Next Article in Journal
Risk Stratification Model for Severe COVID-19 Disease: A Retrospective Cohort Study
Previous Article in Journal
Differential Expression of microRNAs in Serum of Patients with Chronic Painful Polyneuropathy and Healthy Age-Matched Controls
Previous Article in Special Issue
The Clinical Utility of Soluble Serum Biomarkers in Autoimmune Pancreatitis: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Monoclonal Antibody-Based Approaches in the Management of Bacterial Sepsis

by
Kusum Kharga
1,
Lokender Kumar
1,2,* and
Sanjay Kumar Singh Patel
3,*
1
School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, Himachal Pradesh, India
2
Cancer Biology Laboratory, Raj Khosla Centre for Cancer Research, Shoolini University, Solan 173229, Himachal Pradesh, India
3
Department of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea
*
Authors to whom correspondence should be addressed.
Submission received: 3 February 2023 / Revised: 21 February 2023 / Accepted: 27 February 2023 / Published: 2 March 2023
(This article belongs to the Special Issue Immunoglobulins in Inflammation 2.0)

Abstract

:
Sepsis is a life-threatening condition characterized by an uncontrolled inflammatory response to an infectious agent and its antigens. Immune cell activation against the antigens causes severe distress that mediates a strong inflammatory response in vital organs. Sepsis is responsible for a high rate of morbidity and mortality in immunosuppressed patients. Monoclonal antibody (mAb)-based therapeutic strategies are now being explored as a viable therapy option for severe sepsis and septic shock. Monoclonal antibodies may provide benefits through two major strategies: (a) monoclonal antibodies targeting the pathogen and its components, and (b) mAbs targeting inflammatory signaling may directly suppress the production of inflammatory mediators. The major focus of mAb therapies has been bacterial endotoxin (lipopolysaccharide), although other surface antigens are also being investigated for mAb therapy. Several promising candidates for mAbs are undergoing clinical trials at present. Despite several failures and the investigation of novel targets, mAb therapy provides a glimmer of hope for the treatment of severe bacterial sepsis and septic shock. In this review, mAb candidates, their efficacy against controlling infection, with special emphasis on potential roadblocks, and prospects are discussed.

1. Introduction

The definition of sepsis has been a concern of constant evolution and fine-tuning [1]. With advancing knowledge on the pathogenic mechanism of sepsis, currently “sepsis is well-defined as a serious, potentially life threatening, organic dysfunction initiated by an inadequate or dysregulated host response to infection” [2,3]. According to a global analysis, approximately 48.9 million people are estimated to be affected by sepsis yearly and 11 million deaths are estimated to occur by sepsis [4]. Despite the advances in understanding the pathology of sepsis and the development of its treatments, sepsis remains the leading cause of mortality worldwide [5]. Sepsis is a multifaceted disorder involving inflammation and anti-inflammation disbalance leading to the unregulated widespread release of inflammatory mediators, cytokines, and pathogen-related molecules leading to systemwide organ dysfunction [6]. Although our knowledge of the etiology, pathophysiology, and immunology has improved drastically over the past few years, the knowledge regarding the successful management of the same remains limited [1]. Early diagnosis followed by immediate treatment entails the success rate of treatment in sepsis [7]. Currently, treatment for sepsis and septic shock deeply relies on fluid resuscitation and other general supportive measures along with broad-spectrum antibiotics administration [8].
With the advent of antibiotics in the 20th century, they remain [9] the mainstay for the treatment of bacterial infection [10]. However, empirical use of broad-spectrum antibiotics has been associated with increased mortality and the development of antibiotic resistance [11]. The selective pressure of the unsupervised use of antibiotics has already driven bacteria to develop resistant genes against antibiotics [12]. The rise of antibiotic resistance is notably one of the biggest threats of the 21st century leading to therapeutic failure in the field of infectious diseases [13]. In fact, the dawn of extensively pandrug-resistant (PDR), multidrug-resistant (MDR), and extensively drug-resistant (XDR) strains of ESKAPE pathogens (Acinetobacter baumannii, Enterobacter spp., Enterococcus faecium, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Staphylococcus aureus) combined with the rising gap between the development of antibiotic resistance and novel antibiotics has compelled researchers to shift focus on devising alternative innovations for combating bacterial infection [14]. Of such strategies, monoclonal antibiotics (mAbs) stand out as a promising avenue. Serum treatment was one of the effective ways used to treat bacterial infections early in the 1890s. However, limited spectrum and safety concerns such as allergy and cross-reaction led to its discontinuation [15]. The development of hybridoma technology in 1975 revolutionized the field of research and medicine, garnering the 1984 Nobel prize recognition in physiology and medicine [16,17]. With the introduction of hybridoma technology, mAbs have had a profound impact on immunotherapy, providing large-scale production of pure antibodies with improved specificity and reduced immunogenicity [16].
The major obstacle hindering the successful translation of drug candidates in sepsis is the complexity of disease progression that leads to a heterogeneous population with variable underlying clinical presentation, comorbidities, and prognosis abilities [18]. Therefore, monoclonal antibodies targeted against a particular sepsis biomarker present a viable therapeutic option [18]. Monoclonal antibodies may be beneficial in the treatment of sepsis by either directly impeding the growth of the pathogen or by immunomodulation. Initially overshadowed by viral infection and cancer, the application of mAbs in the treatment of bacterial infection is fairly a new approach with only a few FDA-approved drugs [15]. However, it has garnered considerable attention and several promising candidates for mAbs are undergoing clinical trials at present. The present manuscript aims to provide a comprehensive journey of establishing mAbs as a potential alternative therapy for sepsis by highlighting milestone achievements that were ratified by the Federal and Drug Administration (FDA) and those that failed clinical trials but have significantly contributed to our knowledge base.

2. Pathophysiology of Sepsis

Sepsis is a multifaceted chaos of wavering balance between inflammation and anti-inflammation leading to the unregulated widespread release of inflammatory mediators, cytokines, and pathogen-related molecules [6]. This dysregulated host response further activates coagulation and complements cascades that often result in death accompanied by multiple organ dysfunction [19].
The initial activation of the host immune system is mediated by the binding of invading pathogens by pattern-recognition receptors (PRR) on the surface of antigen-presenting cells (APCs) [20]. The PRR-like toll-like receptors (TLR), C-type lectin receptors (CLR’s), retenoic-inducible gene I (RIG-I), and nucleotide-binding oligomerization domain (Nod)-like receptors recognize pathogen-associated molecular patterns (PAMPs) [21,22] and/or host-derived damaged associated molecular patterns (DAMPs) [23]. Murine and mouse models of sepsis revealed that TLR 2, 3, 4, 7 and 9 are involved in the pathogenesis of sepsis by mediating the host’s innate immune response [24,25,26]. Upon recognition of PAMP and damps by TLRs, TLRs initiate transcription of genes involved in inflammation and adaptive immunity through activation of transcription factors such as nuclear factor-κB (NF-κB), activator protein (AP)-1, and mitogen-activated protein kinase (MAPK). This activation of “early genes” leads to the production of pro-inflammatory cytokines such as interleukins IL-8, 12,18, interferons (INFs), and tumor necrosis factor alpha (TNF-α). The pro-inflammatory cytokines thus produced, downstream recruit a torrent of other inflammatory cytokines and chemokines. Components of adaptive immunity are suppressed during the process [27] (Figure 1).
This overwhelming cytokine storm may initially be beneficial in the prognosis of sepsis; however, it ultimately results in progressive organ failure and finally death [6]. In fact, patients in the later stages of sepsis display “immunoparalysis”, a state of chronic immunosuppression [28]. Immunoparalysis is an adjudication of enhanced apoptosis, pyroptosis of immune cells merged with the exhaustion of T cells that marks a patient’s vulnerability to secondary nosocomial infections and reactivation of viruses [18,29,30]. The clinical decline of multiple organs and the development of intravascular thrombosis have been associated with the overproduction or inadequate degradation of immune cell-extracellular traps originally forged to trap and devour pathogens [30,31].
Although sepsis is recognized as an interplay of dysregulated host immune response to infection, clinical representation of the same remains highly individualized making clinical diagnostics a challenge [1]. This may be due to factors such as genetics and the knowledge gap in our understanding of sepsis pathophysiology [32].

3. Current Therapeutic Approaches to Treat Sepsis

Current treatment of sepsis consists mainly of antimicrobial and all-purpose supporting treatment [33]. Early diagnosis followed by immediate treatment entails the success rate of treatment in sepsis [7].

3.1. Antimicrobial Therapy

Antibiotic treatment still remains the cornerstone of the treatment of bacterial infection despite increasing antibiotic resistance. It has been demonstrated that administering the proper antibiotics during the initial phase (1–3 h window) considerably lowers mortality [34,35,36]. The administration of empirical broad-spectrum antibiotics is the primary strategy employed to contain the bacterial infection up until the active microorganism has been determined [33]. However, knowledge of local epidemiological data and the antibiotic resistance pattern of circulating causative bacteria is of utmost importance for launching an effective antibiotics regime [37,38,39]. Antibiotic treatment choices are limited as a result of the selective pressure caused by the unregulated use of antibiotics, which has caused bacteria to acquire antibiotic-resistance genes [40].

3.2. Immunotherapy

With growing antibiotic resistance and the rising gap between the development of antibiotic resistance and novel antibiotics, the focus has been shifted to devising alternative novel innovations for combating bacterial infection. One such strategy is revisiting the antibody-based therapeutic approach which was once deemed harmful.

3.2.1. Immunoglobulins (Ig)

Ig are heterodimeric glycoproteins secreted by differentiated plasma cells and form the part of the natural defense system of the host. They are composed of two heavy (H) and two light chains (L) [41]. Functionally they can be divided into antigen-recognizing variable domains and constant (C) domains involved in activating the complement system and binding Fc regions of other immunoglobulins. Based on the composition of the constant domain, immunoglobulins are defined into five classes: IgM, IgA, IgG, IgE, and IgD. IgG is further subclassified into IgG1, IgG2, IgG3, and IgG4l; similarly, IgA into IgA1 and IgA2. All subclasses each represent unique biological functions [42].

3.2.2. Intravenous Immunoglobulins

Driven by the hypothesis that sepsis-induced immunosuppression could be neutralized by stimulating the immune response or substitution of the individual immune system component, the therapeutic application of polyvalent intravenous immunoglobulins (IVIg) was postulated [43]. The rationale is the ability of the immunoglobulins to (i) recognize and neutralize pathogens and associated toxins, (ii) anti-apoptotic effect on immune cells, and (iii) inhibit transcription [43]. Although the application of IVIg has been found to significantly reduce the sequential organ failure assessment scores (SOFA), apoptosis [44], and disseminated intravascular coagulation (DIC) [45] while increasing the blood level of immunoglobulins [46], it was unable to reduce mortality in sepsis patients [47]. The meta-analysis carried out by Cui and his group in 2019 revealed reduced mortality among adult patients with sepsis by intravenous administration of IgM-enriched immunoglobulin (IVIgGM). However, the treatment effect tended to diminish or become less consistent when the study was narrowed down to a specific indicator of sepsis, invalidating the large-scale use of IVIgGM as a therapeutic treatment [48].

3.2.3. Monoclonal Antibodies

Most therapeutic drugs targeted against sepsis are liable to failure, probably due to the wrong conception that a single therapeutic strategy would suffice to counteract in equal measure all the heterogeneous population with variable clinical presentation, comorbidities, and prognosis abilities [49]. Over the years, studies have been directed toward finding more appropriate biomarkers by conducting more homogeneous sub-population studies in order to develop better targeted drug candidates [50]. As such, monoclonal antibodies (mAbs) targeted against a single target may potentially be the game changer in the treatment of sepsis. With cancer [51] and viral [52,53] infections taking the majority of the limelight, the use of mAbs in the treatment of bacterial infection is still very young with only three FDA-approved drugs so far [15]. Monoclonal antibodies may provide benefits through two major strategies: (1) monoclonal antibodies targeting the pathogen and its components, and (2) mAbs targeting inflammatory signaling may directly suppress the production of inflammatory mediators.

Monoclonal Antibodies Targeting Pathogen and Its Components

Monoclonal antibodies can impede bacterial burden via different mechanisms including direct toxin neutralization, inhibiting virulence factors, complement depositions, and enhanced opsonization (Figure 2) [54].
i.
Toxin Neutralization
Among the array of virulence factors produced by bacterial pathogens to induce infection, secreted toxins play a pivotal role in the induction of infection in targeted host cells located at the sites of infection or distantly [41]. Numerous pathogenic bacteria such as Clostridium botulinum, Clostridium difficile, Clostridium tetani, Bacillus anthracis, Corynebacterium diphtheriae, Vibrio Cholera, Escherichia coli, etc., act by secreting toxins [55,56,57,58,59,60]. Currently, toxins have also been associated with the development of persister cells [61]. Therefore, sequestering toxins though mAbs provides a feasible alternative to reduce infection. In fact, all three FDA-approved mAbs for the treatment of bacterial infections fall under this category.
Raxibacumab (Abthrax®), a human mAb raised against the protective antigen (PA) of Bacillus anthracis, was approved by the FDA on 14 December 2012 as the treatment therapy for post-exposure inhalation anthrax [62]. During inhalation of anthrax, B. anthracis spores deposited in the alveolar spaces of the lungs germinate over a period of 2–43 days into actively dividing bacilli, resulting in bacteremia [63]. Spore germination results in gastrointestinal and oropharyngeal ulceration followed by local edema, necrosis, perforation, and sepsis [64]. These actively dividing bacilli produce tripartite anthrax toxin: (i) protective antigen (PA), (ii) a protease lethal factor (LF), and (iii) an adenylate cyclase edema factor (EF). PA binds to the mammalian cellular receptors and facilitates the translocation of LF and EF into the cells. During the initial stages of infection, the toxins interfere with the signaling pathways and normal functioning of the immune system while the later stage is marked by vascular collapse [65]. Raxibacumab binds to the PA component of the tripartite anthrax toxin and inhibits LF and EF cellular internalization, thereby neutralizing the toxin and preventing the progression of the disease [66]. A study by Cui and his team examined the therapeutic efficacy of raxibacumab in a rat model of anthrax sepsis. Sepsis was first induced using the Bacillus anthracis lethal toxin (LeTx), and raxibacumab was then given post-induction at a dosage of 1–10X of PA. During the beginning of sepsis and 3, 6, 9, or 12 h later, the impact of PA-MAb was examined. Raxibacumab was found to be effective when given to rats for up to 6 h, enhancing their survival and highlighting the efficacy of raxibacumab to manage sepsis [67]. Following further rigorous studies on other animal models [68], raxibacumab was approved for treating inhalation anthrax under the ‘animal rule’ where no other therapeutic option is available [63,66].
Based on the same mechanism of neutralizing anthrax toxin by binding and disarming PA another novel mAb, obiltoxaximab (Anthim®), a chimeric mAb was developed by Elusys Therapeutics [69]. Under the US FDA animal rule, in March of 2016, obiltoxaximab in combination with antibiotics was permitted for the treatment of inhalation anthrax where other treatment options are unavailable [69]. The animal-to-human dose was selected and justification was provided by comparing different animal [70] and human studies [71], where obiltoxaximab at 16 mg/kg was found to exhibit a favorable tolerability and safety profile following intramuscular and intravenous exposure. When Yamamoto and his team investigated the impact of a single intravenous or intramuscular dose of 2–16 mg/kg obiltoxaximab at various time intervals relative to B. anthracis spore exposure, they showed the effectiveness of obiltoxaximab in toxin neutralization pre- and post-exposure administration [72]. This study showed that obiltoxaximab, when given as a 5-day treatment, was able to protect 89–100% of rabbits either given alone or in combination with levofloxacin 9 h after the challenge. Comparatively, only 33% of rabbits were protected by levofloxacin monotherapy. Coherently, a single intramuscular dose of obiltoxaximab at 16 mg/kg given to cynomolgus macaques prior to the onset of bacteremia resulted in 100% survival when given 1–3 days prior to exposure and 38–100% when given 18–24 h following exposure, demonstrating the efficacy of obiltoxaximab as a potent therapeutic agent by inhibiting bacterial spread, ameliorating toxemia, and promoting the survival of the animals. However, administration of obiltoxaximab after the commencement of bacteremia resulted in lowered survival rate (25–50%).
Bezlotoxumab (Zinplava®), a human mAb targeting toxin B of Clostridium difficile was approved by the FDA for the treatment of recurrent C. difficile infection (CDI) [73]. A single molecule of bezlotoxumab neutralizes toxin B by binding to two homologous epitopes within the N-terminal half of the combined repetitive oligopeptide (CROP) domain present within toxin B through its two Fab regions, inhibiting the binding of toxin B to target colonocytes [74]. During gut infection, the translocation of bacteria and their virulence factors due to diminished vascular permeability has been implicated in the development of sepsis. In a study conducted to evaluate the protective effect of bezlotoxumab against systemic infection in mice models, it was demonstrated that bezlotoxumab administered at a dose of 10 mg/kg before and after 24 h of infection could prevent systemic infection and atrophy of the thymus. Models of mice treated with bezlotoxumab showed normal levels of CD4+ and CD8+ cells, which otherwise would reflect immune suppression observed during sepsis [75]. During two large randomized, double-blind trials, intravenous infusion of bezlotoxumab at 10 mg/kg dosage exhibited significant benefit in patients with one or more predefined risks such as age, immunocompromise, history of CDI, and severe CDI. However, an inexplicably increased risk of heart failure was observed in subjects with underlying congestive heart failure [76].
ii.
Enhanced Killing via ADCC and/or Opsonic Phagocytosis
Most of the time, mAbs are unable to cause direct bacterial killing and depend on the cooperation of the complement system (complement-dependent cytotoxicity—CDC) and/or phagocytic cells (antibody-dependent cellular cytotoxicity—ADCC) [15,77]. A mAb 514G3 eradicates S. aureus by enhancing opsonization [78]. S. aureus causes life-threatening conditions and is adept at immune evasion [79]. The cell wall protein A (SpA) of S. aureus tightly binds to most immunoglobulin subclasses via their Fc region thereby neutralizing the effector function of antibodies [78]. Anti-SpA mAb 514G3 binds specifically to S. aureus surface protein SpA and provides a platform for loading phagocytic cells. Phagocytic cells bind to the Fc domain of anti-SpA mAb 514G3 through their Fcγ receptors and clear S. aureus [78]. To determine the protective effect of 514G3 in S. aureus bacteremia, Varshney and his group established the mouse sepsis model using MRSA. Before MRSA infection, mice models were treated with 10 mg of 514G3, whereas control mice were treated with 10 mg of the VH3/IgG3-k isotype or just the vehicle. On day 14, it was revealed that 60% of mice treated with 514G3 survived, whereas none of the mice in the control group or receiving vehicle alone survived [78].
Similarly, to tackle the clinically important Pseudomonas aeruginosa, a bivalent, bispecific human immunoglobulin G1κ monoclonal antibody MEDI3902 biS4aPA was developed. MEDI3902 binds to both the Pseudomonal proteins PcrV responsible for host cell cytotoxicity and exopolysaccharide Psl involved in adherence to tissue and colonization [80]. In preclinical studies involving murine and rabbit pneumonia models and in models of thermal and bacteremia, MEDI3902 was demonstrated to reduce bacterial burden, preserve integrity of pulmonary tissue and prevent dissemination of bacteria to the spleen and kidneys [81,82]. MEDI3902 also exhibited a synergistic effect with different antibiotics against different clinical strains of P. aeruginosa [81]. In a Phase 1 study conducted in healthy adults, MEDI3902 showed that no treatment-emergent adverse events (TEAEs) were observed [80]. MEDI3902, when administered at 250, 750, and 1500 mg/kg doses, exhibited linear pharmacokinetics whereas dosages between 1500 and 3000 mg exhibited non-linear pharmacokinetics. Serum cytotoxicity antibody and opsonophagocytic killing activity were in correlation to the serum concentration of MEDI3902, encouraging further studies. More recently, the safety, efficacy, and pharmacokinetics of MEDI3902 were evaluated in P. aeruginosa-colonized, mechanically ventilated ICU patients [83]. During this randomized double-blind study, pharmacokinetic data showed a low MEDI3902 serum concentration with 500 mg. Patients randomized between 1500 mg and placebo reflected confirmed P. aeruginosa pneumonia in 22.4% of patients receiving 1500 mg MEDI3902 and 18.1% of patients receiving placebo. At 21 days post-treatment with 1500 mg, the mean serum concentration of MEDI3902 was 9.46 µg/mL. The study revealed that MEDI3902 was unsuccessful in reducing nosocomial P. aeruginosa pneumonia in mechanically ventilated patients.
In a similar account, to minimize the casualty of very-low-birth-weight (VLBW) infants due to S. aureus-associated sepsis, pagibaximab, a murine/human chimeric mAb was developed against lipoteichoic acid (LTA) of S. aureus [84]. LTA is highly conserved on the cell wall of S. aureus, which allows S. aureus to evade phagocytosis and sustain bacterial survival by inducing cytokine cascade upon stimulation of TLRs [85]. Pagibaximab was found to be promising in preclinical studies. Pagibaximab, in initial in vitro and in vivo studies, was established to be opsonic, promote phagocytosis, and inhibit the activation of cytokine cascade. However, the Phase III clinical trial, most conclusively, demonstrated a non-significant reduction in sepsis by pagibaximab compared to placebo [86]. The failure of pagibaximab in preventing staphylococcal infection was attributed to the unavailability of LTA for the binding of the mAb (LTA may be present beneath the surface of the cell wall in some S. aureus isolates) [87]. The failure of pagibaximab highlighted the importance of selecting the target and the appropriate animal model [86].
iii.
Inhibiting Virulence Factors
Bacterial growth and survival can be directly hindered by mAbs targeted against the virulence factors present on the surface of the bacteria. KB001-A is PEGylated mAb fragment directed against the Type III secretion system (TTSS) of P. aeruginosa [88]. P. aeruginosa is an avid opportunistic pathogen that is generally found to inhabit the airways of lungs affected by cystic fibrosis (CF) [89]. TTSS of P. aeruginosa facilitates the release of exotoxins into the cytoplasm of host cells and extracellular spaces, leading to the cytotoxicity of P. aeruginosa in host epithelial cells, neutrophils, and macrophages [90]. KB001-A is a modified, recombinant, anti- P. aeruginosa PcrV Fab’ antibody that inhibits the function of TTSS by selectively binding to the PcrV [91]. PcrV is a protein located at the tip of TTSS that is crucial for the transport of Pseudomonal exotoxins into the host cells [92]. A study conducted to evaluate the safety efficacy, pharmacodynamics, and pharmacokinetic (PK) properties of KB001 in 27 CF patients with chronic P. aeruginosa infection revealed that KB001-A had a satisfactory safety profile with an average serum half-life of 11.9 days [91]. No significant difference was observed between the KB001-A treated patients when compared to placebo subjects. However, the 28th day revealed a reduction in the sputum myeloperoxidase, IL-8, IL-1, elastase, and neutrophil counts in the group treated with KB001-A at 10 mg/kg in a dose-dependent manner, indicating a viable alternate option to antibiotic treatment.
Outer membrane proteins (OMPs) are a major source of virulence in gram-negative bacteria making them a potential candidate for the development of mAb [93,94,95,96,97]. Located abundantly on the outer membrane, OMPs are proteinaceous components mostly composed of β-barrel [98]. Proper folding and integration of these β-barrel proteins are highly essential for the viability and pathogenesis of gram-negative bacteria. It is catalyzed by the β-barrel assembly machinery (BAM) [99]. Recent studies reveal that mAb, MAB1, raised against BamA, the central component of the BAM complex was able to retard E. coli growth by exerting direct bactericidal activity [100,101]. MAB1 binds to and interferes with the protein folding activity of BamA.
iv.
Targeting the Biofilm
Biofilm is the most powerful armor forged by bacteria to survive hostile environments [102]. During biofilm formation, the planktonic bacteria enter a sessile mode of lifestyle [103]. Matrix-encased biofilm insulates bacteria and aids in escaping the deleterious effect of immune macrophages and antibiotics and is thus the reason for chronic infection [104]. Some bacterial populations within the biofilm develop to tolerate higher levels of antibiotics and are called persister cells [105]. Following the erosion of biofilm, these persister cells resume planktonic lifestyles, translocate to different areas, colonize them, and restore the original bacterial population [106]. Therefore, biofilms providing an unlimited reservoir of bacteria have emerged as an attractive target for mAbs.
Extracellular DNA (eDNA) is one of the key components of biofilm and serves as a scaffold for the other biofilm components [107]. eDNA in turn is held in place by various bacterial secreted proteins, most prominent belonging to the DNABII family consisting of an integration host factor (IHF) and histone-like (HU) proteins that are conserved among both gram-negative and gram-positive bacteria. Both of these proteins bind to eDNA in a nonspecific manner. Antibody labeling revealed that the IHF was located at the anchoring nodes in the matrix [108]. Therefore, a native human monoclonal antibody TRL1068 was developed that could disrupt the biofilm by binding to and sequestering the scaffold proteins DNABII [109]. In vitro studies revealed TRL1068 could disrupt the established biofilm at 1.2 µg/mL over 12 h. In vivo efficacy of TRL1068 was established in animal models infected with antibiotic-resistant of S. aureus and Acinetobacter baumannii, where TRL1068 was demonstrated to significantly reduce mature biofilm in combination with antibiotics daptomycin, vancomycin, and imipenem [109,110], encasing the potential use of TRL1068 as adjunct therapy against difficult-to-treat bacterial infections. Currently, a clinical trial is recruiting patients for the evaluation of safety and efficacy in prosthetic joint infections.
The efficacy of mAb in disrupting biofilm was further investigated by Jurcisek and his group [111]. In this study, two mAbs, one raised against DNABII protein and another against type IV pilus (T4P) of non-typeable Haemophilus influenzae (NTHI), were tested to disrupt biofilms composed of two different genera of bacteria, wherein NTHI was allied with another clinically important respiratory tract pathogen (Burkholderia cenocepacia, S. aureus, P. aeruginosa, S. pneumoniae, or Moraxella catarrhalis). These monoclonals were tested individually as well as in the form of a cocktail. The study revealed that mAb against NTHI type IV pilus (T4P) was effective only against biofilms formed by single species of NTHI and not on biofilms formed by other species singly. Nonetheless, NTHI-directed mAbs were able to disrupt biofilms composed of two different genera of species. On the contrary, mAbs against DNABII protein were adept at disrupting both multi-species biofilms and all single-species biofilms. The highest release of pathogens following the disruption of multi-species biofilms was achieved by a 1:1 cocktail of both mAbs. This study marked the potential use of a mAb cocktail as an alternate therapy to bacterial infections.
Another study highlighting the efficacy of mAb in inhibiting the biofilm of Staphylococcus epidermidis was demonstrated by Lyu et al. [112]. YycFG, a two-component signal transduction system (TCS), is pivotal for biofilm formation in S. epidermidis [113]. During the study, Lyu et al. evaluated the efficacy of mAbs 2F3 and 1H1, in preventing S. epidermidis biofilm formation. mAbs 2F3 and 1H1 are directed against the histidine kinase YycG extracellular domain (YycGex) [112]. It was revealed that these mAbs were able to inhibit S. epidermidis biofilm formation in a dose-dependent manner. 2F3 and 1H1 administered at a concentration of 120 µg/mL were able to reduce 78.3% and 93.1% biofilm, respectively, as compared to the normal mouse IgG control. Treatment with mAbs 2F3 and 1H1 also exhibited reduced initial adherence and synthesis of polysaccharide intracellular adhesin. Further, a marked reduction in the transcription level of genes responsible for encoding proteins involved in S. epidermidis biofilm formation was observed. This study is an indicator of the YycGex domain as a potential candidate for vaccine development to prevent S. epidermidis biofilm infections.

mAbs Targeting Inflammatory Signaling to Suppress the Production of Inflammatory Mediators and Control Sepsis Progression

As mentioned in earlier sections, sepsis is the result of unregulated hyperinflammation and immune suppression in response to bacterial infections. Therefore, controlling the intermediate mediators of the inflammatory pathway poses an appealing target for developing mAb as a therapeutic agent against sepsis (Figure 3).
During sepsis, neutrophils play a crucial role as first responders; however, they are rendered highly dysfunctional [114]. ADAM17, a transmembrane protease belonging to a disintegrin and metalloproteinase family, is involved in the regulation of various signaling pathways [115]. By conditionally knocking out ADAM17, it was established that the excessive ADAM17 activity in leukocytes has a negative effect on the host response [116]. With the predetermined efficacy of ADAM17 knocking out leukocytes in increasing neutrophil recruitment and reducing bacterial spread during polymicrobial sepsis [117], Mishra et al. investigated the efficacy of ADAM17 mAb MEDI3622 against a murine model of polymicrobial sepsis [118]. This work demonstrated significant sepsis survival following delivery of ADAM17 mAb MEDI3622 both prior to and following infection induction, establishing ADAM17 as the prospective therapeutic target for sepsis control. ADAM17 mAb MEDI3622 also improved survival when administered in combination with antibiotics.
Immunoparalysis due to heightened induced cell death is the hallmark of later stages of sepsis and therefore immunostimulation may prove to be beneficial in controlling sepsis [20]. Following the success in the treatment of cancer, recently there has been an increasing interest in immune checkpoint blockers (ICBs) as candidates for the treatment of bacterial sepsis [119]. In cancer, following the encounter with a foreign antigen, the cytotoxic T lymphocytes (CTLs) expand vigorously [120]. After resolving the inflammation and antigen clearance, the programmed cell death protein 1 (PD-1) receptors on the surface of cytotoxic lymphocytes bind to their ligands, PD-1L and PD-L2, generating negative co-stimulatory signals in order to suppress the CTL expansion [120]. Several studies have documented the enhanced expansion of PD-1 during sepsis. Subjects who died from multiple organ failure resulting from sepsis exhibited increased PD-L1 on macrophages and other APCs [121]. Additionally, a gradual increase in the level of PD-L1 was also seen in monocytes based on the increasing severity of sepsis, supporting the idea that unusual activation of the PD-1/PD-L1 pathway is a major cause of immunoparalysis in sepsis patients [122,123]. Nivolumab is a mAb targeting drug that has proven to be successful at immunomodulation during the treatment of cancer [124]. Nivolumab acts by blocking PD-1 inhibitory pathways, allowing T cells to proliferate. Watanabe et al. found that a single dose of 960 mg of nivolumab was well tolerated and sufficient for maintaining blood concentration levels of nivolumab in a multicenter, open-label phase ½ research on patients with sepsis-induced immunosuppression [125]. Nivolumab 480 mg and nivolumab 960 mg both enhanced immune system indices over time. Additionally, nivolumab and meropenem combination therapy demonstrated that early administration of nivolumab at 6 mg/kg can improve bacterial sepsis when lone antibiotics fail [119], emphasizing the significance of using accurate biomarkers to categorize patients and developing precision medicine in sepsis.
Adrenomedullin (ADM), a 52 amino acid-containing peptide, is required for regulating the endothelial barrier function and vascular tone [126]. Vascular leakage and vasodilation are crucial in the progression of septic shock, where leakage of the vascular membrane is attributed to disrupted endothelial integrity [127]. The level of ADM has been found to rise during sepsis and correlate with the severity of sepsis. Adrecizumab (HAM 8101) is a non-neutralizing mAb directed against the ADM N-terminal that inhibits ADM activity partially. Adrecizumab acts by suppressing the level of ADM in blood concentration. Preclinical studies in the sepsis model demonstrated the efficacy of adrecizumab in improving the endothelial barrier function and reducing mortality induced by sepsis-associated organ failure [128,129]. Following the success in the preclinical trial, Laterre et al. evaluated the safety of adrecizumab by conducting a double-blind, randomized biomarker-guided human trial. During the study, subjects were randomly allocated in a 1:1:2 ratio depending on adrecizumab dosage (adrecizumab 2 mg/kg, 4 mg/kg, and placebo) [130,131]. It was revealed that both doses were well tolerated with no overt signs of harm.
Anaphylatoxins C3a, C4a, and C5a are upregulated due to complement activation during the initial stages of sepsis. Of these, C5a is of particular importance as it possesses pro-inflammatory and chemotactic properties, is spasmogenic, and its sustained level in serum leads to excessive inflammation in sepsis. Disproportionate activation of C5a in sepsis exacerbates systemic inflammation, and leads to apoptosis of immune cells and neutrophil exhaustion, making C5a an attractive pharmacological target. The mouse model of sepsis unveiled the therapeutic potential of blocking C5a or C5aR, where it was revealed that the C5ar1-deficient mice were able to significantly survive mild to moderate sepsis. The decreased mortality was directly associated with improved pathogen clearance and preserved liver function. C5ar1-deficient mice were also shown to have an increased level of pro-inflammatory interferon γ and decreased level of anti-inflammatory cytokine IL-10, validating the role of C5a as a mediator of immunosuppression observed during sepsis.
Vilobelimab (IFX-1) is a chimeric mAb of the IgG4 class developed against the human complement component C5a. A multicenter, randomized, placebo-controlled, double-blind, phase-2a trial involving adult patients suffering from severe sepsis or septic shock exhibiting early onset of infection-associated organ dysfunction was conducted by Bauer et al. in eleven multidisciplinary ICUs across Germany. The study disclosed that subjects displaying severe sepsis and septic shock upon receiving vilobelimab treatment selectively neutralized C5a in a dose-dependent manner exclusive of interference with the normal formation of the membrane attack complex (MAC) nor safety issue. Another mAb avdoralimab directed against C5aR1 inhibits the signaling of C5a via its receptor. A recent study by Carvelli et al. in a randomized controlled trial in patients with COVID-associated pneumonia demonstrated no significant improvement in patients following treatment with avdoralimab [132].

Antibody-Antibiotic Conjugate

Another prospective application of mAbs in the treatment of bacterial infection is through the targeted delivery of antibiotics. The AAC is drafted based on the design of antibody-drug conjugates (ADC) that has previously contributed significantly to the treatment of cancer [133]. AAC consists of a mAb directed against a selective target attached to an antibiotic payload through a cleavable and non-cleavable linker [134]. This innovative approach takes advantage of the selectivity and safety of the antibody to administer more potent antibiotics with reduced off-targets. A successful formulation developed based on this mechanistic is DSTA4637S or THIOMAB™, directed against S. aureus. DSTA4637S is an AAC containing an engineered human IgG1 anti-S. aureus mAb MSTA3852A explicitly for O-Antigen bacterial LPS, and a novel rifamycin antibiotic, dmDNA31 (4-dimethylamino piperidino- hydroxybenzoxazino rifamycin), conjugated through a valine-citrulline (vc) linker cleavable by a protease. DSTA4637S-bound S. aureus upon ingestion by phagocytic cells is cleaved through intracellular cathepsin releasing dmDNA31 that kills intracellular S. aureus [135] (Figure 4), providing hope for the treatment of intracellular pathogens. Overall, mAbs can potentially treat bacterial infections with a few FDA-approved drugs (Table 1).

4. Conclusions

Despite advancements made in the field of science and technology, bacterial sepsis and septic shock remain major health concerns claiming millions of lives every year all over the world. Current therapeutic strategies for treating sepsis still consist of early administration of antibacterial drugs and a general support system aimed towards the restoration of the patient’s hemostasis. Defense against bacterial infection has been dominated by the use of small molecules such as antibiotics over the past decades. However, the escalation of antimicrobial resistance has dwindled the therapeutic efficacy of the antibiotic arsenal to a great extent mandating venture for alternate strategies. Fueled by success stories in the treatment of cancer, mAb has garnered significant attraction as a viable alternative therapy to antibiotics. mAb offers several advantages over antibiotics such as target specificity, reduced immunogenicity, longer half-life, and low probability of developing resistance, making it a plausible alternate therapy for the treatment of sepsis. However, the implementation of mAbs in combating bacterial sepsis has been a rough journey with many pitfalls. With the majority of candidates being inept in clinical translation despite successful preclinical animal studies, mAbs as the treatment of sepsis is still far from being realized. The clinical translation of mAbs in sepsis is also limited by the lack of knowledge regarding the underlying complex physiopathology of the disease itself. Regardless of the increasing understanding of the etiology and pathophysiology of sepsis, researchers are still unable to narrow down the targets for the efficient development of mAbs among the heterogeneous population. Moreover, because mAbs are target specific and need accurate pathogen identification to exert their therapeutic effect, using mAbs in the first hour after exposure is questionable. Furthermore, the limitation of successful clinical translation can be attributed to the lack of an animal model that truly represents the sepsis progression in humans. The safety concerns involving mAbs also remain debatable. The development of mAbs is an expensive process requiring huge investments. Despite the aforementioned drawbacks, mAbs hold huge therapeutic potential as was evidenced by their success in battling cancer. With respect to treating bacterial infection, the approval of three mAbs formulations; raxibacumab, obiltoxaximab, and bezlotoxumab, represents a tiny step toward realizing the therapeutic potential of mAbs against sepsis, encouraging further investigation into this arena.

Author Contributions

Conceptualization, L.K. and S.K.S.P.; data curation, K.K., L.K., and S.K.S.P.; writing—original draft preparation, K.K., L.K., and S.K.S.P.; writing—review and editing, L.K. and S.K.S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Prem Kumar Khosla, Atul Khosla, and Ashish Khosla from Shoolini University, Solan for providing financial support and necessary facilities. This work was supported by the KU Research Professor Program of Konkuk University.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jarczak, D.; Kluge, S.; Nierhaus, A. Sepsis—Pathophysiology and Therapeutic Concepts. Front. Med. 2021, 8, 628302. [Google Scholar] [CrossRef] [PubMed]
  2. Salomão, R.; Ferreira, B.L.; Salomão, M.C.; Santos, S.S.; Azevedo, L.C.P.; Brunialti, M.K.C. Sepsis: Evolving Concepts and Challenges. Braz. J. Med. Biol. Res. 2019, 52, e8595. [Google Scholar] [CrossRef] [PubMed]
  3. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.-D.; Craig, M.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
  4. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, Regional, and National Sepsis Incidence and Mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. World Health Organisation. Global Report on the Epidemiology and Burden of Sepsis: Current Evidence, Identifying Gaps and Future Directions; World Health Organisation: Geneva, Switzerland, 2020; ISBN 9789240010789. [Google Scholar]
  6. Chousterman, B.G.; Swirski, F.K.; Weber, G.F. Cytokine Storm and Sepsis Disease Pathogenesis. Semin. Immunopathol. 2017, 39, 517–528. [Google Scholar] [CrossRef]
  7. Nauclér, P.; Huttner, A.; van Werkhoven, C.H.; Singer, M.; Tattevin, P.; Einav, S.; Tängdén, T. Impact of Time to Antibiotic Therapy on Clinical Outcome in Patients with Bacterial Infections in the Emergency Department: Implications for Antimicrobial Stewardship. Clin. Microbiol. Infect. 2021, 27, 175–181. [Google Scholar] [CrossRef] [PubMed]
  8. Shahrami, B.; Sharif, M.; Sefidani Forough, A.; Najmeddin, F.; Arabzadeh, A.A.; Mojtahedzadeh, M. Antibiotic Therapy in Sepsis: No next Time for a Second Chance! J. Clin. Pharm. Ther. 2021, 46, 872–876. [Google Scholar] [CrossRef] [PubMed]
  9. Gungor, H.; Ekici, M.; Onder Karayigit, M.; Turgut, N.H.; Kara, H.; Arslanbas, E. Zingerone Ameliorates Oxidative Stress and Inflammation in Bleomycin-Induced Pulmonary Fibrosis: Modulation of the Expression of TGF-Β1 and INOS. Naunyn. Schmiedebergs. Arch. Pharmacol. 2020, 393, 1659–1670. [Google Scholar] [CrossRef]
  10. Cook, M.A.; Wright, G.D. The Past, Present, and Future of Antibiotics. Sci. Transl. Med. 2022, 14, eabo7793. [Google Scholar] [CrossRef]
  11. Rhee, C.; Kadri, S.S.; Dekker, J.P.; Danner, R.L.; Chen, H.C.; Fram, D.; Zhang, F.; Wang, R.; Klompas, M.; CDC Prevention Epicenters Program. Prevalence of Antibiotic-Resistant Pathogens in Culture-Proven Sepsis and Outcomes Associated With Inadequate and Broad-Spectrum Empiric Antibiotic Use. JAMA Netw. Open 2020, 3, e202899. [Google Scholar] [CrossRef]
  12. Yen, C.F.; Cutrell, J.B. Antimicrobial Ethicists: Making Ethics Explicit in Antimicrobial Stewardship. Antimicrob. Steward. Healthc. Epidemiol. 2021, 1, e17. [Google Scholar] [CrossRef]
  13. Members, I.U.S.S. Acinetobacter Baumannii Infections Among Patients at Military Medical Facilities Treating. MMWR Morb. Mortal. Wkly. Rep. 2011, 53, 1063–1066. [Google Scholar]
  14. Prasad, N.K.; Seiple, I.B.; Cirz, R.T.; Rosenberg, O.S. Leaks in the Pipeline: A Failure Analysis of Gram-Negative Antibiotic Development from 2010 to 2020. Antimicrob. Agents Chemother. 2022, 66, e00054-22. [Google Scholar] [CrossRef] [PubMed]
  15. Cavaco, M.; Castanho, M.A.R.B.; Neves, V. The Use of Antibody-Antibiotic Conjugates to Fight Bacterial Infections. Front. Microbiol. 2022, 13, 666. [Google Scholar] [CrossRef]
  16. Mitra, S.; Tomar, P.C. Hybridoma Technology; Advancements, Clinical Significance, and Future Aspects. J. Genet. Eng. Biotechnol. 2021, 19, 159. [Google Scholar] [CrossRef] [PubMed]
  17. Moraes, J.Z.; Hamaguchi, B.; Braggion, C.; Speciale, E.R.; Cesar, F.B.V.; da Silva Soares, G.D.F.; Osaki, J.H.; Pereira, T.M.; Aguiar, R.B. Hybridoma Technology: Is It Still Useful? Curr. Res. Immunol. 2021, 2, 32–40. [Google Scholar] [CrossRef]
  18. Nakamori, Y.; Park, E.J.; Shimaoka, M. Immune Deregulation in Sepsis and Septic Shock: Reversing Immune Paralysis by Targeting PD-1/PD-L1 Pathway. Front. Immunol. 2021, 11, 624279. [Google Scholar] [CrossRef]
  19. Iba, T.; Levy, J.H. Sepsis-Induced Coagulopathy and Disseminated Intravascular Coagulation. Anesthesiology 2020, 132, 1238–1245. [Google Scholar] [CrossRef]
  20. van der Poll, T.; Shankar-Hari, M.; Wiersinga, W.J. The Immunology of Sepsis. Immunity 2021, 54, 2450–2464. [Google Scholar] [CrossRef]
  21. Mogensen, T.H. Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [Green Version]
  22. Raymond, S.L.; Holden, D.C.; Mira, J.C.; Stortz, J.A.; Loftus, T.J.; Mohr, A.M.; Moldawer, L.L.; Moore, F.A.; Larson, S.D.; Efron, P.A. Microbial Recognition and Danger Signals in Sepsis and Trauma. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2564–2573. [Google Scholar] [CrossRef] [PubMed]
  23. Gong, T.; Liu, L.; Jiang, W.; Zhou, R. DAMP-Sensing Receptors in Sterile Inflammation and Inflammatory Diseases. Nat. Rev. Immunol. 2020, 20, 95–112. [Google Scholar] [CrossRef] [PubMed]
  24. Jian, W.; Gu, L.; Williams, B.; Feng, Y.; Chao, W.; Zou, L. Toll-like Receptor 7 Contributes to Inflammation, Organ Injury, and Mortality in Murine Sepsis. Anesthesiology 2019, 131, 105–118. [Google Scholar] [CrossRef] [PubMed]
  25. Williams, B.; Neder, J.; Cui, P.; Suen, A.; Tanaka, K.; Zou, L.; Chao, W. Toll-like Receptors 2 and 7 Mediate Coagulation Activation and Coagulopathy in Murine Sepsis. J. Thromb. Haemost. 2019, 17, 1683–1693. [Google Scholar] [CrossRef] [PubMed]
  26. Krivan, S.; Kapelouzou, A.; Vagios, S.; Tsilimigras, D.I.; Katsimpoulas, M.; Moris, D.; Aravanis, C.V.; Demesticha, T.D.; Schizas, D.; Mavroidis, M.; et al. Increased Expression of Toll-like Receptors 2, 3, 4 and 7 MRNA in the Kidney and Intestine of a Septic Mouse Model. Sci. Rep. 2019, 9, 4010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Font, M.D.; Thyagarajan, B.; Khanna, A.K. Sepsis and Septic Shock—Basics of Diagnosis, Pathophysiology and Clinical Decision Making. Med. Clin. N. Am. 2020, 104, 573–585. [Google Scholar] [CrossRef]
  28. Hotchkiss, R.S.; Moldawer, L.L.; Opal, S.M.; Reinhart, K.; Turnbull, I.R.; Vincent, J.L. Sepsis and Septic Shock. Nat. Rev. Dis. Prim. 2016, 2, 16045. [Google Scholar] [CrossRef] [Green Version]
  29. Poulin, L.F.; Lasseaux, C.; Chamaillard, M. Understanding the Cellular Origin of the Mononuclear Phagocyte System Sheds Light on the Myeloid Postulate of Immune Paralysis in Sepsis. Front. Immunol. 2018, 9, 823. [Google Scholar] [CrossRef]
  30. Cao, C.; Yu, M.; Chai, Y. Pathological Alteration and Therapeutic Implications of Sepsis-Induced Immune Cell Apoptosis. Cell Death Dis. 2019, 10, 782. [Google Scholar] [CrossRef] [Green Version]
  31. Wen, X.; Xie, B.; Yuan, S.; Zhang, J. The “Self-Sacrifice” of ImmuneCells in Sepsis. Front. Immunol. 2022, 13, 833479. [Google Scholar] [CrossRef]
  32. Rubio, I.; Osuchowski, M.F.; Shankar-Hari, M.; Skirecki, T.; Winkler, M.S.; Lachmann, G.; La Rosée, P.; Monneret, G.; Venet, F.; Bauer, M.; et al. Current Gaps in Sepsis Immunology: New Opportunities for Translational Research. Lancet Infect. Dis. 2019, 19, e422–e436. [Google Scholar] [CrossRef] [PubMed]
  33. Polat, G.; Ugan, R.A.; Cadirci, E.; Halici, Z. Sepsis and Septic Shock: Current Treatment Strategies and New Approaches Sepsis ve Septik Şok: Mevcut Tedavi Stratejileri ve Yeni Yaklaşımlar. Eurasian J. Med. 2017, 49, 53–58. [Google Scholar] [CrossRef] [PubMed]
  34. Niederman, M.S.; Baron, R.M.; Bouadma, L.; Calandra, T.; Daneman, N.; DeWaele, J.; Kollef, M.H.; Lipman, J.; Nair, G.B. Initial Antimicrobial Management of Sepsis. Crit. Care 2021, 25, 307. [Google Scholar] [CrossRef] [PubMed]
  35. Schuttevaer, R.; Alsma, J.; Brink, A.; van Dijk, W.; de Steenwinkel, J.E.M.; Lingsma, H.F.; Melles, D.C.; Schuit, S.C.E. Appropriate Empirical Antibiotic Therapy and Mortality: Conflicting Data Explained by Residual Confounding. PLoS ONE 2019, 14, e0225478. [Google Scholar] [CrossRef] [PubMed]
  36. Richter, D.C.; Briegel, J.; Pletz, M.W.; Zimmermann, S. Bacterial Sepsis Diagnostics and Calculated Antibiotic. Anaesthesist 2019, 68 (Suppl. S1), 40–62. [Google Scholar] [CrossRef] [Green Version]
  37. Esposito, S.; De Simone, G.; Boccia, G.; De Caro, F.; Pagliano, P. Sepsis and septic shock: New definitions, new diagnostic and therapeutic approaches. J. Glob. Antimicrob. Resist. 2017, 10, 204–212. [Google Scholar] [CrossRef]
  38. Tessema, B.; Lippmann, N.; Knüpfer, M.; Sack, U. Antibiotic Resistance Patterns of Bacterial Isolates from Neonatal Sepsis Patients at University Hospital of Leipzig, Germany. Antibiotics 2021, 10, 323. [Google Scholar] [CrossRef]
  39. Pokhrel, B.; Koirala, T.; Shah, G.; Joshi, S.; Baral, P. Bacteriological Profile and Antibiotic Susceptibility of Neonatal Sepsis in Neonatal Intensive Care Unit of a Tertiary Hospital in Nepal. BMC Pediatr. 2018, 18, 208. [Google Scholar] [CrossRef]
  40. CDC. Antibiotic Resistance Threats in the United States, 2019; U.S. Department of Health and Human Services, CDC: Atlanta, GA, USA, 2019.
  41. Sun, Y.; Huang, T.; Hammarström, L.; Zhao, Y. The Immunoglobulins: New Insights, Implications, and Applications. Annu. Rev. Anim. Biosci. 2020, 8, 145–169. [Google Scholar] [CrossRef] [Green Version]
  42. Napodano, C.; Marino, M.P.; Stefanile, A.; Pocino, K.; Scatena, R.; Gulli, F.; Rapaccini, G.L.; Delli Noci, S.; Capozio, G.; Rigante, D.; et al. Immunological Role of IgG Subclasses. Immunol. Investig. 2020, 50, 427–444. [Google Scholar] [CrossRef]
  43. Jarczak, D.; Kluge, S.; Nierhaus, A. Use of Intravenous Immunoglobulins in Sepsis Therapy—A Clinical View. Int. J. Mol. Sci. 2020, 21, 5543. [Google Scholar] [CrossRef] [PubMed]
  44. Hagiwara, J.; Yamada, M.; Motoda, N.; Yokota, H. Intravenous Immunoglobulin Attenuates Cecum Ligation and Puncture-Induced Acute Lung Injury by Inhibiting Apoptosis of Alveolar Epithelial Cells. J. Nippon Med. Sch. 2020, 87, 129–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Takahashi, G.; Shibata, S. The Study of Usefulness of Low-Dose IgG for Patients with Septic Disseminated Intravascular Coagulation. Biomark Med. 2020, 14, 1189–1196. [Google Scholar] [CrossRef] [PubMed]
  46. Berlot, G.; Scamperle, A.; Istrati, T.; Dattola, R.; Longo, I.; Chillemi, A.; Baronio, S.; Quarantotto, G.; Zanchi, S.; Roman-Pognuz, E.; et al. Kinetics of Immunoglobulins in Septic Shock Patients Treated With an IgM- and IgA-Enriched Intravenous Preparation: An Observational Study. Front. Med. 2021, 8, 1–6. [Google Scholar] [CrossRef] [PubMed]
  47. Welte, T.; Dellinger, R.P.; Ebelt, H.; Ferrer, M.; Opal, S.M.; Singer, M.; Vincent, J.L.; Werdan, K.; Loeches, I.M.; Almirall, J.; et al. Efficacy and Safety of Trimodulin, a Novel Polyclonal Antibody Preparation, in Patients with Severe Community-Acquired Pneumonia: A Randomized, Placebo-Controlled, Double-Blind, Multicenter, Phase II Trial ( CIGMA Study ). Intensive Care Med. 2018, 44, 438–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Cui, J.; Wei, X.; Lv, H.; Li, Y.; Li, P.; Chen, Z.; Liu, G. The Clinical Efficacy of Intravenous IgM-Enriched Immunoglobulin ( Pentaglobin ) in Sepsis or Septic Shock: A Meta-Analysis with Trial Sequential Analysis. Ann. Intensive Care 2019, 9, 27. [Google Scholar] [CrossRef] [Green Version]
  49. Vignon, P.; Laterre, P.F.; Daix, T.; François, B. New Agents in Development for Sepsis: Any Reason for Hope? Drugs 2020, 80, 1751–1761. [Google Scholar] [CrossRef]
  50. Pierrakos, C.; Velissaris, D.; Bisdorff, M.; Marshall, J.C.; Vincent, J. Biomarkers of Sepsis: Time for a Reappraisal. Crit. Care 2020, 24, 287. [Google Scholar] [CrossRef]
  51. Cheng, J.; Chen, G.; Lv, H.; Xu, L.; Liu, H.; Chen, T.; Qu, L.; Wang, J.; Cheng, L.; Hu, S.; et al. CD4-Targeted T Cells Rapidly Induce Remissions in Mice with T Cell Lymphoma. Biomed Res. Int. 2021, 2021, 6614784. [Google Scholar] [CrossRef]
  52. Caskey, M.; Klein, F.; Nussenzweig, M.C. Broadly Neutralizing Anti-HIV-1 Monoclonal Antibodies in the Clinic. Nat. Med. 2019, 25, 547–553. [Google Scholar] [CrossRef]
  53. Mokhtary, P.; Pourhashem, Z.; Mehrizi, A.A.; Sala, C. Recent Progress in the Discovery and Development of Monoclonal Antibodies against Viral Infections. Biomedicines 2022, 10, 1861. [Google Scholar] [CrossRef] [PubMed]
  54. Zurawski, D.V.; McLendon, M.K. Monoclonal Antibodies as an Antibacterial Approach against Bacterial Pathogens. Antibiotics 2020, 9, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Dong, M.; Masuyer, G.; Stenmark, P. Botulinum and Tetanus Neurotoxins. Annu. Rev. Biochem. 2019, 88, 811–837. [Google Scholar] [CrossRef] [PubMed]
  56. Jarmo, O.; Veli-Jukka, A.; Eero, M. Treatment of Clostridioides (Clostridium) Difficile Infection. Ann. Med. 2020, 52, 12–20. [Google Scholar] [CrossRef]
  57. Yang, N.J.; Isensee, J.; Neel, D.V.; Quadros, A.U.; Zhang, H.-X.B.X.B.; Lauzadis, J.; Liu, S.M.; Shiers, S.; Belu, A.; Palan, S.; et al. Anthrax Toxins Regulate Pain Signaling and Can Deliver Molecular Cargoes into ANTXR2+ DRG Sensory Neurons. Nat. Neurosci. 2022, 25, 168–179. [Google Scholar] [CrossRef]
  58. Crisan, C.V.; Hammer, B.K. The Vibrio Cholerae Type VI Secretion System: Toxins, Regulators and Consequences. Environ. Microbiol. 2020, 22, 4112–4122. [Google Scholar] [CrossRef] [Green Version]
  59. García, A.; Fox, J.G. A One Health Perspective for Defining and Deciphering Escherichia Coli Pathogenic Potential in Multiple Hosts. Comp. Med. 2021, 71, 3–45. [Google Scholar] [CrossRef]
  60. Will, R.C.; Ramamurthy, T.; Sharma, N.C.; Veeraraghavan, B.; Sangal, L.; Haldar, P.; Pragasam, A.K.; Vasudevan, K.; Kumar, D.; Das, B.; et al. Spatiotemporal Persistence of Multiple, Diverse Clades and Toxins of Corynebacterium Diphtheriae. Nat. Commun. 2021, 12, 1500. [Google Scholar] [CrossRef]
  61. Paul, P.; Sahu, B.R.; Suar, M. Plausible Role of Bacterial Toxin–Antitoxin System in Persister Cell Formation and Elimination. Mol. Oral Microbiol. 2019, 34, 97–107. [Google Scholar] [CrossRef]
  62. Sakari, M.; Laisi, A.; Pulliainen, A.T. Exotoxin-Targeted Drug Modalities as Antibiotic Alternatives. ACS Infect. Dis. 2022, 8, 433–456. [Google Scholar] [CrossRef]
  63. Mazumdar, S. Raxibacumab. MAbs 2009, 1, 531–538. [Google Scholar] [CrossRef]
  64. Kummerfeldt, C.E. Raxibacumab: Potential Role in the Treatment of Inhalational Anthrax. Infect. Drug Resist. 2014, 7, 101–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Moayeri, M.; Leppla, S.H.; Vrentas, C.; Pomerantsev, A. Anthrax Pathogenesis. Ann. Rev. Microbiol. 2015, 69, 185–208. [Google Scholar] [CrossRef] [PubMed]
  66. Tsai, C.W.; Morris, S. Approval of Raxibacumab for the Treatment of Inhalation Anthrax under the US Food and Drug Administration “Animal Rule”. Front. Microbiol. 2015, 6, 1320. [Google Scholar] [CrossRef] [Green Version]
  67. Cui, X.; Li, Y.; Moayeri, M.; Choi, G.H.; Subramanian, G.M.; Li, X.; Haley, M.; Fitz, Y.; Feng, J.; Banks, S.M.; et al. Late Treatment with a Protective Antigen-Directed Monoclonal Antibody Improves Hemodynamic Function and Survival in a Lethal Toxin-Infused Rat Model of Anthrax Sepsis. J. Infect. Dis. 2005, 191, 422–434. [Google Scholar] [CrossRef] [PubMed]
  68. Corey, A.; Migone, T.S.; Bolmer, S.; Fiscella, M.; Ward, C.; Chen, C.; Meister, G. Bacillus Anthracis Protective Antigen Kinetics in Inhalation Spore-Challenged Untreated or Levofloxacin/ Raxibacumab-Treated New Zealand White Rabbits. Toxins 2013, 5, 120–138. [Google Scholar] [CrossRef] [PubMed]
  69. Greig, S.L. Obiltoxaximab: First Global Approval. Drugs 2016, 76, 823–830. [Google Scholar] [CrossRef]
  70. Henning, L.N.; Comer, J.E.; Stark, G.V.; Ray, B.D.; Tordoff, K.P.; Knostman, K.A.B.; Meister, G.T. Development of an Inhalational Bacillus Anthracis Exposure Therapeutic Model in Cynomolgus Macaques. Clin. Vaccine Immunol. 2012, 19, 1765–1775. [Google Scholar] [CrossRef] [Green Version]
  71. 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.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Yamamoto, B.J.; Shadiack, A.M.; Carpenter, S.; Sanford, D.; Henning, L.N.; Gonzales, N.; O’Connor, E.; Casey, L.S.; Serbina, N.V. Obiltoxaximab Prevents Disseminated Bacillus Anthracis Infection and Improves Survival during Pre- and Postexposure Prophylaxis in Animal Models of Inhalational Anthrax. Antimicrob. Agents Chemother. 2016, 60, 5796–5805. [Google Scholar] [CrossRef] [Green Version]
  73. Gerding, D.N.; Kelly, C.P.; Rahav, G.; Lee, C.; Dubberke, E.R.; Kumar, P.N.; Yacyshyn, B.; Kao, D.; Eves, K.; Ellison, M.C.; et al. Bezlotoxumab for Prevention of Recurrent Clostridium Difficile Infection in Patients at Increased Risk for Recurrence. Clin. Infect. Dis. 2018, 67, 649–656. [Google Scholar] [CrossRef]
  74. Orth, P.; Xiao, L.; Hernandez, L.D.; Reichert, P.; Sheth, P.R.; Beaumont, M.; Yang, X.; Murgolo, N.; Ermakov, G.; Dinunzio, E.; et al. Mechanism of Action and Epitopes of Clostridium Difficile Toxin B-Neutralizing Antibody Bezlotoxumab Revealed by X-Ray Crystallography. J. Biol. Chem. 2014, 289, 18008–18021. [Google Scholar] [CrossRef] [Green Version]
  75. Mileto, S.J.; Hutton, M.L.; Walton, S.L.; Das, A.; Ioannidis, L.J.; Ketagoda, D.; Quinn, K.M.; Denton, K.M.; Hansen, D.S.; Lyras, D. Bezlotoxumab Prevents Extraintestinal Organ Damage Induced by Clostridioides Difficile Infection. Gut Microbes 2022, 14, 2117504. [Google Scholar] [CrossRef]
  76. Johnson, S.; Gerding, D.N. Bezlotoxumab. Clin. Infect. Dis. 2019, 68, 699–704. [Google Scholar] [CrossRef] [Green Version]
  77. Lu, L.L.; Suscovich, T.J.; Fortune, S.M.; Alter, G. Beyond Binding: Antibody Effector Functions in Infectious Diseases. Nat. Rev. Immunol. 2018, 18, 46–61. [Google Scholar] [CrossRef]
  78. Varshney, A.K.; Kuzmicheva, G.A.; Lin, J.B.; Sunley, K.M.; Bowling, R.A.; Kwan, T.Y.; Mays, H.R.; Rambhadran, A.; Zhang, Y.; Martin, R.L.; et al. A Natural Human Monoclonal Antibody Targeting Staphylococcus Protein A Protects against Staphylococcus Aureus Bacteremia. PLoS ONE 2018, 13, e0190537. [Google Scholar] [CrossRef] [Green Version]
  79. Cheung, G.Y.C.; Bae, J.S.; Otto, M. Pathogenicity and Virulence of Staphylococcus Aureus. Virulence 2021, 12, 547–569. [Google Scholar] [CrossRef] [PubMed]
  80. Ali, S.O.; Yu, X.Q.; Robbie, G.J.; Wu, Y.; Shoemaker, K.; Yu, L.; Digiandomenico, A.; Keller, A.E.; Anude, C.; Bellamy, T.; et al. Phase 1 Study of MEDI3902, an Investigational Anti—Pseudomonas Aeruginosa PcrV and Psl Bispeci Fi c Human Monoclonal Antibody, in Healthy Adults. Clin. Microbiol. Infect. 2018, 25, 629.e1–629.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. 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, 262ra155. [Google Scholar] [CrossRef] [PubMed]
  82. Le, H.N.; Quetz, J.S.; Tran, V.G.; Le, V.T.M.; Aguiar-Alves, F.; Pinheiro, M.G.; Cheng, L.; Yu, L.; Sellman, B.R.; Stover, C.K.; et al. MEDI3902 Correlates of Protection against Severe Pseudomonas Aeruginosa Pneumonia in a Rabbit Acute Pneumonia Model. Antimicrob. Agents Chemother. 2018, 62, e02565-17. [Google Scholar] [CrossRef] [Green Version]
  83. Chastre, J.; François, B.; Bourgeois, M.; Komnos, A.; Ferrer, R.; Rahav, G.; De Schryver, N.; Lepape, A.; Koksal, I.; Luyt, C.E.; et al. Safety, Efficacy, and Pharmacokinetics of Gremubamab (MEDI3902), an Anti-Pseudomonas Aeruginosa Bispecific Human Monoclonal Antibody, in P. Aeruginosa-Colonised, Mechanically Ventilated Intensive Care Unit Patients: A Randomised Controlled Trial. Crit. Care 2022, 26, 355. [Google Scholar] [CrossRef]
  84. Weisman, L.E.; Fischer, G.W.; Thackray, H.M.; Johnson, K.E.; Schuman, R.F.; Mandy, G.T.; Stratton, B.E.; Adams, K.M.; Kramer, W.G.; Mond, J.J. Safety and Pharmacokinetics of a Chimerized Anti-Lipoteichoic Acid Monoclonal Antibody in Healthy Adults. Int. Immunopharmacol. 2009, 9, 639–644. [Google Scholar] [CrossRef]
  85. Weisman, L.E.; Thackray, H.M.; Garcia-Prats, J.A.; Nesin, M.; Schneider, J.H.; Fretz, J.; Kokai-Kun, J.F.; Mond, J.J.; Kramer, W.G.; Fischer, G.W. Phase 1/2 Double-Blind, Placebo-Controlled, Dose Escalation, Safety, and Pharmacokinetic Study of Pagibaximab (BSYX-A110), an Antistaphylococcal Monoclonal Antibody for the Prevention of Staphylococcal Bloodstream Infections, in Very-Low-Birth-Weight Neon. Antimicrob. Agents Chemother. 2009, 53, 2879–2886. [Google Scholar] [CrossRef] [Green Version]
  86. Patel, M.; Kaufman, D.A. Anti-Lipoteichoic Acid Monoclonal Antibody (Pagibaximab) Studies for the Prevention of Staphylococcal Bloodstream Infections in Preterm Infants. Expert Opin. Biol. Ther. 2015, 15, 595–600. [Google Scholar] [CrossRef]
  87. Reichmann, N.T.; Piçarra Cassona, C.; Monteiro, J.M.; Bottomley, A.L.; Corrigan, R.M.; Foster, S.J.; Pinho, M.G.; Gründling, A. Differential Localization of LTA Synthesis Proteins and Their Interaction with the Cell Division Machinery in Staphylococcus Aureus. Mol. Microbiol. 2014, 92, 273–286. [Google Scholar] [CrossRef] [Green Version]
  88. 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, 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]
  89. Malhotra, S.; Hayes, D.; Wozniak, D.J. Cystic Fibrosis and Pseudomonas Aeruginosa: The Host-Microbe Interface. Clin. Microbiol. Rev. 2019, 32, e00138-18. [Google Scholar] [CrossRef]
  90. Armentrout, E.I.; Kundracik, E.C.; Rietsch, A.; Division, P.; Sinai, C.; Angeles, L. Cell-Type-Specific Hypertranslocation of Effectors by the Pseudomonas Aeruginosa Type III Secretion System. Mol. Microbiol. 2022, 115, 305–319. [Google Scholar] [CrossRef]
  91. Milla, C.E.; Chmiel, J.F.; Accurso, F.J.; Vandevanter, D.R.; Konstan, M.W.; Yarranton, G.; Geller, D.E. Anti-PcrV Antibody in Cystic Fibrosis: A Novel Approach Targeting Pseudomonas Aeruginosa Airway Infection. Pediatr. Pulmonol. 2014, 49, 650–658. [Google Scholar] [CrossRef] [Green Version]
  92. Pelletier, J.P.R.; Mukhtar, F. Passive Monoclonal and Polyclonal Antibody Therapies. In Immunologic Concepts in Transfusion Medicine; Elsevier: Amsterdam, The Netherlands, 2020; pp. 251–348. ISBN 9780323675093. [Google Scholar]
  93. Nie, D.; Hu, Y.; Chen, Z.; Li, M.; Hou, Z.; Luo, X.; Mao, X.; Xue, X. Outer Membrane Protein A (OmpA) as a Potential Therapeutic Target for Acinetobacter Baumannii Infection. J. Biomed. Sci. 2020, 27, 26. [Google Scholar] [CrossRef] [Green Version]
  94. Xu, C.; Soyfoo, D.M.; Wu, Y.; Xu, S. Virulence of Helicobacter Pylori Outer Membrane Proteins: An Updated Review. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
  95. Rodrigues, C.; Rodrigues, S.C.; Duarte, F.V.; Costa, P.M.; Costa, P.M. The Role of Outer Membrane Proteins in UPEC Antimicrobial Resistance: A Systematic Review. Membranes 2022, 12, 981. [Google Scholar] [CrossRef] [PubMed]
  96. Xu, L.; Wang, M.; Yuan, J.; Wang, H.; Li, M.; Zhang, F.; Tian, Y.; Yang, J.; Wang, J.; Li, B. The KbvR Regulator Contributes to Capsule Production, Outer Membrane Protein Biosynthesis, Antiphagocytosis, and Virulence in Klebsiella Pneumoniae. Infect. Immun. 2021, 89, e00016-21. [Google Scholar] [CrossRef] [PubMed]
  97. Uppalapati, S.R.; Sett, A.; Pathania, R. The Outer Membrane Proteins OmpA, CarO, and OprD of Acinetobacter Baumannii Confer a Two-Pronged Defense in Facilitating Its Success as a Potent Human Pathogen. Front. Microbiol. 2020, 11, 589234. [Google Scholar] [CrossRef]
  98. Horne, J.E.; Brockwell, D.J.; Radford, S.E. Role of the Lipid Bilayer in Outer Membrane Protein Folding in Gram-Negative Bacteria. J. Biol. Chem. 2020, 295, 10340–10367. [Google Scholar] [CrossRef]
  99. Lee, J.; Tomasek, D.; Santos, T.M.; May, M.D.; Meuskens, I.; Kahne, D. Formation of a β-Barrel Membrane Protein Is Catalyzed by the Interior Surface of the Assembly Machine Protein BamA. eLife 2019, 8, e49787. [Google Scholar] [CrossRef]
  100. 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] [Green Version]
  101. 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]
  102. Rather, M.A.; Gupta, K.; Mandal, M. Microbial Biofilm: Formation, Architecture, Antibiotic Resistance, and Control Strategies. Braz. J. Microbiol. 2021, 52, 1701–1718. [Google Scholar] [CrossRef]
  103. Roy, S.; Chowdhury, G.; Mukhopadhyay, A.K.; Dutta, S.; Basu, S. Convergence of Biofilm Formation and Antibiotic Resistance in Acinetobacter Baumannii Infection. Front. Med. 2022, 9, 793615. [Google Scholar] [CrossRef]
  104. Kolpen, M.; Kragh, K.N.; Enciso, J.B.; Faurholt-Jepsen, D.; Lindegaard, B.; Egelund, G.B.; Jensen, A.V.; Ravn, P.; Mathiesen, I.H.M.; Gheorge, A.G.; et al. Bacterial Biofilms Predominate in Both Acute and Chronic Human Lung Infections. Thorax 2022, 77, 1015–1022. [Google Scholar] [CrossRef] [PubMed]
  105. Geißler, P.; Wollanke, B. Biofilm Formation in Persistent Infections and Its Role in the Pathogenesis of Equine Recurrent Uveitis (ERU)—A Literature Review. Pferdeheilkunde 2021, 37, 225–233. [Google Scholar] [CrossRef]
  106. Rumbaugh, K.P.; Sauer, K. Biofilm Dispersion. Nat. Rev. Microbiol. 2020, 18, 571–586. [Google Scholar] [CrossRef] [PubMed]
  107. 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] [PubMed]
  108. Goodman, S.D.; Obergfell, K.P.; Jurcisek, J.A.; Novotny, L.A.; Downey, J.S.; Ayala, E.A.; Tjokro, N.; Li, B.; Justice, S.S.; Bakaletz, L.O. Biofilms Can Be Dispersed by Focusing the Immune System on a Common Family of Bacterial Nucleoid-Associated Proteins. Mucosal Immunol. 2011, 4, 625–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Heidecker, M.; Yang, Y.; Simon, R.J.; Tenorio, E.; Ellsworth, S.; Leighton, A.; Ryser, S.; Gremmelmaier, N.K.; Kauvar, M. A High-Affinity Native Human Antibody Disrupts Biofilm from Staphylococcus Aureus Bacteria and Potentiates Antibiotic Efficacy in a Mouse Implant Infection Model. Antimicrob. Agents Chemother. 2016, 60, 2292–2301. [Google Scholar] [CrossRef]
  110. Xiong, Y.Q.; Estellés, A.; Li, L.; Abdelhady, W.; Gonzales, R.; Bayer, A.S.; Tenorio, E.; Leighton, A.; Ryser, S.; Kauvar, L.M. A Human Biofilm-Disrupting Monoclonal Antibody Potentiates Antibiotic Efficacy in Rodent Models of Both Staphylococcus Aureus and Acinetobacter Baumannii Infections. Antimicrob. Agents Chemother. 2017, 61, e00904-17. [Google Scholar] [CrossRef] [Green Version]
  111. Jurcisek, J.A.; Hofer, L.K.; Goodman, S.D.; Bakaletz, L.O. Monoclonal Antibodies That Target Extracellular DNABII Proteins or the Type IV Pilus of Nontypeable Haemophilus Influenzae (NTHI) Worked Additively to Disrupt 2-Genera Biofilms. Biofilm 2022, 4, 100096. [Google Scholar] [CrossRef]
  112. Lyu, Z.; Shang, Y.; Wang, X.; Wu, Y.; Zheng, J.; Liu, H.; Gong, T.; Ye, L.; Qu, D. Monoclonal Antibodies Specific to the Extracellular Domain of Histidine Kinase YycG of Staphylococcus Epidermidis Inhibit Biofilm Formation. Front. Microbiol. 2020, 11, 1839. [Google Scholar] [CrossRef]
  113. Xu, T.; Wu, Y.; Lin, Z.; Bertram, R.; Götz, F.; Zhang, Y.; Qu, D. Identification of Genes Controlled by the Essential YycFG Two-Component System Reveals a Role for Biofilm Modulation in Staphylococcus Epidermidis. Front. Microbiol. 2017, 8, 724. [Google Scholar] [CrossRef] [Green Version]
  114. Liu, L.; Sun, B. Neutrophil Pyroptosis: New Perspectives on Sepsis. Cell. Mol. Life Sci. 2019, 76, 2031–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wang, K.; Xuan, Z.; Liu, X.; Zheng, M.; Yang, C.; Wang, H. Immunomodulatory Role of Metalloproteinase ADAM17 in Tumor Development. Front. Immunol. 2022, 13, 1059376. [Google Scholar] [CrossRef] [PubMed]
  116. Long, C.; Hosseinkhani, M.R.; Wang, Y.; Sriramarao, P.; Walcheck, B. ADAM17 Activation in Circulating Neutrophils Following Bacterial Challenge Impairs Their Recruitment. J. Leukoc. Biol. 2012, 92, 667–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Mishra, H.K.; Johnson, T.J.; Seelig, D.M.; Walcheck, B. Targeting ADAM17 in Leukocytes Increases Neutrophil Recruitment and Reduces Bacterial Spread during Polymicrobial Sepsis. J. Leukoc. Biol. 2016, 100, 999–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Mishra, H.K.; Ma, J.; Mendez, D.; Hullsiek, R.; Pore, N.; Walcheck, B. Blocking Adam17 Function with a Monoclonal Antibody Improves Sepsis Survival in a Murine Model of Polymicrobial Sepsis. Int. J. Mol. Sci. 2020, 21, 6688. [Google Scholar] [CrossRef]
  119. Gillis, A.; Ben Yaacov, A.; Agur, Z. A New Method for Optimizing Sepsis Therapy by Nivolumab and Meropenem Combination: Importance of Early Intervention and CTL Reinvigoration Rate as a Response Marker. Front. Immunol. 2021, 12, 6881. [Google Scholar] [CrossRef]
  120. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 Pathway: Current Researches in Cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar]
  121. Boomer, J.S.; To, K.; Chang, K.C.; Takasu, O.; Osborne, D.F.; Walton, A.H.; Bricker, T.L.; Jarman, S.D.; Kreisel, D.; Krupnick, A.S.; et al. Immunosuppression in Patients Who Die of Sepsis and Multiple Organ Failure. Surv. Anesthesiol. 2012, 56, 272–273. [Google Scholar] [CrossRef] [Green Version]
  122. Fu, Y.; Wang, D.; Wang, S.; Zhang, Q.; Liu, H.; Yang, S.; Xu, Y.; Ying, B. Blockade of Macrophage-Associated Programmed Death 1 Inhibits the Pyroptosis Signalling Pathway in Sepsis. Inflamm. Res. 2021, 70, 993–1004. [Google Scholar] [CrossRef]
  123. Shao, R.; Fang, Y.; Yu, H.; Zhao, L.; Jiang, Z.; Li, C.S. Monocyte Programmed Death Ligand-1 Expression after 3-4 Days of Sepsis Is Associated with Risk Stratification and Mortality in Septic Patients: A Prospective Cohort Study. Crit. Care 2016, 20, 124. [Google Scholar] [CrossRef] [Green Version]
  124. Hakenberg, O.W. Nivolumab for the Treatment of Bladder Cancer. Expert Opin. Biol. Ther. 2017, 17, 1309–1315. [Google Scholar] [CrossRef] [PubMed]
  125. Watanabe, E.; Nishida, O.; Kakihana, Y.; Odani, M.; Okamura, T.; Harada, T.; Oda, S. Pharmacokinetics, Pharmacodynamics, and Safety of Nivolumab in Patients with Sepsis-Induced Immunosuppression: A Multicenter, Open-Label Phase 1/2 Study. Shock 2020, 53, 686–694. [Google Scholar] [CrossRef] [PubMed]
  126. Geven, C.; Peters, E.; Schroedter, M.; Struck, J.; Bergmann, A.; McCook, O.; Radermacher, P.; Kox, M.; Pickkers, P. Effects of the Humanized Anti-Adrenomedullin Antibody Adrecizumab (HAM8101) on Vascular Barrier Function and Survival in Rodent Models of Systemic Inflammation and Sepsis. Shock 2018, 50, 648–654. [Google Scholar] [CrossRef] [PubMed]
  127. Pickkers, P. Adrenomedullin and Adrenomedullin- Targeted Therapy As Treatment Strategies Relevant for Sepsis. Front. Immunol. 2018, 9, 292. [Google Scholar] [CrossRef]
  128. Blet, A.; Deniau, B.; Geven, C.; Sadoune, M.; Caillard, A.; Kounde, P.R.; Polidano, E.; Pickkers, P.; Samuel, J.L.; Mebazaa, A. Adrecizumab, a Non-Neutralizing Anti-Adrenomedullin Antibody, Improves Haemodynamics and Attenuates Myocardial Oxidative Stress in Septic Rats. Intensive Care Med. Exp. 2019, 7, 25. [Google Scholar] [CrossRef]
  129. Caironi, P.; Latini, R.; Struck, J.; Hartmann, O.; Bergmann, A.; Maggio, G.; Cavana, M.; Tognoni, G.; Pesenti, A.; Gattinoni, L.; et al. Circulating Biologically Active Adrenomedullin (Bio-ADM) Predicts Hemodynamic Support Requirement and Mortality during Sepsis. Chest 2017, 152, 312–320. [Google Scholar] [CrossRef]
  130. Thiele, C.; Simon, T.P.; Szymanski, J.; Daniel, C.; Golias, C.; Hartmann, O.; Struck, J.; Martin, L.; Marx, G.; Schuerholz, T. Effects of the Non-Neutralizing Humanized Monoclonal Anti-Adrenomedullin Antibody Adrecizumab on Hemodynamic and Renal Injury in a Porcine Two-Hit Model. Shock 2020, 54, 810–818. [Google Scholar] [CrossRef]
  131. van Lier, D.; Picod, A.; Marx, G.; Laterre, P.F.; Hartmann, O.; Knothe, C.; Azibani, F.; Struck, J.; Santos, K.; Zimmerman, J.; et al. Effects of Enrichment Strategies on Outcome of Adrecizumab Treatment in Septic Shock: Post-Hoc Analyses of the Phase II Adrenomedullin and Outcome in Septic Shock 2 Trial. Front. Med. 2022, 9, 1058235. [Google Scholar] [CrossRef]
  132. Carvelli, J.; Meziani, F.; Dellamonica, J.; Cordier, P.; Allardet-Servent, J.; Fraisse, M.; Velly, L.; Barbar, S.D.; Lehingue, S.; Guervilly, C.; et al. Avdoralimab (Anti-C5aR1 MAb) Versus Placebo in Patients With Severe COVID-19: Results From a Randomized Controlled Trial (FOR COVID Elimination [FORCE]). Crit. Care Med. 2022, 50, 1788–1798. [Google Scholar] [CrossRef]
  133. Mariathasan, S.; Tan, M.W. Antibody–Antibiotic Conjugates: A Novel Therapeutic Platform against Bacterial Infections. Trends Mol. Med. 2017, 23, 135–149. [Google Scholar] [CrossRef]
  134. Deng, R.; Zhou, C.; Li, D.; Cai, H.; Sukumaran, S.; Carrasco-Triguero, M.; Saad, O.; Nazzal, D.; Lowe, C.; Ramanujan, S.; et al. Preclinical and Translational Pharmacokinetics of a Novel THIOMABTM Antibody-Antibiotic Conjugate against Staphylococcus Aureus. MAbs 2019, 11, 1162–1174. [Google Scholar] [CrossRef] [PubMed]
  135. Peck, M.; Rothenberg, M.E.; Deng, R.; Lewin-Koh, N.; She, G.; Kamath, A.V.; Carrasco-Triguero, M.; Saad, O.; Castro, A.; Teufel, L.; et al. A Phase 1, Randomized, Single-Ascending-Dose Study To Investigate the Safety, Tolerability, and Pharmacokinetics of DSTA4637S, an Anti-Staphylococcus aureus Thiomab Antibody-Antibiotic Conjugate, in Healthy Volunteers. Antimicrob. Agents Chemother. 2019, 63, e02588-18. [Google Scholar] [CrossRef] [Green Version]
  136. Giacobbe, D.R.; Dettori, S.; Di Bella, S.; Vena, A.; Granata, G.; Luzzati, R.; Petrosillo, N.; Bassetti, M. Bezlotoxumab for Preventing Recurrent Clostridioides Difficile Infection: A Narrative Review from Pathophysiology to Clinical Studies. Infect. Dis. Ther. 2020, 9, 481–494. [Google Scholar] [CrossRef]
  137. Francois, B.; Garcia, M.S.; Eggimann, P.; Dequin, P.-F.; Laterre, P.-F.; Huberlant, V.; Soria, L.V.; Boulain, T.; Bretonnière, C.; Pugin, J.; et al. Session: Efficacy, Pharmacokinetics (PK), and Safety Profile of Suvratoxumab (MEDI4893), a Staphylococcus aureus Alpha Toxin (AT)-Neutralizing Human Monoclonal Antibody in Mechanically Ventilated Patients in Intensive Care Units; Results of the Phase 2 SAATELLITE Study Conducted by the Public-Private COMBACTE Consortium. Open Forum. Infect. Dis. 2019, 6 (Suppl. S2), S66. [Google Scholar]
  138. Tkaczyk, C.; Semenova, E.; Shi, Y.Y.; Rosenthal, K.; Oganesyan, V.; Warrener, P.; Stover, C.K.; Sellman, B.R. Alanine Scanning Mutagenesis of the MEDI4893 (Suvratoxumab) Epitope Reduces Alpha Toxin Lytic Activity In Vitro and Staphylococcus Aureus Fitness in Infection Models. Antimicrob. Agents Chemother. 2018, 62, e01033-18. [Google Scholar] [CrossRef] [Green Version]
  139. Tkaczyk, C.; Hamilton, M.M.; Sadowska, A.; Shi, Y.; Chang, C.S.; Chowdhury, P.; Buonapane, R.; Xiao, X.; Warrener, P.; Mediavilla, J.; et al. Targeting Alpha Toxin and ClfA with a Multimechanistic Monoclonal- Antibody-Based Approach for Prophylaxis of Serious Staphylococcus Aureus Disease. mBio 2016, 7, e00528-16. [Google Scholar] [CrossRef] [Green Version]
  140. Que, Y.A.; Lazar, H.; Wolff, M.; François, B.; Laterre, P.F.; Mercier, E.; Garbino, J.; Pagani, J.L.; Revelly, J.P.; Mus, E.; et al. Assessment of Panobacumab as Adjunctive Immunotherapy for the Treatment of Nosocomial Pseudomonas Aeruginosa Pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1861–1867. [Google Scholar] [CrossRef] [Green Version]
  141. Koch, H.; Emrich, T.; Jampen, S.; Wyss, M.; Gafner, V.; Lazar, H.; Rudolf, M.P. Development of a 4-Valent Genotyping Assay for Direct Identification of the Most Frequent Pseudomonas Aeruginosa Serotypes from Respiratory Specimens of Pneumonia Patients. J. Med. Microbiol. 2014, 63, 508–517. [Google Scholar] [CrossRef] [PubMed]
  142. Magyarics, Z.; Leslie, F.; Bartko, J.; Rouha, H.; Luperchio, S.; Schörgenhofer, C.; Schwameis, M.; Derhaschnig, U.; Lagler, H.; Stiebellehner, L.; et al. Randomized, Double-Blind, Placebo-Controlled, Single- Ascending-Dose Study of the Penetration of a Monoclonal Antibody Combination (ASN100) Targeting Staphylococcus Aureus Cytotoxins in the Lung Epithelial Lining Fluid of Healthy Volunteers. Antimicrob. Agents Chemother. 2019, 63, e00350-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Rouha, H.; Weber, S.; Janesch, P.; Maierhofer, B.; Gross, K.; Dolezilkova, I.; Mirkina, I.; Visram, Z.C.; Malafa, S.; Stulik, L.; et al. Disarming Staphylococcus Aureus from Destroying Human Cells by Simultaneously Neutralizing Six Cytotoxins with Two Human Monoclonal Antibodies. Virulence 2018, 9, 231–247. [Google Scholar] [CrossRef] [Green Version]
  144. Nagy, C.F.; Mondick, J.; Serbina, N.; Casey, L.S.; Carpenter, S.E.; French, J.; Guttendorf, R. Animal-to-Human Dose Translation of Obiltoxaximab for Treatment of Inhalational Anthrax Under the US FDA Animal Rule. Clin. Transl. Sci. 2017, 10, 12–19. [Google Scholar] [CrossRef]
  145. Nagy, C.F.; Leach, T.S.; King, A.; Guttendorf, R. Safety, Pharmacokinetics, and Immunogenicity of Obiltoxaximab After Intramuscular Administration to Healthy Humans. Clin. Pharmacol. Drug Dev. 2018, 7, 652–660. [Google Scholar] [CrossRef] [Green Version]
  146. François, B.; Mercier, E.; Gonzalez, C.; Asehnoune, K.; Nseir, S. Safety and Tolerability of a Single Administration of AR-301, a Human Monoclonal Antibody, in ICU Patients with Severe Pneumonia Caused by Staphylococcus Aureus: First-in-Human Trial. Intensive Care Med. 2018, 44, 1787–1796. [Google Scholar] [CrossRef]
  147. Perez-Ruiz, E.; Minute, L.; Otano, I.; Alvarez, M.; Ochoa, M.C.; Belsue, V.; de Andrea, C.; Rodriguez-Ruiz, M.E.; Perez-Gracia, J.L.; Marquez-Rodas, I.; et al. Prophylactic TNF Blockade Uncouples Efficacy and Toxicity in Dual CTLA-4 and PD-1 Immunotherapy. Nature 2019, 569, 428–432. [Google Scholar] [CrossRef]
  148. Geven, C.; van Lier, D.; Blet, A.; Peelen, R.; ten Elzen, B.; Mebazaa, A.; Kox, M.; Pickkers, P. Safety, Tolerability and Pharmacokinetics/Pharmacodynamics of the Adrenomedullin Antibody Adrecizumab in a First-in-Human Study and during Experimental Human Endotoxaemia in Healthy Subjects. Br. J. Clin. Pharmacol. 2018, 84, 2129–2141. [Google Scholar] [CrossRef] [Green Version]
  149. Laterre, P.F.; Pickkers, P.; Marx, G.; Wittebole, X.; Meziani, F.; Dugernier, T.; Huberlant, V.; Schuerholz, T.; François, B.; Lascarrou, J.B.; et al. Safety and Tolerability of Non-Neutralizing Adrenomedullin Antibody Adrecizumab (HAM8101) in Septic Shock Patients: The AdrenOSS-2 Phase 2a Biomarker-Guided Trial. Intensive Care Med. 2021, 47, 1284–1294. [Google Scholar] [CrossRef]
  150. Vlaar, A.P.J.; Witzenrath, M.; van Paassen, P.; Heunks, L.M.A.; Mourvillier, B.; de Bruin, S.; Lim, E.H.T.; Brouwer, M.C.; Tuinman, P.R.; Saraiva, J.F.K.; et al. Anti-C5a Antibody (Vilobelimab) Therapy for Critically Ill, Invasively Mechanically Ventilated Patients with COVID-19 (PANAMO): A Multicentre, Double-Blind, Randomised, Placebo-Controlled, Phase 3 Trial Alexander. Lancet Respir. Med. 2022, 10, 1137–1146. [Google Scholar] [CrossRef]
  151. Bauer, M.; Weyland, A.; Marx, G.; Bloos, F.; Weber, S.; Weiler, N.; Kluge, S.; Diers, A.; Simon, T.P.; Lautenschläger, I.; et al. Efficacy and Safety of Vilobelimab (IFX-1), a Novel Monoclonal Anti-C5a Antibody, in Patients With Early Severe Sepsis or Septic Shock—A Randomized, Placebo-Controlled, Double-Blind, Multicenter, Phase IIa Trial (SCIENS Study). Crit. Care Explor. 2021, 3, e0577. [Google Scholar] [CrossRef]
Figure 1. Immunopathogenesis of sepsis. The initial host response is triggered by the binding of bacterial virulence factors to the various pattern recognition receptors such as toll-like receptors (TLR), C-type lectin receptors (CLRs), nucleotide-binding oligomerization domain (Nod)-like receptors and retenoic-inducible gene I (RIG-I). The pro-inflammatory response is marked by the activation of leukocytes which further activates components of the complement and coagulation systems and vascular endothelium via secretion of mediators such as cytokines, reactive oxygen species, and proteases. This led to release of damaged associated molecular patterns (DAMPs) that further exacerbate the pro-inflammatory response. The anti-inflammatory response is represented by impaired function of immune cells, abnormal level of apoptosis, exhaustion of B cells, T cells, and dendritic cells due to negative regulation of TLR signaling and inhibition of pro-inflammatory genes leading to “immunoparalysis”.
Figure 1. Immunopathogenesis of sepsis. The initial host response is triggered by the binding of bacterial virulence factors to the various pattern recognition receptors such as toll-like receptors (TLR), C-type lectin receptors (CLRs), nucleotide-binding oligomerization domain (Nod)-like receptors and retenoic-inducible gene I (RIG-I). The pro-inflammatory response is marked by the activation of leukocytes which further activates components of the complement and coagulation systems and vascular endothelium via secretion of mediators such as cytokines, reactive oxygen species, and proteases. This led to release of damaged associated molecular patterns (DAMPs) that further exacerbate the pro-inflammatory response. The anti-inflammatory response is represented by impaired function of immune cells, abnormal level of apoptosis, exhaustion of B cells, T cells, and dendritic cells due to negative regulation of TLR signaling and inhibition of pro-inflammatory genes leading to “immunoparalysis”.
Biomedicines 11 00765 g001
Figure 2. Examples of mAbs that target bacteria with their general mechanism of action.
Figure 2. Examples of mAbs that target bacteria with their general mechanism of action.
Biomedicines 11 00765 g002
Figure 3. Immunomodulatory targets for the development of mAb to control the progression of sepsis.
Figure 3. Immunomodulatory targets for the development of mAb to control the progression of sepsis.
Biomedicines 11 00765 g003
Figure 4. Mechanism of killing intracellular S. aureus by DSTA4637S. DSTA4637S binds to S. aureus, enhancing phagocytosis. Following internalization by host cells, it fuses with the phagolysosome where lysosomal cathepsins cleave the vc linker releasing the antibiotic dmDNA31. dmDNA31 is responsible for killing the intracellular pathogen.
Figure 4. Mechanism of killing intracellular S. aureus by DSTA4637S. DSTA4637S binds to S. aureus, enhancing phagocytosis. Following internalization by host cells, it fuses with the phagolysosome where lysosomal cathepsins cleave the vc linker releasing the antibiotic dmDNA31. dmDNA31 is responsible for killing the intracellular pathogen.
Biomedicines 11 00765 g004
Table 1. Monoclonal-antibody-based biologics that are FDA approved or in clinical trials for the treatment of sepsis.
Table 1. Monoclonal-antibody-based biologics that are FDA approved or in clinical trials for the treatment of sepsis.
NameTargetTarget BacteriaClass of ImmunoglobulinMechanism of ActionPhaseTrial IDReferences
Monoclonal antibodies targeted against the pathogen and pathogen-associated factors
514G3Staphylococcus aureus Protein A (SpA)HumanIgG3Enhanced opsonization Phase IINCT02357966[78]
RaxibacumabBacillus anthracis toxinHumanIgG1Toxin neutralizationApprovedNCT02016963[63,66,68]
Bezlotoxumab(Zinplava®) (MK-6072, MBL-CDB1, or MDX-1388).C. difficile toxin BHumanIgG1Toxin neutralizationApprovedNCT05304715[73,74,136]
MEDI4893 SuvratoxumabStaphylococcus aureus Alpha toxin (AT)HumanIgG1Toxin neutralizationPhase IIINCT05331885[137,138,139]
TRL1068DNABII proteins such as integration host factor (IHF) and histone-like (HU) proteins of both gram-negative and gram-positive bacteriaHumanIgGBiofilm disruptionPreclinical/Phase INCT04763759[107,109,110]
PanobacumabLPS O-polysaccharide moiety of P. aeruginosa O11HumanIgM/κComplement-dependent enhanced opsonophagocytosis Phase IINCT00851435[140,141]
ASN100
Combination of ASN-1 and ASN-2
Staphylococcus aureus alpha-hemolysin (Hla) and leukocidins LukSF-PV, HlgAB, HlgCB, LukED, and LukGH (LukAB)HumanIgG1/κToxin neutralizationPhase II, terminatedNCT02940626[142,143]
ObiltoxaximabBacillus anthracis (Protective antigen—PA) IgG1Toxin neutralizationApprovedNCT03088111[69,71,144,145]
Tosatoxumab
(Salvecin)
AR-301
Staphylococcus aureus alpha toxinHumanIgG1 ʎToxin neutralizationPhase IIINCT03816956[146]
Pagibaximab-BSYX-A110Staphylococcus epidermidis (Lipoteichoic acid—LTAMouse/Human chimericIgGEnhanced opsonizationPhase IINCT00631800[85,86]
KB001-APseudomonas aeruginosa Type III secretion systemRecombinant IgG lacking Fc regionInhibiting the activity of TTSSPhase IINCT01695343[88,91]
MEDI3902, GremubamabPseudomonas aeruginosa
PcrV protein and Psl exopolysaccharide
Human bispecificIgGComplement-dependent enhanced opsonophagocytosisPhase INCT02255760[83]
Monoclonal antibodies targeting inflammatory signaling that directly suppress the production of inflammatory mediators
MEDI3622ADAM17 transmembrane proteaseHuman IgG1ADAM17 function-blockingPreclinical [115,118]
Nivolumab(opdivo®)Protein Domain 1 (PD-1)Human IgG4Inhibiting PD-1/PD-L1 pathwayPhase INCT02960854[119,125,147]
AdrecizumabAdrenomedullin (ADM)Humanized IgG1/κBlocking adrenomedullin (ADM)Phase INCT03083171[131,148,149]
Vilobelimab (IFX-1)C5aChimeric IgG4Blocking C5aPhase IINCT02246595[150,151]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kharga, K.; Kumar, L.; Patel, S.K.S. Recent Advances in Monoclonal Antibody-Based Approaches in the Management of Bacterial Sepsis. Biomedicines 2023, 11, 765. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11030765

AMA Style

Kharga K, Kumar L, Patel SKS. Recent Advances in Monoclonal Antibody-Based Approaches in the Management of Bacterial Sepsis. Biomedicines. 2023; 11(3):765. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11030765

Chicago/Turabian Style

Kharga, Kusum, Lokender Kumar, and Sanjay Kumar Singh Patel. 2023. "Recent Advances in Monoclonal Antibody-Based Approaches in the Management of Bacterial Sepsis" Biomedicines 11, no. 3: 765. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines11030765

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