1,660
Views
0
CrossRef citations to date
0
Altmetric
Review

The case for complement component 5 as a target in neurodegenerative disease

, , , , & ORCID Icon
Pages 97-109 | Received 14 Oct 2022, Accepted 03 Feb 2023, Published online: 17 Feb 2023

References

  • Morgan BP, Harris CL. Complement, a target for therapy in inflammatory and degenerative diseases. Nat Rev Drug Discov. 2015;14(12):857–877.
  • Ricklin D, Hajishengallis G, Yang K, et al. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010;11:785–797.
  • Bajic G, Degn SE, Thiel S, et al. Complement activation, regulation, and molecular basis for complement‐related diseases. EMBO J. 2015;34:2735–2757.
  • Carroll MC, Fischer MB. Complement and the immune response. Curr Opin Immunol. 1997;9:64–69.
  • Wagner E. Therapeutic potential of complement modulation. Nat Rev Drug Discov. 2010;9:43–56.
  • 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.
  • Hajishengallis G, Reis ES, Mastellos DC, et al. Novel mechanisms and functions of complement. Nat Immunol. 2017;18:1288–1298.
  • Mollnes TE, Kirschfink M. Strategies of therapeutic complement inhibition. Mol Immunol. 2006;43:107–121.
  • Dunkelberger JR. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20:34–50.
  • Freeley S, Kemper C, Le FG. The “ins and outs” of complement-driven immune responses. Immunol Rev. 2016;274:16–32.
  • Harris CL. Tailoring anti-complement therapeutics. Biochem Soc Trans. 2002;30:1019–1026.
  • Reis ES, Mastellos D C, Yancopoulou D, et al. Applying complement therapeutics to rare diseases. Clin Imm. 2015;161(2):225–240.
  • Kirschfink M. Targeting complement in therapy. Immunol Rev. 2001;180:177–189.
  • Sjöberg AP, Trouw LA, Blom AM. Complement activation and inhibition: a delicate balance. Trends Immunol. 2009;30:83–90.
  • Thurman JM, Holers VM. The Central Role of the Alternative Complement Pathway in Human Disease. J Immunol. 2006;176:1305–1310.
  • Carroll M. The complement system in regulation of adaptive immunity. Nat Immunol. 2004;5:981–986.
  • Hawksworth OA, Coulthard LG, Woodruff TM. Complement in the fundamental processes of the cell. Mol Immunol. 2017;84:17–25.
  • Ricklin D. Complement in immune and inflammatory disorders: therapeutic interventions. J Immunol. 2013;190:3839–3847.
  • McGeer P, Lee M, McGeer E. A review of human diseases caused or exacerbated by aberrant complement activation. Neurobiol Aging. 2016;52:12–22.
  • Harris CL, Pouw RB, Kavanagh D, et al. Developments in anti-complement therapy; from disease to clinical trial. Mol Immunol. 2018;102:89–119.
  • Mastellos DC, Ricklin D, Lambris JD. Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov. 2019;18:707–729.
  • Ricklin D, Lambris JD. New milestones ahead in complement-targeted therapy. Semin Immunol. 2016;28:208–222.
  • Morgan BP. The membrane attack complex as an inflammatory trigger. Immunobiology. 2016;221:747–751.
  • Triantafilou K, Hughes TR, Triantafilou M, et al. The complement membrane attack complex triggers intracellular Ca2+ fluxes leading to NLRP3 inflammasome activation. J Cell Sci. 2013;126:2903–2913.
  • Fearon DT, Carter RH. The CD19/CR2/TAPA-1 Complex of B Lymphocytes: linking Natural to Acquired Immunity. Annu Rev Immunol. 1995;13:127–149.
  • Taylor BES, Appelboom G, Zilinyi R, et al. Role of the complement cascade in cerebral aneurysm formation, growth, and rupture. Neuroimmunol Neuroinflamm. 2015;2:93–101.
  • Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation. 2020;17:354.
  • Harris CL, Heurich M, de Cordoba SR, et al. The complotype: dictating risk for inflammation and infection. Trends Immunol. 2012;33:513–521.
  • de Córdoba SR, Harris CL, Morgan BP, et al. Lessons from functional and structural analyses of disease-associated genetic variants in the complement alternative pathway. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2011;1812:12–22.
  • Carpanini SM, Torvell M, Morgan BP. Therapeutic inhibition of the complement system in diseases of the central nervous system. Front Immunol. 2019;10:362.
  • Chamberlain JL, Huda S, Whittam DH, et al. Role of complement and potential of complement inhibitors in myasthenia gravis and neuromyelitis optica spectrum disorders: a brief review. J Neurol. 2021;268:1643–1664.
  • Iatropoulos P, Daina E, Curreri M, et al. Cluster analysis identifies distinct pathogenetic patterns in c3 glomerulopathies/immune complex–Mediated membranoproliferative GN. J Am Soc Nephrol. 2018;29:283–294.
  • Wadhwa M, Prabhakar A, Anand JP, et al. Complement activation sustains neuroinflammation and deteriorates adult neurogenesis and spatial memory impairment in rat hippocampus following sleep deprivation. Brain Behav Immun. 2019;82:129–144.
  • Coulthard LG, Woodruff TM. Is the Complement Activation Product C3a a Proinflammatory Molecule? Re-evaluating the Evidence and the Myth. J Immunol. 2015;194:3542–3548.
  • Fredslund F, Laursen NS, Roversi P, et al. Structure of and influence of a tick complement inhibitor on human complement component 5. Nat Immunol. 2008;9:753–760.
  • Jendza K, Kato M, Salcius M, et al. A small-molecule inhibitor of C5 complement protein. Nat Chem Biol. 2019;15:666–668.
  • Law SKA, Dodds AW. The internal thioester and the covalent binding properties of the complement proteins C3 and C4. Protein Sci. 1997;6:263–274.
  • Jore MM, Johnson S, Sheppard D, et al. Structural basis for therapeutic inhibition of complement C5. Nat Struct Mol Biol. 2016;23:378–386.
  • Li XX, Lee JD, Kemper C, et al. The complement receptor C5aR2: a powerful modulator of innate and adaptive immunity. J Immunol. 2019;202:3339–3348.
  • Pandey S, Maharana J, Li XX, et al. Emerging Insights into the structure and function of complement C5a receptors. Trends Biochem Sci. 2020;45:693–705.
  • Reis ES, Chen H, Sfyroera G, et al. C5a receptor-dependent cell activation by physiological concentrations of desarginated C5a: insights from a novel label-free cellular assay. J Immunol. 2012;189:4797–4805.
  • Dmytrijuk A, Robie-Suh K, Cohen MH, et al. FDA report: eculizumab (Soliris®) for the treatment of patients with paroxysmal nocturnal hemoglobinuria. Oncologist. 2008;13:993–1000.
  • Licht C, Greenbaum LA, Muus P, et al. Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2-year extensions of phase 2 studies. Kidney Int. 2015;87:1061–1073.
  • Howard JF, Utsugisawa K, Benatar M, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol Internet]. 2017 [cited 2022 Jul 14];16:976–986. Available from http://www.thelancet.com/article/S1474442217303691/fulltext
  • Bonifati DM, Kishore U. Role of complement in neurodegeneration and neuroinflammation. Mol Immunol. 2007;44:999–1010.
  • Morgan BP. Complement in the pathogenesis of Alzheimer’s disease. Semin Immunopathol Springer Verlag; Jan. 2018;1:113–124.
  • Hakobyan S, Harding K, Aiyaz M, et al. Complement biomarkers as predictors of disease progression in Alzheimer’s disease. J Alzheimers Dis. 2016;54:707–716.
  • Krance SH, Wu CY, Zou Y, et al. The complement cascade in Alzheimer’s disease: a systematic review and meta-analysis. Mol Psychiatry. 2019;26:5532–5541.
  • Benoit ME, Tenner AJ. Complement protein C1q-mediated neuroprotection is correlated with regulation of neuronal gene and MicroRNA expression. J Neurosci. 2011;31:3459–3469.
  • Zhou J, Fonseca MI, Pisalyaput K, et al. Complement C3 and C4 expression in C1q sufficient and deficient mouse models of Alzheimer’s disease. J Neurochem. 2008;106:2080–2092.
  • Stephan AH, Madison DV, Mateos JM, et al. A dramatic increase of C1q protein in the CNS during normal aging. J Neurosci. 2013;33:13460–13474.
  • Woodruff TM, Tenner Yazan AJ, Sanderson SD, et al. Alzheimer’s disease performance in murine models of pathology and enhances behavioral treatment with a C5aR antagonist decreases. J Immunol. 2009;183:1375–1383. Available from: http://www.jimmunol.org/content/183/2/1375
  • Carvalho K, Schartz ND, Balderrama-Gutierrez G, et al. Modulation of C5a–C5aR1 signaling alters the dynamics of AD progression. J Neuroinflammation. 2022;19:1–21. Available from: https://jneuroinflammation.biomedcentral.com/articles/10.1186/s12974-022-02539-2
  • Matsuoka Y, Picciano M, Malester B, et al. Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer’s disease. Am J Pathol. 2001;158:1345–1354.
  • Fonseca MI, Chu SH, Berci AM, et al. Contribution of complement activation pathways to neuropathology differs among mouse models of Alzheimer’s disease. J Neuroinflammation. 2011;8:1–12.
  • Benoit ME, Clarke EV, Morgado P, et al. Complement protein C1q directs macrophage polarization and limits inflammasome activity during the uptake of apoptotic cells. J Immunol. 2012;188:5682–5693.
  • Fonseca M, McGuire S, Counts S, et al. Complement activation fragment C5a receptors, CD88 and C5L2, are associated with neurofibrillary pathology. J Neuroinflammation. 2013;10:25.
  • Ager RR, Fonseca MI, Chu SH, et al. Microglial C5aR (CD88) expression correlates with amyloid-β deposition in murine models of Alzheimer’s disease. J Neurochem. 2010;113:389–401.
  • Linnartz B, Kopatz J, Tenner AJ, et al. Sialic acid on the neuronal glycocalyx prevents complement C1 binding and complement receptor-3-mediated removal by microglia. J Neurosci. 2012;32:946–952.
  • Fonseca MI, Chu SH, Hernandez MX, et al. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J Neuroinflammation. 2017;14:1–15.
  • Fraser DA, Pisalyaput K, Tenner AJ. C1q enhances microglial clearance of apoptotic neurons and neuronal blebs, and modulates subsequent inflammatory cytokine production. J Neurochem. 2010;112:733–743.
  • Fonseca MI, Ager RR, Chu S-H, et al. Treatment with a C5aR antagonist decreases pathology and enhances behavioral performance in murine models of Alzheimer’s disease. J Immunol. 2009;183:1375–1383.
  • Bohlson SS, Fraser DA, Tenner AJ. Complement proteins C1q and MBL are pattern recognition molecules that signal immediate and long-term protective immune functions. Mol Immunol. 2007;44:33–43.
  • Ryman D, Gao Y, Lamb BT. Genetic loci modulating amyloid-beta levels in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2008;29:1190–1198.
  • Carpanini SM, Torvell M, Bevan RJ, et al. Terminal complement pathway activation drives synaptic loss in Alzheimer’s disease models. Acta Neuropathol Commun. 2022;10:1–16. Available from: https://actaneurocomms.biomedcentral.com/articles/10.1186/s40478-022-01404-w
  • Mantovani S, Gordon R, Macmaw JK, et al. Elevation of the terminal complement activation products C5a and C5b-9 in ALS patient blood. J Neuroimmunol. 2014;276:213–218.
  • Xie CB, Qin L, Li G, et al. Complement membrane attack complexes assemble NLRP3 inflammasomes triggering IL-1 activation of IFN-γ-primed human endothelium. Circ Res. 2019;124:1747–1759.
  • Carpanini SM, Harwood JC, Baker E, et al. The impact of complement genes on the risk of late-onset Alzheimer’s disease. Genes (Basel). 2021;12:443.
  • Jun G, Naj AC, Beecham GW, et al. Meta-analysis confirms CR1, CLU, and PICALM as Alzheimer disease risk loci and reveals interactions with APOE genotypes. Arch Neurol. 2010;67:1473–1484.
  • Lambert JC, Heath S, Even G, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet. 2009;41:1094–1099.
  • Grewal RP, Morgan TE, Finch CE. C1qB and clusterin mRNA increase in association with neurodegeneration in sporadic amyotrophic lateral sclerosis. Neurosci Lett. 1999;271:65–67.
  • Kawamata T, Akiyama H, Yamada T, et al. Immunologic reactions in amyotrophic lateral sclerosis brain and spinal cord tissue. Am J Pathol. 1992;140:691.
  • Donnenfeld H, Kascsak RJ, Bartfeld H. Deposits of IgG and C3 in the spinal cord and motor cortex of ALS patients. J Neuroimmunol. 1984;6:51–57.
  • Sta M, Rmr S-S, Casula M, et al. Innate and adaptive immunity in amyotrophic lateral sclerosis: evidence of complement activation. Neurobiol Dis. 2011;42:211–220.
  • Yamada T, Mcgeer PL, Mcgeer EG. Lewy bodies in Parkinson’s disease are recognized by antibodies to complement proteins. Acta Neuropathol. 1992;84:100–104.
  • McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:S3–S7.
  • Singhrao SK, Neal JW, Morgan BP, et al. Increased complement biosynthesis by microglia and complement activation on neurons in Huntington’s disease. Exp Neurol. 1999;159:362–376.
  • Hodges A, Strand AD, Aragaki AK, et al. Regional and cellular gene expression changes in human Huntington’s disease brain. Hum Mol Genet. 2006;15:965–977. Available from: https://academic.oup.com/hmg/article/15/6/965/582386
  • Woodruff TMTM, Costantini KJ, Crane JW, et al. The complement factor C5a contributes to pathology in a rat model of amyotrophic lateral sclerosis. J Immunol. 2008;181:8727–8734.
  • Lee JD, Kumar V, Fung JNT, et al. Pharmacological inhibition of complement C5a-C5a1 receptor signalling ameliorates disease pathology in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Br J Pharmacol. 2017;174:689–699.
  • Woodruff T, Woodruff TM, Crane JW, et al. Therapeutic activity of C5a receptor antagonists in a rat model of neurodegeneration. FASEB J. 2006;20:1407–1417. Available from: https://onlinelibrary.wiley.com/doi/full/10.1096/fj.05-5814com
  • Moller T. Neuroinflammation in Huntington’s disease. J Neural Transm. 2010;117:1001–1008.
  • Heneka MT, Carson MJ, Khoury J, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405. Available from: http://www.sciencedirect.com/science/article/pii/S1474442215700165
  • Frank-Cannon TC, Alto LT, McAlpine FE, et al. Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener. 2009;4:47.
  • Hooten KG, Beers DR, Zhao W, et al. Protective and toxic neuroinflammation in amyotrophic lateral sclerosis. Neurotherapeutics. 2015;12:364–375. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4404435/pdf/13311_2014_Article_329.pdf
  • DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. J Neurochem. 2016;139:136–153.
  • Kölliker-Frers R, Udovin L, Otero-Losada M, et al. Neuroinflammation: an integrating overview of reactive-neuroimmune cell interactions in health and disease. Mediators Inflamm. 2021;2021:1–20.
  • Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009;8: 382–397. Available from: http://www.sciencedirect.com/science/article/pii/S1474442209700626
  • Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.
  • Harry GJ, Kraft AD. Neuroinflammation and microglia: considerations and approaches for neurotoxicity assessment. Expert Opin Drug Metab Toxicol. 2008;4: 1265–1277. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18798697
  • Hong H, Kim BS, Im HI. Pathophysiological role of neuroinflammation in neurodegenerative diseases and psychiatric disorders. Int Neurourol J. 2016;20:S2–S7.
  • Russo I, Barlati S, Bosetti F. Effects of neuroinflammation on the regenerative capacity of brain stem cells. J Neurochem. 2011;116:947–956.
  • Alexander JJ. The complement cascade: yin-Yang in neuroinflammation–neuro-protection and -degeneration. J Neurochem. 2008;107:1169–1187.
  • Orsini F, de Blasio D, Zangari R, et al. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci. 2014;0:380.
  • Warwick CA, Keyes AL, Woodruff TM, et al. The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. J Biol Chem. 2021;297:101085.
  • Alawieh A, Langley EF, Weber S, et al. Identifying the role of complement in triggering neuroinflammation after traumatic brain injury. J Neurosci. 2018;38:2519–2532.
  • Alexander JJ, Anderson AJ, Barnum SR, et al. The complement cascade: yin–Yang in neuroinflammation – neuro-protection and -degeneration. J Neurochem. 2008;107:1169–1187. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/j.1471-4159.2008.05668.x
  • Mallah K, Couch C, Alshareef M, et al. Complement mediates neuroinflammation and cognitive decline at extended chronic time points after traumatic brain injury. Acta Neuropathol Commun. 2021;9:72.
  • Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation. 2018;15:24.
  • Brennan FH, Anderson AJ, Taylor SM, et al. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation. 2012;9:1–13. Available from: https://jneuroinflammation.biomedcentral.com/articles/10.1186/1742-2094-9-137
  • Crehan H, Hardy J, Pocock J. Microglia, Alzheimer‘s disease, and complement. Int J Alzheimers Dis. 2012;2012:983640.
  • Laudisi F, Spreafico R, Evrard M, et al. Cutting edge: the NLRP3 inflammasome links complement-mediated inflammation and IL-1β release. J Immunol. 2013;191:1006–1010.
  • Roy ER, Wang B, Wan YW, et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J Clin Invest. 2020;130:1912–1930. Available from. DOI:10.1172/JCI133737DS1
  • Möller T, Nolte C, Burger R, et al. Mechanisms of C5a and C3a complement fragment-induced [Ca2+](i) signaling in mouse microglia. J Neurosci. 1997;17:615–624.
  • An XQ, Xi W, Gu CY, et al. Complement protein C5a enhances the β-amyloid-induced neuro-inflammatory response in microglia in Alzheimer’s disease. médecine/sciences. 2018;34:116–120.
  • Hschner CNHK S. Complement factor C5a and epidermal growth factor trigger the activation of outward potassium currents in cultured murine microglia. Neuroscience. 1996;73:1109–1120.
  • M Persson MPEHLR. The complement-derived anaphylatoxin C5a increases microglial GLT-1 expression and glutamate uptake in a TNF-alpha-independent manner. Eur J Neurosci. 2009;29:267–274.
  • Miller AM, Stella N. Microglial cell migration stimulated by ATP and C5a involve distinct molecular mechanisms: quantification of migration by a novel near-infrared method. Glia. 2009;57:875–883.
  • Yang C, Yang L, Liu Y. Soluble complement complex C5b-9 promotes microglia activation. J Neuroimmunol. 2014;267:16–19.
  • Schatz-Jakobsen JA, Zhang Y, Johnson K, et al. Structural basis for eculizumab-mediated inhibition of the complement terminal pathway. J Immunol. 2016;197:337–344.
  • Rother RP, Rollins SA, Mojcik CF, et al. Discovery and development of the complement inhibitor eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol. 2007;25:1256–1264.
  • Muppidi S, Utsugisawa K, Benatar M, et al. Long-term safety and efficacy of eculizumab in generalized myasthenia gravis. Muscle Nerve. 2019;60:14–24.
  • Pittock SJ, Lennon VA, McKeon A, et al. Eculizumab in AQP4-IgG-positive relapsing neuromyelitis optica spectrum disorders: an open-label pilot study. Lancet Neurol. 2013;12:554–562.
  • Mantegazza R, O’Brien FL, Yountz M, et al. Consistent improvement with eculizumab across muscle groups in myasthenia gravis. Ann Clin Transl Neurol. 2020;7:1327–1339.
  • Siddiqi ZA, Nowak RJ, Mozaffar T, et al. Eculizumab in refractory generalized myasthenia gravis previously treated with rituximab: subgroup analysis of REGAIN and its extension study. Muscle Nerve. 2021;64:662–669.
  • Howard JF, Karam C, Yountz M, et al. Long-term efficacy of eculizumab in refractory generalized myasthenia gravis: responder analyses. Ann Clin Transl Neurol. 2021;8:1398–1407.
  • Socié G, Caby-Tosi MP, Marantz JL, et al. Eculizumab in paroxysmal nocturnal haemoglobinuria and atypical haemolytic uraemic syndrome: 10-year pharmacovigilance analysis. Br J Haematol. 2019;185:297–310.
  • Zuber J, Le Quintrec M, Krid S, et al. Eculizumab for atypical hemolytic uremic syndrome recurrence in renal transplantation. Am J Transplant. 2012;12:3337–3354.
  • Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med. 2013;368:2169–2181.
  • Palace J, Wingerchuk DM, Fujihara K, et al. Benefits of eculizumab in AQP4+ neuromyelitis optica spectrum disorder: subgroup analyses of the randomized controlled phase 3 PREVENT trial. Mult Scler Relat Disord. 2021;47:102641.
  • Zhu W, Wang Z, Hu S, et al. Human C5-specific single-chain variable fragment ameliorates brain injury in a model of NMOSD. Neurol Neuroimmunol Neuroinflammat. 2019;6:561.
  • Nishimura JI, Ando K, Masuko M, et al. Tesidolumab (LFG316) for treatment of C5-variant patients with paroxysmal nocturnal hemoglobinuria. Haematologica. 2022;107:1483–1488.
  • Roversi P, Ryffel B, Togbe D, et al. Bifunctional lipocalin ameliorates murine immune complex-induced acute lung injury. J Biol Chem. 2013;288:18789–18802.
  • Nunn MA, Sharma A, Paesen GC, et al. Complement inhibitor of C5 activation from the soft tick ornithodoros moubata. J Immunol. 2005;174:2084–2091.
  • Barratt-Due A, Thorgersen EB, Lindstad JK, et al. Ornithodoros moubata complement inhibitor is an equally effective C5 inhibitor in pigs and humans. J Immunol. 2011;187:4913–4919.
  • Hepburn NNJ, Williams AS, Nunn MA, et al. In vivo characterization and therapeutic efficacy of a C5-specific inhibitor from the soft tick Ornithodoros moubata. J of Bio Chemistry. 2007;282:8292–8299.
  • Sezin T, Murthy S, Attah C, et al. Dual inhibition of complement factor 5 and leukotriene B4 synergistically suppresses murine pemphigoid disease. JCI Insight. 2019;4:15.
  • Eskandarpour M, Chen YH, Nunn MA, et al. Leukotriene B4 and its receptor in experimental autoimmune uveitis and in human retinal tissues: clinical severity and LTB4 dependence of retinal Th17 cells. Am J Pathol. 2021;191:320–334.
  • Sadik CD, Rashid H, Hammers CM, et al. Evaluation of nomacopan for treatment of bullous pemphigoid: a phase 2a nonrandomized controlled trial. JAMA Dermatol. 2022;158:641–649.
  • McNamara LA, Topaz N, Wang X, et al. High risk for invasive meningococcal disease among patients receiving eculizumab (Soliris) despite receipt of meningococcal vaccine. MMWR Morb Mortal Wkly Rep. 2019;66:734–737.
  • Konar M, Granoff DM. Eculizumab treatment and impaired opsonophagocytic killing of meningococci by whole blood from immunized adults. Blood. 2017;130:891–899.
  • Gäckler A, Kaulfuß M, Rohn H, et al. Failure of first meningococcal vaccination in patients with atypical haemolytic uraemic syndrome treated with eculizumab. Nephrol Dialysis Transplantation. 2020;35:298–303.
  • Granoff DM, Kim H, Topaz N, et al. Differential effects of therapeutic complement inhibitors on serum bactericidal activity against non-groupable meningococcal isolates recovered from patients treated with eculizumab. Haematologica. 2019;104:e340–e344.
  • Lewis LA, Ram S. Meningococcal disease and the complement system. Virulence. 2014;5:98–126.
  • Muri L, Ispasanie E, Schubart A, et al. Alternative complement pathway inhibition abrogates pneumococcal opsonophagocytosis in vaccine-naïve, but not in vaccinated individuals. Front Immunol. 2021;12:732146.
  • Ispasanie E, Muri L, Schubart A, et al. Alternative complement pathway inhibition does not abrogate meningococcal killing by serum of vaccinated individuals. Front Immunol. 2021;12:4198.
  • Makurvet FD. Biologics vs. Small Molecules: Drug Costs and Patient Access. Med Drug Discov. 2021;9:100075.
  • Levy AR, Chen P, Johnston K, et al. Quantifying the economic effects of ravulizumab versus eculizumab treatment in patients with atypical hemolytic uremic syndrome. J Med Econ. 2022;25:249–259.
  • Wang Y, Johnston K, Popoff E, et al. A US cost-minimization model comparing ravulizumab versus eculizumab for the treatment of atypical hemolytic uremic syndrome. J Med Econ. 2020;23:1503–1515.
  • Biologics: SS. An update and challenge of their pharmacokinetics. Curr Drug Metab. 2014;15:271–290.
  • Mantaj J, Vllasaliu D. Recent advances in the oral delivery of biologics. Pharm J. 2020;304:7933.
  • Oo C, Kalbag SS. Leveraging the attributes of biologics and small molecules, and releasing the bottlenecks: anew wave of revolution in drug development. Expert Review of Clinical Pharmacology. 2016;9:747–749.
  • Smith AJ. New horizons in therapeutic antibody discovery: opportunities and challenges versus small-molecule therapeutics. SLAS Discovery. 2015;20:437–453.
  • Adami G, Saag KG, Chapurlat RD, et al. Balancing benefits and risks in the era of biologics. Ther Adv Musculoskelet Dis. 2019;11.
  • Zelek WM, Morgan BP. Targeting complement in neurodegeneration: challenges, risks, and strategies. Trends Pharmacol Sci. 2022;43:615–628.
  • Scott DE, Bayly AR, Abell C, et al. Small molecules, big targets: drug discovery faces the protein-protein interaction challenge. Nat Rev Drug Discov. 2016;15:533–550.
  • Zhang M, Yang XY, Tang W, et al. Discovery and structural modification of 1-phenyl-3-(1-phenylethyl)urea derivatives as inhibitors of complement. ACS Med Chem Lett. 2012;3:317–321.
  • Mishra R, Rana S. A rational search for discovering potential neutraligands of human complement fragment 5a (hC5a). Bioorg Med Chem. 2019;27:115052.
  • Wang L, Wang N, Zhang W, et al. Therapeutic peptides: current applications and future directions. Signal Transduction and Targeted Therapy. 2022;7:48.
  • Albazli K, Kaminski HJ, Howard J. Complement inhibitor therapy for myasthenia gravis. Front Immunol. 2020;11:917.
  • Ricardo A, Arata M, DeMarco S, et al. Preclinical evaluation of RA101495, a potent cyclic peptide inhibitor of C5 for the treatment of paroxysmal nocturnal hemoglobinuria. Blood. 2015;126:939–939.
  • Ricardo A, Arata M, DeMarco SJ, et al. Development of RA101348, a potent cyclic peptide inhibitor of C5 for complement-mediated diseases. Blood. 2014;124:2936–2936.
  • Howard JF, Nowak RJ, Wolfe GI, et al. Clinical effects of the self-administered subcutaneous complement inhibitor zilucoplan in patients with moderate to severe generalized myasthenia gravis: results of a phase 2 randomized, double-blind, placebo-controlled, multicenter clinical trial. JAMA Neurol. 2020;77:582–592.
  • Macpherson A, Laabei M, Ahdash Z, et al. The allosteric modulation of complement c5 by knob domain peptides. Elife. 2021;10:1–49.
  • Macpherson A, Scott-Tucker A, Spiliotopoulos A, et al. Isolation of antigen-specific, disulphide-rich knob domain peptides from bovine antibodies. PLoS Biol. 2020;18:e3000821.
  • Kusner LL, Yucius K, Sengupta M, et al. Investigational RNAi therapeutic targeting C5 is efficacious in pre-clinical models of myasthenia gravis. Mol Ther Methods Clin Dev. 2019;13:484–492.
  • Finch AM, Wong AK, Paczkowski NJ, et al. Low-molecular-weight peptidic and cyclic antagonists of the receptor for the complement factor C5a. J Med Chem. 1999;42:1965–1974.
  • MND | brisbane, QLD | alsonex [Internet]. [cited 2022 Oct 11]. Available from: https://www.alsonex.com.au/The-technology.
  • Taylor N. ANZCTR - Registration. Australia New Zealand Clinical Trial Registry. 2019. https://www.anzctr.org.au/Trial/Registration/Trial.
  • Hernandez MXM, Namiranian P, Nguyen E, et al. C5a increases the injury to primary neurons elicited by fibrillar amyloid beta. ASN Neuro. 2017;9:1–10. Available from: https://us.sagepub.com/en-us/nam/open-access-at-sage
  • Fella E, Sokratous K, Papacharalambous R, et al. Pharmacological stimulation of phagocytosis enhances amyloid plaque clearance; evidence from a transgenic mouse model of ATTR neuropathy. Front Mol Neurosci. 2017;10:138.
  • Guo Q, Cheng J, Zhang J, et al. Delayed post-injury administration of C5a improves regeneration and functional recovery after spinal cord injury in mice. Clin Exp Immunol. 2013;174:318–325.
  • Mukherjee P, Thomas S, Pasinetti GM. Complement anaphylatoxin C5a neuroprotects through regulation of glutamate receptor subunit 2 in vitro and in vivo. J Neuroinflammation. 2008;5:1–7.