51
|
Elfadil D, Elkhatib WF, El-Sayyad GS. Promising advances in nanobiotic-based formulations for drug specific targeting against multidrug-resistant microbes and biofilm-associated infections. Microb Pathog 2022; 170:105721. [PMID: 35970290 DOI: 10.1016/j.micpath.2022.105721] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Antimicrobial agents and alternative strategies to combat bacterial infections have become urgent due to the rapid development of multidrug-resistant bacteria caused by the misuse and overuse of antibiotics, as well as the ineffectiveness of antibiotics against difficult-to-treat infectious diseases. Nanobiotics is one of the strategies being explored to counter the increase in antibiotic-resistant bacteria. Nanobiotics are antibiotic molecules encapsulated in nanoparticles or artificially engineered pure antibiotics that are ≤ 100 nm in size in at least one dimension. Formulation scientists recognize nanobiotic delivery systems as an effective strategy to overcome the limitations associated with conventional antibiotic therapy. This review highlights the general mechanisms by which nanobiotics can be used to target resistant microbes and biofilm-associated infections. We focus on the design elements, properties, characterization, and toxicity assessment of organic nanoparticles, inorganic nanoparticle and molecularly imprinted polymer-based nano-formulations that can be designed to improve the efficacy of nanobiotic formulation.
Collapse
Affiliation(s)
- Dounia Elfadil
- Biology and Chemistry Department, Hassan II University of Casablanca, Morocco
| | - Walid F Elkhatib
- Microbiology and Immunology Department, Ain Shams University, African Union Organization St., Abbassia, Cairo, 11566, Egypt; Department of Microbiology and Immunology, Galala University, New Galala City, Suez, Egypt.
| | - Gharieb S El-Sayyad
- Department of Microbiology and Immunology, Galala University, New Galala City, Suez, Egypt; Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| |
Collapse
|
52
|
Reig S, Le Gouellec A, Bleves S. What Is New in the Anti–Pseudomonas aeruginosa Clinical Development Pipeline Since the 2017 WHO Alert? Front Cell Infect Microbiol 2022; 12:909731. [PMID: 35880080 PMCID: PMC9308001 DOI: 10.3389/fcimb.2022.909731] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
The spread of antibiotic-resistant bacteria poses a substantial threat to morbidity and mortality worldwide. Carbapenem-resistant Pseudomonas aeruginosa (CRPA) are considered “critical-priority” bacteria by the World Health Organization (WHO) since 2017 taking into account criteria such as patient mortality, global burden disease, and worldwide trend of multi-drug resistance (MDR). Indeed P. aeruginosa can be particularly difficult to eliminate from patients due to its combinatory antibiotic resistance, multifactorial virulence, and ability to over-adapt in a dynamic way. Research is active, but the course to a validated efficacy of a new treatment is still long and uncertain. What is new in the anti–P. aeruginosa clinical development pipeline since the 2017 WHO alert? This review focuses on new solutions for P. aeruginosa infections that are in active clinical development, i.e., currently being tested in humans and may be approved for patients in the coming years. Among 18 drugs of interest in December 2021 anti–P. aeruginosa development pipeline described here, only one new combination of β-lactam/β-lactamase inhibitor is in phase III trial. Derivatives of existing antibiotics considered as “traditional agents” are over-represented. Diverse “non-traditional agents” including bacteriophages, iron mimetic/chelator, and anti-virulence factors are significantly represented but unfortunately still in early clinical stages. Despite decade of efforts, there is no vaccine currently in clinical development to prevent P. aeruginosa infections. Studying pipeline anti–P. aeruginosa since 2017 up to now shows how to provide a new treatment for patients can be a difficult task. Given the process duration, the clinical pipeline remains unsatisfactory leading best case to the approval of new antibacterial drugs that treat CRPA in several years. Beyond investment needed to build a robust pipeline, the Community needs to reinvent medicine with new strategies of development to avoid the disaster. Among “non-traditional agents”, anti-virulence strategy may have the potential through novel and non-killing modes of action to reduce the selective pressure responsible of MDR.
Collapse
Affiliation(s)
- Sébastien Reig
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), Institut de Microbiologie, Bioénergies et Biotechnologie (IM2B), Aix-Marseille Université-CNRS, UMR7255, Marseille, France
- *Correspondence: Sébastien Reig, ; Sophie Bleves,
| | - Audrey Le Gouellec
- Laboratoire Techniques de l’Ingénierie Médicale et de la Complexité (UMR5525), Centre National de la Recherche Scientifique, Université Grenoble Alpes, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, Grenoble, France
| | - Sophie Bleves
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), Institut de Microbiologie, Bioénergies et Biotechnologie (IM2B), Aix-Marseille Université-CNRS, UMR7255, Marseille, France
- *Correspondence: Sébastien Reig, ; Sophie Bleves,
| |
Collapse
|
53
|
Pérez de la Lastra JM, Anand U, González-Acosta S, López MR, Dey A, Bontempi E, Morales delaNuez A. Antimicrobial Resistance in the COVID-19 Landscape: Is There an Opportunity for Anti-Infective Antibodies and Antimicrobial Peptides? Front Immunol 2022; 13:921483. [PMID: 35720330 PMCID: PMC9205220 DOI: 10.3389/fimmu.2022.921483] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
Although COVID-19 has captured most of the public health attention, antimicrobial resistance (AMR) has not disappeared. To prevent the escape of resistant microorganisms in animals or environmental reservoirs a "one health approach" is desirable. In this context of COVID-19, AMR has probably been affected by the inappropriate or over-use of antibiotics. The increased use of antimicrobials and biocides for disinfection may have enhanced the prevalence of AMR. Antibiotics have been used empirically in patients with COVID-19 to avoid or prevent bacterial coinfection or superinfections. On the other hand, the measures to prevent the transmission of COVID-19 could have reduced the risk of the emergence of multidrug-resistant microorganisms. Since we do not currently have a sterilizing vaccine against SARS-CoV-2, the virus may still multiply in the organism and new mutations may occur. As a consequence, there is a risk of the appearance of new variants. Nature-derived anti-infective agents, such as antibodies and antimicrobial peptides (AMPs), are very promising in the fight against infectious diseases, because they are less likely to develop resistance, even though further investigation is still required.
Collapse
Affiliation(s)
- José M. Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), San Cristóbal de la Laguna, Spain
| | - Uttpal Anand
- CytoGene Research & Development LLP, Barabanki, Uttar Pradesh, India
| | - Sergio González-Acosta
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), San Cristóbal de la Laguna, Spain
| | - Manuel R. López
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), San Cristóbal de la Laguna, Spain
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Elza Bontempi
- National Interuniversity Consortium of Materials Science and Technology (INSTM) and Chemistry for Technologies Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, Brescia, Italy
| | - Antonio Morales delaNuez
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), San Cristóbal de la Laguna, Spain
| |
Collapse
|
54
|
Wang H, Chen D, Lu H. Anti-bacterial monoclonal antibodies: next generation therapy against superbugs. Appl Microbiol Biotechnol 2022; 106:3957-3972. [PMID: 35648146 DOI: 10.1007/s00253-022-11989-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 12/19/2022]
Abstract
Prior to the nineteenth century, infectious disease was one of the leading causes of death. Human life expectancy has roughly doubled over the past century as a result of the development of antibiotics and vaccines. However, the emergence of antibiotic-resistant superbugs brings new challenges. The side effects of broad-spectrum antibiotics, such as causing antimicrobial resistance and destroying the normal flora, often limit their applications. Furthermore, the development of new antibiotics has lagged far behind the emergence and spread of antibiotic resistance. On the other hand, the genome complexity of bacteria makes it difficult to create effective vaccines. Therefore, novel therapeutic agents in supplement to antibiotics and vaccines are urgently needed to improve the treatment of infections. In recent years, monoclonal antibodies (mAbs) have achieved remarkable clinical success in a variety of fields. In the treatment of infectious diseases, mAbs can play functions through multiple mechanisms, including toxins neutralization, virulence factors inhibition, complement-mediated killing activity, and opsonic phagocytosis. Toxins and bacterial surface components are good targets to generate antibodies against. The U.S. FDA has approved three monoclonal antibody drugs, and there are numerous candidates in the preclinical or clinical trial stages. This article reviews recent advances in the research and development of anti-bacterial monoclonal antibody drugs in order to provide a valuable reference for future studies in this area. KEY POINTS: • Novel drugs against antibiotic-resistant superbugs are urgently required • Monoclonal antibodies can treat bacterial infections through multiple mechanisms • There are many anti-bacterial monoclonal antibodies developed in recent years and some candidates have entered the preclinical or clinical stages of development.
Collapse
Affiliation(s)
- Hui Wang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Huili Lu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
55
|
MDR Pumps as Crossroads of Resistance: Antibiotics and Bacteriophages. Antibiotics (Basel) 2022; 11:antibiotics11060734. [PMID: 35740141 PMCID: PMC9220107 DOI: 10.3390/antibiotics11060734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 01/27/2023] Open
Abstract
At present, antibiotic resistance represents a global problem in modern medicine. In the near future, humanity may face a situation where medicine will be powerless against resistant bacteria and a post-antibiotic era will come. The development of new antibiotics is either very expensive or ineffective due to rapidly developing bacterial resistance. The need to develop alternative approaches to the treatment of bacterial infections, such as phage therapy, is beyond doubt. The cornerstone of bacterial defense against antibiotics are multidrug resistance (MDR) pumps, which are involved in antibiotic resistance, toxin export, biofilm, and persister cell formation. MDR pumps are the primary non-specific defense of bacteria against antibiotics, while drug target modification, drug inactivation, target switching, and target sequestration are the second, specific line of their defense. All bacteria have MDR pumps, and bacteriophages have evolved along with them and use the bacteria’s need for MDR pumps to bind and penetrate into bacterial cells. The study and understanding of the mechanisms of the pumps and their contribution to the overall resistance and to the sensitivity to bacteriophages will allow us to either seriously delay the onset of the post-antibiotic era or even prevent it altogether due to phage-antibiotic synergy.
Collapse
|
56
|
López-Cano A, Martínez-Miguel M, Guasch J, Ratera I, Arís A, Garcia-Fruitós E. Exploring the impact of the recombinant Escherichia coli strain on defensins antimicrobial activity: BL21 versus Origami strain. Microb Cell Fact 2022; 21:77. [PMID: 35527241 PMCID: PMC9082834 DOI: 10.1186/s12934-022-01803-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/20/2022] [Indexed: 12/24/2022] Open
Abstract
The growing emergence of microorganisms resistant to antibiotics has prompted the development of alternative antimicrobial therapies. Among them, the antimicrobial peptides produced by innate immunity, which are also known as host defense peptides (HDPs), hold great potential. They have been shown to exert activity against both Gram-positive and Gram-negative bacteria, including those resistant to antibiotics. These HDPs are classified into three categories: defensins, cathelicidins, and histatins. Traditionally, HDPs have been chemically synthesized, but this strategy often limits their application due to the high associated production costs. Alternatively, some HDPs have been recombinantly produced, but little is known about the impact of the bacterial strain in the recombinant product. This work aimed to assess the influence of the Escherichia coli strain used as cell factory to determine the activity and stability of recombinant defensins, which have 3 disulfide bonds. For that, an α-defensin [human α-defensin 5 (HD5)] and a β-defensin [bovine lingual antimicrobial peptide (LAP)] were produced in two recombinant backgrounds. The first one was an E. coli BL21 strain, which has a reducing cytoplasm, whereas the second was an E. coli Origami B, that is a strain with a more oxidizing cytoplasm. The results showed that both HD5 and LAP, fused to Green Fluorescent Protein (GFP), were successfully produced in both BL21 and Origami B strains. However, differences were observed in the HDP production yield and bactericidal activity, especially for the HD5-based protein. The HD5 protein fused to GFP was not only produced at higher yields in the E. coli BL21 strain, but it also showed a higher quality and stability than that produced in the Origami B strain. Hence, this data showed that the strain had a clear impact on both HDPs quantity and quality.
Collapse
|
57
|
Palliyil S, Mawer M, Alawfi SA, Fogg L, Tan TH, De Cesare GB, Walker LA, MacCallum DM, Porter AJ, Munro CA. Monoclonal Antibodies Targeting Surface-Exposed Epitopes of Candida albicans Cell Wall Proteins Confer In Vivo Protection in an Infection Model. Antimicrob Agents Chemother 2022; 66:e0195721. [PMID: 35285676 PMCID: PMC9017365 DOI: 10.1128/aac.01957-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Monoclonal antibody (mAb)-based immunotherapies targeting systemic and deep-seated fungal infections are still in their early stages of development, with no licensed antifungal mAbs currently being available for patients at risk. The cell wall glycoproteins of Candida albicans are of particular interest as potential targets for therapeutic antibody generation due to their extracellular location and key involvement in fungal pathogenesis. Here, we describe the generation of recombinant human antibodies specifically targeting two key cell wall proteins (CWPs) in C. albicans: Utr2 and Pga31. These antibodies were isolated from a phage display antibody library using peptide antigens representing the surface-exposed regions of CWPs expressed at elevated levels during in vivo infection. Reformatted human-mouse chimeric mAbs preferentially recognized C. albicans hyphal forms compared to yeast cells, and increased binding was observed when the cells were grown in the presence of the antifungal agent caspofungin. In J774.1 macrophage interaction assays, mAb pretreatment resulted in the faster engulfment of C. albicans cells, suggesting a role of the CWP antibodies as opsonizing agents during phagocyte recruitment. Finally, in a series of clinically predictive mouse models of systemic candidiasis, our lead mAb achieved improved survival (83%) and a several-log reduction of the fungal burden in the kidneys, similar to the levels achieved for the fungicidal drug caspofungin and superior to the therapeutic efficacy of any anti-Candida mAb reported to date.
Collapse
Affiliation(s)
- Soumya Palliyil
- Scottish Biologics Facility, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Mark Mawer
- Scottish Biologics Facility, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Sami A. Alawfi
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Lily Fogg
- Scottish Biologics Facility, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Tyng H. Tan
- Scottish Biologics Facility, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Giuseppe Buda De Cesare
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Louise A. Walker
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Donna M. MacCallum
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Andrew J. Porter
- Scottish Biologics Facility, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeengrid.7107.1, Aberdeen, United Kingdom
| |
Collapse
|
58
|
Mindt BC, DiGiandomenico A. Microbiome Modulation as a Novel Strategy to Treat and Prevent Respiratory Infections. Antibiotics (Basel) 2022; 11:antibiotics11040474. [PMID: 35453224 PMCID: PMC9029693 DOI: 10.3390/antibiotics11040474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023] Open
Abstract
Acute and chronic lower airway disease still represent a major cause of morbidity and mortality on a global scale. With the steady rise of multidrug-resistant respiratory pathogens, such as Pseudomonas aeruginosa and Klebsiella pneumoniae, we are rapidly approaching the advent of a post-antibiotic era. In addition, potentially detrimental novel variants of respiratory viruses continuously emerge with the most prominent recent example being severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To this end, alternative preventive and therapeutic intervention strategies will be critical to combat airway infections in the future. Chronic respiratory diseases are associated with alterations in the lung and gut microbiome, which is thought to contribute to disease progression and increased susceptibility to infection with respiratory pathogens. In this review we will focus on how modulating and harnessing the microbiome may pose a novel strategy to prevent and treat pulmonary infections as well as chronic respiratory disease.
Collapse
|
59
|
Cernooka E, Rumnieks J, Zrelovs N, Tars K, Kazaks A. Diversity of the lysozyme fold: structure of the catalytic domain from an unusual endolysin encoded by phage Enc34. Sci Rep 2022; 12:5005. [PMID: 35322067 PMCID: PMC8943055 DOI: 10.1038/s41598-022-08765-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/10/2022] [Indexed: 12/02/2022] Open
Abstract
Endolysins are bacteriophage-encoded peptidoglycan-degrading enzymes with potential applications for treatment of multidrug-resistant bacterial infections. Hafnia phage Enc34 encodes an unusual endolysin with an N-terminal enzymatically active domain and a C-terminal transmembrane domain. The catalytic domain of the endolysin belongs to the conserved protein family PHA02564 which has no recognizable sequence similarity to other known endolysin types. Turbidity reduction assays indicate that the Enc34 enzyme is active against peptidoglycan from a variety of Gram-negative bacteria including the opportunistic pathogen Pseudomonas aeruginosa PAO1. The crystal structure of the catalytic domain of the Enc34 endolysin shows a distinctive all-helical architecture that distantly resembles the α-lobe of the lysozyme fold. Conserved catalytically important residues suggest a shared evolutionary history between the Enc34 endolysin and GH73 and GH23 family glycoside hydrolases and propose a molecular signature for substrate cleavage for a large group of peptidoglycan-degrading enzymes.
Collapse
Affiliation(s)
- Elina Cernooka
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga, 1067, Latvia
| | - Janis Rumnieks
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga, 1067, Latvia
| | - Nikita Zrelovs
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga, 1067, Latvia
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga, 1067, Latvia.,Faculty of Biology, University of Latvia, Jelgavas 1, Riga, 1004, Latvia
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga, 1067, Latvia.
| |
Collapse
|
60
|
Berini F, Orlandi V, Gornati R, Bernardini G, Marinelli F. Nanoantibiotics to fight multidrug resistant infections by Gram-positive bacteria: hope or reality? Biotechnol Adv 2022; 57:107948. [PMID: 35337933 DOI: 10.1016/j.biotechadv.2022.107948] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022]
Abstract
The spread of antimicrobial resistance in Gram-positive pathogens represents a threat to human health. To counteract the current lack of novel antibiotics, alternative antibacterial treatments have been increasingly investigated. This review covers the last decade's developments in using nanoparticles as carriers for the two classes of frontline antibiotics active on multidrug-resistant Gram-positive pathogens, i.e., glycopeptide antibiotics and daptomycin. Most of the reviewed papers deal with vancomycin nanoformulations, being teicoplanin- and daptomycin-carrying nanosystems much less investigated. Special attention is addressed to nanoantibiotics used for contrasting biofilm-associated infections. The status of the art related to nanoantibiotic toxicity is critically reviewed.
Collapse
Affiliation(s)
- Francesca Berini
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Viviana Orlandi
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Giovanni Bernardini
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Flavia Marinelli
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| |
Collapse
|
61
|
Murugaiyan J, Kumar PA, Rao GS, Iskandar K, Hawser S, Hays JP, Mohsen Y, Adukkadukkam S, Awuah WA, Jose RAM, Sylvia N, Nansubuga EP, Tilocca B, Roncada P, Roson-Calero N, Moreno-Morales J, Amin R, Kumar BK, Kumar A, Toufik AR, Zaw TN, Akinwotu OO, Satyaseela MP, van Dongen MBM. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics (Basel) 2022; 11:200. [PMID: 35203804 PMCID: PMC8868457 DOI: 10.3390/antibiotics11020200] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic resistance, and, in a broader perspective, antimicrobial resistance (AMR), continues to evolve and spread beyond all boundaries. As a result, infectious diseases have become more challenging or even impossible to treat, leading to an increase in morbidity and mortality. Despite the failure of conventional, traditional antimicrobial therapy, in the past two decades, no novel class of antibiotics has been introduced. Consequently, several novel alternative strategies to combat these (multi-) drug-resistant infectious microorganisms have been identified. The purpose of this review is to gather and consider the strategies that are being applied or proposed as potential alternatives to traditional antibiotics. These strategies include combination therapy, techniques that target the enzymes or proteins responsible for antimicrobial resistance, resistant bacteria, drug delivery systems, physicochemical methods, and unconventional techniques, including the CRISPR-Cas system. These alternative strategies may have the potential to change the treatment of multi-drug-resistant pathogens in human clinical settings.
Collapse
Affiliation(s)
- Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - P. Anand Kumar
- Department of Veterinary Microbiology, NTR College of Veterinary Science, Sri Venkateswara Veterinary University, Gannavaram 521102, India;
| | - G. Srinivasa Rao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517502, India;
| | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France;
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | | | - John P. Hays
- Department of Medical Microbiology, Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Yara Mohsen
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria 21544, Egypt;
- Infectious Disease Clinical Pharmacist, Antimicrobial Stewardship Department, International Medical Center Hospital, Cairo 11511, Egypt
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - Wireko Andrew Awuah
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Ruiz Alvarez Maria Jose
- Research Coordination and Support Service, National Institute of Health (ISS) Viale Regina -Elena, 299, 00161 Rome, Italy;
| | - Nanono Sylvia
- Infectious Diseases Institute (IDI), College of Health Sciences, Makerere University, Kampala 7072, Uganda;
| | | | - Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Natalia Roson-Calero
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Javier Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Rohul Amin
- James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh;
| | - Ballamoole Krishna Kumar
- Nitte (Deemed to be University), Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangalore 575018, India;
| | - Abishek Kumar
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Abdul-Rahman Toufik
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Thaint Nadi Zaw
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Oluwatosin O. Akinwotu
- Department of Microbiology and Biotechnology Centre, Maharaja Sayajirao University of Baroda, Vadodara 390002, India;
- Environmental and Biotechnology Unit, Department of Microbiology, University of Ibadan, 200132 Ibadan, Nigeria
| | | | | |
Collapse
|
62
|
Patangia DV, Anthony Ryan C, Dempsey E, Paul Ross R, Stanton C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022; 11:e1260. [PMID: 35212478 PMCID: PMC8756738 DOI: 10.1002/mbo3.1260] [Citation(s) in RCA: 231] [Impact Index Per Article: 115.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
It is well established that the gut microbiota plays an important role in host health and is perturbed by several factors including antibiotics. Antibiotic-induced changes in microbial composition can have a negative impact on host health including reduced microbial diversity, changes in functional attributes of the microbiota, formation, and selection of antibiotic-resistant strains making hosts more susceptible to infection with pathogens such as Clostridioides difficile. Antibiotic resistance is a global crisis and the increased use of antibiotics over time warrants investigation into its effects on microbiota and health. In this review, we discuss the adverse effects of antibiotics on the gut microbiota and thus host health, and suggest alternative approaches to antibiotic use.
Collapse
Affiliation(s)
- Dhrati V. Patangia
- School of MicrobiologyUniversity College CorkCorkIreland
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| | | | - Eugene Dempsey
- School of MicrobiologyUniversity College CorkCorkIreland
| | - Reynolds Paul Ross
- School of MicrobiologyUniversity College CorkCorkIreland
- APC MicrobiomeCorkIreland
| | - Catherine Stanton
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| |
Collapse
|
63
|
Nanayakkara AK, Boucher HW, Fowler VG, Jezek A, Outterson K, Greenberg DE. Antibiotic resistance in the patient with cancer: Escalating challenges and paths forward. CA Cancer J Clin 2021; 71:488-504. [PMID: 34546590 DOI: 10.3322/caac.21697] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/23/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Infection is the second leading cause of death in patients with cancer. Loss of efficacy in antibiotics due to antibiotic resistance in bacteria is an urgent threat against the continuing success of cancer therapy. In this review, the authors focus on recent updates on the impact of antibiotic resistance in the cancer setting, particularly on the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.). This review highlights the health and financial impact of antibiotic resistance in patients with cancer. Furthermore, the authors recommend measures to control the emergence of antibiotic resistance, highlighting the risk factors associated with cancer care. A lack of data in the etiology of infections, specifically in oncology patients in United States, is identified as a concern, and the authors advocate for a centralized and specialized surveillance system for patients with cancer to predict and prevent the emergence of antibiotic resistance. Finding better ways to predict, prevent, and treat antibiotic-resistant infections will have a major positive impact on the care of those with cancer.
Collapse
Affiliation(s)
- Amila K Nanayakkara
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, University of Texas Southwestern, Dallas, Texas
| | - Helen W Boucher
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts
| | - Vance G Fowler
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Amanda Jezek
- Infectious Diseases Society of America, Arlington, Virginia
| | - Kevin Outterson
- CARB-X, Boston, Massachusetts
- Boston University School of Law, Boston, Massachusetts
| | - David E Greenberg
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, University of Texas Southwestern, Dallas, Texas
- Department of Microbiology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
64
|
Li W, Thian ES, Wang M, Wang Z, Ren L. Surface Design for Antibacterial Materials: From Fundamentals to Advanced Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100368. [PMID: 34351704 PMCID: PMC8498904 DOI: 10.1002/advs.202100368] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/27/2021] [Indexed: 05/14/2023]
Abstract
Healthcare-acquired infections as well as increasing antimicrobial resistance have become an urgent global challenge, thus smart alternative solutions are needed to tackle bacterial infections. Antibacterial materials in biomedical applications and hospital hygiene have attracted great interest, in particular, the emergence of surface design strategies offer an effective alternative to antibiotics, thereby preventing the possible development of bacterial resistance. In this review, recent progress on advanced surface modifications to prevent bacterial infections are addressed comprehensively, starting with the key factors against bacterial adhesion, followed by varying strategies that can inhibit biofilm formation effectively. Furthermore, "super antibacterial systems" through pre-treatment defense and targeted bactericidal system, are proposed with increasing evidence of clinical potential. Finally, the advantages and future challenges of surface strategies to resist healthcare-associated infections are discussed, with promising prospects of developing novel antimicrobial materials.
Collapse
Affiliation(s)
- Wenlong Li
- Department of BiomaterialsState Key Lab of Physical Chemistry of Solid SurfaceCollege of MaterialsXiamen UniversityXiamen361005P. R. China
| | - Eng San Thian
- Department of Mechanical EngineeringNational University of SingaporeSingapore117576Singapore
| | - Miao Wang
- Department of BiomaterialsState Key Lab of Physical Chemistry of Solid SurfaceCollege of MaterialsXiamen UniversityXiamen361005P. R. China
| | - Zuyong Wang
- College of Materials Science and EngineeringHunan UniversityChangsha410082P. R. China
| | - Lei Ren
- Department of BiomaterialsState Key Lab of Physical Chemistry of Solid SurfaceCollege of MaterialsXiamen UniversityXiamen361005P. R. China
| |
Collapse
|
65
|
Breiman RF, Blau DM, Mutevedzi P, Akelo V, Mandomando I, Ogbuanu IU, Sow SO, Madrid L, El Arifeen S, Garel M, Thwala NB, Onyango D, Sitoe A, Bassey IA, Keita AM, Alemu A, Alam M, Mahtab S, Gethi D, Varo R, Ojulong J, Samura S, Mehta A, Ibrahim AM, Rahman A, Vitorino P, Baillie VL, Agaya J, Tapia MD, Assefa N, Chowdhury AI, Scott JAG, Gurley ES, Kotloff KL, Jambai A, Bassat Q, Tippett-Barr BA, Madhi SA, Whitney CG. Postmortem investigations and identification of multiple causes of child deaths: An analysis of findings from the Child Health and Mortality Prevention Surveillance (CHAMPS) network. PLoS Med 2021; 18:e1003814. [PMID: 34591862 PMCID: PMC8516282 DOI: 10.1371/journal.pmed.1003814] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 10/14/2021] [Accepted: 09/14/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The current burden of >5 million deaths yearly is the focus of the Sustainable Development Goal (SDG) to end preventable deaths of newborns and children under 5 years old by 2030. To accelerate progression toward this goal, data are needed that accurately quantify the leading causes of death, so that interventions can target the common causes. By adding postmortem pathology and microbiology studies to other available data, the Child Health and Mortality Prevention Surveillance (CHAMPS) network provides comprehensive evaluations of conditions leading to death, in contrast to standard methods that rely on data from medical records and verbal autopsy and report only a single underlying condition. We analyzed CHAMPS data to characterize the value of considering multiple causes of death. METHODS AND FINDINGS We examined deaths identified from December 2016 through November 2020 from 7 CHAMPS sites (in Bangladesh, Ethiopia, Kenya, Mali, Mozambique, Sierra Leone, and South Africa), including 741 neonatal, 278 infant, and 241 child <5 years deaths for which results from Determination of Cause of Death (DeCoDe) panels were complete. DeCoDe panelists included all conditions in the causal chain according to the ICD-10 guidelines and assessed if prevention or effective management of the condition would have prevented the death. We analyzed the distribution of all conditions listed as causal, including underlying, antecedent, and immediate causes of death. Among 1,232 deaths with an underlying condition determined, we found a range of 0 to 6 (mean 1.5, IQR 0 to 2) additional conditions in the causal chain leading to death. While pathology provides very helpful clues, we cannot always be certain that conditions identified led to death or occurred in an agonal stage of death. For neonates, preterm birth complications (most commonly respiratory distress syndrome) were the most common underlying condition (n = 282, 38%); among those with preterm birth complications, 256 (91%) had additional conditions in causal chains, including 184 (65%) with a different preterm birth complication, 128 (45%) with neonatal sepsis, 69 (24%) with lower respiratory infection (LRI), 60 (21%) with meningitis, and 25 (9%) with perinatal asphyxia/hypoxia. Of the 278 infant deaths, 212 (79%) had ≥1 additional cause of death (CoD) beyond the underlying cause. The 2 most common underlying conditions in infants were malnutrition and congenital birth defects; LRI and sepsis were the most common additional conditions in causal chains, each accounting for approximately half of deaths with either underlying condition. Of the 241 child deaths, 178 (75%) had ≥1 additional condition. Among 46 child deaths with malnutrition as the underlying condition, all had ≥1 other condition in the causal chain, most commonly sepsis, followed by LRI, malaria, and diarrheal disease. Including all positions in the causal chain for neonatal deaths resulted in 19-fold and 11-fold increases in attributable roles for meningitis and LRI, respectively. For infant deaths, the proportion caused by meningitis and sepsis increased by 16-fold and 11-fold, respectively; for child deaths, sepsis and LRI are increased 12-fold and 10-fold, respectively. While comprehensive CoD determinations were done for a substantial number of deaths, there is potential for bias regarding which deaths in surveillance areas underwent minimally invasive tissue sampling (MITS), potentially reducing representativeness of findings. CONCLUSIONS Including conditions that appear anywhere in the causal chain, rather than considering underlying condition alone, markedly changed the proportion of deaths attributed to various diagnoses, especially LRI, sepsis, and meningitis. While CHAMPS methods cannot determine when 2 conditions cause death independently or may be synergistic, our findings suggest that considering the chain of events leading to death can better guide research and prevention priorities aimed at reducing child deaths.
Collapse
Affiliation(s)
- Robert F. Breiman
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
- Emory Global Health Institute, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| | - Dianna M. Blau
- Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Portia Mutevedzi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Victor Akelo
- US Centers for Disease Control and Prevention-Kenya, Kisumu, Kenya
| | - Inacio Mandomando
- Centro de Investigação em Saúde de Manhiça [CISM], Manhica, Mozambique
- Instituto Nacional de Saúde [INS], Manhiça, Mozambique
| | | | - Samba O. Sow
- Centre pour le Développement des Vaccins (CVD-Mali), Ministère de la Santé, Bamako, Mali
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lola Madrid
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Shams El Arifeen
- International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh
| | - Mischka Garel
- Emory Global Health Institute, Emory University, Atlanta, Georgia, United States of America
| | - Nana Bukiwe Thwala
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | | | - Antonio Sitoe
- Centro de Investigação em Saúde de Manhiça [CISM], Manhica, Mozambique
| | | | - Adama Mamby Keita
- Centre pour le Développement des Vaccins (CVD-Mali), Ministère de la Santé, Bamako, Mali
| | - Addisu Alemu
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Muntasir Alam
- International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh
| | - Sana Mahtab
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Dickson Gethi
- Kenya Medical Research Institute (KEMRI) Center for Global Health Research, Kisumu, Kenya
| | - Rosauro Varo
- Centro de Investigação em Saúde de Manhiça [CISM], Manhica, Mozambique
- Universitat de Barcelona, Barcelona, Spain
| | | | | | - Ashka Mehta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | | | - Afruna Rahman
- International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh
| | - Pio Vitorino
- Centro de Investigação em Saúde de Manhiça [CISM], Manhica, Mozambique
| | - Vicky L. Baillie
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Janet Agaya
- Kenya Medical Research Institute (KEMRI) Center for Global Health Research, Kisumu, Kenya
| | - Milagritos D. Tapia
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nega Assefa
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | | | - J. Anthony G. Scott
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Emily S. Gurley
- International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Karen L. Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Division of Infectious Disease and Tropical Pediatrics, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Amara Jambai
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça [CISM], Manhica, Mozambique
- ISGlobal, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital de Sant Joan de Deu, University of Barcelona, Barcelona, Spain
- Consorcio de Investigacion Biomedica en Red de Epidemiologia y Salud Publica (CIBERESP), Madrid, Spain
| | | | - Shabir A. Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Cynthia G. Whitney
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
- Emory Global Health Institute, Emory University, Atlanta, Georgia, United States of America
| | | |
Collapse
|
66
|
Kamuyu G, Suen Cheng Y, Willcocks S, Kewcharoenwong C, Kiratisin P, Taylor PW, Wren BW, Lertmemongkolchai G, Stabler RA, Brown J. Sequential Vaccination With Heterologous Acinetobacter baumannii Strains Induces Broadly Reactive Antibody Responses. Front Immunol 2021; 12:705533. [PMID: 34394105 PMCID: PMC8363311 DOI: 10.3389/fimmu.2021.705533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Antibody therapy may be an alternative treatment option for infections caused by the multi-drug resistant (MDR) bacterium Acinetobacter baumannii. As A. baumannii has multiple capsular serotypes, a universal antibody therapy would need to target conserved protein antigens rather than the capsular polysaccharides. We have immunized mice with single or multiple A. baumannii strains to induce antibody responses to protein antigens, and then assessed whether these responses provide cross-protection against a collection of genetically diverse clinical A. baumannii isolates. Immunized mice developed antibody responses to multiple protein antigens. Flow cytometry IgG binding assays and immunoblots demonstrated improved recognition of both homologous and heterologous clinical strains in sera from mice immunized with multiple strains compared to a single strain. The capsule partially inhibited bacterial recognition by IgG and the promotion of phagocytosis by human neutrophils. However, after immunization with multiple strains, serum antibodies to protein antigens promoted neutrophil phagocytosis of heterologous A. baumannii strains. In an infection model, mice immunized with multiple strains had lower bacterial counts in the spleen and liver following challenge with a heterologous strain. These data demonstrate that antibodies targeting protein antigens can improve immune recognition and protection against diverse A. baumannii strains, providing support for their use as an antibody therapy.
Collapse
Affiliation(s)
- Gathoni Kamuyu
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| | - Yat Suen Cheng
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| | - Sam Willcocks
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Chidchamai Kewcharoenwong
- Cellular and Molecular Immunology Unit, Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Pattarachai Kiratisin
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok, Thailand
| | - Peter W Taylor
- School of Pharmacy, University College London, London, United Kingdom
| | - Brendan W Wren
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Ganjana Lertmemongkolchai
- Cellular and Molecular Immunology Unit, Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand.,Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Richard A Stabler
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Jeremy Brown
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| |
Collapse
|
67
|
Avershina E, Shapovalova V, Shipulin G. Fighting Antibiotic Resistance in Hospital-Acquired Infections: Current State and Emerging Technologies in Disease Prevention, Diagnostics and Therapy. Front Microbiol 2021; 12:707330. [PMID: 34367112 PMCID: PMC8334188 DOI: 10.3389/fmicb.2021.707330] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022] Open
Abstract
Rising antibiotic resistance is a global threat that is projected to cause more deaths than all cancers combined by 2050. In this review, we set to summarize the current state of antibiotic resistance, and to give an overview of the emerging technologies aimed to escape the pre-antibiotic era recurrence. We conducted a comprehensive literature survey of >150 original research and review articles indexed in the Web of Science using "antimicrobial resistance," "diagnostics," "therapeutics," "disinfection," "nosocomial infections," "ESKAPE pathogens" as key words. We discuss the impact of nosocomial infections on the spread of multi-drug resistant bacteria, give an overview over existing and developing strategies for faster diagnostics of infectious diseases, review current and novel approaches in therapy of infectious diseases, and finally discuss strategies for hospital disinfection to prevent MDR bacteria spread.
Collapse
Affiliation(s)
- Ekaterina Avershina
- Department of Biotechnology, Inland Norway University of Applied Sciences, Hamar, Norway
- Laboratory or Postgenomic Technologies, Izmerov Research Institute of Occupational Health, Moscow, Russia
| | - Valeria Shapovalova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Centre for Strategic Planning of FMBA of Russia, Moscow, Russia
| | - German Shipulin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Centre for Strategic Planning of FMBA of Russia, Moscow, Russia
| |
Collapse
|
68
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a challenge for ongoing efforts to combat antimicrobial-resistant (AMR) bacterial infections. As we learn more about COVID-19 disease and drug stewardship evolves, there is likely to be a lasting impact of increased use of antimicrobial agents and antibiotics, as well as a lack of consistent access to health care across many populations. Sexually transmitted infections have been underreported during the pandemic and are often caused by some of the most drug-resistant pathogens. In their recent article in mBio, Parzych et al. (E. M. Parzych, S. Gulati, B. Zheng, M. A. Bah, et al., mBio 12:e00242-21, 2021, https://doi.org/10.1128/mBio.00242-21) focus on protection against Neisseria gonorrhoeae infection via in vivo delivery of an antigonococcal DNA-encoded antibody that has been modified for increased complement activation. Nucleic acid approaches are highly adaptable and could be tremendously beneficial for personalized strategies to combat AMR pathogens.
Collapse
|
69
|
Fatoba AJ, Adeleke VT, Maharaj L, Okpeku M, Adeniyi AA, Adeleke MA. Immunoinformatics Design of Multiepitope Vaccine Against Enterococcus faecium Infection. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10245-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
70
|
Yamawaki T, Nakakido M, Ujiie K, Aikawa C, Nakagawa I, Tsumoto K. Characterization of a putative maltodextrin-binding protein of Streptococcus pyogenes, SPs0871 and the development of a VHH inhibitor. Biochem Biophys Res Commun 2021; 565:1-7. [PMID: 34077827 DOI: 10.1016/j.bbrc.2021.05.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 11/30/2022]
Abstract
Streptococcus pyogenes causes a wide range of human infections. Currently, antibiotics are the main treatment for S. pyogenes infection, but serious anti-microbial resistance requires alternative treatment options. To develop a novel strategy for treatment, we physicochemically characterized SPs0871, a putative maltose/maltodextrin-binding protein that is thought to have important roles in the pathogenesis of invasive streptococci. We obtained a variable domain of heavy chain of heavy-chain antibody, the smallest unit of an antibody, which specifically binds to SPs0871. Although the VHH completely inhibited the binding of maltodextrins to SPs0871, the inhibition did not lead to growth suppression of the bacteria. Our results provide important insights for development of VHH as an anti-streptococcal therapeutic.
Collapse
Affiliation(s)
- Tsukushi Yamawaki
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Makoto Nakakido
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Kan Ujiie
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Chihiro Aikawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan; Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
71
|
Sebbane F, Lemaître N. Antibiotic Therapy of Plague: A Review. Biomolecules 2021; 11:724. [PMID: 34065940 PMCID: PMC8151713 DOI: 10.3390/biom11050724] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Plague-a deadly disease caused by the bacterium Yersinia pestis-is still an international public health concern. There are three main clinical forms: bubonic plague, septicemic plague, and pulmonary plague. In all three forms, the symptoms appear suddenly and progress very rapidly. Early antibiotic therapy is essential for countering the disease. Several classes of antibiotics (e.g., tetracyclines, fluoroquinolones, aminoglycosides, sulfonamides, chloramphenicol, rifamycin, and β-lactams) are active in vitro against the majority of Y. pestis strains and have demonstrated efficacy in various animal models. However, some discrepancies have been reported. Hence, health authorities have approved and recommended several drugs for prophylactic or curative use. Only monotherapy is currently recommended; combination therapy has not shown any benefits in preclinical studies or case reports. Concerns about the emergence of multidrug-resistant strains of Y. pestis have led to the development of new classes of antibiotics and other therapeutics (e.g., LpxC inhibitors, cationic peptides, antivirulence drugs, predatory bacteria, phages, immunotherapy, host-directed therapy, and nutritional immunity). It is difficult to know which of the currently available treatments or therapeutics in development will be most effective for a given form of plague. This is due to the lack of standardization in preclinical studies, conflicting data from case reports, and the small number of clinical trials performed to date.
Collapse
Affiliation(s)
- Florent Sebbane
- Univ. Lille, Inserm, CNRS, Institut Pasteur Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nadine Lemaître
- Univ. Lille, Inserm, CNRS, Institut Pasteur Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, F-59000 Lille, France
- Laboratoire de Bactériologie-Hygiène, Centre Hospitalier Universitaire Amiens Picardie, UR 4294, Agents Infectieux, Résistance et Chimiothérapie (AGIR), Université de Picardie Jules Verne, F-80000 Amiens, France
| |
Collapse
|
72
|
Arato V, Raso MM, Gasperini G, Berlanda Scorza F, Micoli F. Prophylaxis and Treatment against Klebsiella pneumoniae: Current Insights on This Emerging Anti-Microbial Resistant Global Threat. Int J Mol Sci 2021; 22:4042. [PMID: 33919847 PMCID: PMC8070759 DOI: 10.3390/ijms22084042] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Klebsiella pneumoniae (Kp) is an opportunistic pathogen and the leading cause of healthcare-associated infections, mostly affecting subjects with compromised immune systems or suffering from concurrent bacterial infections. However, the dramatic increase in hypervirulent strains and the emergence of new multidrug-resistant clones resulted in Kp occurrence among previously healthy people and in increased morbidity and mortality, including neonatal sepsis and death across low- and middle-income countries. As a consequence, carbapenem-resistant and extended spectrum β-lactamase-producing Kp have been prioritized as a critical anti-microbial resistance threat by the World Health Organization and this has renewed the interest of the scientific community in developing a vaccine as well as treatments alternative to the now ineffective antibiotics. Capsule polysaccharide is the most important virulence factor of Kp and plays major roles in the pathogenesis but its high variability (more than 100 different types have been reported) makes the identification of a universal treatment or prevention strategy very challenging. However, less variable virulence factors such as the O-Antigen, outer membrane proteins as fimbriae and siderophores might also be key players in the fight against Kp infections. Here, we review elements of the current status of the epidemiology and the molecular pathogenesis of Kp and explore specific bacterial antigens as potential targets for both prophylactic and therapeutic solutions.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., via Fiorentina 1, 53100 Siena, Italy; (V.A.); (M.M.R.); (G.G.); (F.B.S.)
| |
Collapse
|
73
|
Spirescu VA, Chircov C, Grumezescu AM, Andronescu E. Polymeric Nanoparticles for Antimicrobial Therapies: An Up-To-Date Overview. Polymers (Basel) 2021; 13:724. [PMID: 33673451 PMCID: PMC7956825 DOI: 10.3390/polym13050724] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the many advancements in the pharmaceutical and medical fields and the development of numerous antimicrobial drugs aimed to suppress and destroy pathogenic microorganisms, infectious diseases still represent a major health threat affecting millions of lives daily. In addition to the limitations of antimicrobial drugs associated with low transportation rate, water solubility, oral bioavailability and stability, inefficient drug targeting, considerable toxicity, and limited patient compliance, the major cause for their inefficiency is the antimicrobial resistance of microorganisms. In this context, the risk of a pre-antibiotic era is a real possibility. For this reason, the research focus has shifted toward the discovery and development of novel and alternative antimicrobial agents that could overcome the challenges associated with conventional drugs. Nanotechnology is a possible alternative, as there is significant evidence of the broad-spectrum antimicrobial activity of nanomaterials and nanoparticles in particular. Moreover, owing to their considerable advantages regarding their efficient cargo dissolving, entrapment, encapsulation, or surface attachment, the possibility of forming antimicrobial groups for specific targeting and destruction, biocompatibility and biodegradability, low toxicity, and synergistic therapy, polymeric nanoparticles have received considerable attention as potential antimicrobial drug delivery agents. In this context, the aim of this paper is to provide an up-to-date overview of the most recent studies investigating polymeric nanoparticles designed for antimicrobial therapies, describing both their targeting strategies and their effects.
Collapse
Affiliation(s)
- Vera Alexandra Spirescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| |
Collapse
|
74
|
Stoica C, Cox G. Old problems and new solutions: antibiotic alternatives in food animal production. Can J Microbiol 2021; 67:427-444. [PMID: 33606564 DOI: 10.1139/cjm-2020-0601] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The antimicrobial resistance crisis is a Global Health challenge that impacts humans, animals, and the environment alike. In response to increased demands for animal protein and by-products, there has been a substantial increase in the use of antimicrobial agents in the animal industry. Indeed, they are extensively used to prevent, control, and (or) treat disease in animals. In addition to infection control, in-feed supplementation with antimicrobials became common practice for growth promotion of livestock. Unfortunately, the global overuse of antimicrobials has contributed to the emergence and spread of resistance. As such, many countries have implemented policies and approaches to eliminate the use of antimicrobials as growth promoters in food animals, which necessitates the need for alternate and One Health strategies to maintain animal health and welfare. This review summarizes the antimicrobial resistance crisis from Global Health and One Health perspectives. In addition, we outline examples of potential alternate strategies to circumvent antimicrobial use in animal husbandry practices, including antivirulence agents, bacteriophages, and nutritional measures to control bacterial pathogens. Overall, these alternate strategies require further research and development efforts, including assessment of efficacy and the associated development, manufacturing, and labor costs.
Collapse
Affiliation(s)
- Celine Stoica
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.,Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Georgina Cox
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.,Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
75
|
Wang N, Qi F, Yu H, Yestrepsky BD, Larsen SD, Shi H, Ji J, Anderson DW, Li H, Sun H. Physicochemical properties and formulation development of a novel compound inhibiting Staphylococcus aureus biofilm formation. PLoS One 2021; 16:e0246408. [PMID: 33556134 PMCID: PMC7870075 DOI: 10.1371/journal.pone.0246408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/18/2021] [Indexed: 11/25/2022] Open
Abstract
The emergence of antibiotic resistance over the past several decades has given urgency to new antibacterial strategies that apply less selective pressure. A new class of anti-virulence compounds were developed that are active against methicillin-resistant Staphylococcus aureus (MRSA), by inhibiting bacterial virulence without hindering their growth to reduce the selective pressure for resistance development. One of the compounds CCG-211790 has demonstrated potent anti-biofilm activity against MRSA. This new class of anti-virulence compounds inhibited the gene expression of virulence factors involved in biofilm formation and disrupted the biofilm structures. In this study, the physicochemical properties of CCG-211790, including morphology, solubility in pure water or in water containing sodium dodecyl sulfate, solubility in organic solvents, and stability with respect to pH were investigated for the first time. Furthermore, a topical formulation was developed to enhance the therapeutic potential of the compound. The formulation demonstrated acceptable properties for drug release, viscosity, pH, cosmetic elegance and stability of over nine months.
Collapse
Affiliation(s)
- Nan Wang
- Department of Mechanical and Aerospace Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Feng Qi
- Department of Mechanical and Aerospace Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Haqing Yu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Bryan D. Yestrepsky
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Scott D. Larsen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Honglan Shi
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, United States of America
| | - Juan Ji
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - David W. Anderson
- Ivogen Inc. (Subsidiary of Nanova, Inc.), Columbia, Missouri, United States of America
| | - Hao Li
- Department of Mechanical and Aerospace Engineering, University of Missouri, Columbia, Missouri, United States of America
- * E-mail: (HS); (HL)
| | - Hongmin Sun
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail: (HS); (HL)
| |
Collapse
|
76
|
Mourenza Á, Gil JA, Mateos LM, Letek M. Novel Treatments and Preventative Strategies Against Food-Poisoning Caused by Staphylococcal Species. Pathogens 2021; 10:91. [PMID: 33498299 PMCID: PMC7909252 DOI: 10.3390/pathogens10020091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
Staphylococcal infections are a widespread cause of disease in humans. In particular, S. aureus is a major causative agent of infection in clinical medicine. In addition, these bacteria can produce a high number of staphylococcal enterotoxins (SE) that may cause food intoxications. Apart from S. aureus, many coagulase-negative Staphylococcus spp. could be the source of food contamination. Thus, there is an active research work focused on developing novel preventative interventions based on food supplements to reduce the impact of staphylococcal food poisoning. Interestingly, many plant-derived compounds, such as polyphenols, flavonoids, or terpenoids, show significant antimicrobial activity against staphylococci, and therefore these compounds could be crucial to reduce the incidence of food intoxication in humans. Here, we reviewed the most promising strategies developed to prevent staphylococcal food poisoning.
Collapse
Affiliation(s)
- Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
| | - José A. Gil
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Luis M. Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain; (Á.M.); (J.A.G.)
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
| |
Collapse
|
77
|
Streicher LM. Exploring the future of infectious disease treatment in a post-antibiotic era: A comparative review of alternative therapeutics. J Glob Antimicrob Resist 2021; 24:285-295. [PMID: 33484895 DOI: 10.1016/j.jgar.2020.12.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic resistance is projected to be one of the greatest healthcare challenges of the 21st century. As the efficacy of these critical drugs wanes and the discovery of new antibiotics stagnates, exploration of alternative therapies could offer a much needed solution. Although numerous alternative therapies are currently under investigation, three in particular appear poised for long-term success, namely antimicrobial oligonucleotides, monoclonal antibodies and phage therapy. Antimicrobial oligonucleotides could conceivably offer the greatest spectrum of activity while having the lowest chance of unrecoverable resistance. Bacteriophages, while most susceptible to resistance, are inexhaustible, inexpensive and exceptionally adept at eliminating biofilm-associated infections. And although monoclonal antibodies may have limited access to such recalcitrant bacteria, these agents are uniquely able to neutralise exotoxins and other diffusible virulence factors. This comparative review seeks to illuminate these promising therapies and to encourage the scientific and financial support necessary to usher in the next generation of infectious disease treatment.
Collapse
|
78
|
Min TT, Yamabhai M. Human Hexa-Histidine-Tagged Single-Chain Variable Fragments for Bioimaging of Bacterial Infections. ACS OMEGA 2021; 6:762-774. [PMID: 33458528 PMCID: PMC7808144 DOI: 10.1021/acsomega.0c05340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
The single-chain variable fragment (scFv) of monoclonal antibodies is a promising recombinant nanostructure for various medical applications, including bioimaging and targeted therapy. While numerous scFv antibodies against eukaryotic cell surface proteins (especially cancer biomarkers) have been generated and engineered to suit various purposes, only a few specific scFv against bacterial cell surfaces have been developed, especially those of human origin. Recent incidents of emerging multidrug-resistant pathogenic bacteria and the realization of the importance of a balanced microbiota on the health of the host has led to more interests in the development of recombinant antibacterial antibodies as a detection probe or targeted therapy for bacterial infections. This study reports the generation of two specific human antibacterial scFv using phage display antibody technology. The recombinant scFv fragments of about 30 kDa and a diameter of 5 nm were produced and purified from engineered Escherichia coli that can enhance cytosolic disulfide bond formation. As a proof of principle, Propionibacterium acnes and Pseudomonas aeruginosa were used as model Gram-positive and Gram-negative bacteria, respectively. Specificity at the strain and species level to both planktonic and biofilm forms of these bacteria were demonstrated in various assay formats, namely, ELISA, flow cytometry, western blot, immunofluorescence, and electron microscopy via the hexa-histidine tag. This recombinant scFv generation platform can be applied for other bacteria, and since the scFv obtained has a benefit of being a human origin, it could be conveniently engineered for various therapeutic or theranostic applications with minimized adverse immunoreaction.
Collapse
|
79
|
Cacciatore I, Marinelli L. Patent evaluation of US2019338018 (A1) 2019-11-07 (antibody fragments for the treatment of biofilm-related disorders). Expert Opin Ther Pat 2020; 30:907-909. [PMID: 32970473 DOI: 10.1080/13543776.2020.1828864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION To date, microbial infections are also difficult to eradicate due to the increasing capability of bacteria to form a biofilm. In the era of antibiotic resistance, antibody-based approaches represent great promise in curing infective pathogens. The authors of US2019338018 patent propose a method for the treatment of biofilm-related disorders by using specific antibody fragments. AREAS COVERED The US2019338018 patent reports antibody fragments, pharmaceutical composition that contains it, and their application for the treatment of biofilm-linked disorders. Proof concept and preclinical results show that mAb mIhfB4NTHI Fab caused robust eradication of the biofilm in the middle ear lumen of chinchillas affected by Hemophilus influenzae infection. EXPERT OPINION Fab fragments of the US2019338018 patent are new in a general concept to treat bacterial biofilms and biofilm-linked disorders. However, pre-clinical data are only shown for the treatment with Fab fragments of infections caused by H. influenzae in the middle ear of chinchillas. There are no clinical trials that demonstrate that the treatment with Fab fragments may induce a disruption of biofilm produced by H. influenzae or other pathogens and an anti-inflammatory response in infected patients.
Collapse
Affiliation(s)
- Ivana Cacciatore
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara , Chieti, Italy
| | - Lisa Marinelli
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara , Chieti, Italy
| |
Collapse
|
80
|
Hotinger JA, May AE. Antibodies Inhibiting the Type III Secretion System of Gram-Negative Pathogenic Bacteria. Antibodies (Basel) 2020; 9:antib9030035. [PMID: 32726928 PMCID: PMC7551047 DOI: 10.3390/antib9030035] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogenic bacteria are a global health threat, with over 2 million infections caused by Gram-negative bacteria every year in the United States. This problem is exacerbated by the increase in resistance to common antibiotics that are routinely used to treat these infections, creating an urgent need for innovative ways to treat and prevent virulence caused by these pathogens. Many Gram-negative pathogenic bacteria use a type III secretion system (T3SS) to inject toxins and other effector proteins directly into host cells. The T3SS has become a popular anti-virulence target because it is required for pathogenesis and knockouts have attenuated virulence. It is also not required for survival, which should result in less selective pressure for resistance formation against T3SS inhibitors. In this review, we will highlight selected examples of direct antibody immunizations and the use of antibodies in immunotherapy treatments that target the bacterial T3SS. These examples include antibodies targeting the T3SS of Pseudomonas aeruginosa, Yersinia pestis, Escherichia coli, Salmonella enterica, Shigella spp., and Chlamydia trachomatis.
Collapse
|
81
|
Antibacterial Drug Development: A New Approach Is Needed for the Field to Survive and Thrive. Antibiotics (Basel) 2020; 9:antibiotics9070412. [PMID: 32679767 PMCID: PMC7400211 DOI: 10.3390/antibiotics9070412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
It is often said that the marketplace for new antibiotics is broken. This notion is supported by the observation that many recently-approved antibiotics to treat drug-resistant bacteria have failed commercially in a spectacular fashion. Today, companies with peak market-cap values in excess of USD 500 million to 1 billion prior to product launch regularly sell for pennies on the dollar a few years after market introduction. It is possible, however, that the market is not as broken as we perceive. That is, in the collective mind of the clinician, recently-approved antibiotics may be too-poorly differentiated to justify their broad use and inordinate cost relative to those already existing. Perhaps we in the antibacterial drug development field must change our way of thinking if we are to survive and thrive. Rather than reflexively developing new β-lactam-β-lactamase inhibitor combinations for every new enzyme that evades our current inhibitors, we should focus discovery and development efforts on agents that revolutionize how we potentiate antibiotics. To this end, there has been renewed interest in phage therapies, virulence inhibitors, bacterial growth rate modulators, monoclonal antibodies, and other approaches to augment antibiotic effects. Herein, we suggest that the unmet medical need is less about adding poorly-differentiated antibiotics to our armamentarium and more about the need for innovation in how we augment antibiotic regimen effects.
Collapse
|
82
|
Drayton M, Kizhakkedathu JN, Straus SK. Towards Robust Delivery of Antimicrobial Peptides to Combat Bacterial Resistance. Molecules 2020; 25:molecules25133048. [PMID: 32635310 PMCID: PMC7412191 DOI: 10.3390/molecules25133048] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial peptides (AMPs), otherwise known as host defence peptides (HDPs), are naturally occurring biomolecules expressed by a large array of species across the phylogenetic kingdoms. They have great potential to combat microbial infections by directly killing or inhibiting bacterial activity and/or by modulating the immune response of the host. Due to their multimodal properties, broad spectrum activity, and minimal resistance generation, these peptides have emerged as a promising response to the rapidly concerning problem of multidrug resistance (MDR). However, their therapeutic efficacy is limited by a number of factors, including rapid degradation, systemic toxicity, and low bioavailability. As such, many strategies have been developed to mitigate these limitations, such as peptide modification and delivery vehicle conjugation/encapsulation. Oftentimes, however, particularly in the case of the latter, this can hinder the activity of the parent AMP. Here, we review current delivery strategies used for AMP formulation, focusing on methodologies utilized for targeted infection site release of AMPs. This specificity unites the improved biocompatibility of the delivery vehicle with the unhindered activity of the free AMP, providing a promising means to effectively translate AMP therapy into clinical practice.
Collapse
Affiliation(s)
- Matthew Drayton
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada;
| | - Jayachandran N. Kizhakkedathu
- Department of Pathology and Laboratory Medicine, and Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Life Sciences Centre, Vancouver, BC V6T 1Z3, Canada;
| | - Suzana K. Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada;
- Correspondence: ; Tel.: +1-604-822-2537
| |
Collapse
|
83
|
Hansford KA. Nontraditional Antibiotics—Challenges and Triumphs. Antibiotics (Basel) 2020; 9:antibiotics9040169. [PMID: 32283718 PMCID: PMC7235707 DOI: 10.3390/antibiotics9040169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 11/24/2022] Open
Affiliation(s)
- Karl A Hansford
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland, St Lucia 4072, Australia
| |
Collapse
|