IAI Accepts, published online ahead of print on 10 November 2014 Infect. Immun. doi:10.1128/IAI.02550-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Toxin-mediated paracellular transport of antitoxin antibodies facilitates protection against

2

Clostridium difficile infection

3 4

Z. Zhanga, X. Chenb, L. Hernandeza, P. Liparia, A. Flatterya, S.-C. Chena, S. Kramera, J. D.

5

Polishooka, F. Racinea, H. Capea, C. P. Kellyb, A. G. Theriena#

6 7

Merck Research Laboratories, Merck & Co, Inc, Kenilworth, NJa; Division of Gastroenterology,

8

Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MAb

9 10

Running head: Paracellular transport of antibodies in the gut

11 12

#

Address Correspondence to Alex G. Therien, [email protected]

13

The exotoxins TcdA and TcdB are the major virulence factors of Clostridium difficile.

14

Circulating neutralizing antitoxin antibodies are protective in C. difficile infection (CDI) as

15

demonstrated, in part, by the protective effects of actoxumab and bezlotoxumab, which

16

bind to and neutralize TcdA and TcdB, respectively. The question of how systemic IgG

17

antibodies neutralize toxins in the gut lumen remains unresolved, although it has been

18

suggested that the Fc-receptor FcRn may be involved in active antibody transport across

19

the gut epithelium. In this study, we demonstrate that genetic ablation of FcRn and excess

20

irrelevant human IgG has no impact on actoxumab/bezlotoxumab-mediated protection in

21

murine and hamster models of CDI, suggesting that Fc-dependent transport of antibodies

22

across the gut wall is not required for efficacy. Tissue distribution studies in hamsters

23

suggest, rather, that the transport of antibodies depends on toxin-induced damage to the

24

gut lining. In an in vitro two-dimensional culture system that mimics the architecture of the

25

intestinal mucosal epithelium, toxins on the apical side of epithelial cell monolayers are

26

neutralized by basolateral antibodies, and antibody transport across the cell layer is

27

dramatically increased upon addition of toxin to the apical side. Similar data were obtained

28

with F(ab’)2 fragments, which lack an Fc domain, consistent with FcRn-independent

29

paracellular, rather than transcellular, transport of antibodies. Kinetic studies show that

30

initial damage caused by apical toxin is required for efficient neutralization by basolateral

31

antibodies. These data may represent a general mechanism of humoral response-mediated

32

protection against enteric pathogens.

33 34

35

Introduction

36

The enteric pathogen Clostridium difficile is a gram-positive, anaerobic, spore-forming

37

bacterium. C. difficile infections (CDI) cause diarrhea, pseudomembranous colitis and in some

38

severe cases, colonic rupture and death (1). In recent years, CDI-associated morbidity and

39

mortality has increased significantly and the disease poses a significant healthcare threat in the

40

United States and globally (2).

41

The major virulence factors of C. difficile are the Rho-inactivating toxins A and B (TcdA and

42

TcdB) that consist of large single-chain proteins with similar multidomain structures and

43

functions (3-5). It is thought that both toxins bind to target mammalian cells (typically gut

44

epithelial cells) at least in part through their C-terminal receptor-binding domains (combined

45

repetitive oligopeptide, or CROP domains) and become internalized via receptor-mediated

46

endocytosis (6, 7). Following internalization, acidification of the endosome leads to a

47

conformational change within the toxins that results in translocation of the glucosyltransferase

48

domain (GTD) across the endosomal membrane and auto-cleavage via the cysteine protease

49

domain (8-10). This releases the GTD into the cytoplasm where it inactivates rho-type GTPase

50

by covalent glucosylation, resulting in disruption of the cytoskeleton, changes in cellular

51

morphology and eventually, cell death (11, 12). The resulting disruption of the gut epithelial

52

barrier leads to the symptoms of the disease, which are exacerbated by toxin-mediated

53

recruitment of a pro-inflammatory host immune response (3-5, 13).

54

Although standard of care antibiotic therapy with metronidazole, vancomycin or fidaxomicin is

55

often effective in resolving primary cases of CDI, ~25% of patients develop one or more

56

recurrent episode of CDI even after an initial cure (1, 14). Multiple lines of evidence suggest that

57

adaptive humoral immune responses against the C. difficile toxins are protective in both primary

58

and recurrent CDI. Kyne et al. first showed that circulating levels of anti-TcdA IgG were

59

positively correlated with a lower rate of primary CDI in colonized patients (15) and with a

60

lower rate of recurrence among patients who had suffered a primary episode of CDI (16). A

61

more recent study has shown that anti-TcdB IgG levels also correlate with protection against

62

CDI recurrence (17). While correlative in nature, these studies provided the impetus to test the

63

hypothesis that antitoxin antibodies might be protective in CDI, and this has now been

64

demonstrated in multiple animal models (18-22). More significantly for human disease, passive

65

immunotherapy with the antitoxin neutralizing antibody combination consisting of actoxumab

66

and bezlotoxumab (specific for TcdA and TcdB, respectively) has been shown to reduce CDI

67

recurrence in human patients (23). The combination of actoxumab and bezlotoxumab, both fully-

68

human IgG1 antibodies, is currently in phase III clinical development for the prevention of

69

recurrent CDI.

70

Despite the evidence that circulating antitoxin antibodies are protective in CDI, the question of

71

how systemic IgG antibodies bind to and neutralize toxins presumably located largely in the gut

72

lumen remains unanswered. Previous studies have shown that the neonatal immunoglobulin

73

receptor FcRn mediates specific transport of IgG antibodies across the gut wall (24, 25) and

74

plays a role in antibody-mediated protection against infections of the gastrointestinal tract in

75

mice (26, 27). In this study, we use both in vitro and in vivo model systems to explore the

76

mechanism through which neutralizing antitoxin IgG antibodies cross the gut epithelium and

77

neutralize C. difficile toxins located in the lumen of the gut, thereby protecting the host against

78

CDI. We show that trans-epithelial neutralization depends on non-specific paracellular transport

79

of the antibodies rather than specific transcellular Fc-receptor-mediated transport mechanisms,

80

and that toxin-induced damage paradoxically facilitates the transport of (and toxin neutralization

81

by) antitoxin antibodies.

82 83

Materials and Methods

84

Toxins and cells

85

Purified TcdA and TcdB from strain VPI 10463 were purchased from Native Antigen

86

(Oxfordshire, UK). T84 cells were cultured in DMEM:F12 medium (ATCC) supplemented with

87

5% fetal bovine serum (FBS) and Penicillin-Streptomycin (P/S). Caco-2 cells were cultured in

88

MEM (ATCC) supplemented with 10%FBS, nonessential amino acids, sodium-bicarbonate and

89

P/S. MDCK cells were cultured in DMEM (Life Tech.) supplemented with 10%FBS plus P/S.

90

Mouse primary CDI model

91

The mouse antibiotic-associated CDI model has previously been described (28). Briefly, 8 week

92

old, female, C57BL6 wildtype or FcRn-/- mice (purchased from Jackson Laboratory) were orally

93

administrated with an antibiotic mixture of kanamycin (40 mg/kg/d), gentamicin (3.5 mg/kg/d),

94

colistin (4.2mg/kg/d), metronidazole (21.5 mg/kg/d), and vancomycin (4.5 mg/kg/d) for 3 days

95

followed, 2 days later, by a single dose of clindamycin (10 mg/kg) intraperitoneally 24 hour prior

96

to challenge with 1x105 cfu C. difficile (strain VPI 10463) by gavage. Actoxumab and/or

97

bezlotoxumab were administered intraperitoneally to mice each at a dose of 250 µg, given 1 day

98

prior to challenge with C. difficile. Statistical analysis was carried out using the Logrank test

99

with Bonferroni correction on GraphPad Prism 6 software (GraphPad Sotware, San Diego, CA).

100

Mouse recurrent CDI model

101

Mice were treated with oral vancomycin (50 mg/kg, administered by gavage) once daily for 5

102

days following C. difficile challenge. Actoxumab and bezlotoxumab in combination were

103

administered intraperitoneally each at a dose of 250 µg, given 1 day prior to challenge with C.

104

difficile and again 1 day after discontinuing vancomycin therapy. Statistical significance was

105

determined using the Logrank test with Bonferroni correction on GraphPad Prism 6 software

106

(GraphPad Sotware, San Diego, CA).

107

Hamster CDI model

108

C. difficile B1 spores (generously provided by D. Gerding, Hines VA, Hines, IL) were prepared

109

from confluent cultures grown anaerobically on agar plates for 12 days. Cells were washed from

110

the surface of the agar in an aerobic environment using sterile ice cold diH2O, then left on ice for

111

3 hrs. Spores were placed in microfuge tubes and were separated from vegetative cells by

112

centrifugation at 13,000 rpm for 3 min at 4°C, then further purified by serial washes in ice cold

113

diH2O and repeat centrifugation. Spores were resuspended in diH2O, quantified by anaerobically

114

incubating aliquots on Clostridium difficile Selective Agar (CDSA) supplemented with 0.12ml

115

10% sodium taurocholate in sterile water, and aliquots stored at -80°C. Male Golden Syrian

116

hamsters approximately 100g (Charles River Laboratories) were preconditioned for C. difficile

117

susceptibility by oral administration of clindamycin at 30 mg/kg five days prior to infectious

118

spore challenge. On Day 0, hamsters were infected with a saline suspension containing ~50

119

spores of toxigenic C. difficile strain B1. Hamsters were divided into 3 treatment groups of 6

120

animals each, plus one group of infected controls (n=2). Treatment groups included

121

actoxumab/bezlotoxumab at 50 mg/kg each subcutaneously (SC) once-daily (qd) for 4 days

122

beginning 5 h after infectious challenge; non-specific human IgG (huIgG; Equitech-Bio., Inc.)

123

administered at 2 g/kg SC qd for 4 days beginning 4 h after infectious challenge; or a

124

combination of the two above treatment regimens (actoxumab/bezlotoxumab + huIgG).

125

Hamsters were monitored at least twice-daily for morbidity, mortality and signs of disease

126

including diarrhea (“wet tail”), body weight loss, lethargy, hunched posture or distended

127

abdomen. Animals were euthanized if judged to be in a moribund state or if weight loss

128

exceeded 20%. Statistical significance was determined using the Logrank test with Bonferroni

129

correction on GraphPad Prism 6 software (GraphPad Sotware, San Diego, CA).

130

Tissue distribution of actoxumab/bezlotoxumab in hamsters

131

Male Golden Syrian hamsters were preconditioned with clindamycin on Day -5 relative to

132

infectious spore challenge, as described above. On Day 0, hamsters were divided into 2 groups,

133

one left untreated and the second infected with ~50 spores of C. difficile strain B1. At 5 hours

134

after spore challenge infected and uninfected hamsters received a single SC dose of

135

actoxumab/bezlotoxumab at 50 mg/kg each. Hamsters were euthanized at timepoints after

136

administration of antibodies and gastrointestinal tract (GI) from the stomach to the rectum was

137

collected. The GI tract was sectioned into duodenum, jejunum, ileum, cecum and ascending

138

colon. Cecum contents were collected undiluted, and contents of other GI sections were

139

obtained from the lumen by flushing each section with 1ml sterile phosphate buffered saline

140

(PBS). GI tissue was then thoroughly rinsed with sterile PBS, weighed and homogenized in

141

sterile PBS containing 10% glycerol (5 mL for cecum and 1.5 mL for other GI sections).

142

Concentration of human IgG in each tissue or lumenal contents was determined using a human

143

IgG quantitation ELISA (Bethyl Laboratories). Statistical significance was determined by first

144

converting the data to log of antibody concentration, carrying out unpaired, two-tailed t-tests

145

comparing healthy versus CDI hamsters, and applying the Bonferroni correction for multiple

146

comparisons.

147

Immunohistochemistry

148

Hamsters were preconditioned with clindamycin and infected (or not) with C. difficile strain B1

149

spores as described above and dosed with actoxumab/bezlotoxumab (50 mg/kg of each antibody)

150

subcutaneously, once 5h after spore challenge and a second time 24h later. Cecum tissues were

151

collected 24h after the second dose of actoxumab/bezlotoxumab or vehicle from healthy,

152

untreated hamsters (unchallenged and dosed with vehicle), healthy, treated hamsters

153

(unchallenged with C. difficile spores and dosed with actoxumab/bezlotoxumab) or diseased,

154

treated hamsters (challenged with C. difficile spores and dosed with actoxumab/bezlotoxumab),

155

and fixed with 10% neutral buffered formalin for 22h and subsequently processed for formalin

156

fixed paraffin embedded sections. Localization of injected actoxumab/bezlotoxumab was carried

157

out using a goat anti-human IgG antibody from Jackson ImmunoResearch (West Grove, PA)

158

followed by sequential incubation with a goat HRP polymer and diaminobenzidine in a

159

SuperPictureTM Polymer Detection Kit (Invitrogen, Grand Island, NY). Tissue sections were

160

counterstained with Hematoxylin (Biocare Medical, Concord, CA) for structural analysis.

161

Generation of (Fab’)2 fragments of actoxumab and bezlotoxumab

162

F(ab’)2 fragments of actoxumab and bezlotoxumab were generated using the Pierce F(ab’)2

163

Preparation Kit (Thermo Scientific, Waltham, MA, USA) according to the manufacturer's

164

instructions. Fc fragments were removed from F(ab’)2 by gel filtration chromatography using a

165

Superdex-S200 column (GE Healthcare Life Sciences, Piscataway, NJ, USA) pre-equilibrated

166

with PBS. Five ml fractions were collected and the peaks corresponding to the F(ab’)2 fragment

167

were pooled. F(ab’)2 purity was ascertained under reducing and non-reducing conditions by

168

SDS-PAGE, using 4–12% Novex Tris-Glycine gels (Life Technologies, Grand Island, NY,

169

USA), followed by Coomassie brilliant blue staining. No contamination of samples by uncleaved

170

IgG was detectable (Fig. S4A and B)

171

Two-dimensional cell culture and transepithelial electrical resistance (TER) measurements

172

To establish the two-dimension culture system, cells were seeded on the well insert (BD Falcon

173

HTS 24-multiwell insert, 351181; Bedford, MA, USA) at 0.5 - 1.0x105 cells per well, with 250

174

l medium in the apical chamber and 800 l medium in the basolateral chamber. Cells were

175

cultured for ≥14 days at 37oC (in 5% CO2) to ensure full differentiation and confluency, as

176

assessed by transepithelial electrical resistance (TER) measurements plateauing at ≥600 Ω.cm2

177

(Caco-2) or ≥1000 Ω.cm2 (T84), measured using the Epithelial Volt-Ohm Meter Millicell ERS-2

178

(EMD Millipore, Billerica, MA, USA). For fast-growing MDCK cells, maximal TER (plateauing

179

at ≥600Ωcm2) was achieved 1-2 days after seeding. For assessments of toxin neutralization,

180

actoxumab or bezlotoxumab (or F(ab’)2 fragments thereof) were added to a final concentration of

181

50 g/ml to the apical chamber or 100 g/ml to the basolateral chamber, either 18h before or

182

immediately before addition of toxin to the apical chamber (see figure legends). TER

183

measurements were obtained at different time points either immediately before (t = 0h) or at

184

various time points after addition of various concentrations of TcdA or TcdB to either the apical

185

or basolateral chambers. TER measurements were normalized to values obtained in the absence

186

of toxin at each time point to account for minor time-dependent variability. Ten l samples were

187

taken from the apical chamber either immediately before addition of the toxin (t = 0h) or at

188

various time points after addition of toxin (see figure legends) for quantitation of antibody

189

concentration. Fifty l samples were taken from the basolateral chamber 48h after addition of

190

toxin for quantitation of toxin concentration (see below). Statistical significance was determined

191

by matched two-way ANOVA with Tukey’s multiple comparison test using GraphPad Prism 6

192

software (GraphPad Sotware, San Diego, CA).

193

ELISAs

194

ELISAs were carried out according to standard methodology. Briefly, high-protein-binding

195

ELISA plates (Fisher Scientific, Waltham, MA, USA) were coated with capture antibody

196

(chicken anti-human F(ab’)2 secondary antibody, from Fisher SA1-72043) at 10 g/ml and

197

blocked using blocking buffer (Fisher Scientific). Samples isolated from apical chamber of two-

198

dimensional culture system (see above) were diluted in blocking buffer and added to wells, with

199

purified actoxumab, bezlotoxumab or F(ab’)2 fragments thereof used as standards. Wells were

200

washed and HRP-linked secondary antibody (300 ng/ml; Fisher Scientific) in blocking buffer

201

was added for detection of bound antibodies. Following extensive washing, HRP substrate

202

solution (Fisher Scientific) was added and luminescent signal was read on a SpectraMax M4

203

instrument. Quantification of toxins in the basolateral chamber was carried out by ELISA using a

204

kit from tgcBIOMICS (Bingen, Germany) and following the manufacturer’s instructions.

205

Statistical significance was determined by matched two-way ANOVA with Tukey’s multiple

206

comparison test using GraphPad Prism 6 software (GraphPad Sotware, San Diego, CA).

207

Ethics Statement

208

All procedures in animals were performed in accordance with the highest standards for the

209

humane handling, care and treatment of research animals, and adhered to the National Research

210

Council’s “Guide for Care and Use of Laboratory Animals, 8th Edition”. Mouse studies were

211

approved by the Beth Israel Deaconess Medical Center (BIDMC) Institutional Animal Care and

212

Use Committee (protocol number 101-2013). Hamsters studies were approved by the Merck &

213

Co., Inc. (in Kenilworth, NJ) Institutional Animal Care and Use Committee (protocol number

214

0280-12).

215 216

Results

217

Role of Fc-mediated transport in protection against C. difficile infection.

218

The neonatal Fc receptor FcRn has previously been implicated in transport of IgG from the

219

systemic to the lumenal side of the gut wall (24, 25, 29) and has been shown to play a role in

220

IgG-mediated protection against some forms of bacterial infections (26, 27). To assess whether

221

these observations extend to C. difficile infections, we evaluated the human IgG1 antibody

222

combination actoxumab/bezlotoxumab in a murine CDI model comparing FcRn knockout mice

223

with WT littermates. Since efficacy of actoxumab/bezlotoxumab had not previously been

224

demonstrated in mice, we first administered actoxumab, bezlotoxumab, or a combination of both

225

antibodies intraperitoneally to WT mice 24h before challenge with the toxigenic C. difficile

226

strain VPI 10463. The antibody combination provided excellent protection against mortality in

227

this model, while the individual antibodies alone were only partially protective (Fig. 1A).

228

Furthermore, in both primary (Fig 1B) and recurrent (Fig 1C) CDI paradigms (see “Materials

229

and Methods”), a single administration of actoxumab/bezlotoxumab increased survival to the

230

same extent in WT mice compared to FcRn-null mice, demonstrating that the FcRn receptor does

231

not play a significant role in antibody-mediated protection in this murine model. These data,

232

however, do not rule out the possibility that other Fc-mediated transport mechanisms may be

233

involved in the efficacy of actoxumab/bezlotoxumab. To address this, we used the gold standard

234

Syrian hamster model of CDI, in which actoxumab/bezlotoxumab has previously been

235

demonstrated to be protective (18). Since targeted genetic deletion is not possible in hamsters,

236

and to cover any and all possible Fc-mediated activities of actoxumab/bezlotoxumab, we co-

237

dosed the antibody combination in a therapeutic paradigm (see “Materials and Methods”) with or

238

without a 40-fold excess of irrelevant human IgG (which contains ~63% IgG1; data not shown),

239

reasoning that such antibodies should compete with actoxumab/bezlotoxumab for binding to Fc

240

receptors. As shown in Fig. 1D, co-dosing with excess human IgG had no significant impact on

241

actoxumab/bezlotoxumab-mediated protection, suggesting that such protection is independent of

242

Fc-mediated transport mechanisms.

243

Tissue distribution of actoxumab/bezlotoxumab in the hamster CDI model

244

To gain insight into whether and to what extent systemic antibodies reach the site of infection in

245

the lumen of the gut, we characterized the intestinal tissue distribution of systemically

246

administered actoxumab and bezlotoxumab in hamsters. Hamsters challenged with or without a

247

toxigenic strain of C. difficile (strain B1; see (18)) were injected with a single dose of

248

actoxumab/bezlotoxumab at 50 mg/kg. Samples of intestinal tissues were collected from both

249

infected and uninfected animals at various time points after challenge and levels of human IgG

250

were measured by ELISA in lumenal contents and in the washed whole gut tissues. While no

251

significant differences were observed in actoxumab/bezlotoxumab levels within the intestinal

252

tissues between infected and uninfected hamsters (Fig. 2A), levels of actoxumab/bezlotoxumab

253

in the GI lumen were significantly higher in infected hamsters, in particular in the cecum, where

254

antibody levels were nearly undetectable in healthy animals (Figs. 2B and S1). Increased

255

antibody transport across the gut wall in diseased hamsters was confirmed at the cellular level

256

using immunohistochemistry (Figs. 2C, S1C). In healthy hamsters, antibodies are primarily

257

located in the subepithelial space (including the lamina propria) of the mucosa, with no staining

258

in the epithelial layer, which acts as a barrier preventing leakage of antibodies into the gut lumen.

259

Conversely, in hamsters infected with C. difficile, the actoxumab/bezlotoxumab signal is present

260

throughout the mucosa, including parts of the epithelial layer, which exhibits significant

261

damage/sloughing, allowing antibodies to enter the intestinal lumen.. Together, these data show

262

that transport of systemic antibodies to the gut lumen is significantly facilitated by damage to the

263

gut epithelium mediated by C. difficile toxins.

264

Transepithelial toxin neutralization by antibodies in a two-dimensional culture system

265

To better understand the protective role of circulating antitoxin antibodies against toxins in the

266

gut lumen, we used a two-dimensional cell culture system wherein apical and basolateral

267

compartments are separated by a single monolayer of differentiated epithelial cells (30-33). The

268

system mimics the polarized nature of the intact intestinal mucosal epithelium which separates

269

the gut lumen (apical side) from the subepithelial/systemic space (basolateral side). The integrity

270

of the epithelial layer is monitored by measuring the transepithelial electrical resistance (TER); a

271

drop in TER indicates that the integrity of the epithelial monolayer has been compromised (31).

272

For studies aimed at understanding how neutralizing antibodies present on the basolateral/

273

systemic side can neutralize toxin on the apical/lumenal side, we first demonstrated that TcdA

274

and TcdB added to the apical chamber cause significant time- and concentration-dependent

275

decreases in TER in the colonic epithelial cell line, Caco-2 (Fig. 3A and B). To confirm that the

276

epithelial monolayer was fully differentiated and polarized, we replicated the previously-

277

published observation (33) that colonic epithelial cells are more sensitive to TcdB applied to the

278

basolateral side compared to the apical side, whereas sensitivity to TcdA is comparable on both

279

sides (Fig. S2). The effects of the neutralizing antibodies actoxumab and bezlotoxumab on toxin-

280

induced damage are shown in Fig. 3C,D, E and F for Caco-2 cells (and in Fig. S3 for T84 cells).

281

When applied to the same side (apical) as the toxins, the antibodies show a strong neutralizing

282

effect on the toxins as shown by rightwards shifts in the concentration response curves of the

283

toxins (shown 24h after addition of toxin; Figs. 3E and F). Significant, though smaller, shifts are

284

also observed when antibodies are added to the basolateral/systemic side of the epithelium,

285

approximating the protection afforded by systemically-circulating neutralizing antibodies in the

286

context of CDI. To assess the potential role of Fc-mediated transport in transepithelial

287

neutralization, we generated F(ab’)2 fragments of actoxumab and bezlotoxumab, which lack an

288

Fc region. The F(ab’)2 fragments, which were first shown to be devoid of contamination by

289

uncleaved antibody and fully neutralizing in a toxin induced cell death assay (34) (Fig. S4),

290

neutralized the effects of toxins at least as efficiently as intact antibodies (Figs. 3E and F),

291

demonstrating that the Fc regions of IgG molecules on the basolateral/systemic side of the

292

epithelial layer are not necessary for transepithelial toxin neutralization. Supporting this notion,

293

addition of excess irrelevant human IgG to the basolateral chamber had no impact on the

294

transepithelial neutralization of toxin (Fig. S5).

295

Mechanism of antibody transport across the epithelial monolayer

296

Neutralization of apical toxin by basolateral antibodies strongly suggests that antibodies are

297

transported to the apical side. To confirm this, we measured the extent to which antibody applied

298

to the basolateral chamber translocates to the apical chamber in the presence and absence of

299

toxin on the apical side. Similar to the TER assays described above, 100 g/ml antibody was

300

added to the basolateral side of a confluent monolayer of Caco-2 cells 18h prior to addition of

301

buffer or toxin at different concentrations, and antibody concentration in the apical chamber was

302

measured by ELISA at different time points. As shown in figure 4A and B (and in Fig. S6A and

303

B for T84 cells), transport of actoxumab and bezlotoxumab into the apical chamber is minimal

304

18h after addition of antibodies to the basolateral chamber (at t = 0h with respect to the time of

305

toxin addition). Transport of antibodies into the apical chamber increases with time in the

306

presence of toxin and this effect is dependent on the concentration of toxin added to the apical

307

chamber. Indeed, while the concentration of antibodies on the apical side is 40% and >20% in the presence of 64 ng/ml TcdA or 256 ng/ml TcdB,

310

respectively. The dependence of antibody transport on toxin is presumed to result from toxin-

311

dependent disruptions in the epithelial barrier function leading to increased non-specific

312

paracellular leakage of antibodies to the apical side, analogous to the high levels of antibodies

313

observed in C. difficile infected hamsters versus healthy hamsters (Fig. 2). Importantly, transport

314

of F(ab’)2 fragments of actoxumab and bezlotoxumab was at least as high as intact antibody in

315

the presence and absence of toxin (Fig. 4C and D), confirming that Fc-dependent transport

316

mechanisms such as FcRn are not involved in antibody transport in this system. Indeed,

317

concentrations of F(ab’)2 were consistently higher in the apical chamber, particularly at high

318

toxin concentration. This may be due to (i) the smaller size of F(ab’)2 fragments allowing them

319

to leak through paracellular gaps more efficiently and/or (ii) a slightly higher molar

320

concentration of F(ab’)2 added to the basolateral chamber compared to intact antibodies (since

321

100 g/ml each of intact antibodies and F(ab’)2 fragments were used in these experiments, and

322

the molecular mass of F(ab’)2 is ~1.5-fold lower than intact antibody) .

323

Antibody-induced recovery of epithelial monolayer following toxin-induced damage

324

In order to better understand how toxin neutralization might protect the gut epithelium in the

325

context of the gut wall (where damage is repaired much faster than in isolated epithelial cells

326

owing to the presence of other cells and growth factors in the intact gut wall (35)), we assessed

327

transepithelial neutralization and transport in the two-dimensional culture assay using MDCK

328

cells, which proliferate at a much faster rate in vitro than gut epithelial cells. Similar to Caco-2

329

and T84 cells (Figs. 3 and S3), we observed a robust TcdA-dependent decrease in TER and

330

significant neutralization of apical TcdA by basolateral actoxumab (Fig. 5A and B). Interestingly,

331

the protection afforded by actoxumab was biphasic, with a significant yet incomplete effect at 6h

332

and complete neutralization (except for the highest concentration of TcdA) at 24h. We confirmed

333

that toxin is indeed fully neutralized at 24h by transferring the contents of the apical chambers at

334

this timepoint (TcdA concentrations up to 64 ng/ml) to intact MDCK and Caco-2 monolayers,

335

and showing limited effects on TER after 24h (data not shown). These data are consistent with

336

the notion that toxin on the apical side must first cause damage to the epithelial layer (shown by

337

a drop in TER at 6h) in order for the antibody on the basolateral side to fully neutralize apical

338

toxin.

339

In parallel with these TER measurements, we assessed transport of antibody to the apical side

340

24h after addition of various concentration of toxin (Figs. 5C and S6C). Surprisingly, the apical

341

concentration of actoxumab was

Toxin-mediated paracellular transport of antitoxin antibodies facilitates protection against Clostridium difficile infection.

The exotoxins TcdA and TcdB are the major virulence factors of Clostridium difficile. Circulating neutralizing antitoxin antibodies are protective in ...
1MB Sizes 0 Downloads 7 Views