You are on page 1of 35

Review

pubs.acs.org/CR

Orally Absorbed Cyclic Peptides


Daniel S. Nielsen,†,‡ Nicholas E. Shepherd,†,‡ Weijun Xu,†,‡ Andrew J. Lucke,†,§ Martin J. Stoermer,*,†
and David P. Fairlie*,†,‡

Division of Chemistry and Structural Biology, and ‡Australian Research Council Centre of Excellence in Advanced Molecular
Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia

ABSTRACT: Peptides and proteins are not orally bioavailable in mammals, although a
few peptides are intestinally absorbed in small amounts. Polypeptides are generally too
large and polar to passively diffuse through lipid membranes, while most known active
transport mechanisms facilitate cell uptake of only very small peptides. Systematic
evaluations of peptides with molecular weights above 500 Da are needed to identify
parameters that influence oral bioavailability. Here we describe 125 cyclic peptides
containing four to thirty-seven amino acids that are orally absorbed by mammals.
Cyclization minimizes degradation in the gut, blood, and tissues by removing cleavable
N- and C-termini and by shielding components from metabolic enzymes. Cyclization
also folds peptides into bioactive conformations that determine exposure of polar atoms
to solvation by water and lipids and therefore can influence oral bioavailability. Key
chemical properties thought to influence oral absorption and bioavailability are analyzed,
including molecular weight, octanol−water partitioning, hydrogen bond donors/
acceptors, rotatable bonds, and polar surface area. The cyclic peptides violated to different degrees all of the limits traditionally
considered to be important for oral bioavailability of drug-like small molecules, although fewer hydrogen bond donors and
reduced flexibility generally favored oral absorption.

CONTENTS 4.7. Nepadutant 8104


4.8. Bouvardin 8104
1. Introduction 8095 4.9. Pristinamycin and Related Antibiotics 8105
1.1. Absorption 8095 4.10. 1-NMe3 and Related Cyclic Hexapeptides 8105
1.2. Predicting Absorption 8095 4.11. Cyclo-[Arg-Arg-Arg-Arg-NaphthylAla-Phe] 8107
1.3. Metabolism 8096 4.12. Kahalalide F 8107
1.4. Oral Bioavailability versus Oral Activity 8096 5. Cyclic Heptapeptides 8107
1.5. Formulation and Pharmacokinetics 8097 5.1. Sanguinamide A and Danamides 8107
1.6. Review Scope 8097 5.2. Rhizonin A 8108
2. Cyclic Tetrapeptides 8098 5.3. Microcystin LR 8108
2.1. CJ-15208 8098 5.4. YM254890 8108
2.2. Apicidin and Chlamydocin 8098 5.5. CHEC-7 8108
2.3. Beauveriolides 8098 5.6. Polymyxin B1 and B2 8108
2.4. HIV Fusion Inhibitors 8098 5.7. Bacitracin A 8109
3. Cyclic Pentapeptides 8099 6. Cyclic Octapeptides 8109
3.1. DMP-728 8099 6.1. PF1022A and Emodepside 8109
3.2. Cyclochlorotine and Astin C 8099 6.2. WH1Fungin 8109
3.3. BL3020-1 8099 6.3. α-Amanitin 8109
3.4. Romidepsin 8100 6.4. Griselimycin and Synthetic Derivatives 8110
3.5. Largazole 8100 6.5. Dihydromycoplanecin and Mycoplanecin 8110
3.6. Complement C5aR Antagonists 8100 7. Cyclic Nona- and Deca-Peptides 8110
3.7. Actinomycin D 8100 7.1. CHEC-9 8110
3.8. Leucine Cyclic Peptides 8101 7.2. AFPep 8111
4. Cyclic Hexapeptides 8101 7.3. Antamanide and Cyclolinopeptide 8111
4.1. Desmopressin 8101 7.4. Cyclopeptolide 1 8112
4.2. Melanotan II 8101 7.5. Permetin A 8112
4.3. Oxytocin 8102 7.6. Synthetic N-Methyl β-Strand Decapeptides 8112
4.4. Anidulafungin, Caspofungin, and Micafun-
gin 8102
4.5. Somatostatin, Octreotide, and Analogues 8102 Received: December 20, 2016
4.6. Beauvericin and Enniatins 8104 Published: May 25, 2017

© 2017 American Chemical Society 8094 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

7.7. Surotomycin 8112 antibodies.18−20 They all need to be given by injection, with few
8. Cyclic Undecapeptides 8112 peptides known to be orally absorbed and hardly any being
8.1. Cyclosporin A and Synthetic Derivatives 8112 truly orally bioavailable. There is now growing interest in
8.2. THR-123 8114 developing molecules with molecular weights between those of
9. Cyclic Dodeca- and Tridecapeptides 8114 conventional drugs and antibodies.18 Here we describe some
9.1. Cerulide 8114 examples of orally absorbed peptides, all being cyclic peptides
9.2. L-Phenylalanine-Dipicolinate Macrocycle 8114 of 4−37 amino acid residues.
10. Cyclic Tetradecapeptides and Beyond 8115 1.1. Absorption
10.1. Conotoxins and Synthetic Derivatives 8115
10.2. Duramycin 8115 When a drug is administered by oral, buccal, sublingual, nasal,
10.3. Kalata B1 and Other Cyclotides 8116 dermal, intramuscular, subcutaneous, pulmonary, and rectal
10.4. Stapled α-Helix 8116 routes, it must be absorbed to enter the bloodstream where it is
11. Influences on Oral Bioavailability 8116 systemically circulated, distributed into tissues, metabolized,
12. Conclusions and Future Prospects 8118 cleared, and excreted. Absorption is the process by which a
Author Information 8120 drug moves unchanged from the site of administration to the
Corresponding Author 8120 site of measurement in the organism.21 Absorption from the
ORCID 8120 gastrointestinal (GI) tract after oral administration occurs
Present Address 8120 mainly from the small intestine. Small finger-like folds (villi)
Notes 8120 coating its surface generate a much larger surface area (200 m2)
Biographies 8120 for compound absorption compared to the stomach (1 m2).21
Acknowledgments 8120 The small intestine environment (pH 6−7.5) aids absorption
Abbreviations 8120 because many molecules are uncharged at this pH and can
References 8121 passively diffuse through intestinal membranes. Passive
diffusion is the most common way drugs and many nutrients
are absorbed from the GI tract into plasma. Passive diffusion of
1. INTRODUCTION drugs is most commonly transcellular (through cell membranes,
Proteins and peptides are the largest group of naturally > 90% of drugs) rather than paracellular (through tight
occurring mediators of biological and cellular processes, ranging junctions between enterocytes, 5−10% of drugs). Charged and
in size from small peptide hormones to large multidomain hydrophilic molecules such as peptides are not well absorbed
polypeptides and proteins.1 Proteins bind with high specificity via passive diffusion mechanisms. Membrane-bound transporter
and potency irrespective of whether interactions are localized to proteins also mediate absorption of amino acids, very small
one or more small binding pockets (“hot spots”)2−4 or more peptides, nucleosides, sugars, ions and hydrophilic molecules
dispersed across larger surface areas. When protein function is via facilitated diffusion and active transport.22−26 Studies on
localized, protein−protein interactions (PPIs) can potentially peptide and protein transport across gastrointestinal mucosal
be interfered with by small drug-like molecules.5 However, membranes is still in relative infancy, understanding still
more often than not the bioactive protein interfaces span large confined mainly to single amino acids and small peptides and
surfaces and rely upon multiple weak contacts for affinity and the biotin, transferrin, and glucose transporters. The human
selective recognition. In these cases, an alternative approach to intestinal peptide transporter SLC15A1 is one example of a
modulating PPIs is to mimic one of the interacting protein proton-dependent protein that transports very small peptides
surfaces by downsizing it to smaller peptides or peptidomi- (2−4 residues) via facilitated diffusion.27 SLC15A1 also
metics. These are larger than small molecule drugs, frequently facilitates absorption of certain peptide mimetics and peptide-
require molecular constraints to stabilize structure in water, and like drugs such as ACE inhibitors and β-lactam antibiotics.28
are usually not orally bioavailable.6−13 Conversely, some transporters in enterocytes (e.g., P-
During the last 20 years the pharmaceutical industry has glycoproteins) act as a barrier to absorption,29,30 actively
almost universally adopted drug-like property filters, such as the expelling peptides and drugs31 back into the GI tract.
“rule-of-five” (RO5)14−16 and related parameters.17 These have 1.2. Predicting Absorption
guided the design and development of orally bioavailable
modulators of macromolecules, deliberately focusing attention Predicting oral absorption of drugs in animals is complex and
on small molecules restricted to MW < 500. In the era of experimental measurements are low throughput. Attempts have
postgenomics, transcriptomics, and proteomics, it is time to been made to develop in vitro or ex vivo methods to assess
focus more attention on larger modulators of proteins that can membrane permeability. Although no in vitro or ex vivo model
span larger surfaces, access new therapeutic mechanisms of correlates well with in vivo parameters, some methods have
action, and provide greater target specificity.18 This has been allowed estimates of relative passive membrane permeability.
spectacularly demonstrated by antibodies and some proteins However, others factors (membrane uptake via endocytosis,
that have spearheaded a paradigm shift in the pharmaceutical transporter proteins, protein-binding, stability to proteolytic
industry toward larger therapeutic molecules.19 Proteins and and oxidative/reductive enzymes in the gut, intestine, tissues,
polypeptides are expensive to manufacture, chemically unstable plasma and liver, clearance rate and first pass metabolism) also
(degraded by pH, heat, oxidation, and proteases), difficult to contribute to oral bioavailability.
store, very flexible in water, immunogenic, have low membrane The parallel artificial membrane permeability assay
permeability, and poor oral bioavailability. Despite these (PAMPA)32 has been used widely in drug discovery as a high
limitations, many peptides and polypeptides are in clinical throughput assay to estimate passive diffusion across a
trials and a few are registered drugs, mainly naturally occurring membrane. The artificial membrane lacks transporter proteins
peptides, their semisynthetic derivatives, cyclic peptides, or and so only estimates permeability via passive diffusion.
8095 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Caco-2 cells are heterogeneous human epithelial colorectal flavin-containing monooxygenases, monoamine oxidases and
adenocarcinoma cells grown under conditions to mimic NADPH−CYP450 oxidoreductases. At this stage the com-
absorptive cells of the small intestine.33 Their microvilli, pound can be (1) reabsorbed back into the bloodstream,
metabolic enzymes (e.g., peptidases and esterases), transporter sometimes unchanged; (2) metabolized and then absorbed into
proteins, and bile salts better mimic the physiological the bloodstream; or (3) combined with bile salts and excreted
environment.34 PAMPA and Caco-2 assays can indicate back into the GI tract.
whether a compound is passively or actively transported across Metabolism of peptides can occur at all stages, while
epithelial cells. Madin-Darby canine kidney (MDCK) cells traversing the GI tract, in intestinal tissues, in the bloodstream,
isolated from dog kidney cortex35 retain many properties of the in the liver, and other organs and tissues. Peptides can be
kidney tubular epithelium. Morphologically, MDCK cells hydrolyzed by a myriad of proteases in the gut, plasma, and
exhibit apical microvilli, junctional complexes, and lateral cells, for example, pepsin and HCl in the stomach; trypsin,
membrane infoldings36−39 characteristic of transporting epi- chymotrypsin, elastase, and carboxypeptidases in the small
thelia.40 Physiologically, MDCK cells transport sodium and intestine; and, by thrombin, plasmin and clotting factors in
water in an apical-to-basal direction. When grown on blood plasma degrade peptides. The CYP450 monooxygenases
permeable substrates, MDCK cells generate transepithelial and oxidoreductases in the intestinal lining and in the liver
electrical resistance indicating functioning tight junctions.41 catalyze carbon hydroxylation and epoxidation, heteroatom
These properties resemble Caco-2 cell and other intestinal tract oxygenation and release. CYP450 enzymes account for > 75%
cells, making MDCK cells a viable model for measuring in vitro of drug metabolism within the liver.56 Substances with good gut
permeability despite their anatomical origin. Like Caco-2, absorption can still have low oral bioavailability if metabolism in
MDCK cells have uptake and efflux transporter-proteins, but the intestinal lining, liver, plasma, or elsewhere is high, or
their canine origin may endow different affinity, selectivity, and clearance is rapid. It is a misconception that high gut
activity for substrates compared to human proteins and cells. permeability equates to high oral bioavailability. Stability to
Therefore, caution should be exercised in interpreting these metabolic enzymes, especially in the gut, liver, and blood, is
results.42 MDCK monolayers are faster to culture (3−7 days)43 important when predicting oral activity of membrane-
than Caco-2 (14−28 days), making them useful for high permeable compounds.
throughput permeability assays. A derived cell line, MDCKII- Finally, a relatively new consideration is the influence of the
LE (low efflux) was developed from subpopulations of MDCK microbiome on drug and nutrient metabolism. Differences in
cells.44 MDCKII-LE have greatly reduced expression of canine oral bioavailability between people or animals are often
mRNA/protein and low active uptake/efflux properties making attributed to differences in metabolic enzymes or their
them a live cell alternative to the artificial PAMPA assay. There efficiencies that change with age, genetics and environment.
have been many recent reviews of cell and membrane Until now, the influences of symbiotic bacteria inhabiting the
permeability of cyclic peptides and other macrocycles,45−50 so gut on oral drug stability, metabolism and absorption have not
we will not be covering that literature here. While this is of been considered much and this is likely to be important to
importance, it is not the only contributor to oral absorption and study in the future.
oral bioavailability. Many membrane and cell permeable 1.4. Oral Bioavailability versus Oral Activity
compounds show negligible oral bioavailability.
Oral bioavailability (F) is the fraction of an orally administered
Instead of measuring transport in vitro across cells, an Ussing
compound that reaches the systemic circulation intact.
chamber51 can be used to estimate oral absorption by
Absorption alone is not a good predictor of oral bioavailability,
measuring ex vivo membrane transport of ions, nutrients, and
since extensive first-pass effects in the liver and intestine can
drugs across mouse52,53 or rat54,55 intestinal tissue. However,
lead to poor systemic exposure. Thus, blood is usually sampled
such experiments also correlate poorly with oral bioavailability
from the jugular, rather than portal, vein to take into account
(F%) in rodents and humans. Thus, the Ussing chamber has
first pass metabolism. Oral bioavailability is defined as the ratio
become less popular in industry over the past decade.
of the amounts of drug found in plasma after intravenous (iv)
1.3. Metabolism versus per oral (p.o.) dosing, with the iv dose representing
Orally administered compounds face many obstacles en route 100% bioavailability.21
to the plasma. Compounds of high molecular weight, high
AUCpo dose iv
lipophilicity or low solubility are recognized and degraded by F(%) = 100 × ×
metabolic enzymes. Orally ingested compounds are first dose po AUCiv
exposed to digestive amylases in the saliva, where glycosidic
bonds of starch and other carbohydrates are hydrolyzed. Then For clinical trials, drugs normally need to have F > 20%.
in the stomach, acidic (pH 1−2) gastric juice containing Other parameters described in this review that relate to oral
peptidases begins to degrade proteins and other nutrients. bioavailability include the following: the area under the curve
Upon entering the duodenum, the pancreas excretes additional (AUC), used to express the cumulative amount of drug found
enzymes (proteases, lipases, amylases), bile salts and pH- in plasma over a period of time; the clearance rate (CL), the
neutralizing bicarbonate. Next, in the jejunum and ileum (small volume of plasma from which a drug is completely removed per
intestine) the pH is 6−7.5 and most processed nutrients and unit time; the peak serum concentration (Cmax), the time to
drugs are absorbed here. Intestinal P450 enzymes can reach peak serum concentration (Tmax); the half-life (t1/2), time
metabolize compounds even before they enter the bloodstream taken for serum drug concentration to decrease by half its
and thus reduce measurable oral absorption. Once absorbed original amount; and the volume of distribution (VD), the
from the GI tract, compounds enter the hepatic portal vein, apparent volume in which the drug is distributed at steady state.
flow into the liver and are perfused into hepatocytes for first- These parameters provide quantitative estimates of the
pass metabolism by cytochrome P450 (CYP450) enzymes and compound absorbed and surviving first pass metabolism.
8096 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Oral activity is often used as a surrogate to infer oral rats. Pharmacokinetic investigations in rats often involve
bioavailability. Orally dosing an animal and observing a sampling blood at multiple time points from the same animal
therapeutic effect provides only qualitative evidence of oral through a surgically implanted line. This is more difficult for
bioavailability. We have included reports for orally active mice, for which studies usually involve larger groups of animals,
peptides in this review even though many such molecules are so that multiple mice can be sacrificed at each time point.
poorly absorbed; most peptides and proteins are < 1% orally Pharmacokinetic parameters measured by the latter method
bioavailable. We urge caution in drawing robust conclusions provide less accurate results due to variation within animals
about oral bioavailability of molecules from such studies. The (metabolism, total blood, body volume, individual weight, etc.)
observed biological effect could occur by indirect mechanisms, or their environments. It is therefore prudent to carefully
from metabolites or may be due to exceptional potency of trace consider parameters such as dose, species of test animal, and
material absorbed. The main measures of oral activity used in experimental design before drawing conclusions.
this review are the oral dose (mg/kg p.o.) required to induce a 1.6. Review Scope
measurable response; the effective dose for 50% of the maximal
response (ED50); and the lethal oral dose required to kill 50% Most proteins and peptides show negligible oral bioavailability
or 100% of the test population (LD50 and LD100). (F < 1% and usually F < 0.1%). Very few peptides are
sufficiently orally absorbed to produce some physiological effect
1.5. Formulation and Pharmacokinetics in an animal. Cyclic peptides are the most orally bioavailable
When comparing pharmacokinetic parameters for compounds peptides known, but only a handful show F ≥ 10%. Other types
tested in separate studies, it is important to note possible of macrocycles are orally absorbed, but the reader is directed to
differences in experimental design. Compounds are usually other reviews for these.64−67 This review covers only orally
formulated with a solvent, vehicle, or matrix, which can absorbed cyclic peptides reported up until 2016, including
profoundly affect their solubility, rate and location of some that are absorbed in only trace amounts but sufficient to
dissolution, and permeability across intestinal membranes. induce a physiological response. Cyclic peptides have been
Formulation design is a crucial step in drug delivery. Some classified herein according to the number of residues in their
formulations simply increase chemical stability or solubility at macrocyclic portion. For some highly modified cyclic peptide
different pH, in aqueous or lipophilic environments. Other natural products, we have grouped them with cyclic peptides of
formulations are optimized to release the compound at a equivalent macrocycle size (e.g., largazole is grouped with
specific location in the gut, in tissues or in target organs or cells. romidepsin). In cases where a related series of different sized
Often an excipient is added to enhance membrane permeability cyclic peptides were developed for the same target, we have
or oral absorption. Thus, formulation is a key determinant of grouped them together based on the smallest cyclic peptide in
pharmacokinetic parameters, which are highly dependent on the series in order to streamline discussion and avoid repetition.
the conditions under which they are measured. The main focus of this review is on cyclic peptides larger than
Proteins and peptides have been formulated in many ways in four residues because they violate most, often all, rule-of-five
attempts to increase oral absorption57−63 including with (i) (RO5)14−16 and associated17 parameters. We have also
enzyme inhibitors to reduce proteolysis in the GI tract (e.g., included a few representative cyclic tetrapeptides, even though
sodium glycocholate, trypsin inhibitors, camostat mesylate, these usually comply with RO5 parameters. We describe 125
bacitracin, and ovomucoids); (ii) absorption enhancers to cyclic peptides in total for which there is evidence of oral
improve intestinal permeability (e.g., detergents, surfactants, absorption, oral activity, or oral bioavailability. Where possible,
bile salts, and chelating agents); (iii) mucoadhesive polymers to we have indicated relevant doses, formulations, pharmacoki-
improve delivery and permeability (PEGs, P(MAA-g-EG), netic parameters, and pharmacological activities. In some cases,
lectin microparticles, and thiolated polymers); (iv) formulation there was evidence of solid state or solution conformations that
vehicles to protect drug and improve permeability (emulsions, indicated rigidity or flexibility that likely influence absorption.
liposomes, cyclodextrins, microspheres, micelles, and nano- These are referenced to codes in the Protein Data Bank (PDB)
particles). or Cambridge Crystallographic Data Centre (CCDC).
As shown above, absolute oral bioavailability (F%) is defined Our review concludes with an analysis of this compound set
as the dose-corrected ratio of accumulated concentrations in for influences of physicochemical parameters normally
plasma (AUC) following an iv injection (AUCiv: 100% considered to affect oral bioavailability. A recent review68 on
bioavailability by definition) and an oral dose (AUCpo). This bioactive linear and cyclic peptides from the ChEMBL database
dose-corrected absolute value allows direct comparisons to be only examined eight cyclic peptides that were orally
made across different studies conducted at different doses. In administered. For the cyclic peptides that follow, we have
reality, as dose of the test compound is raised, physio-chemical calculated molecular weight (MW), the predicted octanol−
boundaries and saturation of the biological system will water partition coefficient (Molinspiration LogP, miLogP), the
increasingly affect solubility, absorption, active transport number of hydrogen bond donors (HBD) and hydrogen bond
mechanisms, and metabolism. Other pharmacokinetic param- acceptors (HBA) as strictly defined by Lipinski and colleagues,
eters, including AUC and Cmax, are dose-dependent as they are the number of rotatable bonds (RotB), and the topological
expressed in terms of compound concentration in plasma and polar surface area (tPSA), all determined with the aid of the
will therefore depend on the administered dose. Molinspiration webportal (http://www.molinspiration.com).69
The animal model is an important consideration. Biological We plot each of these six parameters against oral bioavailability
variation allows different species, even different strains of the (F%), for those compounds where oral bioavailability has been
same species, to metabolize drugs in different ways, making reported. We then discuss how the findings relate to the RO5
comparisons across species and even strains difficult. For and associated guidelines commonly used to predict oral
technical reasons, it also may not be possible to perform bioavailability of drug-like small molecules. Systematic evalua-
identical experiments in two species, even rodents like mice and tion of more peptides with MW > 500 are needed to determine
8097 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

allowable upper limits for molecular properties (MW, logP, growth. For 3, LD50 was 226 (iv) and > 850 (p.o.) mg/kg in
HBA, HBD, RotB, and PSA) that influence (a) passive mice and 66 (iv) and 141 (p.o.) mg/kg in rats.72 Both 2 and 3
(unassisted) permeability of cells and oral bioavailability and contain one D-residue, a D-pipecolic acid in 2 and a D-proline in
(b) active or facilitated transport mechanisms that promote 3. In solution,73 two intramolecular hydrogen bonds defined a
uptake of peptides > 5 amino acids from the gut. This article γ- and β-turn in 2, but these were replaced in the crystal
takes one important step toward identifying how these structure from chloroform/methanol73 by intermolecular
molecular properties vary in orally absorbed cyclic peptides. interactions (CCDC code: HEWGOG).

2. CYCLIC TETRAPEPTIDES
Cyclic tetrapeptides are generally RO5 compliant, with MW <
500, HBD = 4, ‘HBA’ = 8, and LogP = 0−5 if bearing
hydrophobic side chains. They generally have few rotatable
bonds and a small surface area and so, not surprisingly, they are
also often orally absorbed and orally bioavailable to some
extent. Cyclic tetrapeptides are typically used to stabilize a β-
turn conformation, with proline and D-amino acids often
promoting β-turn formation.
2.1. CJ-15208
Cyclic tetrapeptide CJ-15208 cyclo-(Phe-D-Pro-Phe-Trp) (1,
Figure 1), isolated from the fungus C. serratus ATCC 15502, Figure 2. Apicidin (2) and chlamydocin (3). 2: MW = 624; miLogP =
showed dose-dependent antinociceptive activity in mice 3.4; HBD = 3; HBA = 11; RotB = 12; tPSA = 139 Å2. 3: MW = 527;
measured from 20 to 80 min after oral administration (1 mg/ miLogP = 0.9; HBD = 3; HBA = 10; RotB = 9; tPSA = 137 Å2.
kg p.o.). When given at a higher dose (10 mg/kg p.o.), 1
antagonized a centrally administered selective κ-opioid receptor 2.3. Beauveriolides
agonist, suggesting that 1 is brain permeable.70 This cyclic Beauveriolides (Figure 3) are cyclic tetradepsipeptides, with
tetrapeptide has only one RO5 violation (MW > 500), a D- three amide bonds, one D-residue, and one ester bond, were
proline ring that removes one peptide NH hydrogen bond isolated from the fungus Beauveria sp. FO-6979. Beauveriolides
donor, and hydrophobic side chains, making it suitable for oral are potential antiatherosclerotic agents that reduce cholesteryl
absorption. ester synthesis in mouse macrophages via inhibition of acyl-
CoA:cholesterol acyltransferase (ACAT), leading to a reduction
in lipid droplet formation in macrophages. Lipid accumulation
is associated with the development of atherosclerosis, which can
be studied in apoE- or LDL-receptor knockout mice.
Beauveriolide III (5) was administered to apoE- or LDL-
receptor knockout mice for 2 months (25 mg/kg/day p.o.) and
was orally active in mouse models of atherosclerogenesis by
inhibiting ACAT activity.74 The ester bond in depsipeptides is
not as stable as the amide bond in vivo, but does promote cell
permeability before intracellular ester cleavage by esterases.
Figure 1. Cyclic tetrapeptide CJ15208 (1). MW = 578; miLogP = 2.8;
HBD = 4; HBA = 9; RotB = 6; tPSA = 120 Å2.

2.2. Apicidin and Chlamydocin


Cyclic tetrapeptides 2 and 3 are analogues of 1 and are RO5
compliant except for MW (Figure 2). Apicidin (2) is a cyclic
tetrapeptide metabolite isolated from cultures of F. pallid-
oroseum. It is a histone deacetylase (HDAC) inhibitor and has
been shown to kill protozoa such as Plasmodium sporozoites. Figure 3. Beauveriolide I (4) and III (5). 4: MW = 488; miLogP = 4.1;
When administered in DMSO/PEG400/saline (15:20:65) by HBD = 8; HBA = 8; RotB = 12; tPSA = 114 Å2. 5: MW = 488;
oral gavage, oral bioavailability varied slightly (10 mg/kg p.o.; F miLogP = 4.3; HBD = 3; HBA = 8; RotB = 8; tPSA = 114 Å2.
= 14% (fasting), 19% (nonfasting); Cmax = 235 ng/mL, Tmax =
66 min, t 1/2 = 54 min, V D = 2.5 L/kg, CL = 62
mL.min−1.kg−1)).71 The apicidin-like natural product, chlamy- 2.4. HIV Fusion Inhibitors
docin (3, Figure 2), was found to inhibit P-815 cell growth in A critical step in HIV-1 entry into host cells is the protein−
mastocytoma mouse cells and glial tumor rat cells in vitro protein interaction between CD4 and gp120. Inhibition of this
(ED50 = 0.36 ng/mL). When 3 was administered to tumor- protein−protein interaction (PPI) has been the target of many
inoculated mice (iv and p.o.) there was no antiproliferative research groups.75 A library of cyclic peptides was generated to
activity. The difference between cell and animal findings was join two noncontinuous regions of CD4-containing key
attributed to in vivo inactivation of the epoxide in blood. residues Phe43 and Arg59 that interact with gp120.75 A six
Consistent with this notion, intraperitoneal injections were residue mimic cyclized with succinic acid and ethylenediamine
more effective than intravenous injections in inhibiting tumor gave cyclic heptapeptide-like 6 (Figure 4). Optimization of the
8098 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

linker-length by inserting extra methylene units and replacing


residues Gln-Gly-Ser with a pimelic acid moiety enabled
inhibition of HIV-1 infection by analogue 7 (Figure 4), which
resembles a cyclic tetrapeptide. Despite retention of an
arginine, with its protonated guanidine at physiological pH
usually being an impediment to absorption, 7 still had
reasonable pharmacokinetic properties in rats (1 mg/kg iv:
Cmax = 102 ± 19 ng/mL, VD = 0.6 L/kg; 10 mg/kg p.o. in
water: Cmax = 866 ± 76 ng/mL, Tmax = 18 min, AUC = 21,770
± 63 min·ng/mL, t1/2 = 73 min, CL = 14.5 ± 0.3 mL/min·kg, F Figure 5. DMP-728 (8). MW = 561; miLogP = −2.3; HBD = 9; HBA
= 10%).75 Converting 6 to 7 increased RO5 compliance for = 15; RotB = 8; tPSA = 236 Å2.
MW, HBD, HBA, and RotB, halved the tPSA, and significantly
increased hydrophobicity. Although these changes make 7 3.2. Cyclochlorotine and Astin C
smaller and less polar than 6, and are expected to enhance oral Cyclochlorotine (9, Figure 6) is a hepatotoxic cyclic
bioavailability, no data for 6 has been reported to confirm this pentapeptide that was isolated from the common rice-infecting
point. mold P. islandicum.77,78 Its structure consists of natural as well
as unnatural L-amino acids, including β-phenylglycine and 3,4-
dichloro-L-proline. LD50 values were determined in mice (LD50;
iv 0.3, sc 0.5, and p.o. 6.6 mg/kg), suggesting some oral
absorption.78

Figure 4. HIV fusion inhibitors (6) and cyclic tetrapeptide-like (7). 6:


MW = 775; miLogP = −5.2; HBD = 15; HBA = 22; RotB = 13; tPSA Figure 6. Structures of cyclochlorotine (9) and astin C (10). 9: MW =
= 363 Å2. 7: MW = 488; miLogP = −0.3; HBD = 8; HBA = 11; RotB 572; miLogP = −2.5; HBD = 6; HBA = 12; RotB = 4; tPSA = 177 Å2.
= 9; tPSA = 184 Å2. 10: MW = 570; miLogP = −1.0; HBD = 5; HBA = 11; RotB = 4; tPSA
= 157 Å2.

3. CYCLIC PENTAPEPTIDES Astin C (or Asterin, 10, Figure 6), a close analogue of 9 is a
Cyclic pentapeptides generally have HBD = 5, HBA = 10, LogP plant cyclic peptide isolated from the roots of A. tataricus. The
= 0−5 if bearing hydrophobic side chains, but their molecular crystal structure of 10 from chloroform/methanol (CCDC
weights usually exceed 500, resulting in more rotatable bonds code: WILXEW) showed a single transannular hydrogen bond
and larger surface areas. To be orally absorbed, they often from an Abu NH to the (i, i+3) carbonyl of the β-phenylglycine
require synthetic modifications. carbonyl.79 The remaining hydrogen bond donors and
3.1. DMP-728 acceptors are highly solvent exposed. Inflammatory bowel
disease is characterized by activation of T lymphocytes. Astin C
DMP-728 (8, Figure 5) is a potent and specific antagonist of
has been shown to lower mouse serum concentrations of TNF,
platelet glycoprotein IIb/IIIa complex (GPIIb/IIIa). Like 6 and
IL-4, IL-17, and to induce apoptosis in activated T cells. Daily
7, compound 8 contains an arginine residue that is normally
oral dosing of astin C (2 or 4 mg/kg/day p.o. in 5%
detrimental to oral absorption. It consists of four conventional
methylcellulose in saline) was shown to protect mice against
amino acids, including a D-Abu, residues with N- and C-termini
TNBS-induced colonic inflammation.80
linked by a fifth, unnatural, hydrophobic amino acid, 3-
aminomethybenzoic acid. The pharmacokinetic profile for 8 3.3. BL3020-1
was measured following oral administration with or without α-Melanocyte stimulating hormone (α-MSH) is a 13-residue
absorption enhancers to rats (10 mg/kg iv, 8 mg/kg p.o. with peptide (Ac-Ser-Tyr-Ser-Met-Glu-His-Phe-Arg-Trp-Gly-Lys-
palmitoylcarnitine chloride; Cmax = 0.44 ± 0.21 μg/mL, Tmax = Pro-Val) that stimulates the release of melanin by skin
0.75 h, AUC = 1.38 ± 0.56 μg·h/mL, t1/2 = 2.8 ± 2.1 h, F = melanocytes. α-MSH also binds to the melanocortin 4 receptor
14.6%) and dogs (2 mg/kg iv, 2 mg/kg p.o. with (MC4R), which modulates food intake and energy utilization.81
palmitoylcarnitine chloride; Cmax = 0.41 ± 0.10 μg/mL, Tmax The tetrapeptide sequence His-Phe-Arg-Trp, and other
= 1.0 h, AUC = 1.09 ± 0.22 μg·h/mL, t1/2 = 3.3 ± 0.9 h, F = analogues derived from α-MSH, decrease food intake and
20.5%).76 Absorption enhancers significantly improved phar- elevate energy utilization upon binding to MC4R, making them
macokinetic parameters compared to controls formulated in attractive targets as antiobesity drugs.82 However, they suffer
microcapsules. The MW, LogP, HBD, and tPSA all violate from metabolic instability and poor intestinal permeability. A
limits usually associated with passively diffusing, orally backbone-cyclized α-MSH analogue based on Phe-D-Phe-Arg-
bioavailable, drug-like small molecules. We speculate that the Trp-Gly-NH2, activated MC4R and had increased oral
N-methyl arginine might promote active transport of this bioavailability. One analogue, BL3020-1 (11, Figure 7) was
compound across the intestinal membrane. MC4R selective, had good permeability in the Caco-2 model,
8099 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

and enhanced metabolic stability in rat brush border membrane HDACs. Largazole has been synthesized by multiple routes
vesicles (BBMVs). A single oral dose administered to rats (0.5 allowing many derivatives to be produced.86−91 A crystal
mg/kg p.o. dissolved in water) led to reduced food structure of 13 bound to HDAC 8 (PDB code: 3RQD) shows a
consumption, while repetitive daily oral dosing reduced weight single NH directed to the center of the macrocycle but no
gain in rats.82 Pharmacokinetic parameters measured in rats strong evidence of intramolecular hydrogen bonding.92
following iv administration (1 mg/kg iv in H2O) indicated t1/2 Largazole analogues cocrystallized with HDAC 8 also do not
> 105 min and VD = 2.1 L/kg. Oral administration (10 mg/kg show any intramolecular hydrogen bonds (PDB codes: 4RN1,
p.o. in H2O) to rats showed AUC = 24980 ng·min/mL, Cmax = 4RN2).93 Pharmacokinetic parameters measured in rats showed
202 ± 39 ng/mL, Tmax = 37 ± 10 min, and F = 8.5%. We that 13 was unstable in vivo and rapidly cleared after a single iv
speculate that charged residues in this compound may help dose (10 mg/kg iv in EtOH/DMSO/PEG400/saline (1:1:1:2);
promote intestinal absorption via facilitated transport. Interest- CL = 76 ± 18 L/h·kg, t1/2 = 0.5 ± 0.1 h, AUC = 134 ± 29 μg·
ingly, concentrations of ∼5 ng/mL were also found in brain h/mL, VD = 27 ± 11 L/kg, Cmax = 280 ± 64).94 Largazole 13,
tissue, indicating some ability of 11 to cross the blood brain Boc-L-cysteine-largazole disulfide 14, and disulfide homodimer
barrier.82 15 (Figure 9) were administered orally (50 mg/kg p.o. in
polyethylene glycol/glycerol/EtOH/DMSO 60:15:15:15:10)
to female nude mice.95 Oral activities were measured through
in vivo hyperacetylation of harvested HCT116 tumors. Only 13
produced hyperacetylated histones, however largazole free thiol
was found in tumors from animals orally dosed with 13, 14, or
15, indicating that all three derivatives displayed some oral
absorption in mice.

Figure 7. Structure of BL3020−1 (11). MW = 836; miLogP = −0.8;


HBD = 12; HBA = 18; RotB = 13; tPSA = 287 Å2.

3.4. Romidepsin
Romidepsin (Istodax, FK-228, 12, Figure 8) is a cyclic peptide Figure 9. Structures of largazole (13), its Boc-L-cysteine disulfide
HDAC inhibitor that possesses potent antitumor activity derivative (14), and its disulfide dimer (15). 13: MW = 623; miLogP
against a variety of human cancer cell lines and xenografts.83,84 = 5.3; HBD = 2; HBA = 9; RotB = 12; tPSA = 127 Å2. 14: MW = 716;
miLogP = 2.8; HBD = 4; HBA = 13; RotB = 12; tPSA = 185 Å2. 15:
It contains D-cysteine, D-valine, and (3S,4E)-3-hydroxy-7-
MW = 991; miLogP = 3.6; HBD = 4; HBA = 16; RotB = 11; tPSA =
mercapto-4-heptenoic acid residues. Romidepsin is an RO5- 220 Å2.
compliant and orally active prodrug, reduction leading to a bis-
thiol, one of which covalently bonds to the catalytic zinc ion in
HDAC enzymes. It was administered orally to mice with 3.6. Complement C5aR Antagonists
human prostate tumor xenographs (3 × 50 mg/kg/wk p.o.),84 3D53 (16, Figure 10), later known as PMX53,96 is a derivative
and was also significantly absorbed after oral administration to of the C-terminal turn of human complement protein C5a and
rats (10 mg/kg iv; Cmax = 1829 ± 359 ng/mL, VD = 22 ± 7 L/ is a potent antagonist of the human C5a receptor C5aR1 (IC50
kg, t1/2 = 5.9 ± 1.2 min, AUC = 25409 ± 8767 ng/mL·min.; 50 3 nM against 3 nM C5a).96−100 The Fairlie group designed
mg/kg p.o., AUC = 19712 ± 9168 ng/mL·min, F = 16 ± 3D53 and a range of cyclic pentapeptides97−99 featuring an
7%).83 endocyclic D-cyclohexylalanine and L-arginine that are im-
3.5. Largazole portant for binding, an aromatic residue (L-tryptophan or L-
phenylalanine) that is required for antagonism, an L-proline
Largazole (13, Figure 9) is a cyclic depsipeptide natural product
turn-inducing constraint, and an exocyclic aromatic ring (L-
isolated from Symploca sp. collected from the Florida Keys.85
phenylalanine in 16 or a phenyl propionyl group in 3D624
Largazole is one of the most potent HDAC inhibitors known,
(later called PMX205)) for affinity.99 NMR structural studies of
with activity at low nM concentrations and selectivity for class I
16 and analogues in DMSO-d6 indicated a turn motif.99
Compound 16 is a classic example where oral activity is
observed despite low oral bioavailability, due to a long
residence time on the receptor (t1/2 ≈ 20 h) overcoming low
systemic availability (F = 1−2%, rat).100 This compound and its
analogues display efficacy in vivo in over 20 rat and mouse
models of human disease following oral administration.96
3.7. Actinomycin D
Actinomycin D (17, Figure 11) is an antibiotic natural product
from a family of actinomycins first isolated in 1940.101 It
Figure 8. Structure of romidepsin (12). MW = 541; miLogP = 1.6; features two cyclic pentapeptides, each with an L-proline and
HBD = 4; HBA = 10; RotB = 2; tPSA = 143 Å2. two N-methyl amino acids, bridged by a phenoxazinone linker.
8100 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

p.o. in olive oil: AUC = 442 ng·h/mL, Cmax = 187 ng/mL, F =


4%), 19 (1 mg/kg iv in DMSO: CL = 4.7 mL/min·kg, VD =
0.19 L/kg, t1/2 = 1.0 h; 10 mg/kg p.o. in olive oil; AUC = 6289
ng·h/mL, Cmax = 1900 ng/mL, F = 18%), and 20 (1 mg/kg iv in
DMSO: CL = 24.1 mL/min·kg, VD = 0.75 L/kg, t1/2 = 1.2 h; 10
mg/kg p.o. in olive oil; AUC = 642 ng·h/mL, Cmax = 174 ng/
mL, F = 9%). Despite being larger with more RO5 violations
(Figure 12), 19 had better oral bioavailability than 18, and was
comparable to cyclosporin A (F = 21%) under the same
conditions.106
Figure 10. Structure of 3D53 (16). MW = 896; miLogP = 1.0; HBD =
11; HBA = 18; RotB = 14; tPSA = 273 Å2.

A crystal structure of actinomycin C in complex with


deoxyguanosine showed two intramolecular hydrogen bonds
between the two Val residues keeping the macrocycles close
together to help insert the phenoxazinone into DNA (CCDC
code: ACTDGU10).102 This preorganization minimized
solvent exposure to polar atoms. Similar structures in the
absence of nucleotides (CCDC codes: BEJXET, BRAXGU,
GIDNUC, POHMUU) show stacking of the cycles but the
Val···Val hydrogen bonds remain. These cycle−cycle inter-
actions loosened in crystal structures of 17 bound to short Figure 12. Cyclic pentaleucine (18) and cyclic hexaleucines (19, 20).
18: MW = 566; miLogP = 3.9; HBD = 5; HBA = 10; RotB = 10; tPSA
sequences of DNA (PDB codes: 4HIV, 1I3W).103,104 Actino-
= 145 Å2. 19: MW = 679; miLogP = 4.7; HBD = 6; HBA = 12; RotB =
mycin D suppresses transcription, is cytotoxic and effective in 12; tPSA = 174 Å2. 20: MW = 679; miLogP = 4.7; HBD = 6; HBA =
treating various tumors. It has an estimated bioavailability F = 12; RotB = 12; tPSA = 175 Å2.
5% based on adverse systemic effects after oral versus iv
administration over 2 weeks.105 LD50 values for 17 were
determined in mice (LD50 7.8 mg/kg p.o.) and rats (LD50 iv
0.4, p.o. 7.2 mg/kg). 4. CYCLIC HEXAPEPTIDES
Cyclic hexapeptides and larger macrocycles generally contra-
vene RO5 parameters, with MW > 500, HBD ≥ 6, HBA ≥ 12,
LogP = 0−5 if bearing hydrophobic side chains. They have
more rotatable bonds and larger surface areas than the smaller
cyclic peptides above.
4.1. Desmopressin
Desmopressin (21, Figure 13) was first reported in 1966 as an
analogue of vasopressin (antidiurectic hormone). Its deami-
nated cysteine at position 1 and D-arginine at position 8
enhanced metabolic stability and antidiuretic effects. The
bioavailability of desmopressin in normal healthy adults was
low following intranasal administration (F = 3−5%) and very
low after oral delivery (F = 0.08−0.16%).107−110
4.2. Melanotan II
Figure 11. Structure of actinomycin D, (17). MW = 1255; miLogP =
0.8; HBD = 6; HBA = 28; RotB = 8; tPSA = 360 Å2. Melanotan II (22, Figure 14) is a cyclic hexapeptide analogue
of α-melanocyte-stimulating hormone that was developed as a
tanning agent to help prevent skin cancer. It displayed some
3.8. Leucine Cyclic Peptides
A selection of three all-leucine cyclic peptides (Figure 12),
cyclo-[(L-Leu)5] (18), cyclo-[(L-Leu)6] (19), and cyclo-[(L-
Leu)5(D-Leu)] (20), were synthesized and orally administered
to rats.106 These compounds are at the boundary limits of RO5
guidelines for molecular weight, hydrogen bond donors/
acceptors. Their other properties (LogP, rotatable bonds,
tPSA) depend upon substituents on the cyclic peptide scaffold.
NMR, CD and X-ray structural studies on 18−20 did not show
intramolecular hydrogen bonds, but intermolecular interactions
for 18 were consistent with evidence for aggregation in
solution.106 Despite lacking N-methyl groups or depsipeptide
bonds to reduce the number of hydrogen bond donors, all three
compounds showed oral absorption: 18 (1 mg/kg iv in DMSO: Figure 13. Structure of desmopressin (21). MW = 1069; miLogP =
CL = 13.1 mL/min·kg, VD = 0.36 L/kg, t1/2 = 0.5 h.; 10 mg/kg −4.3; HBD = 18; HBA = 26; RotB = 20; tPSA = 435 Å2.

8101 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

bioavailability after oral administration to rats (0.3 mg/kg iv, administration (250 mg/kg p.o.: AUC0−48h = 23.3, 26.5, 17.7,
6.76 mg/kg p.o., F = 4.6%) despite violating all RO5 and and 6.9 μg·h/mL, respectively; Cmax = 1.6, 1.5, 0.6, and 0.3 μg/
related parameters (Figure 14).111 mL, respectively). The effect of different meal treatments
(fasted, mixed meal, lipid meal, protein meal, or carbohydrate
meal) prior to identical oral dosing was also investigated (250
mg/kg p.o.; AUC0−48h = 21.2 ± 5.8, 8.9 ± 2.6, 7.5 ± 1.8, 8.9 ±
2.8, and 25.2 ± 5.1 μg·h/mL, respectively. Cmax = 1.1 ± 0.3, 0.5
± 0.17, 0.4 ± 0.13, 0.5 ± 0.2, and 1.6 ± 0.3 μg/mL,
respectively).117 Low oral absorption of 24 in humans was
reported in early clincal trials (F = 2−7%).119 A similar
compound, caspofungin (25), displayed poor oral absorption in
rats (50 mg/kg p.o., F < 0.2%).120 Micafungin (26) is an
antifungal agent approved for intravenous use by the USFDA in
2005 and in several other countries. It is a further modified
echinocandin analogue primarily used against Candida
Figure 14. Structure of melanotan II (22). MW = 1024; miLogP = infections in HIV-positive patients. It is an inhibitor of beta-
−0.1; HBD = 16; HBA = 24; RotB = 18; tPSA = 382 Å2. 1,3-D-glucan synthase, essential for fungal cell wall synthesis. It
is poorly orally absorbed.121,122
4.3. Oxytocin
Oxytocin (23, Figure 15) is a disulfide-bridged cyclic
hexapeptide hormone with an additional three amino acids
outside the cycle. This mammalian neurotransmitter and
hormone is associated with numerous physiological responses.
It is currently used in the clinic to induce childbirth and
lactation.112,113 All physicochemical parameters listed in Figure
15 violate guidelines for oral bioavailability. An NMR structure
for oxytocin in water showed two transannular mainchain
hydrogen bonds (Asn NH···OC Tyr, Cys NH···OC Tyr),114
which likely reduce the 3D PSA from the nominal tPSA value.
A crystal structure of a des-amino derivative of oxytocin (PDB
codes: 1XY1, 1XY2; CCDC code: DUPFAV) showed a
hydrogen bond defining a β-turn for the YIQN motif, and a
Tyr NH···OC Asn transannular hydrogen bond along with the
disulfide bond bracing the structure.115 Nevertheless, the oral
bioavailability of oxytocin is very low in rats (F = 0.9%),
requiring it to be injected for efficacy.116

Figure 16. Structures of anidulafungin (24), caspofungin (25), and


micafungin (26). 24: MW = 1140; miLogP = −0.8; HBD = 14; HBA =
24; RotB = 14; tPSA = 377 Å2. 25: MW = 1093; miLogP = −4.6; HBD
= 18; HBA = 25; RotB = 23; tPSA = 412 Å2. 26: MW = 1270; miLogP
= −5.3; HBD = 17; HBA = 32; RotB = 18; tPSA = 510 Å2.

Figure 15. Structure of oxytocin (23). MW = 1007; miLogP = −3.7; 4.5. Somatostatin, Octreotide, and Analogues
HBD = 16; HBA = 24; RotB = 17; tPSA = 400 Å2.
Somatostatin-14 (27) is a growth hormone-inhibiting peptide
resulting from cleavage of its 166 residue preproprotein that is
4.4. Anidulafungin, Caspofungin, and Micafungin differentially expressed in tissues and regulates endocrine
Anidulafungin (LY303366, Eraxis, 24, Figure 16) is a lipid- function. Potential therapeutic applications of somatostatin-14
modified cyclic peptide analogue of echinocandin B.117 were quickly recognized, however its low half-life in plasma (∼2
Pharmacokinetic parameters were measured in female Beagle min) made it an unsuitable drug candidate. Many analogues
dogs following oral and intravenous administration (5 mg/kg (e.g., Figure 17) were developed to improve pharmacokinetic
iv: t1/2 = 15.6 h; CL = 0.10 ± 0.02 L/h·kg, VD = 1.76 ± 0.11 L/ properties. Bicyclic 28 displayed potent biological activity.
kg, AUC0‑∞ = 49409 ± 10286 ng·h/mL; 5 mg/kg p.o.: Cmax = Contracting this cycle by removing nonessential amino acids
307 ± 61 ng/mL, Tmax = 4.7 ± 1.2 h, AUC0‑∞ = 4477 ± 768 ng· gave 29. These analogues, in particular 28, retained potency in
h/mL, F = 9%).118 Region-dependent intestinal absorption and vitro and in rats. Their metabolic stability improved relative to
meal composition effects on Cmax and AUC were found for somatostatin-14, however, no oral bioavailability was re-
dogs given the same dose via oral, duodenal, jejunal or colonic ported.123
8102 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 18. Cyclic somatostatin analogues. Cyclic hexapeptide (30)


and tri-N-methylated analogue (31). 30: MW = 807; miLogP = 0.5;
HBD = 9; HBA = 15; RotB = 11; tPSA = 228 Å2. 31: MW = 849;
miLogP = 1.3; HBD = 6; HBA = 15; RotB = 11; tPSA = 201 Å2.

turn in the macrocyclic ring and 2 hydrogen bonds between


side chains. The α-helical conformations contained 2 internal
and 2 side chain-side chain hydrogen bonds. In the solid state,
crystals of octreotide from aqueous oxalic acid (CCDC code:
YICMUS) contained two hydrogen bonds in linked β-turns
about the FwKT and wKTC motifs which, in conjunction with
the disulfide, produce a very rigid molecule.129 Octreotide
resisted degradation by enzymes and tissue homogenates.
When injected into rats and monkeys, octreotide showed the
greatest inhibition of growth hormone, insulin and glucagon
production. Furthermore, octreotide was better tolerated than
Figure 17. Structures of somatostatin and cyclic analogues (28−29). somatostatin and had ≥20 times longer duration of action.126
Somatostatin (27): MW = 1638; miLogP = −5.4; HBD = 26; HBA =
37; RotB = 26; tPSA = 613 Å2. 28: MW = 1404; miLogP = −0.7; HBD
Octreotide is orally active126,130,131 but the dose-corrected
= 19; HBA = 27; RotB = 19; tPSA = 428 Å2. 29: MW = 894; miLogP systemic oral bioavailability relative to subcutaneous admin-
= 0.1; HBD = 11; HBA = 17; RotB = 9; tPSA = 266 Å2. istration is very low (F = 0.3%).130

Replacing the -Cys-Aha-Cys- motif in 29 with the simpler


-Phe-Pro- motif gave analogue 30. Compounds 27 and 30 both
significantly reduced the effects of growth hormone following
sc injection to rats. Somatostatin-14, 27, and 30 were
administered orally to rats (25 mg/kg p.o.). Somatostatin
failed to lower growth hormone levels, 28 reduced levels at 1 h
but not at 2 h, while 30 reduced levels for at least 3 h. Peptide
28 was cleaved slowly by trypsin at a similar rate to 29.124 To
improve oral bioavailability and other pharmacokinetic proper-
ties, 30 N-methylated analogues of 30 were synthesized.125
Compound 30 and tri-N-methylated analogue 31 (Figure 18)
were administered orally to rats (10 mg/kg p.o.). Oral Figure 19. Somatostatin analogue 32 and octreotide (33). 32: MW =
bioavailability of 30 could not be determined, whereas 31 784; miLogP = −1.8; HBD = 13; HBA = 16; RotB = 10; tPSA = 277
Å2. 33: MW = 1019; miLogP = −2.0; HBD = 15; HBA = 20; RotB =
showed some oral absorption (F = 10%). Other pharmacody-
17; tPSA = 332 Å2.
namic properties showed that 31 had an extended elimination
half-life (74 vs 15 min) and increased steady state VD (3.7 vs 0.3
L/kg) relative to 30.125
Bauer (Sandoz Ltd.) reported a series of somatostatin-14 SDZ CO 611 (Ilatreotide, 34, Figure 20) is a D(+)-maltose
analogues based on a disulfide cyclized hexapeptide 32
N-terminally modified amadori derivative of octreotide 33. SDZ
containing an L-Phe-D-Trp-L-Lys motif (Figure 19).126 Cyclic
CO 611 (34) displayed improved metabolic stability
hexapeptide 32 was much less active than somatostatin-14
when tested in rats and monkeys. To regain activity, D-Phe was supposedly leading to enhanced oral absorption in rats (F =
added to the N-terminus to mimic the natural Phe-6 and 1.1%).132 Intravenous infusion of somatostatin and sc injection
significantly improved in vivo potency. Addition of threonol to of octreotide 33 suppress pancreatic and gastrointestinal
the C-terminus to mimic the natural Thr-12 gave octreotide hormones and accelerate early gastric emptying. Octreotide
(33, Sandostatin, Figure 19). NMR solution structural studies 33 also prolongs mouth to cecum transit time (MCTT). When
of octreotide127 and close analogues128 in DMSO-d6 (PDB administered orally (1 or 5 mg p.o. twice daily) to healthy
codes: 1SOC, 2SOC, 1YL8, and 1YL9) showed a mixture of β- males, 34 was equally effective as sc 33 in suppressing
sheet and partially α-helical motifs. The β-sheet like structures preprandial hormone levels, early gastric emptying and
contained 5 internal hydrogen bonds, 2 transannular and 1 β- prolonging MCTT.133
8103 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 20. SDZ CO 611 (ilatreotide, 34). MW = 1344; miLogP =


−4.9; HBD = 21; HBA = 32; RotB = 23; tPSA = 488 Å2.

4.6. Beauvericin and Enniatins


Beauvericin (35, Figure 21) is a symmetrical cyclic hexa- Figure 21. Beauvericin (35), enniatins A (R = Ile, 36), B (R = Val,
depsipeptide consisting of three repeating units of the 37), and C (R = Leu, 38), and enniatin B1 (39). 35: MW = 784;
dipeptides N-methyl-L-phenylalanine and the D-valine surrogate miLogP = 6.5; HBD = 0; HBA = 12; RotB = 9; tPSA = 140 Å2. 36:
MW = 682; miLogP = 6.0; HBD = 0; HBA = 12; RotB = 9; tPSA =
(R)-hydroxyisovaleric acid. There are no HBD in 35 or its
140 Å2. 37: MW = 640; miLogP = 4.5; HBD = 0; HBA = 12; RotB =
analogues 36−39, few rotatable bonds and low tPSA, but they 6; tPSA = 140 Å2. 38: MW = 682; miLogP = 6.1; HBD = 0; HBA =
do have RO5 violations for MW, HBA, and LogP (Figure 21). 12; RotB = 9; tPSA = 140 Å2. 39: MW = 654; miLogP = 5.0; HBD =
The crystal structure of beauvericin hydrate134 from n-heptane 0; HBA = 12; RotB = 7; tPSA = 140 Å2.
(CCDC code: BEVERC) did not show intramolecular
hydrogen bonds; however, solvent exposure of the hydrogen confirmed oral activity in infants by determining the
bond acceptors was evidenced in the solid state by complex- concentration of 40 in urine 24 h post oral dosage.148
ation of barium salts (CCDC code: BEAVBA).135 Compound
35 was first isolated from B. bassiana136 and inhibited
cytochrome P450 enzymes. Pharmacokinetic data following
oral administration at various doses to rats (0.5, 1.0, 2.0 mg/kg
p.o.: Cmax = 3.4, 5.4, 13.9 mg/L; Tmax = 4.1, 4.3, 5.4 h; CL =
0.29, 0.21, 0.32 mL/min·kg; AUC0‑∞ = 29, 80, 103 h·mg/L; t1/2
= 2.9, 3.6, 3.0 h, respectively)137 but no iv data was reported
making it difficult to estimate oral absorption. LD50 of 35 has
been reported in mice (LD50 ip >10 mg/kg, p.o. >100 mg/
kg).138,139 The beauvericin scaffold is shared by enniatins A-
C140 (Figure 21, 36−38), where L-phenylalanine is substituted
by L-isoleucine (36), L-valine (37), or L-leucine (38),
respectively. Several crystal structures of 37 and 38 have been
reported (CCDC entries DESYIJ, BICMEF, CIKJAH,
ZASQOZ, QEMCAM). As for beauvericin, there were no
intramolecular hydrogen bonds.141,142 However, 37 did Figure 22. Nepadutant (40). MW = 947; miLogP = −1.6; HBD = 11;
complex rubidium143 and potassium144 ions, indicating a HBA = 22; RotB = 11; tPSA = 348 Å2.
solvent accessible polar surface (CCDC codes: MVHIRB10,
PEKFEQ). Pharmacokinetic data were estimated for 37 from in
vitro assays.145 The toxicokinetic profile of enniatin B1 (39, 4.8. Bouvardin
Figure 21) was measured in piglets (0.05 mg/kg iv or p.o. in 2%
Bouvardin (41, Figure 23) is a cyclic hexapeptide containing
EtOH/water). Compound 39 was highly orally bioavailable (F
three N-methyl amides, one O-methylated tyrosine, one D-Ala,
91%) but was cleared rapidly (t1/2 = 0.15 h, CL = 2.00 L/h·kg)
and an additional diphenylether-bridge between two tyrosine
with low oral plasma exposure (AUC = 25.3 ng·h/mL).146
side chains. The crystal structure of 41 from methanol/water
4.7. Nepadutant (CCDC code: BOUVAR) did not show internal hydrogen
Nepadutant (40, Figure 22) is a glycosylated, bicyclic bonds but was in an extended conformation induced by the
hexapeptide tachykinin NK2 receptor antagonist. Cyclisation oxidatively fused tyrosine side chains. This bridged constraint
is achieved through both head-to-tail and side chain-to-side also introduced a cis-amide bond between the two residues.149
chain linkages and an aminohexose is attached to an asparagine It was isolated from B. ternifolia and showed in vitro
side chain. The aminohexose moiety gives the structure cytotoxicity against P388 lymphocytic leukemia and B16
amphiphilic character. Compound 40 was given orally to melanotic melanoma cell lines. Bouvardin and an acetylated
mice (38 mg/kg p.o. in castor oil; Tmax = 2.7 ± 0.7 h, Cmax = deoxybouvardin derivative 42 (Figure 23) had LD50 values of
1401 ± 220 nM, t1/2 = 11.7 ± 0.9 h, AUC = 8463 ± 2635 10 mg/kg (42, ip), 229 mg/kg (41, p.o.), and 20 mg/kg (42,
nmol·h/mL, F = 5%).147 Pediatric phase 1 clinical studies iv) in mice, indicating that 41 is orally absorbed.150 These
8104 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

compounds have few hydrogen bond donors and few rotatable t1/2 = 55 min, AUC0−24h = 4.5 mg·h/kg; 160 mg/kg p.o.: Cmax =
bonds, but they have high polar surface areas and low LogP. 10.4 mg/L, t1/2 = 76.2 min, AUC0−24h = 11.34 mg·h/kg).152

Figure 23. Bouvardin (41) and acetylated derivative 42. 41: MW =


773; miLogP = 0.5; HBD = 5; HBA = 16; RotB = 3; tPSA = 207 Å2.
42: MW = 799; miLogP = 1.2; HBD = 3; HBA = 16; RotB = 5; tPSA Figure 25. Antibacterial drug, NXL103 consisting of linopristin (45)
= 193 Å2. and floprestin (46). 45: MW = 950; miLogP = 1.6; HBD = 4; HBA =
19; RotB = 9; tPSA = 223 Å2. 46: MW = 532; miLogP = 1.5; HBD =
4.9. Pristinamycin and Related Antibiotics 2; HBA = 9; RotB = 1; tPSA = 122 Å2.
Pristinamycin is an oral antibiotic consisting of two synergistic,
but structurally unrelated, components. Separately, the 4.10. 1-NMe3 and Related Cyclic Hexapeptides
compounds exhibit bacteriostatic activity, while the combina- To permeate through membranes, molecules must transition
tion of pristinamycin IA (43) and IIA (44, Figure 24) has from aqueous to hydrophobic and back to aqueous phases.
bactericidal activity. 43 has one L-Pro, one N-methylated amide, Conformational changes might facilitate this process by
a D-Abu, and a 4-oxopiperidine incorporated into its backbone. lowering desolvation energies. To test this idea, molecular
44 bears a hybrid terpenoid/peptide backbone rigidified by modeling, H-D exchange experiments, and permeability
three alkenyl bonds, a 4,5-dehydroproline, and an oxazole ring. measurements were combined to produce passively permeable
A study in an Australian hospital investigated the effectiveness cyclic hexapeptide diastereomers from cyclo-[Leu-Leu-Leu-
of oral pristinamycin in patients with various bacterial Leu-Pro-Tyr]153 (Figure 26). None of the diasteromers had N-
infections.151 Patients were dosed orally with 500−1000 mg methyl groups; however, one compound, cyclo[Leu-D-Leu-Leu-
of 43 and 44 on multiple days. Of 46 patients, five were Leu-D-Pro-Tyr], showed a conformational change between
excluded due to side effects, 10 were cured from infection, and CDCl3 and water by molecular modeling. This diastereomer
21 had infections suppressed. was the least permeable in the initial series even though higher
permeability had previously been reported for the same
peptide.154 In further studies, tri-N-methylation yielded 1-
NMe3 47 which had good PAMPA permeability and oral
bioavailability in rats (1 mg/kg iv: CL = 4.5 mL/min·kg, VD =
1.1 L/kg; 10 mg/kg p.o.: AUC = 10.5 ng·h/mL, Cmax = 852 ng/
mL, F = 28%).155 Analogues of 47 varied a single side chain
substitution to learn effects on pharmacokinetic profiles.156 An
N-methyl-leucine was substituted for N-methyl-serine (48), N-
methyl-threonine (49), N-methyl-aspartate (50), or N-methyl-
lysine (51). Their pharmacokinetic profiles revealed that
increasing side chain polarity reduced oral absorption.
Interestingly, threonine analogue 49 (1 mg/kg iv: CL = 63.6
mL/min·kg, VD = 3.8 L/kg, t1/2 = 1.0 h; 10 mg/kg p.o.: AUC =
201 ng·h/mL, Cmax = 105 ng/mL, F = 24%) displayed similar
Figure 24. Antibacterial combination drug, pristinamycin consisting of
pristinamycins IA (43) and IIA (44). 43: MW = 867; miLogP = 0.7;
pharmacokinetics as 47, while serine analogue 48 had low oral
HBD = 4; HBA = 18; RotB = 7; tPSA = 228 Å2. 44: MW = 526; absorption (1 mg/kg iv: CL = 60.4 mL/min·kg, VD = 4.3 L/kg,
miLogP = 0.7; HBD = 2; HBA = 10; RotB = 1; tPSA = 139 Å2. t1/2 = 1.5 h; 10 mg/kg p.o.: AUC = 42.3 ng·h/mL, Cmax = 9.86
ng/mL, F = 2%). Aspartate analogue 50 (1 mg/kg iv: CL =
56.4 mL/min·kg, VD = 0.36 L/kg, t1/2 = 0.4 h; 10 mg/kg p.o.:
AUC = 18.7 ng·h/mL, Cmax = 17.3 ng/mL, F < 1%) and lysine
NXL103 (XRP2868; Figure 25) is also an antibiotic cocktail analogue 51 (1 mg/kg iv: CL = 10.4 mL/min·kg, VD = 0.9 L/
made of linopristin 45 and floprestin 46 (30:70 mixture). These kg, t1/2 = 1.0 h; 10 mg/kg p.o.: AUC = 18.1 ng·h/mL, Cmax =
compounds replace three hydrogen bond donor NHs with 5 or 18.3 ng/mL, F < 1%) had negligible oral bioavailability (F <
6 membered rings and a depsipeptide ester, 45 also has an N- 1%). NMR structural studies of this series of peptides153−155 in
methyl group. Pharmacokinetic parameters were measured after CDCl3 and DMSO-d6 suggested that the pattern of internal
oral administration at multiple doses to cyclophosphamide- hydrogen bonds was strongly conserved.
induced neutropenic mice infected with strains of S. pneumoniae The same orally bioavailable scaffold (47) inspired further
(NXL103: 10 mg/kg p.o.: Cmax = 0.32 mg/L; 40 mg/kg p.o.: investigations to better understand and improve passive
Cmax = 0.8 mg/L, t1/2 = 21 min; 80 mg/kg p.o.: Cmax = 4 mg/L, permeability of cyclic hexapeptides. N-Methylamino acid-to-
8105 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

enter cells and interact with intracellular targets. β-hydroxy-γ-


amino acids were used to create cyclic hexapeptides based on
47. Oral bioavailability in rats was reported for one compound,
55, displaying promising passive permeability (1 mg/kg iv: CL
= 10 mL/min·kg, VD = 1.1 L/kg, t1/2 = 1.6 h; 5 mg/kg p.o.;
AUC = 1697 ng·h/mL, Cmax = 324 ng/mL, F = 21%).158
Although good permeability was maintained in the less peptidic
55, its oral bioavailability and plasma concentration (AUC)
were worse than for 47. Interestingly, NMR evidence suggested
that, despite variability in the membrane permeability of some
peptoid and statin-type analogues of 47, these compounds still
retained the two internal hydrogen bonds shown in Figure
Figure 26. 1-NMe3 (47) and derivatives (48−51). 47: MW = 755;
27.157,158
miLogP = 4.1; HBD = 3; HBA = 13; RotB = 10; tPSA = 160 Å2. 48: Other groups have used 47 and analogues to investigate
MW = 729; miLogP = 1.8; HBD = 4; HBA = 14; RotB = 9; tPSA = effects of N-methylation,159 correlating amide NH NMR
180 Å2. 49: MW = 743; miLogP = 2.1; HBD = 4; HBA = 14; RotB = temperature coefficients with cell permeability160 and flexi-
9; tPSA = 180 Å2. 50: MW = 757; miLogP = 1.9; HBD = 4; HBA = bility/rigidity.161 A crystal structure of 47 (CCDC code:
15; RotB = 10; tPSA = 197 Å2. 51: MW = 770; miLogP = 2.3; HBD = AHELEG) indicated a saddle-like structure incorporating two
5; HBA = 14; RotB = 12; tPSA = 186 Å2. Dotted lines indicate β-turns at either end, with one close hydrogen bond in each
hydrogen bonds observed in crystal structure of 47, and NMR solution (2.0, 2.5 Å, indicated by dotted lines, Figure 26).159 Novartis
structures of analogues. researchers compared the original scaffold of 47 with its non-N-
methylated counterpart (56, Figure 28) and showed that N-
peptoid substitutions were explored in cyclic hexapeptide/ methylation was important for oral absorption. Permeability
peptoid hybrids (Figure 27). Peptoid substitutions that enhancers did not improve oral bioavailability of 47.
replaced one (52) or two (53) of the three N-methyl amides Pharmacokinetics were measured in mice for non-N-methylated
facilitated permeability across a monolayer of low efflux analogue 56 (3 μM/kg iv: CL = 82 mL/min·kg, VD = 0.8 L·kg,
epithelial MDCK cells. Interestingly, replacing all three N- t1/2 = 0.2 h, AUCiv = 292 nM·h; 10 μM/kg p.o.: AUCpo = 5.7
methyl moieties with peptoid units (54) generated a macro- nM·h, Cmax = 2.9 nM, Tmax = 0.6 h, F = 2%) and 47 (3 μM/kg
cycle that was less permeable.157 iv: CL = 1 mL/min·kg, VD = 0.4 L/kg, t1/2 = 22 h, AUCiv =
β-Hydroxy-γ-amino acids are naturally occurring peptidomi- 29618 nM·h; 10 μM/kg p.o.: AUC = 8967 nM·h, Cmax = 534
metic surrogates that structurally resemble a peptide backbone nM, Tmax = 3.8 h, F = 30%). These striking results highlight
by replacing heteroatoms with carbon to minimize polarity. good oral absorption for 47 in rodents.
These substructures may have evolved to allow molecules to

Figure 28. Non-N-methylated analogue 56 and bis-N-methylated


analogue 57 with a different N-methylation pattern to 47 retained oral
bioavailability. 56: MW = 713; miLogP = 3.3; HBD = 6; HBA = 13;
RotB = 10; tPSA = 186 Å2. 57: MW = 741; miLogP = 3.8; HBD = 4;
HBA = 13; RotB = 10; tPSA = 168 Å2. Dotted lines indicate conserved
hydrogen bonds observed by NMR spectroscopy.

Cyclic hexapeptides based on 47 were examined with


shuffled N-methyl amino acids and inverted stereochemistries.
Correlations between amide NH NMR temperature coef-
ficients, Caco-2 and PAMPA permeability led to cyclic
hexapeptide 57 (Figure 28). Compared to 47, 57 had one
Figure 27. Peptoid substituted analogues 52−54 and β-hydroxy-γ- less N-methyl group and a modified N-methylation pattern but
amino acid analogue 55. 52: MW = 741; miLogP = 3.5; HBD = 3;
HBA = 13; RotB = 10; tPSA = 160 Å2; 53: MW = 727; miLogP = 3.6;
was still orally bioavailable in rats (1 mg/kg iv: CL = 55 mL/
HBD = 3; HBA = 13; RotB = 10; tPSA = 160 Å2; 54: MW = 713; min·kg, VD = 1.1 L/kg, t1/2 = 29 min; 10 mg/kg p.o.: AUC =
miLogP = 3.6; HBD = 3; HBA = 13; RotB = 10; tPSA = 160 Å2; 55: 1003 ng·h/mL, Cmax = 117 ng/mL, F = 33%).141
MW = 793; miLogP = 3.6; HBD = 4; HBA = 14; RotB = 10; tPSA = Peptide 47 was thought to be orally bioavailable due to
180 Å2. Dotted lines indicate conserved hydrogen bonds observed by cyclosporin-like conformational flexibility; however, it showed
NMR spectroscopy. no conformational difference in polar versus nonpolar
8106 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

solvents.161 Several analogues of 47 were synthesized and their


pharmacokinetic properties characterized to investigate con-
formational flexibility versus rigidity. The structure of 47 was
rigidified by introducing a proline (58, Figure 29) or relaxed by
introducing N-methylleucine (59, Figure 29). Both cyclic
hexapeptides 58 (1 mg/kg iv: CL = 11 mL/min·kg, t1/2 = 58
min; 10 mg/kg p.o.: AUC = 4320 ng·h/mL, Cmax = 879 ng/mL,
F = 30%) and 59 (1 mg/kg iv: CL = 10 mL/min·kg, t1/2 = 121
min; 10 mg/kg p.o.: AUC = 3713 ng·h/mL, Cmax = 768 ng·mL,
F = 18%) were appreciably orally bioavailable in rats.161 For
these compounds (59, 47, and 58) with identical HBD, HBA,
and tPSA (Figure 29), reducing the MW (785 > 755 > 725),
the number of rotatable bonds (12 > 10 > 8), and the Figure 30. Cyclic cell-penetrating hexapeptide (60). MW = 969;
hydrophilic surface area (FISA 127.5 > 122.5 > 110.4 Å2) all miLogP = −2.9; HBD = 22; HBA = 24; RotB = 24; tPSA = 422 Å2.
improved oral bioavailability (F% = 18−16 (28155) < 30, under
the same conditions161). These changes had little effect on the absorption. Preclinical toxicity studies determined LD50 in rats
overall structure of the macrocycle, as shown by NMR at 0.38−0.60 mg/kg iv.169
spectroscopy in CDCl3 and DMSO-d6, with two rigidifying
antiparallel β-sheets connected by type 1 β-turns at each end
(dotted lines, Figure 29).

Figure 31. Kahalalide F (61). MW = 1492; miLogP = 3.0; HBD = 15;


HBA = 30; RotB = 34; tPSA = 442 Å2.

Figure 29. Derivatives of 47 with increased (58, left) or decreased (59,


right) rigidity. 58: MW = 725; miLogP = 2.8; HBD = 3; HBA = 13;
RotB = 8; tPSA = 160 Å2. 59: MW = 785; miLogP = 5.3; HBD = 3; 5. CYCLIC HEPTAPEPTIDES
HBA = 13; RotB = 12; tPSA = 160 Å2. Dotted lines indicate conserved 5.1. Sanguinamide A and Danamides
hydrogen bonds observed by NMR spectroscopy.
Sanguinamide A (62, Figure 32) is a thiazole-containing cyclic
heptapeptide isolated from H. sanguineus.170 It has six natural L-
4.11. Cyclo-[Arg-Arg-Arg-Arg-NaphthylAla-Phe] amino acids including two prolines that adopt a cis, trans
configuration, and an isoleucine-derived thiazole.171 No
Cell penetrating peptides (CPPs) are a small group of short,
biological activity is known for sanguinamide A, but it is orally
natural and synthetic, peptides that are often arginine rich,
bioavailable in rats (1 mg/kg iv in DMSO: t1/2 = 23 min, CL =
positively charged, amphiphilic, and able to cross cell
70 mL/min; 10 mg/kg p.o.: Tmax = 60 min, Cmax = 40 nM, F = 7
membranes. How they cross membranes is still not fully
± 4%).171 The three-dimensional structure helped rationalize
resolved but mechanisms include endocytosis and direct
the pharmacokinetic profile of 62. Two amide protons formed
translocation across the membrane.162−164 A series of
intramolecular hydrogen bonds and were shielded from solvent
arginine-rich, cell-penetrating, cyclic hexapeptides were synthe-
by bulky side chains, while two prolines and thiazole removed 3
sized, and their cellular uptake was measured. One analogue,
amide NH protons, lowering the HBD count. NMR studies on
cyclic hexapeptide 60 (Figure 30), was orally administered to
analogues of 62 examined H-D exchange rates and amide
mice and pharmacokinetic parameters were measured (1.5 mg/
temperature coefficients and determined NMR solution
kg iv: Cmax = 12174 nmol/L, t1/2 = 1.02 h, CL = 0.08 mL/min,
structures in d6-DMSO. These data guided introduction of
VD = 7.51 mL; 40 mg/kg p.o.: Cmax = 3156 nmol/L, AUC =
the bulky tert-butylglycine to minimize solvent exposed polarity,
6357 nmol/L, F = 4%) showing it had some oral
resulting in danamide D (63, Figure 32) that was orally
bioavailability.165
bioavailable (1 mg/kg iv in DMSO: CL = 12.5 mL/min·kg, t1/2
4.12. Kahalalide F = 65 min; 10 mg/kg p.o. in olive oil: Cmax = 352 ng/mL, Tmax =
Kahalalide F (61, Figure 31) is the largest depsipeptide in the 240 min, AUC = 2647 ng·h/mL, F = 21 ± 2%). When an N-
family of kahalalides (A-F) and was isolated from the methyl group was removed (danamide F, 64, Figure 32), even
sacoglossan mollusk E. rufescens and the green alga Bryopsis.166 better oral bioavailability was obtained (1 mg/kg iv in DMSO:
Compound 61 induced oncosis in human prostate and breast CL = 23.0 mL/min kg, t1/2 = 97 min; 10 mg/kg p.o. in olive oil:
cancer cells.167 Pharmacokinetic parameters were measured in Cmax = 726 ng/mL, Tmax = 240 min, AUC = 3372 ng·h/mL, F =
Phase I clinical trials (iv: t1/2 = 0.47 h, CL = 11.0 L/h, VD = 7.0 51 ± 9%). This result highlighted the point that N-methylation
L).168 Kahalalide F had LD50 of 300, 980, and 3200 mg/kg p.o. of solvent accessible amides does not necessarily always
in mouse, rat, and rabbit, respectively, suggesting very low oral improve oral bioavailability.172
8107 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

lethal dose (LD100 = 65 mg/kg ip). Distribution studies in


mice177 indicated uptake from the small intestine with
concentration in the liver, lung, and heart.

Figure 32. Sanguinamide A 62 (R1 = H, R2 = Me), danamide D 63


(R1 = Me, R2 = tBu), and danamide F 64 (R1 = H, R2 = tBu). 62: MW
= 722; miLogP = 2.0; HBD = 4; HBA = 13; RotB = 6; tPSA = 170 Å2. Figure 34. Microcystin-LR (66). MW = 995; miLogP = −3.5; HBD =
63: MW = 778; miLogP = 3.6; HBD = 3; HBA = 13; RotB = 7; tPSA 12; HBA = 22; RotB = 16; tPSA = 341 Å2.
= 161 Å2. 64: MW = 764; miLogP = 3.3; HBD = 4; HBA = 13; RotB =
7; tPSA = 170 Å2. Dotted lines represent hydrogen bonds as
determined by NMR spectroscopy.171 5.4. YM254890
YM254890 (67, Figure 35) is a specific Gαq/11 inhibitor
5.2. Rhizonin A produced by an isolate of the chromobacterium sp. QS3666. The
Rhizonin A (65, Figure 33) is cyclo-[N(Me)Ala-N(Me)D- structure is a depsipeptide with 2 ester linkages and a D-
Ala(fur-3-yl)-D-aIle-D-Val-Val-N(Me)Ala(fur-3-yl)-Leu]. It was phenylalanine derivative. Gαq/11, a G protein implicated in
isolated from fungi R. microspores, a known infection in fruits, purine nucleotide-induced platelet aggregation, was inhibited
vegetables and malt products. This cyclic heptapeptide consists by 67 by targeting the transformation of GDP to GTP. In rats,
of L- and D-amino acids, three N-methyl amides and two (2- 67 inhibited ex vivo platelet aggregation and in vitro thrombus
furyl)-alanines in both (R)- and (S)-configurations.173 In formation, suggesting it may lead to a new antithrombotic
crystals of 65 grown from ethyl acetate/hexane, the four agent. It has been used orally to treat mice with acute
bulky side chains including three β-branched residues were at thrombosis and chronic neointima formation after induced
the wide end of the ovoid macrocycle. The other end showed a vascular injury. Internal bleeding in mice was also effectively
transannular hydrogen bond between the Ile NH and D- treated with 67 at 0.03 mg/kg iv or 1 mg/kg p.o., indicating an
(furylalanine)-carbonyl oxygen.173 Rhizonin A 65 was given oral absorption < 3%.178
orally to male albino rats (70, 96, 131, or 180 mg/kg p.o.) to
determine the oral LD50. The lowest dose exceeded LD100 as all
rats died within 10 days.174 There was no data for iv
administration, so oral bioavailability could not be quantified.

Figure 35. YM254890 (67). MW = 960; miLogP = −0.2; HBD = 5;


HBA = 22; RotB = 13; tPSA = 288 Å2.

Figure 33. Rhizonin A (65). MW = 812; miLogP = 2.6; HBD = 4;


HBA = 16; RotB = 10; tPSA = 204 Å2. Dotted line represents 5.5. CHEC-7
hydrogen bond observed in crystal structure. CHEC refers to a series of peptides of various sizes derived
from the natural survival anti-inflammatory protein dermicidin.
5.3. Microcystin LR CHEC-7 (68, Figure 36) reduced the incidence of experimental
autoimmune encephalomyelitis in a disease model when
Microcystin-LR (66, Figure 34) from M. aeruginosa is one of
administered to rats (1.5 mg/kg p.o. or 0.1 mg/kg/day sc).179
the most toxic of over 80 microcystins produced by
cyanobacteria in aquatic environments. Its amino acid sequence 5.6. Polymyxin B1 and B2
is D-alanine, L-leucine, D-methylaspartic acid, variable L-amino Polymyxin B (Figure 37) is a mixture of two natural product
acid, a 3-amino-9-methoxy-2,6,8-trimethyl-10-phenyldeca-4,6- cyclic peptides, polymyxin B1 69 and B2 70, containing multiple
dienoic acid (Adda), D-glutamic acid, and N-methyldehydroa- unusual diaminobutyric acids and a D-Phe residue. Prolonged
lanine. The extensively modified cyclic peptide is highly water- treatment with polymyxin B mixture could not neutralize
soluble and a persistent contaminant that accumulates in fish lipooligosaccharide-induced immune cell activity in mice.
and displays effects on their liver, gastrointestinal tract, and However, modulation of this response was observed by oral
kidneys.175 Acute hepatotoxic effects especially apoptotic administration in drinking water (29.5 mg/L p.o. suspension in
hepatocyte death were observed in mice,176 although the oral water).180 Human patients with liver disease have been treated
toxic dose (LD50 = 11 mg/kg p.o.) is ∼170 fold lower than the with enteric coated oral polymyxin B. Plasma endotoxin and
8108 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 36. CHEC7 (68). MW = 759; miLogP = −5.6; HBD = 3; HBA


= 21; RotB = 9; tPSA = 347 Å2.
Figure 38. Bacitracin A (71). MW = 1437; miLogP = −1.8; HBD =
ammonia levels were reduced, even though polymyxin B itself 20; HBA = 33; RotB = 32; tPSA = 531 Å2.
could not be detected above 0.5 units/mL.181
inhabited the gut of the test animals. Further work on
analogues of 72 in chickens suggested that certain functional
groups, lipophilicity, and PSA influenced anthelmintic activity
in vivo.191

Figure 37. Polymyxin B1 (69) and B2 (70). 69: MW = 1189; miLogP


= −5.6; HBD = 23; HBA = 29; RotB = 28; tPSA = 491 Å2. 70: MW =
1203; miLogP = −5.5; HBD = 23; HBA = 29; RotB = 29; tPSA = 160
Å2. Figure 39. PF1022A (72) and emodepside (73). 72: MW = 949;
miLogP = 7.9; HBD = 0; HBA = 16; RotB = 12; tPSA = 186 Å2. 73:
MW = 1119; miLogP = 7.8; HBD = 0; HBA = 20; RotB = 14; tPSA =
5.7. Bacitracin A 211 Å2.
Bacitracin A (71, Figure 38) is a cyclic thiazoline-containing
peptide from a family of cyclic peptides isolated from the
“Tracy I” strain of B. subtilis. Bacitracins are an intriguing group Emodepside (BY 44-4400, 73, Figure 39), a dimorpholine
of non-ribosomal peptides containing a sequence of alternating derivative of 72, has been approved for use in animals. Four
L - and D-amino acids. Bacitracin A was compared to
different crystal forms have been reported from methanol.192
vancomycin for treating antibiotic-associated diarrhoea. Plasma With no possibility of forming transannular hydrogen bonds,
and urine concentrations were measured to quantify unwanted the carbonyl groups are directed above and below the plane of
systemic absorption. 71 was found in plasma at low non-toxic the macrocycle and are highly solvent exposed, one D-lactate
concentrations after oral administration.182 Another study in side chain occupying the interior (CCDC code: DOMZOW).
which 71 was administered orally to dogs found significant Compound 73 was orally active against nematodes and orally
levels in blood and urine, confirming absorption from the bioavailable (F = 47−54%). It had a long elimination half-life
intestinal tract.183 Bacitracin A has been investigated for the following intravenous administration to rats (t1/2 = 39−51
treatment of vancomycin-resistant E. faecium in humans. In h).189,190 A phase I clinical trial investigating oral dosing of 73
combination with doxycycline, 71 was not efficacious beyond in normal healthy male volunteers is ongoing.193
the 2-week interval during which they were given.184 71 has 6.2. WH1Fungin
oral toxicity in mice (LD50 > 3750 mg/kg p.o.).185 WH1Fungin (74, Figure 40) is a surfactin type lipo-
depsipeptide extracted from B. amyloliquefaciens WH1. As an
6. CYCLIC OCTAPEPTIDES immunoadjuvant, 74 was orally active in mice (0.2 mg) and
6.1. PF1022A and Emodepside enhanced the immune response to coadministered protein
antigens (chicken ovalbumin or glutathione S-transferase).194
PF1022A (72, Figure 39) is a cyclic octapeptide natural product
74 was also shown to reduce blood glucose and increase serum
with anthelmintic activity first isolated in 1992 from the fungus
insulin in non-obese diabetic mice following oral administration
M. sterilia. It contains four N-methyl-L-leucine residues
at two doses (5 or 25 mg/kg p.o. in 100 μL PBS).195
alternating with D-lactic acid or D-phenyllactic acid. PF1022A
(72) was orally active (0.5−2.0 mg/kg p.o.) and effective 6.3. α-Amanitin
against A. galli nematode (ring worm) infections in chickens The poisonous mushrooms of the Amanita genus contain a
(0.5−2.0 mg/kg p.o.)186 and nematode infections in rats (5 or number of related constrained cyclic peptides of MW = 770-
10 mg/kg p.o.),187 sheep (1 mg/kg p.o.),188 and other 920. These are toxic after ingestion and include phallotoxins,
species.189,190 These data gave no insights into intestinal amatoxins, and virotoxins.196,197 Some of these toxins are heat
absorption of 72 following oral dosing, since the nematodes stable and are not destroyed by cooking. Phallotoxins from A.
8109 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 40. WH1Fungin (74). MW = 1064; miLogP = 8.0; HBD = 9;


HBA = 20; RotB = 27; tPSA = 345 Å2.
Figure 41. α-Amanitin (75). MW = 889; miLogP = −4.1; HBD = 13;
phalloides are bicyclic heptapeptides, amatoxins (A. phalloides) HBA = 23; RotB = 7; tPSA = 361 Å2.
are bicyclic octapeptides, and virotoxins (A. virosa) are
monocyclic heptapeptides.198 Despite extensive modifications Pharmacokinetics were improved by simple structural mod-
(e.g., leucine, proline hydroxylation, D-configuration, thia-cross- ifications. Griselimycin contains an N-methyl-D-leucine, one
link) these are ribosomally generated peptides.199 A systematic proline, and two 4-methylproline residues. Modification of the
examination of their oral absorption has not been carried out 4-position of proline gave methyl-griselimycin 77 (3 mg/kg iv:
but they display high toxicity. CL = 35 mL/min·kg, VD = 1.6 L/kg, t1/2 = 144 min; 30 = mg/
α-Amanitin (75, Figure 41) is a member of the group of kg p.o.: AUC = 6600 ng·h/mL, Cmax = 2820 ng/mL, F = 47%),
poisonous peptides called amatoxins; all L-configured bicyclic dimethyl-griselimycin 78 (3 mg/kg iv: t1/2 = 132 min; 30 mg/
octapeptides found in Amanita species of mushrooms. kg p.o.: AUC = 17000 ng·h/mL, Cmax = 4570 ng/mL, F =
Amatoxins contain an oxidized tryptathionine linkage in 59%), (S)-fluoro-griselimycin 79 (3 mg/kg iv: t1/2 = 120 min;
addition to backbone cyclization. Multiple crystal structures 30 mg/kg p.o.: AUC = 5100 ng·h/mL, Cmax = 1490 ng/mL, F =
(PDB codes: 1K83, 2VUM, and 3CQZ)200−202 of α-amanitin 55%), and cyclohexyl-griselimycin 80 (3 mg/kg iv: CL = 18.3
(75) bound to RNA polymerase II showed, in addition to six mL/min·kg, VD = 5.5 L/kg, t1/2 = 258 min; 30 mg/kg p.o.:
hydrogen bonding interactions to the enzyme, two transannular AUC = 23000 ng·h/mL, Cmax = 2620 ng/mL, F = 89%); all
hydrogen bonds and two more describing β-turns within the α- apparently with improved oral bioavailability and pharmacoki-
amanitin framework. Although native unbound α-amanitin netic properties.208 Griselimycin and the cyclohexyl analogue
crystal structures are not available, simple OMe and sulfone 80 have each been crystallized in complex with the sliding
analogues (CCDC entries CAZFIS, CAZFIS10, CAZFOY, clamp of Mycobacterium tuberculosis (PDB code: 5AH2, 5AGU,
CAZFOY10) show similar structures to the protein bound 5AH4, and 5AGV).208 The bound griselimycins showed two
forms above, with two β-turns, one transannular hydrogen internal hydrogen bonds, one γ-turn from the Leu CO
bond is lost in the free form.203,204 Interestingly, it appears that preceding MePro to the Leu NH after it, one transannular
the toxicity of the amanitins can be modulated by the degree hydrogen bond between the Gly NH and the N-Me-Leu
and stereochemistry of oxidation at the bridging sulfur, with the carbonyl group, and one transannular hydrogen bond between
sulfone analogues being the most toxic, while retaining nearly the Gly NH and the N-Me-Leu carbonyl group.
all structural features of the peptidic backbone. α-Amanitin
causes severe liver damage and in some cases death (50−100 6.5. Dihydromycoplanecin and Mycoplanecin
fatalities per year in Europe) through acute liver failure.205 75 is Dihydromycoplanecin A (81, Figure 43) was discovered as an
orally active only in certain species. The lethal oral dose in active metabolite in the urine of dogs and mice treated with the
humans has been estimated at 0.1 mg/kg from accidental antibiotic mycoplanecin A (82, Figure 43). Both 81 and 82 are
deaths. In guinea pigs, identical toxicity following oral, cyclized through a depsipeptide linkage between the C-
intravenous and intraperitoneal administration (LD50 = 0.1 terminus and a threonine residue. Further modifications include
mg/kg) indicated good intestinal absorption. By contrast, four N-methylated amides, 4-methyl and 4-ethylsubstituted
toxicity in mice (LD50 = 0.4−0.8 mg/kg iv) and rats (LD50 = prolines, L-homoleucine and α-hydroxy (81) or α-ketobutanoyl
3.5−4.0 mg/kg iv) is only observed following iv administration. (82) N-terminal caps. No oral pharmacokinetic data has been
Cats and dogs absorb amatoxins following oral administration, reported for 82, but oral toxicity has been reported (LD50 > 3
but the absorption rate is slow. In dogs, the toxic oral dose is g/kg p.o.).210 Pharmacokinetic parameters for 81 have been
five times higher than the toxic intravenous dose (LD50 = 0.5 measured in mice (t1/2 = 0.5 h; 50 mg/kg p.o.: Cmax = 10 μg/
mg/kg p.o. vs 0.1 mg/kg iv). In cats, the toxic oral dose was mL at 2 and 4 h) and dogs (t1/2 = 5.5 h; 25 mg/kg p.o.: Cmax =
more than ten times higher than the toxic intravenous dose.206 9 μg/mL at 3 h; 12.5 mg/kg p.o.: Cmax = 5 μg/mL at 3 h).
6.4. Griselimycin and Synthetic Derivatives Quantification of 81 from urine led to estimates of F = 21% in
mice and 22−25% in dogs.211
Griselimycin (76, Figure 42) is a macrocyclic poly-N-
methylated depsipeptide isolated from Streptomyces bacteria 7. CYCLIC NONA- AND DECA-PEPTIDES
cultures.207 Griselimycin derivatives exhibit antibiotic activity,
oral bioavailability, metabolic stability, and antitubercular 7.1. CHEC-9
activity.208,209 Griselimycin has impressive pharmacokinetics Disulfide-cyclized nonapeptide CHEC-9 (83, Figure 44) is a
for its chemical class and size (3 mg/kg iv: CL = 40 mL/min· putative inhibitor of Heat Shock Protein 70 and secreted
kg, VD = 1.2 L/kg, t1/2 = 120 min; 30 = mg/kg p.o.: AUC = phospholipase A2. Following oral administration to rats (1 mg/
5100 ng·h/mL, C max = 3900 ng/mL, F = 48%). 208 kg p.o. in gelatin solution) it was found in cytosolic fractions of
8110 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

7.2. AFPep
AFPep (84, Figure 45) is a cyclic analogue of the 472−479
fragment from alpha-fetoprotein (AFP). AFP inhibits estrogen-
stimulated growth of immature mouse uterus and estrogen-
dependent proliferation of human breast cancer cells.213 The
linear peptide fragment has a bench life of a few weeks and an
undesirable biphasic dose−response curve. Cyclization, sub-
stitution of proline for hydroxyproline, and addition of
asparagine to the C-terminus gave AFPep (84).214 AFPep
was orally active and arrested growth of human tumor
xenografts in mice (10 μg/mouse p.o.). It also decreased the
incidence and multiplicity of breast cancers in carcinogen-
exposed rats (100 μg/rat p.o.). In these animal models, AFPep
had similar effects when administered via oral, ip or sc
routes.215,216
Figure 42. Natural product griselimycin (76, R1 = R2 = H) and
synthetic derivatives: methyl-griselimycin (77, R1 = Me, R2 = H),
dimethyl-griselimycin (78, R1 = R2 = Me), (S)-fluoro-griselimycin (79,
R1 = F, R2 = H), and cyclohexyl-griselimycin (80, R1 = cyclohexyl, R2
= H). 76: MW = 1113; miLogP = 4.3; HBD = 3; HBA = 22; RotB =
12; tPSA = 256 Å2. 77: MW = 1127; miLogP = 5.6; HBD = 3; HBA =
22; RotB = 12; tPSA = 256 Å2. 78: MW = 1142; miLogP = 5.1; HBD
= 3; HBA = 22; RotB = 12; tPSA = 256 Å2. 79: MW = 1131; miLogP
= 4.3; HBD = 3; HBA = 22; RotB = 12; tPSA = 256 Å2. 80: MW =
1196; miLogP = 7.5; HBD = 3; HBA = 22; RotB = 13; tPSA = 256 Å2.

Figure 45. AFP fragment 472−479: MW = 844; miLogP = −5.1;


HBD = 12; HBA = 22; RotB = 23; tPSA = 350 Å2. AFPep (84): MW =
Figure 43. Dihydromycoplanecin (81) and mycoplanecin (82). 81: 969; miLogP = −5.4; HBD = 17; HBA = 28; RotB = 13; tPSA = 455
MW = 1186; miLogP = 5.2; HBD = 4; HBA = 23; RotB = 16; tPSA = Å2 .
276 Å2. 82: MW = 1184; miLogP = 5.0; HBD = 3; HBA = 23; RotB =
16; tPSA = 273 Å2.
7.3. Antamanide and Cyclolinopeptide
Antamanide (85, Figure 46) is a cyclic decapeptide derived
the frontal cortex tissues. This indicated both oral absorption from the same fungus as α-amanitin 75 (A. phalloides).
from the gut and blood-brain-barrier penetration,212 despite Antamanide protects against phalloidin poisoning198 by block-
violating all RO5 and like parameters (Figure 44). ing the organic anion transporting polypeptide mechanism.
This inhibits the uptake of phalloidins into hepatocytes. Thus,
antamanide functions by a mechanism similar to immunosup-
pressants cyclosporin A and FK506.217 A related hydrophobic
cyclic nonapeptide cyclolinopeptide (86) found in linseed oil
displayed immunosuppressive activity in a mouse model
following oral administration (10 or 100 μg/mouse p.o. in
olive oil).218 A crystal structure of 85 from acetonitrile/water
(CCDC code: ANTAHC10) showed two transannular hydro-
gen bonds in a large bowl-shaped macrocycle. The concave face
displayed most of the polar atoms, including all hydrogen bond
donors. These NH groups were almost entirely shielded by the
hydrophobic side chains.219 Two structures of 86 (CCDC
codes: GIPKAR10, POWWEE) from isopropanol/water and
methanol/water both showed three internal hydrogen
Figure 44. CHEC-9 (83). MW = 917; miLogP = −6.0; HBD = 16; bonds.220,221 One formed a β-turn around residues LIIL and
HBA = 26; RotB = 10; tPSA = 425 Å2. two transannular interactions. A conserved water molecule was
8111 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

tightly held to a polar patch composed of 2 phenylalanine NH


and two proline carbonyl oxygens.

Figure 48. Permetin A (88). MW = 1101; miLogP = −1.8; HBD = 16;


HBA = 24; RotB = 18; tPSA = 386 Å2.

were modified. Their in vitro permeability, metabolic stability


Figure 46. Antamanide (85) and cyclolinopeptide (86). 85: MW =
1147; miLogP = 2.4; HBD = 6; HBA = 20; RotB = 9; tPSA = 256 Å2. and other pharmacokinetic parameters were evaluated in rats
86: MW = 1040; miLogP = 4.6; HBD = 7; HBA = 8; RotB = 13; tPSA (Table 1). These were the first examples of cyclic peptides of
= 244 Å2. comparable size to cyclosporin A (109) with comparable or
greater oral bioavailability.
7.4. Cyclopeptolide 1 7.7. Surotomycin
Cyclopeptolide 1 (87, Figure 47) is a cyclic 10-residue peptide Surotomycin (CB-183315, 108, Figure 49) is a macrocyclic
featuring five N-methyl amides, a depsi-peptide, an O-methyl antimicrobial lipodepsipeptide, containing multiple unusual and
tyrosine, and a pipecolic acid. Isolated from the fungus Septoria D-amino acids. Originally from Cubist Pharmaceuticals, 88 was
sp., 87 was used to treat systemic and vaginal candidosis in rats in development by Merck for treatment of C. dif f icile infections
by twice daily oral dosing for 4 days (8 × 200 mg/kg p.o.). A (CDI) and associated diarrhea. In hamsters, 108 was as
90% cure rate was observed compared to 100% for the effective as vancomycin when administered orally 12 h
antifungal agent Ketoconazole.222 postinfection twice-daily for five consecutive days (2, 10, or
25 mg/kg p.o. in water).226 In clinical trials, orally administered
108 (125 and 250 mg p.o.) was safe and well tolerated in
humans and proved more effective than vancomycin in the
treatment of CDI.227 CDI occur in the gut, therefore low
intestinal absorption and systemic availability is desirable. Low
oral absorption was reported in rats (F < 1%)228 and
pharmacokinetic parameters from clinical trials also indicated
low systemic absorption in humans at various doses (500 mg
p.o.: Cmax = 10.5 ng/mL, AUC0‑∞ = 317 ng h/mL; 4000 mg
p.o.: Cmax = 86.7 ng/mL, AUC0‑∞ = 2572 ng h/mL).229

8. CYCLIC UNDECAPEPTIDES
8.1. Cyclosporin A and Synthetic Derivatives
Figure 47. Cyclopeptolide 1 (87). MW = 1126; miLogP = 2.6; HBD = Cyclosporin A (Cyclosporine, CSA, 109, Figure 50) is an orally
4; HBA = 23; RotB = 12; tPSA = 282 Å2. bioavailable cyclic peptide natural product with a D-Ala and a
butenyl-methyl-L-threonine. It was first isolated from the fungus
7.5. Permetin A T. inf latum in the early 1970s by scientists at Sandoz (now
Novartis). CSA is most often used in the clinic as an injectable
The decadepsipeptide permetin A (88, Figure 48) has two D-
immunosuppressive drug to combat organ transplant rejection,
residues and an unusual β-hydroxyisoheptanoic acid moiety. It
but it is also used as an oral treatment for graft versus host
displayed broad antibacterial activity in vitro. LD50 was
disease. It binds to cyclophilin in lymphocytes.230 CSA has
determined in mice (LD50 36 mg/kg ip; LD50 2100 mg/kg
interesting pharmacokinetics and is a rare example of a large
p.o.).223 From these data, the oral absorption of 88 can be
cyclic peptide (11 residues, MW = 1202 Da) with appreciable
estimated at around 1−2%.
oral bioavailability (F = 20−70% depending upon formulation
7.6. Synthetic N-Methyl β-Strand Decapeptides and species; 22−29% in rat).106,231 CSA has been used as a
Guided by known permeable and orally bioavailable peptides, model for developing peptides into orally deliverable drugs.
researchers from Novartis prepared a library of highly N- The crystal structure of CSA from acetone, and the NMR
methylated cyclic decapeptides (89−107, Table 1).224,225 NMR solution structure determined in CDCl3, both show that it is
studies showed extensive and varied intramolecular hydrogen stabilized by four intramolecular hydrogen bonds between the
bonds. For 94, a crystal structure from dichloromethane/ backbone amide NH and the backbone carbonyl oxygen atoms
heptane (CCDC code: ILOWAJ) showed two beta turns and 2 (CCDC code: DEKSAN).232 In polar solvents like methanol,
transannular hydrogen bonds.224 N-Methylation and stereo- CSA exists in at least four conformations, but it is insoluble in
chemical patterns were preserved while specific side chains water. Co-crystallization of CSA with cyclophillin (PDB code:
8112 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Table 1. Highly N-Methylated Model Cyclic Decapeptides

no. AA 1 + 8 AA 2 + 7 AA 3 + 6 AA 4 + 9 AA 5 + 10 CLa AUCb (iv/p.o.) F% MW miLogP HBD HBA RotB tPSA (Å2)
89 L L L a A 66 256/69 27 1047 3.9 4 20 12 238
90 L A L a A 64 277/28 10 963 1.2 4 20 8 238
91 L L L G P 121 144/4 3 1043 3.3 4 20 12 238
92 A A A G P 56 377/5 1 791 −4.4 4 20 0 288
93 L A L f P 5 4532/767 18 1139 4.3 4 20 12 238
94 L A L p F 7 2206/1006 46 1139 4.3 4 20 12 238
95 L A L p A 30 579/912 130c 987 1.4 4 20 8 238
96 L A L p V 43 379/40 11 1043 2.9 4 20 10 238
97 L F L p A 4 3673/608 17 1139 4.3 4 20 12 238
98 L A L f A 5 5773/487 10 1115 4.2 4 20 12 238
99 L A L l F 1 12368/491 4 1200 6.8 4 20 16 238
100 L A L p F+X 5 3219/1317 40 1140 3.1 4 21 12 251
101 L A L p F+T 16 12368/491 73 1093 2.2 5 21 11 258
102 L G L p F 10 1490/322 22 1111 4.6 4 20 12 238
103 L L G p F 21 723/214 29 1111 3.6 4 20 12 238
104 L A+L L p F 7 2470/788 32 1181 5.6 4 20 14 238
105 L A+D L p F 12 1192/32 2 1184 3.5 5 22 14 276
106 L A+K L p F 8 1700/8 0 1197 3.8 6 21 16 264
107 L A+T L p F 3 4354/728 15 1170 3.7 5 21 13 258
a
Units = mL/min·kg. bUnits = nM·h. X= 3-pyridylalanine. cValue exceeds 100% but has no SD reported.224

Figure 50. Cyclosporin A (109) and KI-306 (110). 109: MW = 1203;


Figure 49. Surotomycin (108). MW = 1681; miLogP = −4.6; HBD = miLogP = 3.6; HBD = 5; HBA = 23; RotB = 15; tPSA = 279 Å2. 110:
25; HBA = 43; RotB = 33; tPSA = 702 Å2. MW = 1391; miLogP = 3.7 (mPEG not counted); HBD = 4; HBA =
29; RotB = 24; tPSA = 347 Å2.
1CWA) revealed a single bound conformation that differed
from its unbound crystal state.233,234 NMR solution structures
of the CSA-cyclophilin complex in aqueous solution, and 13C = 1.8 μg/mL, Tmax = 1.43 h) had higher bioavailability than
enriched CSA bound to cyclophilin, supported the unusual CSA (Sandimmune Neoral, 7 mg/kg p.o.: AUC = 21.4 μg/mL·
binding conformation (PDB codes 1CYA, 1CYB).235,236 When h, Cmax = 1.1 μg/mL, Tmax = 2.6 h).237
bound to cyclophilin, CSA was not stabilized by intramolecular NIM811 (111, Figure 51) is a CSA analogue modified at
hydrogen bonds. Instead, the four-amide protons were position 4 with N-methylisoleucine and a D-Ala at position 8,
orientated toward the exterior of the macrocycle. The existence and has been investigated as a therapeutic agent for the
of multiple different conformers of CSA suggests that it has a treatment of hepatitis C. 111 suppresses hepatitis C virus
flexible structure. This has been hypothesized to play an replication but, unlike CSA, was not immunosuppressive. It was
important role in its unusual passive membrane permeability found to have similar oral bioavailability to CSA in mice (10
and oral bioavailability, although this has not been proven. mg/kg p.o.: Cmax = 2.1 μg/mL, Tmax = 2−5 h, AUC0−24 = 23.7
KI-306 110 (Figure 50) is a monomethoxypoly(ethylene μg/mL·h), rats (10 mg/kg p.o.: Cmax = 1.4 μg/mL, Tmax = 4−8
glycol) modified CSA prodrug developed to improve water h, AUC0−24 = 13.1 μg/mL·h), dogs (20 mg/kg p.o.: Cmax = 5.2
solubility relative to CSA. The oral bioavailability of 110 and μg/mL, Tmax = 1 h, AUC0−24 = 38.2 μg/mL·h), and monkeys
CSA (Sandimmune Neoral solution) were compared in rats. (10 mg/kg p.o.: Cmax = 1.7 μg/mL, Tmax = 3−8 h, AUC0−24 =
PEG-modified 110 (7 mg/kg p.o.: AUC = 32.8 μg/mL·h, Cmax 11.3 μg/mL·h).238
8113 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 51. NIM811 (111). MW = 1203; miLogP = 3.6; HBD = 5;


HBA = 23; RotB = 15; tPSA = 279 Å2.

SCY-635 112 (Figure 52) is a CSA analogue recently


developed as a potent inhibitor of hepatitis C virus RNA
replication and showed oral bioavailability in rats (5 mg/kg
p.o.: Cmax = 900 ng/mL, Tmax = 4.0 h, AUC0‑∞ = 13000 ng·h/
mL, t1/2 = 19.2 h, F = 23.1%) and monkeys (5 mg/kg p.o.: Cmax
= 1810 ng/mL, Tmax = 2.0 h, AUC0‑∞ = 27900 ng·h/mL, t1/2 =
24.6 h, F = 17.7%).239 Alisporivir (also Debio-025, DEB025,
UNIL025, 113, Figure 52) is an orally bioavailable CSA Figure 53. THR-123 (114). MW = 1926; miLogP = −6.2; HBD = 35;
HBA = 49; RotB = 42; tPSA = 833 Å2.
analogue with N-methyl-D-alanine at position 3 and N-ethyl-
valine at position 4.240 Alisporivir completed Phase I and II
clinical trials as a HCV antiviral drug that blocks viral including 3 L-lactic acids and 3 D-alpha-hydroxyisovaleric acids.
replication. It has excellent pharmacokinetic and safety profiles The 36-membered macrocycle is a potassium-selective
and significantly decreased viral load in HCV-infected patients ionophore (LD50 4 mg/kg p.o, rat).244 Unfortunately, there is
(1200 mg/day p.o.: Cmax = 1052 ng/mL, Tmax = 2.0 h, little pharmacokinetic data available for most dodecapeptides.
AUC0−12h = 3858 ng·h/mL).241,242 9.1. Cerulide
The cyclic dodecadepsipeptide cerulide (115, Figure 54)
contains seven L-residues, five D-residues, six depsipeptide
bonds, and six peptide bonds. It is an emetic toxin (induces
vomiting) produced by the bacteria B. cereus.245 Cerulide was
administered (p.o. or ip) to the S. murinus rodents to test for
emetic effects at doses from 2 to 32 μg/kg. The maximal oral
dose (32 μg/kg p.o.) resulted in emesis in 5 of 5 test animals.
115 (50 μg/kg p.o.) in combination with 5-HT3 antagonist,
ondansetron (iv), had no emetic effect. The authors concluded
that cerulide caused emesis through central 5-HT3 receptor
stimulation of the vagus afferent. Cerulide had ED50 12.9 μg/kg
(p.o.) and 9.8 μg/kg (ip) for inducing emesis in S. murinus
rodents.245
9.2. L-Phenylalanine-Dipicolinate Macrocycle
Figure 52. SCY-635 (112) and alisporivir (113). 112: MW = 1322;
miLogP = 2.9; HBD = 6; HBA = 25; RotB = 19; tPSA = 302 Å2. 113: A series of linear and macrocyclic L-phenylalanine-dipicolinic
MW = 1217; miLogP = 3.8; HBD = 5; HBA = 23; RotB = 15; tPSA = acid based compounds were synthesized and tested for anti-
279 Å2. inflammatory activity. Anti-inflammatory activity was evaluated
by oral administration (7 mg/kg p.o.) to rats with carrageenan-

8.2. THR-123
The 16-residue disulfide-bridged cyclic peptide THR-123 (114,
Figure 53) was designed to mimic a loop in the structure of
human BMP7; a protein from the TGF-β superfamily that
binds Alk2/3/6 and antagonizes TGF-β−mediated activity.
The disulfide bridge stabilizes the conformation. Compound
114 was active against kidney fibrosis by inhibiting the Alk3
receptor after oral administration (5−15 mg/kg p.o.) to
mice.243

9. CYCLIC DODECA- AND TRIDECAPEPTIDES


There are a number of cyclic dodecapeptides that are orally
active. One example is the antibiotic valinomycin, which Figure 54. Cereulide (115). MW = 1153; miLog P = 6.6; HBD = 6;
contains 3 L-valines, 3 D-valines, 6 depsipeptide components HBA = 24; RotB = 12; tPSA = 332 Å2.

8114 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

induced paw edema. Two known nonsteroidal anti-inflamma-


tory drugs (NSAIDs), indomethacin and diclofenac, were used
as controls. Compound 116 (Figure 55) caused 72% inhibition
of paw inflammation compared to 82% and 61% for
indomethacin and diclofenac.246

Figure 55. L-Phenylalanine-dipicolinic acid based macrocycle (116).


MW = 1930; miLogP = 8.6; HBD = 12; HBA = 36; RotB = 16; tPSA = Figure 57. α-Conotoxin MII (118) and lipidated analogue (119). 118:
505 Å2. MW = 1711; miLogP = −5.8; HBD = 27; HBA = 45; RotB = 21; tPSA
= 718 Å2. 119: MW = 1837; miLogP = −4.6; HBD = 27; HBA = 45;
RotB = 29; tPSA = 718 Å2.
10. CYCLIC TETRADECAPEPTIDES AND BEYOND orally to rats (0.3 or 3.0 mg/kg p.o.) and produced significant
10.1. Conotoxins and Synthetic Derivatives dose-dependent relief of neuropathic pain.249
Linaclotide (117, Figure 56) is a 14-residue peptide toxin that
is cyclized through three cysteine disulfide bridges. It is a
treatment for irritable bowel syndrome with constipation and
for chronic constipation. 117 was stable in simulated gastric
fluid up to 3 h and was not hydrolyzed by pepsin. Following
oral administration to rats, 117 was found in plasma despite
minimal oral absorption (10 mg/kg p.o.: AUC0−6h = 19.7
ng·h/mL, F = 0.1%).247

Figure 58. Head-to-tail cyclized analogue of α-conotoxin Vc1.1 (120).


MW = 2160; miLogP = −6.3; HBD = 32; HBA = 59; RotB = 21; tPSA
= 909 Å2.

Figure 56. Linaclotide (117). MW = 1527; miLogP = −5.7; HBD =


21; HBA = 36; RotB = 13; tPSA = 574 Å2. 10.2. Duramycin
Duramycin (lancovutide, Moli1901, 121, Figure 59) is a highly
α-Conotoxin MII (118) and lipidated analogue 119 (Figure cross-linked 19-residue peptide with three lanthione bridges
57) are 16-residue bis-disulfide bridged natural peptides that are and one lysoalanine bridge, all derived from L-cysteine. 121 was
orally absorbed. Following oral administration of 118 and a given to rats via pulmonary, intravenous or oral administration.
lipophilic analogue 119 (1 mg/kg p.o.) to male Sprague− Faeces, urine, blood and tissue were collected over a 7-day
Dawley rats, ∼6% of both peptides crossed the GI tract and period. Analysis indicated minimal systemic availability
were found in a variety of body tissues based on radioactive following oral administration with almost 100% of the oral
labeling.248 dose recovered from faeces. Nevertheless, 121 was detected in
A head-to-tail cyclized analogue of α-conotoxin Vc1.1 (120, serum of rats 7 days after oral administration (231 mg/kg
Figure 58) from the cone snail C. victoriae was administered p.o.).250
8115 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Recently a lysine mutant of kalata B1, KB1[T20K] (124,


Figure 60),254 was investigated in an experimental autoimmune
encephalomyelitis model of multiple sclerosis. 124 was
efficacious when administered orally to mice (10 or 20 mg/
kg p.o.) and impeded disease progression at 20 mg/kg p.o.
compared to the control group dosed with an inactive kalata B1
derivative [V10K].255
10.4. Stapled α-Helix
SAH-gp41(626−662) (125) is a doubly stapled 37 residue peptide
reported to be a stable α-helix and resistant to pepsin and
chromtrypsin. The bridging residues are of normal S-
configuration but possess an additional α-methyl group.
When mice were given 125 by oral gavage (10 and 20 mg/
kg p.o.; Figure 61), it was found in plasma in a dose dependent
manner (10 mg/kg p.o.: Cmax = 1.5 μg/mL; 20 mg/kg p.o.: 2.3
μg/mL). The nonstapled linear analogue was not detected in
Figure 59. Duramycin (121). MW = 2013; miLogP = −6.1; HBD = plasma after oral administration.257 We are not aware of any
29; HBA = 48; RotB = 19; tPSA = 760 Å2. other similar helix-constrained peptide reported to show any
significant oral absorption at all.
10.3. Kalata B1 and Other Cyclotides
The cyclotides are a rapidly expanding family of cyclic peptides,
which owe their stability to a remarkably conserved cyclic
cysteine knot (CCK) motif. Cyclotides have a range of
biological activities (e.g., antiviral, antitumor, antibacterial)
and are actively being investigated for their potential in drug
development.251 The prototypic cyclotide, kalata B1 (122,
Figure 60) is a 29-residue peptide from the African medicinal Figure 61. HIV fusion inhibitor helical peptide SAH-gp41(626−666)
plant O. af f inis. Kalata B1 is a uterotonic agent252 and by virtue (125). B = Norleucine, X = (S)-α-methyl(4-pentenyl)alanine. 125:
of its knotted structure is chemically and enzymatically stable MW = 4589; QPLogP = −23.1*; HBD = 72; HBA = 117; RotB = 132;
and can be made into tea by infusing its parent plant with tPSA* = 2173 Å2. “*”: Data calculated with QikProp256 due to
boiling water. This tea is an indigenous medicinal agent used to structures exceeding Molinspiration maximum size.
accelerate childbirth by inducing uterine contractions. Recently,
orally active peptidic bradykinin B1 receptor antagonists were
grafted onto kalata B1 to replace the entire loop 6 unit.253 The
grafted analogue CKB-KAL (123, Figure 60) was the most 11. INFLUENCES ON ORAL BIOAVAILABILITY
potent compound studied and inhibited acetic acid induced Oral bioavailability is a specific term that indicates how much
abdominal contraction (the writhing assay) after injection (1 intact peptide can be measured in the circulation after oral
mg/kg ip) or oral administration (10 mg/kg p.o.). Maximum ingestion. By definition this parameter is dictated not just by
inhibition was similar for both routes (49% ip and 42% p.o.).253 compound absorption through an intestinal membrane but also
All the cysteine cross-links have L-stereochemistry. by compound stability, interactions, and metabolism before
intestinal absorption as well as compound stability, metabolism,
tissue distribution, and clearance after uptake. Most proteins
and peptides simply do not get absorbed from the GI tract, and
those that do are often rapidly metabolized, cleared, or
distributed into tissues. Key problems for peptides are their
high polarity and large polar surface area, large size, low
membrane permeability, high clearance, and rapid metabolism.
Cyclization of peptides frequently aids formation of intra-
molecular hydrogen bonds and orientation of side chains,
properties that can help shield polar atoms from the solvent
medium (water/lipid) and protect against proteolytic degrada-
tion, reduce flexibility, reduce polar surface area, and promote
permeation through cell membranes. We describe 125 cyclic
peptides above that are orally absorbed, orally active or have a
measured oral bioavailability (F%). Physicochemical parameters
traditionally associated with limits on oral bioavailability have
Figure 60. Cyclotides kalata B1 (122), CKB-KAL (123), and
KB1[T20K] (124). 122: MW = 2892; QPLogP = −15.8*; HBD =
been calculated here for each compound (see Figure legends).
42; HBA = 74; RotB = 23; tPSA* = 1207 Å2. 123: MW = 3135; For cyclic peptides with reported oral bioavailability, we now
QPLogP = −17.6*; HBD = 42; HBA = 77; RotB = 26; tPSA* = 1224 plot key molecular properties, such as MW (Figure 62A),
Å2. 124: MW = 2919; QPLogP = −16.6*; HBD = 43; HBA = 74; miLogP (Figure 62B), HBD (Figure 63A), HBA (Figure 63B,
RotB = 26; tPSA* = 1212 Å2. “*”: Data calculated with QikProp256 Figure 64), RotB (Figure 65A) and tPSA (Figure 65B), against
due to structures exceeding Molinspiration maximum size. F%.
8116 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Figure 62. Plots of oral bioavailability (F%) versus (A) molecular


Figure 63. Plots of oral bioavailability (F%) versus (A) hydrogen bond
weight, and (B) octanol−water partition coefficient (miLogP), of
donors (HBD) or (B) hydrogen bond acceptors (HBA), for orally
orally absorbed cyclic peptides 2−117.
absorbed cyclic peptides 12−112.

The plot of molecular weight versus oral bioavailability


(Figure 62A) shows that cyclic peptides with molecular weights value is one of the better predictors of octanol−water partition
ranging from 500 to 1350 can have some oral bioavailability (F coefficients.258 The partition coefficient P (or distribution
= 0.1−10%). Cyclic peptides with higher oral bioavailability coefficient D) is a ratio of hydrophobicity : hydrophilicity and a
clustered into two distinct MW ranges 700−800 and 1000− measure of relative compound solubility in the two solvents. To
1200, the latter possibly skewed by the high number of be orally absorbed, a cyclic peptide must permeate intestinal
decapeptide- and cyclosporine-like cyclic peptides. It is membranes by partitioning into the lipid, but it must not be so
encouraging that 14 cyclic peptides with MW = 500−850 (2, lipophilic that it cannot partition out. The compromise for
8, 12, 19, 31, 39, 47, 49, 55, 57−59, 63, and 64) had > 10% small molecule drugs is LogP = 0−5, with some support for the
oral bioavailability. A further 23 cyclic peptides (73, 76−81, 89, variation −0.4 to 5.6.259
90, 93−98, 100−104, 107, 109, and 112) with MW = 960− Figure 62B shows that most cyclic peptides in this review
1350 had ≥10% oral bioavailability. These data demonstrate with F ≥ 10% oral bioavailability also had miLogP 1−5 (e.g., 2,
that a higher MW (> 500) does not necessarily preclude oral 12, 19, 31, 39, 47, 49, 57, 58, 63, 64, 76, 79, 89, 90, 93−98,
bioavailability for cyclic peptides, even though it is important 100−103, 107, 109, and 112). The extent of oral bioavailability
for most small molecules.17,64,65,123,124 did not correlate with miLogP within this range. Seven orally
Oral bioavailability for each cyclic peptide in this review was bioavailable (F > 10%) cyclic peptides (59, 73, 77, 78, 80, 81,
next plotted against the predicted octanol−water partition and 104) had miLogP > 5. Eleven cyclic peptides with miLogP
coefficient (miLogP) in Figure 62B. The calculated miLogP < 0 had measurable oral bioavailability but this was limited to F
8117 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

with F ≥ 10% oral bioavailability, but two-thirds of the cyclic


peptides now conform to conventional RO5 guidelines. We
conclude that HBA count is somewhat important for oral
bioavailability of cyclic peptides but not as restrictive as the
HBD count.
Rotatable bonds (RotB) reflect the degree of conformational
flexibility in a molecule. RO5 compliant drug-like compounds
typically have ≤ 10 rotatable bonds.17 However, care must be
taken in the use of RotB itself, or as part of any prediction
model for oral bioavailability. First, it is notable that strict RO5
rules describe any bond in a cycle of any ring size as being not
rotatable. However, as cyclic peptide ring size increases, the
energy required to rotate Φ/Ψ bonds within the macrocycle
decreases, reducing the utility of this RotB metric. As observed
above, from cyclic tetrapeptide 2 to cyclosporin A, solution
structures and crystal structures can often differ greatly due to
such rotations or ring flips. Second, studies in rats and humans
have shown that the calculation method, therapeutic class of
compounds, and desired F% all influence the acceptable
number of RotB. In rats, certain therapeutic classes of drugs
Figure 64. Plot of oral bioavailability (F%) versus hydrogen bond had acceptable F% with RotB > 12.261 Recently, a study on the
acceptors (HBA), where the amide nitrogen is not counted as a HBA
oral bioavailability of 1014 molecules in humans found the
atom, for orally absorbed cyclic peptides 12−112.
RotB limit to be 13.262 Figure 65A shows that most of the cyclic
peptides examined here with oral bioavailability had 6−16
< 10% (7, 8, 21−25, 33, 34, 40, and 92). These data suggest rotatable bonds. Compounds 12 (RotB = 2, F = 16%) and 112
that hydrophobicity (and lipophilicity) is likely an important (RotB = 19, F = 23%) were outside this range. Twenty three
contributor to oral bioavailability of these cyclic peptides. This compounds exceeded RotB ≤ 10; compounds 2, 19, 31, 93, 97,
data shows that this parameter is more accommodating for 98, and 107 had F > 10%, while 36, 73, 76, 77, 78, 79, 80, 81,
cyclic peptides (miLogP = 1−8) than it is for small molecules 89, 94, 100, 101, 102, 103, 104, 109, and 112 had F ≥ 20%.
(LogP = 0−5). This is consistent with a degree of conformational rigidity being
Two RO5 criteria that guide oral bioavailability of drug-like favorable for oral bioavailability of cyclic peptides.
small molecules are the numbers of hydrogen bond donors The plot of topological polar surface area (tPSA) versus oral
(HBD) and acceptors (HBA). A small molecule is considered bioavailability (Figure 65B) indicated that large polar surfaces
more likely to be orally bioavailable if it contains ≤ 5 HBD and (tPSA > 300 Å 2) were a barrier to appreciable oral
≤ 10 HBA,14−16 This implies that hydrogen bond donors are bioavailability (F > 10%), likely due to low absorption and
more detrimental to oral bioavailability. Figure 63A shows that high clearance. Fifteen of the smaller cyclic peptides with still
cyclic peptides with 1−6 HBD are much more likely to be large polar surfaces (tPSA 100−205) had F = ≥ 5% (2, 7, 12,
orally bioavailable than those with HBD > 6. A few compounds 18−20, 31, 39, 47, 49, 55, 57, 62, 63, and 64) and three had F
(19, 20, 31, and 112) with HBD = 6 had F = 9−25%, but all > 30% (39, 57, and 64). Twenty four compounds with tPSA
compounds with HBD > 6, except 8, (15%) had F ≤ 10%. If 200−300 were still orally bioavailable with F ≥ 10% (8, 31, 73,
this data set is representative of all cyclic peptides, HBD does 76−81, 89, 93−98, 100−104, 107, 109, and 112), and violate
indeed limit oral bioavailability for this class of molecules. the prediction limit17 for most drug-like small molecules (tPSA
Figure 63B shows that all cyclic peptides examined had HBA < 140 Å2).
> 10, and those with F > 50% had HBA = 12−22. Thus, almost
all of the orally bioavailable cyclic peptides in this review 12. CONCLUSIONS AND FUTURE PROSPECTS
apparently violate the RO5 limit on HBA (Figure 63B), a This compilation is based on data on oral activity, oral
guideline developed from small molecule drugs. This suggests absorption, and oral bioavailability for 125 cyclic peptides,
that optimizing cyclic peptides to reduce HBD, rather HBA, is composed of 4 to 37 amino acids and derivatives. It indicates
more likely to improve oral bioavailability. However, Figure that there are opportunities to expand the molecular weight
63B uses the original definition of HBA,14,15 which was simply range beyond the RO5 limit of small molecules (MW ≤ 500)
the total count of all nitrogen and oxygen atoms, irrespective of for cyclic peptides and still obtain appreciable oral bioavail-
their actual hydrogen bond accepting properties. This is ability. This is encouraging for therapeutic targeting of
important because the amide nitrogen is not a hydrogen protein−protein interactions that may require modulators
bond acceptor due to electron delocalization of the nitrogen with larger surface areas. The cyclic peptides show great
lone pair, indeed protonation always occurs preferentially on variation in the parameters (MW, HBD, HBA, LogP, RotB, and
the amide oxygen, not the nitrogen.260 Thus, if HBA alone is tPSA) conventionally associated with limiting oral bioavail-
re-counted without amide nitrogens, the result changes ability (F%) of drug-like molecules. Such parameters are of
dramatically (Figure 64). As shown, 24 of the cyclic peptides course all interdependent. As molecules get bigger, they either
for which F ≥ 10% now have HBA ≤ 10 (e.g., 2, 7, 8, 12, 19, increase in the number of polar components (increasing HBD
31, 39, 47, 49, 55, 57−59, 63, 64, 89, 93, 95−98, and 102− and HBA) or nonpolar components (increasing LogP),
104). Only 12 cyclic peptides with F ≥ 10% now have HBA > generally become more flexible (increasing RotB), and thus
10 (e.g., 73, 76-81, 100, 101, 107, 109, and 112). Thus, the expose more of their surface area to water (tPSA) and
HBA count is still violated by one-third of the cyclic peptides membranes. The RO5 has been used very successfully to guide
8118 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

development of orally bioavailable small molecule drugs over


the past two decades. However, it is clear from Figures 62−65
that orally bioavailable cyclic peptides can violate all of these
parameters, especially MW, tPSA, HBA, and, but not to the
same extent, HBD, RotB, and LogP. Interestingly, Figures 62,
63, and 65 show this to be true for the four most orally
bioavailable (F > 70%) cyclic peptides (39, 80, 95, and 101),
which respectively had MW > 500 (724, 1196, 987, and 1079),
HBA > 10 (12, 22, 20, and 21), and three had tPSA > 200 Å2
(140, 256, 238, and 258). On the other hand, all four
compounds respectively had HBD ≤ 5 (0, 3, 4, and 5), three
had RotB ≤ 12 (12, 13, 8, and 10), two had milogP < 5 (6.8,
7.5, 1.4, and 2.0) and, without counting amide nitrogens, two
had HBA ≤ 10 (9, 12, 10, and 11). Further systematic
evaluations of new orally bioavailable cyclic peptides with MW
> 500 are needed to support these observations and better
define limits of these parameters on oral bioavailability. This
may better enable prediction and optimization of orally
bioavailable cyclic peptides larger than 5 amino acids.
The above data set supports current and new chemical
strategies to enhance oral absorption. These include reducing
HBD by removing backbone amide NH protons through
substitution with N-alkyl groups, esters, ethers, or hydro-
carbons; incorporating the nitrogen into a heterocycle; or
substituting amides with other surrogates. Thus, cyclic
hexapeptide 39 has 3 depsipeptide linkages, 3 NMe
substituents, and no NH protons; cyclic octapeptide 80 has 1
depsipeptide linkage, 2 NMe substituents, 2 prolines, and 3 NH
protons; while cyclic decapeptides 90 and 101 both have 6
NMe substituents and 4 NH protons. However, these
approaches may not be a panacea to the problem of poor
oral bioavailability. Biological activity is usually dependent on
three-dimensional structure, and N-methylation or insertion of
amide isosteres263 that reduce NH protons (HBD) can alter
peptide structure, which may negatively impact function. For
example, amide protons might be required for direct binding to
a target or for maintaining the bioactive conformation through
intramolecular hydrogen bonds. N-Methylation or amide
isosteres can disrupt either or both of these properties.
Intramolecular hydrogen bonds are often desirable because
they reduce the exposed polar surface of a peptide. Introducing
an amide isostere also normally requires additional synthetic Figure 65. Plots of oral bioavailability (F%) versus: (A) rotatable
steps, the value of which must be weighed up against potential bonds (RotB) or (B) topological polar surface area (tPSA), for orally
absorbed cyclic peptides 12−112.
pharmacokinetic benefits. Ester bonds are present in many
naturally occurring cyclic depsipeptides and are thought to be
evolutionary adaptions that facilitate permeability through Two important considerations missing from the above
biological membranes. Romidepsin 12, largazole 13, kahalalide analysis are three-dimensional polar surface area and metabolic
F 61, and Gq inhibitor YM254890 67 are among many stability. There were only a few reported three-dimensional
examples of depsipeptides that are active in mammalian cells structures for cyclic peptides in this review, but the effect of
and bind to intracellular targets, supporting this hypothesis. three-dimensional structure and conformational preference of
However, ester bonds are also unstable and readily hydrolyzed cyclic peptides in water versus lipid membranes is very
by esterases in cells and in vivo. Although esterification has important. Three-dimensional structure can significantly change
been widely exploited for delivering carboxylic acids as the solvent-exposed polar surface area. Large macrocycles might
prodrugs, ester substitution can also jeopardize in vivo stability. be folded through intramolecular hydrogen bonds that are
For example, orally bioavailable romidepsin 12, largazole 13 shielded from solvent. This could produce much smaller
and kahalalide F 61 have very short half-lives at 37 °C (t1/2 = 6, exposed polar surface areas than suggested by tPSA calculations
0.3, 28 min), probably due to poor in vivo stability of their on 2D structures. Structural compression may enable larger
cleavable ester moieties. In enniatin B1 39, every second amino cyclic peptides (MW) with more polar atoms (HBD/HBA)
acid linkage is a depsipeptide bond. It displays unusually high and larger tPSA (but low 3D-PSA) to be tolerated for oral
oral bioavailability (F = 91%), but also has a very short half-life absorption. Compaction or hydrophobic collapse is a dominant
(t1/2 = 9 min) leading to minimal exposure in plasma (AUC feature of protein folding that enables proteins to shield their
25.3 ng.h/mL). hydrophobic residues in their interiors away from water.
8119 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Conversely, passive diffusion through a lipid membrane would peptides with Professor Fairlie. He conducted postdoctoral research in
be favored by the reverse effect, structural compression to the same lab. In 2017, he is a postdoctoral fellow in Professor Meldal’s
shield polar residues in the interior of a cyclic peptide and group at University of Copenhagen. Research interests are in
expose outer hydrophobic substituents to lipid bilayers. pharmaceutical sciences, natural products, cyclic peptides, organic
Compact structures that undergo minimal change when going synthesis, and drug discovery.
from water through a lipid membrane to water again should pay Nicholas E. Shepherd developed bioactive helical peptide mimics
a lower entropy penalty to overcome this transition. Thus, during his Ph.D. at the University of Queensland. As a postdoctoral
rigidity may be more desirable than flexibility for oral researcher at the University of Tokyo, he identified novel bimetallic
bioavailability, although this brings an accompanying problem catalysts to enantioselectively synthesize pharmaceutically important
of lower aqueous solubility. chiral small molecules. As an ARC DECRA fellow at the University of
The nature and extent of water solvation is likely to be a key Sydney, he used chemical tools to define the structural basis for R/
consideration for oral absorption. Water molecules need to be DNA-binding proteins and epigenetic multiprotein complex function.
stripped away from polar surfaces to facilitate uptake from the He is currently a senior research officer at the University of
gut via passive diffusion or for interaction with proteins that Queensland.
promote active transport. Experimental determination and
comparison of solvation energies can be very helpful for Weijun Xu was a lecturer at the School of Chemical and Life Sciences
predicting oral absorption as well as solubility. The nature of (Singapore Polytechnic), an undergraduate (B.Sc. Hons. Biochemistry,
amino acid substituents and the degree of flexibility in the 2006) and postgraduate of University of Queensland (Ph.D., 2013−
macrocycle not only dictates solvation but also susceptibility to 2016), where he is a research assistant. He was an International
oxidative and degradative metabolism. Amino acid substituents Postgraduate Research Scholar and University of Queensland
and peptide backbone conformation also determine inter- Advantage Scholar with Professor Fairlie on computer-aided molecular
actions with proteins, including transporters and efflux modeling of protein−ligand and protein−protein interactions,
promoters, metabolizing enzymes, albumin, plasma, and cellular involving discovery of ligands for GPCRs, proteases, enzymes, and
proteins. All of these factors contribute to oral bioavailability other proteins involved in human immune systems.
and need to be studied to better understand how metabolism Andrew J. Lucke obtained a Ph.D. in organic chemistry at Griffith
and clearance, in addition to absorption, are affected by University, followed by postdoctoral research in the United Kingdom
structural modifications to cyclic peptides. and Australia. He has published in organic, medicinal, physical, and
This collection of cyclic peptides has the potential to macromolecular chemistry. His recent research has used molecular
stimulate chemists to reach for new horizons in the design, simulation techniques to model interactions between small organic
synthesis, and application to humans of larger, biologically molecules and large biomolecules. He is a molecular modeller at La
active, compounds including macrocycles. To date extensive Trobe University with interests in organic synthesis, medicinal
research has focused on membrane and cell permeability of chemistry (drug design and development), peptidomimetics, and
peptides and macrocycles but, as emphasized in this review, protein molecular dynamics.
membrane permeability is only one of many contributors to Martin Stoermer obtained a B.Sc. Hons (1986) and Ph.D. (1991)
oral bioavailability. More details on solvation, absorption, from University of Sydney in organic synthesis and organometallic
metabolism, tissue distribution, clearance and three-dimen- reaction mechanisms. He worked in drug design at the Centre for
sional structures of cyclic peptides can provide a deeper Drug Design and Development, University of Queensland (1991−
understanding of how to better exploit the different factors that 1995) and in the Institute for Molecular Bioscience since 2001 as a
influence their oral bioavailability and their promise as new oral Senior Research Officer. He has also collaborated with the
therapeutics. pharmaceutical industry while at the Technical University of
Clausthal-Zellerfeld, Germany (1995−1996) and the Victorian College
AUTHOR INFORMATION
of Pharmacy, Monash University (1996−2000).
Corresponding Author
David Fairlie studied at Adelaide, Australian National, New South
*Fax: +61-733462990. E-mail: d.fairlie@imb.uq.edu.au. Wales, Stanford and Toronto Universities. At University of Queens-
ORCID land, he led the Chemistry Group in the Centre for Drug Design and
Martin J. Stoermer: 0000-0003-3445-2104 Development and is Head of the IMB Division of Chemistry and
David P. Fairlie: 0000-0002-7856-8566 Structural Biology. Interests are medicinal/organic chemistry, protein
mimics, and modulators of GPCRs, PPIs and enzymes in
Present Address inflammation, infection, neurodegeneration and cancer. He studies
§
La Trobe Institute of Molecular Sciences, La Trobe University, mechanisms of chemical, immunological and biological reactions,
Melbourne, VIC 3083, Australia. disease development, and drug action.
Notes
The authors declare no competing financial interest. ACKNOWLEDGMENTS
Tables of physicochemical parameters, pharmacological param- We acknowledge grants from the National Health and Medical
eters, formulation vehicles for compounds in this review can be Research Council (Senior Principal Research Fellowships
provided upon request from the authors. 1027369 and 1117017 to D.P.F.) and the Australian Research
Biographies Council (DP150104609, DP130100629, DP160104442, and
CE140100011).
Daniel S. Nielsen obtained a B.Sc and M.Sc from the Faculty of
Pharmaceutical Sciences at University of Copenhagen (2011). He
received a University of Queensland International Ph.D. Scholarship ABBREVIATIONS
(2012) and obtained a Ph.D. (2016) on orally bioavailable cyclic AFP alpha-fetoprotein
8120 DOI: 10.1021/acs.chemrev.6b00838
Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

AFPep alpha-fetoprotein 472−479 cyclic analogue Tmax time to reach the peak serum concentration
Aha 6-aminohexanoic acid TNBS trinitrobenzenesulfonic acid
Alk3 bone morphogenetic protein receptor, type IA tPSA topological polar surface area
AUC area under the curve, the integral of the VD volume of distribution, the apparent volume in
concentration time curve (0-∞ = after a single which the drug is distributed at steady state
dose or τ,ss = at steady state)
BBMV brush border membrane vesicles
BMP7 bone morphogenic protein 7 REFERENCES
CL clearance, the volume of plasma cleared of the
drug per unit time (1) Otvos, L., Jr.; Wade, J. D. Current Challenges In Peptide-Based
Drug Discovery. Front. Chem. 2014, 2 (62), 1−4.
CSA cyclosporin A (2) Clackson, T.; Wells, J. A. A Hot Spot Of Binding Energy In A
Cmax peak serum concentration Hormone-Receptor Interface. Science 1995, 267, 383−6.
C5a complement factor 5a (3) Moreira, I. S.; Fernandes, P. A.; Ramos, M. J. Hot Spots - A
CDI C. dif f icile infection Review Of The Protein-Protein Interface Determinant Amino-Acid
CCK cyclic cysteine knot Residues. Proteins: Struct., Funct., Genet. 2007, 68, 803−12.
CDCl3 deuterochloroform (4) Hall, D. R.; Kozakov, D.; Whitty, A.; Vajda, S. Lessons From Hot
CYP cytochrome P450 enzyme Spot Analysis For Fragment-Based Drug Discovery. Trends Pharmacol.
Da Daltons Sci. 2015, 36, 724−36.
DMSO dimethyl sulfoxide (5) Sheng, C.; Dong, G.; Miao, Z.; Zhang, W.; Wang, W. State-Of-
EtOH ethanol The-Art Strategies For Targeting Protein-Protein Interactions By
Small-Molecule Inhibitors. Chem. Soc. Rev. 2015, 44, 8238−8259.
ED50 dose required to give 50% of the maximal (6) Modell, A. E.; Blosser, S. L.; Arora, P. S. Systematic Targeting Of
response Protein-Protein Interactions. Trends Pharmacol. Sci. 2016, 37, 702−
EM electron microscopy 713.
F% fraction of orally absorbed compound in plasma (7) Giannis, A.; Kolter, T. Peptidomimetics For Receptor Ligands -
GI gastro-intestinal Discovery, Development, And Medical Perspectives. Angew. Chem., Int.
HCV hepatitis C virus Ed. Engl. 1993, 32, 1244−1267.
HDAC Histone deacetylase (8) Fairlie, D. P.; Abbenante, G.; March, D. R. Macrocyclic
HBA hydrogen bond acceptors Peptidomimetics - Forcing Peptides Into Bioactive Conformations.
HBD hydrogen bond donors Curr. Med. Chem. 1995, 2, 654−686.
H-D hydrogen−deuterium exchange (9) Hruby, V. J. Designing Peptide Receptor Agonists And
Antagonists. Nat. Rev. Drug Discovery 2002, 1, 847−858.
HIV human immunodeficiency virus (10) Tyndall, J. D. A.; Pfeiffer, B.; Abbenante, G.; Fairlie, D. P. Over
HCT116 human colon carcinoma cell line 116 One Hundred Peptide-Activated G Protein-Coupled Receptors
5-HT3 5-hydroxytryptamine subtype 3 Recognize Ligands With Turn Structure. Chem. Rev. 2005, 105,
iv intravenous 793−826.
ip intraperitoneal (11) Walensky, L. D.; Bird, G. H. Hydrocarbon-Stapled Peptides:
LD50 dose required to kill 50% of the test population Principles, Practice, And Progress. J. Med. Chem. 2014, 57, 6275−
LD100 dose required to kill 100% of the test population 6288.
MDCK Madin-Darby canine kidney cells (12) Azzarito, V.; Long, K.; Murphy, N. S.; Wilson, A. J. Inhibition Of
MDCKII-LE Madin-Darby canine kidney cells−low efflux A-Helix-Mediated Protein-Protein Interactions Using Designed
mg/kg milligrams per kilogram Molecules. Nat. Chem. 2013, 5, 161−173.
(13) Hill, T. A.; Shepherd, N. E.; Diness, F.; Fairlie, D. P.
MC4R melanocortin 4 receptor Constraining Cyclic Peptides To Mimic Protein Structure Motifs.
Me methyl Angew. Chem., Int. Ed. 2014, 53, 13020−13041.
miLogP Molinspiration octanol−water coefficient (14) Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J.
α-MSH alpha-melanocyte stimulating hormone Experimental And Computational Approaches To Estimate Solubility
MW molecular weight And Permeability In Drug Discovery And Development Settings. Adv.
NH amide proton Drug Delivery Rev. 1997, 23, 3−25.
NK2 neurokinin 2 (15) Lipinski, C. A. Lead- And Drug-Like Compounds: The Rule-Of-
NMR nuclear magnetic resonance Five Revolution. Drug Discovery Today: Technol. 2004, 1, 337−341.
N-Me N-methyl (16) Leeson, P. D.; Springthorpe, B. The Influence Of Drug-Like
NSAIDs nonsteroidal anti-inflammatory drugs Concepts On Decision-Making In Medicinal Chemistry. Nat. Rev.
Drug Discovery 2007, 6, 881−890.
PAMPA parallel artificial membrane permeability assay (17) Veber, D. F.; Johnson, S. R.; Cheng, H. Y.; Smith, B. R.; Ward,
PEG400 polyethylene glycol average molecular weight of K. W.; Kopple, K. D. Molecular Properties That Influence The Oral
400 Bioavailability Of Drug Candidates. J. Med. Chem. 2002, 45, 2615−
p.o. per oral 2623.
RO5 rule-of-five (18) Craik, D. J.; Fairlie, D. P.; Liras, S.; Price, D. The Future Of
RNA ribonucleic acid Peptide-Based Drugs. Chem. Biol. Drug Des. 2013, 81, 136−147.
RotB rotatable bonds (19) Nisen, M. The Best Selling Prescription Drugs In The World
SAH stabilized α helix Last Year. Gen. Eng. Biotechnol. News 2015, Feb 25,
sc subcutaneous https://qz.com/349929/Best-Selling-Drugs-In-The-World/.
SLC15A1 solute carrier family 15 member 1 (20) Vlieghe, P.; Lisowski, V.; Martinez, J.; Khrestchatisky, M.
Synthetic Therapeutic Peptides: Science And Market. Drug Discovery
t1/2 half-life the time taken for drug concentration to
Today 2010, 15, 40−56.
decrease by half its original value (21) Rowland, M.; Tozer, T. N. Clinical Pharmacokinetics: Concepts
TGF-beta transforming growth factor beta And Applications; 3rd ed.; Williams & Wilkins: Baltimore, MD, 1995.

8121 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

(22) Rubio-Aliaga, I.; Daniel, H. Peptide Transporters And Their (42) Goh, L. B.; Spears, K. J.; Yao, D.; Ayrton, A.; Morgan, P.;
Roles In Physiological Processes And Drug Disposition. Xenobiotica Roland Wolf, C.; Friedberg, T. Endogenous Drug Transporters In In
2008, 38, 1022−1042. Vitro And In Vivo Models For The Prediction Of Drug Disposition In
(23) Giacomini, K. M.; Huang, S.-M.; Tweedie, D. J.; et al. The Man. Biochem. Pharmacol. 2002, 64, 1569−1578.
International Transporter Consortium. Membrane Transporters In (43) Volpe, D. A. Drug-Permeability And Transporter Assays In
Drug Development. Nat. Rev. Drug Discovery 2010, 9, 215−236. CACO-2 And MDCK Cell Lines. Future Med. Chem. 2011, 3, 2063−
(24) Brandsch, M. Drug Transport Via The Intestinal Peptide 2077.
Transporter PEPT1. Curr. Opin. Pharmacol. 2013, 13, 881−887. (44) Di, L.; Whitney-Pickett, C.; Umland, J. P.; Zhang, H.; Zhang, X.;
(25) Hillgren, K. M.; Keppler, D.; Zur, A. A.; Giacomini, K. M.; Gebhard, D. F.; Lai, Y.; Federico, J. J.; Davidson, R. E.; Smith, R.; et al.
Stieger, B.; Cass, C. E.; Zhang, L. Emerging Transporters Of Clinical Development Of A New Permeability Assay Using Low-Efflux Mdckii
Importance: An Update From The International Transporter Cells. J. Pharm. Sci. 2011, 100, 4974−4985.
Consortium. Clin. Pharmacol. Ther. 2013, 94, 52−63. (45) Chatterjee, J.; Gilon, C.; Hoffman, A.; Kessler, H. N-
(26) Li, X.; Yu, M.; Fan, W.; Gan, Y.; Hovgaard, L.; Yang, M. Orally Methylation Of Peptides: A New Perspective In Medicinal Chemistry.
Active-Targeted Drug Delivery Systems For Proteins And Peptides. Acc. Chem. Res. 2008, 41, 1331−1342.
Expert Opin. Drug Delivery 2014, 11, 1435−1447. (46) Bockus, A. T.; Mcewen, C. M.; Lokey, R. S. Form And Function
(27) Bhutia, Y. D.; Babu, E.; Ramachandran, S.; Yang, S.; Thangaraju, In Cyclic Peptide Natural Products: A Pharmacokinetic Perspective.
M.; Ganapathy, V. SLC Transporters As A Novel Class Of Tumour Curr. Top. Med. Chem. 2013, 13, 821−836.
Suppressors: Identity, Function And Molecular Mechanisms. Biochem. (47) Whitty, A.; Zhong, M.; Viarengo, L.; Beglov, D.; Hall, D. R.;
J. 2016, 473, 1113−1124. Vajda, S. Quantifying The Chameleonic Properties Of Macrocycles
(28) Boll, M.; Markovich, D.; Weber, W. M.; Korte, H.; Daniel, H.; And Other High-Molecular-Weight Drugs. Drug Discovery Today
Murer, H. Expression Cloning Of A cDNA From Rabbit Small 2016, 21, 712−717.
Intestine Related To Proton-Coupled Transport Of Peptides, Beta- (48) Over, B.; Mccarren, P.; Artursson, P.; Foley, M.; Giordanetto,
Lactam Antibiotics And Ace-Inhibitors. Pfluegers Arch. 1994, 429, F.; Gronberg, G.; Hilgendorf, C.; Lee, M. D.; Matsson, P.; Muncipinto,
146−149. G.; et al. Impact Of Stereospecific Intramolecular Hydrogen Bonding
(29) Dietrich, C. G.; Geier, A.; Oude Elferink, R. P. J. ABC Of Oral On Cell Permeability And Physicochemical Properties. J. Med. Chem.
Bioavailability: Transporters As Gatekeepers In The Gut. Gut 2003, 2014, 57, 2746−2754.
52, 1788−1795. (49) Over, B.; Matsson, P.; Tyrchan, C.; Artursson, P.; Doak, B. C.;
(30) Lown, K. S.; Mayo, R. R.; Leichtman, A. B.; Hsiao, H. L.; Foley, M. A.; Hilgendorf, C.; Johnston, S. E.; Lee, M. D. T.; Lewis, R.
Turgeon, D. K.; Schmiedlin-Ren, P.; Brown, M. B.; Guo, W.; Rossi, S. J.; et al. Structural And Conformational Determinants Of Macrocycle
J.; Benet, L. Z.; et al. Pharmacokinetics And Drug Disposition: Role Of Cell Permeability. Nat. Chem. Biol. 2016, 12, 1065−1074.
Intestinal P-Glycoprotein (MDR1) In Interpatient Variation In The (50) Wang, C. K.; Craik, D. J. Cyclic Peptide Oral Bioavailability:
Oral Bioavailability Of Cyclosporine. Clin. Pharmacol. Ther. 1997, 62, Lessons From The Past. Biopolymers 2016, 106, 901−909.
248−260. (51) Ussing, H. H.; Zerahn, K. Active Transport Of Sodium As The
(31) Schinkel, A. H.; Jonker, J. W. Mammalian Drug Efflux Source Of Electric Current In The Short-Circuited Isolated Frog Skin.
Transporters Of The Atp Binding Cassette (ABC) Family: An Acta Physiol. Scand. 1951, 23, 110−127.
Overview. Adv. Drug Delivery Rev. 2003, 55, 3−29. (52) Kato, Y.; Miyazaki, T.; Kano, T.; Sugiura, T.; Kubo, Y.; Tsuji, A.
(32) Kansy, M.; Senner, F.; Gubernator, K. Physicochemical High Involvement Of Influx And Efflux Transport Systems In Gastro-
Throughput Screening: Parallel Artificial Membrane Permeation Assay intestinal Absorption Of Celiprolol. J. Pharm. Sci. 2009, 98, 2529−
In The Description Of Passive Absorption Processes. J. Med. Chem. 2539.
1998, 41, 1007−1010. (53) Clarke, L. L. A Guide To Ussing Chamber Studies Of Mouse
(33) Hidalgo, T. J.; Raub, T. J.; Borchardt, R. T. Characterization Of Intestine. Am. J. Physiol.-Gastr. L. 2009, 296, G1151−G1166.
The Human Colon Carcinoma Cell Line (CACO-2) As A Model (54) Forner, K.; Roos, C.; Dahlgren, D.; Kesisoglou, F.; Konerding,
System For Intestinal Epithelial Permeability. Gastroenterology 1989, M. A.; Mazur, J.; Lennernäs, H.; Langguth, P. Optimization of the
96, 736−749. Ussing chamber setup with excised rat intestinal segments for
(34) Press, B.; Di Grandi, D. Permeability For Intestinal Absorption: dissolution/permeation experiments of poorly soluble drugs. Drug
CACO-2 Assay And Related Issues. Curr. Drug Metab. 2008, 9, 893− Dev. Ind. Pharm. 2017, 43, 338−346.
900. (55) Dietrich, C. G.; De Waart, D. R.; Ottenhoff, R.; Schoots, I. G.;
(35) Gaush, C. R.; Hard, W. L.; Smith, T. F. Characterization Of An Elferink, R. P. J. O. Increased Bioavailability Of The Food-Derived
Established Line Of Canine Kidney Cells (MDCK). Exp. Biol. Med. Carcinogen 2-Amino-1-Methyl-6-Phenylimidazo[4,5-B]Pyridine In
1966, 122, 931−935. MRP2-Deficient Rats. Mol. Pharmacol. 2001, 59, 974−980.
(36) Rabito, C. A.; Tchao, R.; Valentich, J.; Leighton, J. Distribution (56) Guengerich, F. P. Cytochrome P450 And Chemical Toxicology.
And Characteristics Of The Occluding Junctions In A Monolayer Of A Chem. Res. Toxicol. 2008, 21, 70−83.
Cell Line (MDCK) Derived From Canine Kidney. J. Membr. Biol. (57) Park, K.; Kwon, I. C.; Park, K. Oral protein Delivery: Current
1978, 43, 351−365. Status and Future Prospect. React. Funct. Polym. 2011, 71, 280−287.
(37) Cereijido, M.; Robbins, E. S.; Dolan, W. J.; Rotunno, C. A.; (58) Lee, E.; Lee, J.; Jon, S. A Novel Approach To Oral Delivery Of
Sabatini, D. D. Polarized Monolayers Formed By Epithelial Cells On A Insulin By Conjugating With Low Molecular Weight Chitosan.
Permeable And Translucent Support. J. Cell Biol. 1978, 77, 853−880. Bioconjugate Chem. 2010, 21, 1720−1723.
(38) Misfeldt, D. S.; Hamamoto, S. T.; Pitelka, D. R. Transepithelial (59) Henriksen, K.; Bay-Jensen, A. C.; Christiansen, C.; Karsdal, M.
Transport In Cell Culture. Proc. Natl. Acad. Sci. U. S. A. 1976, 73, A. Oral Salmon Calcitonin - Pharmacology In Osteoporosis. Expert
1212−1216. Opin. Biol. Ther. 2010, 10, 1617−1629.
(39) Leighton, J.; Estes, L. W.; Mansukhani, S.; Brada, Z. A Cell Line (60) Aungst, B. J. Absorption Enhancers: Applications and Advances.
Derived From Normal Dog Kidney (Mdck) Exhibiting Qualities Of AAPS J. 2012, 14, 10−18.
Papillary Adenocarcinoma And Of Renal Tubular Epithelium. Cancer (61) Renukuntla, J.; Vadlapudi, A. D.; Patel, A.; Boddu, S. H.; Mitra,
1970, 26, 1022−1028. A. K. Approaches for Enhancing Oral Bioavailability of Peptides and
(40) Lodish, H., et al. In Mol. Cell. Biol.; 4th ed.; Freeman, W. H.: Proteins. Int. J. Pharm. 2013, 447, 75−93.
New York, 2000. (62) Fox, C. B.; Kim, J.; Le, L. V.; Nemeth, C. L.; Chirra, H. D.;
(41) Frömter, E.; Diamond, J. Route Of Passive Ion Permeation In Desai, T. A. Micro/nanofabricated Platforms for Oral Drug Delivery. J.
Epithelia. Nat. New Biol. 1972, 235, 9−13. Controlled Release 2015, 219, 431−444.

8122 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

(63) Bak, A.; Leung, D.; Barrett, S. E.; Forster, S.; Minnihan, E. C.; (82) Hess, S.; Linde, Y.; Ovadia, O.; Safrai, E.; Shalev, D. E.; Swed,
Leithead, A. W.; Cunningham, J.; Toussaint, N.; Crocker, L. S. A.; Halbfinger, E.; Lapidot, T.; Winkler, I.; Gabinet, Y.; et al. Backbone
Physicochemical and Formulation Developability Assessment for Cyclic Peptidomimetic Melanocortin-4 Receptor Agonist As A Novel
Therapeutic Peptide DeliveryA Primer. AAPS J. 2015, 17, 144−155. Orally Administrated Drug Lead For Treating Obesity. J. Med. Chem.
(64) Giordanetto, F.; Kihlberg, J. Macrocyclic Drugs And Clinical 2008, 51, 1026−1034.
Candidates: What Can Medicinal Chemists Learn From Their (83) Chan, K. K.; Bakhtiar, R.; Jiang, C. Depsipeptide (FR901228,
Properties? J. Med. Chem. 2014, 57, 278−295. Nsc-630176) Pharmacokinetics In The Rat By LC/MS/MS. Invest.
(65) Doak, B. C.; Over, B.; Giordanetto, F.; Kihlberg, J. Oral New Drugs 1997, 15, 195−206.
Druggable Space Beyond The Rule Of 5: Insights From Drugs And (84) Lai, M.-T.; Yang, C.-C.; Lin, T.-Y.; Tsai, F.-J.; Chen, W.-C.
Clinical Candidates. Chem. Biol. 2014, 21, 1115−1142. Depsipeptide (FK228) Inhibits Growth Of Human Prostate Cancer
(66) Matsson, P.; Doak, B. C.; Over, B.; Kihlberg, J. Cell Permeability Cells. Urol. Oncol: Sem. Orig. Invest. 2008, 26, 182−189.
Beyond The Rule Of 5. Adv. Drug Delivery Rev. 2016, 101, 42−61. (85) Taori, K.; Paul, V. J.; Luesch, H. Structure And Activity Of
(67) Matsson, P.; Kihlberg, J. How Big Is Too Big for Cell Largazole, A Potent Antiproliferative Agent From The Floridian
Permeability? J. Med. Chem. 2017, 60, 1662−1664. Marine Cyanobacterium Symploca SP. J. Am. Chem. Soc. 2008, 130,
(68) Santos, G. B.; Ganesan, A.; Emery, F. S. Oral Administration Of 1806−1807.
Peptide-Based Drugs: Beyond Lipinski’s Rule. ChemMedChem 2016, (86) Bowers, A.; West, N.; Taunton, J.; Schreiber, S. L.; Bradner, J.
11, 2245−2251. E.; Williams, R. M. Total Synthesis And Biological Mode Of Action Of
(69) Ertl, P.; Rohde, B.; Selzer, P. Fast Calculation Of Molecular Largazole: A Potent Class I Histone Deacetylase Inhibitor. J. Am.
Polar Surface Area As A Sum Of Fragment-Based Contributions And Chem. Soc. 2008, 130, 11219−11222.
Its Application To The Prediction Of Drug Transport Properties. J. (87) Bowers, A. A.; Greshock, T. J.; West, N.; Estiu, G.; Schreiber, S.
Med. Chem. 2000, 43, 3714−3717. L.; Wiest, O.; Williams, R. M.; Bradner, J. E. Synthesis And
(70) Aldrich, J. V.; Senadheera, S. N.; Ross, N. C.; Ganno, M. L.; Conformation-Activity Relationships Of The Peptide Isosteres Of
Eans, S. O.; Mclaughlin, J. P. The Macrocyclic Peptide Natural Product Fk228 And Largazole. J. Am. Chem. Soc. 2009, 131, 2900−2905.
CJ-15,208 Is Orally Active And Prevents Reinstatement Of (88) Ghosh, A. K.; Kulkarni, S. Enantioselective Total Synthesis Of
Extinguished Cocaine-Seeking Behavior. J. Nat. Prod. 2013, 76, (+)-Largazole, A Potent Inhibitor Of Histone Deacetylase. Org. Lett.
433−438. 2008, 10, 3907−3909.
(71) Shin, B. S.; Chang, H. S.; Park, E. H.; Yoon, C. H.; Kim, H. Y.; (89) Nasveschuk, C. G.; Ungermannova, D.; Liu, X.; Phillips, A. J. A
Kim, J.; Ryu, J. K.; Zee, O. P.; Lee, K. C.; Cao, D.; et al. Concise Total Synthesis Of Largazole, Solution Structure, And Some
Pharmacokinetics Of A Novel Histone Deacetylase Inhibitor, Apicidin, Preliminary Structure Activity Relationships. Org. Lett. 2008, 10,
In Rats. Biopharm. Drug Dispos. 2006, 27, 69−75. 3595−3598.
(72) Stähelin, H.; Trippmacher, A. Cytostatic Activity Of (90) Seiser, T.; Kamena, F.; Cramer, N. Synthesis And Biological
Chlamydocin, A Rapidly Inactivated Cyclic Tetrapeptide. Eur. J. Activity Of Largazole And Derivatives. Angew. Chem., Int. Ed. 2008, 47,
Cancer 1974, 10, 801−808. 6483−6485.
(73) Kranz, M.; Murray, P. J.; Taylor, S.; Upton, R. J.; Clegg, W.; (91) Ying, Y.; Taori, K.; Kim, H.; Hong, J.; Luesch, H. Total
Elsegood, M. R. J. Solution, Solid Phase And Computational Synthesis And Molecular Target Of Largazole, A Histone Deacetylase
Structures Of Apicidin And Its Backbone-Reduced Analogs. J. Pept. Inhibitor. J. Am. Chem. Soc. 2008, 130, 8455−8459.
Sci. 2006, 12, 383−388. (92) Cole, K. E.; Dowling, D. P.; Boone, M. A.; Phillips, A. J.;
(74) Namatame, I.; Tomoda, H.; Ishibashi, S.; Omura, S. Christianson, D. W. Structural Basis Of The Antiproliferative Activity
Antiatherogenic Activity Of Fungal Beauveriolides, Inhibitors Of Of Largazole, A Depsipeptide Inhibitor Of The Histone Deacetylases.
Lipid Droplet Accumulation In Macrophages. Proc. Natl. Acad. Sci. U. J. Am. Chem. Soc. 2011, 133, 12474−12477.
S. A. 2004, 101, 737−742. (93) Decroos, C.; Clausen, D. J.; Haines, B. E.; Wiest, O.; Williams,
(75) Hurevich, M.; Swed, A.; Joubran, S.; Cohen, S.; Freeman, N. S.; R. M.; Christianson, D. W. Variable Active Site Loop Conformations
Britan-Rosich, E.; Briant-Longuet, L.; Bardy, M.; Devaux, C.; Kotler, Accommodate the Binding of Macrocyclic Largazole Analogues to
M.; et al. Rational Conversion Of Noncontinuous Active Region In HDAC8. Biochemistry 2015, 54, 2126−2135.
Proteins Into A Small Orally Bioavailable Macrocyclic Drug-Like (94) Yu, M.; Salvador, L. A.; Sy, S. K.; Tang, Y.; Singh, R. S.; Chen,
Molecule: The HIV-1 CD4:Gp120 Paradigm. Bioorg. Med. Chem. Q. Y.; Liu, Y.; Hong, J.; Derendorf, H.; Luesch, H. Largazole
2010, 18, 5754−5761. Pharmacokinetics In Rats By LC-MS/MS. Mar. Drugs 2014, 12,
(76) Burcham, D. L.; Aungst, B. A.; Hussain, M.; Gorko, M. A.; 1623−1640.
Quon, C. Y.; Huang, S. M. The Effect Of Absorption Enhancers On (95) Salvador, L. A.; Park, H.; Al-Awadhi, F. H.; Liu, Y.; Kim, B.;
The Oral Absorption Of The GP IIb/IIIa Receptor Antagonist, Dmp Zeller, S. L.; Chen, Q. Y.; Hong, J.; Luesch, H. Modulation Of Activity
728, In Rats And Dogs. Pharm. Res. 1995, 12, 2065−2070. Profiles For Largazole-Based Hdac Inhibitors Through Alteration Of
(77) Terao, K.; Ito, E.; Tatsuno, T. Liver Injuries Induced By Prodrug Properties. ACS Med. Chem. Lett. 2014, 5, 905−910.
Cyclochlorotine Isolated From Penicillium Islandicum. Arch. Toxicol. (96) Monk, P. N.; Scola, A.-M.; Madala, P. K.; Fairlie, D. P. Function,
1984, 55, 39−46. Structure And Therapeutic Potential Of Complement C5a Receptors.
(78) Uraguchi, K.; Saito, M.; Noguchi, Y.; Takahashi, K.; Enomoto, Br. J. Pharmacol. 2007, 152, 429−448.
M. Chronic Toxicity And Carcinogenicity In Mice Of The Purified (97) Wong, A. K.; Finch, A. M.; Pierens, G. K.; Craik, D. J.; Taylor, S.
Mycotoxins, Luteoskyrin And Cyclochlorotine. Food Cosmet. Toxicol. M.; Fairlie, D. P. Small Molecular Probes For G-Protein-Coupled C5a
1972, 10, 193−207. Receptors: Conformationally Constrained Antagonists Derived From
(79) Xu, H.-M.; Zeng, G.-Z.; Zhou, W.-B.; He, W.-J.; Tan, N.-H. The C-Terminus Of The Human Plasma Protein C5a. J. Med. Chem.
Astins K-P Six New Chlorinated Cyclopentapeptides From Aster 1998, 41, 3417−3425.
Tataricus. Tetrahedron 2013, 69, 7964−7969. (98) Finch, A. M.; Wong, A. K.; Wadi, S. K.; Paczkowski, N. J.;
(80) Shen, Y.; Luo, Q.; Xu, H. M.; Gong, F. Y.; Zhou, X. B.; Sun, Y.; Fairlie, D. P.; Taylor, S. M. Low Molecular Weight Peptidic And
Wu, X. F.; Liu, W.; Zeng, G. Z.; Tan, N. H.; et al. Mitochondria- Cyclic Antagonists Of The Receptor For The Complement Factor
Dependent Apoptosis Of Activated T Lymphocytes Induced By Astin C5a. J. Med. Chem. 1999, 42, 1965−1974.
C, A Plant Cyclopeptide, For Preventing Murine Experimental Colitis. (99) March, D. R.; Proctor, L. M.; Stoermer, M. J.; Sbaglia, R.;
Biochem. Pharmacol. 2011, 82, 260−268. Abbenante, G.; Reid, R. C.; Wadi, K.; Paczkowski, N.; Tyndall, J. D. A.;
(81) Cummings, D. E.; Schwartz, M. W. Melanocortins And Body Taylor, S. M.; Fairlie, D. P. Potent Cyclic Antagonists Of The
Weight: A Tale Of Two Receptors. Nat. Genet. 2000, 26, 8−9. Complement C5a Receptor On Human Polymorphonuclear Leuko-

8123 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

cytes. Relationships Between Structures And Activity. Mol. Pharmacol. Conformational Flexibility And Receptor Binding. Science 1986, 232,
2004, 65, 868−879. 633−636.
(100) Seow, V.; Lim, J.; Cotterell, A. J.; Yau, M. K.; Xu, W.; Lohman, (116) Goetz, G. H.; Philippe, L.; Shapiro, M. J. Epsa: A Novel
R. J.; Kok, W. M.; Stoermer, M. J.; Sweet, M. J.; Reid, R. C.; et al. Supercritical Fluid Chromatography Technique Enabling The Design
Receptor Residence Time Trumps Drug-Likeness And Oral Of Permeable Cyclic Peptides. ACS Med. Chem. Lett. 2014, 5, 1167−
Bioavailability In Determining Efficacy Of Complement C5a 1172.
Antagonists. Sci. Rep. 2016, 6, 24575. (117) Zornes, L. L.; Stratford, R. E. Development Of A Plasma High-
(101) Waksman, S. A.; Woodruff, H. B. Bacteriostatic And Performance Liquid Chromatographic Assay For LY303366, A
Bactericidal Substances Produced By A Soil Actinomyces. Exp. Biol. Lipopeptide Antifungal Agent, And Its Application In A Dog
Med. 1940, 45, 609−614. Pharmacokinetic Study. J. Chromatogr., Biomed. Appl. 1997, 695,
(102) Jain, S. C.; Sobell, H. M. Stereochemistry Of Actinomycin 381−387.
Binding To DNA. I. Refinement And Further Structural Details Of (118) Li, C.; Fleisher, D.; Li, L.; Schwier, J. R.; Sweetana, S. A.;
The Actinomycin-Deoxyguanosine Crystalline Complex. J. Mol. Biol. Vasudevan, V.; Zornes, L. L.; Pao, L. H.; Zhou, S. Y.; Stratford, R. E.
1972, 68, 1−20. Regional-Dependent Intestinal Absorption And Meal Composition
(103) Lo, Y. S.; Tseng, W. H.; Chuang, C. Y.; Hou, M. H. The Effects On Systemic Availability Of LY303366, A Lipopeptide
Structural Basis Of Actinomycin D-Binding Induces Nucleotide Antifungal Agent, In Dogs. J. Pharm. Sci. 2001, 90, 47−57.
Flipping Out, A Sharp Bend And A Left-Handed Twist In CGG (119) Raasch, R. H. Anidulafungin: Review Of A New Echinocandin
Triplet Repeats. Nucleic Acids Res. 2013, 41, 4284−4294. Antifungal Agent. Expert Rev. Anti-Infect. Ther. 2004, 2, 499−508.
(104) Robinson, H.; Gao, Y. G.; Yang, X.; Sanishvili, R.; Joachimiak, (120) Denning, D. W. Echinocandin Antifungal Drugs. Lancet 2003,
A.; Wang, A. H. Crystallographic Analysis Of A Novel Complex Of 362, 1142.
Actinomycin D Bound To The DNA Decamer CGATCGATCG. (121) Ikeda, F.; Tanaka, S.; Ohki, H.; Matsumoto, S.; Maki, K.;
Biochemistry 2001, 40, 5587−92. Katashima, M.; Barrett, D.; Aoki, Y. Role Of Micafungin In The
(105) Tan, C. T.; Dargeon, H. W.; Burchenal, J. H. The Effect Of Antifungal Armamentarium. Curr. Med. Chem. 2007, 14, 1263−1275.
Actinomycin D On Cancer In Childhood. Pediatrics 1959, 24, 544− (122) Morris, M. I.; Villmann, M. Echinocandins In The Manage-
561. ment Of Invasive Fungal Infections, Part 1. Am. J. Health-Syst. Pharm.
(106) Hill, T. A.; Lohman, R. J.; Hoang, H. N.; Nielsen, D. S.; Scully, 2006, 63, 1693−1703.
C. C.; Kok, W. M.; Liu, L.; Lucke, A. J.; Stoermer, M. J.; Schroeder, C. (123) Veber, D. F.; Holly, F. W.; Nutt, R. F.; Bergstrand, S. J.; Brady,
I.; et al. Cyclic Penta- And Hexa-Leucine Peptides Without N- S. F.; Hisrschmann, R.; Glitzer, M. S.; Saperstein, R. Highly Active
Methylation Are Orally Absorbed. ACS Med. Chem. Lett. 2014, 5, Cyclic And Bicyclic Somatostatin Analogues Of Reduced Ring Size.
1148−1151. Nature 1979, 280, 512−514.
(107) Robson, W. L. M.; Leung, A. K. C.; Norgaard, J. P. The (124) Veber, D. F.; Freidinger, R. M.; Perlow, D. S.; Paleveda, W. J.;
Comparative Safety Of Oral Versus Intranasal Desmopressin For The Holly, F. W.; Strachan, R. G.; Nutt, R. F.; Arison, B. H.; Homnick, C.;
Treatment Of Children With Nocturnal Enuresis. J. Urol. 2007, 178,
Randall, W. C.; et al. A Potent Cyclic Hexapeptide Analogue Of
24−30.
Somatostatin. Nature 1981, 292, 55−58.
(108) Rembratt, A.; Graugaard-Jensen, C.; Senderovitz, T.; Norgaard,
(125) Biron, E.; Chatterjee, J.; Ovadia, O.; Langenegger, D.;
J. P.; Djurhuus, J. C. Pharmacokinetics And Pharmacodynamics Of
Brueggen, J.; Hoyer, D.; Schmid, H. A.; Jelinek, R.; Gilon, C.;
Desmopressin Administered Orally Versus Intravenously At Daytime
Hoffman, A.; et al. Improving Oral Bioavailability Of Peptides By
Versus Night-Time In Healthy Men Aged 55−70 Years. Eur. J. Clin.
Multiple N-Methylation: Somatostatin Analogues. Angew. Chem., Int.
Pharmacol. 2004, 60, 397−402.
(109) Fjellestad-Paulsen, A.; Hoglund, P.; Lundin, S.; Paulsen, O. Ed. 2008, 47, 2595−2599.
Pharmacokinetics Of 1-Deamino-8-D-Arginine Vasopressin After (126) Bauer, W.; Briner, U.; Doepfner, W.; Haller, R.; Huguenin, R.;
Various Routes Of Administration In Healthy Volunteers. Clin. Marbach, P.; Petcher, T. J.; Pless, J. SMS 201−995: A Very Potent And
Endocrinol. 1993, 38, 177−182. Selective Octapeptide Analogue Of Somatostatin With Prolonged
(110) D’agay-Abensour, L.; Fjellestad-Paulsen, A.; Höglund, P.; Ngô, Action. Life Sci. 1982, 31, 1133−1140.
Y.; Paulsen, O.; Rambaud, J. C. Absolute Bioavailability Of An (127) Melacini, G.; Zhu, Q.; Goodman, M. Multiconformational
Aqueous Solution Of 1-Deamino-8-D-Arginine Vasopressin From NMR Analysis Of Sandostatin (Octreotide): Equilibrium Between
Different Regions Of The Gastrointestinal Tract In Man. Eur. J. Clin. Beta-Sheet And Partially Helical Structures. Biochemistry 1997, 36,
Pharmacol. 1993, 44, 473−476. 1233−1241.
(111) Lan, E.-L.; Ugwu, S. O.; Blanchard, J.; Fang, X.; Hruby, V. J.; (128) Spyroulias, G. A.; Galanis, A. S.; Petrou, C. H.; Vahliotis, D.;
Sharma, S. Preformulation Studies With Melanotan-II: A Potential Sotiriou, P.; Nikolopoulou, A.; Nock, B.; Maina, T.; Cordopatis, P. 3D
Skin Cancer Chemopreventive Peptide. J. Pharm. Sci. 1994, 83, 1081− Solution Structure Of [Tyr3]Octreotate Derivatives In DMSO:
1084. Structure Differentiation Of Peptide Core Due To Chelate Group
(112) Meyer-Lindenberg, A.; Domes, G.; Kirsch, P.; Heinrichs, M. Attachment And Biologically Active Conformation. Med. Chem. 2005,
Oxytocin And Vasopressin In The Human Brain: Social Neuro- 1, 487−499.
peptides For Translational Medicine. Nat. Rev. Neurosci. 2011, 12, (129) Pohl, E.; Heine, A.; Sheldrick, G. M.; Dauter, Z.; Wilson, K. S.;
524−538. Kallen, J.; Huber, W.; Pfaffli, P. J. Structure of Octreotide, a
(113) Manning, M.; Misicka, A.; Olma, A.; Bankowski, K.; Stoev, S.; Somatostatin Analogue. Acta Crystallogr., Sect. D: Biol. Crystallogr.
Chini, B.; Durroux, T.; Mouillac, B.; Corbani, M.; Guillon, G. 1995, 51, 48−59.
Oxytocin And Vasopressin Agonists And Antagonists As Research (130) Köhler, E.; Duberow-Drewe, M.; Drewe, J.; Ribes, G.;
Tools And Potential Therapeutics. J. Neuroendocrinol. 2012, 24, 609− Loubatières-Mariani, M. M.; Mazer, N.; Gyr, K.; Beglinger, C.
628. Absorption Of An Aqueous Solution Of A New Synthetic
(114) de Araujo, A. D.; Mobli, M.; Castro, J.; Harrington, A. M.; Somatostatin Analogue Administered To Man By Gavage. Eur. J.
Vetter, I.; Dekan, Z.; Muttenthaler, M.; Wan, J.; Lewis, R. J.; King, G. Clin. Pharmacol. 1987, 33, 167−171.
F.; Brierley, S. M.; Alewood, P. F. Selenoether Oxytocin Analogues (131) Williams, G.; Burrin, J. M.; Ball, J. A.; Joplin, G. F.; Bloom, S.
Have Analgesic Properties In A Mouse Model Of Chronic Abdominal R. Effective And Lasting Growth-Hormone Suppression In Active
Pain. Nat. Commun. 2014, 5, 3165. Acromegaly With Oral Administration Of Somatostatin Analogue SMS
(115) Wood, S. P.; Tickle, I. J.; Treharne, A. M.; Pitts, J. E.; 201−995. Lancet 1986, 328, 774−778.
Mascarenhas, Y.; Li, J. Y.; Husain, J.; Cooper, S.; Blundell, T. L.; (132) Albert, R.; Marbach, P.; Bauer, W.; Briner, U.; Fricker, G.;
Hruby, V. J.; et al. Crystal Structure Of Deamino-Oxytocin: Brums, C.; Pless, J. SDZ CO 611: A Highly Potent Glycated Analog

8124 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Of Somatostatin With Improved Oral Activity. Life Sci. 1993, 53, 517− Experience At An Australian Hospital. Antimicrob. Agents Chemother.
525. 2010, 54, 3949−3952.
(133) Nelson-Piercy, C.; Hammond, P. J.; Gwilliam, M. E.; Khandan- (152) Andes, D.; Craig, W. A. Pharmacodynamics Of A New
Nia, N.; Myers, M. J.; Ghatei, M. A.; Bloom, S. R. Effect Of A New Streptogramin, XRP 2868, In Murine Thigh And Lung Infection
Oral Somatostatin Analog (SDZ CO 611) On Gastric Emptying, Models. Antimicrob. Agents Chemother. 2006, 50, 243−249.
Mouth To Cecum Transit Time, And Pancreatic And Gut Hormone (153) Rezai, T.; Yu, B.; Millhauser, G. L.; Jacobson, M. P.; Lokey, R.
Release In Normal Male Subjects. J. Clin. Endocrinol. Metab. 1994, 78, S. Testing The Conformational Hypothesis Of Passive Membrane
329−336. Permeability Using Synthetic Cyclic Peptide Diastereomers. J. Am.
(134) Geddes, A. J.; Akrigg, D. The Crystal Structure of Hydrated Chem. Soc. 2006, 128, 2510−2511.
Beauvericin. Acta Crystallogr., Sect. B: Struct. Crystallogr. Cryst. Chem. (154) Rezai, T.; Bock, J. E.; Zhou, M. V.; Kalyanaraman, C.; Lokey,
1976, 32, 3164−3171. R. S.; Jacobson, M. P. Conformational Flexibility, Internal Hydrogen
(135) Braden, B.; Hamilton, J. A.; Sabesan, M. N.; Steinrauf, L. K. Bonding, And Passive Membrane Permeability: Successful In Silico
Crystal Structure Of A Beauvericin-Barium Picrate Complex. J. Am. Prediction Of The Relative Permeabilities Of Cyclic Peptides. J. Am.
Chem. Soc. 1980, 102, 2704−2709. Chem. Soc. 2006, 128, 14073−14080.
(136) Plattner, R. D.; Nelson, P. E. Production Of Beauvericin By A (155) White, T. R.; Renzelman, C. M.; Rand, A. C.; Rezai, T.;
Strain Of Fusarium Proliferatum Isolated From Corn Fodder For Mcewen, C. M.; Gelev, V. M.; Turner, R. A.; Linington, R. G.; Leung,
Swine. Appl. Environ. Microbiol. 1994, 60, 3894−3896. S. S. F.; Kalgutkar, A. S.; et al. On-Resin N-Methylation Of Cyclic
(137) Mei, L.; Zhang, L.; Dai, R. An Inhibition Study Of Beauvericin Peptides For Discovery Of Orally Bioavailable Scaffolds. Nat. Chem.
On Human And Rat Cytochrome P450 Enzymes And Its Biol. 2011, 7, 810−817.
Pharmacokinetics In Rats. J. Enzyme Inhib. Med. Chem. 2009, 24, (156) Rand, A. C.; Leung, S. S.; Eng, H.; Rotter, C. J.; Sharma, R.;
753−762. Kalgutkar, A. S.; Zhang, Y.; Varma, M. V.; Farley, K. A.; Khunte, B.;
(138) Streit, E.; Schwab, C.; Sulyok, M.; Naehrer, K.; Krska, R.; et al. Optimizing PK Properties Of Cyclic Peptides: The Effect Of Side
Schatzmayr, G. Multi-Mycotoxin Screening Reveals The Occurrence Chain Substitutions On Permeability And Clearance. MedChemComm
Of 139 Different Secondary Metabolites In Feed And Feed 2012, 3, 1282−1289.
Ingredients. Toxins 2013, 5, 504−523. (157) Schwochert, J.; Turner, R.; Thang, M.; Berkeley, R. F.; Ponkey,
(139) Jestoi, M. Emerging Fusarium-Mycotoxins Fusaproliferin, A. R.; Rodriguez, K. M.; Leung, S. S.; Khunte, B.; Goetz, G.
Beauvericin, Enniatins, And Moniliformin: A Review. Crit. Rev. Food Limberakis, C. et al. Peptide To Peptoid Substitutions Increase Cell
Sci. Nutr. 2008, 48, 21−49. Permeability In Cyclic Hexapeptides. Org. Lett. 2015, 17, 2928−2931.
(140) Ovchinnikov, Y. A.; Ivanov, V. T.; Evstratov, A. V.; Mikhaleva, (158) Bockus, A. T.; Lexa, K. W.; Pye, C. R.; Kalgutkar, A. S.;
I. I.; Bystrov, V. F.; Portnova, S. L.; Balashova, T. A.; Meshcheryakova, Gardner, J. W.; Hund, K. C.; Hewitt, W. M.; Schwochert, J. A.;
Glassey, E.; Price, D. A.; et al. Probing The Physicochemical
E. N.; Tulchinsky, V. M. The Enniatin Ionophores. Conformation And
Boundaries Of Cell Permeability And Oral Bioavailability In Lipophilic
Ion Binding Properties. Int. J. Pept. Protein Res. 1974, 6, 465−498.
Macrocycles Inspired By Natural Products. J. Med. Chem. 2015, 58,
(141) Kratky, C.; Dobler, M. The Crystal Structure of Enniatin B.
4581−4589.
Helv. Chim. Acta 1985, 68, 1798−1803.
(159) Lewis, I.; Schaefer, M.; Wagner, T.; Oberer, L.; Sager, E.;
(142) Tishchenko, G. N.; Zhukhlistova, N. E. Crystal and Molecular
Wipfli, P.; Vorherr, T. A Detailed Investigation On Conformation,
Structure of the Membrane-Active Antibiotic Enniatin C. Crystallogr.
Permeability And PK Properties Of Two Related Cyclohexapeptides.
Rep. 2000, 45, 677−683.
Int. J. Pept. Res. Ther. 2015, 21, 205−221.
(143) Tishchenko, G. N.; Karimov, Z. Structure Of Rubidium(1+)
(160) Wang, C. K.; Northfield, S. E.; Colless, B.; Chaousis, S.;
Complex Of The LDLLDL Analog Of Enniatin B In Crystals.
Hamernig, I.; Lohman, R. J.; Nielsen, D. S.; Schroeder, C. I.; Liras, S.;
Kristallografiya (Russ.) Crystallogr.Rep. 1978, 23, 729−742. Price, D. A.; et al. Rational Design And Synthesis Of An Orally
(144) Zhukhlistova, N. E.; Tishchenko, G. N.; Refaat, L.; Woolfson, Bioavailable Peptide Guided By NMR Amide Temperature
M. M. Crystal And Molecular Structure Of (Enniatin B)2.KI Complex. Coefficients. Proc. Natl. Acad. Sci. U. S. A. 2014, 111, 17504−17509.
Kristallografiya (Russ.) Crystallogr. Rep. 1998, 43, 50−57. (161) Nielsen, D. S.; Lohman, R. J.; Hoang, H. N.; Hill, T. A.; Jones,
(145) Fæste, C. K.; Ivanova, L.; Uhlig, S. In Vitro Metabolism Of The A.; Lucke, A. J.; Fairlie, D. P. Flexibility Versus Rigidity For Orally
Mycotoxin Enniatin B In Different Species And Cytochrome P450 Bioavailable Cyclic Hexapeptides. ChemBioChem 2015, 16, 2289−
Enzyme Phenotyping By Chemical Inhibitors. Drug Metab. Dispos. 2293.
2011, 39, 1768−1776. (162) Bechara, C.; Sagan, S. Cell-Penetrating Peptides: 20 Years
(146) Devreese, M.; Broekaert, N.; De Mil, T.; Fraeyman, S.; De Later, Where Do We Stand? FEBS Lett. 2013, 587, 1693−1702.
Backer, P.; Croubels, S. Pilot Toxicokinetic Study And Absolute Oral (163) Kauffman, W. B.; Fuselier, T.; He, J.; Wimley, W. C.
Bioavailability Of The Fusarium Mycotoxin Enniatin B1 In Pigs. Food Mechanism Matters: A Taxonomy Of Cell Penetrating Peptides.
Chem. Toxicol. 2014, 63, 161−165. Trends Biochem. Sci. 2015, 40, 749−764.
(147) Lecci, A.; Carini, F.; Tramontana, M.; D’aranno, V.; Marinoni, (164) Tashima, T. Intelligent Substance Delivery Into Cells Using
E.; Crea, A.; Bueno, L.; Fioramonti, J.; Criscuoli, M.; Giuliani, S.; et al. Cell-Penetrating Peptides. Bioorg. Med. Chem. Lett. 2017, 27, 121−130.
Nepadutant Pharmacokinetics And Dose-Effect Relationships As (165) Qian, Z.; Martyna, A.; Hard, R. L.; Wang, J.; Appiah-Kubi, G.;
Tachykinin NK2 Receptor Antagonist Are Altered By Intestinal Coss, C.; Phelps, M. A.; Rossman, J. S.; Pei, D. Discovery And
Inflammation In Rodent Models. J. Pharmacol. Exp. Ther. 2001, 299, Mechanism Of Highly Efficient Cyclic Cell-Penetrating Peptides.
247−254. Biochemistry 2016, 55, 2601−2612.
(148) https://clinicaltrials.gov/ct2/show/NCT00655083. (166) Hamann, M. T.; Scheuer, P. J. Kahalalide-F - A Bioactive
(149) Jolad, S. D.; Hoffman, J. J.; Torrance, S. J.; Wiedhopf, R. M.; Depsipeptide From The Sacoglossan Mollusk Elysia-Rufescens And
Cole, J. R.; Arora, S. K.; Bates, R. B.; Gargiulo, R. L.; Kriek, G. R. The Green-Alga Bryopsis Sp. J. Am. Chem. Soc. 1993, 115, 5825−5826.
Bouvardin and Deoxybouvardin, Antitumor Cyclic Hexapeptides from (167) Suarez, Y.; Gonzalez, L.; Cuadrado, A.; Berciano, M.; Lafarga,
Bouvardia ternifolia (Rubiaceae). J. Am. Chem. Soc. 1977, 99, 8040− M.; Munoz, A.; Kahalalide, F. A New Marine-Derived Compound,
8044. Induces Oncosis In Human Prostate And Breast Cancer Cells. Mol.
(150) US NIH Toxnet, Substance Name: Bouvardin, 5-(N-methyl-L- Cancer Ther. 2003, 2, 863−872.
tyrosine)-, acetate (ester). https://chem.nlm.nih.gov/chemidplus/rn/ (168) Rademaker-Lakhai, J. M.; Horenblas, S.; Meinhardt, W.;
86229-75-6. Stokvis, E.; De Reijke, T. M.; Jimeno, J. M.; Lopez-Lazaro, L.; Lopez
(151) Reid, A. B.; Daffy, J. R.; Stanley, P.; Buising, K. L. Use Of Martin, J. A.; Beijnen, J. H.; Schellens, J. H. Phase I Clinical And
Pristinamycin For Infections By Gram-Positive Bacteria: Clinical Pharmacokinetic Study Of Kahalalide F In Patients With Advanced

8125 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

Androgen Refractory Prostate Cancer. Clin. Cancer Res. 2005, 11, (186) Sasaki, T.; Takagi, M.; Yaguchi, T.; Miyadoh, S.; Okada, T.;
1854−1862. Koyama, M. A New Anthelmintic Cyclodepsipeptide, PF1022a. J.
(169) Brown, A. P.; Morrissey, R. L.; Faircloth, G. T.; Levine, B. S. Antibiot. 1992, 45, 692−697.
Preclinical Toxicity Studies Of Kahalalide F, A New Anticancer Agent: (187) Kachi, S.; Ishih, A.; Terada, M. Effects Of Pf1022a On Adult
Single And Multiple Dosing Regimens In The Rat. Cancer Chemother. Angiostrongylus Cantonensis In The Pulmonary Arteries And Larvae
Pharmacol. 2002, 50, 333−340. Migrating Into The Central Nervous System Of Rats. Z. Parasitenkd.
(170) Dalisay, D. S.; Rogers, E. W.; Edison, A. S.; Molinski, T. F. 1995, 81, 631−637.
Structure Elucidation At The Nanomole Scale. 1. Trisoxazole (188) Von Samson-Himmelstjerna, G.; Harder, A.; Sangster, N. C.;
Macrolides And Thiazole-Containing Cyclic Peptides From The Coles, G. C. Efficacy Of Two Cyclooctadepsipeptides, Pf1022a And
Nudibranch Hexabranchus Sanguineus. J. Nat. Prod. 2009, 72, 732− Emodepside, Against Anthelmintic-Resistant Nematodes In Sheep
738. And Cattle. Parasitology 1999, 130, 343−347.
(171) Nielsen, D. S.; Hoang, H. N.; Lohman, R. J.; Diness, F.; Fairlie, (189) Harder, A.; Holden-Dye, L.; Walker, R.; Wunderlich, F.
D. P. Total Synthesis, Structure, And Oral Absorption Of A Thiazole Mechanisms Of Action Of Emodepside. Parasitol. Res. 2005, 97, S1−
Cyclic Peptide, Sanguinamide A. Org. Lett. 2012, 14, 5720−5723. S10.
(172) Nielsen, D. S.; Hoang, H. N.; Lohman, R. J.; Hill, T. A.; Lucke, (190) Harder, A.; Schmitt-Wrede, H.-P.; Krucken, J.; Marinovski, P.;
A. J.; Craik, D. J.; Edmonds, D. J.; Griffith, D. A.; Rotter, C. J.; Ruggeri, Wunderlich, F.; Willson, J.; Amliwala, K.; Holden-Dye, L.; Walker, R.
R. B.; et al. Improving On Nature: Making A Cyclic Heptapeptide Cyclooctadepsipeptides - An Anthelmintically Active Class Of
Orally Bioavailable. Angew. Chem., Int. Ed. 2014, 53, 12059−12063. Compounds Exhibiting A Novel Mode Of Action. Int. J. Antimicrob.
(173) Steyn, P. S.; Tuinman, A. A.; Van Heerden, F. R.; Van Rooyen, Agents 2003, 22, 318−331.
P. H.; Wessels, P. L.; Rabie, C. J. The Isolation, Structure, And (191) Ohyama, M.; Okada, Y.; Takahashi, M.; Sakanaka, O.;
Absolute Configuration Of The Mycotoxin, Rhizonin A, A Novel Matsumoto, M.; Atsumi, K. Structure-Activity Relationship Of
Cyclic Heptapeptide Containing N-Methyl-3-(3-Furyl)Alanine, Pro- Anthelmintic Cyclooctadepsipeptides. Biosci., Biotechnol., Biochem.
duced By Rhizopus Microsporus. J. Chem. Soc., Chem. Commun. 1983, 2011, 75, 1354−1363.
47−49. (192) Baronsky, J.; Bongaerts, S.; Traeubel, M.; Weiss, H.-C.;
(174) Wilson, T.; Rabie, C. J.; Fincham, J. E.; Steyn, P. S.; Schipper, Urbanetz, N. The Study Of Different Solid Forms Of Emodepside.
M. A. Toxicity Of Rhizonin A, Isolated From Rhizopus Microsporus, Eur. J. Pharm. Biopharm. 2009, 71, 88−99.
In Laboratory Animals. Food Chem. Toxicol. 1984, 22, 275−281. (193) https://clinicaltrials.gov/ct2/show/NCT02661178.
(175) Ernst, B.; Hoeger, S. J.; O’brien, E.; Dietrich, D. R. Oral (194) Gao, Z. Q.; Zhao, X. Y.; Lee, S.; Li, J. J.; Liao, H.; Zhou, X. H.;
Toxicity Of The Microcystin-Containing Cyanobacterium Planktothrix Wu, J.; Qi, G. F. WH1fungin A Surfactin Cyclic Lipopeptide Is A
Rubescens In European Whitefish (Coregonus Lavaretus). Aquat. Novel Oral Immunoadjuvant. Vaccine 2013, 31, 2796−2803.
(195) Gao, Z. Q.; Zhao, X. Y.; Yang, T.; Shang, J.; Shang, L.; Mai, H.
Toxicol. 2006, 79, 31−40.
Z.; Qi, G. F. Immunomodulation Therapy Of Diabetes By Oral
(176) Yoshida, T.; Makita, Y.; Nagata, S.; Tsutsumi, T.; Yoshida, F.;
Administration Of A Surfactin Lipopeptide In NOD Mice. Vaccine
Sekijima, M.; Tamura, S.; Ueno, Y. Acute Oral Toxicity Of
2014, 32, 6812−6819.
Microcystin-LR, A Cyanobacterial Hepatotoxin, In Mice. Nat. Toxins
(196) Wieland, T.; Faulstich, H. Amatoxins, Phallotoxins, Phallolysin,
1997, 5, 91−95.
And Antamanide: The Biologically Active Components Of Poisonous
(177) Ito, E.; Kondo, F.; Harada, K. First Report On The
Amanita Mushrooms. Crit. Rev. Biochem. 1978, 5, 185−260.
Distribution Of Orally Administered Microcystin-LR In Mouse Tissue
(197) Faulstich, H.; Buku, A.; Bodenmuller, H.; Wieland, T.
Using An Immunostaining Method. Toxicon 2000, 38, 37−48. Virotoxins: Actin-Binding Cyclic Peptides Of Amanita Virosa
(178) Kawasaki, T.; Taniguchi, M.; Moritani, Y.; Uemura, T.;
Mushrooms. Biochemistry 1980, 19, 3334−3343.
Shigenaga, T.; Takamatsu, H.; Hayashi, K.; Takasaki, J.; Saito, T.; (198) Wong, J. H.; Ng, T. B. In Handbook Of Biologically Active
Nagai, K. Pharmacological Properties Of YM-254890, A Specific Peptides; Abba, J. K., Ed.; Academic Press: New York, 2006; pp 131−
G(Alpha)q/11 Inhibitor, On Thrombosis And Neointima Formation 135.
In Mice. Thromb. Haemostasis 2005, 94, 184−192. (199) Walton, J. D.; Hallen-Adams, H. E.; Luo, H. Ribosomal
(179) Cunningham, T. J.; Greenstein, J.; Yao, L. Uncompetitive Biosynthesis Of The Cyclic Peptide Toxins Of Amanita Mushrooms.
Phospholipase A2 Inhibition By CHEC Sequences Including Oral Biopolymers 2010, 94, 659−664.
Treatment Of Experimental Autoimmune Myeloencephalitis. Open (200) Bushnell, D. A.; Cramer, P.; Kornberg, R. D. Structural Basis of
Enzyme Inhib. J. 2009, 2, 1−7. Transcription: Alpha-Amanitin-RNA Polymerase II Cocrystal at 2.8 Å
(180) Kicielinska, J.; Szczygiel, A.; Rossowska, J.; Anger, N.; Resolution. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 1218−122.
Kempinska, K.; Switalska, M.; Kaszowska, M.; Wietrzyk, J.; (201) Brueckner, F.; Cramer, P. Structural Basis of Transcription
Boratynski, J.; Pajtasz-Piasecka, E. Oral Administration Of Polymyxin Inhibition by Alpha-Amanitin and Implications for RNA Polymerase II
B Modulates The Activity Of Lipooligosaccharide E. Coli B Against Translocation. Nat. Struct. Mol. Biol. 2008, 15, 811−818.
Lung Metastases In Murine Tumor Models. PLoS One 2016, 11, (202) Kaplan, C. D.; Larsson, K. M.; Kornberg, R. D. The RNA
E0148156. Polymerase II Trigger Loop Functions in Substrate Selection and is
(181) Adachi, Y.; Enomoto, M.; Adachi, M.; Suwa, M.; Nagamine, Y.; Directly Targeted by Alpha-Amanitin. Mol. Cell 2008, 30, 547−556.
Nanno, T.; Hashimoto, T.; Inoue, H.; Yamamoto, T. Enteric Coated (203) Wieland, T.; Gotzendorfer, C.; Dabrowski, J.; Lipscomb, W.
Polymyxin B In The Treatment Of Hyperammonemia And N.; Shoham, G. Unexpected Similarity of the Structures of the Weakly
Endotoxemia In Liver Cirrhosis. Gastroenterol. Jpn. 1982, 17, 550−557. Toxic Amanitin (S)-Sulfoxide and the Highly Toxic (R)-Sulfoxide and
(182) Dudley, M. N.; Mclaughlin, J. C.; Carrington, G.; Frick, J. Sulfone As Revealed by Proton Nuclear Magnetic Resonance and X-
Nightingale, C. H.; Quintiliani, R. Oral Bacitracin Vs Vancomycin ray Analysis. Biochemistry 1983, 22, 1264−1271.
Therapy For Clostridium Difficile-Induced Diarrhea. A Randomized (204) Shoham, G.; Lipscomb, W. N.; Wieland, T. Conformations of
Double-Blind Trial. Arch. Intern. Med. 1986, 146, 1101−1104. Amatoxins in the Crystalline State. J. Am. Chem. Soc. 1989, 111, 4791−
(183) Bond, G. C.; Vanderbrook, M. J.; Wiley, J. L.; Nook, M. A. 4809.
Oral Administration Of Bacitracin. Exp. Biol. Med. 1948, 68, 395−400. (205) Mas, A. Mushrooms, Amatoxins And The Liver. J. Hepatol.
(184) Weinstein, M. R.; Dedier, H.; Brunton, J.; Campbell, I.; Conly, 2005, 42, 166−169.
J. M. Lack Of Efficacy Of Oral Bacitracin Plus Doxycycline For The (206) Wieland, T. Peptides Of Poisonous Amanita Mushrooms;
Eradication Of Stool Colonization With Vancomycin-Resistant Springer-Verlag: Berlin, 1986.
Enterococcus Faecium. Clin. Infect. Dis. 1999, 29, 361−366. (207) Terlain, B.; Thomas, J. P. Structure Of Griselimycin,
(185) https://www.drugbank.ca/drugs/DB00626. Polypeptide Antibiotic Extracted Streptomyces Cultures. I. Identi-

8126 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

fication Of The Products Liberated By Hydrolysis. Bull. Soc. Chim. Fr. (225) Fouche, M.; Schafer, M.; Blatter, M.; Berghausen, J.;
1971, 6, 2349−2356. Desrayaud, S.; Roth, H. J. Pharmacokinetic Studies Around The
(208) Kling, A.; Lukat, P.; Almeida, D. V.; Bauer, A.; Fontaine, E.; Mono- And Difunctionalization Of A Bioavailable Cyclic Decapeptide
Sordello, S.; Zaburannyi, N.; Herrmann, J.; Wenzel, S. C.; Konig, C.; Scaffold. ChemMedChem 2016, 11, 1060−1068.
et al. Antibiotics. Targeting Dnan For Tuberculosis Therapy Using (226) Mascio, C. T.; Mortin, L. I.; Howland, K. T.; Van Praagh, A.
Novel Griselimycins. Science 2015, 348, 1106−1112. D.; Zhang, S.; Arya, A.; Chuong, C. L.; Kang, C.; Li, T.; Silverman, J.
(209) Holzgrabe, U. New Griselimycins For Treatment Of A. In Vitro And In Vivo Characterization Of CB-183,315, A Novel
Tuberculosis. Chem. Biol. 2015, 22, 981−982. Lipopeptide Antibiotic For Treatment Of Clostridium Difficile.
(210) Nakajima, M.; Torikata, A.; Ichikawa, Y.; Katayama, T.; Antimicrob. Agents Chemother. 2012, 56, 5023−5030.
Shiraishi, A.; Haneishi, T.; Arai, M. Mycoplanecins, Novel Anti- (227) Snydman, D. R.; Jacobus, N. V.; Mcdermott, L. A. Activity Of
mycobacterial Antibiotics From Actinoplanes Awajinensis Subsp. A Novel Cyclic Lipopeptide, CB-183,315 Against Resistant Clostri-
Mycoplanecinus Subsp. Nov. II. Isolation, Physico-Chemical Charac- dium Difficile And Other Gram Positive Aerobic And Anaerobic
terization And Biological Activities Of Mycoplanecin A. J. Antibiot. Intestinal Pathogens. Antimicrob. Agents Chemother. 2012, 56, 3448−
1983, 36, 961−966. 3452.
(211) Haneishi, T.; Nakajima, M.; Shiraishi, A.; Katayama, T.; (228) Knight-Connoni, V.; Mascio, C.; Chesnel, L.; Silverman, J.
Torikata, A.; Kawahara, Y.; Kurihara, K.; Arai, M.; Arai, T.; Aoyagi, T.; Discovery And Development Of Surotomycin For The Treatment Of
et al. Antimycobacterial Activities In Vitro And In Vivo And Clostridium Difficile. J. Ind. Microbiol. Biotechnol. 2016, 43, 195−204.
Pharmacokinetics Of Dihydromycoplanecin A. Antimicrob. Agents (229) https://clinicaltrials.gov/ct2/show/NCT01597505.
Chemother. 1988, 32, 110−116. (230) Grevel, J.; Nuesch, E.; Abisch, E.; Kutz, K. Pharmacokinetics
(212) Cunningham, T. J.; Greenstein, J. I.; Loewenstern, J.; Of Oral Cyclosporin A (Sandimmun) In Healthy Subjects. Eur. J. Clin.
Degermentzidis, E.; Yao, L. H. Anti-Inflammatory Peptide Regulates Pharmacol. 1986, 31, 211−216.
The Supply Of Heat Shock Protein 70 Monomers: Implications For (231) Jin, M.; Shimada, T.; Shintani, M.; Yokogawa, K.; Nomura, M.;
Aging And Age-Related Disease. Rejuvenation Res. 2015, 18, 136−144. Miyamoto, K. Long-Term Levothyroxine Treatment Decreases The
(213) Mesfin, F. B.; Bennett, J. A.; Jacobson, H. I.; Zhu, S.; Andersen, Oral Bioavailability Of Cyclosporin A By Inducing P-Glycoprotein In
T. T. Alpha-Fetoprotein-Derived Antiestrotrophic Octapeptide. Small Intestine. Drug Metab. Pharmacokinet. 2005, 20, 324−330.
Biochim. Biophys. Acta, Mol. Basis Dis. 2000, 1501, 33−43. (232) Loosli, H.-R.; Kessler, H.; Oschkinat, H.; Weber, H.-P.;
(214) Defreest, L. A.; Mesfin, F. B.; Joseph, L.; Mcleod, D. J.; Petcher, T. J.; Widmer, A. Peptide Conformations. Part 31. The
Stallmer, A.; Reddy, S.; Balulad, S. S.; Jacobson, H. I.; Andersen, T. T.; Conformation Of Cyclosporin A In The Crystal And In Solution. Helv.
Bennett, J. A. Synthetic Peptide Derived From A-Fetoprotein Inhibits Chim. Acta 1985, 68, 682−704.
(233) Pflügl, G.; Kallen, J.; Schirmer, T.; Jansonius, J. N.; Zurini, M.
Growth Of Human Breast Cancer: Investigation Of The Pharmaco-
G.; Walkinshaw, M. D. X-Ray Structure Of A Decameric Cyclophilin-
phore And Synthesis Optimization. J. Pept. Res. 2004, 63, 409−419.
Cyclosporin Crystal Complex. Nature 1993, 361, 91−94.
(215) Bennett, J. A.; Defreest, L.; Anaka, I.; Saadati, H.; Balulad, S.;
(234) Mikol, V.; Kallen, J.; Pflugl, G.; Walkinshaw, M. D. X-Ray
Jacobson, H. I.; Andersen, T. T. Afpep: An Anti-Breast Cancer Peptide
Structure Of A Monomeric Cyclophilin A-Cyclosporin A Crystal
That Is Orally Active. Breast Cancer Res. Treat. 2006, 98, 133−141.
Complex At 2.1Å Resolution. J. Mol. Biol. 1993, 234, 1119−1130.
(216) Mesfin, F. B.; Andersen, T. T.; Jacobson, H. I.; Zhu, S.;
(235) Weber, C.; Wider, G.; Von Freyberg, B.; Traber, R.; Braun, W.;
Bennett, J. A. Development Of A Synthetic Cyclized Peptide Derived
Widmer, H.; Wuthrich, K. The NMR Structure Of Cyclosporin A
From Alpha-Fetoprotein That Prevents The Growth Of Human Breast Bound To Cyclophilin In Aqueous Solution. Biochemistry 1991, 30,
Cancer. J. Pept. Res. 2001, 58, 246−256. 6563−6574.
(217) Fehrenbach, T.; Cui, Y.; Faulstich, H.; Keppler, D. Character- (236) Fesik, S. W.; Gampe, R. T., Jr.; Eaton, H. L.; Gemmecker, G.;
ization Of The Transport Of The Bicyclic Peptide Phalloidin By Olejniczak, E. T.; Neri, P.; Holzman, T. F.; Egan, D. A.; Edalji, R.;
Human Hepatic Transport Proteins. Naunyn-Schmiedeberg's Arch. Simmer, R.; et al. NMR studies of [U-13C]cyclosporin A bound to
Pharmacol. 2003, 368, 415−420. cyclophilin: bound conformation and portions of cyclosporin involved
(218) Siemion, I. Z.; Pçdyczak, A.; Trojnar, J.; Zimecki, M.; in binding. Biochemistry 1991, 30, 6574−6583.
Wieczorek, Z. Immunosuppressive Activity Of Antamanide And (237) Cho, H.; Chung, Y. Water Soluble Cyclosporine Mono-
Some Of Its Analogues. Peptides 1992, 13, 1233−1237. methoxy Poly(Ethyleneglycol) Conjugates As Potential Prodrugs.
(219) Karle, I. L.; Wieland, T.; Schermer, D.; Ottenheym, H. C. J. Arch. Pharmacal Res. 2004, 27, 662−669.
Conformation Of Uncomplexed Natural Antamanide Crystallized (238) Rosenwirth, B.; Billich, A.; Datema, R.; Donatsch, P.;
From CH3CN/H20. Proc. Natl. Acad. Sci. U. S. A. 1979, 76, 1532− Hammerschmid, F.; Harrison, R.; Hiestand, P.; Jaksche, H.; Mayer,
1536. P.; Peichl, P.; et al. Inhibition Of Human Immunodeficiency Virus
(220) Di Blasio, B.; Rossi, F.; Benedetti, E.; Pavone, V.; Pedone, C.; Type 1 Replication By SNZ NIM 811, A Non-Immunosuppressive
Temussi, P. A.; Zanotti, G.; Tancredi, T. Bioactive Peptides: Solid- Cyclosporine Analog. Antimicrob. Agents Chemother. 1994, 38, 1763−
State And Solution Conformation Of Cyclolinopeptide A. J. Am. 1772.
Chem. Soc. 1989, 111, 9089−9098. (239) Hopkins, S.; Scorneaux, B.; Huang, Z.; Murray, M. G.; Wring,
(221) Quail, J. W.; Shen, J.; Reaney, M. J. T.; Sammynaiken, R. S.; Smitley, C.; Harris, R.; Erdmann, F.; Fischer, G.; Ribeill, Y. SCY-
Cyclolinopeptide A Methanol Solvate. Acta Crystallogr., Sect. E: Struct. 635, A Novel Nonimmunosuppressive Analog Of Cyclosporine That
Rep. Online 2009, 65, 1913−1914. Exhibits Potent Inhibition Of Hepatitis C Virus RNA Replication In
(222) Emmer, G.; Grassberger, M. A.; Meingassner, J. G.; Schulz, G.; Vitro. Antimicrob. Agents Chemother. 2010, 54, 660−672.
Schaude, M. Derivatives Of A Novel Cyclopeptolide. 1. Synthesis, (240) Ptak, R. G.; Gallay, P. A.; Jochmans, D.; Halestrap, A. P.;
Antifungal Activity, And Structure-Activity Relationships. J. Med. Ruegg, U. T.; Pallansch, L. A.; Bobardt, M. D.; De Béthune, M.-P.;
Chem. 1994, 37, 1908−1917. Neyts, J.; De Clercq, E.; et al. Inhibition Of Human Immunodeficiency
(223) Takahara, Y.; Takeuchi, Y.; Komura, I.; Hirose, Y.; Murao, S. Virus Type 1 Replication In Human Cells By DEBIO-025, A Novel
Isolation Of A New Peptide Antibiotic, Permetin A, From Bacillus Cyclophilin Binding Agent. Antimicrob. Agents Chemother. 2008, 52,
Circulans. J. Antibiot. 1979, 32, 115−120. 1302−1317.
(224) Fouche, M.; Schafer, M.; Berghausen, J.; Desrayaud, S.; Blatter, (241) Flisiak, R.; Horban, A.; Gallay, P.; Bobardt, M.; Selvarajah, S.;
M.; Piechon, P.; Dix, I.; Martin Garcia, A.; Roth, H. J. Design And Wiercinska-Drapalo, A.; Siwak, E.; Cielniak, I.; Higersberger, J.;
Development Of A Cyclic Decapeptide Scaffold With Suitable Kierkus, J.; et al. The Cyclophilin Inhibitor DEBIO-025 Shows Potent
Properties For Bioavailability And Oral Exposure. ChemMedChem Anti−Hepatitis C Effect In Patients Coinfected With Hepatitis C And
2016, 11, 1048−1059. Human Immunodeficiency Virus. Hepatology 2008, 47, 817−826.

8127 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128
Chemical Reviews Review

(242) Gallay, P.; Lin, K. Profile Of Alisporivir And Its Potential In characterization of known drug databases. J. Comb. Chem. 1999, 1, 55−
The Treatment Of Hepatitis C. Drug Des., Dev. Ther. 2013, 7, 105− 68.
115. (260) Stewart, W. E.; Siddall, T. H. Nuclear Magnetic Resonance
(243) Sugimoto, H.; Lebleu, V. S.; Bosukonda, D.; Keck, P.; Taduri, Studies Of Amides. Chem. Rev. 1970, 70, 517−551.
G.; Bechtel, W.; Okada, H.; William, C.; Bey, P.; Rusckowski, M.; et al. (261) Lu, J. J.; Crimin, K.; Goodwin, J. T.; Crivori, P.; Orrenius, C.;
Activin-Like Kinase 3 Is Important For Kidney Regeneration And Xing, L.; Tandler, P. J.; Vidmar, T. J.; Amore, B. M.; Wilson, A. G. E.;
Reversal Of Fibrosis. Nat. Med. 2012, 18, 396−404. Stouten, P. F. W.; Burton, P. S. Influence Of Molecular Flexibility And
(244) Rose, L.; Jenkins, A. T. A. The Effect Of The Ionophore Polar Surface Area Metrics On Oral Bioavailability In The Rat. J. Med.
Valinomycin On Biomimetic Solid Supported Lipid DPPTE/EPC Chem. 2004, 47, 6104−6107.
Membranes. Bioelectrochemistry 2007, 70, 387−393. (262) Tian, S.; Li, Y.; Wang, J.; Zhang, J.; Hou, T. ADME Evaluation
(245) Agata, N.; Ohta, M.; Mori, M.; Isobe, M. A Novel In Drug Discovery. 9. Prediction Of Oral Bioavailability In Humans
Dodecadepsipeptide, Cereulide, Is An Emetic Toxin Of Bacillus- Based On Molecular Properties And Structural Fingerprints. Mol.
Cereus. FEMS Microbiol. Lett. 1995, 129, 17−19. Pharmaceutics 2011, 8, 841−851.
(246) Amr, A. E.-G. E.; Abo-Ghalia, M. H.; Abdalah, M. M. Synthesis (263) Meanwell, N. E. Synopsis Of Some Recent Tactical Application
Of New (N-A-Dipicolinoyl)-Bis-L-Valyl-L-Phenylalanyl Linear And Of Bioisosteres In Drug Design. J. Med. Chem. 2011, 54, 2529−2591.
Macrocyclic Bridged Peptides As Anti-Inflammatory Agents. Arch.
Pharm. 2007, 340, 304−309.
(247) Busby, R. W.; Bryant, A. P.; Bartolini, W. P.; Cordero, E. A.;
Hannig, G.; Kessler, M. M.; Mahajan-Miklos, S.; Pierce, C. M.; Solinga,
R. M.; Sun, L. J.; et al. Linaclotide, Through Activation Of Guanylate
Cyclase C, Acts Locally In The Gastrointestinal Tract To Elicit
Enhanced Intestinal Secretion And Transit. Eur. J. Pharmacol. 2010,
649, 328−335.
(248) Blanchfield, J. T.; Gallagher, O. P.; Cros, C.; Lewis, R. J.;
Alewood, P. F.; Toth, I. Oral Absorption And In Vivo Biodistribution
Of [Alpha]-Conotoxin MII And A Lipidic Analogue. Biochem. Biophys.
Res. Commun. 2007, 361, 97−102.
(249) Clark, R. J.; Jensen, J.; Nevin, S. T.; Callaghan, B. P.; Adams, D.
J.; Craik, D. J. The Engineering Of An Orally Active Conotoxin For
The Treatment Of Neuropathic Pain. Angew. Chem., Int. Ed. 2010, 49,
6545−6548.
(250) McNulty, M. J.; Hutabarat, R. H.; Findlay, J. W.; Devereux, K.;
Knick, V. C.; Harvey, R. J.; Molina, L. Pharmacokinetics And Tissue
Distribution Of The Nonadecapeptide MOLI-1901 In Rats And Mice.
Xenobiotica 2003, 33, 197−210.
(251) Pinto, M. F. S.; Almeida, R. G.; Porto, W. F.; Fensterseifer, I.
C. M.; Lima, L. A.; Dias, S. C.; Franco, O. L. Cyclotides: From Gene
Structure To Promiscuous Multifunctionality. J. Evidence-Based
Complementary Altern. Med. 2012, 17, 40−53.
(252) Gran, L. On The Effect Of A Polypeptide Isolated From
″Kalata-Kalata″ (Oldenlandia Af f inis DC) On The Oestrogen
Dominated Uterus. Acta Pharmacol. Toxicol. 1973, 33, 400−408.
(253) Wong, C. T. T.; Rowlands, D. K.; Wong, C.-H.; Lo, T. W. C.;
Nguyen, G. K. T.; Li, H.-Y.; Tam, J. P. Orally Active Peptidic
Bradykinin B1 Receptor Antagonists Engineered From A Cyclotide
Scaffold For Inflammatory Pain Treatment. Angew. Chem., Int. Ed.
2012, 51, 5620−5624.
(254) Henriques, S. T.; Craik, D. J. Importance Of The Cell
Membrane On The Mechanism Of Action Of Cyclotides. ACS Chem.
Biol. 2012, 7, 626−636.
(255) Thell, K.; Hellinger, R.; Sahin, E.; Michenthaler, P.; Gold-
Binder, M.; Haider, T.; Kuttke, M.; Liutkeviciute, Z.; Goransson, U.;
Grundemann, C.; et al. Oral Activity Of A Nature-Derived Cyclic
Peptide For The Treatment Of Multiple Sclerosis. Proc. Natl. Acad. Sci.
U. S. A. 2016, 113, 3960−3965.
(256) QikProp, Schrödinger, LLC: New York, 2016; Schrödinger
Release 2016-1.
(257) Bird, G. H.; Madani, N.; Perry, A. F.; Princiotto, A. M.; Supko,
J. G.; He, X.; Gavathiotis, E.; Sodroski, J. G.; Walensky, L. D.
Hydrocarbon Double-Stapling Remedies The Proteolytic Instability Of
A Lengthy Peptide Therapeutic. Proc. Natl. Acad. Sci. U. S. A. 2010,
107, 14093−14098.
(258) Mannhold, R. I.; Poda, G. I.; Ostermann, C.; Tetko, I. V.
Calculation Of Molecular Lipophilicity: State-Of-The-Art And
Comparison Of Log P Methods On More Than 96,000 Compounds.
J. Pharm. Sci. 2009, 98, 861−893.
(259) Ghose, A. K.; Viswanadhan, V. N.; Wendoloski, J. J. A
knowledge-based approach in designing combinatorial or medicinal
chemistry libraries for drug discovery. 1. A qualitative and quantitative

8128 DOI: 10.1021/acs.chemrev.6b00838


Chem. Rev. 2017, 117, 8094−8128

You might also like