1
|
Yanestria SM, Effendi MH, Tyasningsih W, Mariyono M, Ugbo EN. First report of phenotypic and genotypic (bla OXA-61) beta-lactam resistance in Campylobacter jejuni from broilers in Indonesia. Vet World 2023; 16:2210-2216. [PMID: 38152271 PMCID: PMC10750733 DOI: 10.14202/vetworld.2023.2210-2216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/03/2023] [Indexed: 12/29/2023] Open
Abstract
Background and Aim Campylobacter is a zoonotic bacterium that is a major source of foodborne diseases. In humans, most cases of campylobacteriosis are caused by Campylobacter jejuni. Poultry is the main reservoir of Campylobacter for humans, because Campylobacter is part of the normal flora of the digestive tract of poultry. Antimicrobial resistance to several antibiotics in Campylobacter isolated from humans and food animals has increased rapidly. Beta-lactam is an antibiotic with a high prevalence of resistance in Campylobacter. This study aimed to investigate phenotypic and genotypic (blaOXA-61) beta-lactam resistance in C. jejuni from broilers in Indonesia. Materials and Methods A total of 100 samples of broiler intestinal contents were obtained from 10 broiler farms in Pasuruan Regency, Indonesia. Campylobacter jejuni was identified using conventional and polymerase chain reaction (PCR)-based methods. Phenotypic detection of beta-lactam resistance was performed using an antimicrobial susceptibility test with antibiotic disks of aztreonam, ampicillin, and amoxicillin-clavulanic acid. Genotypic detection by PCR was performed using the blaOXA-61 gene, which encodes beta-lactamase. Results Campylobacter jejuni was identified in 23% of the samples. Phenotypically, 100% (23/23) and 73.9% (17/23) C. jejuni isolates had high resistance to aztreonam and ampicillin, respectively, but all isolates were susceptible to amoxicillin-clavulanic acid. Genotypically, all isolates carried blaOXA-61, indicated by the presence of a 372-bp PCR product. Conclusion Campylobacter jejuni is highly resistant to beta-lactams and is a serious threat to human health. Resistance to beta-lactams should be monitored because beta-lactamase genes can be transferred between bacteria. Public awareness must also be increased on the importance of using antibiotics rationally in humans and animals.
Collapse
Affiliation(s)
- Sheila Marty Yanestria
- Doctoral Program in Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga. Jl. Dr. Ir. H. Soekarno, Kampus C Mulyorejo, Surabaya 60115, East Java, Indonesia
| | - Mustofa Helmi Effendi
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Jl. Dr. Ir. H. Soekarno, Kampus C Mulyorejo, Surabaya 60115, East Java, Indonesia
| | - Wiwiek Tyasningsih
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Jl. Dr. Ir. H. Soekarno, Kampus C Mulyorejo, Surabaya 60115, East Java, Indonesia
| | - Mariyono Mariyono
- Bacteriology Laboratory, Balai Besar Veteriner Wates, Jl. Yogyakarta-Wates No. Km. 27, Wates, Yogyakarta 55651, Central Java, Indonesia
| | - Emmanuel Nnabuike Ugbo
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Enugu - Abakaliki Rd, 481101, Abakaliki, Ebonyi, Nigeria
| |
Collapse
|
2
|
Qian C, Wu Q, Ruan Z, Liu F, Li W, Shi W, Ma L, Peng D, Yin H, Yao L, Li Z, Hong M, Xia L. A Visualized Mortality Prediction Score Model in Hematological Malignancies Patients with Carbapenem-Resistant Organisms Bloodstream Infection. Infect Drug Resist 2023; 16:201-215. [PMID: 36644657 PMCID: PMC9833326 DOI: 10.2147/idr.s393932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Background Bloodstream infection (BSI) due to carbapenem-resistant organisms (CROs) has emerged as a worldwide problem associated with high mortality. This study aimed to evaluate the risk factors associated with mortality in HM patients with CROs BSI and to establish a scoring model for early mortality prediction. Methods We conducted a retrospective cohort study at our hematological department from January 2018 to December 2021, including all HM patients with CROs BSI. The outcome measured was death within 30-day of BSI onset. Survivor and non-survivor subgroups were compared to identify predictors of mortality. Univariate and multivariate Cox regression analyses were used to identify prognostic risk factors and develop a nomogram. Results In total, 150 HM patients were included in the study showing an overall 30-day mortality rate of 56%. Klebsiella pneumonia was the dominant episode. Cox regression analysis showed that pre-infection length of stay was >14 days (score 41), Pitt score >4 (score 100), mucositis (score 41), CAR (The ratio of C-reactive protein to albumin) >8.8 (score 57), early definitive therapy (score 44), and long-duration (score 78) were positive independent risk predictors associated with 30-day mortality, all of which were selected into the nomogram. Furthermore, all patients were divided into the high-risk group (≥160 points) or the low-risk group based on the prediction score model. The mortality of the high-risk group was 8 times more than the low-risk group. Kaplan-Meier analysis showed that empirical polymyxin B therapy was associated with a lower 30-day mortality rate, which was identified as a good prognostic factor in the high-risk group. In comparison, empirical carbapenems and tigecycline were poor prognostic factors in a low-risk group. Conclusion Our score model can accurately predict 30-day mortality in HM patients with CROs BSI. Early administration of CROs-targeted therapy in the high-risk group is strongly recommended to decrease mortality.
Collapse
Affiliation(s)
- Chenjing Qian
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Qiuling Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Zhixuan Ruan
- Faculty of Natural, Mathematical and Engineering Sciences, King’s College, London, UK
| | - Fang Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Weiming Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Danyue Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Hua Yin
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Lan Yao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Zixuan Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| | - Mei Hong
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China,Correspondence: Mei Hong; Linghui Xia, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No. 1277, Wuhan, Hubei Province, People’s Republic of China, Tel +8613037137937; +8618627733999, Email ;
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China
| |
Collapse
|
3
|
Empirical antibiotic therapy for difficult-to-treat Gram-negative infections: when, how, and how long? Curr Opin Infect Dis 2022; 35:568-574. [PMID: 36206149 DOI: 10.1097/qco.0000000000000884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW To discuss empirical therapy for severe infections due to Gram-negative bacteria with difficult-to-treat resistance (GNB-DTR) in current clinical practice, focusing in particular on the positioning of novel therapeutic agents and rapid diagnostic tests. RECENT FINDINGS The current era of novel agents active against GNB-DTR and showing differential activity against specific determinants of resistance is an unprecedented scenario, in which the clinical reasoning leading to the choice of the empirical therapy for treating severe GNB-DTR infections is becoming more complex, but it also allows for enhanced treatment precision. SUMMARY Novel agents should be used in line with antimicrobial stewardship principles, aimed at reducing selective pressure for antimicrobial resistance. However, this does not mean that they should not be used. Indeed, excesses in restrictive uses may be unethical by precluding access to the most effective and less toxic treatments for patients with severe GNB-DTR infections. Given these premises (the 'how'), empirical treatment with novel agents should be considered in all patients with risk factors for GNB-DTR and severe clinical presentation of acute infection (the 'when'). Furthermore, empirical novel agents should preferably be continued only for a few hours, until de-escalation, modification, or confirmation (as targeted therapy) is made possible by the results of rapid diagnostic tests (the 'how long').
Collapse
|
4
|
Lynch JP, Clark NM, Zhanel GG. Infections Due to Acinetobacter baumannii-calcoaceticus Complex: Escalation of Antimicrobial Resistance and Evolving Treatment Options. Semin Respir Crit Care Med 2022; 43:97-124. [PMID: 35172361 DOI: 10.1055/s-0041-1741019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Bacteria within the genus Acinetobacter (principally A. baumannii-calcoaceticus complex [ABC]) are gram-negative coccobacilli that most often cause infections in nosocomial settings. Community-acquired infections are rare, but may occur in patients with comorbidities, advanced age, diabetes mellitus, chronic lung or renal disease, malignancy, or impaired immunity. Most common sites of infections include blood stream, skin/soft-tissue/surgical wounds, ventilator-associated pneumonia, orthopaedic or neurosurgical procedures, and urinary tract. Acinetobacter species are intrinsically resistant to multiple antimicrobials, and have a remarkable ability to acquire new resistance determinants via plasmids, transposons, integrons, and resistance islands. Since the 1990s, antimicrobial resistance (AMR) has escalated dramatically among ABC. Global spread of multidrug-resistant (MDR)-ABC strains reflects dissemination of a few clones between hospitals, geographic regions, and continents; excessive antibiotic use amplifies this spread. Many isolates are resistant to all antimicrobials except colistimethate sodium and tetracyclines (minocycline or tigecycline); some infections are untreatable with existing antimicrobial agents. AMR poses a serious threat to effectively treat or prevent ABC infections. Strategies to curtail environmental colonization with MDR-ABC require aggressive infection-control efforts and cohorting of infected patients. Thoughtful antibiotic strategies are essential to limit the spread of MDR-ABC. Optimal therapy will likely require combination antimicrobial therapy with existing antibiotics as well as development of novel antibiotic classes.
Collapse
Affiliation(s)
- Joseph P Lynch
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology; Department of Medicine; The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Nina M Clark
- Division of Infectious Diseases, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
5
|
Swietnicki W. Secretory System Components as Potential Prophylactic Targets for Bacterial Pathogens. Biomolecules 2021; 11:892. [PMID: 34203937 PMCID: PMC8232601 DOI: 10.3390/biom11060892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Bacterial secretory systems are essential for virulence in human pathogens. The systems have become a target of alternative antibacterial strategies based on small molecules and antibodies. Strategies to use components of the systems to design prophylactics have been less publicized despite vaccines being the preferred solution to dealing with bacterial infections. In the current review, strategies to design vaccines against selected pathogens are presented and connected to the biology of the system. The examples are given for Y. pestis, S. enterica, B. anthracis, S. flexneri, and other human pathogens, and discussed in terms of effectiveness and long-term protection.
Collapse
Affiliation(s)
- Wieslaw Swietnicki
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. R. Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
6
|
Stewart AG, Harris PNA, Chatfield MD, Littleford R, Paterson DL. Ceftolozane-tazobactam versus meropenem for definitive treatment of bloodstream infection due to extended-spectrum beta-lactamase (ESBL) and AmpC-producing Enterobacterales ("MERINO-3"): study protocol for a multicentre, open-label randomised non-inferiority trial. Trials 2021; 22:301. [PMID: 33888139 PMCID: PMC8060904 DOI: 10.1186/s13063-021-05206-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/18/2021] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Extended-spectrum beta-lactamase (ESBL) and AmpC-producing Enterobacterales are common causes of bloodstream infection. ESBL-producing bacteria are typically resistant to third-generation cephalosporins and result in a sizeable economic and public health burden. AmpC-producing Enterobacterales may develop third-generation cephalosporin resistance through enzyme hyper-expression. In no observational study has the outcome of treatment of these infections been surpassed by carbapenems. Widespread use of carbapenems may drive the development of carbapenem-resistant Gram-negative bacilli. METHODS This study will use a multicentre, parallel group open-label non-inferiority trial design comparing ceftolozane-tazobactam and meropenem in adult patients with bloodstream infection caused by ESBL or AmpC-producing Enterobacterales. Trial recruitment will occur in up to 40 sites in six countries (Australia, Singapore, Italy, Spain, Saudi Arabia and Lebanon). The sample size is determined by a predefined quantity of ceftolozane-tazobactam to be supplied by Merck, Sharpe and Dohme (MSD). We anticipate that a trial with 600 patients contributing to the primary outcome analysis would have 80% power to declare non-inferiority with a 5% non-inferiority margin, assuming a 30-day mortality of 5% in both randomised groups. Once randomised, definitive treatment will be for a minimum of 5 days and a maximum of 14 days with the total duration determined by treating clinicians. Data describing demographic information, risk factors, concomitant antibiotics, illness scores, microbiology, multidrug-resistant organism screening, discharge and mortality will be collected. DISCUSSION Participants will have bloodstream infection due to third-generation cephalosporin non-susceptible E. coli and Klebsiella spp. or Enterobacter spp., Citrobacter freundii, Morganella morganii, Providencia spp. or Serratia marcescens. They will be randomised 1:1 to ceftolozane-tazobactam 3 g versus meropenem 1 g, both every 8 h. Secondary outcomes will be a comparison of 14-day all-cause mortality, clinical and microbiological success at day 5, functional bacteraemia score, microbiological relapse, new bloodstream infection, length of hospital stay, serious adverse events, C. difficile infection, multidrug-resistant organism colonisation. The estimated trial completion date is December 2024. TRIAL REGISTRATION The MERINO-3 trial is registered under the US National Institute of Health ClinicalTrials.gov register, reference number: NCT04238390 . Registered on 23 January 2020.
Collapse
Affiliation(s)
- Adam G Stewart
- Centre for Clinical Research, Faculty of Medicine, University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Queensland, Australia.
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
| | - Patrick N A Harris
- Centre for Clinical Research, Faculty of Medicine, University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Queensland, Australia
- Department of Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Mark D Chatfield
- Centre for Clinical Research, Faculty of Medicine, University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Queensland, Australia
| | - Roberta Littleford
- Centre for Clinical Research, Faculty of Medicine, University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Queensland, Australia
| | - David L Paterson
- Centre for Clinical Research, Faculty of Medicine, University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Queensland, Australia
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
7
|
Ding Y, Li Z, Xu C, Qin W, Wu Q, Wang X, Cheng X, Li L, Huang W. Fluorogenic Probes/Inhibitors of β-Lactamase and their Applications in Drug-Resistant Bacteria. Angew Chem Int Ed Engl 2020; 60:24-40. [PMID: 32592283 DOI: 10.1002/anie.202006635] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Indexed: 01/08/2023]
Abstract
β-Lactam antibiotics are generally perceived as one of the greatest inventions of the 20th century, and these small molecular compounds have saved millions of lives. However, upon clinical application of antibiotics, the β-lactamase secreted by pathogenic bacteria can lead to the gradual development of drug resistance. β-Lactamase is a hydrolase that can efficiently hydrolyze and destroy β-lactam antibiotics. It develops and spreads rapidly in pathogens, and the drug-resistant bacteria pose a severe threat to human health and development. As a result, detecting and inhibiting the activities of β-lactamase are of great value for the rational use of antibiotics and the treatment of infectious diseases. At present, many specific detection methods and inhibitors of β-lactamase have been developed and applied in clinical practice. In this Minireview, we describe the resistance mechanism of bacteria producing β-lactamase and further summarize the fluorogenic probes, inhibitors of β-lactamase, and their applications in the treatment of infectious diseases. It may be valuable to design fluorogenic probes with improved selectivity, sensitivity, and effectiveness to further identify the inhibitors for β-lactamases and eventually overcome bacterial resistance.
Collapse
Affiliation(s)
- Yang Ding
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Zheng Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Chenchen Xu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Wenjing Qin
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Qiong Wu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Xuchun Wang
- College of Chemistry and Material Engineering, University of Science and Technology of Anhui, Bengbu, 233000, P. R. China
| | - Xiamin Cheng
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China.,Frontiers Science Center for Flexible Electronics (FSCFE), Shaanxi Institute of Flexible Electronics (SIFE) & Shaanxi Institute of Biomedical Materials and Engineering (SIBME), Northwestern Polytechnical University (NPU), Xi'an, 710072, P. R. China
| |
Collapse
|
8
|
Ding Y, Li Z, Xu C, Qin W, Wu Q, Wang X, Cheng X, Li L, Huang W. Fluorogenic Probes/Inhibitors of β‐Lactamase and their Applications in Drug‐Resistant Bacteria. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Yang Ding
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Zheng Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Chenchen Xu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Wenjing Qin
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Qiong Wu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Xuchun Wang
- College of Chemistry and Material Engineering University of Science and Technology of Anhui Bengbu 233000 P. R. China
| | - Xiamin Cheng
- Institute of Advanced Synthesis School of Chemistry and Molecular Engineering Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing 211816 P. R. China
- Frontiers Science Center for Flexible Electronics (FSCFE) Shaanxi Institute of Flexible Electronics (SIFE) & Shaanxi Institute of Biomedical Materials and Engineering (SIBME) Northwestern Polytechnical University (NPU) Xi'an 710072 P. R. China
| |
Collapse
|
9
|
Gangadharappa BS, Sharath R, Revanasiddappa PD, Chandramohan V, Balasubramaniam M, Vardhineni TP. Structural insights of metallo-beta-lactamase revealed an effective way of inhibition of enzyme by natural inhibitors. J Biomol Struct Dyn 2019; 38:3757-3771. [DOI: 10.1080/07391102.2019.1667265] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Bhavya Somalapura Gangadharappa
- Department of Biotechnology, M.S Ramaiah Institute of Technology, Bengaluru, Karnataka, India
- Visvesvaraya Technological University, Belagavi, Karnataka, India
| | | | | | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru, Karnataka, India
| | | | - Teja Priya Vardhineni
- Biotecthology Skill Enhancement Program, Siddaganga Institute of Technology, Tumakuru, Karnataka, India
| |
Collapse
|
10
|
Nguenang GS, Mbaveng AT, Fankam AG, Manekeng HT, Nayim P, Wamba BEN, Kuete V. Tristemma hirtum and Five Other Cameroonian Edible Plants with Weak or No Antibacterial Effects Modulate the Activities of Antibiotics against Gram-Negative Multidrug-Resistant Phenotypes. ScientificWorldJournal 2018; 2018:7651482. [PMID: 29765275 PMCID: PMC5885400 DOI: 10.1155/2018/7651482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 01/21/2023] Open
Abstract
In order to contribute to the fight against infectious diseases, the in vitro antibacterial activity and the antibiotic-potentiating effects of Tristemma hirtum and five other Cameroonian edible plants have been evaluated against Gram-negative multidrug-resistant (MDR) phenotypes. The microdilution method was used to evaluate the bacterial susceptibility of the extracts and their combination to common antibiotics. The phytochemical screening of the extracts was carried out according to standard methods. Phytochemical analysis of the extracts revealed the presence of alkaloids, triterpenes, steroids, and polyphenols, including flavonoids in most of the tested extracts. The entire tested extracts showed moderate (512 μg/mL ≤ MIC ≤ 2048 μg/mL) to weak (MIC > 2048 μg/mL) antibacterial activities against the tested bacteria. Furthermore, extracts of leaf of Tristemma hirtum and pericarps of Raphia hookeri (at their MIC/2 and MIC/4) strongly potentiated the activities of all antibiotics used in the study, especially those of chloramphenicol (CHL), ciprofloxacin (CIP), kanamycin (KAN), and tetracycline (TET) against 70% (7/10) to 100% (10/10) of the tested MDR bacteria, with the modulating factors ranging from 2 to 128. The results of this study suggest that extracts from leaves of Tristemma hirtum and pericarps of Raphia hookeri can be sources of plant-derived products with antibiotic modifying activity.
Collapse
Affiliation(s)
- Gaëlle S. Nguenang
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Armelle T. Mbaveng
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Aimé G. Fankam
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Hermione T. Manekeng
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Paul Nayim
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Brice E. N. Wamba
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Victor Kuete
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
11
|
Activity of Simulated Human Dosage Regimens of Meropenem and Vaborbactam against Carbapenem-Resistant Enterobacteriaceae in an In Vitro Hollow-Fiber Model. Antimicrob Agents Chemother 2018; 62:AAC.01969-17. [PMID: 29133570 DOI: 10.1128/aac.01969-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/30/2017] [Indexed: 01/07/2023] Open
Abstract
The objective of these studies was to evaluate the exposures of meropenem and vaborbactam that would produce antibacterial activity and prevent resistance development in carbapenem-resistant Klebsiella pneumoniae carbapenemase (KPC)-producing Enterobacteriaceae strains when tested at an inoculum of 108 CFU/ml. Thirteen K. pneumoniae isolates, three Enterobacter cloacae isolates, and one Escherichia coli isolate were examined in an in vitro hollow-fiber model over 32 h. Simulated dosage regimens of 1 to 2 g of meropenem with 1 to 2 g of vaborbactam, with meropenem administered every 8 h by a 3-h infusion based on phase 1 or phase 3 patient pharmacokinetic data, were studied in the model. A dosage of 2 g of meropenem in combination with 2 g of vaborbactam was bactericidal against K. pneumoniae, E. cloacae, and E. coli strains, with meropenem-vaborbactam MICs of up to 8 mg/liter. When the vaborbactam exposure was adjusted to the levels observed in patients enrolled in phase 3 trials (24-h free AUC, ∼550 mg · h/liter, versus 320 mg · h/liter in the phase 1 studies), 2 g of meropenem with 2 g of vaborbactam was also bactericidal against strains with meropenem-vaborbactam MICs of 16 mg/liter. In addition, this level of vaborbactam also suppressed the development of resistance observed using phase 1 exposures. In this pharmacodynamic model, exposures similar to 2 g of meropenem in combination with 2 g of vaborbactam administered every 8 h by a 3-h infusion in phase 3 trials produced antibacterial activity and suppressed the development of resistance against carbapenem-resistant KPC-producing strains of Enterobacteriaceae.
Collapse
|
12
|
Activity of Meropenem-Vaborbactam in Mouse Models of Infection Due to KPC-Producing Carbapenem-Resistant Enterobacteriaceae. Antimicrob Agents Chemother 2017; 62:AAC.01446-17. [PMID: 29109160 DOI: 10.1128/aac.01446-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/26/2017] [Indexed: 12/16/2022] Open
Abstract
Meropenem-vaborbactam (Vabomere) is highly active against Gram-negative pathogens, especially Klebsiella pneumoniae carbapenemase (KPC)-producing, carbapenem-resistant Enterobacteriaceae The objective of these studies was to evaluate the efficacy of meropenem alone and in combination with vaborbactam in mouse thigh and lung infection models. Thighs or lungs of neutropenic mice were infected with KPC-producing carbapenem-resistant Enterobacteriaceae, with meropenem MICs ranging from ≤0.06 to 8 mg/liter in the presence of 8 mg/liter vaborbactam. Mice were treated with meropenem alone or meropenem in combination with vaborbactam every 2 h for 24 h to provide exposures comparable to 2-g doses of each component in humans. Meropenem administered in combination with vaborbactam produced bacterial killing in all strains tested, while treatment with meropenem alone either produced less than 0.5 log CFU/tissue of bacterial killing or none at all. In the thigh model, 11 strains were treated with the combination of meropenem plus vaborbactam (300 plus 50 mg/kg of body weight). This combination produced from 0.8 to 2.89 logs of bacterial killing compared to untreated controls at the start of treatment. In the lung infection model, two strains were treated with the same dosage regimen of meropenem and vaborbactam. The combination produced more than 1.83 logs of bacterial killing against both strains tested compared to untreated controls at the start of treatment. Overall, these data suggest that meropenem-vaborbactam may have utility in the treatment of infections due to KPC-producing carbapenem-resistant Enterobacteriaceae.
Collapse
|
13
|
Affiliation(s)
- Dmitrii V. Kalinin
- Institut für Organische Chemie, Universität Hamburg, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Ralph Holl
- Institut für Organische Chemie, Universität Hamburg, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems
| |
Collapse
|
14
|
Moreira CS, Silva ACJA, Novais JS, Sá Figueiredo AM, Ferreira VF, da Rocha DR, Castro HC. Searching for a potential antibacterial lead structure against bacterial biofilms among new naphthoquinone compounds. J Appl Microbiol 2017; 122:651-662. [PMID: 27930849 DOI: 10.1111/jam.13369] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/13/2016] [Accepted: 11/28/2016] [Indexed: 01/10/2023]
Abstract
AIMS The aims of this study were to design, synthesize and to evaluate 2-hydroxy-3-phenylsulfanylmethyl-[1,4]-naphthoquinones against Gram-negative and Gram-positive bacterial strains, including methicillin-resistant Staphylococcus aureus (MRSA) and its biofilm, to probe for potential lead structures. METHODS AND RESULTS Thirty-six new analogues were prepared with good yields using a simple, fast, operational three-procedure reaction and a thiol addition to an ο-quinone methide using microwave irradiation. All compounds were tested against Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, Proteus mirabilis ATCC 15290, Serratia marcescens ATCC 14756, Klebsiella pneumoniae ATCC 4352, Enterobacter cloacae ATCC 23355, Enterococcus faecalis ATCC 29212, S. aureus ATCC 25923, Staphylococcus simulans ATCC 27851, Staphylococcus epidermidis ATCC 12228 and a hospital strain of MRSA. Their antibacterial activity was determined using the disc diffusion method, revealing the activity of 19 compounds, mainly against Gram-positive strains. Interestingly, the minimal inhibitory concentration ranges detected for the hit molecules (32-128 μg ml-1 ) were within Clinical and Laboratory Standards Institute levels. Promisingly, compound 15 affected the MRSA strain, with a reduction of up to 50% in biofilm formation, which is better than vancomycin as biofilm forms a barrier against the antibiotic that avoids its action. CONCLUSIONS After probing 36 naphthoquinones for a potential antibacterial lead structure against the bacterial biofilm, we found that compound 15 should be explored further and also should be structurally modified in the near future to test against Gram-negative strains. SIGNIFICANCE AND IMPACT OF THE STUDY Since vancomycin is one of the last treatment options currently available, and it is unable to inhibit biofilm, the research of new antimicrobials is urgent. In this context, 2-hydroxy-3-phenylsulfanylmethyl-[1,4]-naphthoquinones proved to be a promising lead structure against MRSA and bacterial biofilm.
Collapse
Affiliation(s)
- C S Moreira
- Organic Chemistry Department, Chemistry Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - A C J A Silva
- Molecular and Cell Biology Department, PPBI, Biology Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - J S Novais
- Molecular and Cell Biology Department, PPBI, Biology Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - A M Sá Figueiredo
- Department of Medical Microbiology, Institute of Microbiology Professor Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - V F Ferreira
- Organic Chemistry Department, Chemistry Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - D R da Rocha
- Organic Chemistry Department, Chemistry Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| | - H C Castro
- Molecular and Cell Biology Department, PPBI, Biology Institute, Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Kong B, Joshi T, Belousoff MJ, Tor Y, Graham B, Spiccia L. Neomycin B-cyclen conjugates and their Zn(II) complexes as RNA-binding agents. J Inorg Biochem 2016; 162:334-342. [DOI: 10.1016/j.jinorgbio.2015.11.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/09/2015] [Accepted: 11/30/2015] [Indexed: 11/26/2022]
|
16
|
Walsh CC, Landersdorfer CB, McIntosh MP, Peleg AY, Hirsch EB, Kirkpatrick CM, Bergen PJ. Clinically relevant concentrations of fosfomycin combined with polymyxin B, tobramycin or ciprofloxacin enhance bacterial killing of Pseudomonas aeruginosa, but do not suppress the emergence of fosfomycin resistance. J Antimicrob Chemother 2016; 71:2218-29. [PMID: 27118778 DOI: 10.1093/jac/dkw115] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/09/2016] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Fosfomycin resistance occurs rapidly with monotherapy. This study systematically investigated bacterial killing and emergence of fosfomycin resistance with fosfomycin combinations against Pseudomonas aeruginosa. METHODS Four clinical isolates and a reference strain of P. aeruginosa were employed. Combinations of fosfomycin plus polymyxin B, tobramycin or ciprofloxacin were examined over 24 h using time-kill studies (inocula ∼10(6) cfu/mL) incorporating clinically relevant concentrations (fosfomycin, 30, 150 or 300 mg/L; polymyxin B, 0.5, 1 or 2 mg/L; tobramycin, 0.5, 1.5 or 4 mg/L; ciprofloxacin, 0.5, 1 or 2.5 mg/L). Microbiological response was examined by log changes and population analysis profiles. RESULTS Against susceptible isolates, monotherapy produced varying degrees of initial killing followed by rapid regrowth. Fosfomycin plus polymyxin B or tobramycin produced greater initial killing (up to ∼4 log10 cfu/mL) with many concentrations compared with monotherapy against fosfomycin-susceptible (FOF(S)) isolates. With these combinations, synergy or additivity was observed in 54 (67%) and 49 (60%) of 81 cases (nine combinations across three isolates at three timepoints) for polymyxin B and tobramycin, respectively. Substantial improvements in killing were absent against fosfomycin-resistant (FOF(R)) isolates. For fosfomycin/ciprofloxacin combinations, synergy or additivity was observed against FOF(R) isolates in 33 of 54 (61%) cases (nine combinations across two isolates at three timepoints), while improvements in killing were largely absent against FOF(S) isolates. No combination prevented emergence of fosfomycin resistance. CONCLUSIONS Against P. aeruginosa, fosfomycin in combination with polymyxin B or tobramycin (FOF(S) isolates) or ciprofloxacin (FOF(R) isolates) increased bacterial killing, but did not suppress emergence of fosfomycin resistance.
Collapse
Affiliation(s)
- Clare C Walsh
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| | | | - Michelle P McIntosh
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia Department of Microbiology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Elizabeth B Hirsch
- Department of Pharmacy and Health Systems Sciences, Northeastern University, Boston, MA, USA
| | - Carl M Kirkpatrick
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| | - Phillip J Bergen
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Walsh CC, McIntosh MP, Peleg AY, Kirkpatrick CM, Bergen PJ. In vitro pharmacodynamics of fosfomycin against clinical isolates of Pseudomonas aeruginosa. J Antimicrob Chemother 2015. [PMID: 26209311 DOI: 10.1093/jac/dkv221] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The use of fosfomycin for treatment of systemic infections due to MDR Pseudomonas aeruginosa is increasing. However, pharmacodynamic data for fosfomycin are limited. METHODS Sixty-four clinical isolates of P. aeruginosa (MDR and non-MDR) from two Australian hospitals were collected; 59 isolates were from patients with cystic fibrosis and 5 isolates were from critically ill patients. The in vitro pharmacodynamic properties of fosfomycin (disodium) were investigated via MICs (all isolates) and, for selected isolates, via time-kill kinetics (static and dynamic models; concentration range, 1-1024 mg/L), population analysis profiles (PAPs) and post-antibiotic effect (PAE). Two inocula (∼10(6) and ∼10(8) cfu/mL) were included in static time-kill studies to examine the effect of inocula on bacterial killing. RESULTS MICs ranged from 1 to >512 mg/L, with 61% of isolates considered fosfomycin susceptible (MIC ≤64 mg/L). The MIC distributions for MDR and non-MDR isolates were similar. Baseline PAPs indicated heteroresistance in all isolates tested. Time-kill studies showed moderate (maximum killing ∼3 log10 cfu/mL), time-dependent killing at the low inoculum with regrowth at 24 h. Most concentrations resulted in complete replacement of fosfomycin-susceptible colonies by fosfomycin-resistant colonies. Bacterial killing was virtually eliminated at the high inoculum. The PAE ranged from 0.3 to 5.5 h. CONCLUSIONS These data suggest monotherapy with fosfomycin may be problematic for the treatment of infections caused by P. aeruginosa. Further investigation of fosfomycin combination therapy is warranted.
Collapse
Affiliation(s)
- Clare C Walsh
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| | - Michelle P McIntosh
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia Department of Microbiology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Carl M Kirkpatrick
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| | - Phillip J Bergen
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Serra I, Scorciapino MA, Manzo G, Casu M, Rinaldi AC, Attoub S, Mechkarska M, Conlon JM. Conformational analysis and cytotoxic activities of the frog skin host-defense peptide, hymenochirin-1Pa. Peptides 2014; 61:114-21. [PMID: 25241629 DOI: 10.1016/j.peptides.2014.08.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 10/24/2022]
Abstract
Hymenochirin-1Pa (LKLSPKTKDTLKKVLKGAIKGAIAIASMA-NH2) is a host-defense peptide first isolated from skin secretions of the frog Pseudhymenochirus merlini (Pipidae). A nuclear magnetic resonance structural investigation demonstrates that the peptide has a random coil conformation in water but, in the membrane-mimetic solvent 50% (v/v) trifluoroethanol-water adopts a well-defined conformation characterized by two α-helical domains from residues K6 to G17 and from G21 to M28, with the N-terminal region unfolded. The presence of a GXXXG domain, the most common structural motif found at the interface between interacting trans-membrane helices, between residues 17 and 21, introduces a kink corresponding to a deviation from linearity of 93 ± 31°. Hymenochirin-1Pa shows broad spectrum anti-bacterial activity, including high potency against multidrug-resistant clinical isolates of Staphylococcus aureus, Acinetobacter baumannii, and Stenotrophomonas maltophilia. The peptide also shows high cytotoxic potency against human non-small lung adenocarcinoma A549 cells, breast adenocarcinoma MDA-MB-231 cells, and colorectal adenocarcinoma HT-29 cells but its therapeutic potential as an anti-cancer agent is limited by moderate hemolytic activity against human erythrocytes and lack of selectivity for tumor cells. Increasing cationicity of the peptide by substituting the Asp(9) residue by either L-Lys (K) or D-Lys (k) has relatively minor effects on antimicrobial and anti-tumor potencies but the [D9k] analog is non-hemolytic LC50 > 400 μM. Thus, [D9k]hymenochirin-1Pa may serve as a template for the design of non-toxic antimicrobial agents for use against multidrug-resistant pathogenic bacteria.
Collapse
Affiliation(s)
- Ilaria Serra
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato (CA), Italy
| | - Mariano A Scorciapino
- Department of Biomedical Sciences, Biochemistry Unit, University of Cagliari, Monserrato (CA), Italy.
| | - Giorgia Manzo
- Department of Biomedical Sciences, Biochemistry Unit, University of Cagliari, Monserrato (CA), Italy
| | - Mariano Casu
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato (CA), Italy
| | - Andrea C Rinaldi
- Department of Biomedical Sciences, Biochemistry Unit, University of Cagliari, Monserrato (CA), Italy
| | - Samir Attoub
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Milena Mechkarska
- Department of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine BT52 1SA, Northern Ireland, United Kingdom
| | - J Michael Conlon
- Department of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine BT52 1SA, Northern Ireland, United Kingdom
| |
Collapse
|
19
|
Alsultan AA, Aboulmagd E, Evans BA, Amyes SGB. Clonal diversity of Acinetobacter baumannii from diabetic patients in Saudi Arabian hospitals. J Med Microbiol 2014; 63:1460-1466. [DOI: 10.1099/jmm.0.079640-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Carbapenem-resistant Acinetobacter baumannii (CR-AB) represents a major health-care problem, causing high rates of morbidity and mortality. This study investigated the clonality of CR-AB isolated from diabetic patients from different regions in Saudi Arabia, as well as the relatedness of the β-lactamase genes. A total of 64 non-repetitive CR-AB clinical isolates were collected from 16 different regions in Saudi Arabia from intensive care patients. Isolates were identified phenotypically by the Vitek 2 compact system and genotypically by amplification of the bla
OXA-51-like gene. The target sequences were amplified by PCR and the clonal diversity of the isolates was explored by PFGE. Resistance studies revealed that the prevalence of imipenem and meropenem resistance was 92 % and 96 %, respectively, while the vast majority of the isolates were susceptible to tigecycline and colistin. In addition, bla
VIM and bla
OXA-23 were the most prevalent genes in the isolates under investigation, while ISAba1 was the most dominant insertion sequence. PFGE results showed 13 clusters; clone H was dominant, comprising 20 isolates from four hospitals, followed by clones C and F, comprising 11 isolates each from three and six hospitals, respectively. Moreover, the current study signified the clonal diversity of CR-AB in Saudi Arabia and showed the ability of some clones to infect patients in many different cities.
Collapse
Affiliation(s)
- Abdulrahman A. Alsultan
- Department of Medical Microbiology, College of Medicine, King Faisal University, Al-Ahsa 31982, PO Box 400, Kingdom of Saudi Arabia
| | - Elsayed Aboulmagd
- Department of Microbiology, Faculty of Pharmacy, Alexandria University, El-Khartoum Square, Azarita, Alexandria 21521, Egypt
- Department of Medical Microbiology, College of Medicine, King Faisal University, Al-Ahsa 31982, PO Box 400, Kingdom of Saudi Arabia
| | - Benjamin A. Evans
- Department of Life Sciences, Faculty of Science and Technology, Anglia Ruskin University, East Road, Cambridge CB1 1PT, UK
| | - Sebastian G. B. Amyes
- Department of Medical Microbiology, College of Medicine and Veterinary Medicine, University of Edinburgh, The Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
20
|
Joshi T, Voo ZX, Graham B, Spiccia L, Martin LL. Real-time examination of aminoglycoside activity towards bacterial mimetic membranes using Quartz Crystal Microbalance with Dissipation monitoring (QCM-D). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:385-91. [PMID: 25450807 DOI: 10.1016/j.bbamem.2014.10.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 09/03/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
The rapid increase in multi-drug resistant bacteria has resulted in previously discontinued treatments being revisited. Aminoglycosides are effective "old" antibacterial agents that fall within this category. Despite extensive usage and understanding of their intracellular targets, there is limited mechanistic knowledge regarding how aminoglycosides penetrate bacterial membranes. Thus, the activity of two well-known aminoglycosides, kanamycin A and neomycin B, towards a bacterial mimetic membrane (DMPC:DMPG (4:1)) was examined using a Quartz Crystal Microbalance with Dissipation monitoring (QCM-D). The macroscopic effect of increasing the aminoglycoside concentration showed that kanamycin A exerts a threshold response, switching from binding to the membrane to disruption of the surface. Neomycin B, however, disrupted the membrane at all concentrations examined. At concentrations above the threshold value observed for kanamycin A, both aminoglycosides revealed similar mechanistic details. That is, they both inserted into the bacterial mimetic lipid bilayer, prior to disruption via loss of materials, presumably aminoglycoside-membrane composites. Depth profile analysis of this membrane interaction was achieved using the overtones of the quartz crystal sensor. The measured data is consistent with a two-stage process in which insertion of the aminoglycoside precedes the 'detergent-like' removal of membranes from the sensor. The results of this study contribute to the insight required for aminoglycosides to be reconsidered as active antimicrobial agents/co-agents by providing details of activity at the bacterial membrane. Kanamycin and neomycin still offer potential as antimicrobial therapeutics for the future and the QCM-D method illustrates great promise for screening new antibacterial or antiviral drug candidates.
Collapse
Affiliation(s)
- Tanmaya Joshi
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Zhi Xiang Voo
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Bim Graham
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Leone Spiccia
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia.
| | - Lisandra L Martin
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
21
|
Fair RJ, Tor Y. Antibiotics and bacterial resistance in the 21st century. PERSPECTIVES IN MEDICINAL CHEMISTRY 2014; 6:25-64. [PMID: 25232278 PMCID: PMC4159373 DOI: 10.4137/pmc.s14459] [Citation(s) in RCA: 871] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/24/2014] [Accepted: 06/24/2014] [Indexed: 12/11/2022]
Abstract
Dangerous, antibiotic resistant bacteria have been observed with increasing frequency over the past several decades. In this review the factors that have been linked to this phenomenon are addressed. Profiles of bacterial species that are deemed to be particularly concerning at the present time are illustrated. Factors including economic impact, intrinsic and acquired drug resistance, morbidity and mortality rates, and means of infection are taken into account. Synchronously with the waxing of bacterial resistance there has been waning antibiotic development. The approaches that scientists are employing in the pursuit of new antibacterial agents are briefly described. The standings of established antibiotic classes as well as potentially emerging classes are assessed with an emphasis on molecules that have been clinically approved or are in advanced stages of development. Historical perspectives, mechanisms of action and resistance, spectrum of activity, and preeminent members of each class are discussed.
Collapse
Affiliation(s)
- Richard J Fair
- Department for Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Berlin, Germany
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Borisova M, Gisin J, Mayer C. Blocking peptidoglycan recycling in Pseudomonas aeruginosa attenuates intrinsic resistance to fosfomycin. Microb Drug Resist 2014; 20:231-7. [PMID: 24819062 DOI: 10.1089/mdr.2014.0036] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Gram-negative bacteria recycle as much as half of their cell wall per generation. Here we show that interference with cell wall recycling in Pseudomonas aeruginosa strains results in four- to eight-fold increased susceptibility to the antibiotic fosfomycin, pushing the minimal inhibitory concentration for strains PA14 and PA01 to therapeutically appropriate values of 2-4 and 8-16 mg/L, respectively. A newly discovered metabolic pathway that connects cell wall recycling with peptidoglycan de novo biosynthesis is responsible for the high intrinsic resistance of P. aeruginosa to fosfomycin. The pathway comprises an anomeric cell wall amino sugar kinase (AmgK) and an uridylyl transferase (MurU), which together convert N-acetylmuramic acid (MurNAc) through MurNAc α-1-phosphate to uridine diphosphate (UDP)-MurNAc, thereby bypassing the fosfomycin-sensitive de novo synthesis of UDP-MurNAc. Thus, inhibition of peptidoglycan recycling can be applied as a new strategy for the combinatory therapy against multidrug-resistant P. aeruginosa strains.
Collapse
Affiliation(s)
- Marina Borisova
- Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen , Tübingen, Germany
| | | | | |
Collapse
|
23
|
Qin W, Panunzio M, Biondi S. β-Lactam Antibiotics Renaissance. Antibiotics (Basel) 2014; 3:193-215. [PMID: 27025744 PMCID: PMC4790388 DOI: 10.3390/antibiotics3020193] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/30/2014] [Accepted: 05/04/2014] [Indexed: 11/16/2022] Open
Abstract
Since the 1940s β-lactam antibiotics have been used to treat bacterial infections. However, emergence and dissemination of β-lactam resistance has reached the point where many marketed β-lactams no longer are clinically effective. The increasing prevalence of multidrug-resistant bacteria and the progressive withdrawal of pharmaceutical companies from antibiotic research have evoked a strong reaction from health authorities, who have implemented initiatives to encourage the discovery of new antibacterials. Despite this gloomy scenario, several novel β-lactam antibiotics and β-lactamase inhibitors have recently progressed into clinical trials, and many more such compounds are being investigated. Here we seek to provide highlights of recent developments relating to the discovery of novel β-lactam antibiotics and β-lactamase inhibitors.
Collapse
Affiliation(s)
- Wenling Qin
- ISOF-CNR Department of Chemistry "G. Ciamician", Via Selmi, 2 I-40126 Bologna, Italy.
| | - Mauro Panunzio
- ISOF-CNR Department of Chemistry "G. Ciamician", Via Selmi, 2 I-40126 Bologna, Italy.
| | - Stefano Biondi
- Allecra Therapeutics SAS, 13, rue de Village-Neuf, F-68300 St-Louis, France.
| |
Collapse
|
24
|
Renteria MI, Biedenbach DJ, Bouchillon SK, Hoban DJ, Raghubir N, Sajben P, Mokaddas E. In vitro activity of tigecycline against isolates collected from complicated skin and skin structure infections and intra-abdominal infections in Africa and Middle East countries: TEST 2007-2012. Diagn Microbiol Infect Dis 2014; 79:54-9. [PMID: 24582580 DOI: 10.1016/j.diagmicrobio.2014.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 01/22/2014] [Accepted: 01/23/2014] [Indexed: 11/29/2022]
Abstract
Complicated skin and skin structure infections (cSSSIs) and intra-abdominal infections (IAIs) are problematic due to decreasing therapeutic options available against multidrug-resistant pathogens common among these types of infections. A total of 2245 isolates from African and the Middle Eastern (AfME) countries were collected to determine in vitro activity for tigecycline and comparators during 2007-2012 as part of the Tigecycline Evaluation Surveillance Trial program. Tigecycline was launched in the AfME in 2007 and remains active against a wide range of targeted pathogens worldwide. Isolates were recovered from cSSSI (1990) and IAI (255) from 38 sites in 11 AfME countries. Staphylococcus aureus was the most common species from cSSSI (27.9%), and the methicillin-resistant S. aureus rate was 25%. Enterococcus spp. (7.1%) and Streptococcus agalactiae (2.9%) were other common Gram-positive pathogens represented. Enterobacter spp. (14.5%), Pseudomonas aeruginosa (13.9%), Escherichia coli (11.4%), Klebsiella spp. (10.9%), and Acinetobacter spp. (7.2 %) were the most common Gram-negative species collected. Tigecycline MIC(90) values were 0.25 μg/mL against S. aureus. E. coli and Enterobacter spp. had tigecycline MIC(90) values of 1 and 2 μg/mL, respectively. E. coli was the most frequently collected species from IAI (28.3%), followed by Klebsiella spp. (20.8%), Enterococcus spp. (11.8%), and Stenotrophomonas maltophilia (6.3%). Isolates collected from IAI had the following tigecycline MIC(90) values: E. coli (1 μg/mL), Klebsiella spp. and other Enterobacteriaceae (2 μg/mL), Enterococcus spp. (0.25 μg/mL), and S. maltophilia (1 μg/mL). Tigecycline in vitro activity was observed against a broad spectrum of bacterial species, including strains resistant to other antimicrobial classes.
Collapse
Affiliation(s)
- M I Renteria
- International Health Management Associates, Inc., Schaumburg, IL, USA.
| | - D J Biedenbach
- International Health Management Associates, Inc., Schaumburg, IL, USA
| | - S K Bouchillon
- International Health Management Associates, Inc., Schaumburg, IL, USA
| | - D J Hoban
- International Health Management Associates, Inc., Schaumburg, IL, USA
| | - N Raghubir
- Pfizer Inc., Africa Middle East, Gulf and Levant
| | - P Sajben
- Pfizer Inc., Africa Middle East, Gulf and Levant
| | | |
Collapse
|
25
|
Abstract
AbstractBacterial resistance to commonly used antibiotics is constantly increasing. Bacteria particularly dangerous for human life are methicillin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus faecium and fluoroquinolone-resistant Pseudomonas aeruginosa. Hence, there is an incessant need for developing compounds with new modes of action and seeking alternate drug targets. In this review, the authors discuss the current situation of antibacterial medicines and present data on new antibiotic targets. Moreover, alternatives to antibiotics, such as bacteriophages, antimicrobial peptides and monoclonal antibodies, are presented. The authors also draw attention to the valuable features of natural sources in developing antibacterial compounds. The need to prevent and control infections as well as the reasonable use of currently available antibiotics is also emphasized.
Collapse
|
26
|
Zhao Y, Jiang X. Multiple strategies to activate gold nanoparticles as antibiotics. NANOSCALE 2013; 5:8340-50. [PMID: 23893008 DOI: 10.1039/c3nr01990j] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Widespread antibiotic resistance calls for new strategies. Nanotechnology provides a chance to overcome antibiotic resistance by multiple antibiotic mechanisms. This paper reviews the progress in activating gold nanoparticles with nonantibiotic or antibiotic molecules to combat bacterial resistance, analyzes the gap between experimental achievements and real clinical application, and suggests some potential directions in developing antibacterial nanodrugs.
Collapse
Affiliation(s)
- Yuyun Zhao
- CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, 11 Beiyitiao, ZhongGuanCun, 100190, Beijing, China
| | | |
Collapse
|
27
|
Nielsen EI, Friberg LE. Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs. Pharmacol Rev 2013; 65:1053-90. [PMID: 23803529 DOI: 10.1124/pr.111.005769] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pharmacokinetic-pharmacodynamic (PKPD) modeling and simulation has evolved as an important tool for rational drug development and drug use, where developed models characterize both the typical trends in the data and quantify the variability in relationships between dose, concentration, and desired effects and side effects. In parallel, rapid emergence of antibiotic-resistant bacteria imposes new challenges on modern health care. Models that can characterize bacterial growth, bacterial killing by antibiotics and immune system, and selection of resistance can provide valuable information on the interactions between antibiotics, bacteria, and host. Simulations from developed models allow for outcome predictions of untested scenarios, improved study designs, and optimized dosing regimens. Today, much quantitative information on antibiotic PKPD is thrown away by summarizing data into variables with limited possibilities for extrapolation to different dosing regimens and study populations. In vitro studies allow for flexible study designs and valuable information on time courses of antibiotic drug action. Such experiments have formed the basis for development of a variety of PKPD models that primarily differ in how antibiotic drug exposure induces amplification of resistant bacteria. The models have shown promise for efficacy predictions in patients, but few PKPD models describe time courses of antibiotic drug effects in animals and patients. We promote more extensive use of modeling and simulation to speed up development of new antibiotics and promising antibiotic drug combinations. This review summarizes the value of PKPD modeling and provides an overview of the characteristics of available PKPD models of antibiotics based on in vitro, animal, and patient data.
Collapse
Affiliation(s)
- Elisabet I Nielsen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
28
|
Abstract
INTRODUCTION Natural products have long been instrumental for discovering antibiotics, but many pharmaceutical companies abandoned this field and new antibiotics declined. In contrast, microbial resistance to current antibiotics has approached critical levels. AREAS COVERED This article gives historical perspectives by providing background about present-day economic realities and medical needs for antibiotic research, whose pipeline is mostly focused toward older known agents and newer semi-synthetic derivatives. Future research trends and projected technological developments open many innovative opportunities to discover novel antibacterials and find ways to control pathogenic bacteria without conventional antibiotics that provoke resistance. EXPERT OPINION The successful registration of daptomycin, retapamulin and fidaxomicin indicate the re-emergence of natural products has already begun. Semi-synthetic derivatives from other under-explored classes are progressing. More effort is being put into approaches such as total synthesis, discovery of new structural scaffolds for synthesis, alterations of biosynthetic pathways, combinatorial biosynthesis, new screening targets and new resources from which to isolate natural products. A return to successful screening of actinomycetes depends on solving the rate-limiting dereplication obstacle. Long-term solutions need to come from greater exploration of the massive numbers of uncultured microbes. An ultimate solution to the antibiotic-promoted microbial resistance cycle may lie in finding ways to control bacteria by non-lethal means.
Collapse
|
29
|
Carlet J, Pittet D. Access to antibiotics: a safety and equity challenge for the next decade. Antimicrob Resist Infect Control 2013; 2:1. [PMID: 23305311 PMCID: PMC3599140 DOI: 10.1186/2047-2994-2-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 01/06/2013] [Indexed: 12/02/2022] Open
Abstract
Bacterial resistance to antibiotics is increasing worldwide in healthcare settings and in the community. Some microbial pathogens have become resistant to multiple antibiotics, if not all presently available, thus severely compromising treatment success and contributing to enhanced morbidity, mortality, and resource use. The major driver of resistance is misuse of antibiotics in both human and non-human medicine. Both enhanced access and restricted use in many parts of the world is mandatory. There is an urgent need for an international, integrated, multi-level action to preserve antibiotics in the armamentarium of the 21st century and address the global issue of antimicrobial resistance.
Collapse
Affiliation(s)
- Jean Carlet
- World Alliance Against Antibiotic Resistance (WAAR), 9 rue de la Terrasse, 94000 Creteil, France
- Hôpital St Joseph, 185 rue Raymond Losserand, 75014, Paris, France
| | - Didier Pittet
- Infection Control Programme and WHO Collaboration Centre on Patient Safety, University of Geneva Hospitals and Faculty of Medicine, 4 Rue Gabrielle Perret-Gentil, Geneva, Switzerland
| |
Collapse
|
30
|
Abstract
The "perpetual challenge of infectious diseases" is no better exemplified than by the phenomena of rapid emergence and spread of bacterial resistance. Although β-lactam antimicrobials have thus far been a mainstay of the therapeutic armamentarium for treatment of severe infections, their preeminent position has been challenged by an onslaught of resistance mechanisms in major nosocomial and community-acquired pathogens, including modification of penicillin-binding proteins, production of β-lactamases, overexpression of efflux pumps, and loss of porins. Given this formidable array of resistance mechanisms facing patients and their physicians, one can reasonably ask if there is hope for the future of β-lactams.
Collapse
|
31
|
Cozzone AJ. An insight into future antibacterial therapy. Emerg Microbes Infect 2012; 1:e38. [PMID: 26038407 PMCID: PMC3636425 DOI: 10.1038/emi.2012.35] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/06/2012] [Accepted: 09/17/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Alain J Cozzone
- Institute of Biology and Chemistry of Proteins, University of Lyon, 7 passage du Vercors , 69007 Lyon, France
| |
Collapse
|
32
|
Oleksiewicz MB, Nagy G, Nagy E. Anti-bacterial monoclonal antibodies: Back to the future? Arch Biochem Biophys 2012; 526:124-31. [DOI: 10.1016/j.abb.2012.06.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/01/2012] [Accepted: 06/02/2012] [Indexed: 01/08/2023]
|
33
|
Sharma A, Kumar Arya D, Dua M, Chhatwal GS, Johri AK. Nano-technology for targeted drug delivery to combat antibiotic resistance. Expert Opin Drug Deliv 2012; 9:1325-32. [PMID: 22924701 DOI: 10.1517/17425247.2012.717927] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several microbes have evolved clinically significant resistance against almost every available antibiotic. Yet the development of new classes of antibiotics has lagged far behind our growing need. Frequent and suboptimal use of antibiotics particularly in developing countries aggravated the problem by increasing the rate of resistance. Therefore, developing new and multidimensional strategies to combat microbial infections is warranted. These include i) modification of existing antibiotics, ii) searching new and novel antibiotics, iii) development and improvement of antibiotics carrier system to reduce amount and frequency of antibiotic doses, iv) development of targeted antibiotic delivery systems. Here, the authors discuss trends and development of nano-materials and alternative antimicrobials to solve the problem of antibiotic resistance.
Collapse
|
34
|
Livermore DM. Fourteen years in resistance. Int J Antimicrob Agents 2012; 39:283-94. [PMID: 22386741 DOI: 10.1016/j.ijantimicag.2011.12.012] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 12/29/2011] [Indexed: 12/31/2022]
Abstract
Resistance trends have changed greatly over the 14 years (1997-2011) whilst I was Director of the UK Antibiotic Resistance Monitoring and Reference Laboratory (ARMRL). Meticillin-resistant Staphylococcus aureus (MRSA) first rose, then fell with improved infection control, although with the decline of one major clone beginning before these improvements. Resistant pneumococci too have declined following conjugate vaccine deployment. If the situation against Gram-positive pathogens has improved, that against Gram-negatives has worsened, with the spread of (i) quinolone- and cephalosporin-resistant Enterobacteriaceae, (ii) Acinetobacter with OXA carbapenemases, (iii) Enterobacteriaceae with biochemically diverse carbapenemases and (iv) gonococci resistant to fluoroquinolones and, latterly, cefixime. Laboratory, clinical and commercial aspects have also changed. Susceptibility testing is more standardised, with pharmacodynamic breakpoints. Treatments regimens are more driven by guidelines. The industry has fewer big profitable companies and more small companies without sales income. There is good and bad here. The quality of routine susceptibility testing has improved, but its speed has not. Pharmacodynamics adds science, but over-optimism has led to poor dose selection in several trials. Guidelines discourage poor therapy but concentrate selection onto a diminishing range of antibiotics, threatening their utility. Small companies are more nimble, but less resilient. Last, more than anything, the world has changed, with the rise of India and China, which account for 33% of the world's population and increasingly provide sophisticated health care, but also have huge resistance problems. These shifts present huge challenges for the future of chemotherapy and for the edifice of modern medicine that depends upon it.
Collapse
Affiliation(s)
- David M Livermore
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK.
| |
Collapse
|
35
|
Hassan M, Javadzadeh Y, Lotfipour F, Badomchi R. Determination of comparative minimum inhibitory concentration (MIC) of bacteriocins produced by enterococci for selected isolates of multi-antibiotic resistant Enterococcus spp. Adv Pharm Bull 2011; 1:75-9. [PMID: 24312760 DOI: 10.5681/apb.2011.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 11/19/2011] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION The occurrence of multi-antibiotic resistance among enterococci is a prevalent clinical problem worldwide and continues to get serious due to the lack of efficient therapeutic options by the time. In this regards, prokaryotic antimicrobial peptides with bactericidal or bacteriostatic activity which are directed against bacterial strains closely related to producer strains looks one of the promising alternative to conventional antibiotics. METHODS The antibiotic susceptibility pattern of 20 clinical isolates of enterococci was evaluated and subsequently the isolates were screened for antibacterial activity against three different indicator strains. The efficacy of potential bacteriocinogenic isolates were assayed against multi-antibiotic resistant Enterococcus spp. by comparative minimum inhibitory concentration (MIC). RESULTS Antibiotic resistant pattern of enterococcal isolates demonstrated that multi-antibiotic resistant to conventional antibiotics were significantly high and the prevalent pattern of resistance was combination of gentamicin, streptomycin, chloramphenicol and vancomycin. In addition, the data from comparative MIC showed the noticeable activity of selected potential bacteriocinogenic strains against pathogenic enterococci. CONCLUSION The present survey may address the potential applicability of antimicrobial peptides in alleviating the problems of antibiotic resistance.
Collapse
Affiliation(s)
- Maryam Hassan
- Faculty of Pharmacy and Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|