Epitopes and Mechanism of Action of the Clostridium difficile Toxin A-Neutralizing Antibody Actoxumab

https://doi.org/10.1016/j.jmb.2017.02.010Get rights and content

Highlights

  • Actoxumab targets TcdA from C. difficile.

  • It neutralizes TcdA toxicity by preventing TcdA from attaching to host cells.

  • Actoxumab binds to two identical epitopes within the CROP domain of TcdA.

  • The structural basis for the interaction between actoxumab and TcdA is discussed.

  • Data provide mechanistic basis for actoxumab’s protective effects.

Abstract

The exotoxins toxin A (TcdA) and toxin B (TcdB) are produced by the bacterial pathogen Clostridium difficile and are responsible for the pathology associated with C. difficile infection (CDI). The antitoxin antibodies actoxumab and bezlotoxumab bind to and neutralize TcdA and TcdB, respectively. Bezlotoxumab was recently approved by the FDA for reducing the recurrence of CDI. We have previously shown that a single molecule of bezlotoxumab binds to two distinct epitopes within the TcdB combined repetitive oligopeptide (CROP) domain, preventing toxin binding to host cells. In this study, we characterize the binding of actoxumab to TcdA and examine its mechanism of toxin neutralization. Using a combination of approaches including a number of biophysical techniques, we show that there are two distinct actoxumab binding sites within the CROP domain of TcdA centered on identical amino acid sequences at residues 2162–2189 and 2410–2437. Actoxumab binding caused the aggregation of TcdA especially at higher antibody:toxin concentration ratios. Actoxumab prevented the association of TcdA with target cells demonstrating that actoxumab neutralizes toxin activity by inhibiting the first step of the intoxication cascade. This mechanism of neutralization is similar to that observed with bezlotoxumab and TcdB. Comparisons of the putative TcdA epitope sequences across several C. difficile ribotypes and homologous repeat sequences within TcdA suggest a structural basis for observed differences in actoxumab binding and/or neutralization potency. These data provide a mechanistic basis for the protective effects of the antibody in vitro and in vivo, including in various preclinical models of CDI.

Introduction

Clostridium difficile is a Gram-positive, spore-forming bacterium that infects the gastrointestinal tract of both humans and animals. In humans C. difficile infection (CDI) can cause mild symptoms such as a low-grade fever, watery stools, and minor abdominal cramping, as well as more severe symptoms such as bloody diarrhea, pseudomembrane colitis, toxic megacolon, and death [1]. Individuals whose normal gut flora has been compromised by treatment with antibiotics are most at risk for CDI. Over the past few decades, the incidence of CDI has increased throughout the developed world and is now a major health concern. Most often transmitted in a healthcare facility setting, C. difficile has become the most commonly reported pathogen in hospitals in the United States [2] and causes over 14,000 deaths per year. Currently, CDI is treated with standard of care antibiotics vancomycin, metronidazole, and fidaxomicin. Despite the high efficacy of these agents in treating an initial episode of CDI, 25 to 30% of patients will suffer a recurrence within 3 months [3], with subsequent recurrences occurring at an even higher rate. Thus, there is a great need to develop novel therapies that will reduce the risk of recurrence.

The symptoms of CDI are primarily caused by the exotoxins toxin A (TcdA) and toxin B (TcdB), which are produced by the bacterium during the infection [4], [5], [6], [7]. TcdA and TcdB are structurally similar proteins, each having four separate domains: an amino-terminal glucosyltransferase domain (GTD), internal autoprotease and translocation domains, and a combined repetitive oligopeptide (CROP) domain at the carboxy-terminus. The CROP domains of TcdA and TcdB are composed of multiple short repeats (SRs; 32 in TcdA and 20 in TcdB) interspersed with a smaller number of long repeats (LRs; 7 in TcdA and 4 in TcdB) and have been presumed to play a role in receptor binding [8]. Both toxins bind to intestinal epithelial cells, and possibly other mucosal cells, and are internalized through receptor-mediated endocytosis [9]. The low pH environment of the endosome triggers a conformational change in the protein, resulting in the translocation of the GTD across the endosomal membrane and into the cytoplasm [10], [11], [12]. The autoprotease domain then cleaves the GTD [13], allowing it to diffuse through the cytoplasm and inactivate small GTPases of the Ras superfamily (particularly the Rho subfamily but also Rap and Ras) through covalent glucosylation [14], [15], resulting in actin depolymerization, inflammatory cytokine production, and cell death [16], [17], [18].

While much is known about the trafficking of TcdA and TcdB and their mechanisms of action once internalized into target cells, exactly how the toxins bind to cells and through which receptors is less clear. Because different cell types show different levels of susceptibility to each toxin, it is believed that TcdA and TcdB bind to different receptors. Truncated versions of TcdA and TcdB lacking the CROP domain are still capable of intoxicating cells, albeit with lower potency than intact toxins, showing that regions outside the CROP domain are also involved in receptor binding [19], [20]. Recently, poliovirus receptor-like protein 3, chondroitin sulfate proteoglycan 4, and members of the Wnt receptor frizzled family have been identified as putative cellular receptors for TcdB [21], [22], [23]. The TcdB CROP domain appears to be not necessary for binding to poliovirus receptor-like protein 3 or frizzled family protein members. While the potential receptors for TcdB identified thus far are membrane proteins, the receptor for TcdA is thought to be a cell surface carbohydrate [24]. The LRs in the CROP domain may serve as receptor binding sites, since a crystal structure of a C-terminal fragment of the TcdA CROP domain in complex with α-Gal-(1,3)-β-Gal-(1,4)-β-GlcNAcO(CH2)8CO2CH3 shows binding of the carbohydrate to residues located around the LR regions [25].

The need for more effective treatments for CDI has led to the development of alternative non-antibiotic therapies. Foremost among these are the two fully human monoclonal antibodies actoxumab and bezlotoxumab, which target TcdA and TcdB, respectively. The combination of actoxumab and bezlotoxumab is highly protective in primary and recurrent animal models of CDI [26], [27], [28], [29]. In clinical trials, bezlotoxumab alone or in combination with actoxumab significantly reduced the rate of CDI recurrence when co-administered with standard of care antibiotics [30]. We have previously shown that bezlotoxumab binds to two separate but homologous epitopes centered on LR1 and LR2 within the TcdB CROP domain and prevents toxin binding to host cells [31]. In this study, we characterized the binding of actoxumab to TcdA and show that the antibody binds at two distinct binding sites centered on two of the seven LRs in the TcdA CROP domain and that similar to bezlotoxumab, the antibody neutralizes toxin activity by preventing binding to host cells.

Section snippets

Mechanism of TcdA neutralization by actoxumab

The CROP domain of TcdA has historically been considered to be the primary receptor binding domain of the toxin [32]. Specifically, pockets within the LR regions of the CROP domain have been implicated in mediating the binding of TcdA to carbohydrate moieties on the host cell surface [25]. Since actoxumab is known to bind to the CROP domain of TcdA [26], we assessed the possibility that the mechanism of neutralization of TcdA by actoxumab involves the blockade of TcdA binding to host cells,

Discussion

Actoxumab and bezlotoxumab bind to TcdA and TcdB, respectively, neutralizing the activity of the toxins both in vitro and in vivo [26], [27], [28], [29], [30], [35], [36], [37]. Because of their potential therapeutic use for the prevention of recurrent CDI and their possible use as tools to gain further insight into toxin structure and function, there is much interest in determining the nature of the interactions of the antibodies with their respective toxin. The interaction of bezlotoxumab

Cell lines, antibodies, and purified toxins

Vero and HT29 cells were purchased from the American Type Culture Collection and grown at 37 °C in 5% CO2. Vero cells were maintained in Eagle's minimal essential medium supplemented with 10% fetal bovine serum, 100 U/ml penicillin, and 100 U/ml streptomycin. HT29 cells were maintained in McCoy's 5A Modified Medium supplemented with 10% fetal bovine serum, 2 mM glutamine, 0.75% sodium bicarbonate, 100 U/mL penicillin, and 100 U/mL streptomycin. Actoxumab and bezlotoxumab were generated in humanized

References (51)

  • A. Shen

    Clostridium difficile toxins: mediators of inflammation

    J. Innate Immun.

    (2012)
  • W.K. Smits et al.

    Clostridium difficile infection

    Nat. Rev. Dis. Primers

    (2016)
  • R.N. Pruitt et al.

    Toward a structural understanding of Clostridium difficile toxins A and B

    Front. Cell. Infect. Microbiol.

    (2012)
  • P. Papatheodorou et al.

    Clostridial glucosylating toxins enter cells via clathrin-mediated endocytosis

    PLoS One

    (2010)
  • M. Qa'Dan et al.

    pH-induced conformational changes in Clostridium difficile toxin B

    Infect. Immun.

    (2000)
  • H. Barth et al.

    Low pH-induced formation of ion channels by Clostridium difficile toxin B in target cells

    J. Biol. Chem.

    (2001)
  • G. Pfeifer et al.

    Cellular uptake of Clostridium difficile toxin B. Translocation of the N-terminal catalytic domain into the cytosol of eukaryotic cells

    J. Biol. Chem.

    (2003)
  • J. Reineke et al.

    Autocatalytic cleavage of Clostridium difficile toxin B

    Nature

    (2007)
  • I. Just et al.

    Glucosylation of Rho proteins by Clostridium difficile toxin B

    Nature

    (1995)
  • I. Just et al.

    The enterotoxin from Clostridium difficile (ToxA) monoglucosylates the Rho proteins

    J. Biol. Chem.

    (1995)
  • M. Qa'Dan et al.

    Clostridium difficile toxin B activates dual caspase-dependent and caspase-independent apoptosis in intoxicated cells

    Cell. Microbiol.

    (2002)
  • R. Gerhard et al.

    Glucosylation of Rho GTPases by Clostridium difficile toxin A triggers apoptosis in intestinal epithelial cells

    J. Med. Microbiol.

    (2008)
  • A. Olling et al.

    The repetitive oligopeptide sequences modulate cytopathic potency but are not crucial for cellular uptake of Clostridium difficile toxin A

    PLoS One

    (2011)
  • B. Schorch et al.

    LRP1 is a receptor for Clostridium perfringens TpeL toxin indicating a two-receptor model of clostridial glycosylating toxins

    Proc. Natl. Acad. Sci. U. S. A.

    (2014)
  • P. Yuan et al.

    Chondroitin sulfate proteoglycan 4 functions as the cellular receptor for Clostridium difficile toxin B

    Cell Res.

    (2015)
  • Cited by (32)

    • Novel structural insights for a pair of monoclonal antibodies recognizing non-overlapping epitopes of the glucosyltransferase domain of Clostridium difficile toxin B

      2022, Current Research in Structural Biology
      Citation Excerpt :

      As part of the long-standing efforts of tackling CDAD by developing mAbs, structures of toxin neutralizing Fabs in complex with different domains of TcdA or TcdB have also been reported. These include the crystal structure of a Fab fragment of bezlotoxumab bound to the N-terminal half of the TcdB CROP domain (Orth et al., 2014), and the Fab fragment of actoxumab bound to a portion of the TcdA RBD (Hernandez et al., 2017). Murase et al. published structures of the RBDs of TcdA and TcdB bound by neutralizing and non-neutralizing single-domain antibodies (Murase et al., 2014).

    • Antibacterial lead compounds and their targets for drug development

      2019, Phytochemicals as Lead Compounds for New Drug Discovery
    • New insights for vaccine development against Clostridium difficile infections

      2019, Anaerobe
      Citation Excerpt :

      It is noteworthy that Bezlotoxumab binds to specific amino acids in the N-terminal domain located in the combined repetitive oligopeptides (CROP) domain [55], blocking the binding to host cells of TcdB [56]. X-ray crystal structure studies of complex between the bezlotoxumab and the amino-terminal half of the TcdB CROP domain demonstrated that bezlotoxumab binds to two homologous but distinct epitopes that overlap the LR1 and LR2 [56], thus preventing TcdB binding to host cell surfaces [56] By contrast, epitopes for Actoxumab binds on different faces of the CROP domain, and therefore cannot bind both epitopes with one molecule of antibody due to steric exclusion, preventing simultaneous binding of both epitopes [57]. These observations suggest that the lack of efficacy found in clinical trials for Actoxumab could be due to impaired binding of antibodies to TcdA, more than to the lack of virulence of TcdA, which has been demonstrated to be essential for the manifestation of CDI [58].

    • Deciphering the domain specificity of C. difficile toxin neutralizing antibodies

      2019, Vaccine
      Citation Excerpt :

      The combination of two murine mAbs to the CTD region of TcdA demonstrated significantly more toxin neutralization than the same concentration of the two mAbs individually [23]. A2, the most potent anti-TcdA mAb we had previously identified, binds multiple A/VTGWQTI sites within the CTD [20] and the toxin neutralizing mAb actoxumab binds to two distinct sites in the in the CROPs of the CTD of TcdA [24]. Taken together these data create a picture that suggests that multiple antibody binding events, whether spread across a single domain or across the entire toxin molecule, can produce synergistic protection against intoxication and that the toxin neutralizing benefit of targeting multiple epitopes is a concept that applies to both TcdB and TcdA.

    View all citing articles on Scopus
    6

    Present address: J. Karczewski, Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, 19711, USA.

    7

    Present address: P. Gupta, Merck Exploratory Sciences Center, Cambridge, MA, 02141, USA.

    8

    Present address: A.G. Therien, Inception Sciences, Montreal, Quebec, H4S 2A1, Canada.

    View full text