throbber
Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`Contents lists available at ScienceDirect
`
`Advanced Drug Delivery Reviews
`
`j o u r na l h om e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / a d d r
`
`Oral delivery of macromolecular drugs: Where we are after almost
`100 years of attempts☆
`Elena Moroz 1, Simon Matoori 1, Jean-Christophe Leroux ⁎
`
`ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 15 November 2015
`Received in revised form 11 January 2016
`Accepted 18 January 2016
`Available online 27 January 2016
`
`Keywords:
`Oral drug delivery
`Oral bioavailability
`Permeation enhancers
`Protease inhibitors
`High molecular weight
`Biologics
`
`Contents
`
`Since the first attempt to administer insulin orally in humans more than 90 years ago, the oral delivery of
`macromolecular drugs (N1000 g/mol) has been rather disappointing. Although several clinical pilot studies
`have demonstrated that the oral absorption of macromolecules is possible, the bioavailability remains generally
`low and variable. This article reviews the formulations and biopharmaceutical aspects of orally administered
`biomacromolecules on the market and in clinical development for local and systemic delivery. The most
`successful approaches for systemic delivery often involve a combination of enteric coating, protease inhibitors
`and permeation enhancers in relatively high amounts. However, some of these excipients have induced local
`or systemic adverse reactions in preclinical and clinical studies, and long-term studies are often missing.
`Therefore, strategies aimed at increasing the oral absorption of macromolecular drugs should carefully take
`into account the benefit–risk ratio. In the absence of specific uptake pathways, small and potent peptides that
`are resistant to degradation and that present a large therapeutic window certainly represent the best candidates
`for systemic absorption. While we acknowledge the need for systemically delivering biomacromolecules, it is our
`opinion that the oral delivery to local gastrointestinal targets is currently more promising because of their
`accessibility and the lacking requirement for intestinal permeability enhancement.
`© 2016 Elsevier B.V. All rights reserved.
`
`1.
`2.
`
`3.
`
`.
`.
`
`2.2.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Local delivery
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.1.
`Peptides
`Vancomycin and fidaxomicin .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.1.1.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Linaclotide .
`.
`.
`.
`.
`.
`.
`.
`.
`2.1.2.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Plecanatide and derivatives .
`.
`2.1.3.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Antibodies
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.2.1.
`Antibody delivery .
`.
`.
`.
`.
`.
`Genetically modified antibody-secreting bacteria .
`2.2.2.
`Oral enzyme therapy .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.3.
`Cytoprotective/anti-inflammatory proteins .
`.
`.
`.
`.
`.
`.
`.
`2.4.
`Nucleic acids
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.5.
`.
`Systemic delivery .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.
`Peptides
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.1.
`Octreotide
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.2.
`Desmopressin .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.3.
`Cyclosporine
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`109
`109
`109
`109
`110
`110
`110
`110
`111
`112
`112
`112
`113
`113
`113
`113
`114
`
`Abbreviations: ADH, anti-diuretic hormone; AON, antisense oligonucleotide; 5-CNAC, N-(5-chlorosalicyloyl)-8-aminocaprylic acid; EDTA, ethylenediaminetetraacetate; FDA, Food and
`Drug Administration; GC C, guanylyl cyclase C; GI, gastrointestinal; GLP-1, glucagon-like peptide 1; IBD, inflammatory bowel diseases; ICAM-1, intercellular adhesion molecule 1; MG, my-
`asthenia gravis; NDA, New Drug Application; SNAC, N-(8-[2-hydroxybenzoyl]amino)caprylic acid; tkRNAi, transkingdom RNA interference platform.
`☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Understanding the challenges of beyond-rule-of-5 compounds”
`⁎ Corresponding author at: ETH Zurich, HCI H 301, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland. Tel.: +41 44 633 73 10; fax: +41 44 633 13 14.
`E-mail address: jleroux@ethz.ch (J.-C. Leroux).
`1 These authors contributed equally to this manuscript.
`
`http://dx.doi.org/10.1016/j.addr.2016.01.010
`0169-409X/© 2016 Elsevier B.V. All rights reserved.
`
`Grün. Exhibit 1097
`Grünenthal v. Antecip
`PGR2017-00022
`
`

`

`3.2.
`
`4.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Acyline
`3.1.4.
`.
`.
`.
`.
`.
`.
`.
`.
`Calcitonin .
`3.1.5.
`.
`.
`.
`.
`.
`.
`.
`.
`Semaglutide
`3.1.6.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Proteins
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Insulin .
`.
`3.2.1.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Nucleic acids
`.
`.
`.
`3.3.
`Safety of bioavailability-increasing excipients .
`4.1.
`Protease inhibitors
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.2.
`Permeation enhancers .
`.
`.
`.
`.
`.
`.
`.
`Conclusions .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.
`Acknowledgments
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`E. Moroz et al. / Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`109
`
`114
`115
`115
`116
`116
`116
`117
`117
`117
`118
`118
`118
`
`1. Introduction
`
`The first attempt to deliver insulin orally in humans was undertaken
`as early as 1922, only 1 year after the discovery of insulin by Drs. Fred-
`erick Banting and Charles Best [1], when increasing doses of insulin
`were given orally to a single diabetes patient. The results were negative,
`and already in this first study, the critical challenges of oral protein de-
`livery became apparent: poor and variable absorption, and low efficacy
`compared with subcutaneous injection. Although the interest and ef-
`forts in the oral delivery of biomacromolecules have intensified over
`the past two decades, safely and effectively delivering high-molecular-
`weight substrates via the oral route remains highly challenging for for-
`mulation scientists [2,3].
`The gastrointestinal (GI) tract is a hostile environment for biomacro-
`molecules because it is evolutionarily optimized to break down nutri-
`ents and deactivate pathogens. The highly acidic pH in the stomach
`results in the protonation of proteins and their unfolding, which ex-
`poses more motifs that are recognized by protein-degrading enzymes
`[4]. The enzymes in the stomach (pepsin) and small intestine (e.g.,
`chymotrypsin, amino- and carboxypeptidases, RNases and DNases)
`cleave proteins and nucleic acids into smaller fragments and single
`units [4]. In the colon, enzymatic fermentation processes further de-
`grade biomacromolecules [4]. Because therapeutically active biomacro-
`molecules are equally affected by these processes, the fraction surviving
`these degradation processes is generally low and variable, especially in
`the presence of food [5]. The macromolecular drug needs to overcome
`multiple barriers designed to prevent the entry of dietary and bacterial
`antigens in order to reach the systemic compartment. To access the ep-
`ithelial cell layer, the biomacromolecule firstly needs to diffuse through
`the mucus layer covering the intestinal epithelium [5]. The latter is an-
`other important barrier, as the tight junctions which seal the epithelial
`cells restrict the paracellular transport (i.e., the passage between cells)
`to small molecules and ions (b600 Da) [6]. The transcytotic pathway
`(i.e., the passage across the cell in an endocytotic vesicle) is mediated
`by luminally expressed endocytotic receptors (e.g., vitamin B12 recep-
`tor, transferrin receptor), and therefore necessitates conjugation to the
`respective ligands in order to be exploited in drug delivery [7]. Another
`access point to the systemic compartment is the phagocytotic M-cells of
`Peyer's patches which sample luminal antigens and can take up partic-
`ular substrates in the low micrometer range [8]. However, the propor-
`tion of M-cells in the gut epithelium is small and varies greatly
`between species, which complicates predictions of absorption in
`humans based on animal data [9].
`Not surprisingly, only six biomacromolecules have been approved
`by the Food and Drug Administration (FDA) for oral delivery: two local-
`ly and two systemically delivered peptides, one locally delivered non-
`peptidic macrocycle, and one locally delivered protein mixture. How-
`ever, several orally applied formulations of proteins, peptides, and
`nucleic acids are currently under clinical evaluation. Often, these formu-
`lations contain at least one of the following excipients (Fig. 1): an enter-
`ic coating and/or protease inhibitors to prevent drug degradation and
`
`permeation enhancers to enable paracellular transport of macromole-
`cules [10,11]. Mechanistically, absorption enhancement can be achieved
`by mechanically disrupting tight junctions or the plasma membrane,
`lowering mucus viscosity, and modulating tight junction-regulating sig-
`naling pathways [2]. Additional strategies for the oral delivery of
`biomacromolecules under clinical development include buccal delivery,
`utilizing carrier-mediated transcytosis, and local delivery to GI targets.
`The overwhelming majority of currently approved oral drugs and
`clinical candidates exhibit a molecular weight of b1000 Da [12]. Above
`this threshold, low bioavailability, inter- and intraindividual variability,
`food effects, and long-term safety concerns of bioavailability-enhancing
`excipients remain important challenges of oral delivery despite clear
`advances in knowledge after nearly 90 years of trial and error. In this re-
`view, we address orally applied biomacromolecular therapeutics
`(N1000 Da) already marketed or under clinical investigation for local
`or systemic delivery with an emphasis on the drug formulations and
`the biopharmaceutical aspects. The oral delivery of vaccines will not
`be covered in this manuscript, and the readers are referred to other re-
`cent reviews for more information on this topic [13–16].
`
`2. Local delivery
`
`Macromolecular drugs that act on GI targets increasingly move into
`focus because local delivery avoids the challenges of reaching the sys-
`temic compartments. Advantages of locally delivering high-molecular-
`weight drugs include fewer restrictions regarding drug size and a
`potentially more favorable safety profile due to minimal systemic
`exposure, reduced immunogenicity, and the absence of permeation-
`enhancing excipients [17,18]. To conserve the therapeutic activity in
`the GI tract, several formulation strategies have been employed, such
`as enteric/colon-targeting capsules that protect against the harsh GI en-
`vironment, supplementation with sacrificial proteins that compete for
`degradation, and hindering enzymatic access using polymer conjuga-
`tion or protective antibodies which bind to known cleavage epitopes
`[4,17,19]. The ailments that are targeted by locally acting macromole-
`cules include inflammatory diseases (Crohn's disease, ulcerative colitis),
`metabolic disorders (e.g., exocrine pancreas insufficiency), constipation,
`and infections (Table 1) [4,17].
`
`2.1. Peptides
`
`2.1.1. Vancomycin and fidaxomicin
`Isolated in 1953 from a soil sample in the jungle of the island Borneo,
`the antibiotic vancomycin is produced by the bacterium Amycolatopsis
`orientalis [20]. Vancomycin is a glycosylated tricyclic heptapeptide
`(1449 Da) which contains modified amino acid residues (e.g., chlorinat-
`ed tyrosine) [20–22]. Due to its highly hydrophilic (log P − 3.1) and
`large cyclic structure, vancomycin is only marginally absorbed and me-
`tabolized in the GI tract [23,24]. Intravenously applied vancomycin was
`initially approved by the FDA to treat penicillin-resistant bacterial infec-
`tions in 1958, and it is still indicated for severe infections caused by
`
`

`

`110
`
`E. Moroz et al. / Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`Fig. 1. Strategies for oral delivery of macromolecular drugs that act on local or systemic targets. In buccal or sublingual delivery, the drug targets the buccal or sublingual mucosa, which
`avoids degradation pathways in the GI tract. Often, permeation enhancement is necessary to cross the multilayered buccal epithelium. Mucoadhesive bacteria that secrete the desired
`protein in situ are a novel means of achieving sustained release in the oral cavity. To enhance stability against degrading enzymes, macromolecular drugs can be chemically modified
`by polymer conjugation (proteins), backbone and base modifications (nucleic acids), and cyclization as well as by introducing D-amino acids (peptides). Sacrificial proteins, protease
`inhibitors, and enteric coatings can also be included in the formulation to further improve GI resistance. To achieve meaningful systemic exposure, absorption enhancement by tight
`junction-disrupting excipients is often needed. A novel approach is the in situ production and secretion of therapeutic biomacromolecules by genetically modified bacteria.
`
`susceptible strains of methicillin-resistant staphylococci, Clostridium dif-
`ficile-associated diarrhea, and staphylococcal enterocolitis and for pa-
`tients who are allergic to beta-lactam antibiotics. Vancomycin is
`formulated as gelatin capsules containing 125 or 250 mg of active com-
`pound (package leaflet). In 2011, a non-peptidic macrocyclic macrolide
`antibiotic fidaxomicin (1058 Da, Dificid®, Cubist Pharmaceuticals) with
`minimal systemic absorption and GI metabolism was approved for the
`treatment of C. difficile infections. Fidaxomicin showed lower relapse
`rates compared with vancomycin. In view of its much higher costs,
`fidaxomicin treatment may be most suitable for patients who are at
`highest risk of relapse [25]. Further cyclic peptidic compounds in clinical
`trials for the local treatment of C. difficile infections include the
`lipopeptide surotomycin (1681 Da, phase III, Cubist Pharmaceuticals/
`Merck), the lipoglycodepsipeptide ramoplanin (2554 Da, NTI-851,
`phase IIb, Nanotherapeutics), the thiazolyl peptide LFF-571 (1367 Da,
`phase II but currently removed from pipeline, Novartis), and the
`lantibiotic NVB302 (2115 Da, phase I, Novacta Biosystems Limited)
`[26,27].
`
`2.1.2. Linaclotide
`Linaclotide (1525 Da) is a truncated derivative of Escherichia coli
`heat-stable (ST) enterotoxin that consists of 14 amino acid residues
`and three disulfide bonds [28]. It acts as a guanylyl cyclase C (GC
`C) agonist locally in the small intestine (Fig. 2) [28–30]. In 2012, it re-
`ceived FDA approval (Linzess®, Forest Labs LLC) for the treatment of
`chronic idiopathic constipation and irritable bowel syndrome with con-
`stipation. After oral administration, linaclotide is minimally absorbed,
`and its plasma concentrations are below the limit of quantification
`[31]. Because linaclotide is stable in the stomach, it is formulated as
`hard gelatin capsules containing 0.145 or 0.290 mg of the active com-
`pound (package leaflet). However, it is cleaved in the small intestine
`to a 13-amino acid active metabolite [31], which is eventually degraded
`in the intestine [32].
`
`2.1.3. Plecanatide and derivatives
`The GC C agonist plecanatide (1682 Da, Synergy Pharmaceuticals) is
`a synthetic analog of uroguanylin, a naturally occurring GI regulator of
`
`GC C signaling, with one amino acid substitution that results in stronger
`receptor binding [33]. It is a bicyclic peptide consisting of 16 amino acid
`residues with two disulfide bonds [33]. Plecanatide completed the clin-
`ical phase III for the treatment of chronic idiopathic constipation, and it
`is currently being evaluated in phase III for irritable bowel syndrome
`with constipation. It showed a statistically significant increase in the
`number of complete spontaneous bowel movements per week com-
`pared with placebo (approximately 20% of durable responders at a
`dose of 3 mg per day vs. 12% in the placebo group). In comparison to
`linaclotide, plecanatide showed reduced diarrhea incidence (10% vs.
`20%) [34]. In addition to plecanatide, Synergy Pharmaceuticals is devel-
`oping a more stable analog, dolcanatide (SP-333), to treat opioid-
`induced constipation. Dolcanatide has two D-amino acid substitutions
`at C- and N-terminus in order to improve stability in simulated gastric
`and intestinal fluids (www.synergypharma.com). Dolcanatide has com-
`pleted a randomized, double-blind, placebo-controlled phase II clinical
`trial in patients with constipation who take opioid analgesics for chronic
`pain (NCT01983306). Despite the increased stability in the intestinal
`environment, the dose of dolcanatide needed to achieve a therapeutic
`effect was comparable with that for plecanatide. Furthermore, the
`dolcanatide-mediated activation of GC C ameliorated inflammation in
`a colitis mouse model, promoting its development in the treatment of
`ulcerative colitis [35].
`
`2.2. Antibodies
`
`2.2.1. Antibody delivery
`The vast majority of clinically tested antibodies with GI luminal tar-
`gets inactivate undesired molecules, such as bacterial toxins, cytokines,
`viruses, and virulence factors [17]. In the treatment of inflammatory
`bowel diseases (IBD), capturing TNF-α in the intestinal lumen is a
`promising alternative to systemic treatment with anti-TNF-α antibodies
`(e.g., adalimumab, infliximab and certolizumab pegol) with regard to
`systemic immunosuppression, development of neutralizing antibodies,
`and needle-free administration [36–38]. AVX-470 (Avaximab™-TNF,
`Avaxia Biologics) is an orally administered polyclonal antibody which
`targets luminal TNF-α in the intestine [39]. It is produced by purifying
`
`

`

`Table 1
`Macromolecular treatments for local GI therapy in clinical development (as of January 2016, source: Thomson Reuters Integrity®, ClinicalTrials.gov, and company press releases).
`
`E. Moroz et al. / Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`111
`
`Type
`
`Generic and trade
`name, molecular
`weight
`
`Modification/Formulation
`
`Indication
`
`Vancomycin (Vancocin®),
`1449 Da
`Fidaxomycin (Dificid®),
`1058 Da
`Surotomycin,
`1681 Da
`Ramoplanin, 2554 Da
`LFF-571, 1367 Da
`NVB302, 2115 Da
`
`Linaclotide (Linzess®),
`1525 Da
`
`Cyclic structure, modified amino
`acid residues; gelatin capsule
`
`Non-peptidic macrocycle; tablet
`
`Cyclic structure
`
`Cyclic structure
`Cyclic structure, thiazolyl peptide
`Cyclic structure, thioether bonds
`
`Cyclic structure; gelatin capsule
`
`Peptide
`
`Clostridium difficile-associated diarrhea
`(CDAD) and staphylococcal enterocolitis
`including methicillin-resistant strains
`
`CDAD
`
`CDAD
`
`CDAD
`CDAD
`CDAD
`Chronic idiopathic constipation (CIC)
`and
`irritable bowel syndrome with
`constipation (IBS-C)
`
`Plecanatide, 1682 Da
`
`Cyclic structure
`
`CIC and IBS-C
`
`Dolcanatide (SP-333),
`1682 Da
`
`Cyclic structure, two D-amino
`acid substitutions
`
`Opioid-induced constipation (OIC) and
`ulcerative colitis (UC)
`
`Development
`phase (NCT#)
`
`Marketed
`
`Marketed
`
`Phase III (NCT01597505,
`NCT01598311)
`Phase IIb (N.A.)
`Phase II (NCT01232595)
`Phase I (N.A.)
`
`Marketed
`
`CIC phase III completed in 2015
`(NCT01919697)
`IBS-C phase III ongoing
`(NCT02493452)
`OIC phase II completed in 2014
`(NCT01983306)
`UC phase Ib ongoing
`(N.A.)
`Phase II ongoing
`(N.A.)
`Phase Ia completed in 2014
`(N.A.)
`
`Antibody
`
`Enzyme
`
`AVX-470 (Avaximab™-TNF),
`broad
`
`AG014, 47.8 kDa
`
`Creon® (porcine protease,
`amylase, lipase)
`
`Protein-secreting
`bacteria
`
`AG013, 6.5 kDa
`
`AG011, 18 kDa
`
`Mongersen (GED-0301),
`6952 Da
`
`Alicaforsen, 6368 Da
`
`Nucleic acid
`
`CEQ508
`
`N.A: unknown or non-existent NCT number.
`
`Polyclonal antibody;
`enteric capsule
`Lyophilized Lactococcus lactis secreting
`certolizumab; enteric capsule
`
`Pediatric UC
`
`Inflammatory bowel disease
`
`Enteric capsule
`
`Exocrine pancreatic insufficiency
`
`Marketed
`
`Oral rinsing solution containing 2.0 × 1011
`colony-forming units (CFU)/15 mL of
`Lactococcus lactis secreting human trefoil
`factor 1
`Lactococcus lactis secreting
`anti-inflammatory
`cytokine IL-10; enteric capsule in
`combination
`with enema
`
`UC
`
`Chemotherapy- or radiation-induced
`oral mucositis
`
`Phase Ib completed in 2012
`(NCT00938080)
`
`Phase IIa completed in 2009,
`failed
`(NCT00729872)
`
`Phase II completed in 2014
`Phase I ongoing
`(NCT02367183)
`
`Phase III ongoing
`(NCT02525523)
`
`Phosphorothioate AON; enteric coating
`
`Moderate to severe Crohn's disease
`
`UC, pouchitis
`
`Phosphorothioate AON; nightly enema
`with
`hydroxypropyl methylcellulose
`Suspension of genetically modified E. coli
`expressing
`invasin, listeriolysin, and short hairpin
`RNA targeting
`β-catenin
`
`Familial adenomatous polyposis
`
`Phase I/II ongoing since 2011
`(N.A.)
`
`total antibodies from the milk of dairy cows that are immunized with
`human TNF-α, which results in approximately 0.3–0.9% (m/m) TNF-α-
`specific and mainly IgG1 subtype antibodies (www.avaxiabiologics.
`com). Providing early immunity to neonates, high amounts of colostrum
`antibodies seem to be partially resistant to GI digestion in adults with
`fully developed digestive systems [40–42]. AVX-470 is developed as an
`enteric-coated capsule for treating pediatric ulcerative colitis and is cur-
`rently in phase II of clinical trials. According to the company's website,
`a phase Ib trial (NCT01759056) in 36 ulcerative colitis patients showed
`that the treatment resulted in consistent encouraging trends across mul-
`tiple disease parameters (colonic TNF-α levels, endoscopic index of se-
`verity, serum C-reactive protein levels) after four weeks, and no allergic
`reaction or human anti-bovine antibodies in serum were observed. Bo-
`vine immunoglobulin was detected in stools and displayed TNF-α-bind-
`ing activity, suggesting that the large excess of decoy antibodies in the
`formulation (N99%) protected a fraction of the anti-TNF-α antibodies.
`
`2.2.2. Genetically modified antibody-secreting bacteria
`Intrexon's (formerly ActoGeniX) approach for luminal antibody
`therapy consists of delivering attenuated bacteria to the GI tract,
`where the antibody is secreted in situ. Food-grade Lactococcus lactis
`was genetically modified to secrete certolizumab, an anti-TNF-α
`Fab fragment (47.8 kDa). In an open-label clinical trial in healthy vol-
`unteers, lyophilized bacteria administered in enteric capsules were
`recovered in both the small and large intestine by endoscopic sam-
`pling, and the antibody was secreted by living bacteria in the colon
`lumen (www.dna.com). The treatment was safe and well-tolerated.
`In addition, preclinical studies demonstrated the treatment's efficacy
`in multiple colitis models and showed that L. lactis adhere especially
`to the inflamed mucosa, which increases the concentration of the an-
`tibody in the close proximity of target cells [43]. However, questions
`remain regarding the concentration variability and the intestinal sta-
`bility of the secreted antibody.
`
`

`

`112
`
`E. Moroz et al. / Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`Fig. 2. Schematic representation of the mechanisms of action, primary structure, and diarrhea incidence of the GC C agonists linaclotide, plecanatide, and dolcanatide. The sequences of
`these peptides are based on the physiological agonist uroguanylin and the structurally similar heat-stable Escherichia coli enterotoxin (ST) but include sequence adaptations such as
`unnatural D-amino acids (dN = D-asparagine, dL = D-leucine). Lines above and below the peptide sequences denote the disulfide bonds. Upon binding of an activating ligand, GC C
`converts guanosine triphosphate (GTP) to cyclic guanosine monophosphate (cGMP), increasing the intracellular cGMP concentration and activating the cystic fibrosis trans-membrane
`conductance regulator (CFTR). The resultant efflux of chloride ions out of the epithelial cell leads to a water influx into the intestinal lumen, which normalizes bowel movements.
`Extracellular cGMP further inhibits intestinal nociceptors. Thus, GC C agonists improve constipation and alleviate chronic abdominal pain in patients suffering from irritable bowel
`syndrome with constipation. The incidence of diarrhea, a common treatment-emergent adverse event associated with excessive treatment response, is also indicated for the three
`peptides (dose and regimen in parentheses). The information was collected from [29–34] and the website of Synergy Pharmaceuticals.
`
`2.3. Oral enzyme therapy
`
`2.4. Cytoprotective/anti-inflammatory proteins
`
`Enzyme replacement therapy via the oral route is an important ther-
`apeutic option in metabolic disorders in which a certain enzymatic ac-
`tion in the GI tract is absent. Numerous oral enzyme products are on
`the market for a variety of disorders such as exocrine pancreatic insuffi-
`ciency due to pancreatitis, cystic fibrosis and other conditions, and lac-
`tose,
`fructose, sucrose and histamine intolerance (e.g., Creon®,
`Lacteeze®, Sucraid®, DAOSiN®) [4]. Whereas the only FDA-approved
`enzymatic drug is pancreatin (a mixture of bovine or porcine pancreatic
`amylase, proteases, and lipases), the other oral enzyme products are
`generally under the legal framework of dietary supplements, which
`usually do not require thorough efficacy studies.
`Because these therapeutic enzymes are inactivated in the stomach
`by low pH and pepsin, one or a combination of stabilization strategies
`are generally needed. Creon® (Abbvie), for instance, consists of pan-
`creatin formulated in enteric-coated capsules, whose dissolution is
`triggered by the pH increase in the small intestine. An uncoated formu-
`lation of pancreatin (Viokace®, Aptalis Pharma) which is used to treat
`exocrine pancreatic insufficiency is co-administered with proton
`pump inhibitors to decrease gastric degradation. As an exception,
`sacrosidase and the acid lactase (tilactase) produced by Aspergillus
`oryzae are intrinsically stable and active in acidic conditions [4], and
`they do not require sophisticated delivery systems. However, average
`residence time of lactase in the stomach might be insufficient for com-
`plete digestion of high amounts of lactose [44]. Therefore, a formulation
`of a lactase, which is active in neutral to basic pH of the small intestine,
`was recently developed as a capsule containing enteric-coated pellets
`(Lactosolv®, Sciotec Diagnostic Technologies) [44]. The same approach
`has been successfully applied for other enzyme deficiencies (Xylosolv®
`for fructose and DAOSiN® for histamine intolerance). As an alternative,
`the administration of probiotics that secrete β-galactosidase, which is
`capable of lactose hydrolysis, has been proposed; however, different
`clinical trials in patients with lactose intolerance gave inconsistent effi-
`cacy results [45]. Oral formulations of polymer-conjugated enzymes
`and enzymes in enteric-coated capsules are under preclinical investiga-
`tion for other pathologies such as celiac disease and phenylketonuria [4,
`46–50].
`
`As a consequence of chemotherapy and radiation, cancer patients
`often develop a complication called oral mucositis in which the mouth's
`mucosal lining is destroyed and ulcers form. AG013 (Intrexon Corpora-
`tion) is a bacteria-based system which delivers a mucosa-healing pro-
`tein for the treatment of oral mucositis. The protein, called human
`trefoil factor 1, is involved in mucosal healing and tissue protection via
`multiple mechanisms, including inhibiting apoptosis during cell migra-
`tion and stabilizing the protective mucus layer [51]. The formulation
`consists of an oral rinsing suspension containing attenuated (i.e., unable
`to replicate due to thymidylate synthase deficiency [51]), genetically
`engineered L. lactis which adhere to the buccal mucosa and secrete the
`mucosa-healing factor locally for up to 24 h after administration [52].
`A phase Ib study in head and neck cancer patients undergoing chemo-
`therapy produced positive results, showing a 35% reduction in the dura-
`tion of oral ulcers [51].
`A similar in situ system based on L. lactis which secretes the anti-
`inflammatory cytokine interleukin-10 (AG011, Intrexon) was proposed
`for ulcerative colitis. However, a combination of enema and enteric
`capsules containing interleukin-10-secreting L. lactis did not result in
`mucosal healing in a phase IIa, double-blind, placebo-controlled study
`with 60 ulcerative colitis patients [53]. Recently, another genetically
`modified strain of L. lactis was engineered to deliver the endogenous
`serine protease inhibitor elafin. The expression of this anti-
`inflammatory peptide is lowered in patients with active celiac dis-
`ease, and elafin treatment was effective in decreasing inflammation
`and intestinal permeability in a mouse model of celiac disease [54].
`
`2.5. Nucleic acids
`
`Antisense oligonucleotides (AONs) are short, single-stranded, syn-
`thetic sequences of nucleotides which can hybridize with complemen-
`tary mRNA and consequently decrease the expression of the encoded
`protein [55]. Several GI tract pathologies hold attractive targets for
`local nucleic acid therapy (e.g., IBD or familial adenomatous polyposis)
`[56,57]. Because nucleic acids are readily degraded by the RNases
`and DNases in the GI tract and depurinated in the acidic stomach
`
`

`

`E. Moroz et al. / Advanced Drug Delivery Reviews 101 (2016) 108–121
`
`113
`
`environment, they require stability-enhancing chemical modifications
`of the backbone or bases [58]. Various modifications of the ribose ring
`(e.g., 2′O-(2-methoxyethyl) or 2′F-ANA) not only exhibit enhanced nu-
`clease resistance but also show higher affinity to complementary
`mRNAs and thus improved knockdown efficacy [59,60].
`Mongersen (GED-0301, Celgene) is a 21-mer phosphorothioate AON
`targeting the mRNA of the Smad7 protein, which is overexpressed in
`IBD mucosal tissues and leads to suppression of anti-inflammatory
`TGF-β signaling [61]. Mongersen acts on epithelial and lamina propria
`cells, and its bioavailability after oral administration is minimal [62].
`An enteric-coated oral formulation of mongersen was evaluated in
`166 patients with active Crohn's disease in a phase II trial [61]. Partici-
`pants received 10, 40, or 160 mg of mongersen once daily for 2 weeks.
`In the mongersen groups that received 40 and 160 mg, significantly
`more patients reached clinical remission, the primary end point, after
`four weeks of treatment (55% and 65%, respectively), compared with
`10% in the placebo group. According to the company, follow-up data
`of this trial (e.g., on mucosal healing by endoscopy) will be presented
`in the future.
`Alicaforsen (Atlantic Healthcare) is a 20-mer phosphorothioate AON
`targeting the mRNA of intercellular adhesion molecule 1 (ICAM-1),
`which recruits immune cells to the site of inflammation and is
`overexpressed in the gut epithelium of IBD patients [63]. Although a
`four-week treatment with injectable alicaforsen fell short of inducing

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket