitable X-ray diffractometry peaks can be, for example, at 26° and 31°, which correspond to the (0 0 2) plane, the (1 1 2) plane, and the (3 0 0) plane for carb onate- substituted hydroxyapatite. Other suitable X-ray diffractometry peaks can be, for example, at 16°, 24°, and 33°, which correspond to the octacalcium phosphate mineral phase. Suitable spectra obtained by Fourier transform infrared spectrometry analysis can be, for example, a peak at 450-600 cm‘l, which corresponds to O-P-O bending, and a peak at 900-1200 cm'^, which corresponds to asymmetric P-0 stretch of the PO4^' group of hydroxyapatite. Suitable spectra peaks obtained by Fourier transform infrared spectrometry analysis can be, for example, peaks at 876 cm'^, 1427 cm'^, and 1483 cm'^, which correspond to the carbonate (CO3^‘) group. The peak for HPOd^" can be influenced by adjusting the calcium and phosphate ion concentrations of the mSBF used to prepare the mineral coating. For example, the HPOd^" peak can be increased by increasing the calcium and phosphate concentrations of the mSBF. Alternatively, the HPCM^" peak can be decreased by decreasing the calcium and phosphate concentrations of the mSBF. Another suitable peak obtained by Fourier transform infrared spectrometry analysis can be, for example, a peak obtained for the octacalcium phosphate mineral phase at 1075 cm" ' , which can be influenced by adjusting the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating. For example, the 1075 cm'l peak can be made more distinct by increasing the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating. Alternatively, the 1075 cm'l peak can be made less distinct by decreasing the calcium and phosphate ion concentrations in the simulated body fluid used to prepare the mineral coating.
[00129] Energy dispersive X-ray spectroscopy analysis can also be used to determine the calcium/phosphorus ratio of the mineral coating. For example, the calcium/phosphorus ratio can be increased by decreasing the calcium and phosphate ion concentrations in the mSBF. Alternatively, the calcium/phosphorus ratio may be decreased by increasing the calcium and phosphate ion concentrations in the mSBF. Analysis of the mineral coatings by energy dispersive X-ray spectroscopy allows for determining the level of carbonate (CO3^‘) substitution for PO4^’ and incorporation of HPCM^" into the mineral coatings The mSBF can include calcium and phosphate ions in a ratio ranging from about 10: 1 to about 0.2: 1, including from about 2.5: 1 to about 1 : 1.
[00130] Further, the morphology of the mineral coatings can be analyzed by scanning electron microscopy, for example. Scanning electron microscopy can be used to visualize the morphology of the resulting mineral coatings. The morphology of the resulting mineral coatings can be, for example, a spherulitic microstructure, plate-like microstructure, and/or a net-like microstructure. Suitable average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2 pm to about 42 pm. Suitable average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2 pm to about 4 pm. In some embodiments, average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 2.5 pm to about 4.5 pm. In some embodiments, average diameters of the spherulites of a spherulitic microstructure can range, for example, from about 16 pm to about 42 pm.
[00131] Mineral coated microparticles can be stored for later use, washed and stored for later use, washed and immediately used for adsorption, or immediately used for adsorption without washing. Storage of mineral coated microparticles can include lyophilization.
Methods of Treatment and Prevention of Inflammation [00132] The addition of MCMs to formulated anakinra before subcutaneous administration can significantly improve its ability to inhibit IL-1 induced inflammation.
[00133] With reference to FIG. 4, mice were given a single subcutaneous injection of anakinra or anakinra bound to MCM (DTX020). Serum anakinra concentration was measured after treatment. Intraperitoneal injections of IL-ip were administered 1, 5, and 7 days after treatment and the resulting inhibition of IL-linduced upregulation of IL-6 was examined in serum. DTX020 elevated anakinra concentration 5 and 7 days after administration (left), and inhibited IL-1 induced levels of IL-6 (right) at all timepoints examined (p<0.05), suggesting improved anti-inflammatory activity, mean ± SE.
[00134] MCM-bound anakinra can increase the serum concentration of anakinra for 5 and 7 days after administration when compared to an equivalent dose of anakinra without the addition of MCMs. MCMs can both significantly lower the required dose and decrease the required frequency of dosing of anakinra to inhibit IL-1 induced inflammation and prevent and treat ARDS and other inflammatory conditions. In some cases, a single dose that successfully inhibits inflammation for 7 days can prevent the development of ARDS in high-risk patients. In some cases, a single dose that successfully inhibits inflammation for 1, 2, 3, 4 5, 6 7, 8, 9, 10 or more days can significantly reduce the duration and/or magnitude of a gout flare.
[00135] A combination of anakinra with MCMs can be delivered subcutaneously, not requiring an intravenous (i.v.) line. In some embodiments the subcutaneous injection comprises a single dose injection of anakinra with MCMs. In some embodiments, the subcutaneous single dose injection of anakinra with MCMs can be administered to ARDS patients in conjunction with other concurrent treatments. In some embodiments, the subcutaneous single dose injection of anakinra with MCMs can be administer to gout patients in conjunction with other concurrent treatments.
[00136] In an aspect, provided herein is a method for treating inflammation. The method can include providing a formulation comprising a biologic, admixing the formulation with a mineral coated microparticle (MCM), wherein the MCM adsorbs to the biologic and provides a sustained delivery of biologic when administered in vivo, wherein the inflammation is exemplified by an increase in IL-1 or IL-6 production, and wherein admixed formulation is administered to a subject in need thereof.
[00137] In some embodiments, the method for treating inflammation includes providing a formulation subcutaneously. In some embodiments, the method for treating inflammation includes providing a formulation through localized injection. In some embodiments, the localized injection is into a joint. In some embodiments, the formulation is injected into the synovial cavity of a joint.
[00138] In some embodiments, the inflammation is acute respiratory distress syndrome (ARDS). In some embodiments, the inflammation is associated with graft versus host disease (GVHD). In some embodiments, the inflammation is associated with infection by the SARS- CoV-2 virus. In some embodiments, the inflammation is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy. In some embodiments, the inflammation is chronic or acute inflammation.
[00139] In some embodiments, the inflammation is associate with a gout flare. In some embodiments, the method of treatment reduces the inflammation associated with a gout flare. In some embodiments, the method of treatment reduces the duration of a gout flare. In some embodiments, the method of treatment prevents the formation of urate crystals that induce of gout flare. In some embodiments, the method of treatment prevents inflammation occurring that is caused by urate crystals.
[00140] In some embodiments, the inflammation is systemic. In some embodiments the inflammation is localized in the lungs. In some embodiments the inflammation is localized within a joint.
[00141] In some embodiments, the biologic is interleukin-1 receptor agonist (IL-IRa), or an analog thereof, such as anakinra.
[00142] In some embodiments, the concentration of the MCM in the formulation is the same as the concentration of anakinra. In some embodiments, the concentration of the anakinra is 10X greater than the concentration of the MCMs in the formulation. Suitable ratios of MCM concentration to anakinra concentration can range from 100: 1 to 10: 1, from 1 : 1 to 1 : 10, and from 1 : 10 to 1 : 1000. In some embodiments, suitable concentrations of MCMs can range from 0.1 mg/ml to 10 mg/ml. In some embodiments, suitable concentrations of MCMs can range from 10 ug/ml to 100 ug/ml. In some embodiments, approximately 0.7 mg of MCM is administered subcutaneously per kg of body weight. In some embodiments, approximately 0.1 mg of MCM is administered subcutaneously per kg of body weight. Suitable dosing ranges for MCMs may include 10 ug/kg to 100 ug/ml, 0.1 mg/kg to 1 mg/kg, or 1 mg/kg to 5 mg/kg. In some embodiments, approximately 7 mg of anakinra per kg of body weight is administered subcutaneously. In other embodiments, approximately 1 mg of anakinra per kg of body weight is administered. Suitable dosing ranges of anakinra may include 0.1 mg/kg to 1 mg/kg or 1 mg/kg to 10 mg/kg. [00143] In some embodiments where the formulation is administered into a joint or other localized area, the amount of MCMs or anakinra administered is depends on the volume or mass of tissue where the treatment is administered. For example, the amount of anakinra and MCM in a localized injection used to treat or prevent a gout flare is dependent on the size of the joint. In some embodiments where the formulation is injected into a joint, approximately 1-5 mg of MCMs per ml of synovial fluid within a joint is administered. In some embodiments, approximately 2.5 mg of MCMs per ml of synovial fluid is administered. In some embodiments, approximately 5-50 mg of MCMs per ml of synovial fluid is administered. In some embodiments, approximately 25 mg of MCMs per ml of synovial fluid is administered. In some embodiments, less than 1 mg of MCMs per ml of synovial fluid is administered. In some embodiments, approximately 0.35 mg of MCMs per ml of synovial fluid is administered. In some embodiments for local administration, the ratio of MCMs to anakinra can be 1 : 1.
[00144] In some embodiments, an amount of biologic administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM (e.g., by at least about 50%, at least about 75%, at least about 85%, at least about 90%, or at least about 95%). In some cases, the admixed formulation is provided prophylactically to a subject in need thereof.
[00145] In another aspect, provided herein is a method for reducing the expression of IL-1 and/or IL-6 in vivo, the method comprising a single administration of a formulation to a patient exhibiting elevated serum concentrations of IL-1 and/or IL-6, the formulation comprising a sustained release admixture, wherein the admixture comprises a drug delivery vehicle and IL- IRa, wherein the elevated serum concentration of IL-1 and/or IL-6 is associated with an acute inflammatory event.
[00146] In some embodiments, the serum concentration of IL-1 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 in vivo. In some embodiments, the serum concentration of IL-6 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-6 in vivo. In some embodiments, the serum concentrations of IL-1 and IL-6 are elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 and IL-6 in vivo.
[00147] In some embodiments, no more than one administration of the formulation is performed in a 24 hour, 48 hour, or 72 hour period. In some embodiments, the method further comprises administering a subsequent dose of the formulation after the single administration of the formulation. [00148] In some cases, the serum concentration of IL-1 and/or IL-6 is at least about 2, at least about 5, at least about 10, at least about 20, or at least about 50-times higher than a patient not having the acute inflammatory event.
[00149] The expression IL-1 and/or IL-6 resulting from the single administration can be reduced compared to a single administration of IL-IRa alone (e.g., by at least about 2, 5, 10, 20, or 50-fold). In some cases, the reduction of IL-1 and/or IL-6 serum concentration lasts over a period of 3, 5, or 7 days in vivo. The drug delivery vehicle can be a mineral coated microparticle (MCM).
[00150] In some embodiments, an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
[00151] In another aspect, provided herein is a method for preventing inflammation in vivo, the method comprising administering an admixture of IL-IRa and a mineral coated microparticle (MCM) to a subject having an elevated risk of inflammation.
[00152] In some cases, the subject is at risk of inflammation of immune cells, lymphocytes, or mast cells. In some embodiments, the subject is at risk of respiratory or cardiovascular inflammation.
[00153] In some embodiments, the subject is showing signs of inflammation. In some embodiments, the subject is at an onset of inflammation. In some embodiments, the subject is exhibiting hyperoxia.
[00154] In some embodiments, the subject is experiencing a gout flare. Indications of a gout flare include painful joint swelling and inflammation and/or the presence of urate crystals in the synovial fluid of a joint.
[00155] In some embodiments, the subject is at risk of developing a gout flare. Increased risk can be indicated by high uric acid levels, formation of urate crystals in a joint, or a history of gout flares.
[00156] In some embodiments, the subject has elevated serum concentrations of IL-1 and/or IL-6. The MCM can provide for sustained release of the IL-IRa.
[00157] In some embodiments, the subject has elevated synovial concentrations of IL-1 and/or IL-6 within a joint.
[00158] To summarize, the methods described herein can exhibit a number of advantages over previous therapies for treating ARDS such as associated with COVID-19, including: (1) decreased need for recombinant anakinra, mitigating supply, storage, and stockpiling limitations; (2) does not require i.v. line for delivery, (3) decreased fluid (volume) load relative to anakinra alone, (4) a single dose can be effective for many patients, (5) stability is much greater than altematives, increasing value for field use, and (6) does not require significant medical training for subcutaneous administration. Additionally, the methods described herein can exhibit a number of advantages for treating and preventing gout flares. When used to treat a gout flare by controlling inflammation, the methods described herein can 1) significantly shorten the duration and magnitude of a gout flare, 2) reduce joint damage caused by the gout flare, 3) reduce the number of injections required for treatment, and 4) reduce the amount of biologic required for treatment. When used to prevent gout flares by administering a treatment into a joint, the methods described herein can 1) dramatically reduce the amount of protein required to prevent flares, 2) allow for biologies with short half-lives to be used to prevent gout flares, 3) and lengthen the duration between treatment administration.
[00159] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. It is not intended that the invention be limited by the specific examples provided within the specification. While the invention has been described with reference to the aforementioned specification, the descriptions and illustrations of the embodiments herein are not meant to be construed in a limiting sense. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. Furthermore, it shall be understood that all aspects of the invention are not limited to the specific depictions, configurations or relative proportions set forth herein which depend upon a variety of conditions and variables. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is therefore contemplated that the invention shall also cover any such alternatives, modifications, variations or equivalents. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method for treating inflammation, comprising: a. providing a formulation comprising a biologic; b. admixing the formulation with a mineral coated microparticle (MCM) to provide an admixed formulation, wherein the MCM adsorbs the biologic and provides a sustained delivery of the biologic when administered in vivo, wherein the inflammation is exemplified by an increase in IL-1 or IL-6 production, and wherein the admixed formulation is administered to a subject in need thereof.
2. The method of Claim 1, wherein the inflammation is acute respiratory distress syndrome (ARDS).
3. The method of Claim 1, wherein the inflammation is associated with graft versus host disease (GVHD).
4. The method of Claim 1, wherein the inflammation is associated with infection by the SARS-CoV-2 virus.
5. The method of Claim 1, wherein the inflammation is associated with cytokine storm syndrome.
6. The method of Claim 1, wherein the inflammation is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy.
7. The method of Claim 1, wherein the inflammation is acute inflammation.
8. The method of Claim 1 wherein the biologic is interleukin-1 receptor agonist (IL-IRa), or an analog thereof.
9. The method of Claim 1 wherein the biologic is anakinra or a derivative thereof.
10. The method of Claim 1, wherein an amount of biologic administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM.
11. The method of Claim 1, wherein the admixed formulation is provided prophylactically to a subject in need thereof.
12. A method for reducing the expression of IL-1 or IL-6, the method comprising: administering a single dose of a formulation to a patient exhibiting elevated serum concentrations of IL-1 or IL-6 associated with an acute inflammatory event, wherein the formulation comprises a sustained release admixture, wherein the sustained release admixture comprises a drug delivery vehicle and IL-IRa. The method of Claim 12, wherein the elevated serum concentration of IL-1 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 in vivo. The method of Claim 12, wherein the elevated serum concentration of IL-6 is elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-6 in vivo. The method of Claim 12, wherein the elevated serum concentrations of IL-1 and IL-6 are elevated in association with the acute inflammatory event and a single administration of the formulation reduces the expression of IL-1 and IL-6 in vivo. The method of Claim 12, wherein no more than one administration of the formulation is performed in a 24 hour, 48 hour, or 72 hour period. The method of Claim 16, further comprising administering a subsequent dose of the formulation after the single administration of the formulation. The method of Claim 12, wherein the elevated serum concentration of IL-1 or IL-6 is at least about 2, at least about 5, at least about 10, at least about 20, or at least about 50- times higher than a patient not having the acute inflammatory event. The method of Claim 12, wherein the expression of IL-1 or IL-6 resulting from the single administration is reduced compared to a single administration of IL-IRa alone. The method of Claim 12, wherein the expression of IL-1 or IL-6 is reduced by at least about 2, 5, 10, 20, or 50-fold. The method of Claim 12, wherein the expression of IL-1 or IL-6 lasts over a period of 3, 5, or 7 days in vivo. The method of Claim 12, wherein the acute inflammatory event is acute respiratory distress syndrome (ARDS). The method of Claim 12, wherein the acute inflammatory event is graft versus host disease (GVHD). The method of Claim 12, wherein the acute inflammatory event is associated with infection by the SARS-CoV-2 virus or influenza virus. The method of Claim 12, wherein the acute inflammatory event is associated with treatment by chimeric antigen receptor T-cell (CAR-T) therapy. The method of Claim 12, wherein the drug delivery vehicle is a mineral coated microparticle (MCM). The method of Claim 26, wherein an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM. A method for preventing inflammation, the method comprising administering an admixture of IL-IRa and a mineral coated microparticle (MCM) to a subject having an elevated risk of inflammation. The method of Claim 28, wherein the subject exhibits or is at risk of a pulmonary edema. The method of Claim 28, wherein the subject requires a high fraction of inspired oxygen (FiO2). The method of Claim 28, wherein the subject has been infected with the SARS-Cov-2 virus or influenza virus. The method of Claim 28, wherein the subject has received a transplant. The method of Claim 28, wherein the subject has undergone chimeric antigen receptor T- cell (CAR-T) therapy. The method of Claim 28, wherein the subject is at risk of inflammation of immune cells, lymphocytes, or mast cells. The method of Claim 28, wherein the subject is at risk of respiratory or cardiovascular inflammation. The method of Claim 28, wherein the subject is showing symptoms of inflammation. The method of Claim 28, wherein the subject is at an onset of inflammation. The method of Claim 28, wherein the subject is exhibiting hyperoxia. The method of Claim 28, wherein the subject has elevated serum concentrations of IL-1 or IL-6. The method of Claim 28, wherein no more than one administration of the admixture is performed in a 24 hour, 48 hour, or 72 hour period. The method of Claim 40, further comprising administering a subsequent dose of the admixture after the single administration of the admixture. The method of Claim 28, wherein the MCM provides for sustained release of the IL-IRa. The method of Claim 28, wherein an amount of IL-IRa administered to the subject is reduced when admixed with the MCM compared to when not admixed with an MCM. The method of Claim 1, wherein the inflammation is gout. The method of Claim 12, wherein the acute inflammatory event is associated with gout. A method for reducing the expression of interleukin (IL)- 1 P, tumor necrosis factor (TNF)-a, or IL-6, the method comprising: administering a single dose of a formulation to a patient exhibiting elevated serum concentrations of interleukin (IL)-ip, tumor necrosis factor (TNF)-a or IL-6 associated with an inflammatory event, wherein the formulation comprises a sustained release admixture, and wherein the sustained release admixture comprises a drug delivery vehicle and IL-lRa. The method of Claim 28, wherein the subject exhibits or is at risk of a polyarthritis.
PCT/US2021/048352 2020-09-01 2021-08-31 Methods for treating or preventing inflammatory events WO2022051247A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063073135P 2020-09-01 2020-09-01
US63/073,135 2020-09-01

Publications (1)

Publication Number Publication Date
WO2022051247A1 true WO2022051247A1 (en) 2022-03-10

Family

ID=80492123

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/048352 WO2022051247A1 (en) 2020-09-01 2021-08-31 Methods for treating or preventing inflammatory events

Country Status (1)

Country Link
WO (1) WO2022051247A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100209357A1 (en) * 2006-08-10 2010-08-19 Levitt Roy C Localized Therapy of Lower Airways Inflammatory Disorders with Proinflammatory Cytokine Inhibitors
WO2011066378A2 (en) * 2009-11-24 2011-06-03 Alder Biopharmaceuticals, Inc. Antagonists of il-6 to prevent or treat thrombosis
WO2014194364A1 (en) * 2013-06-05 2014-12-11 Monash University Method of preventing or treating bronchopulmonary dysplasia (bpd) and pulmonary arterial hypertension (pah)
US20190358344A1 (en) * 2018-05-23 2019-11-28 Dianomi Therapeutics, Inc. Mineral coated microparticles for sustained delivery of steroids
US20200164087A1 (en) * 2017-04-03 2020-05-28 Wisconsin Alumni Research Foundation Mineral coated microparticles for sustained delivery of biologically active molecules
US20200170958A1 (en) * 2017-07-05 2020-06-04 Wisconsin Alumni Research Foundation Mineral Coated Microparticles for Co-Delivery of Anti-Inflammatory Molecules with Nucleic Acids to Improve Gene Delivery Outcomes
WO2021167957A1 (en) * 2020-02-20 2021-08-26 Dianomi Therapeutics, Inc. Inorganic coatings for the sustained release of bioactive compounds

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100209357A1 (en) * 2006-08-10 2010-08-19 Levitt Roy C Localized Therapy of Lower Airways Inflammatory Disorders with Proinflammatory Cytokine Inhibitors
WO2011066378A2 (en) * 2009-11-24 2011-06-03 Alder Biopharmaceuticals, Inc. Antagonists of il-6 to prevent or treat thrombosis
WO2014194364A1 (en) * 2013-06-05 2014-12-11 Monash University Method of preventing or treating bronchopulmonary dysplasia (bpd) and pulmonary arterial hypertension (pah)
US20200164087A1 (en) * 2017-04-03 2020-05-28 Wisconsin Alumni Research Foundation Mineral coated microparticles for sustained delivery of biologically active molecules
US20200170958A1 (en) * 2017-07-05 2020-06-04 Wisconsin Alumni Research Foundation Mineral Coated Microparticles for Co-Delivery of Anti-Inflammatory Molecules with Nucleic Acids to Improve Gene Delivery Outcomes
US20190358344A1 (en) * 2018-05-23 2019-11-28 Dianomi Therapeutics, Inc. Mineral coated microparticles for sustained delivery of steroids
WO2021167957A1 (en) * 2020-02-20 2021-08-26 Dianomi Therapeutics, Inc. Inorganic coatings for the sustained release of bioactive compounds

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CLEMENTS ET AL.: "Microparticles Locally Deliver Active Interleukin-1 Receptor Antagonist In Vivo", ADVANCED HEALTHCARE MATERIALS, vol. 7, no. 16, - 4 July 2018 (2018-07-04), XP055914190, Retrieved from the Internet <URL:https://pubmed.ncbi.nlm.nih.gov/29974661> [retrieved on 20211103] *
GHOSH ET AL.: "Treatment of Acute Gouty Arthritis in Complex Hospitalized Patients With Anakinra", ARTHRITIS CARE & RESEARCH, vol. 65, no. 8, 6 May 2013 (2013-05-06), pages 1381 - 1384, XP055914188 *
HUANG ET AL.: "Targeting inflammation and cytokine storm in COVID-19", PHARMACOLOGICAL RESEARCH, 27 June 2020 (2020-06-27), XP055914187, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7320704/pdf/main.pdf> [retrieved on 20211223] *
MATOS ET AL.: "DoD COVID-19 Practice Management Guide: Clinical Management of COVID-19", DEFENSE HEALTH AGENCY, 18 June 2020 (2020-06-18), Retrieved from the Internet <URL:https://appa.dtic.mil/sti/pdfs/AD1097348.pdf> [retrieved on 20211223] *

Similar Documents

Publication Publication Date Title
JP6416841B2 (en) Pharmaceutical formulation
TWI725973B (en) Anti-cgrp antibody formulation
JP2018138578A (en) Rapid-acting insulin compositions
EP3532029B1 (en) Liquid pharmaceutical composition
JP2023166014A (en) Collagen 7 compositions and methods of using the same
WO2021079337A1 (en) Pharmaceutical formulation of anti-her2 antibody and preparation thereof
Jeswani et al. Development and optimization of paclitaxel loaded Eudragit/PLGA nanoparticles by simplex lattice mixture design: Exploration of improved hemocompatibility and in vivo kinetics
JP7170031B2 (en) Mineral-coated microparticles for sustained delivery of biologically active molecules
WO2022051247A1 (en) Methods for treating or preventing inflammatory events
JP2023123738A (en) Treatment of gastrointestinal bleeding in patients with severe von Willebrand disease by administration of recombinant VWF
TW202237151A (en) Treatment of hemophilia with fitusiran
US11660345B2 (en) Method and composition for enhancing the delivery of anti-platelet drugs for the treatment of acute stroke
US20230285550A1 (en) Compositions and methods for vaccine delivery
JP2013520480A (en) Stable composition of factor IX
JP6852049B2 (en) Etanercept composition with improved stability
US20200009212A1 (en) Cyclodextrin-nle3-a(1-7) compositions and their use

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: 21864968

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21864968

Country of ref document: EP

Kind code of ref document: A1