51
|
Correa-Martinez CL, Stollenwerk VB, Kossow A, Schaumburg F, Mellmann A, Kampmeier S. Risk Factors for Long-Term Vancomycin-Resistant Enterococci Persistence-A Prospective Longitudinal Study. Microorganisms 2019; 7:E400. [PMID: 31561632 PMCID: PMC6843193 DOI: 10.3390/microorganisms7100400] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 01/05/2023] Open
Abstract
Vancomycin-resistant enterococci (VRE) are important nosocomial pathogens that require effective infection control measures, representing a challenge for healthcare systems. This study aimed at identifying risk factors associated with prolonged VRE carriage and determining the rate of clearance that allows the discontinuation of contact precautions. During a 2-year study, screening was performed in patients with a history of VRE or at risk of becoming colonized. After bacterial identification and antibiotic susceptibility testing, glycopeptide resistance was confirmed by PCR. Isolates were compared via whole genome sequence-based typing. Risk factors were recorded, and follow-up screening was performed upon readmission, defining patients as long-term carriers if still colonized ≥10 weeks after first detection. Of 1059 patients positive for VRE, carriage status was assessed upon readmission in 463 patients. VRE was cleared in 56.4% of the cases. Risk factors associated with long-term persistence were hospital stays (frequency, length), hemato-oncological disease, systemic treatment with steroids, and use of antibiotics. No specific genotypic clustering was observed in patients with VRE clearance or persistence. VRE clearance is possibly underestimated. The identification of risk factors favoring long-term carriage may contribute to a targeted implementation of infection control measures upon readmission of patients with history of VRE.
Collapse
Affiliation(s)
- Carlos L Correa-Martinez
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Straße 41, 48149 Münster, Germany.
| | - Verena B Stollenwerk
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Straße 41, 48149 Münster, Germany.
| | - Annelene Kossow
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Straße 41, 48149 Münster, Germany.
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, Domagkstraße 10, 48149 Münster, Germany.
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Straße 41, 48149 Münster, Germany.
| | - Stefanie Kampmeier
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Straße 41, 48149 Münster, Germany.
| |
Collapse
|
52
|
Zhang Q, Ma P, Xie J, Zhang S, Xiao X, Qiao Z, Shao N, Zhou M, Zhang W, Dai C, Qian Y, Qi F, Liu R. Host defense peptide mimicking poly-β-peptides with fast, potent and broad spectrum antibacterial activities. Biomater Sci 2019; 7:2144-2151. [PMID: 30882803 DOI: 10.1039/c9bm00248k] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microbial infections have always been serious challenges to human health considering that antibiotics almost inevitably induce microbial resistance. Therefore, it is urgent to develop a new antibacterial agent that is active against drug-resistant bacteria and is less susceptible to microbial resistance. In this work, a series of host defense peptide (HDP) mimicking antibacterial poly-β-peptides were synthesized, characterized and evaluated for their biological activities. The best poly-β-peptide within this study (20 : 80 Bu : DM) displays potent and broad spectrum antibacterial activity against antibiotic-resistant super bugs and low toxicity toward mammalian cells. Moreover, these poly-β-peptides are bactericidal and kill bacteria very fast within 5 min. An antimicrobial resistance test demonstrated that bacteria develop no resistance toward the selected poly-β-peptides even over 1000 generations. Our studies demonstrate that random copolymers of heterochiral poly-β-peptides, without the need for defined secondary structures, can mimic the antimicrobial HDP. These results imply the potential application of these poly-β-peptides as new antimicrobial agents to tackle drug resistant antimicrobial infections.
Collapse
Affiliation(s)
- Qiang Zhang
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Wu ZC, Boger DL. Exploration of the site-specific nature and generalizability of a trimethylammonium salt modification on vancomycin: A-ring derivatives. Tetrahedron 2019; 75:3160-3165. [PMID: 31327878 PMCID: PMC6640857 DOI: 10.1016/j.tet.2019.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Vancomycin analogues bearing an A-ring trimethylammonium salt modification were synthesized and their antimicrobial activity against vancomycin-resistant Enterococci (VRE) was evaluated. The modification increased antimicrobial potency and provided the capability to induce bacteria cell membrane permeabilization, but both properties were weaker than that found with our earlier reported similar C-terminus modification. The results provide further insights on the additive effect and generalizability of the structural and site-specific nature of a peripheral quaternary trimethylammonium salt modification of vancomycin.
Collapse
Affiliation(s)
- Zhi-Chen Wu
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
54
|
Guan D, Chen F, Qiu Y, Jiang B, Gong L, Lan L, Huang W. Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug‐Resistant Bacteria. Angew Chem Int Ed Engl 2019; 58:6678-6682. [DOI: 10.1002/anie.201902210] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Dongliang Guan
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Feifei Chen
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road, Pudong Shanghai 201203 China
| | - Yunguang Qiu
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Bofeng Jiang
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Likun Gong
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road, Pudong Shanghai 201203 China
| | - Lefu Lan
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road, Pudong Shanghai 201203 China
| | - Wei Huang
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| |
Collapse
|
55
|
Guan D, Chen F, Qiu Y, Jiang B, Gong L, Lan L, Huang W. Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug‐Resistant Bacteria. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201902210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Dongliang Guan
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Feifei Chen
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Yunguang Qiu
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Bofeng Jiang
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Likun Gong
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Lefu Lan
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Wei Huang
- CAS Key Laboratory of Receptor ResearchCAS Center for Excellence in Molecular Cell ScienceCenter for Biotherapeutics Discovery ResearchShanghai Institute of Materia MedicaChinese Academy of Sciences 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| |
Collapse
|
56
|
Huang CM, Lyu SY, Lin KH, Chen CL, Chen MH, Shih HW, Hsu NS, Lo IW, Wang YL, Li YS, Wu CJ, Li TL. Teicoplanin Reprogrammed with the N-Acyl-Glucosamine Pharmacophore at the Penultimate Residue of Aglycone Acquires Broad-Spectrum Antimicrobial Activities Effectively Killing Gram-Positive and -Negative Pathogens. ACS Infect Dis 2019; 5:430-442. [PMID: 30599088 DOI: 10.1021/acsinfecdis.8b00317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Lipoglycopeptide antibiotics, for example, teicoplanin (Tei) and A40926, are more potent than vancomycin against Gram-positive (Gram-(+)) drug-resistant pathogens, for example, methicillin-resistant Staphylococcus aureus (MRSA). To extend their therapeutic effectiveness on vancomycin-resistant S. aureus (VRSA), the biosynthetic pathway of the N-acyl glucosamine (Glc) pharmacophore at residue 4 (r4) of teicoplanin pseudoaglycone redirection to residue 6 (r6) was attempted. On the basis of crystal structures, two regioselective biocatalysts Orf2*T (a triple-mutation mutant S98A/V121A/F193Y) and Orf11*S (a single-mutation mutant W163A) were engineered, allowing them to act on GlcNAc at r6. New analogs thereby made show marked antimicrobial activity against MRSA and VRSA by 2-3 orders of magnitude better than teicoplanin and vancomycin. The lipid side chain of the Tei-analogs armed with a terminal mono- or diguanidino group extends the antimicrobial specificity from Gram-(+) to Gram-negative (Gram-(-)), comparable to that of kanamycin. In addition to low cytotoxicity and high safety, the Tei analogs exhibit new modes of action as a result of resensitization of VRSA and Acinetobacter baumannii. The redirection of the biosynthetic pathway for the N-acyl-Glc pharmacophore from r4 to r6 bodes well for large-scale production of selected r6,Tei congeners in an environmentally friendly synthetic biology approach.
Collapse
Affiliation(s)
- Chun-Man Huang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
- Department of Microbiology and Immunology, National Yang-Ming University, 155 Linong Street, Section 2,
Beitou, Taipei 11221, Taiwan
| | - Syue-Yi Lyu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Kuan-Hung Lin
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Chun-Liang Chen
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Mei-Hua Chen
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Hao-Wei Shih
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Ning-Shian Hsu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - I-Wen Lo
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yung-Lin Wang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yi-Shan Li
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Chang-Jer Wu
- National Taiwan Ocean University, 2 Peining Road, Jhongjhong, Keelung 20224, Taiwan
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
- National Chung-Hsing University, 145 Xingda Road, South Taichung 402, Taiwan
| |
Collapse
|
57
|
Dhanda G, Sarkar P, Samaddar S, Haldar J. Battle against Vancomycin-Resistant Bacteria: Recent Developments in Chemical Strategies. J Med Chem 2018; 62:3184-3205. [DOI: 10.1021/acs.jmedchem.8b01093] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Geetika Dhanda
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Paramita Sarkar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Sandip Samaddar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
58
|
Azuma Y, Imai H, Kawaguchi Y, Nakase I, Kimura H, Futaki S. Modular Redesign of a Cationic Lytic Peptide To Promote the Endosomal Escape of Biomacromolecules. Angew Chem Int Ed Engl 2018; 57:12771-12774. [DOI: 10.1002/anie.201807534] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Yusuke Azuma
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | - Haruka Imai
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | | | - Ikuhiko Nakase
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | - Hiroshi Kimura
- Institute of Innovative Research; Tokyo Institute of Technology; 4259 Nagatsuta-cho, Midori-ku Yokohama 226-8503 Japan
| | - Shiroh Futaki
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| |
Collapse
|
59
|
Azuma Y, Imai H, Kawaguchi Y, Nakase I, Kimura H, Futaki S. Modular Redesign of a Cationic Lytic Peptide To Promote the Endosomal Escape of Biomacromolecules. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201807534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Yusuke Azuma
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | - Haruka Imai
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | | | - Ikuhiko Nakase
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| | - Hiroshi Kimura
- Institute of Innovative Research; Tokyo Institute of Technology; 4259 Nagatsuta-cho, Midori-ku Yokohama 226-8503 Japan
| | - Shiroh Futaki
- Institute for Chemical Research; Kyoto University; Uji Kyoto 611-0011 Japan
| |
Collapse
|
60
|
Abdalla MA, McGaw LJ. Natural Cyclic Peptides as an Attractive Modality for Therapeutics: A Mini Review. Molecules 2018; 23:molecules23082080. [PMID: 30127265 PMCID: PMC6222632 DOI: 10.3390/molecules23082080] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/14/2018] [Accepted: 08/19/2018] [Indexed: 01/04/2023] Open
Abstract
Peptides are important biomolecules which facilitate the understanding of complex biological processes, which in turn could be serendipitous biological targets for future drugs. They are classified as a unique therapeutic niche and will play an important role as fascinating agents in the pharmaceutical landscape. Until now, more than 40 cyclic peptide drugs are currently in the market, and approximately one new cyclopeptide drug enters the market annually on average. Interestingly, the majority of clinically approved cyclic peptides are derived from natural sources, such as peptide antibiotics and human peptide hormones. In this report, the importance of cyclic peptides is discussed, and their role in drug discovery as interesting therapeutic biomolecules will be highlighted. Recently isolated naturally occurring cyclic peptides from microorganisms, sponges, and other sources with a wide range of pharmacological properties are reviewed herein.
Collapse
Affiliation(s)
- Muna Ali Abdalla
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa.
- Department of Food Science and Technology, Faculty of Agriculture, University of Khartoum, Khartoum North 13314, Sudan.
| | - Lyndy J McGaw
- Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa.
| |
Collapse
|
61
|
Guan D, Chen F, Liu J, Li J, Lan L, Huang W. Design and Synthesis of Pyrophosphate-Targeting Vancomycin Derivatives for Combating Vancomycin-Resistant Enterococci. ChemMedChem 2018; 13:1644-1657. [PMID: 29920964 DOI: 10.1002/cmdc.201800252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/13/2018] [Indexed: 12/12/2022]
Abstract
As the last resort for intractable Gram-positive bacterial infections, vancomycin is losing efficacy with the emergence of vancomycin-resistant bacteria, especially vancomycin-resistant Enterococci (VRE). To combat this threat, we rationally designed and synthesized 39 novel vancomycin derivatives by respective or combined modifications using metal-chelating, lipophilic, and galactose-attachment strategies for extensive structure-activity relationship (SAR) analysis. In a proposed mechanism, the conjugation of dipicolylamine on the seventh amino acid resorcinol position or C-terminus endowed the vancomycin backbone with binding capacity for the pyrophosphate moiety in lipid II while maintaining the intrinsic binding affinity for the dipeptide terminus of the bacterial cell wall peptidoglycan precursor. The in vitro antibacterial activities were evaluated, and the optimal compounds indicated 16- to 1024-fold higher activity against VRE than that of vancomycin. Compound 11 b (3',5'-bis(dipicolylaminomethyl)tyrosine [1,2,3]triazolylmethoxylethyoxyl ethylaminomethyl-N-decylvancomycin) was found to have particularly potent activity against VRE through synergistic effects brought about by combining two peripheral modifications.
Collapse
Affiliation(s)
- Dongliang Guan
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P.R. China
| | - Feifei Chen
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
| | - Junjie Liu
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, P.R. China
| | - Jian Li
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P.R. China
| | - Lefu Lan
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P.R. China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P.R. China
| | - Wei Huang
- CAS Key Laboratory of Receptor Research, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, P.R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, P.R. China
- Center for Biotherapeutics Discovery Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P.R. China
| |
Collapse
|
62
|
Yarlagadda V, Sarkar P, Samaddar S, Manjunath GB, Mitra SD, Paramanandham K, Shome BR, Haldar J. Vancomycin Analogue Restores Meropenem Activity against NDM-1 Gram-Negative Pathogens. ACS Infect Dis 2018; 4:1093-1101. [PMID: 29726673 DOI: 10.1021/acsinfecdis.8b00011] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
New Delhi metallo-β-lactamase-1 (NDM-1) is the major contributor to the emergence of carbapenem resistance in Gram-negative pathogens (GNPs) and has caused many clinically available β-lactam antibiotics to become obsolete. A clinically approved inhibitor of metallo-β-lactamase (MBL) that could restore the activity of carbapenems against resistant GNPs has not yet been found, making NDM-1 a serious threat to human health. Here, we have rationally developed an inhibitor for the NDM-1 enzyme, which has the ability to penetrate the outer membrane of GNPs and inactivate the enzyme by depleting the metal ion (Zn2+) from the active site. The inhibitor reinstated the activity of meropenem against NDM-1 producing clinical isolates of GNPs like Klebsiella pneumoniae and Escherichia coli. Further, the inhibitor efficiently restored meropenem activity against NDM-1 producing K. pneumoniae in a murine sepsis infection model. These findings demonstrate that a combination of the present inhibitor and meropenem has high potential to be translated clinically to combat carbapenem-resistant GNPs.
Collapse
Affiliation(s)
- Venkateswarlu Yarlagadda
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Paramita Sarkar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sandip Samaddar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Goutham Belagula Manjunath
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Susweta Das Mitra
- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru, Karnataka 560064, India
| | - Krishnamoorthy Paramanandham
- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru, Karnataka 560064, India
| | - Bibek Ranjan Shome
- National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru, Karnataka 560064, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| |
Collapse
|
63
|
Bai PY, Qin SS, Chu WC, Yang Y, Cui DY, Hua YG, Yang QQ, Zhang E. Synthesis and antibacterial bioactivities of cationic deacetyl linezolid amphiphiles. Eur J Med Chem 2018; 155:925-945. [DOI: 10.1016/j.ejmech.2018.06.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/10/2018] [Accepted: 06/22/2018] [Indexed: 10/28/2022]
|
64
|
Abstract
![]()
Glycopeptide
antibiotics (GPAs) are a key weapon in the fight against drug resistant
bacteria, with vancomycin still a mainstream therapy against serious
Gram-positive infections more than 50 years after it was first introduced.
New, more potent semisynthetic derivatives that have entered the clinic,
such as dalbavancin and oritavancin, have superior pharmacokinetic
and target engagement profiles that enable successful treatment of
vancomycin-resistant infections. In the face of resistance development,
with multidrug resistant (MDR) S. pneumoniae and methicillin-resistant Staphylococcus aureus (MRSA) together causing 20-fold more infections than all MDR Gram-negative
infections combined, further improvements are desirable to ensure
the Gram-positive armamentarium is adequately maintained for future
generations. A range of modified glycopeptides has been generated
in the past decade via total syntheses, semisynthetic modifications
of natural products, or biological engineering. Several of these
have undergone extensive characterization with demonstrated in vivo efficacy, good PK/PD profiles, and no reported preclinical
toxicity; some may be suitable for formal preclinical development.
The natural product monobactam, cephalosporin, and β-lactam
antibiotics all spawned multiple generations of commercially and clinically
successful semisynthetic derivatives. Similarly, next-generation glycopeptides
are now technically well positioned to advance to the clinic, if sufficient
funding and market support returns to antibiotic development.
Collapse
Affiliation(s)
- Mark A. T. Blaskovich
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Karl A. Hansford
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Mark S. Butler
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| | - ZhiGuang Jia
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Alan E. Mark
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane, Queensland 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Chemistry Building 68, Cooper Road, Brisbane, Queensland 4072, Australia
| |
Collapse
|
65
|
Amato DV, Amato DN, Blancett LT, Mavrodi OV, Martin WB, Swilley SN, Sandoz MJ, Shearer G, Mavrodi DV, Patton DL. A bio-based pro-antimicrobial polymer network via degradable acetal linkages. Acta Biomater 2018; 67:196-205. [PMID: 29269331 PMCID: PMC6064185 DOI: 10.1016/j.actbio.2017.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/28/2017] [Accepted: 12/12/2017] [Indexed: 11/19/2022]
Abstract
The synthesis of a fully degradable, bio-based, sustained release, pro-antimicrobial polymer network comprised of degradable acetals (PANDA) is reported. The active antimicrobial agent - p-anisaldehyde (pA) (an extract from star anise) - was converted into a UV curable acetal containing pro-antimicrobial monomer and subsequently photopolymerized into a homogenous thiol-ene network. Under neutral to acidic conditions (pH < 8), the PANDAs undergo surface erosion and exhibit sustained release of pA over 38 days. The release of pA from PANDAs was shown to be effective against both bacterial and fungal pathogens. From a combination of confocal microscopy and transmission electron microscopy, we observed that the released pA disrupts the cell membrane. Additionally, we demonstrated that PANDAs have minimal cytotoxicity towards both epithelial cells and macrophages. Although a model platform, these results point to promising pathways for the design of fully degradable sustained-release antimicrobial systems with potential applications in agriculture, pharmaceuticals, cosmetics, household/personal care, and food industries. STATEMENT OF SIGNIFICANCE With the increasing number of patients prescribed immunosuppressants coupled with the rise in antibiotic resistance - life-threatening microbial infections are a looming global threat. With limited success within the antibiotic pipeline, nature-based essential oils (EOs) are being investigated for their multimodal effectiveness against microbes. Despite the promising potential of EOs, difficulties in their encapsulation, limited water solubility, and high volatility limit their use. Various studies have shown that covalent attachment of these EO derivatives to polymers can mitigate these limitations. The current study presents the synthesis of a fully-degradable, sustained release, cytocompatible, pro-antimicrobial acetal network derived from p-anisaldehyde. This polymer network design provides a pathway toward application-specific EO releasing materials with quantitative encapsulation efficiencies, sustained release, and broad-spectrum antimicrobial activity.
Collapse
Affiliation(s)
- Douglas V Amato
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Dahlia N Amato
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Logan T Blancett
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Olga V Mavrodi
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - William B Martin
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Sarah N Swilley
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Michael J Sandoz
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Glenmore Shearer
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Dmitri V Mavrodi
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Derek L Patton
- School of Polymer Science and Engineering, The University of Southern Mississippi, Hattiesburg, MS 39406, United States.
| |
Collapse
|
66
|
Guan D, Chen F, Xiong L, Tang F, Faridoon, Qiu Y, Zhang N, Gong L, Li J, Lan L, Huang W. Extra Sugar on Vancomycin: New Analogues for Combating Multidrug-Resistant Staphylococcus aureus and Vancomycin-Resistant Enterococci. J Med Chem 2018; 61:286-304. [DOI: 10.1021/acs.jmedchem.7b01345] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Dongliang Guan
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | | | - Lun Xiong
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Tang
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | | | - Yunguang Qiu
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Naixia Zhang
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Likun Gong
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jian Li
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lefu Lan
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Wei Huang
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
67
|
Xu Q, Li X, Jin Y, Sun L, Ding X, Liang L, Wang L, Nan K, Ji J, Chen H, Wang B. Bacterial self-defense antibiotics release from organic-inorganic hybrid multilayer films for long-term anti-adhesion and biofilm inhibition properties. NANOSCALE 2017; 9:19245-19254. [PMID: 29188848 DOI: 10.1039/c7nr07106j] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Implant-associated bacterial infections pose serious medical and financial issues due to the colonization and proliferation of pathogens on the surface of the implant. The as-prepared traditional antibacterial surfaces can neither resist bacterial adhesion nor inhibit the development of biofilm over the long term. Herein, novel (montmorillonite/poly-l-lysine-gentamicin sulfate)8 ((MMT/PLL-GS)8) organic-inorganic hybrid multilayer films were developed to combine enzymatic degradation PLL for on-demand self-defense antibiotics release. Small molecule GS was loaded into the multilayer films during self-assembly and the multilayer films showed pH-dependent and linear growth behavior. The chymotrypsin- (CMS) and bacterial infections-responsive film degradation led to the peeling of the films and GS release. Enzyme-responsive GS release exhibited CMS concentration dependence as measured by the size of the inhibition zone and SEM images. Notably, the obtained antibacterial films showed highly efficient bactericidal activity which killed more than 99.9% of S. aureus in 12 h. Even after 3 d of incubation in S. aureus, E. coli or S. epidermidis solutions, the multilayer films exhibited inhibition zones of more than 1.5 mm in size. Both in vitro and in vivo antibacterial tests indicated good cell compatibility, and anti-inflammatory, and long-term bacterial anti-adhesion and biofilm inhibition properties.
Collapse
Affiliation(s)
- Qingwen Xu
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Zhang E, Bai PY, Cui DY, Chu WC, Hua YG, Liu Q, Yin HY, Zhang YJ, Qin S, Liu HM. Synthesis and bioactivities study of new antibacterial peptide mimics: The dialkyl cationic amphiphiles. Eur J Med Chem 2017; 143:1489-1509. [PMID: 29126736 DOI: 10.1016/j.ejmech.2017.10.044] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/28/2017] [Accepted: 10/15/2017] [Indexed: 12/11/2022]
Abstract
The emergence of infectious diseases caused by pathogenic bacteria is widespread. Therefore, it is urgently required to enhance the development of novel antimicrobial agents with high antibacterial activity and low cytotoxicity. A series of novel dialkyl cationic amphiphiles bearing two identical length lipophilic alkyl chains and one non-peptidic amide bond were synthesized and tested for antimicrobial activities against both Gram-positive and Gram-negative bacteria. Particular compounds synthesized showed excellent antibacterial activity toward drug-sensitive bacteria such as S. aureus, E. faecalis, E. coli and S. enterica, and clinical isolates of drug-resistant species such as methicillin-resistant S. aureus (MRSA), KPC-producing and NDM-1-producing carbapenem-resistant Enterobacteriaceae (CRE). For example, the MIC values of the best compound 4g ranged from 0.5 to 2 μg/mL against all these strains. Moreover, these small molecules acted rapidly as bactericidal agents, and functioned primarily by permeabilization and depolarization of bacterial membranes. Importantly, these compounds were difficult to induce bacterial resistance and can potentially combat drug-resistant bacteria. Thus, these compounds can be developed into a new class of antibacterial peptide mimics against Gram-positive and Gram-negative bacteria, including drug-resistant bacterial strains.
Collapse
Affiliation(s)
- En Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, PR China.
| | - Peng-Yan Bai
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - De-Yun Cui
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Wen-Chao Chu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yong-Gang Hua
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Qin Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Hai-Yang Yin
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yong-Jie Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, PR China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, PR China.
| |
Collapse
|
69
|
Challa C, Ravindran J, Konai MM, Varughese S, Jacob J, Kumar BSD, Haldar J, Lankalapalli RS. Expedient Synthesis of Indolo[2,3- b]quinolines, Chromeno[2,3- b]indoles, and 3-Alkenyl-oxindoles from 3,3'-Diindolylmethanes and Evaluation of Their Antibiotic Activity against Methicillin-Resistant Staphylococcus aureus. ACS OMEGA 2017; 2:5187-5195. [PMID: 30023741 PMCID: PMC6044809 DOI: 10.1021/acsomega.7b00840] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/16/2017] [Indexed: 05/09/2023]
Abstract
Easily accessible 3,3'-diindolylmethanes (DIMs) were utilized to generate a focused library of indolo[2,3-b]quinolines (2), chromeno[2,3-b]indoles (3), and 3-alkenyl-oxindoles (4) under 2,3-Dichloro-5,6-dicyano-1,4-benzoquinone (DDQ)-mediated oxidative conditions. DIMs with ortho-NHTosyl (NHTs) phenyl group afforded indolo[2,3-b]quinolines (2), whereas DIMs with ortho-hydroxy phenyl groups yielded chromeno[2,3-b]indoles (3) and 3-alkenyl-oxindoles (4). The mild conditions and excellent yields of the products make this method a good choice to access a diverse library of bioactive molecules from a common starting material. Two optimized compounds 2a and 2n displayed excellent activity against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA). Compound 2a showed the minimum inhibitory concentration values in the concentration between 1 and 4 μg/mL, whereas compound 2n revealed the values of 1-2 μg/mL. Furthermore, both the compounds were highly bactericidal and capable to kill the MRSA completely within 360 min. Collectively, the results suggested that both compounds 2a and 2n possess enormous potential to be developed as anti-MRSA agents.
Collapse
Affiliation(s)
- Chandrasekhar Challa
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jaice Ravindran
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Mohini Mohan Konai
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Sunil Varughese
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jubi Jacob
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - B. S. Dileep Kumar
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Ravi S. Lankalapalli
- Chemical
Sciences and Technology Division and Academy of Scientific
and Innovative Research (AcSIR) and Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science
and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
- E-mail:
| |
Collapse
|
70
|
Liu YW, Shia KS, Wu CH, Liu KL, Yeh YC, Lo CF, Chen CT, Chen YY, Yeh TK, Chen WH, Jan JJ, Huang YC, Huang CL, Fang MY, Gray BD, Pak KY, Hsu TA, Huang KH, Tsou LK. Targeting Tumor Associated Phosphatidylserine with New Zinc Dipicolylamine-Based Drug Conjugates. Bioconjug Chem 2017; 28:1878-1892. [PMID: 28581724 DOI: 10.1021/acs.bioconjchem.7b00225] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A series of zinc(II) dipicolylamine (ZnDPA)-based drug conjugates have been synthesized to probe the potential of phosphatidylserine (PS) as a new antigen for small molecule drug conjugate (SMDC) development. Using in vitro cytotoxicity and plasma stability studies, PS-binding assay, in vivo pharmacokinetic studies, and maximum tolerated dose profiles, we provided a roadmap and the key parameters required for the development of the ZnDPA based drug conjugate. In particular, conjugate 24 induced tumor regression in the COLO 205 xenograft model and exhibited a more potent antitumor effect with a 70% reduction of cytotoxic payload compared to that of the marketed irinotecan when dosed at the same regimen. In addition to the validation of PS as an effective pharmacodelivery target for SMDC, our work also provided the foundation that, if applicable, a variety of therapeutic agents could be conjugated in the same manner to treat other PS-associated diseases.
Collapse
Affiliation(s)
- Yu-Wei Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chien-Huang Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yu-Cheng Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yun-Yu Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Wei-Han Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Jiing-Jyh Jan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Brian D Gray
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kuan-Hsun Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Lun K Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| |
Collapse
|
71
|
Lin S, Liu X, Tan L, Cui Z, Yang X, Yeung KWK, Pan H, Wu S. Porous Iron-Carboxylate Metal-Organic Framework: A Novel Bioplatform with Sustained Antibacterial Efficacy and Nontoxicity. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19248-19257. [PMID: 28558188 DOI: 10.1021/acsami.7b04810] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Sustained drug release plays a critical role in targeting the therapy of local diseases such as bacterial infections. In the present work, porous iron-carboxylate metal-organic framework [MOF-53(Fe)] nanoparticles (NPs) were designed to entrap the vancomycin (Van) drugs. This system exhibited excellent chemical stability under acidic conditions (pH 7.4, 6.5, and 5.5) and much higher drug-loading capability because of the high porosity and large surface area of MOF NPs. The results showed that the drug-loading ratio of Van could reach 20 wt % and that the antibacterial ratio of the MOF-53(Fe)/Van system against Staphylococcus aureus could reach up to 90%. In addition, this MOF-53(Fe)/Van system exhibited excellent biocompatibility because of its chemical stability and sustained release of iron ions. Hence, these porous MOF NPs are a promising bioplatform not only for local therapy of bacterial infections but also for other biomedical therapies for tissue regeneration.
Collapse
Affiliation(s)
- Sha Lin
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University , Wuhan 430062, China
| | - Xiangmei Liu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University , Wuhan 430062, China
| | - Lei Tan
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University , Wuhan 430062, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Xianjin Yang
- School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| | - Kelvin W K Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong 999077, Pokfulam, Hong Kong, China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055, China
| | - Shuilin Wu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University , Wuhan 430062, China
- School of Materials Science & Engineering, Tianjin University , Tianjin 300072, China
| |
Collapse
|
72
|
Rice DR, de Lourdes Betancourt Mendiola M, Murillo-Solano C, Checkley LA, Ferdig MT, Pizarro JC, Smith BD. Antiplasmodial activity of targeted zinc(II)-dipicolylamine complexes. Bioorg Med Chem 2017; 25:2754-2760. [PMID: 28377170 DOI: 10.1016/j.bmc.2017.03.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 12/29/2022]
Abstract
This study measured the antiplasmodial activity of nine zinc-dipicolylamine (ZnDPA) complexes against three strains of Plasmodium falciparum, the causative parasite of malaria. Growth inhibition assays showed significant activity against all tested strains, with 50% inhibitory concentrations between 5 and 600nM and almost no toxic effect against host cells including healthy red blood cells. Fluorescence microscopy studies with a green-fluorescent ZnDPA probe showed selective targeting of infected red blood cells. The results suggest that ZnDPA coordination complexes are promising antiplasmodial agents with potential for targeted malaria treatment.
Collapse
Affiliation(s)
- Douglas R Rice
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Claribel Murillo-Solano
- Department of Tropical Medicine, J Bennett Johnston Building, 1430 Tulane Avenue, Tulane University, New Orleans, LA 70112, USA
| | - Lisa A Checkley
- Department of Biological Science, Galvin Life Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael T Ferdig
- Department of Biological Science, Galvin Life Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, J Bennett Johnston Building, 1430 Tulane Avenue, Tulane University, New Orleans, LA 70112, USA
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, 236 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
73
|
Mao C, Xie X, Liu X, Cui Z, Yang X, Yeung KWK, Pan H, Chu PK, Wu S. The controlled drug release by pH-sensitive molecularly imprinted nanospheres for enhanced antibacterial activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 77:84-91. [PMID: 28532100 DOI: 10.1016/j.msec.2017.03.259] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 11/16/2022]
Abstract
In this study, we prepared pH-sensitive hybrid nanospheres through the implementation of a facile molecularly imprinted polymer (MIP) technique combined with a UV-initiated precipitation polymerization method using vancomycin (VA) for the templates. During the course of this investigation, both 2-hydroxyethyl methacrylate (HEMA) and 2-(diethylamino) ethyl methacrylate (DEAEMA) were utilized as the functional monomers, while ethylene glycol dimethacrylate (EGDMA) was used as a cross-linker. The obtained MIP nanospheres exhibited well-controlled particle size, with a drug loading capacity of about 17%, much higher than that of the non-imprinted polymer (NIP) nanospheres (5%). In addition, the VA loading quantity was closely correlated with the dosage of the cross-linking agent, and the MIP nanospheres exhibited a slower and more controlled VA release rate than the NIP nanospheres. Moreover, these MIP nanospheres were sensitive to pH values, and consequently showed an increasing release rate of VA as the pH level was decreased. The VA-loaded MIP nanospheres showed the higher antibacterial ratio of over 92% against Staphylococcus aureus (S. aureus) while the NIP nanospheres were friendly to S. aureus. These MIP nanospheres can be promising for targeting drug delivery system to achieve specific therapies such as preventing bacterial infections and killing cancer cells without damaging health cells and tissues.
Collapse
Affiliation(s)
- Congyang Mao
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Xianzhou Xie
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Xiangmei Liu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, Tianjin University, Tianjin 300072, China
| | - Xianjin Yang
- School of Materials Science & Engineering, Tianjin University, Tianjin 300072, China
| | - K W K Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Paul K Chu
- Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Shuilin Wu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China; School of Materials Science & Engineering, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
74
|
Sarkar P, Yarlagadda V, Ghosh C, Haldar J. A review on cell wall synthesis inhibitors with an emphasis on glycopeptide antibiotics. MEDCHEMCOMM 2017; 8:516-533. [PMID: 30108769 DOI: 10.1039/c6md00585c] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/18/2017] [Indexed: 01/24/2023]
Abstract
Cell wall biosynthesis inhibitors (CBIs) have historically been one of the most effective classes of antibiotics. They are the most extensively used class of antibiotics and their importance is exemplified by the β-lactams and glycopeptide antibiotics. However, this class of antibiotics has not received impunity from resistance development. In the wake of this predicament, this review presents the progress of CBIs, especially glycopeptide derivatives as antibiotics to confront antibacterial resistance. The various strategies used for the development of CBIs, their clinical status and possible directions in which this field can evolve have also been discussed.
Collapse
Affiliation(s)
- Paramita Sarkar
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Venkateswarlu Yarlagadda
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Chandradhish Ghosh
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Jayanta Haldar
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| |
Collapse
|
75
|
Hassan MM, Ranzoni A, Phetsang W, Blaskovich MAT, Cooper MA. Surface Ligand Density of Antibiotic-Nanoparticle Conjugates Enhances Target Avidity and Membrane Permeabilization of Vancomycin-Resistant Bacteria. Bioconjug Chem 2016; 28:353-361. [DOI: 10.1021/acs.bioconjchem.6b00494] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Marwa M. Hassan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrea Ranzoni
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Wanida Phetsang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mark A. T. Blaskovich
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
76
|
Santos-Beneit F, Ordóñez-Robles M, Martín JF. Glycopeptide resistance: Links with inorganic phosphate metabolism and cell envelope stress. Biochem Pharmacol 2016; 133:74-85. [PMID: 27894856 DOI: 10.1016/j.bcp.2016.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
Abstract
Antimicrobial resistance is a critical health issue today. Many pathogens have become resistant to many or all available antibiotics and limited new antibiotics are in the pipeline. Glycopeptides are used as a 'last resort' antibiotic treatment for many bacterial infections, but worryingly, glycopeptide resistance has spread to very important pathogens such as Enterococcus faecium and Staphylococcus aureus. Bacteria confront multiple stresses in their natural environments, including nutritional starvation and the action of cell-wall stressing agents. These stresses impact bacterial susceptibility to different antimicrobials. This article aims to review the links between glycopeptide resistance and different stresses, especially those related with cell-wall biosynthesis and inorganic phosphate metabolism, and to discuss promising alternatives to classical antibiotics to avoid the problem of antimicrobial resistance.
Collapse
Affiliation(s)
- Fernando Santos-Beneit
- Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Medical School, Newcastle University, NE2 4AX Newcastle upon Tyne, UK
| | - María Ordóñez-Robles
- Department of Biotechnology, Faculty of Natural Sciences and Technology, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Juan F Martín
- Microbiology Area, Department of Molecular Biology, University of León, 24071 León, Spain.
| |
Collapse
|
77
|
Khalifa L, Shlezinger M, Beyth S, Houri-Haddad Y, Coppenhagen-Glazer S, Beyth N, Hazan R. Phage therapy against Enterococcus faecalis in dental root canals. J Oral Microbiol 2016; 8:32157. [PMID: 27640530 PMCID: PMC5027333 DOI: 10.3402/jom.v8.32157] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 12/16/2022] Open
Abstract
Antibiotic resistance is an ever-growing problem faced by all major sectors of health care, including dentistry. Recurrent infections related to multidrug-resistant bacteria such as methicillin-resistant Staphylococcus aureus, carbapenem-resistant Enterobacteriaceae, and vancomycin-resistant enterococci (VRE) in hospitals are untreatable and question the effectiveness of notable drugs. Two major reasons for these recurrent infections are acquired antibiotic resistance genes and biofilm formation. None of the traditionally known effective techniques have been able to efficiently resolve these issues. Hence, development of a highly effective antibacterial practice has become inevitable. One example of a hard-to-eradicate pathogen in dentistry is Enterococcus faecalis, which is one of the most common threats observed in recurrent root canal treatment failures, of which the most problematic to treat are its biofilm-forming VRE strains. An effective response against such infections could be the use of bacteriophages (phages). Phage therapy was found to be highly effective against biofilm and multidrug-resistant bacteria and has other advantages like ease of isolation and possibilities for genetic manipulations. The potential of phage therapy in dentistry, in particular against E. faecalis biofilms in root canals, is almost unexplored. Here we review the efforts to develop phage therapy against biofilms. We also focus on the phages isolated against E. faecalis and discuss the possibility of using phages against E. faecalis biofilm in root canals.
Collapse
Affiliation(s)
- Leron Khalifa
- Institute of Dental Science, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Mor Shlezinger
- Department of Prosthodontics, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Shaul Beyth
- Orthopedic Surgery Complex, Hadassah University Hospital, Jerusalem, Israel
| | - Yael Houri-Haddad
- Department of Prosthodontics, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Shunit Coppenhagen-Glazer
- Institute of Dental Science, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Nurit Beyth
- Department of Prosthodontics, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Ronen Hazan
- Institute of Dental Science, The Hebrew University Hadassah School of Dental Medicine, Jerusalem, Israel;
| |
Collapse
|