You are on page 1of 56

Neuropeptides: Metabolism to Bioactive

Fragments and the Pharmacology


of Their Receptors

Mathias Hallberg
Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug
Dependence, Uppsala University, Biomedical Center, Uppsala, Sweden

Published online in Wiley Online Library (wileyonlinelibrary.com).


DOI 10.1002/med.21323

Abstract: The proteolytic processing of neuropeptides has an important regulatory function and the
peptide fragments resulting from the enzymatic degradation often exert essential physiological roles. The
proteolytic processing generates, not only biologically inactive fragments, but also bioactive fragments that
modulate or even counteract the response of their parent peptides. Frequently, these peptide fragments
interact with receptors that are not recognized by the parent peptides. This review discusses tachykinins,
opioid peptides, angiotensins, bradykinins, and neuropeptide Y that are present in the central nervous
system and their processing to bioactive degradation products. These well-known neuropeptide systems
have been selected since they provide illustrative examples that proteolytic degradation of parent peptides
can lead to bioactive metabolites with different biological activities as compared to their parent peptides.
For example, substance P, dynorphin A, angiotensin I and II, bradykinin, and neuropeptide Y are all
degraded to bioactive fragments with pharmacological profiles that differ considerably from those of
the parent peptides. The review discusses a selection of the large number of drug-like molecules that
act as agonists or antagonists at receptors of neuropeptides. It focuses in particular on the efforts to
identify selective drug-like agonists and antagonists mimicking the effects of the endogenous peptide
fragments formed. As exemplified in this review, many common neuropeptides are degraded to a variety
of smaller fragments but many of the fragments generated have not yet been examined in detail with
regard to their potential biological activities. Since these bioactive fragments contain a small number of
amino acid residues, they provide an ideal starting point for the development of drug-like substances
with ability to mimic the effects of the degradation products. Thus, these substances could provide a rich
source of new pharmaceuticals. However, as discussed herein relatively few examples have so far been
disclosed of successful attempts to create bioavailable, drug-like agonists or antagonists, starting from the
structure of endogenous peptide fragments and applying procedures relying on stepwise manipulations
and simplifications of the peptide structures. C 2014 Wiley Periodicals, Inc. Med. Res. Rev., 00, No. 0, 1–57, 2014

Correspondence to: Mathias Hallberg, Department of Pharmaceutical Biosciences, Division of Biological Re-
search on Drug Dependence, The Beijer Laboratory, Uppsala University, Biomedical Center, Box 591, 75124
Uppsala, Sweden. E-mail: Mathias.hallberg@farmbio.uu.se.

Medicinal Research Reviews, 00, No. 0, 1–57, 2014



C 2014 Wiley Periodicals, Inc.
2 r HALLBERG

Key words: proteolytic processing; neuropeptides; peptidemimetics; neuropeptide fragments; drug-like


molecules; substance P; opioids; dynorphin A; nociception; angiotensin II; bradykinin;
kallidin; neuropeptide Y

1. INTRODUCTION

Neuropeptides are derived from protein precursors (prepropeptides) that usually comprise
100–250 amino acid residues. These prepropeptides which are most often biologically inert are
formed at the ribosomes which are located at the endoplasmic reticulum of peptide-producing
neurons.1 A series of sequence-specific and tissue-specific proteolytic steps, frequently also
with other modifications, subsequently deliver smaller bioactive peptides. The neuropeptides
are defined as chains of amino acids of limited length released from neurons to exert an
effect on target cells. The physiological action of these neuropeptides is not terminated by
specific synaptic reuptake mechanisms, as often is the case for classic neurotransmittors, but
through degradation. This process is mediated by extracellular proteases anchored in the cell
membranes. Neuropeptides and their receptors are expressed not only in neurons but also in
other types of cells in the brain (such as glial cells) and various types of cells in the peripheral
tissues (such as endocrine cells). The degradation of neuroactive peptides by endopeptidases,
aminopeptidases, and carboxypeptidases (which may be more or less specific) can lead to the
formation of fragments that have similar or very different biological activities from those of the
parent peptide.2, 3
This review discusses tachykinins, opioid peptides, angiotensins, bradykinins, and neu-
ropeptide Y and their proteoloytic processing. These well-known neuropeptide systems have
been selected since they represent good examples that illustrate that proteolytic degradation
of parent peptides (e.g., substance P, angiotensin II, etc.) can lead to bioactive metabolites
with other biological profiles than their parent peptides. Such bioactive fragments and their
receptors can frequently be of considerable interest in a drug discovery context. All of these
neuropeptides are present in the central nervous system (CNS). Neuropeptides, and bioac-
tive fragments derived from neuropeptides are not in general suitable as drugs since they are
not bioavailable after oral administration and are not metabolically stable. Thus, the pharma-
cokinetic profiles of small peptides are far from optimal. Neuropeptides and their fragments,
however, can sometimes serve as starting points in design processes aimed at identifying drug-
like substances that interact with their receptors. New selective, metabolically stable drug-like
agonists (peptidemimetics) and antagonists that are able to cross the blood–brain barrier (BBB)
are constantly required.
A representative selection of drug-like molecules that act as agonists or antagonists at recep-
tors of the neuropeptides is discussed herein. The review focuses on the relatively few selective
drug-like ligands that have been identified and found to interact with the receptors/binding sites
of the bioactive degradation products formed from the parent neuropeptides subsequently. The
short degradation fragments have in some cases been successfully utilized and stepwise con-
verted into drug-like entities. The effects in peripheral tissues have attracted the most attention
in several cases.

2. NEUROPEPTIDES, THEIR FRAGMENTS AND PEPTIDEMIMETICS

A. Tachykinins
The most well-known members of the tachykinin family are substance P (SP), neurokinin A
(NKA), and neurokinin B (NKB). The biochemical effects of these substances are mediated by
Medicinal Research Reviews DOI 10.1002/med
METABOLISM TO BIOACTIVE FRAGMENTS r 3
the G-protein-coupled neurokinin receptor subtypes NK1, NK2, and NK3, which are primarily
activated by SP, NKA, and NKB, respectively.4 The neuropeptides, act as neurotransmittors
and neuromodulators and are widely distributed within both the CNS and peripheral tissues.
SP and NKA are the most abundant tachykinins in the brain. The NK1 and NK3 receptors
are expressed at high levels in the adult brain.5 Hemokinin and endokinins represent examples
of new putative tachykinins.6–8 Hemokinin has a similar biological effect to that of SP and is
an agonist primarily for the NK1 receptor subtype.7 The same C-terminal sequence, Phe-X-
Gly-Leu-Met-NH2, is present in SP, neurokinin A, neurokinin B, and hemokinin, where X
is Phe in SP and human hemokinin, Tyr in mouse hemokinin, and Val in neurokinin A and
neurokinin B.
The undecapeptide SP (Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2 ), discov-
ered as a neuropeptide by von Euler and Gaddum in 1931,9 is the most studied member of the
tachykinin family. It is characterized by neutral and lipophilic amino acid side chains in the
C-terminal part and two basic amino acid side chains in the N-terminal part.10 Its role in pain
transmission has been studied in greatest depth.11–13 SP and its NK1 receptor are found in such
areas of the brain as the amygdala, septum, hippocampus, hypothalamus, and periaqueductal
gray. These regions of the brain are associated with anxiety and depression.14–24 Furthermore,
the undecapeptide is considered to be involved in the mediation of emesis and SP is present
in the primary sensory afferent fibers and in the dorsal root ganglia. It is found also in the
dorsal horn of the spinal cord, and is a neuromodulator in the primary afferent fibers and
the unmyelinated C-fibers. Notably, it was reported 20 years ago that the undecapeptide does
not mediate pain through the NK1 receptor.25 SP and the other tachykinins are important not
only in various disorders of the brain and in pain processing, but also in the induction and
progression of several inflammatory responses.26–30 It is not surprising, therefore, that antag-
onists to the NK1, NK2, and NK3 receptors have the potential to treat diverse pathological
conditions. These include not only primary pain and CNS disorders such as depression, anx-
iety, schizophrenia, migraine, and emesis, but also disorders that are related to inflammatory
processes such as asthma, arthritis, psoriasis, and inflammatory bowel diseases.15, 31–33

1. Metabolism of Tachykinins
The SP peptide is formed from at least three distinct gene transcripts, the α-, β-, and
γ - preprotachykinin. The peptide is released from these precursors and its C-terminal residue
is subsequently modified by an amidating enzyme.34 The amidated C-terminal is important for
the stability of the peptide. Several different proteases process and degrade SP in the CNS and
CSF. This gives at least six lysine-containing metabolites SP(1–4), SP(1–6), SP(1–7), SP(1–9),
SP(2–11), and SP(3–11).35, 36 The SP(1–4), SP(1–7), SP(1–9), and SP(3–11) peptides have, for
example, been identified in the striatum37 and in the mouse spinal cord, while SP(1–6), SP(1–7),
and SP(1–9) are the major degradation products, together with phenylalanine and two frag-
ments that do not contain lysine, SP(8–9), SP(10–11).36 Angiotensin-converting enzyme (ACE),
neutral endopeptidase (NEP), and substance P endopeptidase (SPE) are all active in the en-
zymatic processing. Prolyl endopeptidase, which cleaves after prolines, cleaves the Pro4 -Gln5
peptide bond. Post proline dipeptidyl aminopeptidase subsequently removes the dipeptides
Arg1 -Pro2 and Lys3 -Pro4 from SP. The major cleaving sites of ACE are the Phe8 -Gly9 and
Gly9 -Leu10 bonds38 but ACE is also acting as a peptidyl dipeptidase, releasing dipeptides
from the remaining N-terminal fragment. Hence ACE can generate, for example, SP(1–7).38, 39
NEP primarily hydrolyzes SP at the Gln6 -Phe7 , Phe7 -Phe8 , and Gly9 -Leu10 bonds.38, 40 Thus,
SP is enzymatically degraded into several fragments, some of which retain their biological
activity.2, 3, 41, 42

Medicinal Research Reviews DOI 10.1002/med


4 r HALLBERG

Figure 1. Substance P (SP) is not selective but binds preferably to the NK1 receptor. Neurokinin A and B
bind mainly to the NK2 and NK3 receptors, respectively. While the C-terminal fragment of SP activates the NK1
receptor, the N-terminal heptapeptide metabolite substance P (1–7) binds to an unknown receptor, and has often
an opposing and very different biological profile from that of the parent peptide SP.

The C-terminal fragment SP(6–11) has anxiogenic effects after administration to the dorsal
periaqueductal gray in rats.23, 24 The effect is mediated through the NK1 receptor. In contrast,
the N-terminal fragment SP(1–7) did result in the opposite reaction,24, 43 Fig. 1 and Table I.
Notably, the latter effect was reported not to be mediated through the NK1 receptor.44 Thus,
the C-terminal part mimics the effect of the parent neuropeptide SP, while the N-terminal part
does not. The N-terminal heptapeptide SP(1–7) is the major metabolite of substance P in the
rat and has been studied in depth.42, 45, 46 SP can be metabolized to SP(1–7) by SPE, ACE as
well as NEP. SPE-like activity is present in the CSF45 in spinal cord tissue,47 and in various
other areas of the brain.48 Furthermore, the metabolic biotransformation of substance P in
liver microsomes from mouse, rat, and human was recently studied and the data compared.
Five major substance P metabolites, SP(3–11), SP(5–11), SP(6–11), SP(8–11), and SP(1–7) were
identified and quantified.49
Substance P (1–7) administered spinally attenuates thermal hyperalgesia in diabetic mice50
and has several effects that are opposite to those of SP. Thus, SP(1–7) has antinociceptive,51 anti-
inflammatory,52 antihyperalgesic,50 and anxiolytic effects.24, 43 It also attenuates several with-
drawal signs in morphine-dependent rodents53, 54 and the development of morphine tolerance.53
These effects are mediated through a specific receptor for SP(1–7)55–57 that is distinct from any
of the known opioid and tachykinin receptors. It is possible that SP(1–7) acts by interfering
with allosteric sites present on other neuropeptide receptors.
The SP(1–7) receptor has not been cloned nor studied in detail, but considerable efforts
have been devoted to making drug-like compounds with the same attractive pharmacological
profile as SP(1–7), and hence serve as SP(1–7) mimics. Structure-activity relationship studies,
involving an alanine-scan and truncations as well as C- and N-terminal modification of the

Medicinal Research Reviews DOI 10.1002/med


Table I. Primary Actions of the Neuropeptides and the Degradation Products that are Shown in Figs 1, 5, 12, 16, 19, and 22
Peptide Sequence Receptor target Activity type Biological activity
11–13
Substance P Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly- NK1-receptor Agonist Pain transmission, proinflammatory,26–30 nausea,66
Leu-Met-NH2 anxiogenic effects.23, 24
Substance P (1–7) Arg-Pro-Lys-Pro-Gln-Gln-Phe Receptor not determined – Antinociceptive,51 anti-inflammatory,52 antihyperalgesic,50
anxiolytic effects.24, 43
Substance P (6–11) Gln-Phe-Phe-Gly-Leu-Met-NH2 NK1-receptor Agonist Anxiogenic effects, see substance P.23, 24
Dynorphin A Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg- κ-opioid receptor Agonist Dysphoria, relevance in the brain reward system, where κ-opioid
Pro-Lys-Leu-Lys-Trp-Asp-Asn-Gln receptor agonists produce dysphoria.3, 99, 100 Anxiety-like
responses, sedation, analgesic effects.95–97, 101, 102
Dynorphin (1–8) Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile κ-Opioid receptor Agonist See dynorphin A.95–97
Dynorphin A (2–17) Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg-Pro- Receptor not determined – A nonopioid peptide, producing allodynia.103
Lys-Leu-Lys-Trp-Asp-Asn-Gln
Leu-enkephalin Tyr-Gly-Gly-Phe-Leu δ-Opioid receptor (and Agonist Euphoria, relevance in the brain reward system, where
μ-opioid receptor) stimulation of δ/μ-opioid receptors results in euphoria.3, 99, 100
The δ-opioid receptor agonists less powerful antinociceptive
activity, considered less addictive.142
Endomorphin-1 Tyr-Pro-Trp-Phe-NH2 μ-Opioid receptor Agonist μ-Opioid receptor agonists produce euphoria, respiratory
depression, constipation, sedation, dependence and are strong
analgesics.92
Endomorphin-2 Tyr-Pro-Phe-Phe-NH2 μ-Opioid receptor Agonist See endomorphin-1.92
Nociceptin Phe-Gly-Gly-Phe-Thr-Gly-Ala-Arg-Lys- Nociceptin opioid receptor Agonist Antinociception,175, 176 hyperalgesia.172
Ser-Ala-Arg-Lys-Leu-Ala-Asn-Gln
Nociceptin (1–7) Phe-Gly-Gly-Phe-Thr-Gly-Ala Receptor not determined – Antinociception, not hyperalgesia.172
Nociceptin (1–11) Phe-Gly-Gly-Phe-Thr-Gly-Ala-Arg-Lys- Receptor not determined – Antinociception, not hyperalgesia.172
Ser-Ala
Angiotensin I Asp-Arg-Val-Tyr-Ile-His-Pro-Phe-His- – – Essentially inactive.
Leu
Angiotensin II Asp-Arg-Val-Tyr-Ile-His-Pro-Phe AT1 and AT2 receptor Agonist AT1R: Vasoconstricion, cellular growth, proliferation.223 AT2R:
Vasodilation, apoptosis, antigrowth, and antiinflammatory
effects,219–221 neuroprotection,222 promotes neuronal cell
differentiation and nerve regeneration.216–218 Expressed in
disease conditions as heart failure, renal failure, myocardial
infarction, hypertension, and some brain disorders.215, 225–230
METABOLISM TO BIOACTIVE FRAGMENTS

Medicinal Research Reviews DOI 10.1002/med


r 5
6

Table I. Continued
Peptide Sequence Receptor target Activity type Biological activity
Angiotensin (1–7) Asp-Arg-Val-Tyr-Ile-His-Pro Mas receptor Vasodilation, cardioprotection, decreases hypertropy, and
r HALLBERG

fibrosis.281–283
Angiotensin III Arg-Val-Tyr-Ile-His-Pro-Phe AT1 and AT2 receptor Agonist AT1R and AT2R stimulation: Natriuretic effect, see angiotensin
II.285–287
Angiotensin IV Val-Tyr-Ile-His-Pro-Phe AT4 receptor/ Inhibitor Improves memory and learning,288, 291, 293, 297–299 protection
Insulin-regulated against ischemic stroke294, 295 and hyperglycemia.296
aminopeptidase (IRAP)
Kallidin Lys-Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe- Mainly B2 receptor Agonist The receptor constitutively expressed and agonists are
Arg hypotensive, potential cardioprotective, and
proinflammatory.358, 363
Bradykinin Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg Mainly B2 receptor Agonist See kallidin.358, 363

Medicinal Research Reviews DOI 10.1002/med


des-Arg10 -kallidin Lys-Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe Mainly B1 receptor Agonist Receptor inducible and poorly expressed under normal
physiological conditions.359, 378 Upregulated in certain
inflammatory states.362 Activation associated with pain385–388
and inflammation,361 hyperalgesia,382–384 diabetes and
diabetic complications.389
des-Arg9 -bradykinin Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe Mainly B1 receptor Agonist See des-Arg10 -kallidin.
Neuropeptide Y Tyr-Pro-Ser-Lys-Pro-Asp-Asn-Pro-Gly- NPY receptors, nonselective Agonist NPY Y1, Y2, Y4, Y5 receptor stimulation: Involved in body
Glu-Asp-Ala-Pro-Ala-Glu-Asp-Met- weight regulation (Y1 and Y5 receptor stimulation; increased
Ala-Arg-Tyr-Tyr-Ser-Ala-Leu-Arg- food intake and Y2 and Y4 receptor stimulation; appetite
His-Tyr-Ile-Asn-Leu-Ile-Thr-Arg-Gln- inhibition) and energy homeostasis,423, 430 associated with
Arg-Tyr-NH2 anxiety and depression,415, 431, 432 experience of pain,433, 434
alcohol dependence,435, 436 bone formation,437, 438 angina
pectoris,439 and cardiac stability.440
Neuropeptide Y (3–36) Ser-Lys-Pro-Asp-Asn-Pro-Gly-Glu-Asp- NPY receptor (Y2) Agonist Reduces appetite and food intake,428, 429 involved in energy
Ala-Pro-Ala-Glu-Asp-Met-Ala-Arg- homeostasis.430
Tyr-Tyr-Ser-Ala-Leu-Arg-His-Tyr-Ile-
Asn-Leu-Ile-Thr-Arg-Gln-Arg-Tyr-
NH2
METABOLISM TO BIOACTIVE FRAGMENTS r 7

Figure 2. Small ligands with high affinity to the substance P(1–7) binding site. The compounds were identified
after stepwise modifications and truncations of the heptapeptide SP(1–7).

heptapeptide led to improvements of affinity for the SP(1–7) binding sites58 and stronger effects
in behavioural tests than those of SP(1–7).59, 60 The C-terminal part of the heptapeptide was
found most essential for its binding and a primary amide rather than a carboxylic group in the
C-terminal was preferable. By systematic modifications, a series of high affinity binding ligands
were identified, exemplified by the amides 1 and 2,61, 62 Fig. 2. The amide 1 strongly attenuates
diabetic mechanical allodynia och thermal hyperalgesia in mice.63 About 60% of all diabetic
patients experience diabetic neuropathy and the pain of this condition is poorly relieved by
opiates.64 There is a great need for new strategies for the treatment of diabetic neuropathy, and
drug-like SP(1–7) mimetics that are active in the CNS may be a new therapeutic alternative.

2. Drug-like NK receptor antagonists and agonists


The first nonpeptide NK1 antagonist CP-96,345 (3) was reported in 1991,65 Fig. 3. Several
years later, another NK1 antagonist, aprepitant (4) from Merck was launched for prevention of
chemotherapy-induced nausea and vomiting (CINV).66 Notably, aprepitant was initially devel-
oped for pain, but was not more active than aspirin in human studies, thereafter for depression
and subsequently for CINV. Several NK1 receptor antagonists are now being developed67 and
some have been evaluated in phase II trials for treatment of depression with promising out-
comes, including aprepitant,68 L-759274,69 CP-122,721,70 and casopitant.71, 72 Subjects taking
these compounds experienced fewer symptoms of depression in these phase II studies, but a
phase III clinical study of aprepitant unfortunately did not confirm the positive data with this
compound.67 The NK1 receptor antagonist serlopitant has been evaluated in patients with over-
active bladder73 and topical aprepitant in clinical and experimental pruritis.74, 75 Furthermore,
it has been proposed that NK1 antagonist might find a role as anticancer drugs.76
The NK3 receptor may be a relevant target for the treatment of psychiatric disorders, in
particular schizophrenia. Clinical findings with selective NK3 antagonists, however, have not
been convincing.77 Clinical trials of osanetant (5) and talnetant (6)78 have given poor results and
no selective NK3 antagonists are currently being developed.79 Nonselective dual NK1/NK3
antagonists, such as Ro 4583298 (7), however, have recently attracted interest as potential agents
against CNS disorders such as schizophrenia and depression.80
The NK2 receptor has also been a target for treatment of CNS disorders. One NK2
receptor antagonist, saredutant (8), was tested in clinical trials with depression and anxiety as
indications, which led to its development being discontinued. Other NK2 antagonists, however,
are in phase II clinical trials as potential agents to treat various GI disorders such as IBS (e.g.,
ibodutant) and infant colic (e.g., nepadutant).81, 82 It has been claimed that triple tachykinin
antagonists, such as the compound 9, in Fig. 3,82 can be used to treat functional GI disorders
and specifically IBS and functional dyspepsia. Thus, chemical entities antagonizing all three
tachykinin receptors, the NK1, NK2, and the NK3 receptors, with approximately the same
affinity have been created. Although, SP binds to all three tachykinin receptors, it exhibits the
highest affinity at the NK1 receptor.

Medicinal Research Reviews DOI 10.1002/med


8 r HALLBERG

Figure 3. Examples of selective and nonselective NK1, NK2, and NK3 receptor antagonists. Aprepitant pre-
vents chemotherapy-induced nausea and vomiting. Selective and dual NK1/NK3 antagonists as well as NK2
antagonists have attracted considerable interest as potential agents against CNS disorders as depression and
schizophrenia. NK2 antagonists and triple antagonists, such as compound 9, could serve as useful pharmaceu-
ticals for the treatment of GI disorders. A fluorophenyl or chlorophenyl group attached to the central part of the
scaffold is often a characteristic feature of NK antagonists.

In 2004, the first selective and very potent nonpeptide NK1 receptor agonists were
disclosed.83 Among those, the quinoline amide 11, derived from a series of structurally re-
lated NK1 antagonists such as 10 that had been disclosed by Takeda,84 exhibited subpicomolar
affinity to the NK1 receptor, Fig. 4. Compound 11 comprises structural elements frequently rec-
ognized in the NK antagonists. It does not resemble the endogenous agonist, the undecapeptide
SP.

3. In summary
The degradation of the tachykinin substance P is an example of a metabolic process that
produces fragments with very different pharmacological profiles than the parent neuropeptide.
Drug-like molecules mimicking the effect of substance P, such as 11, and the substance P
degradation product, substance P (1–7), such as 2, have been developed. A large number of
selective and nonselective drug-like neurokinin receptor antagonists are known.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 9

Figure 4. The development of an NK1 receptor antagonist to the first series of drug-like NK1 receptor agonists
(2004). The two classes of compounds are structurally very similar.

B. Opioid peptides
1. Opioid neuropeptides
The opioid peptide family contains several members, which bind to three types of opioid
receptor: μ (MOP), δ (DOP), and κ (KOP). These G-protein-coupled receptors (GPCRs)
are well established by pharmacological and molecular biological methods.85 Subtypes of
the various types have been defined, where κ1, κ2, and κ3 are examples of subtypes of the
κ receptor, μ1 and μ2 are subtypes of the μ opioid receptor, and δ1 and δ2 are subtypes of the
δ opioid receptor. The alkaloid morphine, the archetypical nonpeptide opioid is not selective
and activates all three opioid receptors, but activates preferentially the μ receptor. In fact,
morphine and the related nonpeptide opioids were the only known drug-like agonists to pep-
tide receptors until 1995, when Perlman et al. disclosed the second example; the nonpeptide
angiotensin receptor agonists.86, 87 The opioids have been used for hundreds of years, and it is re-
markable that the first endogenous peptides that activate the opioid receptors, the enkephalins,
were not identified and reported until 1975.88
The enkephalins are pentapeptides with a tyrosine residue at their N-terminal (Tyr-Gly-Gly-
Phe-X; X = Leu or Met). More recently, tetrapeptides with opioid activity have been identified,
and have been given the name “endomorphins” (Tyr-Pro-X-Phe-NH2 ; X = Trp or Phe). These
have a C-terminal amide function.89 The enkephalin sequence resides in the N-terminal of most
opioid peptides, including β-endorphin, dynorphin A and B, and α-neoendorphin. The opioid
peptides β-endorphin, the dynorphins and the enkephalins are widely distributed in the brain,
whereas in the spinal cord dynorphins are mainly present in interneurons. Spinal enkephalins
are found primarily in long descending pathways from midbrain to the dorsal horn.90 The
opioid peptides have been attributed to a variety of behavioural processes, such as reward,
dependency, sedation, and stress response but their modulatory actions in pain processing
have probably attracted the greatest interest.91 It is worth noting that the opioid peptides are
produced also in nonneuronal cells, such as endocrine cells and cells of the immune system.
The enkephalins, activate mainly the δ-opioid receptors, while the dynorphins activate
mainly the κ opioid receptors. The selectivity of the dynorphins for the κ-opioid receptor is
mediated by the C-terminal, where Arg7 and Lys11 are important residues. The N-terminal of
the peptide accounts for the opioid activity. Regarding β-endorphin, this peptide seems less
selective than other members of the family and can produce a response through all three re-
ceptors. This response is somewhat stronger for the μ opioid and δ receptor than it is for the
κ opioid receptor. It is still not clear which peptide that is the endogenous ligand of the μ
receptor: the endomorphins label this site with high affinity and specificity and are considered
to be good candidates.89 Stimulation of the μ-opioid receptor leads to euphoria, respiratory
depression, constipation, sedation, dependence, and strong analgesic effects.92 All of the en-

Medicinal Research Reviews DOI 10.1002/med


10 r HALLBERG

Figure 5. The conversion of dynorphin A to Leu-enkephalin is a proteolytic process in which a κ agonist is


transformed into a δ opioid receptor agonist that also has an agonistic effect on the μ opioid receptor. This may
be important in the brain reward system, for example, where κ receptor activation produces dysphoria and δ/μ
opioid receptor activation euphoria. Dynorphin (2–17) is bioactive but acts via an unknown receptor.

dogenous opioid peptides have very similar N-terminals that act as the “message” part of the
neuropeptides, while their C-terminal act as the “address” part and account for the rather
moderate receptor selectivity observed. This property and their low capacity to cross BBB, and
the fact that the endogenous opioid peptides are readily degraded in vivo by various peptidases
make these neuropeptides not useful in clinic to treat pain.93, 94

2. Metabolism of opioid neuropeptides


The endogenous opioid neuropeptides, the dynorphins, the enkephalins, and β-endorphin
are derived from the three genetically different precursors, prodynorphin, proenkephalin, and
proopiomelanocortin. The opioid peptides are often converted by proteolysis into fragments
with retained or modified biological activity. The degradation of dynorphin A(1–17), derived
from preprodynorphin, into dynorphin A(1–8) in vivo provides one example,95 Fig. 5 and
Table I. Both the parent peptide and its metabolite are agonists of the opioid receptors with
selectivity for the κ receptor type.96, 97 The conversion of dynorphin to Leu-enkephalin is a
second example. The latter peptide can be released from dynorphin A, dynorphin B, and
α-neoendorphin. Members of the proprotein convertase family (PC1/3 and PC2) are the major
endopeptidases and carboxypeptidase E (CPE) the major exopeptidase involved in producing
the various dynorphins and in processing these to encephalin.98 Cleavage by both PC1/3 and
PC2 creates Leu-enkephalin-Arg6 -Arg7 and this intermediate is subsequently transformed to
Leu-enkephalin by CPE.
The release of Leu-enkephalin from dynorphin, reflects a conversion of a κ agonist to a
product acting on δ opioid receptors and (less pronounced) through μ opioid receptors. It is
suggested that in brain areas where the κ opioid receptor signals oppose those of δ and μ
opioid receptors, this type of conversion may be of relevance, for example, in the brain reward

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 11

Figure 6. Examples of a selective κ opioid receptor agonist, a selective δ opioid receptor agonist, and a
selective μ opioid receptor agonist. These opioids are often used as research tools—all are amines but they
have very different structures. Nalfurafine is used as a nonaddictive antipruritic drug. Fentanyl and its more
recently developed congeners have largely replaced morphine for anesthesia.

system, where κ agonists produce dysphoria, while stimulation of δ/μ opioid receptors results
in euphoria.3, 99, 100
The enzymatic removal of the N-terminal tyrosine residue from dynorphin A, that induces
anxiety-like responses, sedation, and analgesic effects,101, 102 yields a peptide with 16 amino acid
residues that is bioactive but that does not have the opioid effect of the parent peptide.103 Thus,
the deletion of single residue can have a dramatic effect on the biological response. This frag-
ment was reported to interact with specific sites located on the N-methyl-D-aspartate receptor.
The biotransformation of dynorphin A in striatum of freely moving rats after direct infusion
of the peptide has been investigated. Microdialysis-mass spectrometry technique revealed that
proteolysis of, for example, the Arg7 -Ile8 bond to provide dynorphin A(1–7) and dynorphin
A(8–17) occurred.104 Several different enzymes are involved in the enzymatic processing of the
opioid peptides and as an example dynorphin A is a substrate a) for PC2 to cleave Arg9 -Pro10
where after carboxypeptidase E cleaves the Ile8 -Arg9 bond,105 b) for a thiol-dependent dynor-
phin A converting enzyme with Arg6 -Arg7 as major cleavage site,106 and c) for a thiol-sensitive
metalloprotease, called dynorphin A-17 processing enzyme with Ile8 -Arg9 as the cleavage site.107
Hence, at least three different proteases are engaged in the selective cleavage of peptide bonds
in the central part of dynorphin A.
Inhibition of neprilysin and aminopeptidase N, that are involved in the degradation of
the enkephalins, increases the levels of the neuropeptides and induces analgesic effects in
various animal models of inflammatory and neurophatic pain. Notably, dual enkephalinase
inhibitors and fatty acid hydrolase inhibitors, the latter preventing degradation of endogenous
cannabinols, are in clinical trials.108

3. Drug-like opioid receptor agonists and antagonists


Many drug-like molecules can activate receptors in the opioid system and mimic the effects
of parent opioid peptides and their degradation fragments. Examples of three opioid receptor
selective agonists; a selective κ opioid receptor agonist (12), a selective δ opioid receptor agonist
(13), and a selective μ opioid receptor agonist (14) are presented in Fig. 6. Morphine (15) and
related analogues, including prodrugs such as heroin are the classical examples of opioids, Fig. 7.
Morphine, the most abundant alkaloid in opium was isolated 1804 and is used to treat moderate-
to-severe pain. Activation of the μ opioid receptor gives the strongest analgesia, and this is the
Medicinal Research Reviews DOI 10.1002/med
12 r HALLBERG

Figure 7. Structural simplification of morphine produces the opioid receptor agonist pethidine, which comprises
the 4-phenylpiperidine unit of morphine. The N-terminal part of Leu-enkephalin, essential for its activity, is shown
for comparison.

receptor to which morphine and morphine analogues preferentially bind. These drugs, however,
are also associated with a series of side effects including sedation, constipation, and respiratory
depression. The drugs create euphoria and opioid dependence and withdrawal syndrome can
develop. Thus, μ opioid receptor agonists as morphine provide the best analgesics but also most
side effects. As a consequence, considerable efforts have been devoted to the development of new
better selective chemical entities that do not have undesired side effects or that have fewer. The
biological response to a special opioid is a function of to which receptor or receptor subtypes
it binds, the affinity to the various receptors and the pharmacokinetics and metabolism of the
drug. The in vivo duration of drug action depends not only on macroscopic pharmacokinetic
properties such as plasma half-life and the time needed to equilibrate between the plasma and
the effect compartments, but also on long-lasting target binding and rebinding.109–111
Pethidine (16) is a structurally simplified morphine analogue encompassing the character-
istic pharmacophore scaffold of the morphine opioids, Fig. 7. It was synthesized in 1933 and
was the first synthetic opioid to be made. The morphine opioids can be anticipated to bind to
the receptor sites that are occupied of the N-terminal tyrosine or tyrosine-glycine residues of
the endogenous peptides, for example, 17. It is somewhat remarkable that no drug-like low-
molecular-weight opioid agonist seems to have been developed by stepwise modifications of an
endogenous enkephalin. In contrast, hundreds of bioactive analogues, such as pethidine, have
been prepared derived from morphine as structural template. In fact, the majority of the opioid
analgesics used in clinical practise are μ opioid receptor agonists derived from morphine.112, 113
Following the discovery of pethidine and after the development of a large number of other
related analogues, fentanyl (14) was synthesized by Paul Jansen in 1960. Notably, fentanyl that
is lacking the Ar-C-C-C-N element found in morphine/pethidine analogues is 100 times more
potent than morphine. It is used in clinic and is a good example of powerful and selective
μ opioid receptor agonist.
To make peptidemimetics starting from endogenous peptides is a tremendous challenge.
It is possible that the detailed structural information obtained from the recently reported
high-resolution crystal structure of the μ opioid receptor bound to a morphinan antagonist
(revealing a large solvent-exposed pocket into which the morphinan ligand binds deeply)114 will
improve the understanding of the essential binding interactions and aid the discovery of better
selective opioid drugs.
Considerable efforts have been directed toward developing opioids that do not interfere
with the μ opioid receptor, due to the side effects, such as addiction, that are associated
with this receptor. Activation of the κ opioid receptor results in a weaker analgesic response
and in addition, receptor activation is associated with miosis, sedation, psychotomimesis, and
dysphoria. Nevertheless, several κ selective compounds have now been disclosed, for example,
U-50,488H (18), CI-977,115, 116 HZ2117 and salvinorin A isolated from Salvia divinorum. The
Medicinal Research Reviews DOI 10.1002/med
METABOLISM TO BIOACTIVE FRAGMENTS r 13

Figure 8. The arylacetamides 18 and 19 are subtype selective and serve as selective κ1 agonists, while
pentazocine (20) and bremazocine (21) are κ2 receptor selective. The latter two compounds include the ben-
zomorphane skeleton found in nalfurafine (Fig. 6), but nalfurafine is κ3 receptor selective.

widely abused hallucinogen salvinorin A has a very different chemical structure from those of
other opioids, in that it does not contain nitrogen.118, 119 In fact, it was the first nonnitrogenous
opioid agonist reported. Nalfurafine (TRK-820) (12), with the characteristic Ar-C-C-C-N
scaffold and encompassing a tyrosine element provides a good example of κ agonist that is
very selective, Fig. 6. Nalfurafine is used as an antipruritic drug.120–122 It has been claimed that
this naltrexone-like derivative is the first opioid that does not cause addiction.123 Hence, while
nalfurafine neither exhibits aversive nor additive effects,124 many arylacetamide derivatives such
as U-50488H (18) and U-69,593 (19) cause psychomimetic and aversive reactions.125, 126 These
differences in psychological effects have been postulated to be attributed to affinity preferences
versus various κ receptor subtypes, where arylacetamide derivatives prefer the κ1 receptor
subtype127, 128 and nalfurafine the κ3 receptor subtype.129–133 The benzomorphans pentazocine
(20) and bremazocine (21) are proposed to show high affinity to the κ2 subtype of the κ opioid
receptor,128, 134 Fig. 8. Thus, very structurally diverse compounds activate the three subtypes of
the κ opioid receptor. The recently reported crystal structure of the human κ opioid receptor
in complex with a κ opioid receptor selective antagonist,135 in combination with modeling
of selective agonists related to salvinorin A as well as morphinan-derived antagonists, have
provided essential insights on ligand-receptor interactions useful in future design of even more
selective κ opioid receptor ligands. A large binding cavity with several potential anchoring
points to ligands was revealed, which can explain the acceptance of broad structural diversity
of the κ opioid receptor ligands.135 Furthermore, access to 3D structures of selective ligands to
the subtypes of the κ receptor will be very helpful in future discovery processes.136
Many selective agonists, not only to the μ and κ opioid receptors but also to the δ re-
ceptor, have recently been reported.137–141 The δ opioid receptor agonists have less powerful
antinociceptive activity but have a potential for use as analgesics since they are associated with
relatively few severe side effects and are anticipated in general to be less addictive.142 Among
such agonists, SNC 80 (13) is highly selective for the δ receptor and is 2000-fold selective over the
μ opioid receptor,137, 139 Fig. 6. Another selective δ agonist, TAN-67 (23)143, 144 is structurally

Medicinal Research Reviews DOI 10.1002/med


14 r HALLBERG

Figure 9. From a selective δ opioid receptor antagonist to a subtype selective δ1 opioid receptor agonist.

Figure 10. Opioid receptor antagonists used after overdoses of opioids and to treat alcohol dependence. These
compounds, which are often used as research tools, are characterized by the benzomorphane skeleton, the
vinyl, or cyclopropyl group linked to the basic nitrogen and the tertiary hydroxyl group. These elements are also
present in the δ opioid receptor antagonist naltrindole (Fig. 9).

very different. It has a rotable bond between the two ring systems and contains a quinoline nu-
cleus. TAN-67 has been derived from the rigid and selective δ antagonist, naltrindole (22),141, 145
Fig. 9. The “message (efficacy) – address (selectivity)” concept introduced by Portoghese has
been useful in the design process of opioid agonists and antagonists of this class.146 The crystal
structure of the δ receptor from mouse bound to the subtype-selective antagonist has recently
been reported.147 The lower part of the binding pocket, which is divided into two distinct
regions is highly conserved among the opioid receptors. The upper part, in contrast, contains
divergent residues and confers subtype selectivity. Thus, a structural rationale for the useful
“message-address” concept has been obtained.147
Naloxone (24), often referred to as an opioid antagonist is an inverse agonist and is used
in the clinic to counter the effects overdoses of opioids. In addition, naloxone is added to
the partial opioid agonist buprenorphine, in a mixture called Suboxone, to prevent abuse of
buprenorphine, which is probably the major current use of naloxone. Naltrexone (25) is mainly
used to treat alcohol dependence,148 Fig. 10. In addition, it is added to buprenorphin (in the
mixture called suboxone) to prevent abuse of buprenorphin. This is probably the major current
use of naltrexone.
The compounds described in this section so far activate receptors of opioid peptides
and opioid peptide fragments and are drug-like. In addition, there are a large number of
compounds disclosed in the literature that retain the peptidic character of the endogenous
ligands. These pseudopeptides have served and serve as important research tools and have
improved the understanding of potential bioactive conformations and binding modes of the
opioid peptides to their receptors. Various modifications such as introduction of D-amino
acids, of β-amino acids, of peptide bond bioisosters, and of conformational constraints by

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 15

Figure 11. The ring size of the macrocycles is critical for opioid receptor selectivity.

cyclizations, for example, have been applied. By these procedures, very potent and selective
agonists were provided, exemplified here with the selective μ opioid receptor agonist JOM-6
(26) and the structurally very similar selective δ opioid receptor agonist JOM-13 (27), Fig. 11.
The C-terminal carboxylate has been replaced with a carboxamide in JOM-6, and this favors
μ binding over δ opioid receptor binding.149
The tyrosine residue constitutes the pharmacophore associated with binding and
activation,150–152 while the position of the aromatic ring of phenylalanine is proposed to be
responsible for the observed subtype receptor selectivity149, 153 (for a review, see154 ).

4. Nociceptin
Nociceptin155 also known as “orphanin FQ”,156 is not a classical opioid but is presented here
due to its similarities with the opioid peptides. This neuropeptide of length 17 amino acid
residues includes the sequence Phe-Gly-Gly-Phe in the N-terminal part rather than the Tyr-
Gly-Gly-Phe present in the enkephalins and dynorphins. Thus, a hydroxyl group is the difference
at the N-terminal end. The nociceptin opioid receptor (NOP) sometimes referred to as opiate
receptor-like 1, ORL-1, LC132, OP4 , or NOP1 was cloned in 1994 and identified using a human
cDNA library on the basis of high homology with the classic μ, κ, and δ opioid receptors.157, 158
This GPCR remained an orphan receptor for a year after it was cloned, until the endogenous
neuropeptide agonist nociceptin or orphanin FQ (N/OFQ) was isolated from brain extracts.
The neuropeptide was found to bind with high affinity to the NOP site, but not to the μ, κ,
or δ opioid receptors.155, 156 Nociceptin, its precursor prepronociceptin and the NOP receptor
are localized to the corticolimibic regions and like the μ opioid receptor, δ opioid receptor and
κ opioid receptor, the NOP receptor, with around 60% sequence homology with the opioid
receptors, is negatively coupled to adenylate cyclase, activates potassium channels, and inhibits
calcium channels.159 The peptide is associated with processes that are relevant to pain,160, 161
learning and memory162, 163 as well as stress and anxiety.164, 165 The peptide is present in several
areas known to be involved in pain perception.166 Activation of the receptor produces spinal
analgesia but it also seems to antagonize the effects of opioids.

5. Metabolism of nociceptin
Nociceptin is metabolized by several proteases of different specificity. Aminopeptidase
N cleaves the Phe1 -Gly2 bond delivering phenylalanine and nociceptin(2–17) in
mouse brain slices, whereas endopeptidase 24.15 acts by cleaving nociceptin at the
Ala7 -Arg8 , at the Ala11 -Arg12 , and at the Arg12 -Lys13 bonds.167 The neutral endopeptidase
was reported not to be involved in the metabolism. In mouse spinal cord on the other hand,

Medicinal Research Reviews DOI 10.1002/med


16 r HALLBERG

Figure 12. Nociceptin is degraded to peptide fragments that have similar biological profiles to that of nociceptin,
i.e., hyperalgesia and antinociception, and to bioactive fragments that have opposite or different biological
profiles to those of nociceptin. Such fragments may, for example, interact with an unknown receptor that leads
only to antinociception.

synaptic membranes endopeptidase 24.15 is not involved but here the neutral endopeptidase
delivers nociceptin(1–13), nociceptin(14–17), and phenylalanine as the major metabolites from
nociceptin.168 Furthermore, small amounts of nociceptin(1–7), (1–9), (1–10), (1–11), (2–9), (2–
11), (2–13), (2–17), and (13–17) were detected in the synaptic membranes. Nociceptin is first me-
tabolized to nociceptin(1–13) and (14–17) in rat hippocampus. The nociceptin(1–13) fragment
is subsequently metabolized into nociceptin(1–9) and nociceptin(10–13).169 Nociceptin(2–17)
that is the major metabolite in human plasma undergoes further proteolytical processing at
its N-terminal to produce nociceptin(3–17), (4–17), and (5–17).170 The metabolism pattern of
nociceptin is highly specific for tissue type.
Some of the nociceptin fragments retain biological activity. The N-terminal nociceptin
(1–7), (1–9), and (1–13) fragments modulate nociceptin-induced scratching, biting, and lick-
ing behavior in mice.171 Some nociceptin fragments have similar biological effects as those of
nociceptin, while other fragments have opposite or different effects. It is probable that at least
some of the biological effects attributed to nociceptin, originate from its bioactive fragments.
For example, since nociceptin is known to produce both antinociception and hyperalgesia, it
was suggested that the antinociceptive activity arises after the conversion of the parent pep-
tide. This hypothesis is supported by studies that show that i.c.v. injection of the nociceptin
fragments (1–7) and (1–11) elicits antinociception without causing hyperalgesia,172 Fig. 12
and Table I. Furthermore, studies on nociceptin C-terminal fragments, such as nociceptin
(13–17), demonstrated an induction of nociceptive response in mice spinal cord, whereas
N-terminal fragments of nociceptin exhibited no effect.173, 174 It appears that effects of
N-terminal nociceptin fragments such as (1–7) and (1–11) are mediated through receptors
that are not recognized by the parent compound nociceptin.

6. Drug-like nociceptin receptor agonists and agonists


The nociceptin fragments (1–7) and (1–11) bind to an unknown binding site(s), but no nonpep-
tide ligands are reported to bind to this receptor(s). In contrast, both nonpeptide agonists and
antagonists are known to bind to the nociceptin receptor. Peripheral and spinal administration
of nociceptin results in antinociceptive effects in monkeys.175, 176 Similarly, administration of
RO0646198 (28), a high-affinity, selective nonpeptidic NOP agonist that has been extensively
studied,177, 178 results in antinociception in monkeys and this drug-like molecule was devoid of
respiratory depression, itch/scratching, and reinforcing effects,179 Fig. 13. However, RO0646198
exhibits a very low bioavailability after oral dosing, below 1% in monkeys180 limiting its value
as a potential pharmaceutical agent. More recently, selective NOP receptor agonists such as

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 17

Figure 13. Two nociceptin receptor agonists and one antagonist. The agonists 28 and 29 exhibit anxiolytic-like
activity in rat.

SCH-221510 (29) that is orally active have been disclosed.181 It has been demonstrated that
RO64–6198 exhibits an anxiolytic-like activity in rats182 and more recently a similar effect
was observed also with SCH 221510 that was qualitatively comparable with that achieved
with benzodiazepine.181 Thus, it was suggested that NOP agonists may represent a new class
of anxiolytic agents181 and in addition to that, based on new experimental data, it has been
proposed that mixed MOP/NOP receptor agonists might provide good analgesics since co-
activation of MOP and NOP receptors produce synergistic antinociception with a minimum
of side effects.183, 184 Furthermore, a mixed-action profile of NOP/opioid activity is suggested
to provide a valuable therapy to treat addiction to various abused substances and/or polydrug
addiction.185 Notably, SCH 486757, an orally available NOP receptor agonist whose structure
is very similar to that of SCH 221510, has recently entered human clinical trials for cough.186, 187
Potent selective drug-like NOP receptor antagonists such as Trap-101 (30) have also
been developed.188 Such antagonists may relieve the symptoms of patients with Parkinson’s
disease.189 The crystal structure of the nociceptin/orphanin FQ receptor (NOP receptor) in
complex with a selective antagonist, encompassing a spiropiperidine scaffold and that mimics
the first four N-terminal amino acid residues of a close analogue of nociceptin was recently
reported. Substantial conformational differences between NOP and the classical κ and μ opioid
receptors were observed.190

7. Three-dimensional structures of opioid receptors


The three-dimensional structures of all three classical opioid subtypes, the μ, δ, and κ re-
ceptors, have been determined, as has the structure of the nociceptin/orphanin FQ peptide
receptor.114, 135, 147, 190 The disclosures of the detailed structures of the receptors in complex
with antagonists have given very important information on the interactions of ligands with the
different opioid receptors. In 2000, the first high-resolution three-dimensional crystal structure
of rhodopsin was reported191 and in 2007 the first crystal structure of a GPCR activated by
a diffusible ligand; the β2-adrenergic receptor (β2-AR) appeared in the literature.192 Notably,
the first structure of a GPCR–G-protein complex that was coupled to the Gs protein activat-
ing adenylyl cyclase was recently disclosed.193 It is likely that opioid receptor complexes also
with agonists soon will be available, providing high-resolution insights and that are foreseen to
significantly facilitate the design of the subtype selective potent opioid receptor agonists.

8. In summary
Members of the opioid peptide family, such as the precursor peptide dynorphin A, activate
one type of receptor, the κ opioid receptor (KOP) while one of its degradation products, Leu-
enkephalin activates other receptor types, the δ and μ opioid receptors (DOP and MOP). Thus,

Medicinal Research Reviews DOI 10.1002/med


18 r HALLBERG

Figure 14. Captopril, the first ACE inhibitor in clinic (1978), and aliskiren, the first renin inhibitor in clinic (2007).

the rate of proteolytic degradation in various structures in brain and the pattern of degradation
of opioid peptides are of outmost importance for the biological outcome. Several different
types of proteases operate, and these proteases may be targets for future pharmaceuticals.
The opioid peptidemimetics, such as morphine (isolated 1804), pethidine (1933), and fentanyl
(1960), existed long before the endogenous ligands to the opioid receptors were identified in the
early 1970-ties. No drug-like opioid receptor agonist or antagonist have been developed and
reached clinical trials, where the endogenous opioids have been utilized as structural prototypes
and starting points, although the N-terminal tyrosine element can be recognized in many of
the drug-like opioid compounds. Newly available structural information about the detailed
molecular interactions of selective ligands binding to the three classical opioid receptors and
the nociceptin/orphanin FQ peptide receptor will be useful in the discovery processes aimed at
identifying new subtype selective pharmaceuticals that target the opioid system. Thus, a large
number of receptor-selective drug-like molecules mimicking the effects of opioid fragments at
their receptors have been developed. Opioid receptor antagonists are known, such as 25.

C. Angiotensins
Angiotensin II (Asp1 -Arg2 -Val3 -Tyr4 -Ile5 -His6 -Pro7 -Phe8 ), Ang II, elicits a pronounced hy-
pertensive effect and is a powerful modulator of a variety of cardiovascular functions. It was
observed in the early 1970s that minor modifications of the amino acid residue sequence of
Ang II could produce peptides that block the action of Ang II. These peptides (e.g., sarile and
saralasin) helped to confirm that the renin-angiotensin system (RAS) was a suitable target for
drugs aimed at combating hypertension, in particular.194–198

1. Drug-like ligands interacting with targets in RAS


Angiotensinogen is the precursor protein for Ang II. Thus, the two major proteases, renin and
ACE, responsible for the degradation to Ang II were early considered to be very attractive
drug targets. The ACE inhibitors were developed199, 200 and the first ACE inhibitor, the thiol
compound captopril (31) was introduced onto the market as early as 1978, Fig. 14. However, it
took almost 30 years before an inhibitor of renin, aliskiren (32), was introduced onto the market
in 2007.201–204 The development of efficient renin inhibitors was found difficult primarily due
to the peptidic character of the inhibitors initially studied. These were metabolically unstable
and poorly absorbed, which meant that oral bioavalability was too low. Furthermore, it was
difficult to predict effects in humans from results obtained in animal models.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 19

Figure 15. Losartan, the first AT1R antagonist in clinic; L-163,491, the first selective drug-like AT1R agonist;
M024/C21, the first selective drug-like AT2R agonist; and EMA401, a selective AT2R antagonist undergoing
clinical trials.

The first nonpeptide angiotensin II receptor blocker (ARB) losartan (33), acting as a se-
lective antagonist to the Ang II AT1 receptor (AT1R) was introduced onto the market in 1995,
Fig. 15. Several effective ARBs, collectively known as “sartans”, have subsequently been dis-
closed and are now widely used in clinical practice.205, 206 Efforts to develop single compounds
that can block both the AT1R and the receptor of the potent vasoconstrictor endothelin A are
now in progress.207 Furthermore, LCZ696, a dual antagonist of AT1R and inhibitor of neutral
endopeptidase is in late clinical trials.208 Drug-like compounds able to activate the AT1R with
high selectivity are known; one such (the first to be reported) is L-163,491 (34).209 The other
major Ang II receptor, the AT2 receptor (AT2R) has recently emerged as a new target for drug
therapy.210–212 The AT2R is abundant in fetal tissues but in adults this GPCR remains abun-
dant only in certain tissues such as vascular endothelium213 and the brain.214, 215 Activation of
the AT2 receptor affects neuronal cell differentiation and nerve regeneration.216–218 The AT2R
exerts vasodilatory, antiproliferative and anti-inflammatory effects,219–221 neuroprotection,222
and several peripheral effects mediated through the AT2R oppose those mediated through
the AT1R.223, 224 It is worth noting that the AT2R is re-expressed in some disease condi-
tions such as heart failure, renal failure, myocardial infarction, hypertension, and some brain
disorders.215, 225–230 The first receptor–selective and drug-like AT2R agonists M024/C21 (35),
that exhibits a pronounced anti-inflammatory effect231 was reported in 2004.232 It was recently
suggested that the combination of drug-like selective AT2R agonists with antihypertensive
treatment might lead to vasculoprotective effects even beyond the blood-pressure-reducing
effect.210, 233–235 No selective AT2R agonists, however, have still entered clinical trials. In con-
trast, EMA401 (36), which does not have significant CNS distribution after oral dosing236 and
which is an analogue of the commonly used research tool, the AT2R antagonist PD123319
is in clinical trials and is aimed for combating neuropathic pain.236–238 EMA401 is acting via
peripheral mechanisms. Thus, both of the Ang II receptors, AT1R and AT2R are today serving
as targets for drugs.

2. Metabolism of angiotensins
The aspartyl protease renin liberates the essentially inactive angiotensin I (Ang I) from circu-
lating and tissue angiotensinogen, Fig. 16. Notably, a receptor for renin exits.239 This receptor
named the (pro)renin receptor binds both renin and prorenin, the inactive form of renin. Re-
ceptor binding triggers intracellular signaling.240 The next step, the proteolytic cleavage of

Medicinal Research Reviews DOI 10.1002/med


20 r HALLBERG

Figure 16. Angiotensin II is degraded to several fragments that have either similar biological effects or very
different ones. The degradation products Ang(1–7), which activates the Mas/Ang(1–7) receptor, and Ang(3–8)
(Ang IV), which inhibits the insulin-regulated aminopeptidase (IRAP) are examples of peptides with very different
profiles from that of the parent compound.

angiotensin I to produce Ang II, is mediated mainly by the drug target, the metalloproteinase
ACE. However, cleavage by chymase, carboxypeptidase, cathepsin G, or tonin provide alterna-
tive routes by which Ang II can be generated.241 Furthermore, carboxypeptidase A6 produces
Ang II.242 Ang II can also be produced from the dodecapeptide angiotensin (1–12) Ang (1–12)
by for example, ACE or chymase and accumulating evidence suggests that Ang (1–12) is a
functionally important substrate for the actions of AngII in the brain.243–245
Proteolytic cleavage of the major effector peptide Ang II by glutamyl aminopeptidase A
(AP-A) and membrane alanyl aminopeptidase N (AP-N), result in the sequential removal of sin-
gle amino acid residues from the N-terminal end to form Ang III (Ang II(2–8)) and Ang IV (Ang
II(3–8)), respectively,246 both of which are essential neuropeptide fragments in the CNS.247–250
Ang IV, in particular, plays an important role.251–254 It is noteworthy that Ang IV can be formed
also by the action of aminopeptidases on Ang I before it is converted to Ang II.255 Further-
more, a human Ang II–related peptide, Ang A, has recently been discovered.256 This peptide,
(Ala1 )-Ang II, is formed when the aspartic acid residue of Ang II is decarboxylated256 and it
acts as a full agonist with properties that are similar to those of Ang II.257 The heptapeptide
alamandine (Ala1 -Arg2 -Val3 -Tyr4 -Ile5 -His6 -Pro7 ) has recently been detected in human blood,
and characterized. Alamandine, can be formed by hydrolysis of (Ala1 )-Ang II by ACE2.258
Ang IV as well as the fragment Ang (3–7) is further processed by chymotrypsin and
dipeptidyl carboxypeptidase into inactive fragments and amino acid residues.259–264 Ang

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 21
(3–7) is produced from Ang IV by carboxypeptidase P (Carb-P) and propyl oligopeptidase
(PO) cleavage. Chymotrypsin cleaves peptide bonds adjacent to Val, Tyr, and Ile and this
should be remembered when preparing metabolically stable Ang IV analogues and Ang IV
peptidemimetics.
Processing of both the decapeptide Ang I and the octapeptide Ang II to deliver the bioactive
Ang(1–7) are important metabolic pathways.265, 266 Ang (1–7) is formed from Ang II by the
removal of the C-terminal phenylalanine. This proteolytic step can be performed either by
Carb-P252 or by the exopeptidase ACE2.267 ACE2 probably plays an essential role in the local
renin-angiotensin system of the heart and kidney. Ang (1–7) can be formed from Ang I in
NG108–15 cells by deletion of the C-terminal tripeptide fragment.265 This process is carried
out by prolyl endopeptidase in rat, while NEP generates most of the Ang(1–7) from Ang
I.268 Ang (1–7) is liberated also from Ang (1–9) after the ACE-mediated cleavage of the Phe8 -
His9 dipeptide fragment in the C-terminal.269 Furthermore, aminopeptidase A removes the
Asp1 -Arg2 residues from both Ang I and Ang II,270–273 while aminopeptidase N deletes Asp1 -
Arg2 from Ang I and Arg2 -Val3 from Ang III.270, 274 Arginine-aminopeptidase can process
angiotensin peptides in the myocardium.275 Further, cathepsin A (deamidase) transforms Ang
I to the fragment Ang(1–9).276
The metabolism of Ang II results in the formation of several fragments with biological
activities that differ from that of the parent peptide as is also the case of the tachykinins and
the opioid peptides, Table I. Thus, several of the degradation products in RAS are bioactive.
These bioactive degradation products include Ang(1–7), Ang(1–9), Ang III (Ang(2–8)), Ang
A, and Ang IV (Ang(3–8)). Ang(1–9) and Ang(1–7), for example, potentiate NO liberation
by bradykinin from endothelial cells. Ang(1–9) is significantly more active than Ang(1–7) at
higher concentrations.276 Both Ang(1–9) and Ang(1–7) inhibit cardiomyocyte hypertrophy.277
Ang(1–9), exerts its anti-hypertrophic effects on angiotensin II induced cardiomyocyte hyper-
trophy through AT2R.278 Ang(1–9) also reduces cardiac fibrosis in stroke prone spontaneously
hypertensive rats through AT2R.279 Concerning Ang(1–7), a counter-regulatory axis of the
RAS exists, which functions mainly via ACE2/Ang(1–7)/Mas280–282 and inhibits many disad-
vantageous cardiovascular disease phenotypes.283, 284
Ang III and Ang IV (Ang II(3–8)) are important neuropeptide fragments in the
CNS.246–250, 285–287 Ang A acts similar to Ang II, while Ang IV exhibits very different activ-
ities. Braszko et al. reported in 1988 that intracerebroventricular injection of Ang IV improves
memory and learning in the rat.288 Ang IV affects motor activity, the performance of passive
avoidance, and a conditioned avoidance response. The effects of Ang IV in several animal
models were subsequently reported.248, 254, 289–293 Ang IV protects against ischemic stroke294, 295
and hyperglycemia.296 Several excellent reviews have been published on the role of Ang IV in
the brain.297–299
Many ACE and renin inhibitors and ARBs are in clinical use. They suppress to different
degrees the negative outcomes of AT1R stimulation that are induced mainly by Ang II. However,
in addition to the AT2R, as previously discussed herein, receptors of several degradation
products of Ang I and Ang II as well as proteases are relevant to address for different indications.
Glutamyl aminopeptidase A (AP-A) that delivers Ang III, is one example.300 Two receptor
targets have attracted particular attention in the recent years; the Ang (1–7)/Mas receptor301, 302
and the insulin-regulated aminopeptidase (IRAP), which is believed to be the target for Ang
IV, Fig. 16.

3. Angiotensin (1–7) and drug-like ligands


The heptapeptide angiotensin (1–7) produces vasodilation, and it has antiproliferative and na-
triuretic effects in normal rats. It has antidiuretic effects in water-loaded rats through activation

Medicinal Research Reviews DOI 10.1002/med


22 r HALLBERG

Figure 17. The Mas (Ang (1–7)) receptor agonist AVE-0991 and compound XNT that activates ACE2 and
promotes degradation of Ang II to Ang(1–7).

of Mas, which is a GPCR.301, 303 Ang(1–7) has been demonstrated to antagonize cardiac hyper-
trophy and fibrosis, outcomes that are mediated by the Mas receptor.283, 304 The heptapeptide
ameliorates cardiac remodeling by decreasing hypertrophy and fibrosis,283, 305, 306 and genetic
depletion of the Mas receptor causes dyslipidaemia, insulin resistance, and marked fibrotic
and hypertrophic changes in rat myocardium.307, 308 It has been reported that chronic adminis-
tration of angiotensin(1–7) also prevented diabetes-induced cardiovascular dysfunction.309, 310
Sanofi–Aventis undertook a medicinal chemistry program aimed at discover drug-like ago-
nists to the Mas receptor. This program resulted in AVE-0991 (37),308 which has considerable
structural similarities to the AT2R agonist M024/C21 (35), Figs. 15 and 17. AVE-0991, how-
ever, does not bind to AT2R. One characteristic feature of AVE-0991 is an aldehyde group.
AVE-0991 mimics the action of angiotensin (1–7) in many tissues by its vasodilating and na-
triuretic properties.301, 308 The magnitude of the infarcted area following an infarction induced
by left coronary artery ligation in rats is much smaller when the compound is administrated.311
Pronounced cardioprotective effects of AVE-0991 were also demonstrated in a diabetes rats
model,309, 312 and it mediates antifibrotic actions in a rat model of cirrhosis.313 It has recently
been reported that this Mas receptor agonist facilitates penile erection314 and that the compound
attenuates pulmonary remodeling in a model of asthma.315
Another heptapeptide, alamandine has actions that resemble those produced by angiotensin
(1–7); vasodilation, antifibrosis, anti-hypertensive, and central effects. Alamandine acts through
the Mas-related GPCR, MrgD.258 Interestingly, small molecules have been discovered that
are able to activate ACE2 and in this way promote the degradation of Ang II, and catalyze
the generation of Ang(1–7). Such compounds, such as the lead XNT (38) need to be further
optimized, but have demonstrated beneficial outcomes in animal models.316–319 Thus, this ACE2
activator (38) elicited reductions in blood pressure, improved cardiac function, and reversed the
myocardial and perivascular fibrosis observed in spontaneously hypertensive rats.316 However,
recently it was postulated that the biological effects of 38 is ACE2-independent and should not
be attributed to the activation of this enzyme.320 In summary, the preclinical data, in particular
with the Ang(1–7) peptidemimetic AVE-0991, are promising and the ACE2/Ang(1–7)/Mas
system is an interesting target for cardiovascular and pulmonary disorders, among others.280

4. Angiotensin IV and drug-like ligands


Ang IV is active in the CNS. A specific binding site for Ang IV was identified in 1992, and
was later named the AT4 receptor.321–323 After the discovery of binding sites, systematic SAR
studies were initiated by Wright and Harding,324–326 revealing that the Val-Tyr-Ile tripeptide

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 23

Figure 18. Examples of small compounds that interac with the IRAP/Ang IV receptor. Compounds 39 and 40
are derived from the hexapeptide Ang IV and compound 41 from virtual screening. IRAP inhibitors enhance
cognition.

motif of Ang IV is critical to its binding affinity. Kobori et al. at Taisho Pharmaceuticals filed
patent applications in the late 1990s that covered compounds, such as compound 39, that bind
strongly to Ang IV binding sites in guinea pig hippocampus membranes,327, 328 Fig. 18. Albiston
et al. suggested in 2001 that insulin-regulated aminopeptidase, a zinc-metallopeptidase of the
M1 family is the main target for Ang IV.329 IRAP was identified as cystinyl aminopeptidase
(CAP, EC 3.4.11.3), placental leucine aminopeptidase (P-LAP, soluble human homologue),
oxytocinase, gp160 or vp165.330–333
Considerable efforts were subsequently devoted to identify drug-like compounds that
mimic Ang IV and inhibit IRAP. It was believed that such compounds could provide new
classes of cognitive enhancers that could be used to treat Alzheimer’s disease and other related
disorders.297, 334–337 The proven effects of Ang IV and related peptide analogues on memory
and learning were promising.297–299 Improved cognitive enhancers are needed since the clinical
studies of the cholinesterase inhibitors and N-methyl-D-aspartate antagonists utilized in ther-
apy today have been mostly disappointing.338–341 The major problem encountered in the design
process of IRAP inhibitors was to make compounds able to penetrate the BBB. One approach
to drug-like Ang IV mimics relied on stepwise alterations of Ang IV by various cyclizations
to determine the bioactive conformation of the hexapeptide when it binds to IRAP.342, 343 This
approach took advantage of the knowledge that oxytocin and vasopressin, both of which have
positive effects on memory, are substrates of IRAP.344 These large peptides encompass a macro-
cyclic disulphide at the cleavage site. Potent macrocyclic inhibitors of IRAP have subsequently
been developed, such as 40,345, 346 but these macrocycles are still too peptidic to cross BBB.347
A very promising series of IRAP inhibitors was identified in 2008, after the in silico screening
of 1.5 million commercially available compounds against a model structure homologous to
IRAP.348, 349 These low-molecular-weight benzopyran-based inhibitors, exemplified by 41, act
as cognitive enhancers in rats. The benzopyran class of IRAP inhibitors provides promising
leads for further development.350 It is not known whether the ligands synthesized by Kobori
et al. inhibit IRAP.
Alternative macromolecular targets for Ang IV have been proposed, such as c-Met, a
tyrosine kinase receptor that binds hepatocyte growth factor and that is associated with memory
and learning consolidation.351, 352 No inhibitors of IRAP have still reached Phase I clinical trials.

5. In summary
Ang I and Ang II are degraded to bioactive fragments with pharmacological profiles that
differ from those of the parent peptides. At least two receptors of peptide fragments derived
from Ang II, the Mas receptor, and IRAP are being studied as targets for the systematic
development of drug-like new chemical entities. Furthermore, the concept of interfering with
the ACE2/Ang(1–7)/Mas receptor axis by applying ACE2 activators and manipulating in this

Medicinal Research Reviews DOI 10.1002/med


24 r HALLBERG

Figure 19. Kallidin is degraded to bradykinin, and both kallidin and bradykinin are agonists that bind prefer-
entially to the B2 receptor. Kallidin is degraded also to des-Arg10-kallidin, which has very high affinity for the
B1 receptor. Bradykinin is degradaded to des-Arg9-bradykinin with high affinity to the B1 receptor and to the
inactive bradykinin(1–7). The fragments have biological profiles that differ from those of the parent peptides.

way the protoelytic processing is an interesting approach. Angiotensin II AT1 and AT2 receptor
peptidemimetics, for example, 34 and 35, respectively, and drug-like molecules mimicking the
effects of the fragment Ang(1–7), such as 37 and the fragment Ang IV, such as 40 and 41 at
their receptors, have been developed.

D. Bradykinins
The nonapeptide bradykinin (Arg1 -Pro2 -Pro3 -Gly4 -Phe5 -Ser6 -Pro7 -Phe8 -Arg9 ) is a powerful
vasodilator that increases the permeability of capillaries. It constricts nonvascular smooth
muscle and is an active component in several physiological and pathological processes related to
inflammation and pain. The decapeptide kallidin (Lys1 -Arg2 -Pro3 -Pro4 -Gly5 -Phe6 -Ser7 -Pro8 -
Phe9 -Arg10 ), also known as “lysyl-bradykinin”, has similar effects to those of bradykinin. Both
bradykinin and kallidin play central roles in the CNS after injury and during infection and
inflammation.353 The two main kinin receptors, B1 and B2 354–356 have been cloned and sequenced
in several species.357–360 They belong to the GPCR family and both have been addressed as drug
targets. The B1 receptors play roles in chronic pain and inflammation361, 362 and are inducible
and poorly expressed under normal physiological conditions. B2 receptors, in contrast, are
constitutively expressed in several cell types and mediate vasodilation.363

1. Metabolism of bradykinins
Bradykinin and kallidin are produced by proteolytic processing of the low-molecular-weight
and high-molecular-weight glycoprotein kininogen by tissue or plasma kallikreins.364, 365
Trypsin and plasmin are also able to affect this conversion. Kallikreins are endopeptidases
of trypsin type, and are activated by autoproteolysis or alternatively by other proteases, for
example, after injury. Thus, the kallikreins play roles that are similar to that of renin in
the renin-angiotensin system, but no kallikrein inhibitors have reached the clinical practice.
Kallikrein, bradykinin, and kallidin have all been identified in the CNS, where bradykinin is
most abundant in the hypothalamus and the pituitary.366–368 Bradykinin can be formed from
kallidin by cleavage of the Lys1 -Arg2 bond by aminopeptidases, such as aminopeptidase N,369
Fig. 19 and Table I. It is worth noting that high-molecular-weight kininogen is the precursor
of bradykinin in plasma.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 25
Kallidin and bradykinin are degraded by several types of proteases, primarily by car-
boxypeptidase N (CPN) and ACE.354 Both the endopeptidase 24.15 and NEP are also in-
volved in these proteolytic processes.370 ACE, aminopeptidase P (APP), and CPN, hydrolyze
the Pro7 -Phe8 , Arg1 -Pro2 , and Phe8 -Arg9 bonds, respectively, of bradykinin in humans.371, 372
The combined proteolytic processing of kallidin and bradykinin in human heart membranes
by aminopeptidase M and NEP is an example of a process delivers an inactive metabolite,
bradykinin(1–7).373 Aminopeptidase P is responsible for most of the metabolism of bradykinin
in the rat liver, while both aminopeptidase P and ACE contribute to the degradation in
the rat lung. The inactive bradykinin(1–5) is formed to a large extent both in liver and
lung.374 Although, the proteolytic cleavages most often deliver inactive metabolites,375 arginyl-
carboxypeptidase, the zinc metallopeptidases carboxypeptidases M and N, mediate cleavages
of the Phe9 -Arg10 bond of kallidin and of the Phe8 -Arg9 bond of bradykinin that result in active
metabolites,376 Fig. 19. Plasmin, which is responsible for lysis of the fibrin clot, can also pro-
duce not only bradykinin but also bradykinin(1–8) from high-molecular weight kininogen.377
In fact, bradykinin(1–8), also known as “des-Arg9 -bradykinin”, has a higher affinity for the B1
receptor than bradykinin itself.359 The effect is even more pronounced for des-Arg10 -kallidin,
which has a nanomolar affinity for the B1 receptor. The binding affinities of des-Arg10 -kallidin
and des-Arg9 -bradykinin for the B2 receptor are considerably lower. Thus, the N-terminal Lys
has a strong impact on the affinity with which the peptides bind to the B1 but not to the B2
receptor. Furthermore, the C-terminal Arg is not crucial for binding, while removing of the
two-amino acid residue fragment Phe-Arg at the C-terminal is deleterious. Thus, even with
regard to bradykinin, peptide processing may lead to both inactive smaller peptide fragments
and bioactive fragments, in this example eliciting very different biological responses. In con-
clusion, bradykinin and kallidin are natural endogenous agonists of the B2 receptor while the
degradation products des-Arg9 -bradykinin and des-Arg10 -kallidin are specific agonists of the
B1 receptor, Fig. 19. The B1 receptor is indeed specialized across species to respond to kinin
metabolites, either des-Arg9 -BK, Lys-des-Arg9 -BK or des-Arg10 -KD that have been generated
by arginine carboxypeptidases.356, 378

2. Drug-like bradykinin receptor antagonists and agonists


The interest in pharmaceutical agents that are antagonists to the B1 receptor is partly a result of
the pharmacological results obtained with the peptidic antagonist, des-Arg9 ,Leu8 -bradykinin,
which is an analogue of the des-Arg9 -bradykinin, and related compounds.379, 380 It is notable
that the displacement of Phe for Leu in the 8 position of des-Arg9 -bradykinin results in an-
tagonism, which is in analogy with the outcome when Phe in the 8 position of angiotensin II
is displaced with a nonaromatic lipophilic carbon side chain, c.f. sarile and saralasin, the first
angiotensin II receptor blockers used in the clinic.194, 195 To date, a large number of peptidic
and nonpeptidic selective B1 receptor antagonists have been prepared.381 Activation of the B1
receptor renders an increase in the cytosolic calcium ion concentration, eventually resulting in
chronic and acute inflammatory responses. Notably, the B1 receptor that is synthesized de novo
in response to tissue injury is associated with hyperalgesia382–384 and selective and effective
drug-like antagonists to this inducible receptor may have a potential in future treatment of
chronic inflammation, pain,385–388 diabetes, and diabetic complications.389
The arylsulfonamide 42 is one example of a potent selective B1 antagonist discovered after
extensive structure activity work starting from a nonselective series of compounds, Fig. 20.
It has analgesic properties in animal models after oral administration.390 This selective B1
antagonist does not resemble any fragments or amino acid residues of des-Arg9 -bradykinin,
except for the basic elements. The biphenyl-based B1 antagonists represent the other major class
of nonpeptidic B1 antagonists that have been studied. The cyclopropyl derivative MK-0686

Medicinal Research Reviews DOI 10.1002/med


26 r HALLBERG

Figure 20. Three B1 receptor antagonists. The B1 receptor is the major target for des-Arg10-kallidin and des-
Arg9-bradykinin. MK-0686 has been evaluated in clinic for postherpetic neuralgia, osteoarthritis, and postsurgery
dental pain.

(43), which has been tested in Phase II clinical trials for postherpetic neuralgia, osteoarthritis,
and postsurgery dental pain, is one example from this group of substances. Human PK data
from MK-0686 are available.391 Interestingly, it has recently been reported that B1 receptor
antagonists, such as LF22–0542 (44), a water- soluble arylsulfonamide are promising candidates
for treating diabetic retinopathy after ocular application.392 Common binding motifs of aryl
sulfonamides and biarylmethyl amines have been proposed393 and all data from structure-
activity relationship and homology modeling394, 395 should hopefully promote the discovery of
investigational drugs antagonizing the B1 receptor and that are successful in clinical use.396, 397
The B2 receptor antagonists may be useful after traumatic brain injury.398, 399 The first B2
antagonist to be used in the clinic icatibant (45), has the length of ten amino acid residues and
is injected in connection with acute attacks of the rare hereditary angioedema that may be life
threatening and that is caused by a deficiency of the C1 esterase inhibitor in the complement
system,400–403 Fig. 21. Large programs have been devoted during the past two decades to
developing efficient, orally bioavailable drug-like B2 receptor antagonists.404, 405 The quinoline
derivative FR165649 (46), which has a glycine linker, is one of a large number of selective
B2 antagonists. Remarkably, this molecule can be transformed into the proinflammatory B2
receptor agonist FR190997 (47) by attaching a pyridine unit to the quinoline nucleus via a
methyleneoxy spacer.406–408 The agonist FR190997 induces a hypotensive response in rats that
is blocked by icatibant.409

3. In summary
Kallidin and bradykinin, which bind preferentially to the B2 receptor, are degraded to des-
Arg10-kallidin and des-Arg9-bradykinin, respectively, that both exhibit high affinity to the

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 27

Figure 21. The B2 receptor antagonists icatibant and FR165649, and the B2 receptor agonist FR190997. Icat-
ibant is used in the clinic to treat acute attacks of the rare hereditary angioedema (HAE). The proinflammatory
agonist FR190997 induces a hypotensive response in rats that is blocked by icatibant.

inducible B1 receptor. Thus, the proteolytic processing can result in fragments with biological
profiles that differ significantly from those of the parent peptides. Drug-like agonists and
antagonists have been discovered and most of the nonpeptide drug-like B1 and B2 antagonists
under development have been identified after screening and a subsequent systematic lead-
optimization process.

E. Neuropeptide Y
Neuropeptide Y (NPY, neuropeptide tyrosine) comprises 36 amino acid residues and is a mem-
ber of the pancreatic polypeptide family. This family of neuropeptides includes the structurally
related gastrointestinal hormone peptide YY (PYY, peptide tyrosine tyrosine) and the pancre-
atic polypeptide (PP).410, 411 NPY, is widely distributed in both the central and peripheral ner-
vous system and is involved in several physiological systems.412–415 NPY acts in humans through
four GPCRs; Y1, Y2, Y4, Y5,416–418 which have a relatively low sequence homology.415, 419 PP
is more likely to serve as the endogenous ligand to the Y4 receptor than NPY or PPY, and
this receptor also shows a greater variability than Y1, Y2, and Y5.420 In fact, the Y1, Y2, and
Y5 receptors are the most different GPCRs (with 27–32% overall identity) known that bind to
the same peptide.421 The Y1 and Y2 receptors are mainly responsible for the periphery effects
mediated by NPY.422
The impact of the neuropeptide on body weight regulation has attracted particular
interest.423 Hence, after chronic central administration of NPY to normal rats, a pathophys-
iological profile of similar nature to that observed in connection with human obesity was
developed.424, 425 Fasting leads to higher levels of NPY than normal in brain regions associated
with energy homeostasis, such as the paraventricular nucleus and the arcuate nucleus.426 It is

Medicinal Research Reviews DOI 10.1002/med


28 r HALLBERG

Figure 22. NPY is nonselective. The peptide is degraded to NPY(3–36), which is selective for the Y2 receptor
and has a different biological profile from that of NPY. NPY(3–36) is further degraded to the inactive NPY(3–35)
by elimination of the important tyrosine amide in the C-terminal.

believed that the feeding response is mediated by Y1 and/or the Y5 receptors expressed in
the hypothalamus.427 Peptide YY, occurs in two forms, PYY(1–36) and PYY(3–36), and these
are released when food is consumed. PYY(1–36) is an unselective Y1/Y2 agonist, while the
PYY(3–36) fragment is a highly selective Y2 agonist. Hence, proteolytic processing determines
the level of receptor selectivity. Peripheral administration of PYY(3–36) to humans significantly
reduces appetite and food intake,428, 429 which suggests that the Y2 receptor is also involved
in energy homeostasis.430 Thus, the experimental data suggest that stimulation of the Y1 and
Y5 receptors plays an important role in increased feeding while stimulation of the Y2 and Y4
receptors plays an important role in appetite inhibition. Furthermore, the activation of NPY re-
ceptors is associated with anxiety and depression,415, 431, 432 the experience of pain,433, 434 alcohol
dependence,435, 436 bone formation437, 438 as well as angina pectoris439 and cardiac stability.440

1. Metabolism of neuropeptide Y
Cleavage of the prepro-NPY sequence441 produces pro-NPY, which has a length of 69 amino
acid residues. This sequence contains (i) the 36 residues that will subsequently form NPY, (ii) the
three-peptide unit Gly-Lys-Arg, required for the cleavage and amidation, and (iii) a sequence
of length 30 amino acids that forms the C-flanking peptide of NPY. This latter sequence, often
named CPON, is liberated from the prohormone by a convertase. NPY is degraded by, for ex-
ample, NEP leading to cleavage of the Tyr20 -Tyr21 and the Leu30 -Ile31 bonds, which is consistent
with the specificity of this enzyme.442 Peptide bond cleavages in hippocampal synaptosomes
deliver NPY(1–30) and NPY(31–36). These reactions can be suppressed by pepstatin, which
suggests that an aspartic protease is involved.443 Removal of the N-terminal dipeptide moiety
Tyr1 -Pro2 by dipeptidyl-aminopeptidase IV yields NPY(3–36),444 a fragment that is inactive at
the Y1 receptor subtype but active at the Y2 receptor,445, 446 Fig. 22 and Table I. It is worth
noting that NPY (13–36) is a selective Y2 receptor agonist.447 The NPY/PYY receptor Y2 is
pharmacologically characterized by its ability to retain high affinity for N-terminally truncated
peptide fragments, not only NPY(3–36) and NPY(13–36) but also PYY(3–36) and PYY(13–
36). Thus, the enzymatic processing of the parent NPY and of PYY, can lead to fragments
with receptor subtype selectivity. Not only enzymes that are similar to dipeptidyl peptidase
IV, but also specific endopeptidases (such as meprin and neprilysin-like enzymes), postargi-
nine hydrolysing enzymes, aminopeptidase P, etc., are engaged in the processing. Deletion of
the C-terminal amino acid residue(s) by specific proteases yields products such as NPY3–35,
PYY3–35, NPY3—34, or PYY3–34, that are completely inactive.448
Medicinal Research Reviews DOI 10.1002/med
METABOLISM TO BIOACTIVE FRAGMENTS r 29

Figure 23. Examples of NPY Y5 receptor antagonists. The NPY Y5 receptor antagonists may be useful agents
to control appetite.

2. Drug-like NPY receptor antagonists


The hypothalamic NPY Y5 receptor, in particular, plays an important role in the central
regulation of food intake and energy balance.449–451 Considerable research, therefore, has been
devoted to developing efficient Y5 receptor antagonists, since they may be useful agents to
control appetite. The two NPY Y5 antagonists, MK-0557 (48) and Velneperit (49) suppressed
body weight gain in human clinical trials to a modest but significant degree.452–455 Fig. 23.
Furthermore, the Y5 receptor is expressed in limbic region, which suggests that this receptor is
also involved in mood disorders.456
Blocking the NPY, Y5, and Y1 neuropeptide receptors is one strategy to treat obe-
sity. Stimulating the Y2 and Y4 receptors may offer an alternative approach. Peptidic se-
lective dual Y2/Y4 and Y4 receptor agonists have been developed, such as obinepitide and
TM30338, respectively.457, 458 Obinepitide suppresses food intake in healthy obese subjects (data
from 7TM Pharma). Several selective drug-like Y5 receptor antagonists are known, such as
MK-0557 (48) and velneperit (49), but small-molecule ligands for the Y1 receptor are needed, as
are drug-like ligands for the Y2 and Y4 receptor that have favorable pharmacokinetic profiles.
These are needed not only as research tools but also as potential anti-obesity drugs. Although,
small-molecule Y2 receptor antagonists are available459–462 no nonpeptide Y2 and Y4 agonists
have been reported to date.463 The Y2 receptor is a pre- and postsynaptic receptor regulating
synthesis and release of NPY464 and Y2 receptor antagonists is thought to elevate levels of
NPY in the synapse and be beneficial for some CNS disorders.465, 466
Proteolytic processing may have an important regulatory function, and thus therapy that
relies on selective protease inhibitors may supplement Y5 and Y1 receptor antagonists and Y2
and Y4 receptor agonists in the treatment of obesity in the future.

3. In summary
The nonselective neuropeptide Y that is involved in many physiological systems can be converted
not only to biologically inert metabolites but also to degradation products that are bioactive
and more receptor selective than the parent peptide. NPY(3–36) is an example of a fragment
that exhibits selectivity for the Y2 receptor. Selective drug-like receptor antagonists are reported
Medicinal Research Reviews DOI 10.1002/med
30 r HALLBERG
but no drug-like Y2 or Y4 receptor agonists with potential use as antiobesity drugs are yet
disclosed.

F. New developments in neuropeptide metabolism


Advanced peptidomics-based strategies have recently been utilized to identify peptidases re-
sponsible for the proteolysis of significant bioactive peptides.467 One such strategy was applied
to calcitonin gene-related peptide (CGRP) and the use of a liquid chromatography-tandem
mass spectrometry peptidomics platform to detect CGRP fragments in tissues and reveal the
endogenous cleavage sites was the distinguishing feature of the approach. This and similar
methodologies will significantly facilitate the identification of important bioactive fragments
in the future. A combination of a peptidomics platform with biochemistry and genetics should
allow the discovery of the peptidases responsible for the proteolysis of bioactive peptides. Fur-
thermore, the role of peptide degrading enzymes in lysosomes in controlling the rate of recycling
peptide receptors to the cell surface has attracted interest.468 Thus, it has, for example, been
postulated that an endosomal location of endothelin-converting enzyme-1 (ECE-1) in smooth
muscle cells, may result in degradation of internalized CGRP and perhaps other neuropeptides,
to regulate recycling of GPCRs and resensitization.468 Biased GPCR agonists that possess
pathway-selective efficacy and activate or inhibit only a subset of the signaling pathway of
their receptors have recently been reported and discussed.469 Hence, biased agonists can be
employed to qualitatively change GPCR signaling in vivo. Concerning peptide fragments, the
truncated N-terminal fragment of PTH (bovine parathyroid hormone), bPTH(7–34) is a biased
agonist for the type 1 PTH receptor and antagonizes receptor-G protein coupling but activates
arrestin-dependent signaling. However, further studies are needed to elucidate whether degra-
dation fragments of the neuropeptides discussed in this review are acting as biased agonists.
Interestingly, the β-arrestin-biased AT1R ligand TRV120023 has cardioprotective and func-
tional properties in vivo which are distinct from the AT1R antagonist losartan. It was suggested
that this new class of drugs that is G-protein-independent but β-arrestin selective can provide
an advantage over conventional ARBs by supporting cardiac function and reducing cellular
injury during acute cardiac injury.470–472

3. CONCLUSION

The proteolytic processing of neuropeptides has an important regulatory function and the
peptide fragments resulting from the enzymatic degradation often exert essential physiological
roles. A large number of the peptide fragments display very different activities than their parent
peptides. This is well illustrated in the RAS system, where the short-lived metabolites of the
octapeptide Ang II, Ang IV (Ang(3–8)), and Ang(1–7) are generated. Both of these peptides
exert very different effects from Ang II. Drug-like substances that mimic the biological effects
of these fragments are now being examined as potential pharmaceutics. A metalloprotease is
the target for the Ang II degradation fragment Ang IV and the Mas receptor for the Ang II
degradation fragment Ang(1–7). Drug-like mimetics of the substance P fragment SP(1–7) have
been reported and provide another example from the tachykinin system, but significantly more
efforts have been devoted to finding selective and nonselective NK1, NK2, and NK3 antago-
nists for clinical use. NPY, although not receptor selective, is degraded to the agonistic selective
Y2 receptor NPY(3–36) fragment. No selective drug-like Y2 receptor agonists have been de-
veloped, even though selective drug-like Y2 receptor antagonists are known. The degradation
of dynorphin A (a κ opioid receptor agonist that produces dysphoria) to Leu-enkephalin (that
acts as a δ and μ opioid receptor agonist and induces euphoria) is one example from the opioid
Medicinal Research Reviews DOI 10.1002/med
METABOLISM TO BIOACTIVE FRAGMENTS r 31
system, where the protolytic processing leads to different biological effect profiles. It is worth
noting that with the exception of the opioid research area, most attention has been directed
toward drug-like receptor antagonists as new chemical entities.
Drug-like antagonists such as B2 receptor antagonists could be transformed into B2 recep-
tor agonists by small structural manipulations, and selective AT1 and AT2 receptor agonists
could be generated from nonselective AT1/AT2 receptor agonists, originally derived from the
sartans. Furthermore, opioid receptor agonists have been transformed to antagonists, and opi-
oid antagonists to agonists, and drug-like NK receptor agonists were obtained after minor
structural alterations of NK antagonists. Thus, herein, a representative series of important
drug-like molecules that act as agonists or antagonists at neuropeptide receptors has briefly
been discussed.
As exemplified in this review, many common neuropeptides are degraded into smaller
fragments that are often bioactive but relatively few of these fragments (e.g., substance P(1–7)
and angiotensin II(3–8)) have yet been examined in detail with respect to their biological
activities. Such short bioactive fragments should be less complicated to convert into drug-
like substances mimicking the desired effects of the degradation products (e.g., the substance
P(1–7) mimic 2 and the angiotensin II(3–8) mimic 39) and could provide a potential rich
source of new pharmaceuticals. However, very few examples heretofore have been reported of
successful design of drug-like agonists or antagonists using the endogenous peptide fragments
as starting points. The transformation of peptides to drug-like nonpeptides displaying favorable
pharmacokinetic profiles, through procedures involving rigidifications, displacement of peptide
bonds, replacement of amino acids with other moieties, truncations, etc. remains a challenge.

4. ABBREVIATIONS
ACE angiotensin-converting enzyme ARB angiotensin II receptor blocker AT1R AT1
receptor AT2R AT2 receptor BBB blood–brain barrier CGRP calcitonin gene-related peptide
CINV chemotherapy-induced nausea and vomiting CNS central nervous system CPE car-
boxypeptidase E GPCR G-protein-coupled receptor IRAP insulin-regulated aminopeptidase
NEP neutral endopeptidase NOP nociceptin opioid receptor NPY Neuropeptide Y RAS
renin-angiotensin system SPE substance P endopeptidase

ACKNOWLEDGMENTS

This study was supported by the Kjell and Märta Beijer Foundation and Swedish Research
Council. The author acknowledges Dr. Christer Westerlund, AstraZeneca, Dr. Rolf Johansson,
AstraZeneca, Dr. Ingrid Påhlman, AstraZeneca/Albireo Pharma, Professor Anders Hallberg,
Uppsala University, and Dr. Hanna Andersson, Gothenburg University for carefully scruti-
nizing the manuscript and for constructive criticism, not least concerning the drawing of the
chemical formula.

REFERENCES

1. Burger E. Peptide hormones and neuropeptides. Proteolytic processing of the precursor regulatory
peptides. Arzneimittelforschung 1988;38(5):754–761.
2. Hallberg M, Nyberg F. Neuropeptide conversion to bioactive fragments–an important pathway in
neuromodulation. Curr Protein Pept Sci 2003;4(1):31–44.
3. Nyberg F, Hallberg M. Peptide conversion–a potential pathway modulating G-protein signaling.
Current Drug Targets 2007;8(1):147–154.

Medicinal Research Reviews DOI 10.1002/med


32 r HALLBERG
4. Hokfelt T, Pernow B, Wahren J. Substance P: A pioneer amongst neuropeptides. J Intern Med
2001;249(1):27–40.
5. Hökfelt T, Kuteeva T, Stanie D, Ljungdahl Å. The histochemistry of tachykinin system in the brain.
In: Holzer P, Ed. Tachykinins, Handbook of Experimental Pharmacology. Heidelberg: Springer-
Verlag Berlin; 2004. p 63–120.
6. Zhang Y, Lu L, Furlonger C, Wu GE, Paige CJ. Hemokinin is a hematopoietic-specific tachykinin
that regulates B lymphopoiesis. Nat Immunol 2000;1(5):392–397.
7. Kurtz MM, Wang R, Clements MK, Cascieri MA, Austin CP, Cunningham BR, Chicchi GG, Liu
Q. Identification, localization and receptor characterization of novel mammalian substance P-like
peptides. Gene 2002;296(1–2):205–212.
8. Page NM, Bell NJ, Gardiner SM, Manyonda IT, Brayley KJ, Strange PG, Lowry PJ. Characteriza-
tion of the endokinins: Human tachykinins with cardiovascular activity. Proc Natl Acad Sci USA
2003;100(10):6245–6250.
9. Von Euler US, Gaddum JH. An unidentified depressor substance in certain tissue extracts. J Physiol
1931;72:74–87.
10. Chang MM, Leeman SE, Niall HD. Amino-acid sequence of substance P. Nat New Biol 1971;
232(29):86–87.
11. Zubrzycka M, Janecka A. Substance P: Transmitter of nociception (Minireview). Endocr Regul
2000;34(4):195–201.
12. Severini C, Improta G, Falconieri-Erspamer G, Salvadori S, Erspamer V. The tachykinin peptide
family. Pharmacol Rev 2002;54(2):285–322.
13. Harrison S, Geppetti P. Substance p. Int J Biochem Cell Biol 2001;33(6):555–576.
14. Rimon R, Le Greves P, Nyberg F, Heikkila L, Salmela L, Terenius L. Elevation of substance P-like
peptides in the CSF of psychiatric patients. Biol Psychiatry 1984;19(4):509–516.
15. Kramer MS, Cutler N, Feighner J, Shrivastava R, Carman J, Sramek JJ, Reines SA, Liu G, Snavely
D, Wyatt-Knowles E, Hale JJ, Mills SG, MacCoss M, Swain CJ, Harrison T, Hill RG, Hefti F,
Scolnick EM, Cascieri MA, Chicchi GG, Sadowski S, Williams AR, Hewson L, Smith D, Carlson
EJ, Hargreaves RJ, Rupniak NM. Distinct mechanism for antidepressant activity by blockade of
central substance P receptors. Science 1998;281(5383):1640–1645.
16. Argyropoulos SV, Nutt DJ. Substance P antagonists: Novel agents in the treatment of depression.
Expert Opin Investig Drugs 2000;9(8):1871–1875.
17. Carletti R, Corsi M, Melotto S, Caberlotto L. Down-regulation of amygdala preprotachykinin A
mRNA but not 3H-SP receptor binding sites in subjects affected by mood disorders and schizophre-
nia. Eur J Neurosci 2005;21(6):1712–1718.
18. McLean S. Do substance P and the NK1 receptor have a role in depression and anxiety? Curr Pharm
Des 2005;11(12):1529–1547.
19. Ebner K, Singewald N. The role of substance P in stress and anxiety responses. Amino Acids
2006;31(3):251–272.
20. Ebner K, Muigg P, Singewald G, Singewald N. Substance P in stress and anxiety: NK-1 receptor
antagonism interacts with key brain areas of the stress circuitry. Ann N Y Acad Sci 2008;1144:61–73.
21. Blier P, Gobbi G, Haddjeri N, Santarelli L, Mathew G, Hen R. Impact of substance P receptor an-
tagonism on the serotonin and norepinephrine systems: Relevance to the antidepressant/anxiolytic
response. J Psychiatry Neurosci 2004;29(3):208–218.
22. Santarelli L, Gobbi G, Debs PC, Sibille ET, Blier P, Hen R, Heath MJ. Genetic and pharmacological
disruption of neurokinin 1 receptor function decreases anxiety-related behaviors and increases
serotonergic function. Proc Natl Acad Sci USA 2001;98(4):1912–1917.
23. Duarte FS, Testolin R, De Lima TC. Further evidence on the anxiogenic-like effect of substance P
evaluated in the elevated plus-maze in rats. Behav Brain Res 2004;154(2):501–510.
24. De Araujo JE, Huston JP, Brandao ML. Opposite effects of substance P fragments C (anxiogenic)
and N (anxiolytic) injected into dorsal periaqueductal gray. Eur J Pharmacol 2001;432(1):43–51.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 33
25. Garces YI, Rabito SF, Minshall RD, Sagen J. Lack of potent antinociceptive activity by substance
P antagonist CP- 96,345 in the rat spinal cord. Life Sci 1993;52(4):353–360.
26. Lembeck F, Holzer P. Substance P as neurogenic mediator of antidromic vasodilation and neuro-
genic plasma extravasation. Naunyn-Schmiedeberg’s Arch Pharmacol 1979;310(2):175–183.
27. Levine JD, Dardick SJ, Roizen MF, Helms C, Basbaum AI. Contribution of sensory afferents and
sympathetic efferents to joint injury in experimental arthritis. J Neurosci 1986;6(12):3423–3429.
28. Barnes PJ. Asthma as an axon reflex. Lancet 1986;1(8475):242–245.
29. Mantyh CR, Gates TS, Zimmerman RP, Welton ML, Passaro EP, Jr., Vigna SR, Maggio JE,
Kruger L, Mantyh PW. Receptor binding sites for substance P, but not substance K or neuromedin
K, are expressed in high concentrations by arterioles, venules, and lymph nodules in surgical spec-
imens obtained from patients with ulcerative colitis and Crohn disease. Proc Natl Acad Sci USA
1988;85(9):3235–3239.
30. Barnes PJ, Baraniuk JN, Belvisi MG. Neuropeptides in the respiratory tract. Part I. Am Rev Respir
Dis 1991;144(5):1187–1198.
31. Michaud JC, Alonso R, Gueudet C, Fournier M, Calassi R, Breliere JC, Le Fur G, Soubrie P. Effects
of SR140333, a selective non-peptide NK1 receptor antagonist, on trigemino-thalamic nociceptive
pathways in the rat. Fundam Clin Pharmacol 1998;12(1):88–94.
32. Lowe JA, 3rd, Drozda SE, Snider RM, Longo KP, Zorn SH, Jackson ER, Morrone J, McLean S,
Bryce DK, Bordner J, Nagahisa A, Kanai Y, Suga O, Tsuchiya M. Discovery of CP-96,345 and its
characterization in disease models involving substance P. Regul Pept 1993;46(1–2):20–23.
33. Navari RM, Reinhardt RR, Gralla RJ, Kris MG, Hesketh PJ, Khojasteh A, Kindler H, Grote TH,
Pendergrass K, Grunberg SM, Carides AD, Gertz BJ. Reduction of cisplatin-induced emesis by
a selective neurokinin-1- receptor antagonist. L-754,030 Antiemetic Trials Group. N Engl J Med
1999;340(3):190–195.
34. Eipper BA, Stoffers DA, Mains RE. The biosynthesis of neuropeptides: Peptide alpha-amidation.
Annu Rev Neurosci 1992;15:57–85.
35. Freed AL, Audus KL, Lunte SM. Investigation of the metabolism of substance P at the blood-brain
barrier using capillary electrophoresis with laser-induced fluorescence detection. Electrophoresis
2001;22(17):3778–3784.
36. Sakurada C, Watanabe C, Sakurada T. Occurrence of substance P(1–7) in the metabolism of
substance P and its antinociceptive activity at the mouse spinal cord level. Methods Find Exp Clin
Pharmacol 2004;26(3):171–176.
37. Freed AL, Cooper JD, Davies MI, Lunte SM. Investigation of the metabolism of substance P in
rat striatum by microdialysis sampling and capillary electrophoresis with laser-induced fluorescence
detection. J Neurosci Methods 2001;109(1):23–29.
38. Skidgel RA, Engelbrecht S, Johnson AR, Erdos EG. Hydrolysis of substance p and neurotensin by
converting enzyme and neutral endopeptidase. Peptides 1984;5(4):769–776.
39. Yokosawa H, Endo S, Ogura Y, Ishii S. A new feature of angiotensin-converting enzyme in the
brain: Hydrolysis of substance P. Biochem Biophys Res Commun 1983;116(2):735–742.
40. Matsas R, Fulcher IS, Kenny AJ, Turner AJ. Substance P and [Leu]enkephalin are hydrolyzed by
an enzyme in pig caudate synaptic membranes that is identical with the endopeptidase of kidney
microvilli. Proc Natl Acad Sci USA 1983;80(10):3111–3115.
41. Hall ME, Stewart JM. Substance P and behavior: Opposite effects of N-terminal and C-terminal
fragments. Peptides 1983;4(5):763–768.
42. Sakurada T, Le Greves P, Stewart J, Terenius L. Measurement of substance P metabolites in rat
CNS. J Neurochem 1985;44(3):718–722.
43. De Araujo JE, Silva RC, Huston JP, Brandao ML. Anxiogenic effects of substance P and its 7–
11 C terminal, but not the 1–7 N terminal, injected into the dorsal periaqueductal gray. Peptides
1999;20(12):1437–1443.
44. De Araujo JE, Huston JP, Brandao ML. Opposite effects of substance P fragments C (anxiogenic)
and N (anxiolytic) injected into dorsal periaqueductal gray. Eur J Pharmacol 2001;432(1):43–51.

Medicinal Research Reviews DOI 10.1002/med


34 r HALLBERG
45. Persson S, Le Greves P, Thornwall M, Eriksson U, Silberring J, Nyberg F. Neuropeptide con-
verting and processing enzymes in the spinal cord and cerebrospinal fluid. Prog Brain Research
1995;104:111–130.
46. Zhou Q, Liu Z, Ray A, Huang W, Karlsson K, Nyberg F. Alteration in the brain content of substance
P (1–7) during withdrawal in morphine-dependent rats. Neuropharmacology 1998;37(12):1545–
1552.
47. Karlsson K, Eriksson U, Andren P, Nyberg F. Purification and characterization of substance P
endopeptidase activities in the rat spinal cord. Prep Biochem Biotechnol 1997;27(1):59–78.
48. Zhou Q, Karlsson K, Liu Z, Johansson P, Le Greves M, Kiuru A, Nyberg F. Substance P
endopeptidase-like activity is altered in various regions of the rat central nervous system during
morphine tolerance and withdrawal. Neuropharmacology 2001;41(2):246–253.
49. Pailleux F, Lemoine J, Beaudry F. Investigation of the metabolic biotransformation of substance
P in liver microsomes by liquid chromatography quadrupole ion trap mass spectrometry. Biomed
Chromatogr 2013;27(1):39–47.
50. Carlsson A, Ohsawa M, Hallberg M, Nyberg F, Kamei J. Substance P(1–7) induces antihyperalgesia
in diabetic mice through a mechanism involving the naloxone-sensitive sigma receptors. Eur J
Pharmacol 2010;626(2–3):250–255.
51. Stewart JM, Hall ME, Harkins J, Frederickson RC, Terenius L, Hokfelt T, Krivoy WA. A fragment
of substance P with specific central activity: SP(1–7). Peptides 1982;3(5):851–857.
52. Wiktelius D, Khalil Z, Nyberg F. Modulation of peripheral inflammation by the substance P
N-terminal metabolite substance P1–7. Peptides 2006;27(6):1490–1497.
53. Kreeger JS, Larson AA. The substance P amino-terminal metabolite substance P(1–7), administered
peripherally, prevents the development of acute morphine tolerance and attenuates the expression
of withdrawal in mice. J Pharmacol Exp Ther 1996;279(2):662–667.
54. Zhou Q, Nyberg F. Injection of substance P (SP) N-terminal fragment SP(1–7) into the ventral
tegmental area modulates the levels of nucleus accumbens dopamine and dihydroxyphenylacetic
acid in male rats during morphine withdrawal. Neurosci Lett 2002;320(3):117–120.
55. Botros M, Hallberg M, Johansson T, Zhou Q, Lindeberg G, Frandberg PA, Tomboly C, Toth G, Le
Greves P, Nyberg F. Endomorphin-1 and endomorphin-2 differentially interact with specific binding
sites for substance P (SP) aminoterminal SP1–7 in the rat spinal cord. Peptides 2006;27(4):753–759.
56. Igwe OJ, Kim DC, Seybold VS, Larson AA. Specific binding of substance P aminoterminal hep-
tapeptide [SP(1–7)] to mouse brain and spinal cord membranes. J Neurosci 1990;10(11):3653–3663.
57. Botros M, Johansson T, Zhou Q, Lindeberg G, Tomboly C, Toth G, Le Greves P, Nyberg F,
Hallberg M. Endomorphins interact with the substance P (SP) aminoterminal SP(1–7) binding in
the ventral tegmental area of the rat brain. Peptides 2008;29(10):1820–1824.
58. Fransson R, Botros M, Nyberg F, Lindeberg G, Sandstrom A, Hallberg M. Small peptides
mimicking substance P (1–7) and encompassing a C-terminal amide functionality. Neuropeptides
2008;42(1):31–37.
59. Ohsawa M, Carlsson A, Asato M, Koizumi T, Nakanishi Y, Fransson R, Sandstrom A, Hallberg
M, Nyberg F, Kamei J. The effect of substance P1–7 amide on nociceptive threshold in diabetic
mice. Peptides 2011;32(1):93–98.
60. Zhou Q, Carlsson A, Botros M, Fransson R, Sandstrom A, Gordh T, Hallberg M, Nyberg F. The
C-terminal amidated analogue of the substance P (SP) fragment SP(1–7) attenuates the expression
of naloxone-precipitated withdrawal in morphine dependent rats. Peptides 2009;30(12):2418–2422.
61. Fransson R, Botros M, Skold C, Nyberg F, Lindeberg G, Hallberg M, Sandstrom A. Discov-
ery of dipeptides with high affinity to the specific binding site for substance P1–7. J Med Chem
2010;53(6):2383–2389.
62. Fransson R, Skold C, Kratz JM, Svensson R, Artursson P, Nyberg F, Hallberg M, Sandstrom A.
Constrained H-Phe-Phe-NH2 analogues with high affinity to the substance P 1–7 binding site and
with improved metabolic stability and cell permeability. J Med Chem 2013;56(12):4953–4965.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 35
63. Ohsawa M, Carlsson A, Asato M, Koizumi T, Nakanishi Y, Fransson R, Sandstrom A, Hallberg
M, Nyberg F, Kamei J. The dipeptide Phe-Phe amide attenuates signs of hyperalgesia, allodynia
and nociception in diabetic mice using a mechanism involving the sigma receptor system. Mol Pain
2011;7:85.
64. Feldman EL, Russell JW, Sullivan KA, Golovoy D. New insights into the pathogenesis of diabetic
neuropathy. Curr Opin Neurol 1999;12(5):553–563.
65. Snider RM, Constantine JW, Lowe JA, 3rd, Longo KP, Lebel WS, Woody HA, Drozda SE, Desai
MC, Vinick FJ, Spencer RW, Hess H-J. A potent nonpeptide antagonist of the substance P (NK1)
receptor. Science 1991;251(4992):435–437.
66. Hargreaves R, Ferreira JC, Hughes D, Brands J, Hale J, Mattson B, Mills S. Development of
aprepitant, the first neurokinin-1 receptor antagonist for the prevention of chemotherapy-induced
nausea and vomiting. Ann N Y Acad Sci 2011;1222:40–48.
67. Ebner K, Sartori SB, Singewald N. Tachykinin receptors as therapeutic targets in stress-related
disorders. Curr Pharm Des 2009;15(14):1647–1674.
68. Hale JJ, Mills SG, MacCoss M, Finke PE, Cascieri MA, Sadowski S, Ber E, Chicchi
GG, Kurtz M, Metzger J, Eiermann G, Tsou NN, Tattersall FD, Rupniak NM, Williams AR,
Rycroft W, Hargreaves R, MacIntyre DE. Structural optimization affording 2-(R)-(1-(R)-3, 5-bis
(trifluoromethyl)phenylethoxy)-3-(S)-(4-fluoro)phenyl-4- (3-oxo-1,2,4-triazol-5-yl)methylmorpho-
line, a potent, orally active, long-acting morpholine acetal human NK-1 receptor antagonist.
J Med Chem 1998;41(23):4607–4614.
69. Kramer MS, Winokur A, Kelsey J, Preskorn SH, Rothschild AJ, Snavely D, Ghosh K, Ball WA,
Reines SA, Munjack D, Apter JT, Cunningham L, Kling M, Bari M, Getson A, Lee Y. Demonstra-
tion of the efficacy and safety of a novel substance P (NK1) receptor antagonist in major depression.
Neuropsychopharmacology 2004;29(2):385–392.
70. McLean S, Ganong A, Seymour PA, Bryce DK, Crawford RT, Morrone J, Reynolds LS, Schmidt
AW, Zorn S, Watson J, Fossa A, DePasquale M, Rosen T, Nagahisa A, Tsuchiya M, Heym J. Char-
acterization of CP-122,721; A nonpeptide antagonist of the neurokinin NK1 receptor. J Pharmacol
Exp Ther 1996;277(2):900–908.
71. Minthorn E, Mencken T, King AG, Shu A, Rominger D, Gontarek RR, Han C, Bambal R, Davis
CB. Pharmacokinetics and brain penetration of casopitant, a potent and selective neurokinin-1
receptor antagonist, in the ferret. Drug Metab Dispos 2008;36(9):1846–1852.
72. Ratti E, Bellew K, Bettica P, Bryson H, Zamuner S, Archer G, Squassante L, Bye A, Trist D,
Krishnan KR, Fernandes S. Results from 2 randomized, double-blind, placebo-controlled studies
of the novel NK1 receptor antagonist casopitant in patients with major depressive disorder. J Clin
Psychopharmacol 2011;31(6):727–733.
73. Frenkl TL, Zhu H, Reiss T, Seltzer O, Rosenberg E, Green S. A multicenter, double-blind, random-
ized, placebo controlled trial of a neurokinin-1 receptor antagonist for overactive bladder. J Urol
2010;184(2):616–622.
74. Santini D, Vincenzi B, Guida FM, Imperatori M, Schiavon G, Venditti O, Frezza AM, Berti P,
Tonini G. Aprepitant for management of severe pruritus related to biological cancer treatments: A
pilot study. Lancet Oncol 2012;13(10):1020–1024.
75. Wallengren J. Topical aprepitant in clinical and experimental pruritus. Arch Dermatol 2012;
148(8):957–959.
76. Munoz M, Martinez-Armesto J, Covenas R. NK-1 receptor antagonists as antitumor drugs: A
survey of the literature from 2000 to 2011. Expert Opin Ther Pat 2012;22(7):735–746.
77. Griebel G, Beeske S. Is there still a future for neurokinin 3 receptor antagonists as potential drugs
for the treatment of psychiatric diseases? Pharmacol Ther 2012;133(1):116–123.
78. Liem-Moolenaar M, Gray FA, de Visser SJ, Franson KL, Schoemaker RC, Schmitt JA, Cohen AF,
van Gerven JM. Psychomotor and cognitive effects of a single oral dose of talnetant (SB223412) in
healthy volunteers compared with placebo or haloperidol. J Psychopharmacol 2010;24(1):73–82.

Medicinal Research Reviews DOI 10.1002/med


36 r HALLBERG
79. Malherbe P, Ballard TM, Ratni H. Tachykinin neurokinin 3 receptor antagonists: A patent review
(2005–2010). Expert Opin Ther Pat 2011;21(5):637–655.
80. Malherbe P, Knoflach F, Hernandez MC, Hoffmann T, Schnider P, Porter RH, Wettstein JG,
Ballard TM, Spooren W, Steward L. Characterization of RO4583298 as a novel potent, dual antag-
onist with in vivo activity at tachykinin NK(1) and NK(3) receptors. Br J Pharmacol 2011;162(4):
929–946.
81. Lecci A, Capriati A, Altamura M, Maggi CA. Tachykinins and tachykinin receptors in the gut, with
special reference to NK2 receptors in human. Auton Neurosci 2006;126–127:232–249.
82. Altamura M. Tachykinin NK2 receptor antagonists. A patent review (2006–2010). Expert Opin
Ther Pat 2012;22(1):57–77.
83. Cappelli A, Giuliani G, Pericot Mohr Gl G, Gallelli A, Anzini M, Vomero S, Cupello A, Scarrone
S, Matarrese M, Moresco RM, Fazio F, Finetti F, Morbidelli L, Ziche M. A non-peptide NK1
receptor agonist showing subpicomolar affinity. J Med Chem 2004;47(6):1315–1318.
84. Natsugari H, Ikeura Y, Kiyota Y, Ishichi Y, Ishimaru T, Saga O, Shirafuji H, Tanaka T, Kamo I, Doi
T, Otsuka M. Novel, potent, and orally active substance P antagonists: Synthesis and antagonist ac-
tivity of N-benzylcarboxamide derivatives of pyrido[3,4-b]pyridine. J Med Chem 1995;38(16):3106–
3120.
85. Dhawan BN, Cesselin F, Raghubir R, Reisine T, Bradley PB, Portoghese PS, Hamon M. In-
ternational Union of Pharmacology. XII. Classification of opioid receptors. Pharmacol Rev
1996;48(4):567–592.
86. Perlman S, Costa-Neto CM, Miyakawa AA, Schambye HT, Hjorth SA, Paiva AC, Rivero RA,
Greenlee WJ, Schwartz TW. Dual agonistic and antagonistic property of nonpeptide angiotensin
AT1 ligands: Susceptibility to receptor mutations. Mol Pharmacol 1997;51(2):301–311.
87. Perlman S, Schambye HT, Rivero RA, Greenlee WJ, Hjorth SA, Schwartz TW. Non-peptide
angiotensin agonist. Functional and molecular interaction with the AT1 receptor. J Biol Chem
1995;270(4):1493–1496.
88. Hughes J, Smith TW, Kosterlitz HW, Fothergill LA, Morgan BA, Morris HR. Identification of two
related pentapeptides from the brain with potent opiate agonist activity. Nature 1975;258(5536):577–
580.
89. Zadina JE, Hackler L, Ge LJ, Kastin AJ. A potent and selective endogenous agonist for the mu-
opiate receptor. Nature 1997;386(6624):499–502.
90. Cooper JR, Bloom FE, Roth RH. Biochemical Basis of Neuropharmacology. New York: Oxford
University Press; 1996.
91. Wagner JJ, Chavkin C. Neuropharmacology of endogenous opioid peptides. In: Bloom FE, Kupfer
DJ, Eds. Psychopharmacology: The Fourth Generation of Progress. New York: Raven Press; 1995.
p 519–529.
92. Pasternak GW, Pan YX. Mu opioids and their receptors: Evolution of a concept. Pharmacol Rev
2013;65(4):1257–1317.
93. Witt KA, Gillespie TJ, Huber JD, Egleton RD, Davis TP. Peptide drug modifications to enhance
bioavailability and blood-brain barrier permeability. Peptides 2001;22(12):2329–2343.
94. Gentilucci L, De Marco R, Cerisoli L. Chemical modifications designed to improve peptide stability:
Incorporation of non-natural amino acids, pseudo-peptide bonds, and cyclization. Curr Pharm Des
2010;16(28):3185–3203.
95. Seizinger BR, Hollt V, Herz A. Proenkephalin B (prodynorphin)-derived opioid peptides: Evidence
for a differential processing in lobes of the pituitary. Endocrinology 1984;115(2):662–671.
96. Chavkin C, James IF, Goldstein A. Dynorphin is a specific endogenous ligand of the kappa opioid
receptor. Science 1982;215(4531):413–415.
97. Corbett AD, Paterson SJ, McKnight AT, Magnan J, Kosterlitz HW. Dynorphin and dynorphin are
ligands for the kappa-subtype of opiate receptor. Nature 1982;299(5878):79–81.
98. Hook V, Funkelstein L, Lu D, Bark S, Wegrzyn J, Hwang SR. Proteases for processing proneuropep-
tides into peptide neurotransmitters and hormones. Annu Rev Pharmacol Toxicol 2008;48:393–423.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 37
99. Koob GF. Drug addiction: The yin and yang of hedonic homeostasis. Neuron 1996;16(5):893–896.
100. Hallberg M, Le Greves P, Nyberg F. Neuropeptide processing. In: Lendeckel U, Hooper NM, Eds.
Proteases in Biology and Disease Proteases in the Brain, Vol 3. New York: Springer Science; 2005.
p 203–234.
101. Pfeiffer A, Brantl V, Herz A, Emrich HM. Psychotomimesis mediated by kappa opiate receptors.
Science 1986;233(4765):774–776.
102. Chavkin C. Dynorphin–still an extraordinarily potent opioid peptide. Mol Pharmacol 2013;
83(4):729–736.
103. Laughlin TM, Vanderah TW, Lashbrook J, Nichols ML, Ossipov M, Porreca F, Wilcox GL.
Spinally administered dynorphin A produces long-lasting allodynia: Involvement of NMDA but
not opioid receptors. Pain 1997;72(1–2):253–260.
104. Reed B, Zhang Y, Chait BT, Kreek MJ. Dynorphin A(1–17) biotransformation in striatum of freely
moving rats using microdialysis and matrix-assisted laser desorption/ionization mass spectrometry.
J Neurochem 2003;86(4):815–823.
105. Day R, Lazure C, Basak A, Boudreault A, Limperis P, Dong W, Lindberg I. Prodynorphin pro-
cessing by proprotein convertase 2. Cleavage at single basic residues and enhanced processing in the
presence of carboxypeptidase activity. J Biol Chem 1998;273(2):829–836.
106. Silberring J, Castello ME, Nyberg F. Characterization of dynorphin A-converting enzyme in human
spinal cord. An endoprotease related to a distinct conversion pathway for the opioid heptadecapep-
tide? J Biol Chem 1992;267(30):21324–21328.
107. Berman Y, Ageyeva L, Veksler B, Wood D, Devi LA. Dynorphin A processing enzyme: Tissue
distribution, isolation, and characterization. J Biochem (Tokyo) 1999;125(3):641–647.
108. Roques BP, Fournie-Zaluski MC, Wurm M. Inhibiting the breakdown of endogenous opioids and
cannabinoids to alleviate pain. Nat Rev 2012;11(4):292–310.
109. Vauquelin G, Fierens F, Van Liefde I. Long-lasting angiotensin type 1 receptor binding and pro-
tection by candesartan: Comparison with other biphenyl-tetrazole sartans. J Hypertens Suppl
2006;24(1):S23–S30.
110. Copeland RA. The dynamics of drug-target interactions: Drug-target residence time and its impact
on efficacy and safety. Expert Opin Drug Discov 2010;5(4):305–310.
111. Vauquelin G, Charlton SJ. Long-lasting target binding and rebinding as mechanisms to prolong in
vivo drug action. Br J Pharmacol 2010;161(3):488–508.
112. Breivik H. Opioids in chronic non-cancer pain, indications and controversies. Eur J Pain
2005;9(2):127–130.
113. Johnson RE, Fudala PJ, Payne R. Buprenorphine: Considerations for pain management. J Pain
Symptom Manage 2005;29(3):297–326.
114. Manglik A, Kruse AC, Kobilka TS, Thian FS, Mathiesen JM, Sunahara RK, Pardo L, Weis WI,
Kobilka BK, Granier S. Crystal structure of the μ-opioid receptor bound to a morphinan antagonist.
Nature 2012;485(7398):321–326.
115. Hunter JC, Leighton GE, Meecham KG, Boyle SJ, Horwell DC, Rees DC, Hughes J. CI-977, a
novel and selective agonist for the kappa-opioid receptor. Br J Pharmacol 1990;101(1):183–189.
116. Emmerson PJ, Liu MR, Woods JH, Medzihradsky F. Binding affinity and selectivity of opi-
oids at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp Ther
1994;271(3):1630–1637.
117. Siener T, Cambareri A, Kuhl U, Englberger W, Haurand M, Kogel B, Holzgrabe U. Synthesis and
opioid receptor affinity of a series of 2, 4-diaryl-substituted 3,7-diazabicylononanones. J Med Chem
2000;43(20):3746–3751.
118. Roth BL, Baner K, Westkaemper R, Siebert D, Rice KC, Steinberg S, Ernsberger P, Rothman RB.
Salvinorin A: A potent naturally occurring nonnitrogenous kappa opioid selective agonist. Proc
Natl Acad Sci USA 2002;99(18):11934–11939.

Medicinal Research Reviews DOI 10.1002/med


38 r HALLBERG
119. Harding WW, Tidgewell K, Byrd N, Cobb H, Dersch CM, Butelman ER, Rothman RB, Prisinzano
TE. Neoclerodane diterpenes as a novel scaffold for mu opioid receptor ligands. J Med Chem
2005;48(15):4765–4771.
120. Nagase H, Hayakawa J, Kawamura K, Kawai K, Takezawa Y, Matsuura H, Tajima C, Endo T.
Discovery of a structurally novel opioid kappa-agonist derived from 4,5-epoxymorphinan. Chem
Pharm Bull (Tokyo) 1998;46(2):366–369.
121. Kawai K, Hayakawa J, Miyamoto T, Imamura Y, Yamane S, Wakita H, Fujii H, Kawamura K,
Matsuura H, Izumimoto N, Kobayashi R, Endo T, Nagase H. Design, synthesis, and structure-
activity relationship of novel opioid kappa-agonists. Bioorg Med Chem 2008;16(20):9188–9201.
122. Nakao K, Mochizuki H. Nalfurafine hydrochloride: A new drug for the treatment of uremic pruritus
in hemodialysis patients. Drugs Today (Barc) 2009;45(5):323–329.
123. Nagase H, Nemoto T, Matsubara A, Saito M, Yamamoto N, Osa Y, Hirayama S, Nakajima M,
Nakao K, Mochizuki H, Fujii H. Design and synthesis of KNT-127, a delta-opioid receptor agonist
effective by systemic administration. Bioorg Med Chem Lett 2010;20(21):6302–6305.
124. Tsuji M, Takeda H, Matsumiya T, Nagase H, Narita M, Suzuki T. The novel kappa-opioid receptor
agonist TRK-820 suppresses the rewarding and locomotor-enhancing effects of morphine in mice.
Life Sci 2001;68(15):1717–1725.
125. Mucha RF, Herz A. Motivational properties of kappa and mu opioid receptor agonists studied with
place and taste preference conditioning. Psychopharmacology (Berl) 1985;86(3):274–280.
126. Millan MJ. Kappa-opioid receptors and analgesia. Trends Pharmacol Sci 1990;11(2):70–76.
127. Clark JA, Liu L, Price M, Hersh B, Edelson M, Pasternak GW. Kappa opiate receptor multiplicity:
Evidence for two U50,488-sensitive kappa 1 subtypes and a novel kappa 3 subtype. J Pharmacol
Exp Ther 1989;251(2):461–468.
128. Nock B. κ and ϵ opioid receptor binding. In: Tseng LF, Ed. The Pharmacology of Opioid Peptides.
Singapore: Harwood Academic Publishers; 1995. p 29–56.
129. Endoh T, Matsuura H, Tajima A, Izumimoto N, Tajima C, Suzuki T, Saitoh A, Suzuki T, Narita
M, Tseng L, Nagase H. Potent antinociceptive effects of TRK-820, a novel kappa-opioid receptor
agonist. Life Sci 1999;65(16):1685–1694.
130. Endoh T, Tajima A, Suzuki T, Kamei J, Narita M, Tseng L, Nagase H. Characterization of the
antinociceptive effects of TRK-820 in the rat. Eur J Pharmacol 2000;387(2):133–140.
131. Tsuji M, Yamazaki M, Takeda H, Matsumiya T, Nagase H, Tseng LF, Narita M, Suzuki T. The
novel kappa-opioid receptor agonist TRK-820 has no affect on the development of antinociceptive
tolerance to morphine in mice. Eur J Pharmacol 2000;394(1):91–95.
132. Tsuji M, Takeda H, Matsumiya T, Nagase H, Yamazaki M, Narita M, Suzuki T. A novel kappa-
opioid receptor agonist, TRK-820, blocks the development of physical dependence on morphine in
mice. Life Sci 2000;66(25):PL353–PL358.
133. Mori T, Nomura M, Yoshizawa K, Nagase H, Narita M, Suzuki T. Differential properties between
TRK-820 and U-50,488H on the discriminative stimulus effects in rats. Life Sci 2004;75(20):2473–
2482.
134. Zukin RS, Eghbali M, Olive D, Unterwald EM, Tempel A. Characterization and visualization of rat
and guinea pig brain kappa opioid receptors: Evidence for kappa 1 and kappa 2 opioid receptors.
Proc Natl Acad Sci USA 1988;85(11):4061–4065.
135. Wu H, Wacker D, Mileni M, Katritch V, Han GW, Vardy E, Liu W, Thompson AA, Huang XP,
Carroll FI, Mascarella SW, Westkaemper RB, Mosier PD, Roth BL, Cherezov V, Stevens RC.
Structure of the human kappa-opioid receptor in complex with JDTic. Nature 2012;485(7398):327–
332.
136. Vardy E, Mosier PD, Frankowski KJ, Wu H, Katritch V, Westkaemper RB, Aube J, Stevens RC,
Roth BL. Chemotype-selective modes of action of kappa-opioid receptor agonists. J Biol Chem
2013;288(48):34470–34483.
137. Calderon SN, Rothman RB, Porreca F, Flippen-Anderson JL, McNutt RW, Xu H, Smith LE,
Bilsky EJ, Davis P, Rice KC. Probes for narcotic receptor mediated phenomena. 19. Synthesis

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 39
of (+)-4-[(alpha R)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3- methoxybenzyl]-N,N-
diethylbenzamide (SNC 80): A highly selective, nonpeptide delta opioid receptor agonist. J Med
Chem 1994;37(14):2125–2128.
138. Chang KJ, Rigdon GC, Howard JL, McNutt RW. A novel, potent and selective nonpeptidic delta
opioid receptor agonist BW373U86. J Pharmacol Exp Ther 1993;267(2):852–857.
139. Bilsky EJ, Calderon SN, Wang T, Bernstein RN, Davis P, Hruby VJ, McNutt RW, Rothman RB,
Rice KC, Porreca F. SNC 80, a selective, nonpeptidic and systemically active opioid delta agonist.
J Pharmacol Exp Ther 1995;273(1):359–366.
140. Portoghese PS, Sultana M, Takemori AE. Design of peptidomimetic delta opioid receptor antago-
nists using the message-address concept. J Med Chem 1990;33(6):1714–1720.
141. Takemori AE, Sultana M, Nagase H, Portoghese PS. Agonist and antagonist activities of ligands
derived from naltrexone and oxymorphone. Life Sci 1992;50(20):1491–1495.
142. Varga EV, Navratilova E, Stropova D, Jambrosic J, Roeske WR, Yamamura HI. Agonist-specific
regulation of the delta-opioid receptor. Life Sci 2004;76(6):599–612.
143. Nagase H, Kawai K, Hayakawa J, Wakita H, Mizusuna A, Matsuura H, Tajima C, Takezawa Y,
Endoh T. Rational drug design and synthesis of a highly selective nonpeptide delta-opioid agonist,
(4aS*,12aR*)-4a-(3-hydroxyphenyl)-2-methyl- 1,2,3,4,4a,5,12,12a-octahydropyrido[3,4-b]acridine
(TAN-67). Chem Pharm Bull (Tokyo) 1998;46(11):1695–1702.
144. Nagase H, Yajima Y, Fujii H, Kawamura K, Narita M, Kamei J, Suzuki T. The pharmacological
profile of delta opioid receptor ligands, (+) and (-) TAN-67 on pain modulation. Life Sci 2001;68(19–
20):2227–2231.
145. Portoghese PS, Sultana M, Nagase H, Takemori AE. Application of the message-address concept
in the design of highly potent and selective non-peptide delta opioid receptor antagonists. J Med
Chem 1988;31(2):281–282.
146. Portoghese PS. Bivalent ligands and the message-address concept in the design of selective opioid
receptor antagonists. Trends Pharmacol Sci 1989;10(6):230–235.
147. Granier S, Manglik A, Kruse AC, Kobilka TS, Thian FS, Weis WI, Kobilka BK. Structure of the
delta-opioid receptor bound to naltrindole. Nature 2012;485(7398):400–404.
148. Latt NC, Jurd S, Houseman J, Wutzke SE. Naltrexone in alcohol dependence: A randomised
controlled trial of effectiveness in a standard clinical setting. Med J Aust 2002;176(11):530–534.
149. Mosberg HI, Fowler CB. Development and validation of opioid ligand-receptor interaction models:
The structural basis of mu vs delta selectivity. J Pept Res 2002;60(6):329–335.
150. Podlogar BL, Paterlini MG, Ferguson DM, Leo GC, Demeter DA, Brown FK, Reitz AB. Confor-
mational analysis of the endogenous mu-opioid agonist endomorphin-1 using NMR spectroscopy
and molecular modeling. FEBS Lett 1998;439(1–2):13–20.
151. Borics A, Toth G. Structural comparison of mu-opioid receptor selective peptides confirmed four
parameters of bioactivity. J Mol Graph Model 2010;28(6):495–505.
152. Liu X, Kai M, Jin L, Wang R. Molecular modeling studies to predict the possible binding modes of
endomorphin analogs in mu opioid receptor. Bioorg Med Chem Lett 2009;19(18):5387–5391.
153. Fowler CB, Pogozheva ID, Lomize AL, LeVine H, 3rd, Mosberg HI. Complex of an active mu-
opioid receptor with a cyclic peptide agonist modeled from experimental constraints. Biochemistry
2004;43(50):15796–15810.
154. Gentilucci L. Addressing the interactions between opioid ligands and their receptors – Classic and
nonclassic cases. In: Nyberg F, Ed. Neuropeptides in Neuroprotection and Neuroregeneration.
Boca Raton, FL: CRC Press; 2012. p 229–252.
155. Meunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, Butour JL, Guillemot JC,
Ferrara P, Monsarrat B, Mazarguil H, Vassart G, Parmentier M, Costentin J. Isolation and structure
of the endogenous agonist of opioid receptor- like ORL1 receptor. Nature 1995;377(6549):532–535.
156. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, Grandy DK,
Langen H, Monsma FJ, Jr., Civelli O. Orphanin FQ: A neuropeptide that activates an opioidlike
G protein- coupled receptor. Science 1995;270(5237):792–794.

Medicinal Research Reviews DOI 10.1002/med


40 r HALLBERG
157. Bunzow JR, Saez C, Mortrud M, Bouvier C, Williams JT, Low M, Grandy DK. Molecular cloning
and tissue distribution of a putative member of the rat opioid receptor gene family that is not a mu,
delta or kappa opioid receptor type. FEBS Lett 1994;347(2–3):284–288.
158. Mollereau C, Parmentier M, Mailleux P, Butour JL, Moisand C, Chalon P, Caput D, Vassart G,
Meunier JC. ORL1, a novel member of the opioid receptor family. Cloning, functional expression
and localization. FEBS Lett 1994;341(1):33–38.
159. Meunier J, Mouledous L, Topham CM. The nociceptin (ORL1) receptor: Molecular cloning and
functional architecture. Peptides 2000;21(7):893–900.
160. Meunier JC. Nociceptin/orphanin FQ and the opioid receptor-like ORL1 receptor. Eur J Pharmacol
1997;340(1):1–15.
161. Flores CA, Wang XM, Zhang KM, Mokha SS. Orphanin FQ produces gender-specific modu-
lation of trigeminal nociception: Behavioral and electrophysiological observations. Neuroscience
2001;105(2):489–498.
162. Sandin J, Georgieva J, Schott PA, Ogren SO, Terenius L. Nociceptin/orphanin FQ microinjected
into hippocampus impairs spatial learning in rats. Eur J Neurosci 1997;9(1):194–197.
163. Manabe T, Noda Y, Mamiya T, Katagiri H, Houtani T, Nishi M, Noda T, Takahashi T, Sugimoto
T, Nabeshima T, Takeshima H. Facilitation of long-term potentiation and memory in mice lacking
nociceptin receptors. Nature 1998;394(6693):577–581.
164. Griebel G, Perrault G, Sanger DJ. Orphanin FQ, a novel neuropeptide with anti-stress-like activity.
Brain Res 1999;836(1–2):221–224.
165. Reinscheid R, Civelli O. The orphanin FQ/nociceptin knockout mouse: A behavioral model for
stress responses. Neuropeptides 2002;36(2–3):72.
166. Neal CR, Jr., Mansour A, Reinscheid R, Nothacker HP, Civelli O, Watson SJ, Jr. Localization of
orphanin FQ (nociceptin) peptide and messenger RNA in the central nervous system of the rat. J
Comparative Neurol 1999;406(4):503–547.
167. Montiel JL, Cornille F, Roques BP, Noble F. Nociceptin/orphanin FQ metabolism: Role of
aminopeptidase and endopeptidase 24.15. J Neurochem 1997;68(1):354–361.
168. Sakurada C, Sakurada S, Orito T, Tan-No K, Sakurada T. Degradation of nociceptin (orphanin
FQ) by mouse spinal cord synaptic membranes is triggered by endopeptidase-24.11: An in vitro and
in vivo study. Biochem Pharmacol 2002;64(8):1293–1303.
169. Sandin J, Georgieva J, Silberring J, Terenius L. In vivo metabolism of nociceptin/orphanin FQ in
rat hippocampus. Neuroreport 1999;10(1):71–76.
170. Yu J, Chait BT, Toll L, Kreek MJ. Nociceptin in vitro biotransformation in human blood. Peptides
1996;17(5):873–876.
171. Sakurada T, Sakurada S, Katsuyama S, Hayashi T, Sakurada C, Tan-No K, Johansson H, Sandin
J, Terenius L. Evidence that N-terminal fragments of nociceptin modulate nociceptin- induced
scratching, biting and licking in mice. Neurosci Lett 2000;279(1):61–64.
172. Rossi GC, Leventhal L, Bolan E, Pasternak GW. Pharmacological characterization of orphanin
FQ/nociceptin and its fragments. J Pharmacol Exp Ther 1997;282(2):858–865.
173. Inoue M, Matsunaga S, Rashid MH, Yoshida A, Mizuno K, Sakurada T, Takeshima H, Ueda H.
Pronociceptive effects of nociceptin/orphanin FQ (13–17) at peripheral and spinal level in mice. J
Pharmacol Exp Ther 2001;299(1):213–219.
174. Chen LX, Wang ZZ, Wu H, Fang Q, Chen Y, Wang R. Effects of nociceptin (13–17) in pain
modulation at supraspinal level in mice. Neurosci Lett 2002;331(2):95–98.
175. Ko MC, Naughton NN, Traynor JR, Song MS, Woods JH, Rice KC, McKnight AT. Orphanin
FQ inhibits capsaicin-induced thermal nociception in monkeys by activation of peripheral ORL1
receptors. Br J Pharmacol 2002;135(4):943–950.
176. Ko MC, Wei H, Woods JH, Kennedy RT. Effects of intrathecally administered nociceptin/
orphanin FQ in monkeys: Behavioral and mass spectrometric studies. J Pharmacol Exp Ther
2006;318(3):1257–1264.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 41
177. Wichmann J, Adam G, Rover S, Hennig M, Scalone M, Cesura AM, Dautzenberg FM, Jenck F.
Synthesis of (1S,3aS)-8-(2,3,3a,4,5, 6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.
5]decan-4-one, a potent and selective orphanin FQ (OFQ) receptor agonist with anxiolytic-like
properties. Eur J Med Chem 2000;35(9):839–851.
178. Shoblock JR. The pharmacology of Ro 64–6198, a systemically active, nonpeptide NOP receptor
(opiate receptor-like 1, ORL-1) agonist with diverse preclinical therapeutic activity. CNS Drug Rev
2007;13(1):107–136.
179. Ko MC, Woods JH, Fantegrossi WE, Galuska CM, Wichmann J, Prinssen EP. Behavioral effects of
a synthetic agonist selective for nociceptin/orphanin FQ peptide receptors in monkeys. Neuropsy-
chopharmacology 2009;34(9):2088–2096.
180. Heinig K, Kratochwil N, Bucheli F, Thomae A. Bioanalytics and pharmacokinetics of the noci-
ceptin/orphanin FQ peptide receptor agonist RO0646198 in Wistar rats and Cynomolgus monkeys.
J Chromatogr B Analyt Technol Biomed Life Sci 2010;878(23):2101–2105.
181. Varty GB, Lu SX, Morgan CA, Cohen-Williams ME, Hodgson RA, Smith-Torhan A, Zhang H,
Fawzi AB, Graziano MP, Ho GD, Matasi J, Tulshian D, Coffin VL, Carey GJ. The anxiolytic-like
effects of the novel, orally active nociceptin opioid receptor agonist 8-[bis(2-methylphenyl)methyl]-
3-phenyl-8-azabicyclo[3.2.1]octan-3-ol (SCH 221510). J Pharmacol Exp Ther 2008;326(2):672–682.
182. Jenck F, Wichmann J, Dautzenberg FM, Moreau JL, Ouagazzal AM, Martin JR, Lundstrom K,
Cesura AM, Poli SM, Roever S, Kolczewski S, Adam G, Kilpatrick G. A synthetic agonist at the
orphanin FQ/nociceptin receptor ORL1: Anxiolytic profile in the rat. Proc Natl Acad Sci USA
2000;97(9):4938–4943.
183. Cremeans CM, Gruley E, Kyle DJ, Ko MC. Roles of mu-opioid receptors and nociceptin/orphanin
FQ peptide receptors in buprenorphine-induced physiological responses in primates. J Pharmacol
Exp Ther 2012;343(1):72–81.
184. Lin AP, Ko MC. The therapeutic potential of nociceptin/orphanin FQ receptor agonists as anal-
gesics without abuse liability. ACS Chem Neurosci 2013;4(2):214–224.
185. Zaveri NT. The nociceptin/orphanin FQ receptor (NOP) as a target for drug abuse medications.
Curr Top Med Chem 2011;11(9):1151–1156.
186. McLeod RL, Tulshian DB, Bolser DC, Varty GB, Baptista M, Fernandez X, Parra LE, Zim-
mer JC, Erickson CH, Ho GD, Jia Y, Ng FW, Korfmacher W, Xu X, Veals J, Smith-Torhan A,
Wainhaus S, Fawzi AB, Austin TM, van Heek M, Hey JA. Pharmacological profile of the NOP ag-
onist and cough suppressing agent SCH 486757 (8-[Bis(2-Chlorophenyl)Methyl]-3-(2-Pyrimidinyl)-
8-Azabicyclo[3.2.1]Octan-3-Ol) in preclinical models. Eur J Pharmacol 2010;630(1–3):112–120.
187. McLeod RL, Tulshian DB, Sadeh J. Where are the new cough treatments: A debriefing of recent
clinical proof-of-concept trials with the NOP agonist SCH 486757. Pharmacology 2011;88(1–2):50–
54.
188. Trapella C, Guerrini R, Piccagli L, Calo G, Carra G, Spagnolo B, Rubini S, Fanton G, Hebbes C,
McDonald J, Lambert DG, Regoli D, Salvadori S. Identification of an achiral analogue of J-113397
as potent nociceptin/orphanin FQ receptor antagonist. Bioorg Med Chem 2006;14(3):692–704.
189. Marti M, Trapella C, Morari M. The novel nociceptin/orphanin FQ receptor antagonist Trap-101
alleviates experimental parkinsonism through inhibition of the nigro-thalamic pathway: Positive
interaction with L-DOPA. J Neurochem 2008;107(6):1683–1696.
190. Thompson AA, Liu W, Chun E, Katritch V, Wu H, Vardy E, Huang XP, Trapella C, Guerrini R,
Calo G, Roth BL, Cherezov V, Stevens RC. Structure of the nociceptin/orphanin FQ receptor in
complex with a peptide mimetic. Nature 2012;485(7398):395–399.
191. Palczewski K, Kumasaka T, Hori T, Behnke CA, Motoshima H, Fox BA, Le Trong I, Teller DC,
Okada T, Stenkamp RE, Yamamoto M, Miyano M. Crystal structure of rhodopsin: A G protein-
coupled receptor. Science 2000;289(5480):739–745.
192. Rasmussen SG, Choi HJ, Rosenbaum DM, Kobilka TS, Thian FS, Edwards PC, Burghammer M,
Ratnala VR, Sanishvili R, Fischetti RF, Schertler GF, Weis WI, Kobilka BK. Crystal structure of
the human beta2 adrenergic G-protein-coupled receptor. Nature 2007;450(7168):383–387.

Medicinal Research Reviews DOI 10.1002/med


42 r HALLBERG
193. Rasmussen SG, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, Thian FS, Chae PS, Pardon
E, Calinski D, Mathiesen JM, Shah ST, Lyons JA, Caffrey M, Gellman SH, Steyaert J, Skiniotis G,
Weis WI, Sunahara RK, Kobilka BK. Crystal structure of the beta2 adrenergic receptor-Gs protein
complex. Nature 2011;477(7366):549–555.
194. Khosla MC, Leese RA, Maloy WL, Ferreira AT, Smeby RR, Bumpus FM. Synthesis of some
analogs of angiotensin II as specific antagonists of the parent hormone. J Med Chem 1972;15(8):792–
795.
195. Haber E, George C. Griffith lecture. The role of renin in normal and pathological cardiovascular
homeostasis. Circulation 1976;54(6):849–861.
196. Brunner HR, Gavras H, Laragh JH. Angiotensin-II blockade in man by sar1-ala8-angiotensin II
for understanding and treatment of high blood-pressure. Lancet 1973;2(7837):1045–1048.
197. Streeten DH, Anderson GH, Jr., Dalakos TH. Angiotensin blockade: Its clinical significance. Am
J Med 1976;60(6):817–824.
198. Ogihara T, Yamamoto T, Kumahara Y. Clinical applications of synthetic angiotensin II analogue.
Jpn Circ J 1974;38(11):997–1003.
199. Ondetti MA, Rubin B, Cushman DW. Design of specific inhibitors of angiotensin-converting en-
zyme: New class of orally active antihypertensive agents. Science 1977;196(4288):441–444.
200. Ondetti MA, Cushman DW. Inhibition of the renin-angiotensin system. A new approach to the
therapy of hypertension. J Med Chem 1981;24(4):355–361.
201. Azizi M, Webb R, Nussberger J, Hollenberg NK. Renin inhibition with aliskiren: Where are we
now, and where are we going? J Hypertens 2006;24(2):243–256.
202. Ibrahim MM. RAS inhibition in hypertension. J Hum Hypertens 2006;20(2):101–108.
203. Werner C, Baumhakel M, Teo KK, Schmieder R, Mann J, Unger T, Yusuf S, Bohm M. RAS
blockade with ARB and ACE inhibitors: Current perspective on rationale and patient selection.
Clin Res Cardiol 2008;97(7):418–431.
204. Valentova M, von Haehling S. An overview of recent developments in the treatment of heart failure:
Update from the ESC Congress 2013. Expert Opin Investig Drugs 2014.
205. Wexler RR, Greenlee WJ, Irvin JD, Goldberg MR, Prendergast K, Smith RD, Timmermans PB.
Nonpeptide angiotensin II receptor antagonists: The next generation in antihypertensive therapy. J
Med Chem 1996;39(3):625–656.
206. Paulis L, Steckelings UM, Unger T. Key advances in antihypertensive treatment. Nat Rev Cardiol
2012;9(5):276–285.
207. Kirkby NS, Hadoke PW, Bagnall AJ, Webb DJ. The endothelin system as a therapeutic target in
cardiovascular disease: Great expectations or bleak house? Br J Pharmacol 2008;153(6):1105–1119.
208. Ruilope LM, Dukat A, Bohm M, Lacourciere Y, Gong J, Lefkowitz MP. Blood-pressure reduction
with LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor and neprilysin: A ran-
domised, double-blind, placebo-controlled, active comparator study. Lancet 2010;375(9722):1255–
1266.
209. Kivlighn SD, Huckle WR, Zingaro GJ, Rivero RA, Lotti VJ, Chang RS, Schorn TW, Kevin N,
Johnson RG, Jr., Greenlee WJ, Siegl PKS. Discovery of L-162,313: A nonpeptide that mimics the
biological actions of angiotensin II. Am J Physiol 1995;268(3 Pt 2):R820–R823.
210. Paulis L, Steckelings UM, Unger T. Key advances in antihypertensive treatment. Nat Rev Cardiol
2011;9(5):276–285.
211. Paulis L, Unger T. Novel therapeutic targets for hypertension. Nat Rev Cardiol 2010;7(8):431–441.
212. Gallo-Payet N, Shum M, Baillargeon JP, Langlois MF, Wallinder C, Alterman M, Hallberg A,
Carpentier AC. AT2 receptor agonists: Exploiting the beneficial arm of Ang II signaling. Curr
Hypertens Rev 2012;8(1):47–59.
213. Yamada H, Akishita M, Ito M, Tamura K, Daviet L, Lehtonen JY, Dzau VJ, Horiuchi M. AT2
receptor and vascular smooth muscle cell differentiation in vascular development. Hypertension
1999;33(6):1414–1419.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 43
214. Lenkei Z, Palkovits M, Corvol P, Llorens-Cortes C. Expression of angiotensin type-1 (AT1) and
type-2 (AT2) receptor mRNAs in the adult rat brain: A functional neuroanatomical review. Front
Neuroendocrinol 1997;18(4):383–439.
215. Horiuchi M, Mogi M, Iwai M. The angiotensin II type 2 receptor in the brain. J Renin Angiotensin
Aldosterone Syst 2010;11(1):1–6.
216. Meffert S, Stoll M, Steckelings UM, Bottari SP, Unger T. The angiotensin II AT2 receptor inhibits
proliferation and promotes differentiation in PC12W cells. Mol Cell Endocrinol 1996;122(1):59–67.
217. Laflamme L, Gasparo M, Gallo JM, Payet MD, Gallo-Payet N. Angiotensin II induction of neurite
outgrowth by AT2 receptors in NG108- 15 cells. Effect counteracted by the AT1 receptors. J Biol
Chem 1996;271(37):22729–22735.
218. Lucius R, Gallinat S, Rosenstiel P, Herdegen T, Sievers J, Unger T. The angiotensin II type 2 (AT2)
receptor promotes axonal regeneration in the optic nerve of adult rats. J Exp Med 1998;188(4):661–
670.
219. Steckelings UM, Kaschina E, Unger T. The AT2 receptor–a matter of love and hate. Peptides
2005;26(8):1401–1409.
220. Jones ES, Vinh A, McCarthy CA, Gaspari TA, Widdop RE. AT2 receptors: Functional relevance
in cardiovascular disease. Pharmacol Ther 2008;120(3):292–316.
221. McCarthy CA, Widdop RE, Denton KM, Jones ES. Update on the angiotensin AT(2) receptor.
Curr Hypertens Rep 2013;15(1):25–30.
222. McCarthy CA, Vinh A, Broughton BR, Sobey CG, Callaway JK, Widdop RE. Angiotensin II type
2 receptor stimulation initiated after stroke causes neuroprotection in conscious rats. Hypertension
2012;60(6):1531–1537.
223. de Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T. International union of pharmacology.
XXIII. The angiotensin II receptors. Pharmacol Rev 2000;52(3):415–472.
224. Carey RM, Wang ZQ, Siragy HM. Role of the angiotensin type 2 receptor in the regulation of blood
pressure and renal function. Hypertension 2000;35(1 Pt 2):155–163.
225. Steckelings UM, Rompe F, Kaschina E, Namsolleck P, Grzesiak A, Funke-Kaiser H, Bader M,
Unger T. The past, present and future of angiotensin II type 2 receptor stimulation. J Renin
Angiotensin Aldosterone Syst 2010;11(1):67–73.
226. Carey RM. Angiotensin type-2 receptors and cardiovascular function: Are angiotensin type-2 re-
ceptors protective? Curr Opin Cardiol 2005;20(4):264–269.
227. Savoia C, Touyz RM, Volpe M, Schiffrin EL. Angiotensin type 2 receptor in resistance arteries of
type 2 diabetic hypertensive patients. Hypertension 2007;49(2):341–346.
228. Siragy HM. The angiotensin II type 2 receptor and the kidney. J Renin Angiotensin Aldosterone
Syst 2010;11(1):33–36.
229. Unger T, Jakobsen A, Heroys J, Ralph A, Rees T, Shaw M. Targeting cardiovascular protection:
The concept of dual renin-angiotensin system control. Medscape J Med 2008;10(Suppl:S4).
230. Lemarie CA, Schiffrin EL. The angiotensin II type 2 receptor in cardiovascular disease. J Renin
Angiotensin Aldosterone Syst 2010;11(1):19–31.
231. Rompe F, Artuc M, Hallberg A, Alterman M, Stroder K, Thone-Reineke C, Reichenbach A,
Schacherl J, Dahlof B, Bader M, Alenina N, Schwaninger M, Zuberbier T, Funke-Kaiser H, Schmidt
C, Schunck WH, Unger T, Steckelings UM. Direct angiotensin II type 2 receptor stimulation
acts anti-inflammatory through epoxyeicosatrienoic acid and inhibition of nuclear factor kappaB.
Hypertension 2010;55(4):924–931.
232. Wan Y, Wallinder C, Plouffe B, Beaudry H, Mahalingam AK, Wu X, Johansson B, Holm M,
Botoros M, Karlen A, Pettersson A, Nyberg F, Fandriks L, Gallo-Payet N, Hallberg A, Alterman
M. Design, synthesis, and biological evaluation of the first selective nonpeptide AT2 receptor agonist.
J Med Chem 2004;47(24):5995–6008.
233. Foulquier S, Steckelings UM, Unger T. Impact of the AT(2) receptor agonist C21 on blood pressure
and beyond. Curr Hypertens Rep 2012;14(5):403–409.

Medicinal Research Reviews DOI 10.1002/med


44 r HALLBERG
234. Steckelings UM, Unger T. Angiotensin II type 2 receptor agonists–where should they be applied?
Expert Opin Investig Drugs 2012;21(6):763–766.
235. Foulquier S, Steckelings UM, Unger T. Perspective: A tale of two receptors. Nature 2013;
493(7434):S9.
236. Anand U, Facer P, Yiangou Y, Sinisi M, Fox M, McCarthy T, Bountra C, Korchev YE, Anand P.
Angiotensin II type 2 receptor (AT2 R) localization and antagonist-mediated inhibition of capsaicin
responses and neurite outgrowth in human and rat sensory neurons. Eur J Pain 2013;17(7):1012–
1026.
237. Smith MT, Wyse BD, Edwards SR. Small molecule angiotensin II type 2 receptor (AT(2)R) antago-
nists as novel analgesics for neuropathic pain: Comparative pharmacokinetics, radioligand binding,
and efficacy in rats. Pain Med 2013;14(5):692–705.
238. Rice AS, Dworkin RH, McCarthy TD, Anand P, Bountra C, McCloud PI, Hill J, Cutter G,
Kitson G, Desem N, Raff M. EMA401, an orally administered highly selective angiotensin II type
2 receptor antagonist, as a novel treatment for postherpetic neuralgia: A randomised, double-blind,
placebo-controlled phase 2 clinical trial. Lancet 2014;383((9929)):1637–1647.
239. Nguyen G, Delarue F, Burckle C, Bouzhir L, Giller T, Sraer JD. Pivotal role of the renin/prorenin re-
ceptor in angiotensin II production and cellular responses to renin. J Clin Invest 2002;109(11):1417–
1427.
240. Nguyen G. Renin, (pro)renin and receptor: An update. Clin Sci (Lond) 2011;120(5):169–178.
241. Johnston CI, Risvanis J. Preclinical pharmacology of angiotensin II receptor antagonists: Update
and outstanding issues. Am J Hypertens 1997;10(12 Pt 2):306S–310S.
242. Lyons PJ, Callaway MB, Fricker LD. Characterization of carboxypeptidase A6, an extracellular
matrix peptidase. J Biol Chem 2008;283(11):7054–7063.
243. Ferrario CM, Ahmad S, Nagata S, Simington SW, Varagic J, Kon N, Dell’italia LJ. An evolving
story of angiotensin-II-forming pathways in rodents and humans. Clin Sci (Lond) 2014;126(7):461–
469.
244. Chitravanshi VC, Proddutur A, Sapru HN. Cardiovascular actions of angiotensin-(1–12) in the
hypothalamic paraventricular nucleus of the rat are mediated via angiotensin II. Exp Physiol
2012;97(9):1001–1017.
245. Arakawa H, Kawabe K, Sapru HN. Angiotensin-(1–12) in the rostral ventrolateral medullary pressor
area of the rat elicits sympathoexcitatory responses. Exp Physiol 2013;98(1):94–108.
246. Fyhrquist F, Saijonmaa O. Renin-angiotensin system revisited. J Intern Med 2008;264(3):224–236.
247. Blair-West JR, Carey KD, Denton DA, Madden LJ, Weisinger RS, Shade RE. Possible contribution
of brain angiotensin III to ingestive behaviors in baboons. Am J Physiol Regul Integr Comp Physiol
2001;281(5):R1633–R1636.
248. Wright JW, Miller-Wing AV, Shaffer MJ, Higginson C, Wright DE, Hanesworth JM, Harding JW.
Angiotensin II(3–8) (ANG IV) hippocampal binding: Potential role in the facilitation of memory.
Brain Res Bull 1993;32(5):497–502.
249. Lee J, Chai SY, Mendelsohn FA, Morris MJ, Allen AM. Potentiation of cholinergic trans-
mission in the rat hippocampus by angiotensin IV and LVV-hemorphin-7. Neuropharmacology
2001;40(4):618–623.
250. Cesari M, Rossi GP, Pessina AC. Biological properties of the angiotensin peptides other than an-
giotensin II: Implications for hypertension and cardiovascular diseases. J Hypertens 2002;20(5):793–
799.
251. Hall KL, Venkateswaran S, Hanesworth JM, Schelling ME, Harding JW. Characterization of
a functional angiotensin IV receptor on coronary microvascular endothelial cells. Regul Pept
1995;58(3):107–115.
252. Wright JW, Harding JW. Important role for angiotensin III and IV in the brain renin- angiotensin
system. Brain Res Brain Res Rev 1997;25(1):96–124.
253. Pederson ES, Krishnan R, Harding JW, Wright JW. A role for the angiotensin AT4 receptor subtype
in overcoming scopolamine-induced spatial memory deficits. Regul Pept 2001;102(2–3):147–156.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 45
254. De Bundel D, Smolders I, Yang R, Albiston AL, Michotte Y, Chai SY. Angiotensin IV and LVV-
haemorphin 7 enhance spatial working memory in rats: Effects on hippocampal glucose levels and
blood flow. Neurobiol Learn Mem 2009;92(1):19–26.
255. Ardaillou R, Chansel D. Synthesis and effects of active fragments of angiotensin II. Kidney Int
1997;52(6):1458–1468.
256. Jankowski V, Vanholder R, van der Giet M, Tolle M, Karadogan S, Gobom J, Furkert J, Oksche
A, Krause E, Tran TN, Tepel M, Schuchardt M, Schluter H, Wiedon A, Beyermann M, Bader M,
Todiras M, Zidek W, Jankowski J. Mass-spectrometric identification of a novel angiotensin peptide
in human plasma. Arterioscler Thromb Vasc Biol 2007;27(2):297–302.
257. Yang R, Smolders I, Vanderheyden P, Demaegdt H, Van Eeckhaut A, Vauquelin G, Lukaszuk A,
Tourwe D, Chai SY, Albiston AL, Nahmias C, Walther T, Dupont AG. Pressor and renal hemo-
dynamic effects of the novel angiotensin A peptide are angiotensin II type 1A receptor dependent.
Hypertension 2011;57(5):956–964.
258. Lautner RQ, Villela DC, Fraga-Silva RA, Silva N, Verano-Braga T, Costa-Fraga F, Jankowski
J, Jankowski V, Sousa F, Alzamora A, Soares E, Barbosa C, Kjeldsen F, Oliveira A, Braga J,
Savergnini S, Maia G, Peluso AB, Passos-Silva D, Ferreira A, Alves F, Martins A, Raizada M,
Paula R, Motta-Santos D, Kemplin F, Pimenta A, Alenina N, Sinisterra R, Bader M, Campagnole-
Santos MJ, Santos RA. Discovery and characterization of alamandine: A novel component of the
renin-angiotensin system. Circ Res 2013;112(8):1104–1111.
259. Banegas I, Prieto I, Vives F, Alba F, de Gasparo M, Segarra AB, Hermoso F, Duran R, Ramirez M.
Brain aminopeptidases and hypertension. J Renin Angiotensin Aldosterone Syst 2006;7(3):129–134.
260. Reudelhuber TL. The renin-angiotensin system: Peptides and enzymes beyond angiotensin II. Cur-
rent Opin Nephrol Hypertens 2005;14(2):155–159.
261. Saavedra JM. Brain and pituitary angiotensin. Endocrine Rev 1992;13(2):329–380.
262. Speth RC, Brown TE, Barnes RD, Wright JW. Brain angiotensinergic activity: The state of our
current knowledge. Proc West Pharmacol Soc 2003;46:11–15.
263. Unger T, Badoer E, Ganten D, Lang RE, Rettig R. Brain angiotensin: Pathways and pharmacology.
Circulation 1988;77(6 Pt 2):I40–I54.
264. Johnston CI. Biochemistry and pharmacology of the renin-angiotensin system. Drugs 1990;39(Suppl
1):21–31.
265. Chappell MC, Tallant EA, Brosnihan KB, Ferrario CM. Processing of angiotensin peptides by
NG108–15 neuroblastoma x glioma hybrid cell line. Peptides 1990;11(2):375–380.
266. Greene LJ, Spadaro AC, Martins AR, Perussi De Jesus WD, Camargo AC. Brain endo-
oligopeptidase B: A post-proline cleaving enzyme that inactivates angiotensin I and II. Hypertension
1982;4(2):178–184.
267. Ferrario CM, Chappell MC. Novel angiotensin peptides. Cell Mol Life Sci 2004;61(21):2720–2727.
268. Yamamoto K, Chappell MC, Brosnihan KB, Ferrario CM. In vivo metabolism of angiotensin I by
neutral endopeptidase (EC 3.4.24.11) in spontaneously hypertensive rats. Hypertension 1992;19(6
Pt 2):692–696.
269. Vauquelin G, Michotte Y, Smolders I, Sarre S, Ebinger G, Dupont A, Vanderheyden P. Cellular
targets for angiotensin II fragments: Pharmacological and molecular evidence. J Renin Angiotensin
Aldosterone Syst 2002;3(4):195–204.
270. Mentlein R, Roos T. Proteases involved in the metabolism of angiotensin II, bradykinin, calci-
tonin gene-related peptide (CGRP), and neuropeptide Y by vascular smooth muscle cells. Peptides
1996;17(4):709–720.
271. Ryan JW. Peptidase enzymes of the pulmonary vascular surface. Am J Physiol 1989;257(2 Pt
1):L53–L60.
272. Ahmad S, Ward PE. Role of aminopeptidase activity in the regulation of the pressor activity of
circulating angiotensins. J Pharmacol Exp Ther 1990;252(2):643–650.

Medicinal Research Reviews DOI 10.1002/med


46 r HALLBERG
273. Robertson MJ, Cunoosamy MP, Clark KL. Effects of peptidase inhibition on angiotensin receptor
agonist and antagonist potency in rabbit isolated thoracic aorta. Br J Pharmacol 1992;106(1):166–
172.
274. Vaghy PL, Russell JS, Lantry LE, Stephens RE, Ward PE. Angiotensin and bradykinin metabolism
by peptidases identified in cultured human skeletal muscle myocytes and fibroblasts. Peptides
1995;16(8):1367–1373.
275. Petrov VV, Fagard RH, Lijnen PJ. Arginine-aminopeptidase in rat cardiac fibroblastic cells partic-
ipates in angiotensin peptide turnover. Cardiovasc Res 2004;61(4):724–735.
276. Jackman HL, Massad MG, Sekosan M, Tan F, Brovkovych V, Marcic BM, Erdos EG. Angiotensin
1–9 and 1–7 release in human heart: Role of cathepsin A. Hypertension 2002;39(5):976–981.
277. Flores-Munoz M, Godinho BM, Almalik A, Nicklin SA. Adenoviral delivery of angiotensin-(1–7)
or angiotensin-(1–9) inhibits cardiomyocyte hypertrophy via the mas or angiotensin type 2 receptor.
PLoS One 2012;7(9):e45564.
278. Flores-Munoz M, Smith NJ, Haggerty C, Milligan G, Nicklin SA. Angiotensin1–9 antagonises pro-
hypertrophic signalling in cardiomyocytes via the angiotensin type 2 receptor. J Physiol 2011;589(Pt
4):939–951.
279. Flores-Munoz M, Work LM, Douglas K, Denby L, Dominiczak AF, Graham D, Nicklin SA.
Angiotensin-(1–9) attenuates cardiac fibrosis in the stroke-prone spontaneously hypertensive rat via
the angiotensin type 2 receptor. Hypertension 2012;59(2):300–307.
280. Ferreira AJ, Bader M, Santos RA. Therapeutic targeting of the angiotensin-converting enzyme
2/Angiotensin-(1–7)/Mas cascade in the renin-angiotensin system: A patent review. Expert Opin
Ther Pat 2012;22(5):567–574.
281. Santos RA, Ferreira AJ, Verano-Braga T, Bader M. Angiotensin-converting enzyme 2, angiotensin-
(1–7) and Mas: New players of the renin-angiotensin system. J Endocrinol 2013;216(2):R1–R17.
282. Santos RA, Ferreira AJ. Simoes ESAC. Recent advances in the angiotensin-converting enzyme
2-angiotensin(1–7)-Mas axis. Exp Physiol 2008;93(5):519–527.
283. Grobe JL, Mecca AP, Lingis M, Shenoy V, Bolton TA, Machado JM, Speth RC, Raizada MK,
Katovich MJ. Prevention of angiotensin II-induced cardiac remodeling by angiotensin-(1–7). Am J
Physiol Heart Circ Physiol 2007;292(2):H736–H742.
284. Gurley SB, Allred A, Le TH, Griffiths R, Mao L, Philip N, Haystead TA, Donoghue M, Breitbart
RE, Acton SL, Rockman HA, Coffman TM. Altered blood pressure responses and normal cardiac
phenotype in ACE2-null mice. J Clin Invest 2006;116(8):2218–2225.
285. Reaux A, Fournie-Zaluski MC, Llorens-Cortes C. Angiotensin III: A central regulator of vaso-
pressin release and blood pressure. Trends Endocrinol Metab 2001;12(4):157–162.
286. Yugandhar VG, Clark MA. Angiotensin III: A physiological relevant peptide of the renin an-
giotensin system. Peptides 2013;46:26–32.
287. Bosnyak S, Jones ES, Christopoulos A, Aguilar MI, Thomas WG, Widdop RE. Relative affinity of
angiotensin peptides and novel ligands at AT1 and AT2 receptors. Clin Sci (Lond) 2011;121(7):297–
303.
288. Braszko JJ, Kupryszewski G, Witczuk B, Wisniewski K. Angiotensin II-(3–8)-hexapeptide affects
motor activity, performance of passive avoidance and a conditioned avoidance response in rats.
Neuroscience 1988;27(3):777–783.
289. Lee J, Albiston AL, Allen AM, Mendelsohn FA, Ping SE, Barrett GL, Murphy M, Morris MJ,
McDowall SG, Chai SY. Effect of I.C.V. injection of AT4 receptor ligands, NLE1-angiotensin IV
and LVV-hemorphin 7, on spatial learning in rats. Neuroscience 2004;124(2):341–349.
290. Wright JW, Clemens JA, Panetta JA, Smalstig EB, Weatherly LA, Kramar EA, Pederson ES,
Mungall BH, Harding JW. Effects of LY231617 and angiotensin IV on ischemia-induced deficits in
circular water maze and passive avoidance performance in rats. Brain Res 1996;717(1–2):1–11.
291. Wright JW, Stubley L, Pederson ES, Kramar EA, Hanesworth JM, Harding JW. Contribu-
tions of the brain angiotensin IV-AT4 receptor subtype system to spatial learning. J Neurosci
1999;19(10):3952–3961.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 47
292. Braszko JJ, Wielgat P, Walesiuk A. Effect of D(3) dopamine receptors blockade on the cognitive
effects of angiotensin IV in rats. Neuropeptides 2008;42(3):301–309.
293. Gard PR, Naylor C, Ali S, Partington C. Blockade of pro-cognitive effects of angiotensin IV and
physostigmine in mice by oxytocin antagonism. Eur J Pharmacol 2012;683(1–3):155–160.
294. Pham V, Albiston AL, Downes CE, Wong CH, Diwakarla S, Ng L, Lee S, Crack PJ, Chai SY.
Insulin-regulated aminopeptidase deficiency provides protection against ischemic stroke in mice. J
Neurotrauma 2012;29(6):1243–1248.
295. Faure S, Chapot R, Tallet D, Javellaud J, Achard JM, Oudart N. Cerebroprotective effect of
angiotensin IV in experimental ischemic stroke in the rat mediated by AT(4) receptors. J Physiol
Pharmacol 2006;57(3):329–342.
296. Wong YC, Sim MK, Lee KO. Des-aspartate-angiotensin-I and angiotensin IV improve glu-
cose tolerance and insulin signalling in diet-induced hyperglycaemic mice. Biochem Pharmacol
2011;82(9):1198–1208.
297. Chai SY, Yeatman HR, Parker MW, Ascher DB, Thompson PE, Mulvey HT, Albiston AL. De-
velopment of cognitive enhancers based on inhibition of insulin-regulated aminopeptidase. BMC
Neurosci 2008;9(Suppl 2):S14.
298. Albiston AL, Diwakarla S, Fernando RN, Mountford SJ, Yeatman HR, Morgan B, Pham V,
Holien JK, Parker MW, Thompson PE, Chai SY. Identification and development of specific in-
hibitors for insulin-regulated aminopeptidase as a new class of cognitive enhancers. Br J Pharmacol
2011;164(1):37–47.
299. Wright JW, Harding JW. Brain renin-angiotensin–a new look at an old system. Prog Neurobiol
2011;95(1):49–67.
300. Wright JW, Mizutani S, Harding JW. Focus on Brain Angiotensin III and Aminopeptidase A in
the Control of Hypertension. Int J Hypertens 2012;2012:124758.
301. Pinheiro SV, Simoes e Silva AC, Sampaio WO, de Paula RD, Mendes EP, Bontempo ED, Pesquero
JB, Walther T, Alenina N, Bader M, Bleich M, Santos RA. Nonpeptide AVE 0991 is an angiotensin-
(1–7) receptor Mas agonist in the mouse kidney. Hypertension 2004;44(4):490–496.
302. Ferreira AJ, Murca TM, Fraga-Silva RA, Castro CH, Raizada MK, Santos RA. New cardiovascular
and pulmonary therapeutic strategies based on the Angiotensin-converting enzyme 2/angiotensin-
(1–7)/mas receptor axis. Int J Hypertens 2012;2012:147825.
303. Santos RA, Campagnole-Santos MJ, Andrade SP. Angiotensin-(1–7): An update. Regul Pept
2000;91(1–3):45–62.
304. Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP, de Buhr I, Heringer-Walther S,
Pinheiro SV, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss
HP, Speth R, Walther T. Angiotensin-(1–7) is an endogenous ligand for the G protein-coupled
receptor Mas. Proc Natl Acad Sci USA 2003;100(14):8258–8263.
305. Grobe JL, Der Sarkissian S, Stewart JM, Meszaros JG, Raizada MK, Katovich MJ. ACE2 over-
expression inhibits hypoxia-induced collagen production by cardiac fibroblasts. Clin Sci (Lond)
2007;113(8):357–364.
306. Santos RA, Castro CH, Gava E, Pinheiro SV, Almeida AP, Paula RD, Cruz JS, Ramos AS, Rosa
KT, Irigoyen MC, Bader M, Alenina N, Kitten GT, Ferreira AJ. Impairment of in vitro and in vivo
heart function in angiotensin-(1–7) receptor MAS knockout mice. Hypertension 2006;47(5):996–
1002.
307. Yamada K, Iyer SN, Chappell MC, Ganten D, Ferrario CM. Converting enzyme determines plasma
clearance of angiotensin-(1–7). Hypertension 1998;32(3):496–502.
308. Wiemer G, Dobrucki LW, Louka FR, Malinski T, Heitsch H. AVE 0991, a nonpeptide mimic of
the effects of angiotensin-(1–7) on the endothelium. Hypertension 2002;40(6):847–852.
309. Benter IF, Yousif MH, Cojocel C, Al-Maghrebi M, Diz DI. Angiotensin-(1–7) prevents diabetes-
induced cardiovascular dysfunction. Am J Physiol Heart Circ Physiol 2007;292(1):H666–H672.
310. Singh K, Singh T, Sharma PL. Beneficial effects of angiotensin (1–7) in diabetic rats with cardiomy-
opathy. Ther Adv Cardiovasc Dis 2011;5(3):159–167.

Medicinal Research Reviews DOI 10.1002/med


48 r HALLBERG
311. Ferreira AJ, Jacoby BA, Araujo CA, Macedo FA, Silva GA, Almeida AP, Caliari MV, Santos RA.
The nonpeptide angiotensin-(1–7) receptor Mas agonist AVE-0991 attenuates heart failure induced
by myocardial infarction. Am J Physiol Heart Circ Physiol 2007;292(2):H1113–H1119.
312. Ebermann L, Spillmann F, Sidiropoulos M, Escher F, Heringer-Walther S, Schultheiss HP, Tschope
C, Walther T. The angiotensin-(1–7) receptor agonist AVE0991 is cardioprotective in diabetic rats.
Eur J Pharmacol 2008;590(1–3):276–280.
313. Lubel JS, Herath CB, Tchongue J, Grace J, Jia Z, Spencer K, Casley D, Crowley P, Sievert W, Burrell
LM, Angus PW. Angiotensin-(1–7), an alternative metabolite of the renin-angiotensin system, is
up-regulated in human liver disease and has antifibrotic activity in the bile-duct-ligated rat. Clin Sci
(Lond) 2009;117(11):375–386.
314. da Costa Goncalves AC, Fraga-Silva RA, Leite R, Santos RA. AVE 0991, a non-peptide Mas-
receptor agonist, facilitates penile erection. Exp Physiol 2013;98(3):850–855.
315. Rodrigues-Machado MG, Magalhaes GS, Cardoso JA, Kangussu LM, Murari A, Caliari MV,
Oliveira ML, Cara DC, Noviello ML, Marques FD, Pereira JM, Lautner RQ, Santos RA,
Campagnole-Santos MJ. AVE 0991, a non-peptide mimic of angiotensin-(1–7) effects, attenuates
pulmonary remodelling in a model of chronic asthma. Br J Pharmacol 2013;170(4):835–846.
316. Hernandez Prada JA, Ferreira AJ, Katovich MJ, Shenoy V, Qi Y, Santos RA, Castellano RK,
Lampkins AJ, Gubala V, Ostrov DA, Raizada MK. Structure-based identification of small-
molecule angiotensin-converting enzyme 2 activators as novel antihypertensive agents. Hypertension
2008;51(5):1312–1317.
317. Fraga-Silva RA, Sorg BS, Wankhede M, Dedeugd C, Jun JY, Baker MB, Li Y, Castellano RK,
Katovich MJ, Raizada MK, Ferreira AJ. ACE2 activation promotes antithrombotic activity. Mol
Med 2010;16(5–6):210–215.
318. Kulemina LV, Ostrov DA. Prediction of off-target effects on angiotensin-converting enzyme 2. J
Biomol Screen 2011;16(8):878–885.
319. Murca TM, Almeida TC, Raizada MK, Ferreira AJ. Chronic activation of endogenous angiotensin-
converting enzyme 2 protects diabetic rats from cardiovascular autonomic dysfunction. Exp Physiol
2012;97(6):699–709.
320. Haber PK, Ye M, Wysocki J, Maier C, Haque SK, Batlle D. Angiotensin-converting enzyme 2-
independent action of presumed angiotensin-converting enzyme 2 activators: Studies in vivo, ex
vivo, and in vitro. Hypertension 2014;63(4):774–782.
321. Swanson GN, Hanesworth JM, Sardinia MF, Coleman JK, Wright JW, Hall KL, Miller-Wing AV,
Stobb JW, Cook VI, Harding EC, Harding JW. Discovery of a distinct binding site for angiotensin
II (3–8), a putative angiotensin IV receptor. Regul Pept 1992;40(3):409–419.
322. Harding JW, Cook VI, Miller-Wing AV, Hanesworth JM, Sardinia MF, Hall KL, Stobb JW,
Swanson GN, Coleman JK, Wright JW, Harding EC. Identification of an AII(3–8) [AIV] binding
site in guinea pig hippocampus. Brain Res 1992;583(1–2):340–343.
323. de Gasparo M, Husain A, Alexander W, Catt KJ, Chiu AT, Drew M, Goodfriend T, Harding JW,
Inagami T, Timmermans PB. Proposed update of angiotensin receptor nomenclature. Hypertension
1995;25(5):924–927.
324. Sardinia MF, Hanesworth JM, Krebs LT, Harding JW. AT4 receptor binding characteristics: D-
amino acid- and glycine-substituted peptides. Peptides 1993;14(5):949–954.
325. Sardinia MF, Hanesworth JM, Krishnan F, Harding JW. AT4 receptor structure-binding relation-
ship: N-terminal-modified angiotensin IV analogues. Peptides 1994;15(8):1399–1406.
326. Krishnan R, Hanesworth JM, Wright JW, Harding JW. Structure-binding studies of the adrenal
AT4 receptor: Analysis of position two- and three-modified angiotensin IV analogs. Peptides
1999;20(8):915–920.
327. Kobori T, Goda K, Sugimoto K, Ota T, Tomisawa K. Preparation of peptide derivatives as an-
giotensin IV receptor agonists. WO 97/03093 A1 1997.
328. Kobori T, Goda K, Sugimoto K, Ota T, Tomisawa K. Preparation of amino acid derivatives as
angiotensin IV receptor agonists. WO 98/05624 A1 1998.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 49
329. Albiston AL, McDowall SG, Matsacos D, Sim P, Clune E, Mustafa T, Lee J, Mendelsohn FA,
Simpson RJ, Connolly LM, Chai SY. Evidence that the angiotensin IV (AT(4)) receptor is the
enzyme insulin- regulated aminopeptidase. J Biol Chem 2001;276(52):48623–48626.
330. De Bundel D, Smolders I, Vanderheyden P, Michotte Y. Ang II and Ang IV: Unraveling the mecha-
nism of action on synaptic plasticity, memory, and epilepsy. CNS Neurosci Ther 2008;14(4):315–339.
331. Tsujimoto M, Mizutani S, Adachi H, Kimura M, Nakazato H, Tomoda Y. Identification of human
placental leucine aminopeptidase as oxytocinase. Arch Biochem Biophys 1992;292(2):388–392.
332. Rogi T, Tsujimoto M, Nakazato H, Mizutani S, Tomoda Y. Human placental leucine aminopep-
tidase/oxytocinase. A new member of type II membrane-spanning zinc metallopeptidase family. J
Biol Chem 1996;271(1):56–61.
333. Rasmussen TE, Pedraza-Diaz S, Hardre R, Laustsen PG, Carrion AG, Kristensen T. Structure
of the human oxytocinase/insulin-regulated aminopeptidase gene and localization to chromosome
5q21. Eur J Biochem 2000;267(8):2297–2306.
334. Wolfe MS. Therapeutic strategies for Alzheimer’s disease. Nature Rev 2002;1(11):859–866.
335. Hallberg M. Targeting the insulin-regulated aminopeptidase/AT4 receptor for cognitive disorders.
Drug News Perspect 2009;22(3):133–139.
336. Gard PR. Cognitive-enhancing effects of angiotensin IV. BMC Neurosci 2008;9(Suppl 2):S15.
337. Wright JW, Harding JW. The angiotensin AT4 receptor subtype as a target for the treatment of
memory dysfunction associated with Alzheimer’s disease. J Renin Angiotensin Aldosterone Syst
2008;9(4):226–237.
338. Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst Rev (Online)
2006;(1):CD005593.
339. Cosman KM, Boyle LL, Porsteinsson AP. Memantine in the treatment of mild-to-moderate
Alzheimer’s disease. Expert Opin Pharmacother 2007;8(2):203–214.
340. Doraiswamy PM, Xiong GL. Pharmacological strategies for the prevention of Alzheimer’s disease.
Expert Opin Pharmacother 2006;7(1):1–10.
341. Raina P, Santaguida P, Ismaila A, Patterson C, Cowan D, Levine M, Booker L, Oremus M.
Effectiveness of cholinesterase inhibitors and memantine for treating dementia: Evidence review for
a clinical practice guideline. Ann Intern Med 2008;148(5):379–397.
342. Axen A, Andersson H, Lindeberg G, Ronnholm H, Kortesmaa J, Demaegdt H, Vauquelin G,
Karlen A, Hallberg M. Small potent ligands to the insulin-regulated aminopeptidase (IRAP)/AT(4)
receptor. J Pept Sci 2007;13(7):434–444.
343. Axen A, Lindeberg G, Demaegdt H, Vauquelin G, Karlen A, Hallberg M. Cyclic insulin-regulated
aminopeptidase (IRAP)/AT4 receptor ligands. J Pept Sci 2006;12(11):705–713.
344. Wallis MG, Lankford MF, Keller SR. Vasopressin is a physiological substrate for the insulin-
regulated aminopeptidase IRAP. American J Physiol 2007;293(4):E1092–E1102.
345. Andersson H, Demaegdt H, Vauquelin G, Lindeberg G, Karlen A, Hallberg M, Erdelyi M, Hallberg
A. Disulfide cyclized tripeptide analogues of angiotensin IV as potent and selective inhibitors of
insulin-regulated aminopeptidase (IRAP). J Med Chem 2010;53(22):8059–8071.
346. Andersson H, Demaegdt H, Johnsson A, Vauquelin G, Lindeberg G, Hallberg M, Erdelyi M, Karlen
A, Hallberg A. Potent macrocyclic inhibitors of insulin-regulated aminopeptidase (IRAP) by olefin
ring-closing metathesis. J Med Chem 2011;54(11):3779–3792.
347. Andersson H, Hallberg M. Discovery of inhibitors of insulin-regulated aminopeptidase as cognitive
enhancers. Int J Hypertens 2012;2012:789671.
348. Thunnissen MM, Nordlund P, Haeggstrom JZ. Crystal structure of human leukotriene A(4) hydro-
lase, a bifunctional enzyme in inflammation. Nat Struct Biol 2001;8(2):131–135.
349. Albiston AL, Morton CJ, Ng HL, Pham V, Yeatman HR, Ye S, Fernando RN, De Bundel D, Ascher
DB, Mendelsohn FA, Parker MW, Chai SY. Identification and characterization of a new cognitive
enhancer based on inhibition of insulin-regulated aminopeptidase. FASEB J 2008;22(12):4209–4217.

Medicinal Research Reviews DOI 10.1002/med


50 r HALLBERG
350. Mountford SJ, Albiston AL, Charman WN, Ng L, Holien JK, Parker MW, Nicolazzo JA,
Thompson PE, Chai SY. Synthesis, structure-activity relationships and brain uptake of a novel
series of benzopyran inhibitors of insulin-regulated aminopeptidase. J Med Chem 2014;57(4):1368–
1377.
351. Wright JW, Yamamoto BJ, Harding JW. Angiotensin receptor subtype mediated physiologies and
behaviors: New discoveries and clinical targets. Prog Neurobiol 2008;84(2):157–181.
352. Yamamoto BJ, Elias PD, Masino JA, Hudson BD, McCoy AT, Anderson ZJ, Varnum MD, Sardinia
MF, Wright JW, Harding JW. The angiotensin IV analog Nle-Tyr-Leu-psi-(CH2-NH2)3–4-His-
Pro-Phe (norleual) can act as a hepatocyte growth factor/c-Met inhibitor. J Pharmacol Exp Ther
2010;333(1):161–173.
353. Couture R, Harrisson M, Vianna RM, Cloutier F. Kinin receptors in pain and inflammation. Eur
J Pharmacol 2001;429(1–3):161–176.
354. Regoli D, Barabe J. Pharmacology of bradykinin and related kinins. Pharmacol Rev 1980;32(1):1–
46.
355. Hall JM. Bradykinin receptors: Pharmacological properties and biological roles. Pharmacol Ther
1992;56(2):131–190.
356. Leeb-Lundberg LM, Marceau F, Muller-Esterl W, Pettibone DJ, Zuraw BL. International union
of pharmacology. XLV. Classification of the kinin receptor family: From molecular mechanisms to
pathophysiological consequences. Pharmacol Rev 2005;57(1):27–77.
357. McEachern AE, Shelton ER, Bhakta S, Obernolte R, Bach C, Zuppan P, Fujisaki J, Aldrich
RW, Jarnagin K. Expression cloning of a rat B2 bradykinin receptor. Proc Natl Acad Sci USA
1991;88(17):7724–7728.
358. Hess JF, Borkowski JA, Young GS, Strader CD, Ransom RW. Cloning and pharmacological charac-
terization of a human bradykinin (BK- 2) receptor. Biochem Biophys Res Commun 1992;184(1):260–
268.
359. Menke JG, Borkowski JA, Bierilo KK, MacNeil T, Derrick AW, Schneck KA, Ransom RW, Strader
CD, Linemeyer DL, Hess JF. Expression cloning of a human B1 bradykinin receptor. J Biol Chem
1994;269(34):21583–21586.
360. Hess JF, Hey PJ, Chen TB, O’Brien J, Omalley SS, Pettibone DJ, Chang RS. Molecular cloning
and pharmacological characterization of the canine B1 and B2 bradykinin receptors. Biol Chem
2001;382(1):123–129.
361. Duchene J, Lecomte F, Ahmed S, Cayla C, Pesquero J, Bader M, Perretti M, Ahluwalia A. A novel
inflammatory pathway involved in leukocyte recruitment: Role for the kinin B1 receptor and the
chemokine CXCL5. J Immunol 2007;179(7):4849–4856.
362. Calixto JB, Medeiros R, Fernandes ES, Ferreira J, Cabrini DA, Campos MM. Kinin B1 recep-
tors: Key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J
Pharmacol 2004;143(7):803–818.
363. Heitsch H. The therapeutic potential of bradykinin B2 receptor agonists in the treatment of cardio-
vascular disease. Expert Opin Investig Drugs 2003;12(5):759–770.
364. Campbell DJ. The kallikrein-kinin system in humans. Clin Exp Pharmacol Physiol 2001;
28(12):1060–1065.
365. Erdos EG. Kinins, the long march–a personal view. Cardiovasc Res 2002;54(3):485–491.
366. Kariya K, Yamauchi A, Sasaki T. Regional distribution and characterization of kinin in the CNS
of the rat. J Neurochem 1985;44(6):1892–1897.
367. Chao J, Chao L, Swain CC, Tsai J, Margolius HS. Tissue kallikrein in rat brain and pituitary: Re-
gional distribution and estrogen induction in the anterior pituitary. Endocrinology 1987;120(2):475–
482.
368. Campbell DJ, Kladis A, Duncan AM. Bradykinin peptides in kidney, blood, and other tissues of
the rat. Hypertension 1993;21(2):155–165.
369. Proud D, Kaplan AP. Kinin formation: Mechanisms and role in inflammatory disorders. Annu Rev
Immunol 1988;6:49–83.

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 51
370. Rosenbaum C, Cardozo C, Lesser M. Degradation of lysylbradykinin by endopeptidase 24.11 and
endopeptidase 24.15. Peptides 1995;16(3):523–525.
371. Dendorfer A, Wolfrum S, Wagemann M, Qadri F, Dominiak P. Pathways of bradykinin degradation
in blood and plasma of normotensive and hypertensive rats. Am J Physiol Heart Circ Physiol
2001;280(5):H2182–H2188.
372. Kuoppala A, Lindstedt KA, Saarinen J, Kovanen PT, Kokkonen JO. Inactivation of bradykinin by
angiotensin-converting enzyme and by carboxypeptidase N in human plasma. Am J Physiol Heart
Circ Physiol 2000;278(4):H1069–H1074.
373. Kokkonen JO, Kuoppala A, Saarinen J, Lindstedt KA, Kovanen PT. Kallidin- and bradykinin-
degrading pathways in human heart: Degradation of kallidin by aminopeptidase M-like activity
and bradykinin by neutral endopeptidase. Circulation 1999;99(15):1984–1990.
374. Griswold JA, Beall CV, Baker CR, Jr., Little DT, Little GH, Behal FJ. Bradykinin metabolism in
the liver and lung of the rat. J Surg Res 1996;66(1):12–20.
375. Bralet J, Schwartz JC. Vasopeptidase inhibitors: An emerging class of cardiovascular drugs. Trends
Pharmacol Sci 2001;22(3):106–109.
376. Marceau F. Kinin B1 receptors: A review. Immunopharmacology 1995;30(1):1–26.
377. Molinaro G, Gervais N, Adam A. Biochemical basis of angioedema associated with recombinant
tissue plasminogen activator treatment: An in vitro experimental approach. Stroke 2002;33(6):1712–
1716.
378. Marceau F, Hess JF, Bachvarov DR. The B1 receptors for kinins. Pharmacol Rev 1998;50(3):357–
386.
379. Perkins MN, Campbell E, Dray A. Antinociceptive activity of the bradykinin B1 and B2 receptor
antagonists, des-Arg9, [Leu8]-BK and HOE 140, in two models of persistent hyperalgesia in the rat.
Pain 1993;53(2):191–197.
380. Rupniak NM, Boyce S, Webb JK, Williams AR, Carlson EJ, Hill RG, Borkowski JA, Hess JF.
Effects of the bradykinin B1 receptor antagonist des-Arg9[Leu8]bradykinin and genetic disruption
of the B2 receptor on nociception in rats and mice. Pain 1997;71(1):89–97.
381. Whalley ET, Figueroa CD, Gera L, Bhoola KD. Discovery and therapeutic potential of kinin
receptor antagonists. Expert Opin Drug Discov 2012;7(12):1129–1148.
382. Perkins MN, Kelly D. Induction of bradykinin B1 receptors in vivo in a model of ultra-violet
irradiation-induced thermal hyperalgesia in the rat. Br J Pharmacol 1993;110(4):1441–1444.
383. Davis AJ, Perkins MN. Induction of B1 receptors in vivo in a model of persistent inflammatory
mechanical hyperalgesia in the rat. Neuropharmacology 1994;33(1):127–133.
384. Gabra BH, Sirois P. Role of bradykinin B(1) receptors in diabetes-induced hyperalgesia in
streptozotocin-treated mice. Eur J Pharmacol 2002;457(2–3):115–124.
385. Dressen D, Garofalo AW, Hawkinson J, Hom D, Jagodzinski J, Marugg JL, Neitzel ML, Pleiss
MA, Szoke B, Tung JS, Wone DW, Wu J, Zhang H. Preparation and optimization of a series
of 3-carboxamido-5-phenacylaminopyrazole bradykinin B1 receptor antagonists. J Med Chem
2007;50(21):5161–5167.
386. Su DS, Lim JL, Tinney E, Wan BL, Murphy KL, Reiss DR, Harrell CM, O’Malley SS, Ransom RW,
Chang RS, Pettibone DJ, Yu J, Tang C, Prueksaritanont T, Freidinger RM, Bock MG, Anthony
NJ. 2-Aminobenzophenones as a novel class of bradykinin B1 receptor antagonists. J Med Chem
2008;51(13):3946–3952.
387. Huang H, Player MR. Bradykinin B1 receptor antagonists as potential therapeutic agents for pain.
J Med Chem 2010;53(15):5383–5399.
388. Fincham CI, Bressan A, Paris M, Rossi C, Fattori D. Bradykinin receptor antagonists–a review of
the patent literature 2005–2008. Expert Opin Ther Pat 2009;19(7):919–941.
389. Catanzaro OL, Dziubecki D, Obregon P, Rodriguez RR, Sirois P. Antidiabetic efficacy of bradykinin
antagonist R-954 on glucose tolerance test in diabetic type 1 mice. Neuropeptides 2010;44(2):187–
189.

Medicinal Research Reviews DOI 10.1002/med


52 r HALLBERG
390. Barth M, Bondoux M, Luccarini JM, Peyrou V, Dodey P, Pruneau D, Massardier C, Paquet JL.
From bradykinin B2 receptor antagonists to orally active and selective bradykinin B1 receptor
antagonists. J Med Chem 2012;55(6):2574–2584.
391. Tang C, Carr BA, Poignant F, Ma B, Polsky-Fisher SL, Kuo Y, Strong-Basalyga K, Nor-
cross A, Richards K, Eisenhandler R, Carlini EJ, Di Marco CN, Kuduk SD, Yu NX,
Raab CE, Rushmore T, Frederick CB, Bock MG, Prueksaritanont T. CYP2C75-involved
autoinduction of metabolism in rhesus monkeys of methyl 3-chloro-3’-fluoro-4’-{(1R)-1-
[({1-[(trifluoroacetyl)amino]cyclopropyl}carbonyl)a mino]ethyl}-1,1’-biphenyl-2-carboxylate (MK-
0686), a bradykinin B1 receptor antagonist. J Pharmacol Exp Ther 2008;325(3):935–946.
392. Pouliot M, Talbot S, Senecal J, Dotigny F, Vaucher E, Couture R. Ocular application of the kinin B1
receptor antagonist LF22–0542 inhibits retinal inflammation and oxidative stress in streptozotocin-
diabetic rats. PLoS One 2012;7(3):e33864.
393. Kuduk SD, Di Marco CN, Chang RK, Wood MR, Kim JJ, Schirripa KM, Murphy KL, Ransom
RW, Tang C, Torrent M, Ha S, Prueksaritanont T, Pettibone DJ, Bock MG. 5-Piperazinyl pyridine
carboxamide bradykinin B1 antagonists. Bioorg Med Chem Lett 2006;16(10):2791–2795.
394. Ha SN, Hey PJ, Ransom RW, Harrell CM, Jr., Murphy KL, Chang R, Chen TB, Su DS, Markowitz
MK, Bock MG, Freidinger RM, Hess FJ. Binding modes of dihydroquinoxalinones in a homology
model of bradykinin receptor 1. Biochem Biophys Res Commun 2005;331(1):159–166.
395. Su DS, Markowitz MK, DiPardo RM, Murphy KL, Harrell CM, O’Malley SS, Ransom RW, Chang
RS, Ha S, Hess FJ, Pettibone DJ, Mason GS, Boyce S, Freidinger RM, Bock MG. Discovery of a
potent, non-peptide bradykinin B1 receptor antagonist. J Am Chem Soc 2003;125(25):7516–7517.
396. Chen JJ, Johnson EJ. Targeting the bradykinin B1 receptor to reduce pain. Expert Opin Ther Targets
2007;11(1):21–35.
397. Congreve M, Langmead CJ, Mason JS, Marshall FH. Progress in structure based drug design for
G protein-coupled receptors. J Med Chem 2011;54(13):4283–4311.
398. Marmarou A, Nichols J, Burgess J, Newell D, Troha J, Burnham D, Pitts L. Effects of the bradykinin
antagonist Bradycor (deltibant, CP-1027) in severe traumatic brain injury: Results of a multi-
center, randomized, placebo-controlled trial. American Brain Injury Consortium Study Group. J
Neurotrauma 1999;16(6):431–444.
399. Ivashkova Y, Svetnitsky A, Mayzler O, Pruneau D, Benifla M, Fuxman Y, Cohen A, Artru AA,
Shapira Y. Bradykinin B2 receptor antagonism with LF 18–1505T reduces brain edema and im-
proves neurological outcome after closed head trauma in rats. J Trauma 2006;61(4):879–885.
400. Wirth K, Hock FJ, Albus U, Linz W, Alpermann HG, Anagnostopoulos H, Henk S, Breipohl G,
Konig W, Knolle J, Schölkens BA. Hoe 140 a new potent and long acting bradykinin-antagonist:
In vivo studies. Br J Pharmacol 1991;102(3):774–777.
401. Hock FJ, Wirth K, Albus U, Linz W, Gerhards HJ, Wiemer G, Henke S, Breipohl G, Konig W,
Knolle J, Schölkens BA. Hoe 140 a new potent and long acting bradykinin-antagonist: In vitro
studies. Br J Pharmacol 1991;102(3):769–773.
402. Bork K, Frank J, Grundt B, Schlattmann P, Nussberger J, Kreuz W. Treatment of acute edema
attacks in hereditary angioedema with a bradykinin receptor-2 antagonist (Icatibant). J Allergy Clin
Immunol 2007;119(6):1497–1503.
403. Aberer W, Maurer M, Reshef A, Longhurst H, Kivity S, Bygum A, Caballero T, Bloom B, Nair N,
Malbran A. Open-label, multicenter study of self-administered icatibant for attacks of hereditary
angioedema. Allergy 2013;69(3):305–314.
404. Fortin JP, Marceau F. Advances in the development of bradykinin receptor ligands. Curr Top Med
Chem 2006;6(13):1353–1363.
405. Gibson C, Schnatbaum K, Pfeifer JR, Locardi E, Paschke M, Reimer U, Richter U, Scharn D,
Faussner A, Tradler T. Novel small molecule bradykinin B2 receptor antagonists. J Med Chem
2009;52(14):4370–4379.
406. Aramori I, Zenkoh J, Morikawa N, Asano M, Hatori C, Sawai H, Kayakiri H, Satoh S, Inoue T,
Abe Y, Sawada Y, Mizutani T, Inamura N, Nakahara K, Kojo H, Oku T, Notsu Y. Nonpeptide

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 53
mimic of bradykinin with long-acting properties at the bradykinin B2 receptor. Mol Pharmacol
1997;52(1):16–20.
407. Asano M, Hatori C, Sawai H, Johki S, Inamura N, Kayakiri H, Satoh S, Abe Y, Inoue T, Sawada
Y, Mizutani T, Oku T, Nakahara K. Pharmacological characterization of a nonpeptide bradykinin
B2 receptor antagonist, FR165649, and agonist, FR190997. Br J Pharmacol 1998;124(3):441–446.
408. Hayashi I, Ishihara K, Kumagai Y, Majima M. Proinflammatory characteristics of a nonpeptide
bradykinin mimic, FR190997, in vivo. Br J Pharmacol 2001;133(8):1296–1306.
409. Majima M, Hayashi I, Inamura N, Fujita T, Ogino M. A nonpeptide mimic of bradykinin blunts
the development of hypertension in young spontaneously hypertensive rats. Hypertension 2000;35(1
Pt 2):437–442.
410. Cerda-Reverter JM, Larhammar D. Neuropeptide Y family of peptides: Structure, anatomical
expression, function, and molecular evolution. Biochem Cell Biol 2000;78(3):371–392.
411. Sundstrom G, Larsson TA, Brenner S, Venkatesh B, Larhammar D. Evolution of the neuropeptide
Y family: New genes by chromosome duplications in early vertebrates and in teleost fishes. Gen
Comp Endocrinol 2008;155(3):705–716.
412. Tatemoto K, Carlquist M, Mutt V. Neuropeptide Y–a novel brain peptide with structural similarities
to peptide YY and pancreatic polypeptide. Nature 1982;296(5858):659–660.
413. Parker E, Van Heek M, Stamford A. Neuropeptide Y receptors as targets for anti-obesity drug
development: Perspective and current status. Eur J Pharmacol 2002;440(2–3):173–187.
414. Redrobe JP, Dumont Y, Fournier A, Quirion R. The neuropeptide Y (NPY) Y1 receptor subtype
mediates NPY-induced antidepressant-like activity in the mouse forced swimming test. Neuropsy-
chopharmacology 2002;26(5):615–624.
415. Silva AP, Cavadas C, Grouzmann E. Neuropeptide Y and its receptors as potential therapeutic drug
targets. Clin Chim Acta 2002;326(1–2):3–25.
416. Blomqvist AG, Herzog H. Y-receptor subtypes–how many more? Trends Neurosci 1997;20(7):294–
298.
417. Larhammar D, Wraith A, Berglund MM, Holmberg SK, Lundell I. Origins of the many NPY-family
receptors in mammals. Peptides 2001;22(3):295–307.
418. Larhammar D, Salaneck E. Molecular evolution of NPY receptor subtypes. Neuropeptides
2004;38(4):141–151.
419. Michel MC, Beck-Sickinger A, Cox H, Doods HN, Herzog H, Larhammar D, Quirion R, Schwartz
T, Westfall T. XVI. International Union of Pharmacology recommendations for the nomenclature of
neuropeptide Y, peptide YY, and pancreatic polypeptide receptors. Pharmacol Rev 1998;50(1):143–
150.
420. Hoyle CH. Neuropeptide families and their receptors: Evolutionary perspectives. Brain Res
1999;848(1–2):1–25.
421. Larhammar D. Evolution of neuropeptide Y, peptide YY and pancreatic polypeptide. Regul Pept
1996;62(1):1–11.
422. Mitic K, Stanojevic S, Kustrimovic N, Vujic V, Dimitrijevic M. Neuropeptide Y modulates functions
of inflammatory cells in the rat: Distinct role for Y1, Y2 and Y5 receptors. Peptides 2011;32(8):1626–
1633.
423. Chee MJ, Colmers WF. Y eat? Nutrition 2008;24(9):869–877.
424. Zarjevski N, Cusin I, Vettor R, Rohner-Jeanrenaud F, Jeanrenaud B. Chronic intracerebroventric-
ular neuropeptide-Y administration to normal rats mimics hormonal and metabolic changes of
obesity. Endocrinology 1993;133(4):1753–1758.
425. Vettor R, Zarjevski N, Cusin I, Rohner-Jeanrenaud F, Jeanrenaud B. Induction and reversibility
of an obesity syndrome by intracerebroventricular neuropeptide Y administration to normal rats.
Diabetologia 1994;37(12):1202–1208.
426. Sahu A, Kalra PS, Kalra SP. Food deprivation and ingestion induce reciprocal changes in neu-
ropeptide Y concentrations in the paraventricular nucleus. Peptides 1988;9(1):83–86.

Medicinal Research Reviews DOI 10.1002/med


54 r HALLBERG
427. Fetissov SO, Kopp J, Hokfelt T. Distribution of NPY receptors in the hypothalamus. Neuropeptides
2004;38(4):175–188.
428. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE,
Low MJ, Ghatei MA, Cone RD, Bloom SR. Gut hormone PYY(3–36) physiologically inhibits food
intake. Nature 2002;418(6898):650–654.
429. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA, Bloom SR.
Inhibition of food intake in obese subjects by peptide YY3–36. N Engl J Med 2003;349(10):941–948.
430. Sainsbury A, Schwarzer C, Couzens M, Herzog H. Y2 receptor deletion attenuates the type 2
diabetic syndrome of ob/ob mice. Diabetes 2002;51(12):3420–3427.
431. Kask A, Harro J, von Horsten S, Redrobe JP, Dumont Y, Quirion R. The neurocircuitry and
receptor subtypes mediating anxiolytic-like effects of neuropeptide Y. Neurosci Biobehav Rev
2002;26(3):259–283.
432. Heilig M. The NPY system in stress, anxiety and depression. Neuropeptides 2004;38(4):213–224.
433. Taiwo OB, Taylor BK. Antihyperalgesic effects of intrathecal neuropeptide Y during inflammation
are mediated by Y1 receptors. Pain 2002;96(3):353–363.
434. Yalamuri SM, Brennan TJ, Spofford CM. Neuropeptide Y is analgesic in rats after plantar incision.
Eur J Pharmacol 2013;698(1–3):206–212.
435. Thorsell A, Rimondini R, Heilig M. Blockade of central neuropeptide Y (NPY) Y2 receptors
reduces ethanol self-administration in rats. Neurosci Lett 2002;332(1):1–4.
436. Thiele TE, Koh MT, Pedrazzini T. Voluntary alcohol consumption is controlled via the neuropeptide
Y Y1 receptor. J Neurosci 2002;22(3):RC208.
437. Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, Herzog H.
Hypothalamic Y2 receptors regulate bone formation. J Clin Invest 2002;109(7):915–921.
438. Sousa DM, Baldock PA, Enriquez RF, Zhang L, Sainsbury A, Lamghari M, Herzog H.
Neuropeptide Y Y1 receptor antagonism increases bone mass in mice. Bone 2012;51(1):8–16.
439. Gullestad L, Bjuro T, Aaberge L, Apelland T, Skardal R, Kjekshus E, Nordlander M, Ablad B,
Pernow J. The effect of a neuropeptide Y Y1 receptor antagonist in patients with angina pectoris.
Eur Heart J 2003;24(12):1120–1127.
440. Ilebekk A, Bjorkman JA, Nordlander M. Influence of endogenous neuropeptide Y (NPY)
on the sympathetic-parasympathetic interaction in the canine heart. J Cardiovasc Pharmacol
2005;46(4):474–480.
441. Minth CD, Bloom SR, Polak JM, Dixon JE. Cloning, characterization, and DNA sequence
of a human cDNA encoding neuropeptide tyrosine. Proc Natl Acad Sci USA 1984;81(14):
4577–4581.
442. Medeiros Mdos S, Turner AJ. Metabolism and functions of neuropeptide Y. Neurochem Res
1996;21(9):1125–1132.
443. Stenfors C, Hellman U, Silberring J. Characterization of endogenous neuropeptide Y in rat
hippocampus and its metabolism by nanospray mass spectrometry. J Biol Chem 1997;272(9):
5747–5751.
444. Unniappan S, McIntosh CH, Demuth HU, Heiser U, Wolf R, Kieffer TJ. Effects of dipeptidyl
peptidase IV on the satiety actions of peptide YY. Diabetologia 2006;49(8):1915–1923.
445. Mentlein R, Dahms P, Grandt D, Kruger R. Proteolytic processing of neuropeptide Y and peptide
YY by dipeptidyl peptidase IV. Regul Pept 1993;49(2):133–144.
446. Mentlein R. Dipeptidyl-peptidase IV (CD26)–role in the inactivation of regulatory peptides. Regul
Pept 1999;85(1):9–24.
447. Labelle M, St-Pierre S, Savard R, Boulanger Y. Solution structure of neuropeptide tyrosine 13–36,
a Y2 receptor agonist, as determined by NMR. Eur J Biochem 1997;246(3):780–785.
448. Abid K, Rochat B, Lassahn PG, Stocklin R, Michalet S, Brakch N, Aubert JF, Vatansever B,
Tella P, De Meester I, Grouzmann E. Kinetic study of neuropeptide Y (NPY) proteolysis in

Medicinal Research Reviews DOI 10.1002/med


METABOLISM TO BIOACTIVE FRAGMENTS r 55
blood and identification of NPY3–35: A new peptide generated by plasma kallikrein. J Biol Chem
2009;284(37):24715–24724.
449. Kamiji MM, Inui A. Neuropeptide y receptor selective ligands in the treatment of obesity. Endocr
Rev 2007;28(6):664–684.
450. Ladyman SR, Woodside B. Regulation of maternal food intake and mother-pup interactions by the
Y5 receptor. Physiol Behav 2009;97(1):91–97.
451. Marsh DJ, Hollopeter G, Kafer KE, Palmiter RD. Role of the Y5 neuropeptide Y receptor in
feeding and obesity. Nat Med 1998;4(6):718–721.
452. Erondu N, Gantz I, Musser B, Suryawanshi S, Mallick M, Addy C, Cote J, Bray G, Fujioka K,
Bays H, Hollander P, Sanabria-Bohorquez SM, Eng W, Langstrom B, Hargreaves RJ, Burns HD,
Kanatani A, Fukami T, MacNeil DJ, Gottesdiener KM, Amatruda JM, Kaufman KD, Heymsfield
SB. Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in
overweight and obese adults. Cell Metab 2006;4(4):275–282.
453. Erondu N, Addy C, Lu K, Mallick M, Musser B, Gantz I, Proietto J, Astrup A, Toubro S, Rissannen
AM, Tonstad S, Haynes WG, Gottesdiener KM, Kaufman KD, Amatruda JM, Heymsfield SB.
NPY5R antagonism does not augment the weight loss efficacy of orlistat or sibutramine. Obesity
(Silver Spring) 2007;15(8):2027–2042.
454. George M, Rajaram M, Shanmugam E. New and emerging drug molecules against obesity. J
Cardiovasc Pharmacol Ther 2014;19(1):65–76.
455. Fichtner M, Lee E, Tomlinson E, Scott D, Cornelius P, Patterson TA, Carpino PA. Discovery and
evaluation of spirocyclic derivatives as antagonists of the neuropeptide Y5 receptor. Bioorg Med
Chem Lett 2012;22(8):2738–2743.
456. Walker MW, Wolinsky TD, Jubian V, Chandrasena G, Zhong H, Huang X, Miller S,
Hegde LG, Marsteller DA, Marzabadi MR, Papp M, Overstreet DH, Gerald CP, Craig
DA. The novel neuropeptide Y Y5 receptor antagonist Lu AA33810 [N-[[trans-4-[(4,5-
dihydro[1]benzothiepino[5,4-d]thiazol-2-yl)amino]cyclohexyl]me thyl]-methanesulfonamide] exerts
anxiolytic- and antidepressant-like effects in rat models of stress sensitivity. J Pharmacol Exp Ther
2009;328(3):900–911.
457. Melnikova I, Wages D. Anti-obesity therapies. Nat Rev 2006;5(5):369–370.
458. Cooke D, Bloom S. The obesity pipeline: Current strategies in the development of anti-obesity
drugs. Nat Rev 2006;5(11):919–931.
459. Andres CJ, Antal Zimanyi I, Deshpande MS, Iben LG, Grant-Young K, Mattson GK, Zhai W.
Differentially functionalized diamines as novel ligands for the NPY2 receptor. Bioorg Med Chem
Lett 2003;13(17):2883–2885.
460. Lunniss GE, Barnes AA, Barton N, Biagetti M, Bianchi F, Blowers SM, Caberlotto L, Emmons A,
Holmes IP, Montanari D, Norris R, Walters DJ, Watson SP. The identification and optimisation
of novel and selective diamide neuropeptide Y Y2 receptor antagonists. Bioorg Med Chem Lett
2009;19(15):4022–4025.
461. Brothers SP, Saldanha SA, Spicer TP, Cameron M, Mercer BA, Chase P, McDonald P, Wahlestedt
C, Hodder PS. Selective and brain penetrant neuropeptide y y2 receptor antagonists discovered by
whole-cell high-throughput screening. Mol Pharmacol 2010;77(1):46–57.
462. Jablonowski JA, Chai W, Li X, Rudolph DA, Murray WV, Youngman MA, Dax SL, Nepomuceno
D, Bonaventure P, Lovenberg TW, Carruthers NI. Novel non-peptidic neuropeptide Y Y2 receptor
antagonists. Bioorg Med Chem Lett 2004;14(5):1239–1242.
463. Sato N, Ogino Y, Mashiko S, Ando M. Modulation of neuropeptide Y receptors for the treatment
of obesity. Expert Opin Ther Pat 2009;19(10):1401–1415.
464. Gehlert DR, Beavers LS, Johnson D, Gackenheimer SL, Schober DA, Gadski RA. Expression
cloning of a human brain neuropeptide Y Y2 receptor. Mol Pharmacol 1996;49(2):224–228.
465. Morales-Medina JC, Dumont Y, Bonaventure P, Quirion R. Chronic administration of the Y2 re-
ceptor antagonist, JNJ-31020028, induced anti-depressant like-behaviors in olfactory bulbectomized
rat. Neuropeptides 2012;46(6):329–334.

Medicinal Research Reviews DOI 10.1002/med


56 r HALLBERG
466. Swanson DM, Wong VD, Jablonowski JA, Shah C, Rudolph DA, Dvorak CA, Seierstad M, Dvorak
LK, Morton K, Nepomuceno D, Atack JR, Bonaventure P, Lovenberg TW, Carruthers NI. The
discovery and synthesis of JNJ 31020028, a small molecule antagonist of the Neuropeptide Y Y(2)
receptor. Bioorg Med Chem Lett 2011;21(18):5552–5556.
467. Kim YG, Lone AM, Nolte WM, Saghatelian A. Peptidomics approach to elucidate the proteolytic
regulation of bioactive peptides. Proc Natl Acad Sci USA 2012;109(22):8523–8527.
468. McNeish AJ, Roux BT, Aylett SB, Van Den Brink AM, Cottrell GS. Endosomal proteolysis regulates
calcitonin gene-related peptide responses in mesenteric arteries. Br J Pharmacol 2012;167(8):1679–
1690.
469. Gesty-Palmer D, Yuan L, Martin B, Wood WH, 3rd, Lee MH, Janech MG, Tsoi LC, Zheng
WJ, Luttrell LM, Maudsley S. beta-arrestin-selective G protein-coupled receptor agonists engender
unique biological efficacy in vivo. Mol Endocrinol 2013;27(2):296–314.
470. Kim KS, Abraham D, Williams B, Violin JD, Mao L, Rockman HA. beta-Arrestin-biased AT1R
stimulation promotes cell survival during acute cardiac injury. Am J Physiol Heart Circ Physiol
2012;303(8):H1001–H1010.
471. Boerrigter G, Soergel DG, Violin JD, Lark MW, Burnett JC, Jr. TRV120027, a novel beta-arrestin
biased ligand at the angiotensin II type I receptor, unloads the heart and maintains renal function
when added to furosemide in experimental heart failure. Circ Heart Fail 2012;5(5):627–634.
472. Monasky MM, Taglieri DM, Henze M, Warren CM, Utter MS, Soergel DG, Violin JD, Solaro RJ.
The beta-arrestin-biased ligand TRV120023 inhibits angiotensin II-induced cardiac hypertrophy
while preserving enhanced myofilament response to calcium. Am J Physiol Heart Circ Physiol
2013;305(6):H856–H866.

Mathias Hallberg, Associate Professor in Pharmacology at Uppsala University, Sweden, received


his PhD in 2005 with a thesis on the effects of anabolic androgenic steroids on the brain. His research
covers the involvement of peptidergic mechanisms in chronic pain and drug dependence as well as
the neuroendocrine regulation of cognitive behavior. His current activities include the development
of drug-like peptidemimetics of bioactive degradation products in the renin angiotensin and the
tachykinin systems. Peptidemimetics of substance P (1–7), attenuating neuropathic pain in animal
models, and angiotensin (3–8) (Ang IV) with potential as cognitive enhancers were recently
developed. He received the “Best Teacher Award” at Uppsala University in 2013.

Medicinal Research Reviews DOI 10.1002/med

You might also like