WO2018111903A1 - Préparations salines de peptides splunc1 - Google Patents

Préparations salines de peptides splunc1 Download PDF

Info

Publication number
WO2018111903A1
WO2018111903A1 PCT/US2017/065862 US2017065862W WO2018111903A1 WO 2018111903 A1 WO2018111903 A1 WO 2018111903A1 US 2017065862 W US2017065862 W US 2017065862W WO 2018111903 A1 WO2018111903 A1 WO 2018111903A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
sodium channel
pharmaceutical composition
homolog
Prior art date
Application number
PCT/US2017/065862
Other languages
English (en)
Inventor
Timothy Morgan CROWDER
David W. Scott
John Clayton TAYLOR
Original Assignee
Spyryx Biociences, 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 Spyryx Biociences, Inc. filed Critical Spyryx Biociences, Inc.
Priority to CA3046361A priority Critical patent/CA3046361A1/fr
Priority to EP17832396.0A priority patent/EP3554527A1/fr
Publication of WO2018111903A1 publication Critical patent/WO2018111903A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/12Mucolytics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids

Definitions

  • the present invention relates to optimized peptides that are specialized non-naturally occurring peptides with improved ability to bind to sodium channels and inhibit activation of the sodium channels.
  • the invention further relates to methods for regulating sodium absorption and fluid volume and treating disorders responsive to modulating sodium absorption by activity of sodium channels.
  • Epithelial mucosal surfaces are lined with fluids whose volume and composition are precisely controlled.
  • a thin film of airway surface liquid helps protect mammalian airways from infection by acting as a lubricant for efficient mucus clearance (Hobbs et al, J. Physiol. 591: 4377 (2013), Knowles et al, J. Clin. Invest. 109:571 (2002)).
  • This layer moves cephalad during mucus clearance and excess liquid that accumulates as two airways converge is eliminated by Na + - modulated airway surface liquid absorption with Na + passing through the epithelial Na + channel (ENaC) (Hobbs et al, J. Physiol.
  • ENaC must be cleaved by intracellular furin-type proteases and/or extracellular channel activating proteases (CAPs) such as prostasin to be active and to conduct Na + (Planes et al., Curr. Top. Dev. Biol. 78:23 (2007);
  • CAPs extracellular channel activating proteases
  • ENaC can also be cleaved and activated by exogenous serine proteases such as trypsin, an action that is attenuated by the protease inhibitor aprotinin (Vallet et al., Nature 389:607 (1997)).
  • serine proteases such as trypsin
  • aprotinin an action that is attenuated by the protease inhibitor aprotinin
  • the Short Palate Lung and Nasal epithelial Clone (SPLUNC1) protein comprises up to 10% of the total protein in the airway surface liquid and can readily be detected in both nasal lavage and tracheal secretions (Bingle, C. D., and Craven, C. J. (2002) PLUNC: a novel family of candidate host defense proteins expressed in the upper airways and nasopharynx Hum Mol Genet 11, 937 ; Campos, M. A., et al. (2004) Purification and characterization of PLUNC from human tracheobronchial secretions Am J Respir Cell Mol Biol 30, 184; Lindahl, M., Stahlbom, B., and Lipson, C.
  • SPLUNC1 Short Palate Lung and Nasal epithelial Clone
  • SPLUNC1 appears to be a volume sensing molecule since it can be secreted onto the mucosal surface of the superficial epithelia where ENaC is expressed (Bartlett et al, J. Leukoc. Biol. 83: 1201 (2008); Bingle et al, J. Pathol. 205:491 (2005)). Furthermore, SPLUNC1 has been demonstrated to contain a subdomain that functions as an inhibitor of ENaC through its N-terminal domain.
  • the present invention discloses novel specialized non-naturally occurring peptides that mimic the properties of SPLUNC1 in regulation of sodium channels by binding to and inhibiting ion transport to regulate sodium absorption and fluid volume and treat disorders responsive to modulating sodium absorption.
  • the present disclosure is based, in part, on the surprising finding that hypertonic formulations containing ENaC inhibitory peptides are more effective in vitro and in vivo compared to corresponding isotonic formulations.
  • formulations comprising ENaC inhibitory peptide SPX-101 in hypertonic saline significantly improved airway surface liquid (ASL) height in CFTR-mutant human bronchial epithelial cells in comparison to formulations in isotonic saline.
  • a formulation of SPX- 101 in 3% saline significantly increased ASL height by over 250% compared to the effect obtained using SPX-101 in isotonic saline.
  • the results demonstrate that hypertonic saline and SPX-101 are synergistic in increasing ASL height, and that the formulation produce an effect that was greater than additive compared to the effect conferred by hypertonic saline alone or SPX-101 alone.
  • the present invention is based, in part, on the design of specialized non-naturally occurring peptides to regulate the activity of sodium channels.
  • the invention relates to a method of inhibiting the activation of a sodium channel, comprising contacting a sodium channel with a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the sodium channel is an epithelial sodium channel (ENaC).
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • Another aspect of the invention relates to a method of inhibiting sodium absorption through a sodium channel, comprising contacting the sodium channel with a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • a further aspect of the invention relates to a method of increasing the volume of fluid lining an epithelial mucosal surface, comprising contacting a sodium channel present on the epithelial mucosal surface with a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • Another aspect of the invention relates to a method of reducing the level of a sodium channel present on the surface of a cell, comprising contacting the sodium channel with a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • a further aspect of the invention relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • Another aspect of the invention relates to a method of regulating salt balance, blood volume, and/or blood pressure in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the specialized non-naturally occurring peptide or a functional fragment thereof binds to the sodium channel.
  • Another aspect of the invention relates to use of a specialized non- naturally occurring peptide or a functional fragment thereof that mimics the sodium channel binding domain of a PLUNC protein and binds to a sodium channel, wherein cleavage of the sodium channel by a protease is inhibited when bound to the peptide, for regulating salt balance, blood volume, and/or blood pressure in a subject in need thereof.
  • Another aspect of the disclosure relates to a kit comprising the peptide of the invention.
  • Another aspect of the invention relates to the use of a specialized non- naturally occurring peptide or a functional fragment thereof for the preparation of a medicament to treat a disorder responsive to inhibition of sodium absorption in a subject in need thereof.
  • FIG. 1 shows the sequence of S18 (SEQ ID NO:l).
  • the residues of subsequence A are essential for ENaC interaction, while the residues of subsequence B were found not to contribute.
  • FIGs. 2A-2B show the 1 st half (LPVPLDQT (SEQ ID NO: 113) but not the remainder (DQTLPLNVNP (SEQ ID NO: 114) of S18 is required for inhibition of ENaC and preservation of ASL height.
  • FIG. 3A shows that S18 (SEQ ID NO:l) and
  • aaLPVPLDQTLPLNVNPaa (SEQ ID NO:2) have equal potency and efficacy, despite SEQ ID NO:2 being flanked by D-alanines.
  • Fig 3B shows that SI 8 and
  • aaLPVPLDQTaa (SEQ ID NO:3) have equal potency and efficacy, despite (SEQ ID NO:3) being flanked by D-alanines.
  • Fig. 3A also shows the relative potency of a sample of peptides, including aaLPNlePLDQTaa (SEQ ID NO:5), which displays increased potency, relative to S18 or SEQ ID NO:4.
  • FIG. 4 shows an additional comparison of S18 and SEQ ID NO:5, and increased potency of SEQ ID NO: 5.
  • FIGs. 5A-5U show the results of experiments analyzing the effects of various peptides on internalization of alpha-ENaC in HEK293T cells and effects on cell viability when GFP-tagged alpha-ENaC is co-expressed with beta and gamma ENaC.
  • EC50 values are shown at the bottom of several of the figures.
  • ADG is a negative control peptide with the sequence: NH3-ADGGLLLLNNPPPPQTVV-NH2 (SEQ ID NO:143).
  • the peptides used in these experiments were S18 (SEQ ID NO:l), SEQ ID NO:2, SEQ ID NO:141, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:128, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, SEQ ID NO:134, SEQ ID NO:
  • peptides of SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 16, SEQ ID NO:17, SEQ ID NO:18, and SEQ ID NO:136 each with an aa cap at both the N- and the C-terminus.
  • amiloride which inhibits ENaC by another mechanism and does not reduce the amount of functional receptor on the surface of cells.
  • FIG. 6 shows the results of an experiment analyzing the effects of various peptides on internalization of alpha-ENaC in HEK293T cells when GFP-tagged alpha-ENaC is co-expressed only with gamma ENaC and no beta-ENaC.
  • SEQ ID NO: 143 negative control peptide
  • water vehicle
  • SEQ ID NO:l water
  • SEQ ID NO:128 SEQ ID NO:128
  • SEQ ID NO:l AND SEQ ID NO: 128 are effective in reducing alpha-ENaC when beta- ENaC is co-expressed, no effect is observed in this experiment when beta-ENaC is not present.
  • FIG. 7A shows the results of an experiment analyzing the effect on percent survival of ENaC-Tg C57BL:FVB mice after treatment with the S18 peptide (SEQ ID NO:l).
  • S18 100 mM solution
  • FIG. 7B shows the results of an experiment analyzing the effect on percent survival of ENaC-Tg C57BL:FVB mice after treatment with inhaled SEQ ID NO: 142 or SEQ ID NO: 128.
  • FIGs. 7C and 7D show the results of an experiment analyzing the effect on percent survival of ENaC-Tg C57BL:C3H mice after treatment with SEQ ID NO: 129 or SEQ ID NO: 128.
  • FIG. 7E shows the results of an experiment analyzing the effect on percent survival (left panel) and weight gain (right panel) of ENaC-Tg C57BL:C3H mice after treatment with a once daily dose of SEQ ID NO: 128.
  • FIG. 7F shows the results of an experiment analyzing the effect on percent survival of ENaC-Tg C57BL:C3H mice after treatment with SEQ ID NO: 127, SEQ ID NO: 134, or SEQ ID NO: 136.
  • FIGs. 8A-8C are graphs showing the results of an experiment analyzing the effect of SEQ ID NO:128 (aaLPIPLDQTaa) on CFTRinhibitor-172 (CFTRinh-172) induced slowing of tracheal mucus velocity (TMV) in sheep.
  • SEQ ID NO: 128 was found to reverse CFTRinh induced slowing of TMV. The reversal was dose dependent.
  • Fig. 8B also shows treatment with amiloride, which inhibits ENaC by another mechanism and does not reduce the amount of functional receptor on the surface of cells.
  • FIG. 9 shows that SPX-101 binds selectively to ENaC.
  • A The chemical structure of SPX-101.
  • B Representative western blot images of biotin- tagged SPX-101 pulldowns from ENaC transfected HEK293T cells.
  • FIG. 10 shows that SPX-101 induces internalization of ENaC.
  • A HBECs from healthy donors were treated with 1 and 10 ⁇ SPX-101, 100 ⁇ amiloride, or 10 ⁇ control peptide. Surface proteins were labeled with Sulfo-NHS- biotin and enriched by streptavidin pulldown. Enriched lysates and input control were western blotted for ⁇ , ⁇ , and ⁇ ENaC and total protein in the blots was assessed by Ponceau S staining.
  • FIG. 11 shows that SPX-101 decreases ENaC current. Amiloride- sensitive current was determined in healthy (A) and CF (B) HBECs two hours after administration of SPX-101. Data are inclusive of five independent experiments for both healthy and CF HBECs. Significance was determined by Wilcoxon Matched Pairs Test, (p-value - ** ⁇ 0.005, *** ⁇ 0.0005).
  • FIG. 12 shows that SPX-101 increases survival of ENaC-Tg mice.
  • A Kaplan-Meier survival curve of PENaC-Tg mice treated once-daily with 0.18, 0.9, or 1.8 mg/kg SPX-101, 1.8 mg/kg control peptide, or saline starting two days after birth.
  • B Weight gain in mice described in (A).
  • C Leukocyte composition in the BALF of mice treated in (A).
  • FIG. 13 shows that SPX-101 increases mucus transport in PENaC-Tg mice. Mucus transport was assessed as velocity (A), directionality (B), and the product of those two measurements deemed mucus transport index (MTI) (C).
  • FIG. 14 shows that SPX-101 restores tracheal mucus velocity in an ovine model of CF.
  • SPX-101 1-4 mg/kg
  • control peptide 4 mg/kg
  • 0.9% isotonic saline was administered via nebulization and TMV measured hourly for eight hours (A).
  • sheep were nebulized with amiloride (0.06 mg/kg) formulated in isotonic (0.9%) or hypertonic saline (4.2%) (B), 1 mg/kg SPX-101 in hypertonic saline (4.2%) (C), and 2 mg/kg SPX-101 in hypertonic saline (D).
  • Data for 0.9% saline is the same in A-D.
  • Data for 4.2% saline is the same in B-D.
  • the data for 1 mg/kg SPX-101 in 0.9% saline is the same in A and C and the data for 2 mg/kg SPX-101 in 0.9% saline is the same in A and D. All dosing was conducted in at least three independent sheep.
  • FIG. 15 shows a dose-dependent effect of saline in presence or absence of SPX-101 on ASL height maintained in vitro by CFTR-mutant human bronchial epithelial cells.
  • the data show that treating cells with a formulation of SPX-101 in hypertonic saline solution significantly increased ASL height compared to a control formulation of SPX-lOlin isotonic saline. More importantly, the effect of the formulation of SPX-101 in hypertonic saline solution on ASL height was greater than additive (i.e. , synergistic) compared to the effect achieved via treatment with hypertonic saline alone or SPX-101 alone.
  • additive i.e. , synergistic
  • FIG. 16 shows the in vivo effect of formulations containing SPX-101 (1 mg/kg) in hypertonic saline (2.7%, 4.2% or 7.0% saline) or normal saline over a 12-hour period in restoring tracheal mucus velocity (TMV) in sheep that were treated with a CFTR inhibitor (CFTRinh-172).
  • Amino acids are represented herein in the manner recommended by the IUPAC-IUB Biochemical Nomenclature Commission, or (for amino acids) by either the one-letter code, or the three letter code, both in accordance with 37 C.F.R. ⁇ 1.822 and established usage.
  • the total of ten or less additional amino acids includes the total number of additional amino acids on both ends added together.
  • the term "materially altered,” as applied to peptides of the invention, refers to an increase or decrease in binding activity (e.g. , to a sodium channel or specialized non-naturally occurring peptide) of at least about 50% or more as compared to the activity of a peptide consisting of the recited sequence.
  • modulate refers to enhancement (e.g. , an increase) or inhibition (e.g. , a decrease) in the specified level or activity.
  • the term "enhance” or “increase” refers to an increase in the specified parameter of at least about 1.25-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 8- fold, 10-fold, twelve-fold, or even fifteen-fold.
  • inhibitor or “reduce” or grammatical variations thereof as used herein refers to a decrease or diminishment in the specified level or activity of at least about 15%, 25%, 35%, 40%, 50%, 60%, 75%, 80%, 90%, 95% or more. In particular embodiments, the inhibition or reduction results in little or essentially no detectible activity (at most, an insignificant amount, e.g. , less than about 10% or even 5%).
  • contact refers to bringing the specialized non-naturally occurring peptide and the sodium channel in sufficiently close proximity to each other for one to exert a biological effect on the other.
  • contact means binding of the specialized non-naturally occurring peptide to the sodium channel.
  • a "therapeutically effective” amount as used herein is an amount that provides some improvement or benefit to the subject. Alternatively stated, a
  • therapeutically effective amount is an amount that will provide some alleviation, mitigation, or decrease in at least one clinical symptom in the subject. Those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject.
  • fragment as applied to a peptide, will be understood to mean an amino acid sequence of reduced length relative to a reference peptide or amino acid sequence and comprising, consisting essentially of, and/or consisting of an amino acid sequence of contiguous amino acids identical to the reference peptide or amino acid sequence. Such a peptide fragment according to the invention may be, where appropriate, included in a larger polypeptide of which it is a constituent.
  • such fragments can comprise, consist essentially of, and/or consist of peptides having a length of at least about 4, 5, 6, 7, 8, 9, 10, or more consecutive amino acids of a peptide or amino acid sequence according to the invention. In other embodiments, such fragments can comprise, consist essentially of, and/or consist of peptides having a length of less than about 10, 9, 8, 7, 6, 5, 4, or less consecutive amino acids of a peptide or amino acid sequence according to the invention.
  • protein and “polypeptide” are used interchangeably and encompass both peptides and proteins, unless indicated otherwise.
  • a "fusion protein” is a polypeptide produced when two heterologous nucleotide sequences or fragments thereof coding for two (or more) different polypeptides not found fused together in nature are fused together in the correct translational reading frame.
  • Illustrative fusion polypeptides include fusions of a peptide of the invention (or a fragment thereof) to all or a portion of glutathione-S-transferase, maltose-binding protein, or a reporter protein (e.g. , Green Fluorescent Protein, ⁇ - glucuronidase, ⁇ -galactosidase, luciferase, etc.), hemagglutinin, c-myc, FLAG epitope, etc.
  • a “functional” peptide or “functional fragment” is one that substantially retains at least one biological activity normally associated with that peptide (e.g. , binding to or inhibiting a sodium channel).
  • the "functional" peptide or “functional fragment” substantially retains all of the activities possessed by the unmodified peptide.
  • substantially retains biological activity, it is meant that the peptide retains at least about 20%, 30%, 40%, 50%, 60%, 75%, 85%, 90%, 95%, 97%, 98%, 99%, or more, of the biological activity of the native polypeptide (and can even have a higher level of activity than the native peptide).
  • a “nonfunctional" peptide is one that exhibits little or essentially no detectable biological activity normally associated with the peptide (e.g. , at most, only an insignificant amount, e.g. , less than about 10% or even 5%).
  • Biological activities such as protein binding and sodium channel inhibitory activity can be measured using assays that are well known in the art and as described herein.
  • a first aspect of the invention relates to the ability of specialized non- naturally occurring peptides to bind to a sodium channel and prevent activation of the sodium channel, thereby inhibiting the flow of sodium ions.
  • one aspect of the present invention relates to a method of inhibiting the activation of a sodium channel, comprising contacting (e.g., binding) a sodium channel with a specialized non-naturally occurring peptide or a functional fragment thereof.
  • the sodium channel is an epithelial sodium channel (ENaC), e.g., human ENaC, or a non-human mammalian ENaC.
  • the sodium channel is one that is similar in sequence and/or structure to ENaC, such as acid-sensing ion channels (ASIC).
  • ASIC acid-sensing ion channels
  • the inhibition of sodium channel activation can be measured by any method known in the art or disclosed herein, including, without limitation, measuring sodium flow or change in potential across a membrane, across a cell, or across a natural or artificial lining.
  • the inhibition can be at least about 20%, e.g. , at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%.
  • the method of inhibiting the activation of a sodium channel can be carried out, e.g., on an isolated sodium channel, a sodium channel in an artificial membrane, or a sodium channel in a cell.
  • the sodium channel is present in an isolated cell, e.g. , a cultured primary cell or cell line.
  • the isolated cell is part of an epithelial cell culture, e.g. , a natural or artificial epithelial lining, e.g. , a cell culture in a device (such as an Ussing chamber) in which characteristics such as ion flow and/or potential can be measured across lining.
  • the cell is part of an isolated tissue or a tissue culture.
  • the cell can be present in an animal, e.g. , an animal that is a disease model or a subject in need of treatment.
  • the step of contacting (e.g. , binding) the sodium channel with a specialized non-naturally occurring peptide comprises delivering the specialized non-naturally occurring peptide or a functional fragment or homolog thereof to a cell comprising the sodium channel.
  • the term "homolog” is used to refer to a polypeptide which differs from the disclosed specialized non-naturally occurring peptide by modifications to the specialized non-naturally occurring peptide, but which significantly retains a biological activity of the disclosed non-naturally occurring peptide. Minor modifications include, without limitation, changes in one or a few amino acid side chains, changes to one or a few amino acids (including deletions, insertions, and substitutions), changes in stereochemistry of one or a few atoms, and minor
  • substantially retains refers to a fragment, homolog, or other variant of a peptide that retains at least about 20% of the activity of the naturally occurring peptide (e.g. , binding to a sodium channel), e.g. , about 30%, 40%, 50% or more.
  • Other biological activities, depending on the peptide may include enzyme activity, receptor binding, ligand binding, induction of a growth factor, a cell signal transduction event, etc.
  • the method comprises delivering to a cell comprising a sodium channel an isolated specialized non-naturally occurring peptide.
  • the specialized non-naturally occurring peptide comprises, consists essentially of, or consists of the disclosed specialized non-naturally occurring peptide or a functional fragment thereof.
  • the isolated specialized non-naturally occurring peptide comprises, consists essentially of, or consists of an amino acid sequence that is at least 70% identical, e.g. , at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the publicly known amino acid sequence or a functional fragment thereof.
  • the peptides comprise a portion of the natural amino acid sequence of a PLUNC protein with one or more conservative substitutions with natural or non-natural amino acids and/or one or more additions of non-natural amino acids. Conservative substitutions are described below. In some embodiments, the peptides comprise one or more terminal modifications as described below.
  • Non- limiting examples of peptides of the invention are disclosed in Table 1 below.
  • the peptides of the invention may comprise one or more additional residues at the amino- and/or carboxyl-terminal ends.
  • the one or more additional residues are D-alanines.
  • a peptide may comprise one or two D-alanines at the amino- and/or carboxyl-terminal ends.
  • the disclosure further relates to formulations comprising specialized non-naturally occurring peptides of the disclosure (e.g., SEQ ID NO: 127, SEQ ID NO: 128) and saline.
  • the formulation comprises an ENaC inhibitory peptide and hypertonic saline.
  • Hypertonic saline refers to any saline solution with a concentration of sodium chloride (NaCl) higher than physiological concentration (about 0.9 (w/v)).
  • the formulations of the disclosure may include, e.g., about 1%- 10% saline; about 2%-8% saline; about 3%-7% saline, including all sub-ranges in between, e.g.
  • the formulations of the disclosure may include, e.g., about 1.2%, 1.4%, 1.6%, 1.8%, 2.0%, 2.2%, 2.4%, 2.6%, 2.8%, 3.0%, 3.2%, 3.4%, 3.6%, 3.8%, 4.0%, 3.0%, 4.2%, 4.4%, 4.6%, 4.8%, 5.0%, 5.2%, 5.4%, 5.6%, 5.8%, 6.0%, 6.2%, 6.4%, 6.6%, 6.8%, 7.0%, 7.2%, 7.4%, 7.6%, 7.8%, 8.0%, 8.5%, 9%, or a greater % of NaCl, e.g., about 15% NaCl.
  • the formulations of the disclosure may comprise two or more ingredients such that the total tonicity of the solution exceeds the physiological concentration of saline (about 0.9% NaCl).
  • the ingredients may include, e.g., sodium chloride and an osmolality adjusting agent selected from the group consisting of mannitol, xylitol, sorbitol, isomaltol, glucose, lactose, dextrose, sucrose, trehalose, maltose, glycerin, propylene glycol, ethylene glycol, glycerol, glycine, dimethylsulphoxide, calcium chloride, sodium sulfate, magnesium chloride, sodium gluconate, sodium pyruvate, pentosane polysulfate, and a cyclodextrin.
  • formulations include, e.g. , a solution comprising 0.9% sodium chloride and 1% sodium phosphate (wherein the total Na content exceeds that of isotonic saline) or a solution of 0.9% sodium chloride and 1% potassium chloride (wherein the total CI content exceeds that of isotonic saline).
  • normal saline means a solution containing 0.9% (w/v) NaCl (also referred herein as isotonic saline).
  • a D-alanine
  • Nle Norleucine
  • HYP 4-hydroxyproline
  • DHP 3,4-dehydro-L-proline
  • Ahp aminoheptanoic acid
  • 2PP (2R,5S)-5-phenyl- pyrrolidine-2-carboxylic acid
  • MS L-a-methylserine
  • mV N-methylvaline
  • the specialized non-naturally occurring peptides of the invention also include functional portions or fragments.
  • the length of the fragment is not critical as long as it substantially retains the biological activity of the peptide (e.g. , sodium channel binding activity).
  • Illustrative fragments comprise at least about 4, 5, 6, 7, 8, 9, 10, or more contiguous amino acids of a specialized non-naturally occurring peptide. In other embodiments, the fragment comprises no more than about 10, 9, 8, 7, 6, 5, or 4 contiguous amino acids of a specialized non-naturally occurring peptide.
  • the present invention also encompasses fusion polypeptides comprising a specialized non-naturally occurring peptides peptide or a functional fragment thereof.
  • the fusion protein can comprise a reporter molecule.
  • the fusion protein can comprise a polypeptide that provides a function or activity that is the same as or different from the activity of the peptide, e.g. , a targeting, binding, or enzymatic activity or function.
  • amino acid substitutions may be based on any characteristic known in the art, including the relative similarity or differences of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • the hydropathic index of amino acids may be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (see, Kyte and Doolittle, /. Mol. Biol. 157: 105 (1982); incorporated herein by reference in its entirety). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
  • Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte and Doolittle, id.), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (- 3.5); lysine (-3.9); and arginine (-4.5).
  • the hydropathic index of the amino acid may be considered when modifying the peptides specifically disclosed herein.
  • hydrophilicity of the amino acid may be considered when identifying additional peptides beyond those specifically disclosed herein.
  • Peptides (and fragments thereof) of the invention include peptides that have at least about 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or higher amino acid sequence identity with the peptide sequences disclosed herein.
  • Sequence identity or similarity may be determined using standard techniques known in the art, including, but not limited to, the local sequence identity algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the sequence identity alignment algorithm of Needleman & Wunsch, /. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Natl. Acad. Sci.
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments. It can also plot a tree showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng & Doolittle, /. Mol. Evol. 35:351 (1987); the method is similar to that described by Higgins & Sharp, CABIOS 5:151 (1989).
  • Another example of a useful algorithm is the BLAST algorithm, described in Altschul et al, J. Mol. Biol. 215:403 (1990) and Karlin et al, Proc. Natl. Acad. Sci. USA 90:5873 (1993).
  • a particularly useful BLAST program is the WU-BLAST-2 program which was obtained from Altschul et al. , Meth. Enzymol. , 266:460 (1996);
  • WU-BLAST-2 uses several search parameters, which are preferably set to the default values.
  • the parameters are dynamic values and are established by the program itself depending upon the composition of the particular sequence and composition of the particular database against which the sequence of interest is being searched; however, the values may be adjusted to increase sensitivity.
  • a percentage amino acid sequence identity value is determined by the number of matching identical residues divided by the total number of residues of the "longer" sequence in the aligned region.
  • the "longer” sequence is the one having the most actual residues in the aligned region (gaps introduced by WU-Blast-2 to maximize the alignment score are ignored).
  • the alignment may include the introduction of gaps in the sequences to be aligned.
  • sequences which contain either more or fewer amino acids than the peptides specifically disclosed herein it is understood that in one embodiment, the percentage of sequence identity will be determined based on the number of identical amino acids in relation to the total number of amino acids.
  • sequence identity of sequences shorter than a sequence specifically disclosed herein will be determined using the number of amino acids in the shorter sequence, in one embodiment. In percent identity calculations relative weight is not assigned to various manifestations of sequence variation, such as insertions, deletions, substitutions, etc.
  • identities are scored positively (+1) and all forms of sequence variation including gaps are assigned a value of "0," which obviates the need for a weighted scale or parameters as described below for sequence similarity calculations.
  • Percent sequence identity can be calculated, for example, by dividing the number of matching identical residues by the total number of residues of the "shorter" sequence in the aligned region and multiplying by 100. The "longer" sequence is the one having the most actual residues in the aligned region.
  • Peptides and fragments of the invention can be modified for in vivo use by the addition, at the amino- and/or carboxyl-terminal ends, of a blocking agent to facilitate survival of the relevant polypeptide in vivo.
  • a blocking agent to facilitate survival of the relevant polypeptide in vivo.
  • Such blocking agents can include, without limitation, additional related or unrelated peptide sequences that can be attached to the amino and/or carboxyl terminal residues of the peptide to be administered. This can be done either chemically during the synthesis of the peptide or by recombinant DNA technology by any suitable methods.
  • one or more non-naturally occurring amino acids can be added to the termini.
  • blocking agents such as pyroglutamic acid or other molecules known in the art can be attached to the amino and/or carboxyl terminal residues, or the amino group at the amino terminus or carboxyl group at the carboxyl terminus can be replaced with a different moiety.
  • the peptide terminus can be modified, e.g. , by acetylation of the N-terminus and/or amidation of the C-terminus.
  • the peptides can be covalently or noncovalently coupled to pharmaceutically acceptable "carrier" proteins prior to administration.
  • the peptides or fragments thereof of the invention are administered directly to a subject.
  • the compounds of the invention will be suspended in a pharmaceutically-acceptable carrier (e.g. , physiological saline) and administered orally or by intravenous infusion, or administered subcutaneously, intramuscularly, intrathecally, intraperitoneally, intrarectally, intravaginally, intranasally, intragastrically, intratracheally, or intrapulmonarily.
  • a pharmaceutically-acceptable carrier e.g. , physiological saline
  • the intratracheal or intrapulmonary delivery can be accomplished using a standard nebulizer, jet nebulizer, wire mesh nebulizer, dry powder inhaler, or metered dose inhaler.
  • Suitable dosages are in the range of 0.01 ⁇ g/kg-2.0 g/kg. Wide variations in the needed dosage are to be expected in view of the variety of peptides, fragments, and homologs available and the differing efficiencies of various routes of administration. For example, oral administration would be expected to require higher dosages than administration by i.v. injection.
  • Variations in these dosage levels can be adjusted using standard empirical routines for optimization as is well understood in the art.
  • Administrations can be single or multiple (e.g. , 2-, 3-, 4-, 6-, 8-, 10-; 20-, 50-, 100-, 150-, or more fold).
  • Encapsulation of the peptides, fragments, and homologs in a suitable delivery vehicle e.g. , polymeric microparticles or implantable devices
  • the methods of the present invention can be practiced with any of the peptides disclosed herein or their functional fragments.
  • the methods of the invention are practiced with S18 (SEQ ID NO: 1) or a functional fragment thereof.
  • the methods of the invention are practiced with S8 (SEQ ID NO: 120) or a functional fragment thereof.
  • the methods of the invention are practiced with any of the peptides disclosed in U.S. 2012/0115795, U.S. 2016/0102121 or U.S. 9,127,040 (each of which is incorporated herein by reference in its entirety).
  • the peptides, or fragments thereof can be targeted to specific cells or tissues in vivo.
  • Targeting delivery vehicles including liposomes and targeted systems are known in the art.
  • a liposome can be directed to a particular target cell or tissue by using a targeting agent, such as an antibody, soluble receptor or ligand, incorporated with the liposome, to target a particular cell or tissue to which the targeting molecule can bind.
  • a targeting agent such as an antibody, soluble receptor or ligand
  • Targeting liposomes are described, for example, in Ho et al, Biochemistry 25:5500 (1986); Ho et al, J. Biol. Chem. 262:13979 (1987); Ho et al, J. Biol. Chem. 262:13973 (1987); and U.S. Pat. No. 4,957,735 to Huang et al, each of which is incorporated herein by reference in its entirety).
  • Another aspect of the invention relates to a method of inhibiting sodium absorption through a sodium channel, comprising contacting (e.g., binding) the sodium channel with a specialized non-naturally occurring peptide or fragment thereof. Inhibition of sodium absorption can be measured by any technique known in the art or disclosed herein.
  • Another aspect of the invention relates to a method of increasing the volume of fluid lining an epithelial mucosal surface, comprising contacting (e.g., binding) a sodium channel present on the epithelial mucosal surface with a specialized non-naturally occurring peptide or a functional fragment or homolog thereof.
  • the volume of fluid lining an epithelial mucosal surface can be measured by any technique known in the art or disclosed herein.
  • a further aspect of the invention relates to a method of reducing the level of a sodium channel present on the surface of a cell, comprising contacting (e.g., binding) the sodium channel with a specialized non-naturally occurring peptide or a functional fragment or homolog thereof.
  • An additional aspect of the invention relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a specialized non-naturally occurring peptide or a functional fragment or homolog thereof.
  • the invention encompasses a method for treating a symptom of a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising administering a peptide comprising a sequence selected from SEQ ID NOS:2-128 to the subject.
  • the disorder in the methods of the invention can be, in non-limiting examples, a lung disorder (e.g.
  • cystic fibrosis cystic fibrosis, non-cystic fibrosis bronchiectasis, chronic obstructive pulmonary disease, acute or chronic bronchitis, or asthma
  • a gastrointestinal disorder e.g. , inflammatory bowel disease
  • kidney disorder e.g., a kidney disorder, or a cardiovascular disorder.
  • Another aspect of the invention relates to a method of regulating salt balance, blood volume, and/or blood pressure in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a specialized non-naturally occurring peptide or a functional fragment or homolog thereof.
  • a third aspect of the invention relates to products that can be used to carry out the methods disclosed herein.
  • one aspect of the invention relates to a peptide comprising the sequence:
  • Xi is leucine or a conservative substitution with a natural or non-natural amino acid
  • X2 is proline or a conservative substitution with a natural or non-natural amino acid
  • X 3 is valine or a conservative substitution with a natural or non-natural amino acid
  • X 4 is proline or a conservative substitution with a natural or non-natural amino acid
  • X5 is leucine or a conservative substitution with a natural or non-natural amino acid
  • X 6 is aspartic acid or a conservative substitution with a natural or non-natural amino acid
  • X 7 is glutamine or a conservative substitution with a natural or non-natural amino acid
  • Xg is threonine or a conservative substitution with a natural or non-natural amino acid; or a functional fragment thereof.
  • Another aspect of the invention relates to a peptide comprising the sequence:
  • Xi is leucine, norleucine, or valine
  • X2 is proline, 4-hydroxyproline, (2R,5S)-5-phenyl-pyrrolidine-2-carboxylic acid, or 3,4- dehydro-L-proline;
  • X 3 is valine, leucine, norleucine, or N-methylvaline
  • X 4 is proline, 4-hydroxyproline, (2R,5S)-5-phenyl-pyrrolidine-2-carboxylic acid, or 3,4- dehydro-L-proline;
  • X5 is leucine, norleucine, or valine
  • X 6 is aspartic acid or glutamic acid
  • X 7 is glutamine or asparagine; and Xg is threonine, serine, or L-a-methylserine;or a functional fragment thereof.
  • a further aspect of the invention relates to a peptide comprising the sequence: Xi-X 2 -X 3 -X4-X5-X6-X7-X8-X9-Xio-Xii-Xi2-Xi3-Xi4-Xi5(SEQ ID NO: 118) wherein:Xi is leucine or a conservative substitution with a natural or non-natural amino acid; X 2 is proline or a conservative substitution with a natural or non-natural amino acid;
  • X3 is valine or a conservative substitution with a natural or non-natural amino acid
  • X 4 is proline or a conservative substitution with a natural or non-natural amino acid
  • X 5 is leucine or a conservative substitution with a natural or non-natural amino acid
  • X 6 is aspartic acid or a conservative substitution with a natural or non-natural amino acid
  • X 7 is glutamine or a conservative substitution with a natural or non-natural amino acid
  • Xg is threonine or a conservative substitution with a natural or non-natural amino acid
  • X9 is threonine or a conservative substitution with a natural or non-natural amino acid
  • X10 is leucine or a conservative substitution with a natural or non-natural amino acid
  • X11 is proline or a conservative substitution with a natural or non-natural amino acid
  • X12 is asparagine or a conservative substitution with a natural or non-natural amino acid
  • Xi3 is valine or a conservative substitution with a natural or non-natural amino acid;
  • Xi 4 is asparagine or a conservative substitution with a natural or non-natural amino acid
  • Xi5 is proline or a conservative substitution with a natural or non-natural amino acid
  • a functional fragment thereof
  • the peptide comprises a sequence selected from the group consisting of SEQ ID NOS:4-142 and SEQ ID NO: 144. In one embodiment, the peptide comprises the sequence of SEQ ID NO:5 or SEQ ID NO: 122. In one embodiment, the peptide comprises the sequence of SEQ ID NO:2 (aaLPVPLDQTLPLNVNPaa) or SEQ ID NO: 119. In one embodiment, the peptide comprises the sequence of SEQ ID NO: 127 or SEQ ID NO: 128 (aaLPIPLDQTaa).
  • the peptides of the invention comprise at least one modified terminus, e.g. , to protect the peptide against degradation.
  • the N-terminus is acetylated and/or the C-terminus is amidated.
  • the peptide comprises the sequence of any one of SEQ ID NOS:10-127, 129, 130, 133, 134, or 136-140, further comprising one or two D-alanines at the amino- and/or carboxyl-terminal ends.
  • the peptides of the invention comprise at least one non- natural amino acid (e.g. , 1, 2, 3, or more) or at least one terminal modification (e.g. , 1 or 2). In some embodiments, the peptide comprises at least one non-natural amino acid and at least one terminal modification. [0082] In certain embodiments, the peptide mimics the sodium channel binding domain of a PLUNC protein.
  • the sodium channel binding domain is the minimal fragment of the PLUNC protein required to have substantially the same binding activity to the sodium channel as the full length PLUNC protein.
  • the term “substantially the same binding activity” refers to an activity that is at least about 50% of the binding activity of the full length protein, e.g., at least about 60%, 70%, 80%, or 90% of the binding activity.
  • the peptide has at least the same binding activity as the full length PLUNC protein.
  • the sodium channel is ENaC, e.g. , human ENaC.
  • the sodium channel is one that is similar in sequence and/or structure to ENaC, such as acid-sensing ion channels (ASIC).
  • the peptides of the present invention can optionally be delivered in conjunction with other therapeutic agents.
  • the additional therapeutic agents can be delivered concurrently with the peptides of the invention.
  • the word "concurrently” means sufficiently close in time to produce a combined effect (that is, concurrently can be simultaneously, or it can be two or more events occurring within a short time period before or after each other).
  • the specialized non-naturally occurring peptide is delivered to a patient concurrently with a compound that modulates the function of the cystic fibrosis transmembrane conductance regulator (CFTR) where the combined activity of the specialized non-naturally occurring peptide and the CFTR-targeted agent have superior activity to the CFTR-targeted agent alone.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the specialized non- naturally occurring peptide is delivered to a patient concurrently with a mucolytic compound where the combined activity of the specialized non-naturally occurring peptide and the mucolytic agent have superior activity to the mucolytic agent alone.
  • the specialized non-naturally occurring peptide is delivered to a patient concurrently with a long acting B-agonist compound (LABA) where the combined activity of the specialized non-naturally occurring peptide and the LABA agent have superior activity to the LABA alone.
  • the specialized non-naturally occurring peptide is delivered to a patient concurrently with a glucocorticoid agonist where the combined activity of the specialized non-naturally occurring peptide and the glucocorticoid agent have superior activity to the glucocorticoid alone.
  • kits comprising the peptide of the invention and useful for carrying out the methods of the invention.
  • the kit may further comprise additional reagents for carrying out the methods (e.g. , buffers, containers, additional therapeutic agents) as well as instructions.
  • additional reagents for carrying out the methods (e.g. , buffers, containers, additional therapeutic agents) as well as instructions.
  • the invention provides pharmaceutical formulations and methods of administering the same to achieve any of the therapeutic effects (e.g. , modulation of sodium absorption) discussed above.
  • the pharmaceutical formulation may comprise any of the reagents discussed above in a pharmaceutically acceptable carrier, e.g. , a specialized non- naturally occurring peptide or functional fragment thereof.
  • pharmaceutically acceptable it is meant a material that is not biologically or otherwise undesirable, i.e., the material can be administered to a subject without causing any undesirable biological effects such as toxicity.
  • the formulations of the invention can optionally comprise medicinal agents, pharmaceutical agents, carriers, adjuvants, dispersing agents, diluents, and the like.
  • the peptides of the invention can be formulated for administration in a pharmaceutical carrier in accordance with known techniques. See, e.g., Remington, The Science And Practice of Pharmacy (9 th Ed. 1995).
  • the peptide (including the physiologically acceptable salts thereof) is typically admixed with, inter alia, an acceptable carrier.
  • the carrier can be a solid or a liquid, or both, and is preferably formulated with the peptide as a unit-dose formulation, for example, a tablet, which can contain from 0.01 or 0.5% to 95% or 99% by weight of the peptide.
  • One or more peptides can be incorporated in the formulations of the invention, which can be prepared by any of the well-known techniques of pharmacy.
  • a further aspect of the invention is a method of treating subjects in vivo, comprising administering to a subject a pharmaceutical composition comprising a peptide of the invention in a pharmaceutically acceptable carrier, wherein the pharmaceutical composition is administered in a therapeutically effective amount.
  • Administration of the peptides of the present invention to a human subject or an animal in need thereof can be by any means known in the art for administering compounds.
  • the formulations of the invention include those suitable for oral, rectal, topical, buccal (e.g. , sub-lingual), vaginal, parenteral (e.g., subcutaneous, intramuscular including skeletal muscle, cardiac muscle, diaphragm muscle and smooth muscle, intradermal, intravenous, intraperitoneal), topical (i.e., both skin and mucosal surfaces, including airway surfaces), intranasal, transdermal, intraarticular, intrathecal, and inhalation administration, administration to the liver by intraportal delivery, as well as direct organ injection (e.g. , into the liver, into the brain for delivery to the central nervous system, into the pancreas, or into a tumor or the tissue surrounding a tumor).
  • the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular peptide which is being used.
  • the carrier will typically be a liquid, such as sterile pyrogen-free water, sterile normal saline, hypertonic saline, pyrogen-free phosphate-buffered saline solution, bacteriostatic water, or Cremophor EL[R] (BASF, Parsippany, N.J.).
  • the carrier can be either solid or liquid.
  • the peptide can be administered in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions.
  • Peptides can be encapsulated in gelatin capsules together with inactive ingredients and powdered carriers, such as glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium saccharin, talcum, magnesium carbonate and the like.
  • inactive ingredients examples include red iron oxide, silica gel, sodium lauryl sulfate, titanium dioxide, edible white ink and the like.
  • Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric- coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising the compound in a flavored base, usually sucrose and acacia or tragacanth; and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
  • Formulations of the present invention suitable for parenteral administration comprise sterile aqueous and non-aqueous injection solutions of the peptide, which preparations are preferably isotonic with the blood of the intended recipient. These preparations can contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient.
  • Aqueous and non-aqueous sterile suspensions can include suspending agents and thickening agents.
  • the formulations can be presented in unit/dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze- dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or water-for-injection immediately prior to use.
  • sterile liquid carrier for example, saline or water-for-injection immediately prior to use.
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the kind previously described.
  • an injectable, stable, sterile composition comprising a peptide of the invention, in a unit dosage form in a sealed container.
  • the peptide or salt is provided in the form of a lyophilizate which is capable of being reconstituted with a suitable pharmaceutically acceptable carrier to form a liquid composition suitable for injection thereof into a subject.
  • the unit dosage form typically comprises from about 1 mg to about 10 grams of the peptide or salt.
  • a sufficient amount of emulsifying agent which is pharmaceutically acceptable can be employed in sufficient quantity to emulsify the peptide or salt in an aqueous carrier.
  • emulsifying agent is phosphatidyl choline.
  • Formulations suitable for rectal administration are preferably presented as unit dose suppositories. These can be prepared by admixing the peptide with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers which can be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
  • Formulations suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • Formulations suitable for transdermal administration can also be delivered by iontophoresis (see, for example, Tyle, Pharm. Res. 3:318 (1986)) and typically take the form of an optionally buffered aqueous solution of the peptides.
  • Suitable formulations comprise citrate or bis/tris buffer (pH 6) or ethanol/water and contain from 0.1 to 0.2M of the compound.
  • the peptide can alternatively be formulated for nasal administration or otherwise administered to the lungs of a subject by any suitable means, e.g. , administered by an aerosol suspension of respirable particles comprising the peptide, which the subject inhales.
  • the respirable particles can be liquid or solid.
  • aerosol includes any gas-borne suspended phase, which is capable of being inhaled into the bronchioles or nasal passages.
  • aerosol includes a gas-borne suspension of droplets, as can be produced in a metered dose inhaler or nebulizer, or in a mist sprayer. Aerosol also includes a dry powder composition suspended in air or other carrier gas, which can be delivered by insufflation from an inhaler device, for example.
  • Aerosols of liquid particles comprising the peptide can be produced by any suitable means, such as with a pressure- driven aerosol nebulizer or an ultrasonic nebulizer, as is known to those of skill in the art. See, e.g. , U.S. Patent No. 4,501,729. Aerosols of solid particles comprising the peptide can likewise be produced with any solid particulate medicament aerosol generator, by techniques known in the pharmaceutical art.
  • peptide in a local rather than systemic manner, for example, in a depot or sustained-release formulation.
  • the present invention provides liposomal formulations of the peptides disclosed herein and salts thereof.
  • the technology for forming liposomal suspensions is well known in the art.
  • the peptide or salt thereof is an aqueous-soluble salt
  • the peptide or salt using conventional liposome technology, the same can be incorporated into lipid vesicles.
  • the peptide or salt will be substantially entrained within the hydrophilic center or core of the liposomes.
  • the lipid layer employed can be of any conventional composition and can either contain cholesterol or can be cholesterol-free.
  • the salt can be substantially entrained within the hydrophobic lipid bilayer which forms the structure of the liposome.
  • the liposomes which are produced can be reduced in size, as through the use of standard sonication and homogenization techniques.
  • the liposomal formulations containing the peptides disclosed herein or salts thereof can be lyophilized to produce a lyophilizate which can be reconstituted with a pharmaceutically acceptable carrier, such as water, to regenerate a liposomal suspension.
  • a pharmaceutical composition can be prepared containing the water-insoluble peptide, such as for example, in an aqueous base emulsion.
  • the composition will contain a sufficient amount of
  • emulsifying agent to emulsify the desired amount of the peptide.
  • Particularly useful emulsifying agents include phosphatidyl cholines and lecithin.
  • the peptide is administered to the subject in a therapeutically effective amount, as that term is defined above.
  • Dosages of pharmaceutically active peptides can be determined by methods known in the art, see, e.g., Remington's
  • the therapeutically effective dosage of any specific peptide will vary somewhat from peptide to peptide, and patient to patient, and will depend upon the condition of the patient and the route of delivery. As a general proposition, a dosage from about 0.1 mg/kg to about 2 g/kg will have therapeutic efficacy, with all weights being calculated based upon the weight of the peptide, including the cases where a salt is employed. In certain cases, toxicity concerns at the higher level can restrict intravenous dosages to a lower level such as up to about 10 mg/kg, with all weights being calculated based upon the weight of the peptide, including the cases where a salt is employed.
  • a dosage from about 0.1 mg/kg to about 2 mg/kg, from about 0.5 mg/kg to about 2 g/kg, from about 1 mg/kg to about 2 g/kg, from about 10 mg/kg to about 2 g/kg, from about 100 mg/kg to about 2 g/kg, from about 0.5 g/kg to about 2 g/kg, from about 1 g/kg to about 2 g/kg, from about 0.1 mg/kg to about 1 g/kg, from about 0.5 mg/kg to about 1 g/kg, from about 1 mg/kg to about 1 g/kg, from about 10 mg/kg to about 1 g/kg, from about 100 mg/kg to about 1 g/kg, from about 0.5 g/kg to about 1 g/kg, from 0.1 mg/kg to about 0.5 g/kg, from 0.5 mg/kg to about 0.5 g/kg, from 1 mg/kg to about 0.5 g/kg, from 10 mg/kg to about 0.5 g/kg, from about
  • a dosage of about 1 mg/kg can be employed for inhalation, intranasal, oral, intravenous, or intramuscular administration.
  • a dosage of about 2 mg/kg can be employed for inhalation, intranasal, oral, intravenous, or intramuscular administration.
  • a dosage of about 4 mg/kg can be employed for inhalation, intranasal, oral, intravenous, or intramuscular administration.
  • a dosage from about 0.5 mg/kg to 5 mg/kg can be employed for intramuscular injection.
  • dosages are about 1 ⁇ /kg to 50 ⁇ /kg, and more particularly to about 22 ⁇ /kg and to 33 ⁇ /kg of the peptide for intravenous or oral administration, respectively.
  • more than one administration e.g. , two, three, four, or more administrations
  • time intervals e.g. , hourly, daily, weekly, monthly, etc.
  • the present invention finds use in veterinary and medical applications. Suitable subjects include both avians and mammals, with mammals being preferred.
  • avian as used herein includes, but is not limited to, chickens, ducks, geese, quail, turkeys, and pheasants.
  • mammal as used herein includes, but is not limited to, humans, bovines, ovines, caprines, equines, felines, canines, lagomorphs, etc. Human subjects include neonates, infants, juveniles, and adults.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a Palate Lung and Nasal epithelial Clone (PLUNC) protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel, and wherein the PLUNC protein, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • PLUNC Palate Lung and Nasal epithelial Clone
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g., about 4.2% NaCl.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a formulation as described above (e.g. , a PLUNC protein or a functional fragment thereof or homolog thereof and saline) and a pharmaceutically acceptable carrier and optionally a pharmaceutically acceptable osmolality adjusting agent.
  • a formulation as described above e.g. , a PLUNC protein or a functional fragment thereof or homolog thereof and saline
  • a pharmaceutically acceptable carrier e.g. a pharmaceutically acceptable carrier and optionally a pharmaceutically acceptable osmolality adjusting agent.
  • the PLUNC protein, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 0.9%, e.g. , about 4.2% NaCl.
  • the osmolality adjusting agent is one or more selected from the group consisting of mannitol, xylitol, sorbitol, isomaltol, glucose, lactose, dextrose, sucrose, trehalose, maltose, glycerin, propylene glycol, ethylene glycol, glycerol, glycine, dimethylsulphoxide, calcium chloride, sodium sulfate, magnesium chloride, sodium gluconate, sodium pyruvate, pentosane polysulfate, and a cyclodextrin.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising at least one polypeptide comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO: 141, SEQ ID NO: 129, SEQ ID NO: 130, SEQ ID NO: 128, SEQ ID NO: 131, SEQ ID NO: 132, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 135, SEQ ID NO: 136, SEQ ID NO: 137, SEQ ID NO: 138, SEQ ID NO: 131, SEQ ID NO: 139; SEQ ID NO: 140, SEQ ID NO: 144, SEQ ID NO: 127, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 136, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:7, or
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising at least one peptide comprising, consisting essentially of, or consisting of, the sequence set forth in SEQ ID NO: 127 or SEQ ID NO: 128, wherein the peptide is formulated in a saline solution containing a salt concentration of at least 1%, e.g. , at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, at least 13%, at least 14%, at least 15%, at least 16%, at least 17%, at least 18%, at least 19%, or at least 20%.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g., about 4.2% NaCl.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide comprising a modified sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof which is formulated in a saline solution containing a salt concentration of at least 1%.
  • the PLUNC protein or the functional fragment or homolog thereof is modified to reduce or eliminate the inactivation of the polypeptide at an acidic pH, e.g. , by the addition of a blocking agent to facilitate survival of the polypeptide in vivo and/or by fusing to a protein that increases the stability of the polypeptide.
  • the modified PLUNC protein or the functional fragment or homolog thereof is modified at the N-terminus via acetylation. In some aspects, the modified PLUNC protein or the functional fragment or homolog thereof comprises at least one non-natural amino acid or at least one terminal modification, e.g. , at least one D-alanine at the N-terminus and/or the C-terminus.
  • the disclosure provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide comprising a modified sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof which is formulated in a saline solution containing a salt concentration of at least 1%, wherein the formulation is to be administered by inhalation, intranasally, intravenously, intramuscularly, intraocularly, transdermally, or orally, preferably by inhalation, e.g. , via metered dose, inhaler, nebulizer, or in a mist sprayer.
  • the disclosure provides for a kit comprising, in one or more packages, a polypeptide comprising a modified sodium channel binding domain of the PLUNC protein, or a functional fragment, or homolog thereof and reagents for formulation of the PLUNC protein or the fragment or homolog thereof in a saline solution containing a salt concentration of at least 1%.
  • the disclosure relates to a method of inhibiting activity of a sodium channel, comprising contacting the sodium channel with a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g., at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, at least 13%, at least 14%, at least 15%, at least 16%, at least 17%, at least 18%, at least 19%, or at least 20%.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the polypeptide, or the functional fragment,
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the disclosure relates to a method of inhibiting activity of a sodium channel, comprising contacting the sodium channel with a pharmaceutical composition comprising at least one polypeptide comprising, consisting essentially of, or consisting of, the amino acid sequence set forth in SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO: 141, SEQ ID NO: 129, SEQ ID NO: 130, SEQ ID NO: 128, SEQ ID NO: 131, SEQ ID NO: 132, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 135, SEQ ID NO: 136, SEQ ID NO: 137, SEQ ID NO: 138, SEQ ID NO: 131, SEQ ID NO: 139; SEQ ID NO: 140, SEQ ID NO: 144, SEQ ID NO: 127, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO:
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the activity of the sodium channel is inhibited by at least 20%, especially by at least 50%, preferably by at least 60%, and particularly preferably by at least 90%.
  • the inhibition of activity by the pharmaceutical composition of the disclosure is mediated by internalization of the sodium channel, e.g. , internalization of one or more subunits of ENaC, e.g. , a, ⁇ , or ⁇ ENaC.
  • the disclosure relates to a method of inhibiting sodium absorption through a sodium channel, comprising contacting the sodium channel with a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the disclosure relates to a method of increasing the volume of fluid lining an epithelial mucosal surface, comprising contacting a sodium channel present on the epithelial mucosal surface with a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel, and wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • increasing the volume of fluid lining an epithelial mucosal surface increases the activity of and/or expression level of cystic fibrosis transmembrane conductance regulator (CFTR).
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the disclosure relates to a method of reducing the level of a sodium channel present on the surface of a cell, comprising contacting the sodium channel with a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel, and wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g., about 4.2% NaCl.
  • the disclosure relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel, and wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the disorder is a lung disorder, e.g.
  • the disorder is a gastrointestinal disorder, e.g., constipation, or inflammatory bowel disease (IBD). In some aspects, the disorder is a kidney disorder.
  • the disclosure relates to a method of regulating salt balance and/or fluid volume regulation in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject, and wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the disclosure relates to a method for treating a symptom of a lung disorder, a gastrointestinal disorder, a kidney disorder, or a cardiovascular disorder in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject, and wherein the polypeptide, or the functional fragment, or homolog thereof is formulated in a saline solution containing a salt concentration of at least 1%, e.g.
  • the salt concentration in the formulation is between 2% and 10%, particularly between 3% and 8%, and especially between 4% and 7%.
  • the salt concentration in the formulation is at least 4.2%, e.g. , about 4.2% NaCl.
  • the lung disorder is cystic fibrosis, non- cystic fibrosis bronchiectasis, chronic obstructive pulmonary disease, acute or chronic bronchitis, or asthma.
  • the gastrointestinal disorder includes constipation or inflammatory bowel disease.
  • the disclosure relates to a method of inhibiting activity of a sodium channel, comprising contacting the sodium channel with: a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • the pharmaceutical composition and the hypertonic saline solution is administered simultaneously.
  • the hypertonic saline solution is administered after the administration of the pharmaceutical composition.
  • the disclosure relates to a method of inhibiting sodium absorption through a sodium channel, comprising contacting the sodium channel with: a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel
  • a hypertonic saline solution e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • the disclosure relates to a method of increasing the volume of fluid lining an epithelial mucosal surface, comprising contacting a sodium channel present on the epithelial mucosal surface with: a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel
  • a hypertonic saline solution e.g. , a saline solution containing a salt concentration of about 4.2% NaC
  • the disclosure relates to a method of reducing the level of a sodium channel present on the surface of a cell, comprising contacting the sodium channel with: a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to the sodium channel
  • a hypertonic saline solution e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • the disclosure relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject: a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel; and a hypertonic saline solution, e.g. , a saline solution containing a
  • the disclosure relates to a method of regulating salt balance and/or fluid volume regulation in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a pharmaceutical composition comprising: a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising: a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject; and a hypertonic saline solution, e.g. ,
  • the disclosure relates to a method for treating a symptom of a lung disorder, a gastrointestinal disorder, a kidney disorder, or a cardiovascular disorder in a subject in need thereof, comprising administering to the subject: a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject; and a hypertonic saline solution, e.g. , a saline solution containing a salt concentration of about 4.2% NaCl.
  • a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel present on the surface of a cell in the subject
  • the disclosure relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject: a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel; and a compound that modulates the function of cystic fibrosis transmembrane conductance regulator (CFTR), e.g.
  • CFTR cystic fibrosis transmembrane conductance regulator
  • the pharmaceutical composition and the compound are delivered to the subject concurrently.
  • the pharmaceutical composition and the compound are delivered to the subject simultaneously.
  • the disclosure relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject: a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel; and a mucolytic compound, e.g.
  • the disclosure relates to a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject: a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof, wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel; and a long acting beta-agonist compound (LAB A), e.g. , a LABA compound selected from the group consisting of albuterol, levalbuterol, formoterol, and salmeterol.
  • LAB A long acting beta-agonist compound
  • the disclosure provides for a method of treating a disorder responsive to inhibition of sodium absorption across an epithelial mucosal surface in a subject in need thereof, comprising delivering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a polypeptide comprising a sodium channel binding domain of a PLUNC protein, or a functional fragment, or homolog thereof and a
  • osmolality adjusting agent e.g. , a compound selected from mannitol, xylitol, sorbitol, isomaltol, glucose, lactose, dextrose, sucrose, trehalose, maltose, glycerin, propylene glycol, ethylene glycol, glycerol, glycine, dimethylsulphoxide, calcium chloride, sodium sulfate, magnesium chloride, sodium gluconate, sodium pyruvate, pentosane polysulfate, and a cyclodextrin), wherein the PLUNC protein, or the functional fragment, or homolog thereof binds to a sodium channel.
  • osmolality adjusting agent e.g. , a compound selected from mannitol, xylitol, sorbitol, isomaltol, glucose, lactose, dextrose, sucrose, trehalose, maltose,
  • Tissue procurement and cell culture Cells were harvested by enzymatic digestion from human bronchial tissue as previously described under a protocol approved by the UNC School of Medicine IRB (Tarran et al. , J. Gen. Physiol. 127:591 (2006)). Human excess donor lungs and excised recipient lungs were obtained at the time of lung transplantation from portions of main stem or lumbar bronchi and cells were harvested by enzymatic digestion. All preparations were maintained at an air-liquid interface in a modified bronchial epithelial medium and used 2-5 weeks after seeding on 12 mm T-Clear inserts (Corning Costar) coated with human placental type VI collagen (Sigma). Phosphate buffered saline (PBS) was used for washing human bronchial epithelial culture mucosal surfaces.
  • PBS Phosphate buffered saline
  • human bronchial epithelial cultures were bathed serosally in a modified Ringer solution containing (mM): 116 NaCl, 10 NaHC0 3 , 5.1 KC1, 1.2 CaCl 2 , 1.2 MgCl 2 , 20 TES, 10 glucose, pH 7.4).
  • human bronchial epithelial cultures were maintained in a modified BEGM growth medium which contained 24 mM NaHCC>3 gassed with 5% CO2.
  • Perfluorocarbon (FC-77) was obtained from 3M and had no effect on ASL height as previously reported.
  • alpha-ENaC HEK293T cells were transfected with gfp- ocENaC where gfp was fused at the N-terminus of ENaC and unlabeled ENaC and yENaC using lipofectamine in 384 well plates as per the manufacturer's instructions and as published previously (see, e.g. , Hobbs et al. Am J Physiol Lung Cell Mol Physiol. 2013
  • Fig. 6 shows the results of an experiment analyzing the effects of various peptides on internalization of alpha-ENaC in HEK293T cells when GFP-tagged alpha-ENaC is co-expressed only with gamma ENaC and no beta- ENaC.
  • SEQ ID NO: 143 negative control peptide
  • water vehicle
  • SEQ ID NO: l water
  • SEQ ID NO: 128 SEQ ID NO: 128
  • SEQ ID NO: l AND SEQ ID NO: 128 are effective in reducing alpha-ENaC when beta-ENaC is co-expressed, no effect is observed in this experiment when beta-ENaC is not present.
  • PENaC-Tg mice are disease free at birth, but soon develop obstructive lung disease (Zhou, Z., et al. Am J Respir Crit Care Med 178, 1245-1256 (2008)).
  • C57BL:FVB and C57BL:C3H mixed strains have 90% and 50% mortality, respectively, and the reproducibility of the disease on these backgrounds is sufficiently high that reliable data are produced with n of about 8-10 animals/group.
  • PENaC-Tg mouse responds to therapeutic interventions in a fashion similar to CF/COPD in humans, and the development of lung disease can be prevented following inhibition of lung disease at birth (Livraghi, A., et al. J Immunol 182, 4357-4367 (2009)).
  • SI 8- derived peptides or vehicle were dosed one to three times a day by intranasal instillation ( ⁇ /g body weight) for 14 days in parallel cohorts of mice. Mice were weighed daily and, if a diuretic effect was found, the volume excreted was replaced by sub-cutaneous injections of sterile saline. At the end of the treatment, mice were sacrificed for phenotypic analysis.
  • SPLUNCl derived peptides (namely, SI 8; FIG. 1) can inhibit ENaC with EC 50 in the sub micromolar range for up to 24 h following a single dose (Hobbs et al, 2013). While S18 is resistant to proteolysis and heat-stable, we tested whether we could reduce the size of SI 8, increase its stability and/or increase its potency. Any of these actions would increase its utility as a drug.
  • LPVPLDQT (SEQ ID NO: 113); FIG. 1).
  • DQT charged mid-region
  • DQTLPLNVNP subsequence B
  • FIG. 2 LPVPLDQT (SEQ ID NO: 113) retained similar activity to SI 8, whilst DQTLPLNVNP (SEQ ID NO: 114) was inactive.
  • LPVPLDQT (SEQ ID NO: 113) could inhibit ENaC-led fluid absorption, this peptide was more susceptible to proteolytic degradation.
  • Peptides were added mucosally to HBECs at 30 ⁇ and ASL height measured 3 h later by XZ-confocal microscopy.
  • FIG. 2A shows the comparison of the N- terminal region of S18 (LPVPLDQT) (SEQ ID NO: 113) (SEQ ID NO: 113) could inhibit ENaC-led fluid absorption, this peptide was more susceptible to proteolytic degradation.
  • Peptides were added mucosally to HBECs at 30 ⁇ and ASL height measured 3 h later by XZ-confocal microscopy.
  • FIG. 2A shows the comparison of the N- terminal region of S18 (LPVPLDQT) (SEQ ID NO: 113)
  • aaLPNlePLDQTaa shows a log-fold increase in potency as compared to SI 8
  • SEQ ID NO:9 shows aaLPVPLDQSaa
  • SEQ ID NO:6 shows diminished potency and efficacy respectively
  • FIG. 3B Other sequences included in FIG. 3B are SEQ ID NO:4 (aaNlePVPLDQTaa), SEQ ID NO:7 (aaLPVPLDNTaa) and SEQ ID NO:8 (aaLPVPLEQTaa).
  • ADG is a negative control peptide with the sequence: NH3 - ADGGLLLLNNPPPPQTVV-NH2 (SEQ ID NO: 143).
  • the peptides used in these experiments were S18 (SEQ ID NO: l), SEQ ID NO:2, SEQ ID NO: 141, SEQ ID NO: 129, SEQ ID NO: 130, SEQ ID NO: 128, SEQ ID NO: 131, SEQ ID NO: 132, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 135, SEQ ID NO: 136, SEQ ID NO: 137, SEQ ID NO: 138, SEQ ID NO: 131, SEQ ID NO: 139; SEQ ID NO: 140, SEQ ID NO: 144 and SEQ ID NO: 127.
  • SEQ ID NO: 128 did not have a diuretic effect.
  • Fig. 7F shows that treatment with SEQ ID NO: 127 increased percent survival of ENaC-Tg C57BL:C3H mice. Sequences of other peptides used in the experiment were SEQ ID NO: 129, SEQ ID NO: 134, and SEQ ID NO: 136.
  • TMV Tracheal Mucus Velocity: TMV was measured in vivo by a roentgenographic technique. Between 8 and 10 radiopaque Teflon/bismuth trioxide disks, 1-mm diameter, 0.8-mm thick and 1.5-2mg in weight, were insufflated onto the trachea. A modified suction catheter connected to a source of continuous compressed air at a flow of 3-4 L/min was used to introduce the particles via the endotracheal tube. The catheter remained within the endotracheal tube only during particle delivery, so that no contact with the tracheal surface is made. The cephalad-axial velocities of the individual disks were recorded on videotape from a portable image intensifier unit.
  • Aerosols were generated using an Airlife medication nebulizer.
  • a dosimetry system activated by a piston respirator Harmonic Apparatus Respiratory Pump, Holliston, MA
  • the nebulizer was connected to a T-piece with one end attached to the respiratory pump and the other to the animal's tracheal tube.
  • Nebulized aerosols were delivered directly into the trachea only during inspiration at a frequency of 20 breaths / min and at a tidal volume of 500 mL.
  • Agents Ten (10) mg of CFTRinh-172 was dissolved in 0.3 mL of DMSO. The solution was vortexed and then 2.7 mL of ethanol was added bringing the final nebulized solution of CFTRinh to 10 mg/3 mL. The solution was stirred for 10 min before dosing to assure that the CFTRinh was completely dissolved. A new nebulizer was used for each CFTRinh challenge. SEQ ID NO: 128 was dissolved in 3mL 0.9% saline to achieve final doses of 4 mg/kg, 2 mg/kg, and 1 mg/kg and the total amount delivered to the sheep.
  • Study Protocol After a 10 minute period on the humidifier, a baseline TMV was obtained. Immediately following the baseline, the sheep were challenged with an aerosol of 10 mg of CFTRinh. TMV was measured hourly for 4h. Immediately after the 4h TMV measurement, 3 mL of 0.9% normal saline (vehicle) or the different doses of SEQ ID NO: 128 were given by nebulization. TMV was then measured from 5-12h following treatment.
  • Results The results of SEQ ID NO: 128 treatments on the CFTRinh response are shown in Figs. 8A-8C.
  • the average response for the saline treated group between 5-12h was 50 + 0.5%. All treatments significantly improved this response: 73.7 + 0.7% with 1 mg/kg; 88.4 + 0.6% with 2 mg/kg; and 103.0 + 0.6 4 mg/kg.
  • the treatment responses were significantly different from each other (P ⁇ 0.001).
  • Cystic Fibrosis lung disease is caused by the loss of function of the cystic fibrosis transmembrane conductance regulator (CFTR) combined with hyperactivation of the epithelial sodium channel (ENaC).
  • ENaC is responsible for movement of sodium.
  • Hyperactivation of ENaC which creates an osmotic gradient that pulls fluid out of the airway, contributes to reduced airway hydration causing mucus dehydration, decreased mucociliary clearance, and recurrent, acute bacterial infections.
  • ENaC represents a therapeutic target to treat all CF patients independent of their underlying CFTR mutation.
  • SPX-101 binds selectively to ENaC and promotes internalization of the ⁇ , ⁇ , and y subunits. Removing ENaC from the membrane with SPX-101 causes a significant decrease in amiloride-sensitive current. The peptide increases survival of ENaC transgenic mice to >90% with once-daily dosing by inhalation. SPX-101 increased mucus transport in the ENaC mouse model as well as the sheep model of CF.
  • Cystic fibrosis is a recessive genetic disease affecting more than 70,000 people globally.
  • the disease is caused by mutations in the gene for the cystic fibrosis transmembrane conductance regulator (CFTR) protein, which result in lack of expression, or expression of dysfunctional, protein (1).
  • CFTR controls transepithelial secretion of ClTHCCV onto the airway surface.
  • CF is also characterized by hyperactivation of the epithelial sodium channel (ENaC) (2, 3). This hyperactivation causes an excess of sodium to be absorbed via ENaC, resulting in an osmotic gradient that draws water away from the airway surface.
  • ENaC epithelial sodium channel
  • SPLUNC1 Short Palate, Lung, and Nasal Clone 1
  • SI 8 18 amino acid domain
  • S18 was found to regulate ENaC in a pH-independent manner (27, 28). This discovery underscored the therapeutic potential of using such an ENaC-regulating peptide for the treatment of CF lung disease.
  • SPX-101 induces internalization of oc/ /yENaC in normal and CF human bronchial epithelial cells (HBECs), providing a novel mechanism of action for the regulation of water movement in the airway.
  • the peptide increases survival of ENaC-Tg mice, increases mucociliary clearance in these mice, and increases tracheal mucus velocity in an ovine model of CF.
  • SPX-101 represents a novel peptide promoter of ENaC internalization which we propose has the potential to serve as a new therapeutic for the treatment of all patients with CF.
  • HBECs used in these studies were isolated at Spyryx Biosciences and purchased from Lonza (Walkersville, MD). Cells were grown as previously described (31). HEK293T cells were purchased from ATCC (Manassas, VA).
  • Vt Transepithelial voltage
  • R resistance
  • SPX-101 The structure of SPX-101 is shown in Figure 9A.
  • HEK293T cells were mock-transfected or transfected with ccENaC-GFP, ENaC-FLAG, and yENaC-HA. Twenty-four hours after transfection, cells were lysed and incubated with biotin- tagged SPX-101. Interacting proteins were isolated on streptavidin agarose and detected by western blot. As seen in Figure 9B, all three ENaC subunits were detected in the pulldown. Similar results were previously reported with SPLUNCl and S18 binding to ENaC (26, 28).
  • SPX-101 promotes internalization of ENaC [0174] Previously, it was reported that SPLUNC1 induced internalization of ENaC (32). To determine if SPX-101 had a similar effect, we treated primary HBECs derived from healthy donors with SPX-101, amiloride, or an alphabetized control peptide. As seen in Figure 10A, SPX-101 induced internalization of all three ENaC subunits. SPX-101 did not alter expression of ATP12A demonstrating that these effects were ENaC-specific. As expected, amiloride and the control peptide had no effect on ENaC levels in the plasma membrane. We next determined the duration of this effect in HBECs from healthy and CF donors.
  • SPX-101 induced a rapid and durable internalization of all three ENaC subunits out to eight hours, the longest time point analyzed ( Figures lOB/C). These data demonstrate that SPX-101, but not amiloride, promotes internalization of all three ENaC subunits.
  • ENaC-Tg mice have increased proportions of disease-associated neutrophils and eosinophils in their bronchoalveolar lavage fluid (BALF) as compared to healthy wild-type mice (37).
  • SPX-101 treatment decreased the number of neutrophils and eosinophils in the BALF ( Figure 12C).
  • an alphabetized control peptide did not cause changes in mouse survival or BALF leukocyte composition ruling out an osmotic effect of the peptide as the driving force behind these events.
  • SPX-101 increases mucociliary clearance in flENaC-Tg mice
  • SPX-101 increases tracheal mucus velocity in a sheep model of Cystic Fibrosis
  • SPX-101 binds selectively to ENaC (Figure 1) to induce internalization of ⁇ / ⁇ / ⁇ ENaC ( Figures lOA-lOC), and a decrease in amiloride-sensitive current (Figure 3).
  • SPX-101 increases survival of ENaC-Tg mice while reducing neutrophil and eosinophil infiltration into the lung with a once-daily dosing regimen (Figure 12C).
  • a single intranasal dose of the peptide increases mucociliary clearance velocity and directionality in ENaC-Tg mice ( Figures 13A-13C).
  • the peptide restores tracheal mucus velocity in an ovine model of CF ( Figures 14A, 14C, and 14D).
  • SPX-101 is an optimized, first-in-class ENaC-effecting therapeutic capable of restoring durable mucus transport.
  • SPLUNC1 In healthy lungs, SPLUNC1 is secreted by airway epithelia creating an autocrine control mechanism of ENaC membrane concentration (4, 26, 38). This function is lost in CF due to a conformational change in SPLUNC1 that occurs in the acidified CF airway.
  • S18 a peptide derived from SPLUNCl's N-terminus, S18, was shown to retain ENaC-regulatory function at acidic pH (27).
  • SPX-101 we optimized S 18 to enhance drug-like properties and deliverability into the lung via nebulization. The reduced size of SPX- 101 increases the dose density and reduces nebulization time, an important consideration for CF patients who spend multiple hours a day receiving therapy.
  • the hyperactivation of ENaC in the CF lung has been shown to occur from both an increase in channel number (39), as well as an increase in open probability (Po) of the channel (40, 41).
  • the CF lung contains increased levels of proteases which can activate ENaC (40, 42).
  • Inhibitors such as amiloride decrease ENaC hyperactivation by reducing Po, but the channels and activating proteases are still present in the airway. This scenario ensures that ENaC hyperactivation rapidly resumes when the inhibitor is washed away.
  • SPX-101 causes a durable reduction of ENaC hyperactivation by removing channels from the apical membrane of the cell and functionally blocking sodium absorption.
  • SPX-101 has a durable effect due to the peptide's ability to internalize ENaC.
  • Highlighting the advantage afforded by this mechanism of action is the ability of SPX-101 to inhibit amiloride-sensitive current.
  • the ability of SPX-101 to maintain its reduction in ENaC current occurred even after the peptide was washed away because the number of channels on the surface of the cell has been reduced.
  • the PENaC-Tg mouse model develops CF-like lung disease that is characterized by mucus plugging of the upper airway and increased neutrophil and eosinophil levels in the BAL fluid (29, 37).
  • 45% of PENaC-Tg mice survived through two weeks of age in the absence of therapeutic intervention.
  • Zhou and colleagues demonstrated that amiloride increased survival of PENaC-Tg mice to -80% but this required thrice daily administration (37).
  • any compound to be beneficial in CF lung disease it must increase MCC. It was previously reported that the PENaC-Tg mice have impaired mucus transport (29). We observed an -50% reduction in velocity of fluorescent microspheres along the trachea of PENaC-Tg mice compared to wild-type littermate mice. The mucus velocity in these mice was significantly restored to 77% of wild- type levels by a single dose of SPX-101. In addition to the increase in velocity, SPX-101 also corrected directionality of mucus movement to wild- type levels. This restoration of mucus transport suggests that SPX-101 is likely augmenting mucus hydration in this model. Important future studies will address this effect of SPX-101, as well as its effects on mucus composition and rheology.
  • SPX-101 also increased mucus transport in a large animal model.
  • the ovine model based on CFTRinhl72 nebulization provides an opportunity to understand a compound's effects in a complex airway (29).
  • Our data in this model demonstrate that SPX-101 provides a sustained increase in TMV for the duration of the experiment and reached 100% at the highest dose tested.
  • a control peptide did not increase TMV, again ruling out changes in airway osmolality as the driver of the observed effect.
  • SPX-101 has demonstrated in CF HBECs a greater than 60% reduction in amiloride-sensitive current through its mechanism of internalizing ENaC (Figure 3). This reduction restores the amiloride-sensitive current to approximately wild-type levels in HBECs from healthy donors. Additionally, the duration of SPX-101's internalization effect in these cells was shown to exceed 8 hours ( Figures 1 OA- IOC). This finding corroborates the results with SPX-101 in the sheep model, which showed full drug response beyond 8 hours ( Figures 14A, 14C, and 14D). Fortunately, SPX-101's effect on ENaC is independent of the CF-causing mutations, which means it represents a unique opportunity to treat all CF patients.
  • SPX-101 fully differentiates itself from previous ENaC-targeting therapeutics through its mechanism of action and duration of effect. Not simply a channel blocker, SPX-101 replaces the natural ENaC-inhibiting properties of endogenous SPLUNC1 that have been lost in the CF airway.
  • SPX-101 achieves a durable increase in mucus transport in multiple animal models, demonstrating its potential to provide meaningful, clinical benefit to CF patients.
  • SPX-101 has the potential to also address other diseases associated with MCC defects, such as non-CF bronchiectasis, COPD, and severe asthma.
  • DF508/DF508 were used.
  • Cells were harvested by enzymatic digestion from human bronchial tissue as previously described.
  • Human excess donor lungs and excised recipient lungs were obtained at the time of lung transplantation from portions of main stem or lumbar bronchi and cells were harvested by enzymatic digestion. All preparations were maintained at an air-liquid interface in a modified bronchial epithelial medium and used 2-5 weeks after seeding on 12 mm T-Clear inserts (Corning Costar) coated with human placental type VI collagen
  • PBS Phosphate buffered saline
  • Ringer containing rhodamine-dextran (2 mg/ml; Invitrogen) was added to human bronchial epithelial culture mucosal surfaces.
  • Perfluorocarbon was added mucosally to prevent evaporation of the airway surface liquid and the culture placed in a chamber containing 100 ⁇ Ringer on the stage of a Leica SP8 confocal microscope with a 63X glycerol immersion objective.
  • results are presented in FIG. 15.
  • the results show a surprising effect when hypertonic saline was used as vehicle for SPX-101 compared to the effect conferred by isotonic saline.
  • use of a formulation of SPX-101 in 3% saline increased ASL height by over 250% compared to SPX-101 in isotonic saline (about 12 urn with 10 mM SPX-101 in 0.9% saline compared to about 30 ⁇ with the same dose of SPX-101 in 3.0% saline).
  • the combined effects of hypertonic saline and SPX-101 were synergistic, e.g. , greater than additive.
  • Aerosolized amiloride dose effect on nasal bioelectric properties, pharmacokinetics, and effect on sputum expectoration in patients with cystic fibrosis. J Aerosol Med 1997; 10: 147-158.
  • Nonantibiotic macrolides prevent human neutrophil elastase-induced mucus stasis and airway surface liquid volume depletion. Am J Physiol Lung Cell Mol Physiol 2013; 304: L746-756.

Abstract

La présente invention concerne des préparations pharmaceutiques de peptides spécialisés d'origine non naturelle destinés à se lier à des canaux sodiques et à inhiber l'activation desdits canaux sodiques. L'invention concerne, en outre, des méthodes de régulation de l'absorption du sodium et d'un volume de fluide, et de traitement de troubles sensibles à la modulation de l'absorption du sodium par modulation de la liaison de peptides spécialisés d'origine non naturelle aux canaux sodiques.
PCT/US2017/065862 2016-12-13 2017-12-12 Préparations salines de peptides splunc1 WO2018111903A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA3046361A CA3046361A1 (fr) 2016-12-13 2017-12-12 Preparations salines de peptides splunc1
EP17832396.0A EP3554527A1 (fr) 2016-12-13 2017-12-12 Préparations salines de peptides splunc1

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662433549P 2016-12-13 2016-12-13
US62/433,549 2016-12-13

Publications (1)

Publication Number Publication Date
WO2018111903A1 true WO2018111903A1 (fr) 2018-06-21

Family

ID=61006312

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/065862 WO2018111903A1 (fr) 2016-12-13 2017-12-12 Préparations salines de peptides splunc1

Country Status (3)

Country Link
EP (1) EP3554527A1 (fr)
CA (1) CA3046361A1 (fr)
WO (1) WO2018111903A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111317712A (zh) * 2020-02-27 2020-06-23 南京致中生物科技有限公司 一种盐酸左氧氟沙星注射液及其制备方法
WO2023141458A1 (fr) * 2022-01-18 2023-07-27 Emory University Traitement de la bronchiectasie

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4501729A (en) 1982-12-13 1985-02-26 Research Corporation Aerosolized amiloride treatment of retained pulmonary secretions
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US4957735A (en) 1984-06-12 1990-09-18 The University Of Tennessee Research Corporation Target-sensitive immunoliposomes- preparation and characterization
US20080260863A1 (en) * 2007-04-20 2008-10-23 Pre Holding, Inc. Compositions for mucociliary clearance and methods for administering same
US20120115795A1 (en) 2009-05-29 2012-05-10 Robert Tarran Regulation of Sodium Channels by PLUNC Proteins
WO2013043720A1 (fr) * 2011-09-20 2013-03-28 The University Of North Carolina At Chapel Hill Régulation de canaux sodiques par des protéines plunc
US20160102121A1 (en) 2014-10-08 2016-04-14 The University Of North Carolina At Chapel Hill Peptide Inhibitors of Sodium Channels
WO2017147128A1 (fr) * 2016-02-22 2017-08-31 The University Of North Carolina At Chapel Hill Inhibiteurs peptidiques de canaux calciques

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US4501729A (en) 1982-12-13 1985-02-26 Research Corporation Aerosolized amiloride treatment of retained pulmonary secretions
US4957735A (en) 1984-06-12 1990-09-18 The University Of Tennessee Research Corporation Target-sensitive immunoliposomes- preparation and characterization
US20080260863A1 (en) * 2007-04-20 2008-10-23 Pre Holding, Inc. Compositions for mucociliary clearance and methods for administering same
US20120115795A1 (en) 2009-05-29 2012-05-10 Robert Tarran Regulation of Sodium Channels by PLUNC Proteins
WO2013043720A1 (fr) * 2011-09-20 2013-03-28 The University Of North Carolina At Chapel Hill Régulation de canaux sodiques par des protéines plunc
US9127040B2 (en) 2011-09-20 2015-09-08 The University Of North Carolina At Chapel Hill Regulation of sodium channels by PLUNC proteins
US20160102121A1 (en) 2014-10-08 2016-04-14 The University Of North Carolina At Chapel Hill Peptide Inhibitors of Sodium Channels
WO2016057795A1 (fr) * 2014-10-08 2016-04-14 The University Of North Carolina At Chapel Hill Inhibiteurs peptidiques améliorés de canaux sodiques
WO2017147128A1 (fr) * 2016-02-22 2017-08-31 The University Of North Carolina At Chapel Hill Inhibiteurs peptidiques de canaux calciques

Non-Patent Citations (86)

* Cited by examiner, † Cited by third party
Title
"Remington, The Science And Practice of Pharmacy", 1995
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
ALTHAUS M: "ENaC inhibitors and airway re-hydration in cystic fibrosis: state of the art", CURR MOL PHARMACOL, vol. 6, 2013, pages 3 - 12
ALTON EW; ROGERS DF; LOGAN-SINCLAIR R; YACOUB M; BARNES PJ; GEDDES DM: "Bioelectric properties of cystic fibrosis airways obtained at heart-lung transplantation", THORAX, vol. 47, 1992, pages 1010 - 1014
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403
ALTSCHUL ET AL., METH. ENZYMOL., vol. 266, 1996, pages 460
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389
APP EM; KING M; HELFESRIEDER R; KOHLER D; MATTHYS H: "Acute and long-term amiloride inhalation in cystic fibrosis lung disease. A rational approach to cystic fibrosis therapy", AM REV RESPIR DIS, vol. 141, 1990, pages 605 - 612
BANGEL N; DAHLHOFF C; SOBCZAK K; WEBER WM; KUSCHE-VIHROG K: "Upregulated expression of ENaC in human CF nasal epithelium", J CYST FIBROS, vol. 7, 2008, pages 197 - 205, XP022664597, DOI: doi:10.1016/j.jcf.2007.07.012
BARTLETT ET AL., J. LEUKOC. BIOL., vol. 83, 2008, pages 1201
BENNETT WD; WU J; FULLER F; BALCAZAR JR; ZEMAN KL; DUCKWORTH H; DONN KH; O'RIORDAN TG; BOUCHER RC; DONALDSON SH: "Duration of action of hypertonic saline on mucociliary clearance in the normal lung", JAPPL PHYSIOL, vol. 118, 2015, pages 1483 - 1490
BINGLE ET AL., J. PATHOL., vol. 205, 2005, pages 491
BINGLE, C. D.; CRAVEN, C. J.: "PLUNC: a novel family of candidate host defense proteins expressed in the upper airways and nasopharynx", HUM MOL GENET, vol. 11, 2002, pages 937, XP002254846, DOI: doi:10.1093/hmg/11.8.937
BRIDGES ET AL., AM. J. PHYSIOL. LUNG CELL. MOL. PHYSIOL., vol. 281, 2001, pages L16
BURROWS EF; SOUTHERN KW; NOONE PG: "Sodium channel blockers for cystic fibrosis", COCHRANE DATABASE SYST REV, 2014, pages CD005087
CAMPOS, M. A. ET AL.: "Purification and characterization of PLUNC from human tracheobronchial secretions", AM J RESPIR CELL MOL BIOL, vol. 30, 2004, pages 184
CANTIN AM; HARTL D; KONSTAN MW; CHMIEL JF: "Inflammation in cystic fibrosis lung disease: Pathogenesis and therapy", J CYST FIBROS, vol. 14, 2015, pages 419 - 430
CHAMBERS ET AL., RESPIR. PHYSIOL. NEUROBIOL., vol. 159, 2007, pages 256
CHRAIBI ET AL., J. GEN. PHYSIOL., vol. 111, 1998, pages 127
CIOFU O; HANSEN CR; HOIBY N: "Respiratory bacterial infections in cystic fibrosis", CURR OPIN PULM MED, vol. 19, 2013, pages 251 - 258
DAVID W SCOTT ET AL: "SPX-101 Is a Novel Epithelial Sodium Channel-targeted Therapeutic for Cystic Fibrosis That Restores Mucus Transport", THE AMERICAN REVIEW OF RESPIRATORY DISEASE, AMERICAN THORACIC SOCIETY, US, vol. 196, no. 6, 15 September 2017 (2017-09-15), pages 734 - 744, XP009503564, ISSN: 0003-0805, DOI: 10.1164/RCCM.201612-2445OC *
DEVEREUX ET AL., NUCL. ACID RES., vol. 12, 1984, pages 387
DONALDSON ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 8338
DONALDSON SH; BENNETT WD; ZEMAN KL; KNOWLES MR; TARRAN R; BOUCHER RC: "Mucus clearance and lung function in cystic fibrosis with hypertonic saline", N ENGL J MED, vol. 354, 2006, pages 241 - 250, XP055285335, DOI: doi:10.1056/NEJMoa043891
DONALDSON SH; BOUCHER RC: "Sodium channels and cystic fibrosis", CHEST, vol. 132, 2007, pages 1631 - 1636
DONALDSON SH; HIRSH A; LI DC; HOLLOWAY G; CHAO J; BOUCHER RC; GABRIEL SE: "Regulation of the epithelial sodium channel by serine proteases in human airways", J BIOL CHEM, vol. 277, 2002, pages 8338 - 8345
EUR. RES.J., vol. S59, 2015, pages PA2054
FELLNER RC; TERRYAH ST; TARRAN R: "Inhaled protein/peptide-based therapies for respiratory disease", MOL CELL PEDIATR, vol. 3, 2016, pages 16
FENG; DOOLITTLE, J. MOL. EVOL., vol. 35, 1987, pages 351
FULCHER ML; GABRIEL S; BURNS KA; YANKASKAS JR; RANDELL SH: "Well-differentiated human airway epithelial cell cultures", METHODS MOL MED, vol. 107, 2005, pages 183 - 206
GAILLARD EA; KOTA P; GENTZSCH M; DOKHOLYAN NV; STUTTS MJ; TARRAN R: "Regulation of the epithelial Na+ channel and airway surface liquid volume by serine proteases", PFLUGERS ARCH, vol. 460, 2010, pages 1 - 17, XP019848547
GAILLARD ET AL., PFLEUGERS ARCH, vol. 460, 2010, pages 1 - 17
GANDERTON; JONES, DRUG DELIVERY TO THE RESPIRATORY TRACT, 1987
GARCIA-CABALLERO A; RASMUSSEN JE; GAILLARD E; WATSON MJ; OLSEN JC; DONALDSON SH; STUTTS MJ; TARRAN R: "SPLUNC1 regulates airway surface liquid volume by protecting ENaC from proteolytic cleavage", PROC NATL ACAD SCI USA, vol. 106, 2009, pages 11412 - 11417
GARLAND AL; WALTON WG; COAKLEY RD; TAN CD; GILMORE RC; HOBBS CA; TRIPATHY A; CLUNES LA; BENCHARIT S; STUTTS MJ: "Molecular basis for pH-dependent mucosal dehydration in cystic fibrosis airways", PROC NATL ACAD SCI U S A, vol. 110, 2013, pages 15973 - 15978
GARLAND ET AL., PROC NATL ACAD SCI USA., vol. 110, no. 40, 1 October 2013 (2013-10-01), pages 15973 - 15978
GONDA, CRITICAL REVIEWS IN THERAPEUTIC DRUG CARRIER SYSTEMS, vol. 6, 1990, pages 273 - 313
GRAHAM A; HASANI A; ALTON EW; MARTIN GP; MARRIOTT C; HODSON ME; CLARKE SW; GEDDES DM: "No added benefit from nebulized amiloride in patients with cystic fibrosis", EUR RESPIR J, vol. 6, 1993, pages 1243 - 1248, XP001063214
HAQ IJ; GRAY MA; GARNETT JP; WARD C; BRODLIE M: "Airway surface liquid homeostasis in cystic fibrosis: pathophysiology and therapeutic targets", THORAX, vol. 71, 2016, pages 284 - 287
HIGGINS; SHARP, CABIOS, vol. 5, 1989, pages 151
HIRSH AJ: "Altering airway surface liquid volume: inhalation therapy with amiloride and hyperosmotic agents", ADV DRUG DELIV REV, vol. 54, 2002, pages 1445 - 1462, XP002478966, DOI: doi:10.1016/S0169-409X(02)00161-8
HIRSH AJ; SABATER JR; ZAMURS A; SMITH RT; PARADISO AM; HOPKINS S; ABRAHAM WM; BOUCHER RC: "Evaluation of second generation amiloride analogs as therapy for cystic fibrosis lung disease", J PHARMACOL EXP THER, vol. 311, 2004, pages 929 - 938, XP002437463, DOI: doi:10.1124/jpet.104.071886
HO ET AL., BIOCHEMISTRY, vol. 25, 1986, pages 5500
HO ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 13973
HO ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 13979
HOBBS ET AL., AM J PHYSIOL LUNG CELL MOL PHYSIOL., vol. 305, no. 12, December 2013 (2013-12-01), pages L990 - L1001
HOBBS ET AL., J. PHYSIOL., vol. 591, 2013, pages 4377
HOFMANN T; SENIER I; BITTNER P; HULS G; SCHWANDT HJ; LINDEMANN H: "Aerosolized amiloride: dose effect on nasal bioelectric properties, pharmacokinetics, and effect on sputum expectoration in patients with cystic fibrosis", J AEROSOL MED, vol. 10, 1997, pages 147 - 158
HURT K; BILTON D: "Inhaled interventions in cystic fibrosis: mucoactive and antibiotic therapies", RESPIRATION, vol. 88, 2014, pages 441 - 448, XP055364921, DOI: doi:10.1159/000369533
KARLIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873
KNOWLES ET AL., J. CLIN. INVEST., vol. 109, 2002, pages 571
KNOWLES M; GATZY J; BOUCHER R: "Increased bioelectric potential difference across respiratory epithelia in cystic fibrosis", N ENGL J MED, vol. 305, 1981, pages 1489 - 1495
KNOWLES M; GATZY J; BOUCHER R: "Relative ion permeability of normal and cystic fibrosis nasal epithelium", J CLIN INVEST, vol. 71, 1983, pages 1410 - 1417
KYTE; DOOLITTLE, J. MOL. BIOL., vol. 157, 1982, pages 105
LENNOX ALISON ET AL: "SPX-101 Is a Promising and Novel Nebulized ENaC Inhibitor", THE AMERICAN REVIEW OF RESPIRATORY DISEASE, AMERICAN THORACIC SOCIETY, US, vol. 196, no. 6, 15 September 2017 (2017-09-15), pages 671 - 672, XP009503563, ISSN: 0003-0805, Retrieved from the Internet <URL:http://www.atsjournals.org/doi/abs/10.1164/rccm.201705-0928ED> DOI: 10.1164/RCCM.201705-0928ED *
LINDAHL, M.; STAHLBOM, B.; TAGESSON, C.: "Identification of a new potential airway irritation marker, palate lung nasal epithelial clone protein, in human nasal lavage fluid with two-dimensional electrophoresis and matrix-assisted laser desorption/ionization-time of flight", ELECTROPHORESIS, vol. 22, 2001, pages 1795, XP002405356, DOI: doi:10.1002/1522-2683(200105)22:9<1795::AID-ELPS1795>3.0.CO;2-J
LIVRAGHI, A. ET AL., J IMMUNOL, vol. 182, 2009, pages 4357 - 4367
MALL M; GRUBB BR; HARKEMA JR; O'NEAL WK; BOUCHER RC: "Increased airway epithelial Na+ absorption produces cystic fibrosis-like lung disease in mice", NAT MED, vol. 10, 2004, pages 487 - 493, XP002313818, DOI: doi:10.1038/nm1028
MALL MA; GRAEBER SY; STAHL M; ZHOU-SUCKOW Z: "Early cystic fibrosis lung disease: Role of airway surface dehydration and lessons from preventive rehydration therapies in mice", INT J BIOCHEM CELL BIOL, vol. 52, 2014, pages 174 - 179
MYERBURG ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 27942
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
O'RIORDAN TG; DONN KH; HODSMAN P; ANSEDE JH; NEWCOMB T; LEWIS SA; FLITTER WD; WHITE VS; JOHNSON MR; MONTGOMERY AB: "Acute hyperkalemia associated with inhalation of a potent ENaC antagonist: Phase 1 trial of GS-9411", J AEROSOL MED PULM DRUG DELIV, vol. 27, 2014, pages 200 - 208
PEARSON; LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PLANES ET AL., CURR. TOP. DEV. BIOL., vol. 78, 2007, pages 23
PUCHELLE E; BAJOLET O; ABELY M: "Airway mucus in cystic fibrosis", PAEDIATR RESPIR REV, vol. 3, 2002, pages 115 - 119
RAEBURN ET AL., J. PHARMACOL. TOXICOL. METH., vol. 27, 1992, pages 143
RIORDAN JR: "CFTR function and prospects for therapy", ANNU REV BIOCHEM, vol. 77, 2008, pages 701 - 726
RODGERS HC; KNOX AJ: "The effect of topical benzamil and amiloride on nasal potential difference in cystic fibrosis", EUR RESPIR J, vol. 14, 1999, pages 693 - 696
ROLLINS BM; GARCIA-CABALLERO A; STUTTS MJ; TARRAN R: "SPLUNC1 expression reduces surface levels of the epithelial sodium channel (ENaC) in Xenopus laevis oocytes", CHANNELS (AUSTIN, vol. 4, 2010, pages 255 - 259
ROSSIER, PROC. AM. THORAC. SOC., vol. 1, 2004, pages 4
SHTEINBERG M; ELBORN JS: "Use of inhaled tobramycin in cystic fibrosis", ADV THER, vol. 32, 2015, pages 1 - 9, XP035438364, DOI: doi:10.1007/s12325-015-0179-3
SMITH; WATERMAN, ADV. APPL. MATH., vol. 2, 1981, pages 482
STUTTS MJ; CANESSA CM; OLSEN JC; HAMRICK M; COHN JA; ROSSIER BC; BOUCHER RC: "CFTR as a cAMP-dependent regulator of sodium channels", SCIENCE, vol. 269, 1995, pages 847 - 850
TAN ET AL., J PHYSIOL., vol. 592, no. 23, 1 December 2014 (2014-12-01), pages 5251 - 5268
TARRAN ET AL., J. GEN. PHYSIOL., vol. 127, 2006, pages 591
TARRAN R; REDINBO MR: "Mammalian short palate lung and nasal epithelial clone 1 (SPLUNC1) in pH-dependent airway hydration", INT J BIOCHEM CELL BIOL, vol. 52, 2014, pages 130 - 135
TARRAN R; SABATER JR; CLARKE TC; TAN CD; DAVIES CM; LIU J; YEUNG A; GARLAND AL; STUTTS MJ; ABRAHAM WM: "Nonantibiotic macrolides prevent human neutrophil elastase-induced mucus stasis and airway surface liquid volume depletion", AM J PHYSIOL LUNG CELL MOL PHYSIOL, vol. 304, 2013, pages L746 - 756
TYLE: "Pharm. Res.", vol. 3, 1986, pages: 318
VALLET ET AL., NATURE, vol. 389, 1997, pages 607
VAN GOOR F; HADIDA S; GROOTENHUIS PD; BURTON B; CAO D; NEUBERGER T; TURNBULL A; SINGH A; JOUBRAN J; HAZLEWOOD A: "Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770", PROC NATL ACAD SCI U S A, vol. 106, 2009, pages 18825 - 18830
VAN GOOR F; HADIDA S; GROOTENHUIS PD; BURTON B; STACK JH; STRALEY KS; DECKER CJ; MILLER M; MCCARTNEY J; OLSON ER: "Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809", PROC NATL ACAD SCI USA, vol. 108, 2011, pages 18843 - 18848, XP055200039, DOI: doi:10.1073/pnas.1105787108
WIELPUTZ MO; EICHINGER M; ZHOU Z; LEOTTA K; HIRTZ S; BARTLING SH; SEMMLER W; KAUCZOR HU; PUDERBACH M; MALL MA: "vivo monitoring of cystic fibrosis-like lung disease in mice by volumetric computed tomography", EUR RESPIR J, vol. 38, 2011, pages 1060 - 1070
YANG C; CHILVERS M; MONTGOMERY M; NOLAN SJ: "Dornase alfa for cystic fibrosis", COCHRANE DATABASE SYST REV, vol. 4, 2016, pages CD001127
ZHOU Z; DUERR J; JOHANNESSON B; SCHUBERT SC; TREIS D; HARM M; GRAEBER SY; DALPKE A; SCHULTZ C; MALL MA: "The ENaC-overexpressing mouse as a model of cystic fibrosis lung disease", J CYST FIBROS, vol. 10, no. 2, 2011, pages 172 - 182
ZHOU Z; TREIS D; SCHUBERT SC; HARM M; SCHATTERNY J; HIRTZ S; DUERR J; BOUCHER RC; MALL MA: "Preventive but not late amiloride therapy reduces morbidity and mortality of lung disease in betaENaC-overexpressing mice", AM J RESPIR CRIT CARE MED, vol. 178, 2008, pages 1245 - 1256
ZHOU, Z. ET AL., AM J RESPIR CRIT CARE MED, vol. 178, 2008, pages 1245 - 1256

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111317712A (zh) * 2020-02-27 2020-06-23 南京致中生物科技有限公司 一种盐酸左氧氟沙星注射液及其制备方法
WO2023141458A1 (fr) * 2022-01-18 2023-07-27 Emory University Traitement de la bronchiectasie

Also Published As

Publication number Publication date
EP3554527A1 (fr) 2019-10-23
CA3046361A1 (fr) 2018-06-21

Similar Documents

Publication Publication Date Title
Scott et al. SPX-101 is a novel epithelial sodium channel–targeted therapeutic for cystic fibrosis that restores mucus transport
DK2760463T3 (en) REGULATION OF SODIUM CHANNELS USING PLUNC PROTEINS
KR101328851B1 (ko) 개선된 특성을 가지는 재구성된 계면활성제
US7951781B2 (en) Methods and compositions related to PLUNC surfactant polypeptides
KR20120028892A (ko) 계면 활성 단백질 b (sp-b) 및 계면 활성 단백질 c (sp-c)의 유사체를 함유하는 개량된 재구성 계면 활성제 조성물
KR102122088B1 (ko) 재구성된 폐 계면활성제
US20230107377A1 (en) Synthetic lung surfactant with enhanced stability and effectiveness
US20190071468A1 (en) Peptide inhibitors of sodium channels
WO2018111903A1 (fr) Préparations salines de peptides splunc1
US20200031866A1 (en) Peptide inhibitors of sodium channels
Wu et al. CFTR dysfunction leads to defective bacterial eradication on cystic fibrosis airways
WO2024050300A2 (fr) Tensioactifs synthétiques pour inhiber une infection à coronavirus
CA3178840A1 (fr) Methode de traitement d&#39;un trouble inflammatoire
Bukovnik Biophysical studies of m2glyr modified sequences: The effect of electrostatics on ion channel selectivity
WO2015118546A1 (fr) Compositions mucolytiques et leurs utilisations

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: 17832396

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3046361

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017832396

Country of ref document: EP

Effective date: 20190715