WO2023218278A1 - Thérapie génique à long terme pour vessie hyperactive - Google Patents

Thérapie génique à long terme pour vessie hyperactive Download PDF

Info

Publication number
WO2023218278A1
WO2023218278A1 PCT/IB2023/054387 IB2023054387W WO2023218278A1 WO 2023218278 A1 WO2023218278 A1 WO 2023218278A1 IB 2023054387 W IB2023054387 W IB 2023054387W WO 2023218278 A1 WO2023218278 A1 WO 2023218278A1
Authority
WO
WIPO (PCT)
Prior art keywords
maxi
uro
oab
aspects
present disclosure
Prior art date
Application number
PCT/IB2023/054387
Other languages
English (en)
Inventor
Cornelia HAAG-MOLKENTELLER
Salim Khalil MUJAIS
Original Assignee
Urovant Sciences Gmbh
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 Urovant Sciences Gmbh filed Critical Urovant Sciences Gmbh
Publication of WO2023218278A1 publication Critical patent/WO2023218278A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0083Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the administration regime
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA

Definitions

  • the present invention relates generally to the field of gene therapy to improve one or more symptoms related to overactive bladder.
  • Overactive bladder is a chronic and sometimes debilitating condition of the lower urinary tract.
  • the function of the lower urinary tract is to store and periodically release urine. This requires the orchestration of storage and micturition reflexes which involve a variety of afferent and efferent neural pathways, leading to modulation of central and peripheral neuroeffector mechanisms, and resultant coordinated regulation of sympathetic and parasympathetic components of the autonomic nervous system as well as somatic motor pathways. These proximally regulate the contractile state of bladder (detrusor) and urethral smooth muscle, and urethral sphincter striated muscle.
  • BOTOX® onabotulinumtoxinA
  • BOTOX is administered via multiple intradetrusor injections into the bladder wall approximately every 6-months.
  • BOTOX has been associated with urinary tract infections (UTIs), dysuria, and urinary retention.
  • UTIs urinary tract infections
  • Other side effects are consistent with the mechanism of action of botulinum toxin and may include asthenia, generalized muscle weakness, diplopia, ptosis, dysphagia, dysphonia, dysarthria, urinary incontinence, and breathing difficulties. These symptoms have been reported hours to weeks after injection.
  • the administration of onabotulinumtoxinA, a bacterial toxin can trigger an immune system response in a significant number of patients, leading to the development of neutralizing antibodies and to resistance to the treatment.
  • the present disclosure provides a long-term gene therapy method of treating overactive bladder (OAB) in a subject in need thereof comprising the intradetrusoral administration of a single dose of 50 mg or 100 mg of URO-902, wherein the administration of the single dose achieves a sustained therapeutic effect of at least one symptom of OAB lasting for at least 48 weeks after the administration of the single dose.
  • OAB overactive bladder
  • the present disclosure also provides a long-term gene therapy method of treating overactive bladder (OAB) in a subject in need thereof comprising the intradetrusoral administration of a single dose of 50 mg or 100 mg of URO-902, wherein the administration of the single dose achieves a sustained therapeutic effect of at least one symptom of OAB lasting for at least 12 months, at least about 15 months, at least about 18 months, or at least about 24 months after the administration of the single dose.
  • OAB overactive bladder
  • a long-term gene therapy method of treating overactive bladder (OAB) in a subject in need thereof comprising the intradetrusoral administration of a single dose of a composition comprising an isolated nucleic acid encoding a Maxi-K potassium channel polypeptide of SEQ ID NO: 8 or 15, wherein the dose is at least about 50 mg, wherein the administration of the single dose achieves a sustained therapeutic effect of at least one symptom of OAB lasting for at least 48 weeks after the administration of the single dose.
  • the at least one symptom of OAB is selected from the group consisting of average daily number of micturitions, average daily number of urinary urgency episodes, average daily number of urgency urinary incontinence (UUI) episodes, OAB symptom bother score, patient global impression, nocturia, and any combination thereof.
  • the isolated nucleic acid encoding the Maxi-K potassium channel polypeptide comprises an open reading frame (ORF) set forth in SEQ ID NOS: 51, 52, or 53.
  • the isolated nucleic acid is a vector.
  • the vector comprises a sequence set forth in SEQ ID NOS: 16, 49, or 50.
  • the composition is administered by injection. In some aspects, the injection is intramuscular injection.
  • the intramuscular injections are administered at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more injection sites.
  • the injection sites are in the bladder detrusor muscle.
  • the volume of each injection is about 0.5 ml, about 1 ml, about 1.5 ml, or about 2 ml.
  • the injection sites are about 0.5 cm, about 1 cm, about 1.5 cm, or about 2 cm apart.
  • the injections are administered at a depth of injection of about 2 mm, 2.5 mm, 3 mm, 3.5 mm or 4 mm.
  • the subject is a human.
  • the human is a female.
  • the human is a male.
  • the human is 65 years or older.
  • human is under the age of 65 years.
  • the subject experiences three or fewer UUI episodes per day.
  • the subject experiences more than three UUI episodes per day.
  • the subject is treatment naive for OAB.
  • the subject is treatment experienced for OAB.
  • the subject's previous treatment experience for OAB comprised the use of onabotulinumtoxinA.
  • the use of onabotulinumtoxinA resulted in an immune response.
  • the immune response comprises onabotulinumtoxinA-neutralizing antibodies.
  • the subject is resistant or non-responsive to onabotulinumtoxinA therapy.
  • the subject has received previous oral pharmacologic therapy for OAB.
  • the previous oral pharmacologic therapy for OAB failed to treat OAB in the subject and/or failed to alleviate one or more symptoms of OAB in the subject.
  • the composition comprising an isolated nucleic acid encoding a Maxi-K potassium channel polypeptide of SEQ ID NO: 8 or 15 is URO-902.
  • the dose is about 50 mg, about 100 mg, about 150 mg, about 200 mg, or about 250 mg.
  • the sustained therapeutic effect is observed for at least about 12 months, at least about 15 months, at least about 18 months, or at least about 24 months.
  • FIG. 1 is a line plot of the least squares means change from baseline in average daily number of micturitions.
  • the results for the placebo group are depicted by the light grey dashed line
  • the results for the URO-902 24 mg group are depicted by the grey solid line
  • the results for the URO-902 48 mg group are depicted by the black solid line.
  • the data from the following time points is shown: baseline, week 2, week 6, and week 12.
  • FIG. 2 is a line plot that shows the least squares mean change from baseline in average daily number of urgency episodes.
  • the results for the placebo group are depicted by the light grey dashed line
  • the results for the URO-902 24 mg group are depicted by the grey solid line
  • the results for the URO-902 48 mg group are depicted by the black solid line.
  • the data from the following time points is shown: baseline, week 2, week 6, and week 12.
  • FIG. 3 is a line plot that shows the least squares mean change from baseline in average daily number of UUI episodes.
  • the results for the placebo group are depicted by the light grey dashed line
  • the results for the URO-902 24 mg group are depicted by the grey solid line
  • the results for the URO-902 48 mg group are depicted by the black solid line.
  • the data from the following time points is shown: baseline, week 2, week 6, and week 12.
  • FIG. 4 is a bar graph that shows the proportion of patient global impression of change responders for the placebo, URO-902 24 mg, and URO-902 48 mg groups.
  • FIG. 5 shows patient disposition in a clinical trial to determine the effect 24 mg and 48 mg doses of URO-902 in patients with OAB.
  • FIG. 6 shows patient baseline demographics and clinical characteristics (ITT-E Population).
  • FIG. 7 is a summary of safety (Safety Population).
  • FIG. 8 is a line plot showing change from baseline in Mean Daily Number of Micturitions.
  • LS least square; BL, baseline; Nominal P ⁇ 0.05 vs placebo: (*) URO-902 24 mg, (+) URO-902 48 mg. Efficacy data were not collected from patients who received additional OAB treatments after week 24.
  • FIG. 9 is a line plot showing change from baseline in Mean Daily Number of Urgency Episodes. An urgency episode is defined as the “need to urinate immediately” as indicated in the bladder diary.
  • LS least square; BL, baseline; Nominal P ⁇ 0.05 vs placebo: (*) URO-902 24 mg, (+) URO-902 48 mg. Efficacy data were not collected from patients who received additional OAB treatments after week 24.
  • FIG. 10 is a line plot showing change from baseline in Mean Daily Number of UUI Episodes. LS, least square; BL, baseline; UUI, urge urinary incontinence. Efficacy data were not collected from patients who received additional OAB treatments after week 24.
  • FIG. 11 shows number of patients at each time point (ITT-E Population). Includes all patients who were randomized and received study drug. BL, baseline; ITT-E, intent-to-treat exposed
  • FIG. 12 is a schematic representation showing differences between the long-term gene therapy treatment of OAB with URO-902 and treatment with botulinum neurotoxin A (onabotulinumtoxinA).
  • FIG. 13 is a schematic representation of the clinical trial described in Example 4.
  • FIG. 14 corresponds to the data presented in FIGS. 8, 9 and 10, but including placebo values after week 24. As indicated in FIGS. 8, 9, and 10 such data is not included after week 24 in those figures because patients from the placebo group were offered the possibility of receiving OAB treatments after week 24. Thus, the placebo group pre-week 24 and post-week 24 are different and not comparable.
  • compositions and methods of gene therapy for the long-term treatment of overactive bladder (OAB) and symptoms thereof are provided.
  • a primary goal of the compositions and methods disclosed herein is to restore normal smooth muscle function with reduced intervention and with less side effects with respect to state of the art treatments.
  • the present disclosure provides compositions ("Maxi-K compositions of the present disclosure”) comprising at least one polynucleotide that contains at least one open reading frame encoding a polypeptide comprising a subunit of the Maxi-K channel (Maxi-K), e.g., a Maxi-K alpha-subunit, a beta-subunit, or any combination thereof, suitable for administration to smooth muscle (e.g., the detrusor muscle of the bladder wall), to a subject in need thereof having OAB and symptoms thereof.
  • the Maxi-K composition of the present disclosure is URO-902.
  • the Maxi-K channel polypeptide e.g., the alpha subunit of the Maxi-K channel (hSlo) are expressed in smooth muscle cells of the target tissue, e.g., the bladder wall.
  • the resulting Maxi-K activity in the target tissue significantly alleviates, treats, or prevents the symptoms of OAB.
  • compositions and methods An important characteristic of the disclosed compositions and methods is that, advantageously with respect to conventional therapeutic interventions, they can be used for chronic diseases such as OAB that otherwise would require the continued administration of a drug, or multiple administrations over a certain time-period.
  • the disclosed gene therapy methods comprising the administration of a Maxi-K composition (e.g., URO-902) would require a single administration, e.g., once every 24 weeks, 36 weeks, or 48 weeks, or a series of administrations at long time intervals (several months to a year).
  • adherence to treatment issues that are prevalent in chronic diseases, particularly in subjects having severe symptoms can be obviated.
  • the disclosed methods of treatment show unprecedented duration of the therapeutic effect in the treatment of OAB resulting from gene therapy using URO-902, a vector comprising a polynucleotide encoding the alpha subunit of Maxi-K.
  • the therapeutic effect of a single administration of URO-902 in the bladder wall lasts at least for 12 months, and the data suggest that the duration of the effect may be much longer.
  • compositions and methods are suitable not only for the treatment of nerve induced smooth muscle dysfunctions (neurogenic dysfunction in overactive bladder), as is the case with botulinum neurotoxins, but also for the treatment of non-nerve induced smooth muscle dysfunction (non-neurogenic dysfunction in overactive bladder).
  • the disclosure includes aspects in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process.
  • the disclosure includes aspects in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.
  • the compositions and methods of this disclosure as described herein can employ, unless otherwise indicated, techniques and descriptions of molecular biology (including recombinant techniques), cell biology, biochemistry, immunochemistry and ophthalmic techniques, which are within the skill of those who practice in the art.
  • Such techniques include, e.g., methods for observing and analyzing smooth muscle function in a subject, cloning and propagation of recombinant virus, formulation of a pharmaceutical compositions, and biochemical purification and immunochemistry. Specific illustrations of suitable techniques can be had by reference to the examples herein. However, equivalent conventional procedures can, of course, also be used.
  • Nucleotides are referred to by their commonly accepted single-letter codes. Unless otherwise indicated, nucleic acids are written left to right in 5' to 3' orientation. Nucleotides are referred to herein by their commonly known one-letter symbols recommended by the IUPAC- IUB Biochemical Nomenclature Commission. Accordingly, A represents adenine, C represents cytosine, G represents guanine, T represents thymine, and U represents uracil.
  • Amino acids are referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Unless otherwise indicated, amino acid sequences are written left to right in amino to carboxy orientation.
  • the term "about” as used herein refers to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within one or more than one standard deviation per the practice in the art. Alternatively, “about” can mean a range of up to 20%. For example, “about,” if applied to a 100 mg dose of a Maxi-K compositions of the present disclosure (e.g., URO-902), would encompass doses between 80 mg and 120 mg of the Maxi-K composition of the present disclosure. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value.
  • Administered in combination means that two or more therapeutic agents, e.g., a Maxi-K composition of the present disclosure (e.g., URO-902), and a second agent, are administered to a subject at the same time or within an interval such that there can be an overlap of an effect of each agent on the patient.
  • the administrations of the agents are spaced sufficiently closely together such that a combinatorial (e.g., a synergistic) effect is achieved.
  • Simultaneous administration is not necessary for a therapy to be considered a combination therapy.
  • a combination therapy does not require simultaneous administration of two or more therapeutic agents. Instead, any additional treatment while the transgene is effectively being expressed in the target tissue is considered a combination therapy.
  • amino acid substitution refers to replacing an amino acid residue present in a parent or reference sequence (e.g., a wild type Maxi-K sequence) with another amino acid residue.
  • An amino acid can be substituted in a parent or reference sequence (e.g., a wild type Maxi-K polypeptide sequence), for example, via chemical peptide synthesis or through recombinant methods known in the art. Accordingly, a reference to a "substitution at position X" refers to the substitution of an amino acid present at position X with an alternative amino acid residue.
  • substitution patterns can be described according to the schema AnY, wherein A is the single letter code corresponding to the amino acid naturally or originally present at position n, and Y is the substituting amino acid residue.
  • substitution patterns can be described according to the schema An(YZ), wherein A is the single letter code corresponding to the amino acid residue substituting the amino acid naturally or originally present at position n, and Y and Z are alternative substituting amino acid residues that can replace A.
  • substitutions are conducted at the nucleic acid level, i.e., substituting an amino acid residue with an alternative amino acid residue is conducted by substituting the codon encoding the first amino acid with a codon encoding the second amino acid.
  • the term “approximately,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain aspects, the term “approximately” refers to a range of values that fall within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
  • a smooth muscle dysfunction such as overactive bladder
  • the term "associated with” means that the symptom, measurement, characteristic, or status in question is linked to the diagnosis, development, presence, or progression of that dysfunction.
  • An association can, but need not, be causatively linked to the disease.
  • a smooth muscle dysfunction e.g., poor bladder control
  • a lesion e.g., spinal cord injury
  • a neurodegenerative development e.g., multiple sclerosis
  • Conservative amino acid substitution is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, or histidine), acidic side chains (e.g., aspartic acid or glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, or cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, or tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, or
  • amino acid substitution is considered to be conservative.
  • a string of amino acids can be conservatively replaced with a structurally similar string that differs in order and/or composition of side chain family members.
  • Non-conservative amino acid substitutions include those in which (i) a residue having an electropositive side chain (e.g., Arg, His or Lys) is substituted for, or by, an electronegative residue (e.g., Glu or Asp), (ii) a hydrophilic residue (e.g., Ser or Thr) is substituted for, or by, a hydrophobic residue (e.g., Ala, Leu, He, Phe or Vai), (iii) a cysteine or proline is substituted for, or by, any other residue, or (iv) a residue having a bulky hydrophobic or aromatic side chain (e.g., Vai, His, He or Trp) is substituted for, or by, one having a smaller side chain (e.g., Ala or Ser) or no side chain (e.g., Gly).
  • an electropositive side chain e.g., Arg, His or Lys
  • an electronegative residue e.g., Glu or As
  • amino acid substitutions can be readily identified by persons of ordinary skill in the art.
  • a substitution can be taken from any one of D-alanine, glycine, beta-alanine, L-cysteine and D-cysteine.
  • a replacement can be any one of D-lysine, arginine, D-arginine, homo-arginine, methionine, D-methionine, ornithine, or D- ornithine.
  • substitutions in functionally important regions that can be expected to induce changes in the properties of isolated polypeptides are those in which (i) a polar residue, e.g., serine or threonine, is substituted for (or by) a hydrophobic residue, e.g., leucine, isoleucine, phenylalanine, or alanine; (ii) a cysteine residue is substituted for (or by) any other residue; (iii) a residue having an electropositive side chain, e.g., lysine, arginine or histidine, is substituted for (or by) a residue having an electronegative side chain, e.g., glutamic acid or aspartic acid; or (iv) a residue having a bulky side chain, e.g., phenylalanine, is substituted for (or by) one not having such a side chain, e.g., glycine.
  • a polar residue e.g
  • conserved refers to nucleotides or amino acid residues of a polynucleotide sequence or polypeptide sequence, respectively, that are those that occur unaltered in the same position of two or more sequences being compared. Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.
  • two or more sequences are said to be “completely conserved” or “identical” if they are 100% identical to one another.
  • two or more sequences are said to be “highly conserved” if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another.
  • two or more sequences are said to be “highly conserved” if they are about 70% identical, about 80% identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another.
  • two or more sequences are said to be "conserved” if they are at least 30% identical, at least 40% identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some aspects, two or more sequences are said to be “conserved” if they are about 30% identical, about 40% identical, about 50% identical, about 60% identical, about 70% identical, about 80% identical, about 90% identical, about 95% identical, about 98% identical, or about 99% identical to one another. Conservation of sequence can apply to the entire length of a polynucleotide or polypeptide or can apply to a portion, region or feature thereof.
  • Detrusor As used herein, the term “detrusor” or “detrusor muscle” refers to the muscle of the bladder. By “intradetrusorally” is meant into the detrusor muscle. In some aspects, the compositions disclosed herein are injected intradetrusorally (i.e., in the detrusor muscle).
  • Detrusor overactivity refers to the occurrence of involuntary detrusor muscle contractions, e.g., during filling cystometry. These contractions, which can be spontaneous or provoked, are unable to be suppressed by the patient. They can take a wave (phasic) form, of variable duration and amplitude, on the cystometrogram. Urgency is generally associated in women with normal bladder sensation though contractions can be asymptomatic or can be interpreted as a normal desire to void. Urinary incontinence may or may not occur. A gradual increase in detrusor pressure without subsequent decrease is best regarded as a change in compliance.
  • Detrusor overactivity is defined by the International Continence Society (ICS) as follows: Detrusor overactivity is a urodynamic observation characterized by involuntary detrusor contractions during the filling phase that can be spontaneous or provoked (Abrams P et al., Urology 2003, 62(Supplement 5B): 28-37 and 40- 42).
  • Effective Amount As used herein, the term "effective amount" of a Maxi-K composition of the present disclosure (e.g., URO-902) in any dosage form, pharmaceutical composition, or formulation, is that amount sufficient to effect beneficial or desired results. In some aspects, the beneficial or desired results are, for example, clinical results, and, as such, an "effective amount” depends upon the context in which it is being applied. The term “effective amount” can be used interchangeably with “effective dose,” “therapeutically effective amount,” or “therapeutically effective dose.”
  • Expression vector' is a polynucleotide which, when introduced into an appropriate host cell, can be transcribed and translated into a Maxi-K polypeptide of the present disclosure (e.g, hSlo).
  • Polynucleotides encoding a Maxi-K polypeptide can be transfected into target cells (e.g., a smooth muscle cell in a target tissue, or a stem cell for subsequent administration to the target tissue) by any means known in the art, and be transcribed and translated into a Maxi-K polypeptide of the present disclosure (e.g., hSlo) in the target tissue.
  • target cells e.g., a smooth muscle cell in a target tissue, or a stem cell for subsequent administration to the target tissue
  • Such transfection methods are widely known in the state of the art.
  • homology refers to the overall relatedness between polymeric molecules, e.g., between nucleic acid molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Generally, the term “homology” implies an evolutionary relationship between two molecules. Thus, two molecules that are homologous will have a common evolutionary ancestor. In the context of the present disclosure, the term homology encompasses both to identity and similarity.
  • polymeric molecules are considered to be "homologous" to one another if at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the monomers in the molecule are identical (exactly the same monomer) or are similar (conservative substitutions).
  • the term “homologous” necessarily refers to a comparison between at least two sequences (polynucleotide or polypeptide sequences).
  • hSlo' The terms "Maxi-K alpha subunit,” “hSlo,” and “hSlol” are used interchangeably throughout the present specification.
  • Identity refers to the overall monomer conservation between polymeric molecules, e.g., between polypeptide molecules or polynucleotide molecules (e.g. DNA molecules and/or RNA molecules).
  • Calculation of the percent identity of two polynucleotide sequences can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes).
  • the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence.
  • the nucleotides at corresponding nucleotide positions are then compared.
  • Suitable software programs are available from various sources, and for alignment of both protein and nucleotide sequences.
  • One suitable program to determine percent sequence identity is bl2seq, part of the BLAST suite of program available from the U.S. government's National Center for Biotechnology Information BLAST web site (blast.ncbi.nlm.nih.gov).
  • B12seq performs a comparison between two sequences using either the BLASTN or BLASTP algorithm.
  • BLASTN is used to compare nucleic acid sequences
  • BLASTP is used to compare amino acid sequences.
  • Sequence alignments can be conducted using methods known in the art such as MAFFT, Clustal (ClustalW, Clustal X or Clustal Omega), MUSCLE, etc.
  • Different regions within a single polynucleotide or polypeptide target sequence that aligns with a polynucleotide or polypeptide reference sequence can each have their own percent sequence identity. It is noted that the percent sequence identity value is rounded to the nearest tenth. For example, 80.11, 80.12, 80.13, and 80.14 are rounded down to 80.1, while 80.15, 80.16, 80.17, 80.18, and 80.19 are rounded up to 80.2. It also is noted that the length value will always be an integer.
  • sequence alignments can be generated by integrating sequence data with data from heterogeneous sources such as structural data (e.g., crystallographic protein structures), functional data (e.g., location of mutations), or phylogenetic data.
  • a suitable program that integrates heterogeneous data to generate a multiple sequence alignment is T-Coffee, available at www.tcoffee.org, and alternatively available, e.g, from the EBI. It will also be appreciated that the final alignment used to calculate percent sequence identity can be curated either automatically or manually.
  • isolated refers to a substance or entity (e.g., polypeptide, polynucleotide, vector, cell, or composition that is in a form not found in nature) that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting).
  • Isolated substances e.g., nucleotide sequence or protein sequence
  • Isolated substances and/or entities can be separated from at least about 10%, at least about 15%, at least about 20%, at least 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least 95%, or more of the other components with which they were initially associated.
  • isolated substances are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure.
  • a substance is “pure” if it is substantially free of other components.
  • substantially isolated means that the compound is substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compound of the present disclosure.
  • Substantial separation can include compositions containing at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about
  • a polynucleotide, vector, polypeptide, cell, or any composition disclosed herein which is "isolated” is a polynucleotide (e.g., a nucleic acid encoding a Maxi-K polypeptide), vector, polypeptide, cell, or composition that is in a form not found in nature.
  • Isolated polynucleotides, vectors, polypeptides, or compositions include those that have been purified to a degree that they are no longer in a form in which they are found in nature.
  • a polynucleotide, vector, polypeptide, or composition that is isolated is substantially pure.
  • isolated nucleic acid refers to any type of isolated nucleic acid, it can notably be natural or synthetic, DNA or RNA, single or double stranded. In particular, where the nucleic acid is synthetic, it can comprise nonnatural modifications of the bases or bonds, in particular for increasing the resistance to degradation of the nucleic acid.
  • the modifications notably encompass capping its ends or modifying the 2' position of the ribose backbone so as to decrease the reactivity of the hydroxyl moiety, for instances by suppressing the hydroxyl moiety (to yield a 2'-deoxyribose or a 2'-deoxyribose-2'-fluororibose), or substituting the hydroxyl moiety with an alkyl group, such as methyl group (to yield a 2'-O-methyl-ribose.)
  • Modulate smooth muscle contraction' is intended to include the capacity to inhibit or stimulate smooth muscle contraction to various levels, e.g., which allows for the treatment of targeted states.
  • the language is also intended to include the inducement of relaxation of smooth muscle, e.g., total relaxation, and the contraction of smooth muscle that is in relaxed state and it is desired to have the muscle in a more contracted state.
  • mutation refers to the deletion, insertion, or substitution of any nucleotide, by chemical, enzymatic, or any other means, in a nucleic acid encoding a Maxi-K polypeptide (e.g., URO-902, which encodes hSlo) such that the amino acid sequence of the resulting polypeptide is altered at one or more amino acid residues.
  • a mutation in a nucleic acid sequence disclosed herein results in an amino acid substitution.
  • the mutation of a codon in a nucleic acid sequence disclosed herein wherein the resulting codon is a synonymous codon does not result in an amino acid substitution.
  • the nucleic acid sequences disclosed herein can be codon optimized by introducing one or more synonymous codon changes. Such codon optimization can, for example, (i) improve protein yield in recombinant protein expression, or (ii) improve the stability, half-life, or other desirable property of an mRNA or a DNA encoding a binding molecule disclosed herein, wherein such mRNA or DNA is administered to a subject in need thereof. [0075] Nocturia.
  • nocturia refers to a complaint of interruption of sleep one or more times because of the need to micturate. Each void is preceded and followed by sleep.
  • compositions and methods disclosed herein can be used to treat, prevent, or ameliorate nocturia.
  • Overactive bladder refers to urinary urgency, usually accompanied by frequency and nocturia, with or without urgency urinary incontinence, in the absence of urinary tract infection or other obvious pathology.
  • the term “overactive bladder” is defined by the International Continence Society (ICS) as follows: Overactive bladder (OAB) is a symptom complex consisting of urgency with or without urge incontinence, usually with frequency and nocturia, in the absence of local pathologic or hormonal factors (Abrams P et al., Urology 2003, 61(1): 37-49; Abrams P et al., Urology 2003, 62(Supplement 5B): 28-37 and 40-42). Synonyms of overactive bladder (OAB) include "urge syndrome” and "urge frequency syndrome”.
  • the compositions and methods disclosed herein can be used to treat, prevent, or ameliorate overactive bladder.
  • patient refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained professional for a particular disease or condition.
  • patient also encompasses any a human or non-human mammal affected or likely to be affected with a smooth muscle dysfunction such as overactive bladder.
  • composition refers to a preparation which is in such form as to permit the biological activity of the active ingredient (e.g., a Maxi-K composition of the present disclosure such as URO-902) to be effective, and which contains no additional components that are unacceptably toxic to a subject to which the composition would be administered.
  • a Maxi-K composition of the present disclosure such as URO-902
  • Such composition can be sterile.
  • compositions, and/or dosage forms are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • approval by a regulatory agency of the Federal or state governments (or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia) for use in animals, and more particularly in humans implies that those compounds, materials, compositions, and/or dosage forms are pharmaceutically acceptable.
  • Compounds, materials, compositions, and/or dosage forms that are generally acceptable as safe for therapeutically purposes are "therapeutically acceptable.”
  • Compounds, materials, compositions, and/or dosage forms that are generally acceptable as safe for diagnostic purposes are "diagnostically acceptable.”
  • compositions refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
  • Excipients can include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration.
  • antiadherents antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration.
  • Excipients that are generally accepted as safe for therapeutic purposes are "therapeutically acceptable excipients.”
  • compositions described herein also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid).
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are used.
  • compositions are found in Remington's Pharmaceutical Sciences, 17 th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth (eds.), Wiley-VCH, 2008, and Berge et al., Journal of Pharmaceutical Science, 66, 1-19 (1977), each of which is incorporated herein by reference in its entirety.
  • Pharmaceutically acceptable solvate' means a compound of the disclosure wherein molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered.
  • solvates can be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. When water is the solvent, the solvate is referred to as a "hydrate.”
  • Polynucleotide refers to polymers of nucleotides of any length, including ribonucleotides, deoxyribonucleotides, analogs thereof, or mixtures thereof. This term refers to the primary structure of the molecule. Thus, the term includes triple-, double- and single-stranded deoxyribonucleic acid ("DNA”), as well as triple-, double- and single-stranded ribonucleic acid (“RNA”). It also includes modified, for example by alkylation, and/or by capping, and unmodified forms of the polynucleotide.
  • DNA triple-, double- and single-stranded deoxyribonucleic acid
  • RNA triple-, double- and single-stranded ribonucleic acid
  • the polynucleotide comprises a DNA or an RNA (e.g., an mRNA), which can be synthetic.
  • the synthetic DNA or an RNA e.g., an mRNA, comprises at least one unnatural nucleobase.
  • T bases in the sequences disclosed herein are present in DNA, whereas the T bases would be replaced by U bases in corresponding RNAs.
  • Polypeptide The terms "polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer can comprise modified amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids such as homocysteine, ornithine, p-acetylphenylalanine, D-amino acids, and creatine), as well as other modifications known in the art.
  • a polypeptide can be a single polypeptide or can be a multi -molecular complex such as a dimer, trimer or tetramer.
  • the term "preventing” refers to partially or completely delaying onset of an disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular disease, disorder, and/or condition; partially or completely delaying progression from a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
  • Prophylactic Prophylactic.
  • prophylactic refers to a therapeutic or course of action used to prevent the onset of a disease or condition, or to prevent or delay a symptom associated with disease related to smooth muscle dysfunction.
  • compositions and methods disclosed herein can be applied prophylactically.
  • Prophylaxis refers to a measure taken to maintain health and prevent or delay the onset of a disease or condition related to smooth muscle dysfunction or to mitigate its extent and/or severity of the symptoms.
  • a prophylactic use of a therapeutic agent disclosed herein corresponds to that amount sufficient to effect beneficial or desired results.
  • any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • Similarity refers to the overall relatedness between polymeric molecules, e.g. between polynucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art.
  • Smooth muscle is intended to include smooth muscle sensitive to the Maxi-K compositions of the present disclosure (e.g., URO-902). Smooth muscle is sensitive to a Maxi-K composition of the present disclosure if the transgenically expressed Maxi-K polypeptide modulates the contraction of the smooth muscle.
  • Smooth muscle dysfunction' As used herein the term smooth muscle dysfunction related to any disease, condition, symptom, or sequelae that can be treated, prevented, or ameliorated by the transgenic expression of the Maxi-K compositions of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • Subject' By “subject” or “individual” or “animal” or “patient” or “mammal,” is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired. Mammalian subjects are not limited to humans. In certain aspects, the mammal is a human subject. In some aspects, the subject is a human patient. In a particular aspect, a subject is a human patient with a smooth muscle dysfunction. [0095] Substantially. As used herein, the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • Susceptible to An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms.
  • an individual who is susceptible to a disease, disorder, and/or condition can be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some aspects, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
  • Therapeutic agent is used in a broad sense to include a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902) that can provide a significant therapeutic benefit to a subject in need thereof, in particular, a subject suffering from a smooth muscle dysfunction such as overactive bladder.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902
  • therapeutic agent also encompasses prophylactic agents comprising a composition disclosed herein, wherein the therapeutic agent is administered, e.g., parenterally, topically, or via instillation.
  • the therapeutic agent is administered via injection into the bladder wall.
  • the therapeutic agent is administered via instillation into the subject's bladder.
  • Therapeutic agents of the present disclosure include not only agents that treat smooth muscle dysfunction such as overactive bladder, but also agents that can ameliorate and/or prevent any symptom associated with the presence of such dysfunction.
  • therapeutic agent would include, for example, agents that can reduce or suppress a particular symptom caused by the smooth muscle dysfunction.
  • Target tissue refers to any one or more tissue types of interest in which the delivery of a therapeutic and/or prophylactic agent of the present disclosure would result in a desired biological and/or pharmacological effect.
  • target tissues of interest include specific tissues, organs, and systems or groups thereof.
  • the target tissue can be any tissue comprising smooth muscle, e.g., bladder wall tissue.
  • Treating, treatment, therapy refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, reducing incidence of one or more symptoms or features of disease, or any combination thereof.
  • a treatment comprising a Maxi-K composition of the present disclosure can be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition, and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of, e.g., (i) decreasing the risk of developing a pathology associated with the disease, disorder, and/or condition, (ii) delaying the onset of the disease, disorder, and/or condition, or a pathology associated with said disease, disorder, and/or condition, or (iii) mitigating the symptoms and/or sequels of the disease, disorder, and/or condition or a pathology associated with said disease, disorder, and/or condition.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902
  • a treatment comprising a Maxi-K composition of the present disclosure can be administered to a subject who does not exhibit signs
  • treatment comprising a Maxi-K composition of the present disclosure can be administered to a subject who exhibits an increase in severity of one or more OAB symptoms and/or who is inadequately managed by existing treatments.
  • treatment refers to countering the effects caused as a result of the disease or pathological condition of interest in a subject including (i) inhibiting the progress of the disease or pathological condition, in other words, slowing or stopping the development or progression thereof, or one or more symptoms of such disorder or condition; (ii) relieving the disease or pathological condition, in other words, causing said disease or pathological condition, or the symptoms thereof, to regress; (iii) stabilizing the disease or pathological condition or one or more symptoms of such disorder or condition, (iv) reversing the disease or pathological condition or one or more symptoms of such disorder or condition to a normal state, (v) preventing the disease or pathological condition or one or more symptoms of such disorder or condition, and (vi) any combination thereof.
  • treatment period means the period of time during which the compositions of the disclosure, e.g., URO-902, show clinical efficacy after administration to a subject.
  • the treatment period can be from about 2 weeks to about 10 years.
  • the efficacy and/or safety of the compositions of the disclosure can be assessed by measuring certain parameters and calculating the changes from baseline over the treatment period. For example, indicating that treatment with URO-902 shows long-term efficacy during a 48 week treatment period means that statistically significant improvements from baseline (e.g., clinically relevant reductions in mean daily micturitions, urgency episodes, and/or UUI episodes) are observed 48 weeks after the administration of a single dose of URO-902.
  • the terms "treatment period” and “duration of effect” are used interchangeably in the present disclosure.
  • ug, uM, uL As used herein, the terms “ug,” “uM,” and “uL” are used interchangeably with “pg,” “pM,” and “pL” respectively.
  • Urge incontinence refers to a complaint of involuntary loss of urine.
  • Urgency urinary incontinence refers to a complaint of involuntary loss of urine associated with urgency.
  • Urinary urgency refers to a complaint of a sudden, compelling desire to void which is difficult to defer.
  • Urinary frequency refers to a complaint by the patient who considers that he/she voids too often by day.
  • Vector is a nucleic acid molecule, in particular self-replicating, which transfers an inserted nucleic acid molecule into and/or between host cells.
  • the term includes vectors that function primarily for insertion of DNA or RNA into a cell (e.g., chromosomal integration), replication of vectors that function primarily for the replication of DNA or RNA, and expression vectors that function for transcription and/or translation of the DNA or RNA.
  • a nucleic acid DNA or RNA, such as an mRNA
  • a binding molecule disclosed herein can take place in vitro (e.g., during recombinant protein production), whereas in other cases it can take place in vivo (e.g., administration of an mRNA to a subject), or ex vivo (e.g., DNA or RNA introduced into an autologous or heterologous cells for administration to a subject in need thereof).
  • vectors that provide more than one of the functions as described.
  • the term "vector” also refers in general to any nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • vector refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are replicated along with the host genome.
  • certain vectors, expression vectors are capable of directing the expression of genes to which they are operably linked.
  • the present disclosure provides methods of long-term gene therapy for treating overactive bladder and symptoms thereof.
  • the methods disclosed herein relate to long-term gene therapy comprising the administration of Maxi-K compositions of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902) encoding a hSlol protein, e.g., of SEQ ID NO: 8 or 15, to treat or to prevent overactive bladder and symptoms thereof in a subject in need thereof.
  • Maxi-K compositions of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902
  • a hSlol protein e.g., of SEQ ID NO: 8 or 15
  • Maxi-K channel and “BK channel” are used interchangeably herein. Structurally, Maxi-K channels are composed of alpha and beta subunits. Four alpha subunits form the pore of the channel, and these alpha subunits are encoded by a single Slol gene (also called Slo, hSlo, potassium calcium-activated channel subfamily M alpha 1, or KCNMA1).
  • Slol gene also called Slo, hSlo, potassium calcium-activated channel subfamily M alpha 1, or KCNMA1
  • compositions of the present disclosure comprise, e.g., (a) one or more polynucleotides encoding one or more Maxi-K polypeptides, and domains or combination of domains thereof (according to the domain boundaries known in the art); (b) one or more polynucleotides encoding one or more Maxi-K polypeptide sequences presented in TABLE 1 (e.g., Maxi-K alpha subunits, Maxi-K beta subunits, or combinations thereof), or fragments, isoforms, mutants, variants, or derivatives thereof or any combination thereof; (c) one or more polynucleotides encoding fusions or chimeric proteins comprising Maxi-K polypeptides disclosed herein, e.g., a Maxi-K alpha subunit genetically fused to a non-Maxi-K polypeptide conferring a desirable property, or
  • long-term gene therapy refers to the treatment of a subject (e.g., a subject with OAB) with a Maxi-K composition of the present disclosure, e.g., URO-902, wherein the treatment achieves long-term efficacy, e.g., clinically relevant reduction from baseline in a symptom of OAB selected from the group consisting of mean daily micturitions, urgency episodes, UUI episodes, and combinations thereof.
  • the long-term efficacy of the treatments disclosed herein refers to a clinically relevant reduction from baseline in a symptom of OAB that lasts for at least 6-months (24 weeks), 12-months (48 weeks), or longer, e.g., 18-months, 24-months, 30-months, or 36-months following the administration of a single dose of a Maxi-K composition of the present disclosure, e.g., URO-902.
  • the Maxi-K composition of the present disclosure comprises a pVAX-hSlo vector of SEQ ID NO: 16. In some aspects, the Maxi-K composition of the present disclosure comprises a pVAX-hSlo vector of SEQ ID NO: 49. In some aspects, the Maxi-K composition of the present disclosure comprises a pVAX-hSlo vector of SEQ ID NO: 50. In some aspects, the Maxi-K composition of the present disclosure comprises URO-902.
  • the present disclosure provide long term relief to patients suffering from OAB, reducing the invasiveness of the procedure and reducing adverse effects with respect to other methods known in the art, such as the administration of botulinum toxin to the detrusor muscle via intramuscular injection.
  • botulinum toxin which causes presynaptic denervation, requires multiple injections, repeated every few months, and since it is an exogenous protein (a bacterial toxin), the emergence of neutralizing antibodies and undesired side effects is frequent.
  • the present disclosure relates to the administration of a vector encoding an endogenous protein, i.e., a subunit of a channel that naturally occurs in the human body and therefore does not cause an immune reaction or the development of neutralizing antibodies as in the case of botulinum neurotoxin.
  • an endogenous protein i.e., a subunit of a channel that naturally occurs in the human body and therefore does not cause an immune reaction or the development of neutralizing antibodies as in the case of botulinum neurotoxin.
  • the administration of the claimed compositions can have long lasting effects over symptoms of OAB.
  • the relief caused by the administration of a Maxi-K composition of the present disclosure is observed at least 6 months after a single administration.
  • the length of this effect which was fully unexpected and unprecedented, has been observed at longer time ranges. Additional follow up of the results obtained in the clinical trials described in the application has shown that the therapeutic effects persist much longer, e.g., at least 48 weeks.
  • the data shows a stabilization of the observed effects, which indicates
  • the present disclosure provides a long-term gene therapy method to treat OAB comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to a subject in need thereof, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • a hSlol e.g., of SEQ ID NO: 8 or 15
  • the present disclosure provides a long-term gene therapy method to treat OAB comprising administering a URO-902 to a subject in need thereof by injection into the subject's urinary bladder, e.g., by direct injection into the detrusor muscle.
  • the present disclosure provides a long-term gene therapy method to prevent OAB comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to a subject in need thereof, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • a hSlol e.g., of SEQ ID NO: 8 or 15
  • the present disclosure provides a long-term gene therapy method to prevent OAB comprising administering URO-902 to a subject in need thereof by injection into the subject's urinary bladder, e.g., by direct injection into the detrusor muscle.
  • the present disclosure provides a long-term gene therapy method to treat or ameliorate at least one symptom of OAB comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to a subject in need thereof, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • the present disclosure provides a long-term gene therapy method to treat or ameliorate at least one symptom of OAB comprising administering URO-902 to a subject in need thereof by injection into the subject's urinary bladder, e.g., by direct injection into the detrusor muscle.
  • a long-term gene therapy method to reduce urgency and/or frequency of urination comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to a subject in need thereof, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • a hSlol e.g., of SEQ ID NO: 8 or 15
  • a long-term gene therapy method to reduce urgency and/or frequency of urination associated with OAB comprising administering URO-902 to a subject in need thereof by injection into the subject's urinary bladder, e.g., by direct injection into the detrusor muscle.
  • the present disclosure also provides a long-term gene therapy method to reduce UUI (urge urinary incontinence), e.g., associated with OAB, comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to a subject in need thereof, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • a hSlol e.g., of SEQ ID NO: 8 or 15
  • a subject in need thereof e.g., by injection, implantation, or instillation into the subject's urinar
  • the present disclosure also provides a long-term gene therapy method to reduce UUI associated with OAB, comprising administering URO-902 to a subject in need thereof by injection into the subject's urinary bladder, e.g., by direct injection into the detrusor muscle.
  • the present disclosure provides a long-term gene therapy method to treat OAB or a symptom thereof comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO-902) to a subject in need thereof, e.g., by injection into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle), wherein the treatment achieves at least one of changes (i) to (v) from baseline over a treatment period (e.g., 24-weeks, 48-weeks, 18-months, 24-months, 30- months, 36-months, or longer): (i) a decrease in average daily number of micturitions; (ii) a decrease in average daily number of urgency episodes; (iii) a decrease in average daily number of UUI episodes; (iv) a decrease in OAB symptom bother score; (v) an increase (improvement) on a Maxi-K composition of
  • the treatment achieves at least two of changes (i) to (v) over the treatment period, at least three of changes (i) to (v) over the treatment period, at least four of changes (i) to (v) over the treatment period, or all of changes (i) to (v) over the treatment period.
  • the treatment achieves changes (i) and (ii) over the treatment period.
  • the treatment achieves changes (i), (ii), (iv), and (v) over the treatment period.
  • the treatment achieves any combination of changes (i), (ii), (iii), (iv), and (v) over the treatment period where a person of ordinary skill can contemplate the combinatorial combinations.
  • the subject is a human.
  • the human is a female.
  • the human is a male.
  • the human is 65 years or older.
  • the human is under the age of 65 years.
  • the subject is treatment naive for OAB. In some aspects of the long-term gene therapy methods of the present disclosure, the subject is treatment experienced for OAB. In some aspects of the long-term gene therapy methods of the present disclosure, the subject's previous treatment experience for OAB comprised use of onabotulinumtoxinA. In some aspects of the long-term gene therapy methods of the present disclosure, the subject's previous treatment experience for OAB comprised an immune reaction to onabotulinumtoxinA. In some aspects of the long-term gene therapy methods of the present disclosure, the subject's previous treatment experience for OAB comprised the development of neutralizing antibodies against onabotulinumtoxinA.
  • the subject's previous treatment experience for OAB comprised the development of resistance to onabotulinumtoxinA.
  • the long-term gene therapy methods of the present disclosure can be administered to a subject that has had a negative reaction to treatment with onabotulinumtoxinA, developed neutralizing antibodies against onabotulinumtoxinA, developed resistance to onabotulinumtoxinA treatment, or in general has experienced side effects, lack of efficacy, or any other fact that makes the patient ineligible for onabotulinumtoxinA treatment or would make a clinician recommend non-administration of onabotulinumtoxinA treatment or discontinuing onabotulinumtoxinA treatment.
  • the subject has received previous oral pharmacologic therapy for OAB.
  • the subject has been non-responsive to the oral pharmacologic therapy or has experience side effects that would make a clinician recommend non-administration of the oral pharmacologic therapy or discontinuing the oral pharmacologic therapy.
  • the present disclosure also provides a long-term gene therapy method to increase the number and/or activity of Maxi-K channels in the detrusor smooth muscle cell membrane in a subject in need thereof comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to the subject, e.g., by injection, implantation, or instillation into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • a hSlol e.g., of SEQ ID NO: 8 or 15
  • the present disclosure also provides a long-term gene therapy method to increase the number of Maxi-K channels in the detrusor smooth muscle cell membrane in a subject in need thereof comprising administering URO-902 to the subject, e.g., by injection into the subject's urinary bladder (e.g., by direct injection into the detrusor muscle).
  • the Maxi-K composition of the present disclosure comprises a vector of SEQ ID NO: 16. In other aspects, the Maxi-K composition of the present disclosure comprises a vector of SEQ ID NO: 49. In other aspects, the Maxi-K composition of the present disclosure comprises a vector of SEQ ID NO: 50. In some aspects, the Maxi-K composition of the present disclosure comprises a combination thereof. In some aspects, the Maxi-K composition of the present disclosure is URO-902.
  • the Maxi-K composition of the present disclosure comprises a polynucleotide comprising a nucleic acid sequence of SEQ ID NO: 51, 52 or 53, wherein the nucleic acid sequence encodes a Maxi-K alpha subunit (hSlol) protein.
  • the Maxi-K composition of the present disclosure comprises a polynucleotide sequence comprising a nucleic acid sequence encoding a Maxi-K alpha subunit (hSlol) protein of SEQ ID NO: 54, 55, or 56.
  • hSlol Maxi-K alpha subunit
  • the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) protein comprising a Glycine amino acid at position 23. In some aspects, the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) protein comprising a Serine amino acid at position 23. In some aspects, the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) proterin comprising an Arginine amino acid at position 366. In some aspects, the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) protein comprising a Glycine amino acid at position 366.
  • the Maxi-K composition of the present disclosure encodes a
  • Maxi-K alpha subunit (hSlol) protein comprising a Glycine amino acid at position 23 and an Arginine amino acid at position 366, e.g., a Maxi-K alpha subunit of SEQ ID NO: 54.
  • the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) protein comprising a Glycine amino acid at position 23 and a Glycine amino acid at position 366, e.g., a Maxi-K alpha subunit of SEQ ID NO: 55.
  • the Maxi-K composition of the present disclosure encodes a Maxi-K alpha subunit (hSlol) protein comprising a Serine amino acid at position 23 and an Glycine amino acid at position 366, e.g., a Maxi-K alpha subunit of SEQ ID NO: 56.
  • hSlol Maxi-K alpha subunit
  • the Maxi-K composition of the present disclosure is a pVAX- hSlo construct derived from a pVAX-hSlo disclosed herein comprising at least a silent mutation which results in the expression of a Maxi-K alpha subunit polypeptide disclosed herein.
  • the Maxi-K composition of the present disclosure comprises a polynucleotide (e.g., a vector or an ORF) having at least about 70%, at least about 75%, at least 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% sequence identity to a Maxi- K-encoding polynucleotide sequence disclosed herein, wherein the polynucleotide encodes a hSlol protein, e.g., of SEQ ID NO: 8 or 15.
  • a polynucleotide e.g., a vector or an ORF having at least about 70%, at least about 75%, at least 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% sequence identity to a Maxi- K-encoding polynucleotide sequence disclosed herein, wherein the polynucleotide encodes a hSlol protein, e.g., of SEQ ID NO:
  • the Maxi-K compositions of the present disclosure encoding a hSlol protein can be administered using gene transfer techniques known in the art (e.g., naked DNA or mRNA, plasmids, viral vectors, or gene editing technologies such as CRISPR), resulting in the expression of a Maxi-K polypeptide (e.g., hSlo) in the target tissue.
  • gene transfer techniques e.g., naked DNA or mRNA, plasmids, viral vectors, or gene editing technologies such as CRISPR
  • delivery of a Maxi-K composition of the present disclosure to a subject in need thereof can be referred to as gene therapy.
  • the present disclosure provides long-term gene therapy methods to treat a smooth muscle dysfunction (e.g., overactive bladder) in a subject in need thereof comprising administering a Maxi-K composition of the present disclosure, i.e., at least one dose of a composition comprising an isolated nucleic acid encoding a Maxi-K potassium channel polypeptide (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50) encoding a hSlol, e.g., of SEQ ID NO: 8 or 15, to the subject, wherein the expression of the Maxi-K potassium channel polypeptide in smooth muscle cells of the subject modulates smooth muscle contractility.
  • a Maxi-K potassium channel polypeptide e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50
  • hSlol e.g., of SEQ ID NO: 8 or 15
  • Maxi-K potassium channel polypeptide or “Maxi-K polypeptide” are used interchangeably and refer, e.g., to Maxi-K alpha subunit (hSlo) or a fragment, variant, mutant, or derivative thereof.
  • the Maxi-K polypeptide is an hSlo of SEQ ID NO: 8.
  • the Maxi-K polypeptide is an hSlo of SEQ ID NO: 15.
  • the term "administered,” as applied to a Maxi-K polypeptide of the present disclosure does not refer to the administration of a recombinant polypeptide. Instead, it refers to the administration of a Maxi-K composition comprising a nucleic acid comprising a polynucleotide encoding a Maxi-K polypeptide (e.g., hSlo).
  • the Maxi-K polypeptide is a fragment, e.g., a Maxi-K functional fragment (e.g., an hSlo fragment).
  • the terminal "functional fragment” refers, .e.g., to an hSlo subunit that when incorporated in a Maxi-K channel results in a multimer with potassium channel activity.
  • the Maxi-K polypeptide functional fragment e.g., a functional fragment of a hSlol of SEQ ID NO:8 or 15, retains at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about
  • the Maxi-K polypeptide functional fragment exhibits an increase in activity with respect to the activity of the full sequence Maxi-K polypeptide
  • variant refers to a Maxi-K polypeptide sequence, e.g., an hSlo protein, that possesses some modification of a structural property of the native protein.
  • Specific variants of hSlo are, for example, SEQ ID NOS: 54, 55 or 56.
  • the variant is the result of naturally occurring alternative splicing.
  • the Maxi-K polypeptide e.g., hSlo
  • Exemplary splice variant forms (isoforms) of the Maxi-K alpha and beta subunits are included in TABLE 1.
  • an isolated nucleic acid encoding a Maxi-K potassium channel polypeptide of the present disclosure comprises a nucleic acid sequence disclosed in TABLE 1 or a fragment thereof capable of expressing a functional Maxi-K polypeptide, e.g., a hSlo subunit that when incorporated in a Maxi-K channel results in a multimer with potassium channel activity.
  • an isolated nucleic acid encoding the Maxi-K polypeptide of the present disclosure comprises a nucleic acid sequence disclosed in TABLE 1 (or a fragment thereof capable of assembling into a functional Maxi-K channel).
  • a Maxi-K polypeptide of the present disclosure can be at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% or about 100% identical to the wild sequence of a human Maxi-K polypeptide, e.g., a wild type Maxi-K polypeptide sequence disclosed in TABLE 1.
  • the smooth muscle dysfunction e.g., overactive bladder
  • the term idiopathic refers to a medical disease or condition having no known associated disease or cause, wherein the disease or condition is characterized by altered smooth muscle contractility.
  • the smooth muscle dysfunction e.g., overactive bladder
  • is neurogenic i.e., the smooth muscle dysfunction is due to a disease or injury of the central nervous system or peripheral nerves not involved in bladder smooth muscle control, for example, neurogenic bladder, spinal cord injury, or neurodegenerative diseases. Any condition that impairs bladder and bladder outlet afferent and efferent signaling can cause neurogenic bladder.
  • the neurogenic smooth muscle dysfunction is cause by a neurodegenerative disease, e.g., Parkinson's disease or multiple sclerosis.
  • the smooth muscle dysfunction e.g., overactive bladder, is non-neurogenic, i.e., it is not caused by pathological changes in smooth muscle innervation.
  • the Maxi-K compositions of the present disclosure e.g., URO- 902 have an onset of action of about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks following their administration.
  • “Onset of action,” as used herein, refers to the duration of time it takes for the physiological effect of a composition of the present disclosure, e.g., URO-902, to come to prominence upon administration.
  • the duration of effect of the Maxi-K compositions of the present disclosure is observed for at least about 24 weeks, at least about 36 weeks, at least about 48 weeks, at least about 6 months, at least about 9 months, at least about 12 months (1 year), at least about 15 months, at least about 18 months, at least about 21 months, at least 24 months (2 years), or more than 24 months.
  • the duration of effect of at least 6 months to at least about 24 months is observed after a single administration of a Maxi-K composition of the present disclosure, e.g., a vector disclosed herein that encodes hSlo, such as URO-902.
  • the duration of effect of the Maxi-K compositions of the present disclosure is observed for about 24 weeks, about 36 weeks, about 48 weeks, about 6 months, about 9 months, about 12 months (1 year), about 15 months, about 18 months, about 21 months, or about 24 months (2 years).
  • the duration of effect of about 6 months to about 24 months is observed after a single administration of a Maxi-K composition of the present disclosure, e.g., a vector disclosed herein that encodes hSlo, such as URO-902.
  • the duration of effect (for example, the duration of a therapeutic effect such as the improvement in at least a symptom of overactive bladder or detrusor overactivity) of a Maxi-K composition of the present disclosure, e.g., a vector disclosed herein that encodes hSlo, such as URO-902, for a dose of about 24 mg (24,000 mcg), about 48 mg, about 100 mg, about 150 mg, about 200 mg or about 250 mg is at least about 6 months, about 9 months, about 12 months (1 year), about 15 months, about 18 months, about 21 months, or about 24 months (2 years).
  • the duration of effect of about 6 months to about 24 months is observed after a single administration of a dose of a Maxi-K composition of the present disclosure, e.g., a vector disclosed herein that encodes hSlo, such as URO-902, between about 24 mg and about 250 mg.
  • the isolated nucleic acid sequence encoding a Maxi-K polypeptide of the present disclosure is a DNA, e.g., a naked DNA.
  • the isolated nucleic acid sequence encoding a Maxi-K polypeptide of the present disclosure is an RNA, for example, an mRNA (e.g., a naked RNA).
  • naked nucleic acid e.g., a “naked DNA” or a “naked RNA” is defined herein as a nucleic acid, e.g., a DNA or an RNA, not contained in a non-viral vector, e.g., a liposome or nanoparticle.
  • Maxi-K polynucleotide compositions of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a smooth muscle cell e.g., a bladder smooth muscle cells, for example in the detrusor
  • procedures known to one skilled in the art such as electroporation, DEAE Dextran, monocationic liposome fusion, polycationic liposome fusion, protoplast fusion, polynucleotide (e.g., DNA)-coated microprojectile bombardment, creation of an in vivo electrical field, injection with recombinant replication-defective viruses, homologous recombination, nanoparticles, and naked polynucleotide (e.g., DNA) transfer by, for example, intravesical instill
  • the Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a delivery agent e.g., a lipidoid, a liposome, a lipoplex, a lipid nanoparticle, a polymeric compound, a peptide, a protein, a cell, a nanoparticle mimic, a nanotube, or a conjugate.
  • the isolated nucleic acid or vector is incorporated into a cell in vivo, in vitro, or ex vivo.
  • the cell can be a stem cell, a muscle cell, or a fibroblast transfected with a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding an hSlo protein, e.g., of SEQ ID NO: 8 or 15.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • an hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a target cell or target tissue e.g., via direct injection into smooth muscle in the urinary bladder wall
  • the long-term gene therapy methods of the present disclosure provide for delivery of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, to bladder wall smooth muscle by injection (e.g., by direct injection into the detrusor muscle), implantation, or instillation.
  • the injection is intramuscular injection, in particular, injection into the smooth muscle of the bladder.
  • injections are administered at 1, 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, or more injection sites.
  • the Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • an hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • the dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding an hSlo protein, e.g., of SEQ ID NO: 8 or 15, is a single unit dose.
  • the single dose is administered in multiple injections sites, for example, in multiple injection sites in the detrusor muscle. In other words, if for example the single dose contains 100 mg of URO-902, and the single dose is administered in 20 different injection sites in the detrusor, each injection site will receive 5 mg of URO-902.
  • the single dose is injected into the detrusor muscle in 10 to 50 injections sites, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 43, 44, 45, 46, 47, 48, 49 or 50 injection sites.
  • the single dose is injected into the detrusor muscle in 10 to 20, 15 to 25, 20 to 30, 25 to 35, 30 to 40, 35 to 45, or 40 to 50 injections sites.
  • the dose of a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902 encoding an hSlo protein, e.g., of SEQ ID NO: 8 or 15, is about 24 mg of the composition (e.g., a naked nucleic acid, a plasmid, or a vector).
  • the dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding an hSlo protein, e.g., of SEQ ID NO: 8 or 15, is about 48 mg of the composition (e.g., a naked nucleic acid, a plasmid, or a vector).
  • the dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, comprises at least about 5 mg, at least about 10 mg, at least about 15 mg, at least about 20 mg, at least about 25 mg, at least about 30 mg, at least about 35 mg, at least about 40 mg, at least about 50 mg, at least about 75 mg, at least about 100 mg, at least about 125 mg, at least about 150 mg, at least about 175 mg, at least about 200 mg, at least about 225 mg, at least about 250 mg, at least 275 mg, at least about 300 mg, at least about 325 mg, at last about 350 mg, at least about 375 mg, at least about 400 mg, at least about 425 mg, at least about 450 mg, at least about 475 mg, or at least about 500
  • the dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, is between about 5 mg and about 10 mg, or between about 10 mg and about 15 mg, or between about 15 mg and about 20 mg, or between about 20 mg and about 25 mg, or between about 25 mg and about 30 mg, or between about 30 mg and about 35 mg, or between about 35 mg and about 40 mg, or between about 40 mg and about 45 mg, or between about 45 mg and about 50 mg, or between about 50 mg and about 75 mg, or between about 75 mg and about 100 mg, or between about 100 mg and about 125 mg, or between about 125 mg and about 150 mg, or between about 150 mg and about 175 mg, or between about 175 mg and about 200 mg, or between about 200 mg and about 225 mg, or between about 225
  • the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 24 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 48 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 50 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 75 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 100 mg of URO-902.
  • the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 125 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 150 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 175 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 200 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 225 mg of URO-902.
  • the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 250 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 275 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 300 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 325 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 350 mg of URO-902.
  • the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 375 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 400 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 425 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 450 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 475 mg of URO-902. In some aspects, the single dose of Maxi-K composition used in the long-term gene therapy methods of the present disclosure comprises about 500 mg of URO-902.
  • Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • an hSlo protein e.g., of SEQ ID NO: 8 or 15
  • the limiting factor in the administration of the Maxi-K composition of the present disclosure has been the solubility of the compositions.
  • the dose of Maxi-K composition of the present disclosure can be above 50 mg, 100 mg, 200 mg, 300 mg, 400 mg or 500 mg.
  • the Maxi-K compositions can be optimized to improve their solubility and/or to reduce precipitation and/or precipitation using methods known in the art, for example by incorporating (e.g., conjugating) hydrophilic polymers such as polyethylene glycols or polyglycerols in the delivery system..
  • the total dose of Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding an hSlo protein, e.g., of SEQ ID NO: 8 or 15, can be administered in a single administration (e.g., a single injection) or in multiple administrations (e.g., multiple injections).
  • the multiple injection are administered simultaneously (for example, within a short period of time, e.g., within 30 minutes, an hour, two hours, or the same day), wherein in other aspects a substantial period of time elapses between injection (e.g., one or more days between injections).
  • multiple doses are administered, for example, every month, every two months, every three months, every four months, every five months or every six months.
  • multiple doses are administered, every six months, every nine months, every 12 months, every 18 months, every 24 months, or more than every 24 months.
  • a subject with a urinary bladder smooth muscle dysfunction can receive a single total dose (unit dose) of, e.g., 16 mg, or 24 mg, or 48 mg, or 50 mg, or 100 mg, or higher doses as disclosed (e.g., up to 500 mg) of a Maxi-K composition of the present disclosure (e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902) administered, e.g., as multiple intramuscular injections (e.g., at 20-30 sites, or higher or lower numbers of sites as disclosed above) into the bladder wall (e.g., a target site below or inferior to the bladder midline).
  • a Maxi-K composition of the present disclosure e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K
  • a subject with a urinary bladder smooth muscle dysfunction can receive a single total dose (unit dose) of, e.g., 16 mg, or 24 mg, or 48 mg, or 50 mg, or 100 mg, or higher doses as disclosed (e.g., up to 500 mg) of a Maxi-K composition of the present disclosure (e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902) administered, e.g., as multiple intramuscular injections (e.g., at 20-30 sites, or higher or lower numbers of sites as disclosed above) into the detrusor muscle.
  • a Maxi-K composition of the present disclosure e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-90
  • a subject with a urinary bladder smooth muscle dysfunction can receive a single total dose (unit dose) of, e.g., 16 mg, or 24 mg, or 48 mg, or 50 mg, or 100 mg, or higher doses as disclosed (e.g., up to 500 mg) of a Maxi-K composition of the present disclosure (e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902) administered, e.g., as multiple intramuscular injections (e.g., at 20-30 sites, or higher or lower numbers of sites as disclosed above) into the trigone.
  • a Maxi-K composition of the present disclosure e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902
  • a Maxi-K composition of the present disclosure e.g., a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902
  • a plasmid such as a pVAX plasmid comprising a polynucleotide sequence encoding a Maxi-K alpha subunit, e.g., URO-902
  • injection sites e.g., at least about 10, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, or at least about 50 injections
  • the bladder wall e.g., detrusor muscle
  • the injection target site comprises the bladder base, the posterior and lateral bladder wall, or both.
  • the target site below (or inferior to) the bladder midline is selected from the regions consisting of the bladder base, the posterior and lateral bladder wall, the bladder base exclusive of the trigone, the bladder base exclusive of the trigone and the bladder neck, the trigone only, and the bladder neck only.
  • the bladder midline corresponds to approximately 2-3 cm above an imaginary line intersecting the trigone above the ureteral orifices.
  • a Maxi-K composition of the present disclosure e.g., a pVAX- hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a hSlo protein
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a hSlo protein
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a hSlo protein
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a Maxi-K composition of the present disclosure
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a Maxi-K composition of the present disclosure
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a Maxi-K composition of the present disclosure
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a Maxi-K composition of the present disclosure
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a hSlo protein
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a hSlo protein
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the injection sites are in the trigone. In some aspects, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the injection sites are in the detrusor.
  • about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the injection sites are in the lower part of the bladder wall. In some aspects, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the injection sites are in the base of the bladder.
  • the injections are located equidistantly in a grid pattern.
  • the distance between injection sites is at least about 0.5 cm, at least about 0.75 cm, at least about 1 cm, at least about 1.25 cm, at least about 1.5 cm, at least about 1.75 cm, or at least about 2 cm.
  • the distance between injection sites is about 0.5 cm, about 0.75 cm, about 1 cm, about 1.25 cm, about 1.5 cm, about 1.75 cm, or about 2 cm.
  • the depth of injection is about 1.5 mm, about 2 mm, about 2.5 mm, about 3.0 mm, about 3.5 mm, or about 4.0 mm into the detrusor, i.e., the needle is inserted approximately 1.5 mm, 2 mm, 2.5 mm, 3 mm, 3.5 mm, or 4 mm into the detrusor.
  • the depth of injection is about 1.5 mm, about 2 mm, about 2.5 mm, about 3.0, about 3.5 mm, or about 4.0 mm into the trigone, i.e., the needle is inserted approximately 1.5 mm, 2 mm,
  • the depth of injection is about
  • the needle is inserted approximately 1.5 mm, 2 mm, 2.5 mm, 3.0 mm, 3.5 mm, or 4 mm into the bladder wall.
  • the injection volume is about 0.5 ml, about 0.6 ml, about 0.7 ml, about 0.8 ml, about 0.9 ml, about 1 ml. about 1.1 ml, about 1.2 ml, about 1.3 ml, about 1.4 ml, or about 1.5 ml of a solution comprising a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e g., of SEQ ID NO: 8 or 15.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e g., of SEQ ID NO: 8 or 15.
  • a subject with a urinary bladder smooth muscle dysfunction can receive a total dose of, e.g., 16 mg, 24 mg, 48 mg, 50 mg, 100 mg, or higher of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, administered as, e.g., approximately 20 to 30 intramuscular injections into the lower part of the bladder wall. See, e.g., U.S. Prov. Appl.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • hSlo protein e.g., of SEQ ID NO: 8 or 15
  • instillation refers to a procedure during which a tube (e.g., a catheter) is first inserted into the bladder, and a medication is infused through so that it can coat the inside of the bladder for a short time.
  • the administration by instillation is conducted in an empty bladder.
  • the patient is mildly dehydrated to increase absorption of the instilled composition by the bladder.
  • the volume of solution instilled inside the bladder is at least about 50 ml, at least about 60 ml, at least about 70 ml, at least about 80 ml, at least about 90 ml, at least about 100 ml, at least about 110 ml, at least about 120 ml, at least about 130 ml, at least about 140 ml, at least about 150 ml, at least about 160 ml, at least about 170 ml, at least about 180 ml, at least about 190 ml, at least about 200 ml, at least about 210 ml, at least about 220 ml, at least about 230 ml, at least about 240 ml, at least about 250 ml, at least about 260 ml, at least about 270 ml, at least about 280 ml, at least about 290, or at least about 300 ml.
  • the solution instilled inside the bladder is held for at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 20 minutes, at least about 25 minutes, at least about 30 minutes, at least about 35 minutes, at least about 40 minutes, at least about 45 minutes, at least about 50 minutes, at least about 55 minutes, or at least about 60 minutes, before being emptied.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO- 902
  • a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50 such as URO- 902
  • an hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO- 902 a Maxi-K composition of the present disclosure
  • This disclosure also provides long-term gene therapy methods of treating a patient having or being at risk of having a disease or disorder related to smooth muscle dysfunction, e.g., overactive bladder, comprising administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, to the patient if a determination of the potential clinical effect of the administration of the Maxi-K composition according to the long-term gene therapy methods disclosed herein indicates that the patient can benefit from treatment with the Maxi-K composition.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a therapeutic agent comprising a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, to the patient if analysis of a sample obtained from the patient indicates that the patient would benefit from such treatment (e.g., because of upregulation or downregulation in the expression of Maxi-K in the sample).
  • a sample is obtained from the patient and is submitted for functional or genetic testing, for example, to a clinical laboratory.
  • a therapeutic agent comprising a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding
  • the disclosure also provides long-term gene therapy methods of treating a patient having or at risk of having a disease or disorder related to smooth muscle dysfunction, e.g., overactive bladder, comprising (a) measuring muscle tone and/or Maxi-K expression in a sample obtained from a patient having or at risk of having a disease or disorder; (b) determining whether the patient can benefit from the treatment with a therapeutic agent comprising a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, based on the presence/absence of normal muscle tone and/or Maxi-K expression levels; and, (c) advising a healthcare provider to administer the therapeutic agent to the patient if the muscle tone and/or Maxi-K expression levels are abnormal.
  • muscle tone is evaluated via surrogate measurements that are indicative of an altered muscle tone (e.
  • a clinical laboratory e.g., a genetic testing laboratory
  • clinician determining smooth muscle function will advise the healthcare provider or health care benefits provider as to whether the patient can benefit from a long-term gene therapy treatment with a particular Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15.
  • a particular Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15.
  • the clinical laboratory can advise the healthcare provider (e.g., a medical doctor or hospital) or healthcare benefits provider (e.g., a benefits administrator or a health care insurance company) as to whether the patient can benefit from the initiation, cessation, or modification of a long-term gene therapy treatment with a particular Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15.
  • a healthcare provider e.g., a medical doctor or hospital
  • healthcare benefits provider e.g., a benefits administrator or a health care insurance company
  • results of a test procedure determining the presence or absence of a smooth muscle dysfunction, risk of occurrence of a smooth muscle dysfunction, or presence or absence of a symptom related to a smooth muscle dysfunction conducted according to methods known in the art can be submitted to a healthcare provider or a healthcare benefits provider for determination of whether the patient's insurance will cover treatment with a certain Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15.
  • a certain Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15.
  • urodynamic studies can be used to assess the severity of the dysfunction, the response or lack of response to treatment with a Maxi-K composition of the present disclose (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, or to stratify a population of patients.
  • a Maxi-K composition of the present disclose e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • the disclosure provides a long-term gene therapy method of treating a patient having a smooth muscle dysfunction, e.g., overactive bladder, or at risk of having a smooth muscle dysfunction, wherein the method comprises (i) diagnosing, e.g., in a genetic testing laboratory or by a clinician, the presence or absence of a smooth muscle dysfunction or presence or absence of a symptom associated with such smooth muscle dysfunction; and (ii) advising a healthcare provider to administer or a health benefits provider to authorize the administration of a particular Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, to the patient if the diagnosis indicates that the patient can benefit from the long-term gene therapy treatment with the Maxi-K composition.
  • a particular Maxi-K composition of the present disclosure e.g., a pVAX-hS
  • the long-term gene therapy treatment method can comprise: (i) diagnosing, e.g., in a genetic testing laboratory or by a clinician, the presence or absence of a smooth muscle dysfunction, e.g., overactive bladder, or presence or absence of a symptom associated with such smooth muscle dysfunction; (ii) determining whether the diagnosis indicates that the patient can benefit from the treatment with a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15; and (iii) advising a healthcare provider the adjust the dosage or a health benefits provider to authorize the adjustment of the dosage of the Maxi-K composition of the present disclosure if indicated, e.g., to
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a Maxi-K compositions of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO- 902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • a standard treatment combination treatment
  • a Maxi-K composition of the present disclosure e.g., a pVAX- hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a Maxi-K composition of the present disclosure e.g., a pVAX- hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15
  • URO-902 a Maxi-K composition of the present disclosure
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a specific Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15, among several potential Maxi-K compositions of the present disclosure as options for treatment;
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a hSlo protein e.g., of SEQ ID NO: 8 or 15;
  • a Maxi-K composition disclosed herein e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • a healthcare provider, healthcare benefits provider, or counselor can provide treatment advice and/or lifestyle advice as part of a treatment.
  • a subject in response to the identification of a smooth muscle dysfunction treatable with a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15, a subject can be advised, e.g., to adjust his or her diet, to cease smoking, or to cease or reduce the ingestion of alcohol, in addition to being administered a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902) encoding a hSlo protein, e.g., of SEQ ID NO: 8 or 15.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, such as URO-902
  • the long-term gene therapy methods of the present disclosure are used to treat or alleviate a symptom of overactive bladder (OAB) syndrome or detrusor overactivity by introducing into bladder smooth muscle cells of the subject a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50), e.g., via injection into the bladder wall (e.g., detrusor muscle) and in particular, specific locations in the bladder wall (e.g., the trigone).
  • the nucleic acid is expressed in the bladder smooth cells such that bladder smooth muscle tone is regulated; thus, the regulation of bladder smooth muscle tone results in less heightened contractility of smooth muscle in the subject.
  • the single dose of Maxi-K composition of the present disclosure can comprise about 25 mg, about 50 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, or about 250 mg.
  • the dose of Maxi-K composition of the present disclosure that can be administered in a single dose injected, for example, in one of more sites (e.g., 20 to 30 sites) in the bladder wall muscle (e.g., the detrusor muscle) can be high as necessary to obtain the desired pharmacological effects as long as toxicity or adverse effects are absent.
  • the present disclosure provides Maxi-K compositions (e.g., URO-902) that can be administered, for example, according to the long-term gene therapy methods disclosed above.
  • the Maxi-K composition comprises a vector, e.g., pVAX, comprising an ORF encoding a Maxi-K alpha subunit (e.g., a wild type Maxi-K alpha subunit or Maxi-K mutant subunit disclosed herein).
  • the pVAX vector sequence comprises a sequence of SEQ ID NO: 10.
  • the pVAX vector sequence comprises a sequence with at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98% or at least about 99% identity to SEQ ID NO: 10.
  • the pVAX sequence comprises a substitution of G for A at position 2 of SEQ ID NO: 10, an additional G at position 5 of SEQ ID NO: 10, a substitution of T for C at position 1158 of SEQ ID NO: 10, a missing A at position 2092 of SEQ ID NO: 10, a substitution of T for C at position 2493 of SEQ ID NO: 10, or a combination thereof.
  • Promoters suitable for the practice of the long-term gene therapy methods of the present disclosure include, but are not limited to, constitutive promoters, tissue-specific promoters, and inducible promoters.
  • the promoter is a smooth muscle promoter.
  • the promoter is a muscle cell promoter.
  • the promoter is not an urothelium specific expression promoter.
  • expression of a Maxi-K polynucleotide sequence of the present disclosure is regulated by an inducible promoter.
  • the promoter is a smooth-muscle-specific promoter.
  • An example of a smooth-muscle- specific promoter is SM22a.
  • the vector comprises a SM22a promoter sequence, which can include but is not limited to sequences such as SEQ ID NO: 9.
  • the vector comprises a promoter that is a human cytomegalovirus intermediate-early promoter (CMEV) sequence, which can include but is not limited to sequences such as SEQ ID NO: 1.
  • CMEV human cytomegalovirus intermediate-early promoter
  • the vector comprises a T7 priming site, which can include but is not limited to sequences such as SEQ ID NO: 2.
  • the recombinant virus and/or plasmid used to express a Maxi-K polypeptide of the disclosure comprises a polyA (polyadenylation) sequence, such as those provided herein (e.g., a BGH polyA sequence).
  • a polyA sequence polyadenylation sequence
  • the present disclosure provides for a sequence comprising BGH polyA sequence, or portion of a BGH polyA sequence.
  • polyA sequences comprising a combination of one or more polyA sequences or sequence elements.
  • no polyA sequence is used.
  • one or more polyA sequences may be referred to as untranslated regions (UTRs), 3'UTRs, or termination sequences.
  • a BGH polyA can include but is not limited to sequences such as SEQ ID NO: 3.
  • the vector comprises a sequence encoding a replication origin sequence, such as those provided herein.
  • the origin of replication is a pUC origin of replication, e.g., SEQ ID NO: 4.
  • the vector can also comprise a selectable marker, e.g., a nucleic acid encoding resistance to kanamycin such as SEQ ID NO: 5.
  • a single point mutation in the alpha, or pore-forming, subunit of the human Maxi- K channel is more efficient in reducing smooth muscle dysfunction, e.g., detrusor overactivity (DO) in urinary bladder smooth muscle, than the wild type Maxi-K alpha subunit gene.
  • a single point mutation at nucleotide position 1054 of the Maxi-K alpha subunit gene that results in a substitution of a Threonine (T) for a Serine (S) at position 352 of the amino acid sequence (T352S) causes increased current of the Maxi-K channel at lower intracellular calcium ion concentrations when compared to the channels expressed by the non-mutated gene.
  • the single mutation improves conductivity in high glucose of high oxidative stress environments compared to genes having multiple mutations.
  • the Maxi-K alpha subunit encoded the T352S mutant e.g., incorporation into a pVAX to yield a pVAX-hSlo-T352S construct
  • the Maxi-K polynucleotide encoding Maxi-K alpha subunit comprises a point mutation at nucleic acid position 1054 when numbered in accordance with SEQ ID NO: 7.
  • This point mutation results in an amino acid substitution at position 352 of the Maxi-K alpha subunit when numbered in accordance with SEQ ID NO: 7.
  • the point mutation is a substitution of a Serine (S) for a Threonine (T) (e.g., T352S). Additional description of the mutation of T352S, including generation of this mutant, is described in U.S. Appl. No. 17/294,296 and Int'l Pub. No. W02020/100089, each of which is incorporated by reference herein. See also U.S. Appl. Pubis. 2008/0269159 and 2018/0126003; and WO2013151665A2, which are herein incorporated by reference in their entireties.
  • compositions comprising a cell, e.g., a smooth muscle cell or a stem cell, which expresses an exogenous DNA or RNA (e.g., mRNA) sequence encoding a protein involved in the regulation of smooth muscle tone, e.g., a Maxi-K polypeptide such as a Maxi-K alpha subunit.
  • exogenous means any DNA or RNA (e.g., an mRNA) that is introduced into an organism or cell.
  • the Maxi-K composition of the present disclosure comprises a cell, e.g., a muscle cell or a stem cell.
  • the muscle cell or stem cell is autologous.
  • the muscle cell or stem cell is allogeneic.
  • the Maxi-K sequences disclosed in the patents and application publications above can also be used as Maxi-K compositions of the disclosure, for the manufacture of such compositions, and for the treatment of smooth muscle dysfunctions as disclosed herein.
  • the Maxi-K sequences disclosed in the incorporated patents and application publications can be used in plasmids/vectors, e.g., for naked administration, in viral vectors, or in any system known in the art that can effectively introduce a nucleic acid into a host cell for expression in such host cell (e.g., a smooth muscle cell).
  • the present disclosure also provides pharmaceutical compositions comprising a Maxi-K composition of the present disclosure (e.g., URO-902).
  • a Maxi-K composition of the present disclosure can be administered with a delivery agent, e.g., a lipidoid, a liposome, a lipoplex, a lipid nanoparticle, a polymeric compound, a peptide, a protein, a cell, a nanoparticle mimic, a nanotube, or a conjugate.
  • a delivery agent e.g., a lipidoid, a liposome, a lipoplex, a lipid nanoparticle, a polymeric compound, a peptide, a protein, a cell, a nanoparticle mimic, a nanotube, or a conjugate.
  • a pharmaceutical composition is a formulation containing one or more active ingredients as well as one or more excipients, carriers, stabilizers or bulking agents, which is suitable for administration to a human patient to achieve a desired diagnostic result or therapeutic or prophylactic effect (e.g., to treat overactive bladder).
  • a pharmaceutical composition comprising a Maxi-K composition of the present disclosure can be formulated as a lyophilized (i.e. freeze-dried) or vacuum dried power that can be reconstituted with saline or water prior to administration to a patient.
  • the pharmaceutical composition comprising a Maxi-K composition of the present disclosure can be formulated as an aqueous solution.
  • a pharmaceutical composition comprising a Maxi-K composition of the present disclosure can contain a cryoprotectant, e.g., sucrose.
  • compositions comprising a Maxi-K composition of the present disclosure can include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the composition comprising a Maxi-K composition of the present disclosure must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the pharmaceutical compositions comprising a Maxi-K composition of the present disclosure can be included in a container, pack or dispenser together with instructions for administration.
  • a Maxi-K composition of the present disclosure can be encapsulated in nanoparticles.
  • the pharmaceutical composition comprising a Maxi-K composition of the present disclosure comprises substances which increase the viscosity of the suspension, such as sucrose or dextran, in the amount from about 1% to about 30% percent, such as about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 15%, about 20%, about 25%, or about 30 % (w/v).
  • the sucrose is about 2% to 10% (w/v).
  • the sucrose is about 10% to about 20% (w/v).
  • the sucrose is about 20% (w/v).
  • the sucrose is about 2% (w/v).
  • the sucrose is about 3% (w/v).
  • the sucrose is about 5% (w/v).
  • kits and articles of manufacture comprising Maxi-K compositions of the present disclosure (e.g., URO-902).
  • Packaged Maxi-K compositions of the present disclosure in kits can facilitate the application of the Maxi-K compositions to a subject in need thereof.
  • the kit comprises a Maxi-K polynucleotide of the disclosure (e.g., URO-902).
  • the kit comprises cells transfected with a Maxi-K composition of the present disclosure.
  • the kit comprises (i) a Maxi-K composition of the present disclosure (e.g., URO-902), and (ii) instructions for use, e.g., according to the long-term gene therapy methods disclosed herein.
  • the instructions can be in any desired form, including but not limited to a label, printed on a kit insert, printed on one or more containers, as well as electronically stored instructions provided on an electronic storage medium, such as a computer readable storage medium that permits the user to integrate the information and calculate a control dose.
  • kits and articles of manufacture can be affixed to packaging material or can be included as a package insert. While the instructions are typically written or printed materials they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated. Such media include, but are not limited to, electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. As used herein, the term "instructions" can include the address of an internet site that provides the instructions.
  • the kit comprises a Maxi-K composition of the present disclosure (e.g., URO-902), in one or more containers (e.g., each container comprising a unit dose, or a unit dose split among multiple containers).
  • the kit contains all the components necessary and/or sufficient to administer a Maxi-K composition of the present disclosure (e.g., URO-902), including vials or other container with the Maxi-K composition of the present disclosure, syringes, needles, controls, directions for performing assays, or any combination thereof.
  • URO-902 e.g., URO-902
  • a kit comprises: (a) a recombinant plasmid provided herein, e.g., pVAX-hSlo, and (b) instructions to administer to cells or an individual a therapeutically effective amount of the recombinant plasmid.
  • the kit comprises pharmaceutically acceptable salts or solutions for administering a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the kit can further comprise instructions for suitable operational parameters in the form of a label or a separate insert.
  • the kit may have standard instructions informing a physician or laboratory technician to prepare a dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the kit can further comprise a standard or control information so that a patient sample can be compared with the control information standard to determine if the test amount of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902) is a therapeutic amount.
  • a Maxi-K composition of the present disclosure e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902
  • the kit could further comprise devices for administration, such as a syringe, filter needle, extension tubing, cannula, or any combination thereof.
  • devices for administration such as a syringe, filter needle, extension tubing, cannula, or any combination thereof.
  • kit or article of manufacture can comprise multiple vials, each one of them containing a single dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the kit or article of manufacture can comprise one or more vials, each one of them comprising more than one dose of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the article of manufacture is a bag containing a solution of a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the article of manufacture is a bottle (e.g., a glass bottle or a plastic bottle) containing a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902).
  • the article of manufacture is a bag containing a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902) in powder form for reconstitution in an appropriate solvent.
  • the article of manufacture is a bottle (e.g., a glass bottle or a plastic bottle) containing a Maxi-K composition of the present disclosure (e.g., a pVAX-hSlo vector of SEQ ID NO: 16, 49, or 50, e.g., URO-902) in powder form for reconstitution in an appropriate solvent.
  • URO-902 (pVAX-hSlo) is a GMP manufactured double-stranded deoxyribonucleic acid (DNA)-plasmid vector based gene therapy product for the treatment of OAB containing a cDNA insert encoding the pore-forming a subunit of the human smooth muscle Maxi-K channel, hSlo.
  • the Maxi-K channel is involved in smooth muscle relaxation. The increased number of Maxi-K channels in the bladder detrusor smooth muscle cells associated with effective URO-902 treatment can improve this condition.
  • Phase 1 (Study ION-301) and Phase 2 (Study ION-04 ED) studies evaluating single intracavemous injections of URO-902 have been completed in male subjects with ED.
  • Single intracavemous injections of URO-902 were well tolerated in male subjects with ED (Studies ION-301 and ION-04 ED).
  • the majority of adverse events reported were mild to moderate in severity and not treatment-related. Only two SAEs were reported in each study and all were unrelated to study treatment.
  • the objectives of this study were (1) to evaluate the efficacy of a single dose of URO-902 24 mg and 48 mg (administered via intradetrusor injection), compared with placebo, in subjects with OAB and UUI up to 48 weeks post-dose, and (2) to evaluate the safety and tolerability of a single dose of URO-902 24 mg and 48 mg (administered via intradetrusor injection), compared with placebo, in subjects with OAB and UUI up to 48 weeks post-dose.
  • This study has no formal statistical primary endpoint hypothesis.
  • Study endpoints included efficacy endpoint, safety endpoints (e.g., adverse events), and other endpoints (e.g., hSlo cDNA concentrations in blood or urine).
  • Efficacy endpoint included, e.g., change from baseline at Week 12 in average daily number of UUI episodes; change from baseline at Week 12 in average daily number of micturitions; change from baseline at Week 12 in average daily number of urinary incontinence (UI) episodes; change from baseline at Week 12 in average daily number of urgency episodes; proportion of subjects achieving > 50%, > 75%, and 100% reduction from baseline at Week 12 in UUI episodes per day; change from baseline at Week 12 in average volume voided per micturition; health outcomes parameters (e.g., change from baseline at Week 12 in total summary score from the Urinary Incontinence-Specific Quality-of-Life Instrument (I-QOL), change from baseline at Week 12 in OAB Questionnaire (OAB-q) scores, or overall change of bladder symptoms based on the Patient Global Impression of
  • URO-902 (24 mg or 48 mg) was administered as intradetrusor injections via cystoscopy. A single treatment of URO-902 24 mg was administered to subjects in Cohort 1. Dose escalation was conducted only after unblinded review of safety data from all subjects in Cohort 1 up to Week 6. Study treatment at the higher dose (URO-902 48 mg) began only after it was deemed safe to proceed to Cohort 2.
  • Randomization/Stratification' Subjects were enrolled into two cohorts. In both cohorts, subjects were randomized in a 2:1 ratio to receive either URO-902 (24 mg or 48 mg) or placebo. Each cohort was randomized separately, and enrollment was sequential, starting with Cohort 1 (URO-902 24 mg and placebo) and followed by Cohort 2 (URO-902 48 mg and placebo). At the Randomization Visit, subjects in both Cohort 1 and Cohort 2 were randomized centrally to receive either a single treatment of URO-902 or matching placebo. Randomization was stratified by baseline UUI episodes per day and presence or absence of DO.
  • Cohort 1 was stratified by baseline UUI episodes per day ( ⁇ 3 vs >3) and presence or absence of detrusor overactivity, and cohort 2 was stratified by baseline UUI episodes per day and previous use of onabotulinumtoxinA (botulinum neurotoxin A).
  • Visit Schedule' Study visits were identical for Cohorts 1 and 2. Subjects were evaluated during a 2-week screening period for eligibility (Days -35 to -21). Eligible subjects were randomized to treatment at the Randomization Visit (Day -14 to Day -7) within each cohort; however, subjects were administered the study treatment via cystoscopy on Day 1. All subjects were evaluated at scheduled post-treatment clinic visits at Weeks 2, 6, 12, 18, and 24, or until the subject exits the study. Afterwards, two follow-up telephone visits were performed at Week 36 and Week 48.
  • Additional OAB treatment Starting at Week 24, subjects could request and be prescribed additional OAB treatment(s) at the clinical discretion of the investigator. Subjects who received additional OAB treatment(s) at Week 24 or after were only followed to assess adverse events at any future telephone visits (Week 36 and/or Week 48). No efficacy assessments was performed once a subject was prescribed an additional OAB treatment.
  • ITT-E intent-to-treat exposed
  • ITT-E modified
  • the safety population consisted of all subjects who received the study medication and were used to assess treatment-emergent adverse events and other safety evaluations based on actual treatment received.
  • ITT-E was used for demographics, baseline characteristics, and efficacy analyses up to Week 24.
  • the ITT-E population consisted of all subjects randomized and treated subjects from Cohorts 1 and 2.
  • ITT-E (modified) which consisted of subjects in the ITT-E who did not receive additional OAB treatment(s) after Week 24, was used to evaluate efficacy after Week 24.
  • Interim analyses was conducted when > 50% of subjects in Cohort 1 and/or when > 50% of subjects in Cohort 2 had completed at least 12 weeks of follow-up post-randomization (or prematurely exited the study prior to Week 12).
  • a planned interim analysis was performed to evaluate the objectives of the protocol at Week 12, after all subjects in Cohorts 1 and 2 had completed the Week 12 Visit (or prematurely exited the study prior to Week 12). The final analysis was performed after all subjects have completed the study. Data from completed URO-902 12-week interim analysis are summarized in EXAMPLE 2 and data from completed URO-902 48-week analysis are summarized in EXAMPLE 3.
  • CSH Cochran-Mantel-Haenszel
  • FIG. 5 A schematic representation of the study is provided in FIG. 5.
  • Eligible subjects were randomized to treatment within each cohort at the Randomization Visit; however, subjects were administered study treatment via cystoscopy on Day 1. All subjects were evaluated at scheduled post-treatment clinic visits at Weeks 2, 6, 12, 18, and 24, or until the subject exited the study. Afterwards, two follow-up telephone visits for assessment of safety were performed at Week 36 and Week 48. Subjects were enrolled into two cohorts, with approximately the same number of subjects randomized into each cohort. In both cohorts, subjects were randomized in a 2:1 ratio to receive either URO-902 (24 mg or 48 mg) or placebo.
  • Each cohort was randomized separately, and enrollment was sequential, starting with Cohort 1 (URO 902 24 mg and placebo) and followed by Cohort 2 (URO-902 48 mg and placebo). Subjects in both Cohort 1 and Cohort 2 were randomized centrally (Days 14 to 7) to receive either a single treatment of URO-902 or matching placebo. Randomization was stratified by baseline UUI episodes per day and presence or absence of DO.
  • human bladder surface area (average bladder volume of 400 mL) is 263.5 cm 2 . Therefore, the approximate dose relationship of human to rat bladder is 20: 1.
  • doses up to 25 mg by direct injection into the bladder wall/detrusor muscle were well tolerated.
  • the equivalent doses in rat were 0.222 to 0.480 mg for the highest human dose of 24 mg and 0.148 to 0.240 mg for the lower human dose of 16 mg which were given as a single administration by multiple direct bladder injections into the detrusor muscle. No clinically meaningful safety signals were identified at either the 16 mg or 24 mg dose in Study ION-03.
  • a starting dose of 24 mg was initially tested in the planned Phase 2a clinical study URO-902-2001 to evaluate the safety and efficacy of URO-902 in subjects with OAB and UUI.
  • Study URO-902-2001 had a dose-escalation design. Based on the unblinded review of observed safety data from all subjects in Cohort 1 (URO-902 24 mg) up to Week 6, the DSMB made the recommendation to proceed with Cohort 2.
  • End of Study Definition The end of the study was defined as the date of the last visit or last scheduled procedure (Week 48) shown in the schedule of activities for the last subject in the study. A subject was considered to have completed the study if she was treated, had not been discontinued for any reason, attended the scheduled exit visit of the cohort she was enrolled in, and was properly discharged from the study.
  • Study population The study was conducted in female subjects with OAB and UUI. Specific inclusion and exclusion criteria are specified below. Prospective approval of protocol deviations to recruitment and enrollment criteria, also known as protocol waivers or exemptions, were not permitted.
  • Inclusion Criteria Subjects met all of the following inclusion criteria to be eligible for participation in this study. (1) Capable of giving written informed consent, which includes compliance with the requirements and restriction listed in the consent form; (2) subject is female, aged 40 to 76 years old, at screening; (3) subject has symptoms of OAB (frequency and urgency) with UUI for a period of at least 6 months prior to screening, determined by documented subject history; (4) subject experiences > 1 episode of UUI per day (i.e., a total of > 3 UUI episodes over the 3-day subject bladder diary completed during the screening period); (5) subject experiences urinary frequency, defined as an average of > 8 micturitions (toilet voids) per day (i.e., a total of > 24 micturitions over the 3-day subject bladder diary completed during the screening period); (6) subject has not been adequately managed with > 1 oral or transdermal pharmacologic therapies for the treatment of their OAB symptoms (e.g., anticholinergics, beta-3 agonist, etc.), in the opinion of the investigator (not adequately managed
  • OAB
  • Subjects were excluded from participating in the study for any one of the following criteria assessed during the screening period and at the Randomization Visit: (1) subject has symptoms of OAB due to any known neurological reason (e.g., spinal cord injury, multiple sclerosis, cerebrovascular accident, Alzheimer's disease, Parkinson's disease, etc.); (2) subject has a predominance of stress incontinence in the opinion of the investigator, determined by subject history; (3) subject currently uses or plans to use medications or therapies to treat symptoms of OAB, including nocturia (subjects previously receiving these medications must have discontinued their use prior to the start of the Screening Visit as follows: for desmopressin, at least one day prior; for anticholinergic therapy, at least 14 days prior; for intravesical anticholinergic therapy, at least 4 weeks prior; for P3 agonists, at least 14 days prior); (4) subjects who have previously been treated with onabotulinumtoxinA (or any other toxin) for urological indications (subjects who have been treated with on urological indications (subject
  • Study Drugs Administered All eligible subjects enrolled into the study received a single double-blind treatment of either URO-902 or placebo based on the cohort they were enrolled in. URO-902 (24 mg or 48 mg) or matching placebo were administered as intradetrusor injections via cystoscopy. For Cohort 1, a single treatment of URO-902 24 mg or placebo was administered. Based on the unblinded review of observed safety data from all subjects in Cohort 1 up to Week 6, the DSMB made the recommendation to proceed with Cohort 2. Study treatment at the higher dose (URO-902 48 mg) began only after the DSMB had recommended it was safe to proceed to Cohort 2.
  • Cohort 1 URO-902 24 mg or matching placebo (phosphate buffered saline with 20% sucrose [PBS-20%]).
  • Cohort 2 URO-902 48 mg or matching placebo (PBS- 20%). TABLE 2A provides a summary on study drugs.
  • Day of Treatment Criteria For each subject in Cohort 1 or Cohort 2, a single treatment was administered on Day 1 after fulfillment of the following "day of treatment criteria" : (a) negative urine dipstick reagent strip test (for nitrates and leukocyte esterase); (b) if evaluated, negative urinalysis/urine culture/sensitivity results for a possible UTI have been reviewed; (c) subject is asymptomatic for a UTI, in the opinion of the investigator; (d) no presence of bladder stones prior to or at cystoscopy, and (e) investigator continues to deem that no condition or situation exists which, in the investigator's opinion, puts the subject at significant risk from receiving URO-902.
  • Treatment Administration If a subject was taking any anticoagulants or antiplatelet drugs, it was discussed with the subject's primary care physician (or internist, cardiologist, etc.), as deemed clinically necessary by the investigator, if the subject could discontinue these drugs for 2-3 days prior to the intradetrusor injections treatment and on the day of treatment. Subjects on an anticoagulant and/or anti-platelet therapy were managed appropriately to decrease the risk of bleeding, per the clinical judgment of the investigator.
  • a flexible or rigid cystoscope was used for administration of study treatment. Per local site practice lubricating gel was used to insert the cystoscope.
  • the bladder was instilled with a sufficient amount of saline to visualize the study injections.
  • One 12-mL syringe prefilled with 12 mL of study medication and one 1-mL syringe prefilled with PBS-20% were prepared and ready for treatment administration.
  • the injection needle was primed with approximately 0.5 mL of study drug.
  • the 12 mL of study drug were administered as 20 injections, each approximately 0.6 mL. Under direct visualization via cystoscopy, injections were distributed evenly across the detrusor wall and spaced approximately 1 cm apart, avoiding the bladder dome and trigone.
  • the needle To administer study medication (from the 12-mL syringe), the needle should be inserted approximately 2 mm into the detrusor for each injection. For the final injection site, a sufficient amount of PBS-20% (from the 1-mL syringe) was pushed through the injection needle to ensure delivery of the remaining amount of study medication. After injections were administered, the saline used for visualization was not be drained from the bladder to allow subjects to demonstrate the ability to void prior to leaving the clinic. Subjects remained in the clinic for at least 30 minutes for observation, and until a spontaneous void occurred. Subjects were instructed to contact the study site to report any adverse events that occurred within 48 hours following administration of study treatment.
  • Preparation/Handling/Storage When URO-902 and placebo were shipped to the clinical site, the site stored both products at -20°C. The day prior to administrations, the product was thawed and stored in the refrigerator at 2°C to 8°C overnight. The product was not be re-frozen after thawing. Study medication (URO-902 or placebo) could remain in the refrigerator (2°C to 8°C, in the original vial) for up to 14 days.
  • Urodynamic Parameters Urodynamic assessments was only performed at baseline after confirmation of subject eligibility during the Randomization Visit or at Day 1 (prior to treatment administration). A historical urodynamic study, performed no more than 90 days prior to the first day of screening, served as the baseline urodynamic assessment if the criteria detailed below were satisfied. At Week 12, all subjects underwent a second urodynamic assessment. The historical urodynamic study may be substituted for the baseline urodynamic assessment, if the following 3 criteria were met: (1) historical urodynamic study was obtained no more than 90 days prior to the first day of screening, (2) historical urodynamic results were available for evaluation by the central reader, and (3) subject was not being treated with any OAB medication or had discontinued OAB treatment.
  • the following urodynamic parameters were measured: (a) cystometric volume at 1 st sensation to void (CVl st sen), (b) maximum cystometric capacity (MCC), (c) maximum detrusor pressure during the storage phase (Pdetmax), (d) presence/absence of the first involuntary detrusor contraction (IDC) and, if present: volume at first IDC (VpmaxiDc) and maximum detrusor pressure during the first IDC (PmaxiDc). Additional related instructions were provided in the study manual.
  • Urine and blood samples for hSlo cDNA assessment were collected pre-treatment from subjects on Day 1 (treatment administration), Week 6 follow-up clinic visit and Week 24 follow-up clinic/exit visit.
  • the number of UUI episodes was defined as the number of times a subject had marked "urge” as the main reason for the leakage as indicated on the Bladder Diary; regardless of whether more than one reason for leakage in addition to "urge” was checked. Average daily number of UUI episodes was calculated using the daily entries in the Bladder Diary, which was completed prior to each study visit. Average daily number of UUI episodes was calculated as the total number of UUI episodes that occurred on a Diary Day divided by the number of Diary Days in the Bladder Diary.
  • a micturition was defined as "Urinated in Toilet.” Average daily micturitions at each study visit were calculated in the same manner as described above for UUI episodes. Urinary incontinence was defined as having any reason for leakage or “Accidental Urine Leakage.” An urgency episode was defined as the "Need to Urinate Immediately.”
  • Baseline was defined as the diary assessments collected during the screening period for all diary related efficacy endpoints and results of the questionnaires collected at the Day 1 Visit for all health outcome endpoints.
  • Baseline was defined as the diary assessments collected during the screening period for all diary related efficacy endpoints and results of the questionnaires collected at the Day 1 Visit for all health outcome endpoints.
  • continuous change from baseline endpoints e.g., change from baseline in average daily number of UUI episodes, change from baseline in average daily number of micturitions, change from baseline in average daily number of UI episodes, change from baseline in average daily number of urgency episodes, change from baseline in average volume voided per micturition, change from baseline in average LQOL total summary score, change from baseline in OAB-q score, and change from baseline in PGLC score
  • MMRM mixed model for repeated measures
  • This model corrected for dropout and accounted for the fact that measurements taken on the same subject over time tended to be correlated, by using all available information on subjects within the same covariate set to derive an estimate of the treatment effect for a drop-out free population.
  • the proportion of subjects who had > 50% reduction from baseline in UUI episodes at Week 12 was calculated for each treatment group.
  • responder analyses was also calculated for subjects who achieved > 75% and 100% decrease in episodes of UUI at Week 12 relative to baseline.
  • the Cochran-Mantel-Haenszel (CMH) method was utilized to compare the proportion of responders between the active and placebo groups.
  • Study design This was a prespecified, 12-week interim analysis of the 48-week multicenter, randomized, double-blind, placebo-controlled, dose-escalation study described in Example 1. Women aged 40-79 years with OAB and urge urinary incontinence (LTUI) who were not adequately managed with oral OAB medications were randomly assigned to receive singledose URO-902 24 mg, URO-902 48 mg, or placebo administered by intradetrusor injection via cystoscopy under local anesthesia. Safety and tolerability was assessed by adverse events (AEs) and post-void residual (PVR) urine.
  • AEs adverse events
  • PVR post-void residual
  • a 3-day bladder diary (e.g., voiding diary) was used to collect information to assess the exploratory efficacy endpoints related to the number of LUI, micturition, urgency, and UI episodes per day as well as one 24-hrs. volume voided of urine.
  • a goal of the study was to estimate the treatment effect. No formal hypothesis testing was conducted, and p-values are presented for descriptive purposes. Placebo data was pooled from both cohorts for summaries.
  • the safety population consisted of all subjects who received the study medication.
  • the ITT-E population consisted of all subjects randomized and treated subjects from Cohorts 1 and 2.
  • Results Of the 80 subjects randomized, 68 completed week 12, and 74 were included in the intent-to-treat population. Mean (SD) age was 64.7 (7.1) years, and 13.5% had prior treatment with onabotulinumtoxinA (see TABLE 2B).
  • TABLE 3 provides demographic and baseline characteristics for the ITT-E population for the following outcomes: UUI episodes, daily micturitions, urinary incontinence episodes, urgency episodes, and volume voided per micturition.
  • FIG. 14 shows the same data presented in FIGS. 8-11, but also includes data corresponding to the placebo group after 24 weeks.
  • placebo patients were offered the possibility to start OSB treatment, so the number of subjects on that group changed, becoming a different population compared to the population used as control during weeks 1 to 24.
  • Patients in the placebo group that chose not to start treatment were likely to have less severe symptoms or had seen a spontaneous improvement in their condition, therefore introducing a bias after week 24.
  • the study objective is to determine an optimal dose of URO-902 in terms of efficacy and safety by comparing a single dose of URO-902 50 mg and 100 mg (administered via intradetrusor injection) with placebo, in participants with symptoms of OAB and UUI up to 48 weeks post-dose.
  • Primary Efficacy Endpoints are (i) change from baseline at Week 12 in the average daily number of urgency episodes, and (ii) change from baseline at Week 12 in average daily number of UUI episodes.
  • Secondary Efficacy Endpoints are (i) change from baseline at Week 12 in average daily number of micturitions; (ii) change from baseline at Week 12 in average volume voided per micturition; (iii) change from baseline at Week 12 in OAB Questionnaire (OAB-q) scores (sub-scores and total score separately); (iv) change from baseline at Week 12 in total summary score from the urinary Incontinence-Specific Quality-of-Life Instrument (I-QOL); (v) overall change of overactive bladder symptoms based on the Patient Global Impression of Change (PGI-C) scale score at Week 12; (vi) change from baseline at Weeks 2, 6, 12, 18, 24, 36, and 48 in average daily number of micturitions; (vii) change from baseline at Weeks 2, 6, 12, 18, 24, 36, and 48 in OAB Questionnaire (OAB-q) symptom bother score; (viii) change from baseline at Weeks 2, 6, 12, 18, 24, 36, and 48 in I-QOL total summary score; (ix) change from baseline at Weeks 2, 6,
  • Exploratory Efficacy Endpoints are (i) proportion of participants with > 10- point reduction in OAB-q symptom bother score at Weeks 2, 6, 12, 18, 24, 36, and 48; (ii) proportion of participants with > 10-point increase (participants who achieve minimally important difference [MID]) in I-QOL total score at Weeks 2, 6, 12, 18, 24, 36, and 48; (iii) proportion of participants who answered “Much Better” or “Moderately Better” on the PGI-C at Weeks 2, 6, 12, 18, 24, 36, and 48; (iv) proportion of participants with zero incontinence episodes per day at Weeks 2, 6, 12, 18, 24, 36, and 48; (v) proportion of participants with zero night-time voids per 24 hours at Weeks 2, 6, 12, 18, 24, 36, and 48; (vi) change from baseline at Weeks 2, 6, 12, 18, 24, 36, and 48 in average daily number of total urinary incontinence (UI) episodes; (vii) change from baseline at Weeks 2, 6, 12, 18, 24, 36, and 48 in average daily number of U
  • Safety Endpoints include adverse events, serious adverse events, physical examination, vital signs (pulse rate, blood pressure, and body temperature), hematology and clinical chemistry, post-void residual (PVR) urine volume, clean intermittent catheterization (CIC) use, electrocardiograms (ECGs), concomitant medications and concurrent procedures.
  • Endpoints include hSlo cDNA concentrations in blood and urine, and immunogenicity.
  • Randomization and Dosing Visit participants will be randomized centrally to receive either a single treatment of URO-902 (50 mg or 100 mg) or matching placebo. Randomization will be stratified by age, gender, and by baseline UUI episodes per day (i.e., ⁇ 3 versus > 3 UUI episodes per day). Enrollment for participants with previous onabotulinumtoxinA use for a urological indication will be capped at 25% of the total participant population. Potential participants will undergo a screening period of up to 5 weeks (Days -35 to -1), during which they will be assessed for eligibility (Visit 1).
  • Eligible participants will be randomized at Baseline (Visit 2) and will be administered the study treatment via cystoscopy on the same day 1 (Day 1; Visit 2). All participants will be evaluated at scheduled post-treatment visits at Weeks 2, 6, 12, 18, 24, 36, and 48 (Visits 3 to 9, respectively), or until the participant exits the study. Participants who complete the Week 24 visit (Visit 7) may choose to either roll over into an open-label extension study or continue in this study through to study completion at Week 48 (Visit 9) when they will complete all assessments specified for the End-of-Study visit. See FIG. 13.
  • Treatment will be based on the following criteria (i) 24 weeks or longer has elapsed since the previous administration of study drug, and (ii) the investigator deems retreatment appropriate, and (iii) participant has not experienced a PVR > 200 mL during the study, and (iv) one of the following symptom criteria: >8 micturitions per day (i.e., a total of > 24 micturitions over the 3-day participant bladder diary) or >1 episode of UUI per day. Participants who do not wish to enroll in the open-label extension study can request and receive additional OAB treatment(s) at the clinical discretion of the investigator, at or after Week 24 (Visit 7). Participants who receive additional OAB treatment(s) will be followed through Week 48.
  • the study is intended to have 165 evaluable participants (55 per treatment group) at Week 12 (primary analysis time point); assuming a dropout rate of approximately 20%, it is estimated that 207 participants will be randomized.
  • the study will involve approximately 70 sites in North America.
  • the duration of the study is up to 53 weeks (5-week screening and randomization period, treatment administration on Day 1, and 48-week double-blind post- treatment/follow-up period).
  • the following analysis populations will be evaluated: Safety, and Intent-to-treat-Exposed (ITT-E).
  • the safety population will consist of all participants who received the study drug and will be used to assess treatment-emergent adverse events and other safety evaluations based on actual treatment received.
  • the ITT-E population will be used for demographics, baseline characteristics, and primary efficacy analyses; the ITT-E population will consist of all participants who were randomized and received study treatment.
  • a primary analysis will be conducted when all participants have completed at least 12 weeks of follow-up post treatment- (or prematurely exited the study prior to Week 12).
  • the final analysis will be performed after all participants have completed the study (at Week 48) or exit the study prematurely (including when they may rollover into the open -label extension study from Week 24 onwards). Details of the primary and final analyses, together with any sensitivity analyses, will be described in the Statistical Analysis Plan. This study has two coprimary endpoints. Descriptive statistics will be used to evaluate the efficacy and safety endpoints. For continuous efficacy endpoints, estimates of least squares means, standard error, and 95% confidence intervals (CI) will be presented for each treatment group. P-values from comparisons to placebo may be provided for the two primary endpoints; other p-values will be considered nominal.
  • the point estimate of the treatment difference and 95% confidence interval for the change from baseline at each visit for each continuous efficacy variables relative to placebo will be analyzed using a mixed-effect model for repeated measures (MMRM) method.
  • the analysis model will include terms for baseline value as a covariate, in addition to the terms for treatment, visit, and treatment by visit interaction.
  • the proportion of participants who achieve > 50% reduction from baseline in urgency episodes at Week 12 will be calculated for each treatment group.
  • responder analyses will also be calculated for participants who achieve > 75% and 100% decrease in episodes of urgency at Week 12 relative to baseline.
  • the Cochran-Mantel-Haenszel (CMH) method will be utilized to compare the proportion of responders between the active and placebo groups by adjusting for the stratification factors. Data for all visits will also be presented. Similar responder analyses will be performed for reductions in UUI.
  • the CMH method will also be used for the responder analyses of micturition frequency, quality of life questionnaire scores (OAB-q and I-QOL), PGI-C, night-time voids, and urinary incontinence.
  • OAB-q and I-QOL quality of life questionnaire scores
  • PGI-C night-time voids
  • urinary incontinence For safety variables, data from all participants who received study drug will be included. The incidence of adverse events will be summarized. The change from baseline in PVR urine volume will be
  • Sample Size Estimation The sample size for this study was based on a 2-sample t-test with effect size estimated using data from the Urovant Phase 2a study, URO-902-2001, as well as placebo data from a Phase 3 onabotulinumtoxinA OAB study, for the follow two primary endpoints: (i) for urgency, the improvement from baseline is assumed to be 2.3 episodes per day comparing to placebo, with standard deviation (SD) of 3.0 episodes; the power to detect the signal will be > 0.97; (ii) for UUI, the improvement over placebo is assumed to be 1.71 episodes per day comparing to placebo, with a SD of 3.2 episodes; the power to detect the signal will be 0.80.
  • SD standard deviation
  • DSMB Data and Safety Monitoring Board
  • Inclusion Criteria (1) participant is male or female aged > 18 years old at the Screening Visit (Visit 1); (2) participant is capable of giving written informed consent, which includes compliance with the requirements and restriction listed in the consent form; (3) participant has symptoms of OAB (frequency and urgency) with UUI for a period of at least 3 months prior to the Screening Visit (Visit 1), determined by documented participant history (urodynamic evaluation is not required): a.
  • OAB frequency and urgency
  • Participant experiences an average of > 1 episode of UUI per day (i.e., a total of > 3 UUI episodes over the 3-day participant bladder diary completed during the screening and randomization period), and ⁇ 12 episodes of UUI per day; (4) participant experiences urinary frequency, defined as an average of > 8 micturitions (toilet voids) per day (i.e., a total of > 24 micturitions over the 3-day participant bladder diary completed during the screening and randomization period); (5) for females of reproductive potential: Agrees to remain abstinent or use (or have their male partner use) an acceptable method of birth control each time the patient has intercourse from the Screening Visit until completion of the End of Study (EOS) Visit; (6) sexually active males with female partners must ensure that a double barrier method of contraception is used each time the patient has intercourse from the Screening Visit until completion of the EOS Visit; (7) for females of reproductive potential: Agrees not to donate ova (eggs) until at least 1 month after completion of the EOS Visit
  • participant has symptoms of OAB due to any known neurological reason (e.g, spinal cord injury, multiple sclerosis, cerebrovascular accident, Alzheimer’s disease, Parkinson’s disease, etc.); (2) participant has a predominance of stress incontinence in the opinion of the investigator, determined by participant history; (3) participant currently uses or plans to use medications or therapies to treat symptoms of OAB, including nocturia.
  • neurological reason e.g, spinal cord injury, multiple sclerosis, cerebrovascular accident, Alzheimer’s disease, Parkinson’s disease, etc.
  • participant has a predominance of stress incontinence in the opinion of the investigator, determined by participant history
  • participant currently uses or plans to use medications or therapies to treat symptoms of OAB, including nocturia.
  • participant previously receiving these medications must have discontinued their use to achieve sufficient washout prior to beginning recording of symptoms into the Screening Bladder Diary as follows: for desmopressin, at least one day prior; for anticholinergic therapy, at least 14 days prior; for intravesical anticholinergic therapy, at least 4 weeks prior; for P3 agonists, at least 14 days prior; (4) participants who have previously been treated with: o onabotulinumtoxinA for urological indications within 6 months of starting the Screening Bladder Diary (participants treated with onabotulinumtoxinA or other toxins for non-urological indications are eligible, regardless of when treated), any other toxin for urological indications, regardless of when treated , or intravesical pharmacologic agent (e.g, capsaicin, resiniferatoxin) within 12 months of beginning recording of symptoms into the Screening Bladder Diary; (5) participant currently uses CIC or indwelling catheter to manage their urinary incontinence; (6) participant has history or
  • Suspicious urine cytology abnormalities require that urothelial malignancy is ruled out to the satisfaction of the investigator according to local site practice; (13) participant is male with previous or current diagnosis of prostate cancer or a prostate specific antigen (PSA) level of > 10 ng/mL at screening. Patients with a PSA level of > 4 ng/mL but ⁇ 10 ng/mL must have prostate cancer ruled out to the satisfaction of the investigator according to local site practice; (14) participant has evidence of urethral and/or bladder outlet obstruction, in the opinion of the investigator at screening or Randomization/Day 1; (15) participant has a post-void residual (PVR) urine volume of > 100 mL at any time during Screening (before randomization).
  • PVR post-void residual
  • the PVR measurement can be repeated once on the same day; the participant is to be excluded if the repeated measure is above 100 mL; (16) participant has had urinary retention or an elevated PVR urine volume that has been treated with an intervention (such as catheterization), within 6 months of the Screening Visit (Visit 1).
  • transient voiding difficulties as a result of surgical procedures that resolved within 24 hours are not exclusionary; (17) participant has a 24-hour total volume of urine voided > 3000 mL, collected over 24 consecutive hours during the 3 -day Screening Bladder Diary collection period prior to Randomization/Day 1; (18) participant has a history of 3 or more urinary tract infections (UTIs) within 6 months of the Screening Visit (Visit 1) or is taking prophylactic antibiotics to prevent chronic UTIs. Participants with a current acute UTI during screening can be treated appropriately and are eligible; (19) participant has a serum creatinine level > 2 times the upper limit of normal at screening; (20) participant has current or previous uninvestigated hematuria.
  • UTIs urinary tract infections
  • Participants with investigated hematuria may enter the study if urological/renal pathology has been ruled out to the satisfaction of the investigator; (21) participant has hemophilia, or other clotting factor deficiencies, or disorders that cause bleeding diathesis; (22) participant cannot withhold any antiplatelet, anticoagulant therapy or medications with anticoagulant effects for 3 days prior to Randomization/Day 1.
  • some medications may need to be withheld for > 3 days, per clinical judgment of the investigator; (23) females who are pregnant, nursing or planning a pregnancy during the study, and either males, or females of childbearing potential who are unable or unwilling to use a reliable form of contraception during the study; (24) participant has a known allergy or sensitivity to URO-902, anesthetics, or antibiotics to be used during the study; (25) participant needs a walking aid on a permanent basis;
  • participant is currently participating in or has previously participated in another therapeutic study within 30 days of screening (or longer if local requirements specify). Participant has been previously treated with URO-902 in clinical trials (formerly known as AMaxi-K or pVAX/ASlo);
  • participant has a history or current evidence of any condition, therapy, laboratory abnormality or other circumstances that might, in the opinion of the investigator, confound the results of the study, interfere with the participant’s ability to comply with the study procedure, or make participation in the study not in the participant’s best interest.
  • URO-902 is a clear and colorless sterilized drug product solution supplied for intradetrusor injections.
  • URO-902 plasmid is dissolved in PBS-2%. The solution is filtered and filled into a sterilized vial and capped with a sterilized gray stopper. Each vial contains 10 mL at a concentration of 2.5 mg/mL and 5.0 mg/mL, respectively. This equates to 25 mg and 50 mg of URO-902 per vial.
  • URO-902 will be provided in vials in identical packaging to placebo. Each vial will contain 10 mL of study drug solution and will be labeled as required per regulatory requirement.
  • a flexible or rigid cystoscope will be used for administration of study treatment. Per local site practice lubricating gel will be used to insert the cystoscope.
  • the bladder will be instilled with a sufficient amount of saline to visualize the study injections.
  • One or two syringes of appropriate size to hold a minimum of 20 mL of study drug and one syringe of appropriate size to hold a minimum of 1 mL saline flushing solution will be prepared and ready for treatment administration (refer to Section 6.5 as needed).
  • the injection needle will be primed with approximately 0.5 mL of study drug.
  • the 20 mL of study drug should be administered as 20 injections, each approximately 1.0 mL.
  • injections Under direct visualization via cystoscopy, injections will be distributed evenly across the detrusor wall and spaced approximately 1 cm apart, avoiding the bladder dome and trigone.
  • the needle To administer study drug from the syringe, the needle should be inserted approximately 3 mm into the detrusor for each injection.
  • a sufficient amount of saline from the 1-mL syringe will be pushed through the injection needle to ensure delivery of the remaining amount of study drug.
  • Sample Size Determination The sample size for this study was calculated to be a total of 207 enrolled participants, i.e., 69 randomized and treated participants for each treatment arm (50 mg URO-902, 100 mg URO-902, and placebo). Assuming a 20% drop out rate, approximately 55 evaluable participants in each treatment arm will give enough power to assess the two co-primary endpoints.
  • ITT-E intent-to-treat-exposed
  • the ITT-E population will be used for demographics, baseline characteristics, and efficacy analyses; the ITT-E population will consist of all participants who were randomized and received study treatment.
  • the safety population will consist of all participants who received the study drug and will be used to assess treatment-emergent adverse events and other safety evaluations based on actual treatment received.
  • a separate safety population, safety (modified), will be created to assess safety excluding participants who received additional OAB medication at Week 24 or later.

Abstract

La présente divulgation concerne des méthodes de thérapie génique à action prolongée pour traiter une vessie hyperactive (OAB) et des symptômes de celle-ci, ou pour améliorer des symptômes de celle-ci, comprenant l'administration d'URO-902, une composition pharmaceutique comprenant un vecteur codant pour la sous-unité alpha du canal Maxi-K. L'administration d'une dose unique d'URO-902 par injection dans le muscle lisse de la vessie de patients souffrant d'une OAB provoquée par des réductions cliniquement pertinentes à partir de la ligne de base dans des mictions quotidiennes moyennes, des épisodes d'urgence, et des épisodes d'incontinence urinaire d'urgence qui commencent à la semaine 2 après l'injection, ont atteint un effet maximal à environ 12 semaines, et persistent pendant 48 semaines après l'injection de la dose unique d'URO-902.
PCT/IB2023/054387 2022-05-12 2023-04-27 Thérapie génique à long terme pour vessie hyperactive WO2023218278A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263341357P 2022-05-12 2022-05-12
US63/341,357 2022-05-12

Publications (1)

Publication Number Publication Date
WO2023218278A1 true WO2023218278A1 (fr) 2023-11-16

Family

ID=86497559

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/054387 WO2023218278A1 (fr) 2022-05-12 2023-04-27 Thérapie génique à long terme pour vessie hyperactive

Country Status (1)

Country Link
WO (1) WO2023218278A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080269159A1 (en) 2007-04-27 2008-10-30 Board Of Trustees Of The University Of Arkansas Compositions and methods for regulation of smooth muscle cells and blood pressure
WO2013151665A2 (fr) 2012-04-02 2013-10-10 modeRNA Therapeutics Polynucléotides modifiés destinés à la production de protéines associées à une maladie humaine
US20170136106A1 (en) 2015-11-17 2017-05-18 Allergan, Inc. Botulinum toxin administration for treatment of neurogenic detrusor overactivity associated urinary incontinence
US20170258878A1 (en) 2016-02-22 2017-09-14 Allergan, Inc. Bladder injeciton paradigm for administration of botulinum toxins
US20180126003A1 (en) 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
WO2018209351A1 (fr) 2017-05-12 2018-11-15 Ion Channel Innovations, Llc Compositions et méthodes de traitement du syndrome de la vessie hyperactive idiopathique et de l'hyperactivité du détrusor
WO2020100089A2 (fr) 2018-11-14 2020-05-22 Urovant Sciences Gmbh Compositions et méthodes pour le traitement d'un dysfonctionnement des muscles lisses

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080269159A1 (en) 2007-04-27 2008-10-30 Board Of Trustees Of The University Of Arkansas Compositions and methods for regulation of smooth muscle cells and blood pressure
WO2013151665A2 (fr) 2012-04-02 2013-10-10 modeRNA Therapeutics Polynucléotides modifiés destinés à la production de protéines associées à une maladie humaine
US20170136106A1 (en) 2015-11-17 2017-05-18 Allergan, Inc. Botulinum toxin administration for treatment of neurogenic detrusor overactivity associated urinary incontinence
US20170258878A1 (en) 2016-02-22 2017-09-14 Allergan, Inc. Bladder injeciton paradigm for administration of botulinum toxins
US20180126003A1 (en) 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
WO2018209351A1 (fr) 2017-05-12 2018-11-15 Ion Channel Innovations, Llc Compositions et méthodes de traitement du syndrome de la vessie hyperactive idiopathique et de l'hyperactivité du détrusor
US20200276333A1 (en) * 2017-05-12 2020-09-03 Ion Channel Innovations, Llc Compositions and methods for treating idiopathic overactive bladder syndrome and detrusor overactivity
WO2020100089A2 (fr) 2018-11-14 2020-05-22 Urovant Sciences Gmbh Compositions et méthodes pour le traitement d'un dysfonctionnement des muscles lisses
US20220133849A1 (en) * 2018-11-14 2022-05-05 Urovant Sciences Gmbh Compositions and methods for the treatment of smooth muscle dysfunction

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
"Genome Analysis: A Laboratory Manual", 2007
"Pharmaceutical Salts: Properties, Selection, and Use", 2008, WILEY-VCH
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
"The Dictionary of Cell and Molecular Biology", 1999, ACADEMIC PRESS
ABRAMS P ET AL., UROLOGY, vol. 61, no. 28-37, 2003, pages 37 - 49
BERGE ET AL., JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19
CHAPPLE ET AL.: "Terminology report from the International Continence Society (ICS) Working Group on Underactive Bladder (UAB", NEUROLOGY AND URODYNAMICS, vol. 37, 2018, pages 2928 - 2931
GAIT: "Oligonucleotide Synthesis: A Practical Approach", 1984, IRL PRESS
MOUNT, BIOINFORMATICS: SEQUENCE AND GENOME ANALYSIS, 2004
NELSONCOXLEHNINGER: "Oxford Dictionary Of Biochemistry And Molecular Biology", 2000, OXFORD UNIVERSITY PRESS
RUI ZHANG ET AL: "OnabotulinumtoxinA for neurogenic detrusor overactivity and dose differences: a systematic review", INTERNATIONAL BRAZ J UROL, vol. 41, no. 2, 1 April 2015 (2015-04-01), pages 207 - 219, XP055339454, DOI: 10.1590/S1677-5538.IBJU.2015.02.05 *
SAMBROOKRUSSELL: "Condensed Protocols from Molecular Cloning: A Laboratory Manual", 2006
SAMBROOKRUSSELL: "The Concise Dictionary of Biomedicine and Molecular Biology", 2002, COLD SPRING HARBOR LABORATORY PRESS
STRYER, L.: "Biochemistry", 1995, W.H. FREEMAN

Similar Documents

Publication Publication Date Title
JP6579629B2 (ja) 筋障害を相殺するための手段と方法
AU2013246034B2 (en) Compositions and methods for inhibiting the activity of LAR family phosphatases
TW201521718A (zh) 低頻率格拉替雷(glatiramer)醋酸鹽之治療
US20190046662A1 (en) Compositions and Methods for Treating Neuropathic Pain
US20230293725A1 (en) Compositions and methods for treating idiopathic overactive bladder syndrome and detrusor overactivity
CA3093844A1 (fr) Micro-arn et obesite
US11077110B2 (en) Compositions and methods for treating and preventing metabolic disorders
US10898550B2 (en) Compositions and methods of treating root avulsion injury
WO2023218278A1 (fr) Thérapie génique à long terme pour vessie hyperactive
US20220133849A1 (en) Compositions and methods for the treatment of smooth muscle dysfunction
TW202408594A (zh) 用於膀胱過動之長期基因療法
US11389507B2 (en) Use of interleukin-4-inducing principle of Schistosoma mansoni eggs for treating pain, interstitial cystitis and/or overactive bladder
JP2017521408A (ja) 無毒性破傷風毒素cフラグメント(ttc)を使用して筋量を増加させる方法
CN112142856B (zh) 一种用于改善脑卒中兴奋性毒性损伤的特异性降解ndrg2靶向肽及其应用
EP1855665B1 (fr) Utilisation de derives de phenylacetyle pour la production d'un medicament employe dans le traitement de la maladie de von hippel-lindau (vhl)
WO2023023189A2 (fr) Surexpression de la clustérine dans la maladie d'alzheimer
CN117925817A (en) Application of PPIC gene in preparing medicine for preventing and treating idiopathic pulmonary fibrosis
CA3218054A1 (fr) Produits et methodes pour induire un saut d'exon 2 du gene dmd dans le traitement de la dystrophie musculaire

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: 23725807

Country of ref document: EP

Kind code of ref document: A1