Next Article in Journal
Shared Patterns of Brain Functional Connectivity for the Comorbidity between Migraine and Insomnia
Previous Article in Journal
Initial Biological Assessment of Upconversion Nanohybrids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plague Prevention and Therapy: Perspectives on Current and Future Strategies

by
Raysa Rosario-Acevedo
,
Sergei S. Biryukov
,
Joel A. Bozue
and
Christopher K. Cote
*
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA
*
Author to whom correspondence should be addressed.
Submission received: 1 September 2021 / Revised: 27 September 2021 / Accepted: 4 October 2021 / Published: 9 October 2021
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Plague, caused by the bacterial pathogen Yersinia pestis, is a vector-borne disease that has caused millions of human deaths over several centuries. Presently, human plague infections continue throughout the world. Transmission from one host to another relies mainly on infected flea bites, which can cause enlarged lymph nodes called buboes, followed by septicemic dissemination of the pathogen. Additionally, droplet inhalation after close contact with infected mammals can result in primary pneumonic plague. Here, we review research advances in the areas of vaccines and therapeutics for plague in context of Y. pestis virulence factors and disease pathogenesis. Plague continues to be both a public health threat and a biodefense concern and we highlight research that is important for infection mitigation and disease treatment.

1. Plague History and Disease Presentation

Yersinia pestis is a Gram-negative coccobacillus that is the etiologic agent of plague, an acute infectious disease that has played an important role in human history [1,2,3,4,5]. This bacterium has been responsible for three devastating pandemics that led to millions of deaths [6,7,8]. The first pandemic, referred to as the “Justinian Plague”, occurred in 541 AD, originated in Central Africa, and spread to the Mediterranean region [7]. The “Black Death” of 1347 was the second major pandemic and occurred during the 14th century, originating in Central Asia and eventually reaching the Middle East, North Africa, and Europe. The third pandemic took place in 1855 in Yunnan, China and spread globally via infected rats on ships likely originating from Hong Kong [7,9,10]. This bacterium has impacted human civilization throughout history and continues to be of concern [5].
Dissemination of Y. pestis among wild rodents and humans takes place in two different cycles, enzootic and epizootic [11]. Enzootic refers to when the infection, at low levels, cycles naturally among wild rodents (e.g., rats, squirrels, marmots, and prairie dogs). Sporadically, other susceptible species can be infected, causing an outbreak—this is referred to as epizootic plague. In this disease cycle, animals become infected and ultimately die as fleas from infected natural reservoir species begin to seek other blood sources and the probability of human transmission is increased [11].
Y. pestis can cause three forms of plague disease: bubonic, septicemic, and pneumonic (Figure 1). Historically, the bubonic form of plague has been the most common and can be transmitted to humans by a bite from an infected flea or by handling an animal infected with Y. pestis [12,13,14,15]. After transmission, the Y. pestis bacteria disseminate to the nearest lymph node where they colonize and subsequently proliferate. The incubation time ranges from approximately 2–8 days and during that period patients will experience flu-like symptoms, such as chills, fever and headaches [16,17,18,19]. As the bacteria continue to multiply, the lymph nodes become swollen and extremely painful. Infected lymph nodes (referred to as buboes when attributable to plague) are generally found in specific areas of the body, such as the neck, under the chin, armpits and groin. If left untreated, these buboes can become necrotic and cause hemorrhaging ultimately leading to lethal disease in approximately 50–60% of cases [20]. A culture isolated from a blood sample or fluid from the swollen lymph node may be necessary for the diagnosis and identification of Y. pestis [17]. Several classes of antibiotics are effective in treating bubonic plague and if treatment is initiated promptly, antibiotics can significantly increase survival rates [19,21,22]. Aminoglycosides, such as streptomycin and gentamicin, tetracyclines, and the fluoroquinolone ciprofloxacin are some effective antibiotics for bubonic plague treatment [20,23,24].
In other more severe cases, the bacteria can enter the blood stream directly and multiply, causing septicemic plague. The increased number of bacteria in the blood causes the release of endotoxins, leading to ischemic necrosis. In those cases, Y. pestis directly infects the blood without developing bubonic plague symptoms (resulting in primary septicemic plague) [25]. Furthermore, septicemia may induce intravascular coagulation that may lead to gangrene of the extremities. The prognosis for these patients is poor, with mortality rates reaching 100% in untreated individuals. Even when the appropriate treatment is administered, mortality rates remain high [26]
A third manifestation of plague, pneumonic plague, can be transmissible either person-to-person or by secondary pneumonia, resulting from the colonization of the lungs with Y. pestis via hematogenous spreading [1,27]. If droplets containing plague bacteria are inhaled, the result can be primary pneumonic plague. Secondary pneumonic plague may be associated with primary septicemic plague or as a complication of bubonic plague. In the United States, approximately 5–10% of patients develop secondary pneumonic plague [28]. Symptoms of plague include chills, fever, body pains, headache, weakness, dizziness, and chest discomfort [29]. These rather non-descript symptoms mirror many other infectious diseases, which can lead to frequent misdiagnosis and subsequent poor disease outcomes for the patients. Blood cultures or a sputum sample can be used to diagnose pneumonic plague [30]. Pneumonic plague is considered highly contagious and is nearly 100% lethal if appropriate treatment is not administered within the first 20 h after onset of symptoms [30].
Due to rapid spread, high fatality rates, and the ability to transmit via aerosols, Y. pestis has been classified as a Tier 1 select agent by the United States Department of Health and Human Services and is considered an agent of biothreat concern [31,32]. Numerous studies have been performed in mice and other species in order to elucidate Y. pestis pathogenesis and pneumonic plague disease progression in animal models of human disease [25,33,34,35]. It has been reported that during the first 36 h post infection in mice, there is an increase in bacterial replication in the lungs with no appreciable changes in cytokines or chemokines levels [36,37]. In this pre-inflammatory phase, it has been shown that Y. pestis suppresses the host immune response in order to facilitate pulmonary infection [38,39]. After 36 h, the pro-inflammatory phase begins, where there is an upregulation of cytokine or chemokine levels, resulting in a critical point that can lead to death [40].

2. Yersinia pestis Virulence Plasmids

Y. pestis must be able to survive in and infect very diverse hosts, including both fleas and mammals. There are numerous essential virulence factors and effector proteins that are either encoded on the chromosome of Y. pestis or carried on its plasmids. It is believed that Y. pestis evolved from the genetically related Yersinia species Yersinia pseudotuberculosis (usually an intestinal pathogen) through the process of gaining plasmids, most likely several thousands of years ago [4,41,42].
Y. pestis carries three plasmids, two of which are unique to this species: pMT1 (or pFra), which encodes the F1 capsular antigen and pPCP (or pPst or pPla), which carries the gene for the virulence factor plasminogen activator. The third plasmid is common to the human pathogenic Yersiniae and is known as pCD1 (calcium dependence), pYV (Yersinia virulence), or pLcr (low calcium response). This plasmid is responsible for the synthesis of a number of anti-host factors and is an absolute requirement for virulence.
The pMT1/pFra plasmid (100–110 kb) carries two important Y. pestis virulence factors involved with survival/infection of these two different hosts: the murine toxin Ymt and the F1 capsular protein. Ymt is a phospholipase D which has toxic properties for mice and rats but not for other animals [43,44,45]. More importantly, Ymt is required for survival within the midgut of the flea by protecting it against some components of plasma [46]. In addition, the caf operon encoding for the F1 capsular antigen protein (referred to here as F1) is also located on pMT1. The F1 antigen has been described both as capsular and fimbrial-like as it is composed of fibers [47,48,49,50]. The F1 protein is produced abundantly in vivo during infection of a mammal and in vitro when grown at 37 °C. F1 is thought to protect Y. pestis from uptake by host phagocytic cells [51]. However, naturally derived and genetically engineered F1-negative Y. pestis strains have been described and remain virulent in animal models of plague [52,53].
The other Y. pestis specific plasmid (pPCP/pPst/pPla) is 9.5 kb and carries genes that encode for pesticin, pesticin immunity protein, and plasminogen activator (Pla) [54]. Pla appears to be a multifunctional protein and belongs to the omptin family of enterobacterial surface proteins. One of the functions of Pla during infection is to cleave numerous host substrates, such as those involved in coagulation and fibrinolysis [55]. During bubonic plague, Pla activates host fibrolysis to allow bacterial dissemination from the site of infection [56,57]. Furthermore, this protein is believed to alter the host immune response during pneumonic plague to allow outgrowth of the bacteria within the lungs [58,59]. In addition to its ability to cleave proteins, it has a role in mediating bacterial adhesion to host cells and extracellular matrices [60,61,62,63,64].
The final plasmid is pCD1/pYV/pLcr (68–75 kb), which is common to all human pathogenic Yersiniae, is essential for virulence, and carries genes encoding for the Type III secretion system (T3SS) and effector proteins known as Yops (Yersinia outer protein). The T3SS forms a syringe-like structure (injectisome) made up of 27 proteins, including the LcrV protein, which is both a structural and effector protein and is critical for virulence [65,66,67,68,69,70,71,72,73,74,75,76]. The injectisome is able to “inject” host cells when in close contact with Yops. Two of the Yops (YopB and YopD) function to transport the other six effector proteins into the host cell (YopO, YopH, YopM, YopT, YopJ, and YopE). Toxic activities of the Yops include disruption of the cytoskeleton, interference with phagocytic activity, prevention of proinflammatory cytokine synthesis, inhibition of the oxidative burst, and induction of programmed cell death (apoptosis). The overall effect of the Yops is to block phagocytic uptake of the bacteria by host macrophages and polymorphonuclear leukocytes [77,78,79,80,81,82].

3. Outbreak Prevention

Despite the efforts of public health agencies to monitor and prevent this disease, reports of plague outbreaks continue to exist in various parts of the word [83,84,85]. The United States, China, India, Vietnam, Democratic Republic of the Congo, Peru, and Madagascar are among the countries that have confirmed plague cases annually and their respective climates, public health infrastructure, surveillance programs, and socio-economic differences make outbreak prevention strategies and severity of outbreaks unique in each case [86,87,88]. Madagascar is currently the most affected country with the highest numbers of recently reported cases of plague, representing approximately 75% of the reported cases in the world [87,89,90]. Differences in weather and temperature patterns affect the abundance of fleas [89,91] which in turn influences the start of annual transmission that generally arises between the months of September to April in Madagascar [92]. Given the right conditions, it may be possible for person-to-person transmission mediated by human ectoparasites—especially in underdeveloped but densely populated areas [93]. A total of 2417 suspect plague cases, including 209 deaths, were reported by the Word Health Organization (WHO) from August to November in 2017 in Madagascar [94]. Generally, bubonic plague cases in Madagascar are commonly concentrated in rural areas, while pneumonic cases are more prevalent in urban locations [95]. In 2017, the majority (77%; 1854 total) reported were clinically classified as pneumonic plague whereas 15% (355) were classified as bubonic plague. Only one of the reported cases was diagnosed as septicemic plague while the remaining 207 cases remain unclassified [94]. Control and prevention of plague cases in Madagascar has proven to be challenging. Most of the prevention strategies are focused on the rodent host and the flea vectors [91]. Inappropriate use of chemical insecticides inadvertently selects for flea resistance, which leads to the survival of Y. pestis-infected fleas [91]. Food handling/storage procedures, community sanitation standards, and environmental factors of the household and its surroundings are parameters that can increase human interactions with rodent reservoirs [96]. Importantly, distribution of supplies and equipment including hand washing facilities, sanitation standards, disinfectants (e.g., laundry soap, chlorine powder, disinfectant sprays), and personal protective equipment contribute to an efficient plague prevention strategy [94].

4. Current Antibiotic Therapies

Due to the rapid onset and rather non-descript disease characteristics of plague, the key to positive patient outcomes, particularly after infection with aerosolized bacteria, is early diagnosis and rapid implementation of appropriate medical countermeasures. The gold-standard for plague diagnostics continues to be culture-positive samples (normally blood or sputum samples) but other molecular diagnostic assays have been used or continue to be developed [2].
If diagnosed accurately and early after infection, human plague cases can be controlled by the appropriate administration of antimicrobial drugs, including aminoglycoside, tetracyclines, fluroquinolones, and sulfonamides [91,97,98,99,100,101]. Generally, successful treatment of Y. pestis infections requires early recognition and the administration of an effective antibiotic during the first 24 h after the onset of symptoms that often present within 24 to 48 h post-infection [18]. According to the United States Centers for Disease Control and Prevention (CDC), the majority of human plague cases can be treated successfully with antibiotics [18]. The most effective antibiotics against Y. pestis are aminoglycosides, such as streptomycin and gentamicin, and accordingly, streptomycin is the first-line antibiotic for treatment of plague in most cases; both can be administered and are recommended for all adults, including pregnant women, immunocompromised patients, and children (although reduced dosages may be warranted in some cases) [102]. Chloramphenicol is another suitable agent for bubonic or septicemic plague and can be administered in conjunction with aminoglycosides [18,102]. Doxycycline and tetracycline are acceptable alternate antimicrobial agents as primary treatment for patients with uncomplicated plague and can also be used in conjunction with other antibiotics or in patients intolerant of aminoglycosides [18]. Fluoroquinolones, such as ciprofloxacin, have been demonstrated to have pharmacokinetic properties that make them suitable for plague therapy. Ciprofloxacin possesses bactericidal activity and its efficacy has been tested in vitro, in vivo, and in patients with bubonic plague [103,104]. Individuals in close contact with people afflicted with pneumonic plague, exposed to Y. pestis infected fleas, or who have been handling body fluids or tissues infected with Y. pestis should receive prophylactic antibiotic therapy.
The emergence of multidrug-resistant (MDR) strains of bacterial pathogens, including Y. pestis, is one of the most critical issues facing public health due to the difficulty in treatment, the high cost associated with medical care (particularly in developing countries) and increased mortality rates associated with the drug-resistant phenotypes [105]. This increased incidence of antibiotic resistance is likely due to the close proximity between humans and rodents, the extensive use of antibiotics in animal husbandry, and/or the presence of antibiotics in contaminated hospital waste. In addition to being the most recent epicenter of plague infections, Madagascar is also a focus for MDR Y. pestis strains. In 1995, two different strains of naturally occurring antibiotic-resistant Y. pestis were isolated in Madagascar. The first one, Y. pestis 17/95 was isolated from a 16-year-old boy, and the second strain 16/95 was isolated from a 14-year-old boy; both cases were diagnosed with bubonic plague [97,106,107]. Galimand et al. determined that strain 17/95 isolated from the city of Ambalavao was resistant to eight antimicrobial agents, including those for therapy and prophylaxis—while strain 16/95, isolated from the city of Ambohimahasoa, demonstrated high levels of streptomycin-resistance [97,106]. The multiple antibiotic resistance genes in both MDR strains of Y. pestis were all located on plasmids belonging to different incompatibility (Inc) groups [108]. Y. pestis strain 17/95 encoded MDR genes on pIP1202 in the Inc6-C group of plasmids, while Y. pestis strain 16/95 harbored MDR genes on pIP1203, which belongs to the IncP group of plasmids [97]. In 1998, a novel doxycycline-resistant strain was isolated from the spleen of a rat (Rattus norvegicus) in Antananavo, Madagascar [109]. To date, these characterized MDR Y. pestis strains were isolated from distinct hosts, at different times, and from distant locations [109]. In addition, Cabanel, et al., 2018 cited that independent horizontal transfer may be the reason for unrelated plasmids among these MDR strains. Other drug-resistant strains and antimicrobial resistance mechanisms have been reported and all aspects of drug-resistance in Y. pestis must remain an important research priority [110,111].

5. Vaccines

Due to the relative ease of transmissibility, rapid course of disease, non-descript clinical signs and symptoms, high mortality, and antibiotic resistance potential, effective vaccine strategies are needed. An effective and safe plague vaccine is important from a public health perspective but also in context of national biodefense strategies [2,33,112,113]. The first plague vaccines, developed late in the 19th century, consisted of killed whole cells of Y. pestis [114].
An immunogenic and somewhat less-reactogenic licensed vaccine (USP) containing a formalin-killed highly virulent 195/P strain of Y. pestis, was effective in preventing or ameliorating bubonic disease, as seen by the low incidence of plague cases in military personnel serving in Vietnam. However, the extent of efficacy remains in question, and it is thought that the protection was almost entirely based upon titers to the F1 capsular antigen [115,116,117,118]. In vivo data suggested that this vaccine might not offer optimal protection against pneumonic plague [119,120,121,122,123,124,125]. Such vaccines may not protect against genetically engineered or naturally occurring F1-negative strains, which often maintain significant virulence despite the loss of capsule. The killed whole-cell preparation vaccines, in the absence of frequent boosting, failed to provide long-term protection against bubonic plague.
Live bacterial vaccines are protective, and in many cases, contain a nearly complete array of native antigens—thereby reducing the chances of breakthrough infections. However, live attenuated vaccines are also considered more reactogenic than other vaccination approaches, and may cause safety concerns in certain subsections of the community (e.g., elderly or immune-compromised), and depending upon the vaccination strategy may elicit only short-term immunity [114]. Candidate live vaccines have included recombinant Y. pestis, Y. pseudotuberculosis, and Salmonella strains [126,127,128].
Live attenuated Y. pestis vaccines, such as the EV strain (derived and attenuated in the 1920s by serial passaging a virulent Y. pestis parent strain isolated from a patient identified as EV), have been in use in various parts of the world for decades [129]. Evidence demonstrated that these live vaccine strategies were able to protect against pneumonic and bubonic plague and induced high antibody titers [119,130]. Unfortunately, these vaccines may have severe side effects and systemic reactions in non-human primates [119] and humans [131], and only induce short-lived protection that require annual boosters [132,133].
Recently, additional live attenuated vaccine strategies using attenuated Y. pestis strains are also being developed and their corresponding immune responses evaluated [134,135]. Bubeck and Dube achieved significant protection when using a ΔyopH live vaccine strain [136] and Bozue et al., demonstrated that a Y. pestis ΔyscN mutant strain also offered significant protection against challenge with a fully virulent strain of Y. pestis [134,137]. Follow-on studies further characterized the vaccine potential of ΔyscN strains of Y. pestis as well as a strain lacking both the pigmentation (pgm) locus and the pPst virulence plasmid [138,139,140,141]. In addition, promising results were obtained with Y. pseudotuberculosis based live vaccines that conferred protection and immunological memory [142,143,144,145].
Besides whole-cell based vaccines, considerable work on subunit vaccine strategies has been completed. With recombinant DNA technology and improved protein chemistry, antigens can be purified for subunit vaccine development. Protein-based subunit vaccines have the potential of increased stability and consistency between vaccine preparations along with reduction in adverse effects that are often correlated with live vaccines [146]. Both the United States and the United Kingdom have focused attention on the development of subunit vaccines based on the fraction 1 (F1) capsular antigen and the low calcium response protein V (LcrV) antigens [123,147,148] (Figure 2). As discussed earlier, Y. pestis expresses a capsule antigen F1, encoded by the caf1 gene, which has been shown to have significant antiphagocytic activity [51,149,150,151,152,153]. LcrV is a secreted virulence protein which is essential for survival in the host, acting as an immunosuppressive factor [65]. Early work demonstrated a protective effect associated with active vaccination with V fractions [154,155,156,157]. Several groups reported that anti-V antibodies may be protective by promoting phagocytosis of bacteria and reducing bacteria-induced cytotoxicity [158,159,160]. In addition to being a protective antigen, LcrV was later shown to be a multi-factorial protein that is an important structural component of the T3SS (injectisome) and a secreted protein that is trafficked into eukaryotic cells; furthermore, the LcrV antigen can regulate aspects of the host immune response extracellularly by induction of anti-inflammatory IL-10 [65,66,67,68,69,70,71,72,73,74,75,76,156]. Unfortunately, important differences have been documented between LcrV proteins isolated from various Y. pestis strains which may hamper protective efficacy of subunit vaccines that rely on a specific polymorph for induction of immunological protection but may have a limited cross-reactive response [161]. Nevertheless, both F1 and V have been studied as components of subunit vaccines and have been shown to confer significant protection against bubonic and pneumonic plague induced by encapsulated strains of Y. pestis, with few documented concerns about tolerability or safety [155,157,162,163,164,165].
The primary subunit vaccine candidate in the United States is the recombinant F1-V, a fusion protein of the F1 capsular antigen and the lcrV gene product [166,167,168,169,170]. The United Kingdom pursued a similar strategy for vaccination but retained the F1 and V immunogens as separate proteins [171,172,173]. PharmAthene further advanced this concept and developed a recombinant dual antigen vaccine for plague, composed of rF1 + rV, named RypVax™ [174,175]. A truncated LcrV antigen, rV10, developed by Schneewind and colleagues in 2011 was also assessed [176]. Both vaccines, rF1 + rV and rV10, were tested and demonstrated efficacy against pneumonic plague infection in mice, guinea pigs and Cynomolgus macaques [176,177] but not in African green monkeys [125]. Further investigations of enhancing immunogenicity or delivery of these subunit vaccines have been attempted or are ongoing by several groups [178,179,180,181,182,183,184,185].
The two-component vaccines have been variably effective and are vulnerable to breakthrough infection. Researchers found that the combination of these subunits significantly enhances protection against bubonic and pneumonic plague in different animal models [176,186,187]. The protection afforded by this vaccine strategy against F1-negative strains relies entirely on the LcrV antigen component of the F1-V fusion protein. Since there is evidence for V heterogeneity within Yersinia species, the potential exists that naturally occurring or engineered strains harboring altered LcrV antigens could overcome F1-V-induced immunity [161].
Other vaccine strategies have attempted to identify vaccine antigens that would protect animals against non-encapsulated strains. Andrews demonstrated that active vaccination against YopD could protect mice against an F1-negative strain of Y. pestis [188]. Later studies by Ivanov et al. further characterized the protective efficacy of YopD and created fusion proteins of YopBD and YopBDE [189]. Ivanov et al. achieved protection with both active and passive vaccination strategies using these proteins, but these data suggested that anti-Yop immune responses were most protective against non-encapsulated Y. pestis [189]. While there has been progress in vaccine development, the need remains for other reliably protective and readily deployable countermeasures against plague.

6. Antibodies

There has been significant interest in and effort towards identifying antibody therapies in animal models of plague. In 1963, Lawton and coworkers identified protection associated with anti-LcrV polyclonal serum passively administered to mice. Importantly, it was determined that the protection associated with this polyclonal serum could not be completely attributable to the LcrV antigen due to the likelihood of other Yersinia proteins present in the material used to generate the sera [190]. Later, Motin et al. demonstrated with more modern techniques that a highly purified preparation of the V protein could be produced and used for polyclonal sera generation [190]. These anti-sera directed against native LcrV antigen or recombinant LcrV antigens were able to provide substantial passive protection against Y. pestis infection.
Table 1 and Table 2 and the following paragraphs provide summary information extracted from noteworthy publications describing particularly effective monoclonal antibodies in mouse models of plague. Hill et al. demonstrated that a monoclonal antibody directed against the LcrV could protect against a fully virulent strain of Y. pestis and went on to demonstrate which regions of the LcrV antigen appeared to be protective epitopes [156]. Quenee et al. and Amemiya et al. further elaborated on the importance of the specific binding sites on protective anti-LcrV monoclonal antibodies [191,192]. The mAb 7.3 was shown to protect macrophages from Y. pestis-induced cell death and also to promote phagocytosis of the bacteria [158,193]. Importantly, much of the protection afforded by mAb 7.3 can be attributed to the blocking of the T3SS machinery. When anti-V antibodies block this injectisome, the immunomodulation and anti-phagocytic Yop effectors are not efficiently secreted, thus blocking cytotoxic phenotypes and promoting phagocytosis of the bacterial cells [158]. This concept was further supported by Eisele and Anderson when they demonstrated that both blocking the T3SS as well as inducing phagocytosis are required from an anti-LcrV mAb in order to optimally protect against pneumonic plague in a mouse model of disease [194]. Novel Adenovirus-mediated delivery has also been used to demonstrate the protection associated with anti-LcrV antigen mAbs [195].
Partly due to the known heterogeneity amongst LcrV protein sequences from different bacterial isolates [161,196,197,198], a multi-antigen strategy was postulated to be required to optimize protection. Anderson et al. also demonstrated that it was possible to passively protect mice against either bubonic or pneumonic plague [199] with anti-F1 antibodies. These anti-F1 monoclonal antibodies, however, would not protect against F1-negative (non-encapsulated) strains of Y. pestis (Figure 2). Since immunity to F1 is not sufficient to protect in all cases, there was renewed focus on the incorporation of the LcrV antigen or other proteins for multifactorial strategies.
Combinations of F1 and LcrV monoclonal antibodies were shown to protect mice against both bubonic and pneumonic plague either prophylactically or as post-exposure therapy [200,201]. More recent efforts have continued to examine strategies combining anti-F1 and anti-LcrV antibodies [202]. Xiao et al. identified three human mAbs: m252 for anti-F1, and m253 and m254 for anti-LcrV. They found that anti-F1 human mAb (m252) provided better protection in mice than anti-LcrV human mAbs. However, an apparent synergistic effect was found when they combined all three antibodies. Another three anti-F1 (F5C10, F6E5, and F2H5) mAbs were tested against subcutaneous challenge with Y. pestis 141 and showed different protection levels. Liu et al. demonstrated that mAb F2H5 from a mouse hybridoma provides complete protection from bubonic plague in BALB/c mice [203]. Altogether, it would be feasible that these mAbs against F1 or LcrV can be utilized as a potential treatment or prophylaxis for humans against plague. Additional anti-Y. pestis human antibodies have recently been produced and are currently being evaluated for therapeutic application [204].

7. Advantages of Monoclonal Antibodies (mAbs) and Final Perspectives

Currently, there are only a few mAbs licensed for use against bacterial infectious agents. To date, mAb against Clostridium difficile and Bacillus anthracis toxins are licensed in Europe and the USA [205,206,207]. Other work has focused on mAbs against Staphylococcus aureus or Pseudomonas aeruginosa [208]. The renewal of interest in mAb therapies is even more relevant given the current era of worrisome multidrug resistance in bacterial isolates of clinical importance. Several clinically relevant bacteria such as Burkholderia pseudomallei and Burkholderia mallei are naturally resistant to many antibiotics [209]. Examples of emerging MDR bacteria include but are not limited to S. aureus, C. difficile, P. aeruginosa, Acinetobacter baumeii, Enterococci, etc [210,211,212,213]. This MDR trait has been observed with Y. pestis, and the lack of effective therapies for such an infection could result in catastrophic loss of life, whether from a naturally emerging outbreak or an intentional attack utilizing purposefully engineered bacteria.
Recent strategies have focused on nimble and rapid platforms that can be leveraged when combatting outbreaks and emerging threats. Advances in rapid mAb discovery, optimization, and production make this an attractive strategy that could significantly decrease the time required to get a product to civilian populations in the midst of an outbreak or to military personnel deployed to areas where endemic disease could be an issue and an effective vaccine has yet to be identified. Other more novel strategies involve administering nucleic acids that encode the protective mAb to patients. In that case, a nucleic acid encoding the immunoglobulin may be manufactured or obtained from a cDNA library or nucleic acid isolated from any tissue or cells expressing the antibody. These nucleic acids can be cloned into a suitable vector and administered into an infected patient [202].
These concepts have been useful recently during West African Ebola virus outbreaks [214]. In this epidemic, mAbs were demonstrated to be important novel treatment strategies. Products such as ZMapp® and others were critical in the response to the Ebola virus epidemic [214]. Viruses of recent or continual concern including HIV, Zika, and COVID-19 have also been targets of these novel strategies [215,216,217,218,219,220]. Monoclonal antibodies will likely be of the utmost importance moving forward when developing novel medical countermeasures against existing and rapidly emerging infectious diseases.

Funding

This review article was written with funding provided by the U.S. Defense Threat reduction Agency (DTRA) CB10392.

Acknowledgments

We thank William Discher for assistance with illustrations. Opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endorsed by the U.S. Army.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pechous, R.D.; Sivaraman, V.; Stasulli, N.M.; Goldman, W.E. Pneumonic plague: The darker side of Yersinia pestis. Trends Microbiol. 2016, 24, 190–197. [Google Scholar] [CrossRef] [PubMed]
  2. Demeure, C.E.; Dussurget, O.; Fiol, G.M.; Le Guern, A.S.; Savin, C.; Pizzarro-Cerdá, J. Yersinia pestis and plague: An updated view on evolution, virulence determinants, immune subversion, vaccination, and diagnostics. Genes Immun. 2019, 20, 357–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Stenseth, N.C.; Atshabar, B.B.; Begon, M.; Belmain, S.R.; Bertherat, E.; Carniel, E.; Gage, K.L.; Leirs, H.; Rahalison, L. Plague: Past, present, and future. PLoS Med. 2008, 5, e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Achtman, M.; Zurth, K.; Morelli, G.; Torrea, G.; Guiyoule, A.; Carniel, E. Yersinia pestis, the cause of plague, is a recently emerged clone of Yersinia pseudotuberculosis. Proc. Natl. Acad. Sci. USA 1999, 96, 14043–14048. [Google Scholar] [CrossRef] [Green Version]
  5. Glatter, K.A.; Finkelman, P. History of the plague: An ancient pandemic for the age of COVID-19. Am. J. Med. 2021, 134, 176–181. [Google Scholar] [CrossRef]
  6. Brubaker, R.R. Yersinia pestis. Mol. Med. Microbiol. 2015, 3, 1845–1865. [Google Scholar]
  7. Frith, J. The history of plague—Part 1. The three great pandemics. J. Mil. Vet. Health 2012, 20, 11–16. [Google Scholar]
  8. Barbieri, R.; Signoli, M.; Chevé, D.; Costedoat, C.; Tzortzis, S.; Aboudharam, G.; Raoult, D.; Drancourt, M. Yersinia pestis: The natural history of plague. Clin. Microbiol. Rev. 2020, 34, e00044-19. [Google Scholar] [CrossRef]
  9. Drancourt, M.; Raoult, D. Molecular history of plague. Clin. Microbiol. Infect. 2016, 22, 911–915. [Google Scholar] [CrossRef] [Green Version]
  10. Bramanti, B.; Dean, K.R.; Walløe, L.; Stenseth, N.C. The third plague pandemic in Europe. Proc. Biol. Sci. 2019, 286, 20182429. [Google Scholar] [CrossRef]
  11. Eisen, R.J.; Gage, K.L. Adaptive strategies of Yersinia pestis to persist during inter-epizootic and epizootic periods. Vet. Res. 2009, 40, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Raoult, D.; Mouffok, N.; Bitam, I.; Piarroux, R.; Drancourt, M. Plague: History and contemporary analysis. J. Infect. 2013, 66, 18–26. [Google Scholar] [CrossRef] [PubMed]
  13. Melman, S.D.; Ettestad, P.E.; VinHatton, E.S.; Ragsdale, J.M.; Takacs, N.; Onischuk, L.M.; Leonard, P.M.; Master, S.S.; Lucero, V.S.; Kingry, L.C. Human case of bubonic plague resulting from the bite of a wild Gunnison’s prairie dog during translocation from a plague-endemic area. Zoonoses Public Health 2018, 65, e254–e258. [Google Scholar] [CrossRef] [Green Version]
  14. Dai, R.; Wei, B.; Xiong, H.; Yang, X.; Peng, Y.; He, J.; Jin, J.; Wang, Y.; Zha, X.; Zhang, Z.; et al. Human plague associated with Tibetan sheep originates in marmots. PLoS Negl. Trop. Dis. 2018, 12, e0006635. [Google Scholar] [CrossRef] [Green Version]
  15. Richgels, K.L.; Russell, R.E.; Bron, G.M.; Rocke, T.E. Evaluation of Yersinia pestis transmission pathways for sylvatic plague in prairie dog populations in the Western U.S. Ecohealth 2016, 13, 415–427. [Google Scholar] [CrossRef] [PubMed]
  16. CDC. Fatal laboratory-acquired infection with an attenuated Yersinia pestis Strain—Chicago, Illinois, 2009. MMWR Morb. Mortal. Wkly. Rep. 2011, 60, 201–205. [Google Scholar]
  17. Worsham, P.L.; McGovern, T.W.; Vietri, N.J.; Friedlander, A.M.; Bozue, J. Plague. In Textbook of Military Medicine: Medical Aspects of Biological Warfare; Bozue, J.A., Cote, C.K., Glass, P.J., Eds.; Borden Institute US Army Medical Department Center and School Health Readiness Center of Excellence: Houston, TX, USA, 2018; pp. 247–284. [Google Scholar]
  18. Yang, R. Plague: Recognition, treatment, and prevention. J. Clin. Microbiol. 2017, 56, e01519-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Nikiforov, V.V.; Gao, H.; Zhou, L.; Anisimov, A. Plague: Clinics, diagnosis and treatment. Adv. Exp. Med. Biol. 2016, 918, 293–312. [Google Scholar]
  20. Dennis, D.T.; Gage, K.L.; Gratz, N.G.; Poland, J.D.; Tikhomirov, E. Plague Manual: Epidemiology, Distribution, Surveillance and Control; No. WHO/CDS/CSR/EDC/99.2; World Health Organization: Geneva, Switzeralnd, 1999. [Google Scholar]
  21. Sebbane, F.; Lemaitre, N. Antibiotic therapy of plague: A review. Biomolecules 2021, 11, 724. [Google Scholar] [CrossRef]
  22. Bossi, P.; Tagnell, A.; Baka, A.; Loock, F.V.; Hendriks, J.; Werner, A.; Maidhof, H.; Gouvras, G. Bichat guidelines for the clinical management of plague and bioterrorism-related plague. Euro Surveill 2004, 9, E5–E6. [Google Scholar] [CrossRef]
  23. Lemaître, N.; Ricard, I.; Pradel, E.; Foligné, B.; Courcol, R.; Simonet, M.; Sebbane, F. Efficacy of ciprofloxacin-gentamicin combination therapy in murine bubonic plague. PLoS ONE 2012, 7, e52503. [Google Scholar] [CrossRef] [Green Version]
  24. Boulanger, L.L.; Ettestad, P.; Fogarty, J.D.; Dennis, D.T.; Romig, D.; Mertz, G. Gentamicin and tetracyclines for the treatment of human plague: Review of 75 cases in New Mexico, 1985–1999. Clin. Infect. Dis. 2004, 38, 663–669. [Google Scholar] [CrossRef]
  25. Lawrenz, M.B. Model systems to study plague pathogenesis and develop new therapeutics. Front. Microbiol. 2010, 1, 119. [Google Scholar] [CrossRef] [Green Version]
  26. Dillard, R.L.; Juergens, A.L. Plague; StatPearls: Treasure Island, FL, USA, 2020. [Google Scholar]
  27. Cleri, D.J.; Marton, R.; Rabbat, M.; Vernaleo, J. Pneumonia Caused by Yersinia pestis: Plague Pneumonia, in Community-Acquired Pneumonia; Marrie, T.J., Ed.; Springer: Boston, MA, USA, 2001; pp. 777–799. [Google Scholar]
  28. Kool, J. Risk of person-to-person transmission of pneumonic plague. Clin. Infect. Dis. 2005, 40, 1166–1172. [Google Scholar] [CrossRef] [PubMed]
  29. Dennis, D.T.; Mead, P.S. Plague. Trop. Infect. Dis. 2006, 1, 471–481. [Google Scholar]
  30. Gage, K.L.; Beard, C.B. 126—Plague. In Infectious Diseases, 4th ed.; Cohen, J., Powderly, W.G., Opal, S.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1078–1084.e1. [Google Scholar]
  31. Wagar, E. Bioterrorism and the role of the clinical microbiology laboratory. Clin. Microbiol. Rev. 2016, 29, 175–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ansari, I.; Grier, G.; Byers, M. Deliberate release: Plague—A review. J. Biosaf. Biosecur. 2020, 2, 10–22. [Google Scholar] [CrossRef]
  33. Du, Z.; Wang, X. Pathology and pathogenesis of Yersinia pestis. Adv. Exp. Med. Biol. 2016, 918, 193–222. [Google Scholar]
  34. Hewitt, J.A.; Lanning, L.L.; Campbell, J.L. The African green monkey model of pneumonic plague and US Food and Drug Administration approval of antimicrobials under the animal rule. Clin. Infect. Dis. 2020, 70 (Suppl. S1), S51–S59. [Google Scholar] [CrossRef]
  35. Warren, R.; Lockman, H.; Barnewall, R.; Krile, R.; Bermeo Blanco, O.; Vasconcelos, D.; Price, J.; House, R.V.; Bolanowksi, M.A.; Fellows, P. Cynomolgus macaque model for pneumonic plague. Microb. Pathog. 2011, 50, 12–22. [Google Scholar] [CrossRef]
  36. Bubeck, S.S.; Cantwell, A.M.; Dube, P.H. Delayed inflammatory response to primary pneumonic plague occurs in both outbred and inbred mice. Infect. Immun. 2007, 75, 697–705. [Google Scholar] [CrossRef] [Green Version]
  37. Lathem, W.W.; Crosby, S.D.; Miller, V.L.; Goldman, W.E. Progression of primary pneumonic plague: A mouse model of infection, pathology, and bacterial transcriptional activity. Proc. Natl. Acad. Sci. USA 2005, 102, 17786–17791. [Google Scholar] [CrossRef] [Green Version]
  38. Pechous, R.D.; Sivaraman, V.; Price, P.A.; Stasulli, N.M.; Goldman, W.E. Early host cell targets of Yersinia pestis during primary pneumonic plague. PLoS Pathog. 2013, 9, e1003679. [Google Scholar] [CrossRef]
  39. Banerjee, S.K.; Crane, S.D.; Pechous, R.D. A dual role for the plasminogen activator protease during the preinflammatory phase of primary pneumonic plague. J. Infect. Dis. 2020, 222, 407–416. [Google Scholar] [CrossRef] [PubMed]
  40. Pechous, R.D.; Broberg, C.A.; Stasulli, N.M.; Miller, V.L.; Goldman, W.E. In vivo transcriptional profiling of Yersinia pestis reveals a novel bacterial mediator of pulmonary inflammation. mBio 2015, 6, e02302-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Zhou, D.; Yang, R. Molecular Darwinian evolution of virulence in Yersina pestis. Infect. Immun. 2020, 77, 2242–2250. [Google Scholar] [CrossRef] [Green Version]
  42. Skurnik, M.; Peippo, A.; Ervelä, E. Characterization of the O-antigen gene clusters of Yersinia pseudotuberculosis and the cryptic O-antigen gene cluster of Yersinia pestis shows that the plague bacillus is most closely related to and has evolved from Y. pseudotuberculosis serotype O:1b. Mol. Microbiol. 2000, 37, 316–330. [Google Scholar] [CrossRef] [PubMed]
  43. Rudolph, A.E.; Stuckey, J.A.; Zhao, Y.; Matthews, H.R.; Patton, W.A.; Moss, I.; Dixon, J.E. Expression, characterization, and mutagenesis of the Yersinia pestis murine toxin, a phospholipase D superfamily member. J. Biol. Chem. 1999, 274, 11824–11831. [Google Scholar] [CrossRef] [Green Version]
  44. Schar, M.; Meyer, K.F. Studies on immunization against plague. XV. The pathophysiologic action of the toxin of Pasteurella pestis in experimental animals. Schweiz Z Pathol. Bakteriol. 1956, 19, 51–70. [Google Scholar]
  45. Bergdoll, M.S.; Enterotoxins, T.C.; Montie, S.J.A. Microbial Toxins; Academic Press: New York, NY, USA, 1970; Volume 3, pp. 265–326. [Google Scholar]
  46. Hinnebusch, B.J.; Rudolph, A.E.; Cherepanov, P.; Dixon, J.E.; Schwan, T.G. Role of Yersinia murine toxin in survival of Yersinia pestis in the midgut of the flea vector. Science 2002, 296, 733–735. [Google Scholar] [CrossRef]
  47. Chen, T.H.; Crocker, T.T.; Meyer, K.F. Electron microscopic study of the extracellular materials of Pasteurella pestis. J. Bacteriol. 1956, 72, 851–857. [Google Scholar]
  48. Davis, K.J.; Fritz, D.L.; Pitt, M.L.; Welkos, S.L.; Worsham, P.L.; Friedlander, A.M. Pathology of experimental pneumonic plague produced by fraction 1-positive and fraction 1-negative Yersinia pestis in African green monkeys (Cercopithecus aethiops). Arch. Pathol. Lab. Med. 1996, 120, 156–163. [Google Scholar]
  49. Engelsberg, E.; Levy, J.B. Studies on immunization against plague. VI. Growth of Pasteurella pestis and the production of the envelope and other soluble antigens in a casein hydrolyzate mineral glucose medium. J. Bacteriol. 1954, 67, 438–449. [Google Scholar] [CrossRef] [Green Version]
  50. Runco, L.M.; Myrczek, S.; Bliska, J.B.; Thanassi, D.G. Biogenesis of the fraction 1 capsule and analysis of the ultrastructure of Yersinia pestis. J. Bacteriol. 2008, 190, 3381–3385. [Google Scholar] [CrossRef] [Green Version]
  51. Du, Y.; Rosqvist, R.; Forsberg, Å. Role of fraction 1 antigen of Yersinia pestis in inhibition of phagocytosis. Infect. Immun. 2002, 70, 1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Welkos, S.L.; Davis, K.M.; Pitt, L.M.; Worsham, P.L.; Friedlander, A.M. Studies on the contribution of the F1 capsule-associated plasmid pFra to the virulence of Yersinia pestis. Contrib. Microbiol. Immunol. 1995, 13, 299–305. [Google Scholar] [PubMed]
  53. Winter, C.C.; Cherry, W.B.; Moody, M.D. An unusual strain of Pasteurella pestis isolated from a fatal human case of plague. Bull. World Health Organ. 1960, 23, 408–409. [Google Scholar]
  54. Sebbane, F.; Uversky, V.N.; Anisimov, A.P. Yersinia pestis plasminogen activator. Biomolecules 2020, 10, 1554. [Google Scholar] [CrossRef] [PubMed]
  55. Caulfield, A.J.; Lathem, W.W. Substrates of the plasminogen activator protease of Yersinia pestis. Adv. Exp. Med. Biol. 2012, 954, 253–260. [Google Scholar]
  56. Degen, J.L.; Bugge, T.H.; Goguen, J.D. Fibrin and fibrinolysis in infection and host defense. J. Thromb. Haemost. 2007, 5 (Suppl. S1), 24–31. [Google Scholar] [CrossRef]
  57. Sodeinde, O.A.; Subrahmanyam, Y.V.; Stark, K.; Quan, T.; Bao, Y.; Goguen, J.D. A surface protease and the invasive character of plague. Science 1992, 258, 1004–1007. [Google Scholar] [CrossRef]
  58. Caulfield, A.J.; Walker, M.E.; Gielda, L.M.; Lathem, W.W. The Pla protease of Yersinia pestis degrades fas ligand to manipulate host cell death and inflammation. Cell Host Microbe 2014, 15, 424–434. [Google Scholar] [CrossRef] [Green Version]
  59. Lathem, W.W.; Price, P.A.; Miller, V.L.; Goldman, W.E. A plasminogen-activating protease specifically controls the development of primary pneumonic plague. Science 2007, 315, 509–513. [Google Scholar] [CrossRef]
  60. Kienle, Z.; Emödy, L.; Svanborg, C.; O’Toole, P.W. Adhesive properties conferred by the plasminogen activator of Yersinia pestis. J. Gen. Microbiol. 1992, 138, 1679–1687. [Google Scholar] [CrossRef] [Green Version]
  61. Lähteenmäki, K.; Virkola, R.; Sarén, A.; Emödy, L.; Korhonen, T.K. Expression of plasminogen activator Pla of Yersinia pestis enhances bacterial attachment to the mammalian extracellular matrix. Infect. Immun. 1998, 66, 5755–5762. [Google Scholar] [CrossRef] [Green Version]
  62. Lobo, L.A. Adhesive properties of the purified plasminogen activator Pla of Yersinia pestis. FEMS Microbiol. Lett. 2006, 262, 158–162. [Google Scholar] [CrossRef] [Green Version]
  63. Cowan, C.; Jones, H.A.; Kaya, Y.H.; Perry, R.D.; Straley, S.C. Invasion of epithelial cells by Yersinia pestis: Evidence for a Y. pestis-specific invasin. Infect. Immun. 2000, 68, 4523–4530. [Google Scholar] [CrossRef] [Green Version]
  64. Lähteenmäki, K.; Kukkonen, M.; Korhonen, T.K. The Pla surface protease/adhesin of Yersinia pestis mediates bacterial invasion into human endothelial cells. FEBS Lett. 2001, 504, 69–72. [Google Scholar] [CrossRef] [Green Version]
  65. Fields, K.A.; Straley, S.C. LcrV of Yersinia pestis enters infected eukaryotic cells by a virulence plasmid-independent mechanism. Infect. Immun. 1999, 67, 4801–4813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Derewenda, U.; Mateja, A.; Devedjiev, Y.; Routzahn, K.M.; Evdokimov, A.G.; Derewenda, Z.S.; Waugh, D.S. The structure of Yersinia pestis V-antigen, an essential virulence factor and mediator of immunity against plague. Structure 2004, 12, 301–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Price, S.B.; Cowan, C.; Perry, R.D.; Straley, S.C. The Yersinia pestis V antigen is a regulatory protein necessary for Ca2(+)-dependent growth and maximal expression of low-Ca2+ response virulence genes. J. Bacteriol. 1991, 173, 2649–2657. [Google Scholar] [CrossRef] [Green Version]
  68. Hamad, M.A.; Nilles, M.L. Structure-function analysis of the C-terminal domain of LcrV from Yersinia pestis. J. Bacteriol. 2007, 189, 6734–6739. [Google Scholar] [CrossRef] [Green Version]
  69. DiMezzo, T.L.; Ruthel, G.; Brueggemann, E.E.; Hines, H.B.; Ribot, W.J.; Chapman, C.E.; Powell, B.S.; Welkos, S.L. In vitro intracellular trafficking of virulence antigen during infection by Yersinia pestis. PLoS ONE 2009, 4, e6281. [Google Scholar] [CrossRef] [PubMed]
  70. Sing, A.; Roggenkamp, A.; Geiger, A.M.; Heesemann, J. Yersinia enterocolitica evasion of the host innate immune response by V antigen-induced IL-10 production of macrophages is abrogated in IL-10-deficient mice. J. Immunol. 2002, 168, 1315–1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Perry, R.D.; Haddix, P.; Atkins, E.B.; Soughers, T.K.; Straley, S.C. Regulation of expression of V antigen and outer membrane proteins in Yersinia pestis. Contrib. Microbiol. Immunol. 1987, 9, 173–178. [Google Scholar] [PubMed]
  72. Perry, R.D.; Harmon, P.A.; Bowmer, W.S.; Straley, S.C. A low-Ca2+ response operon encodes the V antigen of Yersinia pestis. Infect. Immun. 1986, 54, 428–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nilles, M.L.; Williams, A.W.; Skrzypek, E.; Straley, S.C. Yersinia pestis LcrV forms a stable complex with LcrG and may have a secretion-related regulatory role in the low-Ca2+ response. J. Bacteriol. 1997, 179, 1307–1316. [Google Scholar] [CrossRef] [Green Version]
  74. Fields, K.A.; Nilles, M.L.; Cowan, C.; Straley, S.C. Virulence role of V antigen of Yersinia pestis at the bacterial surface. Infect. Immun. 1999, 67, 5395–5408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Philipovskiy, A.V.; Cowan, C.; Wulff-Strobel, C.R.; Burnett, S.H.; Kerschen, E.J.; Cohen, D.A.; Kaplan, A.M.; Straley, S.C. Antibody against V antigen prevents Yop-dependent growth of Yersinia pestis. Infect. Immun. 2005, 73, 1532–1542. [Google Scholar] [CrossRef] [Green Version]
  76. Nilles, M.L.; Fields, K.A.; Straley, S.C. The V antigen of Yersinia pestis regulates Yop vectorial targeting as well as Yop secretion through effects on YopB and LcrG. J. Bacteriol. 1998, 180, 3410–3420. [Google Scholar] [CrossRef] [Green Version]
  77. Rosqvist, R.; Forsberg, A.; Rimpiläinen, M.; Bergman, T.; Wolf-Watz, H. The cytotoxic protein YopE of Yersinia obstructs the primary host defence. Mol. Microbiol. 1990, 4, 657–667. [Google Scholar] [CrossRef]
  78. Bliska, J.B.; Black, D.S. Inhibition of the Fc receptor-mediated oxidative burst in macrophages by the Yersinia pseudotuberculosis tyrosine phosphatase. Infect. Immun. 1995, 63, 681–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Fällman, M.; Andersson, K.; Håkansson, S.; Magnusson, K.E.; Stendahl, O.; Wolf-Watz, H. Yersinia pseudotuberculosis inhibits Fc receptor-mediated phagocytosis in J774 cells. Infect. Immun. 1995, 63, 3117–3124. [Google Scholar] [CrossRef] [Green Version]
  80. Grosdent, N.; Maridonneau-Parini, I.; Paule Sory, M.; Cornelis, G.R. Role of Yops and adhesins in resistance of Yersinia enterocolitica to phagocytosis. Infect. Immun. 2002, 70, 4165–4176. [Google Scholar] [CrossRef] [Green Version]
  81. Visser, L.G.; Annema, A.; van Furth, R. Role of Yops in inhibition of phagocytosis and killing of opsonized Yersinia enterocolitica by human granulocytes. Infect. Immun. 1995, 63, 2570–2575. [Google Scholar] [CrossRef] [Green Version]
  82. Anderson, D.M.; Schneewind, O. Yersinia enterocolitica type III secretion: An mRNA signal that couples translation and secretion of YopQ. Mol. Microbiol. 1999, 31, 1139–1148. [Google Scholar] [CrossRef]
  83. Heitzinger, K.; Impouma, B.; Farham, B.L.; Hambion, E.L.; Lukoya, C.; Machingaidze, C.; Rakotonjanabelo, L.A.; Yao, M.; Diallo, B.; Djingarey, M.H.; et al. Using evidence to inform response to the 2017 plague outbreak in Madagascar: A view from the WHO African Regional Office. Epidemiol. Infect. 2018, 147, 1–5. [Google Scholar] [CrossRef] [Green Version]
  84. Nguyen, V.K.; Parra-Rojas, C.; Hernandez-Vargas, E.A. The 2017 plague outbreak in Madagascar: Data descriptions and epidemic modelling. Epidemics 2018, 25, 20–25. [Google Scholar] [CrossRef] [PubMed]
  85. Vallès, X.; Stenseth, N.C.; Demeure, C.; Horby, P.; Mead, P.S.; Cabanillas, O.; Ratsitorahina, M.; Rajerison, M.; Andrianaivoarimanana, V.; Ramasindrazana, B.; et al. Human plague: An old scourge that needs new answers. PLoS Negl. Trop. Dis. 2020, 14, e0008251. [Google Scholar] [CrossRef]
  86. Carniel, E. Plague today. Med. Hist. Suppl. 2008, 52, 115–122. [Google Scholar] [CrossRef] [Green Version]
  87. Bertherat, E. Plague around the world, 2010–2015. Wkly. Epidemiol. Rec. 2016, 91, 89–93. [Google Scholar]
  88. Bevins, S.N.; Baroch, J.A.; Nolte, D.L.; Zhang, M.; He, H. Yersinia pestis: Examining wildlife plague surveillance in China and the USA. Integr. Zool. 2012, 7, 99–109. [Google Scholar] [CrossRef] [PubMed]
  89. Alderson, J.; Quastel, M.; Wilson, E.; Bellamy, D. Factors influencing the re-emergence of plague in Madagascar. Emerg. Top. Life Sci. 2020, 4, 411–421. [Google Scholar] [PubMed]
  90. Randremanana, R.; Andrianaivoarimanana, V.; Nikolay, B.; Ramasindrazana, B.; Paireau, J.; Ten Bosch, Q.A.; Rakotondramanga, J.M.; Rahajandraibe, S.; Rahelinirina, S.; Rakotomanana, F.; et al. Epidemiological characteristics of an urban plague epidemic in Madagascar, August–November, 2017: An outbreak report. Lancet Infect. Dis. 2019, 19, 537–545. [Google Scholar] [CrossRef] [Green Version]
  91. Andrianaivoarimanana, V.; Piola, P.; Wagner, D.M.; Rakotomanana, F.; Maheriniaina, V.; Andrianalimanana, S.; Chanteau, S.; Rahalison, L.; Ratsitorahina, M.; Rajerison, M. Trends of human plague, Madagascar, 1998–2016. Emerg. Infect. Dis. 2019, 25, 220–228. [Google Scholar] [CrossRef] [Green Version]
  92. Rabaan, A.A.; Al-Ahmed, S.H.; Alsuliman, S.A.; Aldrazi, F.A.; Alfouzan, W.A.; Haque, S. The rise of pneumonic plague in Madagascar: Current plague outbreak breaks usual seasonal mould. J. Med. Microbiol. 2019, 68, 292–302. [Google Scholar] [CrossRef]
  93. Barbieri, R.; Drancourt, M.; Raoult, D. The role of louse-transmitted diseases in historical plague pandemics. Lancet Infect. Dis. 2021, 21, e17–e25. [Google Scholar] [CrossRef]
  94. WHO. Madagascar Plague Outbreak: External Situation; Report #14; WHO: Geneva, Switzerland, 2017. [Google Scholar]
  95. Bragazzi, N.L.; Mahroum, N. Google trends predicts present and future plague cases during the plague outbreak in Madagascar: Infodemiological study. JMIR Public Health Surveill 2019, 5, e13142. [Google Scholar] [CrossRef] [Green Version]
  96. Kilonzo, B.S.; Mvena, Z.S.; Machangu, R.S.; Mbise, T.J. Preliminary observations on factors responsible for long persistence and continued outbreaks of plague in Lushoto district, Tanzania. Acta Trop. 1997, 68, 215–227. [Google Scholar] [CrossRef]
  97. Galimand, M.; Guiyoule, A.; Gerbaud, G.; Rasoamanana, B.; Chanteau, S.; Carniel, E.; Courvalin, P. Multidrug resistance in Yersinia pestis mediated by a transferable plasmid. N. Engl. J. Med. 1997, 337, 677–680. [Google Scholar] [CrossRef] [PubMed]
  98. Godfred-Cato, S.; Cooley, K.M.; Fleck-Derderian, S.; Becksted, H.A.; Russell, Z.; Meaney-Delman, D.; Mead, P.S.; Nelson, C.A. Treatment of human plague: A systematic review of published aggregate data on antimicrobial efficacy, 1939–2019. Clin. Infect. Dis. 2020, 70 (Suppl. S1), S11–S19. [Google Scholar] [CrossRef] [PubMed]
  99. Kugeler, K.J.; Mead, P.S.; Campbell, S.B.; Nelson, C.A. Antimicrobial treatment patterns and illness outcome among United States patients with plague, 1942–2018. Clin. Infect. Dis. 2020, 70 (Suppl. S1), S20–S26. [Google Scholar] [CrossRef] [PubMed]
  100. Nelson, C.A.; Fleck-Derderian, S.; Cooley, K.M.; Meaney-Delman, D.; Becksted, H.A.; Russell, Z.; Renaud Bertherat, E.; Mead, P.S. Antimicrobial treatment of human plague: A systematic review of the literature on individual cases, 1937–2019. Clin. Infect. Dis. 2020, 70 (Suppl. S1), S3–S10. [Google Scholar] [CrossRef]
  101. Oyston, P.C.; Williamson, E.D. Prophylaxis and therapy of plague. Expert Rev. Anti-Infect. Ther. 2013, 11, 817–829. [Google Scholar] [CrossRef]
  102. Dennis, D.T. Plague as a Biological Weapon. In Bioterrorism and Infectious Agents: A New Dilemma for the 21st Century; Fong, I.W., Alibek, K., Eds.; Springer New York: New York, NY, USA, 2009; pp. 37–70. [Google Scholar]
  103. Wendte, J.M.; Ponnusamy, D.; Reiber, D.; Blair, J.L.; Clinkenbeard, K.D. In vitro efficacy of antibiotics commonly used to treat human plague against intracellular Yersinia pestis. Antimicrob. Agents Chemother. 2011, 55, 3752–3757. [Google Scholar] [CrossRef] [Green Version]
  104. Apangu, T.; Griffith, K.; Abaru, J.; Candini, G.; Apio, H.; Okoth, F.; Okello, R.; Kaggwa, J.; Acayo, S.; Ezama, G.; et al. Successful treatment of human plague with oral ciprofloxacin. Emerg. Infect. Dis. 2017, 23, 553–555. [Google Scholar] [CrossRef]
  105. Welch, T.J.; Fricke, W.F.; McDermott, P.F.; White, D.G.; Rosso, M.L.; Rasko, D.A.; Mammel, M.K.; Eppinger, M.; Rosovitz, M.L.; Wagner, D.; et al. Multiple antimicrobial resistance in plague: An emerging public health risk. PLoS ONE 2007, 2, e309. [Google Scholar] [CrossRef] [Green Version]
  106. Galimand, M.; Carniel, E.; Courvalin, P. Resistance of Yersinia pestis to antimicrobial agents. Antimicrob. Agents Chemother. 2006, 50, 3233–3236. [Google Scholar] [CrossRef] [Green Version]
  107. Guiyoule, A.; Gerbaud, G.; Buchrieser, C.; Galimand, M.; Rahalison, L.; Chanteau, S.; Couvalin, P.; Carniel, E. Transferable plasmid-mediated resistance to streptomycin in a clinical isolate of Yersinia pestis. Emerg. Infect. Dis. 2001, 7, 43–48. [Google Scholar] [CrossRef] [Green Version]
  108. Popowska, M.; Krawczyk-Balska, A. Broad-host-range IncP-1 plasmids and their resistance potential. Front. Microbiol. 2013, 4, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Cabanel, N.; Bouchier, C.; Rajerison, M.; Carniel, E. Plasmid-mediated doxycycline resistance in a Yersinia pestis strain isolated from a rat. Int. J. Antimicrob. Agents 2018, 51, 249–254. [Google Scholar] [CrossRef] [PubMed]
  110. Dai, R.; He, J.; Zha, X.; Wang, Y.; Zhang, X.; Gao, H.; Yang, X.; Li, J.; Xin, Y.; Wang, Y.; et al. A novel mechanism of streptomycin resistance in Yersinia pestis: Mutation in the rpsL gene. PLoS Negl. Trop. Dis. 2021, 15, e0009324. [Google Scholar] [CrossRef] [PubMed]
  111. Taitt, C.R.; Leski, T.A.; Chen, A.; Berk, K.L.; Dorsey, R.W.; Gregory, M.J.; Sozhamannan, S.; Frey, K.G.; Dutt, D.L.; Vora, G.J. A survey of antimicrobial resistance determinants in category a select agents, exempt strains, and near-neighbor species. Int. J. Mol. Sci. 2020, 21, 1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Smiley, S.T. Current challenges in the development of vaccines for pneumonic plague. Expert Rev. Vaccinines 2008, 7, 209–221. [Google Scholar] [CrossRef] [PubMed]
  113. Titball, R.W.; Williamson, E.D. Vaccination against bubonic and pneumonic plague. Vaccine 2001, 19, 4175–4184. [Google Scholar] [CrossRef]
  114. Sun, W.; Singh, A.K. Plague vaccine: Recent progress and prospects. NPJ Vaccines 2019, 4, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Cavanaugh, D.C. K.F. Meyer’s work on plague. J. Infect. Dis. 1974, 129 (Suppl. S1), S10–S12. [Google Scholar] [CrossRef]
  116. Meyer, K.F.; Smith, G.; Foster, L.E.; Marshall, J.D., Jr.; Cavanaugh, D.C. Plague immunization. IV. Clinical reactions and serologic response to inoculations of Haffkine and freeze-dried plague vaccine. J. Infect. Dis. 1974, 129, S30–S36. [Google Scholar] [CrossRef]
  117. Bartelloni, P.J.; Marshall, J.D., Jr.; Cavanaugh, D.C. Clinical and serological responses to plague vaccine U.S.P. Mil. Med. 1973, 138, 720–722. [Google Scholar] [CrossRef]
  118. Cavanaugh, D.C.; Elisberg, B.L.; Llewellyn, C.H.; Marshall, J.D., Jr.; Rust, J.H., Jr.; Williams, J.E.; Meyer, K.F. Plague immunization. V. Indirect evidence for the efficacy of plague vaccine. J. Infect. Dis. 1974, 129 (Suppl. S1), S37–S40. [Google Scholar] [CrossRef]
  119. Meyer, K.F.; Smith, G.; Foster, L.; Brookman, M.; Sung, M. Live, attenuated Yersinia pestis vaccine: Virulent in nonhuman primates, harmless to guinea pigs. J. Infect. Dis. 1974, 129, S85–S112. [Google Scholar] [CrossRef]
  120. Titball, R.W.; Williamson, E.D. Yersinia pestis (plague) vaccines. Expert Opin. Biol. Ther. 2004, 4, 965–973. [Google Scholar] [CrossRef]
  121. Marshall, J.D., Jr.; Bartelloni, P.J.; Cavanaugh, D.C.; Kadull, P.J.; Meyer, K.F. Plague immunization. II. Relation of adverse clinical reactions to multiple immunizations with killed vaccine. J. Infect. Dis. 1974, 129, S19–S25. [Google Scholar] [CrossRef]
  122. Russell, P.; Eley, S.M.; Hibbs, S.E.; Manchee, R.J.; Stagg, A.J.; Titball, R.W. A comparison of Plague vaccine, USP and EV76 vaccine induced protection against Yersinia pestis in a murine model. Vaccine 1995, 13, 1551–1556. [Google Scholar] [CrossRef]
  123. Andrews, G.P.; Health, D.G.; Anderson, G.W.; Welkos, S.L.; Friedlander, A.M. Fraction 1 capsular antigen (F1) purification from Yersinia pestis CO92 and from an Escherichia coli recombinant strain and efficacy against lethal plague challenge. Infect. Immun. 1996, 64, 2180–2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Smith, H.; Packman, L.P. A filtered non-toxic plague vaccine which protects guinea-pigs and mice. Br. J. Exp. Pathol. 1966, 47, 25–34. [Google Scholar] [PubMed]
  125. Pitt, L.M. Non-human primates as a model for pneumonic plague: Animal models and correlates of protection for plague. In Proceedings of the Plague Vaccines Workshop, Gaithersburg, MD, USA, 13–14 October 2004. [Google Scholar]
  126. Branger, C.G.; Torres-Escobar, A.; Sun, W.; Perry, R.; Fetherston, J.D.; Roland, K.; Curtiss, R. Oral vaccination with LcrV from Yersinia pestis KIM delivered by live attenuated Salmonella enterica serovar Typhimurium elicits a protective immune response against challenge with Yersinia pseudotuberculosis and Yersinia enterocolitica. Vaccine 2009, 27, 5363–5370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Branger, C.G.; Sun, W.; Torres-Escobar, A.; Perry, R.; Roland, K.L.; Fetherston, J.; Curtiss, R. Evaluation of Psn, HmuR and a modified LcrV protein delivered to mice by live attenuated Salmonella as a vaccine against bubonic and pneumonic Yersinia pestis challenge. Vaccine 2010, 29, 274–282. [Google Scholar] [CrossRef] [Green Version]
  128. Branger, C.G.; Fetherston, J.D.; Perry, R.D.; Curtiss, R. Oral vaccination with different antigens from Yersinia pestis KIM delivered by live attenuated Salmonella typhimurium elicits a protective immune response against plague. Adv. Exp. Med. Biol. 2007, 603, 387–399. [Google Scholar]
  129. Girard, G.; Robic, J. Current status of the plague in Madagascar and vaccinal prophylaxis with the aid of the EV virus-vaccine. Bull. Soc. Path. Exot. 1942, 35, 43–49. [Google Scholar]
  130. Feodorov, V.A.; Lyapina, A.M.; Ulianova, O.V.; Lyapina, E.P.; Sayapina, L.V.; Lyapin, M.N.; Shcherbakov, A.A.; Telepnev, M.V.; Motin, V.L. Serologic markers for long-term immunity in humans vaccinated with live Yersinia pestis EV NIIEG. Procedia Vaccinol. 2012, 6, 10–13. [Google Scholar] [CrossRef] [Green Version]
  131. Meyer, K.F.; Cavanaugh, D.C.; Bartelloni, P.J.; Marshall, J.D., Jr. Plague immunization. I. Past and present trends. J. Infect. Dis. 1974, 129, S13–S18. [Google Scholar] [CrossRef]
  132. Feodorova, V.A.; Sayapina, L.V.; Corbel, M.J.; Motin, V.L. Russian vaccines against especially dangerous bacterial pathogens. Emerg. Microbes Infect. 2014, 3, e86. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, X.; Zhang, X.; Zhou, D.; Yang, R. Live-attenuated Yersinia pestis vaccines. Expert Rev. Vaccines 2013, 12, 677–686. [Google Scholar] [CrossRef]
  134. Sun, W.; Roland, K.L.; Curtiss, R. Developing live vaccines against plague. J. Infect. Dev. Ctries. 2011, 5, 614–627. [Google Scholar] [CrossRef] [PubMed]
  135. Sun, W.; Curtiss, R. Rational considerations about development of live attenuated Yersinia pestis vaccines. Curr. Pharm. Biotechnol. 2013, 14, 878–886. [Google Scholar] [CrossRef] [PubMed]
  136. Bubeck, S.S.; Dube, P.H. Yersinia pestis CO92 delta yopH is a potent live, attenuated plague vaccine. Clin. Vaccine Immunol. 2007, 14, 1235–1238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Bozue, J.; Cote, C.K.; Webster, W.; Bassett, A.; Tobery, S.; Little, S.; Swietnicki, W. A Yersinia pestis YscN ATPase mutant functions as a live attenuated vaccine against bubonic plague in mice. FEMS Microbiol. Lett. 2012, 332, 113–121. [Google Scholar] [CrossRef]
  138. Culbreth, M.J.; Biryukov, S.S.; Shoe, J.L.; Dankmeyer, J.L.; Hunter, M.; Klimko, C.P.; Rosario-Acevedo, R.; Fetterer, D.P.; Moreau, A.M.; Welkos, S.L.; et al. The use of analgesics during vaccination with a live attenuated Yersinia pestis vaccine alters the resulting immune response in mice. Vaccines 2019, 7, 205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Cote, C.K.; Biryukov, S.S.; Klimko, C.P.; Shoe, J.L.; Hunter, M.; Rosario-Acevedo, R.; Fetterer, D.P.; Moody, K.L.; Meyer, J.R.; Rill, N.O.; et al. Protection elicited by attenuated Live Yersinia pestis vaccine strains against lethal infection with virulent, Y. pestis. Vaccines 2021, 9, 161. [Google Scholar] [CrossRef]
  140. Jenkins, A.L.; Worsham, P.L.; Welkos, S.L. A strategy to verify the absence of the pgm locus in Yersinia pestis strain candidates for select agent exemption. J. Microbiol. Methods 2009, 77, 316–319. [Google Scholar] [CrossRef]
  141. Welkos, S.; Pitt, M.L.; Martinez, M.; Friedlander, A.; Vogel, P.; Tammariello, R. Determination of the virulence of the pigmentation-deficient and pigmentation-/plasminogen activator-deficient strains of Yersinia pestis in non-human primate and mouse models of pneumonic plague. Vaccine 2002, 20, 2206–2214. [Google Scholar] [CrossRef]
  142. Demeure, C.E.; Derbise, A.; Carniel, E. Oral vaccination against plague using Yersinia pseudotuberculosis. Chem. Biol. Interact. 2017, 267, 89–95. [Google Scholar] [CrossRef] [PubMed]
  143. Singh, A.K.; Curtiss, R., 3rd; Sun, W. A recombinant attenuated Yersinia pseudotuberculosis vaccine delivering a Y. pestis YopE(Nt138)-LcrV fusion elicits broad protection against plague and yersiniosis in mice. Infect. Immun. 2019, 87, e00296-19. [Google Scholar] [CrossRef] [PubMed]
  144. Sun, W.; Sanapala, S.; Rahav, H.; Curtiss, R., 3rd. Oral administration of a recombinant attenuated Yersinia pseudotuberculosis strain elicits protective immunity against plague. Vaccine 2015, 33, 6727–6735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Sun, W.; Sanapala, S.; Henderson, J.C.; Sam, S.; Olinzock, J.; Trent, M.S.; Curtiss, R., 3rd. LcrV delivered via type III secretion system of live attenuated Yersinia pseudotuberculosis enhances immunogenicity against pneumonic plague. Infect. Immun. 2014, 82, 4390–4404. [Google Scholar] [CrossRef] [Green Version]
  146. Hervé, C.; Laupèze, B.; Del Giudice, G.; Didierlaurent, A.M.; Tavares Da Silva, F. The how’s and what’s of vaccine reactogenicity. NPJ Vaccines 2019, 4, 39. [Google Scholar] [CrossRef] [Green Version]
  147. Williamson, E.D.; Eley, S.M.; Stagg, A.J.; Green, M.; Russell, P.; Titball, R.W. A sub-unit vaccine elicits IgG in serum, spleen cell cultures and bronchial washings and protects immunized animals against pneumonic plague. Vaccine 1997, 15, 1079–1084. [Google Scholar] [CrossRef]
  148. Jones, S.M.; Day, F.; Stagg, A.J.; Williamson, E.D. Protection conferred by a fully recombinant sub-unit vaccine against Yersinia pestis in male and female mice of four inbred strains. Vaccine 2000, 19, 358–366. [Google Scholar] [CrossRef]
  149. Al-Jawdah, A.D.; Ivanova, I.G.; Waller, H.; Perkins, N.D.; Lakey, J.H.; Peters, D.T. Induction of the immunoprotective coat of Yersinia pestis at body temperature is mediated by the Caf1R transcription factor. BMC Microbiol. 2019, 19, 68. [Google Scholar] [CrossRef]
  150. Galyov, E.E.; Smirnov, O.Y.; Karlishev, A.V.; Volkovoy, K.I.; Denesyuk, A.I.; Nazimov, I.V.; Rubtsov, K.S.; Abramov, V.M.; Dalvadyanz, S.M.; Zavyalov, V.P. Nucleotide sequence of the Yersinia pestis gene encoding F1 antigen and the primary structure of the protein. FEBS Lett. 1990, 277, 230–232. [Google Scholar] [CrossRef] [Green Version]
  151. Karlyshev, A.V.; Galyov, E.E.; Smirnov, O.Y.; Guzayev, A.P.; Abramov, V.M.; Zavyalov, V.P. A new gene of the ƒ1 operon of Y. pestis involved in the capsule biogenesis. FEBS Lett. 1992, 297, 77–80. [Google Scholar] [CrossRef] [Green Version]
  152. Donavan, J.E.; Ham, D.; Fukui, G.M.; Surgalla, M.J. Role of the capsule of Pasteurella Pestis in bubonic plague in the guinea pig. J. Infect. Dis. 1961, 109, 154–157. [Google Scholar] [CrossRef] [PubMed]
  153. Vorontsov, E.D.; Dubichev, A.G.; Serdobintsev, L.N.; Naumov, A.V. Association-dissociation processes and supermolecular organisation of the capsule antigen (protein F1) of Yersinia pestis. Biomed. Sci. 1990, 1, 391–396. [Google Scholar] [PubMed]
  154. Burrows, T.W.; Bacon, G.A. The effects of loss of different virulence determinants on the virulence and immunogenicity of strains of Pasteurella pestis. Br. J. Exp. Pathol. 1958, 39, 278–291. [Google Scholar] [PubMed]
  155. Anderson, G.W., Jr.; Leary, S.E.; Williamson, E.D.; Titball, R.; Welkos, S.L.; Wosham, P.L.; Friendlander, A.M. Recombinant V antigen protects mice against pneumonic and bubonic plague caused by F1-capsule-positive and -negative strains of Yersinia pestis. Infect. Immun. 1996, 64, 4580–4585. [Google Scholar] [CrossRef] [Green Version]
  156. Hill, J.; Leary, S.E.; Griffin, K.F.; Williamson, E.D.; Titball, R.W. Regions of Yersinia pestis V antigen that contribute to protection against plague identified by passive and active immunization. Infect. Immun. 1997, 65, 4476–4482. [Google Scholar] [CrossRef] [Green Version]
  157. Leary, S.E.; Williamson, E.D.; Griffin, K.F.; Russell, P.; Eley, R.W.; Titball, R.W. Active immunization with recombinant V antigen from Yersinia pestis protects mice against plague. Infect. Immun. 1995, 63, 2854–2858. [Google Scholar] [CrossRef] [Green Version]
  158. Weeks, S.; Hill, J.; Friendlander, A.; Welkos, S. Anti-V antigen antibody protects macrophages from Yersinia pestis -induced cell death and promotes phagocytosis. Microb. Pathog. 2002, 32, 227–237. [Google Scholar] [CrossRef]
  159. Pettersson, J.; Holmström, A.; Hill, J.; Leary, S.; Frithz-Lindsten, E.; von Euler-Matell, A.; Carlsson, E.; Titball, R.; Forsberg, A.; Wolf-Watz, H. The V-antigen of Yersinia is surface exposed before target cell contact and involved in virulence protein translocation. Mol. Microbiol. 1999, 32, 961–976. [Google Scholar] [CrossRef]
  160. Sawa, T.; Yahr, T.L.; Ohara, M.; Kurahashi, K.; Gropper, M.A.; Wiener-Kronish, J.P.; Frank, D.W. Active and passive immunization with the Pseudomonas V antigen protects against type III intoxication and lung injury. Nat. Med. 1999, 5, 392–398. [Google Scholar] [CrossRef]
  161. Anisimov, A.P.; Dentovskaya, A.V.; Panfertsev, E.A.; Svetoch, T.E.; Kopylov, P.K.; Segelke, B.W.; Zemla, A.; Telepnev, M.V.; Motin, V.L. Amino acid and structural variability of Yersinia pestis LcrV protein. Infect. Genet. Evol. 2010, 10, 137–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Simpson, W.J.; Thomas, R.E.; Schwan, T.G. Recombinant capsular antigen (fraction 1) from Yersinia pestis induces a protective antibody response in BALB/c mice. Am. J. Trop. Med. Hyg. 1990, 43, 389–396. [Google Scholar] [CrossRef] [PubMed]
  163. Carr, S.; Miller, J.; Leary, S.E.; Bennett, A.M.; Ho, A.; Williamson, E.D. Expression of a recombinant form of the V antigen of Yersinia pestis, using three different expression systems. Vaccine 1999, 18, 153–159. [Google Scholar] [CrossRef]
  164. Chichester, J.A.; Musiychuk, K.; Farrance, C.E.; Mett, V.; Lyons, J.; Mett, V.; Yusibov, V. A single component two-valent LcrV-F1 vaccine protects non-human primates against pneumonic plague. Vaccine 2009, 27, 3471–3474. [Google Scholar] [CrossRef] [PubMed]
  165. Williamson, E.D.; Vesey, P.M.; Gillhespy, K.J.; Eley, S.M.; Green, M.; Titball, R.W. An IgG1 titre to the F1 and V antigens correlates with protection against plague in the mouse model. Clin. Exp. Immunol. 1999, 116, 107–114. [Google Scholar] [CrossRef]
  166. Powell, B.S.; Andrews, G.P.; Enama, J.T.; Jenderk, S.; Bolt, C.; Worsham, P.; Pullen, J.K.; Ribot, W.; Hines, H.; Smith, L.; et al. Design and testing for a nontagged F1-V fusion protein as vaccine antigen against bubonic and pneumonic plague. Biotechnol. Prog. 2005, 21, 1490–1510. [Google Scholar] [CrossRef] [Green Version]
  167. Goodin, J.L.; Nellis, D.F.; Powell, B.S.; Vyas, V.V.; Enama, J.T.; Wang, L.C.; Clark, P.K.; Giardina, S.L.; Adamovicz, J.J.; Michiel, D.F. Purification and protective efficacy of monomeric and modified Yersinia pestis capsular F1-V antigen fusion proteins for vaccination against plague. Protein Exp. Purif. 2007, 53, 63–79. [Google Scholar] [CrossRef] [Green Version]
  168. Fellows, P.; Adamovicz, J.; Hartings, J.; Sherwood, R.; Mega, W.; Brasel, T.; Barr, E.; Holland, L.; Lin, W.; Rom, A.; et al. Protection in mice passively immunized with serum from cynomolgus macaques and humans vaccinated with recombinant plague vaccine (rF1V). Vaccine 2010, 28, 7748–7756. [Google Scholar] [CrossRef]
  169. Glynn, A.; Roy, C.J.; Powell, B.S.; Adamovicz, J.J.; Freytag, L.C.; Clements, J.D. Protection against aerosolized Yersinia pestis challenge following homologous and heterologous prime-boost with recombinant plague antigens. Infect. Immun. 2005, 73, 5256–5261. [Google Scholar] [CrossRef] [Green Version]
  170. Heath, D.G.; Anderson, G.W.; Mauro, J.M.; Welkos, S.L.; Andrews, G.P.; Adamovicz, J.; Friedlander, A.M. Protection against experimental bubonic and pneumonic plague by a recombinant capsular F1-V antigen fusion protein vaccine. Vaccine 1998, 16, 1131–1137. [Google Scholar] [CrossRef]
  171. Williamson, E.D.; Eley, S.M.; Stagg, A.J.; Green, M.; Russell, P.; Titball, R.W. A single dose sub-unit vaccine protects against pneumonic plague. Vaccine 2000, 19, 566–571. [Google Scholar] [CrossRef]
  172. Williamson, E.D.; Eley, S.M.; Griffin, K.F.; Green, M.; Russell, P.; Leary, S.E.; Oyston, P.C.; Easterbrokk, T.; Reddin, K.M.; Robinson, A. A new improved sub-unit vaccine for plague: The basis of protection. FEMS Immunol. Med. Microbiol. 1995, 12, 223–230. [Google Scholar] [CrossRef] [PubMed]
  173. Elvin, S.J.; Williamson, E.D. The F1 and V subunit vaccine protects against plague in the absence of IL-4 driven immune responses. Microb. Pathog. 2000, 29, 223–230. [Google Scholar] [CrossRef]
  174. Sun, W. Plague Vaccines: Status and Future. Adv. Exp. Med. Biol. 2016, 918, 313–360. [Google Scholar]
  175. Quenee, L.E.; Schneewind, O. Plague vaccines and the molecular basis of immunity against Yersinia pestis. Hum. Vaccine 2009, 5, 817–823. [Google Scholar] [CrossRef]
  176. Quenee, L.E.; Ciletti, N.A.; Elli, D.; Hermanas, T.M.; Schneewind, O. Prevention of pneumonic plague in mice, rats, guinea pigs and non-human primates with clinical grade rV10, rV10-2 or F1-V vaccines. Vaccine 2011, 29, 6572–6583. [Google Scholar] [CrossRef] [Green Version]
  177. Williamson, E.D.; Packer, P.J.; Waters, E.L.; Simpson, D.D.; Hartings, J.; Twenhafel, N.; Pitt, M.L. Recombinant (F1+V) vaccine protects cynomolgus macaques against pneumonic plague. Vaccine 2011, 29, 4771–4777. [Google Scholar] [CrossRef]
  178. Amemiya, K.; Meyers, J.L.; Rogers, T.E.; Fast, R.L.; Bassett, A.D.; Worsham, P.L.; Powell, B.S.; Norris, S.L.; Krieg, A.M.; Adamovicz, J.J. CpG oligodeoxynucleotides augment the murine immune response to the Yersinia pestis F1-V vaccine in bubonic and pneumonic models of plague. Vaccine 2009, 27, 2220–2229. [Google Scholar] [CrossRef]
  179. D’Arco, C.; McCormick, A.A.; Arnaboldi, P.M. Single-dose intranasal subunit vaccine rapidly clears secondary sepsis in a high-dose pneumonic plague infection. Vaccine 2021, 39, 1435–1444. [Google Scholar] [CrossRef]
  180. Kilgore, P.B.; Sha, J.; Anderson, J.A.; Motin, V.L.; Chopra, A.K. A new generation needle- and adjuvant-free trivalent plague vaccine utilizing adenovirus-5 nanoparticle platform. NPJ Vaccines 2021, 6, 21. [Google Scholar] [CrossRef] [PubMed]
  181. Hamzabegovic, F.; Goll, J.B.; Hooper, W.F.; Frey, S.; Gelber, C.E.; Abate, G. Flagellin adjuvanted F1/V subunit plague vaccine induces T cell and functional antibody responses with unique gene signatures. NPJ Vaccines 2020, 5, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Del Prete, G.; Santi, L.; Andrianaivoarimanana, V.; Amedei, A.; Domarle, O.; D’Elios, M.M.; Arntzen, C.J.; Rahalison, L.; Mason, H.S. Plant-derived recombinant F1, V, and F1-V fusion antigens of Yersinia pestis activate human cells of the innate and adaptive immune system. Int. J. Immunopathol. Pharmacol. 2009, 22, 133–143. [Google Scholar] [CrossRef] [Green Version]
  183. Jones, T.; Adamovicz, J.J.; Cyr, S.L.; Bolt, C.R.; Bellerose, N.; Pitt, L.M.; Lowell, G.H.; Burt, D.S. Intranasal Protollin/F1-V vaccine elicits respiratory and serum antibody responses and protects mice against lethal aerosolized plague infection. Vaccine 2006, 24, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  184. Goodin, J.L.; Powell, B.S.; Enama, J.T.; Raab, R.W.; McKown, R.L.; Coffman, G.L.; Andrews, G.P. Purification and characterization of a recombinant Yersinia pestis V-F1 “Reversed” fusion protein for use as a new subunit vaccine against plague. Protein Exp. Purif. 2011, 76, 136–144. [Google Scholar] [CrossRef]
  185. Yamanaka, H.; Hoyt, T.; Yang, X.; Golden, S.; Bosio, C.M.; Crist, K.; Becker, T.; Maddaloni, M.; Pascual, D.W. A nasal interleukin-12 DNA vaccine coexpressing Yersinia pestis F1-V fusion protein confers protection against pneumonic plague. Infect. Immun. 2008, 76, 4564–4573. [Google Scholar] [CrossRef] [Green Version]
  186. Jones, S.; Griffin, K.; Hodgson, I.; Williamson, E. Protective efficacy of a fully recombinant plague vaccine in the guinea pig. Vaccine 2003, 21, 3912–3918. [Google Scholar] [CrossRef]
  187. Williamson, E.D.; Flick-Smith, H.C.; Lebutt, C.; Rowland, C.A.; Jones, S.M.; Waters, E.L.; Gwyther, R.J.; Miller, J.; Packer, P.J.; Irving, M. Human immune response to a plague vaccine comprising recombinant F1 and V antigens. Infect. Immun. 2005, 73, 3598–3608. [Google Scholar] [CrossRef] [Green Version]
  188. Andrews, G.P.; Strachan, S.T.; Benner, G.E.; Sample, A.K.; Anderson, G.W., Jr.; Adamovicz, J.J.; Welkos, S.L.; Pullen, J.K.; Friedlander, A.M. Protective efficacy of recombinant Yersinia outer proteins against bubonic plague caused by encapsulated and nonencapsulated Yersinia pestis. Infect. Immun. 1999, 67, 1533–1537. [Google Scholar] [CrossRef] [Green Version]
  189. Ivanov, M.I.; Noel, B.L.; Rampersaud, R.; Mena, P.; Benach, J.L.; Bliska, J.B. Vaccination of mice with a Yop translocon complex elicits antibodies that are protective against infection with F1- Yersinia pestis. Infect. Immun. 2008, 76, 5181–5190. [Google Scholar] [CrossRef] [Green Version]
  190. Motin, V.L.; Nakajima, R.; Smirnov, G.B.; Brubaker, R.R. Passive immunity to yersiniae mediated by anti-recombinant V antigen and protein A-V antigen fusion peptide. Infect. Immun. 1994, 62, 4192–4201. [Google Scholar] [CrossRef] [Green Version]
  191. Quenee, L.E.; Berube, B.J.; Segal, J.; Elli, D.; Ciletti, N.A.; Anderson, D.; Schneewind, O. Amino acid residues 196-226 of LcrV represent a plague protective epitope. Vaccine 2010, 28, 1870–1876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Amemiya, K.; Dankmeyer, J.L.; Keasey, S.L.; Trevino, S.R.; Wormald, M.M.; Halasohoris, S.A.; Ribot, W.J.; Fetterer, D.P.; Cote, C.K.; Worsham, P.L.; et al. Binding sites of anti-LcrV monoclonal antibodies are more critical than the avidities and affinities for passive protection against Yersinia pestis infection in a bubonic plague model. Antibodies 2020, 9, 37. [Google Scholar] [CrossRef] [PubMed]
  193. Cowan, C.; Philipovskiy, A.V.; Wulff-Strobel, C.R.; Ye, Z.; Straley, S.C. Anti-LcrV antibody inhibits delivery of Yops by Yersinia pestis KIM5 by directly promoting phagocytosis. Infect. Immun. 2005, 73, 6127–6137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Eisele, N.A.; Anderson, D.M. Dual-function antibodies to Yersinia pestis LcrV required for pulmonary clearance of plague. Clin. Vaccine Immunol. 2009, 16, 1720–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Sofer-Podesta, C.; Ang, J.; Hackett, N.R.; Senina, S.; Perlin, D.; Crystal, R.G.; Boyer, J.L. Adenovirus-mediated delivery of an anti-V antigen monoclonal antibody protects mice against a lethal Yersinia pestis challenge. Infect. Immun. 2009, 77, 1561–1568. [Google Scholar] [CrossRef] [Green Version]
  196. Miller, N.C.; Quenee, L.E.; Elli, D.; Ciletti, N.A.; Schneewind, O. Polymorphisms in the lcrV gene of Yersinia enterocolitica and their effect on plague protective immunity. Infect. Immun. 2012, 80, 1572–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Roggenkamp, A.; Geiger, A.M.; Leitritz, L.; Kessler, A.; Heesemann, J. Passive immunity to infection with Yersinia spp. mediated by anti-recombinant V antigen is dependent on polymorphism of V antigen. Infect. Immun. 1997, 65, 446–451. [Google Scholar] [CrossRef] [Green Version]
  198. Daniel, C.; Dewitte, A.; Poiret, S.; Marceau, M.; Simonet, M.; Marceau, L.; Descombes, G.; Boutillier, D.; Bennaceur, N.; Bontemps-Gallo, S.; et al. Polymorphism in the yersinia LcrV antigen enables immune escape from the protection conferred by an LcrV-secreting Lactococcus lactis in a pseudotuberculosis mouse model. Front. Immunol. 2019, 10, 1830. [Google Scholar] [CrossRef] [Green Version]
  199. Anderson, G.W., Jr.; Worsham, P.L.; Bolt, C.R.; Andrews, G.P.; Welkos, S.L.; Friedlander, A.M. Protection of mice from fatal bubonic and pneumonic plague by passive immunization with monoclonal antibodies against the F1 protein of Yersinia pestis. Am. J. Trop. Med. Hyg. 1997, 56, 471–473. [Google Scholar] [CrossRef]
  200. Hill, J.; Copse, C.; Leary, S.; Stagg, A.J.; Williamson, E.D.; Titball, R.W. Synergistic protection of mice against plague with monoclonal antibodies specific for the F1 and V antigens of Yersinia pestis. Infect. Immun. 2003, 71, 2234–2238. [Google Scholar] [CrossRef] [Green Version]
  201. Hill, J.; Eyles, J.E.; Elvin, S.J.; Healey, G.D.; Lukaszewski, R.A.; Titball, R.W. Administration of antibody to the lung protects mice against pneumonic plague. Infect. Immun. 2006, 74, 3068–3070. [Google Scholar] [CrossRef] [Green Version]
  202. Xiao, X.; Zhu, Z.; Dankmeyer, J.L.; Wormald, M.M.; Fast, R.L.; Worsham, P.L.; Cote, C.K.; Amemiya, K.; Dimitrov, D.S. Human anti-plague monoclonal antibodies protect mice from Yersinia pestis in a bubonic plague model. PLoS ONE 2010, 5, e13047. [Google Scholar] [CrossRef] [Green Version]
  203. Liu, W.; Ren, J.; Zhang, J.; Song, X.; Lui, S.; Chi, X.; Chen, Y.; Wem, Z.; Li, J.; Chen, W. Identification and characterization of a neutralizing monoclonal antibody that provides complete protection against Yersinia pestis. PLoS ONE 2017, 12, e0177012. [Google Scholar] [CrossRef]
  204. Lillo, A.M.; Velappan, N.; Kelliher, J.M.; Watts, A.J.; Merriman, S.P.; Vuyisich, G.; Lilley, L.M.; Coombs, K.E.; Mastren, T.; Teshima, M.; et al. Development of anti-Yersinia pestis human antibodies with features required for diagnostic and therapeutic applications. Immunotargets Ther. 2020, 9, 299–316. [Google Scholar] [CrossRef] [PubMed]
  205. Navalkele, B.D.; Chopra, T. Bezlotoxumab: An emerging monoclonal antibody therapy for prevention of recurrent Clostridium difficile infection. Biologics 2018, 12, 11–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Humphreys, D.P.; Wilcox, M.H. Antibodies for treatment of Clostridium difficile infection. Clin. Vaccine Immunol. 2014, 21, 913–923. [Google Scholar] [CrossRef]
  207. Chen, Z.; Moayeri, M.; Purcell, R. Monoclonal antibody therapies against anthrax. Toxins 2011, 3, 1004–1019. [Google Scholar] [CrossRef] [Green Version]
  208. Pelfrene, E.; Mura, M.; Cavaleiro Sanches, A.; Cavaleri, M. Monoclonal antibodies as anti-infective products: A promising future? Clin. Microbiol. Infect. 2019, 25, 60–64. [Google Scholar] [CrossRef] [PubMed]
  209. Schweizer, H.P. Mechanisms of antibiotic resistance in Burkholderia pseudomallei: Implications for treatment of melioidosis. Future Microbiol. 2012, 7, 1389–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Jarrad, A.M.; Karoli, T.; Blaskovich, M.A.; Lyras, D.; Cooper, M.A. Clostridium difficile drug pipeline: Challenges in discovery and development of new agents. J. Med. Chem. 2015, 58, 5164–5185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Merriman, H. Chapter 13—Infectious diseases. In Acute Care Handbook for Physical Therapists, 4th ed.; Paz, J.C., West, M.P., Eds.; W.B. Saunders: St. Louis, MO, USA, 2014; pp. 313–334. [Google Scholar]
  212. Santajit, S.; Indrawattana, N. Mechanisms of antimicrobial resistance in ESKAPE pathogens. Biomed. Res. Int. 2016, 2016, 2475067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Pachori, P.; Gothalwal, R.; Gandhi, P. Emergence of antibiotic resistance Pseudomonas aeruginosa in intensive care unit; a critical review. Genes Dis. 2019, 6, 109–119. [Google Scholar] [CrossRef] [PubMed]
  214. González-González, E.; Alvarez, M.M.; Márquez-Ipiña, A.R.; Trujillo-de Santiago, G.; Rodríguez-Martínez, L.M.; Annabi, N.; Khademhosseini, A. Anti-Ebola therapies based on monoclonal antibodies: Current state and challenges ahead. Crit. Rev. Biotechnol. 2017, 37, 53–68. [Google Scholar] [CrossRef]
  215. Bailey, M.J.; Broecker, F.; Freyn, A.W.; Choi, A.; Brown, J.A.; Federova, N.; Simon, V.; Lim, J.K.; Evans, M.J.; García-Sastre, A.; et al. Human monoclonal antibodies potently neutralize Zika virus and select for escape mutations on the lateral ridge of the envelope protein. J. Virol. 2019, 93, e00405-19. [Google Scholar] [CrossRef] [Green Version]
  216. Marovich, M.; Mascola, J.R.; Cohen, M.S. Monoclonal antibodies for prevention and treatment of COVID-19. JAMA 2020, 324, 131–132. [Google Scholar] [CrossRef] [PubMed]
  217. Jahanshahlu, L.; Rezaei, N. Monoclonal antibody as a potential anti-COVID-19. Biomed. Pharmacother. 2020, 129, 110337. [Google Scholar] [CrossRef]
  218. Lülf, S.; Matz, J.; Rouyez, M.C.; Järviluoma, A.; Saksela, K.; Benichou, S.; Geyer, M. Structural basis for the inhibition of HIV-1 Nef by a high-affinity binding single-domain antibody. Retrovirology 2014, 11, 24. [Google Scholar] [CrossRef] [Green Version]
  219. Cruz-Teran, C.; Tiruthani, K.; McSweeney, M.; Ma, A.; Pickles, R.; Lai, S.K. Challenges and opportunities for antiviral monoclonal antibodies as COVID-19 therapy. Adv. Drug Deliv. Rev. 2021, 169, 100–117. [Google Scholar] [CrossRef] [PubMed]
  220. Pecetta, S.; Finco, O.; Seubert, A. Quantum leap of monoclonal antibody (mAb) discovery and development in the COVID-19 era. Semin. Immunol. 2020, 50, 101427. [Google Scholar] [CrossRef]
Figure 1. Y. pestis infection can lead to three forms of plague. Bubonic plague can result from flea bites or handling infected animals and can result in a severe localized infected lymph node referred to as a buboe (green). Septicemic plague can result as a secondary issue arising from bubonic disease or if the bacteria are introduced directly to the blood stream (red). Pneumonic plague is the most severe form of the disease and can result in person-to-person spread if untreated and initiated by the deposition of Y. pestis bacteria into the respiratory tract of individuals (blue).
Figure 1. Y. pestis infection can lead to three forms of plague. Bubonic plague can result from flea bites or handling infected animals and can result in a severe localized infected lymph node referred to as a buboe (green). Septicemic plague can result as a secondary issue arising from bubonic disease or if the bacteria are introduced directly to the blood stream (red). Pneumonic plague is the most severe form of the disease and can result in person-to-person spread if untreated and initiated by the deposition of Y. pestis bacteria into the respiratory tract of individuals (blue).
Biomedicines 09 01421 g001
Figure 2. Schematic of Antibodies that target Y. pestis and are effective at ameliorating disease. Y. pestis is a Gram-negative bacterium that expresses multiple potential targets that have been exploited for the development of novel medical countermeasures. Antibodies generated by active vaccination or administered via passive immunization have been successful at protecting cell culture and animals from Y. pestis infection. These targets include The F1 capsular antigen (yellow structures depicted in the left panel) and the LcrV (V antigen) component of the T3SS injectisome. The capsule, while not absolutely necessary for virulence, has been the target of both vaccine and therapeutic strategies. The LcrV protein has been shown to be essential for virulence and has been a successful target in both active and passive immunization studies and is often combined with the F1 capsular antigen (i.e., the F1-V vaccine or mAb cocktail experiments). (Left Panel) In most cases, the Y. pestis bacterium will interact directly with host target cells via the T3SS injectisome and a robust anti-phagocytic capsule will also be present; thus, both anti-F1 and anti-V antibodies are potentially effective. (Right Panel) Non-encapsulated strains of Y. pestis can be found in nature or engineered in the laboratory. In cases of infection caused by non-encapsulated Y. pestis, the antibodies to the F1 capsular antigen are no longer effective and the protective immune response generated by a vaccine or the effective therapeutic mAb relies solely on the anti-V antibodies. A lipopolysaccharide structure (LPS) often observed in Y. pestis grown at 37 °C is depicted in blue in both panels. Other protective antigens have been identified and other novel antigen targets will almost certainly be identified in ongoing research efforts.
Figure 2. Schematic of Antibodies that target Y. pestis and are effective at ameliorating disease. Y. pestis is a Gram-negative bacterium that expresses multiple potential targets that have been exploited for the development of novel medical countermeasures. Antibodies generated by active vaccination or administered via passive immunization have been successful at protecting cell culture and animals from Y. pestis infection. These targets include The F1 capsular antigen (yellow structures depicted in the left panel) and the LcrV (V antigen) component of the T3SS injectisome. The capsule, while not absolutely necessary for virulence, has been the target of both vaccine and therapeutic strategies. The LcrV protein has been shown to be essential for virulence and has been a successful target in both active and passive immunization studies and is often combined with the F1 capsular antigen (i.e., the F1-V vaccine or mAb cocktail experiments). (Left Panel) In most cases, the Y. pestis bacterium will interact directly with host target cells via the T3SS injectisome and a robust anti-phagocytic capsule will also be present; thus, both anti-F1 and anti-V antibodies are potentially effective. (Right Panel) Non-encapsulated strains of Y. pestis can be found in nature or engineered in the laboratory. In cases of infection caused by non-encapsulated Y. pestis, the antibodies to the F1 capsular antigen are no longer effective and the protective immune response generated by a vaccine or the effective therapeutic mAb relies solely on the anti-V antibodies. A lipopolysaccharide structure (LPS) often observed in Y. pestis grown at 37 °C is depicted in blue in both panels. Other protective antigens have been identified and other novel antigen targets will almost certainly be identified in ongoing research efforts.
Biomedicines 09 01421 g002
Table 1. Monoclonal antibodies shown to protect mice in bubonic plague models (subcutaneous challenge with bacteria).
Table 1. Monoclonal antibodies shown to protect mice in bubonic plague models (subcutaneous challenge with bacteria).
Antigen amAb DescriptionAntibody/Therapeutic AdministrationY. pestis Challenge Strain dChallenge Dose (LD50)Ref.
RouteConcentrationSchedule
F1; LcrV; F1V + LcrVhuman IgG1IP500 µg24 h pre–120 h postCO9225–40202
LcrVmouseIP350 µg24 h preGB12156
LcrV bmouse IgG1, IgG2aIP1–500 µg24 h preCO92 or C1221–39192
LcrV; F1 + LcrVmouse IgG1IP0.7–100 µg4 h pre–96 postGB9.6–91,000200
LcrVmouse IgG1IP200 µg1 h preCO9220191
F1 cmouse IgG1IP125–500 µg6 h or 24 h preCO9248–54199
F1not reportedIV100 µg24 h pre141600203
a, unless otherwise noted mAbs tested in BALB/c female mice; b, tested in BALB/c and Swiss Webster mice; c, tested in Swiss Webster female mice; d, bacteria delivered via subcutaneous injection.
Table 2. Monoclonal antibodies shown to protect mice in pneumonic plague models (intranasal instillation or exposure to aerosolized bacteria).
Table 2. Monoclonal antibodies shown to protect mice in pneumonic plague models (intranasal instillation or exposure to aerosolized bacteria).
Antigen amAb DescriptionAntibody/Therapeutic AdministrationY. pestis Challenge Strain fChallenge Dose (LD50)Ref.
RouteConcentrationSchedule
LcrVmouse IgG1IP35 µg4 h pre–96 h postGB88156
LcrV bmouse IgG1IP200 or 400 µg1 h preCO92 g15–20194
LcrV cmouse IgG2bIP, IV100–500 µg, 1011 pu e94 h pre–24 h postCO92 g363–9080195
F1 dmouse IgG1IP125–500 µg6 h or 24 h preCO9229–74199
F1 + LcrVmouse IgG1IT77.5 µg (each)2 h postGB27200
a, unless otherwise noted mAbs tested in BALB/c female mice; b, tested in C57BL/6 female mice; c, tested in BALB/c female and C57BL/6 male mice; d, tested in Swiss Webster female mice; e, antibodies delivered as hybridoma supernatant or via adenovirus vector; f, unless otherwise noted aerosolized bacteria were delivered; g, intranasal instillation was used for challenge
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosario-Acevedo, R.; Biryukov, S.S.; Bozue, J.A.; Cote, C.K. Plague Prevention and Therapy: Perspectives on Current and Future Strategies. Biomedicines 2021, 9, 1421. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9101421

AMA Style

Rosario-Acevedo R, Biryukov SS, Bozue JA, Cote CK. Plague Prevention and Therapy: Perspectives on Current and Future Strategies. Biomedicines. 2021; 9(10):1421. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9101421

Chicago/Turabian Style

Rosario-Acevedo, Raysa, Sergei S. Biryukov, Joel A. Bozue, and Christopher K. Cote. 2021. "Plague Prevention and Therapy: Perspectives on Current and Future Strategies" Biomedicines 9, no. 10: 1421. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9101421

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop