You are on page 1of 6

Drug Discovery Stories

Cite This: ACS Pharmacol. Transl. Sci. 2019, 2, 485−490 pubs.acs.org/ptsci

Discovery of the Migraine Prevention Therapeutic Aimovig


(Erenumab), the First FDA-Approved Antibody against a G‑Protein-
Coupled Receptor
Chadwick Terence King, Colin V. Gegg, Sylvia Nai-Yu Hu, Hsieng Sen Lu, Brian M. Chan,
Kelly A. Berry, David W. Brankow, Tom J. Boone, Nebojsa Kezunovic, Matt R. Kelley, Licheng Shi,
and Cen Xu*
Amgen Research, 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: In 2018, the United States Food and Drug Administration (FDA) approved Aimovig (erenumab) for the
prevention of migraine. Erenumab is the first FDA approved antibody therapeutic against a G-protein-coupled receptor, the
canonical receptor of calcitonin gene related peptide (CGRP-R). A novel, epitope-focused antigen was created to reconstruct
Downloaded via 103.165.20.202 on March 18, 2023 at 18:04:52 (UTC).

the extracellular domains of the CGRP-R in a stable conformation. Successful inoculation of XenoMouse animals and careful
screening yielded multiple candidate molecules for high potency and exquisite selectivity toward the CGRP-R over related
receptors. These efforts led to the discovery of erenumab which has demonstrated the desired efficacy and safety profiles in
multiple clinical studies for the prevention of migraine. The innovation developed in the discovery of erenumab furthers the
ability to target G-coupled protein receptors using antibody approaches.

■ INTRODUCTION
Migraine, a leading cause of disability, affects near 15% of the
agonists, considered the standard of care for acute migraine for
several decades.9 In addition, intravenous infusion of CGRP in
world’s population and is characterized by debilitating migraineurs triggers migraine-like attacks.10 Collectively, these
headache pain, nausea, and increased sensitivity to light and data supported a substantive role for CGRP in migraine
sound.1 Historically, migraine attacks have been difficult to pathology and that antagonizing the action of CGRP may be of
prevent due to poor patient adherence and a large number of therapeutic benefit.
nonresponders to treatments repurposed from other indica- With the discovery of the major role the CGRP pathway
tions.2,3 To answer this unmet medical need, Aimovig plays in migraine, small-molecule antagonists of the CGRP-R,
(erenumab) was specifically designed and developed, leverag- known as “gepants”, were developed. Multiple gepants were
ing Amgen’s leading biotechnology expertise. Erenumab tested in the clinic, and these compounds demonstrated
(erenumab-aooe in the United States) is the only Food and efficacy in the reversal of acute migraine attacks. This clinical
Drug Administration (FDA)-approved monoclonal antibody proof of concept brought further validation of CGRP
(mAb) against the canonical receptor of calcitonin gene- antagonism for the treatment of migraine.11−15
related peptide (CGRP). It is potent and specific for its target,
clinically efficacious, and well-tolerated by patients.4 In this
article, we seek to summarize the key innovations that led to
■ USE OF ANTIBODIES INSTEAD OF SMALL
MOLECULES
the discovery of this agent. Despite their efficacy and generally good safety profile, the

■ CGRP RECEPTOR ANTAGONISM AND MIGRAINE


Migraine is a complex neurological disease involving central
first-generation gepants had limited success clinically due to
hepatoxicity concerns after extended use.16,17 In 2011, the
development of telcagepant, the most advanced small-molecule
and peripheral nervous systems. Studies on the interaction of CGRP-R antagonist, was terminated.18 The site of action of
sensory nerves and cranial blood vessels in the trigeminovas- the gepants was originally assumed to be in the central nervous
cular system revealed that CGRP, a potent vasodilatory peptide system. However, calculation of brain concentration based on
is involved in the headache pain occurring during a migraine the clinically efficacious exposure and the physical-chemical
attack.5 CGRP is a 37 amino acid (AA) neuropeptide that properties of these antagonists suggested that clinical efficacy is
binds to several different G-protein-coupled receptors most likely peripherally driven.14,19 Therefore, we hypothe-
(GPCRs) in the calcitonin receptor family, including CGRP, sized that a peripherally restrictive antagonist antibody
amylin, calcitonin, and adrenomedullin receptors, with the targeting the CGRP-R would be therapeutically efficacious
CGRP-R showing the highest binding affinity, making it the for migraine treatment. Monoclonal antibodies provide several
canonical CGRP-R.6,7 CGRP-Rs are present in both the central potential advantages over small molecules, including high
and peripheral nervous systems, as well as in smooth muscle affinity, high specificity (less off-target toxicity) through
cells surrounding cerebral, meningeal, and dural arteries.8
CGRP plasma levels increase during migraine5 and normalize Received: August 2, 2019
after administration of triptans, serotonin 5-HT1B,1D receptor Published: September 3, 2019

© 2019 American Chemical Society 485 DOI: 10.1021/acsptsci.9b00061


ACS Pharmacol. Transl. Sci. 2019, 2, 485−490
ACS Pharmacology & Translational Science Drug Discovery Stories

Figure 1. CGRP and related receptors of the calcitonin family. The CGRP receptor is similar in composition to several other members of the
calcitonin family of receptors (receptor name listed above diagrams). These receptors are formed by a complex of the calcitonin-like receptor
(CLR) or calcitonin receptor (CTR) with receptor-activity-modifying proteins (RAMPs 1−3), predominantly binding ligands CGRP,
adrenomedullin (AM), and amylin (AMY). Due to the close similarity between receptor complexes, these agonists cross-react with other receptors
of the family.

binding to a unique epitope, and prolonged plasma half-life effects.24 These functional indications call for an exquisite
enabling less frequent dosing. In addition, limited CNS selectivity to the CGRP-R. However, components shared
exposure can reduce potential liability associated with between these related receptors present additional challenges
CGRP-R blockade behind the blood brain barrier. This where asymmetric binding of an antagonist antibody to either
approach was later supported by positron-emission tomog- component alone would result in lack of selectivity.


raphy (PET) imaging biodistribution data of an efficacious
dose of telcagepant in migraineurs, which demonstrated that DESIGN CRITERIA OF AN ANTIBODY AGAINST
CGRP-R antagonism in the periphery is sufficient for migraine THE CGRP-R
pain relief.20


At the time of designing our strategy, it was known that CGRP
CHALLENGES OF TARGETING THE CGRP-R WITH interacted with the CGRP-R through two distinct regions: the
AN ANTIBODY CGRP C-terminal domain, conferring high-affinity binding,
and the CGRP N-terminal domain, driving agonist activities.25
GPCRs represent about 35% of targets of prescription drugs The CGRP C-terminal domain interacts with a high-affinity
approved by the FDA.21 Despite the significant therapeutic binding region formed at the distal end of the CLR-RAMP1
opportunities in this target class, none of these drugs were extracellular domains. The CGRP N-terminal domain drives
antibody therapeutics prior to the approval of erenumab. This agonist activity, through an interaction with a “classical”
scarcity of antibody therapeutics is due to the technical agonist binding region formed in the CGRP-R by the
challenges associated with generating functional antibodies to transmembrane α-helices of the CLR heterodimer, with no
GPCRs. Generally, the success rates of antibody discovery direct interaction with RAMP1.26,27 The range of interaction
campaigns increased with the availability of the soluble forms between CGRP and its receptor presents alternate strategies
of antigens, and maintaining the structural and functional for designing antibody antagonists: (1) Blocking the agonist
accuracy of the soluble antigen is critical. This is difficult in the
binding region from interacting with the CGRP N-terminus.
case of GPCRs, as they contain multiple transmembrane
This strategy may result in less-selective inhibitors as it does
domains, causing their structural stability to depend on the
not involve RAMP1, as demonstrated by small-molecule
lipid layers of the cell membrane. Full-length membrane-
antagonists that interact within the transmembrane region.26
associated forms of GPCRs can also be used for antibody
discovery, but historically these had lower success rates due to (2) Directly blocking the high-affinity binding area so that the
the inherently poor immunogenic and antigenic characteristics. CGRP is blocked from associating with the receptor. We
For the CGRP-R, there are added challenges in that the decided that such blocking of the high-affinity binding region
receptor is a heterodimeric complex of the calcitonin receptor- was likely the most effective strategy and the best epitope
like receptor (CLR), a class B GPCR, and the receptor activity- would be an extracellular region of CGRP-R that includes both
modifying protein 1 (RAMP1), a type 1 transmembrane RAMP1 and CLR, as this unique combination in the epitope
protein; both proteins are required for CGRP binding and would likely provide selectivity over the AM and AMY
subsequent complexing with the Gs-protein heterotrimer, the receptors.
main transducer protein for this receptor. The CGRP-R is An important additional consideration in our design strategy
closely related to adrenomedullin (AM) and amylin (AMY) is that CGRP has a very high affinity for the CGRP-R with a
receptors of the calcitonin receptor family, shown in Figure 1. dissociation constant (Kd) of ∼15 pM.28 Therefore, we
The two components of the CGRP-R, CLR and RAMP1, are expected that for an antibody to be effective in vivo it should
also individually found in AM1 (CLR + RAMP2), AM2 (CLR bind to the receptor in the low picomolar range.
+ RAMP3), and AMY1 (CTR + RAMP1) receptors.22 These The design criteria for an anti-CGRP-R antagonist antibody
receptors demonstrate sensitivity to CGRP;6 however, neither were therefore strict, requiring innovative solutions. The
AM1 nor AMY1 have been linked to migraine pathophysiol- antibody would need to block a very specific binding region
ogy. In contrast, circulating levels of amylin are raised in on the receptor with selectivity for CGRP-R over related
response to meal ingestion, and the peptide potently inhibits receptor family members, along with high potency and affinity
gastric emptying and gastric acid secretion,22,23 while necessary to be therapeutically relevant. To tackle this
adrenomedullin has been shown to have a remarkable range formidable challenge, we focused on developing novel and
of actions, from regulating cellular growth and differentiation, specific immunogens to conduct large-scale antibody discovery
through modulating hormone secretion, to antimicrobial campaigns.
486 DOI: 10.1021/acsptsci.9b00061
ACS Pharmacol. Transl. Sci. 2019, 2, 485−490
ACS Pharmacology & Translational Science Drug Discovery Stories

Figure 2. Design, purification, and competition binding assay of the soluble CGRPR-ECD Fc fusion protein immunogen. (A) Space-filling model
of the theoretical structure of the CGRP-R ECD-Fc immunogen. This protein was constructed through fusing the N-terminal extracellular domains
of RAMP1 and CLR (yellow and purple) to human IgG1 Fc domain (green). A 5× glycine linker was inserted between the extracellular domains
and Fc region to confer additional flexibility and rotational freedom (orange). Coexpression of the two receptor components will generate a
heterodimeric protein of 81.5 kDa. (B) MALDI-TOF mass data for native sample of CGRPR ECD-Fc immunogen. Peak 1 (76.1 kDa) represents
the homodimer of RAMP1 ECD-Fc, peak 2 (86.3 kDa) represents the heterodimer of CGRP-R ECD-Fc (CLR + RAMP1), and peak 3 (99.1 kDa)
represents CLR ECD-Fc. (C) Results of a fluorescence activated cell sorting (FACS)-based inhibition of ligand binding study, plotted as percent
inhibition. Measured as percent reduction in binding of ALEXA-647 CGRP after 1 h of preincubation at room temperature with increasing
concentrations of purified CGRP-R (ECD)-Fc. An Fc-fusion protein OPG-Fc was used as a negative control.

■ NOVEL ANTIGEN DESIGN: RECONSTRUCTING THE


EXTRACELLULAR DOMAIN OF THE CGRP-R
provided confidence that the heterodimer was likely replicating
the structure of the native CGRP-R ligand binding sites
between CLR and RAMP1.
In addition to the conventional cellular and cell-membrane
immunogen preparations, we specifically designed and
produced a novel soluble protein immunogen containing
only the extracellular domains (ECDs) of the CGRP-R,
■ ANTIBODY DISCOVERY
XenoMouse animals, a transgenic mouse strain that has fully
seeking to recapitulate the unique heterodimeric structure of human immunoglobulin genes,29 were used to generate human
this receptor by coexpressing the extracellular regions of both CGRP-R antibodies. The advantage of fully human antibodies
CLR and RAMP1. Achieving a structurally stable heterodimer as therapeutic molecules is the relatively lower rates of
was essential to represent the unique heterodimeric epitope for immunogenicity.30
immunization. Our approach was to express each individual To ensure maximal diversity and identification of the highest
ECD component as a fusion protein to an antibody Fc domain. quality candidates, two antibody generation campaigns were
Fc domains are expected to form dimers when coexpressed to run using XenoMouse animals: the first inoculating with the
promote the reassembly of CLR and RAMP1. In addition, a soluble CGRP-R ECD-Fc immunogen and the second with a
linker region was inserted between the ECD and Fc domains combination of CGRP-R expressing cells and their cell-
to decrease the rotational constraint on the ECDs and promote
membrane preparations as immunogens.
native reassembly of the CLR and RAMP-1 domains (Figure
The first campaign focused on the soluble protein
2A).
immunogen, with a pool of immune repertoires from the
The two components, CLR ECD-Fc and RAMP1 ECD-Fc,
best-responding animals used for hybridoma generation.
were coexpressed to generate heterodimer Fc proteins. A
Approximately 100 000 of these hybridoma clones were
complicating factor in producing this immunogen was the
uncontrolled dimerization of the Fc domains. This process screened for binding using Chinese hamster ovary (CHO)
produced three species: a CLR ECD homodimer, a RAMP-1 cells stably expressing CGRP-R and counter-screened on CHO
ECD homodimer, and the CGRP-R ECD-heterodimer. cells stably expressing the AM1 receptor. A total of 1092
Applying conventional chromatography purification strategies hybridomas were identified with selective binding to CGRP-R.
allowed separation of the smaller RAMP1-ECD-Fc homo- These 1092 hybridomas were then screened for the ability to
dimers, but the CLR ECD-Fc homodimers and CGRP-R ECD- block CGRP-induced signaling in a cell-based cAMP assay, the
Fc heterodimers could not be completely separated due to key second messenger of CGRP-R action. Antagonist activity
their size similarity (Figure 2B). Overall, we estimated the final of each hybridoma supernatant was expressed as a percent
purified sample to be 40% CGRP-R ECD-Fc and 60% inhibition of cAMP production induced by 1 nM of CGRP. Of
homodimeric species. the panel of CGRP-R specific binders, 28% achieved >50%
To assess whether the partially purified CGRP-R ECD-Fc inhibition in this assay.
heterodimer structurally recapitulated ligand binding, it was The campaign with cells and cell-membrane immunogens
tested for activity in a fluorescence activated cell sorting was far less productive for generating specific binders and
(FACS)-based CGRP ligand binding assay. The CGRPR ECD- antagonists compared to results with the CGRP-R ECD-Fc
Fc sample was incubated with a labeled form of CGRP soluble immunogen. In this case, approximately 103 000
(ALEXA-647 CGRP) and added to cells expressing CGRP-R. hybridomas were screened and a total of 119 binders of
A reduction in the amount of ALEXA-647 CGRP binding to CGRP-R were identified. This panel of binders was then
the cells was then calculated as the percent inhibition. ECD screened in the CGRP signaling assay using the same
complex demonstrated concentration-dependent inhibition of conditions as the campaign above, with only 2.5% of the
the binding of CGRP to the CGRP-R (Figure 2C). These data panel achieving >50% inhibition of CGRP signaling.
487 DOI: 10.1021/acsptsci.9b00061
ACS Pharmacol. Transl. Sci. 2019, 2, 485−490
ACS Pharmacology & Translational Science Drug Discovery Stories

The immunization of XenoMouse animals with the soluble Table 1. Potency and Selectivity of Representative
CGRP-R ECD-Fc protein resulted in robust target-specific Antibodies
immune responses compared to results with the native cell-
based immunogens. The dramatically different outcome of
these two screening campaigns clearly indicates the benefit of
the epitope-focused soluble immunogen strategy. The soluble
CGRP-R ECD-Fc heterodimer immunogen generated signifi-
cantly stronger immune responses in XenoMouse animals,
allowing identification of a 10-fold larger specific binder panel
than a comparably scaled campaign with the cellular
immunogens. The soluble immunogen also gave rise to a
much higher frequency of antibody antagonists specific to
CGRP-R, critically important for campaign success. This larger
panel enabled stringent refinement of the lead candidates to
only those highly selective antagonists. Figure 3 summarizes

human CGRP-R over the other members of this receptor


family. Taken together, these results indicated the design goal
had been achieved.

■ CONFIRMATION OF HETEROMETRIC
INTERACTIONS
We inferred that the antibody drug candidates identified must
bind to a heterodimeric region on the CGRP-R formed by the
CLR and RAMP1 components. To test this hypothesis, we
conducted protease protection assay to map the binding
regions. After generating a disulfide peptide map of the CGRP-
R ECD-Fc protein (method described in ref 31), peptides
produced through proteolytic digestion using AspN (which
cleaves after aspartic acid and some glutamic acid residues)
were analyzed by LC-MS to determine the sequences of
peptide fragments of CLR and RAMP1. Protection assays were
then performed with the presence of an anti-CGRP-R
Figure 3. Functional selectivity screening of most potent CGRP-R
antagonists. Large panel of 167 of the most potent CGRP-R antibody. The difference between the peptide maps indicated
antagonist hybridomas (>70% inhibition) were tested for functional the region(s) of the CGRP-R ECD Fc protein that were
activity in AMY1 (CTR+RAMP1) and AM1 (CLR+RAMP2) cell- protected from proteolytic digestion by AspN when bound to
based assays. The majority of samples tested showed little to no the anti-CGRP-R antibody (method described in ref 32). A
activity. A subset of the most selective hybridomas were then representative result of these studies is shown in Table 2. The
recombinantly expressed for potency determination.
Table 2. Result of AspN Proteolytic Digestion of the CGRP-
R ECD-Fc Fusion Proteina
the functional selectivity of this panel. In these screens the
most potent set of 167 CGRP-R antagonists (>70% inhibition) sequence protection assay − HPLC
were tested for selectivity against AMY1 and AM1 receptors. peptide origin location chromatogram peak height
The majority of candidates were functionally inactive at these C5 CLR D55-P67/D86- decreased
related receptors, indicating a high degree of selectivity, and H110
the advantage of using an epitope focused immunization C6 CLR D8-Y24 decreased
strategy with the soluble antigen. Immunization with CGRP-R C7 CLR E25-Q32/D48- decreased
N54
ECD-Fc protein yielded almost the entirety of the lead panel of R1 RAMP1 D32-A44 decreased
candidate antibodies. R2 RAMP1 E12-V20/D45- decreased
High-throughput screening performed with hybridoma A51
supernatants identified a lead candidate panel. From this a
Change to peptide peak height as measured by HPLC chromato-
subset, we generated clonal and purified antibody samples for gram are reported in the last column. Reduction in peak height
potency determination. Potency and selectivity data for a indicates a protection.
representative set of four sequence-unique and functionally
selective CGRP-R antagonist antibodies is shown in Table 1,
which summarizes the activity profiles. These four candidates study identified peptide fragments from both CLR and
were shown to have dissociation constants (Kd) in the double- RAMP1 that are protected by the binding of anti-CGRP-R
digit pM range (compared to a Kd of CGRP at 20 pM when Ab from AspN digestion. Our analysis indicated that on CLR
tested side by side in these studies, data not shown) and there were multiple cleavage sites protected by the bound
antagonist IC50’s in the single-digit nM range in cell-based antibody; these included regions between D55-P67/D86-
cAMP signaling assay. Furthermore, the binding and functional H110, D8-Y24, and E25-Q32/D48-N54. On RAMP1, the
antagonistic activities were shown to be exquisitely selective to results indicated that the antibody protected multiple cleavage
488 DOI: 10.1021/acsptsci.9b00061
ACS Pharmacol. Transl. Sci. 2019, 2, 485−490
ACS Pharmacology & Translational Science Drug Discovery Stories

sites, including regions between D32-A44 and E12-V20/D45- (10) Lassen, L., Haderslev, P., Jacobsen, V., Iversen, H., Sperling, B.,
A51. The data, although with limited resolution, provided and Olesen, J. (2002) CGRP May Play a Causative Role in Migraine.
direct evidence that regions on both CLR and RAMP1 are Cephalalgia 22, 54−61.
closely associated with the anti-CGRP-R antibody when (11) Connor, K. M., Aurora, S. K., Loeys, T., Ashina, M., Jones, C.,
bound. The binding of the antibody to the unique complex Giezek, H., Massaad, R., Williams-diaz, A., Lines, C., Ho, T. W., et al.
(2011) Headache 51, 73.
of CGRP-R ECD-Fc likely contributes to the specificity over
(12) Hewitt, D. J., Aurora, S. K., Dodick, D. W., Goadsby, P. J., Ge,
other related receptors in the receptor family.


Y., Bachman, R., Taraborelli, D., Fan, X., Assaid, C., Lines, C., et al.
(2011) Randomized Controlled Trial of the CGRP Receptor
CONCLUSION Antagonist MK-3207 in the Acute Treatment of Migraine. Cephalalgia
In this article, we highlight the key innovations that led to the 31 (6), 712−722.
discovery of erenumab, a preventive therapy for migraine. With (13) Ho, T. W, Ferrari, M. D, Dodick, D. W, Galet, V., Kost, J., Fan,
the aim of developing a first-in-class molecule, we generated a X., Leibensperger, H., Froman, S., Assaid, C., Lines, C., et al. (2008)
Effi Cacy and Tolerability of MK-0974 (Telcagepant), a New Oral
fully human, highly potent monoclonal antibody antagonist of
Antagonist of Calcitonin Gene-Related Peptide Receptor, Compared
the human CGRP-R, a complex GPCR formally only targeted with Zolmitriptan for Acute Migraine : A Randomised, Placebo-
by small-molecule approaches. Epitope-focused design of the Controlled, Parallel-Treatment Trial. Lancet 372 (9656), 2115−2123.
specific antigen led to a panel of candidate antibodies with (14) Olesen, J., Diener, H.-C., Husstedt, I. W., Goadsby, P. J., Hall,
desired potency and specificity. Clinical trials for erenumab D., Meier, U., Pollentier, S., and Lesko, L. M. (2004) Calcitonin
demonstrated safety and efficacy for the prevention of episodic Gene−Related Peptide Receptor Antagonist BIBN 4096 BS for the
and chronic migraine.33−36 The high potency and exquisite Acute Treatment of Migraine. N. Engl. J. Med. 350 (11), 1104−1110.
specificity of erenumab were key in this successful develop- (15) Marcus, R., Goadsby, P. J., Dodick, D., Stock, D., Manos, G.,
ment in the clinic as the first FDA approved antibody therapy and Fischer, T. Z. (2014) BMS-927711 for the Acute Treatment of
for the prevention of migraine. The innovation implemented in Migraine: A Double-Blind, Randomized, Placebo Controlled, Dose-
the discovery of erenumab furthers the ability to target GPCRs Ranging Trial. Cephalalgia 34 (2), 114−125.
therapeutically using antibody technology. We look forward to (16) Gonzalez-Hernandez, A., Marichal-Cancino, B. A.,
the application of this strategy to the development of many MaassenVanDenBrink, A., and Villalon, C. M. (2018) Expert Opinion
on Drug Metabolism & Toxicology Side Effects Associated with
agents in the near future.


Current and Prospective Antimigraine Pharmacotherapies. Expert
Opin. Drug Metab. Toxicol. 14 (1), 25−41.
AUTHOR INFORMATION (17) Edvinsson, L., Haanes, K. A., Warfvinge, K., and Krause, Di. N.
Corresponding Author (2018) CGRP as the Target of New Migraine Therapies - Successful
*E-mail: cenx@amgen.com. Translation from Bench to Clinic. Nat. Rev. Neurol. 14 (6), 338−350.
(18) Zhang, Y., Edvinsson, L., Michelson, D., Lines, C., Fan, X.,
Notes Pearlman, E., Koppenhaver, J., Connor, K. M., Goadsby, P. J., and Ho,
The authors declare the following competing financial T. W. (2014) Randomized Controlled Trial of the CGRP Receptor
interest(s): All authors are or were employed by Amgen Inc. Antagonist Telcagepant for Migraine Prevention. Neurology 83 (11),


958−966.
REFERENCES (19) Iovino, M., Feifel, U., Yong, C. L., Wolters, J. M., and
Wallenstein, G. (2004) Safety, Tolerability and Pharmacokinetics of
(1) Vos, T., Abajobir, A. A., Abbafati, C., Abbas, K. M., Abate, K. H., BIBN 4096 BS, the First Selective Small Molecule Calcitonin Gene-
Abd-Allah, F., Abdulle, A. M., Abebo, T. A., Abera, S. F., Aboyans, V., Related Peptide Receptor Antagonist, Following Single Intravenous
et al. (2017) Global, Regional, and National Incidence, Prevalence, Administration in Healthy Volunteers. Cephalalgia 24 (8), 645−656.
and Years Lived with Disability for 328 Diseases and Injuries for 195 (20) Bormans, G., Bell, I. M., Li, C.-C., Sur, C., De Lepeleire, I.,
Countries, 1990−2016: A Systematic Analysis for the Global Burden
Blanchard, R., Evelhoch, J. L., de Groot, T., Williams, M., Miller, P.,
of Disease Study 2016. Lancet 390 (10100), 1211−1259.
et al. (2013) In Vivo Quantification of Calcitonin Gene-Related
(2) Hepp, Z., Bloudek, L. M., and Varon, S. F. (2014) Systematic
Peptide Receptor Occupancy by Telcagepant in Rhesus Monkey and
Review of Migraine Prophylaxis Adherence and Persistence. J. Manag.
Care Pharm. 20 (1), 22−33. Human Brain Using the Positron Emission Tomography Tracer
(3) Lipton, R. B., Bigal, M. E., Diamond, M., Freitag, F., Reed, M. L., [11C]MK-4232. J. Pharmacol. Exp. Ther. 347 (2), 478−486.
and Stewart, W. F. (2007) Migraine Prevalence, Disease Burden, and (21) Sriram, K., and Insel, P. A. (2018) G Protein-Coupled
the Need for Preventive Therapy. Neurology 68, 343−349. Receptors as Targets for Approved Drugs: How Many Targets and
(4) Summary of Product Characteristics; Amgen Inc., 2017. How Many Drugs? Mol. Pharmacol. 93 (4), 251−258.
(5) Goadsby, P. J., Edvinsson, L., and Ekman, R. (1988) Release of (22) Hay, D. L., Poyner, D. R., and Walker, C. S. Calcitonin
Vasoactive Peptides in the Extracerebral Circulation of Humans and receptors. http://www.guidetopharmacology.org/GRAC/
the Cat during Activation of the Trigeminovascular System. Ann. FamilyDisplayForward?familyId=11.
Neurol. 23 (2), 193−196. (23) Roberson, S. W. The Islet Amyloid Polypeptide Hormone
(6) Hay, D. L., and Walker, C. S. (2017) CGRP and Its Receptors. Amylin: Its Function, Effects and Uses in Medicine. Loyola
Headache 57 (4), 625−636. Marymount University Undergraduate Library Research Award, 2013.
(7) Ho, T. W., Edvinsson, L., and Goadsby, P. J. (2010) CGRP and (24) Hinson, J. P., Kapas, S., and Smith, D. M. (2000)
Its Receptors Provide New Insights into Migraine Pathophysiology. Adrenomedullin, a Multifunctional Regulatory Peptide. Endocr. Rev.
Nat. Rev. Neurol. 6 (10), 573−582. 21 (2), 138−167.
(8) Edvinsson, L. (2015) CGRP Receptor Antagonists and (25) Banerjee, S., Evanson, J., Harris, E., Lowe, S. L, Thomasson, K.
Antibodies against CGRP and Its Receptor in Migraine Treatment. A, and Porter, J. E (2006) Identification of Specific Calcitonin-like
Br. J. Clin. Pharmacol. 80 (2), 193−199. Receptor Residues Important for Calcitonin Gene-Related Peptide
(9) Goadsby, P. J., and Edvinsson, L. (1993) The Trigeminovascular High Affinity Binding (BMC Pharmacology). BMC Pharmacol. 6, 9.
System and Migraine: Studies Characterizing Cerebrovascular and (26) Zartman, C. B., Bell, I. M., Gallicchio, S. N., Graham, S. L.,
Neuropeptide Changes Seen in Humans and Cats. Ann. Neurol. 33 Kane, S. A., Mallee, J. J., Rutledge, R. Z., Salvatore, C. A., Vacca, J. P.,
(1), 48−56. and Williams, T. M. (2011) Identification of a Novel RAMP-

489 DOI: 10.1021/acsptsci.9b00061


ACS Pharmacol. Transl. Sci. 2019, 2, 485−490
ACS Pharmacology & Translational Science Drug Discovery Stories

Independent CGRP Receptor Antagonist. Bioorg. Med. Chem. Lett. 21


(22), 6705−6708.
(27) Liang, Y.-L., Khoshouei, M., Deganutti, G., Glukhova, A.,
Koole, C., Peat, T. S., Radjainia, M., Plitzko, J. M., Baumeister, W.,
Miller, L. J., et al. (2018) Cryo-EM Structure of the Active, Gs-Protein
Complexed, Human CGRP Receptor. Nature 561, 492.
(28) Edvinsson, L., Alm, R., Shaw, D., Rutledge, R. Z., Koblan, K. S.,
Longmore, J., and Kane, S. A. (2002) Effect of the CGRP Receptor
Antagonist BIBN4096BS in Human Cerebral, Coronary and Omental
Arteries and in SK-N-MC Cells. Eur. J. Pharmacol. 434 (1−2), 49−53.
(29) Mendez, M. J., Green, L. L., Corvalan, J. R. F., Jia, X., Maynard-
currie, C. E., Yang, X., Gallo, M. L., Louie, D. M., Lee, D. V., and
Erickson, K. L. (1997) Functional Transplant of Megabase Human
Immunoglobulin Loci Recapitulate Antibody Response in Mice. Nat.
Genet. 15, 146.
(30) Foltz, I. N., Gunasekaran, K., and King, C. T. (2016) Discovery
and Bio-Optimization of Human Antibody Therapeutics Using the
XenoMouse® Transgenic Mouse Platform. Immunol. Rev. 270 (1),
51−64.
(31) Merewether, L. A., Le, J., Jones, M. D., Lee, R., Shimamoto, G.,
and Lu, H. S. (2000) Development of Disulfide Peptide Mapping and
Determination of Disulfide Structure of Recombinant Human
Osteoprotegerin Chimera Produced in Escherichia Coli. Arch.
Biochem. Biophys. 375, 101−110.
(32) Suckau, D., Köhl, J., Karwath, G., Schneider, K., Casaretto, M.,
Bitter-Suermann, D., and Przybylski, M. (1990) Molecular Epitope
Identification by Limited Proteolysis of an Immobilized Antigen-
Antibody Complex and Mass Spectrometric Peptide Mapping. Proc.
Natl. Acad. Sci. U. S. A. 87 (24), 9848−9852.
(33) Sun, H., Dodick, D. W., Silberstein, S., Goadsby, P. J., Reuter,
U., Ashina, M., Saper, J., Cady, R., Chon, Y., Dietrich, J., and Lenz, R.
(2016) Safety and Efficacy of AMG 334 for Prevention of Episodic
Migraine: A Randomised, Double-Blind, Placebo-Controlled, Phase 2
Trial. Lancet Neurol. 15 (4), 382−390.
(34) Tepper, S., Ashina, M., Reuter, U., Brandes, J. L., Doležil, D.,
Silberstein, S., Winner, P., Leonardi, D., Mikol, D., and Lenz, R.
(2017) Safety and Efficacy of Erenumab for Preventive Treatment of
Chronic Migraine: A Randomised, Double-Blind, Placebo-Controlled
Phase 2 Trial. Lancet Neurol. 16 (6), 425−434.
(35) Goadsby, P. J., Reuter, U., Hallström, Y., Broessner, G., Bonner,
J. H., Zhang, F., Sapra, S., Picard, H., Mikol, D. D., and Lenz, R. A.
(2017) A Controlled Trial of Erenumab for Episodic Migraine. N.
Engl. J. Med. 377 (22), 2123−2132.
(36) Dodick, D. W., Ashina, M., Brandes, J. L., Kudrow, D., Lanteri-
Minet, M., Osipova, V., Palmer, K., Picard, H., Mikol, D. D., and Lenz,
R. A. (2018) ARISE: A Phase 3 Randomized Trial of Erenumab for
Episodic Migraine. Cephalalgia 38 (6), 1026−1037.

490 DOI: 10.1021/acsptsci.9b00061


ACS Pharmacol. Transl. Sci. 2019, 2, 485−490

You might also like