Abstract
With the awareness that immune-inflammatory crosstalk is at the heart of many disorders, the desire for novel immunomodulatory strategies in the therapy of such diseases has grown dramatically. As a prime initiator and important modulator of immunological and inflammatory processes, the complement system has emerged as an attractive target for early and upstream intervention in inflammatory diseases and has moved into the spotlight of drug discovery. While prevalent conditions such as age-related macular degeneration have attracted the most attention, the diverse array of complement-mediated pathologies, with distinct underlying mechanisms, demands a multifaceted arsenal of therapeutic strategies. Fortunately, efforts in recent years have not only introduced the first complement inhibitors to the clinic but also filled the pipelines with promising candidates. With a focus on immunomodulatory strategies, this review discusses complement-directed therapeutic concepts and highlights promising candidate molecules.
Many concepts of modern drug discovery have deep roots in immunology research, with Paul Ehrlich’s notions of the “magic bullet” and his “side-chain theory” being among the most prominent examples (1). The connection between these two disciplines has even intensified in recent years in view of biological drugs such as mAb’s. Therapeutic immune modulation is increasingly recognized as a promising strategy for tackling inflammatory diseases, yet is in need of more selective modulators that help restore the immune balance and resolve inflammation (2). Fueled by the discovery of new functional roles, immune crosstalk mechanisms, and a growing number of disease associations (see accompanying review (3)), complement has emerged as focal point of interest in immunomodulatory and anti-inflammatory strategies (4). The variety of intervention points and high number of extracellular targets within the complement cascade (Fig. 1), the availability of potent natural inhibitor templates, and a spike in structure-function insight have all contributed to rapid advances in the field of complement-related drug discovery. While only two drugs with connection to complement are currently available in the clinic, ongoing research efforts have produced a plethora of innovative and diverse drug candidates that demonstrate great promise in many clinical conditions. In the following sections, we provide an overview of current therapeutic strategies and highlight drug candidates that are in preclinical or clinical development.
The therapeutic arsenal to tackle complement-related diseases
In view of the diversity and impact of complement-related disorders (3), potent complement inhibitors are desired yet a “one-size-fits-all” solution for therapeutic management is improbable. Fortunately, complement offers many intervention points from pattern recognition molecules that detect pathogen-and damage-associated surface structures, proteases that drive the activation cascade, opsonins that mark target cells for elimination, to anaphylatoxins that attract immune cells and mediate inflammatory responses (Fig. 1). With purified or recombinant C1 inhibitor concentrates (C1-INH; Table I) and the anti-C5 antibody Eculizumab (Alexion), first complement-directed drugs have meanwhile entered the clinic. C1-INH is a host serine protease inhibitor that was first recognized for its ability to regulate the activity of the C1 complex, yet also acts at the level of mannose binding lectin-associated serine proteases (MASP), thereby preventing complement initiation via the classical and lectin pathways (CP and LP, respectively). In fact, the specificity of C1-INH is even broader and includes serine proteases of the kinin, coagulation and fibrinolytic systems; in addition, it may exert anti-inflammatory effects through non-protease-directed mechanisms (5). Whereas the primary indication of C1-INH preparations, i.e., as a substitution therapy in hereditary angioedema (HAE), is likely mediated by the kinin rather than the complement system, the availability of a CP/LP inhibitor in the clinic enables exploring the drug in complement-related diseases. Indeed, C1-INH has shown promising effects in several disease models including myocardial infarction (6), transplantation (7), and, recently, in a mouse model of type I diabetes (8); clinical trials have been initiated for the attenuation of thromboinflammatory responses in trauma (ClinicalTrials.gov identifier NCT01275976) and kidney transplantation (NCT01147302, NCT01134510). In contrast to C1-INH, Eculizumab is complement-specific as this mAb binds to C5 and prevents its activation by the convertase and, consequently, the generation of C5a and the membrane attack complex (MAC). Eculizumab was originally approved for the orphan disease paroxysmal nocturnal hemoglobinuria (PNH), in which it prevents intravascular lysis of insufficiently protected erythrocytes with high efficacy. Meanwhile, indications for Eculizumab have been extended to include atypical hemolytic uremic syndrome (aHUS) where it was shown to improve clinical parameters like renal function or microangiopathy (9). Despite its high cost, the availability of this potent terminal pathway inhibitor has allowed off-label use of the drug, perhaps most noticeably for the treatment of hemolytic uremic syndrome caused by shiga toxin-producing E. coli during a recent outbreak in Europe (10). In addition, Eculizumab has been tested in various disease models and is currently being evaluated in clinical trials ranging from age-related macular degeneration (AMD) to transplantation (Table I).
Table I.
Compound Name | Manufacturer | Complement Target | Compound Class | Phase/Indication1 |
---|---|---|---|---|
C1 inhibitor2 (Cinryze, Berinert, Cetor, Ruconest) | ViroPharma, Lev/CSL Behring, Sanquin, Pharming/Santarus | C1r, C1s, MASP1, MASP2, other proteases | Regulator (purified or recombinant) | P4 (HAE), P3 (trauma), P1/2 (TP) |
Nafamostat2 (FUT-175, Futhan) | Torii, SK Chemicals | C1s, FD, other proteases | Small molecule | P4 (pancreatitis) P3 (acute kidney injury) |
[MASP2 program] | Omeros | MASP-2 | mAb | PC (AMD, TP) |
FCFD4514S, TNX-234 | Genentech | FD | mAb | P1 (AMD) |
TA106 | Alexion | FB | mAb | N/A |
Bikaciomab, NM9308 | Novelmed | FB | mAb | N/A |
NM9401 | Novelmed | Properdin | mAb | N/A |
CVF, HC3-1496 | InCode | C3 | Protein (recombinant) | PC (IRI, inflammation) |
Compstatin3 POT-4/AL-78898A Cp40/AMY-101 APL-1 |
Potentia/Alcon Amyndas Apellis |
C3, C3-conv | Peptide | P2 (AMD) PC (PNH, TP, Hemodialysis, aHUS, DDD) PC (Asthma, COPD) |
sCR1, CDX-1135 | Celldex | C3-conv, C4b, C3b | Regulator (recombinant) | PC/P1 (DDD) |
Mirococept, APT070 | (MRC)4 | C3-conv, C4b, C3b | Regulator (targeted) | P1/2 (TP) |
ALXN1102/ALXN11037 (TT30) | Alexion | C3-conv, C3b | Regulator (targeted) | P1 (PNH) |
rFH | [Ophtherion, Taligen]8 | C3-conv, C3b | Regulator (recombinant) | N/A |
mini-FH, AMY-201 | Amyndas | C3-conv, C3b | Regulator (engineered) | PC (PNH) |
5C6, AMY-301 | Amyndas | FH (recruitment) | Peptide | PC |
Eculizumab (Soliris) | Alexion | C5 | mAb | P4 (PNH, aHUS), P2/3 (STEC-HUS), P2 (ANCA-Vasculitis, AMD, CAD, chronic hemolysis), P1/2 (neuromyelitis optica), P1 (TP) |
Mubodina | Adienne | C5 | mAb (minibody) | PC (aHUS, DDD) |
Ergidina | Adienne | C5 | mAb (minibody, targeted) | PC (TP) |
ARC1905 | Ophthotech | C5 | Aptamer | P1 (AMD) |
LFG316 | Novartis, MorphoSys | (C5)6 | (mAb) | P1 (AMD) |
MEDI7814 | MedImmune, AstraZeneca | C5/C5a | mAb | P1 (COPD) |
NOX-D19 | Noxxon | C5a | Aptamer (Spiegelmer) | PC (Sepsis) |
IFX-1, CaCP29 | InflaRx | (C5a)6 | mAb | P1 (Sepsis) |
PMX53, PMX205 | Cephalon/Teva5 | C5aR | Peptidomimetic | N/A |
CCX168 | ChemoCentryx | C5aR | Small molecule | P2 (ANCA-Vasculitis) |
ADC-1004 | Alligator | C5aR | Protein | PC (IRI) |
Anti-C5aR-151, NN8209 Anti-C5aR-215, NN8210 |
Novo Nordisk | C5aR | mAb | P1 (RA) |
Imprime PGG | Biothera | CR3, other targets | Soluble beta-glucan | P1-P3 (cancer; add-on) |
Clinical development: PC = preclinical, P1–P3 = clinical trials phase 1–3, P4 = marketed (phase 4); Preclinical indications extracted from company websites etc. and may be subject to change.
Abbreviations: aHUS, atypical hemolytic uremic syndrome; AMD, age-related macular degeneration; ANCA, anti-neutrophil cytoplasmic antibodies; COPD, chronic obstructive pulmonary disease; DDD, dense deposit disease; HAE, hereditary angioedema; IRI, ischemia/reperfusion injury; PNH, paroxysmal nocturnal hemoglobinuria; RA, rheumatoid arthritis; STEC-HUS, hemolytic uremic syndrome caused by shiga toxin-releasing E. coli; TP, transplantation.
C1 inhibitor and Nafamostat also inhibit proteases outside the complement network; clinical effects may be not or partially complement-related
POT-4 is developed by Potentia/Alcon for AMD; Apelllis develops POT-4 for other indications; Amyndas develops advanced compstatin analogs
Clinical trials with Mirococept are performed by the UK Medical Research Council (MRC) using a government-sponsored grant
Cephalon has recently been acquired by Teva; no plans for the clinical development of PMX had been disclosed by Teva at the time of publication
Targets have not been officially disclosed for LFG316 and IFX-1
ALXN1103 is a subcutaneous formulation of ALXN1102/TT30
Recombinant FH (preclinically indicated for AMD) had been developed by Optherion and Taligen before both companies left the market
The pursuit of extending the arsenal of complement inhibitors for pathway or tissue-specific prevention of complement attack has produced various attractive candidates that cover a wide range of targets and applications. Traditionally, selective blockage of the CP has focused on its major activating serine protease C1s, yet attempts to arrive at small molecule inhibitors were hampered by poor specificity and pharmacokinetics (11). More recently, structure-guided discovery approaches and PEG-ylation were able to improve such shortcomings and may lead to clinical C1s inhibitors (12, 13). In addition to small molecule approaches, antibody-centered strategies to prevent pattern recognition by C1q have also shown promise; for example, the single chain antibody fragment scFv(QuVHVL) that binds to the globular heads of C1q was shown to block recognition of both IgG and C-reactive protein and to largely reduce CP activation by apoptotic cells (14).
Novel candidates have also emerged for LP-associated targets such as the MASP family. For example, Omeros runs a preclinical program that involves MASP-2-specific antibodies with potential application in aHUS and other conditions ranging from ischemic diseases to AMD (15). Indeed, anti-MASP-2 mAb’s have shown beneficial effects in mouse models of myocardial and gastrointestinal ischemia-reperfusion injury (IRI) (16). In addition, phage-display library screening using sunflower trypsin inhibitor as a template, produced peptides that are monospecific for MASP-1 and MASP-2, respectively, and inhibit the LP with nanomolar activity (17). Finally, the therapeutic potential of the endogenous LP regulator MBL/ficolin-associated protein-1 (MAP-1), which displaces MASP proteins from the mannose-binding lectin (MBL) complex, has been demonstrated in mouse models of myocardial infarction (18). Given the proposed roles of MASPs in the control of the alternative pathway (AP) and in linking to the coagulation system via activation of prothrombin (19), MASP-directed therapies may develop into interesting candidates for the treatment of multifaceted diseases.
Owing to the role of the AP as major amplifier of complement responses, contributing up to some 80% of activity even in the case of CP-mediated initiation (20), strategies that lead to comprehensive complement inhibition by targeting at the level of C3 have been actively pursued in recent years (21). Currently, the peptide compstatin and its analogs are the only inhibitors that act on native C3; by preventing activation of C3 by convertases, compstatin blocks C3b opsonization, amplification and generation of effectors. Despite specificity for human/primate C3, compstatin analogs have been tested in models ranging from sepsis to hemodialysis-induced thromboinflammation (11, 22, 23). In a primate model of AMD, intravitreal compstatin suppressed or reversed drusen formation (24), and a compstatin analog (AL-78898A, Alcon) is in clinical development for the treatment of AMD. The same analog (termed APL-1) is developed by Apellis for asthma and COPD. While peptidic drugs are often hampered by rapid plasma elimination, recent optimization efforts have produced compstatin analogs (e.g., AMY-101; Amyndas) that feature subnanomolar target affinities and plasma half-lifes of up to 12 hours (25), which is expected to facilitate systemic applications. In addition to compstatin, antibodies against C3b, factor B (FB), and factor D (FD) have been disclosed. For example, mAb S77 (Genentech) was shown to prevent convertase formation by blocking the binding area of FB on C3b (26), and the anti-C3b mAb 3E7 demonstrated high efficacy in a model of PNH (27). Genentech also developed an mAb against FD (FCFD4514S), which is being tested in clinical trials for dry AMD (NCT01229215) (28). A fragment of the anti-FB mAb 1379 (29) had been disclosed by Taligen (TA106), yet no development plans have been revealed since its acquisition by Alexion. More recently, properdin emerged as an attractive therapeutic target, and antibodies against this modulator showed efficacy in models of arthritis and abdominal aortic aneurism (30, 31). Though not a classical “inhibitor”, the C3 homolog cobra venom factor (CVF) also targets C3 by forming long-lasting C3 convertases that rapidly deplete C3 stores (32). Therapeutic C3 depletion by CVF and its humanized form (HC3-1496, InCode) has shown efficacy in disease models including AMD (33) and transplantation (34). Of note, HC3-1497 does not act as a C5 convertase (in contrast to some forms of native CVF), thereby alleviating toxicity concerns due to direct generation of massive C5a levels (32). Still, it will need to be further evaluated what effect instant solution activation of complement may have on diseased tissue and which indications may benefit most from depletion strategies.
Whereas these concepts aim at preventing formation of the convertases, our body possesses formidable inhibitors, i.e., the regulators of complement activation (RCA) family, to deal with those potent enzyme complexes and prevent AP amplification. Their potency, specificity, and modular architecture (consisting of 4–30 complement control protein domains; CCP) render RCA attractive templates for inhibitor development (21, 35). For example, soluble complement receptor 1 (sCR1; TP10, Avant) has been evaluated for various diseases (36, 37) and the program has meanwhile been continued by Celldex (CDX-1135) with focus on renal pathologies; plans for a pilot study with CDX-1135 for the treatment of DDD in 2013 have been announced (38). A truncated, membrane-targeted form of CR1 (Mirococept, APT070) (39) was recently shown to improve early transplant function in a pilot study of kidney transplantation, and is scheduled to enter formal clinical trials (40). Owing to its strong disease associations in AMD and kidney disorders (3), the direct use of recombinant factor H (FH) for therapeutic purposes has been considered yet is not actively pursued at this point. Hence, recent strategies focus on engineered protein therapeutics based on the regulatory domains of FH. Among those, TT30 (ALXN1102, Alexion) is the most established candidate. TT30 combines the regulatory CCP1-5 of FH with the N-terminal four domains of CR2 that bind to iC3b, C3dg and C3d. Thereby, TT30 inhibits AP activity (by accelerating the decay of convertases and acting as a cofactor for the factor I-mediated degradation of C3b to prevent further C3/C5 convertase formation) on sites of ongoing complement activation that typically accumulate these downstream opsonins (41, 42). TT30 and its rodent homologs have shown promising results for AP-specific complement inhibition in disease models ranging from AMD to PNH (41, 43, 44), and TT30 is now being evaluated in phase I trials for PNH (NCT01335165). This targeting approach has been extended to combine the N-terminal regions of CR1 and CR2 (TT32), thereby producing an inhibitor with activities for both CP/LP and AP (by acting on C4b and C3b, respectively) that has been tested in an arthritis model (45). Based on insight into the structure of FH, a streamlined FH derivative that directly links the regulatory and targeting domains was recently disclosed (mini-FH; AMY-201, Amyndas) (46). Despite a size reduction by 70%, mini-FH preserved functional activities compared to FH and showed a unique targeting profile towards sites of ongoing activation (similar to TT30), self-cells (via binding to glycosaminoglycans) and markers of oxidative stress-induced damage (i.e., recognition of malondialdehyde); in models of PNH, mini-FH showed activities exceeding those of FH and other inhibitors (46). Inspired by complement evasion strategies employed by human pathogens (47), an approach for protecting biomaterial surfaces has been described, in which surfaces are coated with an FH-binding peptide (5C6; AMY-301, Amyndas) that recruits host FH and prevents AP amplification (48). Recently, this approach was expanded to transplantation-relevant cell surfaces and combined with apyrase immobilization to produce anti-thromboinflammatory coatings (49).
The therapeutic value of inhibiting at the level of C5, simultaneously blocking the generation of C5a and MAC, remains a major focus of complement-targeted drug discovery (21, 50). For example, an mAb against mouse C5 (BB5.1) with anti-hemolytic activity was disclosed in 1987 (51) and subsequently used in disease models (52–54), spearheading a development that resulted in human-specific C5 antibodies (55) and, eventually, the clinical availability of Eculizumab (see above). An scFv form of the antibody (Pexelizumab, Alexion) was evaluated for use in myocardial infarction and cardiopulmonary bypass surgery, among other models (56, 57), but appears to be discontinued after those studies did not meet expectations (58). Though the target has not been disclosed, the mAb LFG316 (Novartis) that is being tested in phase II trials for dry AMD (NCT01527500) is likely directed against C5. In addition, Adienne is developing neutralizing minibodies against C5, in which an scFv fragment is linked to the IgG hinge region. A rat homolog of this minibody showed promising results in models of posttransplant IRI (59), and the human minibody (Mubodina) received orphan drug status for the treatment of aHUS and DDD. Ergidina (Adienne), a Mubodina derivative to which an RGD tail peptide was added to target the minibody to endothelial cells (50), has been positioned for transplant-related IRI. This targeting approach has been extended for use in rodent models of arthritis by fusing a peptide-tag specific for the synovial microvascular endothelium to the minibody; the resulting molecule (MT07) resolved inflammation by reducing recruitment of polymorphonuclear cells, cytokine release and tissue injury (60). C5-neutralizing entities have also been developed on the basis of aptamers (ARC1905, Ophthotech); while a phase I trial for AMD in combination with the anti-VEGF mAb Lucentis has been completed in 2011 with positive safety results (NCT00709527)(61), no details about the future development of this compound have been released. Finally, nature provides potent C5 inhibitors such as SSL7 from S. aureus (62) or the tick-derived OmCI, latter of which was shown to prevent terminal pathway activation and partially reduce inflammatory markers in an ex vivo human/porcine whole blood model of sepsis and completely prevented the development of experimental myasthenia gravis in rats (63, 64). As with all exogenous natural products, however, potential immunogenicity concerns need to be carefully considered and may render a direct use of these inhibitors challenging.
Instead of inhibiting the entire terminal pathway, the selective blockage of either C5a-mediated signaling or MAC formation is desired under certain clinical conditions (50). While MAC-directed therapy is the road less traveled, it produced promising concepts for the treatment of PNH or AMD based on soluble or membrane-targeted forms of the regular CD59 (65, 66). Given the strong pro-inflammatory and modulatory activities of C5a signaling, therapeutic intervention at the level of C5a or the C5a receptor (C5aR; CD88) remains a focal area. Neutralizing antibodies against C5a have demonstrated protective effects in experimental sepsis (67), and this promising therapeutic concept appears to apply to the use of mAb IFX-1 (CaCP29, InflaRx; target has not been officially disclosed) that has been evaluated in phase I trials (NCT01319903). In addition, two strategies have been developed that exploit structurally complementary molecules as C5a inhibitors; whereas NOX-D19 (Noxxon) is based on spiegelmer technology (i.e., biostable RNA aptamers), antisense peptides (C5aIP, AcPepA) have been tested in islet transplantation and sepsis (68, 69). As a GPCR, the C5aR represents a druggable target and several small molecule antagonists have indeed been developed over the years (11, 50). Among those, the peptidomimetic C5aR antagonist PMX53 (originally developed by Promics) is the most widely used candidate and has shown promise in disease models ranging from sepsis, cancer and IRI to inflammatory bowel disease, arthritis and pregnancy-related complications. Clinical studies have been conducted with PMX53 for use in psoriasis and rheumatoid arthritis (50, 70); while the compound demonstrated beneficial safety profiles, it did not significantly reduce synovial inflammation in the arthritis study (71). A derivative of the compound with increased metabolic stability and ability to cross the blood-brain barrier (PMX205) has shown promising effects in a mouse model of Alzheimer’s disease (72) and has recently been tested in models of periodontitis and colitis (73, 74). The technology has undergone several acquisitions, and future plans for the clinical development of PMX53 or PMX205 have not been revealed. A structurally similar antagonist (JPE-1375) has been discontinued after operations of the developing company (Jerini Ophthalmic) had been closed in 2009. The only small molecule C5aR antagonist under clinical development, to the best of our knowledge, remains CCX168 (ChemoCentryx), which is evaluated in phase II for anti-neutrophil cytoplasmic antibodies (ANCA)-associated renal vasculitis (NCT01363388). Alongside synthetic antagonists, blockage of C5aR signaling can also be achieved by protein therapeutics. Novo Nordisk is developing two anti-C5aR antibodies (Table 1) and is conducting phase I trials for rheumatoid arthritis (NCT01223911, NCT01611688). A protein antagonist based on the immune evasion protein CHIPS from S. aureus (ADC-1004, Alligator Bioscience) had been tested in a myocardial IRI model, in which it reduced infarct size by 21% (75), but the company has announced a delay in further development of this compound. Compared with C5aR, therapeutic modulation of C3a receptor (C3aR) has proven challenging and a C3aR antagonist candidate (SB 290157) showed partial agonism (76). Still, SB 290157 continues to being tested in experimental disease studies, primarily in rodents, and the compound has shown modulatory effect in models of intracerebral hemorrhage and metabolic dysfunction, among others (77, 78).
Translation into complement-centered immunomodulatory strategies
Ongoing research on underlying disease mechanisms using clinically relevant models will continue to be of high importance for validating the involvement of complement in a specific disease and for selecting the most promising indications. These studies will also guide target selection, which is a balancing act between comprehensive amelioration of complement-induced damage, maintenance of physiological complement activities, treatment cost, and other factors. The ideal level of intervention will be different for each disease and may even vary within groups of affected patients. As a major mediator of immune responses, C5a signaling can be an attractive target to tame inflammation without affecting opsonization or lytic functions of complement. In other diseases, it may be more important to disrupt the amplification cycle to avoid perpetual opsonization and immune cell recognition. Finally, in disorders with known initiator, the upstream inhibition of a single pathway may be preferred. The complexity and intricate consequences of choosing an appropriate target level for therapeutic complement inhibition is nicely illustrated in the case of PNH. Since MAC-induced lysis is the hallmark symptom of this disease, inhibition needs to occur at or upstream of C5 cleavage, thereby excluding the C5a-C5aR axis. C5-directed therapy using Eculizumab potently prevents intravascular lysis and presents a highly effective (yet costly) treatment option. However, studies have shown that some PNH patients under Eculizumab show insufficient success and remain dependent on transfusion. In these patients, uncontrolled AP amplification on PNH erythrocytes in the absence of CD55 can lead to an accumulation of opsonized cells in circulation, which are recognized by immune cells and become subject to C3-dependent extravascular hemolysis (79). Upstream intervention at the level of C3 may therefore provide a valuable alternative to Eculizumab, especially for patients with poor response to the treatment. The study of PNH patients treated with Eculizumab also established that the effects of complement-directed therapy reaches beyond prevention of lysis and may beneficially shape other clinical aspects including lower risk of thrombosis and normalization of immune parameters (79, 80).
The clinical availability of complement inhibitors and a growing number of drug trials have also shed more light on the important question of safety. With long-term data for chronic Eculizumab treatment available, the results suggest good safety and tolerability for this C5-directed strategy (81, 82). Similarly, the various C1-INH preparations share a favorable safety/tolerability profile when used in the typical dosage regimen for the treatment and/or prophylaxis of HAE. Finally, clinical trials conducted so far with inhibitors acting at various levels ranging from C3 to C5aR (e.g., sCR1, PMX53, compstatin, ARC1905 etc.; Table I) did not reveal toxicity or other major complications. Increased susceptibility to infection still remains the biggest concern in the context of complement inhibition. Indeed, while patients with a primary deficiency in proximal components may develop a higher risk for infection with certain pathogens such as Streptococcus pneumonia, immune complex diseases or systemic lupus erythematosus-like symptoms, complete lack of one of the terminal component primarily raises the susceptibility to neisserial infections (83, 84); patients receiving Eculizumab are therefore prophylactically vaccinated against Neisseria meningitis. On a functional level, even downstream effectors such as C5a have been shown to critically contribute to the protection against certain pathogens such as S. aureus (85), thereby emphasizing the notion that no inhibition level is a priori to be considered “safe” or “unsafe”. Importantly, though, severe infections in patients with primary complement deficiencies are predominantly seen in childhood with a significant improvement in adulthood; it has therefore been concluded that the antibacterial functions of complement becomes less critical once adaptive immunity is fully developed and high IgG titers are reached (86). Moreover, in the case of C3, even low residual plasma levels, as for example observed in patients with nephritic factors that deplete the C3 store, appear to confer a considerable level of protection. In this context, one open question is whether full complement inhibition can be achieved (e.g., due to rapid turnover of some components) or even needs to be achieved. In diseases based on a chronically imbalanced complement system (e.g., as a consequence of RCA polymorphisms), it may be enough to tip the balance into the normal range. In addition, even C3 inhibition does not lead to complete complement impairment as it does not interrupt opsonization with CP/LP components, and direct activation of C3 and C5 by non-convertase proteases remains intact in most inhibitory regimens. Finally, safety considerations are also dependent on the timeframe and localization of inhibitor administration, with potential concerns being less pronounced when inhibitors are administered locally (e.g., intravitreal injection in AMD) and/or for a limited timeframe (e.g., in sepsis). While the final “verdict” on the safety of complement-targeted therapy is still out, and certainly needs to carefully be reassessed for each compound, the current picture is encouraging.
One of the emerging strategies that may further add to the selectivity and safety of complement-directed therapies is drug targeting. As a majority of complement activation and amplification occurs on the surface of foreign or diseased cells, and given the importance of peripherally synthesized complement, a directed intervention at the site of major activity is considered preferable in many situations. Approaches that target specific surfaces via coating (e.g., 5C6), membrane anchoring (e.g., Mirococept) or cell/tissue-specific peptide tags (e.g., Ergidina) have shown great promise and will be interesting to follow through clinical development. The same is true for the concept of directing regulatory entities towards sites of ongoing AP activity (as marked by accumulating iC3b/C3dg) using FH- or CR2-derived recognition domains (e.g., in mini-FH or TT30). In latter case, comparative studies impressively demonstrated the efficacy benefit of the targeted compound in attenuating IRI-induced damage on affected tissue while retaining systemic complement activity (87).
Novel therapeutic concepts increasingly attempt to exploit the crosstalk between complement and systems of innate/adaptive immunity and coagulation during pathological events. For example, as complement and the Toll-like receptor (TLR) system show cooperative effects during confrontation with danger patterns, their combined attenuation was hypothesized to be advantageous (88). Indeed, synergistic beneficial effects of concerted inhibition of complement (using compstatin or anti-C5) and TLR (via anti-CD14) have been shown in whole blood models of sepsis (89) and might have great implications for SIRS (88) and other inflammatory diseases such as periodontitis. The newly discovered negative feedback loop in the C5aR-FcγR crosstalk via dectin-1 and galactosylated IgG, on the other hand, may influence the development of glyocoengineered intravenous immunoglobulin preparations similar to those used as anti-inflammatory treatment in autoimmune disorders (90, 91). In some situations, such as during infection, the explicit stimulation of C5a mediation via C5aR agonists such as peptide EP67 may lead to a beneficial enhancement of immune responses (92); the same agonist has also been suggested as potential adjuvant due to its ability to drive Th1-mediated immune responses (93). Even in cases of complement-inhibitory monotherapy, one should consider that associated physiological systems may be affected and aim for a comprehensive analysis. In the case of sepsis, for example, compstatin administered to baboons 6 hours after a sublethal dose of E. coli not only prevented complement activation but also influenced inflammatory and thrombotic parameters, leading to significant organ protection (23). Especially in complex disorders like sepsis, there may be several routes (e.g., targeting complement, coagulation or downstream inflammation) that lead to similar results as they provide the body with sufficient free resources to cope with the remaining complications. In many aspects, tipping the balance from an excessive back to an effective complement response may be an essential and highly promising strategy for restoring immune homeostasis and resolving inflammation.
Conclusions
In coming back to the drug discovery concepts coined by Paul Ehrlich and other pioneers in immunology, it appears unlikely that there will be one “magic bullet” to treat all complement-related diseases. Rather, we may aim for a diverse armory of therapeutics and for carefully tailoring the treatment strategies for each disease. The commonalities within many complement-related pathologies indicate that, once on the market for one disease, a new complement-targeted drug may also benefit patients with other disorders, as the continuous expansion of indications for both Eculizumab and C1-INH have already demonstrated. In any case, the pipelines of academic research and biopharmaceutical companies look highly promising, as they are already filled with a diverse panel of intriguing candidates, rendering the goal of a complement therapeutic “toolbox” ever more likely. Finally, the increasing use of diverse and highly specific complement inhibitors for the dissection and exploration of disease mechanisms may not only reveal novel candidates with therapeutic potential but also help discover even more fascinating crosstalk mechanisms between complement and other branches of immunity.
Acknowledgments
We thank Dr. Robert A. DeAngelis and Dr. Edimara Reis for critically reading the manuscript and for their valuable discussion.
This work was supported by National Institutes of Health grants AI003040, AI068730, AI072106, AI097805, EY020633, GM097747 and DE021685.
Abbreviations used in this article
- aHUS
atypical hemolytic uremic syndrome
- AMD
age-related macular degeneration
- ANCA
anti-neutrophil cytoplasmic antibodies
- AP
alternative pathway
- C1-INH
C1 inhibitor
- C3aR
C3a receptor
- C5aR
C5a receptor
- CP
classical pathway
- CR
complement receptor
- CVF
cobra venom factor
- DDD
dense deposit disease
- FB
factor B
- FD
factor D
- FH
factor H
- HAE
hereditary angioedema
- IRI
ischemia/reperfusion injury
- LP
lectin pathway
- MAC
membrane attack complex
- MASP
MBL-associated serine proteases
- MBL
mannose-binding lectin
- PNH
paroxysmal nocturnal hemoglobinuria
- RCA
regulator of complement activation
- RGD
arginine-glycine-aspartic acid
- scFv
single chain variable fragment
- TLR
Toll-like receptor
Footnotes
Disclosures
D.R. and J.D.L. are the inventors of patents and/or patent applications that describe the use of complement inhibitors for therapeutic purposes. J.D.L. is the founder of Amyndas Biotherapeutics and Amyndas Pharmaceuticals, which are developing complement inhibitors for clinical applications.
References
- 1.Drews J. Paul Ehrlich: magister mundi. Nat Rev Drug Discov. 2004;3:797–801. doi: 10.1038/nrd1498. [DOI] [PubMed] [Google Scholar]
- 2.Hussell T. Immune modulatory strategies: a playground with a swing and a seesaw. Expert Rev Anti Infect Ther. 2012;10:249–251. doi: 10.1586/eri.12.12. [DOI] [PubMed] [Google Scholar]
- 3.Ricklin D, Lambris JD. Complement in immune and inflammatory disorders: pathophysiological mechanisms. J Immunol. 2013 doi: 10.4049/jimmunol.1203487. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Markiewski MM, Lambris JD. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am J Pathol. 2007;171:715–727. doi: 10.2353/ajpath.2007.070166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zeerleder S. C1-inhibitor: more than a serine protease inhibitor. Semin Thromb Hemost. 2011;37:362–374. doi: 10.1055/s-0031-1276585. [DOI] [PubMed] [Google Scholar]
- 6.Lu F, Fernandes SM, Davis AE., 3rd The effect of C1 inhibitor on myocardial ischemia and reperfusion injury. Cardiovasc Pathol. 2013;22:75–80. doi: 10.1016/j.carpath.2012.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tillou X, Poirier N, Le Bas-Bernardet S, Hervouet J, Minault D, Renaudin K, Vistoli F, Karam G, Daha M, Soulillou JP, Blancho G. Recombinant human C1-inhibitor prevents acute antibody-mediated rejection in alloimmunized baboons. Kidney Int. 2010;78:152–159. doi: 10.1038/ki.2010.75. [DOI] [PubMed] [Google Scholar]
- 8.Koulmanda M, Uknis ME, Strom TB. C1 esterase inhibitor reverses invasive insulitis and hyperglycemia in a model of type 1 diabetes. XXIV International Complement Workshop; Chania, Greece. 2012. [Google Scholar]
- 9.Schmidtko J, Peine S, El-Housseini Y, Pascual M, Meier P. Treatment of Atypical Hemolytic Uremic Syndrome and Thrombotic Microangiopathies: A Focus on Eculizumab. Am J Kidney Dis. 2013;61:289–299. doi: 10.1053/j.ajkd.2012.07.028. [DOI] [PubMed] [Google Scholar]
- 10.Orth-Holler D, Riedl M, Wurzner R. Inhibition of terminal complement activation in severe Shiga toxin-associated HUS - perfect example for a fast track from bench to bedside. EMBO Mol Med. 2011;3:617–619. doi: 10.1002/emmm.201100169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Qu H, Ricklin D, Lambris JD. Recent developments in low molecular weight complement inhibitors. Mol Immunol. 2009;47:185–195. doi: 10.1016/j.molimm.2009.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Subasinghe NL, Khalil E, Travins JM, Ali F, Ballentine SK, Hufnagel HR, Pan W, Leonard K, Bone RF, Soll RM, Crysler CS, Ninan N, Kirkpatrick J, Kolpak MX, Diloreto KA, Eisennagel SH, Huebert ND, Molloy CJ, Tomczuk BE, Gaul MD. Design and synthesis of polyethylene glycol-modified biphenylsulfonyl-thiophene-carboxamidine inhibitors of the complement component C1s. Bioorg Med Chem Lett. 2012;22:5303–5307. doi: 10.1016/j.bmcl.2012.06.030. [DOI] [PubMed] [Google Scholar]
- 13.Subasinghe NL, Travins JM, Ali F, Huang H, Ballentine SK, Marugan JJ, Khalil E, Hufnagel HR, Bone RF, DesJarlais RL, Crysler CS, Ninan N, Cummings MD, Molloy CJ, Tomczuk BE. A novel series of arylsulfonylthiophene-2-carboxamidine inhibitors of the complement component C1s. Bioorg Med Chem Lett. 2006;16:2200–2204. doi: 10.1016/j.bmcl.2006.01.036. [DOI] [PubMed] [Google Scholar]
- 14.Duvall MR, Hwang HY, Boackle RJ. Specific inhibition of the classical complement pathway with an engineered single-chain Fv to C1q globular heads decreases complement activation by apoptotic cells. Immunobiology. 2010;215:395–405. doi: 10.1016/j.imbio.2009.05.010. [DOI] [PubMed] [Google Scholar]
- 15.Dempulos GA, Dudler T, Schwaeble WJ U. S. P. Office. Methods for treating conditions associated with MASP-2 dependent complement activation. Omeros Corp., University of Leicester; U.S.A: 2012. [Google Scholar]
- 16.Schwaeble WJ, Lynch NJ, Clark JE, Marber M, Samani NJ, Ali YM, Dudler T, Parent B, Lhotta K, Wallis R, Farrar CA, Sacks S, Lee H, Zhang M, Iwaki D, Takahashi M, Fujita T, Tedford CE, Stover CM. Targeting of mannan-binding lectin-associated serine protease-2 confers protection from myocardial and gastrointestinal ischemia/reperfusion injury. Proc Natl Acad Sci U S A. 2011;108:7523–7528. doi: 10.1073/pnas.1101748108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Heja D, Harmat V, Fodor K, Wilmanns M, Dobo J, Kekesi KA, Zavodszky P, Gal P, Pal G. Monospecific inhibitors show that both mannan-binding lectin-associated serine protease-1 (MASP-1) and -2 Are essential for lectin pathway activation and reveal structural plasticity of MASP-2. J Biol Chem. 2012;287:20290–20300. doi: 10.1074/jbc.M112.354332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pavlov VI, Skjoedt MO, Siow Tan Y, Rosbjerg A, Garred P, Stahl GL. Endogenous and natural complement inhibitor attenuates myocardial injury and arterial thrombogenesis. Circulation. 2012;126:2227–2235. doi: 10.1161/CIRCULATIONAHA.112.123968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Krarup A, Wallis R, Presanis JS, Gal P, Sim RB. Simultaneous activation of complement and coagulation by MBL-associated serine protease 2. PLoS One. 2007;2:e623. doi: 10.1371/journal.pone.0000623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Harboe M, Ulvund G, Vien L, Fung M, Mollnes TE. The quantitative role of alternative pathway amplification in classical pathway induced terminal complement activation. Clin Exp Immunol. 2004;138:439–446. doi: 10.1111/j.1365-2249.2004.02627.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ricklin D, Lambris JD. Progress and trends in complement therapeutics. Adv Exp Med Biol. 2013;734:1–22. doi: 10.1007/978-1-4614-4118-2_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kourtzelis I, Markiewski MM, Doumas M, Rafail S, Kambas K, Mitroulis I, Panagoutsos S, Passadakis P, Vargemezis V, Magotti P, Qu H, Mollnes TE, Ritis K, Lambris JD. Complement anaphylatoxin C5a contributes to hemodialysis-associated thrombosis. Blood. 2010;116:631–639. doi: 10.1182/blood-2010-01-264051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Silasi-Mansat R, Zhu H, Popescu NI, Peer G, Sfyroera G, Magotti P, Ivanciu L, Lupu C, Mollnes TE, Taylor FB, Kinasewitz G, Lambris JD, Lupu F. Complement inhibition decreases the procoagulant response and confers organ protection in a baboon model of Escherichia coli sepsis. Blood. 2010;116:1002–1010. doi: 10.1182/blood-2010-02-269746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chi ZL, Yoshida T, Lambris JD, Iwata T. Suppression of drusen formation by compstatin, a peptide inhibitor of complement C3 activation, on cynomolgus monkey with early-onset macular degeneration. Adv Exp Med Biol. 2010;703:127–135. doi: 10.1007/978-1-4419-5635-4_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Qu H, Ricklin D, Bai H, Chen H, Reis ES, Maciejewski M, Tzekou A, Deangelis RA, Resuello RR, Lupu F, Barlow PN, Lambris JD. New analogs of the clinical complement inhibitor compstatin with subnanomolar affinity and enhanced pharmacokinetic properties. Immunobiology. 2012 doi: 10.1016/j.imbio.2012.06.003. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Katschke KJ, Jr, Stawicki S, Yin J, Steffek M, Xi H, Sturgeon L, Hass PE, Loyet KM, Deforge L, Wu Y, van Lookeren Campagne M, Wiesmann C. Structural and functional analysis of a C3b-specific antibody that selectively inhibits the alternative pathway of complement. J Biol Chem. 2009;284:10473–10479. doi: 10.1074/jbc.M809106200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lindorfer MA, Pawluczkowycz AW, Peek EM, Hickman K, Taylor RP, Parker CJ. A novel approach to preventing the hemolysis of paroxysmal nocturnal hemoglobinuria: both complement-mediated cytolysis and C3 deposition are blocked by a monoclonal antibody specific for the alternative pathway of complement. Blood. 2010;115:2283–2291. doi: 10.1182/blood-2009-09-244285. [DOI] [PubMed] [Google Scholar]
- 28.Katschke KJ, Jr, Wu P, Ganesan R, Kelley RF, Mathieu MA, Hass PE, Murray J, Kirchhofer D, Wiesmann C, van Lookeren Campagne M. Inhibiting alternative pathway complement activation by targeting the factor D exosite. J Biol Chem. 2012;287:12886–12892. doi: 10.1074/jbc.M112.345082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Thurman JM, Kraus DM, Girardi G, Hourcade D, Kang HJ, Royer PA, Mitchell LM, Giclas PC, Salmon J, Gilkeson G, Holers VM. A novel inhibitor of the alternative complement pathway prevents antiphospholipid antibody-induced pregnancy loss in mice. Mol Immunol. 2005;42:87–97. doi: 10.1016/j.molimm.2004.07.043. [DOI] [PubMed] [Google Scholar]
- 30.Kimura Y, Zhou L, Miwa T, Song WC. Genetic and therapeutic targeting of properdin in mice prevents complement-mediated tissue injury. J Clin Invest. 2010;120:3545–3554. doi: 10.1172/JCI41782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhou HF, Yan H, Stover CM, Fernandez TM, Rodriguez de Cordoba S, Song WC, Wu X, Thompson RW, Schwaeble WJ, Atkinson JP, Hourcade DE, Pham CT. Antibody directs properdin-dependent activation of the complement alternative pathway in a mouse model of abdominal aortic aneurysm. Proc Natl Acad Sci U S A. 2012;109:E415–422. doi: 10.1073/pnas.1119000109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vogel CW, Fritzinger DC. Cobra venom factor: Structure, function, and humanization for therapeutic complement depletion. Toxicon. 2010;56:1198–1222. doi: 10.1016/j.toxicon.2010.04.007. [DOI] [PubMed] [Google Scholar]
- 33.Fritzinger DC, Dean R, Meschter C, Wong K, Halter R, Borlak J, St John WD, Vogel CW. Complement depletion with humanized cobra venom factor in a mouse model of age-related macular degeneration. Adv Exp Med Biol. 2010;703:151–162. doi: 10.1007/978-1-4419-5635-4_11. [DOI] [PubMed] [Google Scholar]
- 34.Chen Song S, Zhong S, Xiang Y, Li JH, Guo H, Wang WY, Xiong YL, Li XC, Chen Shi S, Chen XP, Chen G. Complement inhibition enables renal allograft accommodation and long-term engraftment in presensitized nonhuman primates. Am J Transplant. 2011;11:2057–2066. doi: 10.1111/j.1600-6143.2011.03646.x. [DOI] [PubMed] [Google Scholar]
- 35.Ricklin D. Manipulating the mediator: Modulation of the alternative complement pathway C3 convertase in health, disease and therapy. Immunobiology. 2012;217:1057–1066. doi: 10.1016/j.imbio.2012.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li JS, Jaggers J, Anderson PA. The use of TP10, soluble complement receptor 1, in cardiopulmonary bypass. Expert Rev Cardiovasc Ther. 2006;4:649–654. doi: 10.1586/14779072.4.5.649. [DOI] [PubMed] [Google Scholar]
- 37.Rioux P. TP-10 (AVANT Immunotherapeutics) Curr Opin Investig Drugs. 2001;2:364–371. [PubMed] [Google Scholar]
- 38.Celldex Therapeutics. SEC Filing Report. 2013 Jan 7; available from: http://ir.celldextherapeutics.com/secfiling.cfm?filingID=1104659-13-878&CIK=744218.
- 39.Smith RA. Targeting anticomplement agents. Biochem Soc Trans. 2002;30:1037–1041. doi: 10.1042/bst0301037. [DOI] [PubMed] [Google Scholar]
- 40.Sacks S, Karegli J, Farrar CA, Asgari E, Schwaeble W, Zhou W, Smith RA. Targeting complement at the time of transplantation. Adv Exp Med Biol. 2013;734:247–255. doi: 10.1007/978-1-4614-4118-2_17. [DOI] [PubMed] [Google Scholar]
- 41.Holers VM, Rohrer B, Tomlinson S. CR2-Mediated Targeting of Complement Inhibitors: Bench-to-Bedside Using a Novel Strategy for Site-Specific Complement Modulation. Adv Exp Med Biol. 2013;734:137–154. doi: 10.1007/978-1-4614-4118-2_9. [DOI] [PubMed] [Google Scholar]
- 42.Fridkis-Hareli M, Storek M, Mazsaroff I, Risitano AM, Lundberg AS, Horvath CJ, Holers VM. Design and development of TT30, a novel C3d-targeted C3/C5 convertase inhibitor for treatment of human complement alternative pathway-mediated diseases. Blood. 2011;118:4705–4713. doi: 10.1182/blood-2011-06-359646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Risitano AM, Notaro R, Pascariello C, Sica M, del Vecchio L, Horvath CJ, Fridkis-Hareli M, Selleri C, Lindorfer MA, Taylor RP, Luzzatto L, Holers VM. The complement receptor 2/factor H fusion protein TT30 protects paroxysmal nocturnal hemoglobinuria erythrocytes from complement-mediated hemolysis and C3 fragment. Blood. 2012;119:6307–6316. doi: 10.1182/blood-2011-12-398792. [DOI] [PubMed] [Google Scholar]
- 44.Rohrer B, Coughlin B, Bandyopadhyay M, Holers VM. Systemic Human CR2-Targeted Complement Alternative Pathway Inhibitor Ameliorates Mouse Laser-Induced Choroidal Neovascularization. J Ocul Pharmacol Ther. 2012;28:402–409. doi: 10.1089/jop.2011.0212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Holers M, Banda N, Mehta G, Fridkis-Hareli M, Or E, Storek M, Altman R, Johnson K, Katti S. The human complement receptor type 2 (CR2)/CR1 fusion protein TT32, a targeted inhibitor of the classical and alternative pathway C3 convertases, prevents arthritis in active immunization and passive transfer models and acts by CR2-dependent targeting of CR1 regulatory activity. Immunobiology. 2012;217:1210. [Google Scholar]
- 46.Schmidt CQ, Bai H, Lin Z, Risitano AM, Barlow PN, Ricklin D, Lambris JD. Rational engineering of a novel complement regulator-based therapeutic affords triple targeting of host surfaces and pathway-specific inhibition of complement activation. Immunobiology. 2012;217:1197. [Google Scholar]
- 47.Lambris JD, Ricklin D, Geisbrecht BV. Complement evasion by human pathogens. Nat Rev Microbiol. 2008;6:132–142. doi: 10.1038/nrmicro1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wu YQ, Qu H, Sfyroera G, Tzekou A, Kay BK, Nilsson B, Nilsson Ekdahl K, Ricklin D, Lambris JD. Protection of nonself surfaces from complement attack by factor H-binding peptides: implications for therapeutic medicine. J Immunol. 2011;186:4269–4277. doi: 10.4049/jimmunol.1003802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Nilsson PH, Ekdahl KN, Magnusson PU, Qu H, Iwata H, Ricklin D, Hong J, Lambris JD, Nilsson B, Teramura Y. Autoregulation of thromboinflammation on biomaterial surfaces by a multicomponent therapeutic coating. Biomaterials. 2012;34:985–994. doi: 10.1016/j.biomaterials.2012.10.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Woodruff TM, Nandakumar KS, Tedesco F. Inhibiting the C5-C5a receptor axis. Mol Immunol. 2011;48:1631–1642. doi: 10.1016/j.molimm.2011.04.014. [DOI] [PubMed] [Google Scholar]
- 51.Frei Y, Lambris JD, Stockinger B. Generation of a monoclonal antibody to mouse C5 application in an ELISA assay for detection of anti-C5 antibodies. Mol Cell Probes. 1987;1:141–149. doi: 10.1016/0890-8508(87)90022-3. [DOI] [PubMed] [Google Scholar]
- 52.Strey CW, Markiewski M, Mastellos D, Tudoran R, Spruce LA, Greenbaum LE, Lambris JD. The proinflammatory mediators C3a and C5a are essential for liver regeneration. J Exp Med. 2003;198:913–923. doi: 10.1084/jem.20030374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wang Q, Rozelle AL, Lepus CM, Scanzello CR, Song JJ, Larsen DM, Crish JF, Bebek G, Ritter SY, Lindstrom TM, Hwang I, Wong HH, Punzi L, Encarnacion A, Shamloo M, Goodman SB, Wyss-Coray T, Goldring SR, Banda NK, Thurman JM, Gobezie R, Crow MK, Holers VM, Lee DM, Robinson WH. Identification of a central role for complement in osteoarthritis. Nat Med. 2011;17:1674–1679. doi: 10.1038/nm.2543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang Y, Rollins SA, Madri JA, Matis LA. Anti-C5 monoclonal antibody therapy prevents collagen-induced arthritis and ameliorates established disease. Proc Natl Acad Sci U S A. 1995;92:8955–8959. doi: 10.1073/pnas.92.19.8955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Rinder CS, Rinder HM, Smith BR, Fitch JC, Smith MJ, Tracey JB, Matis LA, Squinto SP, Rollins SA. Blockade of C5a and C5b-9 generation inhibits leukocyte and platelet activation during extracorporeal circulation. J Clin Invest. 1995;96:1564–1572. doi: 10.1172/JCI118195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Smith PK, Shernan SK, Chen JC, Carrier M, Verrier ED, Adams PX, Todaro TG, Muhlbaier LH, Levy JH PRIMO–CABG II Investigators. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: combined results from the PRIMO-CABG I and II trials. J Thorac Cardiovasc Surg. 2011;142:89–98. doi: 10.1016/j.jtcvs.2010.08.035. [DOI] [PubMed] [Google Scholar]
- 57.Testa L, Van Gaal WJ, Bhindi R, Biondi-Zoccai GG, Abbate A, Agostoni P, Porto I, Andreotti F, Crea F, Banning AP. Pexelizumab in ischemic heart disease: a systematic review and meta-analysis on 15,196 patients. J Thorac Cardiovasc Surg. 2008;136:884–893. doi: 10.1016/j.jtcvs.2007.12.062. [DOI] [PubMed] [Google Scholar]
- 58.Verrier ED, Shernan SK, Taylor KM, Van de Werf F, Newman MF, Chen JC, Carrier M, Haverich A, Malloy KJ, Adams PX, Todaro TG, Mojcik CF, Rollins SA, Levy JH, Investigators PC. Terminal complement blockade with pexelizumab during coronary artery bypass graft surgery requiring cardiopulmonary bypass: a randomized trial. JAMA. 2004;291:2319–2327. doi: 10.1001/jama.291.19.2319. [DOI] [PubMed] [Google Scholar]
- 59.Ferraresso M, Macor P, Valente M, Della Barbera M, D’Amelio F, Borghi O, Raschi E, Durigutto P, Meroni P, Tedesco F. Posttransplant ischemia-reperfusion injury in transplanted heart is prevented by a minibody to the fifth component of complement. Transplantation. 2008;86:1445–1451. doi: 10.1097/TP.0b013e31818a68e2. [DOI] [PubMed] [Google Scholar]
- 60.Macor P, Durigutto P, De Maso L, Garrovo C, Biffi S, Cortini A, Fischetti F, Sblattero D, Pitzalis C, Marzari R, Tedesco F. Treatment of experimental arthritis by targeting synovial endothelium with a neutralizing recombinant antibody to C5. Arthritis Rheum. 2012;64:2559–2567. doi: 10.1002/art.34430. [DOI] [PubMed] [Google Scholar]
- 61.Cousins SW the Ophthotech Study Group. Targeting complement factor 5 in combination with vascular endothelial growth factor (VEGF) inhibition for neovascular age related macular degeneration (AMD): results of a Phase 1 study. Invest Ophthalmol Vis Sci. 2010;51:e-abstract 1251. [Google Scholar]
- 62.Bestebroer J, Aerts PC, Rooijakkers SH, Pandey MK, Kohl J, van Strijp JA, de Haas CJ. Functional basis for complement evasion by staphylococcal superantigen-like 7. Cell Microbiol. 2010;12:1506–1516. doi: 10.1111/j.1462-5822.2010.01486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Barratt-Due A, Thorgersen EB, Lindstad JK, Pharo A, Lissina O, Lambris JD, Nunn MA, Mollnes TE. Ornithodoros moubata complement inhibitor is an equally effective C5 inhibitor in pigs and humans. J Immunol. 2011;187:4913–4919. doi: 10.4049/jimmunol.1101000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hepburn NJ, Williams AS, Nunn MA, Chamberlain-Banoub JC, Hamer J, Morgan BP, Harris CL. In vivo characterization and therapeutic efficacy of a C5-specific inhibitor from the soft tick Ornithodoros moubata. J Biol Chem. 2007;282:8292–8299. doi: 10.1074/jbc.M609858200. [DOI] [PubMed] [Google Scholar]
- 65.Bora NS, Kaliappan S, Jha P, Xu Q, Sivasankar B, Harris CL, Morgan BP, Bora PS. CD59, a complement regulatory protein, controls choroidal neovascularization in a mouse model of wet-type age-related macular degeneration. J Immunol. 2007;178:1783–1790. doi: 10.4049/jimmunol.178.3.1783. [DOI] [PubMed] [Google Scholar]
- 66.Cashman SM, Ramo K, Kumar-Singh R. A non membrane-targeted human soluble CD59 attenuates choroidal neovascularization in a model of age related macular degeneration. PLoS One. 2011;6:e19078. doi: 10.1371/journal.pone.0019078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Czermak BJ, Sarma V, Pierson CL, Warner RL, Huber-Lang M, Bless NM, Schmal H, Friedl HP, Ward PA. Protective effects of C5a blockade in sepsis. Nat Med. 1999;5:788–792. doi: 10.1038/10512. [DOI] [PubMed] [Google Scholar]
- 68.Okada N, Imai M, Okada A, Ono F, Okada H. HMGB1 Release by C5a Anaphylatoxin is an Effective Target for Sepsis Treatment. Clin Exp Pharmacol. 2012;2:1000114. [Google Scholar]
- 69.Tokodai K, Goto M, Inagaki A, Nakanishi W, Ogawa N, Satoh K, Kawagishi N, Sekiguchi S, Nilsson B, Okada N, Okada H, Satomi S. Attenuation of cross-talk between the complement and coagulation cascades by C5a blockade improves early outcomes after intraportal islet transplantation. Transplantation. 2010;90:1358–1365. doi: 10.1097/tp.0b013e3181ffb9f5. [DOI] [PubMed] [Google Scholar]
- 70.Kohl J. Drug evaluation: the C5a receptor antagonist PMX-53. Curr Opin Mol Ther. 2006;8:529–538. [PubMed] [Google Scholar]
- 71.Vergunst CE, Gerlag DM, Dinant H, Schulz L, Vinkenoog M, Smeets TJ, Sanders ME, Reedquist KA, Tak PP. Blocking the receptor for C5a in patients with rheumatoid arthritis does not reduce synovial inflammation. Rheumatology (Oxford) 2007;46:1773–1778. doi: 10.1093/rheumatology/kem222. [DOI] [PubMed] [Google Scholar]
- 72.Fonseca MI, Ager RR, Chu SH, Yazan O, Sanderson SD, LaFerla FM, Taylor SM, Woodruff TM, Tenner AJ. Treatment with a C5aR antagonist decreases pathology and enhances behavioral performance in murine models of Alzheimer’s disease. J Immunol. 2009;183:1375–1383. doi: 10.4049/jimmunol.0901005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Breivik T, Gundersen Y, Gjermo P, Taylor SM, Woodruff TM, Opstad PK. Oral treatment with complement factor C5a receptor (CD88) antagonists inhibits experimental periodontitis in rats. J Periodontal Res. 2011;46:643–647. doi: 10.1111/j.1600-0765.2011.01383.x. [DOI] [PubMed] [Google Scholar]
- 74.Jain U, Woodruff TM, Stadnyk AW. The C5a receptor antagonist PMX205 ameliorates experimentally induced colitis associated with increased IL-4 and IL-10. Br J Pharmacol. 2013;168:488–501. doi: 10.1111/j.1476-5381.2012.02183.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.van der Pals J, Koul S, Andersson P, Gotberg M, Ubachs JF, Kanski M, Arheden H, Olivecrona GK, Larsson B, Erlinge D. Treatment with the C5a receptor antagonist ADC-1004 reduces myocardial infarction in a porcine ischemia-reperfusion model. BMC Cardiovasc Disord. 2010;10:45. doi: 10.1186/1471-2261-10-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mathieu MC, Sawyer N, Greig GM, Hamel M, Kargman S, Ducharme Y, Lau CK, Friesen RW, O’Neill GP, Gervais FG, Therien AG. The C3a receptor antagonist SB 290157 has agonist activity. Immunol Lett. 2005;100:139–145. doi: 10.1016/j.imlet.2005.03.003. [DOI] [PubMed] [Google Scholar]
- 77.Garrett MC, Otten ML, Starke RM, Komotar RJ, Magotti P, Lambris JD, Rynkowski MA, Connolly ES. Synergistic neuroprotective effects of C3a and C5a receptor blockade following intracerebral hemorrhage. Brain Res. 2009;1298:171–177. doi: 10.1016/j.brainres.2009.04.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lim J, Iyer A, Suen JY, Seow V, Reid RC, Brown L, Fairlie DP. C5aR and C3aR antagonists each inhibit diet-induced obesity, metabolic dysfunction, and adipocyte and macrophage signaling. FASEB J. 2012 doi: 10.1096/fj.12-220582. In Press. [DOI] [PubMed] [Google Scholar]
- 79.Risitano AM. Paroxysmal nocturnal hemoglobinuria and other complement-mediated hematological disorders. Immunobiology. 2012;217:1080–1087. doi: 10.1016/j.imbio.2012.07.014. [DOI] [PubMed] [Google Scholar]
- 80.Alfinito F, Ruggiero G, Sica M, Udhayachandran A, Rubino V, Pepa RD, Palatucci AT, Annunziatella M, Notaro R, Risitano AM, Terrazzano G. Eculizumab treatment modifies the immune profile of PNH patients. Immunobiology. 2012;217:698–703. doi: 10.1016/j.imbio.2011.11.009. [DOI] [PubMed] [Google Scholar]
- 81.Hill A, Hillmen P, Richards SJ, Elebute D, Marsh JC, Chan J, Mojcik CF, Rother RP. Sustained response and long-term safety of eculizumab in paroxysmal nocturnal hemoglobinuria. Blood. 2005;106:2559–2565. doi: 10.1182/blood-2005-02-0564. [DOI] [PubMed] [Google Scholar]
- 82.Roth A, Hock C, Konik A, Christoph S, Duhrsen U. Chronic treatment of paroxysmal nocturnal hemoglobinuria patients with eculizumab: safety, efficacy, and unexpected laboratory phenomena. Int J Hematol. 2011;93:704–714. doi: 10.1007/s12185-011-0867-y. [DOI] [PubMed] [Google Scholar]
- 83.Reis SE, Falcao DA, Isaac L. Clinical aspects and molecular basis of primary deficiencies of complement component C3 and its regulatory proteins factor I and factor H. Scand J Immunol. 2006;63:155–168. doi: 10.1111/j.1365-3083.2006.01729.x. [DOI] [PubMed] [Google Scholar]
- 84.Skattum L, van Deuren M, van der Poll T, Truedsson L. Complement deficiency states and associated infections. Mol Immunol. 2011;48:1643–1655. doi: 10.1016/j.molimm.2011.05.001. [DOI] [PubMed] [Google Scholar]
- 85.von Kockritz-Blickwede M, Konrad S, Foster S, Gessner JE, Medina E. Protective role of complement C5a in an experimental model of Staphylococcus aureus bacteremia. J Innate Immun. 2010;2:87–92. doi: 10.1159/000247157. [DOI] [PubMed] [Google Scholar]
- 86.Lachmann PJ, Smith RA. Taking complement to the clinic--has the time finally come? Scand J Immunol. 2009;69:471–478. doi: 10.1111/j.1365-3083.2009.02258.x. [DOI] [PubMed] [Google Scholar]
- 87.Atkinson C, Song H, Lu B, Qiao F, Burns TA, Holers VM, Tsokos GC, Tomlinson S. Targeted complement inhibition by C3d recognition ameliorates tissue injury without apparent increase in susceptibility to infection. J Clin Invest. 2005;115:2444–2453. doi: 10.1172/JCI25208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Barratt-Due A, Pischke SE, Brekke OL, Thorgersen EB, Nielsen EW, Espevik T, Huber-Lang M, Mollnes TE. Bride and groom in systemic inflammation--the bells ring for complement and Toll in cooperation. Immunobiology. 2012;217:1047–1056. doi: 10.1016/j.imbio.2012.07.019. [DOI] [PubMed] [Google Scholar]
- 89.Brekke OL, Waage C, Christiansen D, Fure H, Qu H, Lambris JD, Osterud B, Nielsen EW, Mollnes TE. The Effects of Selective Complement and CD14 Inhibition on the E. coli-Induced Tissue Factor mRNA Upregulation, Monocyte Tissue Factor Expression, and Tissue Factor Functional Activity in Human Whole Blood. Adv Exp Med Biol. 2013;734:123–136. doi: 10.1007/978-1-4614-4118-2_8. [DOI] [PubMed] [Google Scholar]
- 90.Karsten CM, Pandey MK, Figge J, Kilchenstein R, Taylor PR, Rosas M, McDonald JU, Orr SJ, Berger M, Petzold D, Blanchard V, Winkler A, Hess C, Reid DM, Majoul IV, Strait RT, Harris NL, Kohl G, Wex E, Ludwig R, Zillikens D, Nimmerjahn F, Finkelman FD, Brown GD, Ehlers M, Kohl J. Anti-inflammatory activity of IgG1 mediated by Fc galactosylation and association of FcgammaRIIB and dectin-1. Nat Med. 2012;18:1401–1406. doi: 10.1038/nm.2862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ricklin D, Reis ES, Lambris JD. A sweet spot to control complement-induced inflammation. Nat Med. 2012;18:1340–1341. doi: 10.1038/nm.2916. [DOI] [PubMed] [Google Scholar]
- 92.Sheen TR, Cavaco CK, Ebrahimi CM, Thoman ML, Sanderson SD, Morgan EL, Doran KS. Control of methicillin resistant Staphylococcus aureus infection utilizing a novel immunostimulatory peptide. Vaccine. 2011;30:9–13. doi: 10.1016/j.vaccine.2011.10.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Morgan EL, Morgan BN, Stein EA, Vitrs EL, Thoman ML, Sanderson SD, Phillips JA. Enhancement of in vivo and in vitro immune functions by a conformationally biased, response-selective agonist of human C5a: implications for a novel adjuvant in vaccine design. Vaccine. 2009;28:463–469. doi: 10.1016/j.vaccine.2009.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]