1
|
Gao L, Wu H, Feng J, Liu Y, Wang R, Yan L, Lv Q, Jiang Y. In vitro and in vivo activity of 1,2,3,4,6-O-pentagalloyl-glucose against Candida albicans. Antimicrob Agents Chemother 2025; 69:e0177524. [PMID: 39853121 PMCID: PMC11881577 DOI: 10.1128/aac.01775-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/26/2025] Open
Abstract
Invasive fungal infections have become an increasingly serious threat to global human health, underscoring the urgent need for the development of new antifungal drugs. In this study, we found a natural polyphenolic compound 1,2,3,4,6-O-pentagalloyl-glucose (PGG), which is present in various plants and herbs. PGG showed broad-spectrum antifungal activities, enhancing the efficacy of fluconazole. Furthermore, PGG could protect mice against gastrointestinal and systemic infection with Candida albicans. Our mechanistic studies revealed that PGG exerts its antifungal effects partially by binding to the CaEno1 protein to inhibit its activity. As a crucial therapeutic target, Eno1 has been reported to be closely associated with cancer, hypertension, and infectious diseases. Our findings indicated that PGG, a new Eno1 inhibitor, is a potential candidate for further antifungal development.
Collapse
Affiliation(s)
- Lu Gao
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Wu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Feng
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Ruina Wang
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Lan Yan
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Quanzhen Lv
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Palma F, Acunzo M, Della Marca R, Dell'Annunziata F, Folliero V, Chianese A, Zannella C, Franci G, De Filippis A, Galdiero M. Evaluation of antifungal spectrum of Cupferron against Candida albicans. Microb Pathog 2024; 194:106835. [PMID: 39117014 DOI: 10.1016/j.micpath.2024.106835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Candida albicans is an opportunistic yeast accounting for about 50-90 % of all cases of candidiasis in humans, ranging from superficial to systemic potentially life-threatening infections. The presence of several virulence factors, including biofilm, hyphal transition, and proteolytic enzymes production, worsens the fungal infections burden on healthcare system resources. Hence, developing new bioactive compounds with antifungal activity is a pressing urgence for the scientific community. In this perspective, we evaluated the anti-Candida potential of the N-Nitroso-N-phenylhydroxylamine ammonium salt (cupferron) against standard and clinical C. albicans strains. Firstly, the in vitro cytotoxicity of cupferron was checked in the range 400-12.5 μg/mL against human microglial cells (HMC-3). Secondly, its antifungal spectrum was explored via disk diffusion test, broth-microdilution method, and time-killing curve analysis, validating the obtained results through scanning electron microscopy (SEM) observations. Additionally, we evaluated the cupferron impact on the main virulence determinants of Candida albicans. At non-toxic concentrations (100-12.5 μg/mL), the compound exerted interesting anti-Candida activity, registering a minimum inhibitory concentration (MIC) between 50 and 100 μg/mL against the tested strains, with a fungistatic effect until 100 μg/mL. Furthermore, cupferron was able to counteract fungal virulence at MIC and sub-MIC values (50-12.5 μg/mL). These findings may propose cupferron as a new potential antifungal option for the treatment of Candida albicans infections.
Collapse
Affiliation(s)
- Francesca Palma
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Marina Acunzo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Roberta Della Marca
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Federica Dell'Annunziata
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Veronica Folliero
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Italy
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081, Baronissi, Italy
| | - Anna De Filippis
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
| |
Collapse
|
3
|
Dib Ferreira Gremião I, Pereira-Oliveira GR, Pereira SA, Corrêa ML, Borba-Santos LP, Viçosa AL, Garg A, Haranahalli K, Dasilva D, Pereira de Sa N, Matos GS, Silva V, Lazzarini C, Fernandes CM, Miranda K, Artunduaga Bonilla JJ, Nunes AL, Nimrichter L, Ojima I, Mallamo J, McCarthy JB, Del Poeta M. Combination therapy of itraconazole and an acylhydrazone derivative (D13) for the treatment of sporotrichosis in cats. Microbiol Spectr 2024; 12:e0396723. [PMID: 38647345 PMCID: PMC11237696 DOI: 10.1128/spectrum.03967-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
Acylhydrazone (AH) derivatives represent a novel category of anti-fungal medications that exhibit potent activity against Sporothrix sp., both in vitro and in a murine model of sporotrichosis. In this study, we demonstrated the anti-fungal efficacy of the AH derivative D13 [4-bromo-N'-(3,5-dibromo-2-hydroxybenzylidene)-benzohydrazide] against both planktonic cells and biofilms formed by Sporothrix brasiliensis. In a clinical study, the effect of D13 was then tested in combination with itraconazole (ITC), with or without potassium iodide, in 10 cats with sporotrichosis refractory to the treatment of standard of care with ITC. Improvement or total clinical cure was achieved in five cases after 12 weeks of treatment. Minimal abnormal laboratory findings, e.g., elevation of alanine aminotransferase, were observed in four cats during the combination treatment and returned to normal level within a week after the treatment was ended. Although highly encouraging, a larger and randomized controlled study is required to evaluate the effectiveness and the safety of this new and exciting drug combination using ITC and D13 for the treatment of feline sporotrichosis. IMPORTANCE This paper reports the first veterinary clinical study of an acylhydrazone anti-fungal (D13) combined with itraconazole against a dimorphic fungal infection, sporotrichosis, which is highly endemic in South America in animals and humans. Overall, the results show that the combination treatment was efficacious in ~50% of the infected animals. In addition, D13 was well tolerated during the course of the study. Thus, these results warrant the continuation of the research and development of this new class of anti-fungals.
Collapse
Affiliation(s)
- Isabella Dib Ferreira Gremião
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Gabriela Reis Pereira-Oliveira
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Sandro Antonio Pereira
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Maria Lopes Corrêa
- Laboratory of Clinical Research on Dermatozoonoses in Domestic Animals, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | | | - Alessandra Lifsitch Viçosa
- Laboratory of Experimental Pharmacotechnics, Institute of Drug Technology – Farmanguinhos, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Ashna Garg
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, USA
| | - Krupanandan Haranahalli
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, USA
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Deveney Dasilva
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Gabriel S. Matos
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Vanessa Silva
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Cristina Lazzarini
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Caroline Mota Fernandes
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Kildare Miranda
- Laboratory of Cellular Ultrastructure Hertha Meyer, Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jhon Jhamilton Artunduaga Bonilla
- Laboratory of Eukaryotic Glycobiology (LaGE), Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Letícia Nunes
- Laboratory of Eukaryotic Glycobiology (LaGE), Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Laboratory of Eukaryotic Glycobiology (LaGE), Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, USA
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - John Mallamo
- MicroRid Technologies Inc., Dix Hills, New York, USA
| | | | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- MicroRid Technologies Inc., Dix Hills, New York, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Veterans Administration Medical Center, Northport, New York, USA
| |
Collapse
|
4
|
Ahmady L, Gothwal M, Mukkoli MM, Bari VK. Antifungal drug resistance in Candida: a special emphasis on amphotericin B. APMIS 2024; 132:291-316. [PMID: 38465406 DOI: 10.1111/apm.13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Invasive fungal infections in humans caused by several Candida species, increased considerably in immunocompromised or critically ill patients, resulting in substantial morbidity and mortality. Candida albicans is the most prevalent species, although the frequency of these organisms varies greatly according to geographic region. Infections with C. albicans and non-albicans Candida species have become more common, especially in the past 20 years, as a result of aging, immunosuppressive medication use, endocrine disorders, malnourishment, extended use of medical equipment, and an increase in immunogenic diseases. Despite C. albicans being the species most frequently associated with human infections, C. glabrata, C. parapsilosis, C. tropicalis, and C. krusei also have been identified. Several antifungal drugs with different modes of action are approved for use in clinical settings to treat fungal infections. However, due to the common eukaryotic structure of humans and fungi, only a limited number of antifungal drugs are available for therapeutic use. Furthermore, drug resistance in Candida species has emerged as a result of the growing use of currently available antifungal drugs against fungal infections. Amphotericin B (AmB), a polyene class of antifungal drugs, is mainly used for the treatment of serious systemic fungal infections. AmB interacts with fungal plasma membrane ergosterol, triggering cellular ion leakage via pore formation, or extracting the ergosterol from the plasma membrane inducing cellular death. AmB resistance is primarily caused by changes in the content or structure of ergosterol. This review summarizes the antifungal drug resistance exhibited by Candida species, with a special focus on AmB.
Collapse
Affiliation(s)
- Lailema Ahmady
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | - Manisha Gothwal
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | | | - Vinay Kumar Bari
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
5
|
Xie F, Hao Y, Liu Y, Bao J, Wang R, Chi X, Wang T, Yu S, Jin Y, Li L, Jiang Y, Zhang D, Yan L, Ni T. From Synergy to Monotherapy: Discovery of Novel 2,4,6-Trisubstituted Triazine Hydrazone Derivatives with Potent Antifungal Potency In Vitro and In Vivo. J Med Chem 2024; 67:4007-4025. [PMID: 38381075 DOI: 10.1021/acs.jmedchem.3c02292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Invasive fungal infections pose a serious threat to public health and are associated with high mortality and incidence rates. The development of novel antifungal agents is urgently needed. Based on hit-to-lead optimization, a series of 2,4,6-trisubstituted triazine hydrazone compounds were designed, synthesized, and biological evaluation was performed, leading to the identification of compound 28 with excellent in vitro synergy (FICI range: 0.094-0.38) and improved monotherapy potency against fluconazole-resistant Candida albicans and Candida auris (MIC range: 1.0-16.0 μg/mL). Moreover, 28 exhibited broad-spectrum antifungal activity against multiple pathogenic strains. Furthermore, 28 could inhibit hyphal and biofilm formation, which may be related to its ability to disrupt the fungal cell wall. Additionally, 28 significantly reduced the CFU in a mouse model of disseminated infection with candidiasis at a dose of 10 mg/kg. Overall, the triazine-based hydrazone compound 28 with low cytotoxicity, hemolysis, and favorable ADME/T characteristics represents a promising lead to further investigation.
Collapse
Affiliation(s)
- Fei Xie
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Yu Liu
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Junhe Bao
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Ruina Wang
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Xiaochen Chi
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, No.103 Wenhua Road, Shenyang 110016, China
| | - Ting Wang
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Shichong Yu
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Yongsheng Jin
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai 200072, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai 200072, China
| | - Dazhi Zhang
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai 200072, China
| | - Lan Yan
- School of Pharmacy, The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), Naval Medical University, No.325 Guohe Road, Shanghai 200433, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No.1239 Siping Road, Shanghai 200072, China
| |
Collapse
|
6
|
Puumala E, Fallah S, Robbins N, Cowen LE. Advancements and challenges in antifungal therapeutic development. Clin Microbiol Rev 2024; 37:e0014223. [PMID: 38294218 PMCID: PMC10938895 DOI: 10.1128/cmr.00142-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Over recent decades, the global burden of fungal disease has expanded dramatically. It is estimated that fungal disease kills approximately 1.5 million individuals annually; however, the true worldwide burden of fungal infection is thought to be higher due to existing gaps in diagnostics and clinical understanding of mycotic disease. The development of resistance to antifungals across diverse pathogenic fungal genera is an increasingly common and devastating phenomenon due to the dearth of available antifungal classes. These factors necessitate a coordinated response by researchers, clinicians, public health agencies, and the pharmaceutical industry to develop new antifungal strategies, as the burden of fungal disease continues to grow. This review provides a comprehensive overview of the new antifungal therapeutics currently in clinical trials, highlighting their spectra of activity and progress toward clinical implementation. We also profile up-and-coming intracellular proteins and pathways primed for the development of novel antifungals targeting their activity. Ultimately, we aim to emphasize the importance of increased investment into antifungal therapeutics in the current continually evolving landscape of infectious disease.
Collapse
Affiliation(s)
- Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sara Fallah
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Sun Y, Kim S, Shin S, Takemura K, Matos GS, Lazzarini C, Haranahalli K, Zambito J, Garg A, Del Poeta M, Ojima I. SAR study of N'-(Salicylidene)heteroarenecarbohydrazides as promising antifungal agents. Bioorg Med Chem 2024; 100:117610. [PMID: 38306882 PMCID: PMC11984380 DOI: 10.1016/j.bmc.2024.117610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
Clinically available antifungal drugs have therapeutic limitations due to toxicity, narrow spectrum of activity, and intrinsic or acquired drug resistance. Thus, there is an urgent need for new broad-spectrum antifungal agents with low toxicity and a novel mechanism of action. In this context, we have successfully identified several highly promising lead compounds, i.e., aromatic N'-(salicylidene)carbohydrazides, exhibiting excellent antifungal activities against Cryptococcus neoformans, Candida albicans, Aspergillus fumigatus and several other fungi both in vitro and in vivo. Building upon these highly promising results, 71 novel N'-(salicylidene)heteroarenecarbohydrazides 5 were designed, synthesized and their antifungal activities examined against fungi. Based on the SAR study, four highly promising lead compounds, i.e., 5.6a, 5.6b, 5.7b and 5.13a were identified, which exhibited excellent potency against C. neoformans, C. albicans and A. fumigatus, and displayed impressive time-kill profiles against C. neoformans with exceptionally high selectivity indices (SI ≥ 500). These four lead compounds also showed synergy with clinical antifungal drugs, fluconazole, caspofungin (CS) and amphotericin B against C. neoformans. For the SAR study, we also employed quantitative structure-activity relationship (QSAR) analysis by taking advantage of the accumulated data on a large number of aromatic and heteroaromatic N'-(salicylidene)carbohydrazides, which successfully led to rational design and selection of promising compounds for chemical synthesis and biological evaluation.
Collapse
Affiliation(s)
- Yi Sun
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Saerom Kim
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - SeungYoun Shin
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Kathryn Takemura
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Gabriel S Matos
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794-5222, United States
| | - Cristina Lazzarini
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794-5222, United States; Veterans Administration Medical Center, Northport, NY 11768, United States
| | - Krupanandan Haranahalli
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Julia Zambito
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Ashna Garg
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, United States; Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794-5222, United States; Veterans Administration Medical Center, Northport, NY 11768, United States; Division of Infectious Diseases, School of Medicine, Stony Brook University, New York 11794-8434, United States
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, United States; Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, United States.
| |
Collapse
|
8
|
Vishwakarma M, Haider T, Soni V. Update on fungal lipid biosynthesis inhibitors as antifungal agents. Microbiol Res 2024; 278:127517. [PMID: 37863019 DOI: 10.1016/j.micres.2023.127517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/22/2023]
Abstract
Fungal diseases today represent a world-wide problem. Poor hygiene and decreased immunity are the main reasons behind the manifestation of this disease. After COVID-19, an increase in the rate of fungal infection has been observed in different countries. Different classes of antifungal agents, such as polyenes, azoles, echinocandins, and anti-metabolites, as well as their combinations, are currently employed to treat fungal diseases; these drugs are effective but can cause some side effects and toxicities. Therefore, the identification and development of newer antifungal agents is a current need. The fungal cell comprises many lipids, such as ergosterol, phospholipids, and sphingolipids. Ergosterol is a sterol lipid that is only found in fungal cells. Various pathways synthesize all these lipids, and the activities of multiple enzymes govern these pathways. Inhibiting these enzymes will ultimately impede the lipid synthesis pathway, and this phenomenon could be a potential antifungal therapy. This review will discuss various lipid synthesis pathways and multiple antifungal agents identified as having fungal lipid synthesis inhibition activity. This review will identify novel compounds that can inhibit fungal lipid synthesis, permitting researchers to direct further deep pharmacological investigation and help develop drug delivery systems for such compounds.
Collapse
Affiliation(s)
- Monika Vishwakarma
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, M.P., India
| | - Tanweer Haider
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, M.P., India; Amity Institute of Pharmacy, Amity University, Gwalior, M.P., India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, M.P., India.
| |
Collapse
|
9
|
Rollin-Pinheiro R, de Moraes DC, Bayona-Pacheco B, Curvelo JADR, dos Santos-Freitas GMP, Xisto MIDDS, Borba-Santos LP, Rozental S, Ferreira-Pereira A, Barreto-Bergter E. Structural and Functional Alterations Caused by Aureobasidin A in Clinical Resistant Strains of Candida spp. J Fungi (Basel) 2023; 9:1115. [PMID: 37998920 PMCID: PMC10672136 DOI: 10.3390/jof9111115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Candida species are one of the most concerning causative agents of fungal infections in humans. The treatment of invasive Candida infections is based on the use of fluconazole, but the emergence of resistant isolates has been an increasing concern which has led to the study of alternative drugs with antifungal activity. Sphingolipids have been considered a promising target due to their roles in fungal growth and virulence. Inhibitors of the sphingolipid biosynthetic pathway have been described to display antifungal properties, such as myriocin and aureobasidin A, which are active against resistant Candida isolates. In the present study, aureobasidin A did not display antibiofilm activity nor synergism with amphotericin B, but its combination with fluconazole was effective against Candida biofilms and protected the host in an in vivo infection model. Alterations in treated cells revealed increased oxidative stress, reduced mitochondrial membrane potential and chitin content, as well as altered morphology, enhanced DNA leakage and a greater susceptibility to sodium dodecyl sulphate (SDS). In addition, it seems to inhibit the efflux pump CaCdr2p. All these data contribute to elucidating the role of aureobasidin A on fungal cells, especially evidencing its promising use in clinical resistant isolates of Candida species.
Collapse
Affiliation(s)
- Rodrigo Rollin-Pinheiro
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (G.M.P.d.S.-F.); (M.I.D.d.S.X.)
| | - Daniel Clemente de Moraes
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (D.C.d.M.); (B.B.-P.); (J.A.d.R.C.); (A.F.-P.)
| | - Brayan Bayona-Pacheco
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (D.C.d.M.); (B.B.-P.); (J.A.d.R.C.); (A.F.-P.)
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Km 5, Vía Puerto Colombia, Área Metropolitana de Barranquilla, Barranquilla 081007, Colombia
| | - Jose Alexandre da Rocha Curvelo
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (D.C.d.M.); (B.B.-P.); (J.A.d.R.C.); (A.F.-P.)
| | - Giulia Maria Pires dos Santos-Freitas
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (G.M.P.d.S.-F.); (M.I.D.d.S.X.)
| | - Mariana Ingrid Dutra da Silva Xisto
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (G.M.P.d.S.-F.); (M.I.D.d.S.X.)
| | - Luana Pereira Borba-Santos
- Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Sonia Rozental
- Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Antonio Ferreira-Pereira
- Laboratório de Bioquímica Microbiana, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (D.C.d.M.); (B.B.-P.); (J.A.d.R.C.); (A.F.-P.)
| | - Eliana Barreto-Bergter
- Laboratório de Química Biológica de Microrganismos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (G.M.P.d.S.-F.); (M.I.D.d.S.X.)
| |
Collapse
|
10
|
Xavier MO, Poester VR, Trápaga MR, Stevens DA. Sporothrix brasiliensis: Epidemiology, Therapy, and Recent Developments. J Fungi (Basel) 2023; 9:921. [PMID: 37755029 PMCID: PMC10532502 DOI: 10.3390/jof9090921] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Sporotrichosis caused by Sporothrix brasiliensis is an emergent mycosis that is now a worldwide concern. One important step to sporotrichosis control is its correct treatment. However, limitations abound; thus, new antifungals, mainly focused on S. brasiliensis, are urgently needed. We performed a systematic review (following the PRISMA guideline) focused on (1) the global distribution of human and animal sporotrichosis by S. brasiliensis, especially outside of Brazil; (2) appraising therapies tested against this pathogen. We identified sporotrichosis caused by S. brasiliensis reported in five countries (Paraguay, Chile, Argentina, the United Kingdom, and the United States) in addition to Brazil, occurring on three continents, highlighting the epidemiological scenario in Argentina with an important increase in reported cases in recent years. Regarding the antifungal activity of drugs, 25 articles described the in vitro action of 20 unique chemicals and eight repurposed drugs against S. brasiliensis. Only five studies reported in vivo activity against S. brasiliensis (five drugs) using invertebrate and vertebrate models. Sporotrichosis caused by S. brasiliensis has a global impact and it is no longer specifically a Brazilian problem. We review the need for understanding the disease epidemiology, education of clinicians and of the populace, organization of health care delivery to respond to a spreading epidemic, and research on therapy for sporotrichosis.
Collapse
Affiliation(s)
- Melissa Orzechowski Xavier
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil; (M.O.X.); (V.R.P.); (M.R.T.)
- Laboratório de Micologia, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil
| | - Vanice Rodrigues Poester
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil; (M.O.X.); (V.R.P.); (M.R.T.)
- Laboratório de Micologia, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil
| | - Mariana Rodrigues Trápaga
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil; (M.O.X.); (V.R.P.); (M.R.T.)
- Laboratório de Micologia, Faculdade de Medicina (FAMED), Universidade Federal do Rio Grande (FURG), Rio Grande 96200-190, RS, Brazil
| | - David A. Stevens
- California Institute for Medical Research, San Jose, CA 95128, USA
- Division of Infectious Diseases and Geographic Medicine, Stanford University Medical School, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Vaccination with Live or Heat-Killed Aspergillus fumigatus Δ sglA Conidia Fully Protects Immunocompromised Mice from Invasive Aspergillosis. mBio 2022; 13:e0232822. [PMID: 36066100 PMCID: PMC9600187 DOI: 10.1128/mbio.02328-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aspergillus fumigatus causes invasive aspergillosis (IA) in immunocompromised patients, resulting in high mortality rates. Currently, no vaccine formulations to promote immune protection in at-risk individuals have been developed. In this work, we deleted the sterylglucosidase-encoding gene, sglA, in Aspergillus fumigatus and investigated its role in fungal virulence and host vaccine protection. The ΔsglA mutant accumulated sterylglucosides (SGs), newly studied immunomodulatory glycolipids, and exhibited reduced hyphal growth and altered compositions of cell wall polysaccharides. Interestingly, the ΔsglA mutant was avirulent in two murine models of IA and was fully eliminated from the lungs. Both corticosteroid-induced immunosuppressed and cyclophosphamide-induced leukopenic mice vaccinated with live or heat-killed ΔsglA conidia were fully protected against a lethal wild-type A. fumigatus challenge. These results highlight the potential of SG-accumulating strains as safe and promising vaccine formulations against invasive fungal infections. IMPORTANCE Infections by Aspergillus fumigatus occur by the inhalation of environmental fungal spores called conidia. We found that live mutant conidia accumulating glycolipids named sterylglucosides are not able to cause disease when injected into the lung. Interestingly, these animals are now protected against a secondary challenge with live wild-type conidia. Remarkably, protection against a secondary challenge persists even with vaccination with heat-killed mutant conidia. These results will significantly advance the field of the research and development of a safe fungal vaccine for protection against the environmental fungus A. fumigatus.
Collapse
|
12
|
Cui X, Wang L, Lü Y, Yue C. Development and research progress of anti-drug resistant fungal drugs. J Infect Public Health 2022; 15:986-1000. [PMID: 35981408 DOI: 10.1016/j.jiph.2022.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
With the widespread use of immunosuppressive agents and the increase in patients with severe infections, the incidence of fungal infections worldwide has increased year by year. The fungal pathogens Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus cause a total of more than 1 million deaths each year. Long-term use of antifungal drugs can easily lead to fungal resistance, and the prevalence of drug-resistant fungi is a major global health challenge. In order to effectively control global fungal infections, there is an urgent need for new drugs that can exert effective antifungal activity and overcome drug resistance. We must promote the discovery of new antifungal targets and drugs, and find effective ways to control drug-resistant fungi through different ways, so as to reduce the threat of drug-resistant fungi to human life, health and safety. In the past few years, certain progress has been made in the research and development of antifungal drugs. In addition to summarizing some of the antifungal drugs currently approved by the FDA, this review also focuses on potential antifungal drugs, the repositioned drugs, and drugs that can treat drug-resistant bacteria and fungal infections, and provide new ideas for the development of antifungal drugs in the future.
Collapse
Affiliation(s)
- Xiangyi Cui
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Lanlin Wang
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Yuhong Lü
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Changwu Yue
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| |
Collapse
|
13
|
Li L, Wu H, Zhu S, Ji Z, Chi X, Xie F, Hao Y, Lu H, Yang F, Yan L, Zhang D, Jiang Y, Ni T. Discovery of Novel 7-Hydroxy-5-oxo-4,5-dihydrothieno[3,2- b]pyridine-6-carboxamide Derivatives with Potent and Selective Antifungal Activity against Cryptococcus Species. J Med Chem 2022; 65:11257-11269. [PMID: 35922963 DOI: 10.1021/acs.jmedchem.2c00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cryptococcus neoformans and Cryptococcus gattii can cause fatal invasive infections, especially in immunocompromised patients. However, few antifungal drugs are available to help treat cryptococcosis. In this study, by compound library screening, we presented the first report of hit compound P163-0892, which had potent in vitro and in vivo antifungal activity against Cryptococcus spp. In vitro tests showed that P163-0892 was not cytotoxic and had highly selective and strong antifungal activities against Cryptococcus spp. with MIC values less than 1 μg/mL. Synergism of P163-0892 and fluconazole was also observed in vitro. The in vivo antifungal efficacy of P163-0892 was assessed in a wax moth larval fungal infection model, and treatment with 10 mg/kg P163-0892 caused a significant reduction in fungal burden and significant extension of the survival time. Taken together, our data indicate that the hit compound P163-0892 warrants further investigation as a novel anti-Cryptococcus agent.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Hao Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Shuo Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaochen Chi
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Xie
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Lan Yan
- Center for New Drug Research, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
14
|
Caillaud K, Ladavière C. Water‐soluble (poly)acylhydrazones: Syntheses and Applications. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202200064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kilian Caillaud
- Univ Lyon, CNRS, UMR 5223, Ingénierie des Matériaux Polymères Université Claude Bernard Lyon1, INSA Lyon, Université Jean Monnet Villeurbanne Cédex F‐69622 France
| | - Catherine Ladavière
- Univ Lyon, CNRS, UMR 5223, Ingénierie des Matériaux Polymères Université Claude Bernard Lyon1, INSA Lyon, Université Jean Monnet Villeurbanne Cédex F‐69622 France
| |
Collapse
|
15
|
Murphy SE, Bicanic T. Drug Resistance and Novel Therapeutic Approaches in Invasive Candidiasis. Front Cell Infect Microbiol 2022; 11:759408. [PMID: 34970504 PMCID: PMC8713075 DOI: 10.3389/fcimb.2021.759408] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Candida species are the leading cause of invasive fungal infections worldwide and are associated with acute mortality rates of ~50%. Mortality rates are further augmented in the context of host immunosuppression and infection with drug-resistant Candida species. In this review, we outline antifungal drugs already in clinical use for invasive candidiasis and candidaemia, their targets and mechanisms of resistance in clinically relevant Candida species, encompassing not only classical resistance, but also heteroresistance and tolerance. We describe novel antifungal agents and targets in pre-clinical and clinical development, including their spectrum of activity, antifungal target, clinical trial data and potential in treatment of drug-resistant Candida. Lastly, we discuss the use of combination therapy between conventional and repurposed agents as a potential strategy to combat the threat of emerging resistance in Candida.
Collapse
Affiliation(s)
- Sarah E Murphy
- Institute of Infection & Immunity, St George's University of London, London, United Kingdom
| | - Tihana Bicanic
- Institute of Infection & Immunity, St George's University of London, London, United Kingdom.,Clinical Academic Group in Infection and Immunity, St. George's University Hospital National Health Service (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
16
|
Abstract
Lipids play a fundamental role in fungal cell biology, being essential cell membrane components and major targets of antifungal drugs. A deeper knowledge of lipid metabolism is key for developing new drugs and a better understanding of fungal pathogenesis. Here, we built a comprehensive map of the Histoplasma capsulatum lipid metabolic pathway by incorporating proteomic and lipidomic analyses. We performed genetic complementation and overexpression of H. capsulatum genes in Saccharomyces cerevisiae to validate reactions identified in the map and to determine enzymes responsible for catalyzing orphan reactions. The map led to the identification of both the fatty acid desaturation and the sphingolipid biosynthesis pathways as targets for drug development. We found that the sphingolipid biosynthesis inhibitor myriocin, the fatty acid desaturase inhibitor thiocarlide, and the fatty acid analog 10-thiastearic acid inhibit H. capsulatum growth in nanomolar to low-micromolar concentrations. These compounds also reduced the intracellular infection in an alveolar macrophage cell line. Overall, this lipid metabolic map revealed pathways that can be targeted for drug development.
Collapse
|
17
|
DeJarnette C, Meyer CJ, Jenner AR, Butts A, Peters T, Cheramie MN, Phelps GA, Vita NA, Loudon-Hossler VC, Lee RE, Palmer GE. Identification of Inhibitors of Fungal Fatty Acid Biosynthesis. ACS Infect Dis 2021; 7:3210-3223. [PMID: 34786940 DOI: 10.1021/acsinfecdis.1c00404] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fungal fatty acid (FA) synthase and desaturase enzymes are essential for the growth and virulence of human fungal pathogens. These enzymes are structurally distinct from their mammalian counterparts, making them attractive targets for antifungal development. However, there has been little progress in identifying chemotypes that target fungal FA biosynthesis. To accomplish this, we applied a whole-cell-based method known as Target Abundance-based FItness Screening using Candida albicans. Strains with varying levels of FA synthase or desaturase expression were grown in competition to screen a custom small-molecule library. Hit compounds were defined as preferentially inhibiting the growth of the low target-expressing strains. Dose-response experiments confirmed that 16 hits (11 with an acyl hydrazide core) differentially inhibited the growth of strains with an altered desaturase expression, indicating a specific chemical-target interaction. Exogenous unsaturated FAs restored C. albicans growth in the presence of inhibitory concentrations of the most potent acyl hydrazides, further supporting the primary mechanism being inhibition of FA desaturase. A systematic analysis of the structure-activity relationship confirmed the acyl hydrazide core as essential for inhibitory activity. This collection demonstrated broad-spectrum activity against Candida auris and mucormycetes and retained the activity against azole-resistant candida isolates. Finally, a preliminary analysis of toxicity to mammalian cells identified potential lead compounds with desirable selectivities. Collectively, these results establish a scaffold that targets fungal FA biosynthesis with a potential for development into novel therapeutics.
Collapse
Affiliation(s)
- Christian DeJarnette
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee 38163, United States
| | - Chris J. Meyer
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Alexander R. Jenner
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Arielle Butts
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| | - Tracy Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| | - Martin N. Cheramie
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Gregory A. Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis Tennessee 38103, United States
| | - Nicole A. Vita
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee 38163, United States
| | - Victoria C. Loudon-Hossler
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Glen E. Palmer
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
18
|
Sphingolipid Inhibitors as an Alternative to Treat Candidiasis Caused by Fluconazole-Resistant Strains. Pathogens 2021; 10:pathogens10070856. [PMID: 34358009 PMCID: PMC8308474 DOI: 10.3390/pathogens10070856] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/19/2022] Open
Abstract
Candida species are fungal pathogens known to cause a wide spectrum of diseases, and Candida albicans and Candida glabrata are the most common associated with invasive infections. A concerning aspect of invasive candidiasis is the emergence of resistant isolates, especially those highly resistant to fluconazole, the first choice of treatment for these infections. Fungal sphingolipids have been considered a potential target for new therapeutic approaches and some inhibitors have already been tested against pathogenic fungi. The present study therefore aimed to evaluate the action of two sphingolipid synthesis inhibitors, aureobasidin A and myriocin, against different C. albicans and C. glabrata strains, including clinical isolates resistant to fluconazole. Susceptibility tests of aureobasidin A and myriocin were performed using CLSI protocols, and their interaction with fluconazole was evaluated by a checkerboard protocol. All Candida strains tested were sensitive to both inhibitors. Regarding the evaluation of drug interaction, both aureobasidin A and myriocin were synergic with fluconazole, demonstrating that sphingolipid synthesis inhibition could enhance the effect of fluconazole. Thus, these results suggest that sphingolipid inhibitors in conjunction with fluconazole could be useful for treating candidiasis cases, especially those caused by fluconazole resistant isolates.
Collapse
|
19
|
Cairns TC, Zheng X, Zheng P, Sun J, Meyer V. Turning Inside Out: Filamentous Fungal Secretion and Its Applications in Biotechnology, Agriculture, and the Clinic. J Fungi (Basel) 2021; 7:535. [PMID: 34356914 PMCID: PMC8307877 DOI: 10.3390/jof7070535] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/14/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Filamentous fungi are found in virtually every marine and terrestrial habitat. Vital to this success is their ability to secrete a diverse range of molecules, including hydrolytic enzymes, organic acids, and small molecular weight natural products. Industrial biotechnologists have successfully harnessed and re-engineered the secretory capacity of dozens of filamentous fungal species to make a diverse portfolio of useful molecules. The study of fungal secretion outside fermenters, e.g., during host infection or in mixed microbial communities, has also led to the development of novel and emerging technological breakthroughs, ranging from ultra-sensitive biosensors of fungal disease to the efficient bioremediation of polluted environments. In this review, we consider filamentous fungal secretion across multiple disciplinary boundaries (e.g., white, green, and red biotechnology) and product classes (protein, organic acid, and secondary metabolite). We summarize the mechanistic understanding for how various molecules are secreted and present numerous applications for extracellular products. Additionally, we discuss how the control of secretory pathways and the polar growth of filamentous hyphae can be utilized in diverse settings, including industrial biotechnology, agriculture, and the clinic.
Collapse
Affiliation(s)
- Timothy C. Cairns
- Chair of Applied and Molecular Microbiology, Institute of Biotechnology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Xiaomei Zheng
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Ping Zheng
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Jibin Sun
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Vera Meyer
- Chair of Applied and Molecular Microbiology, Institute of Biotechnology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| |
Collapse
|
20
|
Antifungal activity of Acylhydrazone derivatives against Sporothrix spp. Antimicrob Agents Chemother 2021; 65:AAC.02593-20. [PMID: 33593845 PMCID: PMC8092869 DOI: 10.1128/aac.02593-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sporotrichosis is an emerging mycosis caused by members of the genus Sporothrix The disease affects humans and animals, particularly cats, which plays an important role in the zoonotic transmission. Feline sporotrichosis treatment options include itraconazole (ITC), potassium iodide and amphotericin B, drugs usually associated with deleterious adverse reactions and refractoriness in cats, especially when using ITC. Thus, affordable, non-toxic and clinically effective anti-Sporothrix agents are needed. Recently, acylhydrazones (AH), molecules targeting vesicular transport and cell cycle progression, exhibited a potent antifungal activity against several fungal species and displayed low toxicity when compared to the current drugs. In this work, the AH derivatives D13 and SB-AF-1002 were tested against Sporothrix schenckii and Sporothrix brasiliensis Minimal inhibitory concentrations of 0.12 - 1 μg/mL were observed for both species in vitro D13 and SB-AF-1002 showed an additive effect with itraconazole. Treatment with D13 promoted yeast disruption with release of intracellular components, as confirmed by transmission electron microscopy of S. brasiliensis exposed to the AH derivatives. AH-treated cells displayed thickening of the cell wall, discontinuity of the cell membrane and an intense cytoplasmic degeneration. In a murine model of sporotrichosis, treatment with AH derivatives was more efficient than ITC, the drug of choice for sporotrichosis. The results of the preliminary clinical study in cats indicate that D13 is safe and has potential to become a therapeutic option for sporotrichosis when associated to ITC. Our results expand the antifungal broadness of AH derivatives and suggest that these drugs could be exploited to combat sporotrichosis.
Collapse
|
21
|
Toro PM, Peralta F, Oyarzo J, Wilkinson SR, Zavala M, Arancibia R, Moncada-Basualto M, Brito I, Cisterna J, Klahn AH, López C. Evaluation of trypanocidal properties of ferrocenyl and cyrhetrenyl N-acylhydrazones with pendant 5-nitrofuryl group. J Inorg Biochem 2021; 219:111428. [PMID: 33774450 DOI: 10.1016/j.jinorgbio.2021.111428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
Four N-acylhydrazones of general formulae [R1-C(O)-NH-N=C(R2)(5-nitrofuryl)] with (R1 = ferrocenyl or cyrhetrenyl and R2 = H or Me) are synthesized and characterized in solution and in the solid-state. Comparative studies of their stability in solution under different experimental conditions and their electrochemical properties are reported. NMR studies reveal that the four compounds are stable in DMSO‑d6 and complementary UV-Vis studies confirm that they also exhibit high stability in mixtures DMSO:H2O at 37 °C. Electrochemical studies show that the half-wave potential of the nitro group of the N-acylhydrazones is smaller than that of the standard drug nifurtimox and the reduction process follows a self-protonation mechanism. In vitro studies on the antiparasitic activities of the four complexes and the nifurtimox against Trypanosoma cruzi and Trypanosoma brucei reveal that: i) the N-acylhydrazones have a potent inhibitory growth activity against both parasites [EC50 in the low micromolar (in T. cruzi) or even in the nanomolar (in T. brucei) range] and ii) cyrhetrenyl derivatives are more effective than their ferrocenyl analogs. Parallel studies on the L6 rat skeletal myoblast cell line have also been conducted, and the selectivity indexes determined. Three of the four N-acylhydrazones showed higher selectivity towards T. brucei than the standard drug nifurtimox. Additional studies suggest that the organometallic compounds are bioactivated by type I nitroreductase enzymes.
Collapse
Affiliation(s)
- Patricia M Toro
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andrés Bello, Quillota 980, Viña del Mar, Chile.
| | - Francisco Peralta
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Casilla 4059, Valparaíso, Chile
| | - Juan Oyarzo
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Casilla 4059, Valparaíso, Chile
| | - Shane R Wilkinson
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Mónica Zavala
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Rodrigo Arancibia
- Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Mauricio Moncada-Basualto
- Departamento de Química Inorgánica y Analítica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Iván Brito
- Departamento de Química, Facultad de Ciencias Básicas, Universidad de Antofagasta, Avda. Universidad de Antofagasta 02800, Campus Coloso, Antofagasta, Chile
| | - Jonathan Cisterna
- Departamento de Química, Facultad de Ciencias Básicas, Universidad de Antofagasta, Avda. Universidad de Antofagasta 02800, Campus Coloso, Antofagasta, Chile
| | - A Hugo Klahn
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Casilla 4059, Valparaíso, Chile
| | - Concepción López
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Facultat de Química, Universitat de Barcelona, Martí i Franqués 1-11, E-08028 Barcelona, Spain.
| |
Collapse
|
22
|
Bryan AM, You JK, McQuiston T, Lazzarini C, Qiu Z, Sheridan B, Nuesslein-Hildesheim B, Del Poeta M. FTY720 reactivates cryptococcal granulomas in mice through S1P receptor 3 on macrophages. J Clin Invest 2021; 130:4546-4560. [PMID: 32484801 DOI: 10.1172/jci136068] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
FTY720 is a treatment for relapsing remitting multiple sclerosis (MS). It is an analog of sphingosine-1-phosphate (S1P) and targets S1P receptors 1, 3, 4, and 5. Recent reports indicate an association between long-term exposure to FTY720 and cases of cryptococcal infection. Here, we studied the effect of FTY720 and its derivative, BAF312, which only target S1P receptors 1 and 5, in a mouse model of cryptococcal infection. We found that treatment with FTY720, but not with BAF312, led to decreased survival and increased organ burden in mouse cryptococcal granulomas. Both FTY720 and BAF312 caused a profound CD4+ and CD8+ T cell depletion in blood and lungs but only treatment with FTY720 led to cryptococcal reactivation. Treatment with FTY720, but not with BAF312, was associated with disorganization of macrophages and with M2 polarization at the granuloma site. In a cell system, FTY720 decreased phagocytosis and production of reactive oxygen species by macrophages, a phenotype recapitulated in the S1pr3-/- knockout macrophages. Our results suggest that FTY720 reactivates cryptococcosis from the granuloma through a S1P receptor 3-mediated mechanism and support the rationale for development of more-specific receptor modulators for therapeutic use of MS.
Collapse
Affiliation(s)
- Arielle M Bryan
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Jeehyun Karen You
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Cristina Lazzarini
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Zhijuan Qiu
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Brian Sheridan
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA.,Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, USA.,Veterans Affairs Medical Center, Northport, New York, USA
| |
Collapse
|
23
|
The Future of Antifungal Drug Therapy: Novel Compounds and Targets. Antimicrob Agents Chemother 2021; 65:AAC.01719-20. [PMID: 33229427 DOI: 10.1128/aac.01719-20] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fungal infections are a universal problem and are routinely associated with high morbidity and mortality rates in immunocompromised patients. Existing therapies comprise five different classes of antifungal agents, four of which target the synthesis of ergosterol and cell wall glucans. However, the currently available antifungals have many limitations, including poor oral bioavailability, narrow therapeutic indices, and emerging drug resistance resulting from their use, thus making it essential to investigate the development of novel drugs which can overcome these limitations and add to the antifungal armamentarium. Advances have been made in antifungal drug discovery research and development over the past few years as evidenced by the presence of several new compounds currently in various stages of development. In the following minireview, we provide a comprehensive summary of compounds aimed at one or more novel molecular targets. We also briefly describe potential pathways relevant for fungal pathogenesis that can be considered for drug development in the near future.
Collapse
|
24
|
Sertbas M, Ulgen KO. Genome-Scale Metabolic Modeling for Unraveling Molecular Mechanisms of High Threat Pathogens. Front Cell Dev Biol 2020; 8:566702. [PMID: 33251208 PMCID: PMC7673413 DOI: 10.3389/fcell.2020.566702] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
Pathogens give rise to a wide range of diseases threatening global health and hence drawing public health agencies' attention to establish preventative and curative solutions. Genome-scale metabolic modeling is ever increasingly used tool for biomedical applications including the elucidation of antibiotic resistance, virulence, single pathogen mechanisms and pathogen-host interaction systems. With this approach, the sophisticated cellular system of metabolic reactions inside the pathogens as well as between pathogen and host cells are represented in conjunction with their corresponding genes and enzymes. Along with essential metabolic reactions, alternate pathways and fluxes are predicted by performing computational flux analyses for the growth of pathogens in a very short time. The genes or enzymes responsible for the essential metabolic reactions in pathogen growth are regarded as potential drug targets, as a priori guide to researchers in the pharmaceutical field. Pathogens alter the key metabolic processes in infected host, ultimately the objective of these integrative constraint-based context-specific metabolic models is to provide novel insights toward understanding the metabolic basis of the acute and chronic processes of infection, revealing cellular mechanisms of pathogenesis, identifying strain-specific biomarkers and developing new therapeutic approaches including the combination drugs. The reaction rates predicted during different time points of pathogen development enable us to predict active pathways and those that only occur during certain stages of infection, and thus point out the putative drug targets. Among others, fatty acid and lipid syntheses reactions are recent targets of new antimicrobial drugs. Genome-scale metabolic models provide an improved understanding of how intracellular pathogens utilize the existing microenvironment of the host. Here, we reviewed the current knowledge of genome-scale metabolic modeling in pathogen cells as well as pathogen host interaction systems and the promising applications in the extension of curative strategies against pathogens for global preventative healthcare.
Collapse
Affiliation(s)
- Mustafa Sertbas
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey.,Department of Chemical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Kutlu O Ulgen
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
25
|
Fernandes CM, Poeta MD. Fungal sphingolipids: role in the regulation of virulence and potential as targets for future antifungal therapies. Expert Rev Anti Infect Ther 2020; 18:1083-1092. [PMID: 32673125 PMCID: PMC7657966 DOI: 10.1080/14787210.2020.1792288] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The antifungal therapy currently available includes three major classes of drugs: polyenes, azoles and echinocandins. However, the clinical use of these compounds faces several challenges: while polyenes are toxic to the host, antifungal resistance to azoles and echinocandins has been reported. AREAS COVERED Fungal sphingolipids (SL) play a pivotal role in growth, morphogenesis and virulence. In addition, fungi possess unique enzymes involved in SL synthesis, leading to the production of lipids which are absent or differ structurally from the mammalian counterparts. In this review, we address the enzymatic reactions involved in the SL synthesis and their relevance to the fungal pathogenesis, highlighting their potential as targets for novel drugs and the inhibitors described so far. EXPERT OPINION The pharmacological inhibition of fungal serine palmitoyltransferase depends on the development of specific drugs, as myriocin also targets the mammalian enzyme. Inhibitors of ceramide synthase might constitute potent antifungals, by depleting the pool of complex SL and leading to the accumulation of the toxic intermediates. Acylhydrazones and aureobasidin A, which inhibit GlcCer and IPC synthesis, are not toxic to the host and effectively treat invasive mycoses, emerging as promising new classes of antifungal drugs.
Collapse
Affiliation(s)
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, NY, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, NY, USA
- Veterans Administration Medical Center, Northport, NY, USA
| |
Collapse
|
26
|
Li M, Cheng LP, Pang W, Zhong ZJ, Guo LL. Design, Synthesis, and Biological Evaluation of Novel Acylhydrazone Derivatives as Potent Neuraminidase Inhibitors. ACS Med Chem Lett 2020; 11:1745-1750. [PMID: 32944142 DOI: 10.1021/acsmedchemlett.0c00313] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/24/2020] [Indexed: 12/26/2022] Open
Abstract
Neuraminidase (NA) is an important target for current research on anti-influenza drugs. The acylhydrazone derivatives containing the -CONHN=CH- framework have been shown to have good NA inhibitory activity. In this paper, a series of novel acylhydrazone NA inhibitors (9a-9n) were designed and synthesized, and the inhibitory activities against NA were evaluated in vitro. The NA inhibition results showed that compound 9j has the most potent inhibitory activity (IC50 = 0.6 μM) against NA, which is significantly lower than that of the positive control oseltamivir carboxylic acid (OSC) (IC50 = 17.00 μM). Molecular docking analysis indicates that the acylhydrazone group plays an important role in compound 9j, which can bind well to the residues Arg371 and Arg292 in the S1 subsite of NA. The good potency of 9j may be also ascribed to the extending of morpholinyl ring into the 430-cavity. The results of this work may contribute to the development of more potent NA inhibitors to against mutant influenza viruses.
Collapse
Affiliation(s)
- Meng Li
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Li Ping Cheng
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wan Pang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhi Jian Zhong
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Ling Ling Guo
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
27
|
McEvoy K, Normile TG, Poeta MD. Antifungal Drug Development: Targeting the Fungal Sphingolipid Pathway. J Fungi (Basel) 2020; 6:jof6030142. [PMID: 32825250 PMCID: PMC7559796 DOI: 10.3390/jof6030142] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Fungal infections are becoming more prevalent and problematic due to the continual rise of immune deficient patients as well as the progressive development of drug resistance towards currently available antifungal drugs. There has been a significant increase in the development of antifungal compounds with a similar mechanism of action of current drugs. In contrast, there has been very little progress in developing compounds inhibiting totally new fungal targets or/and fungal pathways. This review focuses on novel compounds recently discovered to target the fungal sphingolipids and their metabolizing enzymes.
Collapse
Affiliation(s)
- Kyle McEvoy
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
| | - Tyler G. Normile
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA; (K.M.); (T.G.N.)
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Veterans Administration Medical Center, Northport, NY 11768, USA
- Correspondence: ; Tel.: +1-631-632-4024
| |
Collapse
|
28
|
Preclinical Evaluation of Acylhydrazone SB-AF-1002 as a Novel Broad-Spectrum Antifungal Agent. Antimicrob Agents Chemother 2020; 64:AAC.00946-20. [PMID: 32601165 DOI: 10.1128/aac.00946-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
The incidence of invasive fungal infections is rising due to the increase in susceptible populations. Current clinically available drugs have therapeutic limitations due to toxicity, a narrow spectrum of activity, and, more importantly, the consistent rise of fungal species that are intrinsically resistant or that develop resistance due to prolonged therapy. Thus, there is an urgent need for new broad-spectrum antifungal agents with low toxicity and a novel mechanism of action. We previously reported a new class of potent antifungal compounds, acylhydrazones, that target the fungal sphingolipid pathway. Based upon our initial lead molecules, (E)-N'-(5-bromo-2-hydroxybenzylidene)-2-methylbenzohydrazide and D13, we performed a structure-activity relationship study, synthesizing ca. 300 new compounds. Of these, 5 compounds were identified to be the most promising for further studies, based on their broad-spectrum activity and low toxicity in mammalian cells lines. Among these top 5 lead compounds, we report here the impressive in vivo activity of 2,4-dibromo-N'-(5-bromo-2-hydroxybenzylidene)benzohydrazide (SB-AF-1002) in several models of systemic fungal infection. Our data show that SB-AF-1002 is efficacious and outperforms current standard-of-care drugs in models of invasive fungal infections, such as cryptococcosis, candidiasis, and aspergillosis. Specifically, animals treated with SB-AF-1002 not only survived the infection but also showed a clearing of fungal cells from key organs. Moreover, SB-AF-1002 was very effective in an aspergillosis model as a prophylactic therapy. SB-AF-1002 also displayed acceptable pharmacokinetic properties in mice, similar to those of the parent compound, D13. These results clearly indicate that our novel acylhydrazones constitute a new class of highly potent and efficacious antifungal agents which warrant further development for the treatment of invasive fungal infections.
Collapse
|
29
|
Yu Y, Albrecht K, Groll J, Beilhack A. Innovative therapies for invasive fungal infections in preclinical and clinical development. Expert Opin Investig Drugs 2020; 29:961-971. [DOI: 10.1080/13543784.2020.1791819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Yidong Yu
- Interdisciplinary Center for Clinical Research Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital of Würzburg , Würzburg, Germany
| | - Krystyna Albrecht
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg , Würzburg, Germany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg , Würzburg, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research Laboratory for Experimental Stem Cell Transplantation, Department of Internal Medicine II, University Hospital of Würzburg , Würzburg, Germany
- Department of Pediatrics, University Hospital of Würzburg , Würzburg, Germany
| |
Collapse
|
30
|
Xie F, Ni T, Ding Z, Hao Y, Wang R, Wang R, Wang T, Chai X, Yu S, Jin Y, Jiang Y, Zhang D. Design, synthesis, and in vitro evaluation of novel triazole analogues featuring isoxazole moieties as antifungal agents. Bioorg Chem 2020; 101:103982. [DOI: 10.1016/j.bioorg.2020.103982] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/23/2020] [Accepted: 05/29/2020] [Indexed: 01/17/2023]
|
31
|
Bogdanov AV, Iskhakova KR, Voloshina AD, Sapunova AS, Kulik NV, Terekhova NV, Arsenyev MV, Ziyatdinova GK, Bukharov SV. Ammonium-Charged Sterically Hindered Phenols with Antioxidant and Selective Anti-Gram-Positive Bacterial Activity. Chem Biodivers 2020; 17:e2000147. [PMID: 32349191 DOI: 10.1002/cbdv.202000147] [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] [Received: 03/01/2020] [Accepted: 03/27/2020] [Indexed: 11/10/2022]
Abstract
The increase in the resistance of pathogens, in particular Staphylococcus aureus, to the action of antibiotics necessitates the search for new readily available and non-toxic drugs. In solving this problem, phenolic acylhydrazones have high potential. In this communication, the synthesis of quaternary ammonium compounds containing a differently substituted phenolic moiety has been performed. An initial study of antimicrobial activity showed that these compounds are highly selective against S. aureus and B. cereus. The highest activity (MIC 2.0 μm) was shown by hydrazones containing a catechol fragment. These compounds are more than 3-fold more active against S. aureus and 3-10-fold more active against B. cereus than norfloxacin. Low hemolytic and high antioxidant activities of all new compounds were also established.
Collapse
Affiliation(s)
- Andrei V Bogdanov
- A.E.Arbuzov Institute of Organic and Physical Chemistry, FRC, Kazan Scientific Center of RAS, 8 Arbuzov Str., Kazan, 420088, Russian Federation
| | - Kamilla R Iskhakova
- Kazan National Research Technological University, 68 K. Marx Str., Kazan, 420015, Russian Federation
| | - Alexandra D Voloshina
- A.E.Arbuzov Institute of Organic and Physical Chemistry, FRC, Kazan Scientific Center of RAS, 8 Arbuzov Str., Kazan, 420088, Russian Federation
| | - Anastasia S Sapunova
- A.E.Arbuzov Institute of Organic and Physical Chemistry, FRC, Kazan Scientific Center of RAS, 8 Arbuzov Str., Kazan, 420088, Russian Federation
| | - Natalia V Kulik
- A.E.Arbuzov Institute of Organic and Physical Chemistry, FRC, Kazan Scientific Center of RAS, 8 Arbuzov Str., Kazan, 420088, Russian Federation
| | - Natalia V Terekhova
- A.E.Arbuzov Institute of Organic and Physical Chemistry, FRC, Kazan Scientific Center of RAS, 8 Arbuzov Str., Kazan, 420088, Russian Federation
| | - Maxim V Arsenyev
- G. A. Razuvaev Institute of Organometallic Chemistry, RAS, 49 Tropinin Str., Nizhny, Novgorod, 603950, Russian Federation
| | - Guzel K Ziyatdinova
- Kazan Federal University, 18 Kremlevskaya Str., Kazan, 420008, Russian Federation
| | - Sergey V Bukharov
- Kazan National Research Technological University, 68 K. Marx Str., Kazan, 420015, Russian Federation
| |
Collapse
|
32
|
Li D, She X, Calderone R. The antifungal pipeline: the need is established. Are there new compounds? FEMS Yeast Res 2020; 20:5827531. [DOI: 10.1093/femsyr/foaa023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
Our review summarizes and compares the temporal development (eras) of antifungal drug discovery as well as antibacterial ventures. The innovation gap that occurred in antibacterial discovery from 1960 to 2000 was likely due to tailoring of existing compounds to have better activity than predecessors. Antifungal discovery also faced innovation gaps. The semi-synthetic antibiotic era was followed closely by the resistance era and the heightened need for new compounds and targets. With the immense contribution of comparative genomics, antifungal targets became part of the discovery focus. These targets by definition are absolutely required to be fungal- or even lineage (clade) specific. Importantly, targets need to be essential for growth and/or have important roles in disease and pathogenesis. Two types of antifungals are discussed that are mostly in the FDA phase I–III clinical trials. New antifungals are either modified to increase bioavailability and stability for instance, or are new compounds that inhibit new targets. One of the important developments in incentivizing new antifungal discovery has been the prolific number of publications of global and country-specific incidence. International efforts that champion global antimicrobial drug discovery are discussed. Still, interventions are needed. The current pipeline of antifungals and alternatives to antifungals are discussed including vaccines.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| | - Xiaodong She
- Jiangsu Key laboratory of Molecular Biology for Skin Disease and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS), Nanjing 210029, China
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| |
Collapse
|
33
|
Houšť J, Spížek J, Havlíček V. Antifungal Drugs. Metabolites 2020; 10:metabo10030106. [PMID: 32178468 PMCID: PMC7143493 DOI: 10.3390/metabo10030106] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
We reviewed the licensed antifungal drugs and summarized their mechanisms of action, pharmacological profiles, and susceptibility to specific fungi. Approved antimycotics inhibit 1,3-β-d-glucan synthase, lanosterol 14-α-demethylase, protein, and deoxyribonucleic acid biosynthesis, or sequestrate ergosterol. Their most severe side effects are hepatotoxicity, nephrotoxicity, and myelotoxicity. Whereas triazoles exhibit the most significant drug–drug interactions, echinocandins exhibit almost none. The antifungal resistance may be developed across most pathogens and includes drug target overexpression, efflux pump activation, and amino acid substitution. The experimental antifungal drugs in clinical trials are also reviewed. Siderophores in the Trojan horse approach or the application of siderophore biosynthesis enzyme inhibitors represent the most promising emerging antifungal therapies.
Collapse
|
34
|
New Triazole NT-a9 Has Potent Antifungal Efficacy against Cryptococcus neoformans In Vitro and In Vivo. Antimicrob Agents Chemother 2020; 64:AAC.01628-19. [PMID: 31791946 DOI: 10.1128/aac.01628-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022] Open
Abstract
In the past decades, the incidence of cryptococcosis has increased dramatically, which poses a new threat to human health. However, only a few drugs are available for the treatment of cryptococcosis. Here, we described a leading compound, NT-a9, an analogue of isavuconazole, that showed strong antifungal activities in vitro and in vivo NT-a9 showed a wide range of activities against several pathogenic fungi in vitro, including Cryptococcus neoformans, Cryptococcus gattii, Candida albicans, Candida krusei, Candida tropicalis, Candida glabrata, and Candida parapsilosis, with MICs ranging from 0.002 to 1 μg/ml. In particular, NT-a9 exhibited excellent efficacy against C. neoformans, with a MIC as low as 0.002 μg/ml. NT-a9 treatment resulted in changes in the sterol contents in C. neoformans, similarly to fluconazole. In addition, NT-a9 possessed relatively low cytotoxicity and a high selectivity index. The in vivo efficacy of NT-a9 was assessed using a murine disseminated-cryptococcosis model. Mice were infected intravenously with 1.8 × 106 CFU of C. neoformans strain H99. In the survival study, NT-a9 significantly prolonged the survival times of mice compared with the survival times of the control group or the isavuconazole-, fluconazole-, or amphotericin B-treated groups. Of note, 4 and 8 mg/kg of body weight of NT-a9 rescued all the mice, with a survival rate of 100%. In the fungal-burden study, NT-a9 also significantly reduced the fungal burdens in brains and lungs, while fluconazole and amphotericin B only reduced the fungal burden in lungs. Taken together, these data suggested that NT-a9 is a promising antifungal candidate for the treatment of cryptococcosis infection.
Collapse
|
35
|
Identification of a Phenylthiazole Small Molecule with Dual Antifungal and Antibiofilm Activity Against Candida albicans and Candida auris. Sci Rep 2019; 9:18941. [PMID: 31831822 PMCID: PMC6908612 DOI: 10.1038/s41598-019-55379-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022] Open
Abstract
Candida species are a leading source of healthcare infections globally. The limited number of antifungal drugs combined with the isolation of Candida species, namely C. albicans and C. auris, exhibiting resistance to current antifungals necessitates the development of new therapeutics. The present study tested 85 synthetic phenylthiazole small molecules for antifungal activity against drug-resistant C. albicans. Compound 1 emerged as the most potent molecule, inhibiting growth of C. albicans and C. auris strains at concentrations ranging from 0.25–2 µg/mL. Additionally, compound 1 inhibited growth of other clinically-relevant yeast (Cryptococcus) and molds (Aspergillus) at a concentration as low as 0.50 µg/mL. Compound 1 exhibited rapid fungicidal activity, reducing the burden of C. albicans and C. auris below the limit of detection within 30 minutes. Compound 1 exhibited potent antibiofilm activity, similar to amphotericin B, reducing the metabolic activity of adherent C. albicans and C. auris biofilms by more than 66% and 50%, respectively. Furthermore, compound 1 prolonged survival of Caenorhabditis elegans infected with strains of C. albicans and C. auris, relative to the untreated control. The present study highlights phenylthiazole small molecules, such as compound 1, warrant further investigation as novel antifungal agents for drug-resistant Candida infections.
Collapse
|
36
|
Aerts JMFG, Artola M, van Eijk M, Ferraz MJ, Boot RG. Glycosphingolipids and Infection. Potential New Therapeutic Avenues. Front Cell Dev Biol 2019; 7:324. [PMID: 31867330 PMCID: PMC6908816 DOI: 10.3389/fcell.2019.00324] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
Glycosphingolipids (GSLs), the main topic of this review, are a subclass of sphingolipids. With their glycans exposed to the extracellular space, glycosphingolipids are ubiquitous components of the plasma membrane of cells. GSLs are implicated in a variety of biological processes including specific infections. Several pathogens use GSLs at the surface of host cells as binding receptors. In addition, lipid-rafts in the plasma membrane of host cells may act as platform for signaling the presence of pathogens. Relatively common in man are inherited deficiencies in lysosomal glycosidases involved in the turnover of GSLs. The associated storage disorders (glycosphingolipidoses) show lysosomal accumulation of substrate(s) of the deficient enzyme. In recent years compounds have been identified that allow modulation of GSLs levels in cells. Some of these agents are well tolerated and already used to treat lysosomal glycosphingolipidoses. This review summarizes present knowledge on the role of GSLs in infection and subsequent immune response. It concludes with the thought to apply glycosphingolipid-lowering agents to prevent and/or combat infections.
Collapse
Affiliation(s)
| | - M Artola
- Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - M van Eijk
- Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - M J Ferraz
- Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - R G Boot
- Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
37
|
Abstract
Aspergilli produce conidia for reproduction or to survive hostile conditions, and they are highly effective in the distribution of conidia through the environment. In immunocompromised individuals, inhaled conidia can germinate inside the respiratory tract, which may result in invasive pulmonary aspergillosis. The management of invasive aspergillosis has become more complex, with new risk groups being identified and the emergence of antifungal resistance. Patient survival is threatened by these developments, stressing the need for alternative therapeutic strategies. As germination is crucial for infection, prevention of this process might be a feasible approach. A broader understanding of conidial germination is important to identify novel antigermination targets. In this review, we describe conidial resistance against various stresses, transition from dormant conidia to hyphal growth, the underlying molecular mechanisms involved in germination of the most common Aspergillus species, and promising antigermination targets. Germination of Aspergillus is characterized by three morphotypes: dormancy, isotropic growth, and polarized growth. Intra- and extracellular proteins play an important role in the protection against unfavorable environmental conditions. Isotropically expanding conidia remodel the cell wall, and biosynthetic machineries are needed for cellular growth. These biosynthetic machineries are also important during polarized growth, together with tip formation and the cell cycle machinery. Genes involved in isotropic and polarized growth could be effective antigermination targets. Transcriptomic and proteomic studies on specific Aspergillus morphotypes will improve our understanding of the germination process and allow discovery of novel antigermination targets and biomarkers for early diagnosis and therapy.
Collapse
|
38
|
Sphingolipid biosynthetic pathway is crucial for growth, biofilm formation and membrane integrity of Scedosporium boydii. Future Med Chem 2019; 11:2905-2917. [PMID: 31713454 DOI: 10.4155/fmc-2019-0186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Glycosphingolipids are conserved lipids displaying a variety of functions in fungal cells, such as determination of cell polarity and virulence. They have been considered as potent targets for new antifungal drugs. The present work aimed to test two inhibitors, myriocin and DL-threo-1-Phenyl-2-palmitoylamino-3-morpholino-1-propanol, in Scedosporium boydii, a pathogenic fungus which causes a wide range of disease. Materials & methods: Mass spectrometry, microscopy and cell biology approaches showed that treatment with both inhibitors led to defects in fungal growth and membrane integrity, and caused an increased susceptibility to the current antifungal agents. Conclusion: These data demonstrate the antifungal potential of drugs inhibiting sphingolipid biosynthesis, as well as the usefulness of sphingolipids as promising targets for the development of new therapeutic options.
Collapse
|
39
|
Haranahalli K, Lazzarini C, Sun Y, Zambito J, Pathiranage S, McCarthy JB, Mallamo J, Del Poeta M, Ojima I. SAR Studies on Aromatic Acylhydrazone-Based Inhibitors of Fungal Sphingolipid Synthesis as Next-Generation Antifungal Agents. J Med Chem 2019; 62:8249-8273. [PMID: 31369263 DOI: 10.1021/acs.jmedchem.9b01004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recently, the fungal sphingolipid glucosylceramide (GlcCer) synthesis has emerged as a highly promising new target for drug discovery of next-generation antifungal agents, and we found two aromatic acylhydrazones as effective inhibitors of GlcCer synthesis based on HTP screening. In the present work, we have designed libraries of new aromatic acylhydrazones, evaluated their antifungal activities (MIC80 and time-kill profile) against C. neoformans, and performed an extensive SAR study, which led to the identification of five promising lead compounds, exhibiting excellent fungicidal activities with very large selectivity index. Moreover, two compounds demonstrated broad spectrum antifungal activity against six other clinically relevant fungal strains. These five lead compounds were examined for their synergism/cooperativity with five clinical drugs against seven fungal strains, and very encouraging results were obtained; e.g., the combination of all five lead compounds with voriconazole exhibited either synergistic or additive effect to all seven fungal strains.
Collapse
Affiliation(s)
- Krupanandan Haranahalli
- Institute of Chemical Biology and Drug Discovery , Stony Brook University , Stony Brook , New York 11794-3400 , United States.,Department of Chemistry , Stony Brook University , Stony Brook , New York 11794-3400 , United States
| | - Cristina Lazzarini
- Department of Molecular Genetics and Microbiology , Stony Brook University , Stony Brook , New York 11794-5222 , United States.,Veterans Administration Medical Center , Northport , New York 11768 , United States
| | - Yi Sun
- Department of Chemistry , Stony Brook University , Stony Brook , New York 11794-3400 , United States
| | - Julia Zambito
- Department of Chemistry , Stony Brook University , Stony Brook , New York 11794-3400 , United States
| | - Senuri Pathiranage
- Department of Chemistry , Stony Brook University , Stony Brook , New York 11794-3400 , United States
| | - J Brian McCarthy
- MicroRid Technologies Inc. , 86 Deer Park Road , Dix Hills , New York 11746 , United States
| | - John Mallamo
- MicroRid Technologies Inc. , 86 Deer Park Road , Dix Hills , New York 11746 , United States
| | - Maurizio Del Poeta
- Institute of Chemical Biology and Drug Discovery , Stony Brook University , Stony Brook , New York 11794-3400 , United States.,Department of Molecular Genetics and Microbiology , Stony Brook University , Stony Brook , New York 11794-5222 , United States.,Veterans Administration Medical Center , Northport , New York 11768 , United States.,Division of Infectious Diseases, School of Medicine , Stony Brook University , New York 11794-8434 , United States
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery , Stony Brook University , Stony Brook , New York 11794-3400 , United States.,Department of Chemistry , Stony Brook University , Stony Brook , New York 11794-3400 , United States
| |
Collapse
|
40
|
Nimrichter L, Rodrigues ML, Del Poeta M. Exploiting Lipids to Develop Anticryptococcal Vaccines. CURRENT TROPICAL MEDICINE REPORTS 2019. [DOI: 10.1007/s40475-019-00178-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
41
|
Guilherme FD, Simonetti JÉ, Folquitto LRS, Reis ACC, Oliver JC, Dias ALT, Dias DF, Carvalho DT, Brandão GC, Souza TBD. Synthesis, chemical characterization and antimicrobial activity of new acylhydrazones derived from carbohydrates. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.02.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
42
|
Synthesis, anticancer activity and mechanism of iron chelator derived from 2,6-diacetylpyridine bis(acylhydrazones). J Inorg Biochem 2019; 193:1-8. [DOI: 10.1016/j.jinorgbio.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/01/2019] [Accepted: 01/07/2019] [Indexed: 12/11/2022]
|
43
|
Abstract
Abstract
Purpose of Review
Pervasive fungal infection among the immunocompromised population, in conjunction with a lack of effective treatment options, has demanded further scrutiny. Millions of people are still dying annually from fungal infections. While existing treatment for these fungal infections exists, it is difficult to administer without adverse effects in the immunocompromised and is slowly becoming obsolete due to varying mutation rates and rising resistance in multiple species. Thus, vaccines may be a viable target for preventing and treating fungal infections and addressing the critical challenge of such infections.
Recent Findings
Candida albicans, along with other non-albicans Candida species, is among the more virulent class of fungal specimens considered for vaccine development. C. albicans is responsible for a large percentage of invasive fungal infections among immunocompromised and immunocompetent populations and carries a relatively high mortality rate. In the last decade, a recent increase in infective capacity among Candida species has shed light on the lack of adequate fungal vaccine choices. While roadblocks still exist in the development of antifungal vaccines, several novel targets have been examined and proposed as candidates.
Summary
Success in vaccine development has universal appeal; an anti-Candida vaccine formulation could be modified to work against other fungal infections and thus bolster the antifungal pipeline.
Collapse
|
44
|
Di Mambro T, Guerriero I, Aurisicchio L, Magnani M, Marra E. The Yin and Yang of Current Antifungal Therapeutic Strategies: How Can We Harness Our Natural Defenses? Front Pharmacol 2019; 10:80. [PMID: 30804788 PMCID: PMC6370704 DOI: 10.3389/fphar.2019.00080] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Fungal infections have aroused much interest over the last years because of their involvement in several human diseases. Immunocompromission due to transplant-related therapies and malignant cancer treatments are risk factors for invasive fungal infections, but also aggressive surgery, broad-spectrum antibiotics and prosthetic devices are frequently associated with infectious diseases. Current therapy is based on the administration of antifungal drugs, but the occurrence of resistant strains to the most common molecules has become a serious health-care problem. New antifungal agents are urgently needed and it is essential to identify fungal molecular targets that could offer alternatives for development of treatments. The fungal cell wall and plasma membrane are the most important structures that offer putative new targets which can be modulated in order to fight microbial infections. The development of monoclonal antibodies against new targets is a valid therapeutic strategy, both to solve resistance problems and to support the immune response, especially in immunocompromised hosts. In this review, we summarize currently used antifungal agents and propose novel therapeutic approaches, including new fungal molecular targets to be considered for drug development.
Collapse
Affiliation(s)
- Tomas Di Mambro
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy.,Diatheva s.r.l., Cartoceto, Italy
| | - Ilaria Guerriero
- Takis s.r.l., Rome, Italy.,Veterinary Immunotherapy and Translational Research, Rome, Italy
| | - Luigi Aurisicchio
- Takis s.r.l., Rome, Italy.,Veterinary Immunotherapy and Translational Research, Rome, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy.,Diatheva s.r.l., Cartoceto, Italy
| | - Emanuele Marra
- Takis s.r.l., Rome, Italy.,Veterinary Immunotherapy and Translational Research, Rome, Italy
| |
Collapse
|
45
|
Fabri JHTM, Godoy NL, Rocha MC, Munshi M, Cocio TA, von Zeska Kress MR, Fill TP, da Cunha AF, Del Poeta M, Malavazi I. The AGC Kinase YpkA Regulates Sphingolipids Biosynthesis and Physically Interacts With SakA MAP Kinase in Aspergillus fumigatus. Front Microbiol 2019; 9:3347. [PMID: 30692984 PMCID: PMC6339957 DOI: 10.3389/fmicb.2018.03347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/28/2018] [Indexed: 12/12/2022] Open
Abstract
Sphingolipids (SL) are complex lipids and components of the plasma membrane which are involved in numerous cellular processes, as well as important for virulence of different fungal pathogens. In yeast, SL biosynthesis is regulated by the "AGC kinases" Ypk1 and Ypk2, which also seem to connect the SL biosynthesis with the cell wall integrity (CWI) and the High Osmolarity Glycerol (HOG) pathways. Here, we investigate the role of ypkA Y PK1 in SL biosynthesis and its relationship with the CWI and the HOG pathways in the opportunistic human pathogen Aspergillus fumigatus. We found that ypkA is important for fungal viability, since the ΔypkA strain presented a drastically sick phenotype and complete absence of conidiation. We observed that under repressive condition, the conditional mutant niiA::ypkA exhibited vegetative growth defects, impaired germination and thermosensitivity. In addition, the ypkA loss of function caused a decrease in glycosphingolipid (GSL) levels, especially the metabolic intermediates belonging to the neutral GSL branch including dihydroceramide (DHC), ceramide (Cer), and glucosylceramide (GlcCer), but interestingly a small increase in ergosterol content. Genetic analyzes showed that ypkA genetically interacts with the MAP kinases of CWI and HOG pathways, mpkA and sakA, respectively, while only SakA physically interacts with YpkA. Our results suggest that YpkA is important for fungal survival through the regulation of GSL biosynthesis and cross talks with A. fumigatus MAP kinase pathways.
Collapse
Affiliation(s)
| | - Naiane Lima Godoy
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Marina Campos Rocha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Mansa Munshi
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
| | - Tiago Alexandre Cocio
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Marcia Regina von Zeska Kress
- Departamento de Análises Clínicas Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Anderson Ferreira da Cunha
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Maurizio Del Poeta
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States.,Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY, United States.,Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, United States.,Veterans Administration Medical Center, Northport, NY, United States
| | - Iran Malavazi
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, Brazil
| |
Collapse
|
46
|
Haranahalli K, Honda T, Ojima I. Recent progress in the strategic incorporation of fluorine into medicinally active compounds. J Fluor Chem 2019; 217:29-40. [PMID: 31537946 PMCID: PMC6752223 DOI: 10.1016/j.jfluchem.2018.11.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This account exemplifies our recent progress on the strategic incorporation of fluorine and organofluorine groups to (i) taxoid anticancer agents, (ii) acylhydrazone-based antifungal agents and (iii) inhibitors of matrix metalloproteinase 9 (MMP9) for medicinal chemistry and chemical biology studies. In the case study (i), a series of next-generation fluorotaxoids, bearing m-OCF3 or m-OCF2H group in the C2-benzoate moiety was designed, synthesized and examined for their potencies. A number of these fluorotaxoids possess two orders of magnitude greater potency in different drug-resistant cancer cell lines as compared to paclitaxel. One of these next-generation fluorotaxoids, SB--121205wasselected for detailed mechanistic study against highly paclitaxel-resistant human breast cancer cell line, MCF-7/PTX, which disclosed a unique mechanism of action. Recently, glucosylceramide (GlcCer) synthesis emerged as a promising target for next-generation antifungal agents, especially against cryptococcosis, candidiasis and pulmonary aspergillosis. The HTP screening of compound libraries identified several acylhydrazones as hit compounds. In the case study (ii), fluoro-acylhydrazones containing F, OCF3, OCHF2, o-F/p-OCF3, as well as o-F/p-CF3 functional groups in the ring A and ring B were designed based on these hit compounds, synthesized and examined for their potencies against C. neoformans. A number of those novel fluoro-acylhydrazones exhibited high potency and excellent killing properties. The hemopexin-like domain of matrix metalloproteinases (MMPs) is a highly promising target to circumvent the critical issue in the development of MMP inhibitors for the treatment of various cancers. In the case study (iii), a small optimization library of compounds, based on the OCHF2-containing hit compound, SB-M-001, was generated and evaluated, which identified a fluorine-containing new lead compound, SB-M-103. SB-M-103 was found to inhibit tumor cell growth, migration, and invasion by effectively disrupting the MMP-9 homodimerization.
Collapse
Affiliation(s)
- Krupanandan Haranahalli
- Institute of Chemical Biology & Drug Discovery, Stony Brook University–State University of New York, Stony Brook, NY, 11794-3400, U.S. A
| | - Tadashi Honda
- Department of Chemistry, Stony Brook University–State University of New York, Stony Brook, NY, 11794-3400, U. S. A
- Institute of Chemical Biology & Drug Discovery, Stony Brook University–State University of New York, Stony Brook, NY, 11794-3400, U.S. A
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University–State University of New York, Stony Brook, NY, 11794-3400, U. S. A
- Institute of Chemical Biology & Drug Discovery, Stony Brook University–State University of New York, Stony Brook, NY, 11794-3400, U.S. A
| |
Collapse
|
47
|
Nicola AM, Albuquerque P, Paes HC, Fernandes L, Costa FF, Kioshima ES, Abadio AKR, Bocca AL, Felipe MS. Antifungal drugs: New insights in research & development. Pharmacol Ther 2018; 195:21-38. [PMID: 30347212 DOI: 10.1016/j.pharmthera.2018.10.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The need for better antifungal therapy is commonly accepted in view of the high mortality rates associated with systemic infections, the low number of available antifungal classes, their associated toxicity and the increasing number of infections caused by strains with natural or acquired resistance. The urgency to expand the range of therapeutic options for the treatment of fungal infections has led researchers in recent decades to seek alternative antifungal targets when compared to the conventional ones currently used. Although new potential targets are reported, translating the discoveries from bench to bedside is a long process and most of these drugs fail to reach the patients. In this review, we discuss the development of antifungal drugs focusing on the approach of drug repurposing and the search for novel drugs for classical targets, the most recently described gene targets for drug development, the possibilities of immunotherapy using antibodies, cytokines, therapeutic vaccines and antimicrobial peptides.
Collapse
Affiliation(s)
| | - Patrícia Albuquerque
- Faculty of Ceilândia, University of Brasília, Brazil; Graduate Programme in Microbial Biology, University of Brasília, Brazil
| | - Hugo Costa Paes
- Division of Clinical Medicine, University of Brasília Medical School, Brazil
| | - Larissa Fernandes
- Faculty of Ceilândia, University of Brasília, Brazil; Graduate Programme in Microbial Biology, University of Brasília, Brazil
| | - Fabricio F Costa
- Graduate Programme in Genomic Science and Biotechnology, Catholic University of Brasília, Brazil; MATTER, Chicago, IL, USA; Cancer Biology and Epigenomics Program, Ann & Robert Lurie Children's Hospital of Chicago Research Center, Northwestern University's Feinberg School of Medicine, Chicago, Illinois, USA
| | - Erika Seki Kioshima
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Paraná, Brazil
| | - Ana Karina Rodrigues Abadio
- School for Applied Social and Agricultural Sciences, State University of Mato Grosso, Nova Mutum Campus, Mato Grosso, Brazil
| | | | - Maria Sueli Felipe
- Graduate Programme in Genomic Science and Biotechnology, Catholic University of Brasília, Brazil; Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brazil.
| |
Collapse
|
48
|
Mourad A, Perfect JR. Present and Future Therapy of Cryptococcus Infections. J Fungi (Basel) 2018; 4:jof4030079. [PMID: 29970809 PMCID: PMC6162641 DOI: 10.3390/jof4030079] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 01/05/2023] Open
Abstract
Cryptococcal infections burden the immunocompromised population with unacceptably high morbidity and mortality. This population includes HIV-infected individuals and those undergoing organ transplants, as well as seemingly immunocompetent patients (non-HIV, non-transplant). These groups are difficult to manage with the current therapeutic options and strategies, particularly in resource-limited settings. New trials aimed at providing the best treatment strategies for resource-limited countries that will reduce costs and adverse reactions have focused on decreasing the length of therapy and using more readily accessible antifungal agents such as fluconazole. Furthermore, the emergence of antifungal resistance poses another challenge for successful treatment and may require the development of new agents for improved management. This review will discuss the principles of management, current and future antifungal agents, as well as emerging techniques and future directions of care for this deadly infection.
Collapse
Affiliation(s)
- Ahmad Mourad
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | - John R Perfect
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
49
|
Abstract
Filamentous and dimorphic fungi cause invasive mycoses associated with high mortality rates. Among the fungal determinants involved in the establishment of infection, glycosphingolipids (GSLs) have gained increased interest in the last few decades. GSLs are ubiquitous membrane components that have been isolated from both filamentous and dimorphic species and play a crucial role in polarized growth as well as hypha-to-yeast transition. In fungi, two major classes of GSLs are found: neutral and acidic GSLs. Neutral GSLs comprise glucosylceramide and galactosylceramide, which utilize Δ4-Δ8-9-methyl-sphingadienine as a sphingoid base, linked to a C16-18 fatty acid chain, forming ceramide, and to a sugar residue, such as glucose or galactose. In contrast, acidic GSLs include glycosylinositol phosphorylceramides (GIPCs), composed of phytosphingosine attached to a long or very long fatty acid chain (C18-26) and to diverse and complex glycan groups via an inositol-phosphate linker. GIPCs are absent in mammalian cells, while fungal glucosylceramide and galactosylceramide are present but diverge structurally from their counterparts. Therefore, these compounds and their biosynthetic pathways represent potential targets for the development of selective therapeutic strategies. In this minireview, we discuss the enzymatic steps involved in the production of fungal GSLs, analyze their structure, and address the role of the currently characterized genes in the biology and pathogenesis of filamentous and dimorphic fungi.
Collapse
|