1,303
Views
2
CrossRef citations to date
0
Altmetric
Editorial

Catching the evader: Can monoclonal antibodies interfere with <italic>Staphylococcus aureus</italic> immune escape?

Pages 1-4 | Received 07 Apr 2017, Accepted 07 Apr 2017, Published online: 02 May 2017
This article refers to:
A human monoclonal antibody that specifically binds and inhibits the staphylococcal complement inhibitor protein SCIN

Approximately 20% of human beings harbor Staphylococcus aureus (S. aureus) in their nares or other body sites.Citation1 Most S. aureus carriers never develop infection while others usually suffer from mild skin and soft tissue infections (SSTIs). Under certain circumstances, however, S. aureus can gain access to deeper tissues or the blood and cause life-threatening conditions.Citation2 So far, our most effective response to this aggression has been to poison the bacterium with bactericidal agents. This approach has saved countless lives, but it has also led to a constant arms race between our ability to develop new drugs and the ability of S. aureus to evolve antimicrobial resistance.Citation3 Not only do we have no guarantee that we will eventually win this race against natural selection, but we are also increasingly concerned with the collateral damages that antimicrobial drugs cause to the bacteria of our microbiote and the environment.Citation4,5

We face, thus, the urgent need to develop more sustainable strategies against infection. To put the arms race to an end, a crucial approach is to favor highly specific therapies that interfere only with the cause of the disease while limiting consequences on uninvolved organisms. Although trivial at first glance, this specificity objective has an important evolutionary underpinning: pathogen evolution does not select for severe disease.Citation6 Deep-seated infection, either by leading to the rapid death of the human host or by limiting her social activity, lowers the odds of colonizing new hosts for the S. aureus population. From this standpoint, severe staphylococcal infections that do not favor transmission, such as bacteremia, should be considered as evolutionary accidents caused by bacteria escaping their ecological niche, the human surfaces. Targeting bacteria only outside of their niche should, thus, lower the ecological pressure of therapy given that the bacterial invaders are often already engaged in an evolutionary dead-end: they will eventually be cleared by the immune system, or die with their host.

The ecological niche of S. aureus is distinct from the tissues whose invasion leads to severe disease. Hence, therapeutic specificity can be considered not only from a taxonomic standpoind (the antimicrobial spectrum) but also from an anatomic one, by restricting the therapeutic action to deep tissues and vital organs, while leaving mucosae unaffected. Several technical means can be contemplated to achieve this goal, including targeted drug delivery.Citation7 Nonetheless, the most efficiently targeted antimicrobial strategy is arguably that of our own immune system. In physiologic conditions, immunity rapidly clears bacteria from tissues while allowing the survival of microbiote inhabitants, including S. aureus. A treatment meant to assist the immune system to provide an effective and balanced response to aggression—that is, pathogen eradication without excessive inflammation—should impose minimal selection pressure on transmissible, non-invading bacteria, thus fulfilling the sustainability objective.

Restoring a complete immune activity against S. aureus is difficult, however, due to the exceptional ability of S. aureus to evade both the innate and adaptive immune systems.Citation8 The recognition of S. aureus surface antigens by antibodies is hampered by the staphylococcal protein A, reducing opsonization and phagocytosis. When phagocytosis succeeds, S. aureus still manages to survive within the phagosome,Citation9 disrupts its membrane or subverts the autophagic pathway, eventually killing the phagocyte.Citation10,11 The activation of the complement pathway is hampered by several virulence factors such as the staphylococcal complement inhibitor SCIN.Citation12 Several secreted cytotoxins and leukotoxins activate and lyse immune cells before they even reach the bacteria.Citation13 The versatility of these immune evasion strategies is currently considered a major candidate explanation of the repeated failures of anti-S. aureus vaccine strategies in clinical trials.Citation14

Nonetheless, clinical observations suggest that adaptive immunity does contribute to controlling staphylococcal infections. For instance, patients who develop S. aureus bacteremia with their own colonizing strain are less likely to die than non-colonized patients,Citation15 and patients with high antibody titers against several staphylococcal toxins are less likely to develop severe sepsis during S. aureus infection.Citation16 With the hypothesis that adaptive immunity affords patients protection from severe staphylococcal infections comes the hope to determine the right combination of antigens (in active immunization strategies) or antibodies (in passive immunotherapy) that will mimic or reinforce an effective immunity with preventive or curative objectives. Given that most staphylococcal virulence factors are neither necessary nor sufficient to cause severe infection, however, blocking only one factor is unlikely to afford universal protection.Citation17 Alternatively, we might consider these pathogenic functions in a cumulative fashion, where each of them contributes independently to the probability and severity of infection. A multi-targeted, or polyvalent, intervention might aim at blocking these functions one after another until the probability (severity) of infection becomes low enough to achieve preventive (curative) efficacy.

To follow this research direction toward polyvalent immunotherapy, a crucial task is to rank the potential targets by their contribution to the condition we want to prevent. Many staphylococcal targets have been identified so far, including toxins, surface proteins or quorum sensing mediators. S. aureus toxins such as the α haemolysin (Hla) or the Panton-Valentine leukocidin (PVL), which are involved in lethal necrotizing pneumonia, have been understandably considered major candidates for both passive and active immunization strategies.Citation18 However, concerns have been raised by several authors that immunization against PVL might be ineffective at preventing SSTIs,Citation19 based on the clinical observation that elevated PVL antibody titers did not prevent recurrence of PVL-associated SSTIs and the more worrisome conclusion of animal models that PVL immunization might enhance such infections.Citation20,21 Strikingly, a recent study of toxin production in colonizing, SSTI and bacteremia S. aureus isolates demonstrated that bacteremia isolates were significantly less toxic than their colonizing and SSTI counterparts,Citation22 thus suggesting that toxin production might indeed decrease the ability of S. aureus to reach or survive within the bloodstream.

These results can seem counterintuitive from the usual viewpoint that virulence leads to severe infections. They become coherent, however, if we lean toward an evolutionary perspective and consider that colonization and SSTIs, not bacteremia or lethal pneumonia, contribute to the transmission and the long-term survival of staphylococcal populations. In the context of colonization and superficial infections, pro-inflammatory toxins such as Hla and PVL might have evolved not only as bacterial defense mechanisms, but also as warning signals meant to alert immune cells of tissue invasion and to preserve the host from an uncontrolled, lethal infection. In contrast, releasing the same pro-inflammatory toxins in a vital organ such as the lung, which threatens the host's life without favoring S. aureus transmission, is unlikely to have provided any evolutionary benefit. Although speculative and difficult to test, this dual-faceted interpretation of toxin activity is further supported by animal models in which blocking the inflammatory response induced by the interaction of Hla with its ADAM10 eukaryotic receptor abolished mortality in experimental pneumonia but strongly increased the dermonecrosis area in experimental SSTI.Citation23 Collectively, these observations and their proposed interpretation imply that toxins should be considered with caution when used as immunotherapeutic targets: the curative use of anti-toxin antibodies probably limits inflammatory-induced damages in deep-seated infections,Citation18 however their usefulness for preventing the infection is still uncertain.Citation21

Contrary to pro-inflammatory toxins, non-toxic immune evasion factors have not been involved so far in mechanisms beneficial to the host, lowering the risk that their inhibition might be detrimental. Preclinical models of immunization against protein A,Citation24,25 which captures the constant fragment of immunoglobulins, or the coagulase,Citation26 which coats the bacterium with a phagocytosis-inhibiting fibrin shield, have shown promising results. In this issue of Virulence, Hoekstra et al. describe properties of a neutralizing monoclonal antibody against a major immune evasion factor of S. aureus, the complement inhibitor SCIN.Citation27 The research strategy of the authors stemmed from the clinical observation that patients with the epidermolysis bullosa chronic skin disease, associated with long-term S. aureus colonization, develop bacteremia less frequently than would have been expected given their impaired skin barrier. In these patients, high antibody titers against several S. aureus antigens were hypothesized to confer protection against bacterial invasion. Monoclonal antibodies against S. aureus, including the so-called 6D4 antibody targeting SCIN, were then identified by collecting and screening B-cells of these naturally immunized patients. The 6D4 antibody binds to amino acid residues that overlap with the previously identified active site of SCIN. 6D4 treatment restored both the deposition of C3b of S. aureus surface and the complement-induced lysis of rabbit erythrocytes in the presence of SCIN, hence demonstrating a functional neutralizing activity of the complement-inhibiting function.

In spite of this SCIN-neutralizing activity, Hoekstra et al. prudently suggest that the 6D4 antibody might not be useful for therapeutic purposes given that SCIN-negative isolates still cause disease. This argument is undebatable as long as the 6D4 antibody is to be used alone for therapy. In the framework of polyvalent immunotherapy, however, the 6D4 antibody probably deserves more attention due to its specificity, in vitro efficacy, well-determined interaction with the SCIN active site and, importantly, its primary isolation from patients suspected to exhibit anti-S. aureus protective immunity. Hence, further research should determine whether and to which extent SCIN inhibition contributes to protect against S. aureus infection in vivo. Whether active or passive polyvalent immunotherapy will finally succeed and provide us with efficient and sustainable solutions against S. aureus infections is uncertain. Major technical obstacles must be overcome, such as the limited ability of animal models to predict the clinical efficacy of anti-S. aureus immunization approaches.Citation14 Research groups and industries who have independently developed virulence-inhibiting strategies should also be encouraged to collaborate and examine the therapeutic potential of combined strategies. This research task is difficult and risky, but the long-term potential of its sustainability objective is probably worth the effort. Meanwhile, each contribution to the arsenal of S. aureus immune evasion-inhibiting agents, including the 6D4 antibody, should be regarded as a significant step toward this important objective.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References

  • Weidenmaier C, Goerke C, Wolz C. Staphylococcus aureus determinants for nasal colonization. Trends Microbiol 2012; 20:243-50; PMID:22494802; https://doi.org/10.1016/j.tim.2012.03.004
  • Lowy FD. Staphylococcus aureus infections. N Engl J Med 1998; 339:520-32; PMID:9709046; https://doi.org/10.1056/NEJM199808203390806
  • Cameron DR, Howden BP, Peleg AY. The interface between antibiotic resistance and virulence in staphylococcus aureus and its impact upon clinical outcomes. Clin Infect Dis 2011; 53:576-82; PMID:21865195; https://doi.org/10.1093/cid/cir473
  • Becattini S, Taur Y, Pamer EG. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol Med 2016; 22:458-78; PMID:27178527; https://doi.org/10.1016/j.molmed.2016.04.003
  • Venter H, Henningsen ML, Begg SL. Antimicrobial resistance in healthcare, agriculture and the environment: The biochemistry behind the headlines. Essays Biochem 2017; 61:1-10; PMID:28258225; https://doi.org/10.1042/EBC20160053
  • Johnson RB. Human disease and the evolution of pathogen virulence. J Theor Biol 1986; 122:19-24; PMID:3796006; https://doi.org/10.1016/S0022-5193(86)80222-3
  • Sharma A, Kumar Arya D, Dua M, Chhatwal GS, Johri AK. Nano-technology for targeted drug delivery to combat antibiotic resistance. Expert Opin Drug Deliv 2012; 9:1325-32; PMID:22924701; https://doi.org/10.1517/17425247.2012.717927
  • Fedtke I, Götz F, Peschel A. Bacterial evasion of innate host defenses–the staphylococcus aureus lesson. Int J Med Microbiol 2004; 294:189-94; PMID:15493829; https://doi.org/10.1016/j.ijmm.2004.06.016
  • Kubica M, Guzik K, Koziel J, Zarebski M, Richter W, Gajkowska B, Golda A, Maciag-Gudowska A, Brix K, Shaw L, et al. A potential new pathway for staphylococcus aureus dissemination: The silent survival of s. aureus phagocytosed by human monocyte-derived macrophages. PLoS One 2008; 3:e1409; PMID:18183290; https://doi.org/10.1371/journal.pone.0001409
  • McGuinness WA, Kobayashi SD, DeLeo FR. Evasion of neutrophil killing by staphylococcus aureus. Pathogens 2016; 5:E32; PMID:26999220; https://doi.org/10.3390/pathogens5010032
  • O'Keeffe KM, Wilk MM, Leech JM, Murphy AG, Laabei M, Monk IR, Massey RC, Lindsay JA, Foster TJ, Geoghegan JA, et al. Manipulation of autophagy in phagocytes facilitates staphylococcus aureus bloodstream infection. Infect Immun 2015; 83:3445-57; PMID:26099586; https://doi.org/10.1128/IAI.00358-15
  • Garcia BL, Ramyar KX, Ricklin D, Lambris JD, Geisbrecht BV. Advances in understanding the structure, function, and mechanism of the SCIN and Efb families of staphylococcal immune evasion proteins. Adv Exp Med Biol 2012; 946:113-33; PMID:21948365; https://doi.org/10.1007/978-1-4614-0106-3_7
  • Vandenesch F, Lina G, Henry T. Staphylococcus aureus hemolysins, bi-component leukocidins, and cytolytic peptides: A redundant arsenal of membrane-damaging virulence factors?. Front Cell Infect Microbiol 2012; 2:12; PMID:22919604; https://doi.org/10.3389/fcimb.2012.00012
  • Pier GB. Will there ever be a universal staphylococcus aureus vaccine?. Hum Vaccin Immunother 2013; 9:1865-76; PMID:23793522; https://doi.org/10.4161/hv.25182
  • Wertheim HFL, Vos MC, Ott A, van Belkum A, Voss A, Kluytmans JAJW, van Keulen PHJ, Vandenbroucke-Grauls CMJE, Meester MHM, Verbrugh HA. Risk and outcome of nosocomial staphylococcus aureus bacteraemia in nasal carriers versus non-carriers. Lancet 2004; 364:703-5; PMID:15325835; https://doi.org/10.1016/S0140-6736(04)16897-9
  • Adhikari RP, Ajao AO, Aman MJ, Karauzum H, Sarwar J, Lydecker AD, Johnson JK, Nguyen C, Chen WH, Roghmann MC. Lower antibody levels to staphylococcus aureus exotoxins are associated with sepsis in hospitalized adults with invasive S. aureus infections. J Infect Dis 2012; 206:915-23; PMID:22807524; https://doi.org/10.1093/infdis/jis462
  • Otto M. Novel targeted immunotherapy approaches for staphylococcal infection. Expert Opin Biol Ther 2010; 10:1049-59; PMID:20528609; https://doi.org/10.1517/14712598.2010.495115
  • Diep BA, Le VTM, Badiou C, Le HN, Pinheiro MG, Duong AH, Wang X, Dip EC, Aguiar-Alves F, Basuino L, et al. IVIG-mediated protection against necrotizing pneumonia caused by MRSA. Sci Transl Med 2016; 8:357ra124; PMID:27655850; https://doi.org/10.1126/scitranslmed.aag1153
  • Hermos CR, Yoong P, Pier GB. High levels of antibody to panton-valentine leukocidin are not associated with resistance to staphylococcus aureus-associated skin and soft-tissue infection. Clin Infect Dis 2010; 51:1138-46; PMID:20946065; https://doi.org/10.1086/656742
  • Yoong P, Pier GB. Antibody-mediated enhancement of community-acquired methicillin-resistant Staphylococcus aureus infection. Proc Natl Acad Sci USA 2010; 107:2241-6; PMID:20133867; https://doi.org/10.1073/pnas.0910344107
  • Yoong P. Enhancement of bacterial virulence by antibody neutralization of immune-activating toxins. Virulence 2010; 1:409-13; PMID:21178480; https://doi.org/10.4161/viru.1.5.12705
  • Laabei M, Uhlemann AC, Lowy FD, Austin ED, Yokoyama M, Ouadi K, Feil E, Thorpe HA, Williams B, Perkins M, et al. Evolutionary trade-offs underlie the multi-faceted virulence of staphylococcus aureus. PLoS Biol 2015; 13:e1002229; PMID:26331877; https://doi.org/10.1371/journal.pbio.1002229
  • Becker REN, Berube BJ, Sampedro GR, DeDent AC, Bubeck Wardenburg J. Tissue-specific patterning of host innate immune responses by staphylococcus aureus α-toxin. J Innate Immun 2014; 6:619-31; PMID:24820433; https://doi.org/10.1159/000360006
  • Kim HK, Cheng AG, Kim H-Y, Missiakas DM, Schneewind O. Nontoxigenic protein a vaccine for methicillin-resistant staphylococcus aureus infections in mice. J Exp Med 2010; 207:1863-70; PMID:20713595; https://doi.org/10.1084/jem.20092514
  • Kim HK, Emolo C, DeDent AC, Falugi F, Missiakas DM, Schneewind O. Protein A-specific monoclonal antibodies and prevention of staphylococcus aureus disease in mice. Infect Immun 2012; 80:3460-70; PMID:22825452; https://doi.org/10.1128/IAI.00230-12
  • Thomer L, Emolo C, Thammavongsa V, Kim HK, McAdow ME, Yu W, Kieffer M, Schneewind O, Missiakas D. Antibodies against a secreted product of staphylococcus aureus trigger phagocytic killing. J Exp Med 2016; 213:293-301; PMID:26880578; https://doi.org/10.1084/jem.20150074
  • Hoekstra H, Romero Pastrana F, Bonarius HPJ, van Kessel KPM, Elsinga GS, Kooi N, Groen H, van Dijl JM, Buist G. A human monoclonal antibody that specifically binds and inhibits the staphylococcal complement inhibitor protein SCIN. Virulence 2017. Advance online publication. PMID:28277903; https://doi.org/10.1080/21505594.2017.1294297