Next Article in Journal
Genotypic and Phenotypic Evaluation of Biofilm Production and Antimicrobial Resistance in Staphylococcus aureus Isolated from Milk, North West Province, South Africa
Next Article in Special Issue
Nontraditional Antibiotics—Challenges and Triumphs
Previous Article in Journal
Carbapenem-Resistant but Cephalosporin-Susceptible Pseudomonas aeruginosa in Urinary Tract Infections: Opportunity for Colistin Sparing
Previous Article in Special Issue
Prevalence and Therapeutic Challenges of Fungal Drug Resistance: Role for Plants in Drug Discovery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens

by
Daniel V. Zurawski
* and
Molly K. McLendon
Wound Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
*
Author to whom correspondence should be addressed.
Antibiotics 2020, 9(4), 155; https://doi.org/10.3390/antibiotics9040155
Submission received: 23 February 2020 / Revised: 14 March 2020 / Accepted: 16 March 2020 / Published: 1 April 2020
(This article belongs to the Special Issue Nontraditional Antibiotics—Challenges and Triumphs)

Abstract

:
In the beginning of the 21st century, the frequency of antimicrobial resistance (AMR) has reached an apex, where even 4th and 5th generation antibiotics are becoming useless in clinical settings. In turn, patients are suffering from once-curable infections, with increases in morbidity and mortality. The root cause of many of these infections are the ESKAPEE pathogens (Enterococcus species, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species, and Escherichia coli), which thrive in the nosocomial environment and are the bacterial species that have seen the largest rise in the acquisition of antibiotic resistance genes. While traditional small-molecule development still dominates the antibacterial landscape for solutions to AMR, some researchers are now turning to biological approaches as potential game changers. Monoclonal antibodies (mAbs)—more specifically, human monoclonal antibodies (Hu-mAbs)—have been highly pursued in the anti-cancer, autoimmune, and antiviral fields with many success stories, but antibody development for bacterial infection is still just scratching the surface. The untapped potential for Hu-mAbs to be used as a prophylactic or therapeutic treatment for bacterial infection is exciting, as these biologics do not have the same toxicity hurdles of small molecules, could have less resistance as they often target virulence proteins rather than proteins required for survival, and are narrow spectrum (targeting just one pathogenic species), therefore avoiding the disruption of the microbiome. This mini-review will highlight the current antibacterial mAbs approved for patient use, the success stories for mAb development, and new Hu-mAb products in the antibacterial pipeline.

1. Introduction

Steve Projan (former Vice President, Head of Infectious Diseases and Vaccines at AstraZeneca/MedImmune) has been one of the most ardent supporters of immunotherapies for bacterial infections over the last two decades. His experience, leadership, and knowledge drove a successful program of more than 30 people at MedImmune, Inc. (now AstraZeneca plc) generating bi-specific antibody solutions against bacterial infections (discussed below) and subsequently led to numerous keynote talks at antibacterial meetings. He famously starts many of his talks with the story of the Iditarod, the great dogsled race held every year in Alaska. The Iditarod began as commemoration of events that occurred in 1925 when a diphtheria infection spread through the town of Nome. Diptheria is caused by the bacterium Corynebacterium diphtheriae. At the time, the only solution for this lethal infection was serum that had been isolated from horses injected with diphtheria toxin, a toxin made by C. diphtheriae [1]. Because of harsh weather conditions, the only way to get the serum to Nome before it expired was by dogsled relay from Nenana, located 674 miles (1085 km) from Nome [2]. Estimates suggest 10,000 lives were saved in Nome and surrounding villages because of the heroic efforts of twenty sled dog teams running the serum across the Alaskan wilderness (Figure 1), in just six days, a journey that would normally take more than twenty days [2]. Horse serum, and the antibodies within, was the means of combatting C. diphtheriae infections in this case, but at the time, this method was also used to treat infections caused by bacterial species such as Streptococcus, Neisseria, and Haemophilus [3].
Until the Golden Age of Antibiotics began in the 1940s with the advent of penicillin, delivering passive immunity via horse serum or using bacteriophage therapy were the standards to treat bacterial infection [4,5], and the epic story of the birth of the Iditarod indicates how important serum was at limiting the spread of a bacterial infection outbreak. However, as more antibiotics were discovered and brought to market, it could be argued with good reason that small molecules were the better approach for controlling bacterial infection considering cost and efficacy [5]. Because of this, small molecule-based antibiotics dominated the antibacterial space for the next sixty years and still do. However, with the dawn of multidrug-resistant (MDR) strains and the present day emergence of extensively drug-resistant (XDR) and pandrug-resistant (PDR) strains of the ESKAPEE pathogens (Enterococcus species, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species, and Escherichia coli), it is clear that the Golden Age of Antibiotics is over, and the medical and research communities are seeking alternative solutions to traditional small-molecule antibacterial approaches.
Hybridomas were first discovered in the early 1970s by Kilner and Milstein [6], who went on to earn the Nobel Prize for this work. Since then, many developments have been made to improve the performance of monoclonal antibodies (mAbs) as therapeutics. The first of these was to show that a human cell line could also be used for the hybridoma process [7]. This early advance was born from the concept that fully human antibodies would make better therapeutics as they would less likely be cleared by the human immune system. Over the next decade, researchers discovered additional methods to further this idea. Key advances included identifying fully functional mouse mAbs and then humanizing them [8,9]. Others realized that isolating mAbs directly from patients who were infected and cleared infections could be important tools for identifying human monoclonal antibodies (Hu-mAbs) to neutralize the infectious agent [10]. Modern approaches have shown that once obtained, these Hu-mAbs can be sequenced, generated recombinantly, and generated in large quantities for clinical use [11]. More recent advances also include the development of phage display libraries of engineered Hu-mAbs that can dramatically increase the total number of human antibodies for testing to increase the likelihood of finding a unique antibody to interfere with or delay a disease-state [12]. Molecular biology and recombinant DNA techniques have also allowed researchers to modify different amino acids of the antibody’s structure to improve stability and utility in the host [13]. On the whole, the state of the science has advanced to where three major approaches are typically used to isolate/generate Hu-mAbs for therapeutic use: combinatorial display libraries, humanized mice, or single B cell cloning, and each of these strategies are thoroughly discussed in the review by Walter and Burton [14]. Additionally, others are using adenovirus [15,16] or recombinant DNA [17] to produce antibodies in the body, essentially a vaccination-like approach where the viral-encoded DNA or the naked DNA is then translated into a human antibody. Thus, rather than injecting the Hu-mAbs, the human body essentially becomes a Hu-mAb factory, continuously generating the antibody over a period of time. New data suggest that the recombinant DNA platform approach could also be used for bacterial infections as was shown recently in animals [18]. Nonetheless, the driving agent of protection is the Hu-mAb, whether it is made inside the body or injected intravenously.
Aside from the fact that Hu-mAbs are a human therapeutic product, which, in turn, minimizes toxicity concerns, there are other advantages of Hu-mAbs that make their pursuit a promising antibacterial approach. The first of these is longevity, as Hu-mAbs are not cleared by the host immune system as the half-life is typically 21 days for IgG subtypes [11]. Second, Hu-mAbs used as treatment against one bacterial species confers inherent pathogen specificity that does not disrupt normal bacterial flora in the body. Third, it potentiates both rapid and sustained killing via multiple mechanisms including: direct killing, anti-virulence, neutralization, complement deposition, and opsonization by phagocytes [11,19,20]. Furthermore, mAbs with Fc domains that bind to the host phagocyte receptor FcγRII result in downstream suppression of inflammation and sepsis caused by Gram-negative bacteria [11,21]. Killing bacteria by these multiple mechanisms limits toxic shock and the emergence of resistance. Further, tapping into the full capacity of the immune system allows for a diverse repertoire of cell types and killing machinery to clear bacteria from various locales in the body. It should also be noted that small molecules alone never completely clear bacteria; these chemicals will always require the immune system to help clear the remaining bacteria and infection. Some of the ways mAbs can disrupt bacterial function and survival are presented as examples in Table 1 below, and some of this knowledge originally comes from vaccine-based approaches.

2. Antibacterial Antibodies—Previous Success

From 2002, when the first fully human antibody was approved (HUMIRA®/adalimumab), to 2016, over 40 Hu-mAbs were approved by the United States Food and Drug Administration (FDA) for different diseases and treatments [11]. Most of these are related to cancer and autoimmune disease, and none were for bacterial infection. Of note, with respect to infectious disease, SYNAGIS® (palivizumab) was approved for respiratory syncytial virus RSV in 2004 [36]. In 2016, the first Hu-mAb for antibacterial treatment was approved by the FDA: Anthim® (obiltoxaximab), an injection to treat inhalational anthrax in combination with appropriate antibiotics (often ciprofloxacin). Anthim® was also approved to prevent inhalational anthrax when alternative therapies are not available or not appropriate via the Animal Rule for biothreat organisms [37]. The Animal Rule, put in place by the FDA in 2002, allows for the approval of a drug for biothreat organisms that, if untreated, leads to death or serious disability; therefore, safety and efficacy is based on the results in animal models that best represent the clinical indication being targeted [38]. Using the Animal Rule, efficacy of Anthim® was evaluated using New Zealand white rabbits [39]. Inhalation of Bacillus anthracis spores cause anthrax infection and, because of the ability of the spores to withstand harsh environments, spores can serve as a source for subsequent infection [40]. Another interesting study, performed after approval, showed that Anthim® could also prevent these types of infections, which further bolstered the product’s utility [40]. Later that year, in October of 2016, the FDA approved the second antibody for bacterial infection, Zinplava™ (bezlotoxumab), for Clostridum difficile infections in adults [41]. It is important to understand that this product does not protect from or treat initial or primary C. difficile infection, rather it was approved to reduce the recurrence of infection, which is often seen with C. difficile [41,42]. However, unlike Anthim®, which relied on just one main study in animals and the Animal Rule, Zinplava™ showed both safety and efficacy in multiple animal models [42,43]. FDA approval then followed the traditional path to approval including Phase 1, Phase 2, and Phase 3 trials [41].

3. Previous Failures Lead to Current Success

With the clinical success of Hu-mAbs for other diseases, it is surprising there are not more Hu-mAbs being made for bacterial infections. It is important to understand that research and production of antibodies can be expensive, and there have been some failures in the recent past that may have dampened the enthusiasm of researchers and investors that were considering Hu-mAbs for antibacterial development. One example is KB001-A, a mAb developed by KaloBios for P. aeruginosa infections. This product was made against the Type III secretion system (T3SS), which is required for P. aeruginosa pathogenesis [44]. Even though there were positive data in animal models [44,45] and the mAb was found to be safe in human patients [45], the product was not effective for patients with mechanical ventilation in a Phase 2 trial. Unfortunately, there have also been numerous failures with Hu-mAb treatments being developed for S. aureus, such as tefibazumab, which caused developers to question the monoclonal antibody approach for this bacterial species [46]. These antibodies showed promise in animals, but subsequently failed in Phase 2 efficacy trials [46]. However, there are lessons to be learned from these failures. Using just one antibody against one target, when bacteria secrete or have more than 200-400+ targets on their surface with presumed roles in virulence, may not be sufficient. Further, most approaches do not take into account that bacteria have different lifestyles, and therefore variable surface or secreted protein expression profiles, while residing within the host: vegetative, encapsulated or unencapsulated, biofilm associated, and intracellular, among others [46,47,48,49]. Finally, with respect to the mAb KB-001A, a diagnostic assay was not being used to properly identify patients with P. aeruginosa at the outset of the study. Because other bacterial species can also be the cause of pneumonia and sometimes infections are polymicrobial, KB-001A was only going to be effective with a subpopulation of patients making it difficult to achieve the threshold for success. However, examples below will highlight how companies are now addressing some of these issues like growth state and the inclusion of an onboard diagnostic with the clinical trial, which can lead to success.
Currently, there are 14 Hu-mAb products in development for nosocomial bacterial pathogens (Table 2). This list is the result of literature searches and publicly available company information; however, it is not exhaustive, as some Hu-mAb initiatives may have been unintentionally overlooked. The focus of the field has mainly been on S. aureus and P. aeruginosa, the most prevalent causes of disease with respect to the ESKAPEE pathogens in the Western world (making this more commercially viable), but some newer preclinical approaches have also targeted the difficult-to-treat species such as C. difficile and A. baumannii. All but one of the products target individual pathogens, making them narrow spectrum. The exception is the Hu-mAb being developed for biofilm by Trellis. This antibody targets DNABII, which is conserved amongst multiple bacteria and is required for biofilm formation as bacteria often release DNA into the extracellular milieu when establishing a biofilm [24]. If this product shows success in human trials, it could be very exciting to partner it with antibiotics that are often stymied by biofilms.
Of the Hu-mAbs being clinically evaluated, the products from AstraZeneca PLC (formerly MedImmune) have been extensively evaluated in a number of preclinical models. MEDI4893 was developed for S. aureus and has been shown to be protective in multiple models of pneumonia including ferret, rabbit and mouse [50,51]. The antibody targets the secreted alpha-toxin and prevents the bacterium’s ability to cause apoptosis in cells, which, in turn, prevents lysis and tissue necrosis caused by S. aureus infection. Further, the antibody has been shown to prevent necrosis in other clinical indications such as surgical site and wound infection models [52,53,54]. Recently, MEDI4893 was shown to improve lung function in patients with S. aureus infections in a successful Phase 2 trial, and the results were presented earlier this year (ASM Microbe, 2019, San Francisco). The trial design was relatively simple, where the product was administered intravenously (i.v.) at 2000 or 5000 mg on the first day of enrollment, and was compared to a placebo control. Patients were excluded if they were given antibiotics, and only enrolled if positive for S. aureus. The company is now looking to out-license the product as AstraZeneca PLC has stopped actively resourcing its antibacterial Hu-mAb group.
Aridis Pharmaceuticals, Inc. is also targeting S. aureus and the secreted alpha-toxin with AR-301 (Salvecin®) (Table 2), but there are no published data around this product and the Phase 2 efficacy trial was designed as an adjunct therapy to be used with standard-of-care antibiotics. Like MEDI4893, patients were administered antibody i.v. once upon the day of trial enrollment, however the dose was 20 mg/kg. AR-301 was also successful in a Phase 2 trial for hospital-acquired pneumonia (HAP)/ventilator-associated pneumonia (VAP), and patients were given a 20 mg/kg dose i.v. upon trial enrollment. According to the company, “Patients treated with AR-301 consistently demonstrated less time spent under mechanical ventilation and higher rates of S. aureus eradication as compared to those treated with antibiotics alone. AR-301 was deemed to be safe and well tolerated.” AR-301 has been granted Fast Track designation by the FDA and was given an orphan drug designation by the European Medicines Agency (EMA), and the company is currently enrolling patients for a Phase 3 trial according to its website and clinicaltrials.gov The future of both these two S. aureus products, MEDI4893 and AR-301, could pave the way for other Hu-mAb antibacterial treatments especially with respect to Gram-positive pathogens.
Another product from AstraZeneca PLC (formerly MedImmune) is MEDI3902, which is a bi-specific antibody targeting both the T3SS (PcrV), the same target as the KaloBios product, but in addition, also targets a surface polysaccharide (Psl) [55], which is essential for biofilm formation and virulence of P. aeruginosa [56,57]. MEDI3902 was successful in a Phase I trial [58] and was evaluated for HAP/VAP infections caused by P. aeruginosa in a Phase II trial. Like the other Hu-mAb from AstraZeneca PLC, this product was also dosed i.v., excluded patients on antibiotics, and only patients positive for P. aeruginosa were enrolled. Multiple animal models led to the development of this Hu-mAb [55,59], and aside from the anti-virulence mechanism of action, it was recently shown in vitro that MEDI3902 can steer neutrophils through a “dead zone” to reach and destroy biofilm generated by P. aeruginosa [60], which highlights another positive aspect of infection remediation.
Aridis also has an anti-P. aeruginosa product: AR101 (Aerumab™). This product specifically targets the O-antigen of the O11 serotype of lipopolysaccharide (LPS), which the company states “represents 22% of all P. aeruginosa lung infections”. It was successful in both Phase 1 and Phase 2a trials for HAP/VAP infections. Like AR301, this product was evaluated as adjunct with antibiotics, and has “primary safety endpoints and showed a consistent trend toward improvement in mortality, shorter time to clinical cure of pneumonia, shorter time on mechanical ventilation, and fewer days in the ICU as compared to standard of care antibiotics-alone.” However, this is in stark contrast to Aridis’ AR105 product that failed its Phase 2 trial in September 2019. However, the target of this antibody is alginate, which is required for biofilm formation [61], but it is unclear what other roles this polysaccharide may have with respect to pathogenesis of the organism.
The future for these P. aeruginosa products is unclear. As with the other AstraZeneca PLC product, MEDI3902 will likely have to be out-licensed to a large company willing to invest in a Phase 3 trial, or perhaps the Biomedical Advanced Research and Development Authority (BARDA) will support a trial as this government agency has a track record of supporting late-stage antibacterial development efforts. Being specific to just one set of P. aeruginosa strains, AR101 may be of limited value. While the O11 serotype has been the most virulent and resistant with increased mortality in patients, it may be hard to make the case for limited applicability, as in this recent study, O11 strains only made up 15% of the patients in the trial [62]. In contrast, MEDI3902 recognized PcrV and Psl in the most globally distributed isolates [63].
Another interesting approach being pushed forward by Roche Ltd. (formerly Genentech) is RG7861 (anti-S. aureus TAC, DSTA4637S), which is a THIOMAB™ antibiotic conjugate (TAC) that consists of a human monoclonal antibody directed against a S. aureus protein conjugated to an antibiotic. Anti-S. aureus TAC is designed to bind to the surface of S. aureus bacteria, thereby putting the antibiotic in close proximity to its target in order to enhance killing. The company also reported positive results against intracellular S. aureus, which is an important aspect of the bacterium’s pathogenic lifestyle [64]. A Phase I clinical trial is currently evaluating RG7861 for safety. Because this antibody–antibiotic combination is the first of its kind in the clinic, many will be curious of the outcome of the Phase I and subsequent trials [65]. It should also be noted that antibiotic–antibody conjugate chemistry is a difficult endeavor. Development of this product included identifying the best antibiotic to use, identifying the linker between the antibody and antibiotic, and identifying the right place on the antibody for the linkage [66,67]. However, one could certainly argue that the work is worth the effort, as S. aureus is responsible for thousands of deaths per year, and the product was superior to vancomycin in vivo [67].

4. Future and Conclusions

The rise of AMR is projected to be a tremendous problem, where deaths resulting from bacterial infection, estimated to be as many as 10 million/year by 2050, will surpass deaths from cancer and heart disease combined [68]. As resistance grows in ESKAPEE pathogens [69], innovative, effective treatments are needed, and Hu-mAbs will be an important part of the solution. It is hoped that the successes discussed in this review and advancement of antibody approaches [14] will stimulate the development of more Hu-mAbs to treat bacterial infections. Although there can be large upfront costs to develop Hu-mAbs against bacteria, the success stories of AstraZeneca/MedImmune and Aridis Pharmaceuticals, Inc. products in clinical trials show that investment in Hu-mAbs can lead to development of effective therapeutics. While some products are being used as a standalone therapeutic or as adjuvants with standard-of-care antibiotics to improve patient outcome, it should be noted that these same products could also be used as a prophylactic to prevent infection. A clinical trial has not been designed yet specifically for this purpose, but in the future, it could certainly be another application for Hu-mAb antibacterial technology. One could envision running a trial for patients who are at risk for surgical site infections that do or do not receive Hu-mAbs along with standard-of-care antibiotics, and then monitoring for development of infection.
One drawback of Hu-mAb development is the cost. A significant amount of research is required upfront to identify the best targets on the bacterial surface and sift through many positive binding antibodies to find the most effective at remediating bacterial infection via different mechanisms of action (Table 1). Once found, scaling up can also be a financial burden, as the primary method is CHO cells, and it can cost $10,000–$20,000 per gram of material [70]. However, improvements in culturing and processing are being made to improve yields, and plant-based production is also possible, which can also improve yield [70]. Given that the cost of an MDR infection and staying in the hospital can run as much as $50,000–$100,000 per patient [68], an investment in Hu-mAbs development appears more reasonable. In relation to small molecules, it should be noted that a study found that the average costs were $29,941 per patient treated with meropenem-vaborbactam or $32,294 with ceftazidime-avibactam [71]. Newer antibiotics will demand a higher cost no matter whether antibody or small molecule.
In the future, more developmental, preclinical assays will help to serve better antibody design. Critics have long said that Hu-mAbs will still be subject to resistance by bacteria just like antibiotics, but it is important to understand there is a distinct difference. Antibiotics directly kill bacteria, placing genetic pressure on the organism to survive by mutation; survivors, therefore, are resistant to the drug and are left to reestablish infection in the host. However, because antibodies often target virulence factors and not proteins required for survival, a mutation of these targets is likely to make bacteria non-virulent or less virulent, possibly subject to enhanced immune system clearance. A recent study supported this idea and showed that when the alpha-toxin epitope for MEDI4839 was mutagenized, S. aureus had a reduced fitness cost [72]. Another criticism of this approach is the narrow spectrum of the products, as they largely only target one bacterial species; however, this limitation has been somewhat overcome with the use of onboard diagnostics with clinical trials, where a complement diagnostic antibody or PCR test is used to identify patients infected with a particular bacterial species upfront before treatment. As diagnostics improve, this should facilitate the use of Hu-mAbs as treatments. Further, narrow-spectrum therapeutics hold an advantage in avoiding disrupting the normal microbiome of the patient. Finally, there are some that have said that the efficacy of Hu-mAb therapeutics is not significant enough but, again, this criticism will be addressed in the future as technology improves and more targets are added either via a cocktail or by multivalent antibody engineering. It has already been shown that the addition of just one target with a bispecific antibody improves Hu-mAb efficacy [55,59,60]. Further, there are now pentavalent antibodies and mAb cocktails that are also being explored to improve efficacy [73,74]. One can certainly envision a future, 5–10 years from now, where there could be a Hu-mAb antibody product, cocktail or multivalent molecule, which is developed for each bacterial pathogen that causes difficult-to-treat infections. The antibody mixture would ideally use different mechanisms of action, such as inhibiting bacterial growth or spread while also enhancing clearance by the immune system, (Table 1) thereby putting more pressure on the bacterium. While it is possible that some do not see the need for the large, upfront investment in the defining targets when small molecules are still a cheaper approach, the lack of new drug classes and scaffolds have made finding new small-molecule antibiotics a difficult prospect [5]. In contrast, improvements in antibody technology, coupled with clinical need, could certainly drive more research and funding to the Hu-mAb approach. Ultimately, it could become a faster route than small-molecule development and perhaps revolutionize infectious medicine as was proffered years ago [75].

Author Contributions

D.V.Z. conceptualized the study, conducted the literature review, and wrote the review with M.K.M., who assisted with writing and revision. All authors have read and agreed to the draft version of the manuscript.

Funding

D.Z. and M.M. are funded by the Military Infectious Diseases Research Program of the U.S. Department of Defense.

Conflicts of Interest

The authors declare that the preparation of the manuscript was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Material has been reviewed by the Walter Reed Army Institute of Research (WRAIR). There is no objection to its presentation and/or publication. The opinions and assertions contained herein are the private ones of the authors and are not to be construed as official or reflecting the views of the Department of Defense, the Department of the Army, or any other agency of the U.S. Government.

References

  1. Hubbert, W.R. Comparative Statistics of Antitoxin Horses. A Study of the Records of One Hundred Horses Immunized To Diphtheria Toxin, With Composite of Curves. J. Exp. Med. 1905, 7, 176–182. [Google Scholar] [CrossRef] [PubMed]
  2. Crane, L. In 1925, a Remote Town Was Saved from Lethal Disease by Dogs. Available online: http://www.bbc.com/earth/story/20161014-in-1925-a-remote-town-was-saved-from-lethal-disease-by-dogs (accessed on 13 January 2020).
  3. Casadevall, A.; Scharff, M.D. Serum Therapy Revisited: Animal Models of Infection and Development of Passive Antibody Therapy. Antimicrob. Agents Chemother. 1994, 38, 1695–1702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Casadevall, A. Antibody-based therapies for emerging infectious diseases. Emerg. Infect. Dis. 1996, 2, 200–208. [Google Scholar] [CrossRef] [PubMed]
  5. Lewis, K. Platforms for antibiotic discovery. Nat. Rev. Drug Discov. 2013, 12, 371–387. [Google Scholar] [CrossRef] [PubMed]
  6. Köhler, G.; Milstein, C. Continuous Cultures of Fused Cells Secreting Antibody of predefined specificity. Nature 1975, 256, 495–497. [Google Scholar] [CrossRef] [PubMed]
  7. Zurawski, V.R., Jr.; Haber, E.; Black, P.H. Production of Antibody to Tetanus Toxoid by Continuous Human Lymphoblastoid Cell Lines. Science 1978, 199, 1439–1441. [Google Scholar] [CrossRef]
  8. Riechmann, L.; Clark, M.; Waldmann, H.; Winter, G. Reshaping human antibodies for therapy. Nature 1988, 332, 323–327. [Google Scholar] [CrossRef]
  9. Wright, A.; Shin, S.; Morrison, S.L. Genetically engineered antibodies: Progress and prospects. Crit. Rev. Immunol. 1992, 12, 125–168. [Google Scholar]
  10. Robert-Guroff, M.; Brown, M.; Gallo, R.C. HTLV-III-neutralizing Antibodies in Patients With AIDS and AIDS-related Complex. Nature 1985, 316, 72–74. [Google Scholar] [CrossRef]
  11. Hey, A. History and Practice: Antibodies in Infectious Diseases. Microbiol. Spectr. 2015, 3, 2. [Google Scholar] [CrossRef] [Green Version]
  12. Glanville, J.; Zhai, W.; Berka, J.; Telman, D.; Huerta, G.; Mehta, G.R.; Ni, I.; Mei, L.; Sundar, P.D.; Day, G.M.R. Precise determination of the diversity of a combinatorial antibody library gives insight into the human immunoglobulin repertoire. Proc. Natl. Acad. Sci. USA 2009, 106, 20216–20221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Liu, H.; Nowak, C.; Andrien, B.; Shao, M.; Ponniah, G.; Neill, A. Impact of IgG Fc-Oligosaccharides on Recombinant Monoclonal Antibody Structure, Stability, Safety, and Efficacy. Biotechnol. Prog. 2017, 33, 1173–1181. [Google Scholar] [CrossRef]
  14. Walker, L.M.; Burton, D.R. Passive immunotherapy of viral infections: ‘super-antibodies’ enter the fray. Nat. Rev. Immunol. 2018, 18, 297–308. [Google Scholar] [CrossRef] [PubMed]
  15. Liikanen, I.; Tähtinen, S.; Guse, K.; Gutmann, T.; Savola, P.; Oksanen, M.; Kanerva, A.; Hemminki, A. Oncolytic Adenovirus Expressing Monoclonal Antibody Trastuzumab for Treatment of HER2-Positive Cancer. Mol. Cancer Ther. 2016, 15, 2259–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Limberis, M.P. Intranasal antibody gene transfer in mice and ferrets elicits broad protection against pandemic influenza. Sci. Transl. Med. 2013, 5, 187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Flingai, S.; Plummer, E.M.; Patel, A.; Shresta, S.; Mendoza, J.M.; Broderick, K.E.; Sardesai, N.Y.; Muthumani, K.; Weiner, D.B. Protection Against Dengue Disease by Synthetic Nucleic Acid Antibody Prophylaxis/Immunotherapy. Sci. Rep. 2015, 5, 12616. [Google Scholar] [CrossRef] [PubMed]
  18. Patel, A.; DiGiandomenico, A.; Keller, A.E.; Smith, T.R.F.; Park, D.H.; Ramos, S.; Schultheis, K.; Elliott, S.T.C.; Mendoza, J.; Broderick, K.E. An Engineered Bispecific DNA-encoded IgG Antibody Protects against Pseudomonas aeruginosa in a Pneumonia Challenge Model. Nat. Commun. 2017, 8, 637. [Google Scholar] [CrossRef] [Green Version]
  19. Heesterbeek, D.A.C.; Angelier, M.L.; Harrison, R.A.; Rooijakkers, S.H.M. Complement and Bacterial Infections: From Molecular Mechanisms to Therapeutic Applications. J. Innate Immun. 2018, 10, 455–464. [Google Scholar] [CrossRef]
  20. Storek, K.M.; Auerbach, M.R.; Shi, H.; Garcia, N.K.; Sun, D.; Nickerson, N.N.; Vij, R.; Lin, Z.; Chiang, N.; Schneider, K.; et al. Monoclonal antibody targeting the β-barrel assembly machine of Escherichia coli is bactericidal. Proc. Natl. Acad. Sci. USA 2018, 115, 3692–3697. [Google Scholar] [CrossRef] [Green Version]
  21. Dunn-Siegrist, I.; Leger, O.; Daubeuf, B.; Poitevin, Y.; Dépis, F.; Herren, S.; Kosco-Vilbois, M.; Dean, Y.; Pugin, J.; Elson, G. Pivotal involvement of Fcgamma receptor IIA in the neutralization of lipopolysaccharide signaling via a potent novel anti-TLR4 monoclonal antibody 15C1. J. Biol. Chem. 2007, 282, 34817–34827. [Google Scholar] [CrossRef] [Green Version]
  22. Vij, R.; Lin, Z.; Chiang, N.; Vernes, J.M.; Storek, K.M.; Park, S.; Chan, J.; Meng, Y.G.; Comps-Agrar, L.; Luan, P.; et al. A targeted boost-and-sort immunization strategy using Escherichia coli BamA identifies rare growth inhibitory antibodies. Sci. Rep. 2018, 8, 7136. [Google Scholar] [CrossRef] [PubMed]
  23. Tursi, S.A.; Puligedda, R.D.; Szabo, P.; Nicastro, L.K.; Miller, A.L.; Qiu, C.; Gallucci, S.; Relkin, N.R.; Buttaro, B.A.; Dessain, S.K.; et al. Salmonella Typhimurium biofilm disruption by a human antibody that binds a pan-amyloid epitope on curli. Nat. Commun. 2020, 11, 1007. [Google Scholar] [CrossRef] [Green Version]
  24. Novotny, L.A.; Jurcisek, J.A.; Goodman, S.D.; Bakaletz, L.O. Monoclonal antibodies against DNA-binding tips of DNABII proteins disrupt biofilms in vitro and induce bacterial clearance in vivo. EBioMedicine 2016, 10, 33–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kaizuka, K.; Hosogi, Y.; Hayakawa, M.; Shibata, Y.; Abiko, Y. Human monoclonal antibody inhibits Porphyromonas gingivalis hemagglutinin activity. J. Periodontol. 2003, 74, 38–43. [Google Scholar] [CrossRef] [PubMed]
  26. Mohamed, W.; Sethi, S.; Darji, A.; Mraheil, M.A.; Hain, T.; Chakraborty, T. Antibody targeting the ferritin-like protein controls Listeria infection. Infect. Immun. 2010, 78, 3306–3314. [Google Scholar] [CrossRef] [Green Version]
  27. Bennett, M.R.; Dong, J.; Bombardi, R.G.; Soto, C.; Parrington, H.M.; Nargi, R.S.; Schoeder, C.T.; Nagel, M.B.; Schey, K.L.; Meiler, J.; et al. Human VH1-69 Gene-Encoded Human Monoclonal Antibodies against Staphylococcus aureus IsdB Use at Least Three Distinct Modes of Binding to Inhibit Bacterial Growth and Pathogenesis. mBio 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  28. Singh, K.V.; Pinkston, K.L.; Gao, P.; Harvey, B.R.; Murray, B.E. Anti-Ace monoclonal antibody reduces Enterococcus faecalis aortic valve infection in a rat infective endocarditis model. Pathog. Dis. 2018, 76, 8. [Google Scholar] [CrossRef]
  29. Aye, R.; Weldearegay, Y.B.; Lutta, H.O.; Chuma, F.; Pich, A.; Jores, J.; Meens, J.; Naessens, J. Identification of targets of monoclonal antibodies that inhibit adhesion and growth in Mycoplasma mycoides subspecies mycoides. Vet. Immunol. Immunopathol. 2018, 204, 11–18. [Google Scholar] [CrossRef]
  30. Aguilar, J.L.; Varshney, A.K.; Pechuan, X.; Dutta, K.; Nosanchuk, J.D.; Fries, B.C. Monoclonal antibodies protect from Staphylococcal Enterotoxin K (SEK) induced toxic shock and sepsis by USA300 Staphylococcus aureus. Virulence 2017, 8, 741–750. [Google Scholar] [CrossRef] [Green Version]
  31. Iwamoto, R.; Senoh, H.; Okada, Y.; Uchida, T.; Mekada, E. An antibody that inhibits the binding of diphtheria toxin to cells revealed the association of a 27-kDa membrane protein with the diphtheria toxin receptor. J. Biol. Chem. 1991, 266, 20463–20469. [Google Scholar]
  32. Diago-Navarro, E.; Calatayud-Baselga, I.; Sun, D.; Khairallah, C.; Mann, I.; Ulacia-Hernando, A.; Sheridan, B.; Shi, M.; Fries, B.C. Antibody-Based Immunotherapy To Treat and Prevent Infection with Hypervirulent Klebsiella pneumoniae. Clin. Vaccine Immunol. 2017, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Nielsen, T.B.; Pantapalangkoor, P.; Luna, B.M.; Bruhn, K.W.; Yan, J.; Dekitani, K.; Hsieh, S.; Yeshoua, B.; Pascual, B.; Vinogradov, E.; et al. Monoclonal Antibody Protects Against Acinetobacter baumannii Infection by Enhancing Bacterial Clearance and Evading Sepsis. J. Infect. Dis. 2017, 216, 489–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gulati, S.; Beurskens, F.J.; de Kreuk, B.J.; Roza, M.; Zheng, B.; DeOliveira, R.B.; Shaughnessy, J.; Nowak, N.A.; Taylor, R.P.; Botto, M.; et al. Complement alone drives efficacy of a chimeric antigonococcal monoclonal antibody. PLoS Biol. 2019, 17, e3000323. [Google Scholar] [CrossRef] [Green Version]
  35. Visan, L.; Rouleau, N.; Proust, E.; Peyrot, L.; Donadieu, A.; Ochs, M. Antibodies to PcpA and PhtD protect mice against Streptococcus pneumoniae by a macrophage- and complement-dependent mechanism. Hum. Vaccines Immunother. 2018, 14, 489–494. [Google Scholar] [CrossRef] [Green Version]
  36. Resch, B. Product Review on the Monoclonal Antibody Palivizumab for Prevention of Respiratory Syncytial Virus Infection. Hum. Vaccines Immunother. 2017, 13, 2138–2149. [Google Scholar] [CrossRef] [Green Version]
  37. Nagy, C.F.; Leach, T.S.; Hoffman, J.H.; Czech, A.; Carpenter, S.E.; Guttendorf, R. Pharmacokinetics and Tolerability of Obiltoxaximab: A Report of 5 Healthy Volunteer Studies. Clin. Ther. 2016, 38, 2083–2097. [Google Scholar] [CrossRef] [Green Version]
  38. Allio, T. The FDA Animal Rule and its role in protecting human safety. Exp. Opin. Drug Saf. 2018, 17, 971–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Biron, B.; Beck, K.; Dyer, D.; Mattix, M.; Twenhafel, N.; Nalca, A. Efficacy of ETI-204 monoclonal antibody as an adjunct therapy in a New Zealand white rabbit partial survival model for inhalational anthrax. Antimicrob. Agents Chemother. 2015, 59, 2206–2214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Henning, L.N.; Carpenter, S.; Stark, G.V.; Serbina, N.V. Development of Protective Immunity in New Zealand White Rabbits Challenged with Bacillus anthracis Spores and Treated with Antibiotics and Obiltoxaximab, a Monoclonal Antibody against Protective Antigen. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef] [Green Version]
  41. Wilcox, M.H.; Gerding, D.N.; Poxton, I.R.; Kelly, C.; Nathan, R.; Birch, T.; Cornely, O.A.; Rahav, G.; Bouza, E.; Lee, C.; et al. Bezlotoxumab for Prevention of Recurrent Clostridium difficile Infection. N. Engl. J. Med. 2017, 376, 305–317. [Google Scholar] [CrossRef]
  42. Yang, Z.; Ramsey, J.; Hamza, T.; Zhang, Y.; Li, S.; Yfantis, H.G.; Lee, D.; Hernandez, L.D.; Seghezzi, W.; Furneisen, J.M.; et al. Mechanisms of protection against Clostridium difficile infection by the monoclonal antitoxin antibodies actoxumab and bezlotoxumab. Infect. Immun. 2015, 83, 822–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Džunková, M.; D’Auria, G.; Xu, H.; Huang, J.; Duan, Y.; Moya, A.; Kelly, C.P.; Chen, X. The Monoclonal Antitoxin Antibodies (Actoxumab-Bezlotoxumab) Treatment Facilitates Normalization of the Gut Microbiota of Mice with Clostridium difficile Infection. Front. Cell Infect. Microbiol. 2016, 6, 119. [Google Scholar] [CrossRef] [PubMed]
  44. Baer, M.; Sawa, T.; Flynn, P.; Luehrsen, K.; Martinez, D.; Wiener-Kronish, J.P.; Yarranton, G.; Bebbington, C. An engineered human antibody fab fragment specific for Pseudomonas aeruginosa PcrV antigen has potent antibacterial activity. Infect. Immun. 2009, 77, 1083–1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jain, R.; Beckett, V.V.; Konstan, M.W.; Accurso, F.J.; Burns, J.L.; Mayer-Hamblett, N.; Milla, C.; VanDevanter, D.R.; Chmiel, J.F. KB001-A Study Group. KB001-A, a novel anti-inflammatory, found to be safe and well-tolerated in cystic fibrosis patients infected with Pseudomonas aeruginosa. J. Cyst. Fibros. 2018, 17, 484–491. [Google Scholar] [CrossRef] [PubMed]
  46. Sause, W.E.; Buckley, P.T.; Strohl, W.R.; Lynch, A.S.; Torres, V.J. Antibody-Based Biologics and Their Promise to Combat Staphylococcus aureus Infections. Trends Pharmacol. Sci. 2016, 37, 231–241. [Google Scholar] [CrossRef] [Green Version]
  47. Del Pozo, J.L. Biofilm-related disease. Exp. Rev. Anti Infect. Ther. 2018, 16, 51–65. [Google Scholar] [CrossRef]
  48. Gollan, B.; Grabe, G.; Michaux, C.; Helaine, S. Bacterial Persisters and Infection: Past, Present, and Progressing. Ann. Rev. Microbiol. 2019, 73, 359–385. [Google Scholar] [CrossRef]
  49. Ryser, S.; Tenorio, E.; Estellés, A.; Kauvar, L.M. Human antibody repertoire frequently includes antibodies to a bacterial biofilm associated protein. PLoS ONE 2019, 14, e0219256. [Google Scholar] [CrossRef]
  50. Diep, B.A.; Hilliard, J.J.; Le, V.T.; Tkaczyk, C.; Le, H.N.; Tran, V.G.; Rao, R.L.; Dip, E.C.; Pereira-Franchi, E.P.; Cha, P.; et al. Targeting Alpha Toxin To Mitigate Its Lethal Toxicity in Ferret and Rabbit Models of Staphylococcus aureus Necrotizing Pneumonia. Antimicrob. Agents Chemother. 2017, 61. [Google Scholar] [CrossRef] [Green Version]
  51. Hua, L.; Cohen, T.S.; Shi, Y.; Datta, V.; Hilliard, J.J.; Tkaczyk, C.; Suzich, J.; Stover, C.K.; Sellman, B.R. MEDI4893* Promotes Survival and Extends the Antibiotic Treatment Window in a Staphylococcus aureus Immunocompromised Pneumonia Model. Antimicrob. Agents Chemother. 2015, 59, 4526–4532. [Google Scholar] [CrossRef] [Green Version]
  52. Hilliard, J.J.; Datta, V.; Tkaczyk, C.; Hamilton, M.; Sadowska, A.; Jones-Nelson, O.; O’Day, T.; Weiss, W.J.; Szarka, S.; Nguyen, V.; et al. Anti-alpha-toxin monoclonal antibody and antibiotic combination therapy improves disease outcome and accelerates healing in a Staphylococcus aureus dermonecrosis model. Antimicrob. Agents Chemother. 2015, 59, 299–309. [Google Scholar] [CrossRef] [Green Version]
  53. Ortines, R.V.; Wang, Y.; Liu, H.; Dikeman, D.A.; Pinsker, B.L.; Miller, R.J.; Kim, S.E.; Ackerman, N.E.; Rizkallah, J.F.; Marcello, L.T.; et al. Efficacy of a Multimechanistic Monoclonal Antibody Combination against Staphylococcus aureus Surgical Site Infections in Mice. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
  54. Ortines, R.V.; Liu, H.; Cheng, L.I.; Cohen, T.S.; Lawlor, H.; Gami, A.; Wang, Y.; Dillen, C.A.; Archer, N.K.; Miller, R.J.; et al. Neutralizing Alpha-Toxin Accelerates Healing of Staphylococcus aureus-Infected Wounds in Nondiabetic and Diabetic Mice. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef] [Green Version]
  55. DiGiandomenico, A.; Keller, A.E.; Gao, C.; Rainey, G.J.; Warrener, P.; Camara, M.M.; Bonnell, J.; Fleming, R.; Bezabeh, B.; Dimasi, N.; et al. A multifunctional bispecific antibody protects against Pseudomonas aeruginosa. Sci. Transl. Med. 2014, 6, 262. [Google Scholar] [CrossRef]
  56. Zegans, M.E.; DiGiandomenico, A.; Ray, K.; Naimie, A.; Keller, A.E.; Stover, C.K.; Lalitha, P.; Srinivasan, M.; Acharya, N.R.; Lietman, T.M. Association of Biofilm Formation, Psl Exopolysaccharide Expression, and Clinical Outcomes in Pseudomonas aeruginosa Keratitis: Analysis of Isolates in the Steroids for Corneal Ulcers Trial. JAMA Ophthalmol. 2016, 134, 383–389. [Google Scholar] [CrossRef] [Green Version]
  57. Byrd, M.S.; Pang, B.; Hong, W.; Waligora, E.A.; Juneau, R.A.; Armbruster, C.E.; Weimer, K.E.; Murrah, K.; Mann, E.E.; Lu, H.; et al. Direct evaluation of Pseudomonas aeruginosa biofilm mediators in a chronic infection model. Infect. Immun. 2011, 79, 3087–3095. [Google Scholar] [CrossRef] [Green Version]
  58. Ali, S.O.; Yu, X.Q.; Robbie, G.J.; Wu, Y.; Shoemaker, K.; Yu, L.; DiGiandomenico, A.; Keller, A.E.; Anude, C.; Hernandez-Illas, M.; et al. Phase 1 study of MEDI3902, an investigational anti-Pseudomonas aeruginosa PcrV and Psl bispecific human monoclonal antibody, in healthy adults. Clin. Microbiol. Infect. 2019, 25, 629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Le, H.N.; Tran, V.G.; Vu, T.T.T.; Gras, E.; Le, V.T.M.; Pinheiro, M.G.; Aguiar-Alves, F.; Schneider-Smith, E.; Carter, H.C.; Sellman, B.R.; et al. Treatment Efficacy of MEDI3902 in Pseudomonas aeruginosa Bloodstream Infection and Acute Pneumonia Rabbit Models. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Thanabalasuriar, A.; Scott, B.N.V.; Peiseler, M.; Willson, M.E.; Zeng, Z.; Warrener, P.; Keller, A.E.; Surewaard, B.G.J.; Dozier, E.A.; Korhonen, J.T.; et al. Neutrophil Extracellular Traps Confine Pseudomonas aeruginosa Ocular Biofilms and Restrict Brain Invasion. Cell Host Microbe 2019, 25, 526–536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Ma, L.; Conover, M.; Lu, H.; Parsek, M.R.; Bayles, K.; Wozniak, D.J. Assembly and development of the Pseudomonas aeruginosa biofilm matrix. PLoS Pathog. 2009, 5, e1000354. [Google Scholar] [CrossRef] [Green Version]
  62. Recio, R.; Mancheño, M.; Viedma, E.; Villa, J.; Orellana, M.Á.; Lora-Tamayo, J.; Chaves, F. Predictors of Mortality in Bloodstream Infections Caused by Pseudomonas aeruginosa and Impact of Antimicrobial Resistance and Bacterial Virulence. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  63. Tabor, D.E.; Oganesyan, V.; Keller, A.E.; Yu, L.; McLaughlin, R.E.; Song, E.; Warrener, P.; Rosenthal, K.; Esser, M.; Qi, Y.; et al. Pseudomonas aeruginosa PcrV and Psl, the Molecular Targets of Bispecific Antibody MEDI3902, Are Conserved Among Diverse Global Clinical Isolates. J. Infect. Dis. 2018, 218, 1983–1994. [Google Scholar] [CrossRef] [PubMed]
  64. Abel, J.; Goldmann, O.; Ziegler, C.; Höltje, C.; Smeltzer, M.S.; Cheung, A.L.; Bruhn, D.; Rohde, M.; Medina, E. Staphylococcus aureus evades the extracellular antimicrobial activity of mast cells by promoting its own uptake. J. Innate Immun. 2011, 3, 495–507. [Google Scholar] [CrossRef]
  65. Zhou, C.; Lehar, S.; Gutierrez, J.; Rosenberger, C.M.; Ljumanovic, N.; Dinoso, J.; Koppada, N.; Hong, K.; Baruch, A.; Carrasco-Triguero, M.; et al. Pharmacokinetics and pharmacodynamics of DSTA4637A: A novel THIOMAB™ antibody antibiotic conjugate against Staphylococcus aureus in mice. MAbs 2016, 8, 1612–1619. [Google Scholar] [CrossRef] [Green Version]
  66. Linghu, X.; Segraves, N.L.; Abramovich, I.; Wong, N.; Müller, B.; Neubauer, N.; Fantasia, S.; Rieth, S.; Bachmann, S.; Jansen, M.; et al. Highly Efficient Synthesis of a Staphylococcus aureus Targeting Payload to Enable the First Antibody-Antibiotic Conjugate. Chemistry 2018, 24, 2837–2840. [Google Scholar] [CrossRef] [PubMed]
  67. Lehar, S.M.; Pillow, T.; Xu, M.; Staben, L.; Kajihara, K.K.; Vandlen, R.; DePalatis, L.; Raab, H.; Hazenbos, W.L.; Morisaki, J.H.; et al. Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature 2015, 527, 323–328. [Google Scholar] [CrossRef] [PubMed]
  68. O’Neill, J. Tackling Drug-Resistant Infections Globally: Final Report and Recommendations. 2016. Available online: https://amrreview.org/sites/default/files/160525_Final%20paper_with%20cover.pdf (accessed on 30 December 2019).
  69. World Health Organization. Global Action Plan on Antimicrobial Resistance. 2015. Available online: https://apps.who.int/iris/bitstream/handle/10665/193736/9789241509763_eng.pdf?sequence=1&isAllowed=y (accessed on 30 December 2019).
  70. Buyel, J.F.; Twyman, R.M.; Fischer, R. Very-large-scale production of antibodies in plants: The biologization of manufacturing. Biotechnol. Adv. 2017, 35, 458–465. [Google Scholar] [CrossRef] [PubMed]
  71. Simon, M.S.; Sfeir, M.M.; Calfee, D.P.; Satlin, M.J. Cost-effectiveness of ceftazidime-avibactam for treatment of carbapenem-resistant Enterobacteriaceae bacteremia and pneumonia. Antimicrob. Agents Chemother. 2019. [Google Scholar] [CrossRef] [PubMed]
  72. Tkaczyk, C.; Semenova, E.; Shi, Y.Y.; Rosenthal, K.; Oganesyan, V.; Warrener, P.; Stover, C.K.; Sellman, B.R. Alanine scanning mutagenesis of the MEDI4839 (Suvratoxumab) epitope reduces alpha toxin lytic activity in vitro and S. aureus fitness in infection models. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef] [Green Version]
  73. Riazi, A.; Strong, P.C.; Coleman, R.; Chen, W.; Hirama, T.; van Faassen, H.; Henry, M.; Logan, S.M.; Szymanski, C.M.; Mackenzie, R.; et al. Pentavalent single-domain antibodies reduce Campylobacter jejuni motility and colonization in chickens. PLoS ONE 2013, 8, e83928. [Google Scholar] [CrossRef] [Green Version]
  74. Vu, T.T.T.; Nguyen, N.T.Q.; Tran, V.G.; Gras, E.; Mao, Y.; Jung, D.H.; Tkaczyk, C.; Sellman, B.R.; Diep, B.A. Protective Efficacy of Monoclonal Antibodies Neutralizing Alpha-Hemolysin and Bicomponent Leukocidins in Rabbit Model of Staphylococcus aureus Necrotizing Pneumonia. Antimicrob. Agents Chemother. 2019, 64. [Google Scholar] [CrossRef]
  75. DiGiandomenico, A.; Sellman, B.R. Antibacterial monoclonal antibodies: The next generation? Curr. Opin. Microbiol. 2015, 27, 78–85. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Balto (left), one of the heroes of the race to deliver the antitoxin serum to Nome, AK. Upon his death, he was mounted and can still be found at the Cleveland Museum of Natural History in Cleveland, OH. However, Togo (right) was the true hero of the relay, running over 250 miles in 3 days. He can be found at the Trail Sled Dog Race Headquarters museum in Wasilla, Alaska.
Figure 1. Balto (left), one of the heroes of the race to deliver the antitoxin serum to Nome, AK. Upon his death, he was mounted and can still be found at the Cleveland Museum of Natural History in Cleveland, OH. However, Togo (right) was the true hero of the relay, running over 250 miles in 3 days. He can be found at the Trail Sled Dog Race Headquarters museum in Wasilla, Alaska.
Antibiotics 09 00155 g001
Table 1. How antibodies can disrupt bacterial infection.
Table 1. How antibodies can disrupt bacterial infection.
Antibacterial MechanismReference
Bactericidal[20,22]
Biofilm[23,24]
Iron acquisition[25,26,27]
Attachment/Adhesion[28,29]
Anti-toxin/Anti-virulence[30,31]
Opsonophagocytosis[32,33]
Complement[34,35]
Table 2. Companies currently pursuing Hu-mAb therapy for bacterial infections caused by ESKAPEE pathogens and Clostridum difficile—products and stage of development.
Table 2. Companies currently pursuing Hu-mAb therapy for bacterial infections caused by ESKAPEE pathogens and Clostridum difficile—products and stage of development.
NameBacterial Species TargetedCompanyDevelopment Phase
AR301Staphylococcus aureusAridis PharmaceuticalsPhase 2 Complete
Ongoing Phase 3
MEDI4893Staphylococcus aureusMedimmunePhase 2 Complete
MEDI3902Pseudomonas aeruginosaMedimmunePhase 1 Complete
Ongoing Phase 2
AR101Pseudomonas aeruginosaAridis PharmaceuticalsPhase 1 Complete
Ongoing Phase 2
514G3Staphylococcus aureusXBiotechPhase 2
ARN-100Staphylococcus aureusArsansisPhase 2 Halted
PolyCAbClostridium difficileMicroPharmPhase 1
RG7861Staphylococcus aureusRochePhase 1
TRL1068Biofilm—multiple speciesTrellis BiosciencePreclinical
Entering Phase 1
AR401-mAbAcinetobacter baumanniiAridis PharmaceuticalsPreclinical
VXD-003Acinetobacter baumanniiVaxDynPreclinical
Cd-ISTAbClostridium difficileIntegrated BioTherapeuticsPreclinical
ASN-4Escherichia coli (ST131)Arsansis—Outlicensed to BB100Preclinical
ASN-5K. pneumoniaeArsansis—Outlicensed to BB200Preclinical

Share and Cite

MDPI and ACS Style

Zurawski, D.V.; McLendon, M.K. Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens. Antibiotics 2020, 9, 155. https://doi.org/10.3390/antibiotics9040155

AMA Style

Zurawski DV, McLendon MK. Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens. Antibiotics. 2020; 9(4):155. https://doi.org/10.3390/antibiotics9040155

Chicago/Turabian Style

Zurawski, Daniel V., and Molly K. McLendon. 2020. "Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens" Antibiotics 9, no. 4: 155. https://doi.org/10.3390/antibiotics9040155

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