The Future of Peptide-based Drugs - Chem Biol Drug Des 2013; 81: 136-147

Page created by Terry Clarke
 
CONTINUE READING
The Future of Peptide-based Drugs - Chem Biol Drug Des 2013; 81: 136-147
Chem Biol Drug Des 2013; 81: 136–147
    Special Issue-Review
    Rational Design of Biologics and Peptides

The Future of Peptide-based Drugs
David J. Craik1,*, David P. Fairlie1, Spiros Liras2                of peptides in drug design, it is useful to briefly reflect on
and David Price2                                                   the past, where traditionally peptides were not considered
                                                                   as drug candidates. Table 1 summarizes some historical
1
  Division of Chemistry & Structural Biology, Institute for        trends in drug development technologies, targets and
Molecular Bioscience, The University of Queensland,                product classes, as well as providing a broad overview of
Brisbane, Qld 4072, Australia                                      the changing regulatory environment. We structure the dis-
2
  CVMED Medicinal Chemistry, Pfizer Inc., Worldwide                cussion in this section around the major eras of drug dis-
Medicinal Chemistry, 620 Memorial Drive, Cambridge, MA             covery (chemistry, biologics, and genomics) to illustrate
02139, USA                                                         how they might provide insights into future trends.
*Corresponding author: David J. Craik,
d.craik@imb.uq.edu.au                                              Most drug development in the 20th century can be classi-
                                                                   fied as being in the ’chemistry era’, where leads were gen-
    The suite of currently used drugs can be divided into          erated from small molecule natural products (1), either
    two categories – traditional ’small molecule’ drugs with       based on screening, or rational design processes that
    typical molecular weights of 5000 Da that are not orally bioavailable and need to          the treatment of a wide range of diseases, although only
    be delivered via injection. Due to their small size, con-      approximately 20 new chemical entities (NCEs) have been
    ventional small molecule drugs may suffer from
                                                                   registered each year since 1980 (Annual Reports in Medic-
    reduced target selectivity that often ultimately mani-
                                                                   inal Chemistry 1980–2011) and this number has remained
    fests in human side-effects, whereas protein therapeu-
    tics tend to be exquisitely specific for their targets due     fairly constant despite an increase in the number of drug
    to many more interactions with them, but this comes            targets discovered. Prospective analyses of the types of
    at a cost of low bioavailability, poor membrane perme-         compounds that were successful in the clinic led to broad
    ability, and metabolic instability. The time has now           generalizations that have subsequently been useful in
    come to reinvestigate new drug leads that fit between          guiding design of small molecular weight compounds. The
    these two molecular weight extremes, with the goal of          most widely known of these analyses, that is, the ’rule-of-
    combining advantages of small molecules (cost, con-            five’ (2) noted, among other criteria, a preference for a
    formational restriction, membrane permeability, meta-
                                                                   molecular weight of
The Future of Peptide-based Drugs - Chem Biol Drug Des 2013; 81: 136-147
Peptides in Drug Development

Table 1: Changing trends in drug design

Year              Technological era                  Molecular classes and ⁄ or approaches                   Regulatory environment

1960              Chemistry                          Natural products, screening, rational design            Activity paramount
                                                                                                                    ?
                                                                                                                    ?
                                                                                                                    ?
                                                                                                                    ?
                                                                                                                    ?
1980              Molecular biology                  Biologics (insulin, growth factors, EPO)                       ?
                                                                                                                    ?
2000              Genome & proteomics                New target identification and validation                       y
2020              Peptide drugs?                     Personalized medicine                                   Safety paramount
                  Plant factories?                   Increased specificity
                                                     Cheaper manufacture

drugs are generally referred to as ’biologics’ and include          secreted due to regulation at the protein level. However,
molecules such as insulin, growth factors, and engineered           the potential is clearly there to realize many new drug tar-
antibodies. These proteinaceous molecules disobey every             gets over the next decade or two. Because this review is
one of the rule-of-five parameters and, not surprisingly, are       future looking, we speculate that many of the new targets
generally not suitable for oral delivery. They typically            from sequencing efforts are likely to involve large protein–
require injection (via subcutaneous, intramuscular or intra-        protein interactions, featuring shallow interaction sites that
venous routes) or intranasal delivery. Nevertheless, biolo-         span large surface areas. Traditionally, this target class
gics have been an extremely successful class of                     has not been very tractable for small molecule drugs, but
therapeutics, both economically and in treating certain dis-        is potentially more suitable for peptides due to their larger
eases. Surprisingly, antibodies have been found to persist          surfaces areas and suitability for array technologies (3).
in vivo often for weeks following a single administration           Furthermore, along with genomics technologies, transcri-
and several antibodies and proteins are now in the ’block-          ptomics and proteomics technologies are developing rap-
buster’ category, including adalimumab ⁄ Humira Pen and             idly, becoming more heavily used and are likely to
etanercept ⁄ Enbrel, both for rheumatoid arthritis ($8b sales       contribute to more widespread delineation of protein–pro-
in 2011), infliximab ⁄ Remicade for arthritis and ritux-            tein interactions as targets in drug design. Thus, one pre-
imab ⁄ Rituxan for non-Hodgkin’s B-cell lymphoma ($7b               dicted outcome of these new technologies is a greater
sales in 2011), bevacizumab ⁄ Avastin for colorectal cancer         emphasis on proteins and peptides as prospective drugs.
and trastuzumab ⁄ Herceptin for breast cancer ($5–$6b
sales in 2011). There are other soluble proteins such as            Another predicted outcome of ’omics’ studies in humans
insulin glargine ⁄ Lantus ($4.8b, insulin receptor for diabetes     is a greater emphasis in the future on personalized medi-
type I and II), Neulasta ⁄ pegfilgrastim ($3.5b, G-CSF recep-       cines based on identification of an individual’s genetic
tor for myelosuppression), Epogen ($2.5b, erythropoietin            make-up. Interpatient variations in responses to drugs
receptor for renal anemia), and Avonex ($2.5b, interferon           have long been observed, but only recently have the
beta receptor for multiple sclerosis). Among top selling            genetic tools for understanding the mechanistic basis of
injectable peptides are the 10 amino acid immunomodula-             these variations become available, allowing therapy to
tor Copaxone ⁄ glatiramer acetate ($3b) for multiple sclero-        potentially be tailored accordingly. Again, peptides have
sis,    the     9 aa    gonadotropin       receptor    agonists     great potential here, as they are components of the very
Lupron ⁄ leuprorelin ($1.5b) and Zoladex ⁄ goserelin ($1.1b)        proteins that are differentially expressed between patients
for breast and prostate cancer, endometriosis and fibroids,         and result in the different interpatient responses to drugs.
the 8 aa inhibitor of secretion of growth hormone and               Peptides are also very amenable to site-specific modifica-
other hormones Sandostatin ⁄ octreotide ($1.3b) for acro-           tion that might be used to tailor therapeutics to individual
megaly and cancer, and the two amino acid proteasome                patients. Nevertheless, truly personalized medicine may be
inhibitor Velcade ⁄ bortezomib ($1.5b) for multiple myeloma.        a lot further away than currently mooted for many practical
                                                                    reasons.
Genome sequencing efforts that became possible at the
turn of the 21st century were originally touted as having           One of the drivers of personalized medicine will be the
the potential to lead to a boom in drug development via             need for a more targeted approach to therapy, both from
the identification of a vast range of new targets. Although         efficacy and safety perspectives. This trend toward
genome technologies have indeed led to massive amounts              demands for higher safety in therapeutic products is noted
of gene expression data, this has not yet translated into           in Table 1 and has been responsible for the much longer
large numbers of new validated targets, largely because             time frames now required from drug discovery to drug reg-
most contemporary drug targets are proteins and the field           istration, due to increasingly more stringent safety stan-
of proteomics is still in relative infancy. High expression of      dards imposed by regulatory authorities. This has altered
specific genes also often does not correlate with levels of         the drug development landscape, bringing into favor more
corresponding proteins, which may not be expressed or               biologics like antibodies that feature a common scaffold

Chem Biol Drug Des 2013; 81: 136–147                                                                                              137
Craik et al.

and that can realize higher earnings per unit volume. This
change in emphasis has been further catalyzed by some                                                    Peptides as Drugs
well-published failures among registered small molecule
drugs, including the withdrawal from the market of Vio-                                                  Some of the potential advantages and disadvantages of
xx for well-documented cardiovascular side-effects (4)                                                  peptides as drugs, compared to small molecule drugs, are
within a few years after its introduction for the treatment                                              summarized in Table 2. A primary reason for interest in
of inflammatory conditions. An important reason for the                                                  peptides and proteins is that they bind with exquisite
increasing market share of biologics has been their                                                      specificity to their in vivo targets, resulting in exceptionally
much higher target specificity, which is largely driven by                                               high potencies of action and relatively few off-target side-
their larger size than small molecule drugs. This enables                                                effects. This high degree of selectivity in their interactions
many more, and much stronger, interactions and thus                                                      is the product of millions of years of evolutionary selection
less promiscuity in drug targeting. Balanced against this                                                for complementary shapes and sizes from among a huge
major advantage are their disadvantages, such as their                                                   array of structural and functional diversity. Thus, peptides
lack of membrane permeability, poor oral bioavailability,                                                have been fine-tuned to interact specifically with biological
and generally lower metabolic stability than small mole-                                                 targets, evolving into potent endogenous hormones,
cule drugs.                                                                                              growth factors, neurotransmitters and signaling molecules,
                                                                                                         as well as immunologic and defense agents. In principle,
Following this introduction, we now turn to identifying gaps                                             peptides and proteins could have many valuable applica-
in current therapeutic approaches that need to be filled in                                              tions in medicine, but so far applications of chemically syn-
the future. The two main classes of successful drugs, that                                               thesized peptides have been severely limited by their low
is, small molecules (5000 Da)                                                   systemic stability, high clearance, poor membrane perme-
clearly are separated by a significant gap in molecular                                                  ability, negligible activity when administered orally, and
weight that has not, as yet, been serviced by the pharma-                                                their high costs of manufacture.
ceutical industry, as illustrated in Figure 1. Peptides offer
the potential of filling this gap and represent a class of                                               Despite these generic disadvantages of peptides (and
molecules that have the specificity and potency of larger                                                because of their advantages), more than 100 peptide-based
protein biologics, but are smaller in size and more accessi-                                             drugs have already reached the market. Our analysis of
ble and cheaper to manufacture using chemical methods,                                                   structural data for a well-documented subset of these (6)
thus potentially combining some of the advantages of pro-                                                shows that the majority of current peptide-based products
teins with those of small molecules. This article identifies a                                           are at the smaller end of the size spectrum (8–10 amino
few selected classes of peptides that could be used to fill                                              acids), although peptides up to 45 amino acids are currently
this gap, and in particular, we focus on the future potential                                            on the market (Figure 2). Almost invariably, the current pep-
of cyclic peptides in the size range 5–50 amino acids.                                                   tide therapeutics are delivered via injection. In addition to the
Readers are referred to recent reviews of peptides in drug                                               billion dollar peptide drugs listed above, some other exam-
design for coverage of the existing peptide pharmaceutical                                               ples of success stories include the following: oxytocin (8 aa,
market (5,6) and to focused reviews (7–15) on specific                                                   labor), calcitonin (32 aa, hypercalcemia, osteoporosis), teri-
subclasses of therapeutic peptides.                                                                      paratide (34 aa, parathyroid hormone analog, osteoporosis),
                                                                                                         Fuzeon (36 aa, enfuvirtide, antiretroviral), corticotropin-
                                                                                                         releasing hormone ⁄ factor (41 aa), and growth-hormone-
                 500                                                            5000
       Small                        12 aa
                                              MW (Da)
                                                                               50 aa      Biologics      releasing hormone ⁄ factor (44 aa, lipodystrophy).
                  5 aa                        20 aa                                       >5000 Da
      molecule
                         Mimics                                       Grafts             (eg Insulin,
       drugs
      $40 billion per year, or 10% of the ethical
                                                                                                         pharmaceutical market. This market share is growing
                                                                                                         much faster than that of other pharmaceuticals, and suc-
                                                                                                         cess rates for bringing biologics to market are now about
                                            OUTCOMES                                                     twice that of small molecule drugs. The paradigm shift in
                           of proteins but the stability and bioavailability
                                         of small molecules                                              interest by the pharmaceutical industry toward proteins is
                                                                                                         exemplified by recent successes of recombinant proteins
Figure 1: Schematic illustration of the molecular weight (MW) gap                                        (especially monoclonal antibodies) as blockbuster thera-
between conventional small molecule drugs (5000 Da). Peptides offer the potential to fill this gap if ways                                   predictions for how they might compete with small mole-
of overcoming their intrinsically unfavorable biopharmaceutical                                          cule drugs on the one hand, and biologics on the other,
properties can be developed. Two classes of peptides that are of
                                                                                                         as well as with the range of other new technologies that
particular interest are mimics of secondary structure elements of
proteins as these are often involved in target interactions, and                                         are described in other articles in this themed issue.
cyclic peptide scaffolds that can be used to accept a ’graft’ of a
bioactive peptide sequence, stabilizing it while retaining biological                                    Exenatide (Byetta) is one of the most recent peptides to
activity.                                                                                                reach the market and is used in the treatment of type 2 dia-

138                                                                                                                          Chem Biol Drug Des 2013; 81: 136–147
Peptides in Drug Development

Table 2: Advantages and disadvantages of peptides as drugs              One of the particular advantages of peptides from natural
                                                                        sources, relative to small molecules from natural sources,
Advantages                              Disadvantages
                                                                        is that peptides can be mined both directly (e.g., via phar-
High potency                            Poor metabolic stability        macological screening and extraction from venoms, for
High selectivity                        Poor membrane permeability      example), or detected indirectly from the encoded nucleic
Broad range of targets                  Poor oral bioavailability       acids. A good example of this powerful combination of
Potentially lower toxicity than         High production costs           approaches is the CONCO project (http://www.conco.org),
 small molecules                                                        designed to map the genome, transcriptome and prote-
Low accumulation in tissues             Rapid clearance
                                                                        ome of a venomous marine snail and potentially to exploit
High chemical and biological            Sometimes poor solubility
 diversity
                                                                        the outcomes for human health. One outcome from the
Discoverable at peptide and ⁄ or                                        project so far is a lead molecule, XEP-018, being devel-
 nucleic acid levels                                                    oped for pain control and local anesthesia. We will return
                                                                        to venoms with particular reference to disulfide-rich pep-
                                                                        tides later, but first focus on mining peptides from a variety
betes (16). It is interesting to note that this peptide was             of other natural sources, including bacteria, fungi and
derived originally from the saliva of a lizard, the Gila monster,       plants, with the mining followed by chemical value-adding
thus illustrating that natural sources remain a powerful                approaches to optimize biopharmaceutical properties.
source of peptide leads, just as they did for small molecule
drugs. This 39 amino acid peptide violates all rule-of-five
associated parameters (MW 4187, CLogP -21, HBD 58,                      Mine the Gap?
HBA 67, Rot bonds 135, tPSA 1780: MW = molecular
weight, HBD = hydrogen bond donors, HBA = hydrogen                      The 500–5000 Da gap shown in Figure 1 prompts the
bond acceptors, Rot bonds = rotatable bonds, tPSA = total               question of ’do such molecules exist in nature’ and if so
polar surface area), but can be made orally active (F = 4%,             why have they not been examined previously for their
mice) through biotinylation at Lys12 and Lys27 to enhance               pharmaceutical potential. Of course, the answer is that
plasma protein binding (17,18). In fact, to date nearly all             they do exist, and it has partly been the reluctance of the
orally active peptides have F = 0–5% oral bioavailability,              pharmaceutical industry to engage in peptide-based drug
mainly at the low end of this scale. Nevertheless, exenatide            development that has not seen this spectrum of molecular
has stimulated the development of several related mole-                 weight space more fully explored. But with the current
cules, just reaching the market or in clinical trials (16).             changing environment, driven by the technological and
                                                                        regulatory issues outlined in Table 1, the opportunity exists
We chose to feature exenatide as an example as it nicely                to mine natural sources for bioactive peptides as drug
illustrates what we believe will be a continuing trend of ani-          leads. In doing so, it is important to recognize the deficien-
mal venoms being a particularly rich source of peptide                  cies of peptides in terms of their biopharmaceutical prop-
leads. By their nature, venoms from predatory animals                   erties and perhaps design discovery approaches that will
must be potent and fast acting and require a degree of                  find molecules with intrinsically more favorable properties.
stability of their constituent peptides. These evolutionary
requirements have selected particular classes of peptides               An early, and perhaps the best known, example of a natu-
that make excellent staring points in drug design (19). To              ral peptide in this size range that has succeeded as a drug
illustrate this, Table 3 shows that there are currently six             with a reasonable degree of oral bioavailability is cyclospo-
marketed compounds that are derived from animal ven-                    rin A (CSA) (20). Originally discovered (mined) for its anti-
oms (if we include Gila monster saliva as pseudovenom).                 fungal properties, it has been used widely as an
Of these, ziconotide (Prialt) was the first peptide-based              immunosuppressant and contributed to revolutionizing
drug derived from a marine animal, but several others are               organ transplant therapy. This 11-residue peptide has
in preclinical development.                                             three key structural features that contribute to its favorable

Table 3: Examples of marketed peptide-based drugsa derived from animal venoms

Drug                               Size (aa)         Target disease                  Pharmacological mechanism            Year registered

Captopril (Capoten)                 3                Hypertension                    ACE inhibitor                        1982
Tirofiban (Aggrastat)               3                Anticoagulant                   Platelet inhibitor                   1998
Epifibatide (Integrilin)            7                Acute coronary syndrome,        Anticoagulant                        1998
                                                      unstable angina
Bivalirudinrub (Angiomax)          20                Unstable angina                 Anticoagulant                        2000
Ziconotide (Prialt)                26                Neuropathic pain                N-type Ca channel blocker            2004
Exenatide (Byetta)                 39                Type 2 diabetes                 GLP-1 receptor antagonist            2005
a
    Includes examples derived from venom peptides but not actually peptides themselves.

Chem Biol Drug Des 2013; 81: 136–147                                                                                                139
Craik et al.

biopharmaceutical properties, namely its cyclic backbone        using crystallographic and NMR techniques, there are a
that protects against proteolytic degradation and (along        number of ’slowly inter-converting’ conformers that are in
with its hydrophobic side chains) buries polar groups in        equilibrium in polar solvents such as DMSO, methanol or
the interior of the molecule, incorporation of seven N-         methanol ⁄ water mixtures.
methyl groups that reduce the number of amide hydrogen
bond donors, and four intramolecular hydrogen bonds that        A recent paper from the Lokey group (24) reported Com-
tie up the remaining four amide NH protons to reduce their      pound 1, which has a physicochemical profile that would
hydrogen bonding potential for solvation by water. Future       be viewed as lying outside of traditional oral drug space
mining of natural peptides will likely pay attention to         with a molecular weight of >750 Da and multiple hydrogen
searching for compounds with similar attributes.                bond donors and acceptors that upon initial inspection
                                                                would be expected to limit the ability of the compound to
A cyclic peptide backbone appears to be a particularly          cross the gut wall. Compound 1 was profiled in a range of
valuable attribute, but is a difficult one to mine for,         in vitro assays and progressed to an in vivo study, where
because MS-based proteomics studies are typically not           its pharmacokinetic profile in rats was characterized by
good at sequencing cyclic peptides without prior lineariza-     low clearance and a moderate ability to partition into tis-
tion. Furthermore, until recently, most natural cyclic pep-     sue, leading to a terminal elimination half-life of 2.8 h. The
tides were thought to be biosynthesized via non-ribosomal       absolute oral bioavailability (F) of 1 was determined to be
routes, as is the case for CSA, and this non-genetic route      ~28%, an impressively high value for a peptide (24). It was
does not lend itself to screening at the nucleic acid level,    the most bioavailable of a series of macrocycles designed
because no transcripts exist. However, over the last dec-       with specific N-methylation patterns to simultaneously
ade, increasing numbers of ribosomally synthesized cyclic       reduce the hydrogen bond donor count and to promote
peptides have been reported (21,22), thus opening up the        intramolecular hydrogen bonding networks of the remain-
possibility in future of more rapidly detecting precursors      ing protons and carbonyl lone pairs. These compounds
encoding cyclic peptides. Recent examples include a fam-        were prepared as pharmacokinetic probes and had no
ily of ribosomally biosynthesized cyclic peptides from          associated pharmacology described (24), but the study is
Caryophyllaceae plants that have the typical high stability     nevertheless a seminal paper from an absorption stand-
of cyclic peptides (23). As noted above, having two possi-      point. The ability to understand and predict the influence
ble approaches to mining is a definite advantage that pep-      of intramolecular hydrogen bonding networks on cell per-
tides have over small molecule drugs, and even difficult        meability is an exciting advance in the field.
(cyclic) peptides are now becoming amenable to two-
pronged mining approaches.                                      Similarly, important advances in the understanding of
                                                                membrane permeability have come from the Kessler group
Just as the value of traditional mined products, like baux-     (25), based on their earlier postulation that a combination
ite, iron ore or precious metals, is increased by subse-        of macrocyclization and N-methylation of a peptide may
quent manufacturing (value-adding) steps, significant value     be a general strategy to confer the combination of mem-
has been added to peptide-based drug design by recent           brane permeability and resistance to proteolytic degrada-
studies from the Lokey and Kessler groups (24,25) that          tion that is required to achieve oral bioavailability (28).
have examined the role of key structural features (e.g.,        Much of this thinking had also been influenced by the sys-
cyclization and N-methylation) on biopharmaceutical prop-       tematic evaluation of CSA. Interestingly, CSA and the
erties, as well as earlier detailed analyses (26) linking the   designed hexapeptide 1, as well as other related natural
effects of these and other conformational properties to         products, are structurally populated with lipophilic amino
three dimensional structures and pharmacological activi-
ties. These studies provide valuable information on fea-
tures that enhance stability, cell permeability, and oral
                                                                                 O
bioavailability. For example, investigation of CSA and clo-
                                                                                                  N
sely related analogs suggested that passive transcellular                             N
absorption is a characteristic of macrocycles that can                                H   O
                                                                             N
assume multiple, interconverting conformations with differ-                                   O
                                                                                  O O                 N
ent populations in low and high dielectric environments                                       H
(27). In the aqueous conditions of the gut, the conformer                                     N
                                                                                  N                                  OH
population will be different to the population in the low                                         O
dielectric environment of the membrane interior. Thus,
membrane permeability is driven by a dominant conforma-
tion where intramolecular, transannular hydrogen bonding
                                                                Compound 1: An N-methylated cyclic peptide containing multiple
masks the overall hydrogen bonding potential and reduces        hydrogen bond donors and acceptors, and a molecular weight
overall hydrophilicity, enabling CSA to cross the gut wall.     >750 Da. Compound 1 was determined to have a terminal elimi-
The conformational flexibility of CSA has intrigued many        nation half-life of 2.8 h and an absolute oral bioavailability of
laboratories, and in addition to the conformations identified   ~28% (24).

140                                                                                   Chem Biol Drug Des 2013; 81: 136–147
Peptides in Drug Development

acid side chains, and it is worth noting that naturally           a variety of experiments failed to demonstrate whether the
occurring, N-methylated cyclic peptides typically do not          improvement was due to a transcellular transport mode or
contain charged and polar side chains. This identifies a          an unexpected active transport mechanism. This research
possible design limitation in this space of peptidic macro-       clearly demonstrates the evolution and development of
cycles. Yet, this observation suggests that there may be a        robust in vitro transporter assays that need to take place
large unexplored opportunity for the design of peptidic           in macrocyclic drug space to understand which in vitro
macrocycles with unnatural amino acids, which will provide        systems can be used with greatest predictability of phar-
flexibility in testing a broad range of physicochemical prop-     macokinetics in preclinical species or man.
erties such as logD and pKa. Thus, within this broad strat-
egy, the specific tactics that promote oral bioavailability       Overall, these exemplar studies highlight the fact that the abil-
remain to be determined and it is clear that only a subset        ity of chemists to design and synthesize peptides to explicitly
of this chemical space may be relevant.                           explore structure-property and structure-activity relationships
                                                                  will be extremely valuable in developing the next generation
In other work from the Kessler group, a study on cyclic           of peptide-based drug leads. In this process, medicinal
N-methylated somatostatin analogs related to the Veber–           chemists will evolve to accommodate new skills and thinking.
Hirschmann peptide (Compound 2) generated a library of            A sophisticated understanding of the synthetic challenges
30 compounds with varying degrees of methylation of the           associated with peptides and peptide macrocycles, aided by
secondary amides contained in the starting macrocycle             innovative methods for amino acid functional manipulation
(29). Extensive in vitro evaluation showed that specific          and macrocyclization and enrichment of the pool of unnatural
methylation of D-Trp8, Lys9, and Ph11 gave rise to a large        amino acids synthons, will be essential. In addition, research
enhancement in membrane permeability in a Caco-2 cell             in this field will be clearly propelled by a deeper, physics-dri-
monolayer model, while other methylated derivatives gave          ven understanding of conformation and its impact on perme-
no enhancement. This compound also displayed reason-              ability, pharmacological activity, and overall disposition. A
able oral bioavailability in rat. The optimized peptide with      greater understanding of ADME properties in this space will
regard to cell permeability was also the peptide most effi-       be also essential. Physicochemical properties that affect per-
ciently cyclized using standard peptide coupling condi-           meability, clearance and presumably influence elimination
tions. The authors suggested that the linear precursor            routes are still not well understood. Similar to the small mole-
peptide exhibits a dynamic structure in solution while            cule world, the creation of in silico and statistical models that
undergoing some preorganization to bring the N and C              predict key ADME properties will be a major accelerator as
termini into close proximity to improve the efficiency of the     well. Examples of the types of considerations that might
cyclization reaction. The optimized macrocycle showed no          prove of further benefit in future medicinal ⁄ peptide chemistry
degradation after extensive incubation in rat serum and           optimizations can be extrapolated from considering a pivotal
was stable in simulated gut media. Progression of the lead        study by Veber et al. (30) who effectively extended Lipinski’s
analog to in vivo studies showed an oral bioavailability of       rule-of-five after finding that reduced molecular flexibility [£10
F = 9% and a volume of distribution (VD) of 3.7 L ⁄ kg, with      rotatable bonds (RotB), topological polar surface area
an elimination half-life of 74 min. The relatively high oral      (tPSA < 140 Å2), and £12 hydrogen bond donors + accep-
bioavailability, and in particular the ability to cross the gut   tors (HBD + HBA)] correlated with higher oral bioavailability
wall, was not fully explained and demonstrates the com-           independent of molecular weight (MW) in 1100 small mole-
plexity of the pharmacokinetic profile. The high VD sug-          cule drug candidates. Cell permeation increased with lower
gests that the compound can distribute effectively into           polar surface area and fewer rotatable bonds but, surpris-
tissues and move out of the plasma compartment. The               ingly, not always with higher lipophilicity (CLogP). It therefore
improvement in permeability was not fully understood, and         seems reasonable to propose that peptides might be made
                                                                  more orally bioavailable by: (i) conformationally constraining
                                                                  them to reduce molecular flexibility, (ii) reducing solvent
                         Phe7                                     exposed HBD ⁄ HBA atoms by replacing them or masking
                                                                  them with bulky groups, and (iii) forcing HBD ⁄ HBA atoms to
                                           Trp8                   hydrogen bond with one another. There has been no com-
                    O
         Pro6                   H                                 prehensive or systematic evaluation of peptides of MW 500–
                                N                                 5000 Da to identify upper limits for each parameter (MW,
                         N
                         H                                        RotB, HBA, HBD, PSA, ClogP) that determines cell perme-
                N            O O          N
                        O O         NH                            ability and oral bioavailability, and these studies are likely to
                                H         H
                                N                                 prove valuable feedback in future drug design efforts.
                         N                          NH2
        Phe11            H                Lys9
                                  O
                          HO
                           Thr10                                  Bridging the Gap

Compound 2: Veber–Hirschmann        peptide;   MW     806.9 Da;   The mining and structure-activity studies described in Sec-
CLogP 4.11.                                                       tion 3 are broadly applicable to a wide range of peptides

Chem Biol Drug Des 2013; 81: 136–147                                                                                           141
Craik et al.

but most emphasis to date has been on those smaller                    In the mimicry approach, beta strands have been identified
than 20 amino acids in size. This size range represents the            as important structural elements recognized by enzymes
majority (75%) of existing marketed peptides (Figure 2),               (e.g., proteases) and by other strands that form sheets
but in terms of future developments, one promising area                (e.g., amyloids) and have been successfully mimicked by
will be to bridge the size gap up to 50 amino acids using              both non-peptides and short constrained peptides. They
a variety of technologies, including mimicking elements of             often contain cyclic tripeptides, heterocyclic or other
secondary structures in proteins (helices, turns, strands ⁄ -          organic constraints inserted in a peptide sequence to
sheets) or grafting bioactive peptides onto stable scaffolds.          maintain the peptide backbone in a linear saw-toothed
Both of these latter approaches offer the potential to capi-           strand structure. The resulting constrained strand mimetics
talize on the largely untapped target space of protein–pro-            (32) and sheet mimetics (31,41) are usually maintained in
tein interactions, through conformational constraints that             this conformation when bound to proteins (42). Many have
reduce peptide flexibility, reduced peptide HBD ⁄ HBA                  shown highly potent and very selective enzyme inhibiting
atoms or reduced exposure to their solvation by water,                 or protein antagonizing properties, often leading to cell
and ⁄ or masking of HBD ⁄ HBA atoms by their hydrogen                  permeable and bioavailable compounds. Many are, how-
bonding with one another within a peptide.                             ever, usually small enough to match or only slightly exceed
                                                                       the guiding rule-of-five parameters.
The first approach is based on the finding that many pro-
tein–protein interactions involve recognition of key ele-              Helix mimics, in which a peptide is covalently constrained
ments of secondary structure, and these structural motifs              (43) to adopt an alpha or other helical structure, have
can be mimicked in small, carefully designed, peptides to              become possible by inserting (i) lactam (44,45) or hetero-
recapitulate protein-like biological activities. Examples              cycle (46) bridges between amino acid side chains, (ii) a
include mimetics of beta strands ⁄ sheets (31,32), beta,               hydrocarbon linker between amino acid side chains (47–
gamma and other turns (33), and alpha helices (34) (and                49), (iii) a carbon–carbon (50,51) or carbon–nitrogen (52)
references therein), and these are cartooned in Figure 3.              bond replacement for an intramolecular hydrogen bond at
The second approach is based on discoveries of minipro-                an end of a helix, or (iv) a metal ion clip (53,54). Downsiz-
tein scaffolds in nature that are particularly stable and              ing proteins to bioactive, helix-constrained peptides has
amenable to sequence insertions, or ’grafting’ of bioactive            afforded potent modulators of biological targets, including
peptides. Examples of these scaffolds include knottins                 transcription factors, oncoproteins like HDM2, viral fusion
(35,36) and cyclotides (7,37) from plants, lasso peptides              helix bundles, BCL apoptosis proteins, GPCRs like ORL-1
from bacteria (38) and a variety of animal toxins, but most            involved in pain transmission, Alzheimer’s notch protein,
notably the conotoxins (39,40) from marine cone snail ven-             antibacterial pheromones, and HIV-binding RNA.
oms. The goal of this approach is to introduce a bioactive
peptide sequence into the stable framework, with the aim               Turns are short or large loops, sometimes referred to as
of retaining the bioactivity of the target peptide, but                reverse turns, in proteins and peptides, which have a ten-
enhancing its stability and bioavailability by capturing some          dency to fold by forming intramolecular hydrogen bonds.
of the biopharmaceutical properties of the framework.                  Typically, such folding creates 10-membered (beta turns)
These two approaches are highly complementary, with the                or 7-membered (gamma turns) hydrogen-bonded ’cycles’
mimicry approach optimized for peptides of up to 20 aa                 within a peptide sequence. These are classified by phi and
and the scaffolding approach bridging the gap up to the                psi torsional angles as types I, I¢, II, II¢, III, III¢, IV–VI beta
50 aa entry point of conventional biologics.                           turns or classic and inverse gamma turns (55,56), although
                                                                       there are also other types of turns in peptides. Beta turns

                                                                                 Turn                  Helix
                                                                                                               Strand

Figure 2: Size distributions of peptide drugs on the market in
2010. The distribution reflects the sizes, in terms of number of
amino acids (aa) of 65 separate peptide products on the market
as reported by Vlieghe et al. (6). Note that, in some cases, differ-   Figure 3: Elements of protein secondary structure successfully
ent derivatives of the same peptide are counted as separate prod-      mimicked. The boxes indicate three common elements of second-
ucts if they are formulated differently, marketed by different         ary structure discussed in the text, including helices (blue), strands
companies, or used for different disease indications.                  (yellow), and turns (green).

142                                                                                         Chem Biol Drug Des 2013; 81: 136–147
Peptides in Drug Development

are most common and defined by four, gamma turns by               angiogenic agents for wound healing applications (68),
three, sequential residues in which the ends come into            anti-angiogenic agents with the aim of reducing blood ves-
close proximity. Biological activity of short peptides can be     sel growth in tumors (69), anti-infective agents against
enhanced by stabilizing such turns through cyclization            foot-and-mouth disease virus (70), and orally active pep-
and ⁄ or incorporation of heterocyclic or organic constraints.    tides against inflammatory pain (71). Although so far most
Resulting ’turn mimetics’ are designed either to preserve         attention has been directed toward extracellular targets,
turn-defining phi and psi angles in peptide components or         some of these frameworks offer the potential of delivering
to replace them altogether [reviewed in refs (33,57–60)].         bioactive epitopes to intracellular targets based on their
G-protein-coupled receptors have an especially striking           ability to penetrate cells (72,73). This attribute gives these
tendency to recognize turns of protein ⁄ peptide ligands,         peptides a distinct advantage over classic ’biologic’
and many agonists and antagonists have been developed             agents, which are typically limited to extracellular targets.
based on mimicking such turns (61). Because turns and
loops account for >30% of protein structure, it is not sur-       In some cases, disulfide-rich miniprotein scaffolds have
prising that peptide drugs have been developed around             intrinsic activity that is of pharmaceutical relevance, as in
this concept of mimicking turns, especially the more pre-         the case for example of MVIIA (ziconotide, Prialt) a 25
valent beta turns.                                                amino acid cystine knot peptide from cone snail venom that
                                                                  is used clinically for the treatment of neuropathic pain
By contrast with the protein surface mimic approach, the          (74,75). Ziconotide is administered intrathecally, but the
scaffold approach to peptide-based drug design (7,62) gen-        potential for oral delivery of conotoxin peptides was recently
erally involves larger peptides (20–50 aa) and two distinct       realized with the development of an engineered cyclic ver-
regions of the resultant peptide can be identified – the core     sion of another conotoxin, Vc1.1, that is orally active in a rat
scaffold and the inserted bioactive sequence, as illustrated      model of neuropathic pain (76,77). This perhaps represents
in Figure 4. Disulfide-rich scaffolds have proven to be partic-   a prophetic case for future developments where the lessons
ularly valuable and have applications not only as drugs, but      learned from naturally stable cyclic peptides such as the
as pharmacological probes (39) or imaging agents (63–66).         plant cyclotides can be used to re-engineer bioactive pep-
In keeping with the theme noted above of animal venoms            tides to enhance their stability and oral activity.
and plant toxins being a source of stable disulfide-rich scaf-
folds, we will focus mainly on this class here.
                                                                  Future Directions
In the first such example to produce a therapeutic lead,
Vita et al. (67) used a scorpion toxin scaffold to design a       One important area for the future is the need to consider
grafted molecule that inhibited the CD4-gp120 interaction         new sources of lead materials for drug development. Yeast
associated with HIV infection. In more recent studies, plant      cell surface display methods, ribosomal and other biomi-
cyclotides have been used as frameworks to develop                metic cyclic peptide synthesis methods all could be handy

                                                    A                              B                              C

Figure 4: Schematic illustration
of the miniprotein scaffold
approach to peptide-based drug
design. Panel A highlights poten-
tial sources of target epitopes,
from fragments of proteins, from
bioactive peptides, or from phage
display. Panel B schematically
illustrates a range of disulfide-rich
frameworks, including SFTI-1,
cyclotides and theta-defensins.
Panel C shows the bioactive
epitopes grafted into the stable
frameworks.

Chem Biol Drug Des 2013; 81: 136–147                                                                                         143
Craik et al.

in providing good starting points for hit discovery and opti-   Research Council (NHMRC) Senior Principal Research Fel-
mization.                                                       lowships to DJC (APP1026501) and DPF (APP1027369).

Some key technical hurdles to the development of effec-
tive peptide-based therapeutics will need to be addressed       Conflict of Interest
in the near future. First, the synthesis of small peptides
relies on expensive coupling reagents, resins and pro-          DJC and DPF are inventors on patents relating to peptides
tected amino acids, so cheaper methods for their synthe-        in drug development. SL and DP are employees of a phar-
sis and purification will need to be developed. This might      maceutical company.
be achieved by either chemical synthesis or molecular biol-
ogy techniques (recombinant peptide expression). Second,
modifications will need to be devised for enhancing mem-        References
brane permeability without compromising biologically
active peptide conformations; reducing peptide metabo-           1. Newman D.J., Cragg G.M. (2012) Natural products
lism by intestinal, plasma and cellular proteases, intestinal       as sources of new drugs over the 30 years from
and hepatic cytochrome P450 enzymes and P-glycopro-                 1981 to 2010. J Nat Prod;75:311–335.
teins; and reducing the high clearance rates of peptides.        2. Lipinski C.A. (2000) Drug-like properties and the
Third, as the number of peptides entering clinical trials           causes of poor solubility and poor permeability. J
continues to grow, additional methods will also be devel-           Pharmacol Toxicol Methods;44:235–249.
oped for optimizing their delivery and transport. Currently,     3. Katz C., Levy-Beladev L., Rotem-Bamberger S., Rito
peptides and small molecule drugs are being conjugated              T., Rudiger S.G., Friedler A. (2011) Studying protein-
to antibodies (to improve targeting), to carbohydrates (to          protein interactions using peptide arrays. Chem Soc
improve solubility, protect from degradation or conforma-           Rev;40:2131–2145.
tional rearrangements) to PEGs and lipids (to improve            4. Krumholz H.M., Ross J.S., Presler A.H., Egilman
uptake and permeability).                                           D.S. (2007) What have we learnt from Vioxx?
                                                                    BMJ;334:120–123.
The technical challenges further need to be considered in        5. Danho W., Swistok J., Khan W., Chu X.J., Cheung
light of the financial considerations that are driving a need       A., Fry D., Sun H., Kurylko G., Rumennik L., Cefalu
for a more flexible approach to discovery within the phar-          J., Cefalu G., Nunn P. (2009) Opportunities and chal-
maceutical industry. One recent trend to prime pharma-              lenges of developing peptide drugs in the pharmaceu-
ceutical company pipelines has been to partner an                   tical industry. Adv Exp Med Biol;611:467–469.
increasing proportion of discovery programs to academic          6. Vlieghe P., Lisowski V., Martinez J., Khrestchatisky
institutions in key areas of biology or chemistry where             M. (2010) Synthetic therapeutic peptides: science
leading academics are seen as bringing a competitive                and market. Drug Discov Today;15:40–56.
advantage with deep scientific expertise in novel concepts.      7. Craik D.J., Swedberg J.E., Mylne J.S., Cemazar M.
Examples where there is likely to be increased such part-           (2012) Cyclotides as a basis for drug design. Expert
nering include the emerging fields exploring synthetic biol-        Opin Drug Discov;7:179–194.
ogy and the gut microbiome.                                      8. Madala P.K., Tyndall J.D., Nall T., Fairlie D.P. (2010)
                                                                    Update 1 of: proteases universally recognize beta
Looking to the longer term, we speculate that alternative           strands in their active sites. Chem Rev;110:PR1–
methods of production and delivery of peptide-based                 PR31.
drugs might become more common. For example, the                 9. Loughlin W.A., Tyndall J.D., Glenn M.P., Hill T.A.,
recent discoveries of stable cyclic peptides in plants that         Fairlie D.P. (2010) Update 1 of: beta-strand mimetics.
are produced in high yield and have been shown to be                Chem Rev;110:PR32–PR69.
valuable scaffolds in peptide-based drug design suggest         10. Verdine G.L., Hilinski G.J. (2012) Stapled peptides
the possibility of using plants as production factories for         for intracellular drug targets. Methods Enzy-
high-value peptide-based drugs (78). Several exciting new           mol;503:3–33.
technologies, including sortase-mediated ligation, protein      11. Wittrup K.D., Verdine G.L. (2012) Protein engineering
splicing, and genetic code reprogramming also offer great           for therapeutics, part B. Preface Methods Enzy-
potential for the production of cyclic peptides (79).               mol;503:xiii–xiv.
                                                                12. Robinson J.A. (2011) Protein epitope mimetics as
                                                                    anti-infectives. Curr Opin Chem Biol;15:379–386.
Acknowledgments                                                 13. Bellmann-Sickert K., Beck-Sickinger A.G. (2010) Pep-
                                                                    tide drugs to target G protein-coupled receptors.
Work in our laboratories on peptide-based drug design is            Trends Pharmacol Sci;31:434–441.
funded by a grant from the Australian Research Council          14. Zhang L., Bulaj G. (2012) Converting peptides into drug
(ARC: LP110200213) and National Health and Medical                  leads by lipidation. Curr Med Chem;19:1602–1618.

144                                                                               Chem Biol Drug Des 2013; 81: 136–147
Peptides in Drug Development

15. Horne W.S. (2011) Peptide and peptoid foldamers in              tion: somatostatin analogues. Angew Chem Int Ed
    medicinal chemistry. Expert Opin Drug Dis-                      Engl;47:2595–2599.
    cov;6:1247–1262.                                            30. Veber D.F., Johnson S.R., Cheng H.Y., Smith B.R.,
16. Montanya E. (2012) A comparison of currently avail-             Ward K.W., Kopple K.D. (2002) Molecular properties
    able GLP-1 receptor agonists for the treatment of               that influence the oral bioavailability of drug candi-
    type 2 diabetes. Expert Opin Pharmacother;13:1451–              dates. J Med Chem;45:2615–2623.
    1467.                                                       31. Nowick J.S. (2008) Exploring beta-sheet structure
17. Chae S.Y., Jin C.H., Shin H.J., Youn Y.S., Lee S.,              and interactions with chemical model systems. Acc
    Lee K.C. (2008) Preparation, characterization, and              Chem Res;41:1319–1330.
    application of biotinylated and biotin-PEGylated gluca-     32. Loughlin W.A., Tyndall J.D., Glenn M.P., Fairlie D.P.
    gon-like peptide-1 analogues for enhanced oral deliv-           (2004) Beta-strand mimetics. Chem Rev;104:6085–
    ery. Bioconjug Chem;19:334–341.                                 6117.
18. Jin C.H., Chae S.Y., Son S., Kim T.H., Um K.A.,             33. Fairlie D.P., West M.L., Wong A.K. (1998) Towards
    Youn Y.S., Lee S., Lee K.C. (2009) A new orally                 protein surface mimetics. Curr Med Chem;5:29–62.
    available glucagon-like peptide-1 receptor agonist,         34. Shepherd N.E., Hoang H.N., Abbenante G., Fairlie
    biotinylated exendin-4, displays improved hypoglyce-            D.P. (2005) Single turn peptide alpha helices with
    mic       effects   in    db ⁄ db   mice.    J   Control        exceptional stability in water. J Am Chem
    Release;133:172–177.                                            Soc;127:2974–2983.
19. Halai R., Craik D.J. (2009) Conotoxins: natural prod-       35. Heitz A., Avrutina O., Le-Nguyen D., Diederichsen U.,
    uct drug leads. Nat Prod Rep;26:526–536.                        Hernandez J.F., Gracy J., Kolmar H., Chiche L.
20. Borel J.F. (2002) History of the discovery of cyclospo-         (2008) Knottin cyclization: impact on structure and
    rin and of its early pharmacological development.               dynamics. BMC Struct Biol;8:54.
    Wien Klin Wochenschr;114:433–437.                           36. Gracy J., Le-Nguyen D., Gelly J.C., Kaas Q., Heitz
21. Craik D.J. (2006) Chemistry. Seamless proteins tie              A., Chiche L. (2008) KNOTTIN: the knottin or inhibitor
    up their loose ends. Science;311:1563–1564.                     cystine knot scaffold in 2007. Nucleic Acids
22. Cascales L., Craik D.J. (2010) Naturally occurring cir-         Res;36:D314–D319.
    cular proteins: distribution, biosynthesis and evolution.   37. Gould A., Ji Y., Aboye T.L., Camarero J.A. (2011)
    Org Biomol Chem;8:5035–5047.                                    Cyclotides, a novel ultrastable polypeptide scaffold for
23. Condie J.A., Nowak G., Reed D.W., Balsevich J.J.,               drug discovery. Curr Pharm Des;17:4294–4307.
    Reaney M.J., Arnison P.G., Covello P.S. (2011) The          38. Knappe T.A., Manzenrieder F., Mas-Moruno C., Linne
    biosynthesis of Caryophyllaceae-like cyclic peptides            U., Sasse F., Kessler H., Xie X., Marahiel M.A.
    in Saponaria vaccaria L. from DNA-encoded precur-               (2011) Introducing lasso peptides as molecular scaf-
    sors. Plant J;67:682–690.                                       folds for drug design: engineering of an integrin
24. White T.R., Renzelman C.M., Rand A.C., Rezai T.,                antagonist. Angew Chem Int Ed Engl;50:8714–8717.
    McEwen C.M., Gelev V.M. et al. (2011) On-resin N-           39. Terlau H., Olivera B.M. (2004) Conus venoms: a rich
    methylation of cyclic peptides for discovery of orally          source of novel ion channel-targeted peptides. Phys-
    bioavailable scaffolds. Nat Chem Biol;7:810–817.                iol Rev;84:41–68.
25. Beck J.G., Chatterjee J., Laufer B., Kiran M.U., Frank      40. Adams D.J., Alewood P.F., Craik D.J., Drinkwater R.,
    A.O., Neubauer S., Ovadia O., Greenberg S., Gilon               Lewis R.J. (1999) Conotoxins and their potential phar-
    C., Hoffman A., Kessler H. (2012) Intestinal perme-             maceutical applications. Drug Dev Res;46:219–234.
    ability of cyclic peptides: common key backbone             41. Moriuchi T., Hirao T. (2004) Highly ordered structures
    motifs identified. J Am Chem Soc;134:12125–12133.               of peptides by using molecular scaffolds. Chem Soc
26. Fairlie D.P., Abbenante G., March D. (1995) Macro-              Rev;33:294–301.
    cyclic peptidomimetics: forcing peptides into bioactive     42. Tyndall J.D., Nall T., Fairlie D.P. (2005) Proteases
    conformations. Curr Med Chem;2:672–705.                         universally recognize beta strands in their active
27. Rezai T., Bock J.E., Zhou M.V., Kalyanaraman C.,                sites. Chem Rev;105:973–999.
    Lokey R.S., Jacobson M.P. (2006) Conformational             43. Henchey L.K., Jochim A.L., Arora P.S. (2008) Con-
    flexibility, internal hydrogen bonding, and passive             temporary strategies for the stabilization of peptides
    membrane permeability: successful in silico prediction          in the alpha-helical conformation. Curr Opin Chem
    of the relative permeabilities of cyclic peptides. J Am         Biol;12:692–697.
    Chem Soc;128:14073–14080.                                   44. Harrison R.S., Shepherd N.E., Hoang H.N., Ruiz-
28. Chatterjee J., Gilon C., Hoffman A., Kessler H. (2008)          Gómez G., Hill T.A., Driver R.W., Desai V.S., Young
    N-methylation of peptides: a new perspective in medic-          P.R., Abbenante G., Fairlie D.P. (2010) Downsizing
    inal chemistry. Acc Chem Res;41:1331–1342.                      human, bacterial, and viral proteins to short water-
29. Biron E., Chatterjee J., Ovadia O., Langenegger D.,             stable alpha helices that maintain biological potency.
    Brueggen J., Hoyer D., Schmid H.A., Jelinek R.,                 Proc Natl Acad Sci U S A;107:11686–11691.
    Gilon C., Hoffman A., Kessler H. (2008) Improving           45. Taylor J.W. (2002) The synthesis and study of side-
    oral bioavailability of peptides by multiple N-methyla-         chain lactam-bridged peptides. Biopolymers;66:49–75.

Chem Biol Drug Des 2013; 81: 136–147                                                                                    145
Craik et al.

46. Madden M.M., Muppidi A., Li Z., Li X., Chen J., Lin             coupled receptors recognize ligands with turn struc-
    Q. (2011) Synthesis of cell-permeable stapled peptide           ture. Chem Rev;105:793–826.
    dual inhibitors of the p53-Mdm2 ⁄ Mdmx interactions         62. Henriques S.T., Craik D.J. (2010) Cyclotides as tem-
    via photoinduced cycloaddition. Bioorg Med Chem                 plates in drug design. Drug Discov Today;15:57–64.
    Lett;21:1472–1475.                                          63. Akcan M., Stroud M.R., Hansen S.J., Clark R.J., Daly
47. Walensky L.D., Kung A.L., Escher I., Malia T.J., Bar-           N.L., Craik D.J., Olson J.M. (2011) Chemical re-engi-
    buto S., Wright R.D., Wagner G., Verdine G.L., Kors-            neering of chlorotoxin improves bioconjugation prop-
    meyer S.J. (2004) Activation of apoptosis in vivo by a          erties for tumor imaging and targeted therapy. J Med
    hydrocarbon-stapled BH3 helix. Science;305:1466–                Chem;54:782–787.
    1470.                                                       64. Kimura R.H., Miao Z., Cheng Z., Gambhir S.S.,
48. Schafmeister C.E., Po J., Verdine G.L. (2000) An All-           Cochran J.R. (2010) A dual-labeled knottin peptide
    hydrogen cross-linking system for enhancing the he-             for PET and near-infrared fluorescence imaging of in-
    licity and metabolic stability of peptides. J Am Chem           tegrin expression in living subjects. Bioconjug
    Soc;122:5891–5892.                                              Chem;21:436–444.
49. Kim Y.W., Grossmann T.N., Verdine G.L. (2011) Syn-          65. Miao Z., Ren G., Liu H., Kimura R.H., Jiang L.,
    thesis of all-hydrocarbon stapled alpha-helical pep-            Cochran J.R., Gambhir S.S., Cheng Z. (2009) An
    tides by ring-closing olefin metathesis. Nat                    engineered knottin peptide labeled with 18F for PET
    Protoc;6:761–771.                                               imaging      of   integrin     expression.    Bioconjug
50. Bullock B.N., Jochim A.L., Arora P.S. (2011) Assess-            Chem;20:2342–2347.
    ing helical protein interfaces for inhibitor design. J Am   66. Kimura R.H., Cheng Z., Gambhir S.S., Cochran J.R.
    Chem Soc;133:14220–14223.                                       (2009) Engineered knottin peptides: a new class of
51. Mahon A.B., Arora P.S. (2012) End-capped alpha-                 agents for imaging integrin expression in living sub-
    helices as modulators of protein function. Drug Dis-            jects. Cancer Res;69:2435–2442.
    cov Today Technol;9:e57–e62.                                67. Vita C., Drakopoulou E., Vizzavona J., Rochette S.,
52. Calvo J.C., Choconta K.C., Diaz D., Orozco O.,                  Martin L., Ménez A., Roumestand C., Yang Y.S., Yli-
    Bravo M.M., Espejo F., Salazar L.M., Guzman F.,                 sastigui L., Benjouad A., Gluckman J.C. (1999)
    Patarroyo M.E. (2003) An alpha helix conformational-            Rational engineering of a miniprotein that reproduces
    ly restricted peptide is recognized by cervical carci-          the core of the CD4 site interacting with HIV-1 enve-
    noma patients’ sera. J Med Chem;46:5389–5394.                   lope glycoprotein. Proc Natl Acad Sci U S
53. Kelso M.J., Hoang H.N., Appleton T.G., Fairlie D.P.             A;96:13091–13096.
    (2000) The first solution structure of a single alpha-      68. Chan L.Y., Gunasekera S., Henriques S.T., Worth
    helical turn. A pentapeptide alpha-helix stabilised by          N.F., Le S.J., Clark R.J., Campbell J.H., Craik D.J.,
    a metal clip. J Am Chem Soc;122:10488–10489.                    Daly N.L. (2011) Engineering pro-angiogenic peptides
54. Ma M.T., Hoang H.N., Scully C.C., Appleton T.G.,                using     stable,   disulfide-rich   cyclic    scaffolds.
    Fairlie D.P. (2009) Metal clips that induce unstruc-            Blood;118:6709–6717.
    tured pentapeptides to be alpha-helical in water. J         69. Gunasekera S., Foley F.M., Clark R.J., Sando L.,
    Am Chem Soc;131:4505–4512.                                      Fabri L.J., Craik D.J., Daly N.L. (2008) Engineering
55. Ball J.B., Andrews P.R., Alewood P.F., Hughes R.A.              stabilized vascular endothelial growth factor-A antag-
    (1990) A one-variable topographical descriptor for the          onists: synthesis, structural characterization, and bio-
    beta-turns of peptides and proteins. FEBS                       activity of grafted analogues of cyclotides. J Med
    Lett;273:15–18.                                                 Chem;51:7697–7704.
56. Hutchinson E.G., Thornton J.M. (1994) A revised set         70. Thongyoo P., Bonomelli C., Leatherbarrow R.J., Tate
    of potentials for beta-turn formation in proteins. Pro-         E.W. (2009) Potent inhibitors of beta-tryptase and
    tein Sci;3:2207–2216.                                           human leukocyte elastase based on the MCoTI-II
57. Moradi S., Soltani S., Ansari A.M., Sardari S. (2009)           scaffold. J Med Chem;52:6197–6200.
    Peptidomimetics and their applications in antifungal        71. Wong C.T., Rowlands D.K., Wong C.H., Lo T.W.,
    drug design. AntiInfect Agents Med Chem;8:327–344.              Nguyen G.K., Li H.Y., Tam J.P. (2012) Orally active
58. Marshall G.R. (1993) A hierarchical approach to pep-            peptidic bradykinin B1 receptor antagonists engi-
    tidomimetic design. Tetrahedron;49:3547–3558.                   neered from a cyclotide scaffold for inflammatory pain
59. Jones R.M., Boatman P.D., Semple G., Shin Y.J., Tamura          treatment. Angew Chem Int Ed Engl;51:5620–5624.
    S.Y. (2003) Clinically validated peptides as templates      72. Contreras J., Elnagar A.Y., Hamm-Alvarez S.F.,
    for de novo peptidomimetic drug design at G-protein-            Camarero J.A. (2011) Cellular uptake of cyclotide
    coupled receptors. Curr Opin Pharmacol;3:530–543.               MCoTI-I follows multiple endocytic pathways. J Con-
60. Kee K.S., Jois S.D. (2003) Design of beta-turn based            trol Release;155:134–143.
    therapeutic agents. Curr Pharm Des;9:1209–1224.             73. Cascales L., Henriques S.T., Kerr M.C., Huang Y.H.,
61. Tyndall J.D., Pfeiffer B., Abbenante G., Fairlie D.P.           Sweet M.J., Daly N.L., Craik D.J. (2011) Identifica-
    (2005) Over one hundred peptide-activated G protein-            tion and characterization of a new family of

146                                                                                Chem Biol Drug Des 2013; 81: 136–147
Peptides in Drug Development

    cell-penetrating peptides: cyclic cell-penetrating pep-       orally active conotoxin for the treatment of neuropath-
    tides. J Biol Chem;286:36932–36943.                           ic pain. Angew Chem Int Ed Engl;49:6545–6548.
74. Bowersox S., Mandema J., Tarczy-Hornoch K., Milja-        77. Carstens B.B., Clark R.J., Daly N.L., Harvey P.J.,
    nich G., Luther R.R. (1997) Pharmacokinetics of               Kaas Q., Craik D.J. (2011) Engineering of conotoxins
    SNX-111, a selective N-type calcium channel blocker,          for the treatment of pain. Curr Pharm Des;17:4242–
    in rats and cynomolgus monkeys. Drug Metab Dis-               4253.
    pos;25:379–383.                                           78. Craik D.J., Mylne J.S., Daly N.L. (2010) Cyclotides:
75. Miljanich G.P. (2004) Ziconotide: neuronal calcium            macrocyclic peptides with applications in drug design
    channel blocker for treating severe chronic pain. Curr        and agriculture. Cell Mol Life Sci;67:9–16.
    Med Chem;11:3029–3040.                                    79. Katoh T., Goto Y., Reza M.S., Suga H. (2011) Ribo-
76. Clark R.J., Jensen J., Nevin S.T., Callaghan B.P.,            somal synthesis of backbone macrocyclic peptides.
    Adams D.J., Craik D.J. (2010) The engineering of an           Chem Commun (Camb);47:9946–9958.

Chem Biol Drug Des 2013; 81: 136–147                                                                                 147
You can also read