Novel Approaches to the Treatment of Systemic Anthrax
Transcription
Novel Approaches to the Treatment of Systemic Anthrax
REVIEW ARTICLE Novel Approaches to the Treatment of Systemic Anthrax Andrew W. Artenstein and Steven M. Opal Center for Biodefense and Emerging Pathogens, Department of Medicine, Memorial Hospital of Rhode Island, Pawtucket, and The Warren Alpert Medical School of Brown University, Providence, Rhode Island Systemic anthrax, defined as invasive Bacillus anthracis infection associated with bacterial dissemination or toxin-mediated multi-organ dysfunction, may be secondary to any of the well-described forms of clinical disease. Once a zoonotic disease of major economic importance, it was controlled in the developed world through widespread livestock vaccination [1]. Although systemic anthrax is uncommon in humans, it is highly lethal; of the 250 patients who developed the disease during the twentieth century, more than 90 percent died [2]. It remains a persistent threat, both as a cause of deliberate and natural outbreaks of disease. Eleven cases, five of them fatal, resulted from a series of bioterrorist attacks in the U.S. in 2001 [3]. More recently, a novel form of systemic anthrax associated with injected heroin caused at least 54 infections in the Received 30 October 2011; accepted 9 December 2011. Correspondence: Andrew W. Artenstein, MD, Physician-in-Chief, Department of Medicine, Director, Center for Biodefense and Emerging Pathogens, Memorial Hospital of Rhode Island, Professor of Medicine and Health Services, Policy and Practice, The Warren Alpert Medical School of Brown University, 111 Brewster St. Pawtucket, RI 02860 (artenstein@brown.edu). Clinical Infectious Diseases 2012;54(8):1148–61 Ó The Author 2012. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com. DOI: 10.1093/cid/cis017 1148 d CID 2012:54 (15 April) d Artenstein & Opal U.K. and Europe with an attendant mortality rate of 33 percent [4–7]. Improvements in mortality may be partially attributed to the empiric deployment of antimicrobials with activity against B. anthracis. Current treatment recommendations involve the use of either a quinolone or doxycycline in combination with at least one or two other agents against which the organism is typically susceptible, such as vancomycin, rifampin, carbapenems, penicillin, ampicillin, or chloramphenicol [3]. Clindamycin has been used in combination therapy due to its theoretical inhibition of anthrax toxin synthesis [8] and its demonstrated clinical activity in toxic shock syndromes caused by other Gram-positive organisms [9]. However, no efficacy data for clindamycin have been reported in anthrax, and in combination with ciprofloxacin, it significantly decreased the survival of gamma-irradiated, spore-challenged mice when compared with either drug alone [10]. Despite the use of appropriate antimicrobials and advances in supportive care, individuals with systemic anthrax remain at high risk of death due to the deleterious effects of two secreted exotoxins and other virulence factors [3, 11]. Thus, new therapeutic strategies that target pathogenic events in the disease process are being investigated. This review describes Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 Anthrax continues to generate concern as an agent of bioterrorism and as a natural cause of sporadic disease outbreaks. Despite the use of appropriate antimicrobial agents and advanced supportive care, the mortality associated with the systemic disease remains high. This is primarily due to the pathogenic exotoxins produced by Bacillus anthracis as well as other virulence factors of the organism. For this reason, new therapeutic strategies that target events in the pathogenesis of anthrax and may potentially augment antimicrobials are being investigated. These include anti-toxin approaches, such as passive immune-based therapies; nonantimicrobial drugs with activity against anthrax toxin components; and agents that inhibit binding, processing, or assembly of toxins. Adjunct therapies that target spore germination or downstream events in anthrax intoxication are also under investigation. In combination, these modalities may enhance the management of systemic anthrax. approaches to the treatment of systemic anthrax that may augment antimicrobials. ANTHRAX PATHOGENESIS Anthrax Toxin Activities SEARCH STRATEGY AND SELECTION CRITERIA Virulence Factors and Toxin Formation In order to determine what novel therapeutic strategies should be used against anthrax, it is important to understand the details of its pathogenesis. B. anthracis owes its lethality to its capacity to generate environmentally resistant spores that persist. It has an exocapsule, which allows the organism to escape immune clearance and disseminate, and it secretes two potent exotoxins – lethal toxin (LT) and edema toxin (ET) [1, 12]. After the initial spore uptake by local tissue macrophages, the organism germinates into its vegetative, replicative form [13–15], and rapid generation times contribute to its capacity to overwhelm innate host defenses [12]. The pathogen uses bicarbonate detection via an ATP-binding cassette transporter to sense a permissive host environment [16]. Such environmental cues induce the synthesis of the transacting, virulence gene regulator atxA, which activates the genes for capsular assembly and toxin synthesis [17]. Similar to other bacterial pathogens, the anthrax bacillus senses its own population density by a quorum-sensing apparatus, allowing the coordination of its virulence factors to its rapidly expanding density [18]. The anthrax genome comprises a single, covalently closed chromosome accompanied by two essential virulence plasmids: pXO1 and pXO2 [19]. The smaller, 95.3 kilobase pair (kbp) pXO2 carries the capBCAE gene operon, responsible for the synthesis of the poly-c-D-glutamic acid exocapsule [19]. The capsule contributes to pathogenesis by several distinct mechanisms (Table 1); loss of capsular function by plasmid deletion is the basis of attenuation for the live, Sterne vaccine strain used in veterinary settings [1]. The 184.5 kbp pXO1 plasmid carries the atxA regulatory gene and three others responsible for toxin synthesis [19]. LF is a zinc metalloproteinase enzyme that inactivates MAPKK [59–61]; LT affects numerous cellular targets, impairing both innate and adaptive immune functions (Table 1) [24–28, 33–35]. EF alters the transcriptional programs of target cells by inducing excess intracellular levels of cAMP through calciumand calmodulin-dependent adenylyl cyclase activity [36]. ET causes tissue edema, resulting in local changes associated with cutaneous anthrax lesions and contributing to the pleural effusions and massive fluid shifts seen in patients with systemic disease [37, 62]. ET, like LT, adversely impacts numerous host functions (Table 1). Hypotension, vasodilation, tachycardia, and reduced myocardial performance are characteristic of experimental intoxication with LT; at least additive hemodynamic derangements are noted with the combination of LT and ET in animal models [30, 31, 37, 42]. Shock also accompanied fatal cases of systemic anthrax in the 2001 U.S. attacks and recent U.K. outbreak [5, 6, 62]. The deleterious hemodynamic effects of anthrax intoxication in experimental models occur largely in the absence of inflammatory mediators such as pro-inflammatory cytokines or chemokines [30–33, 35]. However, a more pronounced cytokine response and rapidly progressive, generalized inflammatory response, similar to that seen in other forms of bacteremia and septic shock, is observed following spore challenge or systemic infection with vegetative forms of B. anthracis [14, 15, 34]. This is likely related to the effects of the capsule and exotoxins in concert with TLR 2/6 ligand stimulation by outer membrane components of the organism [43] (Table 1). NOVEL APPROACHES TO THERAPY New approaches to the therapy of systemic anthrax derive from in vitro and animal studies, the latter serving as surrogates for Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1149 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 References were identified through iterative searches of the PubMed database using the terms ‘‘systemic anthrax,’’ ‘‘anthrax lethal toxin,’’ ‘‘anthrax edema toxin,’’ and ‘‘inhibitors’’ of the aforementioned terms. As anthrax toxins were initially identified in the 1950s, most references were retrievable from PubMed. More than 97 percent of the 2989 articles identified were published in English. Articles and references cited therein were selected by their relevance to pathogenesis or potential treatment strategies other than antimicrobials; vaccines or other preventive measures were not considered relevant. Anthrax exotoxins are binary, A/B-type bacterial toxins comprising a nontoxic enzyme (A) moiety—lethal factor (LF) or edema factor (EF), each associated with a common binding (B) component—protective antigen (PA), named for its role in engendering protective immunity in experimental models [44]. In combination, the two components form LT—PA plus LF, and ET—PA plus EF. PA is a pore-forming protein that requires post-translational processing and assembly to become functional (Figure 1). After PA monomers bind to surface receptors and undergo proteolytic processing by host-derived, cell-associated, serine endoproteases of the proprotein convertase (PC) family [49, 50], they form a membrane-associated ‘prepore,’ which is an efficient delivery vehicle for either LF or EF [44, 52]. After receptor-mediated endocytosis and a series of subsequent endosomal events, anthrax toxins are released into the cytosol. 1150 d d Artenstein & Opal B. anthracis Components Macrophages, DCs, Lymphocytes Neutrophils Cardiac Tissue Vascular Effects Pro-inflammatory Cytokines Poly-c-D-glutamic acid capsule [12, 20, 21] Promotes PA binding to cell membranes; potentiates LT activity; aggregates bacterial cells into microcolonies on hepatic endothelium Impairs phagocytosis; hinders immune surveillance of cell surface antigens Increases LT activity by inducing myocardial dysfunction Acts synergistically with LT to induce hypotension May potentiate cytokine generation by LT Lethal Toxin (LT) [22–35] Macrophage cell line cytotoxicity via activation of the cytosolic inflammasome pathway leading to caspase-1mediated pyroptosis; impairs DC maturation, lymphocyte activation, B cell proliferation, and antigen recognition, processing, and presentation to T cells Impairs tissue phagocytosis via effects on actin-based mobility High dose directly impairs myocardial function YCardiac output, Ystroke volume, diastolic dysfunction, vasodilation Promotes low level release of cytokines late in anthrax, secondary to the necrosis of tissue macrophages Edema Toxin (ET) [25, 30, 31, 36–42] Promotes migration of infected macrophages to the lymph nodes; facilitates systemic dissemination of intracellular bacteria; immune evasion via inhibition of T-cell activation [cAMP impairs phagocytosis and oxidative burst [Inotropic and chronotropic effects via [cAMP (indirect) Hypotension; vasodilation; Ystroke volume via Ypreload; diuresis Immune suppression, related to cytokine dysregulation; antiinflammatory effects that limit cytokine release by cAMPinduced COX-2 synthesis Cell wall components [12, 43] Activation via TLR 2/6 Activation via TLR 2/6 Decreased myocardial performance as seen in sepsis YVascular resistance; induces hypotension [Cytokines and chemokines as seen in sepsis Abbreviations: cAMP, cyclic adenosine monophosphate; COS-2, cyclo-oxygenase 2; DCs, dendritic cells; PA, protective antigen; TLR, Toll-like receptor. Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 CID 2012:54 (15 April) Table 1. The Immunologic and Hemodynamic Effects of Bacillus anthracis and Its Toxin Components [12, 20–43] human trials in a disease that rarely occurs in the U.S. The FDA promulgated the ‘‘animal efficacy rule’’ in 2002 to permit the regulatory approval of novel drugs and biological products for life-threatening conditions related to biothreat agents based on data derived from animal studies when well-controlled clinical studies are not feasible [63]. In order for evidence to qualify under the rule, certain provisions must be met: the pathophysiology of the illness must be understood and must justify the use of the product; a favorable effect must either be demonstrated in more than one species with a response predictive of that expected in humans or in a single well-characterized animal model predictive of human responses; animal study endpoints must generally involve either survival or significant morbidity benefits; and pharmacodynamic data must be sufficient to allow for appropriate dose correlation and selection in humans. Separate clinical safety and pharmacokinetic evaluations are required. Hemodynamic Support Maneuvers Illumination of the hemodynamic derangements induced by anthrax exotoxins and structural components suggests a role Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1151 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 Figure 1. Schematic representation of events in the pathogenesis of anthrax and the potential therapeutic opportunities (identified as numbers) that relate to each event. Entities within parentheses represent examples of agents that may inhibit or block a particular pathogenic event (see text): (1) spore germination and bacterial growth (antimicrobials; anthrax phage-derived lysin therapy; quorum-sensing inhibitors, anti-spore antibody, 6-thioguanosine, retrocyclins, CXC chemokines); (2) poly-c-D-glutamic acid exocapsule as a virulence factor (anti-capsule mAbs); (3) bacterial protein synthesis (clindamycin); (4) PA83 binding to either a low- (ANTXR1, formerly tumor endothelial marker 8) or high-affinity (ANTXR2, formerly capillary morphogenesis protein 2) type 1 transmembrane receptor expressed on human cells [44–48] (anti-PA mAbs, soluble ANTXR2 receptor); (5) proteolytic processing of PA83 to PA63 and PA20 by furin or other PCs [49, 50] (IaIp, synthetic furin inhibitors); (6) assembly of the PA prepore on the surface of infected cells via heptamerization or octamerization of proteolytically processed PA monomers [44, 51, 52] (dominant negative PA monomers, cisplatin); (7) LF or EF binding to PA prepore (anti-LF or –EF mAbs); (8) prepore clusters congregate on lipid rafts in a process that may [53] or may not [54] require a co-receptor—LRP6 (dominant negative co-receptor variants) and are internalized in clathrin-coated pits via receptor-mediated endocytosis to the early endosomal compartment [44, 55, 56]; (9) acidification of the endosome results in conformational changes that transform the prepore into a transmembrane, cytosolicdelivery pore that translocates partially denatured LF and EF across its aperture into vesicles that fuse with late endosomal membranes, releasing active toxins into the cytosol, where they exert their deleterious effects on the host [44, 57, 58] (chloroquine, amiodarone, niclosamide); (10) assembly and release of EF/ET and its downstream effects (nifedipine, indomethacin, cromolyn, adefovir); (11) assembly and release of LF/LT and its downstream effects via the inflammasome system (N-acetyl-L-cysteine, auranofin); (12) LF effects via MAPKK pathways (verapamil, dantrolene, protamine, neomycin, quinidine, cisplatin, chemically modified tetracyclines, green tea polyphenols). EF, edema factor; LF, lethal factor; PA83, the initial 83 kD monomeric form of the protective antigen synthesized by B. anthracis; ANTXR1/2, anthrax receptor type 1 and type 2; PA63 and PA20, the 63 kD and 20 kD post-proteolytic forms of protective antigen, respectively. Abbreviations: cAMP, cyclic adenosine monophosphate; CRE, cAMP-response elements; CREB, cAMP response element binding protein; ET, edema toxin; IaIp, inter-alpha-inhibitor proteins; LRP6, low-density lipoprotein receptor-related protein 6; LT, lethal toxin; mAbs, monoclonal antibodies; MAPKK, mitogen-activated protein kinase kinases; PCs, proprotein convertases. 1152 Table 2. Approved Drugs Currently or Previously Used for Indications Other Than Anthrax That Possess Anti-anthrax Toxin Activity d CID 2012:54 (15 April) In vitro Antitoxin Activity Amiodarone [87] LT, murine Mu and CHO cells In vivo Antitoxin Activity Potential Mechanism Increases survival in Blocks PA pore LT-challenged rats formation; raises at high concentrations endosomal pH Current Clinical Uses Cardiac arrhythmias Serious Toxicities Therapeutic Antitoxin Levels Pulmonary fibrosis LT IC50 5 3.5 lM Pneumonitis Thyroid dysfunction Usual serum 5 3.75 lM Toxicity . 5 lM Drug Interactions Drugs that prolong QTc d CYP3A4 inducers or inhibitors may Yor [levels, respectively LT, murine peritoneal Enhances survival in Mu BALB/c mice with pre- or post-LT challenge Rx Raises endosomal pH Anti-malarial Blurred vision 15–30 ng/ml Retinopathy (Anti-toxin effect at achievable serum and tissue levels) Same as Amiodarone Dysrhythmias N-acetyl-Lcysteine [89] (Mucomyst) LT, murine Mu Enhances survival in pretreated, LT challenged BALB/c mice Antioxidant: abrogates cytolysis by reactive oxygen intermediates Acetaminophen toxicity Anaphylactoid reaction ND Nitrates (additive hypotension) Emesis Niclosamide [90] LT, murine Mu NA Blocks PA pore via alterations to endosomal pH Antihelminthica Abdominal pain, anorexia, dysgeusia, diarrhea ND Quinidine [91] LT, rat Mu NA K1 channel inhibition Cardiac arrhythmias Dysrhythmias ND Verapamil [92] LT, murine Mu NA Ca21 channel blockade (LT requires Ca21) Anti-hypertensive Dysrhythmias Nifedipine [93] ET, CHO cells NA Ca21 channel blockade (ET requires Ca21) Anti-hypertensive Dysrhythmias ND Same as Amiodarone Cinchonism ND but .10x usual serum levels needed for antitoxin effect CYP3A4 metabolism as per amiodarone; additive effects with other antihypertensive agents Cerebral ischemia Cerebral ischemia Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 Artenstein & Opal Arrhythmia Chloroquine [88] ND As per verapamil Table 2 continued. In vitro Antitoxin Activity Neomycin [94] LF, murine Mu In vivo Antitoxin Activity NA Potential Mechanism Binds to the polyamine class of glutamate receptors on LF; competitively inhibits substrate cleavage Current Clinical Uses Antimicrobial (topical form) Serious Toxicities Nephrotoxic Therapeutic Antitoxin Levels ND Drug Interactions Additive effects with other nephrotoxic agents Ototoxic Bowel decontamination Prohibitively toxic (oral form) as when used adjunctive Rx for systemically hepatic encephalopathy Protamine [95] Statins [96] (simvistatin, fluvastatin) LF, in cell-free activity assay LT, murine Mu NA NA Zinc chelation vs competitive LF binding Counteracts heparin effects Novel Treatments for Anthrax Disrupts Lipid-lowering posttranslational agents processing of precursors via modulating the Rho-family of GTPase activity; affects trafficking and/or localization of LT once internalized Bleeding Serum levels 5 4–100 lM Neutralizes heparin Anaphylactoid reaction Antitoxin effect at 10 lM Myopathy d CID 2012:54 (15 April) ND CYP3A4 metabolism as per amiodarone ND Additive bleeding risk when used with anticoagulants or antiplatelet agents Hepatic transaminase elevations Acute kidney injury Indomethacin [97] NA Improved ET-associated Reduces ETmorbidity (ie, edema) induced vascular with pretreatment of leakage by rabbits, but no effect possible on survival noted interference with ET action on inflammatory mediators Anti-inflammatory, analgesic Acute kidney injury GI hemorrhage d 1153 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 1154 Table 2 continued. d In vivo Antitoxin Activity Potential Mechanism NA Improved ET-associated Reduces ETmorbidity (ie, edema) induced vascular with pretreatment of leakage by rabbits, but no effect possible on survival noted interference with ET action on inflammatory mediators Cromolyn [97] NA Ketotifen [98] NA d Celecoxib [97] Artenstein & Opal Cisplatin [99] LT, mouse Mu Current Clinical Uses Anti-inflammatory, analgesic Serious Toxicities Therapeutic Antitoxin Levels Drug Interactions Possible increased risk of myocardial infarction and stroke ND Additive bleeding risk when used with anticoagulants or antiplatelet agents Improved ET-associated Inhibits mast cell Anti-allergic (used in morbidity (ie, edema) degranulation, systemic with pretreatment of thereby affecting mastocytosis) rabbits, but no effect histamine activity, on survival noted and thus supporting a role for histamine in ET morbidity None reported ND None reported LT, no direct LT Inhibits mast cell Allergic conjunctivitis inhibition; reduces degranulation, (only approved in vascular leakage thereby affecting ophthalmic form) (LT-induced) in mice; histamine activity extends time to rat death Protects BALB/c mice Inhibits LT cleavage Anti-neoplastic or Fischer rats when of MEKs without given with LT; pre-Rx affecting LF does not protect proteolysis; renders PA unable to form heptamers, thereby impacting LF and EF translocation CNS or respiratory depression ND None reported Nephrotoxicity LT effects are at Cumulative biologically relevant nephrotoxicity doses; however, not with feasible in current aminoglycosides; form because only additive ototoxicity effective when given with loop diuretics concurrently with LT (cisplatin inefficient in presence of competing thiol groups) Ototoxicity Peripheral neuropathy Bone marrow suppression Adefovir [100] EF, mouse Mu NA Adefovir metabolite Antiviral (hepatitis B) binds in the catalytic site of EF Bone marrow suppression EF inhibition at None reported relevant antiviral doses Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 CID 2012:54 (15 April) In vitro Antitoxin Activity FDA-approved but no longer marketed in U.S. a Crystalluria Abbreviations: CHO, Chinese hamster ovary; CNS, central nervous system; CYP, cytochrome; EF, edema factor; ET, edema toxin; GI, gastrointestinal; IC, inhibitory concentration; LF, lethal factor; LT, lethal toxin; MEKs, mitogen-activated protein kinase kinases; Mu, macrophages; NA, not assessed; ND, not determined; PA, protective antigen; QTC, corrected QT interval; Rx, treatment. Additive CNS depression with other agents; cardiovascular toxicity with Ca21 channel blockers Usual serum levels up to 2.5 lM; antitoxin effect at 10 lM Malignant hyperthermia, Muscle weakness Blockade of neuroleptic malignant intracellular Ca21 store mobilization syndrome, chronic muscle spasticity NA Dantrolene [102] LT, mouse Mu Drug Interactions Additive hepatotoxicity with other toxic agents ND Dysrhythmias Antiprotozoal Inhibition of Ca dependent phospholipase A2 activity NA Quinacrine [101] LT, mouse Mu 21 Therapeutic Antitoxin Levels Serious Toxicities Current Clinical Uses Potential Mechanism In vivo Antitoxin Activity In vitro Antitoxin Activity Passive Immunotherapy Toxin-neutralizing antibodies, either in conjunction with antimicrobial agents or as salvage therapy in those not responsive to antimicrobials, target the intoxication responsible for the excessive mortality of systemic disease. Anthrax immune globulin (AIG), an unlicensed polyclonal preparation of antibodies derived from the pooled plasma of healthy, anthrax-vaccinated donors, has been anecdotally used in the management of a small number of cases [67, 68]. Although a clinical trial examining its safety and pharmacodynamics in human volunteers has recently been completed, efficacy studies are lacking. Nonetheless, AIG is a component of the U.S. Strategic National Stockpile and has been recommended for possible early use in conjunction with appropriate antimicrobials and/or surgical debridement in the management of recent cases in Scotland [69]. PA has been a primary focus of passive immunotherapy based on several lines of evidence: it is a shared component of both LT and ET, and thus its neutralization would theoretically derail the pathogenesis of systemic anthrax; it is the major immunogen of the currently licensed U.S. and U.K. anthrax vaccines [1, 70]; and anti-PA antibody has been validated as a serologic correlate of protection in a rabbit inhalational anthrax model [71]. Polyclonal antibodies directed against PA protect guinea pigs from the lethal effects of B. anthracis spores [72]. Polyclonal preparations comprise antibodies against a broad array of microbial epitopes and therefore may be useful in the setting of a laboratory-manipulated binding site mutation, as might occur in a bioterrorism scenario. However, monoclonal antibodies (mAbs) can be engineered to enhance specificity, affinity, and durability; their relative purity may be associated with improved safety profiles [73]. Among mAbs, humanderived products are less likely than their animal-derived counterparts to be associated with hypersensitivity reactions. Multiple, distinct, recombinant, humanized or fully human mAbs directed against PA have demonstrated efficacy in preexposure protection of LT-challenged rats [74–76] and in pre- and post-exposure protection of spore-challenged rabbits [77]. Several products, including raxibacumab, a recombinant human IgG1k mAb that binds PA with high affinity and Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1155 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 Table 2 continued. for specific supportive measures. Volume support with crystalloid infusion appears to worsen mortality in a continuous, LT-infusion rat challenge model, as compared with its beneficial effects in gram-negative bacterial sepsis, perhaps related to endothelial dysfunction [64]. The impact of vasopressors on shock associated with systemic anthrax has received scant attention but has not been shown to affect survival in LTchallenged rats [65]. Additionally, corticosteroids, which are used in cases of sepsis, may have deleterious consequences in anthrax intoxication [66]. Approved Agents for Indications Other Than Anthrax With Antitoxin Activities A number of drugs approved for use in diseases other than anthrax possess activity against anthrax toxins (Table 2). These therapeutic agents generally have well-described pharmacokinetics in humans, and their safety profiles have been established in human studies and/or in post-marketing surveillance. Many are available in oral form, and they could be ready for immediate 1156 d CID 2012:54 (15 April) d Artenstein & Opal use in the event of a biological attack or natural outbreak. However, most have been studied in pathogenesis, not therapeutic models. Several compounds have in vitro activity against either LT/LF or ET/EF. Verapamil [92] and nifedipine [93] act through calcium channel blockade of LT and ET, respectively. Dantrolene [102], used in malignant hyperthermia and chronic spasticity, and quinacrine [101], an antiprotozoal agent, also leverage the calcium dependence of LT but through blockades of calcium mobilization and phospholipase A2 activity, respectively. Others disrupt distinct steps in toxin pathogenesis, such as the competitive binding of LF [94, 95] or EF [100], the processing events in intracellular trafficking [96], or pore formation, which is impaired by raising the endosomal pH [87, 88, 90]. In vivo data derived from small animal experimentation are available for very few agents. N-acetyl-L-cysteine [89], used in acetaminophen toxicity, and the antineoplastic agent cisplatin [99] affect distinct, downstream events in LT pathogenesis. Selected, non-steroidal, anti-inflammatory agents and mast cell stabilizers have been shown to improve edema but not mortality in a rabbit model through their effects on inflammatory mediators incited by ET [97]. Chloroquine, an antimalarial with an established safety profile in humans, has demonstrated both in vitro and in vivo data in support of a potential therapeutic, anti-toxin role in anthrax. It improves the survival of LT-challenged mice at clinically relevant doses and serum/tissue levels when given either before or after systemic intoxication (Table 3) [88]. Additionally, chloroquine modulates the deleterious effect of LT on T cells [103] and demonstrates an enhanced anti-toxin effect when combined with amiodarone [87] or furin inhibitors [104]. However, its potential use may be complicated by the in vitro observation that it may reduce the sporicidal efficiency of macrophages [105]; this will only be resolved through additional animal experimentation. Several experimental agents have anti-toxin activity. Selected, non-antimicrobial, chemically modified tetracyclines competitively inhibit LF-mediated cleavage of MAPKK in vitro at doses achieved in phase I/II human cancer trials [106]. These agents may be active after LF has entered cells. Polyphenols found in green tea non-competitively inhibit the proteolytic activity of LF in vitro and protect rats from LT-induced death, but optimal therapeutic concentrations have not been determined and the mechanism used by the polyphenols is unclear [107]. Conversely, aspirin and other antiplatelet agents are associated with increased mortality in LT-challenged mice and thus may be contraindicated in systemic anthrax [108]. Further discovery of novel, small molecule LF inhibitors is complicated by several issues: as metalloproteinase inhibitors, these compounds may adversely affect the function of essential host metalloproteinases; many candidate drugs appear to lose Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 inhibits binding to its cellular receptor, have been developed for human use [78, 79]. A single bolus of raxibacumab significantly improved survival in prophylactic and therapeutic studies in rabbits and cynomolgus macaques challenged with aerosolized spores [80]. Intravenous raxibacumab, given up to 48 hours after spore challenge and concurrent with bacteremia detection, conferred a significant survival advantage in monkeys; nearly two-thirds of the animals in the mAb group survived [80]. No significant safety concerns were observed among human volunteers achieving drug levels that correlated with survival benefits in two animal models [80]. Since the serum half-life of raxibacumab in humans is significantly longer than that in monkeys, humans may maintain therapeutically effective levels for at least 28 days after infusion. Based on these data, raxibacumab is in the Strategic National Stockpile. Despite successful treatment of established infection in animals, clinical evidence of systemic disease might be an insensitive trigger for the initiation of raxibacumab, as the median length of time from exposure to symptom onset in U.S. anthrax cases was 4 days [62]. Raxibacumab has demonstrated some efficacy when deployed late, after the onset of hemodynamic decompensation in LT-challenged rats [81]. Nonetheless, based on the clinical suspicion of exposure, passive immunotherapy would likely need to be initiated empirically, early, and at the time of combination antimicrobial therapy. Importantly, raxibacumab does not appear to alter the pharmacokinetics of concurrently used antimicrobials or the natural immune response to anthrax infection [80]. Because of the perceived importance of diversity in protective immune responses to anthrax, mAbs directed against other pathogenic elements of the organism have received attention. Both fully human mAbs and humanized chimpanzee mAbs targeting LF demonstrated efficacy in protecting sporechallenged mice and LT-challenged rats, respectively; both may provide additive effects with concurrent anti-PA mAbs [82, 83]. A humanized mAb targeting EF was shown to protect mice against the local and systemic effects of ET [84]. Immunologic approaches targeting the ANTXR2 receptor [85] and capsule [86] represent potential strategies that could be combined with anti-toxin mAbs for passive protection against systemic anthrax. Table 3. Chloroquine Enhances Survival in BALB/c Mice Challenged With Anthrax Lethal Toxin Day 3 Treatment Group Survivors/ Total Day 7 Pa Survivors/ Total Pb CQ controls 5/5 . 5/5 . LT controls 1/17 . 0/17 . (CQx2)c plus LT 8/16 .007 5/16 .018 (CQx1)c plus LT 3/16 .335 2/16 .227 4/16 15/48 .175 .049 4/16 11/48 .044 .054 LT plus (CQx1)c Total CQ-treated plus LT-challenged Abbreviations: CQ, chloroquine; LT, lethal toxin. a Compared with LT controls at day 3 (Fisher’s exact test). b Compared with LT controls at day 7 (Fisher’s exact test). c Position of parentheses, either preceding or after LT, refers to the timing of CQ relative to challenge; and (1 or 2) refers to the number of doses of CQ. Adapted from reference [88], by permission of Oxford University Press. Proprotein Convertase Inhibitors Furin, a member of the proprotein convertase (PC) family of serine proteases, is largely responsible for proteolytic processing of PA at the cell surface [110, 111]. Inter-alpha inhibitor proteins (IaIp) represent a family of endogenous serine protease inhibitors that exist in several isoforms in human plasma and urine [112]. Their broad-spectrum activity during severe inflammatory states against many potentially detrimental proteases found in the plasma, such as complement components, coagulation and fibrinolytic enzymes, and granzymes, and the inverse correlation between plasma IaIp levels and mortality in such states, has fueled interest in IaIp as a possible therapy for bacterial sepsis [113–115]. IaIp have also been shown to inhibit furin and to protect experimental animals when administered up to 24 hours after anthrax spore challenge, enhancing the effect of antimicrobials (Figure 2) [112, 116]. In systemic anthrax, IaIp may inhibit furin-mediated toxin processing and address sepsis-induced functional derangements by blocking excess plasma protease activity. Since IaIp are physiologic molecules and are found in high concentrations in normal human plasma, they are likely to be safe. IaIp are currently undergoing animal challenge studies to confirm their efficacy in established systemic anthrax infection. Other synthetic furin inhibitors have demonstrated potential for use in anthrax intoxication adjunctive therapy [110, 117]. Mutant variants of PA monomers have dominant negative (DN) activity and are able to prevent the assembly of wild-type PA monomers into functional prepores, even when present in low concentrations [22, 118]. DN mutants co-assemble with wild-type PA monomers, disrupting pore formation and thereby blocking the delivery of toxins to the intracellular compartment. Such mutants are protective in a rat intoxication model [118]. The low likelihood that sufficient quantities of DN mutants could be administered in time to improve the outcome in ill patients with systemic anthrax makes it improbable that they will advance to clinical application in their current forms. Other Adjunct Therapy Novel antimicrobial strategies are in development to supplement or replace antibiotics against anthrax bacilli or other gram-positive bacteria with lysins, which are lytic enzymes derived from bacteriophages [119, 120]. Lysin therapy could be used for infections caused by genetically engineered, antibioticresistant B. anthracis strains. Quorum-sensing inhibition represents a potential novel approach to disrupt communication between anthrax bacilli [121]. A number of naturally occurring or synthetic, halogenated furanones have been shown to impair virulence gene expression and growth of B. anthracis [17, 18]. The soluble form of the high affinity anthrax toxin receptor, ANTXR2, has been used as a molecular sponge to adsorb prepores and thus prevent PA binding and assembly on cell membranes [26]. A synthetic, polyvalent inhibitor of PA, based upon the activity of a 12-amino acid peptide discovered in a phage display process, prevents PA pores from translocating EF or LF into the cytosol [122]. Because the transformation of B. anthracis from its infective spore form to its invasive, vegetative form initiates pathogenesis, agents that interfere with the germination process may be useful if deployed early after exposure. The anti-cancer agent 6-thioguanosine, a nucleoside analog of the germinant inosine, blocks spore germination and cytotoxicity in murine macrophages [123]. Selected retrocyclins, synthetic peptides derived from defensin effectors of the macaque innate immune response, have shown both sporicidal and antitoxin activity [124]. Recently, the murine CXC chemokines CXCL9, CXCL10, and CXCL11 were shown to disrupt B. anthracis spore germination and vegetative outgrowth in vitro [125, 126], suggesting a potential role for these effectors in novel therapeutic approaches to anthrax. Auranofin, an organo-gold compound used in the treatment of rheumatoid arthritis, blocks LT-induced caspase-1 synthesis and significantly delays mortality in the Fischer rat model of anthrax intoxication [23]. These findings raise the possibility Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1157 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 potency under physiologic conditions; and drugs must act intracellularly, and therefore, be of high affinity to be therapeutically viable [109]. The availability of crystal structures of experimentally potent LF inhibitors may facilitate the design of future candidates. Dominant Negative Mutants that inflammasome inhibitors or inhibitors of caspase-1 immune products, such as the IL-1 receptor antagonist, may have a role in the management of systemic anthrax [127]. CONCLUSIONS Although antibacterial agents with activity against B. anthracis are effective at clearing bacteremia, optimal therapy for systemic disease will require intervention with toxin inhibitors and perhaps other adjuncts. Some of the more promising existing drugs with anti-toxin activity, such as chloroquine, and endogenous furin inhibitors, such as IaIp, should be rigorously studied in combination with antibacterial agents, anti-PA immune-based therapy, or both in animal models of systemic anthrax. However, it must be acknowledged that host-signaling and immunebased therapies may appear efficacious in animal models of infection and intoxication, yet fail to improve outcomes in clinical trials of severely ill patients [128]. To prove efficacy, 1158 d CID 2012:54 (15 April) d Artenstein & Opal these experimental agents will need to be studied in comparative clinical trials that will treat anthrax victims during an outbreak. Although credible research during epidemics of naturally occurring or intentionally released pathogens is challenging, it must be done in real-time in a systematic and ethical manner and will likely require the development of international, collaborative, clinical research networks [129]. Notes Authors’ contributions. A. W. A. and S. M. O. both contributed to the literature search, the writing of the manuscript, and the construction of Tables and Figures in this work. Acknowledgments. The authors acknowledge Kristina Ward, PharmD, Thomas Hill, and Brian Kurt for information regarding drug pharmacology, and Margo Katz for administrative assistance with the manuscript. Potential conflicts of interest. A. W. A. has no conflicts of interest; S. M. O. has a patent application pending for the use of IaIp as therapy for anthrax. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed. Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 Figure 2. Inter-alpha inhibitor proteins (IaIp) are endogenous inhibitors that prevent the proprotein convertase furin from proteolytically activating PA at the cell surface; IaIp protect against experimental anthrax. A, Concentration-dependent inhibition of furin activity in a fluorometric assay (reproduced with permission from the American Society for Microbiology) [112]; B, IaIp administration immediately before a lethal toxin challenge of BALB/c mice results in significant protection against toxin-induced lung tissue injury, with reduced pulmonary edema as measured by wet/dry weight, and significant protection against toxin-induced injury in the spleen, with reduced splenic pathology as blindly scored by a pathologist (reproduced with permission from the American Society for Microbiology) [112]; C, Kaplan-Meier survival plot of A/J mice injected with a lethal intraperitoneal dose of B. anthracis Sterne strain (ie, lacking the pXO2 plasmid encoding the bacterial capsule) spores and subsequently administered 30 mg/kg of IaIp either 1 h or 24 h later with or without concomitant moxifloxacin or PBS. Mice receiving IaIp in combination with moxifloxacin at either time point after infection demonstrated a significant improvement in survival (86% in the 1-h group; 65% in the 24-h group, P , .001 for both vs PBS controls) as compared with animals receiving either agent alone or PBS control animals (by permission of the Shock Society) [116]; D, Gram-stained lung tissue (200X) from a control group mouse showing alveolar capillary spaces with clumps of Gram-positive bacilli and red blood cells (by permission of the Shock Society) [116]; E, Representative hematoxylin and eosin-stained lung tissue (200X) from an IaIp- and moxifloxacin-treated mouse showing intact airspaces, minimal cellular infiltrates, and an absence of bacterial invasion (by permission of the Shock Society) [116]. Abbreviations: C, control group; PBS, phosphate-buffered saline; W/D, wet-to-dry weight ratio. References Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1159 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 1. Turnbull PC. Anthrax. In: Artenstein AW, ed. Vaccines: a biography. New York, NY: Springer, 2009: 57–71. 2. Holty JE, Bravat DM, Liu H, Olshen RA, McDonald KM, Owens DK. Systematic review: a century of inhalational anthrax cases from 1900 to 2005. Ann Intern Med 2006; 144:270–80. 3. Inglesby TV, O’Toole T, Henderson DA, et al. Anthrax as a biological weapon, 2002: updated recommendations for management. JAMA 2002; 287:2236–52. 4. Ringertz SH, Høiby EA, Jensenius M, et al. Injectional anthrax in a heroin skin-popper. Lancet 2000; 356:1574–5. 5. Ramsay CN, Stirling A, Smith J, et al. An outbreak of infection with Bacillus anthracis in injecting drug users in Scotland. Euro Surveill 2010; 15:pii 19465. Available at: http://www.Eurosurveillance.org. 6. Powell AG, Crozier JE, Hodgson H, Galloway DJ. A case of septicemic anthrax in an intravenous drug user. BMC Infect Dis 2011; 11:21; doi:10.1186/1471-2334-11-21. 7. Radun D, Bernard H, Altmann M, et al. Preliminary case report of fatal anthrax in an injecting drug user in North-Rhine-Westphalia, Germany, December 2009. Euro Surveill 2010; 15:pii 19464. Available at: http://www.Eurosurveillance.org. 8. Bhatnagar R, Friedlander AM. Protein synthesis is required for expression of anthrax lethal toxin cytotoxicity. Infect Immun 1994; 62: 2958–62. 9. Lappin E, Ferguson AJ. Gram-positive toxic shock syndromes. Lancet Infect Dis 2009; 9:281–90. 10. Brook I, Germana A, Giraldo DE, et al. Clindamycin and quinolone therapy for Bacillus anthracis Sterne infection in 60Co-gamma-photonirradiated and sham-irradiated mice. J Antimicrob Chemother 2005; 56:1074–80. 11. Artenstein AW. Anthrax: from antiquity to answers. J Infect Dis 2007; 195:471–3. 12. Spencer RC. Bacillus anthracis. J Clin Path 2003; 56:182–7. 13. Brossier F, Levy M, Mock M. Anthrax spores make an essential contribution to vaccine efficacy. Infect Immun 2002; 70:661–4. 14. Chakrabarty K, Wu W, Booth JL, Duggan ES, Coggeshall KM, Metcalf JP. Bacillus anthracis spores stimulate cytokine and chemokine innate immune responses in human alveolar macrophages through multiple mitogen-activated protein kinase pathways. Infect Immun 2006; 74:4430–8. 15. Pickering AK, Osorio M, Lee GM, Grippe VK, Bray M, Merkel TJ. Cytokine response to infection with Bacillus anthracis spores. Infect Immun 2004; 72:6382–9. 16. Wilson AC, Soyer M, Hoch JA, Parego M. The bicarbonate transporter is essential for Bacillus anthracis lethality. PLoS Pathogens 2008; 4:1–9. 17. Jones MB, Blaser MJ. Detection of a luxS-signaling molecule in Bacillus anthracis. Infection 2003; 71:39–49. 18. Jones MB, Jani R, Ren D, Wood TK, Blaser MJ. Inhibition of Bacillus anthracis growth and virulence-gene expression by inhibitors of quorum-sensing. J Infect Dis 2005; 191:1881–8. 19. Read TD, Peterson SN, Tourassa N, et al. The genome sequence of Bacillus anthracis Ames and comparison with closely related bacteria. Nature 2003; 423:81–8. 20. Jang J, Cho M, Chun JH, et al. The poly-c-D-glutamic acid capsule of Bacillus anthracis enhances lethal toxin. Infect Immun 2011; 79: 3846–54. 21. Piris-Gimenez A, Corre JP, Jouvion G, Candela T, Khun H. Encapsulated Bacillus anthracis interacts closely with liver endothelium. J Infect Dis 2009; 200:1381–9. 22. Terra JK, Cote CK, France B, et al. Cutting edge: resistance to Bacillus anthracis infection mediated by a lethal toxin sensitive allele of NaLp1b/NIrp1b. J Immunol 2010; 184:17–20. 23. Newman ZL, Sirianni N, Mawhinney C, et al. Auranofin protects against anthrax lethal toxin-induced activation of the NIrp1b inflammasome. Antimicrob Agents Chemother 2011; 55:1028–35. 24. Agrawal A, Lingappa J, Leppla SH, et al. Impairment of dendritic cells and adaptive immunity by anthrax lethal toxin. Nature 2003; 424: 329–34. 25. Comer JE, Chopra AK, Peterson JW, Konig R. Direct inhibition of T-lymphocyte activation by anthrax toxins in vivo. Infect Immun 2005; 73:8275–81. 26. Fang H, Xu L, Chen TY, Cyr JM, Frucht DM. Anthrax lethal toxin has direct and potent inhibitory effects on B cell proliferation and immunoglobulin production. J Immunol 2006; 176:6155–61. 27. Baldari CT, Tonello F, Pacani SR, Montecucco C. Anthrax toxins: a paradigm of bacterial immune suppression. Trends Immunol 2006; 27:434–40. 28. During RI, Li W, Hao B, et al. Anthrax lethal toxin paralyzes neutrophil actin-based mobility. J Infect Dis 2005; 192:837–45. 29. Kandadi MR, Hua Y, Ma H, et al. Anthrax lethal toxin suppresses murine cardiomyocyte contractile function and intracellular Ca21 handling via a NADPH oxidase-dependent mechanism. PLoS One 2010; 5:e1335; doi: 10.1371/journal.pone.0013335. 30. Cui X, Li Y, Li X, et al. Bacillus anthracis edema and lethal toxin have different hemodynamic effects but function together to worsen shock and outcome in a rat model. J Infect Dis 2007; 195:572–80. 31. Sweeney DA, Cui Z, Solomon SB, et al. Anthrax lethal and edema toxins produce different patterns of cardiovascular and renal dysfunction and synergistically decrease survival in canines. J Infect Dis 2010; 202:1885–96. 32. Cui X, Moayeri M, Li Y, et al. Lethality during continuous anthrax lethal toxin infusion is associated with circulatory shock but not inflammatory cytokine or nitric oxide release in rats. Am J Physiol Regul Integr Comp Physiol 2004; 286:R699–709. 33. Erwin JL, DaSilva LM, Bavari S, Little SF, Friedlander AM, Chanh TC. Macrophage-derived cell lines do not express proinflammatory cytokines after exposure to Bacillus anthracis lethal toxin. Infect Immun 2001; 69:1175–7. 34. Kim SO, Qing J, Hoebe K, Beutler B, Duesbery NS. Sensitizing anthrax lethal toxin-resistant macrophages to lethal toxin-induced killing by tumor necrosis factor-alpha. J Biol Chem 2003; 278:7413–21. 35. Levy H, Weiss S, Altboum Z, et al. Lethal factor is not required for Bacillus anthracis virulence in guinea pigs and rabbits. Microb Pathog 2011; 51:345–51. 36. Leppla SH. Anthrax toxin edema factor: a bacterial adenylate cyclase that increases cyclic AMP concentrations in eukaryotic cells. Proc Natl Acad Sci U S A 1982; 79:3162–6. 37. Hicks CW, Li Y, Okugawa S, et al. Anthrax edema toxin has cAMPmediated stimulatory effects and high-dose lethal toxin has depressant effects in an isolated perfused rat heart model. Am J Physiol Heart Circ Physiol 2010; 300:H1108–18. 38. Hoover DL, Friedlander AM, Rogers LC, Yoon I-K, Warren RL, Cross AS. Anthrax edema toxin differentially regulates lipopolysaccharideinduced monocyte production of tumor necrosis factor alpha and interleukin-6 by increasing intracellular cyclic AMP. Infect Immun 1994; 62:4432–9. 39. O’Brien J, Friedlander A, Dreier T, Ezzell J, Leppla S. Effects of anthrax toxin components on human neutrophils. Infect Immun 1985; 47:306–10. 40. Crawford MA, Aylott CV, Bordeau RW, Bokoch GM. Bacillus anthracis toxins inhibit human neutrophil NADPH oxidase activity. J Immunol 2006; 176:7557–65. 41. Kim C, Wilcox-Adelman S, Sano Y, Tang WJ, Collier RJ, Park JM. Anti-inflammatory cAMP signaling and cell migration genes co-opted by the anthrax bacillus. Proc Natl Acad Sci U S A 2008; 105:6150–5. 42. Watson LE, Mock J, Lal H, et al. Lethal and edema toxins of anthrax induce distinct hemodynamic dysfunction. Front Biosci 2007; 12: 4670–5. 43. Triantafilou M, Uddin A, Maher S, et al. Anthrax toxin evades Tolllike receptor recognition, whereas its cell wall components trigger activation via TLR2/6 heterodimers. Cell Microbiol 2007; 9:2880–92. 1160 d CID 2012:54 (15 April) d Artenstein & Opal 65. Li Y, Cui X, Su J, et al. Norepinephrine increases blood pressure but not survival with anthrax lethal toxin in rats. Crit Care Med 2009; 37: 1348–54. 66. Moayeri M, Webster JI, Wiggins JF, Leppla SH, Sternberg EM. Endocrine perturbation increases susceptibility of mice to anthrax lethal toxin. Infect Immun 2005; 73:4238–44. 67. Cangene Corporation. Biodefense Products, 2009. Available at: www. cangene.com/biodefense_products.shtml. Accessed 2 February 2012. 68. Walsh JJ, Pesik N, Quinn CP, et al. A case of naturally acquired inhalational anthrax: clinical care and analyses of anti-protective antigen immunoglobulin G and lethal factor. Clin Infect Dis 2007; 44: 968–71. 69. National Services Scotland, Health Protection Scotland. Clinical Guidelines for use of anthrax immune globulin intravenous (human) (AIGIV) in Scotland. Available at: www.hps.scot.nhs.uk/anthrax/ documents/guidelines-anthrax-immune-globulin-v3-2010-0208.pdf. Accessed 2 February 2012. 70. Wright JG, Quinn CP, Shadomy S, Messonnier N. Use of anthrax vaccine in the United States: recommendations of the Advisory Committee on Immunization Practices (ACIP), 2009. MMWR Recomm Rep 2010; 59:1–30. 71. Little SF, Ivins BE, Fellows PF, Pitt ML, Norris SL, Andrews GP. Defining a serological correlate of protection in rabbits for a recombinant anthrax vaccine. Vaccine 2004; 22:422–30. 72. Little SF, Ivins BE, Fellows PF, Friedlander AM. Passive protection by polyclonal antibodies against Bacillus anthracis infection in guinea pigs. Infect Immun 1997; 65:5171–5. 73. Berry JD, Gaudet RG. Antibodies in infectious diseases: polyclonals, monoclonals and niche biotechnology. N Biotechnol 2011; 28:489–501. 74. Maynard JA, Maassen CB, Leppla SH, et al. Protection against anthrax toxin by recombinant antibody fragments correlates with antigen affinity. Nat Biotechnol 2002; 20:597–601. 75. Wild MA, Xin H, Maruyama T, et al. Human antibodies from immunized donors are protective against anthrax toxin in vivo. Nat Biotechnol 2003; 21:1305–6. 76. Sawada-Hirai R, Jiang I, Wang F, et al. Human anti-anthrax protective antigen neutralizing monoclonal antibodies derived from donors vaccinated with anthrax vaccine adsorbed. J Immune Based Ther Vaccines 2004; 2:5; doi: 10.1186/1476-8518-2-5. 77. Mohamed N, Clagett M, Li J, et al. A high-affinity monoclonal antibody to anthrax protective antigen passively protects rabbits before and after aerosolized Bacillus anthracis spore challenge. Infect Immun 2005; 73:795–802. 78. Schneemann A, Manchester M. Anti-toxin antibodies in prophylaxis and treatment of inhalational anthrax. Future Microbiol 2009; 4:35–43. 79. Subramanian GM, Cronin PW, Poley G, et al. A phase I study of PA mAb, a fully human monoclonal antibody against Bacillus anthracis protective antigen, in healthy volunteers. Clin Infect Dis 2005; 41:12–20. 80. Migone TS, Subramanian GM, Zhong J, et al. Raxibacumab for the treatment of inhalational anthrax. N Engl J Med 2009; 361:135–44. 81. Cui X, Li Y, Moayeri M, et al. Late treatment with a protective antigen-directed monoclonal antibody improves hemodynamic function and survival in a lethal toxin-infused rat model of anthrax sepsis. J Infect Dis 2005; 191:422–34. 82. Albrecht MT, Li H, Williamson ED, et al. Human monoclonal antibodies against anthrax lethal factor and protective antigen act independently to protect against Bacillus anthracis infection and enhance endogenous immunity to anthrax. Infect Immun 2007; 75:5425–33. 83. Chen Z, Moayeri M, Crown D, et al. Novel chimpanzee/human monoclonal antibodies that neutralize anthrax lethal factor, and evidence for possible synergy with anti-protective antigen antibody. Infect Immun 2009; 77:3902–8. 84. Chen Z, Moayeri M, Zhao H, Crown D, Leppla SH, Purcell RH. Potent neutralization of anthrax edema toxin by a humanized monoclonal antibody that competes with calmodulin for edema factor binding. Proc Natl Acad Sci U S A 2009; 106:13487–92. Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 44. Young JA, Collier RJ. Anthrax toxin: receptor binding, internalization, pore formation, and translocation. Annu Rev Biochem 2007; 76: 243–65. 45. Bradley KA, Mogridge J, Mourez M, Collier RJ, Young JA. Identification of the cellular receptor for anthrax toxin. Nature 2001; 414:225. 46. Liu S, Leppla SH. Cell surface tumor endothelium marker 8 cytoplasmic tail-independent anthrax toxin binding, proteolytic processing, oligomer formation, and internalization. J Biol Chem 2003; 278:5227–34. 47. Verma K, Gu J, Werner E. Tumor endothelial marker 8 amplifies canonical Wnt signaling in blood vessels. PLoS One 2011; 6:e22334; doi: 10.1371/journal.pone.0022334. 48. Scobie HM, Rainey GJ, Bradley KA, Young JA. Human capillary morphogenesis protein 2 functions as an anthrax toxin receptor. Proc Natl Acad Sci U S A 2003; 100:5170–4. 49. Klimpel KR, Molloy SS, Thomas G, Leppla SH. Anthrax toxin protective antigen is activated by a cell surface protease with the sequence specificity and catalytic properties of furin. Proc Natl Acad Sci U S A 1992; 89:10277–81. 50. Gordon VM, Rehemtulla A, Leppla SH. A role for PACE4 in the proteolytic activation of anthrax toxin protective antigen. Infect Immun 1997; 65:3370–5. 51. Kintzer AF, Thoren KL, Sterling HJ, et al. The protective antigen component of anthrax toxin forms functional octameric complexes. J Mol Biol 2009; 392:614–2. 52. Petosa C, Collier RJ, Klimpel KR, Leppla SH, Liddington RC. Crystal structure of the anthrax toxin protective antigen. Nature 1997; 385:833–8. 53. Wei W, Lu Q, Chaudry GJ, Leppla SH, Cohen SN. The LDL receptorrelated protein LRP6 mediates internalization and lethality of anthrax toxin. Cell 2006; 126:1141–54. 54. Young JJ, Bromberg-White JL, Zylstra C, et al. LRP5 and LRP6 are not required for protective antigen-mediated internalization or lethality of anthrax lethal toxin. PLoS Pathog 2007; 3:e27. 55. Rainey GJ, Wigelsworth DJ, Ryan PL, Scobie HM, Collier RJ, Young JA. Receptor-specific requirements for anthrax toxin delivery into cells. Proc Natl Acad Sci U S A 2005; 102:13278–83. 56. Sun J, Vernier G, Wigelsworth DJ, Collier RJ. Insertion of anthrax protective antigen into liposomal membranes: effects of a receptor. J Biol Chem 2007; 282:1059–65. 57. Brown MJ, Thoren KL, Krantz BA. Charge requirements for proton gradient-driven translocation of anthrax toxin. J Biol Chem 2011; 286:23189–99. 58. Zhang S, Udho E, Wu Z, Collier RJ, Finkelstein A. Protein translocation through anthrax toxin channels formed in planar lipid bilayers. Biophys J 2004; 87:3842–9. 59. Pannifer AD, Wong TY, Schwarzenbacher R, et al. Crystal structure of the anthrax lethal factor. Nature 2001; 414:229–33. 60. Duesbery NS, Webb CP, Leppla SH, et al. Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal factor. Science 1998; 280: 734–7. 61. Vitale G, Pellizzari R, Recchi C, Napolitani G, Mock M, Montecucco C. Anthrax lethal factor cleaves the N-terminus of MAPKKs and induces tyrosine/threonine phosphorylation of MAPKs in cultured macrophages. Biochem Biophys Res Comm 1998; 248:706–11. 62. Jernigan JA, Stephens DS, Ashford DA, et al. Boterrorism-related inhalational anthrax: the first 10 cases reported in the United States. Emerg Infect Dis 2001; 7:933–44. 63. Department of Health and Human Services, Food and Drug Administration. New drug and biological drug products; evidence needed to demonstrate effectiveness of new drugs when human efficacy studies are not ethical or feasible. Fed Regist 2002; 67:37988–98. 64. Sherer K, Li Y, Cui X, et al. Fluid support worsens outcome and negates the benefit of protective antigen-directed monoclonal antibody in a lethal toxin-infused rat Bacillus anthracis shock model. Crit Care Med 2007; 35:1560–7. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. ilomastat block Bacillus anthracis lethal factor activity in viable cells. Infect Immun 2005; 73:7548–57. Dell’Aica I, Dona M, Tonello F, et al. Potent inhibitors of anthrax lethal factor from green tea. EMBO Rep 2004; 5:418–22. Kau JH, Sun DS, Tsai WJ, et al. Antiplatelet activities of anthrax lethal toxin are associated with suppressed p42/44 and p38 mitogen-activated protein kinase pathways in the platelets. J Infect Dis 2005; 192:1465–74. Turk BE. Discovery and development of anthrax lethal factor metalloproteinase inhibitors. Curr Pharm Biotech 2008; 9:24–33. Artenstein AW, Opal SM. Proprotein convertases in health and disease. N Engl J Med 2011; 365:2507–18. Molloy S, Bresnahan P, Leppla S, et al. Human furin is a calciumdependent serine endoprotease that recognizes the sequence Arg-X-XArg and efficiently cleaves anthrax toxin protective antigen. J Biol Chem 1992; 267:16396–402. Opal SM, Artenstein AW, Cristofaro PA, et al. Inter-alpha-inhibitor proteins are endogenous furin inhibitors and provide protection against experimental anthrax intoxication. Infect Immun 2005; 73: 5101–5. Opal SM, Lim YP, Siryaporn E, et al. Longitudinal studies of interalpha inhibitor proteins in severely septic patients: a potential clinical marker and mediator of severe sepsis. Crit Care Med 2007; 35:387–92. Lim YP, Bendelja K, Opal SM, et al. Correlation between mortality and the levels of inter-alpha inhibitors in the plasma of patients with severe sepsis. J Infect Dis 2003; 188:919–26. Ying Z, Chen H, Li YM, et al. Thymosin a1 and ulinastatin-based immunomodulatory strategy for sepsis arising from intra-abdominal infection due to carbapenem-resistant bacteria. J Infect Dis 2008; 198: 723–30. Opal SM, Lim YP, Cristofaro P, et al. Inter-alpha inhibitor proteins as a novel treatment strategy of systemic anthrax infection. Shock 2011; 35:42–4. Jean F, Stella K, Thomas L, et al. Alpha antitrypsin Portland, a bioengineered serpin highly selective for furin: application as an antipathogenic agent. Proc Natl Acad Sci USA 1998; 95:7293–8. Sellman BR, Mourez M, Collier RJ. Dominant-negative mutants of a toxin subunit: an approach to therapy of anthrax. Science 2001; 292:695–7. Schuch R, Nelson D, Fischetti VA. A bacteriolytic agent that detects and kills Bacillus anthracis. Nature 2002; 418:884–9. Fischetti VA. Bacteriophage lysins as effective antibacterials. Curr Opin Microbiol 2008; 11:393–400; doi:10.1016/j.mib.2008.09.012. Asad S, Opal SM. Quorum sensing and the role of cell to cell communication in pathogenesis of invasive infection. Crit Care 2008; 12:236–45. Mourez M, Kane RS, Mogridge J, et al. Designing a polyvalent inhibitor of anthrax toxin. Nat Biotechnol 2001; 19:958–61. Akoachere M, Squires RC, Nour AM, Angelov L, Brojatsch J, Abel-Santos EV. Identification of an in vivo inhibitor of Bacillus anthracis spore germination. J Biol Chem 2007; 282:12112–8. Wang W, Mulakala C, Ward SC, et al. Retrocyclins kill bacilli and germinating spores of Bacillus anthracis and inactivate anthrax lethal toxin. J Biol Chem 2006; 281:32755–64. Crawford MA, Burdick MD, Glomski IJ, et al. Interferon-inducible CXC chemokines directly contribute to host defense against inhalational anthrax in a murine model of infection. PLoS Pathog 2010; 6:e1001199. Crawford MA, Lowe DE, Fisher DJ, et al. Identification of the bacterial protein FtsX as a unique target of chemokine-mediated antimicrobial activity against Bacillus anthracis. Proc Natl Acad Sci U S A 2011; 108: 12159–64. Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 2011; 117:3720–32. Marshall JC, Dietch E, Moldawer LL, et al. Preclinical models of shock and sepsis: what do they tell us? Shock 2006; 24(Suppl 1):1–6. Marshall J, Abraham E, Adikhari N, et al. InFACT: a global critical care research response to H1N1. Lancet 2010; 375:11–13. Novel Treatments for Anthrax d CID 2012:54 (15 April) d 1161 Downloaded from http://cid.oxfordjournals.org/ by guest on September 9, 2014 85. Wycoff KL, Belle A, Deppe D, et al. Recombinant anthrax toxin receptor-Fc fusion proteins produced in plants protect rabbits against inhalational anthrax. Antimicrob Agents Chemother 2011; 55:132–9. 86. Chen Z, Schneerson R, Lovchik J, et al. Pre- and postexposure protection against virulent anthrax infection in mice by humanized monoclonal antibodies to Bacillus anthracis capsule. Proc Natl Acad Sci U S A 2011; 108:739–44. 87. Sanchez AM, Thomas D, Gillespie EJ, et al. Amiodarone and bepridil inhibit anthrax toxin entry into host cells. Antimicrob Agents Chemother 2007; 51:2403–11. 88. Artenstein AW, Opal SM, Cristofaro P, et al. Chloroquine enhances survival in Bacillus anthracis intoxication. J Infect Dis 2004; 190:1655–60. 89. Hanna PC, Kruskal BA, Ezekowitz RA, Bloom BR, Collier RJ. Role of macrophage oxidative burst in the action of anthrax lethal toxin. Mol Med 1994; 1:7–18. 90. Zhu PJ, Hobson P, Southall N, et al. Quantitative high throughput screening identifies inhibitors of anthrax-induced cell death. Biorg Med Chem 2009; 17:5139–45. 91. Newman ZL, Crown D, Leppla SH, Moayeri M. Anthrax lethal toxin activates the inflammasome in sensitive rat macrophages. Biochem Biophys Res Commun 2010; 398:785–9. 92. Bhatnagar R, Singh Y, Leppla SH, Friedlander AM. Calcium is required for the expression of anthrax lethal toxin activity in the macrophage like cell line J774A.1. Infect Immun 1989; 57:2107–14. 93. Kumar P, Ahuja N, Bhatnagar R. Anthrax edema toxin requires influx of calcium for inducing cyclic AMP toxicity in target cells. Infect Immun 2002; 70:4997–5007. 94. Goldman ME, Cregar L, Nguyen D, Simo O, O’Malley S, Humphreys T. Cationic polyamines inhibit anthrax lethal factor protease. BMC Pharmacol 2006; 6:8, doi:10.1186/1471-2210-6-8. 95. Thomas M, Castignetti D. Examination of anthrax lethal factor inhibition by siderophores, small hydroxamates, and protamine. J Microbiol Immunol Infect 2009; 42:284–9. 96. deCathelineau AM, Bokoch GM. Inactivation of Rho GTPases by statins attenuates anthrax lethal toxin activity. Infect Immun 2009; 77: 348–59. 97. Tessier J, Green C, Padgett D, et al. Contributions of histamine, prostanoids, and neurokinins to edema elicited by edema toxin from Bacillus anthracis. Infect Immun 2007; 75:1895–903. 98. Gozes Y, Moayeri M, Wiggins JF, Leppla SH. Anthrax lethal toxin induces ketotifen-sensitive intradermal vascular leakage in certain inbred mice. Infect Immun 2006; 74:1266–72. 99. Moayeri M, Wiggins JF, Lindeman RE, Leppla SH. Cisplatin inhibition of anthrax lethal toxin. Antimicrob Agents Chemother 2006; 50:2658–65. 100. Shen Y, Zhukovskaya NL, Zimmer MI, et al. Selective inhibition of anthrax edema factor by adefovir, a drug for chronic hepatitis B infections. Proc Natl Acad Sci U S A 2004; 101:3242–7. 101. Shin S, Kim YB, Hur GH. Involvement of phospholipase A2 activation in anthrax lethal toxin-induced cytotoxicity. Cell Biol Toxicol 1999; 15:19–29. 102. Shin S, Hur GH, Kim YB, Park KJ, Less WS. Intracellular calcium antagonist protects cultured peritoneal macrophages against anthrax lethal toxin-induced cytotoxicity. Cell Biol Toxicol 2000; 16:137–44. 103. Hirsh MI, Cohen V. Chloroquine prevents T lymphocyte suppression induced by anthrax lethal toxin. J Infect Dis 2006; 194:1003–7. 104. Komiyama T, Swanson JA, Fuller RS. Protection from anthrax toxinmediated killing of macrophages by the combined effects of furin inhibitors and chloroquine. Antimicrob Agents Chemother 2005; 49: 3875–82. 105. Welkos S, Friedlander A, Weeks S, Little S, Mendelson I. In-vitro characterization of the phagocytosis and fate of anthrax spores in macrophages and the effects of anti-PA antibody. J Med Microbiol 2002; 51:821–31. 106. Kocer SS, Walker SG, Zerler B, Golub LM, Simon SR. Metalloproteinase inhibitors, nonantimicrobial chemically modified tetracyclines, and