EP3558337A1 - Procédés de traitement du syndrome du côlon irritable avec des formulations à libération modifiée ou retardée de linaclotide - Google Patents

Procédés de traitement du syndrome du côlon irritable avec des formulations à libération modifiée ou retardée de linaclotide

Info

Publication number
EP3558337A1
EP3558337A1 EP17833050.2A EP17833050A EP3558337A1 EP 3558337 A1 EP3558337 A1 EP 3558337A1 EP 17833050 A EP17833050 A EP 17833050A EP 3558337 A1 EP3558337 A1 EP 3558337A1
Authority
EP
European Patent Office
Prior art keywords
linaclotide
week
baseline
pain
composition
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
EP17833050.2A
Other languages
German (de)
English (en)
Inventor
Ahmad Hashash
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ironwood Pharmaceuticals Inc
Original Assignee
Ironwood Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ironwood Pharmaceuticals Inc filed Critical Ironwood Pharmaceuticals Inc
Publication of EP3558337A1 publication Critical patent/EP3558337A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives

Definitions

  • the invention relates to the use of delayed release pharmaceutical compositions comprising linaclotide to treat a variety of indications, including gastrointestinal (GI) disorders, such as irritable bowel syndrome with constipation (IBS-c) and symptoms associated with GI or non-GI disorders, such as abdominal pain.
  • GI gastrointestinal
  • IBS-c irritable bowel syndrome with constipation
  • abdominal pain symptoms associated with GI or non-GI disorders, such as abdominal pain.
  • IBS irritable bowel syndrome
  • CIC chronic idiopathic constipation
  • IBS-c irritable bowel syndrome with constipation
  • Rome III Diagnostic Criteria for irritable bowel syndrome includes recurrent abdominal pain or discomfort more than 3 days per month over the previous 3 months before the diagnosis, with the onset at least 6 months before the diagnosis, and associated with two or more of the following:
  • c Onset of symptoms is associated with a change in the form or appearance of stool.
  • Rome ⁇ Diagnostic Criteria further defines irritable bowel syndrome with constipation (IBS-c) in a patient as having hard or lumpy stool (Bristol Stool Form Scale 1-2) with at least 25% of BMs and loose or watery stool (Bristol Stool Form Scale 6-7) with less than 25% of BMs.
  • Patients with IBS-c may also report symptoms that include (i) alternation between constipation and normal stools, and (ii) lower abdominal cramping, aching or discomfort that is commonly triggered by eating.
  • U.S. Patents 7,304,036 and 7,371,727 disclose peptides that act as agonists of the guanylate cyclase C (GC-C) receptor for the treatment of gastrointestinal (GI) disorders.
  • GC-C guanylate cyclase C
  • One particular peptide disclosed is linaclotide, which consists of the following amino acid sequence: Cys Cys Glu Tyr Cys Cys Asn Pro Ala Cys Thr Gly Cys Tyr. Linaclotide has the chemical structure of:
  • Linaclotide is orally administered and has been approved in the U.S. by the FDA for the treatment of irritable bowel syndrome with constipation (IBS-c) and chronic idiopathic constipation (CIC).
  • IBS-c irritable bowel syndrome with constipation
  • CIC chronic idiopathic constipation
  • linaclotide has been shown to affect GI physiology including reducing visceral pain, reducing bloating and accelerating GI transit which can lead to increased stool frequency and improved stool consistency.
  • Orally administered linaclotide acts locally by binding to and activating GC-C receptors at the luminal surface of the intestine.
  • the GC-C receptor is a key regulator in mammals of intestinal function and is found throughout the luminal surface of the GI tract.
  • the GC-C receptor responds to the endogenous hormones, guanylin and uroguanylin, and to enteric bacterial peptides from the heat stable enterotoxin family (ST peptide).
  • ST peptide enterotoxin family
  • linaclotide As approved by the FDA, linaclotide is administered in an oral, solid, immediate-release capsule formulation manufactured by filling drug-layered beads into gelatin capsules. Due to the high expression of GC-C receptors throughout GI tract, linaclotide from an immediate release formulation activates the GC-C receptor starting from the upper GI tract, resulting in significant amount of intestinal fluid being brought to the lower GI tract. To reduce or mitigate this effect, delayed release compositions are needed which have targeted release of linaclotide in the distal or lower segment of the gastrointestinal tract. Targeting the lower GI for linaclotide release may help avoid excess fluid secretion but at the same time maintain or improve linaclotide efficacy for treating abdominal and bowel symptoms of GI disorders.
  • compositions for pharmaceutically active agents including the use of enteric coatings or pH responsive polymers.
  • specific components of these compositions vary greatly and depend significantly on the particular pharmaceutically active agent and the desired properties.
  • the formulation must be compatible with the pharmaceutically active agent and also provide the necessary dissolution performance and stability properties.
  • Linaclotide has been previously demonstrated to reduce visceral pain in the GI tract which is thought to be mediated by increasing cGMP. Animal studies have shown that orally administered linaclotide can treat colonic hypersensitivity and hyperalgesia. However, due to the reducing environment of the intestinal tract, it is known that much of the oral linaclotide dose is degraded prior to reaching the distal colon. In human volunteers treated with linaclotide, only about 5% of the oral dose is found in feces.
  • Delayed release (“DR") compositions of linaclotide that target the lower GI may improve linaclotide's efficacy towards relieving pain associated with various GI disorders by allowing for delivery of a higher dose of linaclotide to the colon.
  • Such DR compositions of linaclotide would have the potential to release linaclotide predominantly (or fully) in the lower GI.
  • the DR formulation or composition may have an increased capacity to treat lower GI associated disorders.
  • orally administered linaclotide has also been demonstrated to reduce visceral pain in non-GI tissues, providing further evidence that the mechanism of visceral pain relief via linaclotide is not mediated solely by promoting secretion.
  • a cGMP modulator whose distribution is limited to the GI can relieve pain can be used as a therapy to relieve pain in other parts of the body.
  • linaclotide in order for linaclotide to be a useful therapy for the treatment of visceral pain in non-GI tissues (e.g., ulcerative colitis, diverticulitis, IBS, overactive bladder syndrome, bladder hypersensitivity or colitis induced bladder afferent hyperactivity, etc.) for non-constipated patients, it would be necessary to reduce or eliminate the secretion-promoting effects of linaclotide.
  • a means of at least partially, or completely separating the effect of linaclotide in promoting secretion from that of relieving visceral pain may have a capacity to cause lower incidences of adverse events (such as diarrhea) than the immediate-release dosage form, e.g., because it would cause lower overall intestinal fluid secretion by not activating GC-C receptors in the upper GI. This would occur while maintaining or even improving linaclotide efficacy for treating symptoms of GI disorders, such as pain.
  • the invention relates to a method of treating disorders, such as gastrointestinal (GI) disorders (e.g., IBS-c) or symptoms associated with GI or non-GI disorders (e.g., abdominal pain).
  • GI gastrointestinal
  • IBS-c gastrointestinal
  • non-GI disorders e.g., abdominal pain
  • One aspect of the invention is a method of reducing the incidence or severity of an adverse event associated with administration of linaclotide, comprising orally administering to a subject a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet comprises an enteric coating comprising a pH-sensitive polymer that releases linaclotide in a lower GI of the subject when compared to administration of an equal dose of an immediate release dosage form of linaclotide, and the delayed-release pharmaceutical tablet composition further comprises a therapeutically effective amount of linaclotide to treat constipation and/or pain in the subject.
  • Another aspect of the invention is a method of reducing the incidence or severity of an adverse event associated with administration of linaclotide in a subject, comprising orally administering a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet comprises an enteric coating comprising a pH-sensitive polymer that releases a therapeutically effective amount of linaclotide to treat constipation and/or pain in the subject.
  • Another aspect of the invention is a method of reducing intestinal fluid secretion-promoting effects of linaclotide, comprising orally administering to a subject a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet further comprises an enteric coating comprising a pH-sensitive polymer that releases linaclotide in a lower GI of the subject.
  • Yet another aspect of the invention is a method of treating visceral or abdominal pain in a non-constipated subject, comprising orally administering a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet further comprises an enteric coating and a pH sensitive polymer that releases an amount of linaclotide in a stomach of the subject that is less than an amount effective to induce secretion in the subject.
  • Another aspect of the invention is a method of treating irritable bowel syndrome with constipation (IBS-c) comprising orally administering to a patient in need thereof, a delayed release pharmaceutical tablet composition comprising a therapeutically effective amount of linaclotide.
  • IBS-c irritable bowel syndrome with constipation
  • Still another aspect of the invention is a method of treating abdominal pain comprising orally administering to a patient in need thereof, a delayed release pharmaceutical tablet composition comprising a therapeutically effective amount of linaclotide.
  • Figure 1 is an illustration of three distinct periods of the trial described in Example 18.
  • Figure 2 is a plot of the primary efficacy parameter, 6/12 week APC +1 responder, for the eight treatment groups as described in Example 20.
  • Figures 3A and 3B are a plot of the secondary efficacy parameter, weekly SBM frequency over baseline, for the eight treatment groups as described in Example 20.
  • Figure 4 is a plot of the primary efficacy parameter, weekly abdominal pain change from baseline, for the eight treatment groups as described in Example 20.
  • Figure 5 is a plot of the secondary efficacy parameter, 12-week CSBM frequency over baseline, for the eight treatment groups as described in Example 20.
  • Figures 6A and 6B are a plot of the secondary efficacy parameters, 6/12 and
  • Figures 7A and 7B are a plot of the secondary efficacy parameters, 6/12 and
  • Figure 8 is a plot of the secondary efficacy parameter, 12-week abdominal pain change from baseline, for the eight treatment groups as described in Example 20.
  • Figure 9 is a plot of the secondary efficacy parameter, 12-week SBM frequency over baseline, for the eight treatment groups as described in Example 20.
  • Figure 10 is a plot of the secondary efficacy parameter, weekly SBM frequency over baseline, for the eight treatment groups as described in Example 20.
  • Figure 11 is a plot of the secondary efficacy parameter, 12-week stool consistency (BSFS) change from baseline, for the eight treatment groups as described in Example 20.
  • BSFS 12-week stool consistency
  • Figure 12 is a plot of the secondary efficacy parameters, 12- week abdominal symptom score change over baseline, 12-week abdominal pain change over baseline, 12 week abdominal discomfort change over baseline, and 12-week abdominal bloating change over baseline, for the eight treatment groups as described in Example 20.
  • Figure 13 is a plot of the secondary efficacy parameter, treatment satisfaction, for the eight treatment groups as described in Example 20.
  • Figure 14 is a plot of overall diarrhea TEAE rates for the eight treatment groups as described in Example 20.
  • Figure 15 is a plot of the overall diarrhea and diarrhea leading to drop out (ADO) rates for the eight treatment groups as described in Example 20.
  • Figure 16 is a plot of diarrhea TEAEs by severity for the eight treatment groups as described in Example 20.
  • Figure 17 is a plot of certain efficacy parameters compared with data from the Phase III clinical trial for Linzess®.
  • Figure 18 is a plot of the diarrhea TEAE rates compared with data from the Phase III clinical trial for Linzess®.
  • Figure 1 is a plot of the diarrhea TEAEs by severity compared with data from the Phase III clinical trial for Linzess®.
  • Figure 20A shows the release profile at various pHs for delayed release compositions comprising 100 ⁇ g linaclotide.
  • Figure 20 B shows the effect of altering the ratios of pH-sensitive polymers on release profiles.
  • Figure 21 is a plot of the percent change from baseline in weekly abdominal pain at week 12 for the D 1 composition, the IR composition and placebo.
  • a delayed release composition comprising linaclotide which are used to treat any number of diseases, disorders or symptoms involving constipation and/or pain (e.g., visceral pain, abdominal pain).
  • the methods of treatment comprising orally administering a delayed release pharmaceutical tablet composition comprising linaclotide, as described herein are used to treat irritable bowel syndrome with constipation (IBS-c) in a patient in need thereof.
  • IBS-c irritable bowel syndrome with constipation
  • the patient may be diagnosed with IBS-c according to the Rome Criteria (e.g. Rome III).
  • the methods of treatment comprising orally administering a delayed release pharmaceutical tablet composition comprising linaclotide, as described herein are used to treat abdominal pain in a patient in need thereof.
  • One aspect of the invention is a method of reducing the incidence or severity of an adverse event associated with administration of linaclotide, comprising orally administering to a subject a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet comprises an enteric coating comprising a pH-sensitive polymer that releases linaclotide in a lower GI of the subject when compared to administration of an equal dose of an immediate release dosage form of linaclotide, and the delayed-release pharmaceutical tablet composition further comprises a therapeutically effective amount of linaclotide to treat constipation and/or pain in the subject.
  • Another aspect of the invention is a method of reducing the incidence or severity of an adverse event associated with administration of linaclotide in a subject, comprising orally adrninistering a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet comprises an enteric coating comprising a pH-sensitive polymer that releases a therapeutically effective amount of linaclotide to treat constipation and/or pain in the subject.
  • the reduction in the incidence or severity of the adverse event is a reduction in the incidence or severity of the adverse event compared to administration of an equal dose of an immediate release dosage form of linaclotide.
  • the adverse event is diarrhea.
  • the incidence of the adverse event is reduced.
  • the severity of the adverse event is reduced.
  • Another aspect of the invention is a method of reducing intestinal fluid secretion-promoting effects of linaclotide, comprising orally administering to a subject a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet further comprises an enteric coating comprising a pH-sensitive polymer that releases linaclotide in a lower GI of the subject.
  • Yet another aspect of the invention is a method of treating visceral or abdominal pain in a non-constipated subject, comprising orally administering a delayed-release pharmaceutical tablet composition comprising linaclotide, wherein the tablet further comprises an enteric coating and a pH sensitive polymer that releases an amount of linaclotide in a stomach of the subject that is less than an amount effective to induce secretion in the subject.
  • Another aspect of the invention is a method of treating irritable bowel syndrome with constipation (IBS-c) comprising orally administering to a patient in need thereof, a delayed release pharmaceutical tablet composition comprising a therapeutically effective amount of linaclotide.
  • Still another aspect of the invention is a method of treating abdominal pain comprising orally administering to a patient in need thereof, a delayed release pharmaceutical tablet composition comprising a therapeutically effective amount of linaclotide.
  • the delayed-release pharmaceutical tablet composition comprises a therapeutically effective amount of linaclotide to reduce, prevent or relieve pain or constipation in the subject. In some embodiments, the delayed-release pharmaceutical composition comprises a therapeutically effective amount of linaclotide to reduce, prevent or relieve pain in the subject, but does not affect bowel habit. In some embodiments, the delayed- release pharmaceutical composition provides less than an amount of linaclotide effective to substantially affect bowel habit. In some embodiments, the bowel habit is selected from CSBM rate, SBM rate, or stool consistency.
  • the subject is diagnosed with irritable bowel syndrome with constipation (IBS-c).
  • the delayed-release pharmaceutical tablet composition comprising linaclotide releases less than 50%, less than 40%, less than 25% or less than 10% of the linaclotide in the stomach.
  • the delayed release pharmaceutical tablet composition is administered once daily. In some embodiments, the delayed release pharmaceutical tablet composition is administered once daily in the morning. In some embodiments, the delayed release pharmaceutical tablet composition is administered once daily in the morning at least 30 minutes before breakfast. In some embodiments, the delayed release pharmaceutical tablet composition is administered after the patient has fasted for at least 2 hours. In some embodiments, the delayed release pharmaceutical tablet composition is administered for at least 12 weeks.
  • the acLministering increases the number of complete spontaneous bowel movements (CSBMs) by the patient. In some embodiments, the administering decreases abdominal pain in the patient. In some embodiments, the administering increases the number of complete spontaneous bowel movements (CSBMs) by the patient from baseline by at least one per week during at least 6 out of 12 weeks. In some embodiments, the administering decreases the abdominal pain in the patient from baseline by at least 30% during at least 6 out of 12 weeks.
  • CSBMs complete spontaneous bowel movements
  • the administering improves one or more (e.g., two or more) of the following: abdominal pain, CSBM frequency rate, SBM frequency rate, stool consistency, straining, abdominal discomfort, abdominal bloating, abdominal symptom score, constipation severity, IBS symptom severity, days with use of per-protocol rescue medicine, treatment satisfaction, and assessment of adequate relief.
  • the administering results in improvement in one or more (e.g., two or more) of the following efficacy parameters: weekly change from baseline in abdominal pain, weekly change from baseline in CSBM frequency, 6/12 week APC+1 responder, 6/12 week CSBM +1 responder, 9/12 week CSBM +1 responder, 6/12 week abdominal pain responder, 9/12 week abdominal pain responder, weekly CSBM +1 responder, weekly abdominal pain responder, weekly APC +1 responder, change from baseline in 12- week abdominal pain, change from baseline in 12-week CSBM frequency rate, change from baseline in 12-week SBM frequency rate, change from baseline in 12-week stool consistency, change from baseline in 12-week straining, change from baseline in 12-week abdominal discomfort, change from baseline in 12-week abdominal bloating, change from baseline in 12- week abdominal symptom score, change from baseline in 12-week constipation severity, change from baseline in 12-week IBS symptom severity, change from baseline in 12-week percent of days with use of per-protocol rescue medicine, treatment satisfaction, and assessment of
  • the administering improves, as compared to treatment with an immediate release composition of linaclotide, one or more of the following: abdominal pain, CSBM frequency rate, SBM frequency rate, stool consistency, straining, abdominal discomfort, abdominal bloating, abdominal symptom score, constipation severity, IBS symptom severity, days with use of per-protocol rescue medicine, treatment satisfaction, and assessment of adequate relief.
  • the administering results in improvement, as compared to treatment with an immediate release composition of linaclotide, in one or more of the following efficacy parameters: weekly change from baseline in abdominal pain, weekly change from baseline in CSBM frequency, 6/12 week AP+1 responder, 6/12 week CSBM +1 responder, 9/12 week CSBM +1 responder, 6/12 week abdominal pain responder, 9/12 week abdominal pain responder, weekly CSBM +1 responder, weekly abdominal pain responder, weekly APC +1 responder, change from baseline in 12-week abdominal pain, change from baseline in 12-week CSBM frequency rate, change from baseline in 12-week SBM frequency rate, change from baseline in 12-week stool consistency, change from baseline in 12-week straining, change from baseline in 12-week abdominal discomfort, change from baseline in 2- week abdominal bloating, change from baseline in 12-week abdominal symptom score, change from baseline in 12-week constipation severity, change from baseline in 12-week IBS symptom severity, change from baseline in 12-week percent of days with use of per-protocol rescue
  • the administering results in a decrease in the risk of adverse events in the patient compared to treatment with an immediate release composition of linaclotide. In some embodiments, the administering results in a decrease in the risk of diarrhea in the patient compared to treatment with an immediate release composition of linaclotide In some embodiments, the administering results in a decrease of at least 20% in the risk of diarrhea in the patient compared to treatment with an immediate release composition of linaclotide In some embodiments, the administering results in a decrease in the risk of severe diarrhea in the patient compared to treatment with an immediate release composition of linaclotide.
  • Another aspect of the invention is a method of treating or relieving pain comprising administering to a patient in need thereof, a therapeutically effective amount of a delayed-release pharmaceutical tablet composition as described herein.
  • the pain is selected from visceral pain; diverticulitis pain; pelvic pain; abdominal pain; or pain associated with gastrointestinal disorders, venereal diseases, bladder pain syndrome, or interstitial cystitis.
  • the pain is selected from general abdominal pain, diverticular disease, pain associated with irritable bowel syndrome (IBS), chronic or acute radiation proctopathy (also referred to as radiation proctitis), rectal pain, chronic proctalgia, proctalgia fugax, anal pain, chronic anal fissure, post-operative anal pain, overactive bladder syndrome, stress incontinence, interstitial cystitis, bladder pain syndrome, pain associated with cancer, pain associated with gastrointestinal tract neoplasms, general pelvic pain, endometriosis, orchialgia, chronic prostatitis, prostatodynia, vulvodynia, urethral syndrome, penile pain, perianal pain, and pain associated with ulcerative colitis, ulcerative
  • the method of treating a patient includes administering a delayed release composition comprising a therapeutically effective amount of linaclotide once a day.
  • the composition is administered once a day in the morning.
  • the composition is administered once a day at least 30 minutes before ingestion of food. For example, once a day in the morning at least 30 minutes before breakfast.
  • the composition is administered after the patient has fasted, e.g., after the patient has fasted for at least 2 hours, for at least 4 hours, for at least 8 hours, or for at least 10 hours.
  • the composition is administered for a period of greater than four weeks, (e.g., at least 8 weeks, at least 12 weeks, or at least 26 weeks).
  • the linaclotide is administered each day of the week, at least once a week, at least twice a week, at least three times a week, at least four times a week, at least five times a week or at least six times a week.
  • the method of treating a patient includes orally administering a delayed release composition comprising a therapeutically effective amount of linaclotide, wherein the administering increases the frequency of complete spontaneous bowel movements (CSBMs) by the patient.
  • the CSBM frequency rate is increased to three or more CSBMs per week.
  • the CSBM frequency rate is increased by one or more CSBMs per week compared to a baseline level of CSBMs prior to treatment with delayed release compositions of linaclotide.
  • the frequency of CSBMs by the patient is increased compared to treatment with immediate release compositions of linaclotide.
  • the administering increases the spontaneous bowel movement (SBM) frequency rate for the patient.
  • the frequency of SBMs is increased compared to treatment with immediate release compositions of linaclotide.
  • the method of treating a patient includes administering a delayed release composition comprising a therapeutically effective amount of linaclotide, wherein the administering decreases abdominal pain in the patient.
  • the abdominal pain is decreased by at least 30% (e.g., at least 40%, at least 50%) compared to a baseline level of abdominal pain prior to treatment with delayed release compositions of linaclotide.
  • abdominal pain in the patient is decreased compared to treatment with immediate release compositions of linaclotide.
  • the abdominal pain is decreased by at least at least 30% (e.g., at least 40%, at least 50%) at week 12, after 12 weeks of administration.
  • the method of treating a patient includes administering a delayed release composition comprising a therapeutically effective amount of linaclotide, wherein the administering increases the frequency of complete spontaneous bowel movements (CSBMs) by the patient and decreases abdominal pain in the patient.
  • CSBMs complete spontaneous bowel movements
  • the method of treating a patient includes administering a delayed release composition comprising a therapeutically effective amount of linaclotide, wherein the adininistering improves abdominal symptoms (e.g., pain, discomfort, bloating) and/or bowel symptoms (e.g., CSBMs/per week, SBMs/per week, stool consistency, straining) in a patient with greater than moderate abdominal pain, in a patient with moderate to severe abdominal pain, or in a patient with severe to very severe abdominal pain.
  • abdominal symptoms e.g., pain, discomfort, bloating
  • bowel symptoms e.g., CSBMs/per week, SBMs/per week, stool consistency, straining
  • administration of delayed release compositions comprising linaclotide as described herein may provide one or more of the following advantages: an increase in the number of CSBMs, a decrease in abdominal pain or discomfort, a decrease in bloating, a decrease in constipation severity, an improvement in stool consistency, an increase in SBM frequency, an increase in BM frequency, a decrease in straining during defecation, an improvement in abdominal symptom score, an improvement in IBS symptom severity, a decrease in use of per-protocol rescue medicine, and an improvement is patient assessment of quality of life, treatment satisfaction or adequate relief.
  • the improvement may be an improvement as compared to treatment with immediate release composition of linaclotide.
  • the method of treating a patient includes treating a disorder selected from irritable bowel syndrome (IBS), constipation, irritable bowel syndrome with constipation (IBS-c), irritable bowel syndrome with diarrhea (IBS-d), mixed IBS (IBS- m), un-subtyped IBS (IBS-u), chronic idiopathic constipation (CIC), colon cancer, diverticulitis, ulcerative colitis, a functional gastrointestinal disorder, gastroesophageal reflux disease, functional heartburn, dyspepsia, visceral pain, abdominal pain, gastroparesis, chronic intestinal pseudo-obstruction, colonic pseudo-obstruction, Crohn's disease, inflammatory bowel disease, overactive bladder syndrome, bladder hypersensitivity or colitis induced bladder afferent hyperactivity in a patient in need thereof.
  • IBS irritable bowel syndrome
  • IBS-c irritable bowel syndrome with constipation
  • IBS-d mixed IBS
  • IBS-u un-subtyped IBS
  • the method of treating a patient includes treating a symptom associated with a disorder, such as a GI disorder, or alternatively a non-GI disorder in a patient in need thereof.
  • a disorder such as a GI disorder
  • the treatment may be for abdominal pain, discomfort or bloating, or visceral pain associated with a disorder (GI or non-GI).
  • the patient may be a non-constipated patient.
  • the disorder or symptom being treated is selected from visceral pain; diverticulitis pain; pelvic pain; abdominal pain; or pain associated with gastrointestinal disorders, venereal diseases, bladder pain syndrome, or interstitial cystitis.
  • the disorder or symptom being treated is pain selected from general abdominal pain, diverticular disease, pain associated with irritable bowel syndrome (IBS), chronic or acute radiation proctopathy (also referred to as radiation proctitis), rectal pain, chronic proctalgia, proctalgia fugax, anal pain, chronic anal fissure, post-operative anal pain, overactive bladder syndrome, stress incontinence, interstitial cystitis, bladder pain syndrome, pain associated with cancer, pain associated with gastrointestinal tract neoplasms, general pelvic pain, endometriosis, orchialgia, chronic prostatitis, prostatodynia, vulvodynia, urethral syndrome, penile pain, perianal pain,
  • IBS irritable bowel syndrome
  • the disorder or symptom being treated is a disorder or symptom related to constipation, such as chronic constipation, opioid induced constipation, post-surgical constipation (post-operative ileus), and constipation associated with neuropathic disorders or a combination of symptoms thereof (such as a combination of irritable bowel syndrome and chronic constipation), constipation associated with neuropathic disorders (e.g., constipation associated with Parkinson's Disease), constipation associated with cystic fibrosis or thyroid disease.
  • constipation such as chronic constipation, opioid induced constipation, post-surgical constipation (post-operative ileus), and constipation associated with neuropathic disorders or a combination of symptoms thereof (such as a combination of irritable bowel syndrome and chronic constipation), constipation associated with neuropathic disorders (e.g., constipation associated with Parkinson's Disease), constipation associated with cystic fibrosis or thyroid disease.
  • the disorder or symptom being treated is a disorder or symptom associated with the lower GI (e.g., a lower GI disorder).
  • the methods of treatment described herein are useful for the treatment of diseases or symptoms associated with visceral pain selected from the group consisting of general abdominal pain, diverticular disease, pain associated with irritable bowel syndrome (IBS), chronic or acute radiation proctopathy (also referred to as radiation proctitis), rectal pain, chronic proctalgia, proctalgia fugax, anal pain, chronic anal fissure, post-operative anal pain, overactive bladder syndrome, stress incontinence, interstitial cystitis, bladder pain syndrome, pain associated with cancer, pain associated with gastrointestinal tract neoplasms, general pelvic pain, endometriosis, orchialgia, chronic prostatitis, prostatodynia, vulvodynia, urethral syndrome, penile pain, perianal pain, ulcerative colitis, ulcerative proctitis, and Crohn's disease.
  • the compositions described herein are useful for the treatment of bladder pain syndrome
  • the methods of treatment described herein are useful for the treatment of interstitial cystitis.
  • the compositions described herein are useful for the treatment of endometriosis.
  • the compositions described herein are useful for the treatment of anal pain.
  • a method of treating a disorder comprising administering to a patient in need thereof, a therapeutically effective amount of the composition described herein.
  • the disorder is cancer selected from colorectal/local metastasized colorectal cancer, intestinal polyps, Barrett's esophagus, gastrointestinal tract cancer, lung cancer, cancer or pre-cancerous growths or metastatic growths of epithelial cells, polyps, breast, colorectal, lung, ovarian, pancreatic, prostatic, renal, stomach, bladder, liver, esophageal and testicular carcinoma.
  • methods are provided for preventing a cancer or hyperplasia of the gastrointestinal tract or preventing reocciurence of cancer or hyperplasia of the gastrointestinal tract in a patient in need thereof comprising administering an effective amount of the composition or the oral dosage form to the patient.
  • the cancer or hyperplasia is colorectal cancer, intestinal polyps or pre-cancerous growths or metastatic growths of gastrointestinal epithelial cells.
  • the composition or oral dosage form is administered simultaneously or sequentially with an effective amount of a COX-2 inhibitor.
  • Examples of highly selective and selective COX-2 inhibitors include etoricoxib, rofecoxib, lumiracoxib, valdecoxib, celecoxib (Celebrex®), sulindac, diclofenac, meloxicam and etodolac.
  • Non-selective NSAIDs that inhibit COX-2 include naproxen, ibuprofen, sodium salicylate and diflunisal.
  • the term “prevent” or “preventing” means to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease) or reoccurrence of cancer or hyperplasia, and/or reduce the risk of developing cancer or hyperplasia relative to a patient that has not been treated with a composition described herein.
  • methods are provided for treating gastrointestinal disorders in pediatric patients with the compositions and oral dosage forms described herein.
  • methods are provided for treating gastrointestinal disorders in a pediatric patient diagnosed with one or more gastrointestinal disorders or conditions, wherein the method comprises administering an effective amount of the composition or the oral dosage form to the patient.
  • compositions and oral dosage forms for treating gastrointestinal disorders including, but not limited to, GI motility disorders, irritable bowel syndrome, constipation-predominant irritable bowel syndrome (EBS-c), mixed-type irritable bowel syndrome (IBS-m), chronic constipation, chronic idiopathic constipation, opioid induced constipation, post-surgical constipation (postoperative ileus), constipation associated with neuropathic disorders, constipation associated with cystic fibrosis or thyroid disease, dyspepsia (including functional dyspepsia or non-ulcer dyspepsia), gastroparesis, gastrointestinal motility disorders, functional gastrointestinal disorders, gastroesophageal reflux disease (GERD), inflammatory bowel disease, Crohn's disease, ulcerative colitis, functional heartburn, chronic intestinal pseudo-obstruction (or colonic pseudo-obstruction), visceral pain, abdominal pain, pelvic pain, anal fissure pain, pain associated
  • methods are provided to treat IBS-c, EBS-m or chronic constipation (e.g., chronic idiopathic constipation) in pediatric patients with the compositions and oral dosage forms described herein.
  • methods are provided to treat IBS-c in a pediatric patient in need thereof.
  • methods are provided to treat chronic idiopathic constipation in a pediatric patient in need thereof.
  • a method is for treating or relieving pain comprising administering to a patient in need thereof, a therapeutically effective amount of the composition described herein.
  • the pain is selected from visceral pain; abdominal pain; pelvic pain; or pain associated with gastrointestinal disorders, venereal diseases, bladder pain syndrome, diverticulitis pain, prostatitis, testicular pain, endometriosis, vulvodynia, rectal paiaor interstitial cystitis.
  • the pain is selected from pelvic pain, pain associated with proctitis, anal fissure pain, pain associated with vulvodynia, pain associated with endometriosis, pain associated with fibromyalgia, functional abdominal pain, interstitial cystitis pain, pain associated with venereal disease, diverticulitis, pain associated with diverticulitis, and pain associated with celiac sprue.
  • the effective dose range of linaclotide for adult humans is from 25 ⁇ g to 6 mg per day orally. In some embodiments, the dose range is 15 ⁇ g to 2 mg per day orally. In some embodiments, the dose range for adult humans is 15 ⁇ g to 1 mg per day orally (e.g., 15 ⁇ & 30 ⁇ & 50 ⁇ g, 72 ⁇ 3 ⁇ 4 100 ⁇ 145 ⁇ 3 ⁇ 4 150 ⁇ 200 ⁇ & 250 ⁇ 3 ⁇ 4 290 ⁇ & 300 ⁇ & 350 ⁇ & 400 ⁇ & 450 ⁇ & 500 ⁇ g, 550 ⁇ & 579 ⁇ & 600 ⁇ & 650 ⁇ & 700 ⁇ & 750 ⁇ 3 ⁇ 4 800 ⁇ g, 850 ⁇ g, 900 ⁇ g, 950 ⁇ g or 1 mg).
  • the dose range for adult humans is 30 g to 300 ⁇ g per day orally In some embodiments, the dose range is 100 ⁇ g to 600 ⁇ g per day orally. In some embodiments, the dose is 30 ⁇ & 100 ⁇ g, 150 ⁇ g, 200 ⁇ g, 300 ⁇ g, 400 ⁇ g, 500 ⁇ g or 600 ⁇ g linaclotide per day orally. In some embodiments, the dose is 50 ⁇ g linaclotide per day orally. In some embodiments, the dose is 100 ⁇ g linaclotide per day orally. In some embodiments, the dose is 145 ⁇ g linaclotide per day orally. In some embodiments, the dose is 200 ⁇ g linaclotide per day orally.
  • the dose is 290 ⁇ g linaclotide per day orally. In some embodiments, the dose is 300 ⁇ g linaclotide per day orally. In some embodiments, the dose is 500 g linaclotide per day orally. In some embodiments, the dose is 600 ⁇ g linaclotide per day orally.
  • the effective pediatric dose range of linaclotide is from
  • the effective pediatric dose range of linaclotide is from 0.05 ⁇ g to 100 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 90 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 50 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 25 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 10 ⁇ g per day orally.
  • the effective pediatric dose range of linaclotide is from 0.1 g to 5 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 1 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is from 0.1 ⁇ g to 0.5 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 0.1 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 0.15 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 0.25 ⁇ g per day orally.
  • the effective pediatric dose range of linaclotide is 0.5 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 3.5 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 15 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 36 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 45 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 60 ⁇ g per day orally. In some embodiments, the effective pediatric dose range of linaclotide is 90 ⁇ g per day orally. In some embodiments, the unit dosage form and daily dose are equivalent.
  • the unit dosage form is administered with food at any time of the day, without food at any time of the day, with food after an overnight fast (e.g., with breakfast).
  • the unit dosage form is administered once a day, twice a day or three times a day.
  • one, two or three unit dosage forms will contain the daily oral dose of linaclotide.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician. However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity.
  • compositions are administered as a monotherapy.
  • composition consists essentially of an effective amount of linaclotide.
  • the composition consists of an effective amount of linaclotide.
  • the compositions are directly administered to a patient, for example, in the form of delayed release tablet or delayed release capsule.
  • the compositions are dissolved, disintegrated and/or mixed on or within food or beverage prior to administration to patients (e.g., elderly or pediatric patients).
  • the composition is dissolved or disintegrated in a liquid, solution, or fluid optionally containing stabilizing agent(s), preservative(s), sweetener(s), or the like, etc. prior to administration to a patient (e.g., elderly or pediatric patient).
  • the composition is a multiple dose composition, i.e., containing two, three, five, seven, ten, fifteen, twenty, twenty-five, thirty, forty, fifty, sixty, seventy, eighty, ninety or more daily doses of linaclotide.
  • the compositions are administered as part of a combination therapy.
  • a composition may be used in combination with other drugs or therapies that are used in the treatment, prevention, suppression, and/or amelioration of the diseases or conditions for which compounds of the invention are useful.
  • the linaclotide can be co-administered or co-formulated with other medications.
  • the linaclotide composition can be co-administered with other medications used to treat gastrointestinal disorders including but not limited to acid suppressing agents such as Histamine-2 receptor agonists (H2As) and/or proton pump inhibitors (PPIs).
  • the linaclotide composition can be co-administered with other medications used to treat gastrointestinal disorders including 5-ASAs such as mesalamine.
  • Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the invention.
  • a pharmaceutical unit dosage form containing such other drugs in addition to the compound of the invention may be employed.
  • the pharmaceutical compositions of the present invention include those that also contain one or more other active components, in addition to a compound of invention.
  • Several methods can be used for evaluating the bioactivity of the linaclotide composition, including, but not limited to, immunoassays (e.g., enzyme-linked immunosorbent assay), radioimmuno assays, immunoradiometric assays, gel electrophoresis (e.g., SDS- PAGE), high performance liquid chromatography (HPLC), and/or high performance capillary electrophoresis (HPCE).
  • immunoassays e.g., enzyme-linked immunosorbent assay
  • radioimmuno assays e.g., radioimmuno assays
  • immunoradiometric assays e.g., gel electrophoresis (e.g., SDS- PAGE), high performance liquid chromatography (HPLC), and/or high performance capillary electrophoresis (HPCE).
  • HPLC high performance liquid chromatography
  • HPCE high performance capillary electrophoresis
  • the bioactivity of the composition is assessed by a method comprising fixing linaclotide, incubating linaclotide with guanylate cyclase C (GCC), incubating GCC bound linaclotide with antibodies against GCC, incubating GCC antibody-bound linaclotide with fluorescently labeled antibodies against GCC antibodies, and detecting the linaclotide bound to the GCC antibodies by measuring the fluorescence intensity using a plate reader. The drug concentration can then be calculated based on the fluorescence reading of the solution.
  • GCC guanylate cyclase C
  • the bioactivity of the linaclotide compositions can be assessed and quantified using the following method, though other methods are available.
  • the composition is added to a volumetric flask containing 60 ml of phosphate buffer having a pH of 4.5, and the flask is shaken for 60 minutes. 0.2 ml of the supernatant is then removed, and is added into one or more wells of a 96-well plate that is coated with GC-C receptors. The plate is sealed and incubated at 37°C for 2 nr. At the end of incubation, the sample is removed and the plate is washed with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the bound linaclotide is then incubated for 1 hour, at room temperature, with GC-C (such as is available from Sigma-Aldrich Inc.) labeled with fluorescein isocyanate (FITC) in blocking buffer. After incubation, the well is washed with PBS. The fluorescence intensity of the end product is detected, for example, by using a plate reader. The linaclotide concentration is then calculated based on the fluorescence reading of the solution.
  • GC-C such as is available from Sigma-Aldrich Inc.
  • Delayed release oral dosage forms of linaclotide (collectively, "DR") are provided herein.
  • the delayed release pharmaceutical compositions of the present invention relates to stable, solid, oral dosage forms of linaclotide which exhibit delayed release of linaclotide to the lower gastrointestinal tract.
  • the only approved formulation of linaclotide is a capsule that exhibits immediate release (“IR").
  • IR dosage forms release most or all of the linaclotide contained therein in the upper GI. This, in turn, causes GC-C receptor activation and fluid secretion in both the upper GI and to a lesser extent in the lower GI.
  • the difference between upper and lower GI activation and fluid secretion by the IR dosage form is due, in part, to the fact that linaclotide (once released from the dosage form) undergoes proteolytic digestion and loses some or all capacity to activate GC-C receptors, particularly by the time it reaches the lower GI (such as the ileum, terminal ileum, ileocecal valve, or colon).
  • the linaclotide is present in the composition in an amount between 30 ⁇ g to 1,000 ⁇ g.
  • the linaclotide is present in an amount of about 30 ⁇ g, about 100 ⁇ g, about 300 ⁇ g, about 500 ⁇ g, about 600 g, or about 1 ,000 ⁇ g or the linaclotide is present in an amount of 100 ⁇ & 300 ⁇ g, 500 ⁇ g, 600 ⁇ g, or 1 ,000
  • the composition further comprises between 0% - 2% per weight of an amino acid selected from the group consisting of alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine, or any mixture thereof.
  • the composition further comprises between 0% - 2% or between 0.5% - 1.5% per weight of histidine.
  • the composition further comprises between 0% - 3% per weight of a cation salt selected from the group consisting of calcium, potassium, magnesium, zinc, aluminum, manganese, chromium, cobalt, nickel, barium, and sodium, or any combination or mixture thereof.
  • the composition further comprises between 0% - 3% per weight of a calcium salt.
  • the composition further comprises between 0% - 2% or between 0.2% - 0.8% per weight of calcium chloride dehydrate.
  • the composition further comprises between 0% - 5%, between 1 % - 3%, or between 1 % - 1.88% per weight of polyvinyl alcohol (PVA).
  • PVA polyvinyl alcohol
  • the pH-sensitive polymer has a dissolution pH of at least 6.0, at least 6.5, or at least 7.0.
  • the the pH-sensitive polymer comprises methyl acrylate-methacrylic acid copolymers (e.g., Eudragit®).
  • the pH-sensitive polymer comprises Eudragit SI 00.
  • the pH-sensitive polymer comprises Eudragit LI 00.
  • the pH-sensitive polymer consists essentially of Eudragit SI 00.
  • the pH-sensitive polymer comprises a mixture of Eudragit SI 00 and Eudragit LI 00.
  • the pH-sensitive polymer comprises a mixture of Eudragit S100 and Eudragit L100 at a ratio of between 1 :1 and 6:1 (S100:L100), at a ratio of between 4.5:1 and 5.5:1 (S100:L100), or at a ratio of 4.875:1 (S100:L100) by weight.
  • the delayed release pharmaceutical tablet composition comprises an enteric coated tablet. In some embodiments, the delayed release pharmaceutical tablet composition comprises:
  • PVA polyvinyl alcohol
  • the composition further comprises a protective polymer film or subcoating.
  • the subcoating comprises Opadry II®.
  • the DR dosage forms described herein release most or all of the linaclotide contained therein within the lower GI, such as proximate to the ileocecal valve or within the colon (and less or no release in the stomach, duodenum and/or jejunum). Therefore, the inventive dosage forms have a capacity to achieve lower overall fluid secretion than IR dosage forms in the upper GI, while improving or still maintaining excellent efficacy for treating a disorder (e.g., a GI disorder such as IBS-c or symptoms associated with a disorder, such as abdominal pain). IBS patients report lower left quadrant abdominal pain as a symptom of their disorder, so it is believed that the pain of IBS originates from the colon.
  • a disorder e.g., a GI disorder such as IBS-c or symptoms associated with a disorder, such as abdominal pain.
  • the DR dosage forms are believed to be ideally suited for treating lower Gl-associated diseases and disorders. Because the DR dosage forms will not release any (or a small percentage) of its linaclotide in the stomach and upper GI (which can cause rapid digestion of the linaclotide in the intestine), some preferred embodiments of the DR dosage form will incorporate low doses of linaclotide (as compared to the amounts in the approved IR form) but will maintain the same efficacy levels as the IR in treating GI symptoms.
  • Disorders that are suitable for treatment with the delayed release compositions include irritable bowel syndrome (IBS), constipation, irritable bowel syndrome with constipation (IBS-c), irritable bowel syndrome with diarrhea (IBS-d), mixed IBS (IBS-m), un-subtyped IBS (IBS-u), chronic idiopathic constipation (CIC), diverticulitis, ulcerative colitis, a functional gastrointestinal disorder, gastroesophageal reflux disease, functional heartburn, dyspepsia, visceral pain, abdominal pain, gastroparesis, chronic intestinal pseudo-obstruction, colonic pseudo-obstruction, Crohn's disease, inflammatory bowel disease, overactive bladder syndrome, bladder hypersensitivity or colitis induced bladder afferent hyperactivity in a patient in need thereof.
  • Symptoms that are suitable for treatment with the delayed release compositions include abdominal pain, discomfort or bloating, or visceral pain, for example, in a non-constipated patient.
  • the guanylate cyclase C (GC-C) receptor is a transmembrane receptor that is located on the apical surface of epithelial cells in the stomach, intestine and lower GI.
  • the receptor has an extracellular ligand-binding domain, a single transmembrane region and a C-terminal guanylyl cyclase domain.
  • the intracellular catalytic domain catalyzes the production of cGMP from GTP.
  • cGMP is secreted bi-directionally from the epithelium into the submucosa and lumen. Normally, the pH of the GI tract gradually increases from stomach (pH 1.5-3) to terminal ileum (pH 7-8) before it drops in the colon to pH 5.5-7.0.
  • Linaclotide binds to the intestinal GC-C receptor which is a regulator of fluid and electrolyte balance in the intestine.
  • Linaclotide is a peptide that consists of the amino acid sequence Cysi Cys 2 Glu3 Tyr4 Cyss Cys& Asm Pros Ala9 Cysio Thru Glyi 2 Cysi3 Tyrn. Any desired form of linaclotide may be used in the composition, for example, any pharmaceutically acceptable salt or hydrate of the peptide, any isolated and/or purified form thereof, or any disulfide form thereof. Linaclotide has disulfide bonds between Cysi and Cyse, Cys 2 and Cysio, and Cyss and Cysn.
  • the DR composition comprises enteric-coated tablets comprising an immediate release tablet core and containing a unit dose of linaclotide that dissolves only under pH conditions of the distal segment of intestine.
  • the enteric or functional coating comprises a pH-sensitive polymer.
  • the enteric coating comprises a pH-sensitive polymer that has a dissolution profile of a pH of at least 6.0, for example, a pH of at least 6.2, a pH of at least 6.4, a pH of at least 6.5, a pH of at least 6.6, a pH of at least 6.8, a pH of at least 7.0, a pH of at least 7.2, a pH of at least 7.4, a pH of at least 7.6 or higher.
  • the pH-sensitive polymer is selected from methyl acrylate-methacrylic acid copolymers (e.g. Eudragit®); cellulose acetate succinate (CAS); hydroxy propyl methyl cellulose phthalate (HPMCP); PVA; PVP; PVP-LP, hydroxy propyl methyl cellulose acetate succinate (HPMCAS); polyvinyl acetate phthalate (PVAP); methyl methacrylate-methacrylic acid copolymers; sodium alginate and stearic acid; guar gum; and carbomers.
  • methyl acrylate-methacrylic acid copolymers e.g. Eudragit®
  • CAS cellulose acetate succinate
  • HPMCP hydroxy propyl methyl cellulose phthalate
  • PVA hydroxy propyl methyl cellulose phthalate
  • PVP-LP hydroxy propyl methyl cellulose acetate succinate
  • PVAP polyvinyl acetate phthalate
  • the pH-sensitive polymer comprises a mixture of Eudragit SI 00 and Eudragit LlOO at a ratio of between 1:1 and 6:1 (S100:L100) by weight. In another embodiment, the pH-sensitive polymer comprises a mixture of Eudragit SI 00 and Eudragit LlOO at a ratio of between 4.5:1 and 5.5:1 (S100:L100) by weight. In one particular embodiment, the pH-sensitive polymer comprises a mixture of Eudragit SI 00 and Eudragit LlOO at a ratio of 4.875:1 (S100:L100) by weight.
  • the enteric coating is at least 40 microns in average thickness, for example, at least 45 microns in average thickness, at least 50 microns in average thickness, at least 55 microns in average thickness, at least 60 microns in average thickness, at least 65 microns in average thickness, at least 70 microns in average thickness, at least 75 microns in average thickness, at least 80 microns in average thickness, at least 85 microns in average thickness, at least 90 microns in average thickness, at least 95 microns in average thickness, at least 100 microns in average thickness, at least 105 microns in average thickness, at least 110 microns in average thickness, at least 115 microns in average thickness, or at least 120 microns in average thickness.
  • the enteric coating has an average thickness of between 55 microns and 100 microns. In still another embodiment, the enteric coating has an average thickness of between 65 microns and 95 microns. In a particular embodiment, the enteric coating has an average thickness of about 75 microns and 85 microns.
  • the delayed release composition comprises at least 1.25% (w/w) of PVA, for example, at least 1.49% (w/w) of PVA.
  • the delayed release composition comprises at least 0.44% (w/w) of CaCb, for example, at least 0.71% (w/w) of CaCl 2 .
  • the delayed release composition comprises at least 0.93% (w/w) of histidine, for example, at least 1.49% (w/w) of histidine.
  • the delayed release compositions may include any effective amount of linaclotide.
  • the composition comprises from 0.05 ⁇ g to 6 mg of linaclotide.
  • the composition comprises from 1 ⁇ g to 2 mg of linaclotide.
  • the composition comprises from 25 ⁇ g to 2 mg of linaclotide, for example, from 50 ⁇ g to 1 mg of linaclotide.
  • the composition comprises from 0.1 ⁇ g to 90 ⁇ g of linaclotide.
  • the composition comprises from 0.1 ⁇ g to 45 g of linaclotide.
  • the composition comprises from 0.1 ⁇ g to 25 ⁇ g of linaclotide. In some embodiments, for example, the composition comprises from 30 ⁇ g to 300 ⁇ g of linaclotide. In some embodiments, the composition comprises 0.05 ⁇ g, 0.1 ⁇ g, 0.15 ⁇ g, 0.25 ⁇ 3 ⁇ 4 0.5 ⁇ g, 0.75 ⁇ & 1 ⁇ & 1.5 ⁇ 2 ⁇ g, 2.5 ug, 3 ⁇ & 3.5 ⁇ g, 4 ⁇ 4.5 ⁇ 5 ⁇ & 7.5 ⁇ 9 ⁇ & 10 ⁇ 3 ⁇ 4, 15 ⁇ & 20 ⁇ & 25 3 ⁇ 4 30 ⁇ 35 ⁇ & 36 ⁇ 40 ⁇ g, 45 ⁇ g, 50 ⁇ g, 60 ⁇ g, 72 ⁇ & 75 ⁇ g, 90 ⁇ & 100 ⁇ 3 ⁇ 4 145 g, 150 ⁇ , 200 ⁇ & 250 ⁇ & 290 ⁇ & 300 ⁇ & 350 ⁇ & 400 ⁇ &
  • the composition comprises from 100 ⁇ g to 600 ⁇ g of linaclotide. In some embodiments, the composition comprises 30 ⁇ g, 50 ⁇ g, 75 ⁇ g, 100 ⁇ g, 150 ⁇ g, 290 ⁇ g, 300 ⁇ g, 400 ⁇ g, 500 ⁇ g or 600 ⁇ g of linaclotide. In some embodiments, the composition comprises 9 ⁇ g, 10 ⁇ g, 15 ⁇ g, 36 ⁇ g, 72 ⁇ g, 75 ⁇ g, 145 ⁇ g, 290 ⁇ g, 579 ⁇ g, or 600 ⁇ g of linaclotide.
  • the composition comprises 30 ⁇ g of linaclotide. In some embodiments, the composition comprises 50 ⁇ g of linaclotide. In some embodiments, the composition comprises 100 ⁇ g of linaclotide. In some embodiments, the composition comprises 145 ⁇ g of linaclotide. In some embodiments, the composition comprises 300 ⁇ g of linaclotide. [000108] It has been found, in some embodiments, that the stability of delayed release compositions of linaclotide can be increased or improved by including in the compositions a suitable amount of a sterically hindered primary amine (e.g., amino acid) component, a cation
  • a sterically hindered primary amine e.g., amino acid
  • a suitable amount of a cation e.g., Mg 2+ , Ca 2+ , Zn 2+ ) in the composition increases the stability of the composition against oxidative degradation of linaclotide.
  • inclusion of a suitable amount of a sterically hindered primary amine for an example in the form of an amino acid (e.g., histidine) in the composition increases the stability of the composition against, for example, the nucleophilic addition of formaldehyde or a formaldehyde equivalent to the N-terminus of linaclotide, e.g. by acting as a scavenger, and/or by buffering the composition.
  • inclusion of both a sterically hindered primary amine for an example in the form of an amino acid (e.g., histidine) in the composition increases the stability of the composition against, for example, the nucleophilic addition of formaldehyde or a formaldehyde equivalent to the N-terminus of linaclotide, e.g. by acting as a scavenger, and/or by buffering the composition.
  • inclusion of both a sterically hindered primary amine for an example in the form of an amino acid (e.g., hist
  • a suitable amount of a polymer e.g., polyvinyl pyrrolidone or polyvinyl alcohol
  • inclusion of a suitable amount of a polymer increases the stability of the composition for example by decreasing the mobility and/or reactivity of linaclotide within the composition, e.g., by forming a complex or matrix
  • linaclotide for example, a glassy and/or rigid matrix
  • linaclotide e.g., by vitrification reaction
  • composition can comprise any stabilizing amount of a sterically hindered primary amine component.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 400: 1 and 1 :1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 200:1 and 50:1.
  • the composition can comprise a molar ratio of sterically hindered primary amine (e.g. , amino acid) to linaclotide between 100: 1 and 1 : 100.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100 : 1 and 1 : 1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 90: 1 and 2:1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 80:1 and 5:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 70:1 and 10:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 60: 1 and 20: 1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 50:1 and 30:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 40: 1 and 20: 1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100:1 and 20:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100:1 and 25: 1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100:1 and 30:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100: 1 and 40: 1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100:1 and 50:1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100: 1 and 60: 1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide between 100:1 and 70:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 5:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 10:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 20:1.
  • the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 25:1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 30: 1. In some embodiments, the composition comprises a molar ratio of sterically hindered primary amine to linaclotide of at least 40: 1.
  • Suitable sterically hindered primary amines for inclusion in the delayed release composition are, for example, naturally-occurring amino acids (e.g., alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, meglumine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine), synthetic amino acids (e.g., lanthionine, theanine or 1 -amino cyclohexane), amino sugars (e.g., chitosan or glucosamine), or combination or mixtures thereof.
  • amino acids e.g., alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, le
  • the composition comprises an amino acid selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • the composition comprises an amino acid selected from leucine, isoleucine, asparagine, glutamine, glutamic acid, histidine, cysteine, alanine, serine, threonine, tyrosine, proline, tryptophan, or a combination or mixture thereof.
  • the composition comprises an amino acid selected from leucine, isoleucine, methionine, alanine, or a combination or mixture thereof. In some embodiments, the composition comprises an amino acid selected from leucine, isoleucine, alanine, or a combination or mixture thereof. In some embodiments, the composition comprises an amino acid selected from leucine, isoleucine, methionine, or a combination or mixture thereof. In some embodiments, the composition comprises an amino acid selected from leucine, methionine, alanine, or a combination or mixture thereof. In some embodiments, the composition comprises leucine, methionine, or a mixture thereof.
  • the composition comprises leucine, isoleucine, or a mixture thereof. In some embodiments, the composition comprises leucine, alanine, or a mixture thereof. In some embodiments, the composition comprises leucine. In some embodiments, the composition comprises isoleucine.
  • the composition comprises methionine. In some embodiments, the composition comprises alanine. In some embodiments, the composition comprises histidine.
  • the delayed release composition can comprise any stabilizing amount of a cation (e.g., metal cation).
  • the composition comprises a molar ratio of cation to linaclotide between 300:1 and 1 :1.
  • the composition comprises a molar ratio of cation to linaclotide between 250: 1 and 30:1.
  • the composition can comprise a molar ratio of cation to linaclotide between 100: 1 and 1 :100.
  • the composition comprises a molar ratio of cation to linaclotide between
  • the composition comprises a molar ratio of cation to linaclotide between 90:1 and 2:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 80:1 and 5:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 70:1 and 10: 1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 60:1 and 20:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between
  • the composition comprises a molar ratio of cation to linaclotide between 40: 1 and 20: 1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100:1 and 20:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100 : 1 and 25:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100:1 and 30:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100:1 and 40:1.
  • the composition comprises a molar ratio of cation to linaclotide between 100: 1 and 50:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100:1 and 60:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide between 100: 1 and 70: 1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 5:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 10:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 20:1.
  • the composition comprises a molar ratio of cation to linaclotide of at least 25:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 30:1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 40: 1. In some embodiments, the composition comprises a molar ratio of cation to linaclotide of at least 60:1.
  • any suitable cation(s) can be included in the composition, for example, any suitable metal cation or organic cation.
  • the composition comprises a metal cation selected from calcium, potassium, magnesium, zinc, aluminum, iron, tin, manganese, chromium, cobalt, nickel, barium, sodium, or a combination or mixture thereof.
  • the composition comprises a metal cation selected from calcium, potassium, magnesium, zinc, aluminum, manganese, chromium, cobalt, nickel, barium, sodium, or a combination or mixture thereof.
  • the composition comprises a metal cation selected from aluminum, calcium, potassium, sodium, magnesium, manganese, zinc, or a combination or mixture thereof.
  • the composition comprises a metal cation selected from calcium, magnesium, manganese, zinc, or a combination or mixture thereof. In some embodiments, the composition comprises a divalent metal cation. In some embodiments, the composition comprises a divalent metal cation selected from ⁇ 1 3+ ⁇ Ca 2+ , Mg 2 *, Zn 2+ , Mn 2+ , or a combination or mixture thereof. In some embodiments, the composition comprises Mg 2 *. In some embodiments, the composition comprises Ca + . In some embodiments, the composition comprises Zn 2+ . In some embodiments, the composition comprises aluminum.
  • the metal cation can be added to the composition in any suitable form, for example any pharmaceutically acceptable salt with any appropriate counterion.
  • Suitable metal salts include, for example, calcium chloride, calcium carbonate, calcium acetate, magnesium chloride, magnesium acetate, zinc acetate, zinc chloride, or mixtures thereof.
  • the composition comprises calcium chloride, magnesium chloride, zinc acetate, or any combination or mixture thereof.
  • the composition comprises calcium chloride.
  • the composition comprises magnesium chloride.
  • the composition comprises zinc acetate.
  • Suitable organic cations include, for example, ammonium hydroxide, D-arginine, L-arginine, t-butylamine, calcium acetate hydrate, calcium carbonate, calcium DL-malate, calcium hydroxide, choline, ethanolamine, ethylenediamine, glycine, L-histidine, L-lysine, magnesium hydroxide, N- methyl-D-glucamine, L-omithine hydrochloride, potassium hydroxide, procaine hydrochloride, L-proline, pyridoxine, L-serine, sodium hydroxide, DL-tryptophan, tromethamine, L-tyrosine, L-valine, carnitine, taurine, creatine malate, arginine alpha ketoglutarate, ornithine alpha ketoglutarate, spermine acetate, spermidine chloride, or combinations or mixtures thereof.
  • the organic cation is selected from the group consisting of N-methyl D-glucamine, choline, arginine, lysine, procaine, tromethamine (TRIS), spermine, N-methyl-morpholine, glucosamine, N,N-bis(2- hydroxyethyl) glycine, diazabicycloundecene, creatine, arginine ethyl ester, amantadine, rimantadine, ornithine, taurine, and citrulline, or any combination or mixture thereof.
  • TMS tromethamine
  • the composition can contain any stabilizing amount of a polymer.
  • the composition comprises between 1 and 25 % by weight of a polymer, relative to the total weight of the composition. In some embodiments, the composition comprises between 1 and 10% by weight of a polymer, relative to the total weight of the composition.
  • the composition comprises between 2 and 4 % by weight of a polymer, relative to the total weight of the composition. In some embodiments, the composition comprises between 0.1 and 75 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 55 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 35 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 30 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 25 wt.% of a polymer. In some embodiments, the composition comprises between 1 and 25 wt.% of a polymer.
  • the composition comprises between 5 and 25 wt.% of a polymer. In some embodiments, the composition comprises between 10 and 25 wt.% of a polymer. In some embodiments, the composition comprises between 15 and 25 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 22 wt.% of a polymer. In some embodiments, the composition comprises between 1 and 22 wt.% of a polymer. In some embodiments, the composition comprises between 5 and 22 wt.% of a polymer. In some embodiments, the composition comprises between 10 and 22 wt.% of a polymer. In some embodiments, the composition comprises between 0.1 and 20 wt.% of a polymer.
  • the composition comprises between 1 and 20 wt.% of a polymer. In some embodiments, the composition comprises between 5 and 20 wt.% of a polymer. In some embodiments, the composition comprises between 10 and 20 wt.% of a polymer. In some embodiments, the composition comprises between 0.01 and 15 wt.% of a polymer. In some embodiments, the composition comprises between 0.01 and 10 wt.% of a polymer. In some embodiments, the composition comprises between 0.01 and 5 wt.% of a polymer.
  • the composition comprises between 0.1 and 95 wt.%, for example, between 5 and 95 wt.%, between 15 and 95 wt.%, between 25 and 95 wt.%, between 35 and 95 wt.%, between 45 and 95 wt.%, between 0.1 and 85 wt.%, between 1 and 85 wt.%, between 5 and 85 wt.%, between 1 and 85 wt.%, between 25 and 85 wt.%, between 35 and 85 wt.%, between 0.1 and 80 wt.%, between 1 and 80 wt.%, between 5 and 80 wt.%, between 15 and 80 wt.%, between 25 and 80 wt.%, between 35 and 80 wt.%, between 0.1 and 75 wt.%, between 1 and 75 wt.%, between 5 and 75 wt.%, between 15 and 75 wt.%, between 25 and 75 wt.%, between 35 and 75 wt.%, between 0.1 and
  • the polymer acts as both a stabilizer, protective coating, or as a film forming agent within the delayed release composition.
  • the delayed release composition comprises a molar ratio of polymer (e.g., PVP or PVA) to linaclotide between 80:1 and 300: 1, for example, between 100: 1 and 200: 1, between 110: 1 and 190: 1, or even between 120:1 and 180: 1.
  • the delayed release composition comprises a molar ratio of polymer (e.g., PVP or PVA) to linaclotide greater than about 80:1, for example, greater than about 100:1, or even greater than about 120:1.
  • the delayed release composition comprises a weight ration of polymer (e.g., PVP or PVA) to linaclotide between 10: 1 and 300: 1 , for example, between 80: 1 and 200: 1 , between 100: 1 and 180:1, or even between 1 10:1 and 150:1.
  • the delayed release composition comprises a weight ration of polymer (e.g., PVP or PVA) to linaclotide between 100: 1 and 500:1 , for example, between 200:1 and 400:1, between 250:1 and 350:1, or even between 300:1 and 350:1.
  • Suitable polymers for inclusion in the delayed release compositions are, for example, polyvinyl pyrrolidone (PVP), polyvinyl alcohol (PVA), polyvinyl alcohol low peroxide (PVA-LP), hydroxylpropyl methyl cellulose (HPMC), hydroxylpropyl cellulose (HPC), methyl cellulose, methacrylate polymers, cyclodextrin, dextrin, dextran, polyacrylic acid, chitosan, guar gum, xanthan gum, polyethylene oxide (e.g., polyethylene polypropylene oxide), poly (sodium vinylsulfonate), polyethylene glycol, poly(arginine), poly carbophil, polyvinyl pyrrolidone-co-vinyl acetate, a poloxamer (e.g., Pluronic® products available from BASF), alginate, trehalose, sucrose, inulin, or a combination or mixture thereof.
  • PVP polyvinyl pyrroli
  • the composition comprises a polymer selected from PVP, PVA, methacrylate polymers, cyclodextrin, dextran, polyacrylic acid, chitosan, guar gum, xanthan gum, polyethylene oxide, polyethylene glycol, poly(arginine), poly carbophil, polyvinyl pyrrolidone-co-vinyl acetate, a poloxamer, or a combination or mixture thereof.
  • the composition comprises PVP, PVA, polyethylene oxide, or a mixture thereof.
  • the composition comprises PVP, PVA, or a mixture thereof.
  • the composition comprises PVP.
  • the composition comprises PVA.
  • the delayed release composition comprises two or more stabilizing agents.
  • the composition can include a stabilizing amount of a polymer and a stabilizing amount of a sterically hindered primary amine.
  • the composition can include a stabilizing amount of a polymer and a stabilizing amount of a cation (e.g., metal cation).
  • the composition can include a stabilizing amount of a sterically hindered primary amine and a stabilizing amount of a cation (e.g., metal cation).
  • the composition comprises a stabilizing amount of a polymer, a stabilizing amount of a sterically hindered primary amine, and a stabilizing amount of a cation (e.g., metal cation).
  • the delayed release composition comprises a stabilizing amount of PVP and a stabilizing amount of an amino acid selected from histidine, alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • an amino acid selected from histidine, alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of an amino acid selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of leucine, isoleucine, methionine, alanine, or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of histidine.
  • the delayed release composition comprises a stabilizing amount of PVA and a stabilizing amount of an amino acid selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of an amino acid selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, or a mixture thereof.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of leucine, isoleucine, methionine, alanine, or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of leucine.
  • the delayed release composition comprises a stabilizing amount of PVP and a stabilizing amount of a cation ⁇ e.g. , metal cation).
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of a divalent metal cation.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of Mg 2+ , Ca 2+ , Zn 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of Ca 2+ or a salt thereof.
  • the composition comprises a stabilizing amount of PVP and a stabilizing amount of Mg 2 * or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVP and a stabilizing amount of Zn 2+ or a salt thereof.
  • the delayed release composition comprises a stabilizing amount of PVA and a stabilizing amount of a cation ⁇ e.g., metal cation).
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of a divalent metal cation.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of Mg 2+ , Ca 2+ , Zn 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of Ca + or a salt thereof.
  • the composition comprises a stabilizing amount of PVA and a stabilizing amount of Mg 2"1" or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVA and a stabilizing amount of Zn 2+ or a salt thereof.
  • the delayed release composition comprises a stabilizing amount of an amino acid selected from leucine, isoleucine, methionine, alanine; and a stabilizing amount of a divalent metal cation selected from Mg 2 *, Ca 2+ , Zn 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of an amino acid selected from leucine, and isoleucine; and a stabilizing amount of a divalent metal cation selected from Nig 2 *, Ca 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of an amino acid selected from leucine or methionine; and a stabilizing amount of a divalent metal cation selected from Ca 2+ , Zn 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of leucine and a stabilizing amount of Ca 2+ or a salt thereof.
  • the composition comprises a stabilizing amount of a cation and a stabilizing amount of a sterically hindered primary amine.
  • the composition comprises a cation and a sterically hindered primary amine in a molar ratio of cation: sterically hindered primary amine (e.g., Ca 2+ :leucine) of at least 1.5:1, e.g., at least 2:1, at least 2.5:1 , at least 3:1, at least 4:1, or even at least 5:1 (for example, a molar ratio between 1.5: 1 and 5:1, e.g., between 2:1 and 4: 1).
  • the delayed release composition comprises (i) a stabilizing amount of PVP or PVA, (ii) a stabilizing amount of leucine, isoleucine, methionine, alanine, and (iii) a stabilizing amount of Mg 2"1" , Ca 2+ , Zn 2+ or a salt thereof or a combination or mixture thereof.
  • the composition comprises a stabilizing amount of PVP, a stabilizing amount of leucine, and a stabilizing amount of a metal cation.
  • the composition comprises a stabilizing amount of PVA, a stabilizing amount of histidine, and a stabilizing amount of Ca 2+ or a salt thereof.
  • the composition comprises a stabilizing amount of PVP, a stabilizing amount of leucine, and a stabilizing amount of Mg 2+ or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVP, a stabilizing amount of leucine, and a stabilizing amount of Zn 2+ or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVA, a stabilizing amount of leucine, and a stabilizing amount of Ca 2+ or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVA, a stabilizing amount of leucine, and a stabilizing amount of Mg * or a salt thereof. In some embodiments, the composition comprises a stabilizing amount of PVA, a stabilizing amount of leucine, and a stabilizing amount of Zn + or a salt thereof.
  • the composition comprises (i) between 0.1 and 30 wt.% of a polymer, (ii) a sterically hindered primary amine (e.g., an amino acid) in a molar ratio of primary amine to linaclotide between 100:1 and 10:1, and (iii) a cation (e.g., a metal cation) in a molar ratio of cation to linaclotide between 100: 1 and 40:1.
  • a sterically hindered primary amine e.g., an amino acid
  • a cation e.g., a metal cation
  • the composition comprises (i) between 5 and 25 wt.% of a polymer, (ii) a sterically hindered primary amine (e.g., an amino acid) in a molar ratio of primary amine to linaclotide 100:1 and 30:1 (e.g., between 60:1 and 30: 1 or even between 50:1 and 30: 1), and (iii) a cation (e.g., a metal cation) in a molar ratio of cation to linaclotide between 100: 1 and 60:1.
  • a sterically hindered primary amine e.g., an amino acid
  • a cation e.g., a metal cation
  • the composition comprises (i) between 0.1 and 30 wt.% of a polymer selected from PVP and PVA, (ii) an amino acid selected from leucine, isoleucine, alanine, and methionine in a molar ratio of amino acid to linaclotide between 100:1 and 10:1, and (iii) a metal cation selected from Ca 2+ , Mg 2+ , and Zn 2+ in a molar ratio of cation to linaclotide between 100: 1 and 40:1.
  • the composition comprises (i) between 5 and 25 wt.% of a polymer selected from PVP and PVA, (ii) an amino acid selected from leucine, isoleucine, alanine, and methionine in a molar ratio of amino acid to linaclotide 100:1 and 30:1 (e.g., between 60:1 and 30:1), and (iii) a metal cation selected from Ca 2+ , Mg 2+ , and Zn 2+ in a molar ratio of cation to linaclotide between 100:1 and 60:1.
  • the composition comprises (i) between 0.1 and 30 wt.% (e.g., between 5 and 25 wt.%) of PVP or PVA, (ii) leucine in a molar ratio of leucine to linaclotide between 100:1 and 30:1 (e.g., between 60:1 and 30:1 or even between 50:1 and 30:1), and (iii) Ca 2+ in a molar ratio of Ca 2+ to linaclotide between 100:1 and 60:1.
  • the composition comprises (i) between 45 and 99 wt.% of a polymer, (ii) a sterically hindered primary amine (e.g., an amino acid) in a molar ratio of primary amine to linaclotide between 100:1 and 10:1, and (iii) a cation (e.g., a metal cation) in a molar ratio of cation to linaclotide between 100:1 and 40:1.
  • a sterically hindered primary amine e.g., an amino acid
  • a cation e.g., a metal cation
  • the composition comprises (i) between 45 and 70 wt.% of a polymer, (ii) a sterically hindered primary amine (e.g., an amino acid) in a molar ratio of primary amine to linaclotide 100:1 and 30:1 (e.g., between 60: 1 and 30: 1 or even between 50: 1 and 30:1), and (iii) a cation (e.g., a metal cation) in a molar ratio of cation to linaclotide between 100: 1 and 60:1.
  • a sterically hindered primary amine e.g., an amino acid
  • a cation e.g., a metal cation
  • the composition comprises (i) between 45 and 99 wt.% of a polymer selected from PVP and PVA, (ii) an amino acid selected from leucine, isoleucine, alanine, and methionine in a molar ratio of amino acid to linaclotide between 100: 1 and 10:1, and (iii) a metal cation selected from Ca 2+ , Mg 2+ , and Zn 2+ in a molar ratio of cation to linaclotide between 100: 1 and 40: 1.
  • the composition comprises (i) between 45 and 70 wt.% of a polymer selected from PVP and PVA, (ii) an amino acid selected from leucine, isoleucine, alanine, and methionine in a molar ratio of amino acid to linaclotide 100:1 and 30:1 (e.g., between 60:1 and 30: 1), and (iii) a metal cation selected from Ca 2+ , Mg 2+ , and Zn 2+ in a molar ratio of cation to linaclotide between 100:1 and 60:1.
  • the composition comprises (i) between 45 and 99 wt.% (e.g., between 45 and 70 wt.%) of PVP or PVA, (ii) leucine in a molar ratio of leucine to linaclotide between 100:1 and 30: 1 (e.g., between 60:1 and 30:1 or even between 50: 1 and 30:1), and (iii) Ca 2+ in a molar ratio of Ca 2+ to linaclotide between 100:1 and 60: 1.
  • the delayed release composition may also comprise any one or more filling agents.
  • suitable filling agents include, but are not limited to, starch, calcium carbonate, calcium sulfate, hydroxylpropylmethyl cellulose, fructose, methyl cellulose, dextrates, dextrose, dextran, lactitol, maltose, sucrose, sorbitol, isomalt, pregelatinized starch, dicalcium phosphate, microcrystalline cellulose, mannitol, gelatin, trehalose, erythritol, maltitol, lactose, glucose, or a combination thereof, or a mixture thereof.
  • the filling agent is isomalt. In some embodiments, the filling agent is gelatin. In some embodiments, the filling agent is mannitol. In some embodiments, the filling agent is pregelatinized starch. In some embodiments, the filling agent is microcrystalline cellulose.
  • the delayed release composition (e.g. , delayed release tablet) can comprise any suitable concentration of filling agent.
  • the composition comprises one or more filling agents in a concentration of 0.1-99 % by weight, relative to the total weight of the composition.
  • the composition comprises one or more filling agents in a concentration of 1 -95 wt.% of filling agent(s), relative to the total weight of the composition.
  • the composition comprises one or more filling agents in a concentration of 10-90 wt.% of filling agent(s), relative to the total weight of the composition.
  • the composition comprises one or more filling agents in a concentration of 20-90 wt.% of filling agent(s), relative to the total weight of the composition. In some embodiments, for example, the composition comprises one or more filling agents in a concentration of 25-85 wt.% of filling agent(s), relative to the total weight of the composition. In some embodiments, for example, the composition comprises one or more filling agents in a concentration of 30-80 wt.% of filling agent(s), relative to the total weight of the composition. In some embodiments, for example, the composition comprises one or more filling agents in a concentration of 40-70 wt.% of filling agent(s), relative to the total weight of the composition.
  • the composition comprises one or more filling agents in a concentration of 10-60 wt.% of filling agent(s), relative to the total weight of the composition. In some embodiments, for example, the composition comprises one or more filling agents in a concentration of 20-50 wt.% of filling agent(s), relative to the total weight of the composition. In some embodiments, the composition comprises one or more filling agents in a concentration of at least 20 wt.%, for example, at least 40 wt.%, at least 60 wt.%, at least 70 wt.%, at least 80 wt.%, or at least 90 wt.%, relative to the total weight of the composition.
  • the delayed release composition (e.g., delayed release film) comprises one or more plasticizers.
  • plasticizers include, but are not limited to, polyethylene glycol, propylene glycol, glycerin, glycerol, monoacetin, diacetin, triacetin, dimethyl phthalate, diethyl phthalate, dibutyl phthalate, dibutyl sebacate, triethyl titrate, tributyl citrate, triethyl citrate, triethyl acetyl citrate, castor oil, acetylated monoglycerides, sorbitol or combinations thereof.
  • the concentration of the plasticizer in the formulation may be about 0 to about 30 wt.%, for example, about 1 to about 20 wt.%, about 0 to about 10 wt.%, about 1 to about 5 wt.%, or even 0 to about 4 wt.%.
  • the delayed release composition comprises a film forming agent, a water-soluble polymer, a pH sensitive polymer, biodegradable polymer, or combination thereof.
  • Water soluble, pH sensitive, or biodegradable polymers that may be used in the orally dissolving formulations of the present invention include, but are not limited to, cellulose derivatives, synthetic polymers polyacrylates and natural gums.
  • the water soluble polymers used in the orally dissolving formulations of the present invention may include, but are not limited to, methyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, ethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, carboxymethyl cellulose, cellulose acetate phthalate, cellulose acetate butyrate, amylose, dextran, casein, pullulan, gelatin, pectin, agar, carrageenan, xanthan gum, tragacanth, guar gum, acacia gum, arabic gum, polyethylene glycol, polyethylene oxide, polyvinyl pyrrolidone, polyvinyl alcohol, cyclodextrin, carboxyvinyl polymers, sodium alginate, polyacrylic acid, methylmethacrylate or mixtures thereof.
  • the concentration of the water-soluble polymer in the formulation may be about 20% to about 90%
  • the pH sensitive polymer is Eudagrit® L100 that has a threshold pH (also called dissolution pH) of 6.0.
  • the pH sensitive polymer is Eudagrit® SI 00 that has a threshold pH of 7.0.
  • the pH sensitive polymer is Eudagrit® L-30D that has a threshold pH of 5.6.
  • the pH sensitive polymer is Eudagrit® FS 30D that has a threshold pH of 6.8.
  • the pH sensitive polymer is Eudagrit® L100-55 that has a threshold pH of 5.5.
  • the pH sensitive polymer is Polyvinyl acetate phthalate that has a threshold pH of 5.0. In some embodiments, the pH sensitive polymer is
  • the pH sensitive polymer is Hydroxypropylmethylcellulose phthalate that has a threshold pH of 4.5-4.8.
  • the pH sensitive polymer is Hydroxypropylmethylcellulose phthalate 50 that has a threshold pH of 5.2.
  • the pH sensitive polymer is Hydroxypropylmethylcellulose phthalate 55 that has a threshold pH of 5.4.
  • the pH sensitive polymer is Cellulose acetate trimelliate that has a threshold pH of 4.8.
  • the pH sensitive polymer is Cellulose acetate phthalate that has a threshold pH of 5.0.
  • the delayed release composition comprises a combination of the pH sensitive polymers mentioned above.
  • the delayed release compositions may include one or more disintegrants, lubricants, anti-caking additives, anti-microbial agents, antifoaming agents, emulsifiers, surfactants, buffering agents, and/or coloring agents.
  • Suitable disintegrants include, for example, agar-agar, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, povidone, polacnlin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, clays, other algins, other celluloses, gums, and mixtures thereof.
  • the disintegrant is crospovidone.
  • the disintegrant is croscarmellose sodium.
  • Suitable lubricants include, for example, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil and soybean oil), zinc stearate, ethyl oleate, ethyl laurate, agar, syloid silica gel (AEROSIL® 200, W.R.
  • AEROSIL® 200 ethyl oleate
  • W.R syloid silica gel
  • Suitable anti-caking additives include, for example, calcium silicate, magnesium silicate, silicon dioxide, colloidal silicon dioxide, talc, glyceryl, and mixtures thereof.
  • Suitable anti-microbial additives that may be used, e.g. , as a preservative for the linaclotide compositions, include, for example, benzalkonium chloride, benzethonium chloride, benzoic acid, benzyl alcohol, butyl paraben, cetylpyridinium chloride, cresol, chlorobutanol, dehydroacetic acid, ethylparaben, methylparaben, phenol, phenylethyl alcohol, phenoxyethanol, phenylmercuric acetate, phenylmercuric nitrate, potassium sorbate, propylparaben, sodium benzoate, sodium dehydroacetate, sodium propionate, sorbic acid, thimersol, thymol, and mixtures thereof.
  • composition may also comprise any suitable pharmaceutically acceptable carrier or medium.
  • suitable pharmaceutically acceptable carriers include, for example, any solvents, dispersants, pH buffering agents, coatings, absorption promoting agents, controlled release agents, and one or more inert excipients (e.g., filling agents, starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, disintegrating agents), or the like.
  • compositions can contain any desired additional components, additives, and/or species, for example, surface active additives, dispersing additives, humectants, suspending agents, solubilizers, buffering agents, disintegrants, preservatives, colorants, flavorants, and the like.
  • the composition comprises one or more ion species that interact with linaclotide.
  • the composition can also comprise any suitable pH buffering agent.
  • the pH buffering agent is present in the composition in an amount sufficient to achieve the isoelectric point of linaclotide.
  • the composition can have any desired pH.
  • the composition has a pH of 2 to 5 (for example, a pH of 2 to 4.5, a pH of 2 to 4, a pH of 2.5 to 4, a pH of 2.5 to 3.5, a pH of 2.5 to 3, or even a pH of 3).
  • the composition comprises linaclotide and a hydrolysis product, e.g., a hydrolysis product comprising or having a structure of:
  • the composition can contain any desired concentration of the hydrolysis product. In some embodiments, the composition comprises less than 10 wt.% of the hydrolysis product.
  • the composition comprises less than 7 wt.% of the hydrolysis product.
  • the composition comprises less than 6 wt.% of the hydrolysis product.
  • the composition comprises less than 5 wt.% of the hydrolysis product.
  • the composition comprises less than 4 wt.% of the hydrolysis product.
  • the composition comprises less than 3 wt.% of the hydrolysis product.
  • the composition comprises less than 2 wt.% of the hydrolysis product.
  • the composition comprises less than 1 wt.% of the hydrolysis product.
  • the composition comprises between 0.01 and 10 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0. 1 and 7 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0. 1 and 5 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and
  • the composition comprises between
  • the composition comprises between 0. 1 and 4 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 4 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 1 and 4 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0. 1 and 3 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 3 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 1 and 3 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.1 and 2.5 wt.% of the hydrolysis product.
  • the composition comprises between 0.5 and 2.5 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 1 and 2.5 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.1 and 2 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 2 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 1 and 2 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.1 and 1.5 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 1.5 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.1 and 1 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 1 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 1 wt.% of the hydrolysis product. In some embodiments, the composition comprises between 0.5 and 1 wt.% of the hydro
  • the composition comprises linaclotide and an oxidation product, e.g., an oxidation product comprising or having a structure of:
  • the composition comprises linaclotide and an oxidation product having the depicted structure but wherein oxidation occurs at any one or more of the six depicted cysteinyl sulfurs.
  • the composition can contain any desired concentration of the oxidation product. In some embodiments, the composition comprises less than 10 wt.% of the oxidation product. In some embodiments, the composition comprises less than 7 wt.% of the oxidation product. In some embodiments, the composition comprises less than 6 wt.% of the oxidation product. In some embodiments, the composition comprises less than 5 wt.% of the oxidation product. In some embodiments, the composition comprises less than 4 wt.% of the oxidation product.
  • the composition comprises less than 3 wt.% of the oxidation product. In some embodiments, the composition comprises less than 2 wt.% of the oxidation product. In some embodiments, the composition comprises less than 1 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.01 and 10 wt.% of the oxidation product. In some embodiments, the composition comprises between 0. 1 and 7 wt.% of the oxidation product. In some embodiments, the composition comprises between 0. 1 and 5 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 5 wt.% of the oxidation product.
  • the composition comprises between 1 and 5 wt.% of the oxidation product. In some embodiments, the composition comprises between 0. 1 and 4 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 4 wt.% of the oxidation product. In some embodiments, the composition comprises between 1 and 4 wt.% of the oxidation product. In some embodiments, the composition comprises between 0. 1 and 3 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 3 wt.% of the oxidation product. In some embodiments, the composition comprises between 1 and 3 wt.% of the oxidation product.
  • the composition comprises between 0.1 and 2.5 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 2.5 wt.% of the oxidation product. In some embodiments, the composition comprises between 1 and 2.5 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.1 and 2 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 2 wt.% of the oxidation product. In some embodiments, the composition comprises between 1 and 2 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.1 and 1.5 wt.% of the oxidation product.
  • the composition comprises between 0.5 and 1.5 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.1 and 1 wt.% of the oxidation product. In some embodiments, the composition comprises between 0.5 and 1 wt.% of the oxidation product.
  • the composition comprises linaclotide and an acetylation product, e.g., an acetylation product comprising or having:
  • the composition can contain any desired concentration of the acetylation product. In some embodiments, the composition comprises less than 10 wt.% of the acetylation product. In some embodiments, the composition comprises less than 7 wt.% of the acetylation product. In some embodiments, the composition comprises less than 6 wt.% of the acetylation product. In some embodiments, the composition comprises less than 5 wt.% of the acetylation product. In some embodiments, the composition comprises less than 4 wt.% of the acetylation product. In some embodiments, the composition comprises less than 3 wt.% of the acetylation product. In some embodiments, the composition comprises less than 2 wt.% of the acetylation product.
  • the composition comprises less than 1 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.01 and 10 wt.% of the acetylation product. In some embodiments, the composition comprises between 0. 1 and 7 wt.% of the acetylation product. In some embodiments, the composition comprises between 0. 1 and 5 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.5 and 5 wt.% of the acetylation product. In some embodiments, the composition comprises between 1 and 5 wt.% of the acetylation product. In some embodiments, the composition comprises between 0. 1 and 4 wt.% of the acetylation product.
  • the composition comprises between 0.5 and 4 wt.% of the acetylation product. In some embodiments, the composition comprises between 1 and 4 wt.% of the acetylation product. In some embodiments, the composition comprises between 0. 1 and 3 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.5 and 3 wt.% of the acetylation product. In some embodiments, the composition comprises between 1 and 3 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.1 and 2.5 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.5 and 2.5 wt.% of the acetylation product.
  • the composition comprises between 1 and 2.5 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.1 and 2 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.5 and 2 wt.% of the acetylation product. In some embodiments, the composition comprises between 1 and 2 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.1 and 1.5 wt.% of the acetylation product. In some embodiments, the composition comprises between 0.5 and 1.5 wt.% of the acetylation product. In some embodiments, the composition comprises between
  • the composition comprises between 0.5 and 1 wt.% of the acetylation product.
  • a pharmaceutical composition comprising linaclotide, and one or more peptides selected from:
  • a peptide (“Cys'-IMD”) or a pharmaceutically acceptable salt thereof, wherein the peptide comprises the amino acid structure of: ii. a hydrolysis peptide (“Asp 7 ”) or a pharmaceutically acceptable salt thereof, wherein the peptide comprises the amino acid structure of:
  • peptide (Des-Tyr 14 ) or a pharmaceutically acceptable salt thereof, wherein the peptide comprises the amino acid structure of:
  • a peptide (Cys'-a-Ketone) or a pharmaceutically acceptable salt thereof, wherein the peptide comprises the amino acid structure of: ⁇ Cys— Glu— Tyr— Cys— Cys— Asn— Pro— Ala— Cys— Thr— Gly— Cys— Tyr— OH
  • thea Cys'-oc-Ketone peptide may be present in its hydrated form or a pharmaceutically acceptable salt thereof, wherein the peptide comprises an amino acid structure of:
  • the Cys'-a-Ketone peptide comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about 7% by weight of the composition, less than about 5% by weight of the composition, less than about 4% by weight of the composition, less than about 3% by weight of the composition, less than about 2% by weight of the composition, less than about 1.5% by weight of the composition, or less than about 1% by weight of the composition.
  • the Cys'-a-Ketone peptide comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the Cys'-IMD peptide comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about 7% by weight of the composition, less than about 5% by weight of the composition, less than about 4% by weight of the composition, less than about 3.5% by weight of the composition, less than about 3% by weight of the composition, less than about 2% by weight of the composition, or less than about 1% by weight of the composition.
  • the Cys'-IMD peptide comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the hydrolysis peptide (“Asp 7 ”) comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about 7% by weight of the composition, less than about 5% by weight of the composition, less than about 4% by weight of the composition, less than about 3.5% by weight of the composition, less than about 3% by weight of the composition, less than about 2% by weight of the composition, or less than about 1% by weight of the composition.
  • the hydrolysis peptide (“Asp 7 ”) comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the acetylation peptide (“Cys'-N- Acetyl”) comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about 7% by weight of the composition, less than about 5% by weight of the composition, less than about 4% by weight of the composition, less than about 3.5% by weight of the composition, less than about 3% by weight of the composition, less than about 2% by weight of the composition, or less than about 1% by weight of the composition.
  • the acetylation peptide (“Cys'-N- Acetyl”) comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the linaclotide trisulfide peptide comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about 7% by weight of the composition, less than about 5% by weight of the composition, less than about 4% by weight of the composition, less than about 3.5% by weight of the composition, less than about 3% by weight of the composition, less than about 2% by weight of the composition, or less than about 1% by weight of the composition.
  • the linaclotide trisulfide peptide comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the Des-Tyr 14 peptide comprises less than about 15% by weight of the composition, less than about 10% by weight of the composition, less than about
  • the Des-Tyr 14 peptide comprises from about 0.01% to about 15% by weight of the composition, about 0.05% to about 10% by weight of the composition, about 0.05% to about 7% by weight of the composition or about 0.05% to about 5% by weight of the composition.
  • the composition comprises linaclotide and any desired concentration of multimers. In some embodiments, the composition comprises less than 10 wt.% of multimer(s). In some embodiments, the composition comprises between 0.5 and 1 wt.% of multimer(s).
  • the composition comprises an effective amount of linaclotide and any desired amount of reduced form linaclotide.
  • reduced form linaclotide refers to linaclotide having no disulfide bonds between cysteine amino acids.
  • the composition comprises less than 10 wt.% of reduced form linaclotide. In some embodiments, the composition comprises between 0.5 and 1 wt.% of reduced form linaclotide.
  • the composition comprises an effective amount of linaclotide and any desired amount of scrambled form linaclotide.
  • scrambled form linaclotide refers to linaclotide having disulfide bonds between Cysi and Cysio, between Cysi and Cys ⁇ , between Cysi and Cys5, between Cysi and Cys 2 , between Cys 2 and C s6, between Cys 2 and Cysn, between Cys 2 and Cyss, between Cyss and Cys6, and/or between Cyss and Cysio.
  • the composition comprises between 0.5 and 1 wt.% of scrambled form linaclotide. In some embodiments, the composition comprises less than 10 wt.% of scrambled form linaclotide.
  • the composition comprises a total degradant concentration of less than about 10 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 8 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 7 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 6.5 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 6 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 5.5 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 5 wt.%.
  • the composition comprises a total degradant concentration of less than about 4 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 3 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 2.5 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 2 wt.%. In some embodiments, the composition comprises a total degradant concentration of less than about 1 wt.%.
  • the compositions can be prepared by spray drying, which is a technique used to prepare microparticles (e.g., microcapsules or microspheres) of drugs.
  • Spray-dried peptides generally retain their biological activity upon dissolution and may have useful physical characteristics, including a uniform particle size and a spherical shape.
  • the microparticles prepared by spray drying are often free flowing, which is helpful for pharmaceutical manufacturing processes such as forming tablets and filling capsules. Spray drying processes are also useful because they may be readily scaled up for clinical and commercial manufacturing.
  • the spray buffer comprises HC1, histidine, 1.5% PVA and 0.6% talc. This formulation can be used to produce lower dosing ranges between 36-29 ⁇ g.
  • the composition when administered, will dissolve to release linaclotide in targeted areas of the gastrointestinal tract.
  • the formulation may release the linaclotide over a period of time that is determined by a number of different factors. These factors include the dimensions of the formulation, the concentration of the linaclotide, and how the linaclotide is dispersed throughout the formulation. For example, by varying the thickness and surface area of the formulations the rate of dissolution may be adjusted. A thick formulation will dissolve more slowly than an otherwise similar thin formulation and may be desirable to administer high dosages of linaclotide.
  • the delayed release composition has a disintegration rate of less than about 60 minutes in the targeted pH conditions. In some embodiments, the delayed release composition has a disintegration rate of less than about 30 minutes in the targeted pH conditions. In some embodiments, the delayed release composition has a disintegration rate of less than about 25 minutes. In some embodiments, the delayed release composition has a disintegration rate of less than about 20 minutes. In some embodiments, the delayed release composition has a disintegration rate of less than about 15 minutes. In some embodiments, the delayed release composition has a disintegration rate of less than about 10 minutes. In some embodiments, the delayed release composition disintegrates in less than about 30 minutes after entering a targeted environment.
  • the delayed release composition disintegrates in less than about 25 minutes after entering a targeted environment. In some embodiments, the delayed release composition disintegrates in less than about 20 minutes after entering a targeted environment. In some embodiments, the delayed release composition disintegrates in less than about,15 minutes after entering a targeted environment. [000157] In some embodiments, the delayed release composition releases at least about 75% of the linaclotide contained therein within 60 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 75% of the linaclotide contained therein within 30 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 80% of the linaclotide contained therein within 30 minutes of entering a targeted environment.
  • the delayed release composition releases at least about 85% of the linaclotide contained therein within 30 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 90% of the linaclotide contained therein within 30 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 95% of the linaclotide contained therein within 30 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 99% of the linaclotide contained therein within 30 minutes of entering a targeted environment.
  • the delayed release composition releases at least about 40% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 50% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 60% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 70% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 80% of the linaclotide contained therein within 15 minutes of entering a targeted environment.
  • the delayed release composition releases at least about 85% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 90% of the linaclotide contained therein within 15 minutes of entering a targeted environment. In some embodiments, the delayed release composition releases at least about 95% of the linaclotide contained therein within 15 minutes of entering a targeted environment.
  • the delayed release composition releases at least about 80% of the linaclotide contained therein between about 2 to about 2 hours of entering a targeted environment.
  • the delayed release composition releases at least about
  • the delayed release composition releases at least about 80% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 5. In some embodiments, the delayed release composition releases at least about 85% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 5. In some embodiments, the delayed release composition releases at least about 90% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 5. In some embodiments, the delayed release composition releases at least about 95% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 5. In some embodiments, the delayed release composition releases at least about 99% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 5.
  • the delayed release composition releases at least about 75% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7. In some embodiments, the delayed release composition releases at least about 80% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7. In some embodiments, the delayed release composition releases at least about 85% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7. In some embodiments, the delayed release composition releases at least about 90% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7. In some embodiments, the delayed release composition releases at least about 95% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7. In some embodiments, the delayed release composition releases at least about 99% of the linaclotide contained therein within 30 minutes of contacting a pH greater than 7.
  • the linaclotide DR compositions are formulated for delivery of linaclotide to the ileum, late ileum, or colon. In some embodiments, the linaclotide DR compositions are formulated for delivery of linaclotide to the ileum or ileal region. In some embodiments, the linaclotide DR compositions are formulated for delivery of linaclotide to within the late ileum to the ascending colon (e.g., at or near the ileocecal junction).
  • the composition or oral dosage form is administered to a pediatric patient in need thereof as a tablet, capsule or sachet.
  • a sachet comprising the composition is opened and the contents are sprinkled on or stirred into food, such as applesauce, or into a beverage, such as water.
  • a capsule is swallowed whole with fluid, such as water, or is opened and sprinkled on or stirred into food or a beverage. Tablets may be swallowed whole, may be crushed and stirred into food or a beverage, or may be formulated as a chewable tablet.
  • a subject or patient in whom administration of the pharmaceutical composition is an effective therapeutic regimen for a disease or disorder is preferably a human, but can be any animal, including a laboratory animal in the context of a clinical trial or screening or activity experiment.
  • the methods, compounds and compositions described herein are particularly suited for administration to any animal, particularly a mammal, and including, but by no means limited to, humans, rodents and non-rodents, such as feline or canine subjects, farm animals, such as but not limited to bovine, equine, caprine, ovine, and porcine subjects, wild animals (whether in the wild or in a zoological garden), research animals, such as mice, rats, rabbits, goats, sheep, pigs, dogs, cats, etc., avian species, such as chickens, turkeys, songbirds, etc., e.g., for veterinary medical use.
  • the linaclotide composition may be formulated as a rectal dosage form for rectal administration.
  • Rectal dosage forms include, without limitation, rectal suppositories, rectal foams or aerosols, enemas, rectal gels and rectal ointments.
  • the rectal dosage form may be administered to a patient in need thereof.
  • the rectal dosage form may be administered to a pediatric or geriatric patient.
  • Another aspect of the invention is a method of making a delayed release composition, the method comprising: preparing a linaclotide base, pregranulated filler, and placebo base; and blending and compressing the linaclotide base, pregranulated filler, and placebo base into a tablet.
  • the pregranulated filler is prepared through wet granulation and dried before blending and compressing into a tablet.
  • the method further comprises applying a subcoat to the tablet.
  • the method further comprises applying an enteric or functional coating to the tablet.
  • Another aspect of the invention is a method of making a delayed release composition, the method comprising: preparing an aqueous solution comprising linaclotide, or a pharmaceutically acceptable salt thereof; and applying the aqueous solution to a pharmaceutically acceptable carrier.
  • Delayed release mean that the composition dissolves, melts, disintegrates, liquefies, etc. in a targeted area of the gastrointestinal tract such that substantially all of the linaclotide no longer remains in a formulation, composition, or dosage form. Delayed release compositions include sustained release compositions, gastro-retentive compositions, targeted release compositions (e.g. colonic-release compositions, or compositions that target the ileosecal valve, etc.), extended release compositions and/or combinations thereof.
  • the term “delayed release composition” means that the composition is a dosage form that releases linaclotide at a time other than immediately following oral administration.
  • extended release composition means that the composition is a dosage form that releases linaclotide over an extended period of time after administration. This allows a reduction in dosing frequency compared to immediate release compositions.
  • the "disintegration" and “release” is used herein to mean that the capsule, film, bead, or tablet comprising linaclotide dissolves, melts, disintegrates, liquefies, etc. in the environment of an oral cavity such that substantially all of the linaclotide no longer remains in a formulation form, e.g., a pH greater than 5 or 7, or in a phosphate buffer solution and maintained at 37 ⁇ 1°C.
  • the term "entry into a targeted environment” means contact of the composition within a patient at a targeted organ or segment thereof, or within a segment of the GI intended for linaclotide release, e.g., having a pH greater than 5 or 7.
  • lower gastrointestinal means the distal segment of the gastrointestinal tract, for example, the ileum, terminal ileum, ileocecal valve, or colon.
  • upper gastrointestinal means the proximate segment of the gastrointestinal tract, for example, the stomach, duodenum and/or jejunum.
  • stabilizing agent refers to a polymer, sterically hindered primary amine ⁇ e.g., amino acid), or cation ⁇ e.g., metal cation) component of the composition which is included in the composition in a stabilizing amount.
  • a polymeric stabilizing agent is a polymer that is included in the composition in a stabilizing amount.
  • a sterically hindered primary amine stabilizing agent is a sterically hindered primary amine that is included in the composition in a stabilizing amount.
  • a cationic stabilizing agent is a cation that is included in the composition in a stabilizing amount.
  • stabilizing amount refers to a concentration, within the composition, of a polymer, sterically hindered primary amine (e.g., amino acid), or metal cation component at which the component increases the stability of linaclotide in the composition, as compared to a similar composition not having a stabilizing amount of the same component.
  • the term “substantially all” means at least about 90%, for example, at least about 95% or even at least about 99%.
  • isolated and purified means at least 95 percent pure (for example, at least 96% pure, at least 97% pure, at least 98% pure, or even at least 99% pure), as measured, for example, by chromatographic purity using
  • therapeutically effective amount means the amount of a linaclotide or a pharmaceutically acceptable salt thereof that, when administered to a mammal for treating a state, disorder or condition, is sufficient to effect a treatment (as defined below).
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, sex, weight, physical condition and responsiveness of the mammal to be treated.
  • a therapeutically effective amount of linaclotide, or its pharmaceutically acceptable salt or hydrate can be an amount effective to treat gastrointestinal disorders, including irritable bowel syndrome with constipation.
  • pharmaceutically acceptable means biologically or pharmacologically compatible for in vivo use in animals or humans, and preferably means, approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • the term “treat”, in all its verb forms, is used herein to mean to relieve, alleviate, prevent, and/or manage at least one symptom of a disorder in a subject, the disorder including, for example, a gastrointestinal disorder, such as irritable bowel syndrome with constipation.
  • the term “treat” also denotes, to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease) and/or reduce the risk of developing or worsening a disease.
  • treatment means the act of "treating” as defined above.
  • the term “prevent” refers to the prophylactic treatment of a subject who is at risk of developing a condition (e.g., stress related disorder) resulting in a decrease in the probability that the subject will develop the condition.
  • a condition e.g., stress related disorder
  • adverse event refers to any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with the treatment.
  • one adverse event is diarrhea.
  • additives refers to a pharmaceutically acceptable additive.
  • Pharmaceutically acceptable additives include, without limitation, binders, disintegrants, dispersing additives, lubricants, glidants, antioxidants, coating additives, diluents, surfactants, flavoring additives, humectants, absorption promoting additives, controlled release additives, anti-caking additives, anti-microbial agents (e.g., preservatives), colorants, desiccants, plasticizers and dyes.
  • an “excipient” is any pharmaceutically acceptable additive, filler, binder or agent.
  • stressed conditions refer to 40°C and 75% relative humidity (RH).
  • the terms “about” and “approximately” mean within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend, in part, on how the value is measured or determined, i.e., the limitations of the measurement system.
  • “about” can mean within 1 or more than 1 standard deviation, per practice in the art.
  • “about” with respect to the compositions can mean plus or minus a range of up to 20%, preferably up to 10%.
  • the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2- fold, of a value.
  • Particular values are described in the application and claims, unless otherwise stated the term “about” means within an acceptable error range for the particular value.
  • compositions include linaclotide and other desired pharmaceutically inactive additives, excipients, and/or components (e.g., polymers, sterically hindered primary amines, cations, filling agents, binders, carriers, excipients, diluents, disintegrating additives, lubricants, solvents, dispersants, coating additives, absorption promoting additives, hydrolysis products, formaldehyde imine products, oxidation products, acetylation products, deamidation products, multimers, controlled release additives, anti-caking additives, anti-microbial additives, preservatives, sweetening additives, colorants, flavors, desiccants, plasticizers, dyes, or the like), and no other active pharmaceutical ingredient(s).
  • linaclotide e.g., polymers, sterically hindered primary amines, cations, filling agents, binders, carriers, excipients, diluents, disintegrating additives,
  • Enteric-coated tablets comprising a core immediate release tablet containing a unit dose of linaclotide can be coated with coatings that dissolve only under pH conditions of the distal segment of intestine, so that linaclotide will be released in lower GI tract.
  • Linaclotide or a pharmaceutically acceptable salt thereof may be produced and purified using standard techniques known in the art, e.g., chemical synthesis or recombinant expression followed by purification using standard techniques.
  • Preparation of the linaclotide coating solution for beads Approximately 32 g to 42 g of purified water is mixed with hydrochloric acid to create a solution with a pH between 1.5 and 2.0. The cation, if used, is added to the solution in a quantity to provide the desired concentration, and the solution is mixed for sufficient time to produce a clear solution. The sterically hindered primary amine, if used, is added to the solution in a quantity to provide the desired concentration, and the solution is mixed for sufficient time to produce a clear solution. Other additives, such as antioxidants, are then added, if desired. The pH of the solution is tested, and hydrochloric acid is added, if necessary, to produce a solution having a pH between 1.5 and 2.0. The binder is then added to the solution and the mixture is then stirred for sufficient time to achieve a clear solution. The desired amount of linaclotide is added to the solution and mixed for 30-100 minutes to provide the coating solution.
  • the coating solution comprises linaclotide, histidine, 1.5%
  • Linaclotide can be formulated into a tablet for delayed drug release. Compared to an equal volume of beads, tablets have much smaller specific surface area, which makes them potentially less prone to degradation induced by environmental factors such as humidity, oxidation, deamidation, etc. In addition, the smaller surface area of the tablet can become advantageous when an enteric coating is needed since much less coating material is required to cover the surface of the dosage form.
  • Enteric coatings may be applied in a tablet coating pan, and coatings that are used for delayed release beads can be used for tablets to form delayed release tablets.
  • the amount of coating polymer on the tablet can vary from 5 to 60% (weight gain) depending on the size, shape and surface properties of the tablet.
  • a sub-coat can be applied to the tablets to separate linaclotide from the enteric or functional coat.
  • the granulation solution may be prepared by dissolving PVP, histidine and calcium chloride in water, adjusting solution pH to 2, and dissolving linaclotide. Granulation is performed in a fluid bed by spraying the granulation solution onto filler isomalt. At the end of granulation, dry the granules for 30 min. The granules are then blended with tablet components including isomalt, crospovidone, Mg stearate and talc until uniform, and compressed into tablets.
  • linaclotide core tablets are placed into a pan coater and warmed up to 35°C. Start tablets coating with Eudragit® FS 30 D suspension, keep the product temperature at 28° to 32°C, and atomization air pressure at 3 bar. At the end of coating, discharge the tablets and place them into a circulated air oven and dry for 2 h at 40°C.
  • enteric coatings such as Eudragit® L, S, ethyl cellulose, HPMCAS, PVAP, CAP, CAS, etc. may also be applied to form delayed release tablets at various weight gains.
  • Delayed release compositions comprising linaclotide
  • Delayed release capsules comprising linaclotide may be formulated to target the ileum or colon (e.g., the ileum, late ileum, and/or ascending colon).
  • the composition is formulated to include a pH triggered release based on enteric coating of a linaclotide tablet, capsule or linaclotide coated beads contained in a hard gelatin capsule.
  • the composition may be formulated to further comprise stabilizing additives such as a divalent cation and an amino acid.
  • PVA can be used as binder as well as protective layer in between linaclotide and enteric coating.
  • Linaclotide or linaclotide with PVA overcoat may be coated with an additional enteric coating (e.g. Eudragit® FS30D, Eudragit® SI 00, Eudragit® L100, Eudragit®L100-55, Eudragit® L 30D-55) that dissolves in a pH dependent manner to release at the appropriate pH of 7 in the ileum of the GI tract.
  • the enteric coatings may consist of blends combining different types of Eudragit® - Eudragit® SI 00 / Eudragit® LI 00 in different ratios (e.g.
  • compositions may further comprise other excipients including plasticizing agents such as triethylcitrate.
  • the coatings may further comprising disintegrants as suspended solid to expedite the relevant pH triggered release - resulting in mixed systems as croscarmellose sodium / Eudragit® S.
  • anti-tacking agent e.g., talc, Aerosil® 200 or PlasAcrylTM
  • talc talc
  • Aerosil® 200 or PlasAcrylTM may be used to prevent the beads from sticking.
  • two Eudragit® coatings may be applied to ensure swift release once the desired pH region in the GI tract is reached - including partially neutralized coating systems.
  • Buffering agents such as potassium hydrogen phosphate can be included into one of the two Eudragit® films.
  • Alternative non-Eudragit® pH dependent film coatings include hydroxypropyhnethylcellulose acetate succinate (HPMCAS, e.g. Aqoat® AS-HF), cellulose acetate phthalate (CAP, e.g. Aquateric®) or shellac.
  • Linaclotide, immidazolidinone degradant product (“Cys'-IMD”), and oc-ketone degradant product (“Cys'-oc-Ketone”) can be measured and purified as described in US 2010/0048489, US 2013/0190238, and US 2015/0094272 which are incorporated by reference herein.
  • content and purity of linaclotide may be determined by reverse phase gradient liquid chromatography using an Agilent Series 1100 LC System with Chemstation Rev A.09.03 software or equivalent.
  • a YMC ProTM CI 8 column (dimensions: 3.0 x 150 mm, 3.5 um, 120 A; Waters Corp., Milford, MA) or equivalent is used and is maintained at 40°C.
  • Mobile phase A (MP A) consists of water with 0.1% trifluoroacetic acid while mobile phase B (MPB) consists of 95% acetonitrile:5% water with 0.1% trifluoroacetic acid.
  • Elution of the linaclotide is accomplished with a gradient from 0% to 47% MPB in 28 minutes followed by a ramp to 100% MPB in 4 minutes with a 5 minute hold at 100% MPB to wash the column.
  • Re- equilibration of the column is performed by returning to 0% MPB in 1 minute followed by a 10 minute hold at 100% MPA.
  • the flow rate is 0.6 mL/min and detection is accomplished by UV at 220 nm.
  • Linaclotide release from the coated tablets can be assessed by dissolution testing using a USP XXIV type ⁇ paddle dissolution apparatus (model PTWS, Pharma Test, Hamburg, Germany). The tests are conducted in triplicates, at a paddle speed of 100 rpm in 900 ml dissolution medium maintained at 37.0 ⁇ 0.5°C. Tablets are tested first in 0.1 N hydrochloric acid (pH 1.2) for 30 minutes or 2 hours to simulate gastric residence and then for 6 hours in buffer media of varying pH and ionic composition, akin to small intestinal pH conditions.
  • pH 1.2 hydrochloric acid
  • Linaclotide release may be assessed at pH 6.8-7.4 in two compendial buffer media: 0.067 M mixed sodium and potassium phosphate (Sorensen's) buffer and 0.05 M potassium phosphate buffer as well as in a pH 7.4 multi-electrolyte salt solution (Hanks) buffer which is similar in ionic composition to intestinal fluid. All the buffers are freshly prepared with de-ionized water and de-aerated by sparging with helium prior to use. The amount of linaclotide released from the dosage form can be determined by reverse phase gradient liquid chromatography as described in Example 4.
  • Samplel 81 -64B contains a 100 ⁇ g dose of linaclotide, a subcoat of Opadry® II, and a functional coat of Eudragit® FS 30D and talc.
  • Sample 181-64C contains a 100 ⁇ g dose of linaclotide, a subcoat of Opadry II, and a functional coat of Eudragit® FS
  • Delayed release tablets may be prepared by first preparing the following core tablet components: a placebo base, a linaclotide 750 ⁇ g/225 mg base, and pre-granulated fillers.
  • the tablet components may be prepared into separate granulations for blending before tablet compression.
  • Use of separate tablet components, such as, the placebo base and pregranulated filler base provided, among other things, advantageous properties for stability and release profiles for the tablets.
  • all the tablets components listed in Table 2 could be separately prepared by wet granulation and blended before compression or blended together and processed as a mixture for wet granulation.
  • the tablets components listed in Table 2 could be separately prepared by dry granulation and blended before compression or blended together and processed as a mixture for dry granulation.
  • the tablet components are direct blended for compression.
  • the pregranulated filler base and/or placebo base are prepared through wet granulation and dried before mixing with the 750 ⁇ g/225 mg linaclotide base.
  • the linaclotide base could be prepared by wet granulation processes or by Wurster coating process. This preferred process, exhibited further gains in stability for the tablet by reducing moisture exposure to linaclotide during processing and minimizing residue moisture in the tablet core.
  • Table 3 represents the formulation for the placebo base granulation:
  • Table 3 Formulation of the placebo base granulation
  • the placebo base preparation may be prepared by first dispensing the raw materials of Table 4.
  • Impeller speed 1 (290 rpm, 5.5m/s tip speed), Chopper speed 1(1760 rpm). Tare a poly bag and discharge the completed wet granulation into it. Weigh the granulation. Transfer the wet granulation to the FLM-3 fluid bed for drying. Dry the granulation using the following approximate settings. Dry until the granulation LOD is no more than 1.2% moisture. Discharge the dried granulation into a tared poly bag.
  • Settings are suggested settings only and may be adjusted for optimum drying.
  • Table 5 represents the formulation for the 750 ⁇ g/225mg base granulation:
  • the 750 ⁇ g 225mg base granulation may be prepared by first dispensing the raw materials of Table 6.
  • Table 7 represents the formulation for the pregranulated fillers.
  • the fillers preparation may be prepared by first dispensing the raw materials of Table 8.
  • Table 8 Raw materials for preparation of fillers granulation
  • Impeller speed 1 (290 rpm, 5.5m/s tip speed), Chopper speed 1(1760 rpm).
  • Settings are suggested settings only and may be adjusted for optimum drying.
  • a 25 ⁇ g dose tablet of linaclotide may be prepared with the formulation of Table 9.
  • Preblend Set up an 8 qt blender and add the 750 ⁇ /225 ⁇ 3 ⁇ 4 Base and Placebo Base. Close lids and blend for 10 minutes. Tare a poly bag, and discharge the preblend into it.
  • Sub-Blend A Add 650g of the preblend to a 16 qt v-blender. Add the Pregranulated Fillers for sub-blend A to the 16 qt v-blender. Close lids and blend for 10 minutes. Pass the Magnesium Stearate for sub-blend A through the 40 mesh screen. Add to the 16 qt v-blender. Close lids and blend for 3 minutes. Tare a poly bag, and discharge the blend into it.
  • Sub-Blend B Add 650g of the preblend to a 16 qt v-blender. Add the Pregranulated Fillers for sub-blend B to the 16 qt v-blender. Close lids and blend for 10 minutes. Pass the Magnesium Stearate for sub-blend B through the 40 mesh screen. Add to the 16 qt v-blender. Close lids and blend for 3 minutes. Tare a poly bag, and discharge the blend into it.
  • [000228] Charge the blend into the hopper. Set the turret speed to an appropriate speed. Adjust the die fill amount and compression parameters to yield a tablet with a target weight of 225 mg and with a target hardness of 8-12 kP. All waste material should be collected in the tableting waste poly bag. Start timer and compress tablets.
  • a 10( ⁇ g dose tablet of linaclotide may be prepared with the formulation of Table 12.
  • Preblend Set up an 16 qt blender and add the 750 ⁇ g/225mg Base and Placebo Base. Close lids and blend for 10 minutes. Tare a poly bag, and discharge the preblend into it.
  • [000234] Charge the blend into the hopper. Set the turret speed to an appropriate speed. Adjust the die fill amount and compression parameters to yield a tablet with a target weight of 225 mg and with a target hardness of 8-12 kP. All waste material should be collected in the tableting waste poly bag. Start timer and compress tablets.
  • a 2 ⁇ g dose tablet of linaclotide may be prepared with the formulation of Table 15.
  • Sub-blend A Add the Pregranulated Fillers and Linaclotide base for sub-blend A to a 16 qt v-blender. Close lids and blend for 10 minutes. Pass the Magnesium Stearate for sub-blend A through the 40 mesh screen. Add to the 16 qt v-blender. Close lids and blend for 3 minutes. Tare a poly bag, and discharge the blend into it.
  • Sub-blend B Add the Pregranulated Fillers and Linaclotide Base for sub-blend B to a 16 qt v-blender. Close lids and blend for 10 minutes. Pass the Magnesium Stearate for sub-blend B through the 40 mesh screen. Add to the 16 qt v-blender. Close lids and blend for 3 minutes. Tare a poly bag, and discharge the blend into it.
  • [000240] Charge the blend into the hopper. Set the turret speed to an appropriate speed. Adjust the die fill amount and compression parameters to yield a tablet with a target weight of 225 mg and with a target hardness of 8-12 kP. All waste material should be collected in the tableting waste poly bag. Start timer and compress tablets.
  • Example 8 The 25 ⁇ g tablets of Example 8 may be sub-coated with an Opadry® II sub- coating.
  • the formula of Table 18 represents the amounts needed to prepare the coating material, which is prepared in excess to account for losses in the coating process.
  • Table 18 Sub-Coating formula for 25 ⁇ g tablets
  • Add Opadry® to the water. Calculate the theoretical amount of solution needed to apply a 4.0% weight gain, with 85% theoretical efficiency.
  • the spray gun assembly should consist of 1 x 1 ⁇ 4 JAU spray gun mounted with a 40100 AB liquid spray nozzle and matching air cap.
  • the gun assembly should be mounted as far as possible from the tablet bed, with the spray angle perpendicular to the top third of the bed. Verify initial tablet weight by weighing 100 uncoated tablets. Calculate the average weight of a single tablet by dividing the weight of the tablets by 100. Test the initial tablet moisture by crashing approximately 1 gram of tablets and running LOD for 10 minutes at 105°C. Adjust the pump so that the liquid flow rate is approximately 10 g/min. Prime the lines past the guns and verify that there is no leaking in the lines or gun. Charge the tablets into the coating pan and begin warming for 20 minutes with an inlet temperature of 50°C and airflow of 350 CFM. Jog occasionally during warm-up.
  • target bed temperature begins spraying the coating suspension according to the target process parameters outlined in Table 19 below. Test and report tablet moisture content periodically as desired. Once the theoretical amount of solution has been applied check the tablets for weight gain. When 4% weight gain has been achieved, stop spray and dry tablets for 30 minutes with an inlet temperature of 50°C, reducing pan speed to a minimum or jogging. Discharge the tablets and determine the new weight of the coated tablets.
  • Example 9 The 100 ⁇ g tablets of Example 9 may be sub-coated with an Opadry® ⁇ coating.
  • the formula of Table 20 represents the amounts needed to prepare the coating material, which is prepared in excess to account for losses in the coating process.
  • Add Opadry® to the water. Calculate the theoretical amount of solution needed to apply a 4.0% weight gain, with 85% theoretical efficiency.
  • the Compu-Lab has been set up with the 24 inch pan and plenum assembly.
  • Verify the liquid feed lines are Tygon 17 tubing.
  • Verify the gun assembly is installed in the pan.
  • the spray gun assembly should consist of 2 x 1 ⁇ 4 JAU spray gun mounted with a 40100 AB liquid spray nozzle and matching air cap.
  • the gun assembly should be mounted as far as possible from the tablet bed, with the spray angle perpendicular to the top third of the bed.
  • target bed temperature Once target bed temperature is reached, begin spraying the coating suspension according to the target process parameters outlined in Table 21 below. Test and report tablet moisture content periodically as desired. Once the theoretical amount of solution has been applied check the tablets for weight gain. When 4% weight gain has been achieved, stop spray and dry tablets for 30 minutes with an inlet temperature of 50°C, reducing pan speed to a minimum or jogging. Discharge the tablets and determine the new weight of the coated tablets.
  • the 2i ⁇ g tablets of Example 10 may be sub-coated with an Opadry® II coating.
  • the formula of Table 22 represents the amounts needed to prepare the coating material, which is prepared in excess to account for losses in the coating process.
  • Add Opadry® to the water. Calculate the theoretical amount of solution needed to apply a 4.0% weight gain, with 85% theoretical efficiency.
  • the Compu-Lab has been set up with the 19 inch pan and plenum assembly.
  • Verify the liquid feed lines are Tygon 17 tubing.
  • Verify the gun assembly is installed in the pan.
  • the spray gun assembly should consist of 1 x 1 ⁇ 2 JAU spray gun mounted with a 40100 AB liquid spray nozzle and matching air cap.
  • the gun assembly should be mounted as far as possible from the tablet bed, with the spray angle perpendicular to the top third of the bed.
  • target bed temperature Once target bed temperature is reached, begin spraying the coating suspension according to the target process parameters outlined in Table 23 below. Test and report tablet moisture content periodically as desired. Once the theoretical amount of solution has been applied check the tablets for weight gain. When 4% weight gain has been achieved, stop spray and dry tablets for 30 minutes with an inlet temperature of 50°C, reducing pan speed to a minimum or jogging. Discharge the tablets and determine the new weight of the coated tablets.
  • the tablets of previous examples may be prepared with a functional coating.
  • the formulation of Table 24 represents the amounts needed to prepare the coating material, which is prepared in excess to account for losses in the coating process:
  • Dispense the required quantity of Talc into a suitable sized container Homogenize the talc in the remaining 1/3 of purified water for 10 minutes (or until homogenous) using a Silverson Homogenizer. Pour the talc suspension into the Eudragit® suspension while mixing. Mix for no longer than 5 minutes. Screen the coating suspension through a #30 mesh screen.
  • target bed temperature Once target bed temperature is reached, begin spraying the coating suspension according to the target process parameters outlined in Table 25 below. Test and report tablet moisture content periodically as desired. Once the theoretical amount of solution has been applied check the tablets for weight gain. When 9% weight gain has been achieved, stop spray and dry tablets for 5-10 minutes with an inlet temperature of 40°C, reducing pan speed to a minimum or jogging.
  • An alternative coating may be prepared according to the method of Example 14 but with the formulation of Table 26.
  • the coating of this Example 15 is used as a coating for the DR2 delayed release compositions discussed herein.
  • An organic coating may be provided for the linaclotide tablets of the examples above.
  • the formula of Table 27 was used for the coating of 100 ⁇ g tablets.
  • Table 27 Organic coating material formula for linaclotide tablets.
  • Dispense the required quantity of purified water into a suitable sized container Dispense the required quantity of acetone into a suitable sized container. Begin mixing acetone and add the water. Dispense the required quantity of isopropanol into a suitable sized container. Add isopropanol to the water and acetone to create the diluent mixture. Pour approximately half of the diluent mixture into a second container and resume mixing the first half of the diluent mixture. Dispense the required quantity of Eudragit SI 00 into a suitable sized container. Begin mixing the second half of diluent mixture and add the Eudragit SI 00. Dispense the required quantity of Eudragit LI 00 into a suitable sized container.
  • Delayed release tablets were prepared by first preparing the following core tablet components: a placebo base, a linaclotide 1000 ⁇ g/225 mg base, and pre-granulated fillers.
  • the tablet components were prepared essentially as described above in Example 7, but with slight modifications as described below.
  • the placebo base as described in Table 28 was used to provide core tablets with the components listed in Table 28.
  • a final pH of 2.25 was used for placebo and active base granulations.
  • Table 28 Components for various tablet strengths
  • Table 29 represents the formulation for the placebo base granulation:
  • Table 29 Formulation of the placebo base granulation
  • the placebo base preparation was prepared by first dispensing the raw materials of Table 30.
  • the mix container is tared and 2200 ⁇ 5.0 g of treated water (rather than 2682.1 ⁇ 5.0 g) is added into the container.
  • Table 31 represents the formulation for the 1000 ⁇ g 225mg base granulation: Table 31: Formulation for the 1000 ⁇ g 225mg base granulation
  • the 100( ⁇ g/225mg base granulation may be prepared by first dispensing the raw materials of Table 32.
  • Table 32 Raw materials of the linaclotide base granulation
  • Table 33 represents the formulation for the pregranulated fillers.
  • the fillers preparation are prepared by first dispensing the raw materials of Table 34.
  • Delayed release 225 mg tablets can be prepared with the proportions of excipients as shown in Table 34 and Table 35.
  • the 225 mg tablets of this Example 18 can be prepared at dosage strengths of 30 ⁇ & 100 ⁇ 3 ⁇ 4 or 300 ⁇ g.
  • Two example embodiments, A and B, of the proportions of excipients are provided for each dosage strength.
  • Embodiment A (at each dosage) in Table 34 corresponds to the DR1 formulation in Example 20; embodiment A (at each dosage) in Table 35 corresponds to the DR2 formulation in Example 20.
  • the tablets include a tablet core, a barrier coat, a functional coat, and, in some cases, an aesthetic coat.
  • e Final Opadry may be either Blue 85F99031, Yellow 85F120017, or Orange 85F130136
  • Delayed release 450 mg tablets can be prepared with the proportions of excipients as shown in Table 36 and Table 37.
  • the 450 mg tablets of this Example 19 can be prepared at dosage strengths of 300 or 600 ⁇ g.
  • the tablets include a tablet core, a barrier coat, a functional coat, and an aesthetic coat.
  • e Final Opadry may be either Blue 85F99031, Yellow 85F120017, or Orange 85F130136
  • Purified Water __b massageb a The actual quantity of linaclotide drug su bstance is adjusted based on the purity of the drug substance with a concomitant adjustment of microcrystalline cellulose
  • Linaclotide for the Treatment of Irritable Bowel Syndrome with Constipation (IBS-c)
  • Delayed release linaclotide tablets were administered in a Phase 2b, randomized, double-blind, double-dummy, placebo-controlled, parallel-group, dose-range- finding study of two delayed release formulations ("DR1" and "DR2") of linaclotide administered orally for 12 weeks to patients with IBS-c (the "Trial”).
  • the Trial was a multi-center, multiple-dose study consisting of 3 distinct periods: a screening period, a pretreatment period and a treatment period (see Fig. 1).
  • the study enrolled patients having IBS-c diagnosed using Rome III criteria.
  • Eligible patients were randomized in equal proportions to one of 8 treatments: 30 ⁇ g, 100 ⁇ g, or 300 ⁇ g of the linaclotide DRl formulation; 30 ⁇ 3 ⁇ 4 100 ⁇ & or 300 ⁇ g of the linaclotide DR2 formulation; 290 ⁇ g of the immediate release (IR) formulation of linaclotide; or placebo.
  • Linaclotide IR was included as a positive control to compare safety and efficacy.
  • the DRl delayed release composition was designed to release the linaclotide core tablet in the ileal region.
  • the DR2 delayed release composition was designed to release the linaclotide core tablet within the late ileum to the ascending colon (i.e., at or near the ileocecal junction).
  • a pre-treatment period began.
  • the pre-treatment period was defined as the 14 to 21 calendar days immediately before starting the treatment period.
  • patients provided information regarding daily bowel-movement-related symptom severity, daily abdominal symptom-severity, weekly constipation severity, weekly IBS symptom severity, weekly adequate relief and use of per-protocol rescue medicine or other laxatives, suppositories or enemas on an event-driven basis.
  • Patients who satisfied all of the entry criteria entered the treatment period.
  • the treatment period began with treatment assignment (randomization) and lasted for 12 weeks. Patients were randomly assigned to one of 8 treatments with linaclotide: 30 ⁇ g, 100 ⁇ g, or 300 ⁇ g of the linaclotide DRl formulation; 30 ⁇ g, 100 ⁇ 3 ⁇ 4 or 300 ⁇ g of the linaclotide DR2 formulation; 290 ⁇ g of the IR formulation; or placebo. Patients were instructed to take the study drug once daily in the morning at least 30 minutes before breakfast.
  • APC abdominal pain and constipation
  • a patient was classified as a 6/12 week abdominal pain and constipation (APC) +1 responder if they were a weekly APC +1 responder for > 6 out of the 12 weeks of the treatment period.
  • a weekly APC +1 responder was defined as a patient who meets both the criteria to be a weekly CSBM +1 responder and a weekly abdominal pain responder in a given week.
  • a patient was classified as a 6/12 week CSBM +1 responder if they were a weekly CSBM +1 responder for > 6 out of the 12 weeks of the treatment period.
  • a weekly CSBM +1 responder was defined as a patient who had an increase from baseline of at least 1 in the CSBM weekly rate for a given week.
  • a patient was classified as a 6/12 week abdominal pain responder if they were a weekly abdominal pain responder for > 6 out of the 12 weeks of the treatment period.
  • a weekly abdominal pain responder was defined as a patient who had a decrease from baseline of at least 30% in the mean abdominal pain score for a given week.
  • the results of the Trial demonstrate that administration of the DR1 composition has efficacy across the primary and secondary efficacy parameters.
  • the results of the Trial also demonstrate that administration of the DR2 compositions had no effect on constipation as shown by CSBM and BSFS data (e.g., Figures 5 and 11); however administration of the DR2 compositions did improve symptoms of abdominal pain in the patients (with very low incidence of adverse events), suggesting that the DR2 composition is useful for reducing abdominal symptoms (pain, discomfort, bloating) in non-constipated patients.
  • Figure 2 shows the 6/12 Week APC +1 parameter results.
  • the 300 ⁇ g DR1 composition showed improvement over the comparable IR composition (38.8% vs. 31.8%).
  • Other DR1 and DR2 compositions showed improvement over placebo.
  • Figure 3 shows the weekly change from baseline in SBM frequency.
  • Figure 4 shows the weekly change from baseline in abdominal pain for each week over the 12 week treatment period. Again the 300 ⁇ g DRl composition outperformed the comparable IR composition and other DR compositions outperformed placebo. In addition, the 30 ⁇ g DR2 composition showed a change in abdominal pain levels comparable to the IR composition.
  • Figures 5-13 show the trial results for secondary efficacy parameters, demonstrating the overall improvement in patient outcomes for the delayed release compositions.
  • the Bristol Stool Form Scale has been found to be one of the most reliable and consistent endpoints as a measure of efficacy in IBS-c trials.
  • DRl appears to show dose dependent improvement in BSFS scores.
  • Figure 17 shows selected efficacy parameter results with comparison between the 300 g DRl composition, the 290 ⁇ g IR composition from the Trial, placebo, and the 290 ⁇ g IR composition from the Phase III trial for Linzess® and the placebo from the Linzess® Phase III trial.
  • the DRl composition results compare favorably to the results from the IR composition of the Linzess® Phase III trial.
  • Figure 21 is a plot of the percent change from baseline in weekly abdominal pain at week 12 for the DRl composition, the IR composition and placebo.
  • compositions had lower rates of diarrhea (overall and ADO) compared to the IR composition group, including lower rates of moderate to severe diarrhea (Fig. 16).
  • Figures 18 and 19 show the diarrhea rates with comparison to the Linzess® Phase ⁇ Trial, also showing improvement in adverse diarrhea events for the 300 ⁇ g DRl composition compared to the Linzess® Phase III results.
  • a food effect study with the delayed release compositions of the present invention was undertaken to determine the effect of taking the delayed release compositions on a full stomach (with food) compared to an empty stomach (after fasting).
  • Patients were separated into two groups.
  • Patients in group 1 were given a 300 ⁇ g dose of DRl or DR2 delayed release composition of linaclotide under fed conditions. Specifically, the patients in group 1 were given the composition 30 minutes after the start of a high-fat, high-calorie breakfast with 8 ounces of water.
  • Patients in group 2 were given the same composition (DRl or DR2) but with no food under fasted conditions. Specifically, the patients in group 2 were given the composition after an overnight fast of at least 10 hours with approximately 8 ounces of water. In both groups, no food was consumed for 4 hours after taking the medication. After a seven day washout period, the patients in group 1 were then given the composition under fasted conditions and the patients in group 2 were given the compositions under fed conditions.
  • DRl composition under fasted conditions had no occurrences of diarrhea.
  • Some patients who took the DRl composition under fed conditions had occurrences of diarrhea.
  • the patients who took the DR2 composition had no occurrences of diarrhea under fed or fasted conditions.
  • the DRl and DR2 formulations which are designed to release in the lower GI as compared with the upper GI release in the immediate release formulation, significantly reduces the incidence of diarrhea to the extent where, in the case of the DRl formulation, diarrhea is only observed in fed subjects, and in DR2, no diarrhea was observed in either subject group. Therefore, the site of drug release plays a dramatic role in modulating adverse events such as diarrhea.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des procédés de traitement d'un patient souffrant d'un trouble, tel qu'un trouble gastro-intestinal (GI) ou des symptômes associés à un trouble GI ou non GI, par administration d'une quantité thérapeutiquement efficace d'une composition pharmaceutique à libération retardée comprenant du linaclotide.
EP17833050.2A 2016-12-21 2017-12-21 Procédés de traitement du syndrome du côlon irritable avec des formulations à libération modifiée ou retardée de linaclotide Pending EP3558337A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662437566P 2016-12-21 2016-12-21
PCT/US2017/067814 WO2018119191A1 (fr) 2016-12-21 2017-12-21 Procédés de traitement du syndrome du côlon irritable avec des formulations à libération modifiée ou retardée de linaclotide

Publications (1)

Publication Number Publication Date
EP3558337A1 true EP3558337A1 (fr) 2019-10-30

Family

ID=61022424

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17833050.2A Pending EP3558337A1 (fr) 2016-12-21 2017-12-21 Procédés de traitement du syndrome du côlon irritable avec des formulations à libération modifiée ou retardée de linaclotide

Country Status (10)

Country Link
EP (1) EP3558337A1 (fr)
JP (2) JP2020512292A (fr)
CN (1) CN110475564A (fr)
AU (1) AU2017379082A1 (fr)
BR (1) BR112019012689A2 (fr)
CA (1) CA3048195A1 (fr)
EA (1) EA201991531A1 (fr)
MA (1) MA47112A (fr)
MX (2) MX2019007574A (fr)
WO (1) WO2018119191A1 (fr)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022536494A (ja) * 2019-06-10 2022-08-17 アイアンウッド ファーマシューティカルズ, インコーポレイテッド 下痢型過敏性腸症候群に関連する腹痛の処置
WO2023097309A1 (fr) * 2021-11-29 2023-06-01 Ironwood Pharmaceuticals, Inc. Compositions pharmaceutiques pour le traitement de la douleur viscérale
WO2023196821A2 (fr) * 2022-04-04 2023-10-12 Thomas Jefferson University Ciblage de gucy2c de cellule neuropode pour commander la douleur viscérale et l'appétit

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7304036B2 (en) 2003-01-28 2007-12-04 Microbia, Inc. Methods and compositions for the treatment of gastrointestinal disorders
US7371727B2 (en) 2003-01-28 2008-05-13 Microbia, Inc. Methods and compositions for the treatment of gastrointestinal disorders
MX2020005326A (es) 2008-08-15 2022-03-03 Ironwood Pharmaceuticals Inc Formulaciones que contienen linaclotida para administracion oral.
EP2464373A1 (fr) * 2009-08-13 2012-06-20 Ironwood Pharmaceuticals, Inc. Procédé de modulation de l'effet pharmacodynamique d'agonistes des récepteurs de guanylate cyclase administrés par voie orale
US8933030B2 (en) 2010-02-17 2015-01-13 Ironwwod Pharmaceuticals, Inc. Treatments for gastrointestinal disorders
ES2919136T3 (es) * 2010-08-11 2022-07-22 Ironwood Pharmaceuticals Inc Formulaciones estables de linaclotida
US9708371B2 (en) 2011-08-17 2017-07-18 Ironwood Pharmaceuticals, Inc. Treatments for gastrointestinal disorders
AU2014207680B2 (en) * 2013-01-15 2018-09-06 Ironwood Pharmaceuticals, Inc. Gastro-retentive sustained-release oral dosage form of a bile acid sequestrant
UA119335C2 (uk) * 2013-12-11 2019-06-10 Айронвуд Фармасьютикалз, Інк. Композиції лінаклотиду з затриманим вивільненням
WO2016197042A1 (fr) * 2015-06-05 2016-12-08 Ironwood Pharmaceuticals, Inc. Formulations à libération modifiée ou ciblée de linaclotide

Also Published As

Publication number Publication date
WO2018119191A1 (fr) 2018-06-28
JP2020512292A (ja) 2020-04-23
CN110475564A (zh) 2019-11-19
MX2019007574A (es) 2019-09-04
JP2022093472A (ja) 2022-06-23
AU2017379082A1 (en) 2019-07-04
MX2023008839A (es) 2023-08-14
MA47112A (fr) 2019-10-30
BR112019012689A2 (pt) 2019-11-19
CA3048195A1 (fr) 2018-06-28
EA201991531A1 (ru) 2019-11-29
WO2018119191A9 (fr) 2018-10-18

Similar Documents

Publication Publication Date Title
AU2020202319B2 (en) Delayed release compositions of linaclotide
JP2022093472A (ja) リナクロチドの改変放出製剤または遅延放出製剤による過敏性腸症候群の処置方法
US20220202725A1 (en) Modified or Targeted Release Formulations of Linaclotide
US20230346878A1 (en) Treatment of Abdominal Pain Associated with Diarrhea-Predominant Irritable Bowel Syndrome
US20230142270A1 (en) Delayed Release Compositions of Linaclotide
BR112016013419B1 (pt) Composições de liberação retardada compreendendo um comprimido com revestimento entérico, forma de dosagem unitária e uso

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190716

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40017060

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20201014

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230522