This article was downloaded by: [Lakehead University] On: 18 March 2015, At: 23:59 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Human Vaccines & Immunotherapeutics Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/khvi20

Vaccines against Clostridium difficile a

a

a

Rosanna Leuzzi , Roberto Adamo & Maria Scarselli a

Novartis Vaccines S.r.L.; Siena, Italy Published online: 17 Mar 2014.

Click for updates To cite this article: Rosanna Leuzzi, Roberto Adamo & Maria Scarselli (2014) Vaccines against Clostridium difficile, Human Vaccines & Immunotherapeutics, 10:6, 1466-1477, DOI: 10.4161/hv.28428 To link to this article: http://dx.doi.org/10.4161/hv.28428

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

ReviewReview Human Vaccines & Immunotherapeutics 10:6, 1466–1477; June 2014; © 2014 Landes Bioscience

Vaccines against Clostridium difficile Rosanna Leuzzi1, Roberto Adamo1, and Maria Scarselli1 Novartis Vaccines S.r.L.; Siena, Italy

1

Clostridium difficile infection (CDI) is recognized as a major cause of nosocomial diseases ranging from antibiotic related diarrhea to fulminant colitis. Emergence during the last 2 decades of C. difficile strains associated with high incidence, severity and lethal outcomes has increased the challenges for CDI treatment. A limited number of drugs have proven to be effective against CDI and concerns about antibiotic resistance as well as recurring disease solicited the search for novel therapeutic strategies. Active vaccination provides the attractive opportunity to prevent CDI, and intense research in recent years led to development of experimental vaccines, 3 of which are currently under clinical evaluation. This review summarizes recent achievements and remaining challenges in the field of C. difficile vaccines, and discusses future perspectives in view of newly-identified candidate antigens.

Introduction Clostridium difficile is a gram-positive anaerobic bacterium able to infect either humans or animals1 and commonly found in the environment.2,3 It was isolated for the first time in 1935 from the intestinal flora of neonates and was initially considered a normal non-pathogenic resident of the gut.4 Only in 1970s was C. difficile identified as one of the agents responsible for antibiotic-related diarrhea and pseudomembranous colitis.5 C. difficile can exist as spores: metabolically inactive particles able to survive in soil, water, and on surfaces in clinical settings, due to resistance against common sterilization methods such as high temperatures, ultraviolent light, alcohol.6,7 Spores represent the main vehicle for transmission, infection, and persistence of C. difficile. If ingested, spores can survive in the stomach of infected subjects and subsequently reach the intestine. Here, their fate strongly depends on the environment provided by the host. Spores typically switch from their dormant state to become active vegetative cells in a process termed germination. Germination can occur in response to stimuli including the bile salts, taurocholate, glycocholate and cholate,8,9 amino acids10 and factors present in intestinal epithelial cells.11 However, the action of such germination effectors (germinants) is effectively prevented by another bile acid, chenodeoxycholate, which has 10-fold higher affinity than taurocholate to the C. difficile spores.12 *Correspondence to: Maria Scarselli; Email: [email protected] Submitted: 11/29/2013; Revised: 02/22/2014; Published Online: 03/17/2014 http://dx.doi.org/10.4161/hv.28428

1466

The presence of chenodeoxycholate in association with aerobic conditions likely inhibits germination and growth of the bacterium during its passage through the small intestine. In the large intestine of healthy individuals, spores can persist asymptomatically as resident commensal species and can rapidly degrade any residual bile component, preventing their germination.13 On the contrary, in absence or reduction of the normal commensal flora, a condition typically induced by treatment with wide-spectrum antibiotics, spores become able to germinate into vegetative cells and the absence of natural competitors for nutrients permits C. difficile to colonize empty niches in the colonic tract. Once vegetative cells have been released from the germinant spores, the contact with host epithelial cells triggers the upregulation of bacterial genes responsible for adaptation to the new environment.14,15 The bacterium remodels its surface by exposing adhesins, flagella, and proteolytic enzymes including Cwp84,16 which promotes the maturation of structural components of the bacterial cell wall17 and degrades elements of the host epithelium such as fibronectin, vitronectin, laminin, and fibrinogen.18 It has been suggested that the lytic action targeting host tissues induces the release of nutrients from the damaged epithelium and also promotes toxin diffusion.18 C. difficile cells can indeed cause disease by secreting 2 large enterotoxins, TcdA and TcdB, both able to severely damage the intestinal mucosa.19 These toxins have glycosyltransferase activity and modify small GTPases of the Rho protein family within the host cell, leading to alterations of cytoskeleton, activation of apoptosis and disruption of tight junctions.20 The resulting impairment of intestinal barrier function leads to fluid accumulation, inflammation, and severe intestinal damage.19,21,22 Although mechanisms that regulate toxin production are not completely elucidated, there are evidences that toxin synthesis is enhanced by several stimuli including metabolic stress,23,24 temperature,25 and sub-lethal doses of antibiotics.26-29 Healthy individuals are generally able to mount a robust systemic immunity that limits gut damage induced by the toxins.30,31 On the contrary, elderly or immuno-compromised subjects are prone to a series of symptoms whose severity ranges from mild diarrhea to fulminant pseudomembranous colitis.32 In addition to TcdA and TcdB, up to 35% of C. difficile strains produce a third toxin called CDT or binary toxin,33-36 composed by the ADP-ribosyltransferase subunit CDTa and the binding subunit CDTb.37 CDT binds to the lipolysis-stimulated lipoprotein receptor protein on the host cells,38 and the toxin-receptor complex is internalized into endocytotic vescicles. Subsequently, CDTa is released into the cytosol where it inhibits actin polymerization leading to profound alterations of the cell morphology,39 including formation of microtubule protrusions that trap

Human Vaccines & Immunotherapeutics

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

Keywords: Clostridium difficile, vaccine, CDI

www.landesbioscience.com

more effective than vancomycin to prevent relapses, likely due to higher capability to preserve the intestinal microflora and favor beneficial re-colonization.69 CDI is not limited to healthcare settings.74,75 It has been estimated that 10–37% of all CDI cases are community associated (CA-CDI), with a population incidence of 20–30 cases per 100 000.76,77 Diffusion of CDI imposes a considerable burden on patients in terms of morbidity, mortality, and prolonged hospitalization. Moreover, huge economical costs appear to weight on healthcare systems of developed countries. In Europe, potential costs associated to CDI management have been calculated at around € 3000 million per year,54 while costs directly attributable to CDI in US acute healthcare facilities in 2008 have been estimated at between 1 and $4.8 billion USD.78 Additional costs may derive from recurrent CDI, adverse effects on elderly individuals, as well as from infection treatment and management in LTCFs. Vaccination can represent a valuable strategy to prevent CDI. In the next sections we describe the efforts to identify and characterize suitable vaccine candidates against C. difficile and the state-of-the-art of preventive vaccines currently under clinical evaluation.

C. difficile Vaccines: From Toxoids to Recombinant Peptides TcdA and TcdB are the determinants of CDI; however, the relative role of immunity against the 2 toxins in preventing the C. difficile associated disease has been long discussed. Early studies on purified proteins indicated that, differently from TcdB, TcdA alone was able to reproduce C. difficile mediated disease in animal models79-81 and that anti-TcdA antibodies were necessary and sufficient to ensure complete protection against the clinical signs of infection.30,31 More recently, the role of TcdB in C. difficile virulence82 and the relevance of anti-TcdB antibodies to prevent gastro-intestinal disease83,84 have been re-evaluated, together with the identification of TcdA-negative, TcdB-positive strains responsible for severe clinical symptoms.85-87 Collectively, such evidences support the conclusion that TcdB is a key factor in C. difficile disease and clearly suggests that neither toxin can be ignored in the development of an effective vaccine.88 A further debate in the literature focused on the question of whether a mucosal or systemic antibody response is more important in protection against CDI. Several studies were driven by the assumption that local anti-toxin immunity was necessary to confer protection, since CDI in humans is clearly confined to intestinal tract. However, passive immunization studies demonstrated that circulating anti-toxin antibodies are effective in the treatment of severe CDI,70,89,90 strongly suggesting that antibody-mediated toxin neutralization has an effect at the level of the epithelial barrier. The protective effect of circulating antibodies at the site of damage is likely due to the ability of toxins to subvert the epithelial permeability. C. difficile toxins alter the epithelial barrier by disrupting the cytoskeleton organization and destabilizing the tight junctions.19,20 This peculiar cytotoxic activity suggests that

Human Vaccines & Immunotherapeutics 1467

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

C. difficile on the surface of intestinal epithelial cells40. The genes encoding CDTa and CDTb have been rarely found among isolates recovered from hospitalized patients,41,42 but they are conserved in emerging strains associated with severe virulence.43-45 It is therefore believed that CDT might play an adjunctive role in pathogenesis by enhancing the persistence of the bacterium in the colonized host.46,47 Since the early 2000s cases of CDI-associated disease dramatically increased in the United States,44,46,48 Canada,49-51 and Europe,52-54 accompanied by increase of case-fatality rates.48,55 In the US, the incidence of CDI in acute care hospitals increased from 3.82 per 1000 discharges in the year 2000, to 8.75 per 1000 discharges in 2008.56 A rise of CDI cases has also been observed in Europe. Data from the Communicable Diseases Surveillance Centre (CDSC) reported in England and Wales an increase from 1000 cases of CDI-associated disease in 1990, to 35 500 cases in 2003.54 A survey supported by the European Centre for Disease Prevention and Control (ECDC) revealed that the mean incidence of CDI in Europe passed from 2.45 cases per 10 000 patient days in 2005, to 4.1 cases per 10 000 patient days in 2008.52,53 A hyper-virulent strain called NAP1/027/BI was identified as being partly responsible for the increasing incidence of CDI in healthcare settings, although the reasons for its virulence are under discussion and include increased toxin production,57 higher sporulation efficiency,58 and greater antibiotic resistance.50,59 Moreover, increased antibiotic use, an aging population with more comorbidities, as well as more frequent and accurate testing have been indicated as additional factors contributing to the increased CDI incidence.60 Today C. difficile is the most common pathogen associated with nosocomial infectious diarrhea in hospitalized patients.61,62 The Centers for Disease Control and Prevention (CDC) reported in 2012 that in the US among CDI cases with onset in healthcare facilities, approximately one half had onset in acute care units and the other half in long-term care facilities (LTCF).63 Recovery is complicated by the tendency of disease to relapse. After the initial episode of CDI, a significant minority of patients is subjected to recurrences, resulting either from relapses due to persistent infection by the same strain or re-acquisition of a new strain from the environment.64 The main factors responsible for recurrent CDI are persistence of alterations of intestinal microflora and inability to elicit an effective immune response. Antibiotic therapy for patients with mild to moderate CDI typically consists of oral metronidazole, while vancomycin is recommended for more severe cases or subjects with intolerance to metronidazole.65-67 As antibiotics commonly used for CDI treatment cause alterations of fecal microbiota, their protracted administration prevents the re-establishment of natural resistance to C. difficile and might thereby predispose the host to recurrent infections.68 After the initial episode, up to 33% of patients experience recurrent CDI69-72 and recurrences can reach 45% after a second episode73. This scenario is particularly frequent in healthcare settings, where the major part of the high-risk population is concentrated. A novel antibiotic, fidaxomycin, has recently been approved for CDI treatment.69 Fidaxomicin appears to be

Downloaded by [Lakehead University] at 23:59 18 March 2015

Vaccination with Toxoids The efficacy of anti-CDI vaccines based on the administration of formalin inactivated toxoids A and B has been described for over 3 decades. This vaccine consists of formalin-detoxified toxins A and B obtained by purification from culture of VPI 10463, which is a hyper-productive strain for both toxins. The first evidences demonstrated that the concomitant administration of toxoid A and B protected hamsters from lethal CDI.94,95 After these initial studies many investigations explored the efficacy of toxoid-induced immunity in animal models by the evaluation of parameters influencing the vaccine efficacy such as the level of toxoid purification, the use of adjuvants and the routes of immunization, with a particular focus on the choice of the antigen delivery system. Torres and colleagues evaluated the efficacy of chemically detoxified culture filtrates at different antigen doses and immunization routes in the hamster model.96 They indicated that a combination of mucosal and systemic immunization induced protection against lethal outcome and diarrhea in hamsters. The combined use of mucosal and parenteral administration of both toxoids was further investigated by the use of a partially purified toxoid preparation based on fractionation of culture filtrates with ammonium sulfate.89 Full protection was achieved only in animals vaccinated with a combination of rectal immunization with E. coli heat-labile toxin (LT) adjuvant and intramuscular injection of alum-adjuvanted toxoids. Importantly, the vaccination conferred protection to hamsters although the absence of anti-TcdA antibodies in feces suggesting that circulating antibodies were responsible for the recovery from the disease manifestations.89 Similarly, transcutaneous immunization with toxoid A from culture filtrates has been reported to induce in mice both systemic IgG response and mucosal IgA response in serum and stool.97 Whereas most of the previous studies were based on vaccination with toxoid A preparations, a recent study established that parenteral immunization with purified toxoid B conferred protection to CDI in hamsters infected with a TcdA-negative, TcdBpositive strain.98 Importantly, it indicated that immunization of hamsters with a toxoid B preparation from the conventional toxin

1468

hyper-producer strain protected animals from the challenge by phylogenetically diverse strains. Although the debate on the efficacy of mucosal vs. the systemic antigen delivery is still open, the success of passive immunization70,89,90 supports the relevance of the parenteral delivered vaccines in humans. The use of toxoid-based vaccines in humans has been limited for a long time, despite several studies in animal models having demonstrated the importance of toxin immunity in preventing the lethal outcome of CDI. A partially purified toxoid preparation, based on ammonium sulfate fractionation of TcdA and B was the first vaccine candidate tested in a clinical study.91 Groups of 5 healthy adults were vaccinated intramuscularly with toxoid preparations using one of 3 antigen doses in absence or presence of alum as adjuvant. This preliminary clinical trial showed that vaccination was well tolerated and generated a serum antibody response against both toxins, as measured by ELISA and toxin neutralization assay. Moreover, a mucosal antibody response was also present in half of the vaccinated subjects.91,99 The results obtained in this trial encouraged a pilot study aimed at evaluation of this vaccine in 3 patients with recurrent CDI.100 Two of three vaccinated patients developed neutralizing antibodies against TcdA and B and all discontinued antibiotic treatments with resolution of recurrent diarrhea. Recently, the vaccine has been further optimized with a second generation formulation, based on highly purified formalininactivated alum-adsorbed preparations of TcdA and B.101,102 Preclinical testing in the hamster model demonstrated that intramuscular vaccinations with this toxoid-based formulation confer protection to death and disease symptoms in a dose-dependent manner.101 This highly pure toxoid-based vaccine, aimed at the primary prevention in a healthy population, underwent the phase I clinical trial in healthy adults and elderly volunteers showing a good level of safety and tolerability. Immunogenicity evaluation demonstrated that the vaccine induced a complete seroconversion for TcdA which was achieved at all doses in adults and at the highest vaccine dose in elderly. The TcdB seroconversion was lower, with subjects both in adults and elderly groups reaching 75% of a seroconversion. The antibody response appeared persistent only for TcdA in adult groups whereas the TcdB response declined 6 mo after vaccination.102,103 A phase II trial has recently concluded, which evaluated the immunogenicity, dosage, and immunization schedule in 2 target populations, namely adults at risk for CDI (NCT01230957, clinicaltrials.gov) to assess the primary prevention and infected patients (NCT00772343, clinicaltrials.gov) to estimate the prevention of recurrences.103 To overcome the safety issues associated with the large-scale production of toxoids, such as exposure to toxins and spores, Donald and colleagues have recently proposed a novel recombinant toxoid-based candidate vaccine consisting of genetically modified TcdA and B produced in a non-sporulating strain of C. difficile lacking the genes for the native toxins.104 Although site-directed mutations abrogate cytotoxicity linked to the glucosyl-transferase activity of the toxins, a residual toxicity was observed which has been prevented by formalin treatment. This genetically and chemically detoxified recombinant vaccine induced functional antibodies in the hamster model and

Human Vaccines & Immunotherapeutics

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

early toxin production is sufficient to initiate limited damage to the vasculature, which allows permeation of the immune serum containing neutralizing antibodies. Historically, formalin-inactivated TcdA and TcdB were the first antigen mixture proposed for vaccine use.91 Well-established experience with toxoid-based vaccines against important pathogens such as Clostridium tetani92 and Corynebacterium diphtheriae 93 provided the rationale to extend the same approach to C. difficile, in an attempt to direct the immune response toward the inclusive epitope repertoire of TcdA and TcdB. Subsequent studies provided important information about the nature and localization of toxin-neutralizing epitopes and led to investigations on the use of recombinant engineered toxin fragments as vaccine candidates.

Antigens

Animal model

Route of immunization

Adjuvant

Ref.

Toxoid A and B from culture filtrates Toxoid A and B from culture filtrates

Hamster

subcutaneous

Freund

94, 95

Hamster

Parenteral (i.p., s.c.) + mucosal (i.n., i.g., r.)

CT/RIBI

96

Partially purified toxoid A and B (44%)

Hamster

Parenteral (i.m.) + mucosal (i.n., i.g., r.)

None/Al2O3/LT

90

Toxoid A and B from culture filtrates

Mice

t.c., s.c.

CT

97

Purified toxoid B

Hamster

i.p.

MlipidA/RIBI

98

Highly purified toxoid A and B (>90%)

Hamster

i.m.

Al(OH)3/proprietary

101

Genetically modified toxoid A and B

Hamster

i.m.

AlPO4

104

Downloaded by [Lakehead University] at 23:59 18 March 2015

i.p., intraperitoneal; s.c., subcutaneous; i.n., intranasal; i.g., intragastric; r., rectal; i.m., intramuscular; t.c., transcutaneous.

conferred a partial protection to lethality.104 A phase I clinical trial of this vaccine is currently ongoing, (NCT01706367, clinicaltrials.gov), aiming at the evaluation of the vaccine dosage in healthy adults aged 50 to 85 y. A summary of toxoid-based vaccines tested either in animal models or in clinical studies, are shown in Tables 1 and 2, respectively.

Vaccination with Recombinant Toxin-Based Peptides Together with the development of toxoid-based vaccines, a number of studies have focused on the use of recombinant peptide antigens. The use of recombinant toxin sub-domains is an attractive strategy for the design of a vaccine against CDI for several reasons. First, it allows definition of the main neutralizing epitopes associated with a toxin domain; as a result, a vaccine targeting only the epitopes enhancing neutralizing antibodies may maximize the protective efficacy. Second, it permits to overcome the complexity of manufacturing toxoid preparations which requires purification of large proteins and chemical detoxification with intrinsic risk of incomplete inactivation and variability among consecutive preparations. Moreover, previous experiences with toxoid-based vaccines such as the acellular pertussis vaccine, highlighted that the formalin-based inactivation may alter structural epitopes with negative consequences on immunogenicity and reduced generation of neutralizing antibodies.105,106 Importantly, classical toxoid preparations involve the use of toxin hyper-producer strain VPI 10463 but the question on whether these toxoids are also protective against heterologous strains is still unanswered. Recently the antigenic variation of TcdB in the epidemic strain was reported,107 highlighting possible concerns on the limited cross-protective efficacy of the current vaccine formulations. Pilot studies using recombinant peptides were initially aimed at the identification of protective epitopes contained along the TcdA and B sequences. Both toxins belongs to the Large Clostridial Toxin family, which present 3 distinct functional domains: an N-terminal enzymatic domain consisting of glucosyl-transferase (GT) and cysteine protease (CP) moieties, a central translocation (T) domain that mediates import into host

www.landesbioscience.com

cells and a C-terminal receptor binding domain (RBD) with 38 tandem repeats.108 The first report underlining the ability of repeating units of RBD of TcdA to induce protective immunity was presented by Lyerly and colleagues who demonstrated that subcutaneous immunization with a recombinant peptide comprising 33 repeating units partially protected hamsters from death and diarrhea.109 Over the last 2 decades this concept has been further dissected with a number of papers describing the potential of RBD domain of TcdA in conferring protection against CDI. As for the toxoid formulations, the debate has been centered on whether mucosal or systemic antibody response is more efficient in promoting protection and promising vehicles for vaccine delivery and route of immunization have been explored to address this point. With the aim to induce anti-TcdA immunity in the intestinal tract, RBD sub-domains have been introduced in live attenuated vector strains used as delivery vehicle for inducing mucosal immunity. A large RBD domain fused to a secretion signal was introduced in an attenuated Vibrio cholerae strain; oral inoculation of rabbits with this construct evoked a systemic and mucosal immunity against TcdA and prevented fluid secretions and histological changes in an ileal loop challenge assay.110 With a similar approach, a recombinant fusion protein comprising 14 repeat units of TcdA and the immunogenic fragment C of tetanus toxin, was introduced in an attenuated Salmonella typhimurium strain. Intragastric and intranasal administration of this strain generated a significant anti-TcdA IgG serum response as well as an IgA response in intestinal and pulmonary mucosa.111 The same domain in the purified form was tested after the direct administration to the mice nasal mucosa. This recombinant domain either as histidine tagged protein or fusion protein with the fragment C of tetanus toxin, generated anti-TcdA serum antibodies and, when combined to the mucosal adjuvants LT and LTR72, also a strong mucosal response in the pulmonary and nasal lavages but not at the intestinal surface.112 More recently, Bacillus subtilis spores have been demonstrated to be an alternative oral vector able to deliver toxin fragments to the intestinal mucosa.113 RBD subdomains of both toxins expressed on the outer layer of spore coat induced neutralizing serum IgG and fecal IgA in mice and partially protected hamsters from fatal outcome; strikingly, the authors also presented evidences that antibodies raised against TcdA domain are cross-reactive to TcdB, which is in discrepancy with evidences reported by other groups.114-116

Human Vaccines & Immunotherapeutics 1469

©2014 Landes Bioscience. Do not distribute.

Table 1. Summary of toxoid-based vaccines described in the literature

Antigens

Clinical trial

Vaccination regimen/ target population

Adjuvant

References/ study (clinicaltrial.gov)

Partially purified toxoid A and B (ACAM-CDIFFTM by Acambis)

Phase I

4 doses i.m./healthy adults

None/ Al(OH)3

91, 99

Pilot study

4 doses i.m/patients

None

100

Highly purified toxoid A and B (>90%) (Acambis/Sanofi Pasteur*)

Phase I Phase II

3 doses i.m/ healthy adults, elderly Adults at risk/patients

Al(OH)3

102, 103 NCT01230957/ NCT00772343

Genetically modified full-length TcdA and B (Pfizer)

Phase I

3 doses i.m/ healthy adults, elderly

None/ Al(OH)3

NCT01706367

IC84 recombinant fusion protein (Valneva)

Phase I

3 doses i.m/ healthy adults, elderly

None/ Al(OH)3

NCT01296386

Downloaded by [Lakehead University] at 23:59 18 March 2015

*Acambis has been acquired by Sanofi Pasteur in 2008.

Interestingly, since this modular organization in repeating units presents structural homology with the most powerful mucosal adjuvants such as cholera toxin (CT) and E. coli LT, Castagliuolo and colleagues studied the adjuvant activity of the RBD of TcdA117 (US Patent 5919463). The authors found that a peptide corresponding to 14 repeating units had an adjuvant effect on poorly immunogenic peptides, stimulating systemic response as well as mucosal IgA response after oral and nasal administration.117 More specifically, the profile of IgG subclasses and cytokine release suggests the induction of a mixed Th1/Th2 response with a predominance of the Th1 component. DNA vaccine technology, known to provide humoral and cell-mediated immunity, has been also evaluated as proof of concept for a safe and easily-manufactured vaccine against CDI. A DNA vaccine encoding 92% of RBD of TcdA provided high antibodies titers and protected mice from death after parenteral inoculation of TcdA.118 Similarly, an adenovirus-based vaccination directed against a region spanning 85% of RBD of TcdA was demonstrated to generate a robust humoral and T cellular immune response and to provide protection in the mouse model after challenge with lethal doses of TcdA.119 Although all these studies have been informative in the search for the key attributes for an efficient anti CDI vaccination based on recombinant peptides, an optimal vaccine strategy may also need to be redirected for the inclusion of TcdB fragments. Recently, the search of an efficacious vaccine targeting both toxins prompted novel studies on the use of combined recombinant peptides. Tian and colleagues proposed a fusion protein containing 19 of the 38 repetitive units of TcdA and 23 of the 24 repetitive units of TcdB.120 In mice vaccinated intramuscularly, this fusion protein generated an IgG response against both toxins, with the TcdB portion appearing generally less immunogenic than TcdA. Functional antibodies were analyzed both by in vitro cytotoxicity assay and mouse toxin challenge model, demonstrating that a full protection against TcdA was achieved at all tested doses and in absence of adjuvant, whereas the presence of alum hydroxide was necessary to elicit protection against TcdB. The vaccination of hamsters with the adjuvanted fusion protein protected animals from lethality and reduced the severity of the disease. Finally, when co-administered with alum hydroxide this vaccine was also efficacious in a non-human primate

1470

model generating functional antibodies against both toxins. This recombinant fusion protein, named IC84, has been tested in a phase I clinical trial in healthy adults and elderly subjects (Table 2). The IC84 vaccine, administered at different dosages in absence of adjuvants, showed in both study populations favorable safety and tolerability, and induced antibodies against TcdA and B (NCT01296386, clinicaltrials.gov). With a more traditional approach, the use of RBDs of both toxins were also tested in combination with the immune-adjuvant flagellin of Salmonella typhimurium, with the rationale to stimulate the toll-like receptor 5,121 known to protect mice against C. difficile colitis. Although the adjuvant activity of flagellin contributed to an enhanced systemic and intestinal IgA response against TcdA, the vaccination was not equally efficient against TcdB; moreover, flagellin did not confer an advantage in the animal model since the protective effect of the toxin domains promoted mice survival from lethal challenge even in absence of adjuvants. Recently, flagellin of C. difficile has also been demonstrated to stimulate toll-like receptor 5.122 In contrast to the previous assumption that only binding domain regions induce protective antibodies, recent data115,116 demonstrated that alternative neutralizing epitopes within TcdB are promising vaccine candidates. The complete screening of each subdomain of TcdA and B were recently analyzed for the ability to induce neutralizing antibodies against both toxins. This systematic analysis further confirmed that RBDs of both toxins are protective in an in vitro neutralization assay, but while a minimal protective sub-region of TcdA was identified, the entire RBD of TcdB was necessary to neutralize the TcdB toxicity. Importantly, this screening investigated the presence of protective epitopes within the enzymatic domains, establishing that whereas CP is not immunogenic, the GT domain of TcdB elicited functional antibodies.115 These evidences were confirmed in the hamster model where the co-immunization with the RBD of TcdA and the GT domain of TcdB was demonstrated to be the minimal combination necessary and sufficient to confer protection to lethality and recovery from diarrhea and tissue damage. As reported in previous studies, parenteral immunization resulted in the presence of anti-toxin IgG also in the intestinal tract, likely as consequence of toxin-induced alteration of gut mucosa which allows the permeation of circulating neutralizing antibodies.115

Human Vaccines & Immunotherapeutics

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

Table 2. Summary of vaccines tested in clinical studies

Table 3. Summary of vaccines based on recombinant toxin peptides described in the literature

Antigens

Animal model

Route/vehicles

Adjuvant

Ref.

RBD TcdA- 33 out 38 repeating units

Hamster

subcutaneous

Freund

109

RBD TcdA- 720 aa C-terminal peptide

Rabbit

Oral/V. cholerae vector

None/Cholera toxin

110

RBD TcdA- peptide 2387-2708

Mice

i.n., i.g./ S. typhimurium vector

None

111

RBD TcdA- peptide 2387-2708

Mice

i.n.

None/LT/LTR72

112

RBD TcdA/B peptides 388-2706 and 2137–2366)

Mice, hamster

o./B. subtilis spores

None

113

RBD TcdA- peptide 1839-2710

Mice

DNA vaccine, i.m.

/

118

RBD TcdA- peptide 1870-2680

Mice

adenovirus vaccine, i.m.

/

119

Fusion protein RBD TcdA+Ba

Mice, hamster, monkey

i.m.

None/Al(OH)3

120

RBD TcdA+B- full lenght

Mice

i.n.

FliCb /None/ Al(OH)3/LT192

121

ToxinA/B chimeric protein

Mice, hamster

i.m., i.p.

RBD, CP, GT TcdA+B

Mice, rabbit

DNA vaccine, i.m.

/

125

RBD, CP, GT TcdA+B

Mice, hamster

i.p.

Al(OH)3/MF59

115

Downloaded by [Lakehead University] at 23:59 18 March 2015

c

116

i.p., intraperitoneal; o., oral; i.n., intranasal; i.g., intragastric; i.m., intramuscular. aFusion protein contains 19 of the 38 repetitive units of toxin A and 23 of the 24 repetitive units of toxin B. bFlagellin from Salmonella typhimurium tested as adjuvant both in fusion with toxin domains or as recombinant protein. c Genetically detoxified chimeric protein comprising the full-length toxin B in which the original RBD domain was replaced by the corresponding portion of toxin A.

The potential of the GT domain of TcdB to induce neutralizing antibodies has been also evidenced by an alternative approach based on the use of a genetically detoxified chimeric protein comprising the full-length TcdB with the original RBD domain replaced by the corresponding portion of TcdA.116 This chimeric protein comprises the major protective epitopes of TcdA and B, residing in the RBD and GT domains, respectively, and induced neutralizing antibodies against both toxins. Mice and hamsters vaccinated by parenteral route develop long-term immunity against both toxins and are protected from primary and recurrent CDI, although non-stringent sub-lethal challenge conditions were applied. Notably, this chimeric vaccine, generated with toxin sequences of VPI10463 strain, protects experimental animals from challenge by BI/NAP1/027. This last point evidenced the potential of the GT domain to confer broad protection across diverse strains. Indeed, several studies indicated that the RBD region of TcdB appears variable between different toxinotypes123,124 and recent characterization of TcdB in the epidemic strain 027/BI/NAP1 demonstrated that the RBD region is antigenically variable107 from other strains. The prevalence of immunogenic epitopes in the binding domains of TcdA and enzymatic domain of TcdB has been reported also by Jin and colleagues in a DNA-based vaccination study.125 Moreover, the importance of the GT domain of TcdB in inducing protective antibodies was also revealed by epitope mapping studies on humanized monoclonal antibodies.90 Overall, these evidences suggest a potential advantage in the use of a more conserved region such as the GT domain as vaccine candidate. The mechanism by which anti-GT antibodies mediate protection remains to be determined. However, the extracellular cytotoxic activity of TcdB has been recently described,

www.landesbioscience.com

opening new perspectives in the understanding of its mechanism of action.126,127 Table 3 shows a comprehensive summary of the vaccines based on the use of recombinant peptides.

Surface-Associated Antigens as Vaccine Candidates Antibodies to surface protein antigens have also been associated with reduced CDI recurrence, although the correlation was not as statistically significant as in the case of an anti-toxin response.128-130 These evidences suggest the possibility of adding other vaccine targets such as surface associated proteins and polysaccharides to toxin combinations, in an attempt to reduce gastrointestinal colonization and transmission. Surface proteins Toxin-based vaccinations, although effective in preventing the disease manifestations, are likely unable to prevent C. difficile colonization. A vaccine targeting surface-associated antigens could confer a potential advantage in the control of the disease offering preventive measures against carriage and transmission. Indeed, prevention or reduction of the colonization could limit the transmission in the healthcare facilities where the population at highest risk for CDI resides, and thereby reverse the recently emerging increase in community-acquired infections.75 Moreover, several studies indicate that colonization with non-toxigenic strains is associated with a decreased disease incidence,131 suggesting that a vaccination with non-toxin components could be of added value in the prevention of the disease manifestations.

Human Vaccines & Immunotherapeutics 1471

©2014 Landes Bioscience. Do not distribute.

Recombinant toxin-based peptides

Table 4. Summary of surface-associated antigens described in the literature Surface proteins Antigens

Animal model

Route/vehicles

Adjuvant

Ref.

Crude SLP

Mice, hamster

i.p., i.n.

Al(OH)3/ CT /RIBI/chitosan glutamate/TMC

133

SLP, FliD, Cwp84

Mice

i.n., r., i.g./ PLGA encapsulation

Freund/CT

134

Cwp84

Hamster

s.c., r., i.g

None/ Freund/CT

135

Cwp84

Hamster

i.g./ pectin beads encapsulation

None

136

Glycans

Animal model

Conjugation

Adjuvant

Ref.

PSII

Mice

CRM197

MF59

139

PSII

Rabbit

LTB E. coli

None

143

PSIII

Mice, rabbit

ExoA P. aeruginosa/HSA

Freund

148

Downloaded by [Lakehead University] at 23:59 18 March 2015

i.p., intraperitoneal; s.c., subcutaneous; i.n., intranasal; i.g., intragastric; r., rectal.

After the spore germination, the adherence of C. difficile to the intestinal mucosa is the first step for the settlement of the pathogen in the gut.132 The interaction between bacteria and epithelial cells is a multi-factorial process and requires the involvement of surface-displayed adhesins and virulence factors. A number of evidences suggest that during the infection surfaceexposed antigens are able to induce an immune response. Studies on patient sera revealed the presence of antibodies directed to the flagellar components FliC and FliD, the Cwp66 adhesin, the fibronectin binding protein Fbp68, and the cysteine protease Cwp84.128,129 Moreover, a proteomic-based approach on cell wall proteins surrounding the bacterium, showed that S-layer proteins are immunoreactive with patient sera.130 These evidences confirm the expression of these surface proteins during the course of the disease and suggest that they are possible good candidates for an active immunization aiming at the prevention of bacterial colonization (summary in Table 4). The S-layer protein has been tested as a surface-antigen vaccine in combination with a series of systemic and mucosal adjuvants. The antibody response was variable depending on the vaccination regimen and adjuvant adopted, but overall none of the vaccinations conferred a significant advantage in the survival of hamsters to C. difficile challenge.133 The vaccination with colonization factors is expected to reduce the level of colonization rather than to protect against lethal outcome. On the basis of this consideration Pechiné and colleagues tested several antigen combinations in a human flora-associated mouse model.134 After pilot experiments evaluating the route of choice for an efficient systemic and mucosal immune response, mice were vaccinated by rectal route with combinations of FliD, flagellar preparation, Cwp84, and cell wall extract, showing a significant lowering of the level of colonization compared with control group. More recently, the cysteine protease Cwp84 was also evaluated as vaccine candidate both administered by rectal route135 and encapsulated in pectin beads in an oral vaccine,136 conferring a partial protection from lethality in the hamster model (Table 4). Surface carbohydrates Polysaccharides coating the surface of bacterial pathogens represent an optimal target for eliciting carbohydrate specific

1472

antibodies. Glycans are T cell independent antigens, but they can be turned into molecules able to evoke a T cell memory response following conjugation to a carrier protein. This strategy has found application in the prevention of many deadly infectious diseases.137 Consequently, great attention has been directed in the recent years to the structural analysis of polysaccharides on the surface of C. difficile with the result of identifying 3 glycan structures, named PSI, PSII, and PSIII.138 Among these 3 carbohydrates, PSII was found to be the more abundantly expressed by most of C. difficile ribotypes, including the hypervirulent strain NAP1/027 and other clinical isolates belonging to ribotypes 001, 018, 027, 078, and 126.139,140 Following the discovery of PSII, it was not clear whether PSII was part of a capsule or a surface glycoprotein, or released to the external surface of the bacterium. Antibodies against the conjugated PSII detected the polysaccharide at the surface of the bacterial vegetative cells, thus confirming this molecule as a target for a carbohydrate based vaccine.139 However the sugar coating was not as thick and uniformly distributed as expected for a capsule. Therefore, it can be hypothesized that PSII is expressed by the bacterium either as cell wall-linked polysaccharide not bound to peptidoglycan or as a conjugate with lipoteichoic acids.139 Intriguingly, strain 630 and the hypervirulent strain R20291 can form structured biofilms in vitro and antibodies against the synthetic phosphorylated hexaglycosyl unit detect the presence of PSII in the biofilm matrix.141 As biofilms protect bacteria from multiple stresses, including immune responses, this finding sustains the interest for PSII as component of a glycoconjugate vaccine. Notably, glycoarray analysis showed that specific IgA antibodies in the stools of hospital patients infected with C. difficile can recognize the synthetic PSII hexasaccharide hapten, suggesting that under exposure to PSII the human immune system may mount antibodies against several structural epitopes of PSII.142 The PSII polysaccharide conjugated to diphtheria toxoid CRM197 was tested in Balb/C mice formulated with adjuvant MF59, and elicited high levels of IgG.139 Importantly, in the same study it was evidenced that one single repeating unit phosphorylated at the non-terminal sugar was able to reach IgG levels comparable to the polymer, and the charged phosphate group is critical

Human Vaccines & Immunotherapeutics

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

Surface glycans

Conclusions and Perspectives The prevalence of CDI among elderly and immunocompromised individuals, together with the marked recurrence of the infection, poses substantial challenges to future C. difficile vaccines. Pivotal studies in humans100 indicated that intra-muscular 4 doses immunization with the toxoid mixture after cessation of vancomycin can lead to resolution of recurrent C. difficile associated diarrhea, suggesting that vaccination could be highly recommendable to prevent recurrences in subjects that treated a first episode with antibiotics. However, the advent of fidaxomicin represents an important advance in treatment and prevention of recurrent CDI, enforcing the rationale to focus the vaccine therapy against primary CDI in adults. Three experimental vaccines against C. difficile are

www.landesbioscience.com

currently under clinical evaluation, all of them having as objective the primary prevention of CDI in adults and elderly (Table 2). Both toxoid based and recombinant vaccines have proven to be highly immunogenic in healthy adults, including subjects with age ≥65 y (Ref. 102, NCT01706367 and NCT01296386 studies in clinicaltrials.gov). This suggests the possibility to prevent by vaccination the insurgence of CDI in a high-risk population which includes the elderly, adults with planned hospitalization, LTCF residents and patients with co-morbidity requiring prolonged use of antibiotics. Challenges related to such a vaccination strategy will reside mostly in the ability of inducing in elderly and immunocompromised individuals a rapid, long lasting, and protective immunity. For these reasons, further efforts can still be pursued to optimize vaccine efficacy, including regular booster immunizations in adulthood, development of innovative adjuvants149 and design of accelerated schedules compatible with planned hospitalization and surgery. Despite recent surveys which indicated that effective measurements for infection control resulted in beneficial effects on reducing the CDI transmission in nosocomial environments,150,151 healthcare facilities continue to represent a reservoir for C. difficile re-infection and transmission. This emphasizes the importance of developing multi-component vaccines able to neutralize the effects of both toxins and to reduce the bacterial persistence within the host. Colonization factors such as adhesins, flagellar proteins, S-layer components, and the binary toxin CDT represent excellent auxiliary candidates for next generation vaccines, although the sequence variability among different isolates raises the question of their ability to confer broad cross-protection. Besides the high risk patients in the healthcare facilities, cases of community acquired CDI are increasing.152 To explain the phenomenon, new potential sources of transmission have been considered. Although direct C. difficile transmission from animals to humans has not been definitely proven, direct or indirect contact with colonized animals and foodborne transmission have been indicated as potentials sources of community acquired CDI.153 For this reason, domestic pets and production animals represent a potential novel target population for preventive vaccines. Overall, changing epidemiology poses new challenges to CDI treatment. The promise of new vaccines able to eliminate the distressing diarrheal symptoms paves the way to novel treatment opportunities, as well as to have a potential beneficial impact in reducing environmental contamination. Future optimizations of toxin-based vaccines should include development and use of novel adjuvants able to enhance the immune response in elderly. Moreover, the inclusion of additional structural bacterial antigens could help to limit the C. difficile survival into the host, further reducing the potential sources of infection and relapses in hospital environments. Disclosure of Potential Conflicts of Interest

All the authors are employees of Novartis Vaccines S.r.L. Acknowledgments

We are grateful to Matthew J Bottomley for critical review of the manuscript.

Human Vaccines & Immunotherapeutics 1473

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

to induce IgG antibodies able to recognize the entire native polysaccharide.139 Conjugation of PSII to the enterotoxin B subunit (LTB) of enterotoxigenic Escherichia coli (ETEC)143 also achieved the induction of an immunogenic response against native PSII in rabbits. Glycan structures other than PSII have been also detected on the surface of vegetative C. difficile cells. PSI has been detected in ribotype 027, but not in strains MOH900, and MOH718.144 Subsequent NMR analysis or purification of carbohydrates from a larger collection of clinical isolates139 has not led to identification of this polysaccharide, so PSI appears to be much less conserved than PSII. A glycoarray study to assess the presence of antibodies against PSI in CDI patients revealed that IgA levels to both PSI and PSII were higher in patients with less severe disease compared with asymptomatic controls, indicating that higher antibody levels to these antigens correlate with milder forms of CDI.145 Additionally, sera from healthy horses have been demonstrated to contain natural anti-PSI IgG antibodies detecting both the synthetic nonphosphorylated repeating unit and the native polysaccharide, with a slightly higher recognition of the native PSI polysaccharide. This result suggests that the glycosyl phosphate and the polymeric nature of PSI could be immunologically important to develop a vaccine.146 Antibodies against the third isolated glycan structure, named PSIII, have been detected in the blood of infected patients.147 Intraperitoneal or subcutaneous immunizations of Balc/C mice or rabbits, respectively, with either intact or de-O-acylated LTA fraction of PSIII conjugated to the genetically inactivated P. aeruginosa exoTcdA protein (ExoA) or HSA revealed that it was possible to elicit IgG antibodies recognizing PSIII on C. difficile cells148 rendering this molecule a potential target for vaccine development. It is worthy of note that to date no evidence of protective activity of specific antibodies against C. difficile carbohydrates has been reported. However, glycans such as PSII, which are well exposed on the bacterial surface and sufficiently conserved among the different strains, undoubtedly represent an attractive target for a preventive vaccine. It is reasonable to assume that these carbohydrates, either alone or in combination to surface layer antigens or proteins from flagella, could find application in the control of bacterial gut colonization.

1. Keessen EC, Gaastra W, Lipman LJ. Clostridium difficile infection in humans and animals, differences and similarities. Vet Microbiol 2011; 153:20517; PMID:21530110; http://dx.doi.org/10.1016/j. vetmic.2011.03.020 2. al Saif N, Brazier JS. The distribution of Clostridium difficile in the environment of South Wales. J Med Microbiol 1996; 45:133-7; PMID:8683549; http:// dx.doi.org/10.1099/00222615-45-2-133 3. Zidaric V, Beigot S, Lapajne S, Rupnik M. The occurrence and high diversity of Clostridium difficile genotypes in rivers. Anaerobe 2010; 16:3715; PMID:20541023; http://dx.doi.org/10.1016/j. anaerobe.2010.06.001 4. Hall IC, O’Toole E. Intestinal flora in new-born infants: with a description of anew pathogenic anaerobe, Bacillus difficilis. Am J Dis Child 1935; 49:390-402; http://dx.doi.org/10.1001/ archpedi.1935.01970020105010 5. Bartlett JGMN, Moon N, Chang TW, Taylor N, Onderdonk AB. Role of Clostridium difficile in antibiotic-associated pseudomembranous colitis. Gastroenterology 1978; 75:778-82; PMID:700321 6. Gerding DNMC, Muto CA, Owens RC Jr. Measures to control and prevent Clostridium difficile infection. Clin Infect Dis 2008; 46(Suppl 1):S43-9; PMID:18177221; http://dx.doi.org/10.1086/521861 7. Vonberg RPKE, Kuijper EJ, Wilcox MH, Barbut F, Tüll P, Gastmeier P, van den Broek PJ, Colville A, Coignard B, Daha T, et al.; European C difficile-Infection Control Group; European Centre for Disease Prevention and Control (ECDC). Infection control measures to limit the spread of Clostridium difficile. Clin Microbiol Infect 2008; 14(Suppl 5):2-20; PMID:18412710; http://dx.doi. org/10.1111/j.1469-0691.2008.01992.x 8. Sorg JASA, Sonenshein AL. Bile salts and glycine as cogerminants for Clostridium difficile spores. J Bacteriol 2008; 190:2505-12; PMID:18245298; http://dx.doi.org/10.1128/JB.01765-07 9. Francis MB, Allen CA, Shrestha R, Sorg JA. Bile acid recognition by the Clostridium difficile germinant receptor, CspC, is important for establishing infection. PLoS Pathog 2013; 9:e1003356; PMID:23675301 http://dx.doi.org/10.1371/journal.ppat.1003356 10. Wheeldon LJWT, Worthington T, Lambert PA. Histidine acts as a co-germinant with glycine and taurocholate for Clostridium difficile spores. J Appl Microbiol 2011; (Forthcoming); PMID:21261795; http://dx.doi.org/10.1111/j.1365-2672.2011.04953.x 11. Paredes-Sabja D, Sarker MR. Germination response of spores of the pathogenic bacterium Clostridium perfringens and Clostridium difficile to cultured human epithelial cells. Anaerobe 2011; 17:78-84; PMID:21315167; http://dx.doi.org/10.1016/j. anaerobe.2011.02.001 12. Sorg JASA, Sonenshein AL. Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. J Bacteriol 2010; 192:4983-90; PMID:20675492; http://dx.doi. org/10.1128/JB.00610-10 13. Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006; 47:241-59; PMID:16299351; http://dx.doi. org/10.1194/jlr.R500013-JLR200 14. Scaria J, Janvilisri T, Fubini S, Gleed RD, McDonough SP, Chang YF. Clostridium difficile transcriptome analysis using pig ligated loop model reveals modulation of pathways not modulated in vitro. J Infect Dis 2011; 203:1613-20; PMID:21592991 http://dx.doi. org/10.1093/infdis/jir112 15. Janvilisri T, Scaria J, Chang YF. Transcriptional profiling of Clostridium difficile and Caco-2 cells during infection. J Infect Dis 2010; 202:282-90; PMID:20521945; http://dx.doi.org/10.1086/653484

;

;

1474

16. de la Riva L, Willing SE, Tate EW, Fairweather NF. Roles of cysteine proteases Cwp84 and Cwp13 in biogenesis of the cell wall of Clostridium difficile. J Bacteriol 2011; 193:3276-85; PMID:21531808; http://dx.doi.org/10.1128/JB.00248-11 17. Kirby JM, Ahern H, Roberts AK, Kumar V, Freeman Z, Acharya KR, Shone CC. Cwp84, a surface-associated cysteine protease, plays a role in the maturation of the surface layer of Clostridium difficile. J Biol Chem 2009; 284:34666-73; PMID:19808679; http://dx.doi.org/10.1074/jbc.M109.051177 18. Janoir C, Péchiné S, Grosdidier C, Collignon A. Cwp84, a surface-associated protein of Clostridium difficile, is a cysteine protease with degrading activity on extracellular matrix proteins. J Bacteriol 2007; 189:7174-80; PMID:17693508; http://dx.doi. org/10.1128/JB.00578-07 19. Voth DE, Ballard JD. Clostridium difficile toxins: mechanism of action and role in disease. Clin Microbiol Rev 2005; 18:247-63; PMID:15831824; http://dx.doi.org/10.1128/CMR.18.2.247-263.2005 20. Nusrat A, von Eichel-Streiber C, Turner JR, Verkade P, Madara JL, Parkos CA. Clostridium difficile toxins disrupt epithelial barrier function by altering membrane microdomain localization of tight junction proteins. Infect Immun 2001; 69:132936; PMID:11179295; http://dx.doi.org/10.1128/ IAI.69.3.1329-1336.2001 21. Genth H, Dreger SC, Huelsenbeck J, Just I. Clostridium difficile toxins: more than mere inhibitors of Rho proteins. Int J Biochem Cell Biol 2008; 40:592-7; PMID:18289919; http://dx.doi. org/10.1016/j.biocel.2007.12.014 22. Shen A. Clostridium difficile toxins: mediators of inflammation. J Innate Immun 2012; 4:149-58; PMID:22237401; http://dx.doi. org/10.1159/000332946 23. Yamakawa K, Karasawa T, Ikoma S, Nakamura S. Enhancement of Clostridium difficile toxin production in biotin-limited conditions. J Med Microbiol 1996; 44:111-4; PMID:8642571; http://dx.doi. org/10.1099/00222615-44-2-111 24. Karlsson S, Burman LG, Akerlund T. Induction of toxins in Clostridium difficile is associated with dramatic changes of its metabolism. Microbiology 2008; 154:3430-6; PMID:18957596; http://dx.doi. org/10.1099/mic.0.2008/019778-0 25. Karlsson S, Dupuy B, Mukherjee K, Norin E, Burman LG, Åkerlund T. Expression of Clostridium difficile toxins A and B and their sigma factor TcdD is controlled by temperature. Infect Immun 2003; 71:178493; PMID:12654792; http://dx.doi.org/10.1128/ IAI.71.4.1784-1793.2003 26. Pultz NJ, Donskey CJ. Effect of antibiotic treatment on growth of and toxin production by Clostridium difficile in the cecal contents of mice. Antimicrob Agents Chemother 2005; 49:352932; PMID:16048976; http://dx.doi.org/10.1128/ AAC.49.8.3529-3532.2005 27. Drummond LJ, Smith DG, Poxton IR. Effects of sub-MIC concentrations of antibiotics on growth of and toxin production by Clostridium difficile. J Med Microbiol 2003; 52:1033-8; PMID:14614060; http://dx.doi.org/10.1099/jmm.0.05387-0 28. Gerber M, Walch C, Löffler B, Tischendorf K, Reischl U, Ackermann G. Effect of sub-MIC concentrations of metronidazole, vancomycin, clindamycin and linezolid on toxin gene transcription and production in Clostridium difficile. J Med Microbiol 2008; 57:776-83; PMID:18480337; http://dx.doi. org/10.1099/jmm.0.47739-0 29. Aldape MJ, Packham AE, Nute DW, Bryant AE, Stevens DL. Effects of ciprofloxacin on the expression and production of exotoxins by Clostridium difficile. J Med Microbiol 2013; 62:741-7; PMID:23429695; http://dx.doi.org/10.1099/jmm.0.056218-0

Human Vaccines & Immunotherapeutics

30. Kyne L, Warny M, Qamar A, Kelly CP. Asymptomatic carriage of Clostridium difficile and serum levels of IgG antibody against toxin A. N Engl J Med 2000; 342:390-7; PMID:10666429; http://dx.doi. org/10.1056/NEJM200002103420604 31. Kyne L, Warny M, Qamar A, Kelly CP. Association between antibody response to toxin A and protection against recurrent Clostridium difficile diarrhoea. Lancet 2001; 357:189-93; PMID:11213096; http:// dx.doi.org/10.1016/S0140-6736(00)03592-3 32. Bartlett JG. Narrative review: the new epidemic of Clostridium difficile-associated enteric disease. Ann Intern Med 2006; 145:758-64; PMID:17116920; http://dx.doi. org/10.7326/0003-4819-145-10-200611210-00008 33. Geric B, Johnson S, Gerding DN, Grabnar M, Rupnik M. Frequency of binary toxin genes among Clostridium difficile strains that do not produce large clostridial toxins. J Clin Microbiol 2003; 41:522732; PMID:14605169; http://dx.doi.org/10.1128/ JCM.41.11.5227-5232.2003 34. Stubbs S, Rupnik M, Gibert M, Brazier J, Duerden B, Popoff M. Production of actin-specific ADPribosyltransferase (binary toxin) by strains of Clostridium difficile. FEMS Microbiol Lett 2000; 186:307-12; PMID:10802189; http://dx.doi. org/10.1111/j.1574-6968.2000.tb09122.x 35. Gonçalves C, Decré D, Barbut F, Burghoffer B, Petit JC. Prevalence and characterization of a binary toxin (actin-specific ADP-ribosyltransferase) from Clostridium difficile. J Clin Microbiol 2004; 42:1933-9; PMID:15131151; http://dx.doi. org/10.1128/JCM.42.5.1933-1939.2004 36. Martin H, Willey B, Low DE, Staempfli HR, McGeer A, Boerlin P, Mulvey M, Weese JS. Characterization of Clostridium difficile strains isolated from patients in Ontario, Canada, from 2004 to 2006. J Clin Microbiol 2008; 46:2999-3004; PMID:18650360; http://dx.doi.org/10.1128/JCM.02437-07 37. Barth H, Aktories K, Popoff MR, Stiles BG. Binary bacterial toxins: biochemistry, biology, and applications of common Clostridium and Bacillus proteins. [table of contents.]. Microbiol Mol Biol Rev 2004; 68:373-402; PMID:15353562; http://dx.doi. org/10.1128/MMBR.68.3.373-402.2004 38. Papatheodorou P, Carette JE, Bell GW, Schwan C, Guttenberg G, Brummelkamp TR, Aktories K. Lipolysis-stimulated lipoprotein receptor (LSR) is the host receptor for the binary toxin Clostridium difficile transferase (CDT). Proc Natl Acad Sci U S A 2011; 108:16422-7; PMID:21930894; http://dx.doi. org/10.1073/pnas.1109772108 39. Aktories K, Schwan C, Papatheodorou P, Lang AE. Bidirectional attack on the actin cytoskeleton. Bacterial protein toxins causing polymerization or depolymerization of actin. Toxicon 2012; 60:57281; PMID:22543189; http://dx.doi.org/10.1016/j. toxicon.2012.04.338 40. Schwan C, Stecher B, Tzivelekidis T, van Ham M, Rohde M, Hardt WD, Wehland J, Aktories K. Clostridium difficile toxin CDT induces formation of microtubule-based protrusions and increases adherence of bacteria. PLoS Pathog 2009; 5:e1000626; PMID:19834554; http://dx.doi.org/10.1371/journal. ppat.1000626 41. Geric B, Rupnik M, Gerding DN, Grabnar M, Johnson S. Distribution of Clostridium difficile variant toxinotypes and strains with binary toxin genes among clinical isolates in an American hospital. J Med Microbiol 2004; 53:887-94; PMID:15314196; http://dx.doi.org/10.1099/jmm.0.45610-0 42. Carroll KC, Bartlett JG. Biology of Clostridium difficile: implications for epidemiology and diagnosis. Annu Rev Microbiol 2011; 65:501-21; PMID:21682645; http://dx.doi.org/10.1146/ annurev-micro-090110-102824

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

References

www.landesbioscience.com

56. Honda H, Dubberke ER. The changing epidemiology of Clostridium difficile infection. Curr Opin Gastroenterol 2014; 30:54-62; PMID:24285002; http://dx.doi.org/10.1097/ MOG.0000000000000018 57. Warny M, Pepin J, Fang A, Killgore G, Thompson A, Brazier J, Frost E, McDonald LC. Toxin production by an emerging strain of Clostridium difficile associated with outbreaks of severe disease in North America and Europe. Lancet 2005; 366:107984; PMID:16182895; http://dx.doi.org/10.1016/ S0140-6736(05)67420-X 58. Akerlund T, Persson I, Unemo M, Norén T, Svenungsson B, Wullt M, Burman LG. Increased sporulation rate of epidemic Clostridium difficile Type 027/NAP1. J Clin Microbiol 2008; 46:15303; PMID:18287318; http://dx.doi.org/10.1128/ JCM.01964-07 59. Gerding DN. Clindamycin, cephalosporins, fluoroquinolones, and Clostridium difficile-associated diarrhea: this is an antimicrobial resistance problem. Clin Infect Dis 2004; 38:646-8; PMID:14986247; http:// dx.doi.org/10.1086/382084 60. Eyre DW, Walker AS, Wyllie D, Dingle KE, Griffiths D, Finney J, O’Connor L, Vaughan A, Crook DW, Wilcox MH, et al.; Infections in Oxfordshire Research Database. Predictors of first recurrence of Clostridium difficile infection: implications for initial management. Clin Infect Dis 2012; 55(Suppl 2):S7787; PMID:22752869; http://dx.doi.org/10.1093/cid/ cis356 61. Bartlett JG, Gerding DN. Clinical recognition and diagnosis of Clostridium difficile infection. Clin Infect Dis 2008; 46(Suppl 1):S12-8; PMID:18177217; http://dx.doi.org/10.1086/521863 62. McFarland LV. Evidence-based review of probiotics for antibiotic-associated diarrhea and Clostridium difficile infections. Anaerobe 2009; 15:274-80; PMID:19825425; http://dx.doi.org/10.1016/j. anaerobe.2009.09.002 63. Centers for Disease Control and Prevention (CDC). Vital signs: preventing Clostridium difficile infections. MMWR Morb Mortal Wkly Rep 2012; 61:157-62; PMID:22398844 64. Johnson S. Recurrent Clostridium difficile infection: a review of risk factors, treatments, and outcomes. J Infect 2009; 58:403-10; PMID:19394704; http:// dx.doi.org/10.1016/j.jinf.2009.03.010 65. Bauer MP, Kuijper EJ, van Dissel JT; European Society of Clinical Microbiology and Infectious Diseases. European Society of Clinical Microbiology and Infectious Diseases (ESCMID): treatment guidance document for Clostridium difficile infection (CDI). Clin Microbiol Infect 2009; 15:1067-79; PMID:19929973; http://dx.doi. org/10.1111/j.1469-0691.2009.03099.x 66. Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC, Pepin J, Wilcox MH; Society for Healthcare Epidemiology of America; Infectious Diseases Society of America. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect Control Hosp Epidemiol 2010; 31:431-55; PMID:20307191; http://dx.doi.org/10.1086/651706 67. Cocanour CS. Best strategies in recurrent or persistent Clostridium difficile infection. Surg Infect (Larchmt) 2011; 12:235-9; PMID:21767157; http:// dx.doi.org/10.1089/sur.2010.080 68. Petrella LA, Sambol SP, Cheknis A, Nagaro K, Kean Y, Sears PS, Babakhani F, Johnson S, Gerding DN. Decreased cure and increased recurrence rates for Clostridium difficile infection caused by the epidemic C. difficile BI strain. Clin Infect Dis 2012; 55:351-7; PMID:22523271; http://dx.doi.org/10.1093/cid/ cis430

69. Louie TJ, Miller MA, Mullane KM, Weiss K, Lentnek A, Golan Y, Gorbach S, Sears P, Shue YK; OPT-80-003 Clinical Study Group. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 2011; 364:422-31; PMID:21288078; http://dx.doi.org/10.1056/NEJMoa0910812 70. Lowy I, Molrine DC, Leav BA, Blair BM, Baxter R, Gerding DN, Nichol G, Thomas WD Jr., Leney M, Sloan S, et al. Treatment with monoclonal antibodies against Clostridium difficile toxins. N Engl J Med 2010; 362:197-205; PMID:20089970; http://dx.doi. org/10.1056/NEJMoa0907635 71. Pépin J, Routhier S, Gagnon S, Brazeau I. Management and outcomes of a first recurrence of Clostridium difficile-associated disease in Quebec, Canada. Clin Infect Dis 2006; 42:758-64; PMID:16477549; http://dx.doi.org/10.1086/501126 72. Kufelnicka AM, Kirn TJ. Effective utilization of evolving methods for the laboratory diagnosis of Clostridium difficile infection. Clin Infect Dis 2011; 52:1451-7; PMID:21628487; http://dx.doi. org/10.1093/cid/cir201 73. McFarland LV, Elmer GW, Surawicz CM. Breaking the cycle: treatment strategies for 163 cases of recurrent Clostridium difficile disease. Am J Gastroenterol 2002; 97:1769-75; PMID:12135033; http://dx.doi. org/10.1111/j.1572-0241.2002.05839.x 74. Centers for Disease Control and Prevention (CDC). Severe Clostridium difficile-associated disease in populations previously at low risk--four states, 2005. MMWR Morb Mortal Wkly Rep 2005; 54:1201-5; PMID:16319813 75. Khanna S, Pardi DS, Aronson SL, Kammer PP, Orenstein R, St Sauver JL, Harmsen WS, Zinsmeister AR. The epidemiology of community-acquired Clostridium difficile infection: a population-based study. Am J Gastroenterol 2012; 107:89-95; PMID:22108454; http://dx.doi. org/10.1038/ajg.2011.398 76. Lessa FC, Gould CV, McDonald LC. Current status of Clostridium difficile infection epidemiology. Clin Infect Dis 2012; 55(Suppl 2):S65-70; PMID:22752867; http://dx.doi.org/10.1093/cid/ cis319 77. Shears P, Prtak L, Duckworth R. Hospitalbased epidemiology: a strategy for ‘dealing with Clostridium difficile’. J Hosp Infect 2010; 74:31925; PMID:19726105; http://dx.doi.org/10.1016/j. jhin.2009.07.009 78. Dubberke ER, Olsen MA. Burden of Clostridium difficile on the healthcare system. Clin Infect Dis 2012; 55(Suppl 2):S88-92; PMID:22752870; http:// dx.doi.org/10.1093/cid/cis335 79. Lyerly DM, Saum KE, MacDonald DK, Wilkins TD. Effects of Clostridium difficile toxins given intragastrically to animals. Infect Immun 1985; 47:349-52; PMID:3917975 80. Lima AA, Lyerly DM, Wilkins TD, Innes DJ, Guerrant RL. Effects of Clostridium difficile toxins A and B in rabbit small and large intestine in vivo and on cultured cells in vitro. Infect Immun 1988; 56:582-8; PMID:3343050 81. Mitchell TJ, Ketley JM, Haslam SC, Stephen J, Burdon DW, Candy DC, Daniel R. Effect of toxin A and B of Clostridium difficile on rabbit ileum and colon. Gut 1986; 27:78-85; PMID:3949240; http:// dx.doi.org/10.1136/gut.27.1.78 82. Lyras D, O’Connor JR, Howarth PM, Sambol SP, Carter GP, Phumoonna T, Poon R, Adams V, Vedantam G, Johnson S, et al. Toxin B is essential for virulence of Clostridium difficile. Nature 2009; 458:1176-9; PMID:19252482; http://dx.doi. org/10.1038/nature07822 83. Leav BA, Blair B, Leney M, Knauber M, Reilly C, Lowy I, Gerding DN, Kelly CP, Katchar K, Baxter R, et al. Serum anti-toxin B antibody correlates with protection from recurrent Clostridium difficile infection (CDI). Vaccine 2010; 28:965-9; PMID:19941990; http://dx.doi.org/10.1016/j.vaccine.2009.10.144

Human Vaccines & Immunotherapeutics 1475

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

43. De Almeida MN, Heffernan H, Dervan A, Bakker S, Freeman JT, Bhally H, Taylor SL, Riley TV, Roberts SA. Severe Clostridium difficile infection in New Zealand associated with an emerging strain, PCR-ribotype 244. N Z Med J 2013; 126:9-14; PMID:24126745 44. Barbut F, Decré D, Lalande V, Burghoffer B, Noussair L, Gigandon A, Espinasse F, Raskine L, Robert J, Mangeol A, et al. Clinical features of Clostridium difficile-associated diarrhoea due to binary toxin (actinspecific ADP-ribosyltransferase)-producing strains. J Med Microbiol 2005; 54:181-5; PMID:15673514; http://dx.doi.org/10.1099/jmm.0.45804-0 45. Bacci S, Mølbak K, Kjeldsen MK, Olsen KE. Binary toxin and death after Clostridium difficile infection. Emerg Infect Dis 2011; 17:976-82; PMID:21749757; http://dx.doi.org/10.3201/eid/1706.101483 46. McDonald LC, Killgore GE, Thompson A, Owens RC Jr., Kazakova SV, Sambol SP, Johnson S, Gerding DN. An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med 2005; 353:243341; PMID:16322603; http://dx.doi.org/10.1056/ NEJMoa051590 47. Stewart DB, Berg A, Hegarty J. Predicting recurrence of C. difficile colitis using bacterial virulence factors: binary toxin is the key. J Gastrointest Surg 2013; 17:118-24, discussion 124-5; PMID:23086451; http://dx.doi.org/10.1007/s11605-012-2056-6 48. Zilberberg MD, Shorr AF, Kollef MH. Increase in adult Clostridium difficile-related hospitalizations and case-fatality rate, United States, 2000-2005. Emerg Infect Dis 2008; 14:929-31; PMID:18507904; http://dx.doi.org/10.3201/eid1406.071447 49. Pépin J, Valiquette L, Alary ME, Villemure P, Pelletier A, Forget K, Pépin K, Chouinard D. Clostridium difficile-associated diarrhea in a region of Quebec from 1991 to 2003: a changing pattern of disease severity. CMAJ 2004; 171:466-72; PMID:15337727; http:// dx.doi.org/10.1503/cmaj.1041104 50. Pépin J, Saheb N, Coulombe MA, Alary ME, Corriveau MP, Authier S, Leblanc M, Rivard G, Bettez M, Primeau V, et al. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis 2005; 41:1254-60; PMID:16206099; http://dx.doi. org/10.1086/496986 51. Loo VG, Poirier L, Miller MA, Oughton M, Libman MD, Michaud S, Bourgault AM, Nguyen T, Frenette C, Kelly M, et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 2005; 353:2442-9; PMID:16322602; http:// dx.doi.org/10.1056/NEJMoa051639 52. Bauer MP, Notermans DW, van Benthem BH, Brazier JS, Wilcox MH, Rupnik M, Monnet DL, van Dissel JT, Kuijper EJ; ECDIS Study Group. Clostridium difficile infection in Europe: a hospital-based survey. Lancet 2011; 377:63-73; PMID:21084111; http:// dx.doi.org/10.1016/S0140-6736(10)61266-4 53. Kuijper EJ, Barbut F, Brazier JS, Kleinkauf N, Eckmanns T, Lambert ML, Drudy D, Fitzpatrick F, Wiuff C, Brown DJ, et al. Update of Clostridium difficile infection due to PCR ribotype 027 in Europe, 2008. Euro Surveill 2008; 13:18942; PMID:18761903 54. Kuijper EJ, Coignard B, Tüll P; ESCMID Study Group for Clostridium difficile; EU Member States; European Centre for Disease Prevention and Control. Emergence of Clostridium difficile-associated disease in North America and Europe. Clin Microbiol Infect 2006; 12(Suppl 6):2-18; PMID:16965399; http:// dx.doi.org/10.1111/j.1469-0691.2006.01580.x 55. Karas JA, Enoch DA, Aliyu SH. A review of mortality due to Clostridium difficile infection. J Infect 2010; 61:1-8; PMID:20361997; http://dx.doi. org/10.1016/j.jinf.2010.03.025

1476

98. Siddiqui F, O’Connor JR, Nagaro K, Cheknis A, Sambol SP, Vedantam G, Gerding DN, Johnson S. Vaccination with parenteral toxoid B protects hamsters against lethal challenge with toxin A-negative, toxin B-positive clostridium difficile but does not prevent colonization. J Infect Dis 2012; 205:128-33; PMID:22124129; http://dx.doi.org/10.1093/infdis/ jir688 99. Aboudola S, Kotloff KL, Kyne L, Warny M, Kelly EC, Sougioultzis S, Giannasca PJ, Monath TP, Kelly CP. Clostridium difficile vaccine and serum immunoglobulin G antibody response to toxin A. Infect Immun 2003; 71:1608-10; PMID:12595488; http://dx.doi. org/10.1128/IAI.71.3.1608-1610.2003 100. Sougioultzis S, Kyne L, Drudy D, Keates S, Maroo S, Pothoulakis C, Giannasca PJ, Lee CK, Warny M, Monath TP, et al. Clostridium difficile toxoid vaccine in recurrent C. difficile-associated diarrhea. Gastroenterology 2005; 128:764-70; PMID:15765411; http://dx.doi.org/10.1053/j.gastro.2004.11.004 101. Anosova NG, Brown AM, Li L, Liu N, Cole LE, Zhang J, Mehta H, Kleanthous H. Systemic antibody responses induced by a two-component Clostridium difficile toxoid vaccine protect against C. difficileassociated disease in hamsters. J Med Microbiol 2013; 62:1394-404; PMID:23518659; http://dx.doi. org/10.1099/jmm.0.056796-0 102. Greenberg RN, Marbury TC, Foglia G, Warny M. Phase I dose finding studies of an adjuvanted Clostridium difficile toxoid vaccine. Vaccine 2012; 30:2245-9; PMID:22306375; http://dx.doi. org/10.1016/j.vaccine.2012.01.065 103. Foglia G, Shah S, Luxemburger C, Pietrobon PJ. Clostridium difficile: development of a novel candidate vaccine. Vaccine 2012; 30:4307-9; PMID:22682287; http://dx.doi.org/10.1016/j.vaccine.2012.01.056 104. Donald RG, Flint M, Kalyan N, Johnson E, Witko SE, Kotash C, Zhao P, Megati S, Yurgelonis I, Lee PK, et al. A novel approach to generate a recombinant toxoid vaccine against Clostridium difficile. Microbiology 2013; 159:1254-66; PMID:23629868; http://dx.doi. org/10.1099/mic.0.066712-0 105. Nencioni L, Volpini G, Peppoloni S, Bugnoli M, De Magistris T, Marsili I, Rappuoli R. Properties of pertussis toxin mutant PT-9K/129G after formaldehyde treatment. Infect Immun 1991; 59:625-30; PMID:1702767 106. Ibsen PH. The effect of formaldehyde, hydrogen peroxide and genetic detoxification of pertussis toxin on epitope recognition by murine monoclonal antibodies. Vaccine 1996; 14:359-68; PMID:8735545; http:// dx.doi.org/10.1016/0264-410X(95)00230-X 107. Lanis JM, Heinlen LD, James JA, Ballard JD. Clostridium difficile 027/BI/NAP1 encodes a hypertoxic and antigenically variable form of TcdB. PLoS Pathog 2013; 9:e1003523; PMID:23935501; http:// dx.doi.org/10.1371/journal.ppat.1003523 108. Jank T, Giesemann T, Aktories K. Rho-glucosylating Clostridium difficile toxins A and B: new insights into structure and function. Glycobiology 2007; 17:15R22R; PMID:17237138; http://dx.doi.org/10.1093/ glycob/cwm004 109. Lyerly DM. Vaccination against lethal Clostridium difficile enterocolitis with a nontoxic recombinant peptide of toxin A. Curr Microbiol 1990; 21:29-32; http:// dx.doi.org/10.1007/BF02090096 110. Ryan ET, Butterton JR, Smith RN, Carroll PA, Crean TI, Calderwood SB. Protective immunity against Clostridium difficile toxin A induced by oral immunization with a live, attenuated Vibrio cholerae vector strain. Infect Immun 1997; 65:2941-9; PMID:9199470 111. Ward SJ, Douce G, Figueiredo D, Dougan G, Wren BW. Immunogenicity of a Salmonella typhimurium aroA aroD vaccine expressing a nontoxic domain of Clostridium difficile toxin A. Infect Immun 1999; 67:2145-52; PMID:10225867

Human Vaccines & Immunotherapeutics

112. Ward SJ, Douce G, Dougan G, Wren BW. Local and systemic neutralizing antibody responses induced by intranasal immunization with the nontoxic binding domain of toxin A from Clostridium difficile. Infect Immun 1999; 67:5124-32; PMID:10496886 113. Permpoonpattana P, Hong HA, Phetcharaburanin J, Huang JM, Cook J, Fairweather NF, Cutting SM. Immunization with Bacillus spores expressing toxin A peptide repeats protects against infection with Clostridium difficile strains producing toxins A and B. Infect Immun 2011; 79:2295-302; PMID:21482682; http://dx.doi.org/10.1128/ IAI.00130-11 114. Libby JM, Wilkins TD. Production of antitoxins to two toxins of Clostridium difficile and immunological comparison of the toxins by cross-neutralization studies. Infect Immun 1982; 35:374-6; PMID:6172384 115. Leuzzi R, Spencer J, Buckley A, Brettoni C, Martinelli M, Tulli L, Marchi S, Luzzi E, Irvine J, Candlish D, et al. Protective efficacy induced by recombinant Clostridium difficile toxin fragments. Infect Immun 2013; 81:2851-60; PMID:23716610; http://dx.doi.org/10.1128/IAI.01341-12 116. Wang H, Sun X, Zhang Y, Li S, Chen K, Shi L, Nie W, Kumar R, Tzipori S, Wang J, et al. A chimeric toxin vaccine protects against primary and recurrent Clostridium difficile infection. Infect Immun 2012; 80:2678-88; PMID:22615245; http://dx.doi. org/10.1128/IAI.00215-12 117. Castagliuolo I, Sardina M, Brun P, DeRos C, Mastrotto C, Lovato L, Palù G. Clostridium difficile toxin A carboxyl-terminus peptide lacking ADP-ribosyltransferase activity acts as a mucosal adjuvant. Infect Immun 2004; 72:2827-36; PMID:15102793; http://dx.doi.org/10.1128/ IAI.72.5.2827-2836.2004 118. Gardiner DF, Rosenberg T, Zaharatos J, Franco D, Ho DD. A DNA vaccine targeting the receptorbinding domain of Clostridium difficile toxin A. Vaccine 2009; 27:3598-604; PMID:19464540; http://dx.doi.org/10.1016/j.vaccine.2009.03.058 119. Seregin SS, Aldhamen YA, Rastall DP, Godbehere S, Amalfitano A. Adenovirus-based vaccination against Clostridium difficile toxin A allows for rapid humoral immunity and complete protection from toxin A lethal challenge in mice. Vaccine 2012; 30:1492-501; PMID:22200503; http://dx.doi. org/10.1016/j.vaccine.2011.12.064 120. Tian JH, Fuhrmann SR, Kluepfel-Stahl S, Carman RJ, Ellingsworth L, Flyer DC. A novel fusion protein containing the receptor binding domains of C. difficile toxin A and toxin B elicits protective immunity against lethal toxin and spore challenge in preclinical efficacy models. Vaccine 2012; 30:424958; PMID:22537987; http://dx.doi.org/10.1016/j. vaccine.2012.04.045 121. Ghose C, Verhagen JM, Chen X, Yu J, Huang Y, Chenesseau O, Kelly CP, Ho DD. Toll-like receptor 5-dependent immunogenicity and protective efficacy of a recombinant fusion protein vaccine containing the nontoxic domains of Clostridium difficile toxins A and B and Salmonella enterica serovar typhimurium flagellin in a mouse model of Clostridium difficile disease. Infect Immun 2013; 81:2190-6; PMID:23545305; http://dx.doi. org/10.1128/IAI.01074-12 122. Yoshino Y, Kitazawa T, Ikeda M, Tatsuno K, Yanagimoto S, Okugawa S, Yotsuyanagi H, Ota Y. Clostridium difficile flagellin stimulates tolllike receptor 5, and toxin B promotes flagellininduced chemokine production via TLR5. Life Sci 2013; 92:211-7; PMID:23261530; http://dx.doi. org/10.1016/j.lfs.2012.11.017

Volume 10 Issue 6

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

84. Steele J, Mukherjee J, Parry N, Tzipori S. Antibody against TcdB, but not TcdA, prevents development of gastrointestinal and systemic Clostridium difficile disease. J Infect Dis 2013; 207:323-30; PMID:23125448; http://dx.doi.org/10.1093/ infdis/jis669 85. Drudy D, Fanning S, Kyne L. Toxin A-negative, toxin B-positive Clostridium difficile. Int J Infect Dis 2007; 11:5-10; PMID:16857405; http://dx.doi. org/10.1016/j.ijid.2006.04.003 86. Kuijper EJ, de Weerdt J, Kato H, Kato N, van Dam AP, van der Vorm ER, Weel J, van Rheenen C, Dankert J. Nosocomial outbreak of Clostridium difficile-associated diarrhoea due to a clindamycin-resistant enterotoxin A-negative strain. Eur J Clin Microbiol Infect Dis 2001; 20:528-34; PMID:11681431; http://dx.doi.org/10.1007/ s100960100550 87. van den Berg RJ, Claas EC, Oyib DH, Klaassen CH, Dijkshoorn L, Brazier JS, Kuijper EJ. Characterization of toxin A-negative, toxin B-positive Clostridium difficile isolates from outbreaks in different countries by amplified fragment length polymorphism and PCR ribotyping. J Clin Microbiol 2004; 42:1035-41; PMID:15004050; http://dx.doi.org/10.1128/ JCM.42.3.1035-1041.2004 88. Kuehne SA, Cartman ST, Heap JT, Kelly ML, Cockayne A, Minton NP. The role of toxin A and toxin B in Clostridium difficile infection. Nature 2010; 467:711-3; PMID:20844489; http://dx.doi. org/10.1038/nature09397 89. Giannasca PJ, Zhang ZX, Lei WD, Boden JA, Giel MA, Monath TP, Thomas WD Jr. Serum antitoxin antibodies mediate systemic and mucosal protection from Clostridium difficile disease in hamsters. Infect Immun 1999; 67:527-38; PMID:9916055 90. Marozsan AJ, Ma D, Nagashima KA, Kennedy BJ, Kang YK, Arrigale RR, Donovan GP, Magargal WW, Maddon PJ, Olson WC. Protection against Clostridium difficile infection with broadly neutralizing antitoxin monoclonal antibodies. J Infect Dis 2012; 206:706-13; PMID:22732923; http://dx.doi. org/10.1093/infdis/jis416 91. Kotloff KL, Wasserman SS, Losonsky GA, Thomas W Jr., Nichols R, Edelman R, Bridwell M, Monath TP. Safety and immunogenicity of increasing doses of a Clostridium difficile toxoid vaccine administered to healthy adults. Infect Immun 2001; 69:98895; PMID:11159994; http://dx.doi.org/10.1128/ IAI.69.2.988-995.2001 92. Tetanus vaccine. Wkly Epidemiol Rec 2006; 81:198-208; PMID:16710950 93. Diphtheria vaccine. Wkly Epidemiol Rec 2006; 81:24-32; PMID:16671240 94. Libby JM, Jortner BS, Wilkins TD. Effects of the two toxins of Clostridium difficile in antibioticassociated cecitis in hamsters. Infect Immun 1982; 36:822-9; PMID:7085078 95. Kim PH, Iaconis JP, Rolfe RD. Immunization of adult hamsters against Clostridium difficileassociated ileocecitis and transfer of protection to infant hamsters. Infect Immun 1987; 55:2984-92; PMID:3679541 96. Torres JF, Lyerly DM, Hill JE, Monath TP. Evaluation of formalin-inactivated Clostridium difficile vaccines administered by parenteral and mucosal routes of immunization in hamsters. Infect Immun 1995; 63:4619-27; PMID:7591115 97. Ghose C, Kalsy A, Sheikh A, Rollenhagen J, John M, Young J, Rollins SM, Qadri F, Calderwood SB, Kelly CP, et al. Transcutaneous immunization with Clostridium difficile toxoid A induces systemic and mucosal immune responses and toxin A-neutralizing antibodies in mice. Infect Immun 2007; 75:282632; PMID:17371854; http://dx.doi.org/10.1128/ IAI.00127-07

www.landesbioscience.com

134. Péchiné S, Janoir C, Boureau H, Gleizes A, Tsapis N, Hoys S, Fattal E, Collignon A. Diminished intestinal colonization by Clostridium difficile and immune response in mice after mucosal immunization with surface proteins of Clostridium difficile. Vaccine 2007; 25:3946-54; PMID:17433506; http://dx.doi. org/10.1016/j.vaccine.2007.02.055 135. Péchiné S, Denève C, Le Monnier A, Hoys S, Janoir C, Collignon A. Immunization of hamsters against Clostridium difficile infection using the Cwp84 protease as an antigen. FEMS Immunol Med Microbiol 2011; 63:73-81; PMID:21707776; http://dx.doi. org/10.1111/j.1574-695X.2011.00832.x 136. Sandolo C, Péchiné S, Le Monnier A, Hoys S, Janoir C, Coviello T, Alhaique F, Collignon A, Fattal E, Tsapis N. Encapsulation of Cwp84 into pectin beads for oral vaccination against Clostridium difficile. Eur J Pharm Biopharm 2011; 79:566-73; PMID:21664462; http://dx.doi.org/10.1016/j. ejpb.2011.05.011 137. Berti F, Adamo R. Recent mechanistic insights on glycoconjugate vaccines and future perspectives. ACS Chem Biol 2013; 8:1653-63; PMID:23841819; http://dx.doi.org/10.1021/cb400423g 138. Monteiro MA, Ma Z, Bertolo L, Jiao Y, Arroyo L, Hodgins D, Mallozzi M, Vedantam G, Sagermann M, Sundsmo J, et al. Carbohydrate-based Clostridium difficile vaccines. Expert Rev Vaccines 2013; 12:42131; PMID:23560922; http://dx.doi.org/10.1586/ erv.13.9 139. Adamo R, Romano MR, Berti F, Leuzzi R, Tontini M, Danieli E, Cappelletti E, Cakici OS, Swennen E, Pinto V, et al. Phosphorylation of the synthetic hexasaccharide repeating unit is essential for the induction of antibodies to Clostridium difficile PSII cell wall polysaccharide. ACS Chem Biol 2012; 7:14208; PMID:22620974; http://dx.doi.org/10.1021/ cb300221f 140. Reid CW, Vinogradov E, Li J, Jarrell HC, Logan SM, Brisson JR. Structural characterization of surface glycans from Clostridium difficile. Carbohydr Res 2012; 354:65-73; PMID:22560631; http://dx.doi. org/10.1016/j.carres.2012.02.002 141. Ðapa T, Leuzzi R, Ng YK, Baban ST, Adamo R, Kuehne SA, Scarselli M, Minton NP, Serruto D, Unnikrishnan M. Multiple factors modulate biofilm formation by the anaerobic pathogen Clostridium difficile. J Bacteriol 2013; 195:545-55; PMID:23175653; http://dx.doi.org/10.1128/JB.01980-12 142. Oberli MA, Hecht ML, Bindschädler P, Adibekian A, Adam T, Seeberger PH. A possible oligosaccharide-conjugate vaccine candidate for Clostridium difficile is antigenic and immunogenic. Chem Biol 2011; 18:580-8; PMID:21609839; http://dx.doi. org/10.1016/j.chembiol.2011.03.009 143. Bertolo L, Boncheff AG, Ma Z, Chen YH, Wakeford T, Friendship RM, Rosseau J, Weese JS, Chu M, Mallozzi M, et al. Clostridium difficile carbohydrates: glucan in spores, PSII common antigen in cells, immunogenicity of PSII in swine and synthesis of a dual C. difficile-ETEC conjugate vaccine. Carbohydr Res 2012; 354:79-86; PMID:22533919; http://dx.doi.org/10.1016/j.carres.2012.03.032

144. Ganeshapillai J, Vinogradov E, Rousseau J, Weese JS, Monteiro MA. Clostridium difficile cell-surface polysaccharides composed of pentaglycosyl and hexaglycosyl phosphate repeating units. Carbohydr Res 2008; 343:703-10; PMID:18237724; http://dx.doi. org/10.1016/j.carres.2008.01.002 145. Martin CE, Broecker F, Oberli MA, Komor J, Mattner J, Anish C, Seeberger PH. Immunological evaluation of a synthetic Clostridium difficile oligosaccharide conjugate vaccine candidate and identification of a minimal epitope. J Am Chem Soc 2013; 135:9713-22; PMID:23795894; http://dx.doi. org/10.1021/ja401410y 146. Jiao Y, Ma Z, Hodgins D, Pequegnat B, Bertolo L, Arroyo L, Monteiro MA. Clostridium difficile PSI polysaccharide: synthesis of pentasaccharide repeating block, conjugation to exotoxin B subunit, and detection of natural anti-PSI IgG antibodies in horse serum. Carbohydr Res 2013; 378:1525; PMID:23597587; http://dx.doi.org/10.1016/j. carres.2013.03.018 147. Martin CE, Broecker F, Eller S, Oberli MA, Anish C, Pereira CL, Seeberger PH. Glycan arrays containing synthetic Clostridium difficile lipoteichoic acid oligomers as tools toward a carbohydrate vaccine. Chem Commun (Camb) 2013; 49:715961; PMID:23836132; http://dx.doi.org/10.1039/ c3cc43545h 148. Cox AD, St Michael F, Aubry A, Cairns CM, Strong PC, Hayes AC, Logan SM. Investigating the candidacy of a lipoteichoic acid-based glycoconjugate as a vaccine to combat Clostridium difficile infection. Glycoconj J 2013; 30:843-55; PMID:23974722; http://dx.doi.org/10.1007/s10719-013-9489-3 149. Weinberger B, Grubeck-Loebenstein B. Vaccines for the elderly. Clin Microbiol Infect 2012; 18(Suppl 5):100-8; PMID:22862783; http://dx.doi. org/10.1111/j.1469-0691.2012.03944.x 150. Duerden BI. Contribution of a government target to controlling Clostridium difficile in the NHS in England. Anaerobe 2011; 17:175-9; PMID:21182972; http://dx.doi.org/10.1016/j.anaerobe.2010.12.004 151. Daneman N, Stukel TA, Ma X, Vermeulen M, Guttmann A. Reduction in Clostridium difficile infection rates after mandatory hospital public reporting: findings from a longitudinal cohort study in Canada. PLoS Med 2012; 9:e1001268; PMID:22815656; http://dx.doi.org/10.1371/journal. pmed.1001268 152. Hensgens MP, Keessen EC, Squire MM, Riley TV, Koene MG, de Boer E, Lipman LJ, Kuijper EJ; European Society of Clinical Microbiology and Infectious Diseases Study Group for Clostridium difficile (ESGCD). Clostridium difficile infection in the community: a zoonotic disease? Clin Microbiol Infect 2012; 18:635-45; PMID:22536816; http://dx.doi. org/10.1111/j.1469-0691.2012.03853.x 153. Otten AM, Reid-Smith RJ, Fazil A, Weese JS. Disease transmission model for community-associated Clostridium difficile infection. Epidemiol Infect 2010; 138:907-14; PMID:20092667; http://dx.doi. org/10.1017/S0950268809991646

Human Vaccines & Immunotherapeutics 1477

©2014 Landes Bioscience. Do not distribute.

Downloaded by [Lakehead University] at 23:59 18 March 2015

123. Chaves-Olarte E, Weidmann M, Eichel-Streiber C, Thelestam M. Toxins A and B from Clostridium difficile differ with respect to enzymatic potencies, cellular substrate specificities, and surface binding to cultured cells. J Clin Invest 1997; 100:173441; PMID:9312171; http://dx.doi.org/10.1172/ JCI119698 124. Stabler RA, Dawson LF, Phua LT, Wren BW. Comparative analysis of BI/NAP1/027 hypervirulent strains reveals novel toxin B-encoding gene (tcdB) sequences. J Med Microbiol 2008; 57:7715; PMID:18480336; http://dx.doi.org/10.1099/ jmm.0.47743-0 125. Jin K, Wang S, Zhang C, Xiao Y, Lu S, Huang Z. Protective antibody responses against Clostridium difficile elicited by a DNA vaccine expressing the enzymatic domain of toxin B. Hum Vaccin Immunother 2013; 9:63-73; PMID:23143772; http://dx.doi.org/10.4161/hv.22434 126. Chumbler NM, Farrow MA, Lapierre LA, Franklin JL, Haslam DB, Goldenring JR, Lacy DB. Clostridium difficile Toxin B causes epithelial cell necrosis through an autoprocessing-independent mechanism. PLoS Pathog 2012; 8:e1003072; PMID:23236283; http://dx.doi.org/10.1371/journal.ppat.1003072 127. Farrow MA, Chumbler NM, Lapierre LA, Franklin JL, Rutherford SA, Goldenring JR, Lacy DB. Clostridium difficile toxin B-induced necrosis is mediated by the host epithelial cell NADPH oxidase complex. Proceedings of the National Academy of Sciences of the United States of America 2013. 128. Péchiné S, Janoir C, Collignon A. Variability of Clostridium difficile surface proteins and specific serum antibody response in patients with Clostridium difficile-associated disease. J Clin Microbiol 2005; 43:5018-25; PMID:16207956; http://dx.doi. org/10.1128/JCM.43.10.5018-5025.2005 129. Péchiné S, Gleizes A, Janoir C, Gorges-Kergot R, Barc MC, Delmée M, Collignon A. Immunological properties of surface proteins of Clostridium difficile. J Med Microbiol 2005; 54:193-6; PMID:15673516; http://dx.doi.org/10.1099/jmm.0.45800-0 130. Wright A, Drudy D, Kyne L, Brown K, Fairweather NF. Immunoreactive cell wall proteins of Clostridium difficile identified by human sera. J Med Microbiol 2008; 57:750-6; PMID:18480333; http://dx.doi. org/10.1099/jmm.0.47532-0 131. Shim JK, Johnson S, Samore MH, Bliss DZ, Gerding DN. Primary symptomless colonisation by Clostridium difficile and decreased risk of subsequent diarrhoea. Lancet 1998; 351:633-6; PMID:9500319; http://dx.doi.org/10.1016/S0140-6736(97)08062-8 132. Denève C, Janoir C, Poilane I, Fantinato C, Collignon A. New trends in Clostridium difficile virulence and pathogenesis. Int J Antimicrob Agents 2009; 33(Suppl 1):S24-8; PMID:19303565; http:// dx.doi.org/10.1016/S0924-8579(09)70012-3 133. Ní Eidhin DB, O’Brien JB, McCabe MS, AthiéMorales V, Kelleher DP. Active immunization of hamsters against Clostridium difficile infection using surface-layer protein. FEMS Immunol Med Microbiol 2008; 52:207-18; PMID:18093141; http://dx.doi. org/10.1111/j.1574-695X.2007.00363.x

Vaccines against Clostridium difficile.

Clostridium difficile infection (CDI) is recognized as a major cause of nosocomial diseases ranging from antibiotic related diarrhea to fulminant coli...
696KB Sizes 2 Downloads 3 Views