Compostatin
Compostatin
Yijun Huang
To cite this article: Yijun Huang (2018): Evolution of compstatin family as therapeutic complement
inhibitors, Expert Opinion on Drug Discovery, DOI: 10.1080/17460441.2018.1437139
Article views: 2
REVIEW
CONTACT Yijun Huang huang@alumni.pitt.edu WuXi AppTec Inc., 4701 League Island Boulevard, Philadelphia, PA 19112, USA
© 2018 Informa UK Limited, trading as Taylor & Francis Group
2 Y. HUANG
a. b.
Figure 1. (a) X-ray crystal structure of C3c–compstatin complex (Diffraction data was collected up to 2.4 Å resolution [26]. The figure was rendered by PyMOL from
PDB file 2QKI). The compstatin binding site of C3c is shown as a transparent surface, and selected amino acids which forms hydrogen bonds with compstatin are
shown as white sticks. Compstatin is shown as yellow sticks, and the hydrogen bonds between compstatin and C3c are shown in dash lines. (b) The cartoon
representation of the averaged minimized NMR structure of compstatin (grey) and compstatin in the co-crystal structure (yellow).
EXPERT OPINION ON DRUG DISCOVERY 3
contains 2 × 108 unique clones expressing random 27-mer [30]. The β-turn segment (Gln5-Asp6-Trp7-Gly8) of compstatin
peptides was screened to isolate C3b binding peptides [16]. also forms three hydrogen bonds with the receptor, thus it is a
A peptide (I[CVVQDWGHHRC]TAGHMANLTSHASAI) was iden- determinant for the higher inhibitory activity (Figure 2).
tified as a complement inhibitor (classical pathway: Notably, the side-chain residue of Trp7 is deeply buried
IC50 = 65 µM; alternative pathway: IC50 = 19 µM). The trunca- in a hydrophobic pocket, and forms a hydrogen bond with
tions of the parent peptide led to identify its N-terminal region the main-chain oxygen of Met457. The replacement of Trp7
(marked in bold) as the functional fragment, and this 13-mer by Phe resulted in complete loss of inhibitory activity (Ac-I
cyclic peptide was later named compstatin to fulfill the pro- [CVVQDFGHHRC]T-NH2, IC50 > 400 µM) [30]. Later, compsta-
mise of developing complement inhibitors with therapeutic tin derivatives were expressed and purified from Escherichia
potential. The evolution of compstatin family represents a coli to incorporate non-natural Trp analogs (Figure 3) [33].
typical process using structure-activity relationship studies Compared with the wild-type compstatin (NH2-GI
for the development of peptide drugs. [CVWQDWGAHRC]TN-OH; IC50 = 1.2 µM), the incorporation
The biochemical and functional stability of compstatin in of 6-fuoro-tryptophan (NH2-GI[CV(6fW)QD(6fW)GAHRC]TN-
human blood was studied since it has therapeutic potential as OH; IC50 = 0.43 µM) increased the activity three-fold, while
the complement inhibitor [29]. In vitro biotransformation stu- the incorporation of 5-hydroxy-tryptophan (NH2-GI[CV(5-
dies of compstatin have shown that Ile1 was cleaved at the N OH-W)QD(5-OH-W)GAHRC]TN-OH; IC50 = 33 µM) or 7-aza-
terminus; however, proteolytic processing in blood is blocked tryptophan (NH2-GI[CV(7-aza-W)QD(7-aza-W)GAHRC]TN-
after the first amino acid residue due to the cyclic nature of OH; IC50 = 122 µM) rendered less active compstatin analogs.
compstatin. Acetylation of the amino terminus can sufficiently Similarly, the incorporation of 5-fuoro-tryptophan (Ac-I
block the removal of Ile1 from enzymatic degradation, thus [CVWQD(5fW)GAHRC]T-NH2; IC50 = 0.45 µM) increased the
increases the stability of compstatin in human blood (half-life activity three-fold relative to the wild-type compstatin (Ac-I
of 24 h at 37°C) [29]. In addition, the inhibitory activity of [CVWQDWGAHRC]T-NH2; IC50 = 1.2 µM) [34]. This further
compstatin was increased upon N-acetylation, indicating the suggests that the hydrophobic nature of Trp7 is favorable
participation of the N terminus in binding [30]. No cleavage for the interaction of compstatin with C3, thus contributes
was observed at the C terminus, since it was protected with to the inhibitory activity of compstatin.
the amide incorporated via solid-phase peptide synthesis. The disulfide bridge is essential for structural stability and
Although the introduction of D-amino acid residues is inhibitory activity of compstatin. The linear compstatin
expected to improve a peptide’s stability to proteases, comp- (ICVVQDWGHHRCT, alternative pathway: IC50 > 600 µM) com-
statin analogs incorporating D-amino acids led to dramatic pletely loses the activity [27]. Reduction and alkylation of the
loss of activity [30,31]. two cysteines also result in a loss of inhibitory activity [16]. In
Alanine scanning of compstatin (I[CVVQDWGHHRC]T, addition, the replacement of Cys2 and Cys12 with alanine
IC50 = 12 µM) was extensively investigated to identify the yielded an inactive linear analog [30]. In a recent study, a
contribution of each amino acid residue for its activity. An reduction-resistant cystathionine (Cth) bridge was introduced
alanine scan was performed within the cyclic loop of comp- as an isostere structure to replace the disulfide bond of comp-
statin analog ([CVVQDWGHHRC], the notation is aligned with statin (Figure 4) [35]. Compared with the original compstatin
compstatin sequence) [27]. This cyclic loop still retains the (Y = Z = S; IC50 = 1.9 µM), the δ-Cth-containing analog largely
activity (IC50 = 33 µM), however, studies of short constrained maintains the binding and inhibitory properties (Y = S,
peptides with any additional deletion have shown that 11- Z = CH2; IC50 = 3.1 µM), while the γ-Cth-containing analog
membered peptide between disulfide-linked Cys2 and Cys12 (Y = CH2, Z = S; IC50 > 20 µM) is nearly inactive. This suggests
constitutes a minimum structure required for optimal activity that the sulfur-to-methylene substitution on the C-terminal
[30]. The replacement of His9 by Ala yielded two-fold increase side of the original disulfide is unfavorable for the structural
in the inhibitory activity, while replacements of Val4, His10, and functional properties of compstatin. Although the δ-Cth-
and Arg11 by Ala yielded two-fold decrease in the inhibitory
activity. This suggests that Val4, His9, His10, and Arg11 do not
contribute significantly to binding with C3. However, replace-
ments of Val3, Gln5, Asp6, Trp7 and Gly8 by Ala yielded
dramatic decrease in the inhibitory activity. Furlong and cow-
orkers further studied alanine scanning of the full length
compstatin, confirming that replacements of Val4, His9 and Gly8
Arg11 by Ala resulted in minimal change in the functional Trp7
activity [31]. In contrast, replacements of Val3, Gln5, Trp7,
Gly8 and His10 by Ala yielded analogs that were inactive.
These results combined with the biophysical studies of the Asp6
compstatin structure indicate that a type I β-turn comprising
the segment Gln5-Asp6-Trp7-Gly8 is critical for its activity [32]. Gln5
However, the rest of the peptide is mainly disordered and
amenable to further optimization. The residues Gln5-Asp6-
Trp7-Gly8 substituted with Gly-Pro-Phe-Gly, which has the Figure 2. The β-turn segment (Gln5-Asp6-Trp7-Gly8) of compstatin is indispen-
propensity to form type I β-turn, yielded an inactive analog sable for high inhibitory activity.
4 Y. HUANG
1000-fold increase in both potency (IC50 = 62 nM) and binding Table 1. The six pharmacophore features of compstatin [41].
affinity for C3b (KD = 2.3 nM) over that of the original comp- Radius
statin. Biophysical analysis using surface plasmon resonance ID Feature Center (Å)
and isothermal titration calorimetry suggests that the F1 Aromatic ring Center of mass of W4 indole ring 1.2
F2 Hydrogen-bond Carbonyl oxygen on the Q5 side 0.8
improved binding affinity originates from more favorable acceptor chain
free conformation and stronger hydrophobic interactions. F3 Hydrogen-bond donor Amide nitrogen on the Q5 side chain 0.8
The considerable efforts on the generation of complement F4 Hydrogen-bond Carbonyl oxygen on the D6 side 2.2
acceptor chain
inhibitors led to the discovery of a great number of compsta- F5 Hydrogen-bond donor Indole nitrogen of W7 0.8
tin analogs. A quantitative structure–activity relationship F6 Hydrophobe Centroid of two Sγ atoms of C2 and 0.7
(QSAR) model was generated to correlate key physicochemical C12
and geometrical properties of compstatin analogs with their
inhibitory activities [40]. The number of aromatic bonds in the
non-human primates showed that these analogs have highly
side chain of residue 4 (b_ar_4), the hydrophobic patch size
beneficial plasma half-life values (CP20, half life = 9.3 h; CP30,
near the disulfide bond (hyd_patch_surf), the solvent-accessi-
half life = 10.1 h; CP40, half life = 11.8 h). In addition, there
ble surface area occupied by nitrogen atoms of basic amino
are strong indications that these compstatin analogs actually
acid residues (base_N_surf), and the hydrophobicity of residue
follow a target-driven model since C3 is a highly abundant
4 (polar_4) correlate strongly with the inhibitory activity of
plasma protein.
compstatin analogs (Figure 7). This simple QSAR model is
In 2014, conjugates of complement inhibitor compstatin
highly accurate and statistically significant, thus provides the
and albumin binding molecules were reported [46]. This
basis for the rational design of active compstatin analogs even
study resulted in the most potent C3b ligand with subnano-
in the absence of a ligand-receptor complex. Later, a 3D
molar binding affinity (KD = 150 pM) in Figure 8. The interac-
pharmacophore model was identified as a filter with great
tions of ABM2-CP20 with albumins from different species
sensitivity and specificity to discriminate 82 active compstatin
(human, baboon, bovine, rabbit and mouse) were character-
analogs [41]. Taking advantage of the co-crystal structure of
ized. The competition experiment showed that ABM2-CP20
C3c-compstatin complex, six structural features were built into
binds primarily to site II on HSA. Albumin binding molecule
individual pharmacophore points based on the two most
is able to improve the plasma protein binding of ABM2-CP20,
active compstatin analogs (Table 1). Consistent with the
which suggests its favorable pharmacokinetic property.
QSAR study, the aromatic ring of residue 4 (F1) and the
The site specific PEGylation of compstatins was reported in
hydrophobe on the disulfide bond (F6) were identified as the
an effort to increase the half life in vivo [47]. A two-arm
most critical pharmacophore features in determining the inhi-
branched PEG chain (40 kDa) was conjugated to the lysine ε-
bitory activity. Owing to the enriched knowledge in the struc-
amino group of Ac-CP40-K, and the N-terminal α-amino group
tural features, tremendous efforts have been devoted to the
of CP40, respectively (Figure 9). In a preliminary in vivo study,
computational and rational design of new compstatin variants
the pharmacokinetic profile of pegylated compstatin has
over the years [42–44].
shown a significant feature for the treatment of chronic dis-
log10 IC50;Rel ¼ 0:1573b ar 4 þ 0:0034hyd patch surf eases. The half life is around 5.5 days (increased by over 11
þ0:0058base N surf 0:1266polar 4 folder compared with the non-pegylated peptide CP40) in
1:8572 n¼50; R2 adj ¼ 0:89; Q2 ¼ 0:88; s¼ 0:3023; sCV ¼ 0:3167: baboons indicating a feasible option for clinical
Figure 7. QSAR model of compstatin ([IC50,Rel]: IC50 relative to that of the parent compstatin).
6 Y. HUANG
administration; the flat concentration curve shows its potential step in the activation of the human complement system
for the systematic inhibition of complement activation. makes compstatin an attractive therapeutic candidate for the
treatment of inflammatory diseases. In 2000, compstatin was
evaluated to treat protamine/heparin-induced complement
4. Therapeutic development of compstatins activation, the inflammatory response commonly induced by
cardiac surgery and cardiopulmonary bypass [53]. A combina-
Compstatin has shown high species selectivity to human and
tion of bolus injection and infusion of compstatin at a total
primate complement systems, which limits the preclinical
dose of 21 mg/kg in baboons can completely inhibit the
development in animal models using lower mammalian spe-
generation of C3 activation products without adverse effects
cies [48]. The SPR analysis showed that compstatin binds to C3
on heart rate, blood pressure or hematological parameters. In
proteins from human and baboon, however, it does not bind
2002, compstatin (Ac-I[CVVQDWGHHRC]T-NH2) was used to
to C3 proteins form mouse or rat. In addition, compstatin does
study the role of complement in Escherichia coli induced
not bind to two structural homologs of C3, human C4 and C5.
inflammatory responses [54]. It was found that compstatin
Therefore, the inhibitory action of compstatin on complement
can efficiently reduce Escherichia coli induced oxidative burst
activation is solely due to its activity against C3. Recently, the
and MPO release, as well as the complement activation in
molecular dynamics simulations suggested that the rat C3
whole blood. In addition, IL-8 secretion was abolished by the
protein undergoes local conformational changes, which dis-
combination of compstatin and anti-CD14.
rupt polar and nonpolar interactions with compstatin and
Compstatin has shown great therapeutic potential for the
reduce the stability of the complex [49]. So far, compstatin
treatment of transplantations to reduce complement-
analogs have been shown to be safe and effective in a series
mediated organ injury and xenograft rejection. In 1999, pig
of ex vivo and in vivo experiments, and hold great promise for
kidneys, which were perfused with fresh human blood con-
further clinical investigations [15,50]. As the first-in-class com-
taining compstatin, had significantly longer graft survival [55].
plement inhibitor, compstatin can offer great therapeutic
C3 activation products and terminal complement complex
opportunities with regard to many diseases, in which inap-
were effectively controlled by the administration of compsta-
propriate or uncontrolled activation of complement is
tin. This study suggests that compstatin may be a useful
involved in the disease pathogenesis (Table 2).
clinical candidate to delay or prevent hyperacute rejection. In
Studies using animal models have shown that compstatin
2008, compstatin (Ac-I[CV(1MeW)QDWGAHRC]T-NH2) was
has excellent safety and efficacy profiles as a complement
used to treat xenogeneic instant blood-mediated inflamma-
inhibitor, due to its ability to specifically block the comple-
tory reaction (IBMIR) in a non-human primate model of pan-
ment activation at the C3 level [4,5]. The ability to inhibit a key
creatic islet transplantation [56]. The in vitro study showed
that compstatin significantly reduced the binding of C3b/
Table 2. Studies of compstatin used in disease models. iC3b to the islets, and completely inhibited the immediate
Timeline Disease models Subjects Reference destructive immunoglobulin-triggered complement activation.
1998 Extracorporeal Human blood (in vitro) [59] In a human model of clinical islet transplantation, it has been
circulation shown that compstatin (Ac-I[CV(1MeW)QDWGAHRC]T-NH2)
1999 Transplantation Pig kidney (ex vivo) [55]
2000 Inflammation Baboon (in vivo) [53] can block the antibody-mediated complement attack on the
2002 Inflammation Human blood (in vitro) [54] islets, indicating compstatin as a prime candidate for eliminat-
2007 Age-related macular Human patients (phase 1) [51,66] ing complement damage to the islet graft [57].
degeneration
2008 Transplantation Pig/human islet (in vitro) [56,57] It has been known that the artificial surface and biomater-
2008 Age-related macular Rabbit, monkey (in vivo) [52,67] ials used in many clinical settings can cause whole blood
degeneration inflammatory reaction due to complement and cellular activa-
2010 Sepsis Baboon (in vivo) [69]
2010 Hemodialysis Human blood (ex vivo) [63] tion [58]. Compstatin is a promising therapeutic agent to
2013 Paroxysmal nocturnal Patient-derived erythrocytes (in [47] inhibit complement-associated bioincompatibility reactions in
hemoglobinuria vitro), monkey (in vivo) clinical applications. In 1998, compstatin was evaluated in two
2015 C3 glomerulopathy Human blood (in vitro) [71]
2016 Paroxysmal nocturnal Human patients (phase 1) [74] in vitro models of extracorporeal circulation using whole
hemoglobinuria human blood [59]. Compstatin (I[CWQDWGAHRC]T) effectively
2016 Periodontitis Monkey (in vivo) [72] inhibited the complement activation, preventing the
EXPERT OPINION ON DRUG DISCOVERY 7
activation and binding of polymorphonuclear leukocytes to study showed that complement inhibition with compstatin
the biomaterial surface. It has been shown that complement (Ac-I[CV(1MeW)QDWGAHRCT]I-NH2) reduced erythrocyte bind-
activation on a model biomaterial surface is mainly mediated ing and bacterial C3 opsonization [70]. Therefore, compstatin
by the alternative pathway [60]. Independent of the types of is a potentially important therapeutic agent for blocking the
biomaterial surface, compstatin was found to reduce the acti- harmful effects of complement activation products during the
vation of neutrophils in polymer-exposed blood, thus reduce organ failure stage of severe sepsis.
the risk of biomaterial-mediated inflammatory reactions [61]. Amyndas Pharmaceuticals founded by Professor John
The further study showed that the artificial surface markedly Lambris has devoted to the development of next-generation
induced a broad spectrum of proinflammatory chemokines Compstatins for clinical applications. PNH is chronic, comple-
and growth-factors, which was largely reduced by the treat- ment-mediated, intravascular hemolysis which affects 8,000–
ment of compstatin (Ac-I[CV(1MeW)QDWGAHRC]T-NH2) [62]. 10,000 people in North America and Europe. CP40 demon-
Therefore, it is a promising therapeutic strategy for compsta- strated its ability to efficiently prevent C3 activation and opso-
tins to prevent biomaterials trigged complement activation in nization on PNH erythrocytes in vitro (IC50 ~ 4 µM), while
patients on hemodialysis. In 2010, an ex vivo hemodialysis saturating inhibitor concentration could be reached with
model was used to evaluate compstatins (Ac-I[CV(1MeW) CP40 through repetitive subcutaneous administration in cyno-
QDWGAHRC]T-NH2 and Ac-I[CV(1MeW)QDWSarAHRC]I-NH2, molgus monkeys [47]. In 2014, both the European Medicines
termed as CP10) for the treatment of complement-related Agency (EMA) and the U.S. Food and Drug Administration
adverse effects [63]. It has been shown that compstatins are (FDA) granted AMY-101 (based on CP40) as an orphan drug
able to attenuate the hemodialysis filter fibers induced com- designation for the treatment of PNH. The first-in-human clin-
plement activation, and reduce the tissue factor dependent ical study of AMY-101 has been initiated in 2017
procoagulant activity of polymorphonuclear leukocytes during (NCT03316521). Dense deposit disease (DDD) and C3 glomer-
hemodialysis. ulonephritis (C3GN) are rare forms of glomerulonephritis
Compstatin has been investigating for the treatment of which result from abnormal regulation of the alternative com-
local and systemic inflammation in age-related macular plement pathway. CP40 is able to prevent complement-
degeneration (AMD), which is mediated by the deregulated mediated lysis of sheep erythrocytes in sera from C3 glomer-
action of the alternative pathway of the complement system ulopathy (C3G) patients, which holds the promise to C3G
[64,65]. The complement inhibitors may offer a new paradigm patients as a disease-specific, targeted therapy [71]. In 2016,
to prevent, retard, or even reverse AMD, a leading cause of Amyndas Pharmaceuticals announced that AMY-101 was
global blindness [66]. The treatment of compstatin (Ac-I[CV granted an orphan drug designation by EMA and FDA for
(1MeW)QDWGAHRC]T-NH2) on cynomolgus monkeys with the treatment of C3G. Human periodontitis is a prevalent
early-onset macular degeneration can suppress the formation chronic oral disease for nearly half of adults, which is asso-
of drusen, which contains active complement molecules and ciated with over-activation of complement. CP40 has been
represents a risk factor for dry-type AMD [67]. As one of a few shown to inhibit pre-existing chronic periodontal inflamma-
drugs discovered in academia, compstatin was licensed from tion and osteoclastogenesis in non-human primates, suggest-
the University of Pennsylvania to Potentia Pharmaceuticals in ing its potential as a novel adjunctive anti-inflammatory
2006 in the attempt to enter clinical trials. In 2007, Potentia therapy for treating human periodontitis [72]. In addition,
Pharmaceuticals started a Phase I clinical trial (NCT00473928) CP40 has been under evaluation as a potential treatment of
using a compstatin analog POT-4 for the treatment of neovas- complications of hemodialysis and ABO incompatible kidney
cular AMD [68]. Later Potentia signed a licensing and purchase transplantation [73].
agreement with Alcon to develop its leading drug candidate, Apellis Pharmaceuticals acquired Potenita in 2014 and is
POT-4. In 2012, Alcon completed a Phase II clinical trial devoted to the clinical development of APL-1 (POT-4 formu-
(NCT01157065) using a compstatin analog AL-78898A for the lated for inhaled administration) and APL-2 (PEGylated APL-1,
treatment of exudative AMD. In 2013, Alcon terminated a formulated for subcutaneous and intravitreal administration)
multicenter Phase II clinical trial (NCT01603043) using AL- in several diseases and conditions including COPD and AMD.
78898A for the treatment of geographic atrophy associated Apellis completed phase 1b study of APL-2 for PNH patients
with AMD. Compstatin was administrated via single intravi- with daily subcutaneous administration [74]. On 20 December
treal injection, and the gel deposits were formed to slowly 2016, FDA granted Fast Track designation to the development
release active drug into the vitreous cavity over several program for APL-2 in the treatment of patients with PNH, who
months. continue to experience hemolysis and require RBC transfu-
Compstatin induced complement inhibition was investi- sions despite receiving therapy with eculizumab. On 10
gated as a promising approach for treating sepsis and multiple August 2017, Apellis announced the closing of $60 million
organ failure, which involve massive activation of coagulation series E finacing that would be used to advance trials of
and complement cascades [69]. Compstatins (Ac-I[CV(1MeW) APL-2 in PNH. Based on these encoraging efforts, compstatin
QDWGAHRCT]I-NH2) was administered after Escherichia coli is likely to be further developed for the treatment of PNH.
challenge in a baboon model of sepsis-induced multiple
organ failure. Compstatin infusion rapidly inhibited C3a and
5. Conclusion
C3b generation in septic baboons, reduced sepsis-induced
leucopenia and thrombocytopenia, and lowered the accumu- All in all, compstatin family has shown beneficial effects in a
lation of macrophages and platelets in organs. The further number of disease models, and it holds the promise as a
8 Y. HUANG
versatile therapeutic drug for implications against unregulated great potential for next generation complement therapeutics
complement activation in human and primates. We can fore- to benefit patients in the coming years.
see that more and more studies using compstatin as a valu-
able tool in immunological and translational research for
blocking the effect of the complement cascade. It is exciting Acknowledgments
to see clinical advances of next-generation compstatins in new This manuscript is dedicated to Professor John D Lambris for his contribution
therapeutic implications for a variety of disease conditions. in the discovery and development of compstatin. The author offers his
thanks to Professor Lambris (Department of Pathology & Laboratory
Medicine at University of Pennsylvania, USA) and Professor Daniel Ricklin
(Department of Pharmaceutical Sciences at University of Basel, Switzerland).
6. Expert opinion
Considering long-term treatment in chronic diseases (such as
Funding
PNH, aHUS), there is a critical need to develop therapies with
simple administration and reduced dose schedules. Substantial This manuscript has not been funded.
challenges still remain to develop effective therapeutics which
can maintain systemic complement inhibition, since the circulat-
ing levels of complement proteins are usually high and turnover Declaration of interest
rates are relatively rapid, which require the frequent dosing of The author of this manuscript is an employee of WuXi AppTec Inc. They
complement inhibitors. Therefore, it is very essential for new have no other relevant affiliations or financial involvement with any
generations of compstatin derivatives with favarable features organization or entity with a financial interest in or financial conflict
with the subject matter or materials discussed in the manuscript apart
and feasibility as chronic complement therapeutics such as from those disclosed. Peer reviewers on this manuscript have no relevant
improved pharmacokinetics and suitable dosing routes. Further financial or other relationships to disclose.
study is needed to optimize in vivo performance of compstatins
as complement inhibitors, which will greatly facilitate the devel-
opment of chronic complement therapeutics to modulate the References
immune response in patients with autoimmune and inflamma-
Papers of special note have been highlighted as either of interest (•) or of
tory diseases. In addition, an orally active compstatin formula- considerable interest (••) to readers.
tion would have great advantages over conventional injectable 1. Zipfel PF, Skerka C. Complement regulators and inhibitory proteins.
therapies (such as intravenous injection and infusion) for Nat Rev Immunol. 2009;9(10):729–740.
patients with complement mediated chronic inflammation. 2. Holers VM. The spectrum of complement alternative pathway-
However, oral administration of compstatin still remains chal- mediated diseases. Immunol Rev. 2008;223(1):300–316.
3. Ricklin D, Hajishengallis G, Yang K, et al. Complement: a key
lenge due to proteolytic degradations and poor bioavailability. system for immune surveillance and homeostasis. Nat
There are particular challenges and opportunities for comple- Immunol. 2010;11(9):785–797.
ment based drug development trageting rare diseases, which 4. Wagner E, Frank MM. Therapeutic potential of complement
require tremendous efforts on industry and academic collabar- modulation. Nat Rev Drug Discov. 2010;9(1):43–56.
5. Ricklin D, Lambris JD. Complement-targeted therapeutics. Nat
tion. Eculizumab (marketed as Soliris) is a good example of such
Biotech. 2007;25(11):1265–1275.
case. It was developed by Alexion Pharmaceuticals as the first 6. Makrides SC. Therapeutic inhibition of the complement system.
approved terminal complement inhibitor for the treatment of Pharmacol Rev. 1998;50(1):59–88.
PNH and aHUS, two life-threatening, ultra-rare disorders caused 7. Rother RP, Rollins SA, Mojcik CF, et al. Discovery and develop-
by uncontrolled complement activation. Eculizumab is often ment of the complement inhibitor eculizumab for the treat-
ment of paroxysmal nocturnal hemoglobinuria. Nat Biotech.
considered as the most expensive drug in the United States
2007;25(11):1256–1264.
averaging $18,000 per dose or about $500,000 annually. In 8. Garjau M, Azancot M, Ramos R, et al. Early treatment with
2011, Alexion acquired Taligen Therapeutics in an effort to eculizumab in atypical haemolytic uraemic syndrome. Clin
further develop TT30 (ALXN 1102) [75]. It is critical for drug Kidney J. 2012;5(1):31–33.
development programs to get industry and academic collabar- 9. Ariceta G, Arrizabalaga B, Aguirre M, et al. Eculizumab in the
tion earlier in order to enable moving forward to clinical trials treatment of atypical hemolytic uremic syndrome in infants. Am J
Kidney Dis. 2012;59(5):707–710.
faster. In addition, the development of cost-effective treatment 10. Schrezenmeier H, Höchsmann B. Drugs that inhibit complement.
options is also important for patients with complement Transfus Apher Sci: Off J World Apher Assoc: Off J Eur Soc
mediated chronic diseases. It may need the attention and efforts Haemapher. 2012;46(1):87–92.
from major pharmaceutical companies in order to reduce the 11. Holland MCH, Morikis D, Lambris JD. Synthetic small-molecule
complement inhibitors. Current Opinion Investigational Drugs.
development expense and eventually the cost to patients and
2004;5(11):1164–1173. London, England: 2000.
societies. 12. John L, Dimitrios M. The building blocks of the complement sys-
There are tremendous opportunities for the development tem. In: Dimitrios Morikis and John D. Lambris (eds). Structural
of therapeutics to target complement system, providing safe biology of the complement system. Boca Raton, FL: CRC Press;
and effective treatment for a wide range of diseases. 2005:1–18.
13. Sahu A, Lambris JD. Structure and biology of complement protein
Therapeutic targets and their inhibitors with promising drug
C3, a connecting link between innate and acquired immunity.
properties have been identified over the years [76]. It is ben- Immunol Rev. 2001;180:35–48.
eficial for the field to gain more knowledge from the clinical 14. Mollnes TE, Kirschfink M. Strategies of therapeutic complement
trials invovling any type of complement inhibitors. There is inhibition. Mol Immunol. 2006;43(1–2):107–121.
EXPERT OPINION ON DRUG DISCOVERY 9
15. Ricklin D, Lambris JD. Compstatin: a complement inhibitor on • An interesting article on the development of POT-4 as clinical
its way to clinical application. In: Lambris JD, Editor. Current candidate.
topics in complement Il. Springer-Verlag, New York. 2008. p. 35. Knerr PJ, Tzekou A, Ricklin D, et al. Synthesis and activity of thioether-
273–292. containing analogues of the complement inhibitor compstatin. ACS
16. Sahu A, Kay BK, Lambris JD. Inhibition of human complement Chem Biol. 2011;6(7):753–760.
by a C3-binding peptide isolated from a phage-displayed ran- 36. Klepeis JL, Floudas CA, Morikis D, et al. Integrated computational and
dom peptide library. J Immunol. 1996;157(2):884–891. experimental approach for lead optimization and design of compstatin
• The first C3 binding peptide was reported in this paper. variants with improved activity. J Am Chem Soc. 2003;125(28):8422–
17. Janssen BJC, Gros P. Structural insights into the central com- 8423.
plement component C3. Mol Immunol. 2007;44(1–3):3–10. 37. Mallik B, Katragadda M, Spruce LA, et al. Design and NMR characteriza-
18. Janssen BJC, Huizinga EG, Raaijmakers HC, et al. Structures of comple- tion of active analogues of compstatin containing non-natural amino
ment component C3 provide insights into the function and evolution of acids. J Med Chem. 2005;48(1):274–286.
immunity. Nature. 2005;437(7058):505–511. 38. Magotti P, Ricklin D, Qu H, et al. Structure-kinetic relationship analysis of
19. Gros P, Milder FJ, Janssen BJC. Complement driven by conformational the therapeutic complement inhibitor compstatin. J Mol Recognition.
changes. Nat Rev Immunol. 2008;8(1):48–58. 2009;22(6):495–505.
20. Janssen BJC, Christodoulidou A, McCarthy A, et al. Structure of C3b 39. Qu H, Magotti P, Ricklin D, et al. Novel analogues of the therapeutic
reveals conformational changes that underlie complement activity. complement inhibitor compstatin with significantly improved affi-
Nature. 2006;444(7116):213–216. nity and potency. Mol Immunol. 2011;48(4):481–489.
21. Kolln J, Bredehorst R, Spillner E. Engineering of human complement 40. Mulakala C, Lambris JD, Kaznessis Y. A simple, yet highly
component C3 for catalytic inhibition of complement. Immunol Lett. accurate, QSAR model captures the complement inhibitory
2005;98(1):49–56. activity of compstatin. Bioorg Med Chem. 2007;15(4):1638–
22. Leinhase I, Schmidt OI, Thurman JM, et al. Pharmacological 1644.
complement inhibition at the C3 convertase level promotes 41. Chiu T-L, Mulakala C, Lambris JD, et al. Development of a new
neuronal survival, neuroprotective intracerebral gene expres- pharmacophore model that discriminates active compstatin
sion, and neurological outcome after traumatic brain injury. analogs. Chem Biol Drug Des. 2008;72(4):249–256.
Exp Neurol. 2006;199(2):454–464. • The pharmacophore model of compstatin analogs.
23. Schepers A, de Vries MR, van Leuven CJ, et al. Inhibition of 42. Tamamis P., López de Victoria A, Gorham RD Jr.. et al.
complement component C3 reduces vein graft atherosclerosis Molecular dynamics in drug design: new generations of comp-
in apolipoprotein E3-leiden transgenic mice. Circulation. statin analogs. Chem Biol Drug Des. 2012;79(5):703–718.
2006;114(25):2831–2838. 43. Bellows ML, Fung HK, Taylor MS, et al. New compstatin variants through
24. Katragadda M, Morikis D, Lambris JD. Thermodynamic studies two De Novo protein design frameworks. Biophys J. 2010;98(10):2337–
on the interaction of the third complement component and its 2346.
inhibitor, compstatin. J Biol Chem. 2004;279(53):54987–54995. 44. Lopez de Victoria A., Gorham RD Jr., Bellows-Peterson ML, et al. A new
25. Soulika AM, Holland MC, Sfyroera G, et al. Compstatin inhibits generation of potent complement inhibitors of the compstatin family.
complement activation by binding to the beta-chain of comple- Chem Biol Drug Des. 2011;77(6):431–440.
ment factor 3. Mol Immunol. 2006;43(12):2023–2029. 45. Qu H, Ricklin D, Bai H, et al. New analogs of the clinical comple-
26. Janssen BJC, Halff EF, Lambris JD, et al. Structure of compstatin ment inhibitor compstatin with subnanomolar affinity and
in complex with complement component C3c reveals a new enhanced pharmacokinetic properties. Immunobiology.
mechanism of complement inhibition. J Biol Chem. 2007;282 2013;218:496–505.
(40):29241–29247. •• The development and evaluation of next generation
•• The co-crystal structure of compstatin was reported in this compstatins.
paper. 46. Huang Y, Reis ES, Knerr PJ, et al. Conjugation to albumin-binding
27. Morikis D, Assa-Munt N, Sahu A, et al. Solution structure of comp- molecule tags as a strategy to improve both efficacy and pharma-
statin, a potent complement inhibitor. Protein Sci. 1998;7(3):619– cokinetic properties of the complement inhibitor compstatin.
627. Chem Med Chem. 2014;9(10):2223–2226.
28. Morikis D, Roy M, Sahu A, et al. The structural basis of compstatin 47. Risitano AM, Ricklin D, Huang Y, et al. Peptide inhibitors of C3
activity examined by structure-function-based design of peptide activation as a novel strategy of complement inhibition for the
analogs and NMR. J Biol Chem. 2002;277(17):14942–14953. treatment of paroxysmal nocturnal hemoglobinuria. Blood.
29. Soulika AM, Morikis D, Sarrias MR, et al. Studies of structure- 2014;123(13):2094–2101.
activity relations of complement inhibitor compstatin. J •• The study of next generation compstatins for the PNH disease
Immunol. 2003;171(4):1881–1890. model.
30. Sahu A, Soulika AM, Morikis D, et al. Binding kinetics, struc- 48. Sahu A, Morikis D, Lambris JD. Compstatin, a peptide inhibitor
ture-activity relationship, and biotransformation of the comple- of complement, exhibits species-specific binding to comple-
ment inhibitor compstatin. J Immunol. 2000;165(5):2491–2499. ment component C3. Mol Immunol. 2003;39(10):557–566.
31. Furlong ST, Dutta AS, Coath MM, et al. C3 activation is inhibited by 49. Tamamis P, Morikis D, Floudas CA, et al. Species specificity of
analogs of compstatin but not by serine protease inhibitors or the complement inhibitor compstatin investigated by all-atom
peptidyl alpha-ketoheterocycles. Immunopharmacology. 2000;48 molecular dynamics simulations. Proteins-Structure Funct
(2):199–212. Bioinform. 2010;78(12):2655–2667.
32. Lambris J, Dimitrios M Structure, dynamics, activity, and function of 50. Qu H, Ricklin D, Lambris JD. Recent developments in low molecular
compstatin and design of more potent analogues. In: Dimitrios weight complement inhibitors. Mol Immunol. 2009;47(2–3):185–
Morikis and John D. Lambris (eds). Structural biology of the com- 195.
plement system. Boca Raton, FL: CRC Press; 2005. 317–340. 51. Deschatelets P, Olson P, Francois C. Methods of treating age-
33. Katragadda M, Lambris JD. Expression of compstatin in related macular degeneration and associated diseases with comp-
Escherichia coli: incorporation of unnatural amino acids statin and compstatin analogs. US8168584 B2. 2012. p. 57.
enhances its activity. Protein Expr Purif. 2006;47(1):289–295. 52. Francois C, Deschatelets P, Olson P. Sustained delivery of comp-
34. Katragadda M, Magotti P, Sfyroera G, et al. Hydrophobic effect statin analogs from gels. WO 2009046198 A2.
and hydrogen bonds account for the improved activity of a 53. Soulika AM, Khan MM, Hattori T, et al. Inhibition of heparin/
complement inhibitor, compstatin. J Med Chem. 2006;49 protamine complex-induced complement activation by comp-
(15):4616–4622. statin in baboons. Clin Immunol. 2000;96(3):212–221.
10 Y. HUANG
54. Mollnes TE, Brekke OL, Fung M, et al. Essential role of the C5a receptor in 67. Chi ZL, Yoshida T, Lambris JD, et al. Suppression of Drusen
E-coli-induced oxidative burst and phagocytosis revealed by a novel formation by compstatin, a peptide inhibitor of complement C3
lepirudin-based human whole blood model of inflammation. Blood. activation, on Cynomolgus monkey with early-onset macular
2002;100(5):1869–1877. degeneration, in inflammation and retinal disease. Adv Exp
55. Fiane AE, Mollnes TE, Videm V, et al. Compstatin, a peptide inhibitor of Med Biol. 2010;703:127–135.
C3, prolongs survival of ex vivo perfused pig xenografts. 68. Kaushal S, Grossi F, Francois C, et al. Complement C3 inhibitor
Xenotransplantation. 1999;6(1):52–65. POT-4: clinical safety of intravitreal administration. ARVO
56. Goto M, Tjernberg J, Dufrane D, et al. Dissecting the instant blood- Annual Meeting Abstract; Fort Lauderdale, FL. 2009.
mediated inflammatory reaction in islet xenotransplantation. • The first clinical trial of compstatin.
Xenotransplantation. 2008;15(4):225–234.
57. Tjernberg J, Ekdahl KN, Lambris JD, et al. Acute antibody-mediated 69. Silasi-Mansat R, Zhu H, Popescu NI, et al. Complement inhibi-
complement activation mediates lysis of pancreatic islets cells and tion decreases the procoagulant response and confers organ
may cause tissue loss in clinical islet transplantation. Transplantation. protection in a baboon model of Escherichia coli sepsis. Blood.
2008;85(8):1193–1199. 2010;116(6):1002–1010.
58. Nilsson B, Ekdahl KN, Mollnes TE, et al. The role of complement in 70. Brekke OL, Hellerud BC, Christiansen D, et al. Neisseria menin-
biomaterial-induced inflammation. Mol Immunol. 2007;44(1–3):82–94. gitidis and Escherichia coli are protected from leukocyte pha-
59. Nilsson B, Larsson R, Hong J, et al. Compstatin inhibits complement and gocytosis by binding to erythrocyte complement receptor 1 in
cellular activation in whole blood in two models of extracorporeal human blood. Mol Immunol. 2011;48(15–16):2159–2169.
circulation. Blood. 1998;92(5):1661–1667. 71. Zhang Y, Shao D, Ricklin D, et al. Compstatin analog Cp40
60. Andersson J, Ekdahl KN, Lambris JD, et al. Binding of C3 fragments on inhibits complement dysregulation in vitro in C3 glomerulo-
top of adsorbed plasma proteins during complement activation on a pathy. Immunobiology. 2015;220(8):993–998.
model biomaterial surface. Biomaterials. 2005;26(13):1477–1485. 72. Maekawa T, Briones RA, Resuello RR, et al. Inhibition of pre-
61. Schmidt S, Haase G, Csomor E, et al. Inhibitor of complement, comp- existing natural periodontitis in non-human primates by a
statin, prevents polymer-mediated Mac-1 up-regulation of human neu- locally administered peptide inhibitor of complement C3. J
trophils independent of biomaterial type tested. J Biomed Mater Res. Clin Periodontol. 2016;43(3):238–249.
2003;66A(3:491–499. 73. Mastellos DC, Yancopoulou D, Kokkinos P, et al. Compstatin: a
62. Lappegård KT, Bergseth G, Riesenfeld J, et al. The artificial surface- C3-targeted complement inhibitor reaching its prime for bed-
induced whole blood inflammatory reaction revealed by increases in side intervention. Eur J Clin Invest. 2015;45:423–440.
a series of chemokines and growth factors is largely complement 74. Grossi FV, Bedwell P, Deschatelets P, et al. APL-2, a comple-
dependent. J Biomed Mater Res. 2008;87A(1:129–135. ment C3 inhibitor for the potential treatment of paroxysmal
63. Kourtzelis I, Markiewski MM, Doumas M, et al. Complement anaphyla- nocturnal hemoglobinuria (PNH): phase I data from two com-
toxin C5a contributes to hemodialysis-associated thrombosis. Blood. pleted studies in healthy volunteers. ASH 58th Annual Meeting
2010;116(4):631–639. & Exposition, Abstract # 1251; San Diego, CA. 2016.
64. Bradley DT, Zipfel PF, Hughes AE. Complement in age-related macular •• The study of a leading candidate in clinical trial.
degeneration: a focus on function. Eye. 2011;25(6):683–693.
65. Zhou J, Kim SR, Westlund BS, et al. Complement activation by bisreti- 75. Fridkis-Hareli M, Storek M, Mazsaroff I, et al. Design and devel-
noid constituents of RPE lipofuscin. Invest Ophthalmol Vis Sci. 2009;50 opment of TT30, a novel C3d-targeted C3/C5 convertase inhi-
(3):1392–1399. bitor for treatment of human complement alternative
66. Troutbeck R, Al-Qureshi S, Guymer RH. Therapeutic targeting of the pathway-mediated diseases. Blood. 2011;118(17):4705–4713.
complement system in age-related macular degeneration: a review. Clin 76. Ricklin D, Lambris JD. New milestones ahead in complement-
Experiment Ophthalmol. 2012;40(1):18–26. targeted therapy. Semin Immunol. 2016;28(3):208–222.