WO2023283427A2 - Enteric and delayed release formulations of locally bioavailable drugs (lbd) targeting the colon - Google Patents

Enteric and delayed release formulations of locally bioavailable drugs (lbd) targeting the colon Download PDF

Info

Publication number
WO2023283427A2
WO2023283427A2 PCT/US2022/036519 US2022036519W WO2023283427A2 WO 2023283427 A2 WO2023283427 A2 WO 2023283427A2 US 2022036519 W US2022036519 W US 2022036519W WO 2023283427 A2 WO2023283427 A2 WO 2023283427A2
Authority
WO
WIPO (PCT)
Prior art keywords
therapeutic agent
pharmaceutical composition
release
drug
colon
Prior art date
Application number
PCT/US2022/036519
Other languages
French (fr)
Other versions
WO2023283427A3 (en
Inventor
Katherine SHATZER
Original Assignee
Verdure Biotech, 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 Verdure Biotech, Inc. filed Critical Verdure Biotech, Inc.
Publication of WO2023283427A2 publication Critical patent/WO2023283427A2/en
Publication of WO2023283427A3 publication Critical patent/WO2023283427A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4891Coated capsules; Multilayered drug free capsule shells

Definitions

  • Familial Adenomatous Polyposis is a rare, inherited disorder resulting from a germline mutation in the adenomatous polyposis (APC) gene and characterized by colorectal cancer (Half E et al. Orphanet J Rare Dis. 2009;4:22; Patel SK et al. PLoS One. 2013;8(2):e55802).
  • Colectomy is the most common treatment and is typically performed shortly after the time of diagnosis.
  • Adenomatous polyps appear by about 15 years of age in 50% of people with FAP, and by 35 years of age in 95%. Seventy percent of these individuals have colorectal malignancy by the age of 36 years, the average age of symptomatic diagnosis.
  • Familial Adenomatous Polyposis is a debilitating condition that has a long term, severe impact on the physical and psychological health, independence, and quality of life of an affected patient.
  • the affected patient develops hundreds to thousands of intestinal polyps, many of which become cancerous unless surgically removed. Most patients are asymptomatic for years until the adenomas are large and numerous, resulting in rectal bleeding or even anemia, or cancer develops.
  • Nonspecific symptoms may include, but are not limited to, constipation or diarrhea, abdominal pain, palpable abdominal masses, and weight loss.
  • FAP is broken down into two sub conditions based upon severity, classic and attenuated. Classic is the more severe form of FAP.
  • FAP is inherited in an autosomal dominant manner and results from a germline mutation in the adenomatous polyposis (APC) gene. Most patients, approximately 70%, have a family history of colorectal polyps and cancer (Half E et al. Orphanet J Rare Dis. 2009;4:22).
  • Patients suffering from FAP will present with some extraintestinal manifestations that can include, but are not limited to, osteomas, dental abnormalities (e.g., unerupted teeth, congenital absence of one or more teeth, supernumerary teeth, dentigerous cysts and odontomas), congenital hypertrophy of the retinal pigment epithelium, desmoid tumors, and extracolonic cancers (e.g., duodenum, stomach, skin, thyroid, liver, bile ducts and central nervous system).
  • the extracolonic cancers may be benign or malignant.
  • Desmoid tumors are of particular significance; these are fibrous tumors usually occurring in the tissue covering the intestines and may be provoked by surgery to remove the colon.
  • Desmoid tumors are likely to recur after they have been surgically removed.
  • Mutations in the APC gene cause both classic and attenuated familial adenomatous polyposis (Half E et al. Orphanet J Rare Dis. 2009;4:22). The mutations affect the affected cell’s ability to maintain normal growth and function. Cell overgrowth resulting from mutations in the APC gene leads to the colon polyps seen in FAP. Although most people with mutations in the APC gene will develop colorectal cancer, the number of polyps and the time frame in which they become malignant depend on the location of the mutation in the gene (Attard TM et al.
  • Familial Adenomatous Polyposis is a rare, inherited condition with a reported incidence varying from 1 in 6,000 to 1 in 22,000 individuals. FAP manifests equally in both sexes and is the second most common genetic Colorectal Cancer (CRC) syndrome. CRC is the third leading cause of cancer deaths in the United States (Kinney AY et al.
  • CRC cancer associated morbidity and mortality. Its incidence varies considerably among different populations, with the highest incidence reported from Western and industrialized countries. Worldwide, about 85% of CRCs are considered to be sporadic, while approximately 15% are familial with FAP accounting for less than 1% (Half E et al. Orphanet J Rare Dis. 2009;4:22). Clinically, FAP manifests equally in both sexes by the late teens and in the twenties age group. Individuals with FAP carry a 100% risk of CRC (Half E et al. Orphanet J Rare Dis.2009;4:22; Kinney AY et al.
  • FAP Familial Adenomatous Polyposis
  • Non-steroidal anti-inflammatory drugs are perhaps the most versatile group of drugs prescribed for inflammation, analgesic/antipyretic, and auto-immune diseases (9, 114).
  • Non-selective NSAIDs inhibit the synthesis of prostaglandins, thromboxane, and levuloglandins by blocking arachidonic acid substrates from binding to the cyclooxygenase isozymes (10, 11) Cyclooxygenase-1 (COX-1) and Cyclooxygenase-2 (COX-2).
  • COX-1 performs a distinct physiological homeostatic maintenance role within the vascular system and gastrointestinal tract; its inhibition may lead to gastrointestinal toxicity, cardiovascular toxicity, internal bleeding, and stomach ulcers.
  • the COX-2 enzyme is the inducible, pro-inflammatory cyclooxygenase isoform; it is overexpressed and upregulated in many cancer tissues (116). COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (117) and is used as a prominent predictive marker in inflammatory bowel diseases (118).
  • COXIBs Selective COX-2 inhibitors
  • the COX-2 enzyme which is the inducible, pro-inflammatory cyclooxygenase isoform, is observed to be overexpressed and upregulated in FAP and CRC tissues. COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (6) and is used as a prominent predictive marker in inflammatory bowel diseases. [0015] The COX-2 enzyme was observed to be up-regulated, overly expressed in the polyposis of FAP patients, and to attenuate the progression of these polyposis lesions in the colon. If FAP lesions are treated at an early stage, not only can the FAP patients’ morbidity and mortality rates decline, but their quality of life can be improved (Half E et al.
  • COXIBs selective COX-2 inhibitors
  • NSAIDs nonsteroidal anti- inflammatory drug
  • COXIBs cannot be used clinically to treat FAP.
  • the inability to avoid or prevent these harsh side effects ultimately forced the withdrawal of the drug (i.e., rofecoxib) (Higuchi T et al. Clin Cancer Res. 2003;9(13):4756-60; Kerr DJ et al. N Engl J Med.2007;357(4):360-9) or drug indication (i.e., celecoxib) for FAP from the market (Baron JA et al.
  • COXIBs as a Treatment for Management of Familial Adenomatous Polyposis (FAP)
  • COXIBs e.g., celecoxib, rofecoxib
  • NSAIDs nonsteroidal anti-inflammatory drug
  • COX-2 cyclooxygenase-2
  • COXIBs cannot be used clinically to treat FAP.
  • the inability to avoid or prevent these harsh side effects ultimately forced the withdrawal of the drug (i.e., rofecoxib) (Higuchi T et al. Clin Cancer Res. 2003;9(13):4756-60; Kerr DJ et al. N Engl J Med. 2007;357(4):360-9) or drug indication (i.e., celecoxib) for FAP from the market (Baron JA et al. Lancet.
  • COX-2 is an inducible form of the myeloperoxidases enzyme family that is located on chromosome 1 and found on the nuclear membrane or the luminal side of the endoplasmic reticulum (Chandrasekharan NVS et al. Genome Biology. 2004;5(9):241).
  • Prostaglandin G2 is one of the downstream products catalyzed by COX-2 from plasma membrane arachidonic acid.
  • the prostaglandins are responsible for vasodilation (PGD2, PGE2, PGI2), gastric, renal, platelet aggregation homeostasis (PGI2), mediating fever, pain, and inflammation (PGE2).
  • PGE2 modulates different functions via its binding ability to specific EP receptors.
  • PGE2 to EP1 receptors led to the mobilization of intracellular calcium. Binding of PGE2 to EP2 and EP4 led to the deregulation of the cell proliferation pathway.
  • EP2 and EP4 receptors are coupled to the G protein to activate adenylate cyclase, which increased the intracellular cAMP; intracellular cAMP activates PKA, PI3K, and GSK3 protein kinases that ultimately activate beta-catenin (Arico S et al. The Journal of biological chemistry.2002;277(31):27613- 21). The direct effect of PGE2 binding to EP3 receptors is still unclear.
  • Colonic drug delivery is still an attractive opportunity that offers many advantages, such as lowering the effective dose while achieving high drug concentration at the targeted local environments and reducing systemic side effects (Amidon S et al. AAPS PharmSciTech.2015;16(4):731-41).
  • the two main routes of drug delivery to the colon are rectal and oral.
  • the rectal route mostly in the form of a suppository, has the most straightforward requirements for formulation and yields very consistent results.
  • the rectal route is limited on where a drug can be delivered; the drug cannot be delivered to desired sites such as the cecum, ascending colon, or transverse colon.
  • the oral route has been the primary alternative to deliver drugs targeting these proximal sites within the colon.
  • Oral colonic drug delivery systems are effective in delivering acid label, narrow therapeutic index drugs targeted to various locations within the gastric tract, and low solubility drugs with high efficiency.
  • the oral dosage form is aesthetic and convenient, thus has better patient compliance.
  • the oral colonic delivery systems also have many challenges such as a transit time that is variable between individuals, a release mechanism that is affected by the type of diet and amount of food intake, the physiological and integrity differences of normal and disease stage, colonic pH fluctuation, as well as the microbiomes variability between individuals.
  • There are many methods to formulate drugs targeting the colon ( Figure 2) and each has its advantages and disadvantages. Potential methods to target the colon include emulsion, nanoparticles, microparticles, and liposomes.
  • Nanoemulsions and microemulsions are droplets that contain drug molecules dispersed inside oil droplets and suspended in a water medium (o/w) or drug molecules dispersed inside aqueous droplets and suspended in an oil medium (w/o) (McConnell EL et al. International journal of pharmaceutics. 2008;364(2):213-26).
  • emulsion is the bicontinuous emulsion which is composed of both o/w and w/o droplets, thus capable of capturing both hydrophobic and hydrophilic drugs.
  • the nano and micro prefixes describe the droplet size, with the nano prefix ranging from 1 nm - 200 nm.
  • the micro prefix represents a droplet size ranging from one micron and above.
  • the liquid carrier that disperses the drug usually had the same hydrophobicity as the drug.
  • the immiscibility between the water and oil needs to be stabilized by a surfactant (also known as an emulsifier).
  • the surfactant serves as a thermodynamically isotropic interfacial membrane between the two liquid phases, lowers the surface tension, and creates a physical boundary that prevents the droplets from coalescence.
  • Nanoemulsions and microemulsions are approaches used to improve drug bioavailability.
  • a key component for emulsion systems is the drug release mechanism; the release mechanism is controlled through the skillful selection of surfactants and cosurfactants (Guo Y et al. Molecules. 2018;23(7); Patel SK et al. PLoS One. 2013;8(2):e55802; Rao J et al. J Agric Food Chem.2011;59(9):5026-35) that destabilize at the specific target thus allow the release of drugs.
  • Nanoemulsions (Figure 3) are popular for use with the nasal, dermal, and mucosa routes because of their large surface area that results in increased drug absorption and permeation. Nanoemulsions are also thermodynamically and kinetically stable, enabling easy sterilization. Submicron size droplets produce translucent nanoemulsions that are ideal for intravenous formulations. Emulsions require a large amount of surfactant and can sometimes become unstable depending upon the temperature, pH, and ionic strength of the environment. [0027] A group of researchers recently synthesized a dual mode imaging of 19 F magnetic resonance and near-infrared fluorescence capable nanoemulsion.
  • the PGE2 reduction also inversely correlated with the drug’s concentration.
  • the result suggests that the contrasting agents formulated with the nanoemulsion method did not hinder drug permeation into the cell.
  • the emulsion also was stable for more than two months.
  • the standard way of producing nanoparticles (nm) and microparticles ( ⁇ m) is to include a lipid carrier or polymer matrix where the carrier encapsulates the drug.
  • the two common types are solid and lipid particles.
  • compositions and fabrication techniques are employed to increase the drug loading efficiency, achieve the desired pharmacokinetic profiles, and prevent dose dumping.
  • An emerging trend observed is the use of a multi- particulate system where the raw drug and various sized particles are mixed in a controlled proportion as to achieve the desired drug release profile.
  • Carrier materials also influence drug release from these particles (Sharma S et al. Journal of controlled release : official journal of the Controlled Release Society. 2018;272:97-106; Authors M. Nanoemulsions- Nanoarchitectonics for smart delivery and drug targeting 2016-2017).
  • the nanoparticles are usually formulated via some form of strong force such as high ultrasonication, wet-milling, or high-pressure homogenization.
  • Microparticles are usually formulated via a coacervation method which consists of the emulsion solvent evaporation process, spray drying, and solvent extraction-evaporation method.
  • a coacervation method which consists of the emulsion solvent evaporation process, spray drying, and solvent extraction-evaporation method.
  • Poly-lactic-co-glycolic acid (PLGA), pH-sensitive polymers (polymethracrylate, i.e., Eudragit), cellulosic, and solid-phase anchored silica are examples of commonly used carriers for the formulation of particles (McConnell EL et al. J Drug Target. 2009;17(5):335-63).
  • Liposomes are lipid-based carriers that consist of a lipid bilayer that encapsulates an aqueous drug volume (hydrophilic drug) or hydrophobic drug within their lipid bilayer (Kraft JC et al. J Pharm Sci.2014;103(1):29-52).
  • Liposomes have amphoteric properties that enable them to be used as a carrier for both hydrophilic and hydrophobic drugs. Liposomes are shown to accumulate at inflamed tissues and can be enhanced via surface modification and addition of a targeting ligand (Authors C. Liposome: Nano and microscale drug delivery system. Science Direct.). The surface modifications of liposomes are accomplished by physical anchoring and leveraging on the polarity of the chemical moieties or chemically treating the lipids and covalently bonding the moieties to the lipid ( Figure 6) (Pileri P et al. Br J Cancer. 2016;115(1):40-51).
  • Virosome is a small subset of liposomes which are made of a unilamellar phospholipid membrane but contain viral genetic materials (phospholipid and glycoprotein) that facilitate a high amount of binding specificity.
  • This formulation approach is one of a new emerging trend that leverage on the preexisting knowledge coming from the immunogenicity study accumulated over the years through the vaccine industry (Nguyen TXH et al. Nanomedicine.2016;11(9).).
  • each colonic drug delivery system needs to sufficiently address the differences of the gastric transit time, local microbial content, physiological pH, amount of inflamed tissues with altered drug absorption ability, and mucus thickness that changes the surface topography to achieve drug release at the intended site.
  • the current colonic drug delivery systems have several disadvantages. Intestinal physiology [0034] When compared to the small intestine, the colon is much shorter, being only about 1.5 meters in length.
  • the absorptive surface is much less than the small intestine; however, the colon has the longest transit time.
  • the average transit time for material from ingestion to the colon is about five hours.
  • the minimum dwell time in the colon is three to four hours before defecation (43).
  • the optimal COX inhibitor release target time is, therefore around six hours to ensure passage through the stomach yet allow for release prior to defecation.
  • the microbiome on the surface of the colon wall produces a significant amount of bicarbonates, thus neutralizing the acidic content from the stomach and elevating the colon pH to 6.8 and above. Therefore, a successful pH-sensitive, biodegradable coating is expected not to disperse until the coated dosage form reaches the colon where the pH is at least 6.8.
  • a pH-sensitive drug delivery system must withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ⁇ 0.4) or the ileocecal junction.
  • the pH turns slightly basic again due to the fatty acids produced by the resident microbiomes.
  • the mean pH within a healthy colon has been reported to be 6.37 ⁇ 0.58 in the right colon, 6.61 ⁇ 0.83 in the mid-colon, and 7.04 ⁇ 0.67 in the left colon (Evans DFP et al. Gut.1988;29:1035-41).
  • the pH variations can be as high as two pH units (Fallingborg JC et al. Ailment Pharmacol Ther. 1989;3(6)).
  • a methacrylic acid and methyl methacrylate copolymer are the primary polymers of choice when it comes to pH-sensitive formulations (Lamprecht AS et al. Pharmaceutical Research. 2001;18(6):6).
  • Methracyclic-methacrylate copolymer is commercially known as the Eudragit series from Evonik. Eudragit is used in many coatings for particles, hard gelatin capsules, and tablets.
  • a few commercially available Eudragit coated formulations are Asacol®, Ipocol, and Mesren MR (Fadda HM et al. Pharm Res. 2009;26(2):356-60).
  • a pH-sensitive drug delivery system also faces the intra-individual variances resulting from differences in food intake (Ibekwe VC et al.
  • the drug release is generally achieved by erosion, diffusion, or swelling of different matrices over a predetermined period.
  • Time-based approaches work on the assumption that a dosage form will spend approximately six hours in the stomach and small intestine during the fasted state.
  • the time-dependent approach assumes that gastrointestinal transit time is solely influenced by the gastric emptying. Thus, a lag time can be added for the fed state.
  • the erroneous part of this assumption is in the variability of the amount and type of food intake (Weitschies W et al. International journal of pharmaceutics.2011;417(1-2):216-26)
  • a phase III contraction wave in the stomach squeezes and flushes all material remains in the stomach to the small intestine include submicron particulates.
  • CDSS in brief, are influenced by the small intestinal transit time, which in turn is influenced by the food intake, the time the meal was eaten in relative to the time the drug was taken, the shape and form of CDSS formulations, and the disease stage of the patient.
  • sulfasalazine is a prodrug that successfully employs colonic microbiota to cleave the inactive precursor and release the active mesalazine.
  • Other enzymes produced by the colonic bacteria such as saccharolytic bacteroids and bifidobacterium to break down di-, tri-, and polysaccharides were successfully absorbed by the upper intestinal tract (Gorbach S et al. Gastroenterology.1967(6); Hill M, Drasar BS. The normal colonic bacterial flora. From the bacterial metabolism research lab.1975; Sinha VR et al. Drug Dev Ind Pharm. 2004;30(2):143-50).
  • Anaerobic bacteria are also known to be the main engine that produces polysaccharidases that break down short-chain fatty acids such as butyric, acetic, and propionic acids.
  • the high levels of polysaccharidase producing bacteria mean that attention has turned to polysaccharides as colonic delivery systems; these compounds are cheap, nontoxic, and biodegradable.
  • a selection of polysaccharides can avoid degradation in the small intestine but are used as a substrate by the colonic microbiota. These polysaccharides can be used as coatings or matrix systems.
  • the performance of polysaccharide based colonic delivery systems has been reviewed by many authors (74, 75) and will not be discussed in detail in this review.
  • Amylose is a starch polysaccharide; starch polysaccharides come in many forms, several of which are indigestible by human pancreatic enzymes but act as a food source for colonic bacteria (76, 77) This combination of colon-specific polysaccharide and insoluble polymer (to prevent swelling and premature drug release) has achieved consistent colonic targeting with various drug molecules.
  • McConnell et al., 2008d recently compared a pH-sensitive drug delivery system using Eudragit S to the polysaccharides prodrugs of amylose/ethyl-cellulose coated theophylline pellets that target colonic bacteria.
  • the targeted prodrug released in the colon is far more effective compared to the pH-coated pellets.
  • the study was completed by utilizing gamma scintigraphy and pharmacokinetics data of theophylline (43).
  • a phase III efficacious clinical trial studied eight UC patients using the above comparative approach.
  • the authors noted that the prednisolone prodrug approach has fewer side effects compared to the pH-trigger method and proposed that such a prodrug approach may be useful for maintenance therapy.
  • a colon-targeted delivery system also relies on specific colonic bacteria to cleave prodrug into a drug (78).
  • the microbiome-prodrug approach also incorporates pH sensitive elements for targeting the neutral colonic environment.
  • the above dual mode often utilizes lactulose and acidic pH-responsive cores to help dissolve the tablets upon reaching the colon.
  • the by-product of the prodrugs is usually some form of short-chain fatty acids that facilitates a burst release profile.
  • the CDDS that relied on colonic bacteria has been successfully applied to theophylline, a drug that requires a narrow therapeutic index (43, 79, 80).
  • the technology comprises a mixture of pH-responsive polymer (Eudragit S) and biodegradable polysaccharide (resistant starch).
  • the Eudragit S component in the coating has two functions - preventing the disintegration of the film in the upper gastrointestinal tract and controlling the swelling of polysaccharide.
  • the polysaccharide in the coating resists the digestion by mammalian pancreatic amylase enzymes and is digestible by colonic bacterial enzymes. Once entering the colon, both trigger mechanisms contribute to the dosage form disintegration, ensuring appropriate drug targeting.
  • One MMX® formulation is characterized by a lipophilic matrix dispersed in a hydrophilic structure.
  • the technology is a proprietary blend of liposome and hydrophilic excipients mixed with drug powders and pressed into a tablet. The tablets were then coated with an enteric coat to prevent upper gastrointestinal absorption.
  • the mechanism of drug release obtained by this pharmaceutical formulation concerns the gastro-resistant coating, which avoids the release of the embedded compound until the tablet is exposed to a pH of seven or higher, which is normally reached in the terminal ileum.
  • the activity of the tablet core which consists of hydrophilic excipients (thought to drive the tablet to swell into a viscous gel mass, slowing the release of the drug) and lipophilic excipients (thought to slow the penetration of aqueous fluids into the tablet core), results in a homogenous and prolonged exposure of the whole colonic mucosa to the embedded substance(s).
  • the MMX® delivery system guarantees that active drugs play their therapeutic role directly on the colonic mucosa and minimizes the systemic absorption of the drug. This approach proved to be a successful drug delivery system to the colon in a few clinical trials (182).
  • MMX most successful application was toward low molecular weight chemical entities such as mesalazine, budesonide, and low molecular heparins (82). The success with low molecular weight entities is understandable due to the inner core of water-insoluble gel that formed once the outer pH-sensitive coating had dissolved (83). Regardless, MMX is still not wholly site specific due to a pH burst release mechanism that has significant interindividual differences.
  • a mesalazine release study via gamma scintigraphy images showed release started in the small intestine and ileum, then reached mean maximum plasma concentration in the ileocaecal junction.
  • Therapeutic nanoparticles are colloidal structures with a cargo space for drugs that is segregated from the environment by a hydrophilic, usually polyethylene glycol (PEG), corona that prevents recognition by macrophages and enables long-term circulation in the bloodstream. (183).
  • PEG polyethylene glycol
  • the size of nanoparticles (10–100 nm) permits their extravasation and accumulation in tumor sites. Passive targeting is based on pathophysiological characteristics unique to solid tumors: hypervascularity, irregular vascular architecture, potential for secretion of vascular permeability factors, and absence of effective lymphatic drainage that prevents efficient clearance of macromolecules.
  • Nanoparticles are not generally administered orally mainly because of physiological obstacles; i.e., from the perspective of cellular drug delivery, access to the cytosolic space of eukaryotic cells is restricted primarily to hydrophobic small drugs with a MW ⁇ 500, which have relatively high membrane partition coefficients and permeability constants.
  • nanoparticles can be conjugated with various bioadhesive (e.g., poly(lactic acid (PLA)), [Id., citing 3] P-gp pump- inhibiting (e.g., d- ⁇ -tocopheryl PEG succinate (TPGS))[Id., citing 6], and vitamin [Id., citing 7,8,9,10,11] (e.g., biotin, folic acid, vitamin B12, and transferrin) ligands.
  • bioadhesive e.g., poly(lactic acid (PLA)
  • P-gp pump- inhibiting e.g., d- ⁇ -tocopheryl PEG succinate (TPGS)
  • TPGS d- ⁇ -tocopheryl PEG succinate
  • vitamin e.g., citing 7,8,9,10,11
  • biotin, folic acid, vitamin B12, and transferrin ligands.
  • micellar nanoparticles were labeled with the fluorescent molecule coumarin 6 for all three nanoparticle types.
  • fluorescently labeled nanoparticles were administered orally to rats. After jugular vein cannulation, blood samples were taken at fixed time intervals over 24 h, and the concentration of fluorescent nanoparticles in the plasma was measured by a fluorescence HPLC method.
  • CME clathrin-mediated endocytosis pathway
  • the CME pathway comprises primarily four types of organelles: early, recycling, and late endosomes, and lysosomes. These four classes of endocytic organelles are not preexisting, stable structures but rather are dynamic and difficult to recognize based on their morphology or position in the cytoplasm alone. Early endosomes represent a dynamic network of tubules and vesicles dispersed throughout the cytoplasm. [Id., citing 15,16].
  • Late endosomes are defined as vesicular structures that accumulate and concentrate internalized cargo intended for degradation. [Id., citing 16,17]. Late endosomes degrade their contents progressively, thus providing for the recycling of surviving receptors, and ultimately increase in density as digestible membrane and content are processed, catabolic products are released, and the remaining undigestible material (lysosomal hydrolases, and certain membrane and lipid components) is concentrated. At this stage, they become resting lysosomes, which can be activated again upon fusion with late endosomes. [Id., citing 16,17].
  • a chip-based dynamic cell culture model has been compared to the common static cell culture and mouse model to assess its capability to predict in vivo success more accurately, by using a well-defined poly((methyl methacrylate)-co-(methacrylic acid)) and poly((methyl methacrylate)-co-(2-dimethylamino ethylmethacrylate)) based nanoparticle library (184).
  • concentration of poly-methacrylic acid seemed to influence the rate of cellular uptake (higher concentrations of polymethacrylic acid increases the negative surface charge and decreases the hydrophobicity contributed to the decreasing concentration of the copolymer) (184).
  • Anionic pH-sensitive membrane-disruptive polymers have evolved as a class of bioactive excipients for the cytosolic delivery of therapeutic macromolecules.
  • a large variety of anionic copolymers and analogues of poly (acrylic acid)(PA) was investigated and compared to a cationic PA copolymer.
  • the pH-responsive membrane-disruptive properties were characterized by employing three in vitro models, such as pH dependent shift of pyrene fluorescence, liposome leakage and lysis of red blood cells. The pH-dependent increase of polarity and membrane disruption in the different model systems was in good agreement for all tested PA polymers.
  • the efficacy of polymer-induced membrane disruption was concentration-dependent and significantly affected by the composition of the membrane.
  • the sensitivity of relatively complex membranes of mammalian cells can be ranked between plain diphosphatidylcholine (DPPC) liposomal membranes and the more rigid cholesterol- containing DPPC membranes.
  • DPPC plain diphosphatidylcholine
  • PPA poly(propacrylic acid)
  • PDMAEM disruptive cationic PA polymer
  • the charge density of the nanoparticles may have resulted in lysosomal rupture due to osmotic swelling, thus resulting in the release of drugs into the cytosol (185).
  • Colon Bioavailable Drugs Enterohepatic Recycling (EHR) [0054] The concept of a “recyclable colonic drug” utilizing the enterohepatic recirculating loop to target and deliver drugs to the colon is a recent strategy. Bioavailability is a significant issue in drug development because sufficient drug concentration in the target organ is needed to elicit the desired therapeutic effects (88).
  • the described invention utilizes a modification in the current COXIBs structure by adding a desirable metabolic moiety (i.e., phenolic moiety) to enable the derivatives to undergo first pass effect.
  • a desirable metabolic moiety i.e., phenolic moiety
  • the engineered COXIBs will undergo rigorous first pass metabolism in the liver.
  • the desirable metabolic moiety of COXIBs derivatives is metabolites by hepatic uridine 5'-diphospho- glucuronosyl-transferase (UGTs), and efflux into the small intestine via transporters (e.g., breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 2 (MRP2)).
  • transporters e.g., breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 2 (MRP2)
  • Glucuronidation facilitates EHR. Glucuronidation is one of the most important phase II metabolic pathways of xenobiotics, and endogenous substances (97-99). Glucuronides of some phenolic compounds are good substrates of certain transporters (e.g., BCRP, MRP2), resulting in extensive hepatic secretion into the bile.
  • Glucuronidation capability can be highly different in the liver than in the colon.
  • Glucuronidation is catalyzed by UDP-glucuronosyltransferases (UGTs), which have approximately 24 known isoforms (97-99) with organ-specific distribution and expression levels (100).
  • UGT1A10 is exclusively expressed in the colon
  • UGT1A9 and UGT1A1 are highly expressed in the liver but only poorly expressed in the colon (101).
  • the colon’s glucuronidation capability is low (by as much as 100x slower) compared to the small intestine and the liver (101).
  • the glucuronidation rates of some flavonoids and drugs with phenolic moiety employed here are highly specific for the liver and not for the colon (102, 103).
  • the designed COX-2 inhibitors’ phenolic moiety is synthesized toward UGT1A1 enzymes and showed extensive glucuronidation in the liver and facilitate their biliary excretion.
  • EHR can prolong the drug’s apparent half-life in the GI tract. Due to recycling, drug molecules are repeatedly present in the GI tract, resulting in a prolonged apparent drug half- life in the colon (104, 105). Bile salt molecule, an endogenous compound, is estimated to be recycled within the enterohepatic loops on average about 20 times via EHR (106); although the number of recycling loops and repeated exposure of current locally bioavailable COX-2 inhibitors are still under investigation.
  • Microbial ⁇ -glucosidase Due to the increase in hydrophilicity of the metabolites, the recyclable colonic targeted COX-2 inhibiting drugs are not expected to be reabsorbed by the epithelium enterocytes. There, once reaching the colon, the metabolites of locally bioavailable COX-2 inhibitors would get deconjugated and reverse back into active drugs via microbial ⁇ - glucosidase. The colon has approximately 100 billion to two trillion organisms per gram of luminal content. A variety of microbiomes are highly active in the glycosylation of phenolic glucuronides. A fraction of glucuronided candidates is observed to decouple from the glycolic acid and convert back into the parent compound.
  • Hard gelatin capsules provide an advantage over other solid dosage forms, in that the raw drug can be packed inside the casing thereby reducing the need for excipients and extensive formulations (146, 147).
  • Hard gelatin capsules are hydrogenated collagen made up of abundant proteins that are readily broken down into amino acids and become available for absorption within hours of ingestions (148).
  • the coating of gelatin capsules is challenging. The smooth gelatin surface causes poor adhesion. Extreme pH or organic solvent usage in the polymer film can cause structural deformity and compromise the integrity of the capsules (149, 150).
  • enteric and erosion films for the size nine hard gelatin capsule S9C, 2.71 mm x 8.4 mm with a surface area of 68 mm 2
  • S9C size nine hard gelatin capsule
  • Conventional coating equipment and the specific parameters are also inapplicable to the S9C in small quantities for experimental use.
  • Polymers [0063] Polylactic-co-glycolic acid (PLGA) and Eudragit S100 (ES100) are two commonly used pharmaceutical excipients that are generally recognized as safe (GRAS) by the United States FDA and the European Medicines Agency.
  • biodegradable polymers contain hydrolyzable bonds where the passive hydrolysis or enzyme-catalyzed hydrolysis breaks down the chemical bond between the copolymers. The hydrolysis degradation occurs via a random chain scission process to form oligomers and finally monomers (155). The enzyme-catalyzed hydrolysis also referred to as biodegradation is when a biological system is partially involved in the chemical bond cleavage.
  • Most synthetic biodegradable polymers, PLGA and ES100 mainly undergo passive hydrolysis (156).
  • the degradation rate is determined by its chemical bond, composition, the pH of the environment, the water content, and the swelling rate. Degradation begins with water uptake into the bulk of the polymer matrix, which leads to swelling. Next, hydrolysis occurs resulting in oligomers and monomers degrading from the polymer. Progressively, the degradation changes the microstructure of the bulk, forming pores, where the oligomers and monomers are released. Heterogenous degradation refers to the surface degradation of the polymers whereas homogenous degradation describes a uniform break down of the bulk material throughout its matrix. [0067] Erosion is a more complicated process of polymer degradation in which additional parameters such as the swelling rate and material porosity are involved.
  • Poly (D,L-lactic-co- glycolic acid) three dimensional objects have been shown to exhibit an inverse flux erosion throughout the inner bulk material (154); once degradation begins increases in the degradation rate due to the lactide monomer content serves as an autocatalytic hydrolysis resulting in erosion (155, 157, 158).
  • the PLGA erosion rate can be optimized by adjusting the amount of porosity and the sizes of the micropores within PLGA matrix (159).
  • PLGA is an excellent biodegradable and biocompatible polymer (153, 160). Varying the ratios of lactic to glycolic acid controls the rate of water cleavage. Lactic acid contributes to the rigidity of the polymer backbone, thus increasing the duration that the polymer remains within a biological system (161).
  • PLGA 85:15 grade was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule. PLGA 85:15 also allows for successive coating applications.
  • Eudragit S100 is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio. ES100 pH dependent nature were finely tune by the number of negative charge on the MAA’s COOH functional group at basic condition (162).
  • ES100 is soluble in alkaline digestive fluids by salt formation (163, 164).
  • a disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility (supplemental S4). However, the addition of plasticizer changes the dissolution profile and takes longer for the film to dry (165).
  • Coating is the process of adding an outer layer of material to the surface of a substrate.
  • pharmaceutical coating techniques including conventional pan (involves coating capsules with a sprayed-on solution), fluid bed, dry powder (solvent-less) utilizing an electrostatically charged powder, magnetically assisted impaction, compression, hot melt extrusion, supercritical fluid, and supercell coating technology. These techniques function to mask taste and odor, protect from degradation, control the pharmacokinetic properties of drugs, or a combination of the aforementioned purposes.
  • Fluid applied coated technique substrates sequentially transition through the following stages: fresh runny solution, semisolid, sticky, and dry film. Criteria for a successful coating technique would be to have little or no visual defects, no functionality defects, increase production, and simplify operations. Unfortunately, not a single coating technique above meets all the criteria for all applications. Some coating techniques are more advantageous than others in some applications but less advantageous for other applications.
  • a conventional pan coater is one of the oldest coating technologies available — a thin film forms covering the capsules when a solution is sprayed into a temperature and pressure-controlled chamber. The pan continuously rotates tumbling the capsules; heated air is introduced evaporating the excess sprayed solution.
  • a fluid bed coater ( Figure 7) is similar to the conventional pan with additional air injected below the capsules resulting in a suspended, fluidized bed (85).
  • the design helps separate the capsules to prevent clumping while the coating solution is spraying.
  • a critical disadvantage for conventional pan and fluid bed techniques is the prolonged processing time between successive applications of coats. The mass of the polymer dissolved in the spray solution is limited to 3% of the total capsule’s weight to ensure even drying and prevent intense sustained attrition that leads to abrasion and chipping of the film.
  • Other disadvantages to these techniques are the requirements of a large amount of bulk material, expensive equipment, and a trained technician. The technician must monitor critical parameters to ensure reproducibility between batches such as the spraying rate, dewpoint, mass solution flow, air flow, and temperatures.
  • Liquid solution coatings often include an aqueous or organic solvent to dissolve polymers.
  • An aqueous solvent requires a large amount of energy and high temperatures to evaporate often resulting in an undesirable appearance of the film formed as well as degradation of the drugs.
  • Organic solvents on the other hand are environmentally toxic.
  • Liquid solution coating often requires lengthy processing and drying to altogether remove the solvents. This, in turn, increases manufacturing cost.
  • the dry powder, solvent-less coating techniques such as the electrostatic dry coating relieve manufacturers of the above challenges (86). In general, the mixture of conductive particles and polymers are cast into a disk.
  • An electric current is applied to an adjacent electrode thus ionizing the conductive particles, while an air jet is blown toward grounded capsules, the electrical field and the mechanical forces of the jetted air cause ionized particles to deposit onto the capsules.
  • the voltage, air flow, and powder density together all work to control the thickness, performance, and the appearance of the depositing film.
  • a challenge of this method is the precise parameters required to obtain desirable film as well as avoiding supercharging the substrate that would destroy the drug within the capsule.
  • the cleanliness of the substrate, high impact force, heat generated within the chamber sometime led to uneven thickness, void space, and multilayers deposition of the films.
  • the present invention disclosure provides oral dosage forms (e.g., hard gelatin coated capsules) formulated to dispense their payload in the colon comprising a combination of biodegradable polymers and a coating process.
  • the coating formulations and coating techniques provided reduce high systemic exposure to COX-2 inhibitors (total area under the curve (AUC_total)) that can result in cardiovascular toxicity.
  • the described locally bioavailable COX-2 inhibitors are optimized in design to subjugate microorganisms in the microbiota to reactivate locally bioavailable COX-2 inhibitors metabolites into parent compounds, and thus provide formulations and methods for achieving repeated drug exposure in the colon with minimal systemic exposure.
  • the present invention provides an oral pharmaceutical composition
  • a gelatin capsule containing an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and a therapeutically effective amount of a therapeutic agent disposed within the capsule.
  • the present invention provides an oral pharmaceutical composition
  • a capsule comprising an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and a particulate formulation disposed within the capsule, comprising a plurality of particles each comprising a therapeutically effective amount of a therapeutic agent, and one or more polymers encapsulating the therapeutic agent and which releases the therapeutic agent at a pH above 6.0.
  • the therapeutic agent comprises a structure as defined by any one of Formulas 1-10 or a pharmaceutically acceptable salt thereof.
  • the therapeutic agent is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) ( Figure 1C) or a pharmaceutically acceptable salt thereof.
  • the biodegradable polymer is a poly(lactide-co-glycolide) (PLGA) selected from the group consisting of 95:5 poly(lactide-co-glycolide) 85:15 poly(lactide-co- glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide).
  • the biodegradable polymer is 85:15 poly(lactide-co-glycolide) (PLGA8515).
  • the biodegradable polymer is applied to the capsule to a predefined thickness of 115 ⁇ 35 nm.
  • the inner delayed-release coating further comprises a plurality of pores to control release of the therapeutic agent.
  • the pH-sensitive polymer is Eudragit S100.
  • the pH-sensitive polymer is applied to the capsule to a predefined thickness of 50 ⁇ 15 nm.
  • size of the plurality of particles is about 100 nm to about 2000 nm, inclusive.
  • the one or more polymers comprises 50:50 poly(lactide-co-glycolide) (PLGA5050), Eudragit S100, and/or poly(vinyl alcohol) (PVA).
  • PLGA5050 poly(lactide-co-glycolide)
  • Eudragit S100 Eudragit S100
  • PVA poly(vinyl alcohol)
  • the composition when the composition is orally administered to a subject there is a lag period of at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, or at least about 12 hours before sustained release of the therapeutic agent.
  • sustained release of the therapeutic agent in the colon occurs at a pH of about 6.8.
  • the composition reduces therapeutic dose of a therapeutic agent, and/or reduces total systemic exposure (AUC) of a therapeutic agent, and/or increases local colonic exposure (AUCcolon) of a therapeutic agent, and/or increases concentration of the therapeutic in the colon by lowering AUC 0-24Hours , and increasing local AUC colon , and/or utilizes enterohepatic recycling (EHR), and/or reduces occurrence of on target off organ systemic toxicity associated with administration of the therapeutic agent.
  • the dopant is sodium chloride, sodium percarbonate, sodium bicarbonate, sodium carbonate, or sodium acetate.
  • the dopant configures the polymer to increase its porosity.
  • the microparticle formulation has a theoretical encapsulation efficiency (EE) of 30-45%.
  • EE theoretical encapsulation efficiency
  • a release profile of the particles in vitro is pH dependent.
  • the in vitro pH-dependent release profile of 6A1 microparticles comprises a rapid release of the active in a neutral pH environment.
  • the composition comprises an extended release profile.
  • Tmax is 3 hours.
  • the present invention provides a method of spin coating a capsule, comprising: (i) providing a vacuum spinning plate comprising individual pods; (ii) providing sealed capsules wetted with a mild basic water (e.g., pH 9 NaOH 0.1 mM) loaded into the individual pods within the vacuum spinning plates; (iii) providing an amount of a first polymer dissolved in a solvent; (iv) applying a vacuum; (v) spinning the plate a first time for about 30 seconds at a spinning speed of 100 RPM; (vi) spinning the plate a second time for about 15 minutes at spinning speed of 500 RPM;(vii) providing an amount of a second polymer dissolved in a solvent; (viii) applying a vacuum; (ix) spinning the plate a third time for about 30 seconds at spinning speed of 100 RPM; (x) spinning the plate a fourth time for about 15 minutes at a spinning speed of 500 RPM; (xi) removing the capsule from the spinning plate; and (xii)
  • a mild basic water
  • the method achieves a polymer mass loading of at least about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 10.5%, about 11%, about 11.5%, about 12%, about 12.5%, about 13%, about 13.5%, about 14%, about 14.5%, or about 15%; and/or the method achieves a coating comprising a smoothness and uniformity of surface appearance; and/or the method achieves a predetermined thickness measurement; and/or the method achieves in vitro dissolution; and/or the method achieves a drying time of less than about 1 hour, less than about 30 minutes, less than about 15 minutes, less than about 10 minutes, or less than about 5 minutes.
  • the present invention provides a method for reducing severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: providing the pharmaceutical composition according to claim 1 or 2, wherein the therapeutic agent is a selective COX-2 inhibitor, and administering an effective amount of the pharmaceutical composition orally to the subject.
  • the present invention provides a method for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: providing the pharmaceutical composition according to claim 1 or 2 , wherein the therapeutic agent is a selective COX-2 inhibitor; and administering an effective amount of the pharmaceutical composition orally to the subject.
  • the therapeutic agent that is a selective COX-2 inhibitor is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) or a pharmaceutically acceptable salt thereof.
  • 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) or a pharmaceutically acceptable salt thereof.
  • FIG.1A shows the binding of celecoxib
  • FIG.1B shows the flexibility of the central pyrazole ring, which is in the gate side of the binding pocket.
  • FIG.1C shows the structure of celecoxib, 6A1, and 7A1 attaching an additional moiety did not affect the binding. 6A1 was designed to retain the sulfamoylphenyl group and central heterocycle of celecoxib, but with the addition of a phenolic group.
  • FIG.2 shows current practice for colonic drug delivery systems.
  • FIG.3 shows respective size of different drug formulations.
  • FIGS. 4A-4C shows a comparison of nanoemulsion (FIG. 4A), microemulsion (FIG.4B), and mixture of nanoemulsion and microemulsion (FIG.4C).
  • FIG. 5 shows respective size of a microparticle versus nanoparticle, surface area increases as size of spherical particle decreases.
  • FIGS.6A-6D shows various type of liposomes used for colon drug delivery system (CDDS), including (FIG. 6A) Mannosylated, (FIG. 6B) Ionic, (FIG. 6C) Virosomes, (FIG. 6D) Multilayered liposomes (Vesosomes).
  • CDDS colon drug delivery system
  • FIGS. 7A-7D shows current coating technology for colon drug delivery system (CDDS), including Fluidized Bed (85) (FIG.7A), Electronic dry powder (86) (FIG.7B), Hot Melt Extrusion (87) (FIG.7C), and the vacuum spin coating technique described herein (FIG. 7D).
  • FIGS. 8A-8C shows ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) of validated method ran on a colonic mucosa sample.
  • FIG.8A shows representative extracted-ion chromatogram (XIC) of multiple reaction monitoring (MRM) of all four compounds eluting in the order of 6A1 sulfate, 6A1 glucuronide, internal standard (IS), and 6A1.
  • FIG. 8B shows MRM- sulfate and IS.
  • FIG. 8C shows MRM+ glucuronide, IS, and 6A1.
  • FIG. 9A shows representative product ion spectra of internal standard (IS) in negative mode.
  • FIG. 9B shows representative product ion spectra of internal standard (IS) in positive mode.
  • FIG.9C shows representative product ion spectra of 6A1 in positive mode.
  • FIG.9D shows representative product ion spectra of 6A1 glucuronide.
  • FIG.9E shows representative product ion spectra of 6A1 sulfate.
  • FIGS. 11A-11B shows colonic mucosa and liver tissue concentrations collected from F344 rats two hours after administration of an intravenous dose of 5 mg/Kg 6A1, 6A1 glucuronide, and 6A1 sulfate. Liver and colonic mucosa concentrations were analyzed in triplicates.
  • FIG. 11A shows colonic mucosa has an average ⁇ standard deviation (SD) concentration of 225.94 ⁇ 65.94 ng/g for 6A1, 26.78 ⁇ 17.80 ng/g for 6A1 glucuronide, and 5.12 ⁇ 0.86 ng/g for 6A1 sulfate.
  • FIG. 11B shows liver tissues analyzed in triplicate had an average concentration of 337.60 ⁇ 100.27 ng/g for 6A1, 315.79 ⁇ 81.55 ng/g for 6A1 glucuronide, and 218.14 ⁇ 54.21 ng/g for 6A1 sulfate.
  • FIG.12A shows a schematic of exemplary capsule coating layers.
  • FIG.12B shows an image of an exemplary capsule’s respective size compared to a dime.
  • FIG. 12C shows scanning electron microscope (SEM) topography view of exemplary coated capsules (40X magnification).
  • FIG.12D shows thickness of an exemplary coated capsule obtained from scanning electron microscope (SEM) side view given thickness of 115 ⁇ 35 nm for PLGA 8515 coating and 50 ⁇ 15 nm for ES100 coating (80K X magnification).
  • FIG.13A shows factorial screening design for ES100 solvent system with inputted empirical values.
  • FIG.13B shows ES100 solvent system effect summary correlating the PValue set at 0.05 to logworth value (-log of PValue) of 1.30.
  • FIGS.14A-14B shows predicted value and empirical data input generated models to predict ES100 polymer drying time FIG.14A and mass added FIG.14B with R 2 correlation value above 0.93.
  • the ordinary least square model for predicting the polymer drying times FIG.14A is more powerful than the model used to predict the polymers’ mass load FIG.14B based on the square root of the variance residuals observed RMSE values (0.7071 and 1.4142).
  • RMSE was the square root of the observed variance residuals. The smaller the RMSE value, the closer the predicted value was to the real value.
  • FIGS.15A-15C shows scanning electron microscope (SEM) topography images of dip coating (FIG.15A), vacuum spin (FIG.15B), and fluid coating technique (FIG.15C). All pictures had a 1 mm scale bar at 40X magnification.
  • FIG.17 shows factorial screening design via JMPDoe14 results demonstrating that the ES100 system was optimal at 65: 10: 3.5 acetone : IPA: DI water at 11% polymer weight, and 8.75 minutes drying time has the highest desirability.
  • FIG. 18A shows novel vacuum spin coater (10 Pa vacuum, 500 RPM spin rate) capable of coating 8-16 capsules in 30 minutes (with an 88% success rate)
  • FIG.18B shows blueprint drawing of spinning plates.
  • FIGS.19A-19D show thermogravimetry analysis (TGA) and differential scanning calorimetry (DSC) determination of quality control parameters of exemplary coated capsules.
  • FIG. 19A shows TGA of one month old PLGA versus newly formulated PLGA (9.023% vs 10.286%).
  • FIG. 19B shows TGA of 1 month old versus newly formulated ES100 with no substantial solvent loss displayed by the consistent mass loading and the ratio of solvent (first weight loss) to total solid mass (second weight loss) with respect to temperature compared between two samples (analyzed in triplicates).
  • FIG. 19C shows DSC curve for PLGA 8515.
  • FIG. 19D shows DSC curve for ES100. Both DSC curves show a small glass transition, endothermic peak, and exothermic temperatures are consistent with historical data.
  • FIGS.20A-20C shows in vitro dissolution test performed for 7 coated S9C and one quality control (QC) failed S9C (10% PLGA 8515 inner coat and 10% Eudragit S100 as pH- sensitive coat).
  • QC quality control
  • FIG.20A is an image of a capsule that showed indications of breakage 6 hours after initial PBS buffered exposure.
  • FIG.20C shows observed methyl blue trail leakage into medium 6 hours and 21 minutes after initial PBS buffer exposure.
  • FIG. 20C shows empty capsule shell remained 7 hours and 12 minutes after initial PBS buffer exposure at pH 6.8.
  • the 6A1 concentrations were determined using ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) described herein.
  • FIGS.21A-21N show in vivo and ex vivo imaging study of coated S9C.
  • FIG.21A shows ⁇ -CT IVIS image obtained of capsules packed with barium sulfate (bright spots).
  • FIG. 21B shows 0 hour after gavage, capsules are not visible within the intestinal tract.
  • FIG.21C shows 2 hours after gavage, capsules visible (bright spot) within the stomach.
  • FIG. 21D shows 6 hours after gavage, smaller bright spots observed in colon.
  • FIG.21E shows 8 hours, capsules are not visible within the intestinal tract.
  • FIG. 21G shows ex vivo imaging of uncoated SC9 control animal’s stomach showed methyl blue stains indicate the location of capsules 1 hour after oral gavage.
  • FIG. 21G shows ex vivo imaging of uncoated SC9 control animal’s stomach showed methyl blue stains indicate the location of capsules 1 hour after oral gavage.
  • FIG. 21H shows 3 hours after oral gavage, the blue stain passed the duodenum and was visible in the jejunum.
  • FIG. 21I shows 4 hours after gavage, ex vivo intestine has no visible blue stain of the coated capsules group.
  • FIG.21J shows at 8 hours after gavage, ex vivo cecum had extensive methyl blue staining.
  • FIG. 21K shows at 10 hours, ex vivo showed partial staining of the cecum and complete blue staining of colon.
  • FIGS. 21L- 21N shows 12 hours after coated S9C were given, some remnants of undissolved polymer film fragments and capsule shells were visible and mangled in the forming fecal pellets. Some blue stained droppings were also observed (FIG.39).
  • the 6A1 and its metabolites’ concentrations were determined using the UPLC MSMS method (LLOQ of 2 ng/mL with instruments’ LOD of 0.50 ng/mL) described herein.
  • FIG. 23B shows a dose normalized time point of 6A1-glucuronide between three routes of administration.
  • FIG. 23C shows the Kruskal-Wallis of the one-way ANOVA of 6A1 ranks that suggested there was a significant difference between IV/S9C_coated and S9C_uncoated/S9C_coated. The test did not find any substantial different between the IV and uncoated S9C (oral suspension formulation).
  • FIG. 23D shows a one-way ANOVA statistical run of dose normalized 6A1 glucuronide did not yield any significant difference between the three groups.
  • FIG. 24 shows SEM images of formulations 6, 14, and 15.
  • FIG. 25 shows exemplary wet milling and spontaneous emulsification solvent evaporation processes for fabrication of 6A1 microparticles. 10mg/ml of powder 6A1 in 35% methanol water was micronized with three different sized glassbeads at 1,600 rpm for an hour. Different slurry volumes (0 - 1.0 mL) were added dropwise into different volumes of 5% (w/w) ES100 (Table 7).
  • FIG.27 shows blood concentration time profile of food effect performed for coated sized 9 hard gelatin capsules (S9C).
  • the dose normalized of AUC_total was highest in the uncoated (twice the enteric coats and three times the enteric and delayed coated).
  • the uncoated size 9 capsules contained raw drug powder packed inside the S9C.
  • the enteric coat S9C contained raw drug powder packed inside S9C and a layer of ES100 of approximately 50 nm in thickness.
  • the enteric coat and delayed coat contained raw drug powder packed inside S9C, with a middle layer of approximately 150 nm of PLGA8515, and an outer layer of ES100 of approximately 50 nm in thickness. All animals were fed ad libitum.
  • FIG. 30 shows multiple-oral-dose regimen pharmacokinetic study of 6A1 microparticles formulated for sustained released packed inside the enteric and delayed release coated size 9 hard gelatin capsules (S9C).
  • FIG.32 shows blood concentration time profile of 6A1 across three different size 9 hard gelatin capsule coatings showed the enteric and delayed release coating has the lowest systemic drug circulations (Teal color block, AUCtotal).
  • FIG. 33 shows blood concentration time profile of 6A1 of enteric and delayed release coated size 9 hard gelatin capsule packed with microparticle F1 and F2. The single dose and the via twice daily for four day at 20 mg/Kg had similar results.
  • FIG.34 shows colonic tissue drug concentration of 6A1 with normalized dosage across IV, oral suspension, size 9 (S9C) coated capsules with raw powder, and size 9 capsule with microparticle formulations. The microparticles formulation 1 has the highest colon tissue accumulation of 6A1.
  • FIG.35 shows a mechanism of chemoprevention via Cox-2 Inhibition.
  • FIG.36 shows blank matrix sample injection after 6 upper limit of quantification (ULOQ) injections shown negligible internal standard peak at 4.2 minutes.
  • FIG.37 shows lower limit of quantification (LLOQ) injection using Raptor Restek Biphenyl column.
  • FIG.38 shows exemplary solvents and plasticizer for ES100.
  • FIG. 39 shows Perkin Elmer IVIS Lumina III XRMS imaging parameters and special data analysis.
  • FIG.40 shows blue stained fecal droppings were observed 10 hours post dosed as well as polymer residuals.
  • FIG.40 shows blue stained fecal droppings were observed 10 hours post dosed as well as polymer residuals.
  • peptide is a reference to one or more peptides and equivalents thereof known to those skilled in the art, and so forth.
  • the term “about” means plus or minus 20% of the numerical value of the number with which it is being used. Therefore, for example, about 50% means in the range of 40%-60%, inclusive, i.e., 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, or 50%.
  • active refers to the agent, drug, ingredient, component or constituent of the compositions of the present invention responsible for the intended therapeutic effect.
  • active pharmaceutical ingredient refers to a substance or mixture of substances intended to be used in the manufacture of a drug product and that, when used in the production of a drug, becomes an active ingredient of the drug product. Such substances are intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease or to affect the structure and function of the body.
  • administering includes in vivo administration, as well as administration directly to tissue ex vivo.
  • compositions may be administered systemically (e.g., orally, buccally, parenterally, by inhalation or insufflation (i.e., through the mouth or through the nose), or rectally) in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired, or may be locally administered by means such as, but not limited to, injection, implantation, grafting, topical application, or parenterally.
  • the described invention provides for oral administration.
  • agent refers generally to an active compound(s) that is/are contained in or on the formulation. “Agent” includes a single such compound and is also intended to include a plurality of such compounds.
  • the terms “animal,” “patient,” and “subject” as used herein include, but are not limited to, humans and non-human vertebrates such as wild, domestic and farm animals. According to some embodiments, the terms “animal,” “patient,” and “subject” may refer to humans. According to some embodiments, the terms “animal,” “patient,” and “subject” may refer to non-human mammals.
  • AUC or area under the concentration-time curve of drug absorption and elimination is a measure of total systemic exposure to a drug.
  • the term “batch” as used herein refers to a specific quantity of a drug or other material produced in a process or series of processes so that it is expected to have uniform character and quality, within specified limits.
  • the batch size can be defined either by a fixed quantity or by the amount produced in a fixed time interval.
  • the term “batch formula (composition)” as used herein refers to a complete list of the ingredients and their amounts to be used for the manufacture of a representative batch of the drug product.
  • bioavailable and its other grammatical forms refers to the extent and rate at which an active agent, such as a drug or a metabolite thereof, enters systemic circulation, thereby accessing the site of action. According to some embodiments, bioavailability may be described as the fraction (%) of an administered drug that reaches the systemic circulation.
  • biocompatible refers to a material that is generally non- toxic to the recipient and does not possess any significant untoward effects to the subject and, further, that any metabolites or degradation products of the material are non-toxic to the subject. Typically, a substance that is “biocompatible” causes no clinically relevant tissue irritation, injury, toxic reaction, or immunological reaction to living tissue.
  • biodegradable refers to a material that will erode to soluble species or that will degrade under physiologic conditions to smaller units or chemical species that are, themselves, non-toxic (biocompatible) to the subject and capable of being metabolized, eliminated, or excreted by the subject.
  • biodegradable polymers can be degraded into low-molecular-weight compounds by a biological process, such as by an enzymatic process or a metabolic process in microorganisms. More generally, biodegradable polymers can be degraded under a natural environment or a physiological condition by either of enzymatic degradation or spontaneous hydrolysis.
  • Biodegradable polymers useful in the embodiments of the invention include, but are not limited to, polyesters, poly(lactide), poly(lactide-co-glycolide) (PLGA), poly(caprolactone), poly(hydroxybutyrates), Eudragit S100 (ES100), and the like. (Vroman I, Tighzert L. Biodegradable Polymers. Materials (Basel).
  • carrier as used herein describes a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the compound of the composition of the described invention. Carriers must be of sufficiently high purity and of sufficiently low toxicity to render them suitable for administration to the mammal being treated.
  • the carrier can be inert, or it can possess pharmaceutical benefits.
  • excipient “carrier”, or “vehicle” are used interchangeably to refer to carrier materials suitable for formulation and administration of pharmaceutically acceptable compositions. Carriers and vehicles useful herein include any such materials know in the art which are nontoxic and do not interact with other components.
  • Cmax refers to the maximum concentration or maximum systemic exposure of a drug.
  • C ss or steady state concentration is the time during which the concentration of the drug in the body stays consistent.
  • controlled release is intended to refer to a drug-containing formulation in which the manner and profile of drug release from the formulation are regulated. This refers to immediate as well as non-immediate release formulations, with non-immediate release formulations including, but not limited to, sustained release and delayed release formulations.
  • cyclooxygenase or “cyclooxygenase isoenzymes” as used herein refers to an enzyme (specifically, a family of isozymes, EC 1.14.99.1) that is responsible for the conversion of certain polyunsaturated fatty acids to prostanoids, including prostaglandins, thromboxane, and levuloglandins.
  • the specific reaction catalyzed is the conversion of arachidonic acid to Prostaglandin H2, via a short-living Prostaglandin G2 intermediate.
  • COX-1 Cyclooxygenase-1
  • COX-2 Cyclooxygenase-2
  • COX-1 is responsible for the synthesis of prostaglandin and thromboxane in many types of cells, including the gastrointestinal tract and blood platelets.
  • COX-1 performs a distinct physiological homeostatic maintenance role within the vascular system and gastrointestinal tract. Inhibition of COX-1 may lead to gastrointestinal toxicity, cardiovascular toxicity, internal bleeding, and stomach ulcers.
  • COX-2 plays a major role in prostaglandin biosynthesis in inflammatory cells and in the central nervous system.
  • COX-2 enzyme is an inducible, pro- inflammatory cyclooxygenase isoform that is overexpressed and upregulated in many cancer tissues (116). COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (117) and is used as a prominent predictive marker in inflammatory bowel diseases (118). Prostaglandin synthesis in these sites is an important factor in the development of inflammation and pain. Inhibition of COX can provide relief from the symptoms of inflammation and pain. In particular, COX-2 inhibitors can have analgesic and anti- inflammatory activity by blocking the transformation of arachidonic acid into prostaglandin H2 selectively.
  • cyclooxygenase-2 inhibitor or “COX-2 inhibitor”, which can be used interchangeably herein, refer to molecules which inhibit the COX-2 enzyme regardless of the degree of inhibition of the COX-1 enzyme, and include pharmaceutically acceptable salts of those compounds.
  • a compound is considered a COX-2 inhibitor irrespective of whether the compound inhibits the COX-2 enzyme to an equal, greater, or lesser, degree than the COX-1 enzyme.
  • COX-2-selective refers to a molecule that exhibits selective binding to a COX-2 polypeptide. As used herein, “selective binding” means a preferential binding of one molecule for another in a mixture of molecules.
  • the binding of an active agent to a target molecule can be considered selective if the binding affinity is about 1 ⁇ 10 2 M ⁇ 1 to about 1 ⁇ 10 6 M ⁇ 1 or greater.
  • the term “delayed-release” is used herein in its conventional sense to refer to a drug formulation in which there is a time delay between administration of the formulation and the release of the drug there from. "Delayed release” may or may not involve gradual release of drug over an extended period of time, and thus may or may not be “sustained release.”
  • a delayed- release formulation may comprise a “delayed-release coating” to delay release of the active agent.
  • a delayed-release formulation may additionally or alternatively comprise an enteric coating to delay release of an active agent until the tablet or capsule has passed through the stomach to prevent the active agent from being destroyed or inactivated by the gastric environment or where it may irritate the gastric mucosa.
  • density is used herein to refer to the degree of compactness of a substance, and is measured in mass per unit volume.
  • derivative as used herein means a compound that may be produced from another compound of similar structure in one or more steps. A “derivative” or “derivatives” of a compound retains at least a degree of the desired function of the compound.
  • an alternate term for “derivative” may be “functional derivative.”
  • the term “displosed” and its other grammatical forms as used herein means to put in order, set, make us of, or arrange.
  • the term “dissolution rate” as used herein refers to the amount of a drug that dissolves per unit time. The dissolution rate of a drug may be altered by certain conditions, including, but not limited to, surface area, rotation speed, pH and ionic strength of the dissolution medium.
  • doping as used herein refers to the intentional introduction of impurities (“dopants”) for the purpose of modulating the properties of a material, e.g., a polymer.
  • dopants are introduced to control the releasing profile of drugs.
  • drug load (%) and drug loading capacity” are used interchangeably herein to refer to a ratio of the weight of a drug in microparticles relative to the total weight of the microparticles, expressed as a percentage. It reflects the drug content of the microparticle.
  • drug product refers to a finished dosage form that contains a drug substance, generally, but not necessarily, in association with one or more other ingredients.
  • drug substance refers to an active ingredient intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment or prevention of a disease, or to affect the structure and function of the body, but does not include intermediates used in synthesis of such ingredient.
  • effective amount refers to the amount necessary or sufficient to realize a desired biologic effect.
  • emulsion refers to a two-phase system prepared by combining two immiscible liquid carriers, one of which is disbursed uniformly throughout the other and consists of globules that have diameters equal to or greater than those of the largest colloidal particles. The globule size must be such that the system achieves maximum stability.
  • a basic emulsion contains at least three components, the two immiscible liquid carriers and the emulsifying agent, as well as the active ingredient.
  • Most emulsions incorporate an aqueous phase into a non-aqueous phase (or vice versa).
  • emulsions that are basically non-aqueous, for example, anionic and cationic surfactants of the non-aqueous immiscible system glycerin and olive oil.
  • EHR electronichepatic recycling
  • drugs refers to the circulation of biliary acids, bilirubin, drugs or other substances from the liver to the bile, followed by entry into the small intestine, absorption by the enterocyte and transport back to the liver.
  • EHR of drugs relates to the process by which drugs are conjugated to glucuronic acid in the liver, excreted into bile, metabolized back into the free drug by intestinal bacteria, and the drug is then reabsorbed into plasma. For many drugs that undergo this process, lower doses of drugs can be therapeutically effective because elimination is reduced by the recycling of the drug.
  • enterohepatic circulation can enhance their toxicity, and therefore inhibition of recycling may be protective for those drugs.
  • inhibition of this recycling process can lead to a reduction of the levels of drug and often a reduced therapeutic effect.
  • EHR can slow down the decrease of drug concentrations in the intestinal lumen, and make more unconjugated drug molecules available inside the colonic epithelial cells.
  • entero coating refers to a polymer barrier applied to an oral formulation, such as a capsule or tablet, that prevents its dissolution or disintegration in the gastric environment.
  • An enteric coating may protect an active agent from the acidity of the stomach (pH of 1.2), the stomach from the detrimental effects of an active agent, or to facilitate release of an active agent after the stomach at the near neutral pH of the distal small intestine (pH of 7.5 ⁇ 0.4) or the ileocecal junction.
  • the term “entrapment efficiency (%)” as used herein refers to a ratio of drug retained by a particle relative to the total amount available, expressed as a percentage.
  • the term “erosion rate” refers to the degradation of a polymer over time. Polymers can be degraded actively (by enzymes) or passively (by hydrolysis), resulting in surface or bulk erosion.
  • Surface erosion is a heterogeneous process wherein degradation of the polymer happens at only the surface and the rate is proportional to the surface area.
  • Drug release in surface-eroding systems is often correlated with a predictable erosion rate, which is considered ideal for many drug delivery applications. Erosion begins from the outside and progresses inward. In most cases, thicker systems have longer erosion times, and hydrophilic polymers degrade faster compared with hydrophobic materials. With bulk-eroding systems, degradation is homogenous throughout the material, and the size of the system remains constant in most cases. The drug is released in three stages: burst release from the surface, release from initial degradation of the polymer, and release of residual drug during complete degradation/homogeneous erosion of the polymer.
  • excipient is used herein to include any other agent or compound that may be contained in a formulation that is not the bioactive agent. As such, an excipient should be pharmaceutically or biologically acceptable or relevant (for example, an excipient should generally be non-toxic to the subject). “Excipient” includes a single such compound and is also intended to include a plurality of such compounds.
  • Eudragit S100 or “ES100” as used herein refers to an anionic copolymer composed of methacrylic acid (MAA)-methyl methacrylate (MMA) at a 1:2 ratio.
  • FAP Fluorescence Adenomatous Polyposis
  • APC adenomatous polyposis coli
  • FAP may be diagnosed when a person develops more than 100 adenomatous colon polyps.
  • An adenomatous polyp is an area where normal cells that line the inside of a subject’s colon form a mass on the inside of the intestinal tract.
  • Adenomatous polyps (adenomas) of the colon and rectum are typically benign (noncancerous) growths, but may be precursor lesions to colorectal cancer. Polyps greater than one centimeter in diameter are associated with a greater risk of cancer.
  • Adenomatous polyps appear by about 15 years of age in 50% of people with FAP, and by 35 years of age in 95%. Seventy percent of these individuals have colorectal malignancy by the age of 36 years, the average age of symptomatic diagnosis. Over the lifetime of this illness, there is a 100% cancer risk, typically in the fourth and fifth decades of life. The average age of colorectal cancer onset for attenuated familial adenomatous polyposis is 55 years. Clinically, FAP manifests equally in both sexes by the late teens and in the twenties age group. No true pharmacologic treatment is available for these patients.
  • formulation refers to a listing of the ingredients and composition of the dosage form.
  • impregnate as used herein in its various grammatical forms refers to causing to be infused or permeated throughout; to fill interstices with a substance.
  • leaching or “solvent extraction” as used herein refers to a process in which a component of a mixture is removed (extracted) by exposing the mixture to the action of a solvent in which the component to be removed is soluble. Electrospun fibrous polyurethane scaffolds in tissue engineering. Porogen leaching is a common approach to developing large, three-dimensional, porous scaffolds.
  • the scaffold material is incorporated with a chemically or physically incompatible porogen.
  • the porogen is selectively removed.
  • One consequence of this method of fabrication is the high degree of porosity (typically in the range of 90% or higher), which is necessary to achieve an interconnected network for random porogen organization. Scaffolds with extremely high porosities are advantageous for tissue engineering metabolically active tissues by allowing for more rapid diffusion of nutrients and removal of waste products. High porosities also provide large surface areas per volume to allow cell attachment and proliferation.
  • Lewis acid refers to any molecule or ion that can combine with another molecule or ion by forming a covalent bond with two electrons from the second molecule or ion.
  • An acid is thus an electron acceptor.
  • Hydrogen is the simplest substance that will do this.
  • Lewis base refers to a substance that forms a covalent bond by donating a pair of electrons, neutralization resulting from reaction between the base and the acid with formation of a coordinate covalent bond.
  • long-term release refers to an implant constructed and arranged to deliver therapeutic levels of the active ingredient for at least 7 days, and potentially up to about 30 to about 60 days.
  • Terms such as “long-acting”, “sustained-release” or “controlled release” are used generally to describe a formulation, dosage form, device or other type of technologies used, such as, for example, in the art to achieve the prolonged or extended release or bioavailability of bioactive agent to a subject; it may refer to technologies that provide prolonged or extended release or bioavailability of a bioactive agent to the general systemic circulation or a subject or to local sites of action in a subject including (but not limited to) cells, tissues, organs, joints, regions, and the like.
  • these terms may refer to a technology that is used to prolong or extend the release of a bioactive agent from a formulation or dosage form or they may refer to a technology used to extend or prolong the bioavailability or the pharmacokinetics or the duration of action of a bioactive agent to a subject or they may refer to a technology that is used to extend or prolong the pharmacodynamic effect elicited by a formulation.
  • a “long-acting formulation,” a “sustained release formulation,” or a “controlled release formulation” is a pharmaceutical formulation, dosage form, or other technology that is used to provide long-acting release of a bioactive agent to a subject.
  • long-acting or sustained release formulations comprise a bioactive agent or agents that is/are incorporated or associated with a biocompatible polymer in one manner or another.
  • the agent may be blended homogeneously throughout the polymer or polymer matrix, or the agent may be distributed unevenly (or discontinuously or heterogeneously) throughout the polymer or polymer matrix (as in the case of a bioactive agent-loaded core that is surrounded by a polymer-rich coating or polymer wall forming material as in the case of a microcapsule, nanocapsule, a coated or encapsulated implant, and the like.
  • the term “manufacture” as used herein refers to all operations of receipt of materials, production, packaging, repackaging, labeling, relabeling, quality control, release, storage and distribution of APIs and related controls.
  • the term “material” as used herein refers generally to raw materials (e.g., starting materials, reagents, solvents), process aids, intermediates, APIs, packaging and labeling materials.
  • the term “matrix” as used herein refers to a three-dimensional network of fibers and/or polymers that contains voids (or “pores”) where the woven fibers and/or polymers intersect.
  • microbiome or “gut microbiome” as used herein refers to the genes harbored by the microbial cells that constitute the gut microbiota.
  • microbiota refers to the microbial taxa or groups associated with humans.
  • micronize and its other grammatical forms as used herein refers to a process that reduces particle size to obtain micrometer- and nanometer-size particles.
  • Micronization may be useful, e.g., to improve the bioavailability of poorly soluble APIs by increasing particle surface area and accelerating dissolution rates; to improve formulation homogeneity and to control particle size.
  • the micronization process uses fluid energy, such as a jet mill.
  • a jet mill uses pressurized gas to create high particle velocity and high-energy impact between particles.
  • the process gas is separated from the solid particles after exiting the jet-mill chamber with a cyclone filter.
  • the micronization process uses mechanical particle-size reduction, e.g., using a bead mill. Bead milling uses wet mechanical milling to obtain nanoscale particles.
  • agitator bead mill for example, grinding beads and agitating elements are used to reduce the API particle size through impact and shear; product is separated from the grinding media at the outlet.
  • Process parameters include the formulation (e.g., product viscosity, percent solids, additives to prevent reagglomeration), bead density, bead size, bead-filling ratio, stirrer-shaft speed, and flow rate. If containment is needed, the batch-mixing tank can be placed in an isolator, and the mixture can be pumped to the bead mill, which is outside the isolator but is itself a closed system (www.pharmatech.com/using-micronization-reduce-api-particle-size).
  • the micronization process comprises dry powder grinding. According to some embodiments, the micronization process comprises wet milling.
  • microparticulate composition refers to a composition comprising a microparticulate formulation and, optionally, a pharmaceutically acceptable carrier, where the microparticulate formulation comprises a therapeutic agent and a plurality of microparticles. According to some embodiments, the therapeutic agent is impregnated within the polymer matrix of the microparticles.
  • milling and its other grammatical forms as used herein refers to a process (e.g., a machining process) of grinding, pulverizing, pounding, crushing, pressing, or granulating a solid substance to reduce particle size.
  • minimum effective concentration “minimum effective dose” , or “MEC” are used interchangeably to refer to the minimum concentration of a drug required to produce a desired pharmacological effect in most patients.
  • module as used herein means to regulate, alter, adapt, or adjust to a certain measure or proportion.
  • particles refers to an extremely small constituent, e.g., nanoparticles or microparticles) that may contain in whole or in part at least one therapeutic agent as described herein.
  • microparticle is used herein to refer generally to a variety of substantially structures having sizes from about 10 nm to 2000 microns (2 millimeters) and includes microcapsule, microparticle, nanoparticle, nanocapsule, nanosphere as well as particles, in general, that are less than about 2000 microns (2 millimeters).
  • the particles may contain therapeutic agent(s) in a core surrounded by a coating. Therapeutic agent(s) also may be dispersed throughout the particles. Therapeutic agent(s) also may be adsorbed into the particles.
  • the particles may be of any order release kinetics, including zero order release, first order release, second order release, delayed release, sustained release, immediate release, etc., and any combination thereof.
  • the particles may include, in addition to therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof.
  • the particles may be microcapsules that contain the therapeutic agent in a solution or in a semisolid state.
  • the particles may be of virtually any shape.
  • pharmaceutical composition is used herein to refer to a composition that is employed to prevent, reduce in intensity, cure or otherwise treat a target condition or disease.
  • the phrase “pharmaceutically acceptable carrier” refers to any substantially non-toxic carrier useable for formulation and administration of the composition of the described invention in which the product of the described invention will remain stable and bioavailable.
  • the pharmaceutically acceptable carrier must be of sufficiently high purity and of sufficiently low toxicity to render it suitable for administration to the mammal being treated. It further should maintain the stability and bioavailability of an active agent.
  • the pharmaceutically acceptable carrier can be liquid or solid and is selected, with the planned manner of administration in mind, to provide for the desired bulk, consistency, etc., when combined with an active agent and other components of a given composition.
  • pharmaceutically acceptable salt means those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. When used in medicine the salts should be pharmaceutically acceptable, but non- pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts may be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • salts are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well-known in the art. For example, P. H. Stahl, et al. describe pharmaceutically acceptable salts in detail in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” (Wiley VCH, Zurich, Switzerland: 2002). The salts may be prepared in situ during the final isolation and purification of the compounds described within the present invention or separately by reacting a free base function with a suitable organic acid.
  • Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsufonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate(isethionate), lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate and undecanoate
  • the basic nitrogen- containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl and diamyl sulfates
  • long chain halides such as decy
  • Basic addition salts may be prepared in situ during the final isolation and purification of compounds described within the invention by reacting a carboxylic acid-containing moiety with a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary or tertiary amine.
  • Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as lithium, sodium, potassium, calcium, magnesium and aluminum salts and the like and nontoxic quaternary ammonia and amine cations including ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine and the like.
  • Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine and the like.
  • Pharmaceutically acceptable salts also may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium or magnesium) salts of carboxylic acids may also be made.
  • pharmacodynamics refers to the study of the biochemical, physiologic, and molecular effects of drugs on the body and involves receptor binding (including receptor sensitivity), postreceptor effects, and chemical interactions.
  • pharmacokinetics refers to the study of the time course of absorption, distribution, metabolism, and excretion of a drug.
  • pilot scale refers to the manufacture of either a drug substance or drug product by a procedure fully representative of and simulating that used for full manufacturing scale. In production of microspheres, pilot scale can be, for example, 500 grams. For an API, pilot scale can be, for example 1 kg.
  • poly(lactic-co-glycolic acid) or “PLGA” as used herein refers to a linear copolymer that can be prepared at different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA). Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained, and these are usually identified in regard to the monomers’ ratio used (e.g., PLGA 85:15 identifies a copolymer consisted of 85% lactic acid and 15% glycolic acid).
  • Poly lactic acid contains an asymmetric ⁇ -carbon which is typically described as the D or L form in classical stereochemical terms and sometimes as R and S form, respectively.
  • the enantiomeric forms of the polymer PLA are poly D-lactic acid (PDLA) and poly L-lactic acid (PLLA).
  • PLGA is generally an acronym for poly D,L-lactic-co- glycolic acid where D- and L- lactic acid forms are in equal ratio.
  • Different synthesis mechanisms as known in the art can be used to obtain PLGA having different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA).
  • LA lactic
  • GA glycolic acid
  • Gentile P, Chiono V, Carmagnola I Hatton PV.
  • polymer refers to a large molecule, or macromolecule, composed of many repeated subunits.
  • monomer refers to a molecule that may bind chemically to other molecules to form a polymer.
  • copolymer refers to a polymer derived from more than one species of monomer.
  • polar refers to a molecule in which the positive and negative electrical charges are permanently separated, as opposed to nonpolar molecules in which the charges coincide.
  • Polar molecules ionize in solution and impart electrical conductivity. Water, alcohol and sulfuric acid are polar in nature; most hydrocarbon liquids are not. Carboxyl and hydroxyl groups often exhibit an electric charge.
  • polar molecule refers to a molecule with a positive charge on one end and a negative charge on its other end; or a molecule in which the electrons forming the valency bond are not symmetrically arranged.
  • pore refers to a void or interstices between particles of a solid or in a matrix that permits passages of liquids or gases through the material in either direction.
  • the term “porogen” as used herein refers to a substance used to create pores in a structure.
  • the term “porosity” as used herein refers to the ratio of the volume of pores to the volume of the material as a whole; it , and is usually expressed as a percentage.
  • the term “process” as used herein refers to a series of operations, actions and controls used to manufacture a drug product.
  • production refers to all operations involved in the preparation of an API from receipt of materials through processing and packaging of the API.
  • the term “reduce” or “reducing” as used herein refers to a diminution, a decrease, an attenuation, limitation or abatement of the degree, intensity, extent, size, amount, density, number or occurrence of disorder in individuals at risk of developing the disorder.
  • release and its various grammatical forms, refers to dissolution of an active drug component and diffusion of the dissolved or solubilized species by a combination of the following processes: (1) hydration or swelling of a matrix, (2) erosion of a matrix, (3) diffusion of a solution into the matrix; (4) dissolution of the drug; (5) diffusion of the dissolved drug out of the matrix, (6) pH-responsive release mechanism, and (7) microbially triggered delivery.
  • release may comprise a pH-responsive release mechanism.
  • a pH-sensitive drug delivery system e.g., for oral administration, may withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ⁇ 0.4) or the ileocecal junction.
  • a “pH-sensitive” polymer coating means that the coating material dissolves or elutes under circumstances of more than a given pH value to release an active agent.
  • An enteric coating may be dissolved or eluted at a pH of about 5.5 to initiate drug release, while the enteric coating comprising a pH-sensitive polymer of the present invention is preferably a polymer that dissolves at a higher pH and controls drug release to the colon.
  • salt refers to the compound formed when the hydrogen of an acid is replaced by a metal or its equivalent (e.g., an NH4+ radical). This is typical of the general rule that the reaction of an acid and a base yields a salt and water. Most inorganic acids ionize in water solution.
  • soluble and solubility refer to the property of being susceptible to being dissolved in a specified fluid (solvent).
  • insoluble refers to the property of a material that has minimal or limited solubility in a specified solvent. In a solution, the molecules of the solute (or dissolved substance) are uniformly distributed among those of the solvent.
  • solvate refers to a complex formed by the attachment of solvent molecules to that of a solute.
  • solvent refers to a an inorganic or organic liquid capable of dissolving another substance (termed a “solute”) to form a uniformly dispersed mixture (solution) used as a vehicle for the preparation of solutions or suspensions.
  • solvent evaporation method refers to a technique for preparing drug-loaded particles in which an active agent is dissolved, dispersed or emulsified into an organic polymer solution, which is then emulsified into an external aqueous or oil phase.
  • Particles are formed after solvent evaporation and polymer precipitation. Size of the particles can be controlled by adjusting parameters like manipulating evaporation temperature, controlling the rate of evaporation, manipulating stirring rate, etc.
  • Specification refers to a list of tests, references to analytical procedures, and appropriate acceptance criteria that are numerical limits, ranges or other criteria for the test described that establishes the set of criteria to which material should conform to be considered acceptable for its intended use.
  • conformance to specification means that the material, when tested according to the listed analytical procedures, will meet the listed acceptance criteria.
  • a “suspension” is a dispersion (mixture) in which a finely-divided species is combined with another species, with the former being so finely divided and mixed that it doesn't rapidly settle out. In everyday life, the most common suspensions are those of solids in liquid.
  • sustained release (also referred to as “extended release”) is used herein in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period.
  • symptom refers to a phenomenon that arises from and accompanies a particular disease or disorder and serves as an indication of it.
  • therapeutic agent refers to a drug, molecule, composition or other substance that provides a therapeutic effect.
  • therapeutic agent and “active agent” are used interchangeably.
  • the term “therapeutic component” as used herein refers to a therapeutically effective dosage (i.e., dose and frequency of administration) that eliminates, reduces, or prevents the progression of a particular disease manifestation in a percentage of a population.
  • a therapeutically effective dosage i.e., dose and frequency of administration
  • An example of a commonly used therapeutic component is the ED 50 which describes the dose in a particular dosage that is therapeutically effective for a particular disease manifestation in 50% of a population.
  • the term “therapeutic effect” as used herein refers to a consequence of treatment, the results of which are judged to be desirable and beneficial.
  • a therapeutic effect may include, directly or indirectly, the arrest, reduction, or elimination of a disease manifestation.
  • a therapeutic effect may also include, directly or indirectly, the arrest reduction or elimination of the progression of a disease manifestation.
  • the term “therapeutically effective amount”, “effective amount”, or an “amount effective” is an amount that is sufficient to provide the intended benefit of treatment. Combined with the teachings provided herein, by weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen may be planned which does not cause substantial toxicity and yet is effective to treat the particular subject.
  • the therapeutically effective amount for any particular application may vary depending on such factors as the disease or condition being treated, the particular active agent being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art may determine empirically the effective amount of a particular active agent and/or other therapeutic agent without necessitating undue experimentation.
  • Tmax refers to the time of maximum concentration or maximum systemic exposure of a drug.
  • t 1/2 or half-life refers to the time required to reduce plasma concentration of a drug to one-half of its initial value.
  • treat or “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease, condition or disorder, substantially ameliorating clinical or esthetical symptoms of a condition, substantially preventing the appearance of clinical or esthetical symptoms of a disease, condition, or disorder, and protecting from harmful or annoying symptoms.
  • Treating further refers to accomplishing one or more of the following: (a) reducing the severity of the disorder; (b) limiting development of symptoms characteristic of the disorder(s) being treated; (c) limiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting recurrence of symptoms in patients that were previously asymptomatic for the disorder(s). [0229] As used herein, a “wt.
  • the described invention provides pharmaceutical compositions for delivering a therapeutic agent to the colon of a subject.
  • the described invention provides pharmaceutical compositions to delay and sustain the release of an active agent.
  • the active agent is designed to be bioavailable in the colon (LBD), to realize low systemic drug exposure and to avoid off-organ toxicities.
  • the active agent is a selective COX-2 inhibitor.
  • the selective COX-2 inhibitor is 6A1
  • the first drug delivery system delays release via a coating technique in combination with a coating formulation that includes a pH-sensitive polymer and an erosion polymer for hard gelatin capsules.
  • the second drug delivery system also uses hard gelatin capsules containing 6A1 enterically encapsulated microparticles aiming for a sustained release profile.
  • the coating formulation achieves two objectives: the enteric biodegradable polymer coating will prevent early release of LBD COX-2 inhibitors (6A1) in the stomach that facilitate its absorption in the upper small intestine; the erosion mechanism makes the delayed release in the colon more precise, which could result in a smaller drug dose but higher colonic drug exposure. Delaying the release of 6A1 to the colon eliminates the impact of the inter-subject variability due to uridine 5'-diphospho- glucuronosyl- transferase (UGT) expression in the liver, beta-glucuronidase activities expressed by colonic microflora, and colonic disease states.
  • UTT uridine 5'-diphospho- glucuronosyl- transferase
  • delayed release delivery ensures a sufficient and consistent amount of 6A1 is present in the colon to saturate COX-2 enzymes.
  • the provided microparticle formulations will sustain the release of 6A1 via two mechanisms: slow erosion of the microparticles and accumulation of the microparticles within the targeted site via its enhanced solubility and increased surface area.
  • the provided dosage forms will preclude or minimize systemic distribution of LBD COX-2 inhibitors and allow for rapid attainment and maintenance of adequate therapeutic concentrations in the colon.
  • the biodegradable polymer formulations and coating technique will synergistically function as a successful drug delivery system for treating diseases in the colon.
  • compositions provided herein are intended to fill a void in the understanding of how to create safe COX-2 inhibitors that prevent colonic cancer in FAP patients.
  • the pharmaceutical compositions comprise one or more polymeric coatings to control release, delay release, and/or sustain release of a therapeutic agent.
  • the described invention provides pharmaceutical compositions comprising a particulate formulation containing a plurality of particles.
  • the present invention disclosure provides an oral pharmaceutical composition
  • a capsule comprising: (i) an inner delayed-release coating comprising a biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a therapeutically effective amount of a therapeutic agent disposed within the capsule.
  • the biodegradable polymer is doped to modulate porosity of the polymer.
  • the present invention disclosure provides an oral pharmaceutical composition
  • a capsule comprising: (i) an inner delayed-release coating comprising a biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a plurality of particles disposed within the capsule, each comprising a therapeutically effective amount of a therapeutic agent and one or more polymers encapsulating the therapeutic agent and which release the therapeutic agent at a pH above 6.0.
  • the biodegradable polymer is doped to modulate porosity of the polymer.
  • the pharmaceutical composition when the pharmaceutical composition is orally administered to a subject there is a lag period of at least about 6 hours (e.g., at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, or at least about 12 hours) after administration before sustained release of the therapeutic agent occurs at a pH above 5.5 (e.g., a pH between about 6 and about 8).
  • the described invention provides pharmaceutical compositions to (i) reduce the therapeutic dose of an active agent, (ii) reduce the total systemic exposure (AUC) of an active agent, (iii) increase the local colonic exposure (AUCcolon) of an active agent, (iv) increase the concentration of the active agent in the colon (e.g., lower AUC 0- 24Hours , increase local AUC colon ), and/or (v) reduce the occurrence of on target off organ systemic toxicity (e.g., cardiovascular side effects, such as unstable angina, myocardial infarction, and cardiac thrombus) of an active agent.
  • the pharmaceutical compositions are formulated for enterohepatic recycling (EHR).
  • the described invention provides multi-modal oral delayed-release formulations to sustain therapeutic agent exposure in the colon (lower AUC0-24Hours, increase local AUCcolon).
  • the described invention provides pharmaceutical compositions having a dual release mechanism with pH resistance, and comprising varying particle sizes ranging from 100-2000 nm, inclusive to achieve the highest ratio of drug accumulation in the colon over a given dose.
  • the dual-release mechanism design can be used to achieve maximum efficacy without subjecting the patients to a large dose and potentially reduce the dosing frequency for a therapeutic agent.
  • Eudragit S100 pH-sensitive polymer and PLGA 5050 can be used to formulate oral sustained release nano- and microparticles of therapeutic agent.
  • the described invention provides pharmaceutical compositions that sustained drug release above pH 6.0 and delay drug release for at least about 6 hours after administration.
  • the described invention provides pharmaceutical compositions comprising and microparticle formulations that are highly stable, have a high encapsulation efficiency, and exhibit a sustained release profile.
  • the pharmaceutical compositions can be formulated readily by combining the therapeutic agent (s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the actives of the disclosure to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • compositions for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, alter adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP).
  • fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol
  • cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum trag
  • disintegrating agents can be added, such as, but not limited to, the cross- linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Pharmaceutical preparations that can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, scaled capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers.
  • filler such as, e.g., lactose
  • binders such as, e.g., starches
  • lubricants such as, e.g., talc or magnesium stearate
  • suitable liquids such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the capsule may be formed of any suitable material, be any suitable size, and be either hard or soft.
  • the capsule may comprise gelatin, collagen, and/or cellulose.
  • the capsule may be a hard gelatin capsule, for example, a size nine hard gelatin capsule (S9C).
  • S9C size nine hard gelatin capsule
  • hard gelatin capsules provide an advantage over other solid dosage forms, in that the therapeutic agent can be packed inside the casing as a powder and/or a particulate formulation thereby reducing the need for excipients and extensive formulations (146, 147).
  • Hard gelatin capsules are hydrogenated collagen made up of abundant proteins that are readily broken down into amino acids and become available for absorption within hours of ingestions (148). [0247]
  • the coating of gelatin capsules can be challenging.
  • the smooth gelatin surface can cause poor adhesion. Extreme pH or organic solvent usage in the polymer film can cause structural deformity and compromise the integrity of the capsules (149, 150).
  • the application process of enteric and erosion films for the size nine hard gelatin capsule (S9C, 2.71 mm x 8.4 mm with a surface area of 68 mm 2 ), specifically designed for rodents and guinea pigs, is especially challenging and cumbersome with the conventional methods and dip-coating techniques. Conventional coating equipment and the specific parameters are also inapplicable to the S9C in small quantities for experimental use.
  • the novel coating technique described here is important for preclinical research and pilot studies where the amount of the new chemical entity is usually limited.
  • the pharmaceutical compositions can be formulated with a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is art recognized. It is used to mean any substantially non-toxic carrier conventionally useable for administration of pharmaceuticals in which the therapeutic agents of the present invention will remain stable and bioavailable.
  • the pharmaceutically acceptable carrier must be of sufficiently high purity and of sufficiently low toxicity to render it suitable for administration to the subject being treated. It further should maintain the stability and bioavailability of a therapeutic agent.
  • the pharmaceutically acceptable carrier can be liquid or solid and is selected, with the planned manner of administration in mind, to provide for the desired bulk, consistency, etc., when combined with an active agent and other components of a given composition.
  • Exemplary carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer'
  • Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E. W. Martin, which is incorporated herein by reference in its entirety.
  • the pharmaceutically acceptable carrier is sterile and pyrogen-free water.
  • the pharmaceutically acceptable carrier is Ringer’s Lactate, sometimes known as lactated Ringer’s solution.
  • Additives used with the pharmaceutical compositions described herein include, for example, one or more excipients, one or more antioxidants, one or more stabilizers, one or more preservatives (e.g., including antimicrobial preservatives), one or more pH adjusting and/or buffering agents, one or more tonicity adjusting agents, one or more thickening agents, one or more suspending agents, one or more binding agents, one or more viscosity enhancing agents, one or more sweetening agent and the like, either alone or together with one or more additional pharmaceutical agents, provided that the additional components are pharmaceutically acceptable.
  • preservatives e.g., including antimicrobial preservatives
  • the formulation may include combinations of two or more of the additional components as described herein (e.g., any of 2, 3, 4, 5, 6, 7, 8, or more additional components).
  • the additives include processing agents and drug delivery modifiers and enhancers, such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or more thereof.
  • processing agents and drug delivery modifiers and enhancers such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of
  • antioxidants examples include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin
  • Suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, tragacanth, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium stearate, water, and mineral oil.
  • the formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents.
  • compositions may be formulated so as to provide quick, controlled, sustained, or delayed release of the therapeutic agent after administration to the patient by employing procedures well known in the art.
  • unit dosage forms comprising the pharmaceutical composition and formulations described herein. These unit dosage forms can be stored in a suitable packaging in single or multiple unit dosages and may also be further sterilized and sealed.
  • Particulate Formulation [0254] According to another aspect, the described invention provides a particulate formulation comprising a plurality of particles. [0255] According to some embodiments, the particulate formulation comprises a plurality of particles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each particle, adsorbed onto the particle, or is in a core surrounded by a coating.
  • the particulate formulation comprises a plurality of microparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each microparticle, adsorbed onto the microparticles, or in a core surrounded by a coating.
  • the microparticles are formed by a microencapsulation process such that the outside of the microparticles contains the polymer, and the interior of the microparticles contains the drug in a semifluid state.
  • the particulate formulation comprises a plurality of nanoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each nanoparticle, adsorbed onto the nanoparticles, or in a core surrounded by a coating.
  • the particulate formulation comprises a plurality of picoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each picoparticle, adsorbed onto the picoparticles, or in a core surrounded by a coating.
  • the particulate formulation comprises a plurality of femtoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each femtoparticle, adsorbed onto the femtoparticles, or in a core surrounded by a coating.
  • the particulate formulation comprises a plurality of particles from about 1 nm to about 2000 nm, about 1 nm to about 1000 nm, about 1 nm to about 500 nm, inclusive, about 100 nm to about 300 nm, inclusive, or about 200 nm to about 500 nm, inclusive in particle size.
  • the particulate formulation comprises a plurality of particles comprising a particle size of about 1 nm, about 5 nm, about 10 nm, about 15 nm, about 20 nm, about 25 nm, about 30 nm, about 35 nm, about 40 nm, about 45 nm, about 50 nm, about 55 nm, about 60 nm, about 65 nm, about 70 nm, about 75 nm, about 80 nm, about 85 nm, about 90 nm, about 95 nm, about 100 nm, about 150 nm, about 200 nm, about 250 nm, about 300 nm, about 350 nm, about 400 nm, about 450 nm, about 500 nm, about 550 nm, about 600 nm, about 650 nm, about 700 nm, about 750 nm, about 800 nm, about 850 nm, about 900 nm, about
  • the particles of the particulate formulation are of a uniform distribution of particle size.
  • the uniform distribution of particle size is achieved by a non-emulsion-based homogenization process.
  • the uniform distribution of particle size is achieved by an emulsion-based process to form a uniform emulsion.
  • the particles of the particulate formulation have a varying particle size, e.g., ranging from 100- 2000 nm, inclusive.
  • the therapeutic agent is micronized.
  • the therapeutic agent e.g., micronized therapeutic agent
  • the therapeutic agent is disposed on or in the particles.
  • the therapeutic agent e.g., micronized therapeutic agent
  • the particles are impregnated with the therapeutic agent (e.g., micronized therapeutic agent).
  • the therapeutic agent e.g., micronized therapeutic agent
  • the therapeutic agent is adsorbed onto a surface of the particles.
  • the therapeutic agent e.g., micronized therapeutic agent
  • the particles comprise a matrix.
  • the matrix comprises the therapeutic agent (e.g., micronized therapeutic agent).
  • the matrix is impregnated with the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 5% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 10% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 15% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 20% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 25% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 30% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 35% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 40% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 45% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 50% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 55% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 60% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 63% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 65% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 70% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 75% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 80% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 85% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 90% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles are loaded with an average of at least 95% by weight of the therapeutic agent (e.g., micronized therapeutic agent).
  • the particles have a encapsulation efficiency (EE%) of the therapeutic agent (e.g., micronized therapeutic agent) of about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • microparticles of the present invention comprise about 0.1 ⁇ g to about 2 ⁇ g therapeutic agent (e.g., micronized therapeutic agent).
  • the microparticles of the invention may comprise about 0.1 ⁇ g, about 0.15 ⁇ g, about 0.2 ⁇ g, about 0.25 ⁇ g, about 0.3 ⁇ g, about 0.35 ⁇ g, about 0.4 ⁇ g, about 0.45 ⁇ g, about 0.5 ⁇ g, about 0.55 ⁇ g, about 0.6 ⁇ g, about 0.65 ⁇ g, about 0.7 ⁇ g, about 0.75 ⁇ g, about 0.8 ⁇ g, about 0.85 ⁇ g, about 0.9 ⁇ g, about 0.95 ⁇ g, about 1 ⁇ g, about 1.05 ⁇ g, about 1.1 ⁇ g, about 1.15 ⁇ g, about 1.2 ⁇ g, about 1.25 ⁇ g, about 1.3 ⁇ g, about 1.35 ⁇ g, about 1.4 ⁇ g, about 1.45 ⁇ g, about 1.5 ⁇ g, about 1.55 ⁇ g, about 1.6 ⁇ g, about 1.65 ⁇ g, about 1.7 ⁇ g, about 1.75 ⁇ g, about 1.8 ⁇ g, about
  • the therapeutic agent comprises a locally bioavailable COX-2 inhibitor.
  • the therapeutic agent formulated into the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) ( Figure 1C), or a derivative thereof.
  • the micronized therapeutic agent can be in liquid or solid form.
  • the micronized therapeutic agent is very slightly water soluble, moderately water soluble, or fully water soluble.
  • the therapeutic agent (e.g., micronized therapeutic agent) agent can include salts of the therapeutic agent.
  • the therapeutic agent e.g., micronized therapeutic agent
  • the therapeutic agent can be an acidic, basic, or amphoteric salt; it can be a nonionic molecule, a polar molecule, or a molecular complex capable of hydrogen bonding; or the therapeutic agent (e.g., micronized therapeutic agent) can be included in the compositions in the form of, for example, an uncharged molecule, a molecular complex, a salt, an ether, an ester, an amide, polymer drug conjugate, or other form to provide the effective biological or physiological activity.
  • the particles can be formulated for controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent), e.g., to the colon.
  • the microparticulate formulation is characterized by controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour to 24 hours, e.g., to the colon.
  • the microparticulate formulation is characterized by sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, or at least about 24 hours, e.g., to the colon.
  • the therapeutic agent e.g., micronized therapeutic agent
  • the particles can be of any order release kinetics, including zero order release, first order release, second order release, delayed release, sustained release, immediate release, and a combination thereof.
  • the particles can include any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof.
  • the particles can release a therapeutic agent by any of the following processes: (1) hydration or swelling of a matrix, (2) erosion of a matrix, (3) diffusion of a solution into the matrix; (4) dissolution of the drug; (5) diffusion of the dissolved drug out of the matrix, (6) pH-responsive release mechanism, and/or (7) microbially triggered delivery.
  • release may comprise a pH-responsive release mechanism.
  • a pH-sensitive drug delivery system e.g., microparticle, may withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ⁇ 0.4) or the ileocecal junction.
  • the particles are formulated to deliver a therapeutic agent to the colon.
  • the particles can be formulated for controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent), e.g., to the colon.
  • the microparticulate formulation is characterized by controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour to 24 hours, e.g., to the colon.
  • the microparticulate formulation is characterized by sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, at least about 24 hours, e.g., to the colon.
  • the therapeutic agent e.g., micronized therapeutic agent
  • Any biodegradable polymeric material can be used in the manufacture of particles for release of a therapeutic agent as described herein.
  • Such polymers may be natural or synthetic polymers.
  • the polymer may be selected based on the period of time over which release is desired and/or the location at which release is desired.
  • Exemplary criteria for selection of a polymer(s) for use in the described microparticulate formulations include, without limitation, the type of polymer, the selection of a copolymer, the pH-sensitivity of the polymer or copolymer, the type of co-monomers used in the copolymer, the ratio of the types of monomers used in the co-polymer, the molecular weight of the polymer, the size of the microparticle, and any other criteria used by one of skill in the art to control the release profile of a microparticle.
  • the particles may be polymeric pH-sensitive particles that are insoluble at a stomach low pH of about 1.5 to about 3.5, but are soluble at an intestinal neutral pH of about 6 to about pH 7.4.
  • Exemplary biocompatible biodegradable polymers useful for manufacturing the particles of the invention include, without limitation, a poly(lactide); a poly(glycolide); a poly(lactide-co-glycolide) (PLGA); a poly(lactic acid); a poly(glycolic acid); a poly(lactic acid-co-glycolic acid); poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F); a poly(vinyl alcohol) (PVA); a poly(caprolactone); a poly(orthoester); a polyanhydride; a poly(phosphazene); a polyhydroxyalkanoate; a poly(hydroxybutyrate); a poly(hydroxybutyrate) synthetically derived; a poly(hydroxybutyrate) biologically derived; a polyester synthetically derived; a polyester biologically derived; a poly(lactide-co- caprolactone); a poly(lactide); a
  • the particles may comprise poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the particles may comprise poly(lactide-co-glycolide) (PLGA). According to some embodiments, the particles may comprise poly(lactide-co-glycolide) (PLGA) and poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the particles may comprise Eudragit S100 pH-sensitive polymer and/or PLGA 5050.
  • the amount of lactide and glycolide in the polymer can vary.
  • the biodegradable polymer contains 0 to 100 mole %, 40 to 100 mole %, 50 to 100 mole %, 60 to 100 mole %, 70 to 100 mole %, or 80 to 100 mole % lactide and from 0 to 100 mole %, 0 to 60 mole %, 10 to 40 mole %, 20 to 40 mole %, or 30 to 40 mole % glycolide, wherein the amount of lactide and glycolide is 100 mole %.
  • the biodegradable polymer can be poly(lactide), 95:5 poly(lactide-co- glycolide) 85:15 poly(lactide-co-glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide), where the ratios are mole ratios.
  • the ratios are mole ratios.
  • any combination of the aforementioned biodegradable polymers can be used, including, but not limited to, copolymers, mixtures, or blends thereof.
  • Exemplary particulate formulations for the sustained release of 6A1 are provided in Table 7.
  • the particulate formulation is presented as a solution, an emulsion, a suspension of particles, or a powder (e.g., a lyophilized powder).
  • the particulate formulation can be in powder form for incorporation into a pharmaceutical composition, such as a capsule, of the invention.
  • Polymeric Coatings [0280] According to another aspect, the described invention provides pharmaceutical compositions for delivering a therapeutic agent to the colon of a subject comprising one or more polymeric coatings to control release, delay release, and/or sustain release of a therapeutic agent.
  • Any biodegradable polymeric material can be used in the manufacture of polymeric coatings for controlling, delaying, and/or sustaining the release of a therapeutic agent as described herein.
  • Such polymers may be natural or synthetic polymers.
  • the polymer may be selected based on the period of time over which release is desired and/or the location at which release is desired.
  • Exemplary criteria for selection of a polymer(s) for use in the described polymeric coatings include, without limitation, the type of polymer, the selection of a copolymer, the pH- sensitivity of the polymer or copolymer, the type of co-monomers used in the copolymer, the ratio of the types of monomers used in the co-polymer, the molecular weight of the polymer, the size of the microparticle, and any other criteria used by one of skill in the art to control the release profile of a microparticle.
  • the enteric coating may comprise any suitable pH-sensitive polymer.
  • anionic polymers with carboxyl groups have higher water solubility at basic pH than at acidic pH.
  • the enteric coating is insoluble at a stomach low pH of about 1.5 to about 3.5, but are soluble at an intestinal neutral pH of about 6 to about pH 7.4.
  • These polymers may be used for preventing gastric degradation of an therapeutic agent and to achieve delivery to the colon.
  • Exemplary biocompatible biodegradable polymers useful for manufacturing the particles of the invention include, without limitation, a poly(lactide); a poly(glycolide); a poly(lactide-co-glycolide) (PLGA); a poly(lactic acid); a poly(glycolic acid); a poly(lactic acid-co-glycolic acid); poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F); a poly(vinyl alcohol) (PVA); a poly(caprolactone); a poly(orthoester); a polyanhydride; a poly(phosphazene); a polyhydroxyalkanoate; a poly(hydroxybutyrate); a poly(hydroxybutyrate) synthetically derived; a poly(hydroxybutyrate) biologically derived; a polyester synthetically derived; a polyester biologically derived; a poly(lactide-co- caprolactone); a poly(lactide); a
  • the particles may comprise poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F).
  • the coating may comprise poly(lactide-co-glycolide) (PLGA).
  • the coating may comprise poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F).
  • the particles may comprise Eudragit S100 pH-sensitive polymer, PLGA 8515, and/or PLGA 5050.
  • the amount of lactide and glycolide in the polymer can vary.
  • the biodegradable polymer contains 0 to 100 mole %, 40 to 100 mole %, 50 to 100 mole %, 60 to 100 mole %, 70 to 100 mole %, or 80 to 100 mole % lactide and from 0 to 100 mole %, 0 to 60 mole %, 10 to 40 mole %, 20 to 40 mole %, or 30 to 40 mole % glycolide, wherein the amount of lactide and glycolide is 100 mole %.
  • the biodegradable polymer can be poly(lactide), 95:5 poly(lactide-co- glycolide) 85:15 poly(lactide-co-glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide), where the ratios are mole ratios.
  • Polylactic-co-glycolic acid (PLGA) and Eudragit S100 (ES100) are two commonly used pharmaceutical excipients that are generally recognized as safe (GRAS) by the United States FDA and the European Medicines Agency.
  • ES100 and PLGA polymers may be use for formulating enteric and time delayed coatings.
  • polymer degradation has been defined as the amount of time the material takes to degrade over its useful life time and nondegradation has been defined as breakdown that occurs after its useful life. In another way, the ratio between the time the polymer takes to degrade and the duration of the application of the polymer is used to determine whether or not the material is degradable (154).
  • biodegradable polymers contain hydrolysable bonds where the passive hydrolysis or enzyme-catalyzed hydrolysis breaks down the chemical bond between the copolymers. The hydrolysis degradation occurs via a random chain scission process to form oligomers and finally monomers (155). The enzyme-catalyzed hydrolysis also referred to as biodegradation is when a biological system is partially involved in the chemical bond cleavage.
  • Most synthetic biodegradable polymers, PLGA and ES100 mainly undergo passive hydrolysis (156).
  • the degradation rate is determined by its chemical bond, composition, the pH of the environment, the water content, and the swelling rate. Degradation begins with water uptake into the bulk of the polymer matrix, which leads to swelling. Next, hydrolysis occurs resulting in oligomers and monomers degrading from the polymer. Progressively, the degradation changes the microstructure of the bulk, forming pores, where the oligomers and monomers are released. Heterogeneous degradation refers to the surface degradation of the polymers whereas homogenous degradation describes a uniform break down of the bulk material throughout its matrix. [0290] Erosion is a more complicated process of polymer degradation in which additional parameters such as the swelling rate and material porosity are involved.
  • Poly (D,L-lactic-co- glycolic acid) three dimensional objects have been shown to exhibit an inverse flux erosion throughout the inner bulk material (154); once degradation begins increases in the degradation rate due to the lactide monomer content serves as an autocatalytic hydrolysis resulting in erosion (155, 157, 158).
  • the PLGA erosion rate can be optimized by adjusting the amount of porosity and the sizes of the micropores within PLGA matrix (159).
  • PLGA is an excellent biodegradable and biocompatible polymer (153, 160). Varying the ratios of lactic to glycolic acid controls the rate of water cleavage. Lactic acid contributes to the rigidity of the polymer backbone, thus increasing the duration that the polymer remains within a biological system (161).
  • PLGA 85:15 The primary mechanism for degradation of PLGA is the hydrolysis of the ester bond between the lactic acid and glycolic acid.
  • the PLGA 85:15 grade was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule.
  • PLGA 85:15 also allows for successive coating applications.
  • Eudragit S100 is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio. ES100 pH dependent nature were finely tune by the number of negative charge on the MAA’s COOH functional group at basic condition (162).
  • ES100 is soluble in alkaline digestive fluids by salt formation (163, 164).
  • a disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility ( Figure 38). However, the addition of plasticizer changes the dissolution profile and takes longer for the film to dry (165).
  • the polymeric coating may be applied to the capsule to any suitable thickness.
  • the polymeric coating may be applied to the capsule to a predefined thickness of about 1 nm to about 350 nm.
  • a delayed-release coating may be applied to the capsule to a thickness of about 115 ⁇ 35 nm.
  • the delayed- release coating delays the release of a therapeutic agent by at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, or at least about 24 hours after administration of the pharmaceutical composition to a subject.
  • an enteric coating may be applied to the capsule to a thickness of about 50 ⁇ 15 nm.
  • the enteric coating releases active agent at a pH above 5.5 (e.g., a pH between about 6 and about 8).
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be applied to the capsule to a thickness of about 1 nm, about 2 nm, about 3 nm, about 4 nm, about 5 nm, about 6 nm, about 7 nm, about 8 nm, about 9 nm, about 10 nm, about 11 nm, about 12 nm, about 13 nm, about 14 nm, about 15 nm, about 16 nm, about 17 nm, about 18 nm, about 19 nm, about 20 nm, about 21 nm, about 22 nm, about 23 nm, about 24 nm, about 25 nm, about 26 nm, about 27 nm, about 28 nm, about 29 nm, about 30 nm, about 31 nm, about 32 nm, about 33 nm, about 34 nm, about 35 nm, about 36
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to achieve a polymer a mass loading of about 7% to about 15%.
  • the polymeric coating (e.g., delayed- release coating and/or the enteric coating) may be formulated to achieve a polymer mass loading of about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 10.5%, about 11%, about 11.5%, about 12%, about 12.5%, about 13%, about 13.5%, about 14%, about 14.5%, or about 15%.
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores to control (e.g., accelerate and shorten) the degradation rate of a polymer from days into hours and precisely control its drug to deliver to the colon and/or large intestine.
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 1 nm to about 300 ⁇ m, inclusive.
  • the plurality of pores may be formed by doping and leaching the polymeric coating with a dopan porogen, such as sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), sodium acetate (NaCH3CO3), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat.
  • a dopan porogen such as sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), sodium acetate (NaCH3CO3), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat.
  • a dopan porogen such as sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), sodium acetate (NaCH3CO3), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat.
  • the percentage of the dry mass ratio of dopant porogen to polymer can be about 0.01%, about 0.05%, about 0.1%, about 0.15%, about 0.2%, about 0.25%, about 0.3%, about 0.35%, about 0.4%, about 0.45%, about 0.5%, about 0.55%, about 0.6%, about 0.65%, about 0.7%, about 0.75%, about 0.8%, about 0.85%, about 0.9%, about 0.95%, or about 1%.
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 100 ⁇ m to about 300 ⁇ m, inclusive (e.g., about 100 ⁇ m, about 105 ⁇ m, about 110 ⁇ m, about 115 ⁇ m, about 120 ⁇ m, about 125 ⁇ m, about 130 ⁇ m, about 135 ⁇ m, about 140 ⁇ m, about 145 ⁇ m, about 150 ⁇ m, about 155 ⁇ m, about 160 ⁇ m, about 165 ⁇ m, about 170 ⁇ m, about 175 ⁇ m, about 180 ⁇ m, about 185 ⁇ m, about 190 ⁇ m, about 195 ⁇ m, about 200 ⁇ m, about 205 ⁇ m, about 210 ⁇ m, about 215 ⁇ m, about 220 ⁇ m, about 225 ⁇ m, about 230 ⁇ m, about 235 ⁇ m,
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 1 nm to about 50 nm, inclusive (e.g., about 1 nm, about 2 nm, about 3 nm, about 4 nm, about 5 nm, about 6 nm, about 7 nm, about 8 nm, about 9 nm, about 10 nm, about 11 nm, about 12 nm, about 13 nm, about 14 nm, about 15 nm, about 16 nm, about 17 nm, about 18 nm, about 19 nm, about 20 nm, about 21 nm, about 22 nm, about 23 nm, about 24 nm, about 25 nm, about 26 nm, about 27 nm, about 28 nm, about 29 nm, about 30 nm, about 31 nm, about
  • the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores at a pore density of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%.
  • Therapeutic agent [0304] According to some embodiments, the described invention provides a therapeutic agent, which may be formulated into a particulate formulation or a pharmaceutical composition as described herein for colonic delivery. In certain embodiments, the therapeutic agent comprises a COX-2 inhibitor.
  • the therapeutic agent comprises 6A1 (4- [3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) ( Figure 1C), or a derivative thereof.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises a COX-2 inhibitor.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2- hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) ( Figure 1C), or a derivative thereof.
  • COX-2 inhibitors comprising a structural motif that makes them substrates for hepatic UDP- glucuronosyltransferases (UGTs) and sulfotransferases (SULTs).
  • the structural motif may be selected from the group consisting of a phenolic, an amine, an aliphatic alcohol, a carboxylic acid, or a sulfhydryl.
  • the therapeutic agent may be a locally bioavailable COX- 2 inhibitor comprising a structural motif that will subject the compound to rapid first-pass metabolism before reaching the systemic circulation which may limit its exposure to non- therapeutically targeted organs affected by toxicity, e.g., cardiovascular toxicity.
  • the COX-2 inhibitor may be tailored to have bioavailability only in a specific organ, e.g., the colon.
  • the COX-2 inhibitor is not systemically bioavailable, and therefore will not cause cardiovascular toxicity.
  • the COX-2 inhibitor may be used as a chemotherapeutic and/or a chemopreventive agents in certain targeted organs, e.g., the intestine and/or the colon.
  • the therapeutic agent comprises a locally bioavailable COX-2 inhibitor as described in WO/2017/172159, incorporated herein by reference.
  • the therapeutic agent may comprise a compound as defined by any one of Formulas 1-10 as described in WO/2017/172159.
  • the therapeutic agent may comprise a compound as defined by Formula 1 or any pharmaceutically acceptable salts thereof.
  • the therapeutic agent may comprise a compound as defined by Formula I, wherein R1 may be selected from sulfamyl, substituted sulfamyl, halo, alkyl, alkoxy, hydroxyl, and haloalkyl; and [0310] wherein R 2 may be selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoarylamido, ⁇ , ⁇ -dialkyla- mido, N-alkyl-N- arylamido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio, alkylsulfinyl
  • the therapeutic agent may comprise a compound as defined by Formula I, wherein R 3 may be selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoary larnido, ⁇ , ⁇ -dialkyla- mido, N-alkyl-N- arylarnido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio, alkylsulfinyl, alkyl- sulfonyl, N- alkylsulfamyl, N-arylsulfamyl, arylsulfonyl, ⁇ , ⁇ -dial
  • the therapeutic agent may comprise a compound as defined by Formula I, wherein R4 may be selected from aryl, cycloalkyl, cycloalkenyl and heterocyclic; optionally, wherein R4 may be substituted at a substitutable position with one or more radicals selected from halo, alkylthio, alkylsulfinyl, alkyl, alkylsulfonyl, cyano, carboxyl, alkoxycarbonyl, amido, N- monoalkylamido, N-monoarylamido, N,N- dialkylamido, N-alkyl- N-arylamido, haloalkyl, hydroxyl, alkoxy hydroxyalkyl haloalkoxy, sulfamyl, N- alkylsulfamyl, amino, methylthio, N- alkylarnino, ⁇ , ⁇ -dialkylamino, heterocyclic, nitro and acyl
  • the therapeutic agent may comprise a compound as defined by Formula I, wherein Rl is sulfamyl or substituted sulfamyl, wherein R2 is selected from phenyl with one or more hydroxyl, alkylphenyl with one or more hydroxyl, wherein R 3 is from hydrido, halo, wherein R4 is selected from aryl with substitute of methyl, methoxyl, halo, hydrido, or methylthio and pharmaceutically acceptable salts thereof.
  • a compound as defined by Formula I comprises any one of the compounds listed below, or a derivative thereof.
  • the therapeutic agent may comprise a compound as defined by Formula 2 or any pharmaceutically acceptable salts thereof.
  • R1 is selected from hydroxyl, amino, alkyl, carboxyalkyl, alkoxycarbonyl, aminocarbonyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy,
  • the therapeutic agent may comprise a compound as defined by Formula 3, wherein R 4 is sulfamyl or substituted sulfamyl, wherein R l is from hydrido or halo, wherein R2 is selected from aryl with substitute of methyl, methoxyl, halo, hydrido, or methylthio and pharmaceutically acceptable salts thereof as follows: [0328] wherein R1 is selected from alkyl, carboxyalkyl, alkoxycarbonyl, aminocarbonyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, aralkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, alkylthioalkyl
  • the therapeutic agent may comprise a compound as defined by Formula 4 or any pharmaceutically acceptable salts thereof, wherein R 1 is selected from hydroxyl, alkyl, carboxyalkyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycl
  • Formula 5 comprises compounds of Formula 4 wherein R l is selected from hydroxyl, lower alkyl, carboxyl, lower carboxyalkyl, lower aminocarbonylalkyl, lower alkoxycarbonylalkyl, lower aralkyl, lower alkoxyalkyl, lower aralkoxyalkyl, lower alkylthioalkyl, lower aralkylthioalkyl, lower alkylaminoalkyl, lower aryloxyalkyl, lower arylthioalkyl, lower haloalkyl, lower hydroxylalkyl, lower cycloalkyl, lower cycloalkylalkyl, and aralkyl [0337] when R 2 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazoly
  • R1 when R1 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically-acceptable salt thereof.
  • aromatic moiety such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically-acceptable salt thereof.
  • the therapeutic agent may comprise a compound as defined by Formula 5 or any pharmaceutically acceptable salts thereof.
  • the therapeutic agent may comprise a compound as defined by Formula 6 or any pharmaceutically acceptable salts thereof.
  • R1 is selected from the group consisting of (a) S(O)2CH 3 (b) S(O) 2 NH 2 (c) S(O)2NHC(O)CF 3 (d) S(O)(NH)CH 3 (e) S(O)(NH)NH 2 (f) S(O)(NH)NHC(O)CF 3 (g) P(O)(CH 3 )OH, and (h) P(O)(CH 3 )NH
  • R 2 is selected from the group consisting of (a) C 1-6 alkyl, (b) C 3 , C 4 , C 5 , C 6 , and C 1 cycloalkyl, (c) mono-, di- or tri-substituted phenyl or naphthyl wherein the substituent is selected from the group consisting of, (1) hydrogen, (2) halo, (3) C 1-6 alkoxy, (4) C 1-6 al
  • the therapeutic agent may comprise a compound as defined by Formula 7 or any pharmaceutically acceptable salts thereof, which comprises those compounds of Formula 6, [0350] wherein R l is sulfamyl or substituted sulfamyl, [0351] wherein R 2 is selected from hydroxyl, alkyl, carboxyalkyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkyl, hydroxy
  • the therapeutic agent may comprise a compound as defined by Formula 8 or any pharmaceutically acceptable salts thereof, [0354] wherein R1 is selected from methyl, ethyl, or any other alkyl, [0355] wherein R 2 is selected from halo, hydrogen, methoxyl, methyl, ethyl, trifluoromethyl when R 3 is hydroxyl (exclude position-4), [0356] wherein X is selected from N, O, C.
  • the therapeutic agent may comprise a compound as defined by Formula 9 or any pharmaceutically acceptable salts thereof, [0358] wherein R1 is selected from sulfamyl, substituted sulfamyl, halo, alkyl, alkoxy, hydroxyl, and haloalkyl, [0359] wherein R2 is selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoarylamido, ⁇ , ⁇ -dialkyla- mido, N-alkyl-N- arylamido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio,
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl- pyrazol-1-yl-] benzenesulfonamide) ( Figure 1C), or a derivative thereof.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in any suitable amount.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg to about 1000 mg.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg/kg to about 100 mg/kg.
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg, about 30 mg, about 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, about 41 mg, about 42 mg, about 43 mg
  • the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of at least about 0.001 wt%, at least 0.005 wt%, at least 0.01 wt%, at least 0.05 wt%, at least 0.10 wt%, at least 0.50 wt%, at least 1 wt%, at least 2 wt%, at least 3 wt%, at least 4 wt%, at least 5 wt%, at least 6 wt%, at least 7 wt%, at least 8 wt%, at least 9 wt%, at least 10 wt%, at least 15 wt%, at least 20 wt%, at least 25 wt%, at least 30 wt%, at least 35 wt%, at least 40 wt%, at least 45 wt%, at least 50 wt%, at least 55 wt%, at least 60 wt%, at least
  • the present disclosure investigated three different coating techniques, as described in the Examples, including dip coating, fluidic coating, and vacuum spin coating.
  • the parameters used to judge each technique were: smoothness of the film, reproducibility, and ease of clean up. Of the three, vacuum spin coating yielded the most optimal results.
  • the described invention provides vacuum spin coating techniques and formulations to delay the release of therapeutic agents targeting the colon.
  • the controlled coating technique results in an appropriate coat thickness, which combined with an optimal formulation of biodegradable polymers that will protect and release the therapeutic agent in a programmable manner (e.g., delayed beyond 6 hours and sustained at a pH above 6.0).
  • Vacuum spin coating is one of the most common methods for applying thin films to substrates (166). A wide variety of technology sectors and industries commonly use vacuum spin coating. Vacuum spin coating's advantage is its ability to rapidly and easily produce consistently uniform films, ranging from a few microns down to a nanometer of thickness.
  • the vacuum spin coater apparatus works based on two physical properties. The first was an application of a vacuum that creates a shear force to facilitate the spread of the non-Newtonian polymers throughout the 3-dimensional capsules. Secondly, the capsules were spun. As the capsule accelerates in rotation, it expulses all the undissolved polymer. Once the spin coater's RPM reaches its desired speed and the polymer fluid was sufficiently thinned out, viscous shear drag exactly balances the rotational accelerations, drying begins.
  • the capsule was spun at a constant rate, 500 RPM, and solvent evaporation dominates the coating thinning behavior.
  • the rate of evaporation depends on two factors (a) the difference in the partial pressure of solvent evaporation between the free surface of the liquid layer and the gas layer flowing nearby (surface tension) and (b) the capillary forces developing in the channels between the latex particles.
  • the dissolved polymers solidified at the liquid-air interface in a highly viscous, lowly diffusive layer forming an unbroken film on the skin of the capsule (151).
  • the present invention provides an adaptor that could sidestep the standard low mass loading of 3% and instead use a 10% mass loading.
  • the adaptor may be sized to fit any conventional magnetic spinner and vacuum.
  • the adaptor may be used to coat both the aqueous and organic solvent-based systems because it does not require a spraying nozzle to apply the solvent to the substrate.
  • the spin vacuum adaptor does not need high heat as a curing process since the spinning creates an air vortex within each pod that facilitates solvent evaporation.
  • the vacuum spin coater was also designed to produce individualized small-scale batches consisting of 8 or 16 capsules. Currently, the coating success rate is around 80%, meaning that for every quantity of eight capsules, one will fail the visual, weight, or in vitro dissolution test.
  • the spin vacuum design resolves the shortfalls of the conventional coating techniques mentioned above, which does not have the capability to run pilot testing of a drug or produce individually tailored medicines where the pharmacokinetics of the drugs can be controlled and modified. It was discovered that the thickness of the coating layers was controllable by adjusting two parameters of the spin vacuum coater. First, the total suction applied by the vacuum facilitates the surface creeping of 10% of the material. The force exerted onto the capsule by the vacuum also spreads the formulation evenly to the gap and covers the junction between the cap and body of the capsule.
  • the spinning rate of the machine creates a centrifugal shearing force that facilitates the expulsion of chunky or undissolved polymer and also thins out the coating material.
  • the total suction of the vacuum and spinning rate precisely control the optimal thickness of the PLGA and ES100.
  • the described invention provides a method of spin coating a capsule, comprising: (i) providing a vacuum spinning plate comprising individual pods; (ii) providing sealed capsules wetted with a mild basic water (e.g., pH 9 NaOH 0.1 mM) loaded into the individual pods within the vacuum spinning plates; (iii) providing an amount of a first polymer dissolved in a solvent; (iv) applying a vacuum; (v) spinning the plate a first time for about 30 seconds at a spinning speed of 100 RPM; (vi) spinning the plate a second time for about 15 minutes at spinning speed of 500 RPM; (vii) providing an amount of a second polymer dissolved in a solvent; (viii) applying a vacuum; (ix) spinning the plate a third time for about 30 seconds at spinning speed of 100 RPM; (x) spinning the plate a fourth time for about 15 minutes at a spinning speed of 500 RPM; (xi) removing the capsule from the spinning plate; (xii)
  • a mild basic water e
  • the first polymer is PLGA8515.
  • the solvent for PLGA8515 is methylene chloride.
  • the concentration is 10% w/v solution PLGA8515.
  • a dopant porogen can be added to the premade PLGA polymer that is dissolved in methylene chloride.
  • the dopan porogen can be sodium percarbonate (Na2H 3 CO6), sodium bicarbonate (NaHCO 3 ), sodium carbonate (NaCO 3 ), sodium acetate (NaCH 3 CO 3 ), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat.
  • the concentration of dopant can be 1% v/v to 20% v/v of the final solution.
  • the concentration of dopant can be about 1% v/v, about 2% v/v, about 3% v/v, about 4% v/v, about 5% v/v, about 6% v/v, about 7% v/v, about 8% v/v, about 9% v/v, about 10% v/v, about 11% v/v, about 12% v/v, about 13% v/v, about 14% v/v, about 15% v/v, about 16% v/v, about 17% v/v, about 18% v/v, about 19% v/v, or about 20% v/v.
  • the percentage of the dry mass ratio of dopant porogen to polymer can range from about 0.1% to about 1.0% (e.g., about 0.03% to about 0.07%). According to some embodiments, the percentage of the dry mass ratio of dopant porogen to polymer can be about 0.01%, about 0.05%, about 0.1%, about 0.15%, about 0.2%, about 0.25%, about 0.3%, about 0.35%, about 0.4%, about 0.45%, about 0.5%, about 0.55%, about 0.6%, about 0.65%, about 0.7%, about 0.75%, about 0.8%, about 0.85%, about 0.9%, about 0.95%, or about 1%.
  • sodium citrate at 5% w/w as a doping agent creates a pore size of 180- 250 ⁇ m, inclusive, in the PLGA network. While sodium chloride at room temperature to 400°C can consistently forms a pore size of 17-20 nm, inclusive, in diameter.
  • An exemplary, working sodium chloride salt concentration as dopant is 2.5% v/v to 10% v/v of the final solution (e.g., 37.5 ⁇ L NaCl/methanol into 1462.5 ⁇ L of pre-dissolved PLGA8515/methylene chloride polymers to 75 ⁇ L NaCl/methanol into 1425.0 ⁇ L of pre-dissolved PLGA8515/methylene chloride).
  • the second polymer is Eudragit S100.
  • the solvent for Eudragit S100 is a mixture of acetone: isopropanol alcohol: DI water in the ratio of 86:12:2. In certain embodiments, the concentration is 10% w/v solution Eudragit S100.
  • hard gelatin capsules were packed with raw drug or microparticles; the capsules were then coated with PLGA 8515 to a thickness of 115 ⁇ 35 nm thickness and an outer pH-sensitive Eudragit S100 coat of 50 ⁇ 15 nm thickness to achieve colonic delivery. The new vacuum spin coating technique achieved an 88% success rate (410 of 450 capsules made passed the quality assurance tests).
  • the new coating technique was able to coat high viscosity polymers, PLGA8515 and ES100, with a high mass loading of 10% w/w formulation and sufficiently covered the entire three dimensional nine millimeters hard gelatin capsules (S9C).
  • the novel spin coating technique is an innovative coating approach that had not been accomplished prior to this invention.
  • the vacuum applied to the bottom of the capsules placed inside its respective slot within the coater plate creates a shear force that thinned out the polymers, which are behaving as non-Newtonian fluids.
  • the thinning is due to the entanglement of polymer collapsing in the direction that vacuum is applied (bottom of the plate).
  • the angle at which the capsule sits causes it to spin around as the centrifugal force resulting from the spinning of the coater plate.
  • the centrifugal force is large enough to spin out the large undissolved polymers and creates a vortex of air that facilitates further evaporation.
  • the combination of vacuum and spinning creates a micro-environment that has a conical vortex within each capsule pod.
  • the method of coating capsules according to the invention is advantageous compared to conventional coating methods.
  • the conventional method is for a large quantity of hard capsules filled with a drug to be charged into an apparatus which is then heated and rotated either under vacuum or atmospheric pressure.
  • the invention uniquely allows for small batch production as is desired in research and development labs. Small batches are desirable for the production of personalized medications.
  • the cost of the invention is significantly less.
  • the quality of the capsule coating is superior in enteric and acid-resistant characteristics and the delayed release mechanism compared to conventional coatings rapidly disintegrated in intestinal fluid.
  • the quantities of the coating agent, active ingredients, and fillers can be reduced; the products obtained are of higher quality, and the production cost for both researching laboratory and clinical setting are reduced.
  • the invention allows for the delayed burst release of the capsule to specific region in the GI track is tunable through the controlling the coating thickness and the formulation of polymers.
  • FAP Familial Adenoma Polyposis
  • COXIBs Selective COX-2 inhibitors
  • COXIBs are the most versatile group of drugs prescribed for inflammation, analgesic/antipyretic, and auto-immune diseases and are undeniably effective in the treatment and prevention of polyposis formation.
  • COXIBs that circulate systemically inhibit COX-2 in on target off organs causing possible cardiovascular side effects (e.g., unstable angina, myocardial infarction, and cardiac thrombus) at the doses required for efficacy as a prophylactic (6, 16, 25-27), COXIBs cannot currently be used clinically to treat FAP. COXIBs are taken orally and absorbed through the stomach wall leading to systemic circulation. Additionally, COXIBs do not actively enter the EHR, thus large and frequent doses are required to reach a therapeutic level in the colon. [0383] Celecoxib is a selective COX-2 inhibitor class of NSAIDs that is undeniably effective against FAP.
  • a new COX-2 inhibitor, 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C)
  • EHR enterohepatic recycling
  • EHR delivered a variable quantity of 6A1 to the target tissue, colon, subject to variation amongst individuals UGT enzyme expression and ß-glucuronidase expression within the bacteria microbiomes that inhabit the gut.
  • the present disclosure aims to lower the therapeutic dose and the total systemic exposure (AUC) while increasing concentration of a selective COX-2 inhibitor in the colon by capitalizing upon the complementary interplay between the EHR, coating techniques, and biodegradable polymer formulations.
  • the described invention utilizes EHR as a means to target drug delivery to the colon. New chemical entities that undergo active glucuronidation via phase 2 metabolism are often eliminated early during the research and development phase due to an insufficient quantity of drug going to systemic circulation and ultimately inadequate therapeutic outcomes due to low exposure (39, 40).
  • the present invention focuses on a novel class of COX-2 inhibitors that are actively glucuronidated by the UGT1A1 enzyme, which is a subclass of phase II metabolic enzymes found in abundance in the livers. Because the glucuronides of 6A1 undergo EHR, gut microbiomes can successfully reconvert glucuronides back into aglycone, which becomes bioavailable again after absorption into the colon cells, thus reducing the systemic drug circulation while increasing their exposure in the colon.
  • the described invention provides, According to some embodiments, formulation and coating of 6A1 packed inside hard gelatin capsules to ensure that the initial dose, at the correct concentration, will be delivered to the colon and sufficiently saturate the COX-2 enzymes, yielding a consistent C max and T max value across all populations. [0387] Specific modes of administration will depend on the indication.
  • the selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response.
  • the amount of active agent to be administered is that amount sufficient to provide the intended benefit of treatment.
  • the dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular mammal or human treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician).
  • the pharmaceutical composition may be administered once, for a limited period of time or as a maintenance therapy over an extended period of time, for example until the condition is ameliorated, cured or for the life of the subject.
  • a limited period of time may be for 1 week, 2 weeks, 3 weeks, 4 weeks and up to one year, including any period of time between such values, including endpoints.
  • the pharmaceutical composition may be administered for about 1 day, for about 3 days, for about 1 week, for about 10 days, for about 2 weeks, for about 18 days, for about 3 weeks, or for any range between any of these values, including endpoints.
  • the pharmaceutical composition may be administered for more than one year, for about 2 years, for about 3 years, for about 4 years, or longer.
  • the composition or pharmaceutical composition may be administered once daily, twice daily, three times daily, four times daily or more.
  • the described invention provides use of a particulate formulation or pharmaceutical composition in the preparation of a medicament for reducing the severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP).
  • FAP familial adenomatous polyposis
  • the described invention provides use of a particulate formulation or pharmaceutical composition in the preparation of a medicament for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP).
  • FAP familial adenomatous polyposis
  • the described invention provides a method for reducing severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing a particulate formulation or pharmaceutical composition as described herein, and (b) administering an effective amount of the a particulate formulation or pharmaceutical composition orally to the subject.
  • the described invention provide a method for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing a particulate formulation or pharmaceutical composition as described herein, and (b) administering an effective amount of the pharmaceutical composition orally to the subject.
  • the pharmaceutical composition may be administered with an additional therapeutic agent and/or an additional treatment modality.
  • additional therapeutic agent include, without limitation, non-steroidal anti- inflammatory drugs (NSAIDs), polyamine transporter inhibitors, eIF-5A antagonists, chemotherapeutic agents, radiotherapy, and immunomodulatory agents.
  • the dosing frequency of the pharmaceutical composition and the additional pharmaceutical agent may be adjusted over the course of the treatment based on the judgment of the administering physician.
  • the pharmaceutical composition and the additional therapeutic agent can be administered at different dosing frequency or intervals.
  • the pharmaceutical composition can be administered weekly, while the additional therapeutic agent can be administered more or less frequently.
  • a sustained continuous release formulation of the pharmaceutical composition and/or the additional therapeutic agent may be used.
  • Various formulations and devices for achieving sustained release are known in the art.
  • a combination of the administration configurations described herein can be used.
  • the pharmaceutical composition can be administered daily and the additional therapeutic agent can be administered monthly.
  • the pharmaceutical composition can be administered weekly and the additional therapeutic agent can be administered monthly.
  • Example 1 A sensitive and validated ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) method for quantifying a newly synthesized COX- 2 inhibitor (6A1) and its metabolites in blood, liver, and colonic mucosa of F344 rats Introduction [0400] Studies have shown that cyclooxygenase-2 (COX-2) enzyme is elevated in colorectal polyps.
  • COX-2 cyclooxygenase-2
  • a newly synthesized COX-2 inhibitor, 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide), is comparable in its performance to that of celecoxib (4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol- 1- yl]-benzenesulfona-mide), with a potential of lower on-target off-organ systemic toxicity due to low systemic exposure (113).
  • Acrodisc syringe filter with a polytetrafluoroethylene (PTFE) membrane pore size of 0.2 um and diameter of 25 mm were purchased from Pall Corp. (Port Washington, NY).
  • Oasis solid phase extraction (SPE) 5 cc cassette were purchased from Water Corp. (Milford, MA).
  • the parent compound, 6A1 was synthesized in house and quantified as 98% pure using the UPLC and NMR.
  • 6A1 sulfate and 6A1 glucuronide are biosynthesized using F344 rat microsomes and MDCK II -UGT1A1-MRP2- overexpressed cells.
  • Acetonitrile, methanol, 0.1-5% formic acid in acetonitrile, 0.1 - 5% formic acid in methanol as organic phase, 1 - 2.5 mM ammonium acetate in water, and 0.05 - 1% formic acid in water as aqueous phase were tested as potential mobile phases.
  • This experiment tested both the Restek Raptor biphenyl and the Aquity HSS T3 columns as stationary phases to resolve 6A1, 6A1 sulfate, and 6A1 glucuronide.
  • the gradient elution was as follows: 0.1-.5 min 100% B; 0.5-1.5 min, 65% B; 1.5- 2.5 min, 30% B; 2.0-3.0 min 15% B; 3.0 - 4.0 min, 10% B; 4.0 – 6.0 min, 95% B.
  • the column temperature of 45°C, sample temperature of 20°C, injection volume of 10 ⁇ L, and a flow rate of 0.20 mL/min achieved the best peak sharpness and symmetry.
  • Peak Resolution (time peak 2 eluted – time peak 1 eluted) / (0.5)(width peak 2 + width peak 1 )
  • Mass Spectrometry Conditions [0408] For MS/MS analysis, both positive and negative scan modes were employed to analyze 6A1, 6A1 glucuronide, and 6A1 sulfate. Representative MRM spectra of 6A1, 6A1 sulfate, and 6A1 glucuronide are shown in Figure 8. The analytes’ intensity in positive scan mode is more sensitive for 6A1 and 6A1 glucuronide compared to the negative scan mode.
  • MRM multiple reactions monitoring
  • Standard solutions of 6A1, 6A1 glucuronide, 6A1 sulfate, and IS were prepared separately at 100 ⁇ g/mL in methanol. Each standard solution was then fractioned into multiple mini-Eppendorf tubes and stored at -80°C. Each standard solution was diluted with 1:2 water- methanol solution into the final working solution of 1 ⁇ g/mL on the same of day usage and discarded after each use.
  • the calibration standard solution was prepared via serial dilution of working solution to achieve the following concentrations: 0.25, 0.5, 1, 2, 3.91, 7.81, 15.63, 31.25, 62.5, 125, 250, 500, 1,000, and 1,400 ng/mL.
  • Blood Samples [0410] Blank blood samples (40 ⁇ L) were spiked with 10 ⁇ L of calibration standard solution at different concentrations of analytes and 10 ⁇ L IS solution (50 ng/ml in 50% methanol in water). Each sample was extracted with 1,500 ⁇ L of acetonitrile after vortexing for 3 minutes. All samples were centrifuged at 20,000 x g for 15 minutes at 5°C.
  • the supernatant was then transferred to another tube and evaporated to dryness under a steady stream of air at room temperature.
  • the residue was reconstituted with 40 ⁇ L of 66% methanol in water (2:1), vortexed, and centrifuged again at 20,000 x g for 5 minutes. After centrifugation, 10 ⁇ L of the supernatant was injected into UPLC-MS/MS system for analysis. For pharmacokinetic samples, spiking of the calibration standard solution was omitted.
  • Liver and Colonic Mucosa Samples Tissue Homogenization [0411] Liver tissue and colonic mucosa were excised from the sacrificed rats, washed with cold HBSS solution, and stored at ⁇ 80°C in polypropylene tubes until homogenization. The frozen tissues were thawed, chopped and weighed. Exactly 50 mg of chopped tissue was homogenized in 500 ⁇ L of ice-cold homogenizing solution (pH 7.4) containing 10 mM potassium phosphate, 250 mM sucrose, and 1 mM EDTA dehydrate with a polytron tissue homogenizer. The homogenizer probe and test tubes were chilled at -4°C prior to use.
  • homogenization was paused for 20 seconds after every 30 seconds of homogenization at a medium speed. The homogenization was repeated 3 times until a visually uniform homogenate was obtained. Final tissue extract was stored at approximately ⁇ 80°C prior to analysis. The homogenizer probe was washed sequentially with water, methanol, and water after every homogenization.
  • the eluents were collected and dried under a steady stream of air at room temperature.
  • the residue was reconstituted with 40 ⁇ L of methanol in water (2:1 v/v), vortexed, and centrifuged again at 20,000 x g for 5 min. After centrifugation, 10 ⁇ L of the supernatant was injected into the UPLC-MS/MS system for analysis.
  • the CPS readout of 10 ⁇ L injection of the extracted tissue with 1 mg/ ⁇ L tissue sample was 5.42E 6 over the internal standard’s CPS of 1.66E 6 are normalized to give a ratio of 3.26.
  • the normalized ratio is to avoid instrumental drift between each injection, thus 3.26 divided by the slope of the standard curve gives a concentration of 2,506.95 ng (1 mg/ ⁇ L ⁇ ng/1000 ⁇ L) of 6A1 per gram of colonic mucosa.
  • the LOD decided based on the 5:1 signal-to-noise ratio was 0.5 ng/mL, thus the lower limit of quantification (LLOQ) would be 2 ng/mL.
  • Specificity and Selectivity [0418] The metabolites, sulfate and glucuronide, were detected via negative and positive ion mode. Their respective peaks were not overlaps in the elution time.
  • the base line separation between each analyte to the IS peak were set at equal or greater than 1.5-fold.
  • Accuracy and Precision [0419] The “intra-day” and “inter-day” precision and accuracy of the method were determined with quality control (QC) samples at three different concentrations of 20, 200, and 1000 ng/mL (sextuplet per set) and the lower limit of quantification (LLOQ) of 2 ng/mL.
  • QC quality control
  • LLOQ lower limit of quantification
  • Accuracy is to determine how close the empirical peak analyte normalized by internal standard peak ratios divided by standard curve slope and theoretical nominal value are to each other (Equation 1). This value is expressed in percentage with 100% being the same. Precision is the percent of coefficient of variation within the group and within different group measured (Equation 2).
  • Extraction recovery of 6A1, 6A1 sulfate, and 6A1 glucuronide in different bio- matrices were calculated by plotting the ratio of the peak areas of analyte (6A1, 6A1 glucuronide, and 6A1 sulfate) to IS (Baohuaside) in blank matrix spiked after the extraction procedure divided by the ratio of the peak areas of analyte to internal standard for the same matrix before the sample preparation step of liquid- liquid extraction (LLE) and solid phase extraction (SPE) (Equation 3).
  • Response of non-extracted analyte is the normalized response of all 3 compounds (6A1, 6A1 glucuronide, and 6A1 sulfate) divided by IS (Baohuaside) in neat solution.
  • Stability [0421] Short-term, post-processing (25°C for 8 h), 3 freeze thaw cycles, and long-term ( ⁇ 80°C for 1 month) stability of 6A1, 6A1 sulfate, and 6A1 glucuronide was determined by analyzing three replicates of lower limit of quantification (LLOQ) and three QC samples for all three matrices. The stability sample results were compared to the freshly prepared samples to determine the stability of each condition.
  • LLOQ lower limit of quantification
  • 6A1 Intravenous Injection Solution 0.25 w/v% solution of 6A1 was prepared in 40 v/v% of polyethylene glycol 300 and 60% v/v of ethanol. The solution was sonicated for five minutes until it became clear. The solution was further filtered using a 0.2 ⁇ m PTFE filter disc before injecting it into the rats.
  • Rats were kept in an environmentally controlled room (temperature: 25 ⁇ 2°C, humidity: 50 ⁇ 5%, and 12 h dark- light cycle) for at least 1 week before the experiments.
  • the rats were fed ad libitum.
  • the solution of 6A1 was administered via intravenous injection of the tail vein at a dose of 5.0 mg/Kg.
  • Blood samples (about 20 –50 ⁇ L) were collected into heparinized tubes at 0, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post dosing via tail snip with isoflurane as anesthetic. All collected pharmacokinetic blood samples were stored at ⁇ 80°C until analysis. After a wash out period of 1 week, the rats were given a second dose via intravenous injection of 5 mg/Kg.
  • the HSS T3 column had better analytes retention resulting in enhanced sensitivity using the same gradient elution method.
  • the HSS T3 column also had a lower column pressure (6000 psi) compared to the biphenyl column (14,000 psi).
  • the calibration curves for 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, liver, and colonic mucosa matrixes are linear in the range of 0.5-1,400 ng/mL for blood, 0.5-1,400 ng/g for liver, and 0.5-1,400 ng/g for colonic mucosa matrices (Table 2).
  • Linear regression equations were calculated from the raw data of the Count Per Second (CPS) response versus concentration graph with a weighting factor of 1/X 2 .
  • the resulted coefficient of determination, R 2 for all regression lines were > 0.995 (Table 2).
  • the extraction recovery for liver tissue of LLOQ and three QC samples were between 93.6-95.9% for 6A1, 109-112% for 6A1 glucuronide, and 98-108.6% for 6A1 sulfate (Table 4).
  • the colonic mucosa extraction recovery for the above LLOQ and QC samples were 99.4-103.8% for 6A1, 91.71-98.45% for 6A1 glucuronide, and 94.1-103.9% for 6A1 sulfate (Table 4).
  • the extraction recovery of colonic mucosa had the highest %CV at low concentration (20 ng/g); however, all LLOQ and QC samples were reproducible judging from the small %CV between groups and were within the recommendation of the FDA guidance (Table 4).
  • Matrix Effect An ion enhancement (15.99%) in the response signal was observed for the lower limit of quantification (LLOQ) of colonic mucosa matrix. However, the effect was resolved after the additional application of a 96-well Oasis HLB ⁇ -elution plate. The overall matrix effect for blood samples ranged from 92-108%, 94.5-108.88% for colonic mucosa, and 88- 113% for liver tissue (Table 5). The internal standard matrix effect is negligible thus comprehensive matrix effect was not evaluated.
  • Stability of 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, colonic mucosa, and liver tissue was evaluated by analyzing triplicates of LLQC and QC samples at three different concentrations following 8 h at 25°C bench top (short term stability), 3 freeze thawing cycles, and at ⁇ 80°C for 30 days ( ⁇ 80°C and 25°C). All the samples showed 95–105% recoveries after various stability tests (Table 6).
  • a double peak phenomenon was observed in the pharmacokinetic study of blood concentration-time profiles of 6A1 and 6A1 glucuronide. This indicated that the enterohepatic recirculation may be involved in the disposition of 6A1.
  • a second peak of 6A1 at 258.47 ⁇ 100.35 ng/mL was observed five hours after the dose was administered.
  • the first 6A1 glucuronide peak (at 288.16 ⁇ 94.88 ng/mL) was observed two hours post dose, whereas a second 6A1 glucuronidation peak with concentration of 183.08 ⁇ 52.71 ng/mL was observed at four hours after intravenous injection.
  • Colonic mucosa had an average ⁇ SEM concentration of 225.94 ⁇ 65.94 ng/g for 6A1, 26.78 ⁇ 17.80 ng/g for 6A1 glucuronide, and 5.12 ⁇ 0.86 ng/g for 6A1 sulfate (Figure 11A).
  • Liver tissues had an average concentration of 337.60 ⁇ 100.27 ng/g for 6A1, 315.79 ⁇ 81.55 ng/g for 6A1 glucuronide, and 218.14 ⁇ 54.21 ng/g for 6A1 sulfate (Figure 11B).
  • Table 1 Compound dependent parameters 6A1, 6A1 glucuronide, IS, and 6A1 sulfate parameters in MRM mode for LC-MS/MS
  • Table 2 Linearity of the standard calibration curve of 6A1, 6A1 glucuronide, 6A1 sulfate in blood, colonic mucosa, and liver tissue (Mean ⁇ SD (%CV))
  • Table 3 Intraday and Inter-day accuracy (Mean ⁇ SD) and precision (%CV) data of 6A1, 6A1 glucuronide, and 6A1 sulfate for blood matrix using MRM method at three QC concentrations ⁇ 287 ⁇ 571 ⁇ 141 ⁇ 8.67
  • Table 4 The extraction and recovery (Mean ⁇ SD (%CV)) of 6A1, 6A1 glucuronide, and 6A1 sulfate compounds for blood, colonic mucosa and liver tissue at lower limit of quantification (LLOQ) and three QC concentrations
  • Table 5 Mat
  • Table 6 Stabilities of 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, colonic mucosa, and liver tissue were evaluated by analyzing triplicates of LLQC and QC samples at three different concentrations following A) 8 h at 25°C bench top (short term stability), B) 3 freeze thawing cycles, C) stored at ⁇ 80°C for 30 days ( ⁇ 80°C and 25°C), and D) processed sample, autosampler, 20°C for 24 h A) 8 h at 25°C bench top (short term stability)
  • Example 2 Biodegradable coating formulation and vacuum spinning technique applied to size 9 hard gelatin capsules to deliver a selective COX-2 inhibitor to the colon of F344 rats
  • 6A1 a newly-designed selective COX-2 inhibitor
  • the purpose of this study was to use a newly-designed selective COX-2 inhibitor (6A1) in combination with a coating formulation and technique to deliver a high drug concentration to the colon.
  • Drug powder and image tracers were packed inside size nine hard gelatin capsules (S9C), coated with an inner layer of polylactic glycolic acid (PLGA 85:15) with optimum thickness of 115 ⁇ 35 nm for PLGA 8515 and 50 ⁇ 15 nm for Eudragit S100 (ES100) to serve as delayed-release coating via erosion mechanism and an outer pH-sensitive ES100 layer to serve as an enteric coating.
  • PLGA 85:15 polylactic glycolic acid
  • ES100 Eudragit S100
  • the ex vivo tissue stain showed that the capsule released its contents at the distal gastrointestinal tract, i.e., the colon. Live in vivo images obtained also further validated the S9C released in the colon.
  • the PLGA 85:15 also referred to herein as “PLGA 8515” grade coating was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule. PLGA 85:15 also allows for successive coating applications.
  • Eudragit S100 (ES100) is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio.
  • ES100’s pH dependent nature was finely tuned by the number of negative charges on the MAA’s COOH functional group at basic conditions (162).
  • the hydrophobicity of MMA units was realized by synthesizing co-polymer libraries with a systematic variation.
  • ES100 is soluble in alkaline digestive fluids by salt formation (163, 164).
  • a disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility (Figure 38).
  • Size 9 capsules were purchased from Torpac Inc. (Torpac Custom capsules & capsule fillers, 333 Route 46, Fairfield, NJ 07004, USA).
  • the parent compound, 6A1, and 6A1 sulfate were synthesized in house and quantified at 98% pure using the UPLC and NMR.
  • 6A1 glucuronide was biosynthesized using Hela-UGT1A9 cells, purified, and quantified at 98% pure using the previously established methods.
  • Preparation of Capsules [0444] Hard gelatin capsules were visually inspected for shape and coloration, then weighed and selected for use if their weights were within 2 - 5% of the expected range.
  • Drug powder (with 5% w/w barium sulfate 5% w/w and methyl blue) was packed into the capsules using the funnel and pestle kit sold by the vendor. The total weight of capsules and drug blend was monitored within a 3% range of drug packing of 20 mg. A trace amount of pH 9 water ( ⁇ 2 ⁇ l via a pipette tip) was used to seal the body and cap. The sealed capsules were allowed to dry for 24 hours before any coating materials were applied (Figure 12).
  • the capsule was sufficiently dried to be transferred into an individual micro-Eppendorf vial with the lid left open and allowed to stay in a desiccator for complete drying over 24 hours (Figure 14).
  • Appearance and Quality Control Parameters of Coated Capsules [0447] After each successive coating, a visual inspection of individual capsules was performed using a 3.5X-90X zoom trinocular microscope (AmScope, SM-1TSZ-V203); close observations were made to look for wrinkles, craters, and asymmetry ( Figure 15). By comparing the coated and uncoated S9C, a different sheen was observed. The coated capsules were weighed, and the total mass gain was then used to calculate the estimated thickness. The predicted thickness of the capsule was based on the equation below.
  • the capsules were rejected if they were not within the ⁇ 2.5% of the expected thickness (150 nm) based on mass gained.
  • Thermogravimetric Analysis (TGA) [0448] Films of PLGA and ES100 applied to capsules an hour prior were peeled and stamped to fit the aluminum pan. Approximately 10 ⁇ 1 mg of each polymer film per sample was used. Randomized, triplicate films were tested. The TGA curves (Shimadzu, TGA50-H) acquired from temperature ramping rate were 10 °C min ⁇ 1 from room temperature ( ⁇ 20°C), held for 30 seconds at 30°C, and steady ramping till 500°C. Formulation consistency across all formulation batches was determined via the % weight loss curve.
  • DSC Differential Scanning Calorimetry
  • SEM Scanning Electron Microscopy
  • the in vitro dissolution test for the coated capsules was performed in accordance with the recommendation from UPS compendial dissolution, non-sink conditions (168).
  • the simulated intestinal pH was adjusted via 2.5 ⁇ mol / L of HCl or NaOH from the 2.5 ⁇ mol / L of phosphate buffer. 1 mL of samples were collected every half hour, and the pH exposure and duration were as follows: 1.5 hours at pH 1.2 (stomach), 2.5 hours at pH 4.6 (small intestine), 2 hours at pH 6.8, 2.5 hours at pH 8, and 2.5 hour at pH 6.8.
  • Each capsule was placed within a sinker housing, and the stirrer rate was set at 80 rpm (169) at 37 ⁇ 2 °C.
  • Blood samples (about 20 –50 ⁇ L) were collected into heparinized tubes at 0, 0.5,1, 3, 5, 7, 10, and 24 hours post-dosing via tail snip with isoflurane as an anesthetic. All collected pharmacokinetic blood samples were stored at ⁇ 80 °C until analysis.
  • a Perkin Elmer IVIS Lumina III XRMS in X-ray (40 kVp, 100 mAmps, 10 seconds) and photograph modes was used to image the capsules containing barium sulfate (specific settings and data acquisition can be found in greater details in Figure 39).
  • the X-ray source was a tungsten anode lamp. No hair removal was needed.
  • the animals were anesthetized in an induction chamber with constant isoflurane feeding, weighed, manually transferred to the IVIS Lumina imaging system, and constantly fed isoflurane via the adaptor nose cone built within the imaging chamber.
  • the animals were secured in individual pods. The pod could be rotated via a computer control interface. The position/sedation of animals was controlled via a live camera built-in within the chamber.
  • the IVIS Lumina III XRMS (170) was used to track the location of the capsules and the rats digestive tissues’ were excised at the end of each time point to examine the intestinal tissue of methyl blue stains.
  • the control group used uncoated hard gelatin capsules. In vivo transit time and the capsules’ release profiles were also monitored periodically during the pharmacokinetic study.
  • the prediction profiler was set and selected with maximum desirability of polymer mass loading (7.0 – 15%) and minimal time needed to dry (5.5 – 12 minutes) as constraints.
  • the negative slope of polymer drying time to the increasing amount of acetone (Figure 17A) means that the addition of acetone to the formulation positively impacted the drying time of film; it took less time to dry the coated film.
  • the positive slope of polymer drying time ( Figure 17B) to the increasing IPA within the formulation suggested an increase in the amount of drying time to the film. Water had a negative effect on the formulation drying time but of lesser magnitude compared to IPA.
  • the vacuum applied to the bottom of the capsules placed inside a slot within the coater plate creates a shear force that thins out the polymers, which behave as non- Newtonian fluids.
  • the tinning is due to the entanglement of polymer collapsing in the direction that the vacuum is applied (bottom of the plate).
  • the angle at which the capsule sits causes it to spin around as centrifugal force results from the spinning of the coater plate.
  • the centrifugal force is large enough to spin out the large undissolved polymers and creates a vortex of air that facilitates further evaporation.
  • the combination of the vacuum and the spinning creates a conical vortex within each capsule pod.
  • the optimal PLGA 8515 inner erosion coat was made of 115 ⁇ 35 nm PLGA 8515, and the optimal thickness of the pH-sensitive enteric coat was 50 ⁇ 15 nm Eudragit S100.
  • Different PLGA thicknesses were also investigated.
  • PLGA 8515 layers that were thinner than 100 nm showed insufficient scaffolding support for S9C and had a structurally deformed surface upon successive coating and drying.
  • a PLGA 8515 film thicker than 150 nm failed to release its contents at the targeted time.
  • ES100 thickness also has some impact on the time taken by the capsule to release its contents; however, an imperfection in the coating layer of ES100 was not observed via the vacuum spin coating technique.
  • Thermogravimetry analysis is a measurement of the weight loss curve with the response to temperature ramp.
  • the TGA was used to determine the total solid mass of the polymers’ formulation and its consistency across each formulation batch used for each coating.
  • the first steep drop in the weight loss occurred very early and at a relatively low temperature of 20 to 40 °C.
  • the limited loss was accepted, as the weight loss occurs due to the dehydration of solvents.
  • a second weight loss ⁇ 10% between 370 to 375°C was the final solid mass of the bulk polymers ( Figure 19A-B).
  • both PLGA 8515 and ES100 represented, on average, 9-10% of the formulation by weight.
  • the TGA also provided a general clarification regarding the nature of the polymer formulation decomposition’s nature.
  • the mass loss across each triplicate run for the materials remained consistent with respect to the onset temperature and duration, and the decomposition of both materials reflected in the total weight loss correlated with the decomposition of the materials’ solid weights; thus, the results indicated that the solvents used to dissolve the polymers did not cause any physio-chemical reaction. Data acquired were consistent with published data (151) and in house historical data (Figure 19C-D).
  • DSC Differential Scanning Calorimetry
  • the coated S9C were visible in the stomach of group one and were not visible within the intestinal tract until four hours after gavage (Figure 21B-21C). Many smaller bright spots were observed in the ceca and colons of the remaining animals in group one ( Figure 21D). Eight hours post gavage, the S9C were not visible within the intestinal tract ( Figure 21E-21F). The disappearance of the image tracer can be explained by an insufficient amount of BaSO 4 density concentration at a location to provide positive X-ray contrast.
  • barium (atomic number 56) is an oral X-Ray contrasting agent specifically used to delineate the gastrointestinal tract in both preclinical and clinical settings (175, 176).
  • barium sulfate an inert insoluble barium-complex given as an oral slurry is often formulated to a specific gravity of 1.5 density (mass/oral suspension concentration) for positive X-Ray capturing (177). Due to the depth of the tissue and air gap within the abdominal cavity, the disappearance of the capsule after 8 hours might have been caused by the S9C emptying its contents or becoming diluted with fecal matter thus lowering the barium density below the detectable concentration. [0466] Upon examining the ex vivo tissue samples of the intestinal tract, the methyl blue stains were indicative of the S9C’s location (178, 179).
  • the dose normalized Cmax of oral S9C was 4.09% compared to the Cmax of the 6A1 IV.
  • the first 6A1 glucuronide peak was also observed at 687.08 ⁇ 182.12 ng/mL five hours after oral gavage.
  • the 6A1’s dose normalized AUC_24Hour_S9C was 0.7942 ⁇ 0.1731 nM/mL for S9C dosage form.
  • the AUC_ 24Hour_S9C_Uncoat was 12.00% and 9.35% for the enteric and delayed release coated capsules.
  • the absolute bioavailability (Fapp: AUC_24Hour_S9C Coated /AUC_24Hour_IV) of 6A1 coated capsule was 9.30%
  • the relative bioavailable (FRel: AUC_ 24Hour _ S9C Coated / AUC_ 24Hour _ S9C_Uncoated ) between the uncoated and coated capsule dosage form was 78.0%.
  • the total systemic exposure of the S9C coated dosage form reduced the total systemic exposure to less than 10%.
  • a 2-way ANOVA test was performed with the Tukey post hoc approach for dependent variables (AUC_ 24Hour and time) of which the test yielded significant across all three dosage forms.
  • the S9C colonic tissue concentrations were 1.7 and 2.3 times higher.
  • the 6A1 colon tissue concentration of S9C were 6 times higher than the oral suspension after adjusted for the different in dosage.
  • the size 9 hard gelatin capsules (S9C) packed with the novel selective COX-2 inhibitor (6A1) resulted in consistent coatings that attained a uniform release mechanism targeting the colon.
  • a dose-normalized AUC (ng*hr/mL*mg) indicated that the systemic exposure of 6A1 is 9.04% that of the systemic exposure of celecoxib given oral suspension (145).
  • a separate oral suspension study with a 40 mg/kg BID dose of Celebrex (dose equivalent to an 800 mg/day in human) indicated all CV toxicity signs but also reduced the colonic tissue PGE2 by 58%.
  • the same study also showed a Celebrex concentration of about 556.67 ng/g of colonic tissues. Compared to the same treatment and dosage, 6A1 concentration was 138.75 ng/g in the colon (180).
  • the combination coating approach could overcome the exiting drawbacks associated with their conventional dosage form (i.e., oral suspension).
  • the pharmacokinetic study, the in vivo imaging, and the ex vivo tissue staining, and the colon drug concentration (525.0 ⁇ 26.78 ng/g ) showed that the cumulative approaches were able to deliver more than twice the amount of 6A1 to the colon compared to the intravenous route, three times the amount of 6A1 compared to oral suspension, and similar in magnitude to the concentration needed to have therapeutic effect compared to Celebrex.
  • Example 3 Zero order release of enteric and delayed coatings of S9C to deliver a selective COX-2 inhibitor (6A1) to the colon of F344 rats
  • enterically coated microparticles were formulated and packed inside the coated S9C in place of raw 6A1 powder. It was hypothesized that the combination approach will achieve a zero order sustained delay release of 6A1 in the colon (lower AUC_systemic, increase local AUC_colon).Zero order release kinetics refers to the process of constant drug release from the delivery system.
  • Example 1 established methods for the preparation and purification of 6A1, 6A1- Sulfate, and 6A1-glucuronide.
  • Acetone, dichloromethane, isopropanol, DI water of 99.99% purity, and Polyvinyl alcohol (PVA, Mw ⁇ 31–50 kDa) were purchased from Sigma-Aldrich Corp. (St. Louise, MO, USA).
  • Ester-terminated DL-PLGA of 50:50 and 85:15 were purchased from (Lactel Absorbable Polymers, Birmingham, AL). Evonik R ⁇ HM GmbH (Darmstadt, DE) generously donated the ES100.
  • Capsule sinkers Catalog #3, item PSCAPWHT-XS were purchased from Dissolution Accessories (The Netherlands, De Kreek 12, 4906 BB Oosterhout).
  • Formulation of 6A1 Microparticles [0476] Polymeric pH-sensitive ES100 microparticles loaded with 6A1 were prepared via the wet milling and spontaneous emulsification solvent evaporation methods; the final formulation was a combination of these previously published methods (162, 163, 186) with some modifications. The powder 6A1 was first wet milled at 10 mg/mL in 35% methanol water at 1,600 rpm for an hour.
  • Table 7 Top: DoE of Nanoparticle formulations; Bottom: Leading Formulations from the DOE JMP 14 experiment.
  • In Vitro Dissolution Tests of microparticles packed inside coated S9C [0477] The in vitro dissolution test for the microparticles packed inside coated S9C was performed, in accordance with the recommendation from UPS compendial dissolution, non- sink conditions (168). The simulated intestinal pH was adjusted via 2.5 ⁇ mol/L of HCl or NaOH from the 2.5 ⁇ mol/L of phosphate buffer.
  • a 1.0 mL sample was collected every half hour and the pH exposure/duration were as follows: 1.5 hours at pH 1.2 (stomach), 2.5 hours at pH 4.6 (small intestine), 2 hours at pH 6.8, 2.5 hours at pH 8, and 2.5 hour at pH 6.8.
  • Each capsule was placed within a sinker housing and the stirrer rate was set at 80 rpm (169) at 37 ⁇ 2°C.
  • Samples at an extreme pH were air-dried, reconstituted, and neutralized before analysis.
  • Drug concentration (6A1) was determined using the UPLC MS-MS method that was previously validated after the removal of ES100 via methylene chloride.
  • Formulation 1 (Group 1) has 88.8 mg of 6A1 per gram of materials which correlated to an encapsulation efficiency (EE) of 30%.
  • EE encapsulation efficiency
  • Group 1 multiple dose regimen group needed a total of three capsules per dose and a total of six capsules per rat each day.
  • Formulation 2 has 56.3 mg of 6A1 per gram of particles, thus equating to an EE of 45%.
  • Group 2 needed a total of four capsules per rat per day as the doses given were 20 mg/kg BID.
  • Table 8 Pharmacokinetic dosing schedule of multiple doses of Formulation 1 and Formulation 1 microparticles packed inside S9C (20 mg/Kg dose).
  • Formulation development is a critical step in the process leading toward efficient, safe, and stable commercial products. Methods with high throughput capability provide a considerable advantage by expanding the conditions and parameters required. JMP 14 Software DoE facilitated the development of the pilot formulations. [0485] Fifteen pilot formulations were completed to determine the optimal ratio of 6A1- PLGA 8515-ES particles. Of the fifteen, three leading formulations: number 6, 14, and 15 met both the particle size and delayed release criteria set forth (Table 7, Figure 24, and Figure 25). Noticeably, all three formulations had a high percentage, above 75%, of the surfactant PVA.
  • Formulation 6 had the best performance of all the nanoparticle formulations and was used as a precursor for the preparation of 6A1 microspheres.
  • Scale up 6A1 microparticles were generously made by collaborators (Dr. Gan Yong, Shanghai Tech University, Pudong, Shanghai, China).
  • In Vitro Dissolution Tests [0486] The drug loading (EE%) of pH sensitive microparticles was determined by dissolving the microparticles in an alkaline PBS buffer (187), and then using a liquid-liquid extraction method to remove the 6A1 compound using DMC.
  • the in vitro, pH-dependent release profile of 6A1 from the pH-sensitive microparticles was investigated in a gradual pH- changing buffer to assess its ability to protect the content from the acidity and accomplish an effective delivery in neutral pH values (163).
  • the 6A1 microparticles were rapidly released and emptied their content from the enteric and delayed release coated S9C.
  • One under weighted capsule that had failed the quality assurance method was also tested.
  • Most of the coated capsules released 6A1 microparticles at pH 6.8 ( Figure 26). The result suggested the weight of a capsule was a reliable quality assurance method to qualify each coating batches.
  • the uncoated capsule has AUC _Ave24hour of 3.388 ⁇ 0.6967 ⁇ g.
  • the results are consistent with the previous pharmacokinetic study of food effect in uncoated capsules.
  • the AUC_Ave24hour of enteric-coated capsules was 2.567 ⁇ 0.48 ⁇ g.
  • the enteric coating layer reduced total systemic exposure to 60.6% of uncoated capsules.
  • the AUC _Ave24hour of enteric and delayed release coated capsules had the smallest amount, 1.009 ⁇ 0.1317 ⁇ g ( Figure 28).
  • the uncoated capsule had a Tmax at one-hour post-dose, with the enteric coat, the capsules’ Tmax shifted to two hours post-dose. Compared to the two previous, the enteric delayed release coated capsules’ T max shifted noticeably to five hours post-dose (P ⁇ 0.0001).
  • Single dose pharmacokinetic study of 6A1 microparticles formulated for sustained release using two leading microparticle Formulation 1 (F1) and Formulation 2 (F2) [0489] Again, the AUC_Ave24hour of the twelve subjects in the microparticles at the dose of 5 mg/kg was calculated using the trapezoidal rule.
  • the AUC _Ave24hour of 6A1, 6A1 glucuronide, and 6A1 sulfate were 1.43 ⁇ 0.04 ⁇ g/ml, 0.84 ⁇ 0.25 ⁇ g/mL, and 0.137 ⁇ .06 ⁇ g/mL.
  • the above pharmacokinetic parameters equaled to a dose normalized AUC_Ave24hour of 0.7130 ⁇ M/Hr for F1 and 0.6649 ⁇ M/Hr for F2.
  • the AUC _Ave24hour exhibited the characteristics of a sustained release with the onset of both formulation at around three hours and the C max 0.1854 for F1 and 0.1404 for F2.
  • Familial adenomatous polyposis is a chronic disease; multiple dosage regimens with appropriate drug concentration assist with regressing the onset of inflammation and polyposis formation.
  • a short term (4 day) efficacy study of enterically coated microparticulates packed inside coated S9C attempted to assess the therapeutic drug concentration in the colon and ability to deliver a constant, sustained amount of drug (C max_ss ) to the colon while reducing the total systemic exposure (AUC_average_ss).
  • Formulation 1 has a Css of 1,536.65 ng/mL compared to the 2,444.07 ng/mL for Formulation 2; thus, the colon tissue-specific of 6A1 in Formulation 1 was 37% less than that of Formulation 2 (1,536.6 ng/g compared to 2,444.7 ng/g, P ⁇ 0.012).
  • the P values of 6A1 metabolites (glucuronide and sulfate) in the colonic tissue between groups was insignificant (Figure 31). It was unclear whether the size differences of the particles in Formulation 2 or the larger number of capsules given contributed to such differences. The colon tissue concentration further confirmed that the capsules delivered a sufficient amount of drug to the colon.
  • this study achieved a de novo design of capsule coatings and formulations that delivered a higher concentration of drug to the colon and reduced the systemic drug concentration multifold.
  • the long-term efficacy study using the above approach to compare the 6A1 concentration systemically versus the colon targeted at steady state was in progress to elucidate the pharmacodynamics of 6A1.
  • Glucuronidation was the main driving force for of 6A1 entering into EHR; sulfation has little involvement in the phase two metabolism of 6A1.
  • the completion of these studies using the F344 model will help establish the pharmacokinetic mechanism of such a drug delivery system. Further investigation of the 6A1 pharmacodynamic mechanism is needed to successfully translate the application into clinical care for FAP patients as preventive care.
  • the blood plasma concentration (C max ) and time at C max (T max ) were determined from the observed values and confirmed with GraphPad Prism software.
  • the trapezoidal rule was used to calculate the AUCtotal from time 0 to the last measurable time.
  • lack of a powerful model to address the recycling of 6A1 resulted in the analysis not yielding reliable pharmacokinetic parameters and the need for more empirical data.
  • the pharmacokinetic studies revealed that the relative bioavailability of 6A1 was lower and the colonic tissue exposure higher when administered in the microparticles packed inside the S9C compared to the conventional oral suspension and/or intravenous route. Further analyses also revealed that relative bioavailability and exposure to 6A1 is higher when administered to subjects in the fast condition than subjects in the fed condition.
  • the described formulation comprises a blend of solvent mixtures.
  • the solvent system is composed of acetone, isopropanol alcohol (IPA), and water; it provides for an optimal drying time without a lengthy drying period during processing between each successive coating.
  • IPA isopropanol alcohol
  • the high mass loading of the polymer (10%) prevents the solution from having a running effect.
  • a high ratio of material to solvent and large molecular weight in a formula contributes to a high-viscosity.
  • High viscosity (a thick formulation) yields better coat quality as compared to a low viscosity formulation.
  • the intermolecular forces of a higher viscosity formulation prevents the solution from forming a film that is not subjected to shrinking stress during drying and the resulting breakage.
  • a thinner solution with low viscosity is often used as it enhances adhesion to the substrate; however, the downside is the need for multiple coatings.
  • the basic water spray applied to the capsule surface after sealing and before coating neutralizes the capsule's surface lysine, destabilizing their surface chemistry, and thus promotes better adherent for the polymer and preventing peel-off.
  • a robust, sensitive, and validated UPLC-MS/MS method was successfully used to quantify 6A1, 6A1 glucuronide, and 6A1 sulfate in pharmacokinetic blood, colonic mucosa, and liver tissue samples.
  • the method has a multitude of advantages, such as high sensitivity (LLOQ 2 ng/mL), short analysis time (6 minutes), small sample size (10 ⁇ L), functional recovery with negligible matrix effect, and simple sample preparation. With all the above- mentioned advantages, this method was applied to preclinical pharmacokinetic/pharmacodynamic modeling studies of 6A1.
  • Rats were fed ad libitum and the 6A1 active administered by oral gavage. Different study conditions were investigated, such as food effect, real-time imaging agent mixed with 6A1, and uncoated capsule versus enteric or erosion coated capsules. Food is a confounding factor of capsules emptying from the stomach. Fasted animals have a higher Tmax/Cmax and AUC Average24hourtotal compared to their fed cohort. [0500] Each additional coating applied to the capsules reduced the total systemic exposure of 6A1.
  • the uncoated, enteric coated, and enteric and delayed release coated capsules had an AUC_Ave24hour of 6.55 ⁇ 1.02 ⁇ g/mL.3.97 ⁇ 0.48 ⁇ g/mL, and 1.24 ⁇ .50 ⁇ g/ml.
  • Uncoated capsules AUC _total was comparable to the fed stage of the food study experiment. The results also indicated a 5.25 fold reduction of AUC administered via capsule enteric and a delayed release mechanism (P ⁇ 0.0212). The Tmax was also significant between the three groups. Uncoated capsules had a T max at one-hour post-dose. The enteric coated capsules delayed the Tmax to two hours post-dose.
  • the colonic concentration of 6A1 was comparable between the parallel study of 40 mg/Kg twice daily (3300 ⁇ 300 ng/g) to that of the four-day multiple dosing of coated capsules at 20 mg/Kg (2,682 ng/g for Formulation 1 and 1,916 ng/g for Formulation 2). Since the coated capsule's dosage was reduced by half, the result strongly suggested that lowering the dose could achieve similar colonic concentration.
  • Table 9 A dose normalized comparison of colonic tissue concentration of 6A1 across IV, oral suspension, size 9 capsule (SC9) packing with powder, and microparticles.
  • Glucuronidation levels in the multidose four-day study are the main driving force for 6A1 entering EHR; sulfation has little involvement in the phase two metabolism of 6A1. This conclusion was evidenced by the finding that there is no significant difference (P > 0.05) between the two groups when the glucuronide and sulfate concentrations in liver tissues are compared. For colonic mucosa tissues, the amount of 6A1 accumulation in Formulation 1, which has a smaller microparticulate, was significantly higher than that of Formulation 2. The particle size difference might have contributed to the observed data. The P value of glucuronide concentration between the two groups was insignificant (P > 0.05).
  • PLGA is a linear copolymer that can be prepared at different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA): [0506] Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained: these are usually identified in regard to the monomers’ ratio used (i.e., PLGA 85:15 identifies a copolymer consisted of 85% lactic acid and 15% glycolic acid). [0507] Eudragit S 100 (C8H12O4) is an anionic copolymer based on methacrylic acid and methyl methacrylate (poly(methacylic acid-co-methyl methacrylate) 1:2).
  • the solid polylactic glycolic acid (PLGA8515) is stored at -20°C, and the powdered methyl methacrylate acid (Eudragit S100) is stored at 4°C.
  • PLGA8515 is purchased from Direct Corporation, Birmingham Division (2200 Riverchase Center, Suite 501. Birmingham, AL USA 35244).
  • the solvent for PLGA8515 is methylene chloride. The solvent is used to dissolve 1.5 g of PLGA8515. The PLGA8515 is added slowly to a glass dram vial with 5 mL of premeasured methylene chloride.
  • the mixture is stirred at 60 RPM and another 10 mL of methylene chloride gradually added (in 1 mL dropwise increments) until the final volume is 15 mL. After 30 minutes of stirring, the solution will be visually clear. The final concentration is 10% w/v solution.
  • the PLGA8515 polymer is fractioned into 1.5 mL glass vials, sealed with parafilm, and stored at -20°C. Historical qualification steps (TGA/DSC) indicate that the PLGA8515 polymer is stable when stored under these conditions for up to 2 weeks.
  • the solvent for Eudragit S100 is a mixture of acetone: isopropanol alcohol: DI water in the ratio of 86:12:2.
  • the solvent is used to dissolve 1 g of Eudragit S100.
  • the Eudragit S100 powder is slowly added into 5 mL of solvent in a dram vial and stirred at 60 RPM; the powder is incrementally added allowing for the solution to become clear until all powder is added.
  • the Eudragit S100 will be visually clear after 30 minutes; if not, 5 ⁇ L of 100 micromolar NaOH solution at pH 11 is added and stirred for another 15 minutes. If the solution becomes transparent, an additional 5 mL of solvent is added to the solution until the final volume is 10% w/v. If the solution turns turbid, another 5 ⁇ L of 100 micromolar NaOH solution at pH 11 is added and stirred for another 15 minutes.
  • the Eudragit S100 is fractionated into 1.5 mL vials, the vials wrapped with parafilm, and stored at 4°C. Historical qualification steps (TGA/DSC) indicate polymer is stable in such conditions for up to seven days.
  • TGA/DSC Historical qualification steps
  • Both solutions require quantification with TGA and DSC before use. The solutions should be discarded after each use, or when left standing at room temperature for more than 2 hours.
  • Doping of PLGA8515 Polymers [0512] Doping is a process in which a contaminant or doping agent (dopant) is purposely added into a formulation to form a controllable pore size and pore density. In other words, a dopant is introduced as an artifact.
  • an alkaline salt or neutral salt often is used to stabilize acid terminal polymers and an acidic salt to stabilize basic terminal polymers. It is well established that the porosity of a polymer depends on both the polymer fabrication techniques and the concentration of salts.
  • CaCO3 and NaHCO3 are known dopants used to enhance degradation in a PLGA50 matrix. An alkaline dopant promotes faster polymer degradation without compromising the polymer's integrity (i.e., there is no actual chemical reaction between the porogen and the polymer network).
  • Sodium chloride was first dissolved in methanol at its saturated point of 14 g per 1000 g.
  • the sodium chloride working solution (11.074 ⁇ g/ ⁇ L) is added to a premade PLGA polymer that is dissolved in methylene chloride.
  • the best working sodium chloride salt concentration as dopant found was 2.5% v/v to 10% v/v of the final solution (37.5 ⁇ L NaCl/methanol into 1462.5 ⁇ L of pre-dissolved PLGA8515/methylene chloride polymers to 75 ⁇ L NaCl/methanol into 1425.0 ⁇ L of pre- dissolved PLGA8515/methylene chloride).
  • the percentage of the dry mass ratio of dopant to polymer can range from 0.03% to 0.07%.
  • alkaline dopants porogens include, without limitation sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), and sodium acetate (NaCH3CO3).
  • Other porogens include, without limitation, saccharin, lipoprotein, and small molecule fat.

Landscapes

  • Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The described invention provides enteric and delayed release formulations of locally bioavailable drugs (LBD) targeting the colon, methods for their preparation, and pharmaceutical compositions containing same.

Description

ENTERIC AND DELAYED RELEASE FORMULATIONS OF LOCALLY BIOAVAILABLE DRUGS (LBD) TARGETING THE COLON CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to and benefit of U.S. Provisional Application No. 63/220,384, filed July 9, 2021, which is incorporated by reference herein in its entirety. FIELD OF THE INVENTION [0002] The described invention relates to enteric and delayed release formulations of locally bioavailable drugs (LBD) targeting the colon. BACKGROUND OF THE INVENTION [0003] Considerable effort in the pharmaceutical industry has been focused on the design and development of effective pharmacologic treatments for Familial Adenomatous Polyposis (FAP) . [0004] Familial Adenomatous Polyposis (FAP) is a rare, inherited disorder resulting from a germline mutation in the adenomatous polyposis (APC) gene and characterized by colorectal cancer (Half E et al. Orphanet J Rare Dis. 2009;4:22; Patel SK et al. PLoS One. 2013;8(2):e55802). Colectomy is the most common treatment and is typically performed shortly after the time of diagnosis. Adenomatous polyps appear by about 15 years of age in 50% of people with FAP, and by 35 years of age in 95%. Seventy percent of these individuals have colorectal malignancy by the age of 36 years, the average age of symptomatic diagnosis. Over the lifetime of this illness, there is a 100% cancer risk, typically in the fourth and fifth decades of life. The average age of colorectal cancer onset for attenuated familial adenomatous polyposis is 55 years. No true pharmacologic treatment is available for these patients. (Half E et al. Orphanet J Rare Dis.2009;4:22; Kinney AY et al. Am J Gastroenterol.2007;102(1):153- 62; Arber N et al. Clin Ther.2012;34(3):569-79). Familial Adenomatous Polyposis (FAP) [0005] Familial Adenomatous Polyposis (FAP) is a debilitating condition that has a long term, severe impact on the physical and psychological health, independence, and quality of life of an affected patient. The affected patient develops hundreds to thousands of intestinal polyps, many of which become cancerous unless surgically removed. Most patients are asymptomatic for years until the adenomas are large and numerous, resulting in rectal bleeding or even anemia, or cancer develops. Nonspecific symptoms may include, but are not limited to, constipation or diarrhea, abdominal pain, palpable abdominal masses, and weight loss. FAP is broken down into two sub conditions based upon severity, classic and attenuated. Classic is the more severe form of FAP. FAP is inherited in an autosomal dominant manner and results from a germline mutation in the adenomatous polyposis (APC) gene. Most patients, approximately 70%, have a family history of colorectal polyps and cancer (Half E et al. Orphanet J Rare Dis. 2009;4:22). [0006] Patients suffering from FAP will present with some extraintestinal manifestations that can include, but are not limited to, osteomas, dental abnormalities (e.g., unerupted teeth, congenital absence of one or more teeth, supernumerary teeth, dentigerous cysts and odontomas), congenital hypertrophy of the retinal pigment epithelium, desmoid tumors, and extracolonic cancers (e.g., duodenum, stomach, skin, thyroid, liver, bile ducts and central nervous system). The extracolonic cancers may be benign or malignant. [0007] Desmoid tumors are of particular significance; these are fibrous tumors usually occurring in the tissue covering the intestines and may be provoked by surgery to remove the colon. Desmoid tumors are likely to recur after they have been surgically removed. [0008] Mutations in the APC gene cause both classic and attenuated familial adenomatous polyposis (Half E et al. Orphanet J Rare Dis. 2009;4:22). The mutations affect the affected cell’s ability to maintain normal growth and function. Cell overgrowth resulting from mutations in the APC gene leads to the colon polyps seen in FAP. Although most people with mutations in the APC gene will develop colorectal cancer, the number of polyps and the time frame in which they become malignant depend on the location of the mutation in the gene (Attard TM et al. Am J Gastroenterol.2004;99(4):681-6; Baglioni S et al. Am J Med Genet C Semin Med Genet. 2004;129C(1):35-43; Ashburn JH et al. Current Colorectal Cancer Reports. 2017;13(4):302-9). Epidemiology of Familial Adenomatous Polyposis (FAP) [0009] Familial Adenomatous Polyposis (FAP) is a rare, inherited condition with a reported incidence varying from 1 in 6,000 to 1 in 22,000 individuals. FAP manifests equally in both sexes and is the second most common genetic Colorectal Cancer (CRC) syndrome. CRC is the third leading cause of cancer deaths in the United States (Kinney AY et al. Am J Gastroenterol. 2007;102(1):153-62). Worldwide, CRC is a major cause of cancer associated morbidity and mortality. Its incidence varies considerably among different populations, with the highest incidence reported from Western and industrialized countries. Worldwide, about 85% of CRCs are considered to be sporadic, while approximately 15% are familial with FAP accounting for less than 1% (Half E et al. Orphanet J Rare Dis. 2009;4:22). Clinically, FAP manifests equally in both sexes by the late teens and in the twenties age group. Individuals with FAP carry a 100% risk of CRC (Half E et al. Orphanet J Rare Dis.2009;4:22; Kinney AY et al. Am J Gastroenterol.2007;102(1):153-62; Arber N et al. Clin Ther.2012;34(3):569-79). Treatments for Management of Familial Adenomatous Polyposis (FAP) [0010] Colectomy is the most common treatment for FAP and is typically performed shortly after the time of diagnosis. Total proctocolectomy, either with ileostomy or mucosal proctectomy and ileoanal pouch, eliminates the risk of developing colon and rectal cancer. If subtotal colectomy (i.e., removal of most of the colon, but leaving the rectum) with ileorectal anastomosis is done, the rectal remnant must be inspected frequently, and new polyps must be excised or fulgurated. If new polyps appear too rapidly or prolifically to remove, excision of the rectum and permanent ileostomy are needed (2-4, 7). [0011] After colectomy, patients should have upper endoscopic surveillance at periodic intervals. Protocols for postsurgical lower endoscopic surveillance depend on the type of surgery performed but include annual endoscopic surveillance of remnant anorectal tissue (2- 4). Yearly checkups with a physician should be opportunities to screen for other, extra- intestinal complications. One previous study in the USA reported that slightly more than half of participants who had an FAP diagnosis had recent colorectal screening; rates were lower for at-risk relatives, suggesting underscreening (2-4, 7, 8). Cyclooxygenase-1 (COX-1) and Cyclooxygenase-2 (COX-2) [0012] Non-steroidal anti-inflammatory drugs (NSAIDs) are perhaps the most versatile group of drugs prescribed for inflammation, analgesic/antipyretic, and auto-immune diseases (9, 114). Non-selective NSAIDs inhibit the synthesis of prostaglandins, thromboxane, and levuloglandins by blocking arachidonic acid substrates from binding to the cyclooxygenase isozymes (10, 11) Cyclooxygenase-1 (COX-1) and Cyclooxygenase-2 (COX-2). Certain prostaglandins appear to play a vital role in the carcinogenesis by altering cell adhesion, inhibiting apoptosis, and promoting angiogenesis (107, 115). COX-1 performs a distinct physiological homeostatic maintenance role within the vascular system and gastrointestinal tract; its inhibition may lead to gastrointestinal toxicity, cardiovascular toxicity, internal bleeding, and stomach ulcers. The COX-2 enzyme is the inducible, pro-inflammatory cyclooxygenase isoform; it is overexpressed and upregulated in many cancer tissues (116). COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (117) and is used as a prominent predictive marker in inflammatory bowel diseases (118). Selective COX-2 inhibitors (COXIBs) were designed to prevent the unwanted, adverse gastrointestinal events associated with NSAIDs. [0013] The discovery of COX-2 in the early nineties ignited a frenzy of selective COX-2 inhibitor drugs. For the decades that followed, these drugs boasted of enhanced pharmacological effects and safety. COX-2 is a variant of COX-1 with one amino acid difference. Crystallography revealed that COX-1 and COX-2 crystal structures reside in amino acid number 523. COX-2 has valine in place of isoleucine at position number 523. The difference allows COX-2 to form a deeper hydrophobic binding pocket that accounts for the substrate selectivity (Figure 1). [0014] Additionally, crystallography showed that the sulfamoylphenyl of the selective COX-2 inhibitor, celecoxib, is essential for the selective activity of celecoxib's COX-2 inhibition. Prostaglandins appear to play a vital role in the adenoma-carcinoma sequence by altering cell adhesion, inhibiting apoptosis, and promoting angiogenesis (12). COX-1 performs a distinct physiological homeostatic maintenance role within the systemic vascular and gastrointestinal tract (13). The inhibition of COX-1 leads to gastrointestinal toxicity (14), cardiovascular toxicity, internal bleeding, and stomach ulcers. The COX-2 enzyme, which is the inducible, pro-inflammatory cyclooxygenase isoform, is observed to be overexpressed and upregulated in FAP and CRC tissues. COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (6) and is used as a prominent predictive marker in inflammatory bowel diseases. [0015] The COX-2 enzyme was observed to be up-regulated, overly expressed in the polyposis of FAP patients, and to attenuate the progression of these polyposis lesions in the colon. If FAP lesions are treated at an early stage, not only can the FAP patients’ morbidity and mortality rates decline, but their quality of life can be improved (Half E et al. Orphanet J Rare Dis.2009;4:22; Kinney AY et al. Am J Gastroenterol.2007;102(1):153-62; James AS et al. J Cancer Epidemiol.2012;2012:506410). [0016] Selective COX-2 inhibitors (COXIBs), a subclass of nonsteroidal anti- inflammatory drug (NSAIDs) designed to selectively inhibit cyclooxygenase-2 (COX-2) for the treatment of arthritis, have been shown to effectively reduce the incidence of colorectal cancer in FAP patients (Kinney AY et al. Am J Gastroenterol. 2007;102(1):153-62; 22; Steinbach G et al. N Engl J Med. 2000;342(26), 1946-52; Su LK et al. Am J Hum Genet. 2000;67(3):582-90; Su LK et al. Hum Genet. 2000;106(1):101-7). However, because of cardiovascular side effects (e.g., unstable angina, myocardial infarction, and cardiac thrombus) at the doses required for efficacy as a prophylactic (Arber N et al. Clin Ther.2012;34(3):569- 79; Bertagnolli MM et al. N Engl J Med. 2006;355(9):873-84; Sheng JQ et al. Zhonghua Yi Xue Za Zhi.2006;86(8):526-9; Phillips RK et al.2002;50(6):857-60; Katz JA et al. Pain Med. 2013;14 Suppl 1:S29-34), COXIBs cannot be used clinically to treat FAP. The inability to avoid or prevent these harsh side effects ultimately forced the withdrawal of the drug (i.e., rofecoxib) (Higuchi T et al. Clin Cancer Res. 2003;9(13):4756-60; Kerr DJ et al. N Engl J Med.2007;357(4):360-9) or drug indication (i.e., celecoxib) for FAP from the market (Baron JA et al. Lancet.2008;372(9651):1756-64), leaving many patients at high risk for developing colon cancer without a safe and effective treatment other than colectomy (FitzGerald GA. N Engl J Med.2004; 351(17): 1709-11; Lynch PM et al. Am J Gastroenterol.2010;105(6):1437- 43). COXIBs as a Treatment for Management of Familial Adenomatous Polyposis (FAP) [0017] COXIBs (e.g., celecoxib, rofecoxib), a subclass of nonsteroidal anti-inflammatory drug (NSAIDs) designed to selectively inhibit cyclooxygenase-2 (COX-2) for the treatment of arthritis, effectively reduced the incidence of colorectal cancer in FAP patients (Kinney AY et al. Am J Gastroenterol. 2007;102(1):153-62; Steinbach G et al. N Engl J Med. 2000;342(26):1946-52; Su LK et al. Am J Hum Genet.2000;67(3):582-90; Su LK et al. Hum Genet. 2000;106(1):101-7). However, due to impending cardiovascular side effects (e.g., unstable angina, myocardial infarction, and cardiac thrombus) at the doses required for efficacy as a prophylactic (Arber N et al. Clin Ther.2012;34(3):569-79; Bertagnolli MM et al. N Engl J Med. 2006;355(9):873-84; Sheng JQ et al. Zhonghua Yi Xue Za Zhi. 2006;86(8):526-9; Phillips RK et al. 2002;50(6):857-60; Katz JA et al. Pain Med. 2013;14 Suppl 1:S29-34), COXIBs cannot be used clinically to treat FAP. The inability to avoid or prevent these harsh side effects ultimately forced the withdrawal of the drug (i.e., rofecoxib) (Higuchi T et al. Clin Cancer Res. 2003;9(13):4756-60; Kerr DJ et al. N Engl J Med. 2007;357(4):360-9) or drug indication (i.e., celecoxib) for FAP from the market (Baron JA et al. Lancet. 2008;372(9651):1756-64), leaving many patients at high risk for developing colon cancer without a safe and effective treatment other than colectomy (FitzGerald GA. N Engl J Med. 2004;351(17):1709-11; Lynch PM et al. Am J Gastroenterol.2010;105(6):1437-43). PGE2 Receptor and Its Tumor Genesis in the Context of Compound 6A1 [0018] COX-2 is an inducible form of the myeloperoxidases enzyme family that is located on chromosome 1 and found on the nuclear membrane or the luminal side of the endoplasmic reticulum (Chandrasekharan NVS et al. Genome Biology. 2004;5(9):241). IL1ß, IL6, and TNFalpha are known to regulate the expression of COX-2 in response to growth and inflammations (Rouzer CA et al. J Lipid Res.2009;50 Suppl:S29-34). Prostaglandin G2 is one of the downstream products catalyzed by COX-2 from plasma membrane arachidonic acid. The prostaglandins are responsible for vasodilation (PGD2, PGE2, PGI2), gastric, renal, platelet aggregation homeostasis (PGI2), mediating fever, pain, and inflammation (PGE2). In cancer cells, an increase of COX-2 expression positively correlates to an increase in PGE2 production. PGE2 modulates different functions via its binding ability to specific EP receptors. Binding of PGE2 to EP1 receptors led to the mobilization of intracellular calcium. Binding of PGE2 to EP2 and EP4 led to the deregulation of the cell proliferation pathway. EP2 and EP4 receptors are coupled to the G protein to activate adenylate cyclase, which increased the intracellular cAMP; intracellular cAMP activates PKA, PI3K, and GSK3 protein kinases that ultimately activate beta-catenin (Arico S et al. The Journal of biological chemistry.2002;277(31):27613- 21). The direct effect of PGE2 binding to EP3 receptors is still unclear. Through a concerted effort, the increasing concentration of PGE2 and its unclear binding pathway to different EP receptors disequilibrates the intricate balance between program cell death and cell proliferation that gave rise to tumor genesis and progression (Sobolewski C et al. Int J Cell Biol. 2010;2010:215158). [0019] The direct link between COX-2 and Wnt/ β-catenin remains unclear. However, the T-Cell factor/Lymphoid enhancer factor (TCF/LEF) family promotes COX-2 transcription also responsible for the transduction of Wnt/β-catenin (Araki YSO et al. Cancer research. 2003;63:728-34). In both cancer patients and colon cancer derived cell lines, the overexpression of LEF-1 and Pontin52/TIP49a (proteins of the Wnt pathway), and COX2 enzyme are upregulated (Kolligs FT et al. Digestion.2002;66(3):131-44). A more direct link between Wnt/ β-catenin and COX-2 regulation was established using chondrocytes where the lymphoid enhancer factor 1 (LEF-1/β-catenin) binds to the 3′UTR region of the COX2 genomic locus complex and directly regulates COX2 expression (Carlson MW et al. Molecular Cancer. 2003;2(42)) (Figure 35). [0020] Currently, marketed NSAIDs are not indicated for the treatment and prevention of FAP (2). However, Sulindac (commercially available as Clinoril) and Erlotinib (commercially available as TARCEVA) are sometimes prescribed for FAP post-operatively to prevent polyposis formation and to reduce the occurrence of frequent successive relapses (3, 4). Given the detrimental consequences of FAP as it relates to patient cancer risk and the need for repeated invasive surgical interventions, the goal has been to develop a systemic treatment with low toxicity that has the potential to reduce polyp burden as an adjunct or alternative to surgery. Orally administered drugs [0021] Often orally administered drugs are broadly distributed via systemic circulation before accumulating at the intended target sites. Thus, accumulation of a therapeutic dose at the intended organs (i.e. colon) also requires a very high dose resulting in a dangerously high drug concentration in systemic circulation. In order to prescribe a COX-2 inhibitor as preventive care and treatment for FAP patients, achieving the desired colon tissue concentration requires a very high oral dose that can result in the right COX-2 target but wrong organ toxicity. Colonic Drug Delivery System (CDDS) [0022] Over the last four decades, colonic drug delivery research has yielded inconsistent results due to the variability between normal gastrointestinal physiology and pathophysiological disturbances during different disease stages. Colonic drug delivery is still an attractive opportunity that offers many advantages, such as lowering the effective dose while achieving high drug concentration at the targeted local environments and reducing systemic side effects (Amidon S et al. AAPS PharmSciTech.2015;16(4):731-41). [0023] The two main routes of drug delivery to the colon are rectal and oral. The rectal route, mostly in the form of a suppository, has the most straightforward requirements for formulation and yields very consistent results. However, the rectal route is limited on where a drug can be delivered; the drug cannot be delivered to desired sites such as the cecum, ascending colon, or transverse colon. The oral route has been the primary alternative to deliver drugs targeting these proximal sites within the colon. Oral colonic drug delivery systems are effective in delivering acid label, narrow therapeutic index drugs targeted to various locations within the gastric tract, and low solubility drugs with high efficiency. The oral dosage form is aesthetic and convenient, thus has better patient compliance. The oral colonic delivery systems also have many challenges such as a transit time that is variable between individuals, a release mechanism that is affected by the type of diet and amount of food intake, the physiological and integrity differences of normal and disease stage, colonic pH fluctuation, as well as the microbiomes variability between individuals. [0024] There are many methods to formulate drugs targeting the colon (Figure 2) and each has its advantages and disadvantages. Potential methods to target the colon include emulsion, nanoparticles, microparticles, and liposomes. Among these, nanoparticles and microparticles had the most studied techniques. The respective size of different drug formulations are shown in Figure 3 (Zhang M et al. Inflamm Bowel Dis.2018;24(7):1401-15). [0025] Nanoemulsions and microemulsions (Figure 4) are droplets that contain drug molecules dispersed inside oil droplets and suspended in a water medium (o/w) or drug molecules dispersed inside aqueous droplets and suspended in an oil medium (w/o) (McConnell EL et al. International journal of pharmaceutics. 2008;364(2):213-26). Another type of emulsion is the bicontinuous emulsion which is composed of both o/w and w/o droplets, thus capable of capturing both hydrophobic and hydrophilic drugs. The nano and micro prefixes describe the droplet size, with the nano prefix ranging from 1 nm - 200 nm. The micro prefix represents a droplet size ranging from one micron and above. The liquid carrier that disperses the drug usually had the same hydrophobicity as the drug. The immiscibility between the water and oil needs to be stabilized by a surfactant (also known as an emulsifier). The surfactant serves as a thermodynamically isotropic interfacial membrane between the two liquid phases, lowers the surface tension, and creates a physical boundary that prevents the droplets from coalescence. Nanoemulsions and microemulsions are approaches used to improve drug bioavailability. A key component for emulsion systems is the drug release mechanism; the release mechanism is controlled through the skillful selection of surfactants and cosurfactants (Guo Y et al. Molecules. 2018;23(7); Patel SK et al. PLoS One. 2013;8(2):e55802; Rao J et al. J Agric Food Chem.2011;59(9):5026-35) that destabilize at the specific target thus allow the release of drugs. [0026] Nanoemulsions (Figure 3) are popular for use with the nasal, dermal, and mucosa routes because of their large surface area that results in increased drug absorption and permeation. Nanoemulsions are also thermodynamically and kinetically stable, enabling easy sterilization. Submicron size droplets produce translucent nanoemulsions that are ideal for intravenous formulations. Emulsions require a large amount of surfactant and can sometimes become unstable depending upon the temperature, pH, and ionic strength of the environment. [0027] A group of researchers recently synthesized a dual mode imaging of 19F magnetic resonance and near-infrared fluorescence capable nanoemulsion. The drug portion, Celecoxib, in this case, showed a strong signal within the cytoplasm of macrophages (Patel SK et al. PLoS One. 2013;8(2):e55802). The PGE2 reduction also inversely correlated with the drug’s concentration. The result suggests that the contrasting agents formulated with the nanoemulsion method did not hinder drug permeation into the cell. The emulsion also was stable for more than two months. [0028] The standard way of producing nanoparticles (nm) and microparticles (µm) is to include a lipid carrier or polymer matrix where the carrier encapsulates the drug. The two common types are solid and lipid particles. Various compositions and fabrication techniques are employed to increase the drug loading efficiency, achieve the desired pharmacokinetic profiles, and prevent dose dumping. An emerging trend observed is the use of a multi- particulate system where the raw drug and various sized particles are mixed in a controlled proportion as to achieve the desired drug release profile. Carrier materials also influence drug release from these particles (Sharma S et al. Journal of controlled release : official journal of the Controlled Release Society. 2018;272:97-106; Authors M. Nanoemulsions- Nanoarchitectonics for smart delivery and drug targeting 2016-2017). The nanoparticles are usually formulated via some form of strong force such as high ultrasonication, wet-milling, or high-pressure homogenization. Microparticles are usually formulated via a coacervation method which consists of the emulsion solvent evaporation process, spray drying, and solvent extraction-evaporation method. Poly-lactic-co-glycolic acid (PLGA), pH-sensitive polymers (polymethracrylate, i.e., Eudragit), cellulosic, and solid-phase anchored silica are examples of commonly used carriers for the formulation of particles (McConnell EL et al. J Drug Target. 2009;17(5):335-63). One disadvantage observed through recent clinical trials is the nonspecific extracellular release of the drug at the target site, the drug is released into the extracellular matrix thus the efficacy is also dependent upon the permeability of the drug (Cisterna BAK et al. Nanomedicine. 2016;11(18):2443). Figure 5 demonstrate a comparative size and surface area between nanoparticle and microsphere (Zhang M et al. Inflamm Bowel Dis. 2018;24(7):1401-15). [0029] Liposomes are lipid-based carriers that consist of a lipid bilayer that encapsulates an aqueous drug volume (hydrophilic drug) or hydrophobic drug within their lipid bilayer (Kraft JC et al. J Pharm Sci.2014;103(1):29-52). Liposomes have amphoteric properties that enable them to be used as a carrier for both hydrophilic and hydrophobic drugs. Liposomes are shown to accumulate at inflamed tissues and can be enhanced via surface modification and addition of a targeting ligand (Authors C. Liposome: Nano and microscale drug delivery system. Science Direct.). The surface modifications of liposomes are accomplished by physical anchoring and leveraging on the polarity of the chemical moieties or chemically treating the lipids and covalently bonding the moieties to the lipid (Figure 6) (Pileri P et al. Br J Cancer. 2016;115(1):40-51). Surface modified curcumin liposome in vitro study using colorectal cancer showed enhanced targeted delivery and drug concentration compared to a previously published curcumin liposome formulation (Feng T et al. Int J Nanomedicine. 2017;12:6027- 44). [0030] Since 2016, three completed clinical trials studying combination therapy for the treatment of colorectal cancers using liposomes as carriers had not had their results submitted. The lack of submitted results to the NIH clinical trials website is indicative of inconsistent or undesirable outcomes (Krajewska JBB et al. J. New Trends in Liposome-based Drug Delivery In Colorectal Cancer. Mini-Reviews in Medicinal Chemistry. 2019;19(1)). Furthermore, liposomes targeting the colon lack a practical application due to the complicated pH terrain of the colonic environments that may cause the instability and unspecific release of the liposomes. [0031] Virosome is a small subset of liposomes which are made of a unilamellar phospholipid membrane but contain viral genetic materials (phospholipid and glycoprotein) that facilitate a high amount of binding specificity. This formulation approach is one of a new emerging trend that leverage on the preexisting knowledge coming from the immunogenicity study accumulated over the years through the vaccine industry (Nguyen TXH et al. Nanomedicine.2016;11(9).). [0032] Other techniques for colon drug delivery system (CDDS) as therapy emerged during the last few years as more sophisticated analytical instruments became available (Amidon S et al. AAPS PharmSciTech. 2015;16(4):731-41). Of particular interested is the direct guidance of an external magnetic field to promote magnetic-nanoparticle accumulation and release at the targeted colon site. A group of researchers (Grifantini R et al. Journal of controlled release : official journal of the Controlled Release Society. 2018;280:76-86) combined such techniques with a monoclonal antibody (mAb198.3) to increase therapeutic specificity targeting of the FAT1 gene, and to reduce systemic toxicity. In the study, the researchers adhered magnetic nanoparticles to anti-FAT1 mAb198.3. The formulation showed that mAb198.3, by targeting the FAT1 gene expression, recognized colonic cancer cells at different stages and locations (Pileri P et al. Br J Cancer.2016;115(1):40-51). [0033] Taking into consideration the complexity of the colon, each colonic drug delivery system needs to sufficiently address the differences of the gastric transit time, local microbial content, physiological pH, amount of inflamed tissues with altered drug absorption ability, and mucus thickness that changes the surface topography to achieve drug release at the intended site. Moreover, the current colonic drug delivery systems have several disadvantages. Intestinal physiology [0034] When compared to the small intestine, the colon is much shorter, being only about 1.5 meters in length. The absorptive surface is much less than the small intestine; however, the colon has the longest transit time. The average transit time for material from ingestion to the colon is about five hours. The minimum dwell time in the colon is three to four hours before defecation (43). The optimal COX inhibitor release target time is, therefore around six hours to ensure passage through the stomach yet allow for release prior to defecation. In the colon, the microbiome on the surface of the colon wall produces a significant amount of bicarbonates, thus neutralizing the acidic content from the stomach and elevating the colon pH to 6.8 and above. Therefore, a successful pH-sensitive, biodegradable coating is expected not to disperse until the coated dosage form reaches the colon where the pH is at least 6.8. [0035] There has been much research utilizing pH-sensitive polymers or the erosion rate of polymers to deliver drugs to the colon. The pH-sensitive method, however, has had limited success, because the pH of the gastrointestinal (GI) tract varies depending upon individuals and their diets, and the erosion mechanism depends significantly on water content and the species of healthy microbiomes. pH-responsive release mechanism [0036] The distal colon has a near neutral pH of 6.6 ± 0.5 compared to the proximal small intestine 4.5 ± 0.5; thus, targeting the distal colon via a pH-responsive release mechanism, in principle, is a straight forward solution, but in practice has proven challenging due to the inter- individual pH variations. A pH-sensitive drug delivery system must withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ± 0.4) or the ileocecal junction. Interestingly, once in the colon, the pH turns slightly basic again due to the fatty acids produced by the resident microbiomes. The mean pH within a healthy colon has been reported to be 6.37 ± 0.58 in the right colon, 6.61 ± 0.83 in the mid-colon, and 7.04 ± 0.67 in the left colon (Evans DFP et al. Gut.1988;29:1035-41). In the colon, the pH variations can be as high as two pH units (Fallingborg JC et al. Ailment Pharmacol Ther. 1989;3(6)). A methacrylic acid and methyl methacrylate copolymer are the primary polymers of choice when it comes to pH-sensitive formulations (Lamprecht AS et al. Pharmaceutical Research. 2001;18(6):6). Methracyclic-methacrylate copolymer is commercially known as the Eudragit series from Evonik. Eudragit is used in many coatings for particles, hard gelatin capsules, and tablets. A few commercially available Eudragit coated formulations are Asacol®, Ipocol, and Mesren MR (Fadda HM et al. Pharm Res. 2009;26(2):356-60). A pH-sensitive drug delivery system also faces the intra-individual variances resulting from differences in food intake (Ibekwe VC et al. Pharmaceutical Research. 2008;25:1828-35). During disease stages, the right colon of ulcerative colitis patients is slightly acidic (4.7 ± 0.72) (Nugent SK et al. Gut.2001;48:571-7). In one extreme case, a patient with active ulcerative colitis has a pH of 2.3 in the proximal colonic. Another pH-sensitive pellet coated with Eudragit S failed to release the drug (Basit AW, & McConnell E. I.. Drug Delivery to the Colon. In: Delivery CRiOD, editor. Oral Drug Delivery: New York: Springer.; 2001. p. 386) suggesting that the failure of pH-sensitive dosage forms is influenced by more than just pH (Ibekwe VC et al. J Pharmaceutical Sci.2006;95:2760-76; Hardy JGL, S.W.; Clark, A.G. Enema volume and spreading. International journal of pharmaceutics.1986;31:151-5) but also by residence time at the ileocaecal junction, by feeding status, and by gastrointestinal fluid composition. Thus, commercial drug delivery systems leveraging on pH variation of the digestive tract is still underdeveloped (Pang X et al. Journal of controlled release : official journal of the Controlled Release Society. 2016;222:116-29; Maurer AH et al. Journal of Nuclear Medicine Technology.2016;44(1):12-8). Time-based delivery to the colon [0037] Time-dependent systems attempt to utilize the time delay between dosage form ingestion and colonic arrival to achieve colon-specific targeting. The drug release is generally achieved by erosion, diffusion, or swelling of different matrices over a predetermined period. Time-based approaches work on the assumption that a dosage form will spend approximately six hours in the stomach and small intestine during the fasted state. The time-dependent approach assumes that gastrointestinal transit time is solely influenced by the gastric emptying. Thus, a lag time can be added for the fed state. The erroneous part of this assumption is in the variability of the amount and type of food intake (Weitschies W et al. International journal of pharmaceutics.2011;417(1-2):216-26) When an individual is hungry, a phase III contraction wave in the stomach squeezes and flushes all material remains in the stomach to the small intestine include submicron particulates. When the stomach is full, food prevents the onset of phase III contraction wave. A study had shown that enteric coating tablets remained in the stomach for more than twelve hours (Ibekwe VC et al. Pharmaceutical Research. 2008;25:1828-35). Thus, relying solely on the transit time to deliver the drug to the colon would not be sufficient. [0038] The complexity of a time-based colonic drug delivery system intensifies as the CDSS is designed with the assumption that the small intestinal transit time is around three hours ± one hour. In reality, Davis, Hardy & Fara, 1986 and recently, Fara et al., 2009 demonstrated that the assumption of small intestinal transit time is inaccurate (Katsuma MW et al. J of Pharmaceutical Science.2003;93(5)). Indeed, the amount of time that food remains in the small intestine ranges from half an hour to nine hours, depending on the meal size and type of diet. [0039] The order in which the medicine was taken also influences the amount of time taken to reach the colon. A study showed a tablet taken after a meal stayed in the small intestine significantly longer than the tablet ingested half an hour before a meal. [0040] CDSS, in brief, are influenced by the small intestinal transit time, which in turn is influenced by the food intake, the time the meal was eaten in relative to the time the drug was taken, the shape and form of CDSS formulations, and the disease stage of the patient. All these variables in total make it challenging to predict the individual's colonic arrival time which equates to an insurmountable task for the effort of designing a time-dependent colonic CDSS that would accurately deliver drugs for all patients (Hebden JM et al. Eur J Gastroenterol Hepatol.1999;11(12):1379-85). Microbially Triggered Delivery Mechanism [0041] Although much shorter than the small intestine, the colon has an overwhelming number of resident microbiotas that exist sparsely in the upper gastrointestinal tract. The microbiota is another approach for colonic site-specific drug release design. [0042] Medicinal chemists exploited the large bacterial population in the colon to design their prodrugs. For example, sulfasalazine is a prodrug that successfully employs colonic microbiota to cleave the inactive precursor and release the active mesalazine. Other enzymes produced by the colonic bacteria such as saccharolytic bacteroids and bifidobacterium to break down di-, tri-, and polysaccharides were successfully absorbed by the upper intestinal tract (Gorbach S et al. Gastroenterology.1967(6); Hill M, Drasar BS. The normal colonic bacterial flora. From the bacterial metabolism research lab.1975; Sinha VR et al. Drug Dev Ind Pharm. 2004;30(2):143-50). Anaerobic bacteria are also known to be the main engine that produces polysaccharidases that break down short-chain fatty acids such as butyric, acetic, and propionic acids. [0043] The high levels of polysaccharidase producing bacteria mean that attention has turned to polysaccharides as colonic delivery systems; these compounds are cheap, nontoxic, and biodegradable. A selection of polysaccharides can avoid degradation in the small intestine but are used as a substrate by the colonic microbiota. These polysaccharides can be used as coatings or matrix systems. The performance of polysaccharide based colonic delivery systems has been reviewed by many authors (74, 75) and will not be discussed in detail in this review. Although polysaccharide-based systems show promise for colonic targeting, few have reached market. A system based on amylose mixed with the water-insoluble polymer ethylcellulose has demonstrated positive results in Phase I and II clinical trials. Amylose is a starch polysaccharide; starch polysaccharides come in many forms, several of which are indigestible by human pancreatic enzymes but act as a food source for colonic bacteria (76, 77) This combination of colon-specific polysaccharide and insoluble polymer (to prevent swelling and premature drug release) has achieved consistent colonic targeting with various drug molecules. McConnell et al., 2008d recently compared a pH-sensitive drug delivery system using Eudragit S to the polysaccharides prodrugs of amylose/ethyl-cellulose coated theophylline pellets that target colonic bacteria. The targeted prodrug released in the colon is far more effective compared to the pH-coated pellets. The study was completed by utilizing gamma scintigraphy and pharmacokinetics data of theophylline (43). However, a phase III efficacious clinical trial studied eight UC patients using the above comparative approach. The authors noted that the prednisolone prodrug approach has fewer side effects compared to the pH-trigger method and proposed that such a prodrug approach may be useful for maintenance therapy. [0044] A colon-targeted delivery system (CDDS) also relies on specific colonic bacteria to cleave prodrug into a drug (78). The microbiome-prodrug approach also incorporates pH sensitive elements for targeting the neutral colonic environment. The above dual mode often utilizes lactulose and acidic pH-responsive cores to help dissolve the tablets upon reaching the colon. The by-product of the prodrugs is usually some form of short-chain fatty acids that facilitates a burst release profile. The CDDS that relied on colonic bacteria has been successfully applied to theophylline, a drug that requires a narrow therapeutic index (43, 79, 80). [0045] Given the success of bacterially triggered systems, a new concept in colonic drug targeting was introduced by combining pH-responsive and bacterially triggered mechanisms in a single layer matrix film (81). The technology comprises a mixture of pH-responsive polymer (Eudragit S) and biodegradable polysaccharide (resistant starch). The Eudragit S component in the coating has two functions - preventing the disintegration of the film in the upper gastrointestinal tract and controlling the swelling of polysaccharide. The polysaccharide in the coating resists the digestion by mammalian pancreatic amylase enzymes and is digestible by colonic bacterial enzymes. Once entering the colon, both trigger mechanisms contribute to the dosage form disintegration, ensuring appropriate drug targeting. A gamma scintigraphy study showed that the system provides for colon specificity. Consistent disintegration of tablets coated with the single layer matrix film was observed at the ileocecal junction or colon. [0046] The colonic environment in terms of pH, microbiomes, and disease stages is a highly dynamic environment, which is not adequately studied or understood. There is a great deal of variability in the colonic environment, affected by many factors that need to be considered, with one of the most critical factors being the disease stage, when designing drug delivery applications for the colon. Emerging Combination Release Mechanisms [0047] Recent advancements of CDSS tend to revolve around a combination of release mechanism technologies. [0048] The development of a multi matrix approach (MMX) has gained some success in delivering a high concentration of drugs to the colon (181). One MMX® formulation is characterized by a lipophilic matrix dispersed in a hydrophilic structure. The technology is a proprietary blend of liposome and hydrophilic excipients mixed with drug powders and pressed into a tablet. The tablets were then coated with an enteric coat to prevent upper gastrointestinal absorption. The mechanism of drug release obtained by this pharmaceutical formulation concerns the gastro-resistant coating, which avoids the release of the embedded compound until the tablet is exposed to a pH of seven or higher, which is normally reached in the terminal ileum. After reaching this site, the activity of the tablet core, which consists of hydrophilic excipients (thought to drive the tablet to swell into a viscous gel mass, slowing the release of the drug) and lipophilic excipients (thought to slow the penetration of aqueous fluids into the tablet core), results in a homogenous and prolonged exposure of the whole colonic mucosa to the embedded substance(s). In this way, the MMX® delivery system guarantees that active drugs play their therapeutic role directly on the colonic mucosa and minimizes the systemic absorption of the drug. This approach proved to be a successful drug delivery system to the colon in a few clinical trials (182). [0049] MMX’s most successful application was toward low molecular weight chemical entities such as mesalazine, budesonide, and low molecular heparins (82). The success with low molecular weight entities is understandable due to the inner core of water-insoluble gel that formed once the outer pH-sensitive coating had dissolved (83). Regardless, MMX is still not wholly site specific due to a pH burst release mechanism that has significant interindividual differences. A mesalazine release study via gamma scintigraphy images showed release started in the small intestine and ileum, then reached mean maximum plasma concentration in the ileocaecal junction. The budesonide MMX formulation performed the worst; the MMX tablet disintegration began in the mid-small intestine (79, 84). [0050] Therapeutic nanoparticles are colloidal structures with a cargo space for drugs that is segregated from the environment by a hydrophilic, usually polyethylene glycol (PEG), corona that prevents recognition by macrophages and enables long-term circulation in the bloodstream. (183). The size of nanoparticles (10–100 nm) permits their extravasation and accumulation in tumor sites. Passive targeting is based on pathophysiological characteristics unique to solid tumors: hypervascularity, irregular vascular architecture, potential for secretion of vascular permeability factors, and absence of effective lymphatic drainage that prevents efficient clearance of macromolecules. Nanoparticles are not generally administered orally mainly because of physiological obstacles; i.e., from the perspective of cellular drug delivery, access to the cytosolic space of eukaryotic cells is restricted primarily to hydrophobic small drugs with a MW <500, which have relatively high membrane partition coefficients and permeability constants. [Id., citing 4,5] To increase intestinal uptake, nanoparticles can be conjugated with various bioadhesive (e.g., poly(lactic acid (PLA)), [Id., citing 3] P-gp pump- inhibiting (e.g., d-α-tocopheryl PEG succinate (TPGS))[Id., citing 6], and vitamin [Id., citing 7,8,9,10,11] (e.g., biotin, folic acid, vitamin B12, and transferrin) ligands. The in vivo oral absorption of three types of nanoparticles was investigated: basic nanoparticles, unmodified nanoparticles without a targeting ligand, and nanoparticles linked to a biotin-targeting ligand via either amide or amide–disulfide–amide bonds. The hydrophobic cargo core of micellar nanoparticles was labeled with the fluorescent molecule coumarin 6 for all three nanoparticle types. To study the intestinal absorption, fluorescently labeled nanoparticles were administered orally to rats. After jugular vein cannulation, blood samples were taken at fixed time intervals over 24 h, and the concentration of fluorescent nanoparticles in the plasma was measured by a fluorescence HPLC method. Statistical analysis showed that the pharmacokinetic data obtained for the bare DSPE–PEG2000–NH2 and targeting ligand containing DSPE–PEG2000–biotin did not differ significantly (p < 0.05); i.e., the intestinal absorption for both nanoparticle types was negligible. Interestingly, the DSPE–PEG2000– SS–biotin nanoparticles markedly increased the intestinal absorption in the fasted state, giving a superior oral absorption compared with bare DSPE–PEG2000–NH2 and peptide-linked DSPE–PEG2000–biotin nanoparticles (p < 0.05). [0051] It is known that the process of nanoparticle absorption through the intestinal barrier is energy-dependent endocytosis. [Id., citing 14,15]. The exact mechanism by which nanoparticles are taken up and broken down in the enterocytes is unclear. However, leading theory indicates that the ES100 microparticles follow the clathrin-mediated endocytosis pathway (CME, 183). The CME pathway comprises primarily four types of organelles: early, recycling, and late endosomes, and lysosomes. These four classes of endocytic organelles are not preexisting, stable structures but rather are dynamic and difficult to recognize based on their morphology or position in the cytoplasm alone. Early endosomes represent a dynamic network of tubules and vesicles dispersed throughout the cytoplasm. [Id., citing 15,16]. Late endosomes are defined as vesicular structures that accumulate and concentrate internalized cargo intended for degradation. [Id., citing 16,17]. Late endosomes degrade their contents progressively, thus providing for the recycling of surviving receptors, and ultimately increase in density as digestible membrane and content are processed, catabolic products are released, and the remaining undigestible material (lysosomal hydrolases, and certain membrane and lipid components) is concentrated. At this stage, they become resting lysosomes, which can be activated again upon fusion with late endosomes. [Id., citing 16,17]. [0052] A chip-based dynamic cell culture model has been compared to the common static cell culture and mouse model to assess its capability to predict in vivo success more accurately, by using a well-defined poly((methyl methacrylate)-co-(methacrylic acid)) and poly((methyl methacrylate)-co-(2-dimethylamino ethylmethacrylate)) based nanoparticle library (184). The concentration of poly-methacrylic acid seemed to influence the rate of cellular uptake (higher concentrations of polymethacrylic acid increases the negative surface charge and decreases the hydrophobicity contributed to the decreasing concentration of the copolymer) (184). [0053] Anionic pH-sensitive membrane-disruptive polymers have evolved as a class of bioactive excipients for the cytosolic delivery of therapeutic macromolecules. In one study, a large variety of anionic copolymers and analogues of poly (acrylic acid)(PA) was investigated and compared to a cationic PA copolymer. The pH-responsive membrane-disruptive properties were characterized by employing three in vitro models, such as pH dependent shift of pyrene fluorescence, liposome leakage and lysis of red blood cells. The pH-dependent increase of polarity and membrane disruption in the different model systems was in good agreement for all tested PA polymers. The efficacy of polymer-induced membrane disruption was concentration-dependent and significantly affected by the composition of the membrane. The sensitivity of relatively complex membranes of mammalian cells can be ranked between plain diphosphatidylcholine (DPPC) liposomal membranes and the more rigid cholesterol- containing DPPC membranes. Among the various studied PA polymers, medium and low molecular poly(ethacrylic acid) (PEA) and poly(propacrylic acid) (PPA) were identified as displaying significant pH-dependent disruptive activity. Relative to the disruptive cationic PA polymer (PDMAEM) the ranking is PEA < PPA < PDMAEM. The charge density of the nanoparticles may have resulted in lysosomal rupture due to osmotic swelling, thus resulting in the release of drugs into the cytosol (185). Colon Bioavailable Drugs Enterohepatic Recycling (EHR) [0054] The concept of a “recyclable colonic drug” utilizing the enterohepatic recirculating loop to target and deliver drugs to the colon is a recent strategy. Bioavailability is a significant issue in drug development because sufficient drug concentration in the target organ is needed to elicit the desired therapeutic effects (88). Conventionally, most effort in drug development focuses on improving systemic bioavailability (i.e., increasing systemic exposure) since drugs with good systemic bioavailability means more drug circulating to the target organ (89, 90); however, the approach also increases toxicity across non-targeted organs. One of the major physiological barriers limiting systemic bioavailability is the first pass effect where drugs are absorbed from the intestine and metabolized in the liver, dramatically reducing the concentration of active drug reaching the systemic target organ. Medicinal chemists often try to avoid the first-pass effect as a means of improving bioavailability (91-93). [0055] To achieve colon bioavailability, the described invention utilizes a modification in the current COXIBs structure by adding a desirable metabolic moiety (i.e., phenolic moiety) to enable the derivatives to undergo first pass effect. In other words, the engineered COXIBs will undergo rigorous first pass metabolism in the liver. Once administered orally, the desirable metabolic moiety of COXIBs derivatives is metabolites by hepatic uridine 5'-diphospho- glucuronosyl-transferase (UGTs), and efflux into the small intestine via transporters (e.g., breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 2 (MRP2)). For example, genistein, a soy-isoflavonoid with three hydroxyls, was rapidly metabolized by UGTs in the liver and extensively excreted through BCRP and MRP2 transporters via bile, resulting in a low systemic bioavailability (< 6%) (94-96). Phase II Metabolism [0056] Glucuronidation facilitates EHR. Glucuronidation is one of the most important phase II metabolic pathways of xenobiotics, and endogenous substances (97-99). Glucuronides of some phenolic compounds are good substrates of certain transporters (e.g., BCRP, MRP2), resulting in extensive hepatic secretion into the bile. The secreted glucuronides enter the intestine via the bile and often participate in recycling schemes after being hydrolyzed back to the parent drug by microflora. As described herein, adding a phenolic moiety to the structure of a COX-2 inhibitor thereby promoting it for glucuronidation could enable the inhibitors to undergo glucuronidation in the liver, efflux to the colon; thus, the locally bioavailable COX-2 inhibitors repeatedly enter into EHR, had low systemic exposure, and repeated colonic exposure. [0057] Glucuronidation capability can be highly different in the liver than in the colon. Glucuronidation is catalyzed by UDP-glucuronosyltransferases (UGTs), which have approximately 24 known isoforms (97-99) with organ-specific distribution and expression levels (100). For example, UGT1A10 is exclusively expressed in the colon, whereas UGT1A9 and UGT1A1 are highly expressed in the liver but only poorly expressed in the colon (101). Furthermore, the colon’s glucuronidation capability is low (by as much as 100x slower) compared to the small intestine and the liver (101). The glucuronidation rates of some flavonoids and drugs with phenolic moiety employed here are highly specific for the liver and not for the colon (102, 103). As provided herein, the designed COX-2 inhibitors’ phenolic moiety is synthesized toward UGT1A1 enzymes and showed extensive glucuronidation in the liver and facilitate their biliary excretion. [0058] EHR can prolong the drug’s apparent half-life in the GI tract. Due to recycling, drug molecules are repeatedly present in the GI tract, resulting in a prolonged apparent drug half- life in the colon (104, 105). Bile salt molecule, an endogenous compound, is estimated to be recycled within the enterohepatic loops on average about 20 times via EHR (106); although the number of recycling loops and repeated exposure of current locally bioavailable COX-2 inhibitors are still under investigation. Microbial β-glucosidase [0059] Due to the increase in hydrophilicity of the metabolites, the recyclable colonic targeted COX-2 inhibiting drugs are not expected to be reabsorbed by the epithelium enterocytes. There, once reaching the colon, the metabolites of locally bioavailable COX-2 inhibitors would get deconjugated and reverse back into active drugs via microbial β- glucosidase. The colon has approximately 100 billion to two trillion organisms per gram of luminal content. A variety of microbiomes are highly active in the glycosylation of phenolic glucuronides. A fraction of glucuronided candidates is observed to decouple from the glycolic acid and convert back into the parent compound. The above decoupling phenomenon is common in flavonoids. The recycled parent will be reabsorbed into the colon epithelial cells and resume the EHR cycle. EHR, deconjugation of metabolites, and reabsorption will repeat multiple times until all the drug molecules are eliminated through the feces. Thereby, leveraging on the EHR will minimize the side effects associated with broad distribution of the drugs and the deconjugation of local colon microbiomes will help increase the concentration of drug delivery to the colon. [0060] It has been accepted that for diseases such as FAP and CRC, the balance between the proliferation of pathogenic bacteria that incite inflammation and the probiotic bacteria that help to control the pathogenic microorganism population is disproportionate (107-109)). Studies showed that the pathogenic bacteria possess β-glucuronidase enzymes in CRC patients are many folds more active (80, 110). Coated gelatin capsules [0061] Hard gelatin capsules provide an advantage over other solid dosage forms, in that the raw drug can be packed inside the casing thereby reducing the need for excipients and extensive formulations (146, 147). Hard gelatin capsules are hydrogenated collagen made up of abundant proteins that are readily broken down into amino acids and become available for absorption within hours of ingestions (148). [0062] However, the coating of gelatin capsules is challenging. The smooth gelatin surface causes poor adhesion. Extreme pH or organic solvent usage in the polymer film can cause structural deformity and compromise the integrity of the capsules (149, 150). The application process of enteric and erosion films for the size nine hard gelatin capsule (S9C, 2.71 mm x 8.4 mm with a surface area of 68 mm2), specifically designed for rodents and guinea pigs, is especially challenging and cumbersome with the conventional pilot study and dip-coating technique. Conventional coating equipment and the specific parameters are also inapplicable to the S9C in small quantities for experimental use. Polymers [0063] Polylactic-co-glycolic acid (PLGA) and Eudragit S100 (ES100) are two commonly used pharmaceutical excipients that are generally recognized as safe (GRAS) by the United States FDA and the European Medicines Agency. Both excipients are commercially available with different grades of copolymer blends and have been extensively studied (151, 152). Both PLGA and ES100 are biodegradable materials with abundant preclinical and clinical research that focuses on the drug delivery systems (153). ES100 and PLGA polymers were chosen for this experiment because of their general safety and well-studied use for formulating an enteric and time delayed coatings. [0064] In many fields of science, polymer degradation has been defined as the amount of time the material takes to degrade over its useful life time and nondegradation has been defined as breakdown that occurs after its useful life. In another way, the ratio between the time the polymer takes to degrade and the duration of the application of the polymer is used to determine whether or not the material is degradable (154). PLGA and ES100 used for the coating of enteric and time release capsules undergoes chemical degradation; PLGA also undergoes erosion. [0065] All biodegradable polymers contain hydrolyzable bonds where the passive hydrolysis or enzyme-catalyzed hydrolysis breaks down the chemical bond between the copolymers. The hydrolysis degradation occurs via a random chain scission process to form oligomers and finally monomers (155). The enzyme-catalyzed hydrolysis also referred to as biodegradation is when a biological system is partially involved in the chemical bond cleavage. [0066] Most synthetic biodegradable polymers, PLGA and ES100, mainly undergo passive hydrolysis (156). The degradation rate is determined by its chemical bond, composition, the pH of the environment, the water content, and the swelling rate. Degradation begins with water uptake into the bulk of the polymer matrix, which leads to swelling. Next, hydrolysis occurs resulting in oligomers and monomers degrading from the polymer. Progressively, the degradation changes the microstructure of the bulk, forming pores, where the oligomers and monomers are released. Heterogenous degradation refers to the surface degradation of the polymers whereas homogenous degradation describes a uniform break down of the bulk material throughout its matrix. [0067] Erosion is a more complicated process of polymer degradation in which additional parameters such as the swelling rate and material porosity are involved. Poly (D,L-lactic-co- glycolic acid) three dimensional objects have been shown to exhibit an inverse flux erosion throughout the inner bulk material (154); once degradation begins increases in the degradation rate due to the lactide monomer content serves as an autocatalytic hydrolysis resulting in erosion (155, 157, 158). The PLGA erosion rate can be optimized by adjusting the amount of porosity and the sizes of the micropores within PLGA matrix (159). [0068] PLGA is an excellent biodegradable and biocompatible polymer (153, 160). Varying the ratios of lactic to glycolic acid controls the rate of water cleavage. Lactic acid contributes to the rigidity of the polymer backbone, thus increasing the duration that the polymer remains within a biological system (161). How it was degraded? The primary mechanism for degradation of PLGA is the hydrolysis of the ester bond between the lactic acid and glycolic acid. [0069] The PLGA 85:15 grade was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule. PLGA 85:15 also allows for successive coating applications. [0070] Eudragit S100 (ES100) is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio. ES100 pH dependent nature were finely tune by the number of negative charge on the MAA’s COOH functional group at basic condition (162). The hydrophobicity MMA units were realized by synthesizing co-polymer libraries with a systematic variation. ES100 is soluble in alkaline digestive fluids by salt formation (163, 164). A clinical study shown that a 5% W/W gain of the coated Eudragit S100 tablets (84 ± 4 micron) (64) release its content in the colon in 67% of volunteered subject. [0071] A disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility (supplemental S4). However, the addition of plasticizer changes the dissolution profile and takes longer for the film to dry (165). Therefore, there is a need to develop alternative coating techniques to limit drying time, maximize polymer mass loading to 10%, and limit the use of plasticizer while achieving uniformly smooth, defect free films. Pharmaceutical Coating Techniques [0072] Coating is the process of adding an outer layer of material to the surface of a substrate. There are many pharmaceutical coating techniques including conventional pan (involves coating capsules with a sprayed-on solution), fluid bed, dry powder (solvent-less) utilizing an electrostatically charged powder, magnetically assisted impaction, compression, hot melt extrusion, supercritical fluid, and supercell coating technology. These techniques function to mask taste and odor, protect from degradation, control the pharmacokinetic properties of drugs, or a combination of the aforementioned purposes. Fluid applied coated technique substrates sequentially transition through the following stages: fresh runny solution, semisolid, sticky, and dry film. Criteria for a successful coating technique would be to have little or no visual defects, no functionality defects, increase production, and simplify operations. Unfortunately, not a single coating technique above meets all the criteria for all applications. Some coating techniques are more advantageous than others in some applications but less advantageous for other applications. [0073] A conventional pan coater is one of the oldest coating technologies available — a thin film forms covering the capsules when a solution is sprayed into a temperature and pressure-controlled chamber. The pan continuously rotates tumbling the capsules; heated air is introduced evaporating the excess sprayed solution. A fluid bed coater (Figure 7) is similar to the conventional pan with additional air injected below the capsules resulting in a suspended, fluidized bed (85). The design helps separate the capsules to prevent clumping while the coating solution is spraying. A critical disadvantage for conventional pan and fluid bed techniques is the prolonged processing time between successive applications of coats. The mass of the polymer dissolved in the spray solution is limited to 3% of the total capsule’s weight to ensure even drying and prevent intense sustained attrition that leads to abrasion and chipping of the film. Other disadvantages to these techniques are the requirements of a large amount of bulk material, expensive equipment, and a trained technician. The technician must monitor critical parameters to ensure reproducibility between batches such as the spraying rate, dewpoint, mass solution flow, air flow, and temperatures. [0074] Liquid solution coatings often include an aqueous or organic solvent to dissolve polymers. An aqueous solvent requires a large amount of energy and high temperatures to evaporate often resulting in an undesirable appearance of the film formed as well as degradation of the drugs. Organic solvents on the other hand are environmentally toxic. Liquid solution coating often requires lengthy processing and drying to altogether remove the solvents. This, in turn, increases manufacturing cost. The dry powder, solvent-less coating techniques such as the electrostatic dry coating relieve manufacturers of the above challenges (86). In general, the mixture of conductive particles and polymers are cast into a disk. An electric current is applied to an adjacent electrode thus ionizing the conductive particles, while an air jet is blown toward grounded capsules, the electrical field and the mechanical forces of the jetted air cause ionized particles to deposit onto the capsules. The voltage, air flow, and powder density together all work to control the thickness, performance, and the appearance of the depositing film. A challenge of this method is the precise parameters required to obtain desirable film as well as avoiding supercharging the substrate that would destroy the drug within the capsule. Finally, the cleanliness of the substrate, high impact force, heat generated within the chamber sometime led to uneven thickness, void space, and multilayers deposition of the films. [0075] The present invention disclosure provides oral dosage forms (e.g., hard gelatin coated capsules) formulated to dispense their payload in the colon comprising a combination of biodegradable polymers and a coating process. The coating formulations and coating techniques provided reduce high systemic exposure to COX-2 inhibitors (total area under the curve (AUC_total)) that can result in cardiovascular toxicity. The described locally bioavailable COX-2 inhibitors are optimized in design to subjugate microorganisms in the microbiota to reactivate locally bioavailable COX-2 inhibitors metabolites into parent compounds, and thus provide formulations and methods for achieving repeated drug exposure in the colon with minimal systemic exposure. SUMMARY OF THE INVENTION [0076] According to one aspect, the present invention provides an oral pharmaceutical composition comprising a gelatin capsule containing an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and a therapeutically effective amount of a therapeutic agent disposed within the capsule. [0077] According to another aspect, the present invention provides an oral pharmaceutical composition comprising a capsule comprising an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and a particulate formulation disposed within the capsule, comprising a plurality of particles each comprising a therapeutically effective amount of a therapeutic agent, and one or more polymers encapsulating the therapeutic agent and which releases the therapeutic agent at a pH above 6.0. [0078] According to some embodiments of the oral composition, the therapeutic agent comprises a structure as defined by any one of Formulas 1-10 or a pharmaceutically acceptable salt thereof. According to some embodiments of the oral composition, the therapeutic agent is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C) or a pharmaceutically acceptable salt thereof. According to some embodiments of the oral composition, the biodegradable polymer is a poly(lactide-co-glycolide) (PLGA) selected from the group consisting of 95:5 poly(lactide-co-glycolide) 85:15 poly(lactide-co- glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide). According to some embodiments of the oral composition, the biodegradable polymer is 85:15 poly(lactide-co-glycolide) (PLGA8515). [0079] According to some embodiments, the biodegradable polymer is applied to the capsule to a predefined thickness of 115 ± 35 nm. According to some embodiments the inner delayed-release coating further comprises a plurality of pores to control release of the therapeutic agent. According to some embodiments the pH-sensitive polymer is Eudragit S100. According to some embodiments the pH-sensitive polymer is applied to the capsule to a predefined thickness of 50 ± 15 nm. According to some embodiments size of the plurality of particles is about 100 nm to about 2000 nm, inclusive. According to some embodiments the one or more polymers comprises 50:50 poly(lactide-co-glycolide) (PLGA5050), Eudragit S100, and/or poly(vinyl alcohol) (PVA). According to some embodiments when the composition is orally administered to a subject there is a lag period of at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, or at least about 12 hours before sustained release of the therapeutic agent. According to some embodiments when the composition is orally administered to a subject, sustained release of the therapeutic agent in the colon occurs at a pH of about 6.8. According to some embodiments the composition: reduces therapeutic dose of a therapeutic agent, and/or reduces total systemic exposure (AUC) of a therapeutic agent, and/or increases local colonic exposure (AUCcolon) of a therapeutic agent, and/or increases concentration of the therapeutic in the colon by lowering AUC0-24Hours, and increasing local AUCcolon, and/or utilizes enterohepatic recycling (EHR), and/or reduces occurrence of on target off organ systemic toxicity associated with administration of the therapeutic agent. According to some embodiments the dopant is sodium chloride, sodium percarbonate, sodium bicarbonate, sodium carbonate, or sodium acetate. According to some embodiments the dopant configures the polymer to increase its porosity. According to some embodiments the microparticle formulation has a theoretical encapsulation efficiency (EE) of 30-45%. According to some embodiments a release profile of the particles in vitro is pH dependent. According to some embodiments the in vitro pH- dependent release profile of 6A1 microparticles comprises a rapid release of the active in a neutral pH environment. According to some embodiments the composition comprises an extended release profile. According to some embodiments Tmax is 3 hours. [0080] According to another aspect, the present invention provides a method of spin coating a capsule, comprising: (i) providing a vacuum spinning plate comprising individual pods; (ii) providing sealed capsules wetted with a mild basic water (e.g., pH 9 NaOH 0.1 mM) loaded into the individual pods within the vacuum spinning plates; (iii) providing an amount of a first polymer dissolved in a solvent; (iv) applying a vacuum; (v) spinning the plate a first time for about 30 seconds at a spinning speed of 100 RPM; (vi) spinning the plate a second time for about 15 minutes at spinning speed of 500 RPM;(vii) providing an amount of a second polymer dissolved in a solvent; (viii) applying a vacuum; (ix) spinning the plate a third time for about 30 seconds at spinning speed of 100 RPM; (x) spinning the plate a fourth time for about 15 minutes at a spinning speed of 500 RPM; (xi) removing the capsule from the spinning plate; and (xii) drying the capsule in a desiccator. [0081] According to some embodiments of the method, the method achieves a polymer mass loading of at least about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 10.5%, about 11%, about 11.5%, about 12%, about 12.5%, about 13%, about 13.5%, about 14%, about 14.5%, or about 15%; and/or the method achieves a coating comprising a smoothness and uniformity of surface appearance; and/or the method achieves a predetermined thickness measurement; and/or the method achieves in vitro dissolution; and/or the method achieves a drying time of less than about 1 hour, less than about 30 minutes, less than about 15 minutes, less than about 10 minutes, or less than about 5 minutes. [0082] According to another aspect, the present invention provides a method for reducing severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: providing the pharmaceutical composition according to claim 1 or 2, wherein the therapeutic agent is a selective COX-2 inhibitor, and administering an effective amount of the pharmaceutical composition orally to the subject. According to another aspect, the present invention provides a method for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: providing the pharmaceutical composition according to claim 1 or 2 , wherein the therapeutic agent is a selective COX-2 inhibitor; and administering an effective amount of the pharmaceutical composition orally to the subject. According to some embodiments of these methods, the therapeutic agent that is a selective COX-2 inhibitor is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) or a pharmaceutically acceptable salt thereof. [0083] These and other advantages of the invention will be apparent to those of ordinary skill in the art by reference to the following detailed description and the accompanying drawings. BRIEF DESCRIPTION OF THE DRAWINGS [0084] FIGS. 1A-1C shows a schematic depictions of the structural differences between the substrate-binding channels of Cyclooxygenase-1 (COX-1) and Cyclooxygenase-2 (COX- 2). Ile523 in the COX-1 block the binding of celecoxib (FIG.1A), while Val523 in the COX- 2 form a second deeper binding pocket for celecoxib (FIG. 1B). This figure also shows the flexibility of the central pyrazole ring, which is in the gate side of the binding pocket. FIG.1C shows the structure of celecoxib, 6A1, and 7A1 attaching an additional moiety did not affect the binding. 6A1 was designed to retain the sulfamoylphenyl group and central heterocycle of celecoxib, but with the addition of a phenolic group. [0085] FIG.2 shows current practice for colonic drug delivery systems. [0086] FIG.3 shows respective size of different drug formulations. (42) [0087] FIGS. 4A-4C shows a comparison of nanoemulsion (FIG. 4A), microemulsion (FIG.4B), and mixture of nanoemulsion and microemulsion (FIG.4C). (46) [0088] FIG. 5 shows respective size of a microparticle versus nanoparticle, surface area increases as size of spherical particle decreases. (42) [0089] FIGS.6A-6D shows various type of liposomes used for colon drug delivery system (CDDS), including (FIG. 6A) Mannosylated, (FIG. 6B) Ionic, (FIG. 6C) Virosomes, (FIG. 6D) Multilayered liposomes (Vesosomes). (51) [0090] FIGS. 7A-7D shows current coating technology for colon drug delivery system (CDDS), including Fluidized Bed (85) (FIG.7A), Electronic dry powder (86) (FIG.7B), Hot Melt Extrusion (87) (FIG.7C), and the vacuum spin coating technique described herein (FIG. 7D). [0091] FIGS. 8A-8C shows ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) of validated method ran on a colonic mucosa sample. FIG.8A shows representative extracted-ion chromatogram (XIC) of multiple reaction monitoring (MRM) of all four compounds eluting in the order of 6A1 sulfate, 6A1 glucuronide, internal standard (IS), and 6A1. FIG. 8B shows MRM- sulfate and IS. FIG. 8C shows MRM+ glucuronide, IS, and 6A1. [0092] FIG. 9A shows representative product ion spectra of internal standard (IS) in negative mode. [0093] FIG. 9B shows representative product ion spectra of internal standard (IS) in positive mode. [0094] FIG.9C shows representative product ion spectra of 6A1 in positive mode. [0095] FIG.9D shows representative product ion spectra of 6A1 glucuronide. [0096] FIG.9E shows representative product ion spectra of 6A1 sulfate. [0097] FIG. 10 shows blood concentration time profile pharmacokinetic study of 6A1 compound. Blood concentrations of 6A1 (red circles), 6A1 glucuronide (green squares), and 6A1 sulfate (blue triangle) after intravenous administration of 5.0 mg/Kg in F344 rats (n = 3). Blood sample (10 µL) was spiked into acetonitrile (1000 µL), and IS (40 µL, Baohuaside in methanol, 1000 ng/mL). The mixture was vortexed and centrifuged for 15 min at 15,000 rpm. The supernatant was taken out, air dried, and reconstituted in 40 µL of 60% methanol and centrifuged for another 15 min at the same speed. Then 10 µL of supernatant was injected into the UPLC–MS/MS system. Each point is average of three determinations and the error bars are standard errors of the mean. [0098] FIGS. 11A-11B shows colonic mucosa and liver tissue concentrations collected from F344 rats two hours after administration of an intravenous dose of 5 mg/Kg 6A1, 6A1 glucuronide, and 6A1 sulfate. Liver and colonic mucosa concentrations were analyzed in triplicates. FIG. 11A shows colonic mucosa has an average ± standard deviation (SD) concentration of 225.94 ± 65.94 ng/g for 6A1, 26.78 ± 17.80 ng/g for 6A1 glucuronide, and 5.12 ± 0.86 ng/g for 6A1 sulfate. FIG. 11B shows liver tissues analyzed in triplicate had an average concentration of 337.60 ± 100.27 ng/g for 6A1, 315.79 ± 81.55 ng/g for 6A1 glucuronide, and 218.14 ± 54.21 ng/g for 6A1 sulfate. [0099] FIG.12A shows a schematic of exemplary capsule coating layers. [0100] FIG.12B shows an image of an exemplary capsule’s respective size compared to a dime. [0101] FIG. 12C shows scanning electron microscope (SEM) topography view of exemplary coated capsules (40X magnification). [0102] FIG.12D shows thickness of an exemplary coated capsule obtained from scanning electron microscope (SEM) side view given thickness of 115 ± 35 nm for PLGA 8515 coating and 50 ± 15 nm for ES100 coating (80K X magnification). [0103] FIG.13A shows factorial screening design for ES100 solvent system with inputted empirical values. [0104] FIG.13B shows ES100 solvent system effect summary correlating the PValue set at 0.05 to logworth value (-log of PValue) of 1.30. [0105] FIGS.14A-14B shows predicted value and empirical data input generated models to predict ES100 polymer drying time FIG.14A and mass added FIG.14B with R2 correlation value above 0.93. The ordinary least square model for predicting the polymer drying times FIG.14A is more powerful than the model used to predict the polymers’ mass load FIG.14B based on the square root of the variance residuals observed RMSE values (0.7071 and 1.4142). RMSE was the square root of the observed variance residuals. The smaller the RMSE value, the closer the predicted value was to the real value. [0106] FIGS.15A-15C shows scanning electron microscope (SEM) topography images of dip coating (FIG.15A), vacuum spin (FIG.15B), and fluid coating technique (FIG.15C). All pictures had a 1 mm scale bar at 40X magnification. [0107] FIG. 16 shows study design for the pharmacokinetic, ex vivo, and live imaging study of exemplary enteric and delayed release coated capsules (dose = 5 mg /Kg; n= 12). The control groups (n=2) were gavage uncoated size 9 capsules (S9C) and sacrificed at 1 and 3 hours post dose. Group 1 (n=5) were oral gavage coated S9C and the ex vivo tissues were collected at 4 (n=1), 8 (n=1), 10 (n=1), and 12 (n=2) hours post dose to examine the location and condition of the S9C. Group 2 (n=5 were cage-mates of Group 1 cohort) blood samples were collected at 0, 0.5, 1, 3, 5, 7, 10, and 24 hours post dose (purposefully off set the imaging time so as to reduce unnecessary stress to animals). [0108] FIG.17 shows factorial screening design via JMPDoe14 results demonstrating that the ES100 system was optimal at 65: 10: 3.5 acetone : IPA: DI water at 11% polymer weight, and 8.75 minutes drying time has the highest desirability. [0109] FIG. 18A shows novel vacuum spin coater (10 Pa vacuum, 500 RPM spin rate) capable of coating 8-16 capsules in 30 minutes (with an 88% success rate) [0110] FIG.18B shows blueprint drawing of spinning plates. [0111] FIGS.19A-19D show thermogravimetry analysis (TGA) and differential scanning calorimetry (DSC) determination of quality control parameters of exemplary coated capsules. FIG. 19A shows TGA of one month old PLGA versus newly formulated PLGA (9.023% vs 10.286%). FIG. 19B shows TGA of 1 month old versus newly formulated ES100 with no substantial solvent loss displayed by the consistent mass loading and the ratio of solvent (first weight loss) to total solid mass (second weight loss) with respect to temperature compared between two samples (analyzed in triplicates). FIG. 19C shows DSC curve for PLGA 8515. FIG. 19D shows DSC curve for ES100. Both DSC curves show a small glass transition, endothermic peak, and exothermic temperatures are consistent with historical data. [0112] FIGS.20A-20C shows in vitro dissolution test performed for 7 coated S9C and one quality control (QC) failed S9C (10% PLGA 8515 inner coat and 10% Eudragit S100 as pH- sensitive coat). FIG.20A is an image of a capsule that showed indications of breakage 6 hours after initial PBS buffered exposure. FIG.20C shows observed methyl blue trail leakage into medium 6 hours and 21 minutes after initial PBS buffer exposure. FIG. 20C shows empty capsule shell remained 7 hours and 12 minutes after initial PBS buffer exposure at pH 6.8. The 6A1 concentrations were determined using ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) described herein. [0113] FIGS.21A-21N show in vivo and ex vivo imaging study of coated S9C. FIG.21A shows µ-CT IVIS image obtained of capsules packed with barium sulfate (bright spots). The coated capsules do not appear to interfere with the x-ray imaging agent, BaSO4. FIG. 21B shows 0 hour after gavage, capsules are not visible within the intestinal tract. FIG.21C shows 2 hours after gavage, capsules visible (bright spot) within the stomach. FIG. 21D shows 6 hours after gavage, smaller bright spots observed in colon. FIG.21E shows 8 hours, capsules are not visible within the intestinal tract. FIG. 21F shows 12 hours (n = 3), capsules are untraceable within the intestinal tract. FIG. 21G shows ex vivo imaging of uncoated SC9 control animal’s stomach showed methyl blue stains indicate the location of capsules 1 hour after oral gavage. FIG. 21H shows 3 hours after oral gavage, the blue stain passed the duodenum and was visible in the jejunum. FIG. 21I shows 4 hours after gavage, ex vivo intestine has no visible blue stain of the coated capsules group. FIG.21J shows at 8 hours after gavage, ex vivo cecum had extensive methyl blue staining. FIG. 21K shows at 10 hours, ex vivo showed partial staining of the cecum and complete blue staining of colon. FIGS. 21L- 21N shows 12 hours after coated S9C were given, some remnants of undissolved polymer film fragments and capsule shells were visible and mangled in the forming fecal pellets. Some blue stained droppings were also observed (FIG.39). [0114] FIG.22 shows blood concentration time profile from the pharmacokinetic study of coated S9C (n= 5). The 6A1 and its metabolites’ concentrations were determined using the UPLC MSMS method (LLOQ of 2 ng/mL with instruments’ LOD of 0.50 ng/mL) described herein. [0115] FIG.23A shows dose normalized blood concentration time profile pharmacokinetic studies of 6A1 from intravenous dose (5 mg / Kg, n=4), oral gavage of uncoated S9C (20 mg / Kg, n=5), and oral gavage of coated S9C (20 mg/ Kg, n=8). [0116] FIG. 23B shows a dose normalized time point of 6A1-glucuronide between three routes of administration. [0117] FIG. 23C shows the Kruskal-Wallis of the one-way ANOVA of 6A1 ranks that suggested there was a significant difference between IV/S9C_coated and S9C_uncoated/S9C_coated. The test did not find any substantial different between the IV and uncoated S9C (oral suspension formulation). [0118] FIG. 23D shows a one-way ANOVA statistical run of dose normalized 6A1 glucuronide did not yield any significant difference between the three groups. [0119] FIG. 24 shows SEM images of formulations 6, 14, and 15. SEM images were obtained using the following operating parameters: 5.0 kV voltage, 4 mm working distance, and 10-6 millitorr vacuum. The side views of the capsule halves were captured at an 80,000x magnification [0120] FIG. 25 shows exemplary wet milling and spontaneous emulsification solvent evaporation processes for fabrication of 6A1 microparticles. 10mg/ml of powder 6A1 in 35% methanol water was micronized with three different sized glassbeads at 1,600 rpm for an hour. Different slurry volumes (0 - 1.0 mL) were added dropwise into different volumes of 5% (w/w) ES100 (Table 7). The organic phase containing both 6A1 encapsulated by ES100 was emulsified in 20 mL of 2.5% PVA by microtip sonicator for 30 seconds at 10 µAmps, paused for 30 seconds in ice water (30 cycles). The microparticle was gently stirred for 6 hours, collect via ultracentrifugation (20,000 x g, 10 minutes, 4 degree Celsius). [0121] FIG.26 shows in vitro dissolution results of two 6A1 enteric coated microparticle formulations (F1 and F2 with each n=8) using simulated intestinal pH (1.2, 4.6, 6.8, 8, and 6.8 using 2.5 µmol/L of HCl or NaOH from the 2.5 µmol/L to adjust the 2.5 µmol/L phosphate buffer. 1.0 mL of sample was collected at every half hour until a complete disappearance of the particle (approximately 10 hour). [0122] FIG.27 shows blood concentration time profile of food effect performed for coated sized 9 hard gelatin capsules (S9C). The dose normalized total area under the concentration curve of fasted animal (dose= 20 mg/Kg with n=8 per group) was 45% higher compared to the fed group (n=8). Unpaired T Test between two group yielded insignificant difference (P=0.6426); however, the nonparametric 95% CI analysis of Tmax shown significant difference between fasted (Tmax = 2 hours) and fed state (Tmax = 1 hour). [0123] FIG. 28 shows blood concentration time profile of each successive coating was performed for uncoated and coated size 9 hard gelatin capsules (S9C). The dose normalized of AUC_total (n=8) was highest in the uncoated (twice the enteric coats and three times the enteric and delayed coated). The uncoated size 9 capsules (S9C) contained raw drug powder packed inside the S9C. The enteric coat S9C contained raw drug powder packed inside S9C and a layer of ES100 of approximately 50 nm in thickness. The enteric coat and delayed coat contained raw drug powder packed inside S9C, with a middle layer of approximately 150 nm of PLGA8515, and an outer layer of ES100 of approximately 50 nm in thickness. All animals were fed ad libitum. One-way ANOVA Sidak’s multiple comparison test of AUC_total between groups yield insignificant difference (P = 0.6617); however, the 2-way ANOVA analysis of Tmax (Row Factor P value) shown significant different between groups (P<0.001). [0124] FIG. 29 shows blood concentration time profile pharmacokinetic study of microparticles: Formulation 1 and Formulation 2. Rats were fed ad libitum and given gavage of 5mg/Kg dose. Statistical analysis via student t test suggest there are insignificant differences between the two formulation with p = .8063 [0125] FIG. 30 shows multiple-oral-dose regimen pharmacokinetic study of 6A1 microparticles formulated for sustained released packed inside the enteric and delayed release coated size 9 hard gelatin capsules (S9C). Blood concentration time profile of F344 rats (n=6 per group) dosed with 20 mg/Kg BID for four days were collected at 12, 14, 16, 18, 24, 36, 48, 60, 62, 64, 66, 68, 72, 84, and 89 hours after gavage. [0126] FIG. 31 shows ex vivo liver and colon tissues accumulation of 6A, 6A1 glucuronide, and 6A1 sulfate collected 6 hours post 8th dose for F1 and F2 (n=6). Tissues concentration of 6A1 extracted as described herein. [0127] FIG.32 shows blood concentration time profile of 6A1 across three different size 9 hard gelatin capsule coatings showed the enteric and delayed release coating has the lowest systemic drug circulations (Teal color block, AUCtotal). [0128] FIG. 33 shows blood concentration time profile of 6A1 of enteric and delayed release coated size 9 hard gelatin capsule packed with microparticle F1 and F2. The single dose and the via twice daily for four day at 20 mg/Kg had similar results. [0129] FIG.34 shows colonic tissue drug concentration of 6A1 with normalized dosage across IV, oral suspension, size 9 (S9C) coated capsules with raw powder, and size 9 capsule with microparticle formulations. The microparticles formulation 1 has the highest colon tissue accumulation of 6A1. [0130] FIG.35 shows a mechanism of chemoprevention via Cox-2 Inhibition. [0131] FIG.36 shows blank matrix sample injection after 6 upper limit of quantification (ULOQ) injections shown negligible internal standard peak at 4.2 minutes. [0132] FIG.37 shows lower limit of quantification (LLOQ) injection using Raptor Restek Biphenyl column. [0133] FIG.38 shows exemplary solvents and plasticizer for ES100. [0134] FIG. 39 shows Perkin Elmer IVIS Lumina III XRMS imaging parameters and special data analysis. [0135] FIG.40 shows blue stained fecal droppings were observed 10 hours post dosed as well as polymer residuals. [0136] FIG. 41 shows consistent ratio of solvent (first weight loss) to total solid mass (second weight loss) with respect to temperature. Glass transition, endothermic peak, and exothermic temperatures are consistent with historical data. DETAILED DESCRIPTION OF THE INVENTION List Of Abbreviations A AA: Arachidonic acid AL: Alabama ANOVA: Analysis of variance APC: Adenomatous polyposis coli APC Trial (Pfizer): Adenoma Prevention Clinical Trial APPROVe (Merck): Adenomatous polyp prevention on Vioxx API: Active pharmaceutical ingredient AUC: Area under the curve B BCRP: breast cancer resistance protein C CA: California CE: Collision energy COX-1: Cyclooxygenase -1 COX-2: Cyclooxygenase -2 COXIBs: Selective COX-2 inhibitors CRC: Colorectal Cancer CV: Cardiovascular CDDS: Colonic drug delivery system Cmax: Maximum drug concentration Cmin: Minimum drug concentration Css – steady state concentration CXP: Collision cell exit potential D DE: Delaware DI: Distilled Water DL-PLGA: DL-polylactide-co-glycolic acid DMC: Methylene Chloride DNA: Deoxyribonucleic acid DP: Declustering potential DSC: Differential scanning calorimetry E EE %: Encapsulation efficiency % EHR: Enterohepatic recycling EP: Entrance potential ES100: Evonik Eudragit S100 F FAP: Familial adenomatous polyposis FDA: Food and Drug Administration F344-Pirc Rat: Fisher 344-polyposis in the rat colon species G g: Gravity g/dL: Gram per deciliter GI: Gastro intestinal GRAS: Generally recognized as safe H HCL: Hydrochloric acid I i.e.: In other words IACUC: Institutional animal use and care committee IC50: Half maximal inhibitory concentration IPA: Isopropanol alcohol IVIS lumina: Perkin Elmer IVIS Lumina III XRMS J JMP: Jump statistical discovery from SAS K kDA: kilodalton kV: kilovolt L LBD: Local Bioavailable Drugs LC-MS: Liquid chromatography-mass spectrometry LLE: Liquid-liquid extraction LLOQ: Lower limit of quantification M M.D. Anderson: Monroe Dunaway Anderson mg: milligram mg/kg: milligram per kilogram min: Minute mL: milliliter mm: millimeter MMX: Multimatrix system MO: Missouri MRM: Multiple reaction monitor MRP2: Multidrug resistant protein 2 MS: Mass spectrometer Mw: Molecular weight N NaOH: Sodium hydroxide ng/mL: Nanogram per milliliter NIH: National Institutes of Health Nm: nanoparticle NSAID: Non-steroidal anti-inflammatory drug O ODAC: Oncologic Drugs Advisory Committee P PBS: Phosphate buffered saline PD: Pharmacodynamic PGE-2: Prostaglandin E2 PGI2: Prostacyclin pH: power of hydrogen PK: Pharmacokinetics PLGA: Poly-lactile-co-glycolic acid PVA: Polyvinyl alcohol Q QC: Quality control R RPM: Revolutions per minute S SPE: Solid phase extraction SEM: Scanning electron microscopy T Tg: Temperature of glass transition TGA: Thermogravimetric analysis TLag: Time lag before drug reaches maximum concentration Tmax: Time drug reach maximum concentration U µAmps: micro amperes µg/mL: microgram per milliliter UGT: uridine 5'-diphospho-glucuronosyl- transferase µL: Microliter UM: microparticle UPLC: Ultra-performance liquid chromatography USP: United States Pharmacopoeia and in the Agency’s dissolution methods database Definitions [0137] As used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to a “peptide” is a reference to one or more peptides and equivalents thereof known to those skilled in the art, and so forth. [0138] As used herein, the term “about” means plus or minus 20% of the numerical value of the number with which it is being used. Therefore, for example, about 50% means in the range of 40%-60%, inclusive, i.e., 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, or 50%. [0139] The term “active” as used herein refers to the agent, drug, ingredient, component or constituent of the compositions of the present invention responsible for the intended therapeutic effect. [0140] The term “active pharmaceutical ingredient” (API; or Drug Substance) as used herein refers to a substance or mixture of substances intended to be used in the manufacture of a drug product and that, when used in the production of a drug, becomes an active ingredient of the drug product. Such substances are intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease or to affect the structure and function of the body. [0141] The term “administering” as used herein includes in vivo administration, as well as administration directly to tissue ex vivo. Generally, compositions may be administered systemically (e.g., orally, buccally, parenterally, by inhalation or insufflation (i.e., through the mouth or through the nose), or rectally) in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired, or may be locally administered by means such as, but not limited to, injection, implantation, grafting, topical application, or parenterally. The described invention provides for oral administration. [0142] The term “agent” as used herein refers generally to an active compound(s) that is/are contained in or on the formulation. “Agent” includes a single such compound and is also intended to include a plurality of such compounds. [0143] The terms “animal,” “patient,” and “subject” as used herein include, but are not limited to, humans and non-human vertebrates such as wild, domestic and farm animals. According to some embodiments, the terms “animal,” “patient,” and “subject” may refer to humans. According to some embodiments, the terms “animal,” “patient,” and “subject” may refer to non-human mammals. [0144] The term “AUC” or area under the concentration-time curve of drug absorption and elimination is a measure of total systemic exposure to a drug. [0145] The term “batch” as used herein refers to a specific quantity of a drug or other material produced in a process or series of processes so that it is expected to have uniform character and quality, within specified limits. The batch size can be defined either by a fixed quantity or by the amount produced in a fixed time interval. [0146] The term “batch formula (composition)” as used herein refers to a complete list of the ingredients and their amounts to be used for the manufacture of a representative batch of the drug product. [0147] The term “bioavailable” and its other grammatical forms refers to the extent and rate at which an active agent, such as a drug or a metabolite thereof, enters systemic circulation, thereby accessing the site of action. According to some embodiments, bioavailability may be described as the fraction (%) of an administered drug that reaches the systemic circulation. [0148] The term “biocompatible” as used herein refers to a material that is generally non- toxic to the recipient and does not possess any significant untoward effects to the subject and, further, that any metabolites or degradation products of the material are non-toxic to the subject. Typically, a substance that is “biocompatible” causes no clinically relevant tissue irritation, injury, toxic reaction, or immunological reaction to living tissue. [0149] The term “biodegradable” as used herein refers to a material that will erode to soluble species or that will degrade under physiologic conditions to smaller units or chemical species that are, themselves, non-toxic (biocompatible) to the subject and capable of being metabolized, eliminated, or excreted by the subject. In general, “biodegradable polymers” can be degraded into low-molecular-weight compounds by a biological process, such as by an enzymatic process or a metabolic process in microorganisms. More generally, biodegradable polymers can be degraded under a natural environment or a physiological condition by either of enzymatic degradation or spontaneous hydrolysis. Biodegradable polymers useful in the embodiments of the invention include, but are not limited to, polyesters, poly(lactide), poly(lactide-co-glycolide) (PLGA), poly(caprolactone), poly(hydroxybutyrates), Eudragit S100 (ES100), and the like. (Vroman I, Tighzert L. Biodegradable Polymers. Materials (Basel). 2009;2(2):307-344) [0150] The term "carrier" as used herein describes a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the compound of the composition of the described invention. Carriers must be of sufficiently high purity and of sufficiently low toxicity to render them suitable for administration to the mammal being treated. The carrier can be inert, or it can possess pharmaceutical benefits. The terms "excipient", "carrier", or "vehicle" are used interchangeably to refer to carrier materials suitable for formulation and administration of pharmaceutically acceptable compositions. Carriers and vehicles useful herein include any such materials know in the art which are nontoxic and do not interact with other components. [0151] The term “Cmax” as used herein refers to the maximum concentration or maximum systemic exposure of a drug. [0152] The term “Css” or steady state concentration is the time during which the concentration of the drug in the body stays consistent. [0153] The term “controlled release” is intended to refer to a drug-containing formulation in which the manner and profile of drug release from the formulation are regulated. This refers to immediate as well as non-immediate release formulations, with non-immediate release formulations including, but not limited to, sustained release and delayed release formulations. [0154] The term “cyclooxygenase (COX)” or “cyclooxygenase isoenzymes” as used herein refers to an enzyme (specifically, a family of isozymes, EC 1.14.99.1) that is responsible for the conversion of certain polyunsaturated fatty acids to prostanoids, including prostaglandins, thromboxane, and levuloglandins. The specific reaction catalyzed is the conversion of arachidonic acid to Prostaglandin H2, via a short-living Prostaglandin G2 intermediate. There are two main isozymes, Cyclooxygenase-1 (COX-1) and Cyclooxygenase-2 (COX-2), which differ in their regulation of expression and tissue distribution. COX-1 is responsible for the synthesis of prostaglandin and thromboxane in many types of cells, including the gastrointestinal tract and blood platelets. For example, COX-1 performs a distinct physiological homeostatic maintenance role within the vascular system and gastrointestinal tract. Inhibition of COX-1 may lead to gastrointestinal toxicity, cardiovascular toxicity, internal bleeding, and stomach ulcers. COX-2 plays a major role in prostaglandin biosynthesis in inflammatory cells and in the central nervous system. The COX-2 enzyme is an inducible, pro- inflammatory cyclooxygenase isoform that is overexpressed and upregulated in many cancer tissues (116). COX-2 overexpression is associated with poor survival outcome in colorectal cancer patients (117) and is used as a prominent predictive marker in inflammatory bowel diseases (118). Prostaglandin synthesis in these sites is an important factor in the development of inflammation and pain. Inhibition of COX can provide relief from the symptoms of inflammation and pain. In particular, COX-2 inhibitors can have analgesic and anti- inflammatory activity by blocking the transformation of arachidonic acid into prostaglandin H2 selectively. [0155] The terms “cyclooxygenase-2 inhibitor”, or “COX-2 inhibitor”, which can be used interchangeably herein, refer to molecules which inhibit the COX-2 enzyme regardless of the degree of inhibition of the COX-1 enzyme, and include pharmaceutically acceptable salts of those compounds. Thus, for purposes of the present invention, a compound is considered a COX-2 inhibitor irrespective of whether the compound inhibits the COX-2 enzyme to an equal, greater, or lesser, degree than the COX-1 enzyme. [0156] The term “COX-2-selective” refers to a molecule that exhibits selective binding to a COX-2 polypeptide. As used herein, “selective binding” means a preferential binding of one molecule for another in a mixture of molecules. In certain embodiments, the binding of an active agent to a target molecule can be considered selective if the binding affinity is about 1×102 M−1 to about 1×106 M−1 or greater. [0157] The term “delayed-release” is used herein in its conventional sense to refer to a drug formulation in which there is a time delay between administration of the formulation and the release of the drug there from. "Delayed release" may or may not involve gradual release of drug over an extended period of time, and thus may or may not be "sustained release." A delayed- release formulation may comprise a “delayed-release coating” to delay release of the active agent. A delayed-release formulation may additionally or alternatively comprise an enteric coating to delay release of an active agent until the tablet or capsule has passed through the stomach to prevent the active agent from being destroyed or inactivated by the gastric environment or where it may irritate the gastric mucosa. [0158] The term “density” is used herein to refer to the degree of compactness of a substance, and is measured in mass per unit volume. [0159] The term “derivative” as used herein means a compound that may be produced from another compound of similar structure in one or more steps. A “derivative” or “derivatives” of a compound retains at least a degree of the desired function of the compound. Accordingly, an alternate term for “derivative” may be “functional derivative.” [0160] The term “displosed” and its other grammatical forms as used herein means to put in order, set, make us of, or arrange. [0161] The term “dissolution rate” as used herein refers to the amount of a drug that dissolves per unit time. The dissolution rate of a drug may be altered by certain conditions, including, but not limited to, surface area, rotation speed, pH and ionic strength of the dissolution medium. [0162] The term “doping” as used herein refers to the intentional introduction of impurities (“dopants”) for the purpose of modulating the properties of a material, e.g., a polymer. According to some embodiments, dopants are introduced to control the releasing profile of drugs. [0163] The terms “drug load (%)” and drug loading capacity” are used interchangeably herein to refer to a ratio of the weight of a drug in microparticles relative to the total weight of the microparticles, expressed as a percentage. It reflects the drug content of the microparticle. [0164] The term “drug product” as used herein refers to a finished dosage form that contains a drug substance, generally, but not necessarily, in association with one or more other ingredients. [0165] The term “drug substance” as used herein refers to an active ingredient intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment or prevention of a disease, or to affect the structure and function of the body, but does not include intermediates used in synthesis of such ingredient. [0166] The term “effective amount” refers to the amount necessary or sufficient to realize a desired biologic effect. [0167] The term “emulsion” as used herein refers to a two-phase system prepared by combining two immiscible liquid carriers, one of which is disbursed uniformly throughout the other and consists of globules that have diameters equal to or greater than those of the largest colloidal particles. The globule size must be such that the system achieves maximum stability. Usually, separation of the two phases will occur unless a third substance, an emulsifying agent, is incorporated. Thus, a basic emulsion contains at least three components, the two immiscible liquid carriers and the emulsifying agent, as well as the active ingredient. Most emulsions incorporate an aqueous phase into a non-aqueous phase (or vice versa). However, it is possible to prepare emulsions that are basically non-aqueous, for example, anionic and cationic surfactants of the non-aqueous immiscible system glycerin and olive oil. [0168] The term “enterohepatic recycling (EHR)” refers to the circulation of biliary acids, bilirubin, drugs or other substances from the liver to the bile, followed by entry into the small intestine, absorption by the enterocyte and transport back to the liver. [0169] EHR of drugs relates to the process by which drugs are conjugated to glucuronic acid in the liver, excreted into bile, metabolized back into the free drug by intestinal bacteria, and the drug is then reabsorbed into plasma. For many drugs that undergo this process, lower doses of drugs can be therapeutically effective because elimination is reduced by the recycling of the drug. However, for certain drugs that are mildly toxic to the intestine, this recycling process can enhance their toxicity, and therefore inhibition of recycling may be protective for those drugs. However, for drugs which undergo enterohepatic circulation that are not toxic to the intestine, inhibition of this recycling process can lead to a reduction of the levels of drug and often a reduced therapeutic effect. In general, EHR can slow down the decrease of drug concentrations in the intestinal lumen, and make more unconjugated drug molecules available inside the colonic epithelial cells. [0170] The term “enteric coating” as used herein refers to a polymer barrier applied to an oral formulation, such as a capsule or tablet, that prevents its dissolution or disintegration in the gastric environment. An enteric coating may protect an active agent from the acidity of the stomach (pH of 1.2), the stomach from the detrimental effects of an active agent, or to facilitate release of an active agent after the stomach at the near neutral pH of the distal small intestine (pH of 7.5 ± 0.4) or the ileocecal junction. [0171] The term “entrapment efficiency (%)” as used herein refers to a ratio of drug retained by a particle relative to the total amount available, expressed as a percentage. [0172] The term “erosion rate” refers to the degradation of a polymer over time. Polymers can be degraded actively (by enzymes) or passively (by hydrolysis), resulting in surface or bulk erosion. Surface erosion is a heterogeneous process wherein degradation of the polymer happens at only the surface and the rate is proportional to the surface area. Drug release in surface-eroding systems is often correlated with a predictable erosion rate, which is considered ideal for many drug delivery applications. Erosion begins from the outside and progresses inward. In most cases, thicker systems have longer erosion times, and hydrophilic polymers degrade faster compared with hydrophobic materials. With bulk-eroding systems, degradation is homogenous throughout the material, and the size of the system remains constant in most cases. The drug is released in three stages: burst release from the surface, release from initial degradation of the polymer, and release of residual drug during complete degradation/homogeneous erosion of the polymer. [0173] The term “excipient” is used herein to include any other agent or compound that may be contained in a formulation that is not the bioactive agent. As such, an excipient should be pharmaceutically or biologically acceptable or relevant (for example, an excipient should generally be non-toxic to the subject). “Excipient” includes a single such compound and is also intended to include a plurality of such compounds. [0174] The term “Eudragit S100” or “ES100” as used herein refers to an anionic copolymer composed of methacrylic acid (MAA)-methyl methacrylate (MMA) at a 1:2 ratio. [0175] The term “Familial Adenomatous Polyposis (FAP)” as used herein refers to a rare, inherited condition caused by a defect in the adenomatous polyposis coli (APC) gene. FAP may be diagnosed when a person develops more than 100 adenomatous colon polyps. An adenomatous polyp is an area where normal cells that line the inside of a subject’s colon form a mass on the inside of the intestinal tract. Adenomatous polyps (adenomas) of the colon and rectum are typically benign (noncancerous) growths, but may be precursor lesions to colorectal cancer. Polyps greater than one centimeter in diameter are associated with a greater risk of cancer. If polyps are not removed, they continue to grow and can become cancerous. Adenomatous polyps appear by about 15 years of age in 50% of people with FAP, and by 35 years of age in 95%. Seventy percent of these individuals have colorectal malignancy by the age of 36 years, the average age of symptomatic diagnosis. Over the lifetime of this illness, there is a 100% cancer risk, typically in the fourth and fifth decades of life. The average age of colorectal cancer onset for attenuated familial adenomatous polyposis is 55 years. Clinically, FAP manifests equally in both sexes by the late teens and in the twenties age group. No true pharmacologic treatment is available for these patients. [0176] The term “formulation” as used herein refers to a listing of the ingredients and composition of the dosage form. [0177] The term “impregnate”, as used herein in its various grammatical forms refers to causing to be infused or permeated throughout; to fill interstices with a substance. [0178] The term “leaching” or “solvent extraction” as used herein refers to a process in which a component of a mixture is removed (extracted) by exposing the mixture to the action of a solvent in which the component to be removed is soluble. Electrospun fibrous polyurethane scaffolds in tissue engineering. Porogen leaching is a common approach to developing large, three-dimensional, porous scaffolds. In this established technique, the scaffold material is incorporated with a chemically or physically incompatible porogen. Upon scaffold fabrication via a process such as solvent evaporation or chemical and physical cross-linking, the porogen is selectively removed. One consequence of this method of fabrication is the high degree of porosity (typically in the range of 90% or higher), which is necessary to achieve an interconnected network for random porogen organization. Scaffolds with extremely high porosities are advantageous for tissue engineering metabolically active tissues by allowing for more rapid diffusion of nutrients and removal of waste products. High porosities also provide large surface areas per volume to allow cell attachment and proliferation. However, the mechanical properties of highly porous networks are severely compromised, including a dramatic reduction in the relative stiffness of the porous and solid scaffolds, Ecellular and Esolid, respectively, with increasing porosity, P. Christopher J. Bettinger, et al, in Principles of Tissue Engineering (Third Edition), 2007 [0179] The term “Lewis acid” as used herein refers to any molecule or ion that can combine with another molecule or ion by forming a covalent bond with two electrons from the second molecule or ion. An acid is thus an electron acceptor. Hydrogen is the simplest substance that will do this. [0180] The term “Lewis base” as used herein refers to a substance that forms a covalent bond by donating a pair of electrons, neutralization resulting from reaction between the base and the acid with formation of a coordinate covalent bond. [0181] The term "long-term" release, as used herein, refers to an implant constructed and arranged to deliver therapeutic levels of the active ingredient for at least 7 days, and potentially up to about 30 to about 60 days. Terms such as “long-acting”, “sustained-release” or “controlled release” are used generally to describe a formulation, dosage form, device or other type of technologies used, such as, for example, in the art to achieve the prolonged or extended release or bioavailability of bioactive agent to a subject; it may refer to technologies that provide prolonged or extended release or bioavailability of a bioactive agent to the general systemic circulation or a subject or to local sites of action in a subject including (but not limited to) cells, tissues, organs, joints, regions, and the like. Furthermore, these terms may refer to a technology that is used to prolong or extend the release of a bioactive agent from a formulation or dosage form or they may refer to a technology used to extend or prolong the bioavailability or the pharmacokinetics or the duration of action of a bioactive agent to a subject or they may refer to a technology that is used to extend or prolong the pharmacodynamic effect elicited by a formulation. A “long-acting formulation,” a “sustained release formulation,” or a “controlled release formulation” (and the like) is a pharmaceutical formulation, dosage form, or other technology that is used to provide long-acting release of a bioactive agent to a subject. Generally, long-acting or sustained release formulations comprise a bioactive agent or agents that is/are incorporated or associated with a biocompatible polymer in one manner or another. The agent may be blended homogeneously throughout the polymer or polymer matrix, or the agent may be distributed unevenly (or discontinuously or heterogeneously) throughout the polymer or polymer matrix (as in the case of a bioactive agent-loaded core that is surrounded by a polymer-rich coating or polymer wall forming material as in the case of a microcapsule, nanocapsule, a coated or encapsulated implant, and the like. [0182] The term “manufacture” as used herein refers to all operations of receipt of materials, production, packaging, repackaging, labeling, relabeling, quality control, release, storage and distribution of APIs and related controls. [0183] The term “material” as used herein refers generally to raw materials (e.g., starting materials, reagents, solvents), process aids, intermediates, APIs, packaging and labeling materials. [0184] The term “matrix” as used herein refers to a three-dimensional network of fibers and/or polymers that contains voids (or “pores”) where the woven fibers and/or polymers intersect. The structural parameters of the pores, including the pore size, porosity, pore interconnectivity, tortuosity and surface area, can affect how substances (e.g., fluid, solutes) move in and out of the matrix. [0185] The term “microbiome” or “gut microbiome” as used herein refers to the genes harbored by the microbial cells that constitute the gut microbiota. [0186] The term “microbiota” as used herein refers to the microbial taxa or groups associated with humans. [0187] The term “micronize” and its other grammatical forms as used herein refers to a process that reduces particle size to obtain micrometer- and nanometer-size particles. Micronization may be useful, e.g., to improve the bioavailability of poorly soluble APIs by increasing particle surface area and accelerating dissolution rates; to improve formulation homogeneity and to control particle size. According to some embodiments, the micronization process uses fluid energy, such as a jet mill. A jet mill uses pressurized gas to create high particle velocity and high-energy impact between particles. The process gas is separated from the solid particles after exiting the jet-mill chamber with a cyclone filter. According to some embodiments, the micronization process uses mechanical particle-size reduction, e.g., using a bead mill. Bead milling uses wet mechanical milling to obtain nanoscale particles. In an agitator bead mill, for example, grinding beads and agitating elements are used to reduce the API particle size through impact and shear; product is separated from the grinding media at the outlet. Process parameters include the formulation (e.g., product viscosity, percent solids, additives to prevent reagglomeration), bead density, bead size, bead-filling ratio, stirrer-shaft speed, and flow rate. If containment is needed, the batch-mixing tank can be placed in an isolator, and the mixture can be pumped to the bead mill, which is outside the isolator but is itself a closed system (www.pharmatech.com/using-micronization-reduce-api-particle-size). According to some embodiments, the micronization process comprises dry powder grinding. According to some embodiments, the micronization process comprises wet milling. [0188] The term “microparticulate composition”, as used herein, refers to a composition comprising a microparticulate formulation and, optionally, a pharmaceutically acceptable carrier, where the microparticulate formulation comprises a therapeutic agent and a plurality of microparticles. According to some embodiments, the therapeutic agent is impregnated within the polymer matrix of the microparticles. [0189] The term “milling” and its other grammatical forms as used herein refers to a process (e.g., a machining process) of grinding, pulverizing, pounding, crushing, pressing, or granulating a solid substance to reduce particle size. [0190] The terms “minimum effective concentration”, “minimum effective dose” , or “MEC” are used interchangeably to refer to the minimum concentration of a drug required to produce a desired pharmacological effect in most patients. [0191] The term "modulate" as used herein means to regulate, alter, adapt, or adjust to a certain measure or proportion. [0192] The term “particles” as used herein refers to an extremely small constituent, e.g., nanoparticles or microparticles) that may contain in whole or in part at least one therapeutic agent as described herein. The term “microparticle” is used herein to refer generally to a variety of substantially structures having sizes from about 10 nm to 2000 microns (2 millimeters) and includes microcapsule, microparticle, nanoparticle, nanocapsule, nanosphere as well as particles, in general, that are less than about 2000 microns (2 millimeters). The particles may contain therapeutic agent(s) in a core surrounded by a coating. Therapeutic agent(s) also may be dispersed throughout the particles. Therapeutic agent(s) also may be adsorbed into the particles. The particles may be of any order release kinetics, including zero order release, first order release, second order release, delayed release, sustained release, immediate release, etc., and any combination thereof. The particles may include, in addition to therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof. The particles may be microcapsules that contain the therapeutic agent in a solution or in a semisolid state. The particles may be of virtually any shape. [0193] The term “pharmaceutical composition” is used herein to refer to a composition that is employed to prevent, reduce in intensity, cure or otherwise treat a target condition or disease. [0194] As used herein the phrase “pharmaceutically acceptable carrier” refers to any substantially non-toxic carrier useable for formulation and administration of the composition of the described invention in which the product of the described invention will remain stable and bioavailable. The pharmaceutically acceptable carrier must be of sufficiently high purity and of sufficiently low toxicity to render it suitable for administration to the mammal being treated. It further should maintain the stability and bioavailability of an active agent. The pharmaceutically acceptable carrier can be liquid or solid and is selected, with the planned manner of administration in mind, to provide for the desired bulk, consistency, etc., when combined with an active agent and other components of a given composition. [0195] The term “pharmaceutically acceptable salt” means those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. When used in medicine the salts should be pharmaceutically acceptable, but non- pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts may be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group. By “pharmaceutically acceptable salt” is meant those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well-known in the art. For example, P. H. Stahl, et al. describe pharmaceutically acceptable salts in detail in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” (Wiley VCH, Zurich, Switzerland: 2002). The salts may be prepared in situ during the final isolation and purification of the compounds described within the present invention or separately by reacting a free base function with a suitable organic acid. Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsufonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate(isethionate), lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen- containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained. Examples of acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, hydrobromic acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid. Basic addition salts may be prepared in situ during the final isolation and purification of compounds described within the invention by reacting a carboxylic acid-containing moiety with a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary or tertiary amine. Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as lithium, sodium, potassium, calcium, magnesium and aluminum salts and the like and nontoxic quaternary ammonia and amine cations including ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine and the like. Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine and the like. Pharmaceutically acceptable salts also may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium or magnesium) salts of carboxylic acids may also be made. [0196] The term “pharmacodynamics” as used herein refers to the study of the biochemical, physiologic, and molecular effects of drugs on the body and involves receptor binding (including receptor sensitivity), postreceptor effects, and chemical interactions. [0197] The term “pharmacokinetics” as used herein refers to the study of the time course of absorption, distribution, metabolism, and excretion of a drug. Clinical pharmacokinetics is the application of pharmacokinetic principles to the safe and effective therapeutic management of drugs in an individual patient. [0198] The term “pharmacologic effect”, as used herein, refers to a result or consequence of exposure to an active agent. [0199] The term “pilot scale” as used herein refers to the manufacture of either a drug substance or drug product by a procedure fully representative of and simulating that used for full manufacturing scale. In production of microspheres, pilot scale can be, for example, 500 grams. For an API, pilot scale can be, for example 1 kg. [0200] The term “poly(lactic-co-glycolic acid)” or “PLGA” as used herein refers to a linear copolymer that can be prepared at different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA). Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained, and these are usually identified in regard to the monomers’ ratio used (e.g., PLGA 85:15 identifies a copolymer consisted of 85% lactic acid and 15% glycolic acid). Poly lactic acid contains an asymmetric α-carbon which is typically described as the D or L form in classical stereochemical terms and sometimes as R and S form, respectively. The enantiomeric forms of the polymer PLA are poly D-lactic acid (PDLA) and poly L-lactic acid (PLLA). PLGA is generally an acronym for poly D,L-lactic-co- glycolic acid where D- and L- lactic acid forms are in equal ratio. Different synthesis mechanisms as known in the art can be used to obtain PLGA having different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA). (Gentile P, Chiono V, Carmagnola I, Hatton PV. An overview of poly(lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. Int J Mol Sci.2014;15(3):3640-3659; Makadia HK, Siegel SJ. Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers (Basel).2011;3(3):1377-1397). [0201] The term “polymer” refers to a large molecule, or macromolecule, composed of many repeated subunits. The term “monomer” refers to a molecule that may bind chemically to other molecules to form a polymer. The term “copolymer” as used herein refers to a polymer derived from more than one species of monomer. [0202] The term “polar” as used herein refers to a molecule in which the positive and negative electrical charges are permanently separated, as opposed to nonpolar molecules in which the charges coincide. Polar molecules ionize in solution and impart electrical conductivity. Water, alcohol and sulfuric acid are polar in nature; most hydrocarbon liquids are not. Carboxyl and hydroxyl groups often exhibit an electric charge. [0203] The term “polar molecule” as used herein refers to a molecule with a positive charge on one end and a negative charge on its other end; or a molecule in which the electrons forming the valency bond are not symmetrically arranged. [0204] The term “pore” as used herein refers to a void or interstices between particles of a solid or in a matrix that permits passages of liquids or gases through the material in either direction. [0205] The term “porogen” as used herein refers to a substance used to create pores in a structure. [0206] The term “porosity” as used herein refers to the ratio of the volume of pores to the volume of the material as a whole; it , and is usually expressed as a percentage. [0207] The term “process” as used herein refers to a series of operations, actions and controls used to manufacture a drug product. [0208] The term “production” as used herein refers to all operations involved in the preparation of an API from receipt of materials through processing and packaging of the API. [0209] The term “reduce” or “reducing” as used herein refers to a diminution, a decrease, an attenuation, limitation or abatement of the degree, intensity, extent, size, amount, density, number or occurrence of disorder in individuals at risk of developing the disorder. [0210] The term “release” and its various grammatical forms, refers to dissolution of an active drug component and diffusion of the dissolved or solubilized species by a combination of the following processes: (1) hydration or swelling of a matrix, (2) erosion of a matrix, (3) diffusion of a solution into the matrix; (4) dissolution of the drug; (5) diffusion of the dissolved drug out of the matrix, (6) pH-responsive release mechanism, and (7) microbially triggered delivery. In certain embodiments, release may comprise a pH-responsive release mechanism. A pH-sensitive drug delivery system, e.g., for oral administration, may withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ± 0.4) or the ileocecal junction. As used herein, a “pH-sensitive” polymer coating means that the coating material dissolves or elutes under circumstances of more than a given pH value to release an active agent. An enteric coating may be dissolved or eluted at a pH of about 5.5 to initiate drug release, while the enteric coating comprising a pH-sensitive polymer of the present invention is preferably a polymer that dissolves at a higher pH and controls drug release to the colon. [0211] The term “salt” as used herein refers to the compound formed when the hydrogen of an acid is replaced by a metal or its equivalent (e.g., an NH4+ radical). This is typical of the general rule that the reaction of an acid and a base yields a salt and water. Most inorganic acids ionize in water solution. [0212] The terms “soluble” and “solubility” refer to the property of being susceptible to being dissolved in a specified fluid (solvent). The term “insoluble” refers to the property of a material that has minimal or limited solubility in a specified solvent. In a solution, the molecules of the solute (or dissolved substance) are uniformly distributed among those of the solvent. [0213] The term “solvate” as used herein refers to a complex formed by the attachment of solvent molecules to that of a solute. [0214] The term “solvent” refers to a an inorganic or organic liquid capable of dissolving another substance (termed a “solute”) to form a uniformly dispersed mixture (solution) used as a vehicle for the preparation of solutions or suspensions. [0215] The term “solvent evaporation method” as used herein refers to a technique for preparing drug-loaded particles in which an active agent is dissolved, dispersed or emulsified into an organic polymer solution, which is then emulsified into an external aqueous or oil phase. Particles are formed after solvent evaporation and polymer precipitation. Size of the particles can be controlled by adjusting parameters like manipulating evaporation temperature, controlling the rate of evaporation, manipulating stirring rate, etc. [0216] The term “specification” as used herein refers to a list of tests, references to analytical procedures, and appropriate acceptance criteria that are numerical limits, ranges or other criteria for the test described that establishes the set of criteria to which material should conform to be considered acceptable for its intended use. The term “conformance to specification” means that the material, when tested according to the listed analytical procedures, will meet the listed acceptance criteria. [0217] The terms "subject" or "individual" or "patient" are used interchangeably to refer to a member of an animal species of mammalian origin, including humans. [0218] The terms “sufficient amount” and “sufficient time” as used herein refer to an amount and time needed to achieve the desired result or results. [0219] A “suspension” is a dispersion (mixture) in which a finely-divided species is combined with another species, with the former being so finely divided and mixed that it doesn't rapidly settle out. In everyday life, the most common suspensions are those of solids in liquid. [0220] The term “sustained release” (also referred to as “extended release”) is used herein in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period. [0221] The term “symptom” as used herein refers to a phenomenon that arises from and accompanies a particular disease or disorder and serves as an indication of it. [0222] The term “therapeutic agent” as used herein refers to a drug, molecule, composition or other substance that provides a therapeutic effect. The terms “therapeutic agent” and “active agent” are used interchangeably. [0223] The term “therapeutic component” as used herein refers to a therapeutically effective dosage (i.e., dose and frequency of administration) that eliminates, reduces, or prevents the progression of a particular disease manifestation in a percentage of a population. An example of a commonly used therapeutic component is the ED50 which describes the dose in a particular dosage that is therapeutically effective for a particular disease manifestation in 50% of a population. [0224] The term “therapeutic effect” as used herein refers to a consequence of treatment, the results of which are judged to be desirable and beneficial. A therapeutic effect may include, directly or indirectly, the arrest, reduction, or elimination of a disease manifestation. A therapeutic effect may also include, directly or indirectly, the arrest reduction or elimination of the progression of a disease manifestation. [0225] The term “therapeutically effective amount”, “effective amount”, or an “amount effective” is an amount that is sufficient to provide the intended benefit of treatment. Combined with the teachings provided herein, by weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen may be planned which does not cause substantial toxicity and yet is effective to treat the particular subject. The therapeutically effective amount for any particular application may vary depending on such factors as the disease or condition being treated, the particular active agent being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art may determine empirically the effective amount of a particular active agent and/or other therapeutic agent without necessitating undue experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to some medical judgment. “Dose” and “dosage” are used interchangeably herein. [0226] The term “Tmax” as used herein refers to the time of maximum concentration or maximum systemic exposure of a drug. [0227] The term “t1/2 or half-life” as used herein refers to the time required to reduce plasma concentration of a drug to one-half of its initial value. [0228] The term “treat” or “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease, condition or disorder, substantially ameliorating clinical or esthetical symptoms of a condition, substantially preventing the appearance of clinical or esthetical symptoms of a disease, condition, or disorder, and protecting from harmful or annoying symptoms. Treating further refers to accomplishing one or more of the following: (a) reducing the severity of the disorder; (b) limiting development of symptoms characteristic of the disorder(s) being treated; (c) limiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting recurrence of symptoms in patients that were previously asymptomatic for the disorder(s). [0229] As used herein, a “wt. %” or “weight percent” or “percent by weight” of a component, unless specifically stated to the contrary, refers to the ratio of the weight of the component to the total weight of the composition in which the component is included, expressed as a percentage. Embodiments Pharmaceutical Compositions [0230] According to one aspect, the described invention provides pharmaceutical compositions for delivering a therapeutic agent to the colon of a subject. According to some embodiments, the described invention provides pharmaceutical compositions to delay and sustain the release of an active agent. According to some embodiments, the active agent is designed to be bioavailable in the colon (LBD), to realize low systemic drug exposure and to avoid off-organ toxicities. According to some embodiments, the active agent is a selective COX-2 inhibitor. According to some embodiments, the selective COX-2 inhibitor is 6A1 [0231] To achieve the proposed objectives, two different drug delivery systems were formulated. The first drug delivery system delays release via a coating technique in combination with a coating formulation that includes a pH-sensitive polymer and an erosion polymer for hard gelatin capsules. [0232] The second drug delivery system also uses hard gelatin capsules containing 6A1 enterically encapsulated microparticles aiming for a sustained release profile. [0233] According to According to some embodiments, the coating formulation achieves two objectives: the enteric biodegradable polymer coating will prevent early release of LBD COX-2 inhibitors (6A1) in the stomach that facilitate its absorption in the upper small intestine; the erosion mechanism makes the delayed release in the colon more precise, which could result in a smaller drug dose but higher colonic drug exposure. Delaying the release of 6A1 to the colon eliminates the impact of the inter-subject variability due to uridine 5'-diphospho- glucuronosyl- transferase (UGT) expression in the liver, beta-glucuronidase activities expressed by colonic microflora, and colonic disease states. Additionally, delayed release delivery ensures a sufficient and consistent amount of 6A1 is present in the colon to saturate COX-2 enzymes. [0234] The provided microparticle formulations will sustain the release of 6A1 via two mechanisms: slow erosion of the microparticles and accumulation of the microparticles within the targeted site via its enhanced solubility and increased surface area. In combination with enterohepatic recycling (EHR), the provided dosage forms will preclude or minimize systemic distribution of LBD COX-2 inhibitors and allow for rapid attainment and maintenance of adequate therapeutic concentrations in the colon. According to some embodiments, the biodegradable polymer formulations and coating technique will synergistically function as a successful drug delivery system for treating diseases in the colon. According to some embodiments, pharmaceutical compositions provided herein are intended to fill a void in the understanding of how to create safe COX-2 inhibitors that prevent colonic cancer in FAP patients. [0235] According to some embodiments, the pharmaceutical compositions comprise one or more polymeric coatings to control release, delay release, and/or sustain release of a therapeutic agent. [0236] According to some embodiments, the described invention provides pharmaceutical compositions comprising a particulate formulation containing a plurality of particles. [0237] According to some embodiments, the present invention disclosure provides an oral pharmaceutical composition comprising: (a) a capsule comprising: (i) an inner delayed-release coating comprising a biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a therapeutically effective amount of a therapeutic agent disposed within the capsule. According to some embodiments, the biodegradable polymer is doped to modulate porosity of the polymer. [0238] According to some embodiments, the present invention disclosure provides an oral pharmaceutical composition comprising: (a) a capsule comprising: (i) an inner delayed-release coating comprising a biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a plurality of particles disposed within the capsule, each comprising a therapeutically effective amount of a therapeutic agent and one or more polymers encapsulating the therapeutic agent and which release the therapeutic agent at a pH above 6.0. According to some embodiments, the biodegradable polymer is doped to modulate porosity of the polymer. [0239] According to some embodiments, wherein when the pharmaceutical composition is orally administered to a subject there is a lag period of at least about 6 hours (e.g., at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, or at least about 12 hours) after administration before sustained release of the therapeutic agent occurs at a pH above 5.5 (e.g., a pH between about 6 and about 8). [0240] According to some embodiments, the described invention provides pharmaceutical compositions to (i) reduce the therapeutic dose of an active agent, (ii) reduce the total systemic exposure (AUC) of an active agent, (iii) increase the local colonic exposure (AUCcolon) of an active agent, (iv) increase the concentration of the active agent in the colon (e.g., lower AUC0- 24Hours, increase local AUCcolon), and/or (v) reduce the occurrence of on target off organ systemic toxicity (e.g., cardiovascular side effects, such as unstable angina, myocardial infarction, and cardiac thrombus) of an active agent. [0241] according to some embodiments, the pharmaceutical compositions are formulated for enterohepatic recycling (EHR). [0242] According to some embodiments, the described invention provides multi-modal oral delayed-release formulations to sustain therapeutic agent exposure in the colon (lower AUC0-24Hours, increase local AUCcolon). According to some embodiments, the described invention provides pharmaceutical compositions having a dual release mechanism with pH resistance, and comprising varying particle sizes ranging from 100-2000 nm, inclusive to achieve the highest ratio of drug accumulation in the colon over a given dose. According to some embodiments, the dual-release mechanism design can be used to achieve maximum efficacy without subjecting the patients to a large dose and potentially reduce the dosing frequency for a therapeutic agent. In particular embodiments, Eudragit S100 pH-sensitive polymer and PLGA 5050 can be used to formulate oral sustained release nano- and microparticles of therapeutic agent. According to some embodiments, the described invention provides pharmaceutical compositions that sustained drug release above pH 6.0 and delay drug release for at least about 6 hours after administration. According to some embodiments, the described invention provides pharmaceutical compositions comprising and microparticle formulations that are highly stable, have a high encapsulation efficiency, and exhibit a sustained release profile. [0243] For oral administration, the pharmaceutical compositions can be formulated readily by combining the therapeutic agent (s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the actives of the disclosure to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, alter adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP). If desired, disintegrating agents can be added, such as, but not limited to, the cross- linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [0244] Pharmaceutical preparations that can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, scaled capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration. [0245] According to some embodiments, the capsule may be formed of any suitable material, be any suitable size, and be either hard or soft. According to some embodiments, the capsule may comprise gelatin, collagen, and/or cellulose. [0246] In some embodiments, the capsule may be a hard gelatin capsule, for example, a size nine hard gelatin capsule (S9C). Without wishing to be bound by any particular theory, hard gelatin capsules provide an advantage over other solid dosage forms, in that the therapeutic agent can be packed inside the casing as a powder and/or a particulate formulation thereby reducing the need for excipients and extensive formulations (146, 147). Hard gelatin capsules are hydrogenated collagen made up of abundant proteins that are readily broken down into amino acids and become available for absorption within hours of ingestions (148). [0247] However, the coating of gelatin capsules can be challenging. The smooth gelatin surface can cause poor adhesion. Extreme pH or organic solvent usage in the polymer film can cause structural deformity and compromise the integrity of the capsules (149, 150). The application process of enteric and erosion films for the size nine hard gelatin capsule (S9C, 2.71 mm x 8.4 mm with a surface area of 68 mm2), specifically designed for rodents and guinea pigs, is especially challenging and cumbersome with the conventional methods and dip-coating techniques. Conventional coating equipment and the specific parameters are also inapplicable to the S9C in small quantities for experimental use. The novel coating technique described here is important for preclinical research and pilot studies where the amount of the new chemical entity is usually limited. [0248] According to some embodiments, the pharmaceutical compositions can be formulated with a pharmaceutically acceptable carrier. The phrase “pharmaceutically acceptable carrier” is art recognized. It is used to mean any substantially non-toxic carrier conventionally useable for administration of pharmaceuticals in which the therapeutic agents of the present invention will remain stable and bioavailable. The pharmaceutically acceptable carrier must be of sufficiently high purity and of sufficiently low toxicity to render it suitable for administration to the subject being treated. It further should maintain the stability and bioavailability of a therapeutic agent. The pharmaceutically acceptable carrier can be liquid or solid and is selected, with the planned manner of administration in mind, to provide for the desired bulk, consistency, etc., when combined with an active agent and other components of a given composition. Exemplary carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin, which is incorporated herein by reference in its entirety. According to some embodiments, the pharmaceutically acceptable carrier is sterile and pyrogen-free water. According to some embodiments, the pharmaceutically acceptable carrier is Ringer’s Lactate, sometimes known as lactated Ringer’s solution. [0249] Additives used with the pharmaceutical compositions described herein include, for example, one or more excipients, one or more antioxidants, one or more stabilizers, one or more preservatives (e.g., including antimicrobial preservatives), one or more pH adjusting and/or buffering agents, one or more tonicity adjusting agents, one or more thickening agents, one or more suspending agents, one or more binding agents, one or more viscosity enhancing agents, one or more sweetening agent and the like, either alone or together with one or more additional pharmaceutical agents, provided that the additional components are pharmaceutically acceptable. According to some embodiments, the formulation may include combinations of two or more of the additional components as described herein (e.g., any of 2, 3, 4, 5, 6, 7, 8, or more additional components). [0250] According to some embodiments, the additives include processing agents and drug delivery modifiers and enhancers, such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or more thereof. Other suitable pharmaceutically acceptable excipients are described in Remington's Pharmaceutical Sciences, Mack Pub. Co., New Jersey 18th edition (1996), Handbook of Pharmaceutical Excipients, Pharmaceutical Press and American Pharmacists Association, 5th edition (2006), and Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Philadelphia, 20th edition (2003) and 21st edition (2005). [0251] Examples of pharmaceutically acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. [0252] Some examples of suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, tragacanth, gelatin, syrup, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium stearate, water, and mineral oil. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents. The compositions may be formulated so as to provide quick, controlled, sustained, or delayed release of the therapeutic agent after administration to the patient by employing procedures well known in the art. [0253] Also provided are unit dosage forms comprising the pharmaceutical composition and formulations described herein. These unit dosage forms can be stored in a suitable packaging in single or multiple unit dosages and may also be further sterilized and sealed. Particulate Formulation [0254] According to another aspect, the described invention provides a particulate formulation comprising a plurality of particles. [0255] According to some embodiments, the particulate formulation comprises a plurality of particles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each particle, adsorbed onto the particle, or is in a core surrounded by a coating. [0256] According to some embodiments, the particulate formulation comprises a plurality of microparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each microparticle, adsorbed onto the microparticles, or in a core surrounded by a coating. According to some embodiments, the microparticles are formed by a microencapsulation process such that the outside of the microparticles contains the polymer, and the interior of the microparticles contains the drug in a semifluid state. According to some embodiments, the particulate formulation comprises a plurality of nanoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each nanoparticle, adsorbed onto the nanoparticles, or in a core surrounded by a coating. According to some embodiments, the particulate formulation comprises a plurality of picoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each picoparticle, adsorbed onto the picoparticles, or in a core surrounded by a coating. According to some embodiments, the particulate formulation comprises a plurality of femtoparticles comprising a therapeutic amount of a therapeutic agent, wherein the therapeutic agent is dispersed throughout each femtoparticle, adsorbed onto the femtoparticles, or in a core surrounded by a coating. [0257] According to some embodiments, the particulate formulation comprises a plurality of particles from about 1 nm to about 2000 nm, about 1 nm to about 1000 nm, about 1 nm to about 500 nm, inclusive, about 100 nm to about 300 nm, inclusive, or about 200 nm to about 500 nm, inclusive in particle size. According to some embodiments, the particulate formulation comprises a plurality of particles comprising a particle size of about 1 nm, about 5 nm, about 10 nm, about 15 nm, about 20 nm, about 25 nm, about 30 nm, about 35 nm, about 40 nm, about 45 nm, about 50 nm, about 55 nm, about 60 nm, about 65 nm, about 70 nm, about 75 nm, about 80 nm, about 85 nm, about 90 nm, about 95 nm, about 100 nm, about 150 nm, about 200 nm, about 250 nm, about 300 nm, about 350 nm, about 400 nm, about 450 nm, about 500 nm, about 550 nm, about 600 nm, about 650 nm, about 700 nm, about 750 nm, about 800 nm, about 850 nm, about 900 nm, about 950 nm, about 1000 nm, about 1050 nm, about 1100 nm, about 1150 nm, about 1200 nm, about 1250 nm, about 1300 nm, about 1350 nm, about 1400 nm, about 1450 nm, about 1500 nm, about 1550 nm, about 1600 nm, about 1650 nm, about 1700 nm, about 1750 nm, about 1800 nm, about 1850 nm, about 1900 nm, about 1950 nm, or about 2000 nm. [0258] According to some embodiments, the particles of the particulate formulation are of a uniform distribution of particle size. According to some embodiments, the uniform distribution of particle size is achieved by a non-emulsion-based homogenization process. According to some embodiments, the uniform distribution of particle size is achieved by an emulsion-based process to form a uniform emulsion. According to some embodiments, the particles of the particulate formulation have a varying particle size, e.g., ranging from 100- 2000 nm, inclusive. [0259] According to some embodiments, the therapeutic agent is micronized. [0260] According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) is disposed on or in the particles. According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) is dispersed throughout the particles. According to some embodiments, the particles are impregnated with the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) is adsorbed onto a surface of the particles. According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) is contained within a core of the particles surrounded by a coating. According to some embodiments, the particles comprise a matrix. According to some embodiments, the matrix comprises the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the matrix is impregnated with the therapeutic agent (e.g., micronized therapeutic agent). [0261] According to some embodiments, the particles are loaded with an average of at least 5% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 10% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 15% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 20% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 25% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 30% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 35% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 40% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 45% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 50% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 55% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 60% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 63% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 65% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 70% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 75% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 80% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 85% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 90% by weight of the therapeutic agent (e.g., micronized therapeutic agent). According to some embodiments, the particles are loaded with an average of at least 95% by weight of the therapeutic agent (e.g., micronized therapeutic agent). [0262] According to some embodiments, the particles have a encapsulation efficiency (EE%) of the therapeutic agent (e.g., micronized therapeutic agent) of about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%. [0263] According to some embodiments, microparticles of the present invention comprise about 0.1 µg to about 2 µg therapeutic agent (e.g., micronized therapeutic agent). For example, the microparticles of the invention may comprise about 0.1 µg, about 0.15 µg, about 0.2 µg, about 0.25 µg, about 0.3 µg, about 0.35 µg, about 0.4 µg, about 0.45 µg, about 0.5 µg, about 0.55 µg, about 0.6 µg, about 0.65 µg, about 0.7 µg, about 0.75 µg, about 0.8 µg, about 0.85 µg, about 0.9 µg, about 0.95 µg, about 1 µg, about 1.05 µg, about 1.1 µg, about 1.15 µg, about 1.2 µg, about 1.25 µg, about 1.3 µg, about 1.35 µg, about 1.4 µg, about 1.45 µg, about 1.5 µg, about 1.55 µg, about 1.6 µg, about 1.65 µg, about 1.7 µg, about 1.75 µg, about 1.8 µg, about 1.85 µg, about 1.9 µg, about 1.95 µg, or about 2 µg of the therapeutic agent (e.g., micronized therapeutic agent). [0264] According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) formulated into the pharmaceutical composition for colonic delivery comprises a COX-2 inhibitor. For example, according to some embodiments, the therapeutic agent comprises a locally bioavailable COX-2 inhibitor. (See WO/2016/172159, incorporated herein by reference). [0265] According to some embodiments, the therapeutic agent formulated into the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C), or a derivative thereof. [0266] Various forms of the therapeutic agent can be used, which are capable of being released from the controlled release, delayed release, and/or sustained release system into adjacent tissues or fluids, e.g., of the colon. According to some embodiments, the micronized therapeutic agent can be in liquid or solid form. According to some embodiments, the micronized therapeutic agent is very slightly water soluble, moderately water soluble, or fully water soluble. [0267] According to some embodiments, the therapeutic agent (e.g., micronized therapeutic agent) agent can include salts of the therapeutic agent. As such, the therapeutic agent (e.g., micronized therapeutic agent) agent can be an acidic, basic, or amphoteric salt; it can be a nonionic molecule, a polar molecule, or a molecular complex capable of hydrogen bonding; or the therapeutic agent (e.g., micronized therapeutic agent) can be included in the compositions in the form of, for example, an uncharged molecule, a molecular complex, a salt, an ether, an ester, an amide, polymer drug conjugate, or other form to provide the effective biological or physiological activity. [0268] According to some embodiments, the particles can be formulated for controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent), e.g., to the colon. According to some embodiments, the microparticulate formulation is characterized by controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour to 24 hours, e.g., to the colon. According to some embodiments, the microparticulate formulation is characterized by sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, or at least about 24 hours, e.g., to the colon. [0269] According to some embodiments, the particles can be of any order release kinetics, including zero order release, first order release, second order release, delayed release, sustained release, immediate release, and a combination thereof. In addition to therapeutic agent(s), the particles can include any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof. [0270] According to some embodiments, the particles can release a therapeutic agent by any of the following processes: (1) hydration or swelling of a matrix, (2) erosion of a matrix, (3) diffusion of a solution into the matrix; (4) dissolution of the drug; (5) diffusion of the dissolved drug out of the matrix, (6) pH-responsive release mechanism, and/or (7) microbially triggered delivery. In certain embodiments, release may comprise a pH-responsive release mechanism. A pH-sensitive drug delivery system, e.g., microparticle, may withstand the acidic environment of the stomach (1.2) and deliver a drug at the near neutral pH of the distal small intestine (7.5 ± 0.4) or the ileocecal junction. According to some embodiments, the particles are formulated to deliver a therapeutic agent to the colon. [0271] According to some embodiments, the particles can be formulated for controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent), e.g., to the colon. According to some embodiments, the microparticulate formulation is characterized by controlled release, delayed release, and/or sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour to 24 hours, e.g., to the colon. According to some embodiments, the microparticulate formulation is characterized by sustained release of the therapeutic agent (e.g., micronized therapeutic agent) agent over at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, at least about 24 hours, e.g., to the colon. [0272] Any biodegradable polymeric material can be used in the manufacture of particles for release of a therapeutic agent as described herein. Such polymers may be natural or synthetic polymers. The polymer may be selected based on the period of time over which release is desired and/or the location at which release is desired. [0273] Exemplary criteria for selection of a polymer(s) for use in the described microparticulate formulations include, without limitation, the type of polymer, the selection of a copolymer, the pH-sensitivity of the polymer or copolymer, the type of co-monomers used in the copolymer, the ratio of the types of monomers used in the co-polymer, the molecular weight of the polymer, the size of the microparticle, and any other criteria used by one of skill in the art to control the release profile of a microparticle. According to some embodiments, the particles may be polymeric pH-sensitive particles that are insoluble at a stomach low pH of about 1.5 to about 3.5, but are soluble at an intestinal neutral pH of about 6 to about pH 7.4. [0274] Exemplary biocompatible biodegradable polymers useful for manufacturing the particles of the invention include, without limitation, a poly(lactide); a poly(glycolide); a poly(lactide-co-glycolide) (PLGA); a poly(lactic acid); a poly(glycolic acid); a poly(lactic acid-co-glycolic acid); poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F); a poly(vinyl alcohol) (PVA); a poly(caprolactone); a poly(orthoester); a polyanhydride; a poly(phosphazene); a polyhydroxyalkanoate; a poly(hydroxybutyrate); a poly(hydroxybutyrate) synthetically derived; a poly(hydroxybutyrate) biologically derived; a polyester synthetically derived; a polyester biologically derived; a poly(lactide-co- caprolactone); a poly(lactide-co-glycolide-co-caprolactone); a polycarbonate; a tyrosine polycarbonate; a polyamide (including synthetic and natural polyamides, polypeptides, poly(amino acids) and the like); a polyesteramide; a polyester; a poly(dioxanone); a poly(alkylene alkylate); a polyether (such as polyethylene glycol, PEG, and polyethylene oxide, PEO); polyvinyl pyrrolidone or PVP; a polyurethane; a polyetherester; a polyacetal; a polycyanoacrylate; a poly(oxyethylene)/ poly(oxypropylene) copolymer; a polyacetal, a polyketal; a polyphosphate; a (phosphorous-containing) polymer; a polyphosphoester; a polyhydroxyvalerate; a polyalkylene oxalate; a polyalkylene succinate; and a poly(maleic acid). According to some embodiments, the particles may comprise poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the particles may comprise poly(lactide-co-glycolide) (PLGA). According to some embodiments, the particles may comprise poly(lactide-co-glycolide) (PLGA) and poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the particles may comprise Eudragit S100 pH-sensitive polymer and/or PLGA 5050. [0275] When the biodegradable polymer is poly(lactide-co-glycolide), poly(lactide), or poly(glycolide), the amount of lactide and glycolide in the polymer can vary. For example, according to some embodiments, the biodegradable polymer contains 0 to 100 mole %, 40 to 100 mole %, 50 to 100 mole %, 60 to 100 mole %, 70 to 100 mole %, or 80 to 100 mole % lactide and from 0 to 100 mole %, 0 to 60 mole %, 10 to 40 mole %, 20 to 40 mole %, or 30 to 40 mole % glycolide, wherein the amount of lactide and glycolide is 100 mole %. According to some embodiments, the biodegradable polymer can be poly(lactide), 95:5 poly(lactide-co- glycolide) 85:15 poly(lactide-co-glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide), where the ratios are mole ratios. [0276] It is understood that any combination of the aforementioned biodegradable polymers can be used, including, but not limited to, copolymers, mixtures, or blends thereof. [0277] Exemplary particulate formulations for the sustained release of 6A1 are provided in Table 7. [0278] According to some embodiments, the particulate formulation is presented as a solution, an emulsion, a suspension of particles, or a powder (e.g., a lyophilized powder). [0279] According to some embodiments, the particulate formulation can be in powder form for incorporation into a pharmaceutical composition, such as a capsule, of the invention. Polymeric Coatings [0280] According to another aspect, the described invention provides pharmaceutical compositions for delivering a therapeutic agent to the colon of a subject comprising one or more polymeric coatings to control release, delay release, and/or sustain release of a therapeutic agent. [0281] Any biodegradable polymeric material can be used in the manufacture of polymeric coatings for controlling, delaying, and/or sustaining the release of a therapeutic agent as described herein. Such polymers may be natural or synthetic polymers. The polymer may be selected based on the period of time over which release is desired and/or the location at which release is desired. [0282] Exemplary criteria for selection of a polymer(s) for use in the described polymeric coatings include, without limitation, the type of polymer, the selection of a copolymer, the pH- sensitivity of the polymer or copolymer, the type of co-monomers used in the copolymer, the ratio of the types of monomers used in the co-polymer, the molecular weight of the polymer, the size of the microparticle, and any other criteria used by one of skill in the art to control the release profile of a microparticle. [0283] According to some embodiments, the enteric coating may comprise any suitable pH-sensitive polymer. For example, anionic polymers with carboxyl groups have higher water solubility at basic pH than at acidic pH. According to some embodiments, the enteric coating is insoluble at a stomach low pH of about 1.5 to about 3.5, but are soluble at an intestinal neutral pH of about 6 to about pH 7.4. These polymers may be used for preventing gastric degradation of an therapeutic agent and to achieve delivery to the colon. [0284] Exemplary biocompatible biodegradable polymers useful for manufacturing the particles of the invention include, without limitation, a poly(lactide); a poly(glycolide); a poly(lactide-co-glycolide) (PLGA); a poly(lactic acid); a poly(glycolic acid); a poly(lactic acid-co-glycolic acid); poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F); a poly(vinyl alcohol) (PVA); a poly(caprolactone); a poly(orthoester); a polyanhydride; a poly(phosphazene); a polyhydroxyalkanoate; a poly(hydroxybutyrate); a poly(hydroxybutyrate) synthetically derived; a poly(hydroxybutyrate) biologically derived; a polyester synthetically derived; a polyester biologically derived; a poly(lactide-co- caprolactone); a poly(lactide-co-glycolide-co-caprolactone); a polycarbonate; a tyrosine polycarbonate; a polyamide (including synthetic and natural polyamides, polypeptides, poly(amino acids) and the like); a polyesteramide; a polyester; a poly(dioxanone); a poly(alkylene alkylate); a polyether (such as polyethylene glycol, PEG, and polyethylene oxide, PEO); polyvinyl pyrrolidone or PVP; a polyurethane; a polyetherester; a polyacetal; a polycyanoacrylate; a poly(oxyethylene)/ poly(oxypropylene) copolymer; a polyacetal, a polyketal; a polyphosphate; a (phosphorous-containing) polymer; a polyphosphoester; a polyhydroxyvalerate; a polyalkylene oxalate; a polyalkylene succinate; and a poly(maleic acid). According to some embodiments, the particles may comprise poly(methacrylic acid-co- methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the coating may comprise poly(lactide-co-glycolide) (PLGA). According to some embodiments, the coating may comprise poly(methacrylic acid-co-methyl methacrylate) (Eudragit L, S and F). According to some embodiments, the particles may comprise Eudragit S100 pH-sensitive polymer, PLGA 8515, and/or PLGA 5050. [0285] When the biodegradable polymer is poly(lactide-co-glycolide), poly(lactide), or poly(glycolide), the amount of lactide and glycolide in the polymer can vary. For example, according to some embodiments, the biodegradable polymer contains 0 to 100 mole %, 40 to 100 mole %, 50 to 100 mole %, 60 to 100 mole %, 70 to 100 mole %, or 80 to 100 mole % lactide and from 0 to 100 mole %, 0 to 60 mole %, 10 to 40 mole %, 20 to 40 mole %, or 30 to 40 mole % glycolide, wherein the amount of lactide and glycolide is 100 mole %. According to some embodiments, the biodegradable polymer can be poly(lactide), 95:5 poly(lactide-co- glycolide) 85:15 poly(lactide-co-glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co-glycolide), where the ratios are mole ratios. [0286] Polylactic-co-glycolic acid (PLGA) and Eudragit S100 (ES100) are two commonly used pharmaceutical excipients that are generally recognized as safe (GRAS) by the United States FDA and the European Medicines Agency. Both excipients are commercially available with different grades of copolymer blends and have been extensively studied (151, 152). Both PLGA and ES100 are biodegradable materials with abundant preclinical and clinical research that focuses on the drug delivery systems (153). In certain embodiments, ES100 and PLGA polymers may be use for formulating enteric and time delayed coatings. [0287] In many fields of science, polymer degradation has been defined as the amount of time the material takes to degrade over its useful life time and nondegradation has been defined as breakdown that occurs after its useful life. In another way, the ratio between the time the polymer takes to degrade and the duration of the application of the polymer is used to determine whether or not the material is degradable (154). PLGA and ES100 used for the coating of enteric and time release capsules undergoes chemical degradation; PLGA also undergoes erosion. [0288] All biodegradable polymers contain hydrolysable bonds where the passive hydrolysis or enzyme-catalyzed hydrolysis breaks down the chemical bond between the copolymers. The hydrolysis degradation occurs via a random chain scission process to form oligomers and finally monomers (155). The enzyme-catalyzed hydrolysis also referred to as biodegradation is when a biological system is partially involved in the chemical bond cleavage. [0289] Most synthetic biodegradable polymers, PLGA and ES100, mainly undergo passive hydrolysis (156). The degradation rate is determined by its chemical bond, composition, the pH of the environment, the water content, and the swelling rate. Degradation begins with water uptake into the bulk of the polymer matrix, which leads to swelling. Next, hydrolysis occurs resulting in oligomers and monomers degrading from the polymer. Progressively, the degradation changes the microstructure of the bulk, forming pores, where the oligomers and monomers are released. Heterogeneous degradation refers to the surface degradation of the polymers whereas homogenous degradation describes a uniform break down of the bulk material throughout its matrix. [0290] Erosion is a more complicated process of polymer degradation in which additional parameters such as the swelling rate and material porosity are involved. Poly (D,L-lactic-co- glycolic acid) three dimensional objects have been shown to exhibit an inverse flux erosion throughout the inner bulk material (154); once degradation begins increases in the degradation rate due to the lactide monomer content serves as an autocatalytic hydrolysis resulting in erosion (155, 157, 158). The PLGA erosion rate can be optimized by adjusting the amount of porosity and the sizes of the micropores within PLGA matrix (159). [0291] PLGA is an excellent biodegradable and biocompatible polymer (153, 160). Varying the ratios of lactic to glycolic acid controls the rate of water cleavage. Lactic acid contributes to the rigidity of the polymer backbone, thus increasing the duration that the polymer remains within a biological system (161). The primary mechanism for degradation of PLGA is the hydrolysis of the ester bond between the lactic acid and glycolic acid. In certain embodiments, the PLGA 85:15 grade was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule. PLGA 85:15 also allows for successive coating applications. [0292] Eudragit S100 (ES100) is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio. ES100 pH dependent nature were finely tune by the number of negative charge on the MAA’s COOH functional group at basic condition (162). The hydrophobicity MMA units were realized by synthesizing co-polymer libraries with a systematic variation. ES100 is soluble in alkaline digestive fluids by salt formation (163, 164). A clinical study shown that a 5% W/W gain of the coated Eudragit S100 tablets (84 ± 4 micron) (64) release its content in the colon in 67% of volunteered subject. [0293] A disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility (Figure 38). However, the addition of plasticizer changes the dissolution profile and takes longer for the film to dry (165). Therefore, there is a need to develop alternative coating techniques, such as those provided herein, to limit drying time, maximize polymer mass loading to 10%, and limit the use of plasticizer while achieving uniformly smooth, defect free films. [0294] It is understood that any combination of the aforementioned biodegradable polymers can be used, including, but not limited to, copolymers, mixtures, or blends thereof. [0295] According to some embodiments, the polymeric coating may be applied to the capsule to any suitable thickness. According to some embodiments, the polymeric coating may be applied to the capsule to a predefined thickness of about 1 nm to about 350 nm. [0296] According to some embodiments, a delayed-release coating may be applied to the capsule to a thickness of about 115 ± 35 nm. According to some embodiments, the delayed- release coating delays the release of a therapeutic agent by at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, or at least about 24 hours after administration of the pharmaceutical composition to a subject. [0297] According to some embodiments, an enteric coating may be applied to the capsule to a thickness of about 50 ± 15 nm. According to some embodiments, the enteric coating releases active agent at a pH above 5.5 (e.g., a pH between about 6 and about 8). [0298] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be applied to the capsule to a thickness of about 1 nm, about 2 nm, about 3 nm, about 4 nm, about 5 nm, about 6 nm, about 7 nm, about 8 nm, about 9 nm, about 10 nm, about 11 nm, about 12 nm, about 13 nm, about 14 nm, about 15 nm, about 16 nm, about 17 nm, about 18 nm, about 19 nm, about 20 nm, about 21 nm, about 22 nm, about 23 nm, about 24 nm, about 25 nm, about 26 nm, about 27 nm, about 28 nm, about 29 nm, about 30 nm, about 31 nm, about 32 nm, about 33 nm, about 34 nm, about 35 nm, about 36 nm, about 37 nm, about 38 nm, about 39 nm, about 40 nm, about 41 nm, about 42 nm, about 43 nm, about 44 nm, about 45 nm, about 46 nm, about 47 nm, about 48 nm, about 49 nm, about 50 nm, about 51 nm, about 52 nm, about 53 nm, about 54 nm, about 55 nm, about 56 nm, about 57 nm, about 58 nm, about 59 nm, about 60 nm, about 61 nm, about 62 nm, about 63 nm, about 64 nm, about 65 nm, about 66 nm, about 67 nm, about 68 nm, about 69 nm, about 70 nm, about 71 nm, about 72 nm, about 73 nm, about 74 nm, about 75 nm, about 76 nm, about 77 nm, about 78 nm, about 79 nm, about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86 nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm, about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, about 100 nm, about 105 nm, about 110 nm, about 115 nm, about 120 nm, about 125 nm, about 130 nm, about 135 nm, about 140 nm, about 145 nm, about 150 nm, about 155 nm, about 160 nm, about 165 nm, about 170 nm, about 175 nm, about 180 nm, about 185 nm, about 190 nm, about 195 nm, about 200 nm, about 205 nm, about 210 nm, about 215 nm, about 220 nm, about 225 nm, about 230 nm, about 235 nm, about 240 nm, about 245 nm, about 250 nm, about 255 nm, about 260 nm, about 265 nm, about 270 nm, about 275 nm, about 280 nm, about 285 nm, about 290 nm, about 295 nm, about 300 nm, about 305 nm, about 310 nm, about 315 nm, about 320 nm, about 325 nm, about 330 nm, about 335 nm, about 340 nm, about 345 nm, or about 350 nm. [0299] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to achieve a polymer a mass loading of about 7% to about 15%. According to some embodiments, the polymeric coating (e.g., delayed- release coating and/or the enteric coating) may be formulated to achieve a polymer mass loading of about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 10.5%, about 11%, about 11.5%, about 12%, about 12.5%, about 13%, about 13.5%, about 14%, about 14.5%, or about 15%. [0300] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores to control (e.g., accelerate and shorten) the degradation rate of a polymer from days into hours and precisely control its drug to deliver to the colon and/or large intestine. According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 1 nm to about 300 µm, inclusive. For example, the plurality of pores may be formed by doping and leaching the polymeric coating with a dopan porogen, such as sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), sodium acetate (NaCH3CO3), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat. According to some embodiments, the percentage of the dry mass ratio of dopant porogen to polymer can range from about 0.1% to about 1.0% (e.g., about 0.03% to about 0.07%). According to some embodiments, the percentage of the dry mass ratio of dopant porogen to polymer can be about 0.01%, about 0.05%, about 0.1%, about 0.15%, about 0.2%, about 0.25%, about 0.3%, about 0.35%, about 0.4%, about 0.45%, about 0.5%, about 0.55%, about 0.6%, about 0.65%, about 0.7%, about 0.75%, about 0.8%, about 0.85%, about 0.9%, about 0.95%, or about 1%. [0301] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 100 µm to about 300 µm, inclusive (e.g., about 100 µm, about 105 µm, about 110 µm, about 115 µm, about 120 µm, about 125 µm, about 130 µm, about 135 µm, about 140 µm, about 145 µm, about 150 µm, about 155 µm, about 160 µm, about 165 µm, about 170 µm, about 175 µm, about 180 µm, about 185 µm, about 190 µm, about 195 µm, about 200 µm, about 205 µm, about 210 µm, about 215 µm, about 220 µm, about 225 µm, about 230 µm, about 235 µm, about 240 µm, about 245 µm, about 250 µm, about 255 µm, about 260 µm, about 265 µm, about 270 µm, about 275 µm, about 280 µm, about 285 µm, about 290 µm, about 295 µm, or about 300 µm). [0302] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores having a size of about 1 nm to about 50 nm, inclusive (e.g., about 1 nm, about 2 nm, about 3 nm, about 4 nm, about 5 nm, about 6 nm, about 7 nm, about 8 nm, about 9 nm, about 10 nm, about 11 nm, about 12 nm, about 13 nm, about 14 nm, about 15 nm, about 16 nm, about 17 nm, about 18 nm, about 19 nm, about 20 nm, about 21 nm, about 22 nm, about 23 nm, about 24 nm, about 25 nm, about 26 nm, about 27 nm, about 28 nm, about 29 nm, about 30 nm, about 31 nm, about 32 nm, about 33 nm, about 34 nm, about 35 nm, about 36 nm, about 37 nm, about 38 nm, about 39 nm, about 40 nm, about 41 nm, about 42 nm, about 43 nm, about 44 nm, about 45 nm, about 46 nm, about 47 nm, about 48 nm, about 49 nm, or about 50 nm). [0303] According to some embodiments, the polymeric coating (e.g., delayed-release coating and/or the enteric coating) may be formulated to comprise a plurality of pores at a pore density of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%. Therapeutic agent [0304] According to some embodiments, the described invention provides a therapeutic agent, which may be formulated into a particulate formulation or a pharmaceutical composition as described herein for colonic delivery. In certain embodiments, the therapeutic agent comprises a COX-2 inhibitor. In certain embodiments, the therapeutic agent comprises 6A1 (4- [3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C), or a derivative thereof. In some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises a COX-2 inhibitor. In some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2- hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C), or a derivative thereof. [0305] As systemically available COX-2 inhibitors are generally known to be toxic to the cardiovascular system, the described invention provides for the use of COX-2 inhibitors comprising a structural motif that makes them substrates for hepatic UDP- glucuronosyltransferases (UGTs) and sulfotransferases (SULTs). In certain embodiments, the structural motif may be selected from the group consisting of a phenolic, an amine, an aliphatic alcohol, a carboxylic acid, or a sulfhydryl. Without being bound by theory, incorporating such structural motifs into COX-2 inhibitor compounds may make these compounds unavailable to the systemic circulation, thereby eliminating toxicity (e.g., cardiovascular toxicity) associated with systemic exposure. [0306] In certain embodiments, the therapeutic agent may be a locally bioavailable COX- 2 inhibitor comprising a structural motif that will subject the compound to rapid first-pass metabolism before reaching the systemic circulation which may limit its exposure to non- therapeutically targeted organs affected by toxicity, e.g., cardiovascular toxicity. In certain embodiments, the COX-2 inhibitor may be tailored to have bioavailability only in a specific organ, e.g., the colon. In certain embodiments, the COX-2 inhibitor is not systemically bioavailable, and therefore will not cause cardiovascular toxicity. In certain embodiments, the COX-2 inhibitor may be used as a chemotherapeutic and/or a chemopreventive agents in certain targeted organs, e.g., the intestine and/or the colon. [0307] For example, according to some embodiments, the therapeutic agent comprises a locally bioavailable COX-2 inhibitor as described in WO/2016/172159, incorporated herein by reference. In certain embodiments, the therapeutic agent may comprise a compound as defined by any one of Formulas 1-10 as described in WO/2016/172159. [0308] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 1 or any pharmaceutically acceptable salts thereof.
Figure imgf000076_0001
[0309] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula I, wherein R1 may be selected from sulfamyl, substituted sulfamyl, halo, alkyl, alkoxy, hydroxyl, and haloalkyl; and [0310] wherein R2 may be selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoarylamido, Ν,Ν-dialkyla- mido, N-alkyl-N- arylamido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio, alkylsulfinyl, alkyl- sulfonyl, N- alkylsulfamyl, N-arylsulfamyl, arylsulfonyl, Ν,Ν-dialkylsulfamyl, N-alkyl-N- arylsulfamyl, and heterocyclic; and [0311] wherein R3 may be selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid. [0312] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula I, wherein R3 may be selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoary larnido, Ν,Ν-dialkyla- mido, N-alkyl-N- arylarnido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio, alkylsulfinyl, alkyl- sulfonyl, N- alkylsulfamyl, N-arylsulfamyl, arylsulfonyl, Ν,Ν-dialkylsulfamyl, N-alkyl-N- arylsul- famyl, heterocyclic, heterocycloalkyl, and aralkyl; and [0313] wherein R2 may be selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid. [0314] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula I, wherein R4 may be selected from aryl, cycloalkyl, cycloalkenyl and heterocyclic; optionally, wherein R4 may be substituted at a substitutable position with one or more radicals selected from halo, alkylthio, alkylsulfinyl, alkyl, alkylsulfonyl, cyano, carboxyl, alkoxycarbonyl, amido, N- monoalkylamido, N-monoarylamido, N,N- dialkylamido, N-alkyl- N-arylamido, haloalkyl, hydroxyl, alkoxy hydroxyalkyl haloalkoxy, sulfamyl, N- alkylsulfamyl, amino, methylthio, N- alkylarnino, Ν,Ν-dialkylamino, heterocyclic, nitro and acylarnino; or [0315] wherein R3 and R4 together form nosulfonyl]phenyl)-5-phenylpyrazole-3- carboxylic acid has been prepared from the above described 4-[3-methyl-5- 65 phenyl-lH- pyrazol-1- yl]benzenesulfonamide compound
Figure imgf000077_0001
[0316] wherein R5 may be one or more radicals selected from halo, alkylthio, alkylsulfinyl, alkylsulfonyl, cyano, carboxyl, alkoxycarbonyl, amido, N-monoalkylarnido, N- monoarylamido, alkyl, N,N- dialkylamido, N-alkyl- N-arylarnido, haloalkyl, hydrido, hydroxyl, alkoxy, hydroxyalkyl, haloalkoxy, sulfamyl, N-alkylsulfamyl, amino, alkylamino, heterocyclic, nitro and acylarnino; [0317] wherein R2 and R3 are not identical radicals selected from hydrido, carboxyl and ethoxycarbonyl; [0318] further provided that R2 cannot be carboxyl when R3 is hydrido and when R4 is phenyl; and further provided that R4 is sulfamyl or N-alkylsulfamyl when R1 is halo; or a pharmaceutically-acceptable salt thereof when R2 or R3 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid. [0319] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula I, wherein Rl is sulfamyl or substituted sulfamyl, wherein R2 is selected from phenyl with one or more hydroxyl, alkylphenyl with one or more hydroxyl, wherein R3 is from hydrido, halo, wherein R4 is selected from aryl with substitute of methyl, methoxyl, halo, hydrido, or methylthio and pharmaceutically acceptable salts thereof. [0320] In certain embodiments, a compound as defined by Formula I comprises any one of the compounds listed below, or a derivative thereof.
Figure imgf000078_0001
Figure imgf000079_0001
[0321] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 2 or any pharmaceutically acceptable salts thereof.
Figure imgf000080_0001
[0322] wherein R1 is selected from hydroxyl, amino, alkyl, carboxyalkyl, alkoxycarbonyl, aminocarbonyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy,
aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, aralkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycloalkyl, aryl, aralkyl, halo, alkylamino, aralkylamino, N-alkyl-N-aralkylamino, heteroaralkylamino, N-alkyl-N-heteroaralkylamino, N-alkyl-N- cycloalkylalkylamino, arylcarbonylthio, alkylaminocarbonylthioalkyl, arylcarbonyloxyalkyl, alkoxycarbon yloxyalkyl, alkylaminocarbonyloxyalkyl, alkoxycarbonylthioalkyl, and alkylaminocarbonylthioalkyl [0323] when R2 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid; [0324] wherein R2 is selected from cycloalkyl, cycloalkenyl, aryl and heterocyclo; [0325] wherein R3 is optionally substituted at a substitutable position with one or more radicals independently selected from alkyl, cyano, carboxyl, alkoxycarbonyl, haloalkyl, hydroxyl, hydroxyalkyl, haloalkoxy, amino, aminoalkyl, alkylamino, arylamino, nitro, alkoxyalkyl, alkylsulfinyl, aminosulfonyl, halo, alkoxy and alkylthio when Ri is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid; [0326] wherein R3 is one or more radicals independently selected from alkyl, cyano, carboxyl, alkoxycarbonyl, haloalkyl, hydroxyl, hydroxyalkyl, haloalkoxy, amino, aminoalkyl, alkylamino, arylamino, nitro, alkoxyalkyl, alkylsulfinyl, halo, hydroxysulfonyl, alkylsulfonyl, aminosulfonyl, haloalkylsulfonyl, alkoxy and alkylthio; or a pharmaceutically-acceptable salt thereof. [0327] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 3, wherein R4 is sulfamyl or substituted sulfamyl, wherein Rl is from hydrido or halo, wherein R2 is selected from aryl with substitute of methyl, methoxyl, halo, hydrido, or methylthio and pharmaceutically acceptable salts thereof as follows:
Figure imgf000082_0002
[0328] wherein R1 is selected from alkyl, carboxyalkyl, alkoxycarbonyl, aminocarbonyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, aralkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyl, amino, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycloalkyl, aralkyl, halo, alkylamino, aralkylamino, N-alkyl-N-aralkylamino, heteroaralkylamino N-alkyl-N-hetero aralkylamino, N-alkyl-N-cycloalkylalkylamino, arylcarbonyloxy alkyl, arylcarbonylthio, alkoxyc arbonyloxyalkyl , alkylaminocarbonyloxyalkyl, alkoxycarbonylthioalkyl, and alkylaminocarbonylthioalkyl, [0329] when R2 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid; [0330] wherein R4 is selected from alkyl, hydroxyl, and amino; peptides, or a pharmaceutically- acceptable salt thereof. [0331] It can be understood that within Formula 3 there is a subclass of compounds represented by Formula 4.
Figure imgf000082_0001
[0332] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 4 or any pharmaceutically acceptable salts thereof, wherein R1 is selected from hydroxyl, alkyl, carboxyalkyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycloalkyl, aralkyl, halo, alkylamino, aralkylamino, N- alkyl-N- aralkylamino, heteroaralkylamino, N-alkyl-N- heteroaralkylamino, alkyl-N- cycloalkylalkylamino, arylcarbonyloxy alkyl, arylcarbonylthio, alkoxycarbonyloxyalkyl, alkylaminocarbonyloxyalkyl, alkoxycarbonylthioalkyl, and alkylaminocarbonylthioalkyl [0333] when R2 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid; or [0334] wherein R2 is selected from hydroxyl, alkyl, carboxyalkyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycloalkyl, aralkyl, halo, alkylamino, aralkylamino, N-alkyl-N- aralkylamino, heteroaralkylamino, N-alkyl-N- heteroaralkylamino, alkyl-N-cycloalkylalkylamino, arylcarbonyloxy alkyl, arylcarbonylthio, alkoxycarbonyloxyalkyl, alkylaminocarbonyloxyalkyl, alkoxycarbonylthioalkyl, and alkylaminocarbonylthioalkyl [0335] when R1 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid. [0336] It can be understood that within Formula 4, there is a subclass of compounds represented by Formula 5, which comprises compounds of Formula 4 wherein Rl is selected from hydroxyl, lower alkyl, carboxyl, lower carboxyalkyl, lower aminocarbonylalkyl, lower alkoxycarbonylalkyl, lower aralkyl, lower alkoxyalkyl, lower aralkoxyalkyl, lower alkylthioalkyl, lower aralkylthioalkyl, lower alkylaminoalkyl, lower aryloxyalkyl, lower arylthioalkyl, lower haloalkyl, lower hydroxylalkyl, lower cycloalkyl, lower cycloalkylalkyl, and aralkyl [0337] when R2 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid; or [0338] wherein R2 is optionally substituted at a substitutable position with one or more radicals independently selected from lower alkylsulfinyl, aminosulfonyl, lower alkyl, cyano, carboxyl, lower alkoxycarbonyl. lower haloalkyl, hydroxyl, lower hydroxyalkyl, lower haloalkoxy, amino, lower alkylamino, lower arylamino, lower aminoalkyl, nitro, halo, lower alkoxy and lower alkylthio; [0339] when R1 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically-acceptable salt thereof. [0340] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 5 or any pharmaceutically acceptable salts thereof.
Figure imgf000084_0002
[0341] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 6 or any pharmaceutically acceptable salts thereof.
Figure imgf000084_0001
[0342] In certain embodiments, X-Y-Z is selected from the group consisting of (a) -CH2CH2CH2-, (b) -C(O)CH2CH2-, (c) -CH2CH2C(O)-, (d) -CR (R)-O-C(O)-, (e) -C(O)-O-CR5(R5’), (f) -CH -NR3-CH-, (g) –CR5(R5’)-NR3-C(O)-, (h) -CR4=CR4'-S-, (i) -S-CR4=CR4, (j) -S-N=CH-, (k) - CH=N-S-, (l) -N=CR4-O, (m) -O-CR4=N-, (n) -N=CR4-NH-, (o) -N=CR4-S-, (p) -S-CR4-N-, (q) -C(O)-NR -CR (R ')-, (r) -R3N-CH=CH- provided R1 is not –S, and (s) -R3N-CH=CH- provided R1 is not -S(O) Me, [0343] with Rx may be replaced in any place in the above moiety, optionally, wherein Rx is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically-acceptable salt thereof. [0344] In certain embodiments, when side b is a double bond, and sides a and c are single bonds; and X-Y-Z-is selected from the group consisting of: (a) =CH-O-CH=, (b) =CH-NR3-CH-, (c) N-S-CH=, (d) =CH-S-N=, (e) N-O-CH=, (f) =CH-O-N=, (g) N-S-N= and (h) -N-O-N=, [0345] with any Rx replaced in any place in above moiety, wherein Rx is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically-acceptable salt thereof. [0346] In certain embodiments, when sides a and c are double bonds and side b is a single bond R1 is selected from the group consisting of (a) S(O)2CH3 (b) S(O)2NH2 (c) S(O)2NHC(O)CF3 (d) S(O)(NH)CH3 (e) S(O)(NH)NH2 (f) S(O)(NH)NHC(O)CF3 (g) P(O)(CH3)OH, and (h) P(O)(CH3)NH, R2 is selected from the group consisting of (a) C1-6alkyl, (b) C3, C4, C5, C6, and C1 cycloalkyl, (c) mono-, di- or tri-substituted phenyl or naphthyl wherein the substituent is selected from the group consisting of, (1) hydrogen, (2) halo, (3) C1-6 alkoxy, (4) C1-6 alkylthio, (5) CN, (6) CF3, (7) C1-6 alkyl, (8) N3, (9) -CO2H, (10) -CO2-C1-4alkyl, (11) C(R5)(R6)-OH, (12) -C(R5)(R6)-O-C1-4alkyl, (13) -C1-6alkyl-CO2-Rs, (d) mono-, di- or tri-substituted heteroaryl wherein the heteroaryl is a monocyclic aromatic ting of 5 atoms, said ting having one hetero atom which is S, O, or N and optionally 1, 2, or 3 additionally N atoms; or the heteroaryl is a monocyclic ting of 6 atoms, said ring having one hetero atom which is N, and optionally 1, 2, 3, or 4 additional N atoms; said substituents are selected from the group of (1) hydrogen, (2) halo, including fluoro, chloro, bromo and iodo, (3) C1-6alkyl, (4) Cl-6alkoxy (5) Cl-6alkylthio (6) CN (7) CF3 (8) N3 (9) -C(R5)(R6)-OH, and (10) -C(R5)(R6)-O-C1-4alkyl, R3 is selected from the group consisting of (a) Hydrogen (b) CF3 (c) CN (d) C1-6alkyl, (e) hydroxy C1-6alkyl (f) -C(O)- C1-6 alkyl (a) optionally substituted (1) Cl-5alkyl-Q (2) -C1-3alkyl-O-C1-3alkyl-Q (3) -C1-3alkyl-S-C1-3alkyl-Q (4) -C1-5alkyl-O-Q (5) -C1-5alkyl-S-Q [0347] R4 and R4' are each independently selected from the group consisting of wherein the substituent resides on the alkyl and the substituent is C1-3alky (a) Hydrogen, (b) CF3, (c) CN, (d) C1-6alkyl, (e) –Q, (f) -O-Q, (g) -S-Q, (h) Optionally substituted (l) -C1-5alkyl-Q, (2) -O-C1-5alkyl-Q, (3) -S-C1-5alkyl-Q, (5) -C1-3alkyl-S-C1-3alkyl-Q, (6) -C1-5alkyl-O-Q, (7) -C1-5 alkyl-S-Q, [0348] wherein the substituent resides on the alkyl and the substituent is C1-3alkyl, and R5, R5', R6, R7 and R8 are each independently selected from the group consisting of (a) Hydrogen, (b) C1-6 alkyl, or R5 and R6 and R7 and R8 together with the carbon to which they are attached from a saturate monocyclic carbon ring of 3, 4, 5, 6, or 7 atoms; Q is COOH, COO-C1-4alkyl, tetrazoly-5-yl, C(R7)(R8)(OH) or C(R7)(R8)(O-C1-4alkyl) provided that when X-Y-Z is S-CR4=CR4', then R4 and R4' are other than CF3. [0349] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 7 or any pharmaceutically acceptable salts thereof, which comprises those compounds of Formula 6, [0350] wherein Rl is sulfamyl or substituted sulfamyl, [0351] wherein R2 is selected from hydroxyl, alkyl, carboxyalkyl, aminocarbonylalkyl, alkoxycarbonylalkyl, carboxyl, alkoxy, haloalkoxy, aralkoxy, heteroaralkoxy, cycloalkylalkoxy, alkylthio, heteroaralkylthio, cycloalkylalkylthio, alkoxyalkyl, aralkoxyalkyl, alkylthioalkyl, aralkylthioalkyl, alkylaminoalkyl, aryloxyalkyl, arylthioalkyl, hydroxyalkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocyclo, heterocycloalkyl, aralkyl, halo, alkylamino, aralkylamino, N-alkyl-N- aralkylamino, heteroaralkylamino, N-alkyl-N- heteroaralkylamino, alkyl-N-cycloalkylalkylamino, arylcarbonyloxy alkyl, arylcarbonylthio, alkoxycarbonyloxyalkyl, alkylaminocarbonyloxyalkyl, alkoxycarbonylthioalkyl, and alkylaminocarbonylthioalkyl, [0352] wherein R3 is from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, or a pharmaceutically- acceptable salt thereof.
Figure imgf000089_0001
[0353] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 8 or any pharmaceutically acceptable salts thereof,
Figure imgf000089_0002
[0354] wherein R1 is selected from methyl, ethyl, or any other alkyl, [0355] wherein R2 is selected from halo, hydrogen, methoxyl, methyl, ethyl, trifluoromethyl when R3 is hydroxyl (exclude position-4), [0356] wherein X is selected from N, O, C. [0357] In certain embodiments, the therapeutic agent may comprise a compound as defined by Formula 9 or any pharmaceutically acceptable salts thereof,
Figure imgf000090_0001
[0358] wherein R1 is selected from sulfamyl, substituted sulfamyl, halo, alkyl, alkoxy, hydroxyl, and haloalkyl, [0359] wherein R2 is selected from hydrido, halo, haloalkyl, cyano, nitro, formyl, carboxyl, alkoxycarbonyl, carboxyalkyl, alkoxycarbonylalkyl, amidino, cyanoamidino, arnido, alkoxy, amidoalkyl, N-monoalkylamido, N-monoarylamido, Ν,Ν-dialkyla- mido, N-alkyl-N- arylamido, alkylcarbonyl, alkylcarbo- nylalkyl, hydroxyalkyl, alkylthio, alkylsulfinyl, alkyl- sulfonyl, N- alkylsulfamyl, N-arylsulfamyl, arylsulfonyl, Ν,Ν-dialkylsulfamyl, N-alkyl-N- arylsulfamyl, and heterocyclic, [0360] wherein R3 is selected from any aromatic moiety, such as phenyl, pyridyl, thienyl, oxazolyl, isoxazolyl, benzoxazolyl, thiazolyl, quinolinol, pyridinol, with one or more hydroxyl(s) or with one or more amine(s) that can be conjugated with glucuronic acid or sulphonic acid, and [0361] wherein X, Y, Z is selected from N and C. According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery comprises 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl- pyrazol-1-yl-] benzenesulfonamide) (Figure 1C), or a derivative thereof. [0362] According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in any suitable amount. [0363] According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg to about 1000 mg. [0364] According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg/kg to about 100 mg/kg. [0365] According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, about 25 mg, about 26 mg, about 27 mg, about 28 mg, about 29 mg, about 30 mg, about 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, about 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 46 mg, about 47 mg, about 48 mg, about 49 mg, about 50 mg, about 51 mg, about 52 mg, about 53 mg, about 54 mg, about 55 mg, about 56 mg, about 57 mg, about 58 mg, about 59 mg, about 60 mg, about 61 mg, about 62 mg, about 63 mg, about 64 mg, about 65 mg, about 66 mg, about 67 mg, about 68 mg, about 69 mg, about 70 mg, about 71 mg, about 72 mg, about 73 mg, about 74 mg, about 75 mg, about 76 mg, about 77 mg, about 78 mg, about 79 mg, about 80 mg, about 81 mg, about 82 mg, about 83 mg, about 84 mg, about 85 mg, about 86 mg, about 87 mg, about 88 mg, about 89 mg, about 90 mg, about 91 mg, about 92 mg, about 93 mg, about 94 mg, about 95 mg, about 96 mg, about 97 mg, about 98 mg, about 99 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg. [0366] According to some embodiments, the therapeutic agent formulated into the particulate formulation or the pharmaceutical composition for colonic delivery may be included in an amount of at least about 0.001 wt%, at least 0.005 wt%, at least 0.01 wt%, at least 0.05 wt%, at least 0.10 wt%, at least 0.50 wt%, at least 1 wt%, at least 2 wt%, at least 3 wt%, at least 4 wt%, at least 5 wt%, at least 6 wt%, at least 7 wt%, at least 8 wt%, at least 9 wt%, at least 10 wt%, at least 15 wt%, at least 20 wt%, at least 25 wt%, at least 30 wt%, at least 35 wt%, at least 40 wt%, at least 45 wt%, at least 50 wt%, at least 55 wt%, at least 60 wt%, at least 65 wt%, at least 70 wt%, at least 75 wt%, at least 80 wt%, at least 85 wt%, at least 90 wt%, at least 95 wt% of the total weight of the particulate formulation or the pharmaceutical composition. Methods Coating Technique [0367] The present disclosure investigated three different coating techniques, as described in the Examples, including dip coating, fluidic coating, and vacuum spin coating. The parameters used to judge each technique were: smoothness of the film, reproducibility, and ease of clean up. Of the three, vacuum spin coating yielded the most optimal results. [0368] According to another aspect, the described invention provides vacuum spin coating techniques and formulations to delay the release of therapeutic agents targeting the colon. According to some embodiments, the controlled coating technique results in an appropriate coat thickness, which combined with an optimal formulation of biodegradable polymers that will protect and release the therapeutic agent in a programmable manner (e.g., delayed beyond 6 hours and sustained at a pH above 6.0). Compared to the small intestine, the colon is much shorter with a less absorptive surface; however, the colon has the longest transit time (Half E et al. Orphanet J Rare Dis. 2009;4:22). Accordingly, the invention provides vacuum spin coating techniques and formulations to achieve a coating that can be pH-sensitive to a basic environment and have an erosion mechanism that is uniquely tailored to the water content and microbiome within the colon. [0369] Vacuum spin coating is one of the most common methods for applying thin films to substrates (166). A wide variety of technology sectors and industries commonly use vacuum spin coating. Vacuum spin coating's advantage is its ability to rapidly and easily produce consistently uniform films, ranging from a few microns down to a nanometer of thickness. Organic electronics and nanotechnology heavily rely on spin coating, and they had advanced many of the techniques that are used in other semiconductor industries to a more advanced level. [0370] The vacuum spin coater apparatus works based on two physical properties. The first was an application of a vacuum that creates a shear force to facilitate the spread of the non-Newtonian polymers throughout the 3-dimensional capsules. Secondly, the capsules were spun. As the capsule accelerates in rotation, it expulses all the undissolved polymer. Once the spin coater's RPM reaches its desired speed and the polymer fluid was sufficiently thinned out, viscous shear drag exactly balances the rotational accelerations, drying begins. Next, the capsule was spun at a constant rate, 500 RPM, and solvent evaporation dominates the coating thinning behavior. At this point, the rate of evaporation depends on two factors (a) the difference in the partial pressure of solvent evaporation between the free surface of the liquid layer and the gas layer flowing nearby (surface tension) and (b) the capillary forces developing in the channels between the latex particles. During the evaporation stage, the dissolved polymers solidified at the liquid-air interface in a highly viscous, lowly diffusive layer forming an unbroken film on the skin of the capsule (151). [0371] Currently, no commercially available coating machine exists for gelatin capsules smaller than size five. Compared with conventional coating, the present invention provides an adaptor that could sidestep the standard low mass loading of 3% and instead use a 10% mass loading. The adaptor may be sized to fit any conventional magnetic spinner and vacuum. The adaptor may be used to coat both the aqueous and organic solvent-based systems because it does not require a spraying nozzle to apply the solvent to the substrate. Furthermore, the spin vacuum adaptor does not need high heat as a curing process since the spinning creates an air vortex within each pod that facilitates solvent evaporation. The vacuum spin coater was also designed to produce individualized small-scale batches consisting of 8 or 16 capsules. Currently, the coating success rate is around 80%, meaning that for every quantity of eight capsules, one will fail the visual, weight, or in vitro dissolution test. Most noticeably, this may significantly reduce the amount of processing time and the total waste associated with unused material. The spin vacuum design resolves the shortfalls of the conventional coating techniques mentioned above, which does not have the capability to run pilot testing of a drug or produce individually tailored medicines where the pharmacokinetics of the drugs can be controlled and modified. It was discovered that the thickness of the coating layers was controllable by adjusting two parameters of the spin vacuum coater. First, the total suction applied by the vacuum facilitates the surface creeping of 10% of the material. The force exerted onto the capsule by the vacuum also spreads the formulation evenly to the gap and covers the junction between the cap and body of the capsule. Secondly, the spinning rate of the machine creates a centrifugal shearing force that facilitates the expulsion of chunky or undissolved polymer and also thins out the coating material. Together, the total suction of the vacuum and spinning rate precisely control the optimal thickness of the PLGA and ES100. [0372] According to some embodiments, the described invention provides a method of spin coating a capsule, comprising: (i) providing a vacuum spinning plate comprising individual pods; (ii) providing sealed capsules wetted with a mild basic water (e.g., pH 9 NaOH 0.1 mM) loaded into the individual pods within the vacuum spinning plates; (iii) providing an amount of a first polymer dissolved in a solvent; (iv) applying a vacuum; (v) spinning the plate a first time for about 30 seconds at a spinning speed of 100 RPM; (vi) spinning the plate a second time for about 15 minutes at spinning speed of 500 RPM; (vii) providing an amount of a second polymer dissolved in a solvent; (viii) applying a vacuum; (ix) spinning the plate a third time for about 30 seconds at spinning speed of 100 RPM; (x) spinning the plate a fourth time for about 15 minutes at a spinning speed of 500 RPM; (xi) removing the capsule from the spinning plate; (xii) drying the capsule for about 24 hours in a desiccator (Figure 14). [0373] According to some embodiments, the first polymer is PLGA8515. According to some embodiments, the solvent for PLGA8515 is methylene chloride. In certain embodiments, the concentration is 10% w/v solution PLGA8515. [0374] According to some embodiments, where pore formation is desired, a dopant porogen can be added to the premade PLGA polymer that is dissolved in methylene chloride. According to some embodiments, the dopan porogen can be sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), sodium acetate (NaCH3CO3), sodium chloride (NaCl), saccharin, lipoprotein, or small molecule fat. [0375] According to some embodiments, the concentration of dopant can be 1% v/v to 20% v/v of the final solution. According to some embodiments, the concentration of dopant can be about 1% v/v, about 2% v/v, about 3% v/v, about 4% v/v, about 5% v/v, about 6% v/v, about 7% v/v, about 8% v/v, about 9% v/v, about 10% v/v, about 11% v/v, about 12% v/v, about 13% v/v, about 14% v/v, about 15% v/v, about 16% v/v, about 17% v/v, about 18% v/v, about 19% v/v, or about 20% v/v. [0376] According to some embodiments, the percentage of the dry mass ratio of dopant porogen to polymer can range from about 0.1% to about 1.0% (e.g., about 0.03% to about 0.07%). According to some embodiments, the percentage of the dry mass ratio of dopant porogen to polymer can be about 0.01%, about 0.05%, about 0.1%, about 0.15%, about 0.2%, about 0.25%, about 0.3%, about 0.35%, about 0.4%, about 0.45%, about 0.5%, about 0.55%, about 0.6%, about 0.65%, about 0.7%, about 0.75%, about 0.8%, about 0.85%, about 0.9%, about 0.95%, or about 1%. [0377] For example, sodium citrate at 5% w/w as a doping agent creates a pore size of 180- 250µm, inclusive, in the PLGA network. While sodium chloride at room temperature to 400°C can consistently forms a pore size of 17-20 nm, inclusive, in diameter. An exemplary, working sodium chloride salt concentration as dopant is 2.5% v/v to 10% v/v of the final solution (e.g., 37.5 µL NaCl/methanol into 1462.5 µL of pre-dissolved PLGA8515/methylene chloride polymers to 75 µL NaCl/methanol into 1425.0 µL of pre-dissolved PLGA8515/methylene chloride). [0378] According to some embodiments, the second polymer is Eudragit S100. [0379] According to some embodiments, the solvent for Eudragit S100 is a mixture of acetone: isopropanol alcohol: DI water in the ratio of 86:12:2. In certain embodiments, the concentration is 10% w/v solution Eudragit S100. [0380] In certain embodiments, hard gelatin capsules were packed with raw drug or microparticles; the capsules were then coated with PLGA 8515 to a thickness of 115 ± 35 nm thickness and an outer pH-sensitive Eudragit S100 coat of 50 ± 15 nm thickness to achieve colonic delivery. The new vacuum spin coating technique achieved an 88% success rate (410 of 450 capsules made passed the quality assurance tests). In certain embodiments, the new coating technique was able to coat high viscosity polymers, PLGA8515 and ES100, with a high mass loading of 10% w/w formulation and sufficiently covered the entire three dimensional nine millimeters hard gelatin capsules (S9C). The novel spin coating technique is an innovative coating approach that had not been accomplished prior to this invention. The vacuum applied to the bottom of the capsules placed inside its respective slot within the coater plate creates a shear force that thinned out the polymers, which are behaving as non-Newtonian fluids. The thinning is due to the entanglement of polymer collapsing in the direction that vacuum is applied (bottom of the plate). The angle at which the capsule sits causes it to spin around as the centrifugal force resulting from the spinning of the coater plate. The centrifugal force is large enough to spin out the large undissolved polymers and creates a vortex of air that facilitates further evaporation. The combination of vacuum and spinning creates a micro-environment that has a conical vortex within each capsule pod. [0381] The method of coating capsules according to the invention is advantageous compared to conventional coating methods. The conventional method is for a large quantity of hard capsules filled with a drug to be charged into an apparatus which is then heated and rotated either under vacuum or atmospheric pressure. The invention uniquely allows for small batch production as is desired in research and development labs. Small batches are desirable for the production of personalized medications. When compared to conventional capsule coating equipment, the cost of the invention is significantly less. The quality of the capsule coating is superior in enteric and acid-resistant characteristics and the delayed release mechanism compared to conventional coatings rapidly disintegrated in intestinal fluid. Furthermore, the quantities of the coating agent, active ingredients, and fillers can be reduced; the products obtained are of higher quality, and the production cost for both researching laboratory and clinical setting are reduced. The invention allows for the delayed burst release of the capsule to specific region in the GI track is tunable through the controlling the coating thickness and the formulation of polymers.(191) Use in the Preparation of a Medicament/A method of treating [0382] Familial Adenoma Polyposis (FAP) is a debilitating condition that has a severe impact on the physical and psychological health, independence, and quality of life of an affected patient. Selective COX-2 inhibitors (COXIBs) are the most versatile group of drugs prescribed for inflammation, analgesic/antipyretic, and auto-immune diseases and are undeniably effective in the treatment and prevention of polyposis formation. However, COXIBs that circulate systemically inhibit COX-2 in on target off organs causing possible cardiovascular side effects (e.g., unstable angina, myocardial infarction, and cardiac thrombus) at the doses required for efficacy as a prophylactic (6, 16, 25-27), COXIBs cannot currently be used clinically to treat FAP. COXIBs are taken orally and absorbed through the stomach wall leading to systemic circulation. Additionally, COXIBs do not actively enter the EHR, thus large and frequent doses are required to reach a therapeutic level in the colon. [0383] Celecoxib is a selective COX-2 inhibitor class of NSAIDs that is undeniably effective against FAP. However, on-target off-organ cardiovascular toxicity prevents the use of Celecoxib as a treatment for FAP. A new COX-2 inhibitor, 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C), is less systemically bioavailable due to its ability to undergo enterohepatic recycling (EHR). EHR delivered a variable quantity of 6A1 to the target tissue, colon, subject to variation amongst individuals UGT enzyme expression and ß-glucuronidase expression within the bacteria microbiomes that inhabit the gut. [0384] According to another aspect, the present disclosure aims to lower the therapeutic dose and the total systemic exposure (AUC) while increasing concentration of a selective COX-2 inhibitor in the colon by capitalizing upon the complementary interplay between the EHR, coating techniques, and biodegradable polymer formulations. [0385] According to some embodiments, the described invention utilizes EHR as a means to target drug delivery to the colon. New chemical entities that undergo active glucuronidation via phase 2 metabolism are often eliminated early during the research and development phase due to an insufficient quantity of drug going to systemic circulation and ultimately inadequate therapeutic outcomes due to low exposure (39, 40). The present invention focuses on a novel class of COX-2 inhibitors that are actively glucuronidated by the UGT1A1 enzyme, which is a subclass of phase II metabolic enzymes found in abundance in the livers. Because the glucuronides of 6A1 undergo EHR, gut microbiomes can successfully reconvert glucuronides back into aglycone, which becomes bioavailable again after absorption into the colon cells, thus reducing the systemic drug circulation while increasing their exposure in the colon. However, the concern of excessive inter-subject discrepancies (patient's intrinsic and extrinsic factors) have demonstrated the necessity to directly dose the drug to the colon after initial administration, as the regulatory agency mandated that safe and efficacious drugs should have consistent Cmax, Tmax, and AUC across all populations (46). The variability might have been caused by the differences in the expression of UGTs in the liver and/or beta-glucuronidase activities expressed by colonic microflora, which may further be altered due to colonic diseases. [0386] Furthermore, when exclusively relying on beta-glucuronidase to deconjugate the metabolites, an insufficient amount of the parent drug at the colon tissue exhibits its inhibitory efficacy. On the other hand, rely heavily on the UGT1A1 enzymes to metabolize 6A1, may result in a low amount of bioavailability of metabolites effluxing to the colon or high systemic concentrations. Thus, the ability to deliver the right initial dose would be compromised. Accordingly, the described invention provides, According to some embodiments, formulation and coating of 6A1 packed inside hard gelatin capsules to ensure that the initial dose, at the correct concentration, will be delivered to the colon and sufficiently saturate the COX-2 enzymes, yielding a consistent Cmax and Tmax value across all populations. [0387] Specific modes of administration will depend on the indication. The selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response. The amount of active agent to be administered is that amount sufficient to provide the intended benefit of treatment. The dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular mammal or human treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician). [0388] According to the foregoing embodiments, the pharmaceutical composition may be administered once, for a limited period of time or as a maintenance therapy over an extended period of time, for example until the condition is ameliorated, cured or for the life of the subject. A limited period of time may be for 1 week, 2 weeks, 3 weeks, 4 weeks and up to one year, including any period of time between such values, including endpoints. According to some embodiments, the pharmaceutical composition may be administered for about 1 day, for about 3 days, for about 1 week, for about 10 days, for about 2 weeks, for about 18 days, for about 3 weeks, or for any range between any of these values, including endpoints. According to some embodiments, the pharmaceutical composition may be administered for more than one year, for about 2 years, for about 3 years, for about 4 years, or longer. [0389] According to the foregoing embodiments, the composition or pharmaceutical composition may be administered once daily, twice daily, three times daily, four times daily or more. [0390] According to some embodiments, the described invention provides use of a particulate formulation or pharmaceutical composition in the preparation of a medicament for reducing the severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP). [0391] According to some embodiments, the described invention provides use of a particulate formulation or pharmaceutical composition in the preparation of a medicament for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP). [0392] According to some embodiments, the described invention provides a method for reducing severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing a particulate formulation or pharmaceutical composition as described herein, and (b) administering an effective amount of the a particulate formulation or pharmaceutical composition orally to the subject. [0393] According to some embodiments, the described invention provide a method for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing a particulate formulation or pharmaceutical composition as described herein, and (b) administering an effective amount of the pharmaceutical composition orally to the subject. [0394] According to some embodiments, the pharmaceutical composition may be administered with an additional therapeutic agent and/or an additional treatment modality. Exemplary additional therapeutic agent include, without limitation, non-steroidal anti- inflammatory drugs (NSAIDs), polyamine transporter inhibitors, eIF-5A antagonists, chemotherapeutic agents, radiotherapy, and immunomodulatory agents. The dosing frequency of the pharmaceutical composition and the additional pharmaceutical agent may be adjusted over the course of the treatment based on the judgment of the administering physician. When administered separately, the pharmaceutical composition and the additional therapeutic agent can be administered at different dosing frequency or intervals. For example, the pharmaceutical composition can be administered weekly, while the additional therapeutic agent can be administered more or less frequently. According to some embodiments, a sustained continuous release formulation of the pharmaceutical composition and/or the additional therapeutic agent may be used. Various formulations and devices for achieving sustained release are known in the art. A combination of the administration configurations described herein can be used. According to some embodiments, the pharmaceutical composition can be administered daily and the additional therapeutic agent can be administered monthly. According to some embodiments, the pharmaceutical composition can be administered weekly and the additional therapeutic agent can be administered monthly. [0395] All referenced journal articles, patents, and other publications are incorporated by reference herein in their entirety. [0396] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges which may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention. [0397] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, exemplary methods and materials have been described. All publications mentioned herein are incorporated herein by reference to disclose and described the methods and/or materials in connection with which the publications are cited. EXAMPLES [0398] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric. [0399] Example 1. A sensitive and validated ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) method for quantifying a newly synthesized COX- 2 inhibitor (6A1) and its metabolites in blood, liver, and colonic mucosa of F344 rats Introduction [0400] Studies have shown that cyclooxygenase-2 (COX-2) enzyme is elevated in colorectal polyps. Marketed selective COX-2 inhibitors can successfully reduce polyp formation in the colon via inhibition of Prostaglandin E2 (PGE2) production by COX-2 (111, 112), but its prolonged administration as preventive care is not feasible due to on-target off- organ cardiac toxicity. A newly synthesized COX-2 inhibitor, 6A1 (4-[3-(2-hydroxy- phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide), is comparable in its performance to that of celecoxib (4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol- 1- yl]-benzenesulfona-mide), with a potential of lower on-target off-organ systemic toxicity due to low systemic exposure (113). [0401] The objective of this study was to validate a sensitive, and specific method that could simultaneously quantify 6A1, and its metabolites in three matrices, blood, liver, and colonic mucosa, using high performance liquid chromatography-mass spectrometry (UPLC- MS/MS). The development of this method is important for the preclinical pharmacokinetic and efficacy studies of 6A1. [0402] A sensitive and robust ultra-performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) method was developed and validated to simultaneously quantify 6A1 and its phase-II metabolites (glucuronide and sulfate) in rat blood, liver, and colonic mucosa matrices. A Water’s ACQUITY UPLC HSS T3 (2.1 x 100 mm I.D., 1.8 µm) column with gradient elution was determined to achieve the best peak separations. Both negative electrospray ionization (ESI) and positive ESI were used to quantify 6A1 and its conjugated metabolites. The method’s lower limit of quantification (LLOQ) of all three analytes were 2 ng/mL (or per gram) in blood, liver, and colonic mucosa matrices. Good linearity ranges from 2 – 1,400 ng/mL for blood and 2 – 1,400 ng/g for tissue homogenates. The method was validated as per the Food and Drug Administration guidance with the intra- and inter-day precision of 5 - 14%, accuracy of 96.1 - 111.5%. The matrix effect for blood, colonic mucosa, and liver homogenates was within the acceptable range (0.85 - 1.15). The method was used to study the compound’s pharmacokinetic behavior in F344 rats. Materials and Methods Chemicals and Materials [0403] Liquid chromatography-mass spectrometry (LC-MS) grade acetonitrile, methanol, ethanol, dichloromethane, ammonium acetate, polyethylene glycol 300 bio-ultra, and Baohuoside I (IS) of 99.99% purity were purchased from Sigma-Aldrich Corp. (St. Louise, MO, USA). Acrodisc syringe filter with a polytetrafluoroethylene (PTFE) membrane pore size of 0.2 um and diameter of 25 mm were purchased from Pall Corp. (Port Washington, NY). Oasis solid phase extraction (SPE) 5 cc cassette were purchased from Water Corp. (Milford, MA). The parent compound, 6A1, was synthesized in house and quantified as 98% pure using the UPLC and NMR. 6A1 sulfate and 6A1 glucuronide are biosynthesized using F344 rat microsomes and MDCK II -UGT1A1-MRP2- overexpressed cells. Biosynthesis and Purification of 6A1 Sulfate and Glucuronide [0404] The biosynthesis of 6A1 glucuronide was done using F344 rat liver microsomes as previously published (121, 127, 131, 136). In brief, 10 µM of 6A1 were incubated in rat liver microsomes (~ 0.05 mg/mL protein concentration) for 8 hours at 37°C in a shaking-water bath at 60 rpm resulting in the formation of 63% 6A1 glucuronide. The residual 6A1 from the incubated mixture’s supernatant was removed via liquid-liquid extraction utilizing dichloromethane. The samples were run through the Oasis HybridSPE 5 cc cassette, washed with DI water three time the volume, and the eluents then were washed out using 50% methanol in water. The collected eluents were then lyophilized, and stored in a −80°C freezer. The lyophilized residual was reconstituted with 100 µL of 50% methanol in water as stock solution of 6A1 glucuronide. [0405] MDCK II-UGT1A1-MRP2-overexpressed cells were used to generate the 6A1 sulfate following a previously established and published method (131). In brief, 10 µM of 6A1 in HBSS buffer (pH 7.4) was incubated with MDCK II-UGT1A1-MRP2 cells overnight at 37°C. The 6A1 sulfate was extracted from the buffer using the above described solid phase extraction (SPE) process for glucuronide, except 70% methanol in water was used to wash out eluent. Chromatographic Conditions [0406] Different combinations of mobile and stationary phases were employed to enhance the sensitivity and resolution of detection of 6A1, 6A1 sulfate, and 6A1 glucuronide. Acetonitrile, methanol, 0.1-5% formic acid in acetonitrile, 0.1 - 5% formic acid in methanol as organic phase, 1 - 2.5 mM ammonium acetate in water, and 0.05 - 1% formic acid in water as aqueous phase were tested as potential mobile phases. This experiment tested both the Restek Raptor biphenyl and the Aquity HSS T3 columns as stationary phases to resolve 6A1, 6A1 sulfate, and 6A1 glucuronide. [0407] The gradient elution was as follows: 0.1-.5 min 100% B; 0.5-1.5 min, 65% B; 1.5- 2.5 min, 30% B; 2.0-3.0 min 15% B; 3.0 - 4.0 min, 10% B; 4.0 – 6.0 min, 95% B. The column temperature of 45°C, sample temperature of 20℃, injection volume of 10 µL, and a flow rate of 0.20 mL/min achieved the best peak sharpness and symmetry. The peak resolution, defined as the amount of separation between two adjacent peaks, was calculated using the following equation: Peak Resolution = (time peak2 eluted – time peak1 eluted) / (0.5)(width peak2 + width peak1) Mass Spectrometry Conditions [0408] For MS/MS analysis, both positive and negative scan modes were employed to analyze 6A1, 6A1 glucuronide, and 6A1 sulfate. Representative MRM spectra of 6A1, 6A1 sulfate, and 6A1 glucuronide are shown in Figure 8. The analytes’ intensity in positive scan mode is more sensitive for 6A1 and 6A1 glucuronide compared to the negative scan mode. However, the opposite is true for the 6A1 sulfate metabolite. To improve specificity, the multiple reactions monitoring (MRM) scan type was used. MRM+ transitions from precursor ions to product ions were optimized as m/z 436.3 ^ 326.2 for 6A1, 612.10 ^ 435.8 for 6A1 glucuronide and 515.3 ^ 369.1 for IS, based on their protonated molecules and corresponding product ions (Figure 9A, 9C, 9D). The MRM- transition for 6A1 sulfate is optimized as m/z 513.3 ^ 434.1 and 513.1
Figure imgf000102_0001
369.1 for IS based on the deprotonated molecules and corresponding product ions (Figure 9B and 9E). Compound-dependent parameters in MRM mode for 6A1, 6A1 glucuronide, 6A1 sulfate, and Baohuoside I (IS) are summarized in Table 1. The main parameters for mass spectrum used in the QTRAP 5500 system were as follows: ion-spray voltage, 5.5 kV; temperature, 500°C; curtain gas, 20 psi; gas 1, 20 psi; gas 2, 20 psi; and collision gas, medium. Preparation of Calibration Standards, Quality Control Samples, and Pharmacokinetic Study Samples of 6A1, 6A1 Glucuronide, and 6A1 Sulfate [0409] Standard solutions of 6A1, 6A1 glucuronide, 6A1 sulfate, and IS were prepared separately at 100 µg/mL in methanol. Each standard solution was then fractioned into multiple mini-Eppendorf tubes and stored at -80°C. Each standard solution was diluted with 1:2 water- methanol solution into the final working solution of 1 µg/mL on the same of day usage and discarded after each use. The calibration standard solution was prepared via serial dilution of working solution to achieve the following concentrations: 0.25, 0.5, 1, 2, 3.91, 7.81, 15.63, 31.25, 62.5, 125, 250, 500, 1,000, and 1,400 ng/mL. Blood Samples [0410] Blank blood samples (40 µL) were spiked with 10 µL of calibration standard solution at different concentrations of analytes and 10 µL IS solution (50 ng/ml in 50% methanol in water). Each sample was extracted with 1,500 µL of acetonitrile after vortexing for 3 minutes. All samples were centrifuged at 20,000 x g for 15 minutes at 5°C. The supernatant was then transferred to another tube and evaporated to dryness under a steady stream of air at room temperature. The residue was reconstituted with 40 µL of 66% methanol in water (2:1), vortexed, and centrifuged again at 20,000 x g for 5 minutes. After centrifugation, 10 µL of the supernatant was injected into UPLC-MS/MS system for analysis. For pharmacokinetic samples, spiking of the calibration standard solution was omitted. Liver and Colonic Mucosa Samples Tissue Homogenization [0411] Liver tissue and colonic mucosa were excised from the sacrificed rats, washed with cold HBSS solution, and stored at −80°C in polypropylene tubes until homogenization. The frozen tissues were thawed, chopped and weighed. Exactly 50 mg of chopped tissue was homogenized in 500 µL of ice-cold homogenizing solution (pH 7.4) containing 10 mM potassium phosphate, 250 mM sucrose, and 1 mM EDTA dehydrate with a polytron tissue homogenizer. The homogenizer probe and test tubes were chilled at -4°C prior to use. Homogenization was paused for 20 seconds after every 30 seconds of homogenization at a medium speed. The homogenization was repeated 3 times until a visually uniform homogenate was obtained. Final tissue extract was stored at approximately −80°C prior to analysis. The homogenizer probe was washed sequentially with water, methanol, and water after every homogenization. Extraction of 6A1, 6A1 Glucuronide, 6A1 Sulfate from Tissue Homogenate [0412] Blank colonic mucosa or liver tissue samples (1 mg/µL) were spiked with 10 µL of calibration standard of different concentrations and 10 µL IS solution (Baohuoside 50 ng/mL of 1:1 methanolic water).1,500 µL of cold acetonitrile was added to each sample and vortexed for 3 minutes to precipitate out the protein. All samples were centrifuged at 20,000 x g for 15 minutes at 5°C. The supernatant then was passed thorough the Oasis PRiME solid phase extraction (SPE) 5cc cassette with applied vacuum. The eluents were collected and dried under a steady stream of air at room temperature. The residue was reconstituted with 40 µL of methanol in water (2:1 v/v), vortexed, and centrifuged again at 20,000 x g for 5 min. After centrifugation, 10 µL of the supernatant was injected into the UPLC-MS/MS system for analysis. [0413] Liver tissue and colonic mucosa at the conclusion of the pharmacokinetic study were collected, washed with cold HBSS buffer (pH 7.4), and stored at -80°C until homogenization.50 mg of defrosted, chopped tissue was homogenized in 500 µL of ice-cold homogenization buffer (pH 7.4, containing 10 mM potassium phosphate, 250 mM sucrose, and 1 mM EDTA dehydrate) using a polytron tissue homogenizer with its probes and test tubes previously chilled at -4°C. Tissue homogenates were processed as mentioned in the paragraph above, omitting the addition of calibration standards. Calculation of 6A1, 6A1 Glucuronide, 6A1 Sulfate Concentration in Tissue [0414] Blood and tissue concentrations of 6A1, 6A1 glucuronide, and 6A1 sulfate were calculated using the slope and intercept of the linear regression line (R2 > 0.995) generated from the standard curve of the same matrix. For example, a 392 ng /1000 µL nominal value of 6A1 has a count per second (CPS) raw read out of 5.56E6 with 50 ng of IS of 1.13E6. Back calculation gives the above read out of 378.49 ng/1000 µL, which has a 3.45% bias. The line generated range from 0 to 2000 ng/1000 µL of standard solution gives a slope of 1.3E-3 with R2 >0.995. For colonic mucosa of unknown concentration, the CPS readout of 10 µL injection of the extracted tissue with 1 mg/µL tissue sample was 5.42E6 over the internal standard’s CPS of 1.66E6 are normalized to give a ratio of 3.26. The normalized ratio is to avoid instrumental drift between each injection, thus 3.26 divided by the slope of the standard curve gives a concentration of 2,506.95 ng (1 mg/µL ÷ ng/1000 µL) of 6A1 per gram of colonic mucosa. Method Validation [0415] The linearity and stability, selectivity and specificity, sensitivity and carryover, accuracy and precision, as well as the extraction recovery and matrix effect studies followed US FDA guidance (139). Calibration Curve and Lower limit of quantification (LLOQ) [0416] The least-square linear regression method (1/x2 weight) was used to determine the slope and correlation coefficient of linear line. All linear regression lines were forced to go through origin at (0,0). Table 2 presents data showing the linearity of calibration curves for 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, liver, and colonic mucosa matrices. The LOD decided based on the 5:1 signal-to-noise ratio was 0.5 ng/mL, thus the lower limit of quantification (LLOQ) would be 2 ng/mL. Sensitivity [0417] The lower limit of quantification (LLOQ) samples at 2 ng/mL (n = 6) compared to blank matrix yielded peak/baseline ratio of fivefold. No significant carryover was observed in the double blank samples after six high QC concentration injections (Figure 35). Specificity and Selectivity [0418] The metabolites, sulfate and glucuronide, were detected via negative and positive ion mode. Their respective peaks were not overlaps in the elution time. The base line separation between each analyte to the IS peak were set at equal or greater than 1.5-fold. Accuracy and Precision [0419] The “intra-day” and “inter-day” precision and accuracy of the method were determined with quality control (QC) samples at three different concentrations of 20, 200, and 1000 ng/mL (sextuplet per set) and the lower limit of quantification (LLOQ) of 2 ng/mL. Accuracy is to determine how close the empirical peak analyte normalized by internal standard peak ratios divided by standard curve slope and theoretical nominal value are to each other (Equation 1). This value is expressed in percentage with 100% being the same.
Figure imgf000105_0001
Precision is the percent of coefficient of variation within the group and within different group measured (Equation 2).
Figure imgf000105_0002
Extraction Recovery and Matrix Effect [0420] Extraction recovery of 6A1, 6A1 sulfate, and 6A1 glucuronide in different bio- matrices (blood, liver, and colonic mucosa homogenate) were calculated by plotting the ratio of the peak areas of analyte (6A1, 6A1 glucuronide, and 6A1 sulfate) to IS (Baohuaside) in blank matrix spiked after the extraction procedure divided by the ratio of the peak areas of analyte to internal standard for the same matrix before the sample preparation step of liquid- liquid extraction (LLE) and solid phase extraction (SPE) (Equation 3). Similarly, matrix effects were calculated by dividing the ratio of the peak area of the compound and internal standard spiked into extracted blank matrix by the ratio of the peak area of same compound and internal standard in neat solution at the same concentration (Equation 4). All these experiments and evaluations were performed according to FDA recommended validation procedures (139).
Figure imgf000106_0001
Where: Response of post-extracted spike analyte is the normalized response of all 3 compounds (6A1, 6A1 glucuronide, and 6A1 sulfate) divided by IS (Baohuaside) that were added after the blank matrix has been processed via the LLE and SPE steps. Response of non-extracted analyte is the normalized response of all 3 compounds (6A1, 6A1 glucuronide, and 6A1 sulfate) divided by IS (Baohuaside) in neat solution. Stability [0421] Short-term, post-processing (25°C for 8 h), 3 freeze thaw cycles, and long-term (−80°C for 1 month) stability of 6A1, 6A1 sulfate, and 6A1 glucuronide was determined by analyzing three replicates of lower limit of quantification (LLOQ) and three QC samples for all three matrices. The stability sample results were compared to the freshly prepared samples to determine the stability of each condition. Preparation of the 6A1 Intravenous Injection Solution [0422] 0.25 w/v% solution of 6A1 was prepared in 40 v/v% of polyethylene glycol 300 and 60% v/v of ethanol. The solution was sonicated for five minutes until it became clear. The solution was further filtered using a 0.2 µm PTFE filter disc before injecting it into the rats. In Vivo Pharmacokinetic Study of 6A1 [0423] Male Fischer F344 rats (6–10 weeks, body weight between 250 to 280 g, n = 4) were purchased from Harlan Laboratory (Indianapolis, IN). Rats were kept in an environmentally controlled room (temperature: 25 ± 2°C, humidity: 50 ± 5%, and 12 h dark- light cycle) for at least 1 week before the experiments. The rats were fed ad libitum. The solution of 6A1 was administered via intravenous injection of the tail vein at a dose of 5.0 mg/Kg. Blood samples (about 20 –50 µL) were collected into heparinized tubes at 0, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post dosing via tail snip with isoflurane as anesthetic. All collected pharmacokinetic blood samples were stored at − 80°C until analysis. After a wash out period of 1 week, the rats were given a second dose via intravenous injection of 5 mg/Kg. Animals were sacrificed and tissues (colonic mucosa and liver tissues) were collected two hours post dosing. The procedures were approved by the University of Houston’s Institutional Animal Care and Uses Committee (IACUC). Pharmacokinetic Analyses [0424] The primary pharmacokinetic parameters of interest were the maximum concentration (Cmax), the area under the blood concentration–time curve (Figure 3) from baseline through 24 hours post-dose (AUC0–24H), and duration of time after intravenously dosed until reaching maximum systemic concentration (Tmax). The multiple Cmax/Tmax pairs that were observed are of special interest. Results and Discussion Solid stationary phase and column chemistry [0425] A 1.8 um particle size Raptor Restek biphenyl column and a 1.8 um particle size Acquity HSS T3 column were used to separate 6A1, 6A1 glucuronide, and 6A1 sulfate analytes. The biphenyl column has the ability to retain polar aliphatic/aromatic solutes throughout a wide range of pH, making it the ideal choice (140). However, the peak resolution deteriorated as the pH of the aqueous buffer decreased (141, 142). Concurrently, the biphenyl column's pressure also increases; thus, ACN became the replacement organic phase in place of the MeOH (Figure 37). The HSS T3 column had better analytes retention resulting in enhanced sensitivity using the same gradient elution method. The HSS T3 column also had a lower column pressure (6000 psi) compared to the biphenyl column (14,000 psi). Selectivity, Specificity, and Sensitivity [0426] Interference levels of endogenous compounds were determined via blank matrix injections (n=6). Glycoproteins, glycolipids, and phospholipids are known to interfere with the extraction and recovery of 6A1 and its metabolites from both colonic mucosa and liver tissue samples(143, 144). The addition of pass-through solid phase extraction using Oasis PRiME HLB cartridge after the liquid extraction step resolved low recovery due to endogenous matrix suppression of the ionization in the mass spectrometer. The chromatograms of blank matrix injections after each ULOQ sample exhibited a minor IS carryover peak at 4.15 minutes (Figure 36) equal to roughly about 5% of the lower limit of quantification (LLOQ). However, the IS carry-over peak was negligible to the quantification of analytes in this method. Linearity [0427] Three different matrices- blood, liver, and colonic mucosa- were used to validate the method. The calibration curves for 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, liver, and colonic mucosa matrixes are linear in the range of 0.5-1,400 ng/mL for blood, 0.5-1,400 ng/g for liver, and 0.5-1,400 ng/g for colonic mucosa matrices (Table 2). Linear regression equations were calculated from the raw data of the Count Per Second (CPS) response versus concentration graph with a weighting factor of 1/X2. The resulted coefficient of determination, R2, for all regression lines were > 0.995 (Table 2). Accuracy and Precision [0428] The accuracy assay values for lower limit of quantification (LLOQ) and 3 QCs concentrations ranged from 96.1-111.5% for 6A1, 97.6-103.3% for 6A1 glucuronide, and 99.62-108.14% for 6A1 sulfate (Table 3). These ranges are within ± 15%, the acceptable limit recommended by the FDA. The intra-day and inter-day precision and accuracy were accessed by analyzing the sextuplet LLOQ and QC samples at low, medium, and high concentrations of analytes in blood matrix (Table 3). The precision values ranged from 5.33-13.12% for 6A1, 8.65-14.9% for 6A1 glucuronide, and 5.08-12.3% for 6A1 sulfate (Table 3). Extraction Recovery [0429] The mean extraction recovery for blood matrix analyzed from three replicates of lower limit of quantification (LLOQ) and three QC and LLOQ samples at 2, 20, 200, and 500 ng/mL were between 95.2-99.6% for 6A1, 94.3-100.1% for 6A1 glucuronide, and 96.1-101.3% for 6A1 sulfate (Table 4). The extraction recovery for liver tissue of LLOQ and three QC samples were between 93.6-95.9% for 6A1, 109-112% for 6A1 glucuronide, and 98-108.6% for 6A1 sulfate (Table 4). The colonic mucosa extraction recovery for the above LLOQ and QC samples were 99.4-103.8% for 6A1, 91.71-98.45% for 6A1 glucuronide, and 94.1-103.9% for 6A1 sulfate (Table 4). The extraction recovery of colonic mucosa had the highest %CV at low concentration (20 ng/g); however, all LLOQ and QC samples were reproducible judging from the small %CV between groups and were within the recommendation of the FDA guidance (Table 4). Matrix Effect [0430] An ion enhancement (15.99%) in the response signal was observed for the lower limit of quantification (LLOQ) of colonic mucosa matrix. However, the effect was resolved after the additional application of a 96-well Oasis HLB µ-elution plate. The overall matrix effect for blood samples ranged from 92-108%, 94.5-108.88% for colonic mucosa, and 88- 113% for liver tissue (Table 5). The internal standard matrix effect is negligible thus comprehensive matrix effect was not evaluated. Stability [0431] Stability of 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, colonic mucosa, and liver tissue was evaluated by analyzing triplicates of LLQC and QC samples at three different concentrations following 8 h at 25°C bench top (short term stability), 3 freeze thawing cycles, and at −80°C for 30 days (−80°C and 25°C). All the samples showed 95–105% recoveries after various stability tests (Table 6). In Vivo Pharmacokinetic Study of 6A1 [0432] The established UPLC-MS/MS method was utilized to determine the concentration of 6A1, 6A1 glucuronide, and 6A1 sulfate in a blood pharmacokinetic study and in tissue samples collected from F344 rats (n=4). A single-dose intravenous administration of 6A1 was given at 5 mg/Kg normalized by the rats’ body weight. Figure 10 shows the mean blood concentration time profile of 6A1 and its metabolites. The validated method was able to detect all three compounds at and above the lower limit of quantification (LLOQ) of 2 ng/mL (or 2 ng/g of tissue samples). [0433] A double peak phenomenon was observed in the pharmacokinetic study of blood concentration-time profiles of 6A1 and 6A1 glucuronide. This indicated that the enterohepatic recirculation may be involved in the disposition of 6A1. A second peak of 6A1 at 258.47 ± 100.35 ng/mL was observed five hours after the dose was administered. The first 6A1 glucuronide peak (at 288.16 ± 94.88 ng/mL) was observed two hours post dose, whereas a second 6A1 glucuronidation peak with concentration of 183.08 ± 52.71 ng/mL was observed at four hours after intravenous injection. Based on stability data established for 6A1 glucuronide, it is unlikely for the metabolite to deconjugate to 6A1 in the blood and tissues. Therefore, the double peak phenomenon is most likely the contribution of the enterohepatic recycling mechanism. It is believed that intravenous 6A1 is metabolized in the liver, secreted into the bile as 6A1 glucuronide and 6A1 sulfate. Once in the intestine, the colonic β- glucuronidase hydrolyzed the glucuronide metabolites into 6A1, which is then reabsorbed in the colon which give rise to the second peak of 6A1. The reabsorbed 6A1 again got glucuronidated and gave rise to a second peak of 6A1 glucuronide. The exact mechanism requires further investigation as to determine the recycling ratio and overall systemic exposure to 6A1. When compared to a previous study (145), a dose-normalized AUC (ng*hr/mL*mg) indicated that the systemic exposure of 6A1 is 9.04% that of the systemic exposure of celecoxib. The consistent 6A1 sulfate concentration of around 37.47 ± 17.22 ng/mL throughout the study suggested that metabolic disposition of 6A1 in F344 rats is primarily via glucuronidation. Colonic mucosa had an average ± SEM concentration of 225.94 ± 65.94 ng/g for 6A1, 26.78 ± 17.80 ng/g for 6A1 glucuronide, and 5.12 ± 0.86 ng/g for 6A1 sulfate (Figure 11A). Liver tissues had an average concentration of 337.60 ± 100.27 ng/g for 6A1, 315.79 ± 81.55 ng/g for 6A1 glucuronide, and 218.14 ± 54.21 ng/g for 6A1 sulfate (Figure 11B). [0434] In conclusion, a robust, sensitive, and validated UPLC-MS/MS method was developed and successfully used to quantify 6A1, 6A1 glucuronide, and 6A1 sulfate in pharmacokinetic blood, colonic mucosa, and liver tissue samples. The method has a multitude of advantages, such as, high sensitivity lower limit of quantification (LLOQ) 2 ng/mL, short analysis time (6 minutes), small sampling size (10 µL), good recovery with negligible matrix effect, and simplistic sample processing methods.
Table 1: Compound dependent parameters 6A1, 6A1 glucuronide, IS, and 6A1 sulfate parameters in MRM mode for LC-MS/MS
Figure imgf000111_0001
Table 2: Linearity of the standard calibration curve of 6A1, 6A1 glucuronide, 6A1 sulfate in blood, colonic mucosa, and liver tissue (Mean ± SD (%CV))
Figure imgf000111_0002
Table 3: Intraday and Inter-day accuracy (Mean ± SD) and precision (%CV) data of 6A1, 6A1 glucuronide, and 6A1 sulfate for blood matrix using MRM method at three QC concentrations
Figure imgf000111_0003
± 287 ± 571 ± 141 ± 8.67
Figure imgf000112_0001
Table 4: The extraction and recovery (Mean ± SD (%CV)) of 6A1, 6A1 glucuronide, and 6A1 sulfate compounds for blood, colonic mucosa and liver tissue at lower limit of quantification (LLOQ) and three QC concentrations
Figure imgf000112_0002
Table 5: Matrix Effect (Mean ± SD (%CV)) of 6A1, 6A1 glucuronide, and 6A1 sulfate compounds for blood, colonic mucosa, and liver tissue at lower limit of quantification (LLOQ) and three QC concentrations Blood
Figure imgf000113_0001
Table 6: Stabilities of 6A1, 6A1 glucuronide, and 6A1 sulfate of blood, colonic mucosa, and liver tissue were evaluated by analyzing triplicates of LLQC and QC samples at three different concentrations following A) 8 h at 25°C bench top (short term stability), B) 3 freeze thawing cycles, C) stored at −80°C for 30 days (−80°C and 25°C), and D) processed sample, autosampler, 20°C for 24 h A) 8 h at 25°C bench top (short term stability)
Figure imgf000113_0002
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
[0435] Example 2. Biodegradable coating formulation and vacuum spinning technique applied to size 9 hard gelatin capsules to deliver a selective COX-2 inhibitor to the colon of F344 rats [0436] The purpose of this study was to use a newly-designed selective COX-2 inhibitor (6A1) in combination with a coating formulation and technique to deliver a high drug concentration to the colon. Drug powder and image tracers were packed inside size nine hard gelatin capsules (S9C), coated with an inner layer of polylactic glycolic acid (PLGA 85:15) with optimum thickness of 115 ± 35 nm for PLGA 8515 and 50 ± 15 nm for Eudragit S100 (ES100) to serve as delayed-release coating via erosion mechanism and an outer pH-sensitive ES100 layer to serve as an enteric coating. [0437] Vacuum spin coating technique and parameters, as well as new solvent systems, were also investigated. Locally bioavailable COX-2 inhibitor’s (6A1) pharmacokinetic behavior of S9C capsules were extensively studied in F344 rats. S9C capsules were packed with 6A1 (90% w/w), barium sulfate (5% w/w), and methyl blue (5% w/w), coated and administered via oral gavage to F344 rats. In vivo pharmacokinetic study of coated capsules was performed in conjunction with in vivo imaging. The blood concentrations time profile of the enteric (ES100) and delayed-release (PLGA) coating formulation administered via oral gavage achieved a delayed release beyond 6 hours and above pH 6.8. The ex vivo tissue stain showed that the capsule released its contents at the distal gastrointestinal tract, i.e., the colon. Live in vivo images obtained also further validated the S9C released in the colon. [0438] The PLGA 85:15 (also referred to herein as “PLGA 8515”) grade coating was chosen based on its rapid ability to provide scaffolding support for the hard gelatin capsule after it has become wet in the coating process, thereby preventing collapse of the capsule. PLGA 85:15 also allows for successive coating applications. [0439] Eudragit S100 (ES100) is an anionic copolymer composed of methacrylic acid (MAA)- methyl methacrylate (MMA) at a 1:2 ratio. ES100’s pH dependent nature was finely tuned by the number of negative charges on the MAA’s COOH functional group at basic conditions (162). The hydrophobicity of MMA units was realized by synthesizing co-polymer libraries with a systematic variation. ES100 is soluble in alkaline digestive fluids by salt formation (163, 164). A clinical study shown that a 5% W/W gain of the coated Eudragit S100 tablets (84 ± 4 micron) (64) releases its contents in the colon in 67% of volunteer subjects. [0440] A disadvantage noticed when using ES100 is the brittleness of the film as the total polymer weight increased. As such, the manufacturer recommended as much as 25 % addition of plasticizer into the formulation to increase flexibility (Figure 38). However, the addition of plasticizer changes the dissolution profile and takes longer for the film to dry (165). [0441] Therefore, there is a need to develop alternative coating techniques to limit drying time, maximize polymer mass loading to 10%, and limit the use of plasticizer while achieving uniformly smooth, defect free films. [0442] This research investigated three different coating techniques- dip coating (control), fluidic coating (technique 1), and vacuum spin coating (coating technique 2). The parameters used to judge each technique were: smoothness of the film, reproducibility, and ease of clean up. Of the three, vacuum spin coating yielded the most optimal results. Materials and Methods Materials [0443] Liquid chromatography-mass spectrometry (LC-MS) grade acetone, ethanol, methanol, dichloromethane, isopropanol, DI water, barium sulfate of 99.99% purity were purchased from Sigma-Aldrich Corp. (St. Louise, MO, USA). Ester-terminated DL-PLGA of 85:15 (PLGA 8515) were purchased from (Lactel Absorbable Polymers, Birmingham, AL). Evonik RÖHM GmbH (Darmstadt, DE) generously donated Eudragit S100 (ES100). Methyl blue, certified by the biological stain commission, M5528-25G, was also obtained from Sigma- Aldrich Corp. Size 9 capsules (S9C) were purchased from Torpac Inc. (Torpac Custom capsules & capsule fillers, 333 Route 46, Fairfield, NJ 07004, USA). The parent compound, 6A1, and 6A1 sulfate were synthesized in house and quantified at 98% pure using the UPLC and NMR. 6A1 glucuronide was biosynthesized using Hela-UGT1A9 cells, purified, and quantified at 98% pure using the previously established methods. Preparation of Capsules [0444] Hard gelatin capsules were visually inspected for shape and coloration, then weighed and selected for use if their weights were within 2 - 5% of the expected range. Drug powder (with 5% w/w barium sulfate 5% w/w and methyl blue) was packed into the capsules using the funnel and pestle kit sold by the vendor. The total weight of capsules and drug blend was monitored within a 3% range of drug packing of 20 mg. A trace amount of pH 9 water (~2 µl via a pipette tip) was used to seal the body and cap. The sealed capsules were allowed to dry for 24 hours before any coating materials were applied (Figure 12). Factorial Solvent Design to Dissolve PLGA8515 and ES100 [0445] Full factorial designs were used to optimize the solvent systems used to incorporate PLGA 8515 and ES100 (JMP 14 DOE, SAS Institute Inc.100 SAS Campus Drive, Cary, NC 27513-2414, USA). The three continuous factors were: 50–80% for acetone, 5-15% for isopropanol, and 2-5% for deionized water. The responses were: minimal time takes for a single coat applicant to dry and the maximum optimal polymer weight dissolved in certain solvent systems. Such constraints allow for practical application (single polymer, single application) as well as efficient drying between successive coats (Figure 13). Parameter Use for Coating of Capsules Vacuum Spin Coating [0446] It was important to clean and dry the vacuum spinning plates before coating each batch. The sealed capsules were then wetted with mild basic water (pH 9 NaOH 0.1 mM) and loaded into individual pods within the vacuum spinning plates. 50 µL of PLGA 851510 % solution was added into the individual pod, a vacuum was applied, and the plate was allowed to spin for 30 seconds at low spinning speed (100 RPM). The pod spinning rate was increased to 500 RPM and spun for 15 minutes. A successive coat was applied by adding 50 µL of Eudragit S100, applying vacuum, spinning for 30 seconds at a low spinning rate of 100 RPM, followed by a spin rate of 500 RPM for 15 minutes. The capsule was sufficiently dried to be transferred into an individual micro-Eppendorf vial with the lid left open and allowed to stay in a desiccator for complete drying over 24 hours (Figure 14). Appearance and Quality Control Parameters of Coated Capsules [0447] After each successive coating, a visual inspection of individual capsules was performed using a 3.5X-90X zoom trinocular microscope (AmScope, SM-1TSZ-V203); close observations were made to look for wrinkles, craters, and asymmetry (Figure 15). By comparing the coated and uncoated S9C, a different sheen was observed. The coated capsules were weighed, and the total mass gain was then used to calculate the estimated thickness. The predicted thickness of the capsule was based on the equation below. The capsules were rejected if they were not within the ± 2.5% of the expected thickness (150 nm) based on mass gained.
Figure imgf000119_0001
Thermogravimetric Analysis (TGA) [0448] Films of PLGA and ES100 applied to capsules an hour prior were peeled and stamped to fit the aluminum pan. Approximately 10 ± 1 mg of each polymer film per sample was used. Randomized, triplicate films were tested. The TGA curves (Shimadzu, TGA50-H) acquired from temperature ramping rate were 10 °C min−1 from room temperature (~ 20°C), held for 30 seconds at 30°C, and steady ramping till 500°C. Formulation consistency across all formulation batches was determined via the % weight loss curve. Differential Scanning Calorimetry (DSC) [0449] The DSC equipment (Shimadzu model DSC-60A) was programmed to heat the peeled films from coated samples within a range of room temperature (~ 20°C) to 500°C, at a heating rate of 10°C∙min−1. An aluminum, sealed sample holder was used. Reference sample contained an empty but crimped aluminum pan and lid. Nitrogen was utilized as the flowing gas, at a rate of 10 mL∙min−1. The mass of the analyzed samples was 10 ± 1 mg (167). Scanning Electron Microscopy (SEM) [0450] The topography and thickness of randomized samples was inspected by using a scanning electron microscope (SEM, TM3000 tabletop microscope, Hitachi). Three (3) PLGA+ES coated capsules that passed the mass gain quality control step were chosen at random for every 24 coated capsules and dissected into two halves longitudinally. SEM images were obtained using the following operating parameters: 5.0 kV voltage, 4 mm working distance, and 10-6 millitorr vacuum. The side views of the capsule halves were captured at an 80,000 x magnification and used to measure the coated films’ thickness. Due to the excessive charging of the polymer substrate, the samples were coated with graphene prior to SEM imaging. In Vitro Dissolution of Coated Capsules [0451] The in vitro dissolution test for the coated capsules was performed in accordance with the recommendation from UPS compendial dissolution, non-sink conditions (168). The simulated intestinal pH was adjusted via 2.5 µmol / L of HCl or NaOH from the 2.5 µmol / L of phosphate buffer. 1 mL of samples were collected every half hour, and the pH exposure and duration were as follows: 1.5 hours at pH 1.2 (stomach), 2.5 hours at pH 4.6 (small intestine), 2 hours at pH 6.8, 2.5 hours at pH 8, and 2.5 hour at pH 6.8. Each capsule was placed within a sinker housing, and the stirrer rate was set at 80 rpm (169) at 37 ± 2 °C. Samples at an extreme pH were air-dried, reconstituted, and neutralized before analysis. Drug concentration (6A1) was determined using the UPLC MS-MS method that was previously validated. In Vivo, Ex Vivo, Live Imaging Study of Coated Capsules [0452] Male Fisher F344 rats (6–10 weeks, body weight between 250 to 280 g, n = 12) were used. Rats were kept in an environmentally controlled room (temperature: 25 ± 2 °C, humidity: 50 ± 5%, 12 h dark-light cycle). Capsules were administered at a dose of 5 mg/kg via oral gavage. The study used two groups of fed rats (administered control uncoated capsules vs. coated capsules) that were cage mates. Blood samples (about 20 –50 µL) were collected into heparinized tubes at 0, 0.5,1, 3, 5, 7, 10, and 24 hours post-dosing via tail snip with isoflurane as an anesthetic. All collected pharmacokinetic blood samples were stored at −80 °C until analysis. [0453] A Perkin Elmer IVIS Lumina III XRMS in X-ray (40 kVp, 100 mAmps, 10 seconds) and photograph modes was used to image the capsules containing barium sulfate (specific settings and data acquisition can be found in greater details in Figure 39). The X-ray source was a tungsten anode lamp. No hair removal was needed. The animals were anesthetized in an induction chamber with constant isoflurane feeding, weighed, manually transferred to the IVIS Lumina imaging system, and constantly fed isoflurane via the adaptor nose cone built within the imaging chamber. The animals were secured in individual pods. The pod could be rotated via a computer control interface. The position/sedation of animals was controlled via a live camera built-in within the chamber. At time: 0 (n=1), 2 (n=1), 4 (n=1), 6 (n=1), 8(n=1), 12 (n=3) hours, the IVIS Lumina III XRMS (170) was used to track the location of the capsules and the rats digestive tissues’ were excised at the end of each time point to examine the intestinal tissue of methyl blue stains. The control group used uncoated hard gelatin capsules. In vivo transit time and the capsules’ release profiles were also monitored periodically during the pharmacokinetic study. The imaging group’s (Group 1) animals were cage mates to the pharmacokinetic study group (Group 2) as to eliminate confounding factors (Figure 16). Statistical Analysis of Results [0454] The blood concentration-time profiles of 6A1 of each subject (n=6) were analyzed by a noncompartmental method using WinNonlin® 6.1 (Pharsight Corporation, Mountain View, CA, USA). All statistical analyses were performed using the GraphPad Prism 8.0 program (GraphPad Software Inc., San Diego, CA, USA). Results and Discussion Design and Selection of Solvents for Capsule Coating Solution [0455] A full factorial screening design with an extreme vertexes option was used (Figure 13A). The PValue, representing the likelihood that each solvent was significant, was set to 0.05 resulting in a Logworth of 1.30 (LogWorth is the -log (PValue)). The results indicated that the addition of isopropanol into the acetone:water mixture was significant and could not be omitted (PValue: 0.045; LogWorth: 1.34). There was no confounding factor between the solvents, namely the interaction between IPA:water, acetone:PA, and acetone:water (Figure 13B). However, the concentration ratio of IPA to acetone was more significant than the concentration ratio of IPA to water (Figure 16A). The R2 linear fit of the theoretical predictive model to the empirical values was 1.0 for the time needed to dry using the redesigned solvent mixture (Figure 14A); the R2 of the maximum amount of polymer added to the same solvent mixture was 0.93 (Figure 14B). Together, the predictive models indicate that the most desirable ratio of a solvent system for dissolving 11% of Eudragit S100, with a drying time of 8.75 minutes, was 60:10:3.5 (acetone: IPA: water, Figure 17). Because the water’s PValue falls within the range of a significant 0.05 to an insignificant 0.1, its significance cannot be entirely ignored. The summary output showed no confounding factors between IPA to water and Acetone to Water. Again, the Logworth did not conclusively exclude confounding factors between acetone and water. Based on the cumulative results and the positive desirability simulated, water was included in all future formulations. [0456] The prediction profiler was set and selected with maximum desirability of polymer mass loading (7.0 – 15%) and minimal time needed to dry (5.5 – 12 minutes) as constraints. The negative slope of polymer drying time to the increasing amount of acetone (Figure 17A) means that the addition of acetone to the formulation positively impacted the drying time of film; it took less time to dry the coated film. The positive slope of polymer drying time (Figure 17B) to the increasing IPA within the formulation suggested an increase in the amount of drying time to the film. Water had a negative effect on the formulation drying time but of lesser magnitude compared to IPA. The positive slopes of the profilers (Figure 16D and 16F) demonstrated that by increasing the ratio of acetone and water, an increased amount of polymer could be added to the system. Increasing the ratio of IPA decreased the amount of polymer that can be added (Figure 17E) implying that IPA has negative desirability to the amount of polymer dissolving within the system. [0457] IPA was not excluded from the formulation even though it has an overall negative desirability based upon a visual inspection. A solvent system without IPA was observed to have excessive crater defects and be less smooth compared to the solvent system with 10 % IPA. Thus, IPA was incorporated into all future solvent systems. Determination of Parameters Used for Vacuum Spin Coating of Capsules [0458] Three different coating approaches were investigated. The dip-coating approach suffers from a serious drawback, in that the polymer becomes fuzzy at the holding and capsule contacting points (Figure 15A). The defective non-uniform coat surface contributed to a larger variability in the release of drug powder and tracers contained within the capsule. The vacuum spin coating approach successfully met the surface uniformity and apparent smoothness requirements (Figure 15B). The fluid coating approach also suffered from drawbacks, such as excessive surface wrinkles (Figure 15C). A typical sample of the capsules that were coated by the fluidic coater can be seen in the right SEM image. The cleaning steps, unclogging polymers, and the extended soaking of the capsules led to an excessive amount of wrinkle formation. Thus, the fluid coating approach was abandoned. [0459] The vacuum spin coating technique (Figure 18) achieved an 88% success rate: a total of 36 of the 40 capsules made by this technique passed the in-house quality assurance tests- smoothness of surface appearance, weight gained, thickness measurement, and in vitro dissolution. The approach proved vacuum spin coating was a superior choice for coating S9C (Figure 7A-7D). [0460] The spin coating technique coated high viscosity polymers PLGA8515 and ES100 with a high mass loading of 10% w/w formulation and sufficiently covered the entire three dimensional S9C. The vacuum applied to the bottom of the capsules placed inside a slot within the coater plate creates a shear force that thins out the polymers, which behave as non- Newtonian fluids. The tinning is due to the entanglement of polymer collapsing in the direction that the vacuum is applied (bottom of the plate). The angle at which the capsule sits causes it to spin around as centrifugal force results from the spinning of the coater plate. The centrifugal force is large enough to spin out the large undissolved polymers and creates a vortex of air that facilitates further evaporation. Without being limited to a particular theory, the combination of the vacuum and the spinning creates a conical vortex within each capsule pod. [0461] The optimal PLGA 8515 inner erosion coat was made of 115 ± 35 nm PLGA 8515, and the optimal thickness of the pH-sensitive enteric coat was 50 ± 15 nm Eudragit S100. Different PLGA thicknesses were also investigated. PLGA 8515 layers that were thinner than 100 nm showed insufficient scaffolding support for S9C and had a structurally deformed surface upon successive coating and drying. A PLGA 8515 film thicker than 150 nm failed to release its contents at the targeted time. ES100 thickness also has some impact on the time taken by the capsule to release its contents; however, an imperfection in the coating layer of ES100 was not observed via the vacuum spin coating technique. Determination of Quality Control Parameters of Coated Capsules Thermogravimetry Analysis (TGA) [0462] Thermogravimetry analysis is a measurement of the weight loss curve with the response to temperature ramp. Here, the TGA was used to determine the total solid mass of the polymers’ formulation and its consistency across each formulation batch used for each coating. The first steep drop in the weight loss occurred very early and at a relatively low temperature of 20 to 40 °C. The limited loss was accepted, as the weight loss occurs due to the dehydration of solvents. A second weight loss ~10% between 370 to 375°C was the final solid mass of the bulk polymers (Figure 19A-B). Compared to the solid materials, both PLGA 8515 and ES100 represented, on average, 9-10% of the formulation by weight. The TGA also provided a general clarification regarding the nature of the polymer formulation decomposition’s nature. The mass loss across each triplicate run for the materials remained consistent with respect to the onset temperature and duration, and the decomposition of both materials reflected in the total weight loss correlated with the decomposition of the materials’ solid weights; thus, the results indicated that the solvents used to dissolve the polymers did not cause any physio-chemical reaction. Data acquired were consistent with published data (151) and in house historical data (Figure 19C-D). Differential Scanning Calorimetry (DSC) [0463] DSC was used to determine the integrity of dried polymer solutions for possible chemical oxidation and crystallization via inspection of the glass transition temperature (Tg)(167). Indium was used as the calibration standard for the DSC’s temperature probe (Figure 19). ES100 had a small glass transition point at 175 °C. When overlaid with the ES100 dried film to that of manufacturer-supplied solid powder, the two curves (run in triplicate) had the same pattern consistent with the published data (171). The DSC curves for both the PLGA 8515 solid and dried film presented a glass transition temperature (Tg) of 45°C, which agreed with the literature (161, 172). Both DSC and TGA confirmed that the solvents did not cause oxidation or phase changes to the polymers so as to compromise its integrity (173). In Vitro Dissolution of Coated Size 9 Hard Gelatin Capsules (S9C) [0464] The in vitro, pH-dependent dissolution profile of S9C containing 6A1 was investigated in a buffer with gradually changing pH to assess the ability of coated layers to protect the contents from the acidity and accomplish an effective delayed-release delivery in neutral pH values (163).6A1 was not released in the medium at pH 1.2 and 4.5 (<15% released) (Figure 20), suggesting that the capsules would not release their contents in the stomach. Once the pH reached the value of 6.8, 6A1 was rapidly released within 21 minutes and completely emptied the drug content within two hours. One quality-control capsule that was rejected based on an in-house quality control criteria was also found (red curve, Figure 20). The dissolution test indicated the capsule failed to achieve acid resistance, and the delayed-release mechanism might have been caused by a thinner coating of material being deposited. Most of the coated capsules began the release of 6A1 at a pH of 6.8 and six and a half-hour after initial PBS exposure (Figure 20). The visual methyl blue stain results also confirmed the enteric and delayed-release mechanism of the coated S9C. Methyl blue packed inside coated S9C had spread into the medium beginning approximately 6 hours after initial PBS buffered exposure (Figure 20A). The methyl blue vortex tail was more apparent at 6 hours and 21 minutes (Figure 20B) and the capsule shell of completely emptied S9C were observed at 7 hours and 12 minutes (Figure 20C). Values from a student t test performed using the quantified amount of 6A1 samples collected during the dissolution test yielded insignificant differences within the seven S9C. The concentrations of 6A1 were determined using a previously established and validated UPLC-MS/MS method (174). In vivo and ex vivo imaging study of coated S9C [0465] Coated S9C appeared as bright spots on the µ-CT IVIS images that were obtained by overlaying the X-ray and photograph modes; the coated material did not appear to interfere with the X-ray imaging agent, BaSO4 (Figure 21A). For the first two hours, the coated S9C were visible in the stomach of group one and were not visible within the intestinal tract until four hours after gavage (Figure 21B-21C). Many smaller bright spots were observed in the ceca and colons of the remaining animals in group one (Figure 21D). Eight hours post gavage, the S9C were not visible within the intestinal tract (Figure 21E-21F). The disappearance of the image tracer can be explained by an insufficient amount of BaSO4 density concentration at a location to provide positive X-ray contrast. In general, barium (atomic number 56) is an oral X-Ray contrasting agent specifically used to delineate the gastrointestinal tract in both preclinical and clinical settings (175, 176). Due to barium’s toxicity, barium sulfate, an inert insoluble barium-complex given as an oral slurry is often formulated to a specific gravity of 1.5 density (mass/oral suspension concentration) for positive X-Ray capturing (177). Due to the depth of the tissue and air gap within the abdominal cavity, the disappearance of the capsule after 8 hours might have been caused by the S9C emptying its contents or becoming diluted with fecal matter thus lowering the barium density below the detectable concentration. [0466] Upon examining the ex vivo tissue samples of the intestinal tract, the methyl blue stains were indicative of the S9C’s location (178, 179). In the control group that were given uncoated S9C, one hour after oral gavage, the methyl blue mass was still in the stomach and mixed with chyme (Figure 21G). Three hours after oral gavage, the blue stains had passed the duodenum and were visible in the jejunum in the second control animal (Figure 10H). Contrarily, upon examining the ex vivo gastrointestinal tract of group one, which were given coated S9C four hours after oral gavage, no blue stains were visible from ruptured S9C (Figure 21I). The ex vivo ceca were extensively stained eight hours after oral gavage when examining the gastrointestinal tract (Figure 21J and Figure 40). At 10 hours after oral gavage, ex vivo examination showed partial stains of the cecum and complete blue stains of the colon (Figure 21K). The remaining two animals from group one excreted blue-stained fecal pellets at ten hours, and the remnants of PLGA 8515’s stringy material, as well as capsule shells, were visible at 12 hours after oral gavage (Figure 21L-21N and Figure 37). The ex vivo tissues confirmed that the S9C had released most of its contents in the colon between six and eight hours after oral gavage. Pharmacokinetic study of the coated S9C [0467] The validated UPLC-MS/MS method was used to determine the blood and tissue concentrations of 6A1, 6A1 glucuronide, and 6A1 sulfate in a pharmacokinetic study utilizing F344 rats (n=5). A single oral gavage S9C of 6A1 was given containing a 5 mg/Kg dose normalized by the rats’ body weight (Figure 22). The mean blood concentration time profile of 6A1 and its metabolites detected at and above the lower limit of quantification (LLOQ) of 2 ng/mL (or 2 ng/g of tissue samples). Five of the blood concentration data points below the LLOQ, but above the LOD, were also quantified. The first Cmax/Tmax of coated S9C 6A1 was observed five hours after oral gavage (1320.68 ± 141.20 ng/mL/ 5Hr) which has a Tlag time of four and a half hours compared to the intravenous route previously studied ((n=4) at 5 mg/ Kg dose, 1700 ± 859.5 ng/mL). The dose normalized Cmax of oral S9C was 4.09% compared to the Cmax of the 6A1 IV. The first 6A1 glucuronide peak was also observed at 687.08 ± 182.12 ng/mL five hours after oral gavage. The 6A1 sulfate concentration of 41.20 ± 5.29 ng/mL throughout the study suggested that metabolic disposition of 6A1 in F344 rats was primarily via glucuronidation. [0468] The 6A1’s dose normalized AUC_24Hour_S9C was 0.7942 ± 0.1731 nM/mL for S9C dosage form. Compared to IV AUC_24Hour_IV of 8.489 ± 1.291 nM/mL, the AUC_24Hour_S9C_Uncoat was 12.00% and 9.35% for the enteric and delayed release coated capsules. In other words, the absolute bioavailability (Fapp: AUC_24Hour_S9C Coated /AUC_24Hour_IV) of 6A1 coated capsule was 9.30%, and the relative bioavailable (FRel: AUC_24Hour_S9C Coated / AUC_24Hour_S9C_Uncoated) between the uncoated and coated capsule dosage form was 78.0%. Thus, the total systemic exposure of the S9C coated dosage form reduced the total systemic exposure to less than 10%. A 2-way ANOVA test was performed with the Tukey post hoc approach for dependent variables (AUC_24Hour and time) of which the test yielded significant across all three dosage forms. The Tukey’s multiple comparisons across rows (time of each blood samplings vs concentration) had significant different across three dosage up to 4 hours post dosed, which further support the Tlag time observed from the Cmax/Tmax of 6A1 dose normalized blood concentration time profile. The 6A1 glucuronide AUC_24Hour_S9C_Coated exhibited a similar pattern to that of the 6A1 parent compound but yielded statistically insignificant when a one-way ANOVA analysis were performed across the glucuronide metabolites of all three-dosage form (Figure 23). [0469] Colonic mucosa tissue triplicate samples were also analyzed for three rats that were collected at 10 hours (n=1) and twelve hours after oral gavage of S9C; the average 6A1 tissue concentrations were 400.0 ± 65.94, 482.50 ± 35.86, and 525.0 ± 26.78 ng/g. Compared to the colon tissue concentration of rats via intravenous dosing (225.94 ± 65.94 ng/g), the S9C colonic tissue concentrations were 1.7 and 2.3 times higher. The 6A1 colon tissue concentration of S9C were 6 times higher than the oral suspension after adjusted for the different in dosage. A Holm- Sidak’s multiple comparisons test was used to analyze the three dosage forms of colonic tissue 6A1 concentrations and the output suggested that IV and oral suspension tissue concentration was significant (P = 0.0019); there were also a significant difference between IV and coated S9C (P = 0.0017), but the most dramatic differences were observed between the coated S9C dosage and oral suspension (P < 0.0001). [0470] In conclusion, a new solvent system was discovered and investigated for the polymers in this experiment. The solvent ratio of 65% acetone: 10% IPA: 3.5% water will dissolve up to 11% of Eudragit S100 and achieve the shortest amount of time needed to dry each coated film at 8.75 minutes. Utilizing the vacuum spin coating technique, the size 9 hard gelatin capsules (S9C) packed with the novel selective COX-2 inhibitor (6A1), resulted in consistent coatings that attained a uniform release mechanism targeting the colon. [0471] When compared to a previous study (145), a dose-normalized AUC (ng*hr/mL*mg) indicated that the systemic exposure of 6A1 is 9.04% that of the systemic exposure of celecoxib given oral suspension (145). A separate oral suspension study with a 40 mg/kg BID dose of Celebrex (dose equivalent to an 800 mg/day in human) indicated all CV toxicity signs but also reduced the colonic tissue PGE2 by 58%. The same study also showed a Celebrex concentration of about 556.67 ng/g of colonic tissues. Compared to the same treatment and dosage, 6A1 concentration was 138.75 ng/g in the colon (180). [0472] The combination coating approach could overcome the exiting drawbacks associated with their conventional dosage form (i.e., oral suspension). The pharmacokinetic study, the in vivo imaging, and the ex vivo tissue staining, and the colon drug concentration (525.0 ± 26.78 ng/g ) showed that the cumulative approaches were able to deliver more than twice the amount of 6A1 to the colon compared to the intravenous route, three times the amount of 6A1 compared to oral suspension, and similar in magnitude to the concentration needed to have therapeutic effect compared to Celebrex. [0473] Example 3. Zero order release of enteric and delayed coatings of S9C to deliver a selective COX-2 inhibitor (6A1) to the colon of F344 rats [0474] In this research, enterically coated microparticles were formulated and packed inside the coated S9C in place of raw 6A1 powder. It was hypothesized that the combination approach will achieve a zero order sustained delay release of 6A1 in the colon (lower AUC_systemic, increase local AUC_colon).Zero order release kinetics refers to the process of constant drug release from the delivery system. Previous data from a drug mixed into the rats’ food indicated a steady state would be reached after four days of medication (180); as such, a single dose pharmacokinetic study in conjunction with a short term (4 days) efficacy study was done. F344 rats (n=12) were divided into two groups. Each group was administered 20 mg/Kg doses normalized by the rats’ weight. Blood samples were collected periodically and the blood concentration time profile of normalized dose showed the total systemic exposure (AUC_systemic) remained constant while the AUC_colon increased by two fold upon analyzing the ex vivo colonic mucosa tissues samples. Material and Methods Chemicals and Materials [0475] Example 1 established methods for the preparation and purification of 6A1, 6A1- Sulfate, and 6A1-glucuronide. Acetone, dichloromethane, isopropanol, DI water of 99.99% purity, and Polyvinyl alcohol (PVA, Mw ~ 31–50 kDa) were purchased from Sigma-Aldrich Corp. (St. Louise, MO, USA). Ester-terminated DL-PLGA of 50:50 and 85:15 were purchased from (Lactel Absorbable Polymers, Birmingham, AL). Evonik RÖHM GmbH (Darmstadt, DE) generously donated the ES100. Capsule sinkers, Catalog #3, item PSCAPWHT-XS were purchased from Dissolution Accessories (The Netherlands, De Kreek 12, 4906 BB Oosterhout). Formulation of 6A1 Microparticles [0476] Polymeric pH-sensitive ES100 microparticles loaded with 6A1 were prepared via the wet milling and spontaneous emulsification solvent evaporation methods; the final formulation was a combination of these previously published methods (162, 163, 186) with some modifications. The powder 6A1 was first wet milled at 10 mg/mL in 35% methanol water at 1,600 rpm for an hour. Different volumes of 10 mg/mL of 6A1 ranging from 0 - 1.0 mL were added dropwise into different volumes of 5% (w/w) ES100 ranging from 0 – 1.0 mL (Table 7). The organic phase containing both 6A1 encapsulated by ES100 was emulsified in 20 mL of 2.5% PVA by microtip sonicator for 30 seconds at 10 µAmps, paused for 30 seconds in ice water, and repeated for thirty cycles. The slurry was left on gentle stirring for six hours. During the gentle stirring, the organic phase started to migrate into the greater stabilized aqueous solution and the organic solvent started to evaporate. The migration resulted in microspheres precipitated into the aqueous phase. Ultracentrifugation at 20,000 x g for ten minutes, at four degrees, was used to collect the suspended multi particulates. The collected microspheres were filtered via a paper filter and lyophilized after being washed three times with distilled water.
Table 7: Top: DoE of Nanoparticle formulations; Bottom: Leading Formulations from the DOE JMP 14 experiment.
Figure imgf000130_0001
In Vitro Dissolution Tests of microparticles packed inside coated S9C [0477] The in vitro dissolution test for the microparticles packed inside coated S9C was performed, in accordance with the recommendation from UPS compendial dissolution, non- sink conditions (168). The simulated intestinal pH was adjusted via 2.5 µmol/L of HCl or NaOH from the 2.5 µmol/L of phosphate buffer. A 1.0 mL sample was collected every half hour and the pH exposure/duration were as follows: 1.5 hours at pH 1.2 (stomach), 2.5 hours at pH 4.6 (small intestine), 2 hours at pH 6.8, 2.5 hours at pH 8, and 2.5 hour at pH 6.8. Each capsule was placed within a sinker housing and the stirrer rate was set at 80 rpm (169) at 37 ± 2°C. Samples at an extreme pH were air-dried, reconstituted, and neutralized before analysis. Drug concentration (6A1) was determined using the UPLC MS-MS method that was previously validated after the removal of ES100 via methylene chloride. In vivo pharmacokinetic study food effect on the AUC_systemic of 6A1 and time of capsule release (Cmax/Tmax) [0478] The raw 6A1 powder was packed inside coated S9C at a 20 mg/Kg dose normalized by the rat’s weight. For the single dose pharmacokinetic study, blood samples were taken at 0, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, and 24 hours after gavage. The fasted rats (n=8) were housed inside a special metabolic cage as to prevent coprophagy, food was withheld for twelve hours, and free access was given to water. After the S9C were given, food was given to the fasted group. The fed rats were fed ad libitum and given water. In vivo pharmacokinetic study of each successive coating's effect on the total systemic exposure of 6A1(AUC_systemic) and time of capsule release (Cmax/Tmax) [0479] Similar to the previous in vivo studies, the raw 6A1 powder was packed inside coated S9C at a 20 mg/Kg dose normalized by the rat’s weight. A single dose of uncoated capsule, enteric coated capsules, and enteric & delayed release coated capsules were given to F344 rats via oral gavage. The blood samples were taken at 0, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, and 24 hours after gavage. Single dose pharmacokinetic study of 6A1 microparticles formulated for sustained release using two leading microparticle formulation 1 (F1) and formulation 2 (F2) [0480] The microparticles were packed inside coated S9C at a 20 mg/Kg dose normalized by the rat’s weight. For the single dose pharmacokinetic study, the blood samples were taken at 0, 0.5, 1, 2, 3, 4, 6, 8, 10, 12, and 24 hours after gavage. A four day twice daily pharmacokinetic study was done where the rats were given, twice daily, the microparticles packed inside coated S9C at a 20 mg/Kg dose normalized by the rat’s weight. The blood samples were taken at 12, 14, 16, 18, 24, 36, 48, 60, 62, 64, 66, 68, 72, 84, and 89 hours after gavage. Multiple-Oral-Dose Regimen pharmacokinetic study of 6A1 microparticles formulated for sustained released packed inside the enteric and delayed release coated S9C [0481] The dosing schedule for the multiple oral dose regimen for the four day efficacy study were twice daily (BID) at 12 hours apart. The given dose was 20 mg/Kg normalized per rat’s weight. [0482] Highly detailed dosing information can be found in Table 8. There are two different microparticle blends; Formulation 1 (Group 1) has 88.8 mg of 6A1 per gram of materials which correlated to an encapsulation efficiency (EE) of 30%. Thus, Group 1 multiple dose regimen group needed a total of three capsules per dose and a total of six capsules per rat each day. Formulation 2 has 56.3 mg of 6A1 per gram of particles, thus equating to an EE of 45%. Group 2 needed a total of four capsules per rat per day as the doses given were 20 mg/kg BID. Table 8. Pharmacokinetic dosing schedule of multiple doses of Formulation 1 and Formulation 1 microparticles packed inside S9C (20 mg/Kg dose). Group 1 (n=6) were given formulation
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000133_0002
Figure imgf000134_0001
Statistical Analysis [0483] The blood concentration-time profiles of 6A1 for each subject were analyzed by a noncompartmental method using WinNonlin® 6.1 (Pharsight Corporation, Mountain View, CA, USA). All statistical analyses were performed using the GraphPad Prism 8.0 program (GraphPad Software Inc., San Diego, CA, USA). Tmax and Tlag, the nonparametric 95% CI was calculated for the difference in median values of each group. The dose dependence of Tmax and Tlag, rounded to the nearest scheduled time points, was investigated by the Wilcoxon rank sum test to calculate a median and the difference in medians between dose groups. Results and Discussion Formulation of 6A1 Nanoparticles [0484] Formulation development is a critical step in the process leading toward efficient, safe, and stable commercial products. Methods with high throughput capability provide a considerable advantage by expanding the conditions and parameters required. JMP 14 Software DoE facilitated the development of the pilot formulations. [0485] Fifteen pilot formulations were completed to determine the optimal ratio of 6A1- PLGA 8515-ES particles. Of the fifteen, three leading formulations: number 6, 14, and 15 met both the particle size and delayed release criteria set forth (Table 7, Figure 24, and Figure 25). Noticeably, all three formulations had a high percentage, above 75%, of the surfactant PVA. Of the three, number 6 was the leading formulation for both the in vitro dissolution and the in vivo pharmacokinetic studies. Formulation 6 had the best performance of all the nanoparticle formulations and was used as a precursor for the preparation of 6A1 microspheres. Scale up 6A1 microparticles were generously made by collaborators (Dr. Gan Yong, Shanghai Tech University, Pudong, Shanghai, China). In Vitro Dissolution Tests [0486] The drug loading (EE%) of pH sensitive microparticles was determined by dissolving the microparticles in an alkaline PBS buffer (187), and then using a liquid-liquid extraction method to remove the 6A1 compound using DMC. The in vitro, pH-dependent release profile of 6A1 from the pH-sensitive microparticles was investigated in a gradual pH- changing buffer to assess its ability to protect the content from the acidity and accomplish an effective delivery in neutral pH values (163). The 6A1 microparticles were rapidly released and emptied their content from the enteric and delayed release coated S9C. One under weighted capsule that had failed the quality assurance method was also tested. Most of the coated capsules released 6A1 microparticles at pH 6.8 (Figure 26). The result suggested the weight of a capsule was a reliable quality assurance method to qualify each coating batches. In vivo pharmacokinetic study food effect on the AUC_systemic of 6A1 and time of capsule release (Cmax/Tmax) [0487] A noncompartmental analysis of the blood concentration-time profile for food effect study was analyzed. The AUC_Ave24hour of seven rats during fed and fasted states were calculated using the trapezoidal rule. Fed animals had an AUC_Ave24hour of 6.5 ± 1.02 µg/mL compared to 10.6 ± 1.5 µg/mL during a fasted state. The Tmax of fasted animals was delayed by an hour compared to that of fasted animals, two hours post-dose (Figure 27). Statistical student t test with Welch’s correction indicated that the AUC_Ave24hour between the two group are insignificant (P-value = 0.6445). When compared with the Tmax between groups, the result was different from the conventional understanding of gastric emptying of food in rats, which was influenced by the amount and type of food given. A study showed that a fatty meal delays the gastric emptying of liquids in awake rats, whereas fasting sped up the emptying process (188-190). However, capsule emptying was likely independent of the food effect. The result of fasted animals emptying capsules slower than a fed animal was also confirmed by a study done by Saphier et al., 2010 (171). However, the Hodges–Lehmann nonparametric population’s parameter indicated that there was a significant difference in the Tmax value (p = 0.0253). Simulation of prophylactic treatment in humans required that further pharmacokinetic studies be done with fed ad libitum animals. Multiple t test analyses indicated significant Cmax/Tmax differences between the first 2 hours; understandably, because absorption in the upper GI was influenced by the acidity and food content of the stomach. Pharmacokinetic Study of Each Coating's Effect on the Total Systemic Exposure of 6A1 [0488] The dose normalized AUC_Ave24hour of eight subjects of uncoated, enteric coated, and enteric and delayed release coated capsules were calculated using the trapezoidal rule. The uncoated capsule has AUC_Ave24hour of 3.388 ± 0.6967 µg. The results are consistent with the previous pharmacokinetic study of food effect in uncoated capsules. The AUC_Ave24hour of enteric-coated capsules was 2.567 ± 0.48 µg. The enteric coating layer reduced total systemic exposure to 60.6% of uncoated capsules. The AUC_Ave24hour of enteric and delayed release coated capsules had the smallest amount, 1.009 ± 0.1317 µg (Figure 28). The results indicated that the total systemic drug exposure had been reduced by 5.25 folds via the capsule’s enteric and delayed release mechanism compared to uncoated capsules (P < 0.0212). The uncoated capsule had a Tmax at one-hour post-dose, with the enteric coat, the capsules’ Tmax shifted to two hours post-dose. Compared to the two previous, the enteric delayed release coated capsules’ Tmax shifted noticeably to five hours post-dose (P< 0.0001). Single dose pharmacokinetic study of 6A1 microparticles formulated for sustained release using two leading microparticle Formulation 1 (F1) and Formulation 2 (F2) [0489] Again, the AUC_Ave24hour of the twelve subjects in the microparticles at the dose of 5 mg/kg was calculated using the trapezoidal rule. The AUC_Ave24hour of 6A1, 6A1 glucuronide, and 6A1 sulfate were 1.43 ± 0.04 µg/ml, 0.84 ± 0.25 µg/mL, and 0.137 ± .06 µg/mL. The above pharmacokinetic parameters equaled to a dose normalized AUC_Ave24hour of 0.7130 µM/Hr for F1 and 0.6649 µM/Hr for F2. The AUC_Ave24hour exhibited the characteristics of a sustained release with the onset of both formulation at around three hours and the Cmax 0.1854 for F1 and 0.1404 for F2. Noticeably, the Cmax/Tmax of 6A1 glucuronide (green curve) differed significantly from that of 6A1 (red curve) – 198 ng/mL at six hours versus 305.5 ng/mL at four hours. The pharmacokinetic parameters above suggested microparticles were absorbed, metabolized, and entered the EHR (Figure 29). Interestingly, in a dose normalized analysis using the unpaired student t test, the results indicated that there was no significant difference between the two formulation F1 and F2 (P = 0.8061). Multiple-Oral-Dose Regimen pharmacokinetic study of 6A1 microparticles formulated for sustained released packed inside the enteric and delayed release coated S9C [0490] Familial adenomatous polyposis (FAP) is a chronic disease; multiple dosage regimens with appropriate drug concentration assist with regressing the onset of inflammation and polyposis formation. A short term (4 day) efficacy study of enterically coated microparticulates packed inside coated S9C attempted to assess the therapeutic drug concentration in the colon and ability to deliver a constant, sustained amount of drug (Cmax_ss) to the colon while reducing the total systemic exposure (AUC_average_ss). The mean plasma concentration-time profiles after a q12h oral administration of 20 mg/kg of 6A1 and of its metabolite, 6A1 glucuronide and sulfates, were displayed in Figure 30. Other pharmacokinetic parameters can also be deduced from the experiment, such as 6A1 concentration at a steady state. NCA of Phoenix WinNonlin® 6.1 software (Pharsight Corporation, Mountain View, CA, USA) was used to determine the clearance rate of 6A1 at a steady state was (CLss_F) 107,222.7 mL/hr/kg. There was no significant difference in clearance rate between the two formulations (P > 0.1). Interestingly, Formulation 1 has a Css of 1,536.65 ng/mL compared to the 2,444.07 ng/mL for Formulation 2; thus, the colon tissue-specific of 6A1 in Formulation 1 was 37% less than that of Formulation 2 (1,536.6 ng/g compared to 2,444.7 ng/g, P < 0.012). The P values of 6A1 metabolites (glucuronide and sulfate) in the colonic tissue between groups was insignificant (Figure 31). It was unclear whether the size differences of the particles in Formulation 2 or the larger number of capsules given contributed to such differences. The colon tissue concentration further confirmed that the capsules delivered a sufficient amount of drug to the colon. [0491] There was a significant difference between the amount of 6A1 and 6A1 sulfates accumulated in the liver tissues between the two groups (Figure 31). No appreciable accumulation was detected for either 6A1 or its metabolites when comparing between the single dose and multiple dosing assuming Css at 4 days in the F1 microparticles (AUC_Dose Normalized : 0.5531 vs 0.5232 µM ). However, for 6A1 single dose and multiple dosing of F2 microparticle, there was a significant increase in the steady stage concentration (AUC_Dose Normalized : 0.6868 vs 0.9532 µM ). [0492] In conclusion, this study achieved a de novo design of capsule coatings and formulations that delivered a higher concentration of drug to the colon and reduced the systemic drug concentration multifold. Additionally, the long-term efficacy study using the above approach to compare the 6A1 concentration systemically versus the colon targeted at steady state was in progress to elucidate the pharmacodynamics of 6A1. Glucuronidation was the main driving force for of 6A1 entering into EHR; sulfation has little involvement in the phase two metabolism of 6A1. The completion of these studies using the F344 model will help establish the pharmacokinetic mechanism of such a drug delivery system. Further investigation of the 6A1 pharmacodynamic mechanism is needed to successfully translate the application into clinical care for FAP patients as preventive care. Regardless, there is still no commercial drug for the treatment and prevention of polyposis (FAP) and CRC. Reformulation is crucial to lower systemic toxicity using hard gelatin-coated capsules for the treatment and prevention of FAP and CRC. Therefore, research for the targeted delivery of 6A1 and new delivery strategies will provide a therapeutic benefit to many chronically suffering patients. [0493] The in vivo data suggested that these approaches are the most promising. A combined approach of both pH dependent and time-dependent particulate systems is highly desirable for use in a true colon-specific drug delivery system. The combined approach may reduce the side effects of the drug caused by absorption from the stomach when given in the form of raw drugs. [0494] All analyses were conducted using actual sampling times. The blood plasma concentration (Cmax) and time at Cmax (Tmax) were determined from the observed values and confirmed with GraphPad Prism software. The trapezoidal rule was used to calculate the AUCtotal from time 0 to the last measurable time. Unfortunately, lack of a powerful model to address the recycling of 6A1 resulted in the analysis not yielding reliable pharmacokinetic parameters and the need for more empirical data. Overall, the pharmacokinetic studies revealed that the relative bioavailability of 6A1 was lower and the colonic tissue exposure higher when administered in the microparticles packed inside the S9C compared to the conventional oral suspension and/or intravenous route. Further analyses also revealed that relative bioavailability and exposure to 6A1 is higher when administered to subjects in the fast condition than subjects in the fed condition. Conclusions [0495] The described formulation comprises a blend of solvent mixtures. The solvent system is composed of acetone, isopropanol alcohol (IPA), and water; it provides for an optimal drying time without a lengthy drying period during processing between each successive coating. However, the solvent system did not evaporate as abruptly as 100% acetone, which has caused the film formation to be uneven and blister. The high mass loading of the polymer (10%) prevents the solution from having a running effect. Frequently, a high ratio of material to solvent and large molecular weight in a formula contributes to a high-viscosity. High viscosity (a thick formulation) yields better coat quality as compared to a low viscosity formulation. The intermolecular forces of a higher viscosity formulation prevents the solution from forming a film that is not subjected to shrinking stress during drying and the resulting breakage. In a manufacturing setting, a thinner solution with low viscosity is often used as it enhances adhesion to the substrate; however, the downside is the need for multiple coatings. The basic water spray applied to the capsule surface after sealing and before coating neutralizes the capsule's surface lysine, destabilizing their surface chemistry, and thus promotes better adherent for the polymer and preventing peel-off. [0496] A robust, sensitive, and validated UPLC-MS/MS method was successfully used to quantify 6A1, 6A1 glucuronide, and 6A1 sulfate in pharmacokinetic blood, colonic mucosa, and liver tissue samples. The method has a multitude of advantages, such as high sensitivity (LLOQ 2 ng/mL), short analysis time (6 minutes), small sample size (10 µL), functional recovery with negligible matrix effect, and simple sample preparation. With all the above- mentioned advantages, this method was applied to preclinical pharmacokinetic/pharmacodynamic modeling studies of 6A1. [0497] Fifteen pilot formulations with the help of JMP DoE software were able to produce a leading polymeric pH-sensitive PLGA 8515/ES100 particle loaded with the 6A1 formulation. The microparticle formulation has a theoretical encapsulation efficiency (EE) of 30-45% after redissolving the polymer in methylene chloride to extract the 6A1. [0498] The in vitro pH-dependent release profile of 6A1 microparticles showed a rapid release of drug in a neutral pH environment and the in vivo pharmacokinetic study showed an extended release profile, with the Tmax at 3 hours. The formulation was successfully scaled-up, after synthesis, via collaboration and later used in the four days multiple dosing regimens. Example 4. Pharmacokinetic Studies of 6A1 in F344 Rats [0499] Rats were fed ad libitum and the 6A1 active administered by oral gavage. Different study conditions were investigated, such as food effect, real-time imaging agent mixed with 6A1, and uncoated capsule versus enteric or erosion coated capsules. Food is a confounding factor of capsules emptying from the stomach. Fasted animals have a higher Tmax/Cmax and AUCAverage24hourtotal compared to their fed cohort. [0500] Each additional coating applied to the capsules reduced the total systemic exposure of 6A1. Calculated using the trapezoidal rule, the uncoated, enteric coated, and enteric and delayed release coated capsules had an AUC_Ave24hour of 6.55 ± 1.02 µg/mL.3.97 ± 0.48 µg/mL, and 1.24 ± .50 µg/ml. Uncoated capsules AUC_total was comparable to the fed stage of the food study experiment. The results also indicated a 5.25 fold reduction of AUC administered via capsule enteric and a delayed release mechanism (P < 0.0212). The Tmax was also significant between the three groups. Uncoated capsules had a Tmax at one-hour post-dose. The enteric coated capsules delayed the Tmax to two hours post-dose. The combination of PLGA8515 and ES100 coating delayed the Tmax to five hours post-dose compared to uncoated capsules (P< 0.0001). [0501] The enteric and delayed release formulation administered via oral gavage seems to closely simulate a prophylactic treatment with the highest colon tissue accumulation. In a parallel study, data indicated that blood level Css could be reached after four days of medication at 40 mg/Kg twice a day (180). Accordingly, this research included a short term (four days) efficacy study with coated capsules comprising the leading microparticles. The colonic concentration of 6A1 was comparable between the parallel study of 40 mg/Kg twice daily (3300 ± 300 ng/g) to that of the four-day multiple dosing of coated capsules at 20 mg/Kg (2,682 ng/g for Formulation 1 and 1,916 ng/g for Formulation 2). Since the coated capsule's dosage was reduced by half, the result strongly suggested that lowering the dose could achieve similar colonic concentration. Table 9: A dose normalized comparison of colonic tissue concentration of 6A1 across IV, oral suspension, size 9 capsule (SC9) packing with powder, and microparticles.
Figure imgf000140_0001
[0502] Glucuronidation levels in the multidose four-day study are the main driving force for 6A1 entering EHR; sulfation has little involvement in the phase two metabolism of 6A1. This conclusion was evidenced by the finding that there is no significant difference (P > 0.05) between the two groups when the glucuronide and sulfate concentrations in liver tissues are compared. For colonic mucosa tissues, the amount of 6A1 accumulation in Formulation 1, which has a smaller microparticulate, was significantly higher than that of Formulation 2. The particle size difference might have contributed to the observed data. The P value of glucuronide concentration between the two groups was insignificant (P > 0.05). One can infer that the differences between the two groups were at least in part due to the particle sizes and not the activity of microbiomes or the delivery method. Microbiomes can be ruled out as the causative factor since the glucuronide generated in the liver was secular and the rat groups are cage mates given the opportunity to practice coprophagy, as rats are known to do. Also, the delivery system could not have been the causative factor for the P value variance since the AUC are similar between two groups. [0503] Example 5. Doped Polymer Formulations Formulation of polymers [0504] In the present disclosure, two biodegradable polymers are used for the coating of the capsule. [0505] PLGA is a linear copolymer that can be prepared at different ratios between its constituent monomers, lactic (LA) and glycolic acid (GA):
Figure imgf000141_0001
[0506] Depending on the ratio of lactide to glycolide used for the polymerization, different forms of PLGA can be obtained: these are usually identified in regard to the monomers’ ratio used (i.e., PLGA 85:15 identifies a copolymer consisted of 85% lactic acid and 15% glycolic acid). [0507] Eudragit S 100 (C8H12O4) is an anionic copolymer based on methacrylic acid and methyl methacrylate (poly(methacylic acid-co-methyl methacrylate) 1:2). It was generously donated by Evonik. [0508] The solid polylactic glycolic acid (PLGA8515) is stored at -20°C, and the powdered methyl methacrylate acid (Eudragit S100) is stored at 4°C. PLGA8515 is purchased from Direct Corporation, Birmingham Division (2200 Riverchase Center, Suite 501. Birmingham, AL USA 35244). [0509] The solvent for PLGA8515 is methylene chloride. The solvent is used to dissolve 1.5 g of PLGA8515. The PLGA8515 is added slowly to a glass dram vial with 5 mL of premeasured methylene chloride. The mixture is stirred at 60 RPM and another 10 mL of methylene chloride gradually added (in 1 mL dropwise increments) until the final volume is 15 mL. After 30 minutes of stirring, the solution will be visually clear. The final concentration is 10% w/v solution. The PLGA8515 polymer is fractioned into 1.5 mL glass vials, sealed with parafilm, and stored at -20°C. Historical qualification steps (TGA/DSC) indicate that the PLGA8515 polymer is stable when stored under these conditions for up to 2 weeks. [0510] The solvent for Eudragit S100 is a mixture of acetone: isopropanol alcohol: DI water in the ratio of 86:12:2. The solvent is used to dissolve 1 g of Eudragit S100. The Eudragit S100 powder is slowly added into 5 mL of solvent in a dram vial and stirred at 60 RPM; the powder is incrementally added allowing for the solution to become clear until all powder is added. The Eudragit S100 will be visually clear after 30 minutes; if not, 5 µL of 100 micromolar NaOH solution at pH 11 is added and stirred for another 15 minutes. If the solution becomes transparent, an additional 5 mL of solvent is added to the solution until the final volume is 10% w/v. If the solution turns turbid, another 5 µL of 100 micromolar NaOH solution at pH 11 is added and stirred for another 15 minutes. The Eudragit S100 is fractionated into 1.5 mL vials, the vials wrapped with parafilm, and stored at 4°C. Historical qualification steps (TGA/DSC) indicate polymer is stable in such conditions for up to seven days. [0511] Both solutions require quantification with TGA and DSC before use. The solutions should be discarded after each use, or when left standing at room temperature for more than 2 hours. Doping of PLGA8515 Polymers [0512] Doping is a process in which a contaminant or doping agent (dopant) is purposely added into a formulation to form a controllable pore size and pore density. In other words, a dopant is introduced as an artifact. Degradation of the acid terminals of the polymer causes the medium, its microenvironment, and the surrounding tissue's pH to decrease. Therefore, an alkaline salt or neutral salt often is used to stabilize acid terminal polymers and an acidic salt to stabilize basic terminal polymers. It is well established that the porosity of a polymer depends on both the polymer fabrication techniques and the concentration of salts. [0513] CaCO3 and NaHCO3 are known dopants used to enhance degradation in a PLGA50 matrix. An alkaline dopant promotes faster polymer degradation without compromising the polymer's integrity (i.e., there is no actual chemical reaction between the porogen and the polymer network). [0514] To accelerate and shorten the degradation of PLGA8515 from days into hours and precisely control its capacity to deliver a drug to the large intestine, many salts were investigated. For example, sodium citrate at 5% w/w as a doping agent creates a pore size of 180-250 µm in the PLGA network. Sodium chloride at a temperature of from room temperature to 400°C consistently forms a pore size of 17-20 nm in diameter. [0515] The challenge in this formulation of the present disclosure is the ability to dissolve sodium chloride in methylene chloride. Because both sodium chloride and methylene chloride are polar molecules they not necessarily compatible due. [0516] Sodium chloride was first dissolved in methanol at its saturated point of 14 g per 1000 g. The sodium chloride working solution (11.074µg/µL) is added to a premade PLGA polymer that is dissolved in methylene chloride. [0517] The best working sodium chloride salt concentration as dopant found was 2.5% v/v to 10% v/v of the final solution (37.5 µL NaCl/methanol into 1462.5 µL of pre-dissolved PLGA8515/methylene chloride polymers to 75 µL NaCl/methanol into 1425.0 µL of pre- dissolved PLGA8515/methylene chloride). The percentage of the dry mass ratio of dopant to polymer can range from 0.03% to 0.07%. [0518] Examples of alkaline dopants porogens include, without limitation sodium percarbonate (Na2H3CO6), sodium bicarbonate (NaHCO3), sodium carbonate (NaCO3), and sodium acetate (NaCH3CO3). [0519] Other porogens include, without limitation, saccharin, lipoprotein, and small molecule fat.
Figure imgf000144_0001
Figure imgf000145_0001
[0520] While the present invention has been described with reference to the specific embodiments thereof it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adopt a particular situation, material, composition of matter, process, process step or steps, to the objective spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

Claims

CLAIMS What is claimed is: 1. An oral pharmaceutical composition comprising: (a) a gelatin capsule containing: (i) an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a therapeutically effective amount of a therapeutic agent disposed within the capsule.
2. An oral pharmaceutical composition comprising: (a) a capsule containing: (i) an inner delayed-release coating comprising a doped biodegradable polymer applied to the capsule to a predefined thickness, and (ii) an outer enteric coating comprising a pH-sensitive polymer applied to the capsule to a predefined thickness; and (b) a particulate formulation disposed within the capsule, comprising: (i) a plurality of particles each comprising a therapeutically effective amount of a therapeutic agent, and (ii) one or more polymers encapsulating the therapeutic agent and which releases the therapeutic agent at a pH above 6.0.
3. The oral pharmaceutical composition of claim 1 or claim 2, the therapeutic agent comprising a structure as defined by any one of Formulas 1-10 or a pharmaceutically acceptable salt thereof.
4. The oral pharmaceutical composition of claim 1 or claim 2, wherein the therapeutic agent is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) (Figure 1C) or a pharmaceutically acceptable salt thereof.
5. The oral pharmaceutical composition of claim 1 or claim 2, wherein the biodegradable polymer is a poly(lactide-co-glycolide) (PLGA) selected from the group consisting of 95:5 poly(lactide-co-glycolide) 85:15 poly(lactide-co-glycolide), 75:25 poly(lactide-co-glycolide), 65:35 poly(lactide-co-glycolide), or 50:50 poly(lactide-co- glycolide).
6. The oral pharmaceutical composition of claim 1 or 2, wherein the biodegradable polymer is 85:15 poly(lactide-co-glycolide) (PLGA8515).
7. The oral pharmaceutical composition of claim 6, wherein the biodegradable polymer is applied to the capsule to a predefined thickness of 115 ± 35 nm.
8. The oral pharmaceutical composition of claim 1 or 2, wherein the inner delayed- release coating further comprises a plurality of pores to control release of the therapeutic agent.
9. The oral pharmaceutical composition of claim 1 or 2, wherein the pH-sensitive polymer is Eudragit S100.
10. The oral pharmaceutical composition of claim 6, wherein the pH-sensitive polymer is applied to the capsule to a predefined thickness of 50 ± 15 nm.
11. The oral pharmaceutical composition of claim 2, wherein size of the plurality of particles is about 100 nm to about 2000 nm, inclusive.
12. The oral pharmaceutical composition of claim 2, wherein the one or more polymers comprises 50:50 poly(lactide-co-glycolide) (PLGA5050), Eudragit S100, and/or poly(vinyl alcohol) (PVA).
13. The oral pharmaceutical composition of claim 1 or 2, wherein when the composition is orally administered to a subject there is a lag period of at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, or at least about 12 hours before sustained release of the therapeutic agent.
14. The oral pharmaceutical composition of claim 1 or 2, wherein when the composition is orally administered to a subject, sustained release of the therapeutic agent in the colon occurs at a pH of about 6.8.
15. The oral pharmaceutical composition of claim 1 or 2, wherein the composition (i) reduces therapeutic dose of a therapeutic agent, and/or (ii) reduces total systemic exposure (AUC) of a therapeutic agent, and/or (iii) increases local colonic exposure (AUCcolon) of a therapeutic agent, and/or (iv) increases concentration of the therapeutic in the colon by lowering AUC0-24Hours, and increasing local AUCcolon, and/or (v) utilizes enterohepatic recycling (EHR), and/or (vi) reduces occurrence of on target off organ systemic toxicity associated with administration of the therapeutic agent.
16. The oral pharmaceutical composition according to claim 1 or claim 2, wherein the dopant is sodium chloride, sodium percarbonate, sodium bicarbonate, sodium carbonate, or sodium acetate.
17. The oral pharmaceutical composition according to claim 16, wherein the dopant configures the polymer to increase its porosity.
18. The oral pharmaceutical composition according to claim 2, wherein the microparticle formulation has a theoretical encapsulation efficiency (EE) of 30-45%.
19. The oral pharmaceutical composition according to claim 2, wherein a release profile of the particles in vitro is pH dependent.
20. The oral pharmaceutical composition according to claim 19, wherein the in vitro pH- dependent release profile of 6A1 microparticles comprises a rapid release of the active in a neutral pH environment.
21. The oral pharmaceutical composition according to claim 22, wherein the composition comprises an extended release profile.
22. The oral pharmaceutical composition according to claim 21, wherein Tmax is 3 hours.
23 A method of spin coating a capsule, comprising: (i) providing a vacuum spinning plate comprising individual pods; (ii) providing sealed capsules wetted with a mild basic water (e.g., pH 9 NaOH 0.1 mM) loaded into the individual pods within the vacuum spinning plates; (iii) providing an amount of a first polymer dissolved in a solvent; (iv) applying a vacuum; (v) spinning the plate a first time for about 30 seconds at a spinning speed of 100 RPM; (vi) spinning the plate a second time for about 15 minutes at spinning speed of 500 RPM; (vii) providing an amount of a second polymer dissolved in a solvent; (viii) applying a vacuum; (ix) spinning the plate a third time for about 30 seconds at spinning speed of 100 RPM; (x) spinning the plate a fourth time for about 15 minutes at a spinning speed of 500 RPM; (xi) removing the capsule from the spinning plate; and (xii) drying the capsule in a desiccator.
24. The method of claim 23, wherein the method (i) achieves a polymer mass loading of at least about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 10.5%, about 11%, about 11.5%, about 12%, about 12.5%, about 13%, about 13.5%, about 14%, about 14.5%, or about 15%; and/or (ii) achieves a coating comprising a smoothness and uniformity of surface appearance; and/or (iii) achieves a predetermined thickness measurement; and/or (iv) achieves in vitro dissolution; and/or (v) achieves a drying time of less than about 1 hour, less than about 30 minutes, less than about 15 minutes, less than about 10 minutes, or less than about 5 minutes.
25. A method for reducing severity or incidence of a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing the pharmaceutical composition according to claim 1 or 2, wherein the therapeutic agent is a selective COX-2 inhibitor, and (b) administering an effective amount of the pharmaceutical composition orally to the subject.
26. A method for treating a colorectal cancer in a subject having familial adenomatous polyposis (FAP) comprising: (a) providing the pharmaceutical composition according to claim 1 or 2, wherein the therapeutic agent is a selective COX-2 inhibitor; and (b) administering an effective amount of the pharmaceutical composition orally to the subject.
27. The method of claim 25 or claim 26, wherein the therapeutic agent that is a selective COX-2 inhibitor is 6A1 (4-[3-(2-hydroxy-phenoxymethyl)-5-p-tolyl-pyrazol-1-yl-] benzenesulfonamide) or a pharmaceutically acceptable salt thereof.
PCT/US2022/036519 2021-07-09 2022-07-08 Enteric and delayed release formulations of locally bioavailable drugs (lbd) targeting the colon WO2023283427A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163220384P 2021-07-09 2021-07-09
US63/220,384 2021-07-09

Publications (2)

Publication Number Publication Date
WO2023283427A2 true WO2023283427A2 (en) 2023-01-12
WO2023283427A3 WO2023283427A3 (en) 2024-04-04

Family

ID=84801000

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/036519 WO2023283427A2 (en) 2021-07-09 2022-07-08 Enteric and delayed release formulations of locally bioavailable drugs (lbd) targeting the colon

Country Status (1)

Country Link
WO (1) WO2023283427A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117045607A (en) * 2023-07-13 2023-11-14 广东医科大学 Preparation method and application of bionic tea polyphenol nano-particles

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5686105A (en) * 1993-10-19 1997-11-11 The Procter & Gamble Company Pharmaceutical dosage form with multiple enteric polymer coatings for colonic delivery
US9486467B2 (en) * 2006-06-12 2016-11-08 Ramot At Tel-Aviv University Ltd. Method of treating colorectal cancer that expresses a mutated APC gene by administering erythromycin or tylosin
US8124123B2 (en) * 2007-09-05 2012-02-28 Dow Pharmaceutical Sciences, Inc. Controlled release azithromycin solid dosages forms
WO2016172769A1 (en) * 2015-04-29 2016-11-03 University Of South Australia Compositions and methods for administering antibodies
WO2020236562A1 (en) * 2019-05-17 2020-11-26 Cancer Prevention Pharmaceuticals, Inc. Methods for treating familial adenomatous polyposis

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117045607A (en) * 2023-07-13 2023-11-14 广东医科大学 Preparation method and application of bionic tea polyphenol nano-particles
CN117045607B (en) * 2023-07-13 2024-05-31 广东医科大学 Preparation method and application of bionic tea polyphenol nano-particles

Also Published As

Publication number Publication date
WO2023283427A3 (en) 2024-04-04

Similar Documents

Publication Publication Date Title
EP3280448B1 (en) Abiraterone acetate lipid formulations
Panakanti et al. Impact of excipient interactions on drug bioavailability from solid dosage forms
EP3368014B1 (en) Novel cannabinoid formulations
Liu et al. In vitro and in vivo studies on mucoadhesive microspheres of amoxicillin
Nishimura et al. In vitro and in vivo characteristics of prochlorperazine oral disintegrating film
EP0553178B1 (en) Biphasic release formulations for lipophilic drugs
KR101434334B1 (en) Micellar nanoparticles of chemical substances
JP7269227B2 (en) Niraparib formulation
Yadav et al. Dual coating of swellable and rupturable polymers on Glipizide loaded MCC pellets for pulsatile delivery: Formulation design and in vitro evaluation
Katsuma et al. Effects of absorption promoters on insulin absorption through colon-targeted delivery
Khan et al. Tacrolimus-loaded nanostructured lipid carriers for oral delivery-in vivo bioavailability enhancement
WO2019082171A1 (en) Oral cannabinoid compositions with improved bioavailability
Jain et al. Lectin conjugated gastroretentive multiparticulate delivery system of clarithromycin for the effective treatment of Helicobacter pylori
CN102596252A (en) Gastric retentive pharmaceutical compositions for immediate and extended release of acetaminophen
WO2009130204A2 (en) Improved formulations for poorly permeable active pharmaceutical ingredients
JP2003520788A (en) Compositions for delivery of cortisol antagonists
EA035515B1 (en) Deuterated domperidone compositions and methods for therapy of disorders
Park et al. Colon-targeted delivery of solubilized bisacodyl by doubly enteric-coated multiple-unit tablet
WO2023283427A2 (en) Enteric and delayed release formulations of locally bioavailable drugs (lbd) targeting the colon
JPH11502842A (en) Azathioprine compositions for colon administration
Murakami et al. Factors and dosage formulations affecting the solubility and bioavailability of P-glycoprotein substrate drugs
JP2010502710A (en) Liquid pharmaceutical formulation for oral administration of CGRP antagonist
Yoshida et al. Oral drug delivery systems applied to launched products: value for the patients and industrial considerations
Meola et al. The effect of drug ionization on lipid-based formulations for the oral delivery of anti-psychotics
Shatzer Pharmacokinetic Studies of Enteric and Delayed Release Formulations of Locally Bioavailable Drugs (LBD) Targeting the Colon

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22838470

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE