1
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
2
|
Li X, Dong S, Pan Q, Liu N, Zhang Y. Antibiotic conjugates: Using molecular Trojan Horses to overcome drug resistance. Biomed Pharmacother 2025; 186:118007. [PMID: 40268370 DOI: 10.1016/j.biopha.2025.118007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 04/25/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a global health crisis due to the rapid emergence of multi-drug-resistant bacteria. The paucity of novel antibiotics in the clinical pipeline has exacerbated this issue, thereby warranting the development of new antibacterial therapies. The 'Trojan Horse' strategy entails conjugating antibiotics with bioactive components that not only facilitate the entry of antibiotic molecules into bacterial cells by circumventing the membrane barriers, but also augment the effects of conventional antibiotics against recalcitrant pathogens. These Trojan Horse elements can also serve as a promising tool for repurposing drugs with hitherto unexamined antimicrobial activity, or drugs with limited clinical utility due to considerable toxic side effects. In this review, we have discussed the current state of research on antibiotic conjugates with monoclonal antibodies (mAbs), antimicrobial peptides (AMPs) and the iron-chelating siderophores. The rationale and mechanisms of different antibiotic conjugates have been summarized, and the preclinical and clinical evidence pertaining to the activity of these conjugates against drug-resistant pathogens have been reviewed. Furthermore, the challenges associated with the clinical translation of these novel antimicrobials, and the future research directions have also been discussed. While antibiotic conjugates offer an attractive alternative to conventional antimicrobials, there are several obstacles to their clinical translation. A greater understanding of the mechanisms underlying AMR, and continuing advances in genetic engineering, synthetic biology, and bioinformatics will be crucial in designing more selective, potent, and safe antibiotic conjugates for tackling multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Rehabilitation, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
3
|
Dzigba P, Seth MA, Greenlee-Wacker MC, Swarts BM. Redirecting the host immune response to bacterial infection with antibody-recruiting molecules (ARMs). Curr Opin Chem Biol 2025; 86:102585. [PMID: 40117716 DOI: 10.1016/j.cbpa.2025.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/23/2025]
Abstract
The increasing prevalence of antibiotic resistance, the stagnation of antibiotic development, and the adaptive capacity of bacteria to subvert the host immune response combine to pose significant global health concerns. Consequently, there is an urgent need to develop alternative therapeutic approaches to combat bacterial infections. Antibody-recruiting molecules (ARMs), which are bispecific small molecules that recruit endogenous antibodies to pathogenic cells or viruses, offer a promising avenue to harness the host immune system to target various diseases. In this review, we cover ARM strategies that have been developed for bacterial pathogens, including Gram-positive bacteria, Gram-negative bacteria, and mycobacteria, and we discuss the prospects and challenges of utilizing ARMs as alternatives to traditional antibiotic therapies.
Collapse
Affiliation(s)
- Priscilla Dzigba
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, USA; Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Megan A Seth
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Mallary C Greenlee-Wacker
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA.
| | - Benjamin M Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, USA; Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859, USA.
| |
Collapse
|
4
|
Zhai Y, Wang H, Sun K, Yuan Y, Yin S, Fang J, Zheng W, Wudong G, Liu X, Yang Y, Zhou D, Liu W, Jin Y, Wang A. Enhancing host defense against Brucella: The immune effect exerted by anti-OMP16 monoclonal antibody. Int Immunopharmacol 2025; 148:114142. [PMID: 39930647 DOI: 10.1016/j.intimp.2025.114142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 05/08/2025]
Abstract
Brucellosis is an important zoonotic infection caused by the Gram-negative, facultative intracellular bacterium, Brucella. The disease is widespread and prevalent throughout the world, posing a serious threat to human health, with over 500,000 new cases each year. Combination antibiotic therapy is the current treatment for brucellosis, but owing to its intracellular survival capability, the effectiveness of antibiotics is significantly reduced, Monoclonal antibodies (mAbs) are a promising class of biodrugs with a wide range of applications in the clinical treatment of disease. Brucella outer membrane protein 16 (OMP16) is a crucial virulence factor of Brucella for maintaining outer membrane integrity and survival; thus, it is a potential immunotherapy target. In this study, we demonstrated that an anti-Brucella OMP16 IgM-type monoclonal antibody, B7, was able to bind to and agglutinate Brucella abortus A19, and activate the complement system to kill the bacteria. We found that B7 could enhance phagocytosis and killing against Brucella by activating complement in RAW264.7 macrophages, upregulate intracellular nitric oxide and reactive oxygen species levels to enhance cellular resistance to infection, and induce the expression of relevant immunomodulatory cytokines. The antibody's immunoprotective effect was confirmed in a mouse model of Brucella infection. Our study demonstrated that B7 mAb has the potential to provide a new immunotherapeutic approach for the clinical treatment of brucellosis and for related vaccine and drug research.
Collapse
Affiliation(s)
- Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Hui Wang
- Jiagedaqi District Livestock and Aquaculture Service Center, Jiagedaqi 165000, Heilongjiang, China
| | - Kaihui Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Ye Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Shurong Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Jiaoyang Fang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Weifang Zheng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Gaowa Wudong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Yuanhao Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Wei Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China.
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100 China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100 China.
| |
Collapse
|
5
|
Mateu-Borrás M, Dublin SR, Kang J, Monroe HL, Sen-Kilic E, Miller SJ, Witt WT, Chapman JA, Pyles GM, Nallar SC, Huckaby AB, Yang E, Rocuskie-Marker C, Grund ME, Amin MS, Lukomski S, Snyder GA, Ray K, Lewis GK, Ricke DO, Damron FH, Barbier M. Novel broadly reactive monoclonal antibody protects against Pseudomonas aeruginosa infection. Infect Immun 2025; 93:e0033024. [PMID: 39670709 PMCID: PMC11784295 DOI: 10.1128/iai.00330-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/23/2024] [Indexed: 12/14/2024] Open
Abstract
The incidence of infections attributed to antimicrobial-resistant (AMR) pathogens has increased exponentially over the recent decades reaching 1.27 million deaths worldwide in 2019. Without intervention, these infections are predicted to cause up to 10 million deaths a year and incur costs of up to 100 trillion US dollars globally by 2050. The emergence of AMR bacteria such as the ESKAPEE pathogens, and in particular Pseudomonas aeruginosa and species from the genus Burkholderia, underscores an urgent need for new therapeutic strategies. Monoclonal antibody (mAb) therapy offers a promising alternative to treat and prevent bacterial infections. In this study, we used peptides from highly conserved areas of the bacterial flagellin to generate monoclonal antibodies capable of broad binding to flagellated Gram-negative bacteria. We generated a broadly reactive IgG2bĸ mAb (WVDC-2109) that recognizes P. aeruginosa, Burkholderia sp., and other Gram-negative pathogens of interest. Characterization of the therapeutic potential of this antibody was determined using P. aeruginosa as model. In vitro characterization of WVDC-2109 demonstrated complement-mediated bactericidal activity and enhanced opsonophagocytosis of P. aeruginosa. Prophylactic administration of WVDC-2109 markedly improved survival and outcome in a lethal sepsis model and a sub-lethal murine pneumonia model of P. aeruginosa infection, reducing bacterial burden and inflammation. These findings suggest that WVDC-2109 and similar FliC-targeting antibodies could be valuable in preventing or treating diseases caused by P. aeruginosa as well as other life-threatening diseases of concern.IMPORTANCEAntimicrobial resistance (AMR) costs hundreds of thousands of lives and billions of dollars annually. To protect the population against these infections, it is imperative to develop new medical countermeasures targeting AMR pathogens like P. aeruginosa and Burkholderia sp. The administration of broadly reactive monoclonal antibodies can represent an alternative to treat and prevent infections caused by multi-drug-resistant bacteria. Unlike vaccines, antibodies can provide protection regardless of the immune status of the infected host. In this study, we generated an antibody capable of recognizing flagellin from P. aeruginosa and B. pseudomallei along with other Gram-negative pathogens of concern. Our findings demonstrate that the administration of the monoclonal antibody WVDC-2109 enhances survival rates and outcomes in different murine models of P. aeruginosa infection. These results carry significant implications in the field given that there are no available vaccines for P. aeruginosa.
Collapse
Affiliation(s)
- Margalida Mateu-Borrás
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Spencer R. Dublin
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Jason Kang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Hunter L. Monroe
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Sarah J. Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - William T. Witt
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Joshua A. Chapman
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Gage M. Pyles
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Shreeram C. Nallar
- School of Medicine, Division of Vaccine Research, Institute of Human Virology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Evita Yang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Carleena Rocuskie-Marker
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Megan E. Grund
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Md Shahrier Amin
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Slawomir Lukomski
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Greg A. Snyder
- School of Medicine, Division of Vaccine Research, Institute of Human Virology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Krishanu Ray
- School of Medicine, Division of Vaccine Research, Institute of Human Virology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - George K. Lewis
- School of Medicine, Division of Vaccine Research, Institute of Human Virology, University of Maryland Baltimore, Baltimore, Maryland, USA
| | | | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
6
|
Cao M, Wang S, Zhou S, Yan M, Zou Y, Cui Y, Lou X, Gao Y, Chen Y, Han Z, Qian Y, Chen J, Li X. Development of monoclonal antibodies against P. gingivalis Mfa1 and their protective capacity in an experimental periodontitis model. mSphere 2025; 10:e0072124. [PMID: 39699191 PMCID: PMC11774036 DOI: 10.1128/msphere.00721-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis), a gram-negative, black-pigmented anaerobe, is a major etiological agent and a leading cause of periodontitis. Fimbriae protein Mfa1 is a key virulence factor of P. gingivalis and plays a crucial role in bacterial adhesion, colonization, biofilm formation, and persistent inflammation, making it a promising therapeutic target. However, the role of anti-Mfa1 antibodies and the underlying protective mechanisms remain largely unexplored. Here, we developed and characterized the monoclonal antibodies (mAbs) targeting the Mfa1 protein of P. gingivalis. Function analysis showed that anti-Mfa1 mAbs mediated bacterial agglutination and inhibited P. gingivalis adhesion to saliva-coated hydroxyapatite and host cells. Notably, anti-Mfa1 mAbs significantly reduced bacterial burden and alveolar bone loss in a P. gingivalis-induced experimental periodontitis model. These results show that anti-Mfa1 mAbs can be beneficial in alleviating P. gingivalis infections, and provide important insights for the development of adequate adjuvant treatment regimens for Mfa1-targeted therapeutics. IMPORTANCE Fimbriae (pili) play an important role in bacterial adhesion, invasion of host cells and tissues, and formation of biofilms. Studies have shown that two types of fimbriae of Porphyromonas gingivalis, FimA and Mfa1, are important for colonization and infection through their binding to host tissues and other bacteria. While anti-FimA antibodies have been shown to improve periodontitis, the effect of anti-Mfa1 antibodies on P. gingivalis infection and periodontitis was previously unknown. In this study, we report for the first time that anti-Mfa1 monoclonal antibodies can reduce P. gingivalis infection and improve periodontitis. These findings suggest that Mfa1 represents a promising therapeutic target, and the development of anti-Mfa1 mAbs holds a potential as essential diagnostic and adjunctive therapeutic tools for managing P. gingivalis-related diseases.
Collapse
Affiliation(s)
- Mingya Cao
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Siyu Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Shengke Zhou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Min Yan
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yu Zou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yuan Cui
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Xinyu Lou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yichang Gao
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Ying Chen
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Zijing Han
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yi Qian
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| | - Jingying Chen
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| |
Collapse
|
7
|
Luo L, Li Q, Xing C, Li C, Pan Y, Sun H, Yu X, Wen K, Shen J, Wang Z. Antibody-based therapy: An alternative for antimicrobial treatment in the post-antibiotic era. Microbiol Res 2025; 290:127974. [PMID: 39577369 DOI: 10.1016/j.micres.2024.127974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
The consecutive growth of antimicrobial resistance and the spread of resistance genes worldwide, especially the emergence of superbugs, have made traditional antibiotic-based treatments inadequate to fight bacterial infections. Therefore, new therapeutic modalities for bacterial infections are urgently needed. Antibodies are considered to be an effective alternative to antibiotics. The emergence and advancement of technologies such as hybridoma, antibody purification, transgenic mice, phage display, and protein engineering have enabled the production of large quantities of humanized antibodies with high purity and affinity. Antibodies has achieved remarkable achievements in the field of medicine in the past decades. Antibody-based therapy is expected to be an effective way to treat drug-resistant bacterial infections in the post-antibiotic era due to its merits of high specificity, which leads to no selective pressure on non-target bacteria and could cooperate with antibiotics to enhance the antimicrobial effect. This review first introduces the mechanism of action of antibodies against bacterial infections, then summarizes the reported antimicrobial antibodies according to different targets, discusses the advantages and limitations of the antibody-based therapy for antimicrobial treatment, and finally, the perspectives of antimicrobial antibodies developing have been prospected, providing a reference for the development of new antimicrobial antibodies.
Collapse
Affiliation(s)
- Liang Luo
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Chen Xing
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - Chenglong Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, PR China
| | - Yantong Pan
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - He Sun
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Xuezhi Yu
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China.
| |
Collapse
|
8
|
Rai P, Mehrotra S, Prajapati VK. Exploring immunotherapy to control human infectious diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:389-429. [PMID: 39978973 DOI: 10.1016/bs.apcsb.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Infectious diseases continue to pose significant challenges to global health, especially with the rise of antibiotic resistance and emerging pathogens. Traditional treatments, while effective, are often limited in the face of rapidly evolving pathogens. Immunotherapy, which harnesses and enhances the body's immune response, offers a promising alternative to conventional approaches for the treatment of infectious diseases. By employing use of monoclonal antibodies, vaccines, cytokine therapies, and immune checkpoint inhibitors, immunotherapy has demonstrated considerable potential in overcoming treatment resistance and improving patient outcomes. Key innovations, including the development of mRNA vaccines, use of immune modulators, adoptive cell transfer, and chimeric antigen receptor (CAR)-T cell therapy are paving the way for more targeted pathogen clearance. Further, combining immunotherapy with conventional antibiotic treatment has demonstrated effectiveness against drug-resistant strains, but this chapter explores the evolving field of immunotherapy for the treatment of bacterial, viral, fungal, and parasitic infections. The chapter also explores the recent breakthroughs and ongoing clinical trials in infectious disease immunotherapy.
Collapse
Affiliation(s)
- Praveen Rai
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
9
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024; 53:11303-11320. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 10/23/2024]
Abstract
Antibacterial resistance is a severe threat to modern medicine and human health. To stay ahead of constantly-evolving bacteria we need to expand our arsenal of effective antibiotics. As such, antisense therapy is an attractive approach. The programmability allows to in principle target any RNA sequence within bacteria, enabling tremendous selectivity. In this Tutorial Review we provide guidelines for devising effective antibacterial antisense agents and offer a concise perspective for future research. We will review the chemical architectures of antibacterial antisense agents with a special focus on the delivery and target selection for successful antisense design. This Tutorial Review will strive to serve as an essential guide for antibacterial antisense technology development.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Alexander Lindberg
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Chamani M, Asgari S, Najmeddin A, Pourjavadi A, Amin M, Gholami M, Dorkoosh FA. Antibacterial activity of a silver-incorporated vancomycin-modified mesoporous silica against methicillin-resistant Staphylococcus aureus. J Biomater Appl 2024; 39:439-454. [PMID: 39193668 DOI: 10.1177/08853282241274517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Since conventional antibiotics are almost ineffective on methicillin-resistant Staphylococcus aureus (MRSA) strains, designing their antibacterial alternatives is necessary. Besides, the use of vancomycin is applied for specific detection of the bacteria. Silver-incorporated vancomycin-modified mesoporous silica nanoparticles (MSNs@Van@Ag NPs) were designed for detection and treatment of MRSA bacteria. Mesoporous silica nanoparticles (MSNs) were synthesized through the template method, modified with vancomycin, and finally incorporated with silver nanoparticles (Ag NPs). The MSNs@Van@Ag NPs with a homogenously spherical shape, average size of 50-100 nm, surface area of 955.8 m2/g, and thermal stability up to 200°C were successfully characterized. The amount of Ag incorporated into the MSNs@Van@Ag was calculated at 3.9 ppm and the release amount of Ag was received at 2.92 ppm (75%) after 100 h. The in vitro antibacterial susceptibility test showed the MIC of 100 μg mL-1 for MSNs@Van and 50 μg mL-1 for MSNs@Van@Ag, showing in vitro enhanced effect of Ag and vancomycin in the bactericidal process. An in vivo acute pneumonia model was performed and biochemical assays and pathological studies confirmed the nanomedicine's short-term safety for in vivo application. Cytokine assay using ELISA showed that MSN@Van@Ag causes a reduction of pro-inflammatory cytokines and bacterial proliferation leading to alleviation of inflammatory response.
Collapse
Affiliation(s)
- Mehdi Chamani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadi Asgari
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Ali Najmeddin
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Pourjavadi
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Jagdale P, Verma A, Shah DK. Pulmonary Pharmacokinetics of Antibody and Antibody Fragments Following Systemic and Local Administration in Mice. Pharmaceutics 2024; 16:1259. [PMID: 39458591 PMCID: PMC11510323 DOI: 10.3390/pharmaceutics16101259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/28/2024] Open
Abstract
Objective: This study aimed to investigate the effect of molecular size on the pulmonary pharmacokinetics (PK) of proteins following systemic and local administration in wild-type mice. Methods: A non-cross-reactive antibody trastuzumab, and F(ab')2, Fab, and scFv fragments of this antibody were used for the investigation. Proteins were injected intravenously or via intratracheal instillation, and PK was measured in plasma, lungs, trachea, bronchi, and bronchoalveolar lavage (BAL) using ELISA. Concentrations in BAL were urea normalized. Results: Following systemic administration, the biodistribution coefficient (BC) for lungs, trachea, bronchi, and BAL was 11%, 11%, 15%, and 2% for the antibody; 15%, 7%, 13%, and 8% for F(ab')2; 25%, 17%, 28%, and 46% for Fab; and 14%, 1%, 2%, and 50% for scFv. The antibody exposure in BAL was ~50-fold lower than plasma and ~5-7-fold lower than lung tissues. A tissue-dependent BC vs. molecular size relationship was observed, where distribution in tissues was the highest for Fab (50 kDa), and scFv demonstrated the highest distribution in the BAL. PK data generated following local administration were quite variable; however, local dosing resulted in BAL exposures that were 10-100-fold higher than those achieved after systemic dosing for all proteins. The BAL antibody concentrations were 100-1000-fold higher than plasma concentrations initially, which normalized by day 14. For most proteins, local dosing resulted in higher lung concentrations than trachea and bronchi, opposite to what was observed after systemic dosing. Conclusions: The PK data presented here provide an unprecedented quantitative insight into the effect of molecular size on the pulmonary disposition of proteins following systemic and local administration.
Collapse
Affiliation(s)
| | | | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14214, USA; (P.J.); (A.V.)
| |
Collapse
|
12
|
Sun S, Chen X. Mechanism-guided strategies for combating antibiotic resistance. World J Microbiol Biotechnol 2024; 40:295. [PMID: 39122871 DOI: 10.1007/s11274-024-04106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Bacterial antibiotic resistance has been recognized as a global threat to public health. It challenges the antibiotics currently used in clinical practice and causes severe and often fatal infectious diseases. Fighting against antibiotic-resistant bacteria (ARB) is growing more urgent. While understanding the molecular mechanisms that underlie resistance is a prerequisite, several major mechanisms have been previously proposed including bacterial efflux systems, reduced cell membrane permeability, antibiotic inactivation by enzymes, target modification, and target protection. In this context, this review presents a panel of promising and potential strategies to combat antibiotic resistance/resistant bacteria. Different types of direct-acting and indirect resistance breakers, such as efflux pump inhibitors, antibiotic adjuvants, and oxidative treatments are discussed. In addition, the emerging multi-omics approaches for rapid resistance identification and promising alternatives to existing antibiotics are highlighted. Overall, this review suggests that continued effort and investment in research are required to develop new antibiotics and alternatives to existing antibiotics and translate them into environmental and clinical applications.
Collapse
Affiliation(s)
- Shengwei Sun
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Xueyingzi Chen
- Key Laboratory of Food Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| |
Collapse
|
13
|
MacNair CR, Rutherford ST, Tan MW. Alternative therapeutic strategies to treat antibiotic-resistant pathogens. Nat Rev Microbiol 2024; 22:262-275. [PMID: 38082064 DOI: 10.1038/s41579-023-00993-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 04/19/2024]
Abstract
Resistance threatens to render antibiotics - which are essential for modern medicine - ineffective, thus posing a threat to human health. The discovery of novel classes of antibiotics able to overcome resistance has been stalled for decades, with the developmental pipeline relying almost entirely on variations of existing chemical scaffolds. Unfortunately, this approach has been unable to keep pace with resistance evolution, necessitating new therapeutic strategies. In this Review, we highlight recent efforts to discover non-traditional antimicrobials, specifically describing the advantages and limitations of antimicrobial peptides and macrocycles, antibodies, bacteriophages and antisense oligonucleotides. These approaches have the potential to stem the tide of resistance by expanding the physicochemical property space and target spectrum occupied by currently approved antibiotics.
Collapse
Affiliation(s)
- Craig R MacNair
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
14
|
Chen HC, Pan YL, Chen Y, Yang TH, Hsu ET, Huang YT, Chiang MH. Monoclonal Antibodies as a Therapeutic Strategy against Multidrug-Resistant Bacterial Infections in a Post-COVID-19 Era. Life (Basel) 2024; 14:246. [PMID: 38398755 PMCID: PMC10890110 DOI: 10.3390/life14020246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The development of severe multidrug-resistant bacterial infections has recently intensified because of the COVID-19 pandemic. According to the guidelines issued by the World Health Organization (WHO), routine antibiotic administration is not recommended for patients with supposed or confirmed mild SARS-CoV-2 infection or pneumonia, unless bacterial infection is clinically suspected. However, recent studies have pointed out that the proportion of non-essential antibiotic use in patients infected with SARS-CoV-2 remains high. Therefore, the silent pandemic of antibiotic resistance remains a pressing issue regardless of the present threats presented by the COVID-19 pandemic. To prevent or delay entry into the postulated post-antibiotic era, the long-term advocacy for the rational use of antibiotics, the optimization of infection control procedures, and the development of new antibacterial agents and vaccines should be underscored as vital practices of the antibacterial toolbox. Recently, the development of vaccines and monoclonal antibodies has gradually received attention following the advancement of biotechnology as well as enhanced drug discovery and development in cancer research. Although decent progress has been made in laboratory-based research and promising results have been obtained following clinical trials of some of these products, challenges still exist in their widespread clinical applications. This article describes the current advantages of antibacterial monoclonal antibodies, the development of associated clinical trials, and some perceived future perspectives and challenges. Further, we anticipate the development of more therapeutic agents to combat drug-resistant bacterial infections as well as to increase the resilience of current or novel agents/strategies.
Collapse
Affiliation(s)
- Hsiao-Chun Chen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei City 11490, Taiwan;
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
| | - Ying Chen
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
| | - Tsung-Hsuan Yang
- School of Nursing, National Defense Medical Center, Taipei City 11490, Taiwan;
| | - Erh-Tung Hsu
- School of Medicine, National Defense Medical Center, Taipei City 11490, Taiwan; (E.-T.H.); (Y.-T.H.)
| | - Yu-Ting Huang
- School of Medicine, National Defense Medical Center, Taipei City 11490, Taiwan; (E.-T.H.); (Y.-T.H.)
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
- mProbe Taiwan Inc., Taipei City 105037, Taiwan
| |
Collapse
|
15
|
Pei S, Lai L, Sun W, Lu Z, Hao J, Liu Y, Wu W, Guan S, Su X. Discovery of novel tetrahydrobenzothiophene derivatives as MSBA inhibitors for antimicrobial agents. Bioorg Chem 2024; 142:106932. [PMID: 37913586 DOI: 10.1016/j.bioorg.2023.106932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
The incidence of infections caused by drug-resistant bacteria has been one of the most serious health threats in the past and is substantially increasing in an alarming rate. Therefore, the development of new antimicrobial agents to combat bacterial resistance effectively is urgent. This study focused on the design and synthesis of 40 novel tetrahydrobenzothiophene amide/sulfonamide derivatives and their antibacterial activities were evaluated. Compounds 2p, 6p, and 6 s exhibited significant inhibitory effects on the growth of bacteria. To assess their safety, the cytotoxicity of the compounds was assessed using human normal liver cells, revealing that compound 6p has lower cytotoxicity. A mouse wound healing experiment demonstrated that compound 6p effectively improved wound infection induced by trauma and accelerated the healing process. Compound 6p holds promise as a potential therapeutic agent for combating bacterial infections.
Collapse
Affiliation(s)
- Shuchen Pei
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China.
| | - Lin Lai
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Wanlin Sun
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Zhaoyang Lu
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Jielei Hao
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| | - Yuheng Liu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, PR China; Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Wen Wu
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 404120, PR China.
| | - Shan Guan
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, PR China.
| | - Xiaoyan Su
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, PR China
| |
Collapse
|
16
|
Zhu X, Gong L, Qin Q. Development, methodological evaluation and application of a cell-based TRF assay for analysis of ADCC activity. J Pharm Biomed Anal 2023; 235:115655. [PMID: 37647793 DOI: 10.1016/j.jpba.2023.115655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
Interaction of an antibody with its FcγR plays an important role in effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC). Nowadays altered ADCC activity of an antibody can be achieved by utilizing an effective glyco-engineering strategy, which often involves changes of sugar moieties in Fc part of the antibody, thereby affecting its receptor binding with effector cells. We aimed to construct a cell-based time-resolved fluorescence (TRF) assay for the evaluation of ADCC activity triggered by the antibody drug Trastuzumab (anti-HER2) and T-DM1. The assay was initiated by incubating 2,2':6',2 "-Terpyridine-6,6"-dicarboxylic acid (TDA)-labeled target SK-BR3 cells with the testing antibodies and engineered NK-92 effector cells. After incubation, the target cells were lysed to detect TDA released into the supernatant. Together with added Eu, the TDA in the supernatant formed a stable chelate of EuTDA with high-intensity fluorescence. The ADCC activity was then determined by measuring the fluorescence of EuTDA. Consequently, the method demonstrated good accuracy, precision, linearity, and specificity over methodological assessment and compared well with the Luciferase release assay in terms of the agreement of the achieved results. Using the developed assay, we evaluated the ADCC activity of two glyco-engineered anti-HER-2 antibody-drug conjugates (ADCs) and the results showed that antibody Fc glycosylation modifications influenced antibody ADCC activity to varying degrees. In conclusion, the present assay is able to accurately assess the ADCC activity induced by Trastuzumab (anti-HER2) and T-DM1, and a similar methodology can be applied to other therapeutic antibodies during drug development to help screen for antibodies with desirable ADCC activity.
Collapse
Affiliation(s)
- Xiao Zhu
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai 201203, China; Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210023, China
| | - Likun Gong
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai 201203, China; Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210023, China.
| | - Qiuping Qin
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai 201203, China.
| |
Collapse
|
17
|
Ferguson MR, Delgado KN, McBride S, Orbe IC, La Vake CJ, Caimano MJ, Mendez Q, Moraes TF, Schryvers AB, Moody MA, Radolf JD, Weiner MP, Hawley KL. Use of Epivolve phage display to generate a monoclonal antibody with opsonic activity directed against a subdominant epitope on extracellular loop 4 of Treponema pallidum BamA (TP0326). Front Immunol 2023; 14:1222267. [PMID: 37675118 PMCID: PMC10478084 DOI: 10.3389/fimmu.2023.1222267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Syphilis, a sexually transmitted infection caused by the spirochete Treponema pallidum (Tp), is resurging globally. Tp's repertoire of outer membrane proteins (OMPs) includes BamA (β-barrel assembly machinery subunit A/TP0326), a bipartite protein consisting of a 16-stranded β-barrel with nine extracellular loops (ECLs) and five periplasmic POTRA (polypeptide transport-associated) domains. BamA ECL4 antisera promotes internalization of Tp by rabbit peritoneal macrophages. Methods Three overlapping BamA ECL4 peptides and a two-stage, phage display strategy, termed "Epivolve" (for epitope evolution) were employed to generate single-chain variable fragments (scFvs). Additionally, antisera generated by immunizing mice and rabbits with BamA ECL4 displayed by a Pyrococcus furiosus thioredoxin scaffold (PfTrxBamA/ECL4). MAbs and antisera reactivities were evaluated by immunoblotting and ELISA. A comparison of murine and rabbit opsonophagocytosis assays was conducted to evaluate the functional ability of the Abs (e.g., opsonization) and validate the mouse assay. Sera from Tp-infected mice (MSS) and rabbits (IRS) were evaluated for ECL4-specific Abs using PfTrxBamA/ECL4 and overlapping ECL4 peptides in immunoblotting and ELISA assays. Results Each of the five mAbs demonstrated reactivity by immunoblotting and ELISA to nanogram amounts of PfTrxBamA/ECL4. One mAb, containing a unique amino acid sequence in both the light and heavy chains, showed activity in the murine opsonophagocytosis assay. Mice and rabbits hyperimmunized with PfTrxBamA/ECL4 produced opsonic antisera that strongly recognized the ECL presented in a heterologous scaffold and overlapping ECL4 peptides, including S2. In contrast, Abs generated during Tp infection of mice and rabbits poorly recognized the peptides, indicating that S2 contains a subdominant epitope. Discussion Epivolve produced mAbs target subdominant opsonic epitopes in BamA ECL4, a top syphilis vaccine candidate. The murine opsonophagocytosis assay can serve as an alternative model to investigate the opsonic potential of vaccinogens. Detailed characterization of BamA ECL4-specific Abs provided a means to dissect Ab responses elicited by Tp infection.
Collapse
Affiliation(s)
- Mary R. Ferguson
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | | | | | - Isabel C. Orbe
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Carson J. La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Melissa J. Caimano
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
| | - Qiana Mendez
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Trevor F. Moraes
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Anthony B. Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, United States
| | - Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Michael P. Weiner
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Kelly L. Hawley
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Division of Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT, United States
| |
Collapse
|
18
|
Esposito S, Amirthalingam G, Bassetti M, Blasi F, De Rosa FG, Halasa NB, Hung I, Osterhaus A, Tan T, Torres JP, Vena A, Principi N. Monoclonal antibodies for prophylaxis and therapy of respiratory syncytial virus, SARS-CoV-2, human immunodeficiency virus, rabies and bacterial infections: an update from the World Association of Infectious Diseases and Immunological Disorders and the Italian Society of Antinfective Therapy. Front Immunol 2023; 14:1162342. [PMID: 37256125 PMCID: PMC10226646 DOI: 10.3389/fimmu.2023.1162342] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
Monoclonal antibodies (mABs) are safe and effective proteins produced in laboratory that may be used to target a single epitope of a highly conserved protein of a virus or a bacterial pathogen. For this purpose, the epitope is selected among those that play the major role as targets for prevention of infection or tissue damage. In this paper, characteristics of the most important mABs that have been licensed and used or are in advanced stages of development for use in prophylaxis and therapy of infectious diseases are discussed. We showed that a great number of mABs effective against virus or bacterial infections have been developed, although only in a small number of cases these are licensed for use in clinical practice and have reached the market. Although some examples of therapeutic efficacy have been shown, not unlike more traditional antiviral or antibacterial treatments, their efficacy is significantly greater in prophylaxis or early post-exposure treatment. Although in many cases the use of vaccines is more effective and cost-effective than that of mABs, for many infectious diseases no vaccines have yet been developed and licensed. Furthermore, in emergency situations, like in epidemics or pandemics, the availability of mABs can be an attractive adjunct to our armament to reduce the impact. Finally, the availability of mABs against bacteria can be an important alternative, when multidrug-resistant strains are involved.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gayatri Amirthalingam
- Immunisation and Countermeasures Division, National Infection Service, Public Health England, London, United Kingdom
| | - Matteo Bassetti
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | | | - Natasha B. Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ivan Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Tina Tan
- Division of Infectious Diseases, Feinberg School of Medicine of Northwestern University, Chicago, IL, United States
| | - Juan Pablo Torres
- Department of Pediatrics and Pediatric Surgery, Facultad de Medicina, University of Chile, Santiago, Chile
- Instituto Sistemas Complejos de Ingeniería (ISCI), Santiago, Chile
| | - Antonio Vena
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
19
|
van Strijp JAG. MABTRAINS: Numerous anti-infective modalities ride together. Cell Host Microbe 2023; 31:687-688. [PMID: 37167949 DOI: 10.1016/j.chom.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In this issue of Cell Host & Microbe, Buckley et al. report a biological entity called a mAbtyrin, which combines various antimicrobial functions. The authors demonstrate through in vitro and in vivo experiments that this approach can lead to highly effective antimicrobial action against Staphylococcus aureus.
Collapse
Affiliation(s)
- Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
20
|
Roy S, Roy J, Guo B. Nanomaterials as multimodal photothermal agents (PTAs) against 'Superbugs'. J Mater Chem B 2023; 11:2287-2306. [PMID: 36857688 DOI: 10.1039/d2tb02396b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Superbugs, also known as multidrug-resistant bacteria, have become a lethal and persistent threat due to their unresponsiveness toward conventional antibiotics. The main reason for this is that superbugs can rapidly mutate and restrict any foreign drug/molecule in their vicinity. Herein, nanomaterial-mediated therapies have set their path and shown burgeoning efficiency toward the ablation of superbugs. Notably, treatment modalities like photothermal therapy (PTT) have shown prominence in killing multidrug-resistant bacteria with their ability to generate local heat shock-mediated hyperthermia in such species. However, photothermal treatment has some serious limitations, such as high cost, complexity, and even toxicity to some extent. Hence, it is important to resolve such shortcomings of PTTs as they provide substantial tissue penetration. This is why multimodal PTTs have emerged and taken over this domain of research for the past few years. In this work, we have summarized and critically reviewed such exceptional works of recent times and provided a perspective to enhance their efficiencies. Profoundly, we discuss the design rationales of some novel photothermal agents (PTAs) and shed light on their mechanisms. Finally, challenges for PTT-derived multimodal therapy are presented, and capable synergistic bactericidal prospects are anticipated.
Collapse
Affiliation(s)
- Shubham Roy
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Jhilik Roy
- Department of Physics, Jadavpur University, Kolkata 700032, India
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
| |
Collapse
|
21
|
Yu L, Shang Z, Jin Q, Chan SY, Hong W, Li N, Li P. Antibody-Antimicrobial Conjugates for Combating Antibiotic Resistance. Adv Healthc Mater 2023; 12:e2202207. [PMID: 36300640 DOI: 10.1002/adhm.202202207] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Indexed: 02/03/2023]
Abstract
As the development of new antibiotics lags far behind the emergence of drug-resistant bacteria, alternative strategies to resolve this dilemma are urgently required. Antibody-drug conjugate is a promising therapeutic platform to delivering cytotoxic payloads precisely to target cells for efficient disease treatment. Antibody-antimicrobial conjugates (AACs) have recently attracted considerable interest from researchers as they can target bacteria in the target sites and improve the effectiveness of drugs (i.e., reduced drug dosage and adverse effects), abating the upsurge of antimicrobial resistance. In this review, the selection and progress of three essential blocks that compose the AACs: antibodies, antimicrobial payloads, and linkers are discussed. The commonly used conjugation strategies and the latest applications of AACs in recent years are also summarized. The challenges and opportunities of this booming technology are also discussed at the end of this review.
Collapse
Affiliation(s)
- Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Zifang Shang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518026, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology Chinese Academy of Sciences, Beijing, 100101, China
| | - Qizhe Jin
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Weilin Hong
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Nan Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
22
|
Pharmaceuticals and Personal Care Products in the Environment with Emphasis on Horizontal Transfer of Antibiotic Resistance Genes. CHEMISTRY-DIDACTICS-ECOLOGY-METROLOGY 2022. [DOI: 10.2478/cdem-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Abstract
Pharmaceuticals and personal care products (PPCPs) discharged into environment has several adverse impacts. PPCPs are widely utilised for veterinary as well as cosmetic and personal health reasons. These are members of the expanding class of substances known as Contaminants of Emerging Concern (CECs). Antibiotic resistance in the environment and garbage generated by PPCP endanger life. The World Health Organisation (WHO) now recognises antibiotic resistance as a significant global health problem due to the expected increase in mortality caused by it. In the past ten years, mounting data has led experts to believe that the environment has a significant impact on the development of resistance. For human diseases, the external environment serves as a source of resistance genes. It also serves as a major pathway for the spread of resistant bacteria among various habitats and human populations. Large-scale DNA sequencing methods are employed in this thesis to better comprehend the dangers posed by environmental antibiotic resistance. The quantification of the number is an important step in this process. Metagenomic measurement of the number of antibiotic resistance genes in various contexts is a crucial step in this process. However, it’s also crucial to put this data into a broader context by integrating things like taxonomic information, antibiotic concentrations, and the genomic locations of found resistance genes.
Collapse
|
23
|
Ruest MK, Dennis JJ. The Exploration of Complement-Resistance Mechanisms of Pathogenic Gram-Negative Bacteria to Support the Development of Novel Therapeutics. Pathogens 2022; 11:931. [PMID: 36015050 PMCID: PMC9412335 DOI: 10.3390/pathogens11080931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Resistance to antibiotics in Bacteria is one of the biggest threats to human health. After decades of attempting to isolate or design antibiotics with novel mechanisms of action against bacterial pathogens, few approaches have been successful. Antibacterial drug discovery is now moving towards targeting bacterial virulence factors, especially immune evasion factors. Gram-negative bacteria present some of the most significant challenges in terms of antibiotic resistance. However, they are also able to be eliminated by the component of the innate immune system known as the complement system. In response, Gram-negative bacteria have evolved a variety of mechanisms by which they are able to evade complement and cause infection. Complement resistance mechanisms present some of the best novel therapeutic targets for defending against highly antibiotic-resistant pathogenic bacterial infections.
Collapse
Affiliation(s)
| | - Jonathan J. Dennis
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|