downloaded from //cvi.asm.org/content/cdli/early/2016/01/04/cvi.00611-15.full.pdf · 04.01.2016...

24
1 1 Adenoviral expression of a bispecific VHH-based neutralizing agent targeting protective 2 antigen provides prophylactic protection from anthrax in mice 3 4 5 6 7 8 Mahtab Moayeri 1 , Jacqueline M. Tremblay 2 , Michelle Debatis 2 , Igor P. Dmietriev 3 , Elena A. 9 Kashentseva 3 , Anthony J. Yeh 4 , Gordon Y.C. Cheung 4 , David T. Curiel 3 , Stephen Leppla 1 , Charles B. 10 Shoemaker 2# 11 12 13 14 15 1 Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy 16 and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. 17 2 Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary 18 Medicine, North Grafton, MA, USA. 19 3 Department of Radiation Oncology, Washington University, St. Louis, MO, USA. 20 4 Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy 21 and Infectious Disease, National Institutes of Health, Bethesda, MD, USA. 22 23 24 #Corresponding author 25 Corresponding author: Dr. Charles B. Shoemaker 26 Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary 27 Medicine, 200 Westboro Rd, North Grafton, MA 01536 28 E-mail address: [email protected] 29 Tel. 508-887-4324; Fax 508-839-7911 30 31 32 33 34 35 Running title: Adenovirus VNA gene therapy against anthrax 36 37 CVI Accepted Manuscript Posted Online 6 January 2016 Clin. Vaccine Immunol. doi:10.1128/CVI.00611-15 Copyright © 2016, American Society for Microbiology. All Rights Reserved. on March 30, 2021 by guest http://cvi.asm.org/ Downloaded from

Upload: others

Post on 19-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    1 Adenoviral expression of a bispecific VHH-based neutralizing agent targeting protective 2 antigen provides prophylactic protection from anthrax in mice 3 4 5 6 7 8 Mahtab Moayeri1, Jacqueline M. Tremblay2, Michelle Debatis2, Igor P. Dmietriev3, Elena A. 9 Kashentseva3, Anthony J. Yeh4, Gordon Y.C. Cheung4, David T. Curiel3, Stephen Leppla1, Charles B. 10 Shoemaker2# 11 12 13 14 15 1Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy 16 and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. 17 2Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary 18 Medicine, North Grafton, MA, USA. 19 3Department of Radiation Oncology, Washington University, St. Louis, MO, USA. 20 4Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy 21 and Infectious Disease, National Institutes of Health, Bethesda, MD, USA. 22 23 24 #Corresponding author 25 Corresponding author: Dr. Charles B. Shoemaker 26 Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary 27 Medicine, 200 Westboro Rd, North Grafton, MA 01536 28 E-mail address: [email protected] 29 Tel. 508-887-4324; Fax 508-839-7911 30 31 32 33

    34

    35

    Running title: Adenovirus VNA gene therapy against anthrax 36

    37

    CVI Accepted Manuscript Posted Online 6 January 2016Clin. Vaccine Immunol. doi:10.1128/CVI.00611-15Copyright © 2016, American Society for Microbiology. All Rights Reserved.

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 2

    Abstract (Limit 250) 38

    Bacillus anthracis, the causative agent of anthrax, secretes three polypeptides which form the 39

    bipartite lethal and edema toxins (LT, ET). The common component in these toxins, protective 40

    antigen (PA), is responsible for binding to cellular receptors and translocating the lethal factor 41

    (LF) and edema factor (EF) enzymatic moieties to the cytosol. Antibodies against PA protect 42

    against anthrax. We previously isolated toxin-neutralizing variable domains of camelid heavy-43

    chain only antibodies (VHHs) and demonstrated their in vivo efficacy. In this work, gene 44

    therapy with an adenoviral vector (Ad/VNA2-PA) promoting expression of a bispecific VHH-45

    based neutralizing agent (VNA2-PA) consisting of two linked VHHs targeting different PA 46

    neutralizing epitopes was tested in two inbred mouse strains, BALB/cJ and C57BL/6J, and found 47

    to protect mice against anthrax toxin challenge and anthrax spore infection. Two weeks after a 48

    single treatment with Ad/VNA2-PA, serum VNA2-PA levels remained above 1 µg/ml, with 49

    some as high as 10 mg/ml. Levels were 10-100 fold higher and persisted longer in C57BL/6J 50

    than in BALB/cJ mice. Mice were challenged with a lethal dose of LT or spores at various times 51

    after Ad/VNA2-PA administration. The majority of BALB/cJ mice having serum VNA2-PA 52

    levels >0.1 µg/ml survived LT challenge and 9 of 10 C57BL/6J mice with serum levels >1 µg/ml 53

    survived spore challenge. Our findings demonstrate the potential for genetic delivery of VNAs as 54

    an effective method for providing prophylactic protection from anthrax. We also extend prior 55

    findings of mouse strain-based differences in transgene expression and persistence by adenoviral 56

    vectors. 57

    58

    59

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 3

    Introduction 60

    Bacillus anthracis produces two toxins which are responsible for allowing the bacterium to 61

    establish disease and induce lethality in the host. Lethal toxin (LT) and edema toxin (ET) are 62

    comprised of three proteins: protective antigen (PA), lethal factor (LF) and edema factor (EF). 63

    PA is a receptor-binding component that transports LF (a protease) or EF (an adenylate cyclase) 64

    into cells where they can manifest their catalytic activities through targeting of ubiquitous 65

    substrates. EF targets ATP and converts it to cAMP, resulting in cellular dysfunction and 66

    vascular events that can lead to lethality. LF cleaves the mitogen-activated protein kinase kinase 67

    (MEK) family and rodent NLRP1 inflammasome sensors. LF plays an important role both early 68

    and late in anthrax infection. Early in infection, inactivation of the MEK proteins by cleavage 69

    leads to inhibition of a wide variety of innate immune cell responses which allows the bacterium 70

    to evade the immune system, divide and disseminate. Cleavage of NLRP1 early in infection in 71

    certain inbred rodents results in the activation of the inflammasome, macrophage pyroptosis and 72

    induction of proinflammatory cytokines which induce a protective immune response. Thus, 73

    certain inbred mouse strains are resistant to spore infection, while others are sensitive. Late in 74

    infection, high levels of both anthrax toxins in the blood induce unknown vascular events which 75

    contribute to the death of the host. The use of tissue-specific PA receptor knockout mice has 76

    now identified target tissues for both toxins. While the mechanism of LT-induced death is 77

    unknown, the cardiovascular system is clearly the important target and PA acts as the “gateway” 78

    for all intoxication events (for a review of all preceding information, see (1)). 79

    PA is an 83-kDa polypeptide that binds to receptors expressed in most tissues. It is then 80

    cleaved by cell-surface proteases such as furin to a 63-kDa form that rapidly oligomerizes. 81

    Heptamers or octamers of PA form binding sites for LF and EF (for review, see (1)). Because 82

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 4

    antibiotic treatment of B. anthracis infection is not effective after the anthrax toxins have 83

    accumulated in the blood, the targeting of PA is an important therapeutic approach against the 84

    disease. The majority of neutralizing antibodies against PA act on the receptor binding domain 4 85

    and prevent the toxin interaction with cells. More rarely PA is neutralized through other 86

    mechanisms (2). 87

    Alpacas, camels and llamas are known to produce heavy chain only antibodies (for 88

    review see (3, 4)). Variable domains of camelid heavy chain only antibodies (VHHs) can be 89

    expressed as recombinant proteins which bind to antigen with similar affinity to the whole Ab, 90

    but also have beneficial features which include resistance to high temperature and pH, as well as 91

    the ability to access conformational epitopes in folded structures which are not generally reached 92

    by conventional antibodies (3, 4). Our laboratories have established the efficacy of VHHs 93

    against a variety of toxins (5-11). Linking of two or more neutralizing VHHs which target 94

    different epitopes creates VHH-based neutralizing agents (VNAs), which have proven to be 95

    much improved anti-toxin agents compared to a pool of their component monomers (8-10, 12). 96

    We previously characterized a potent VNA for treatment of anthrax (VNA2-PA), made as a 97

    heterodimer of two VHHs which neutralize PA by different mechanisms. One VHH, JKH-C7, 98

    inhibits translocation of cell-surface generated PA63 oligomer, while the other, JIK-B8, is a 99

    potent receptor blocker with a subnanomolar binding affinity for PA (6). 100

    Gene therapy for in vivo expression of antibodies has had some success (13-17). In this 101

    work we use a recombinant, replication-incompetent human adenovirus serotype 5 (Ad5) vector 102

    that promotes expression and secretion into the serum of the VNA (Ad/VNA2-PA), thereby 103

    passively “immunizing” the mice. We measured antibody (Ab) levels over an 8-week period 104

    following a single bolus injection of the Ad/VNA2-PA. We performed studies in two different 105

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 5

    inbred strains in parallel, and found that robust protective Ab levels were rapidly established in 106

    both strains, but at significantly different levels, and then dissipated at different rates. Challenge 107

    studies done at various times post-treatment showed that mice having serum VNA levels above 1 108

    µg/ml were protected from anthrax infection. Our results show the potential for VNA gene 109

    therapy as an anthrax therapeutic. 110

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 6

    Materials and Methods 111

    Ethics statement. Animal studies were done in accordance with protocols approved by the 112

    Animal Care and Use Committees at NIAID (Protocols LPD8E, LPD9E). 113

    Toxins. Endotoxin-free PA and LF were purified from B. anthracis as previously described (18). 114

    LT is a combination of PA and LF, always used in equal amount. The LF used here is a 115

    recombinant protein having an N-terminal sequence beginning HMAGG. LT concentrations 116

    correspond to the concentration of each toxin protein (i.e., 100 µg/ml of LT is 100 µg/ml PA + 117

    100 µg/ml LF). 118

    Spores. Spores were prepared from the nonencapsulated, toxigenic B. anthracis Ames 35 (A35) 119

    strain (19) by growth on NBY sporulation agar at 37 °C for 24 h followed by 5 days at room 120

    temperature. Plates were inspected by microscopy to verify sporulation and spores gently 121

    washed off with cold sterile water, followed by four additional cycles of sterile water washes and 122

    centrifugation. Preparations were then heat-treated at 75°C for 1 h to kill any remaining 123

    vegetative bacteria. Spore quantification was performed using a Petroff-Hausser counting 124

    chamber (Hausser Scientific, Horsham, PA) and verified by dilution plating. 125

    Ad/VNA2-PA construction and preparation. The generation of recombinant replication 126

    incompetent Ad5-based vectors has been previously described (20). Briefly in a modification 127

    from (7), pShCMV-JGf7 shuttle plasmid was used for subcloning the VNA2-PA coding 128

    sequence (6), under control of the mammalian CMV promoter and followed by the bovine 129

    growth hormone polyA signal. A control vector Ad/VNA-RT was created in a similar manner 130

    with the sequence from two VHHs against ricin A chain (21). This control vector results in 131

    secretion of a ricin reactive Ab with no binding to PA. Both shuttle plasmids were linearized and 132

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 7

    employed for homologous recombination with pAdEasy-1 plasmid and resultant plasmids 133

    containing viral genomes were validated by PCR, restriction analyses and sequencing. The 134

    plasmids were linearized with PacI to release the inverted terminal repeats of the viral genomic 135

    DNA and transfected into 293 cells to rescue replication incompetent Ad/VNA2-PA and 136

    Ad/VNA-RT. These Ad vectors were propagated in 911 cells, purified by centrifugation of CsCl 137

    gradients, dialyzed and titers determined. 138

    Adenovirus and monoclonal administration and bleeds. C57BL/6J or BALB/cJ mice (female, 8 139

    weeks old) were obtained from Jackson Laboratories (Bar Harbor, Maine). Non-replicative 140

    adenoviral vectors were diluted in sterile saline and injected by tail IV (100 µl/mouse, 3x1010 or 141

    1.2 x1011 viral particles/study). In separate groups, BALB/cJ mice were injected with anti-PA 142

    monoclonal 14B7 (100 µg or 10 µg/mouse, IV, 100 µl). Mice were bled by mandibular or tail 143

    vein route at various days post adenoviral vector administration and serum separated using serum 144

    separator tubes (Sarstedt, Newton, NC). 145

    Serum VNA or monoclonal antibody measurement by ELISAs. Levels of the VNA2-PA or 146

    monoclonal 14B7 in sera was measured by ELISA. Immulon II HB Immunoassay 96-well flat 147

    bottom plates (Thermo Scientific, Franklin, MA) were coated with PA in PBS (10 µg/ml) 148

    overnight at room temperature. Plates were washed with PBS and blocked with 100 µl/well of 149

    1% Gelatin (Biorad, Hercules, CA) for 1 h. Sera from each mouse were serially diluted in 150

    triplicate, incubated for 2-3 h, followed by removal and washing 3X with PBS-Tween (1XPBS+ 151

    0.05% Tween 20). For sera from mice with adenoviral vector injections, an HRP-conjugated 152

    anti-E-tag monoclonal antibody (mAb) (Bethyl laboratories, Montgomery, TX) was added to 153

    each well at 1:3000 dilution and incubated for 2 h, followed by washing 5X with PBS-Tween. 154

    For mice with 14B7 injected, a higher percentage of Tween 20 (1.3%) was used in washes and 155

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 8

    an HRP-conjugated anti-mouse secondary (Santa Cruz BT, Santa Cruz, CA) was used at 1:4000. 156

    HRP substrate reagent (R&D systems, Minneapolis, MN) made of a combination of stabilized 157

    hydrogen peroxide mixed with stabilized tetramethylbenzidine) was used for colorimetric 158

    assessment of HRP activity by spectrometry (450 nM). Purified VNA2-PA or 14B7 dilutions 159

    were used to construct standard curves and Ab concentrations were calculated relative to these 160

    curves using GraphPad Prism software. 161

    Toxin and Spore challenge. Mice were challenged with lethal doses of LT or spores at various 162

    times following adenovirus administration. Toxin challenges were performed in BALB/cJ mice 163

    which are known to be LT-sensitive, but are spore-resistant due to harboring an LT-responsive 164

    Nlrp1b locus (22). Spore challenges were performed in the spore-sensitive C57BL/6J strain 165

    which harbors a nonresponsive locus. LT (100 µg/mouse) was injected IP (500 µl) while spores 166

    (5x107 A35 spores/mouse) were injected SC (200 µl) in the scruff of the neck. Mice were 167

    monitored for signs of malaise and survival twice daily for seven days following infection. 168 on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 9

    Results 169

    We tested the efficacy of in vivo adenoviral production of heterodimeric anti-PA VNA2-PA by 170

    injecting BALB/cJ (n=15) and C57BL/6J (n=15) mice with 3x1010 viral particles of Ad/VNA2-171

    PA, and the same number of mice with a control adenoviral vector (Ad/VNA-RT) that produces 172

    an anti-ricin A chain Ab not reactive to PA. The IV route was selected for administration of 173

    vector, as earlier studies with a similar adenoviral vector expressing an anti-botulinum toxin 174

    VNA showed 6- to 7-fold higher VNA levels following IV vs. IP injections, and >30-fold benefit 175

    over the SC route (7). All mice were bled on day 10 and anti-PA VNA titers assessed. BALB/cJ 176

    mice had anti-PA VNA concentrations ranging from 0.9 µg /ml – 2.3 mg/ml, with an average of 177

    419 µg /ml, and a median of 35 µg/ml. These levels were far lower than the 6.2 mg/ml average 178

    (7.03 mg/ml median) measured for C57BL/6J mice (Figure 1A). This is likely due to the fact 179

    that BALB/c inbred mice eliminate cells containing the Ad transgene much more rapidly than do 180

    C57BL/6 strain mice (23, 24). While 4 of 15 BALB/cJ mice had mg/ml levels of VNA, the rest 181

    had 1-100 µg/ml. The lowest level of VNA in C57BL/6J mice was 1 mg/ml, with the majority 182

    of mice having between 5-10 mg/ml of VNA (Figure 1A). Five mice from each strain were 183

    challenged in a blinded fashion on day 11. BALB/cJ mice were challenged with 1xLD100 dose 184

    of anthrax LT (100 µg, IP), while spore-sensitive C57BL/6J mice were challenged with 185

    5xLD100 dose of 5x107 spores. All mice treated with the Ad/VNA2-PA survived, while all 186

    challenged controls which had been treated with control vector succumbed (Figures 1B, 1C). 187

    VNA2-PA serum levels were again assessed on day 18 for all mice and were found to be 188

    reduced, but to very different levels in a mouse strain-dependent manner. In BALB/cJ mice, 189

    11/15 had levels 10 µg/ml (Figure 2A), as will 190

    be discussed later. Because protection against LT bolus challenge has historically required bolus 191

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 10

    administration of at least 50 µg of neutralizing mAb, we challenged the remaining BALB/cJ 192

    mice (n=10), with 1xLD100 of LT on day 19. Of the ten challenged mice, 50% survived 193

    challenge (Figure 2B). It appeared that mice with VNA levels of >1 µg/ml (>18.6 nM) survived, 194

    with one exception, while levels

  • 11

    challenged with anthrax spores (5xLD100 dose of 5x107/mouse) on day 34, there was a 215

    substantial delay in onset of malaise and death in the mice treated with Ad/VNA2-PA, and 40% 216

    of challenged mice survived (Figure 4B). Mice with 1 mg/ml (Figure 5). By 4 weeks, however, the levels of VNA had dropped 224

    precipitously, to the 0.1- 1 µg/ml range for all mice, indicating that the relative net loss of VNA 225

    in BALB/cJ mice occurred in proportion to the initial starting concentration, but at a similar pace 226

    independent of vector dose. Not surprisingly, only 3 of 16 mice challenged with LT survived. 227

    228

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 12

    229

    Discussion: 230

    We tested the ability of an Ad5-based adenoviral vector (Ad/VNA2-PA) expressing a bispecific 231

    VHH-based neutralizing agent (VNA2-PA) consisting of two linked VHHs targeting different 232

    anthrax toxin PA neutralizing epitopes to protect mice against anthrax toxin challenge and 233

    anthrax spore infection. A single treatment with Ad/VNA2-PA, resulted in antibody levels as 234

    high as 10 mg/ml. Levels were higher and persisted longer in C57BL/6J than in BALB/cJ mice. 235

    LT-sensitive BALB/cJ mice having serum VNA2-PA levels >0.1 µg/ml survived LT challenge, 236

    and spore-sensitive C57BL/6J mice with levels >1 µg/ml survived spore challenge. The studies 237

    presented here indicate that adenoviral delivery of VHH-based neutralizing agents, VNAs, can 238

    provide an excellent alternative to standard antibody therapeutics. Sustained levels of VNA with 239

    a single administration of non-replicative Ad5 adenovirus allows the host to combat effects of 240

    toxin or virulence factors for longer periods than repeated administration of purified antibody. 241

    Furthermore, the gene therapy vectors can be used as prophylactic therapeutics if there is a 242

    danger for exposure of large populations to toxic agents. The VHH-based therapeutics can also 243

    be engineered to make VNAs which target multiple toxins or agents in a single product, 244

    delivered with a single inoculation (9). These vectors also allow the possibility to deliver VNAs 245

    to specific tissues sites through vector engineering (25). While Ad5 vector use in humans could 246

    be limited by widespread pre-existing immunity, alternative gene therapy vectors such as 247

    adenoviruses from simian sources or adeno-associated viruses are being developed that may 248

    prove more practical for general use. 249

    Interestingly, the levels of VNA in our current studies were not sustained for as long a 250

    period as those observed for a similarly constructed anti-botulinum antitoxin VNA which was 251

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 13

    delivered at 3x1010, IV, into CD-1 Swiss mice (7). In that study, VNA levels remained at 1-10 252

    mg/ml in half the mice even at 8 weeks post inoculation, although some mice had significantly 253

    lower levels as early as 10 days post inoculation. Thus, the range of serum VNA could vary 254

    from 0.01 ng/ml to >100 µg/ml in the mice at 6-8 weeks after vector administration. The reason 255

    for this is very likely the genetic heterogeneity of CD-1 mice, which are outbred. Genetic factors 256

    have been previously reported to influence the efficiency of Ad infection and/or transgene 257

    production (24, 26, 27) and it is not surprising that these factors would be more variable in the 258

    outbred mice than observed within the two inbred lines used in the current study. The same 259

    genetic factors are expected to exist among humans and thus lead to differential responses to 260

    various forms of antibody and VNA gene therapy. 261

    Our findings that protection against anthrax toxin is possible for weeks after a single 262

    administration of Ad/VNA2-PA suggest that adenoviral anti-anthrax therapeutics are a viable 263

    option as a future therapeutic agent for this disease. The options of administering intranasal 264

    Ad/VNA2-PA vectors, or parenterally administering Ad/VNA2-PA vectors designed to promote 265

    pulmonary VNA expression (28), may result in even more effective therapeutics for anthrax 266

    exposures. Future studies will focus on tissue-specific targeting of VNA gene therapy vehicles 267

    for more efficient neutralization of toxic effects at relevant disease sites. 268

    269

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 14

    270

    Acknowledgements 271

    This research was supported in part by the Intramural Research Program of the National Institute 272

    of Allergy and Infectious Diseases (NIAID) and by National Institutes of Health Grant U54 273

    AI057159 (CBS). The authors thank the staff of the NIAID Comparative Medicine Branch for 274

    assistance with vivarium studies 275

    276

    277

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 15

    Figure Legends 278

    279

    Figure 1. Day 10 sera analyses and Day 11 challenge studies. A) Groups of BALB/cJ or 280

    C57BL/6J mice (n=15/group) were injected with Ad/VNA2-PA or Ad/VNA-RT (3x1010 viral 281

    particles), and VNA2-PA levels were assessed on day 10. Each circle refers to a single mouse, 282

    with the upper panel showing Ad/VNA2-PA groups and lower panel showing the control vector 283

    groups. The dashed lines indicate the average (black) and median (red) Ad/VNA2-PA levels for 284

    each strain. The open boxes indicate the mice that were challenged, with results shown in panels 285

    B and C. B) BALB/cJ mice treated with Ad/VNA2-PA and control mice (n=5/see panel A) were 286

    challenged on day 11 with LT (100 µg, IP) and monitored for malaise and survival. C) 287

    C57BL/6J mice treated with Ad/VNA2-PA and control mice (n=5/group, see panel A) were 288

    challenged with anthrax A35 spores (5x107/mouse) and monitored for malaise and survival. 289

    290

    Figure 2. Day 18 sera analyses and Day 19 challenge study results. A) Day 18 sera from the 291

    mice that survived challenge as well unchallenged mice described in Figure 1 were analyzed for 292

    VNA2-PA levels. Each circle refers to a single mouse, with the upper panel showing Ad/VNA2-293

    PA groups and lower panel the control vector groups. The open boxes indicate the two groups of 294

    n=10 BALB/cJ mice that were challenged, with results shown in panel B. The black filled 295

    rectangles indicate the absence of sera for mice that succumbed in previous challenge. The 296

    dashed lines indicate the average (black) and median (red) Ad/VNA2-PA levels for each strain. 297

    The “D” identifies 4 mice that succumbed to the challenge described in panel B. B) BALB/cJ 298

    mice treated with Ad/VNA2-PA and control mice (n=10/group, see panel A) were challenged on 299

    day 19 with LT (100 µg, IP) and monitored for malaise and survival. 300

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 16

    301

    Figure 3. 14B7 levels in sera and LT challenge outcome. A) BALB/cJ mice (n=5/group) were 302

    injected with either 10 or 100 µg of mAb 14B7 (IV, 100 µl) and bled at 2 h to assess circulating 303

    levels of PA-specific monoclonal. The solid line indicates the average mAb serum level. B) All 304

    mice from panel A were challenged with LT (100 µg, IP), 2.5 h following mAb administration. 305

    Mice were monitored for a week for signs of malaise. 306

    307

    Figure 4. Day 25 and 33 sera analyses and Day 34 spore challenge study results. A) A 308

    comparison of day 18 sera VNA2-PA levels from Figure 2 is shown with levels on day 25 and 309

    33. The dashed line indicates the average Ad/VNA2-PA levels for the ten C57BL/6J mice on 310

    DAY 33. The open box indicates the mice that were challenged on day 34. The “D” identifies 6 311

    mice that succumbed to the challenge described in panel B. B) C57BL/6J mice treated with 312

    Ad/VNA2-PA and control mice (n=10/group, see panel A) were challenged on day 34 with 313

    anthrax A35 spores (5x107/mouse) and monitored for malaise and survival. 314

    315

    Figure 5. Study 2: Week 2 and Week 4 sera analyses and Week 5 LT challenge studies. A) 316

    BALB/cJ mice (n=16/ group) were injected with Ad/VNA2-PA or Ad/VNA-RT (3x1010 viral 317

    particles), and VNA2-PA levels were assessed at 2 weeks and 4 weeks. Results shown are for 318

    the Ad/VNA2-PA group, as Ad/VNA-RT mice never had PA-specific antibodies. The dashed 319

    lines indicates the average Ad/VNA2-PA levels at 2 and 4 weeks. Each circle refers to a single 320

    mouse. “S” identifies 3 mice that survived the challenge shown in B. B) All mice in panel A 321

    were challenged at 5 weeks with LT (100 µg, IP) and monitored for malaise and survival. 322

    323

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 17

    References 324

    325

    1. Moayeri M, Leppla SH, Vrentas C, Pomerantsev A, Liu S. 2015. Anthrax 326 Pathogenesis. Annu Rev Microbiol doi:10.1146/annurev-micro-091014-104523. 327

    328 2. Chen Z, Moayeri M, Purcell R. 2011. Monoclonal antibody therapies against anthrax. 329

    Toxins (Basel) 3:1004-1019. 330 331 3. Hassanzadeh-Ghassabeh G, Devoogdt N, De Pauw P, Vincke C, Muyldermans S. 332

    2013. Nanobodies and their potential applications. Nanomedicine (Lond) 8:1013-1026. 333 334 4. Muyldermans S. 2013. Nanobodies: natural single-domain antibodies. Annu Rev 335

    Biochem 82:775-797. 336 337 5. Kaliberov SA, Kaliberova LN, Buggio M, Tremblay JM, Shoemaker CB, Curiel DT. 338

    2014. Adenoviral targeting using genetically incorporated camelid single variable 339 domains. Lab Invest 94:893-905. 340

    341 6. Moayeri M, Leysath CE, Tremblay JM, Vrentas C, Crown D, Leppla SH, 342

    Shoemaker CB. 2015. A heterodimer of a VHH (variable domains of camelid heavy 343 chain-only) antibody that inhibits anthrax toxin cell binding linked to a VHH antibody 344 that blocks oligomer formation is highly protective in an anthrax spore challenge model. J 345 Biol Chem 290:6584-6595. 346

    347 7. Mukherjee J, Dmitriev I, Debatis M, Tremblay JM, Beamer G, Kashentseva EA, 348

    Curiel DT, Shoemaker CB. 2014. Prolonged prophylactic protection from botulism with 349 a single adenovirus treatment promoting serum expression of a VHH-based antitoxin 350 protein. PLoS One 9:e106422. 351

    352 8. Mukherjee J, Tremblay JM, Leysath CE, Ofori K, Baldwin K, Feng X, Bedenice D, 353

    Webb RP, Wright PM, Smith LA, Tzipori S, Shoemaker CB. 2012. A novel strategy 354 for development of recombinant antitoxin therapeutics tested in a mouse botulism model. 355 PLoS One 7:e29941. 356

    357 9. Sheoran AS, Dmitriev IP, Kashentseva EA, Cohen O, Mukherjee J, Debatis M, 358

    Shearer J, Tremblay JM, Beamer G, Curiel DT, Shoemaker CB, Tzipori S. 2015. 359 Adenovirus vector expressing Stx1/Stx2-neutralizing agent protects piglets infected with 360 Escherichia coli O157:H7 against fatal systemic intoxication. Infect Immun 83:286-291. 361

    362 10. Tremblay JM, Mukherjee J, Leysath CE, Debatis M, Ofori K, Baldwin K, Boucher 363

    C, Peters R, Beamer G, Sheoran A, Bedenice D, Tzipori S, Shoemaker CB. 2013. A 364 single VHH-based toxin-neutralizing agent and an effector antibody protect mice against 365 challenge with Shiga toxins 1 and 2. Infect Immun 81:4592-4603. 366

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 18

    11. Yang Z, Schmidt D, Liu W, Li S, Shi L, Sheng J, Chen K, Yu H, Tremblay JM, 367 Chen X, Piepenbrink KH, Sundberg EJ, Kelly CP, Bai G, Shoemaker CB, Feng H. 368 2014. A novel multivalent, single-domain antibody targeting TcdA and TcdB prevents 369 fulminant Clostridium difficile infection in mice. J Infect Dis 210:964-972. 370

    371 12. Vance DJ, Tremblay JM, Mantis NJ, Shoemaker CB. 2013. Stepwise engineering of 372

    heterodimeric single domain camelid VHH antibodies that passively protect mice from 373 ricin toxin. J Biol Chem 288:36538-36547. 374

    375 13. De BP, Hackett NR, Crystal RG, Boyer JL. 2008. Rapid/sustained anti-anthrax passive 376

    immunity mediated by co-administration of Ad/AAV. Mol Ther 16:203-209. 377 378 14. Sofer-Podesta C, Ang J, Hackett NR, Senina S, Perlin D, Crystal RG, Boyer JL. 379

    2009. Adenovirus-mediated delivery of an anti-V antigen monoclonal antibody protects 380 mice against a lethal Yersinia pestis challenge. Infect Immun 77:1561-1568. 381

    382 15. Tutykhina IL, Sedova ES, Gribova IY, Ivanova TI, Vasilev LA, Rutovskaya MV, 383

    Lysenko AA, Shmarov MM, Logunov DY, Naroditsky BS, Tillib SV, Gintsburg AL. 384 2013. Passive immunization with a recombinant adenovirus expressing an HA (H5)-385 specific single-domain antibody protects mice from lethal influenza infection. Antiviral 386 Res 97:318-328. 387

    388 16. Deal CE, Balazs AB. 2015. Engineering humoral immunity as prophylaxis or therapy. 389

    Curr Opin Immunol 35:113-122. 390 391 17. Suscovich TJ, Alter G. 2015. In situ production of therapeutic monoclonal antibodies. 392

    Expert Rev Vaccines 14:205-219. 393 394 18. Park S, Leppla SH. 2000. Optimized production and purification of Bacillus anthracis 395

    lethal factor. Protein Expr Purif 18:293-302. 396 397 19. Pomerantsev AP, Sitaraman R, Galloway CR, Kivovich V, Leppla SH. 2006. 398

    Genome engineering in Bacillus anthracis using Cre recombinase. Infect Immun 74:682-399 693. 400

    401 20. Luo J, Deng ZL, Luo X, Tang N, Song WX, Chen J, Sharff KA, Luu HH, Haydon 402

    RC, Kinzler KW, Vogelstein B, He TC. 2007. A protocol for rapid generation of 403 recombinant adenoviruses using the AdEasy system. Nat Protoc 2:1236-1247. 404

    405 21. Herrera C, Tremblay JM, Shoemaker CB, Mantis NJ. 2015. Mechanisms of Ricin 406

    Toxin Neutralization Revealed through Engineered Homodimeric and Heterodimeric 407 Camelid Antibodies. J Biol Chem doi:10.1074/jbc.M115.658070. 408

    409 22. Moayeri M, Crown D, Newman ZL, Okugawa S, Eckhaus M, Cataisson C, Liu S, 410

    Sastalla I, Leppla SH. 2010. Inflammasome sensor Nlrp1b-dependent resistance to 411

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • 19

    anthrax is mediated by caspase-1, IL-1 signaling and neutrophil recruitment. PLoS 412 Pathog 6:e1001222. 413

    414 23. Peng Y, Falck-Pedersen E, Elkon KB. 2000. Soluble CD8 attenuates cytotoxic T cell 415

    responses against replication-defective adenovirus affording transprotection of transgenes 416 in vivo. J Immunol 165:1470-1478. 417

    418 24. Peng Y, Falck-Pedersen E, Elkon KB. 2001. Variation in adenovirus transgene 419

    expression between BALB/c and C57BL/6 mice is associated with differences in 420 interleukin-12 and gamma interferon production and NK cell activation. J Virol 75:4540-421 4550. 422

    423 25. Alberti MO, Roth JC, Ismail M, Tsuruta Y, Abraham E, Pereboeva L, Gerson SL, 424

    Curiel DT. 2012. Derivation of a myeloid cell-binding adenovirus for gene therapy of 425 inflammation. PLoS One 7:e37812. 426

    427 26. Gregory SM, Nazir SA, Metcalf JP. 2011. Implications of the innate immune response 428

    to adenovirus and adenoviral vectors. Future Virol 6:357-374. 429 430 27. Muruve DA. 2004. The innate immune response to adenovirus vectors. Hum Gene Ther 431

    15:1157-1166. 432 433 28. Alberti MO, Deshane JS, Chaplin DD, Pereboeva L, Curiel DT, Roth JC. 2013. A 434

    myeloid cell-binding adenovirus efficiently targets gene transfer to the lung and escapes 435 liver tropism. Gene Ther 20:733-741. 436

    437

    on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/

  • on March 30, 2021 by guest

    http://cvi.asm.org/

    Dow

    nloaded from

    http://cvi.asm.org/