1
|
Wang H, Li H, Liu Z, Zhu Z, Cao Y. Activity of thonningianin A against Candida albicans in vitro and in vivo. Appl Microbiol Biotechnol 2024; 108:96. [PMID: 38212967 PMCID: PMC10784352 DOI: 10.1007/s00253-023-12996-1] [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: 09/06/2023] [Revised: 12/17/2023] [Accepted: 12/26/2023] [Indexed: 01/13/2024]
Abstract
Fungal infections are increasing rapidly, and antifungal agents used in clinics are limited. Therefore, novel antifungal agents with high efficiency are urgently required. In this study, we investigated the antifungal activity of thonningianin A (THA), a natural compound that is widely found in plants. We first determined the activity of THA against Candida albicans, one of the most common fungal pathogens, and found that THA showed antifungal activity against all C. albicans tested, including several fluconazole-resistant isolates. THA also inhibits the growth of non-Candida albicans species. In addition, THA displayed antibiofilm activity and could not only inhibit biofilm formation but also destroy mature biofilms. The in vivo antifungal efficacy of THA was confirmed in a Galleria mellonella infection model. Further studies revealed that THA could enhance intracellular reactive oxygen species (ROS) production and regulate the transcription of several redox-related genes. Specifically, caspase activity and expression of CaMCA1, a caspase-encoding gene in C. albicans, were remarkably increased upon THA treatment. Consistent with this, in the presence of THA, the Camca1 null mutant displayed higher survival rates and reduced caspase activity compared to the wild-type or CaMCA1-reintroduced strains, indicating an important role of CaMCA1 in the antifungal activity of THA. Taken together, our results indicate that THA possesses excellent antifungal activity and may be a promising novel antifungal candidate. KEY POINTS: • THA exhibits activity against Candida species, including fluconazole-resistant isolates • THA inhibits biofilm formation and destroys mature biofilm • Elevated ROS production and CaMCA1-mediated caspase activity are involved in the antifungal mechanisms of THA.
Collapse
Affiliation(s)
- Hui Wang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Hui Li
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, 200438, China
| | - ZhiWei Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - ZhenYu Zhu
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - YingYing Cao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.
| |
Collapse
|
2
|
Bédard C, Gagnon-Arsenault I, Boisvert J, Plante S, Dubé AK, Pageau A, Fijarczyk A, Sharma J, Maroc L, Shapiro RS, Landry CR. Most azole resistance mutations in the Candida albicans drug target confer cross-resistance without intrinsic fitness cost. Nat Microbiol 2024; 9:3025-3040. [PMID: 39379635 DOI: 10.1038/s41564-024-01819-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/27/2024] [Indexed: 10/10/2024]
Abstract
Azole antifungals are the main drugs used to treat fungal infections. Amino acid substitutions in the drug target Erg11 (Cyp51) are a common resistance mechanism in pathogenic yeasts. How many and which mutations confer resistance is, however, largely unknown. Here we measure the impact of nearly 4,000 amino acid variants of Candida albicans Erg11 on the susceptibility to six clinical azoles. This was achieved by deep mutational scanning of CaErg11 expressed in Saccharomyces cerevisiae. We find that a large fraction of mutations lead to resistance (33%), most resistance mutations confer cross-resistance (88%) and only a handful of resistance mutations show a significant fitness cost (9%). Our results reveal that resistance to azoles can arise through a large set of mutations and this will probably lead to azole pan-resistance, with little evolutionary compromise. This resource will help inform treatment choices in clinical settings and guide the development of new drugs.
Collapse
Affiliation(s)
- Camille Bédard
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Isabelle Gagnon-Arsenault
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Jonathan Boisvert
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Samuel Plante
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Alexandre K Dubé
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Alicia Pageau
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Anna Fijarczyk
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada
| | - Jehoshua Sharma
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Laetitia Maroc
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Christian R Landry
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada.
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada.
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada.
- PROTEO, Le regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Université Laval, Québec, Québec, Canada.
- Centre de Recherche sur les Données Massives (CRDM), Université Laval, Québec, Québec, Canada.
| |
Collapse
|
3
|
Fu X, Tian X, Lin J, Wang Q, Gu L, Wang Z, Chi M, Yu B, Feng Z, Liu W, Zhang L, Li C, Zhao G. Zeolitic Imidazolate Framework-8 Offers an Anti-Inflammatory and Antifungal Method in the Treatment of Aspergillus Fungal Keratitis in vitro and in vivo. Int J Nanomedicine 2024; 19:11163-11179. [PMID: 39502641 PMCID: PMC11537184 DOI: 10.2147/ijn.s480800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
Background Fungal keratitis is a serious blinding eye disease. Traditional drugs used to treat fungal keratitis commonly have the disadvantages of low bioavailability, poor dispersion, and limited permeability. Purpose To develop a new method for the treatment of fungal keratitis with improved bioavailability, dispersion, and permeability. Methods Zeolitic Imidazolate Framework-8 (ZIF-8) was formed by zinc ions and 2-methylimidazole linked by coordination bonds and characterized by Scanning electron microscopy (SEM), X-ray diffraction (XRD), and Zeta potential. The safety of ZIF-8 on HCECs and RAW 264.7 cells was detected by Cell Counting Kit-8 (CCK-8). Safety evaluation of ZIF-8 on mice corneal epithelium was conducted using the Draize corneal toxicity test. The effects of ZIF-8 on fungal growth, biofilm formation, and hyphae structure were detected by Minimal inhibit concentration (MIC), crystal violet staining, Propidium Iodide (PI) testing, and calcofluor white staining. The anti-inflammatory effects of ZIF-8 on RAW 246.7 cells were evaluated by Quantitative Real-Time PCR Experiments (qPCR) and Enzyme-linked immunosorbent assay (ELISA). Clinical score, Colony-Forming Units (CFU), Hematoxylin-eosin (HE) staining, and immunofluorescence were conducted to verify the therapeutic effect of ZIF-8 on C57BL/6 female mice with fungal keratitis. Results In vitro, ZIF-8 showed outstanding antifungal effects, including inhibiting the growth of Aspergillus fumigatus over 90% at 64 μg/mL, restraining the formation of biofilm, and destroying cell membranes. In vivo, treatment with ZIF-8 reduced corneal fungal load and mitigated neutrophil infiltration in fungal keratitis, which effectively reduced the severity of keratitis in mice and alleviated the infiltration of inflammatory factors in the mouse cornea. In addition, ZIF-8 reduces the inflammatory response by downregulating the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β after Aspergillus fumigatus infection in vivo and in vitro. Conclusion ZIF-8 has a significant anti-inflammatory and antifungal effect, which provides a new solution for the treatment of fungal keratitis.
Collapse
Affiliation(s)
- Xueyun Fu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Ziyi Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Menghui Chi
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Bing Yu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Zhuhui Feng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Wenyao Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lina Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
4
|
Tanwar S, Kalra S, Bari VK. Insights into the role of sterol metabolism in antifungal drug resistance: a mini-review. Front Microbiol 2024; 15:1409085. [PMID: 39464401 PMCID: PMC11502366 DOI: 10.3389/fmicb.2024.1409085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Sterols are essential for eukaryotic cells and are crucial in cellular membranes' structure, function, fluidity, permeability, adaptability to environmental stressors, and host-pathogen interactions. Fungal sterol, such as ergosterol metabolism, involves several organelles, including the mitochondria, lipid droplets, endoplasmic reticulum, and peroxisomes that can be regulated mainly by feedback mechanisms and transcriptionally. The majority of sterol transport in yeast occurs via non-vesicular transport pathways mediated by lipid transfer proteins, which determine the quantity of sterol present in the cell membrane. Pathogenic fungi Candida, Aspergillus, and Cryptococcus species can cause a range of superficial to potentially fatal systemic and invasive infections that are more common in immunocompromised patients. There is a significant risk of morbidity and mortality from these infections, which are very difficult to cure. Several antifungal drugs with different modes of action have received clinical approval to treat fungal infections. Antifungal drugs targeting the ergosterol biosynthesis pathway are well-known for their antifungal activity; however, an imbalance in the regulation and transport of ergosterol could lead to resistance to antifungal therapy. This study summarizes how fungal sterol metabolism and regulation can modulate sterol-targeting antifungal drug resistance.
Collapse
|
5
|
Padilla Suarez EG, Siciliano A, Spampinato M, Maione A, Guida M, Libralato G, Galdiero E. Ecotoxicity and Mutagenicity Assessment of Novel Antifungal Agents VT-1161 and T-2307. Molecules 2024; 29:4739. [PMID: 39407667 PMCID: PMC11477875 DOI: 10.3390/molecules29194739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Antifungal substances are essential for managing fungal infections in humans, animals, and plants, and their usage has significantly increased due to the global rise in fungal infections. However, the extensive application of antifungal agents in pharmaceuticals, personal care products, and agriculture has led to their widespread environmental dissemination through various pathways, such as excretion, improper disposal, and agricultural runoff. Despite advances in wastewater treatment, many antifungal compounds persist in the environment, affecting non-target organisms and contributing to resistance development. This study investigates the environmental impact of two novel antifungal agents, VT-1161 and T-2307, recently introduced as alternatives for treating resistant Candida spp. We assessed their ecotoxicity and mutagenicity using multiple bioassays: immobilization of Daphnia magna, growth inhibition of Raphidocelis subcapitata, luminescence inhibition of Aliivibrio fischeri, and mutagenicity on Salmonella typhimurium strain TA100. Results indicate that both VT-1161 and T-2307 exhibit lower toxicity compared to existing antifungal compounds, with effective concentrations (EC50) causing 50% response ranging from 14.34 to 27.92 mg L-1. Furthermore, both agents were classified as less hazardous based on the Globally Harmonized System of Classification and Labeling of Chemicals. Despite these favorable results, further research is needed to understand their environmental behavior, interactions, and potential resistance development among non-target species. Our findings highlight the importance of comprehensive environmental risk assessments to ensure the sustainable use of new antifungal agents.
Collapse
Affiliation(s)
- Edith Guadalupe Padilla Suarez
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Antonietta Siciliano
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Marisa Spampinato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Angela Maione
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Marco Guida
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giovanni Libralato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Emilia Galdiero
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| |
Collapse
|
6
|
Sun Y, Liu R, Luo Z, Zhang J, Gao Z, Liu R, Liu N, Zhang H, Li K, Wu X, Yin W, Qin Q, Su X, Zhao D, Cheng M. Identification of novel and potent triazoles targeting CYP51 for antifungal: Design, synthesis, and biological study. Eur J Med Chem 2024; 280:116942. [PMID: 39369483 DOI: 10.1016/j.ejmech.2024.116942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Invasive fungal infections (IFIs) are emerging as a serious infectious disease worldwide. Due to the lack of effective antifungal drugs and serious drug resistance, the number of people with low immunity is increasing, leading to high morbidity and mortality. Azole drugs targeting CYP51 are widely used in the treatment of invasive fungal infections. By analyzing representative azole antifungal drugs, the characteristics of pharmacophore were summarized. The binding mode of lead compound Iodiconazole was analyzed, and it was found that the narrow hydrophobic cavity was not fully occupied. Therefore, a series of triazole compounds were designed and synthesized by fragment growth strategy. Most of the compounds showed strong inhibitory activity against pathogenic fungi, among which compound A33 showed excellent inhibitory activity against pathogenic fungi and drug-resistant strains. In addition, the preferred compound A33 can prevent fungal phase transition, the formation of fungal biofilm, and show satisfactory fungicidal activity. In addition, the compound A33 was almost non-toxic to mammalian HUVEC cell. These results strongly suggested that compound A33 was worth further investigation as a potential azole inhibitor.
Collapse
Affiliation(s)
- Yixiang Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Rongrong Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Zirui Luo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Jiachen Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Zixuan Gao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Rui Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Nian Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Haoyu Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Kejian Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Xudong Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Wenbo Yin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Qiaohua Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Xin Su
- The School of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Dongmei Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| |
Collapse
|
7
|
Wu X, Zhang J, Liu R, Sun Y, Gao Z, Zhang G, Luo Z, Li K, Qin Q, Liu N, Zhang H, Su X, Zhao D, Cheng M. Design, synthesis and bioactivity evaluation of triazole antifungal drugs with phenylthiophene structure. Bioorg Chem 2024; 153:107785. [PMID: 39255609 DOI: 10.1016/j.bioorg.2024.107785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/28/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024]
Abstract
Invasive fungal infections have high morbidity and mortality rates and have become one of the most serious threats to human health. In the present study, a series of triazole antifungal derivatives with phenylthiophene backbone were obtained by structural modification of the lead compound using Iodiconazole as the lead compound. Among them, compound 19g is a triazole antifungal compound with 4-chloro-2-fluoro phenylthiophene backbone, which showed optimal antifungal activity against Candida albicans, Cryptococcus neoformans, and Aspergillus, with a MIC80 value of 0.0625 μg/mL. In addition, compounds 19e, 19f, 19g, 19h, 19i and 19k exhibited different levels of inhibitory activity against fluconazole-resistant strains with MIC80 values ranging from 0.0625 μg/mL to 32 μg/mL. Since compound 19g had optimal in vitro antifungal activity, we selected 19g for human liver microsomal stability and CYP enzyme inhibition assays as well as further evaluated the inhibitory activity of compound 19g on normal and cancerous cells in humans. Finally, we verified the inhibitory effect of compound 19g on the filamentation of Candida albicans and determined the mechanism of action by sterol composition analysis.
Collapse
Affiliation(s)
- Xudong Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Jiachen Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Rongrong Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yixiang Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Zixuan Gao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Guoqi Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Zirui Luo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Kejian Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Qiaohua Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Nian Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Haoyu Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xin Su
- The School of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Dongmei Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| |
Collapse
|
8
|
Zhen C, Wang L, Feng Y, Whiteway M, Hang S, Yu J, Lu H, Jiang Y. Otilonium Bromide Exhibits Potent Antifungal Effects by Blocking Ergosterol Plasma Membrane Localization and Triggering Cytotoxic Autophagy in Candida Albicans. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406473. [PMID: 38995235 PMCID: PMC11425263 DOI: 10.1002/advs.202406473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Indexed: 07/13/2024]
Abstract
Candidiasis, which presents a substantial risk to human well-being, is frequently treated with azoles. However, drug-drug interactions caused by azoles inhibiting the human CYP3A4 enzyme, together with increasing resistance of Candida species to azoles, represent serious issues with this class of drug, making it imperative to develop innovative antifungal drugs to tackle this growing clinical challenge. A drug repurposing approach is used to examine a library of Food and Drug Administration (FDA)-approved drugs, ultimately identifying otilonium bromide (OTB) as an exceptionally encouraging antifungal agent. Mechanistically, OTB impairs vesicle-mediated trafficking by targeting Sec31, thereby impeding the plasma membrane (PM) localization of the ergosterol transporters, such as Sip3. Consequently, OTB obstructs the movement of ergosterol across membranes and triggers cytotoxic autophagy. It is noteworthy that C. albicans encounters challenges in developing resistance to OTB because it is not a substrate for drug transporters. This study opens a new door for antifungal therapy, wherein OTB disrupts ergosterol subcellular distribution and induces cytotoxic autophagy. Additionally, it circumvents the hepatotoxicity associated with azole-mediated liver enzyme inhibition and avoids export-mediated drug resistance in C. albicans.
Collapse
Affiliation(s)
- Cheng Zhen
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Li Wang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Yanru Feng
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Malcolm Whiteway
- Department of BiologyConcordia UniversityMontrealQCH4B 1R6Canada
| | - Sijin Hang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Jinhua Yu
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's HospitalSchool of MedicineTongji UniversityNo.1239 Siping RoadShanghai200092China
| |
Collapse
|
9
|
Redrado-Hernández S, Macías-León J, Castro-López J, Belén Sanz A, Dolader E, Arias M, González-Ramírez AM, Sánchez-Navarro D, Petryk Y, Farkaš V, Vincke C, Muyldermans S, García-Barbazán I, Del Agua C, Zaragoza O, Arroyo J, Pardo J, Gálvez EM, Hurtado-Guerrero R. Broad Protection against Invasive Fungal Disease from a Nanobody Targeting the Active Site of Fungal β-1,3-Glucanosyltransferases. Angew Chem Int Ed Engl 2024; 63:e202405823. [PMID: 38856634 DOI: 10.1002/anie.202405823] [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: 04/01/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/11/2024]
Abstract
Invasive fungal disease accounts for about 3.8 million deaths annually, an unacceptable rate that urgently prompts the discovery of new knowledge-driven treatments. We report the use of camelid single-domain nanobodies (Nbs) against fungal β-1,3-glucanosyltransferases (Gel) involved in β-1,3-glucan transglycosylation. Crystal structures of two Nbs with Gel4 from Aspergillus fumigatus revealed binding to a dissimilar CBM43 domain and a highly conserved catalytic domain across fungal species, respectively. Anti-Gel4 active site Nb3 showed significant antifungal efficacy in vitro and in vivo prophylactically and therapeutically against different A. fumigatus and Cryptococcus neoformans isolates, reducing the fungal burden and disease severity, thus significantly improving immunocompromised animal survival. Notably, C. deneoformans (serotype D) strains were more susceptible to Nb3 and genetic Gel deletion than C. neoformans (serotype A) strains, indicating a key role for β-1,3-glucan remodelling in C. deneoformans survival. These findings add new insight about the role of β-1,3-glucan in fungal biology and demonstrate the potential of nanobodies in targeting fungal enzymes to combat invasive fungal diseases.
Collapse
Grants
- PID2022-136362NB-I00 Ministerio de Asuntos Económicos y Transformación Digital, Gobierno de España
- BIO2016-79289-P Ministerio de Economía y Competitividad, Gobierno de España
- PID2019-105223GB-I00 Ministerio de Ciencia, Innovación y Universidades y Agencia Estatal de Investigación, Gobierno de España
- PID2022-136888NB-I00 Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- PID2020-114546RB Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- PID2020-113963RB-I00 Ministerio de Ciencia e Innovación y Agencia Estatal de Investigación, Gobierno de España
- S2017/BMD3691-InGEMICS-CM Comunidad de Madrid
- B29_17R, E34_R17, LMP58_18 and LMP139_21 Gobierno de Aragon
- Nanofungi Precipita (crowdfunding)
- BIOSTRUCTX_5186 FP7 (2007-2013), BioStruct-X
Collapse
Affiliation(s)
- Sergio Redrado-Hernández
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
| | - Javier Macías-León
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Jorge Castro-López
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Ana Belén Sanz
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Elena Dolader
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Maykel Arias
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Andrés Manuel González-Ramírez
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - David Sánchez-Navarro
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
| | - Yuliya Petryk
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Vladimír Farkaš
- Department of Glycobiology, Institute of Chemistry, Center for Glycomics, Slovak Academy of Sciences, 84538, Bratislava, Slovakia
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Irene García-Barbazán
- Mycology Reference Laboratory. National Centre for Microbiology., Health Institute Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Celia Del Agua
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, IIS-Aragón, 50009, Zaragoza, Spain
| | - Oscar Zaragoza
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Mycology Reference Laboratory. National Centre for Microbiology., Health Institute Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Javier Arroyo
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Julián Pardo
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
- Department of Microbiology, Pediatry, Radiology and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Eva M Gálvez
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
- Center for Biomedical Research in Network in Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029, Madrid, Spain
| | - Ramon Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I+D, 50018, Zaragoza, Spain
- Fundación ARAID, 50018, Zaragoza, Spain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200, Copenhagen, Denmark
| |
Collapse
|
10
|
de Sousa Cutrim TA, Barcelos FF, Meireles LM, Rodrigues Gazolla PA, Almeida Lima ÂM, Teixeira RR, Moreira LC, de Queiroz VT, Almeida Barbosa LC, Bezerra Morais PA, do Nascimento CJ, Junker J, Costa AV, Fronza M, Scherer R. Design, synthesis, docking studies and bioactivity evaluation of 1,2,3-triazole eugenol derivatives. Future Med Chem 2024; 16:1883-1897. [PMID: 39157870 PMCID: PMC11486170 DOI: 10.1080/17568919.2024.2385292] [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: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Aim: The design, synthesis, docking studies and evaluation of the in vitro antifungal and cytotoxic properties of eugenol (EUG) containing 1,2,3-triazole derivatives are reported. Most of the derivatives have not been reported.Materials & methods: The EUG derivatives were synthesized, molecular docked and tested for their antifungal activity.Results: The compounds showed potent antifungal activity against Trichophyton rubrum, associated with dermatophytosis. Compounds 2a and 2i exhibited promising results, with 2a being four-times more potent than EUG. The binding mode prediction was similar to itraconazole in the lanosterol-14-α-demethylase wild-type and G73E mutant binding sites. Additionally, the pharmacokinetic profile prediction suggests good gastrointestinal absorption and potential oral administration.Conclusion: Compound 2a is a promising antifungal agent against dermatophytosis caused by T. rubrum.
Collapse
Affiliation(s)
- Thiago Antonio de Sousa Cutrim
- Universidade de Vila Velha, Departamento de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
| | - Fernando Fontes Barcelos
- Universidade de Vila Velha, Programa de Pós-Graduação em Biotecnologia Vegetal, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
| | - Leandra Martins Meireles
- Universidade de Vila Velha, Departamento de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
| | - Poliana Aparecida Rodrigues Gazolla
- Departamento de Química e Física, Grupo de Pesquisa de Estudos Aplicados em Produtos Naturais e Síntese Orgânica (GEAPS), Universidade Federal do Espírito Santo, Alto Universitário, s/n, Alegre, Espírito Santo State, 29500-000, Brazil
| | - Ângela Maria Almeida Lima
- Departamento de Química e Física, Grupo de Pesquisa de Estudos Aplicados em Produtos Naturais e Síntese Orgânica (GEAPS), Universidade Federal do Espírito Santo, Alto Universitário, s/n, Alegre, Espírito Santo State, 29500-000, Brazil
| | - Róbson Ricardo Teixeira
- Departamento de Química, Grupo de Síntese e Pesquisa de Compostos Bioativos (GSPCB), Universidade Federal de Viçosa, Av. P.H. Rolfs, s/nViçosa, Minas Gerais State, 36570-900, Brazil
| | - Luiza Carvalheira Moreira
- Departamento de Química, Grupo de Síntese e Pesquisa de Compostos Bioativos (GSPCB), Universidade Federal de Viçosa, Av. P.H. Rolfs, s/nViçosa, Minas Gerais State, 36570-900, Brazil
| | - Vagner Tebaldi de Queiroz
- Departamento de Química e Física, Grupo de Pesquisa de Estudos Aplicados em Produtos Naturais e Síntese Orgânica (GEAPS), Universidade Federal do Espírito Santo, Alto Universitário, s/n, Alegre, Espírito Santo State, 29500-000, Brazil
| | - Luiz Cláudio Almeida Barbosa
- Departamento de Química, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Belo Horizonte, Minas Gerais State, 31270-901, Brazil
| | - Pedro Alves Bezerra Morais
- Departamento de Química e Física, Grupo de Pesquisa de Estudos Aplicados em Produtos Naturais e Síntese Orgânica (GEAPS), Universidade Federal do Espírito Santo, Alto Universitário, s/n, Alegre, Espírito Santo State, 29500-000, Brazil
| | - Cláudia Jorge do Nascimento
- Departamento de Ciências Naturais, Instituto de Biociências, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Av. Pauster, Rio de Janeiro, Rio de Janeiro State, 22290-240, Brazil
| | - Jochen Junker
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Av. Brasil, 4365, Rio de Janeiro, Rio de Janeiro State, 21040-900, Brazil
| | - Adilson Vidal Costa
- Departamento de Química e Física, Grupo de Pesquisa de Estudos Aplicados em Produtos Naturais e Síntese Orgânica (GEAPS), Universidade Federal do Espírito Santo, Alto Universitário, s/n, Alegre, Espírito Santo State, 29500-000, Brazil
| | - Marcio Fronza
- Universidade de Vila Velha, Departamento de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
- Universidade de Vila Velha, Programa de Pós-Graduação em Biotecnologia Vegetal, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
| | - Rodrigo Scherer
- Universidade de Vila Velha, Departamento de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
- Universidade de Vila Velha, Programa de Pós-Graduação em Biotecnologia Vegetal, Av. Comissário José Dantas de Melo, 21, Vila Velha, Espírito Santo State, 29102-770, Brazil
| |
Collapse
|
11
|
Jäger MC, González-Ruiz V, Joos FL, Winter DV, Boccard J, Degenhardt T, Brand S, Rudaz S, Thompson GR, Odermatt A. Assessment of the potential risk of oteseconazole and two other tetrazole antifungals to inhibit adrenal steroidogenesis and peripheral metabolism of corticosteroids. Front Pharmacol 2024; 15:1394846. [PMID: 39175536 PMCID: PMC11338861 DOI: 10.3389/fphar.2024.1394846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
The triazole antifungals posaconazole and itraconazole can cause pseudohyperaldosteronism with hypertension and hypokalemia, edema, and gynecomastia by inhibiting steroid synthesis and metabolism. Mechanisms underlying pseudohyperaldosteronism include inhibition of adrenal 11β-hydroxylase cytochrome-P450 (CYP) 11B1 and 17α-hydroxylase (CYP17A1) as well as peripherally expressed 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2). To enhance specificity for fungal CYP51, tetrazoles have been developed. This study employed H295R adrenocortical cells and enzyme activity assays to assess the potential risk of oteseconazole and two other tetrazoles, VT-1598 and quilseconazole, to inhibit adrenal steroidogenesis or 11β-HSD2. Steroidomic footprint analyses of H295R cell supernatants using untargeted liquid-chromatography-high-resolution mass-spectrometry (LC-HRMS) indicated overall patterns common to oteseconazole, quilseconazole and itraconazole, as well as similarities between VT-1598 and isavuconazole. Additionally, more specific features of the steroid signatures were observed. Targeted quantification of nine adrenal steroids in supernatants from treated H295R cells revealed an overall inhibition of adrenal steroidogenesis by the three tetrazoles, itraconazole and isavuconazole, providing an explanation for their similar steroidomic pattern. Applying recombinant enzymes indicated that this effect is not due to direct inhibition of steroidogenic enzymes because no or only weak inhibition could be observed. Moreover, oteseconazole and the two other tetrazoles did not inhibit 11β-HSD2, suggesting that they do not pose a risk of pseudohyperaldosteronism. Furthermore, oteseconazole did not alter steroid concentrations in a recent clinical study. Nevertheless, follow-up studies should assess the mechanism underlying the observed overall steroidogenesis inhibition by tetrazoles, itraconazole and isavuconazole, and whether concentrations achievable in a subgroup of susceptible patients might cause adrenal insufficiency and hyperplasia.
Collapse
Affiliation(s)
- Marie-Christin Jäger
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Víctor González-Ruiz
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Friedrich L. Joos
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Denise V. Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Thorsten Degenhardt
- Mycovia Pharmaceuticals Inc., Imperial Business Park, Durham, NC, United States
| | - Steve Brand
- Mycovia Pharmaceuticals Inc., Imperial Business Park, Durham, NC, United States
| | - Serge Rudaz
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - George R. Thompson
- Department of Internal Medicine, Division of Infectious Diseases, University of California–Davis Health, Sacramento, CA, United States
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
12
|
Yan Z, Li Q, Li X, Wang H, Zhao D, Yu H, Guo M, Wang Y, Wang X, Xu H, Mou Y, Hou Z, Guo C. Discovery of Novel α,β-Unsaturated Amide Derivatives as Candidate Antifungals to Overcome Fungal Resistance. J Med Chem 2024. [PMID: 39077891 DOI: 10.1021/acs.jmedchem.4c00593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
In our previous study, coumarin-containing CYP51 inhibitor A32 demonstrated potent antiresistance activity. However, compound A32 demonstrated unsatisfied metabolic stability, necessitating modifications to overcome these limitations. In this study, α,β-unsaturated amides were used to replace the unstable coumarin ring, which increased metabolic stability by four times while maintaining antifungal activity, including activity against resistant strains. Subsequently, the sterol composition analysis and morphological observation experiments indicated that the target of these novel compounds is lanosterol 14α-demethylase (CYP51). Meanwhile, biofilm growth was inhibited and resistance genes (ERG11, CDR1, CDR2, and MDR1) expression was downregulated to find out how the antiresistance works. Importantly, compound C07 demonstrated the capacity to stimulate reactive oxygen species, thus displaying potent fungicidal activity. Moreover, C07 exhibited encouraging effectiveness in vivo following intraperitoneal administration. Additionally, the most potent compound C07 showed satisfactory pharmacokinetic properties and low toxicity. These α,β-unsaturated amide derivatives, particularly C07, are potential candidates for treating azole-resistant candidiasis.
Collapse
Affiliation(s)
- Zhongzuo Yan
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qi Li
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinyu Li
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huanlin Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongze Zhao
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hao Yu
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengbi Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yitong Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hang Xu
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanhua Mou
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhuang Hou
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chun Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
13
|
Wani MY, Srivastava V, El-Said WA, Al-Bogami AS, Ahmad A. Inhibition of apoptosis and biofilm formation in Candida auris by click-synthesized triazole-bridged quinoline derivatives. RSC Adv 2024; 14:21190-21202. [PMID: 38966810 PMCID: PMC11223670 DOI: 10.1039/d4ra03728f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024] Open
Abstract
Candida auris, a recent addition to the Candida species, poses a significant threat with its association to numerous hospital outbreaks globally, particularly affecting immunocompromised individuals. Given its resistance to existing antifungal therapies, there is a pressing need for innovative treatments. In this study, novel triazole bridged quinoline derivatives were synthesized and evaluated for their antifungal activity against C. auris. The most promising compound, QT7, demonstrated exceptional efficacy with a minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) of 0.12 μg mL-1 and 0.24 μg mL-1, respectively. Additionally, QT7 effectively disrupted mature biofilms, inhibiting them by 81.98% ± 8.51 and 89.57 ± 5.47 at MFC and 2× MFC values, respectively. Furthermore, QT7 induced cellular apoptosis in a dose-dependent manner, supported by various apoptotic markers such as phosphatidylserine externalization, mitochondrial depolarization, and reduced cytochrome c and oxidase activity. Importantly, QT7 exhibited low hemolytic activity, highlighting its potential for further investigation. Additionally, the physicochemical properties of this compound suggest its potential as a lead drug candidate, warranting further exploration in drug discovery efforts against Candida auris infections.
Collapse
Affiliation(s)
- Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah 21589 Jeddah Saudi Arabia
| | - Vartika Srivastava
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand South Africa
| | - Waleed Ahmed El-Said
- Department of Chemistry, College of Science, University of Jeddah 21589 Jeddah Saudi Arabia
| | | | - Aijaz Ahmad
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand South Africa
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh Medical Center Pittsburgh PA 15213 USA
| |
Collapse
|
14
|
de Sousa Cutrim TA, Eloy MA, Barcelos FF, Meireles LM, de Freitas Ferreira LC, Reis TA, Gonçalves SS, Lacerda V, Fronza M, Morais PAB, Scherer R. New thymol-derived triazole exhibits promising activity against Trichophyton rubrum. Braz J Microbiol 2024; 55:1287-1295. [PMID: 38453819 PMCID: PMC11153403 DOI: 10.1007/s42770-024-01295-0] [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: 05/10/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Fungal infections have emerged worldwide, and azole antifungals are widely used to control these infections. However, the emergence of antifungal resistance has been compromising the effectiveness of these drugs. Therefore, the objective of this study was to evaluate the antifungal and cytotoxic activities of the nine new 1,2,3 triazole compounds derived from thymol that were synthesized through Click chemistry. The binding mode prediction was carried out by docking studies using the crystallographic structure of Lanosterol 14α-demethylase G73E mutant from Saccharomyces cerevisiae. The new compounds showed potent antifungal activity against Trichophyton rubrum but did not show relevant action against Aspergillus fumigatus and Candida albicans. For T. rubrum, molecules nº 5 and 8 showed promising results, emphasizing nº 8, whose fungicidal and fungistatic effects were similar to fluconazole. In addition, molecule nº 8 showed low toxicity for keratinocytes and fibroblasts, concluding that this compound demonstrates promising characteristics for developing a new drug for dermatophytosis caused by T. rubrum, or serves as a structural basis for further research.
Collapse
Affiliation(s)
- Thiago Antonio de Sousa Cutrim
- Pharmaceutical Sciences Graduate Program, Universidade Vila Velha, Comissário José Dantas de Melo St., 21, Boa Vista, Vila Velha, Espírito Santo, 29102-770, Brazil
| | - Mariana Alves Eloy
- Agrochemical Graduate Program, Federal University of Espírito Santo, Alegre, Espirito Santo, 29500-000, Brazil
| | - Fernando Fontes Barcelos
- Plant Biotechnology Graduate Program, Universidade Vila Velha, Vila Velha, Espírito Santo, 29102-770, Brazil
| | - Leandra Martins Meireles
- Pharmaceutical Sciences Graduate Program, Universidade Vila Velha, Comissário José Dantas de Melo St., 21, Boa Vista, Vila Velha, Espírito Santo, 29102-770, Brazil
| | | | - Tatiana Alves Reis
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sarah Santos Gonçalves
- Center for Research in Medical Mycology, Department of Pathology, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Valdemar Lacerda
- Chemistry Graduate Program, Universidade Federal Do Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Marcio Fronza
- Pharmaceutical Sciences Graduate Program, Universidade Vila Velha, Comissário José Dantas de Melo St., 21, Boa Vista, Vila Velha, Espírito Santo, 29102-770, Brazil
| | - Pedro Alves Bezerra Morais
- Agrochemical Graduate Program, Federal University of Espírito Santo, Alegre, Espirito Santo, 29500-000, Brazil.
| | - Rodrigo Scherer
- Pharmaceutical Sciences Graduate Program, Universidade Vila Velha, Comissário José Dantas de Melo St., 21, Boa Vista, Vila Velha, Espírito Santo, 29102-770, Brazil.
| |
Collapse
|
15
|
Tahghighi A, Azerang P. Click chemistry beyond metal-catalyzed cycloaddition as a remarkable tool for green chemical synthesis of antifungal medications. Chem Biol Drug Des 2024; 103:e14555. [PMID: 38862260 DOI: 10.1111/cbdd.14555] [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: 12/13/2023] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Click chemistry is widely used for the efficient synthesis of 1,4-disubstituted-1,2,3-triazole, a well-known scaffold with widespread biological activity in the pharmaceutical sciences. In recent years, this magic ring has attracted the attention of scientists for its potential in designing and synthesizing new antifungal agents. Despite scientific and medical advances, fungal infections still account for more than 1.5 million deaths globally per year, especially in people with compromised immune function. This increasing trend is definitely related to a raise in the incidence of fungal infections and prevalence of antifungal drug resistance. In this condition, an urgent need for new alternative antifungals is undeniable. By focusing on the main aspects of reaction conditions in click chemistry, this review was conducted to classify antifungal 1,4-disubstituted-1,2,3-triazole hybrids based on their chemical structures and introduce the most effective triazole antifungal derivatives. It was notable that in all reactions studied, Cu(I) catalysts generated in situ by the reduction in Cu(II) salts or used copper(I) salts directly, as well as mixed solvents of t-BuOH/H2O and DMF/H2O had most application in the synthesis of triazole ring. The most effective antifungal activity was also observed in fluconazole analogs containing 1,2,3-triazole moiety and benzo-fused five/six-membered heterocyclic conjugates with a 1,2,3-triazole ring, even with better activity than fluconazole. The findings of structure-activity relationship and molecular docking of antifungal derivatives synthesized with copper-catalyzed azide-alkyne cycloaddition (CuAAC) could offer medicinal chemistry scientists valuable data on designing and synthesizing novel triazole antifungals with more potent biological activities in their future research.
Collapse
Affiliation(s)
- Azar Tahghighi
- Medicinal Chemistry Laboratory, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Parisa Azerang
- Medicinal Chemistry Laboratory, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
16
|
Argüelles JC, Sánchez-Fresneda R, Argüelles A, Solano F. Natural Substances as Valuable Alternative for Improving Conventional Antifungal Chemotherapy: Lights and Shadows. J Fungi (Basel) 2024; 10:334. [PMID: 38786689 PMCID: PMC11122340 DOI: 10.3390/jof10050334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Fungi are eukaryotic organisms with relatively few pathogenic members dangerous for humans, usually acting as opportunistic infections. In the last decades, several life-threatening fungal infections have risen mostly associated with the worldwide extension of chronic diseases and immunosuppression. The available antifungal therapies cannot combat this challenge because the arsenal of compounds is scarce and displays low selective action, significant adverse effects, and increasing resistance. A growing isolation of outbreaks triggered by fungal species formerly considered innocuous is being recorded. From ancient times, natural substances harvested from plants have been applied to folk medicine and some of them recently emerged as promising antifungals. The most used are briefly revised herein. Combinations of chemotherapeutic drugs with natural products to obtain more efficient and gentle treatments are also revised. Nevertheless, considerable research work is still necessary before their clinical use can be generally accepted. Many natural products have a highly complex chemical composition, with the active principles still partially unknown. Here, we survey the field underlying lights and shadows of both groups. More studies involving clinical strains are necessary, but we illustrate this matter by discussing the potential clinical applications of combined carnosic acid plus propolis formulations.
Collapse
Affiliation(s)
- Juan Carlos Argüelles
- Área de Microbiología, Facultad Biología, University Murcia, Campus Espinardo, 30100 Murcia, Spain; (J.C.A.); (R.S.-F.); (A.A.)
| | - Ruth Sánchez-Fresneda
- Área de Microbiología, Facultad Biología, University Murcia, Campus Espinardo, 30100 Murcia, Spain; (J.C.A.); (R.S.-F.); (A.A.)
| | - Alejandra Argüelles
- Área de Microbiología, Facultad Biología, University Murcia, Campus Espinardo, 30100 Murcia, Spain; (J.C.A.); (R.S.-F.); (A.A.)
| | - Francisco Solano
- Departamento Bioquímica, Biología Molecular B & Inmunología, Facultad Medicina, University Murcia, Campus El Palmar, 30112 Murcia, Spain
| |
Collapse
|
17
|
Tan J, Zhang Z, Zheng D, Mu Y, Cao B, Yang J, Han L, Huang X. Structure-activity relationship and biofilm formation-related gene targets of oleanolic acid-type saponins from Pulsatilla chinensis against Candida albicans. Bioorg Chem 2024; 146:107311. [PMID: 38547720 DOI: 10.1016/j.bioorg.2024.107311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
In the course of our investigations of antifungal natural products, the structure-activity relationship and antifungal activities of oleanolic acid-type saponins (1-28) from Pulsatilla chinensis against human and plant pathogenic fungi were elucidated. The analysis of structure-activity relationship of oleanolic acid-type saponins showed that the free carboxyl at C-28 was essential for their antifungal activities; the free hydroxyl group at the C-23 site of oleanolic acid-type saponins played a crucial role in their antifungal activities; the oligosaccharide chain at C-3 oleanolic acid-type saponins showed significant effects on antifungal efficacy and a disaccharide or trisaccharide moiety at position C-3 displayed optimal antifungal activity. The typical saponin pulchinenoside B3 (16, PB3) displayed satisfactory antifungal activity against human and plant pathogenic fungi, especially, C. albicans with an MIC value of 12.5 μg/mL. Furthermore, PB3 could inhibit the biofilm formation of C. albicans through downregulating the expression of the integrated network of biofilm formation-associated transcription factors (Bcr1 Efg1, Ndt80, Brg1, Rob1 and Tec1) and adhesion-related target genes (HWP1, ALS1, and ALS3). Meanwhile, we found that PB3 could effectively destroy the mature biofilm of C. albicans by the oxidative damage and inducing mitochondria-mediated apoptosis in cells.
Collapse
Affiliation(s)
- Junfeng Tan
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Zengguang Zhang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Dan Zheng
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Yu Mu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Bixuan Cao
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Junwei Yang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Li Han
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China.
| | - Xueshi Huang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China; Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China.
| |
Collapse
|
18
|
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
|
19
|
Ni T, Hao Y, Ding Z, Chi X, Xie F, Wang R, Bao J, Yan L, Li L, Wang T, Zhang D, Jiang Y. Discovery of a Novel Potent Tetrazole Antifungal Candidate with High Selectivity and Broad Spectrum. J Med Chem 2024; 67:6238-6252. [PMID: 38598688 DOI: 10.1021/acs.jmedchem.3c02188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Thirty-one novel albaconazole derivatives were designed and synthesized based on our previous work. All compounds exhibited potent in vitro antifungal activities against seven pathogenic fungi. Among them, tetrazole compound D2 was the most potent antifungal with MIC values of <0.008, <0.008, and 2 μg/mL against Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus, respectively, the three most common and critical priority pathogenic fungi. In addition, compound D2 also exhibited potent activity against fluconazole-resistant C. auris isolates. Notably, compound D2 showed a lower inhibitory activity in vitro against human CYP450 enzymes as well as a lower inhibitory effect on the hERG K+ channel, indicating a low risk of drug-drug interactions and QT prolongation. Moreover, with improved pharmacokinetic profiles, compound D2 showed better in vivo efficacy than albaconazole at reducing fungal burden and extending the survival of C. albicans-infected mice. Taken together, compound D2 will be further investigated as a promising candidate.
Collapse
Affiliation(s)
- Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road ,Shanghai 200092, China
| | - Yumeng Hao
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Zichao Ding
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
- Department of Pharmacy, 927th Hospital of Joint Logistics Support Force, 3 Yushui Road ,Puer 665000, China
| | - Xiaochen Chi
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Xie
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Ruina Wang
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Junhe Bao
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai 200433, China
| | - Lan Yan
- School of Pharmacy, 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 200092, China
| | - Ting Wang
- School of Pharmacy, Naval Medical University, 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
- School of Pharmacy, Naval Medical University, 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
| |
Collapse
|
20
|
Iancu VI, Chiriac LF, Paun I, Pirvu F, Dinu C, Kim L, Pascu LF, Niculescu M. Occurrence and distribution of azole antifungal agents in eight urban Romanian waste water treatment plants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 920:170898. [PMID: 38369155 DOI: 10.1016/j.scitotenv.2024.170898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
Azole compounds are utilized to combat fungal infections in plants to protect them and also used for treating mycosis in humans. The LC-MS/MS method is a technique that combines liquid chromatography with tandem mass spectrometry for analysis of twelve azole compounds from wastewater (influent, effluent) and sewage sludge. The compounds were isolated from waste water using automatic extraction in the solid phase. Sludge samples were dried by lyophilization, after which they were subjected to ultrasound extraction with methanol. The quantification limits ranged from 0.3 ng/L (clotrimazole-CLO and prochloraz-PRO) to 1.5 ng/L (tetraconazole-TEB and penconazole-PEN), for wastewater samples and for sewage sludge, the LOQs ranged from 0.1 ng/g to 0.6 ng/g. High concentrations of climbazole-CLI (207-391 ng/L), tebuconazole (92-424 ng/L), and clotrimazole (6.9-93-ng/L) were observed in influent samples of the 8 urban wastewater treatment plants, followed by fluconazole (49.3-76.8 ng/L), and prochloraz (7.3-72 ng/L). The ∑Azoles had a maximum of 676 ng/L in the Galati effluent, followed by the Bucharest station 357 ng/L, and 345 ng/L in the Braila effluent. The highest value of the daily mass loading (input) level was observed for climbazole, 265 mg/day/1000 in Iasi station, followed by tebuconazole, 238 mg/day/1000 people in the Bucharest station, and 203 mg/day/1000 people for climbazole in the Targoviste station. The daily mass emission presented values between 0.7 and 247 mg/day/1000 people. The highest emissions were observed for climbazole, 247 mg/day/1000 people in Braila station; 174 mg/day/1000 people in the Iasi station and 129 mg/day/1000 people in the Bucharest station. The concentrations of climbazole detected in the effluent can present a high risk for the plants Lemna minor and Navicula pelliculosa. Clotrimazole may present a high risk to the plant Desmodesmus subspicatus and to the invertebrate Daphnia magna. PRO may present high risk to the invertebrate Mysidopsis Bahia.
Collapse
Affiliation(s)
- Vasile-Ion Iancu
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania.
| | - Laura-Florentina Chiriac
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Iuliana Paun
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Florinela Pirvu
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Cristina Dinu
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Lidia Kim
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Luoana Florentina Pascu
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| | - Marcela Niculescu
- National Research and Development Institute for Industrial Ecology-ECOIND, Drumul Podu Dambovitei Street, 71 -73, Sector 6, Bucharest, Romania
| |
Collapse
|
21
|
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
|
22
|
Paluch E, Bortkiewicz O, Widelski J, Duda-Madej A, Gleńsk M, Nawrot U, Lamch Ł, Długowska D, Sobieszczańska B, Wilk KA. A Combination of β-Aescin and Newly Synthesized Alkylamidobetaines as Modern Components Eradicating the Biofilms of Multidrug-Resistant Clinical Strains of Candida glabrata. Int J Mol Sci 2024; 25:2541. [PMID: 38473787 DOI: 10.3390/ijms25052541] [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: 12/30/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The current trend in microbiological research aimed at limiting the development of biofilms of multidrug-resistant microorganisms is increasingly towards the search for possible synergistic effects between various compounds. This work presents a combination of a naturally occurring compound, β-aescin, newly synthesized alkylamidobetaines (AABs) with a general structure-CnTMDAB, and antifungal drugs. The research we conducted consists of several stages. The first stage concerns determining biological activity (antifungal) against selected multidrug-resistant strains of Candida glabrata (C. glabrata) with the highest ability to form biofilms. The second stage of this study determined the activity of β-aescin combinations with antifungal compounds and alkylamidobetaines. In the next stage of this study, the ability to eradicate a biofilm on the polystyrene surface of the combination of β-aescin with alkylamidobetaines was examined. It has been shown that the combination of β-aescin and alkylamidobetaine can firmly remove biofilms and reduce their viability. The last stage of this research was to determine the safety regarding the cytotoxicity of both β-aescin and alkylamidobetaines. Previous studies on the fibroblast cell line have shown that C9 alkylamidobetaine can be safely used as a component of anti-biofilm compounds. This research increases the level of knowledge about the practical possibilities of using anti-biofilm compounds in combined therapies against C. glabrata.
Collapse
Affiliation(s)
- Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-376 Wroclaw, Poland
| | - Olga Bortkiewicz
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-376 Wroclaw, Poland
| | - Jarosław Widelski
- Department of Pharmacognosy with Medicinal Plants Garden, Lublin Medical University, 20-093 Lublin, Poland
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-376 Wroclaw, Poland
| | - Michał Gleńsk
- Department of Pharmacognosy and Herbal Medicines, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Urszula Nawrot
- Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Łukasz Lamch
- Department of Engineering and Technology of Chemical Processes, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Daria Długowska
- Department of Engineering and Technology of Chemical Processes, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Beata Sobieszczańska
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-376 Wroclaw, Poland
| | - Kazimiera A Wilk
- Department of Engineering and Technology of Chemical Processes, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| |
Collapse
|
23
|
Vanzolini T, Magnani M. Old and new strategies in therapy and diagnosis against fungal infections. Appl Microbiol Biotechnol 2024; 108:147. [PMID: 38240822 PMCID: PMC10799149 DOI: 10.1007/s00253-023-12884-8] [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: 06/26/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024]
Abstract
Fungal infections represent a serious global health threat. The new emerging pathogens and the spread of different forms of resistance are now hardly challenging the tools available in therapy and diagnostics. With the commonly used diagnoses, fungal identification is often slow and inaccurate, and, on the other hand, some drugs currently used as treatments are significantly affected by the decrease in susceptibility. Herein, the antifungal arsenal is critically summarized. Besides describing the old approaches and their mechanisms, advantages, and limitations, the focus is dedicated to innovative strategies which are designed, identified, and developed to take advantage of the discrepancies between fungal and host cells. Relevant pathways and their role in survival and virulence are discussed as their suitability as sources of antifungal targets. In a similar way, molecules with antifungal activity are reported as potential agents/precursors of the next generation of antimycotics. Particular attention was devoted to biotechnological entities, to their novelty and reliability, to drug repurposing and restoration, and to combinatorial applications yielding significant improvements in efficacy. KEY POINTS: • New antifungal agents and targets are needed to limit fungal morbidity and mortality. • Therapeutics and diagnostics suffer of delays in innovation and lack of targets. • Biologics, drug repurposing and combinations are the future of antifungal treatments.
Collapse
Affiliation(s)
- Tania Vanzolini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy.
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| |
Collapse
|
24
|
Taylor MB, Warwick AR, Skophammer R, Boyer JM, Geck RC, Gunkelman K, Walson M, Rowley PA, Dunham MJ. yEvo: A modular eukaryotic genetics and evolution research experience for high school students. Ecol Evol 2024; 14:e10811. [PMID: 38192907 PMCID: PMC10771926 DOI: 10.1002/ece3.10811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 11/26/2023] [Indexed: 01/10/2024] Open
Abstract
The resources for carrying out and analyzing microbial evolution experiments have become more accessible, making it possible to expand these studies beyond the research laboratory and into the classroom. We developed five connected, standards-aligned yeast evolution laboratory modules, called "yEvo," for high school students. The modules enable students to take agency in answering open-ended research questions. In Module 1, students evolve baker's yeast to tolerate an antifungal drug, and in subsequent modules, investigate how evolved yeasts adapted to this stressful condition at both the phenotype and genotype levels. We used pre- and post-surveys from 72 students at two different schools and post-interviews with students and teachers to assess our program goals and guide module improvement over 3 years. We measured changes in student conceptions, confidence in scientific practices, and interest in STEM careers. Students who participated in yEvo showed improvements in understanding of activity-specific concepts and reported increased confidence in designing a valid biology experiment. Student experimental data replicated literature findings and has led to new insights into antifungal resistance. The modules and provided materials, alongside "proof of concept" evaluation metrics, will serve as a model for other university researchers and K - 16 classrooms interested in engaging in open-ended research questions using yeast as a model system.
Collapse
Affiliation(s)
- M. Bryce Taylor
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
- Program in BiologyLoras CollegeDubuqueIowaUSA
| | - Alexa R. Warwick
- Department of Fisheries and WildlifeMichigan State UniversityEast LansingMichiganUSA
| | | | | | - Renee C. Geck
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Kristin Gunkelman
- Department of Teacher EducationMichigan State UniversityEast LansingMichiganUSA
| | - Margaux Walson
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Paul A. Rowley
- Department of Biological SciencesUniversity of IdahoMoscowIdahoUSA
| | - Maitreya J. Dunham
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
25
|
Revtovich S, Lyfenko A, Tkachev Y, Kulikova V, Koval V, Puchkov V, Anufrieva N, Solyev P, Morozova E. Anticandidal Activity of In Situ Methionine γ-Lyase-Based Thiosulfinate Generation System vs. Synthetic Thiosulfinates. Pharmaceuticals (Basel) 2023; 16:1695. [PMID: 38139821 PMCID: PMC10748059 DOI: 10.3390/ph16121695] [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: 10/19/2023] [Revised: 11/19/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Candida albicans and non-albicans Candida species are a common cause of human mucosal infections, as well as bloodstream infections and deep mycoses. The emergence of resistance of Candida spp. to antifungal drugs used in practice requires the search for new antimycotics. The present study unravels the antifungal potential of the synthetic dialk(en)ylthiosulfinates in comparison with an enzymatic in situ methionine γ-lyase-based thiosulfinate generation system (TGS). The kinetics of the TGS reaction, namely, the methionine γ-lyase-catalyzed β-elimination of S-alk(en)yl-L-cysteine sulfoxides, was investigated via 1H NMR spectroscopy for the first time, revealing fast conversion rates and the efficient production of anticandidal dialk(en)ylthiosulfinates. The anticandidal potential of this system vs. synthetic thiosulfinates was investigated through an in vitro assay. TGS proved to be more effective (MIC range 0.36-1.1 μg/mL) than individual substances (MIC range 0.69-3.31 μg/mL). The tested preparations had an additive effect with the commercial antimycotics fluconazole, amphotericin B and 5-flucytosine demonstrating a fractional inhibitory coefficient index in the range of 0.5-2 μg/mL. TGS can be regarded as an attractive candidate for the targeted delivery of antimycotic thiosulfinates and for further implementation onto medically implanted devices.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pavel Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (S.R.); (A.L.); (Y.T.); (V.K.); (V.K.); (V.P.); (N.A.)
| | - Elena Morozova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia; (S.R.); (A.L.); (Y.T.); (V.K.); (V.K.); (V.P.); (N.A.)
| |
Collapse
|
26
|
Freitas CG, Felipe MS. Candida albicans and Antifungal Peptides. Infect Dis Ther 2023; 12:2631-2648. [PMID: 37940816 PMCID: PMC10746669 DOI: 10.1007/s40121-023-00889-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
Candida albicans, a ubiquitous opportunistic fungal pathogen, plays a pivotal role in human health and disease. As a commensal organism, it normally resides harmlessly within the human microbiota. However, under certain conditions, C. albicans can transition into a pathogenic state, leading to various infections collectively known as candidiasis. With the increasing prevalence of immunocompromised individuals and the widespread use of invasive medical procedures, candidiasis has become a significant public health concern. The emergence of drug-resistant strains further complicates treatment options, highlighting the urgent need for alternative therapeutic strategies. Antifungal peptides (AFPs) have gained considerable attention as potential candidates for combating Candida spp. infections. These naturally occurring peptides possess broad-spectrum antimicrobial activity, including specific efficacy against C. albicans. AFPs exhibit several advantageous properties, such as rapid killing kinetics, low propensity for resistance development, and diverse mechanisms of action, making them promising alternatives to conventional antifungal agents. In recent years, extensive research has focused on discovering and developing novel AFPs with improved efficacy and selectivity against Candida species. Advances in biotechnology and synthetic peptide design have enabled the modification and optimization of natural peptides, enhancing their stability, bioavailability, and therapeutic potential. Nevertheless, several challenges must be addressed before AFPs can be widely implemented in clinical practice. These include optimizing peptide stability, enhancing delivery methods, overcoming potential toxicity concerns, and conducting comprehensive preclinical and clinical studies. This commentary presents a short overview of candidemia and AFP; articles and reviews published in the last 10 years were searched on The National Library of Medicine (National Center for Biotechnology Information-NIH-PubMed). The terms used were C. albicans infections, antimicrobial peptides, antifungal peptides, antifungal peptides mechanisms of action, candidemia treatments and guidelines, synthetic peptides and their challenges, and antimicrobial peptides in clinical trials as the main ones. Older publications were cited if they brought some relevant concept or helped to bring a perspective into our narrative. Articles older than 20 years and those that appeared in PubMed but did not match our goal to bring updated information about using antifungal peptides as an alternative to C. albicans infections were not considered.
Collapse
Affiliation(s)
- Camila G Freitas
- Higher Education Course in Food Technology, Instituto Federal de Brasília (IFB), Brasília, DF, Brazil
- Genomic Sciences and Biotechnology Graduate Program, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil
| | - Maria Sueli Felipe
- Genomic Sciences and Biotechnology Graduate Program, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil.
- Universidade de Brasília (UNB), Brasília, DF, Brazil.
| |
Collapse
|
27
|
Tu S, Zhang K, Wang N, Chu J, Yang L, Xie Z. Comparative study of posaconazole and voriconazole for primary antifungal prophylaxis in patients with pediatric acute leukemia. Sci Rep 2023; 13:18789. [PMID: 37914820 PMCID: PMC10620160 DOI: 10.1038/s41598-023-46328-0] [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: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 11/03/2023] Open
Abstract
Posaconazole and voriconazole are commonly used for preventing invasive fungal disease (IFD), but few studies compared posaconazole with voriconazole for primary antifungal prophylaxis (PAP) in pediatric acute leukemia. To compare posaconazole with voriconazole for PAP in pediatric acute leukemia. This retrospective observational study enrolled pediatric patients with non-M3 acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL) between December 2017 and November 2019 in the Second Affiliated Hospital of Anhui Medical University. The patients received voriconazole or posaconazole for PAP. The primary outcome was the breakthrough of IFD. The secondary outcome was the overall survival (OS) and IFD-free survival of patients. A total of the 275 patients were enrolled, of which 120 patients taking voriconazole (43.6%) and 155 patients taking posaconazole (56.4%). The breakthrough of IFD occurred in 19 (15.8%) patients taking voriconazole and in 12 (7.7%) patients taking posaconazole (P = 0.035). There was no significant differences in IFD-free survival (P = 0.336) or OS (P = 0.069) between the patients taking voriconazole and posaconazole. In the subgroup of AML patients, the OS of patients taking posaconazole was better than those receiving voriconazole (P = 0.017). Posaconazole and voriconazole were comparable for PAP in patients with pediatric acute leukemia regarding the OS and IFD-free survival, but posaconazole might achieve a lower IFD breakthrough rate.
Collapse
Affiliation(s)
- Songji Tu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China.
| | - Kunlong Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China
| | - Ningling Wang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China
| | - Jinhua Chu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China
| | - Linhai Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China
| | - Zhiwei Xie
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Economic and Technological Development Zone, Shushan District, Hefei, 230601, China
| |
Collapse
|
28
|
De Francesco MA. Drug-Resistant Aspergillus spp.: A Literature Review of Its Resistance Mechanisms and Its Prevalence in Europe. Pathogens 2023; 12:1305. [PMID: 38003770 PMCID: PMC10674884 DOI: 10.3390/pathogens12111305] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Infections due to the Aspergillus species constitute an important challenge for human health. Invasive aspergillosis represents a life-threatening disease, mostly in patients with immune defects. Drugs used for fungal infections comprise amphotericin B, triazoles, and echinocandins. However, in the last decade, an increased emergence of azole-resistant Aspergillus strains has been reported, principally belonging to Aspergillus fumigatus species. Therefore, both the early diagnosis of aspergillosis and its epidemiological surveillance are very important to establish the correct antifungal therapy and to ensure a successful patient outcome. In this paper, a literature review is performed to analyze the prevalence of Aspergillus antifungal resistance in European countries. Amphotericin B resistance is observed in 2.6% and 10.8% of Aspergillus fumigatus isolates in Denmark and Greece, respectively. A prevalence of 84% of amphotericin B-resistant Aspergillus flavus isolates is reported in France, followed by 49.4%, 35.1%, 21.7%, and 20% in Spain, Portugal, Greece, and amphotericin B resistance of Aspergillus niger isolates is observed in Greece and Belgium with a prevalence of 75% and 12.8%, respectively. The prevalence of triazole resistance of Aspergillus fumigatus isolates, the most studied mold obtained from the included studies, is 0.3% in Austria, 1% in Greece, 1.2% in Switzerland, 2.1% in France, 3.9% in Portugal, 4.9% in Italy, 5.3% in Germany, 6.1% in Denmark, 7.4% in Spain, 8.3% in Belgium, 11% in the Netherlands, and 13.2% in the United Kingdom. The mechanism of resistance is mainly driven by the TR34/L98H mutation. In Europe, no in vivo resistance is reported for echinocandins. Future studies are needed to implement the knowledge on the spread of drug-resistant Aspergillus spp. with the aim of defining optimal treatment strategies.
Collapse
Affiliation(s)
- Maria Antonia De Francesco
- Department of Molecular and Translational Medicine, Institute of Microbiology, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
29
|
Murcia-Galán RA, Durán SM, Leal-Pinto SM, Roa-Cordero MV, Vargas JD, Herrera LV, Muñoz-Castro A, MacLeod-Carey D, Naranjo TW, Rodríguez-Kessler PL, Hurtado JJ. Antifungal activity of Co(II) and Cu(II) complexes containing 1,3-bis(benzotriazol-1-yl)-propan-2-ol on the growth and virulence traits of fluconazole-resistant Candida species: synthesis, DFT calculations, and biological activity. BMC Chem 2023; 17:135. [PMID: 37817173 PMCID: PMC10563319 DOI: 10.1186/s13065-023-01037-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Relevant virulence traits in Candida spp. are associated with dimorphic change and biofilm formation, which became an important target to reduce antifungal resistance. In this work, Co(II) complexes containing a benzotriazole derivative ligand showed a promising capacity of reducing these virulence traits. These complexes exhibited higher antifungal activities than the free ligands against all the Candida albicans and non-albicans strains tested, where compounds 2 and 4 showed minimum inhibitory concentration values between 15.62 and 125 μg mL-1. Moreover, four complexes (2-5) of Co(II) and Cu(II) with benzotriazole ligand were synthesized. These compounds were obtained as air-stable solids and characterized by melting point, thermogravimetric analysis, infrared, Raman and ultraviolet/visible spectroscopy. The analysis of the characterization data allowed us to identify that all the complexes had 1:1 (M:L) stoichiometries. Additionally, Density Functional Theory calculations were carried out for 2 and 3 to propose a probable geometry of both compounds. The conformer Da of 2 was the most stable conformer according to the Energy Decomposition Analysis; while the conformers of 3 have a fluxional behavior in this analysis that did not allow us to determine the most probable conformer. These results provide an important platform for the design of new compounds with antifungal activities and the capacity to attack other target of relevance to reduce antimicrobial resistance.
Collapse
Affiliation(s)
- Ricardo A. Murcia-Galán
- Grupo de Investigación en Química Inorgánica, Catálisis y Bioinorgánica, Departamento de Química, Universidad de los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia
| | - Sandra M. Durán
- Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Calle 70 No. 55-210, Bucaramanga, Colombia
| | - Sandra M. Leal-Pinto
- Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Calle 70 No. 55-210, Bucaramanga, Colombia
| | - Martha V. Roa-Cordero
- Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Calle 70 No. 55-210, Bucaramanga, Colombia
| | - Jose D. Vargas
- Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Calle 70 No. 55-210, Bucaramanga, Colombia
| | - Laura V. Herrera
- Grupo Sistema Estomatognático Y Morfofisiología (SEMF), Departamento de Ciencias Básicas, Universidad Santo Tomás Seccional Bucaramanga, Carrera 27 No. 180-395, Bucaramanga, Colombia
| | - Alvaro Muñoz-Castro
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Bellavista 7, 8420524 Santiago, Chile
| | - Desmond MacLeod-Carey
- Facultad de Ingeniería, Instituto de Ciencias Químicas Aplicadas, Inorganic Chemistry and Molecular Materials Center, Universidad Autónoma de Chile, El Llano Subercaseaux 2801, Santiago, Chile
| | - Tonny W. Naranjo
- Experimental and Medical Micology Group, Corporación para Investigaciones Biológicas (CIB), 050010 Medellin, Colombia
- Facultad de Medicina, Universidad Pontificia Bolivariana, 050034 Medellín, Colombia
| | - Peter L. Rodríguez-Kessler
- Centro de Investigaciones en Óptica A.C., Loma del Bosque 115, Col. Lomas del Campestre, 37150 León, Guanajuato México
| | - John J. Hurtado
- Grupo de Investigación en Química Inorgánica, Catálisis y Bioinorgánica, Departamento de Química, Universidad de los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia
| |
Collapse
|
30
|
Alqarihi A, Kontoyiannis DP, Ibrahim AS. Mucormycosis in 2023: an update on pathogenesis and management. Front Cell Infect Microbiol 2023; 13:1254919. [PMID: 37808914 PMCID: PMC10552646 DOI: 10.3389/fcimb.2023.1254919] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those with hematologic malignancy or transplantation, Diabetes Mellitus (DM) and diabetic ketoacidosis (DKA) and immunocompetent patients with severe wounds. The recent SARS COV2 epidemy in India has resulted in a tremendous increase in MCR cases, typically seen in the setting of uncontrolled DM and corticosteroid use. In addition to the diversity of affected hosts, MCR has pleiotropic clinical presentations, with rhino-orbital/rhino-cerebral, sino-pulmonary and necrotizing cutaneous forms being the predominant manifestations. Major insights in MCR pathogenesis have brought into focus the host receptors (GRP78) and signaling pathways (EGFR activation cascade) as well as the adhesins used by Mucorales for invasion. Furthermore, studies have expanded on the importance of iron availability and the complex regulation of iron homeostasis, as well as the pivotal role of mycotoxins as key factors for tissue invasion. The molecular toolbox to study Mucorales pathogenesis remains underdeveloped, but promise is brought by RNAi and CRISPR/Cas9 approaches. Important recent advancements have been made in early, culture-independent molecular diagnosis of MCR. However, development of new potent antifungals against Mucorales remains an unmet need. Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals. Reversal of underlying immunosuppression, if feasible, rapid DKA correction and in selected patients, surgical debulking are crucial for improved outcomes.
Collapse
Affiliation(s)
- Abdullah Alqarihi
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
31
|
Salama EA, Eldesouky HE, Elgammal Y, Abutaleb NS, Seleem MN. Lopinavir and ritonavir act synergistically with azoles against Candida auris in vitro and in a mouse model of disseminated candidiasis. Int J Antimicrob Agents 2023; 62:106906. [PMID: 37392947 PMCID: PMC10528984 DOI: 10.1016/j.ijantimicag.2023.106906] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/14/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
INTRODUCTION AND OBJECTIVES The emergence of Candida auris has created a global health challenge. Azole antifungals are the most affected antifungal class because of the extraordinary capability of C. auris to develop resistance against these drugs. Here, we used a combinatorial therapeutic approach to sensitize C. auris to azole antifungals. METHODS AND RESULTS We have demonstrated the capability of the HIV protease inhibitors lopinavir and ritonavir, at clinically relevant concentrations, to be used with azole antifungals to treat C. auris infections both in vitro and in vivo. Both lopinavir and ritonavir exhibited potent synergistic interactions with the azole antifungals, particularly with itraconazole against 24/24 (100%) and 31/34 (91%) of tested C. auris isolates, respectively. Furthermore, ritonavir significantly interfered with the fungal efflux pump, resulting in a significant increase in Nile red fluorescence by 44%. In a mouse model of C. auris systemic infection, ritonavir boosted the activity of lopinavir to work synergistically with fluconazole and itraconazole and significantly reduced the kidney fungal burden by a 1.2 log (∼94%) and 1.6 log (∼97%) CFU, respectively. CONCLUSION Our results urge further comprehensive assessment of azoles and HIV protease inhibitors as a novel drug regimen for the treatment of serious invasive C. auris infections.
Collapse
Affiliation(s)
- Ehab A Salama
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Hassan E Eldesouky
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Yehia Elgammal
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Nader S Abutaleb
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Mohamed N Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia.
| |
Collapse
|
32
|
Roy M, Karhana S, Shamsuzzaman M, Khan MA. Recent drug development and treatments for fungal infections. Braz J Microbiol 2023; 54:1695-1716. [PMID: 37219748 PMCID: PMC10484882 DOI: 10.1007/s42770-023-00999-z] [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: 11/25/2022] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Fungal infections are now becoming a hazard to individuals which has paved the way for research to expand the therapeutic options available. Recent advances in drug design and compound screening have also increased the pace of the development of antifungal drugs. Although several novel potential molecules are reported, those discoveries have yet to be translated from bench to bedside. Polyenes, azoles, echinocandins, and flucytosine are among the few antifungal agents that are available for the treatment of fungal infections, but such conventional therapies show certain limitations like toxicity, drug interactions, and the development of resistance which limits the utility of existing antifungals, contributing to significant mortality and morbidity. This review article focuses on the existing therapies, the challenges associated with them, and the development of new therapies, including the ongoing and recent clinical trials, for the treatment of fungal infections. Advancements in antifungal treatment: a graphical overview of drug development, adverse effects, and future prospects.
Collapse
Affiliation(s)
- Madhura Roy
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Sonali Karhana
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Md Shamsuzzaman
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Sahqra, Kingdom of Saudi Arabia
| | - Mohd Ashif Khan
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
33
|
Jäger MC, Joos FL, Winter DV, Odermatt A. Characterization of the interferences of systemic azole antifungal drugs with adrenal steroid biosynthesis using H295R cells and enzyme activity assays. Curr Res Toxicol 2023; 5:100119. [PMID: 37637492 PMCID: PMC10458698 DOI: 10.1016/j.crtox.2023.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/29/2023] Open
Abstract
Azole antifungals, designed to inhibit fungal CYP51, have a liability to inhibit human CYP enzymes. Whilst drug-metabolizing CYPs are covered in preclinical safety assessment, those metabolizing endogenous bioactive molecules are usually not. Posaconazole and itraconazole were recently found to cause pseudohyperaldosteronism with hypokalemia and hypertension by inhibiting CYP11B1-dependent adrenal cortisol biosynthesis. Because this was overlooked in preclinical safety assessment, the present study tested whether applying adrenal carcinoma H295R cells could have predicted this liability and whether other systemic triazole antifungals interfere with adrenal steroidogenesis. Forskolin-stimulated H295R cells were exposed to systemic triazole antifungals that are currently used, and key adrenal steroids were quantified by UHPLC-MS/MS. To support the findings from the H295R model, activity assays for steroidogenic enzymes were performed. The analysis of the steroid profiles and product/substrate ratios predicted the CYP11B1 and CYP11B2 inhibition by posaconazole and itraconazole. Comparison of their steroid profiles allowed distinguishing their effects and suggested inhibition of adrenal androgen synthesis by posaconazole but not itraconazole, which was confirmed by CYP17A1 17,20-lyase activity measurements. In line with clinical observations, there was no evidence from these experiments for an inhibition of either CYP11B1/2 or CYP17A1 by voriconazole, fluconazole or isavuconazole. However, itraconazole and isavuconazole exerted an overall inhibition of steroidogenesis by a mechanism warranting further investigations. In conclusion, analyses of steroid profiles from the H295R assay and product/substrate ratios provide important information on the interference of a chemical with adrenal steroidogenesis and the underlying mechanism. This approach facilitates prioritization of further investigations, including enzyme expression and activity studies.
Collapse
Affiliation(s)
- Marie-Christin Jäger
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Friedrich L. Joos
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Denise V. Winter
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
34
|
Sushmitha TJ, Rajeev M, Kathirkaman V, Shivam S, Rao TS, Pandian SK. 3-Hydroxy coumarin demonstrates anti-biofilm and anti-hyphal efficacy against Candida albicans via inhibition of cell-adhesion, morphogenesis, and virulent genes regulation. Sci Rep 2023; 13:11687. [PMID: 37468600 DOI: 10.1038/s41598-023-37851-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023] Open
Abstract
Candida albicans, a common fungus of human flora, can become an opportunistic pathogen and causes invasive candidiasis in immunocompromised individuals. Biofilm formation is the prime cause of antibiotic resistance during C. albicans infections and treating biofilm-forming cells is challenging due to their intractable and persistent nature. The study intends to explore the therapeutic potential of naturally produced compounds by competitive marine bacteria residing in marine biofilms against C. albicans biofilm. To this end, 3-hydroxy coumarin (3HC), a compound identified from the cell-free culture supernatant of the marine bacterium Brevundimonas abyssalis, was found to exhibit anti-biofilm and anti-hyphal activity against both reference and clinical isolates of C. albicans. The compound demonstrated significant inhibitory effects on biofilms and impaired the yeast-to-hyphal transition, wrinkle, and filament morphology at the minimal biofilm inhibitory concentration (MBIC) of 250 µg mL-1. Intriguingly, quantitative PCR analysis of 3HC-treated C. albicans biofilm revealed significant downregulation of virulence genes (hst7, ume6, efg1, cph1, ras1, als1) associated with adhesion and morphogenesis. Moreover, 3HC displayed non-fungicidal and non-toxic characteristics against human erythrocytes and buccal cells. In conclusion, this study showed that marine biofilms are a hidden source of diverse therapeutic drugs, and 3HC could be a potent drug to treat C. albicans infections.
Collapse
Affiliation(s)
- T J Sushmitha
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Meora Rajeev
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
- Department of Biological Sciences and Bioengineering, Inha University, Inharo 100, Incheon, 22212, Republic of Korea
| | - Vellaisamy Kathirkaman
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Singh Shivam
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India
| | - Toleti Subba Rao
- School of Arts and Sciences, Sai University, OMR, Paiyanur, Tamil Nadu, 603105, India
| | - Shunmugiah Karutha Pandian
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
35
|
Javid S, Ather H, Hani U, Siddiqua A, Asif Ansari SM, Shanmugarajan D, Yogish Kumar H, Arivuselvam R, Purohit MN, Kumar BRP. Discovery of Novel Myristic Acid Derivatives as N-Myristoyltransferase Inhibitors: Design, Synthesis, Analysis, Computational Studies and Antifungal Activity. Antibiotics (Basel) 2023; 12:1167. [PMID: 37508263 PMCID: PMC10376843 DOI: 10.3390/antibiotics12071167] [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: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, N-Myristoyltransferase (NMT) has been identified as a new target for the treatment of fungal infections. It is observed that at present, there are increased rates of morbidity and mortality due to fungal infections. Hence, a series of novel myristic acid derivatives were designed via molecular docking studies and ADMET studies by targeting NMT (N-Myristoyltransferase). The designed myristic acid derivatives were synthesized by converting myristic acid into myristoyl chloride and coupling it with aryl amines to yield corresponding myristic acid derivatives. The compounds were purified and characterized via FTIR, NMR and HRMS spectral analyses. In this study, we carried out a target NMT inhibition assay. In the NMT screening assay results, the compounds 3u, 3m and 3t showed better inhibition compared to the other myristic acid derivatives. In an in vitro antifungal evaluation, the myristic acid derivatives were assessed against Candida albicans and Aspergillus niger strains by determining their minimal inhibitory concentrations (MIC50). The compounds 3u, 3k, 3r and 3t displayed superior antifungal capabilities against Candida albicans, and the compounds 3u, 3m and 3r displayed superior antifungal capabilities against Aspergillus niger compared to the standard drug FLZ (fluconazole). Altogether, we identified a new series of antifungal agents.
Collapse
Affiliation(s)
- Saleem Javid
- Department of Pharmaceutical Chemistry, Farooqia College of Pharmacy, Mysore 570 015, Karnataka, India
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| | - Hissana Ather
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Ayesha Siddiqua
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | | | - Dhivya Shanmugarajan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| | - Honnavalli Yogish Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| | - Rajaguru Arivuselvam
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| | - Madhusudan N Purohit
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysore, JSS Academy of Higher Education & Research, Mysore 570 015, Karnataka, India
| |
Collapse
|
36
|
Zhu P, Li Y, Guo T, Liu S, Tancer RJ, Hu C, Zhao C, Xue C, Liao G. New antifungal strategies: drug combination and co-delivery. Adv Drug Deliv Rev 2023; 198:114874. [PMID: 37211279 DOI: 10.1016/j.addr.2023.114874] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
The growing occurrence of invasive fungal infections and the mounting rates of drug resistance constitute a significant menace to human health. Antifungal drug combinations have garnered substantial interest for their potential to improve therapeutic efficacy, reduce drug doses, reverse, or ameliorate drug resistance. A thorough understanding of the molecular mechanisms underlying antifungal drug resistance and drug combination is key to developing new drug combinations. Here we discuss the mechanisms of antifungal drug resistance and elucidate how to discover potent drug combinations to surmount resistance. We also examine the challenges encountered in developing such combinations and discuss prospects, including advanced drug delivery strategies.
Collapse
Affiliation(s)
- Ping Zhu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Yan Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Ting Guo
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Simei Liu
- Department of Traditional Chinese Medicine, Chongqing College of Traditional Chinese Medicine, Chongqing 402760, China; Institute of Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Robert J Tancer
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Changhua Hu
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China
| | - Chengzhi Zhao
- Chongqing Health Center for Women and Children, Chongqing, 400700, PR China.
| | - Chaoyang Xue
- Public Health Research Institute and Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Guojian Liao
- State Key Laboratory of Silkworm Genome Biology, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400700, China.
| |
Collapse
|
37
|
Wu W, Jiang T, Lin H, Chen C, Wang L, Wen J, Wu J, Deng Y. The Specific Binding and Promotion Effect of Azoles on Human Aldo-Keto Reductase 7A2. Metabolites 2023; 13:metabo13050601. [PMID: 37233642 DOI: 10.3390/metabo13050601] [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: 03/30/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Human AKR 7A2 broadly participates in the metabolism of a number of exogenous and endogenous compounds. Azoles are a class of clinically widely used antifungal drugs, which are usually metabolized by CYP 3A4, CYP2C19, and CYP1A1, etc. in vivo. The azole-protein interactions that human AKR7A2 participates in remain unreported. In this study, we investigated the effect of the representative azoles (miconazole, econazole, ketoconazole, fluconazole, itraconazole, voriconazole, and posaconazole) on the catalysis of human AKR7A2. The steady-state kinetics study showed that the catalytic efficiency of AKR7A2 enhanced in a dose-dependent manner in the presence of posaconazole, miconazole, fluconazole, and itraconazole, while it had no change in the presence of econazole, ketoconazole, and voriconazole. Biacore assays demonstrated that all seven azoles were able to specifically bind to AKR7A2, among which itraconazole, posaconazole, and voriconazole showed the strongest binding. Blind docking predicted that all azoles were apt to preferentially bind at the entrance of the substrate cavity of AKR7A2. Flexible docking showed that posaconazole, located at the region, can efficiently lower the binding energy of the substrate 2-CBA in the cavity compared to the case of no posaconazole. This study demonstrates that human AKR7A2 can interact with some azole drugs, and it also reveals that the enzyme activity can be regulated by some small molecules. These findings will enable a better understanding of azole-protein interactions.
Collapse
Affiliation(s)
- Wanying Wu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Tianqing Jiang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Haihui Lin
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Chao Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Lingling Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
38
|
Younus Wani M, Saeed Saleh Alghamidi M, Srivastava V, Ahmad A, Aqlan FM, Saad Al-Bogami A. Click synthesis of pyrrolidine-based 1,2,3-triazole derivatives as antifungal agents causing cell cycle arrest and apoptosis in Candida auris. Bioorg Chem 2023; 136:106562. [PMID: 37119782 DOI: 10.1016/j.bioorg.2023.106562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
The emergence of multidrug-resistant fungal pathogens such as Candida auris is one of the major reasons WHO has declared fungal infections as a public health threat. Multidrug resistance, high mortality rates, frequent misidentification, and involvement in hospital outbreaks of this fungus demand the development of novel therapeutic drugs. In this direction, we report the synthesis of novel pyrrolidine-based 1,2,3-triazole derivatives using Click Chemistry (CC) and evaluation of their antifungal susceptibility against C. auris following Clinical and Laboratory Standards Institute (CLSI) guidelines. The fungicidal activity of the most potent derivative (P6) was further quantitatively confirmed by the MUSE cell viability assay. For insight mechanisms, the effect of the most active derivative on cell cycle arrest was studied using MuseTM Cell Analyzer and apoptotic mode of cell death was determined by studying phosphatidylserine externalization and mitochondrial depolarization. In vitro susceptibility testing and viability assays showed that all the newly synthesized compounds have antifungal activity with P6 being the most potent derivative. Cell cycle analysis revealed that P6 arrested the cells in S-phase in a concentration dependent manner and the apoptotic mode of cell death was confirmed by the movement of cytochrome c from mitochondria to cytosol with membrane depolarization. The hemolytic assay confirmed the safe use of P6 for further in vivo studies.
Collapse
Affiliation(s)
- Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia.
| | | | - Vartika Srivastava
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - Aijaz Ahmad
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Infection Control, Charlotte Maxeke Johannesburg Academic Hospital National Health Laboratory Service, South Africa
| | - Faisal M Aqlan
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia
| | - Abdullah Saad Al-Bogami
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia
| |
Collapse
|
39
|
Kavaliauskas P, Grybaitė B, Vaickelionienė R, Sapijanskaitė-Banevič B, Anusevičius K, Kriaučiūnaitė A, Smailienė G, Petraitis V, Petraitienė R, Naing E, Garcia A, Mickevičius V. Synthesis and Development of N-2,5-Dimethylphenylthioureido Acid Derivatives as Scaffolds for New Antimicrobial Candidates Targeting Multidrug-Resistant Gram-Positive Pathogens. Antibiotics (Basel) 2023; 12:antibiotics12020220. [PMID: 36830130 PMCID: PMC9952208 DOI: 10.3390/antibiotics12020220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
The growing antimicrobial resistance to last-line antimicrobials among Gram-positive pathogens remains a major healthcare emergency worldwide. Therefore, the search for new small molecules targeting multidrug-resistant pathogens remains of great importance. In this paper, we report the synthesis and in vitro antimicrobial activity characterisation of novel thiazole derivatives using representative Gram-negative and Gram-positive strains, including tedizolid/linezolid-resistant S. aureus, as well as emerging fungal pathogens. The 4-substituted thiazoles 3h, and 3j with naphthoquinone-fused thiazole derivative 7 with excellent activity against methicillin and tedizolid/linezolid-resistant S. aureus. Moreover, compounds 3h, 3j and 7 showed favourable activity against vancomycin-resistant E. faecium. Compounds 9f and 14f showed broad-spectrum antifungal activity against drug-resistant Candida strains, while ester 8f showed good activity against Candida auris which was greater than fluconazole. Collectively, these data demonstrate that N-2,5-dimethylphenylthioureido acid derivatives could be further explored as novel scaffolds for the development of antimicrobial candidates targeting Gram-positive bacteria and drug-resistant pathogenic fungi.
Collapse
Affiliation(s)
- Povilas Kavaliauskas
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute for Genome Sciences, School of Medicine, University of Maryland, 655 W. Baltimore Street, Baltimore, MD 21201, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
| | - Birutė Grybaitė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | - Rita Vaickelionienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | | | - Kazimieras Anusevičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
- Correspondence: ; Tel.: +370-646-21841
| | - Agnė Kriaučiūnaitė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | - Gabrielė Smailienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | - Vidmantas Petraitis
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
| | - Rūta Petraitienė
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
| | - Ethan Naing
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
| | - Andrew Garcia
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
| | - Vytautas Mickevičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| |
Collapse
|
40
|
Tezcan EF, Demirtas Y, Cakar ZP, Ulgen KO. Comprehensive genome-scale metabolic model of the human pathogen Cryptococcus neoformans: A platform for understanding pathogen metabolism and identifying new drug targets. FRONTIERS IN BIOINFORMATICS 2023; 3:1121409. [PMID: 36714093 PMCID: PMC9880062 DOI: 10.3389/fbinf.2023.1121409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Introduction: The fungal priority pathogen Cryptococcus neoformans causes cryptococcal meningoencephalitis in immunocompromised individuals and leads to hundreds of thousands of deaths per year. The undesirable side effects of existing treatments, the need for long application times to prevent the disease from recurring, the lack of resources for these treatment methods to spread over all continents necessitate the search for new treatment methods. Methods: Genome-scale models have been shown to be valuable in studying the metabolism of many organisms. Here we present the first genome-scale metabolic model for C. neoformans, iCryptococcus. This comprehensive model consists of 1,270 reactions, 1,143 metabolites, 649 genes, and eight compartments. The model was validated, proving accurate when predicting the capability of utilizing different carbon and nitrogen sources and growth rate in comparison to experimental data. Results and Discussion: The compatibility of the in silico Cryptococcus metabolism under infection conditions was assessed. The steroid and amino acid metabolisms found in the essentiality analyses have the potential to be drug targets for the therapeutic strategies to be developed against Cryptococcus species. iCryptococcus model can be applied to explore new targets for antifungal drugs along with essential gene, metabolite and reaction analyses and provides a promising platform for elucidation of pathogen metabolism.
Collapse
Affiliation(s)
- Enes Fahri Tezcan
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Yigit Demirtas
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| | - Zeynep Petek Cakar
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey,*Correspondence: Kutlu O. Ulgen,
| |
Collapse
|
41
|
Rakhshan A, Rahmati Kamel B, Saffaei A, Tavakoli-Ardakani M. Hepatotoxicity Induced by Azole Antifungal Agents: A Review Study. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2023; 22:e130336. [PMID: 38116543 PMCID: PMC10728840 DOI: 10.5812/ijpr-130336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/31/2023] [Accepted: 02/21/2023] [Indexed: 12/21/2023]
Abstract
Context Fungal infections are very common, and several medications are used to treat them. Azoles are prescribed widely to treat fungal infections. In addition to therapeutic effects, any drug can be accompanied by side effects in patients. One of the most important complications in this regard is liver injury. Therefore, hepatotoxicity induced by azole antifungal drugs were reviewed in this study. Evidence Acquisition English scientific papers were evaluated to review the effects of hepatotoxicity by azole antifungal agents, and the related studies' results were summarized using a table. The systematic search was implemented on electronic databases, including PubMed, Google Scholar, and Science Direct. Original articles and review articles that were published before April 1, 2022, were included in the study. Those articles without available full text or non-English articles were excluded. Also, articles that reported pediatric data were excluded. Results Most studies have reported the effects of hepatotoxicity by azole antifungal agents, and their mechanisms have been described. Conclusions Clinical evaluations regarding the hepatotoxicity of antifungal agents provided in the literature were reviewed. Therefore, it is recommended to prescribe these drugs with caution in high-risk patients suffering from liver diseases, and patients should be monitored for hepatotoxicity. However, more research is needed to evaluate the hepatotoxicity of azole antifungal agents and select appropriate drugs according to cost-effectiveness and the side effects' profiles, relying on lower incidence of this liver complication.
Collapse
Affiliation(s)
- Amin Rakhshan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bardia Rahmati Kamel
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Saffaei
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Xu H, Mou YH, Guo MB, Zhang R, Yan ZZ, An R, Wang X, Su X, Hou Z, Guo C. Discovery of novel selenium-containing azole derivatives as antifungal agents by exploiting the hydrophobic cleft of CYP51. Eur J Med Chem 2022; 243:114707. [DOI: 10.1016/j.ejmech.2022.114707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022]
|
43
|
Terconazole, an Azole Antifungal Drug, Increases Cytotoxicity in Antimitotic Drug-Treated Resistant Cancer Cells with Substrate-Specific P-gp Inhibitory Activity. Int J Mol Sci 2022; 23:ijms232213809. [PMID: 36430288 PMCID: PMC9696874 DOI: 10.3390/ijms232213809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
Abstract
Azole antifungal drugs have been shown to enhance the cytotoxicity of antimitotic drugs in P-glycoprotein (P-gp)-overexpressing-resistant cancer cells. Herein, we examined two azole antifungal drugs, terconazole (TCZ) and butoconazole (BTZ), previously unexplored in resistant cancers. We found that both TCZ and BTZ increased cytotoxicity in vincristine (VIC)-treated P-gp-overexpressing drug-resistant KBV20C cancer cells. Following detailed analysis, low-dose VIC + TCZ exerted higher cytotoxicity than co-treatment with VIC + BTZ. Furthermore, we found that VIC + TCZ could increase apoptosis and induce G2 arrest. Additionally, low-dose TCZ could be combined with various antimitotic drugs to increase their cytotoxicity in P-gp-overexpressing antimitotic drug-resistant cancer cells. Moreover, TCZ exhibited P-gp inhibitory activity, suggesting that the inhibitory activity of P-gp plays a role in sensitization afforded by VIC + TCZ co-treatment. We also evaluated the cytotoxicity of 12 azole antifungal drugs at low doses in drug-resistant cancer cells. VIC + TCZ, VIC + itraconazole, and VIC + posaconazole exhibited the strongest cytotoxicity in P-gp-overexpressing KBV20C and MCF-7/ADR-resistant cancer cells. These drugs exerted robust P-gp inhibitory activity, accompanied by calcein-AM substrate efflux. Given that azole antifungal drugs have long been used in clinics, our results, which reposition azole antifungal drugs for treating P-gp-overexpressing-resistant cancer, could be employed to treat patients with drug-resistant cancer rapidly.
Collapse
|
44
|
Taylor MB, Skophammer R, Warwick AR, Geck RC, Boyer JM, Walson M, Large CRL, Hickey ASM, Rowley PA, Dunham MJ. yEvo: experimental evolution in high school classrooms selects for novel mutations that impact clotrimazole resistance in Saccharomyces cerevisiae. G3 (BETHESDA, MD.) 2022; 12:jkac246. [PMID: 36173330 PMCID: PMC9635649 DOI: 10.1093/g3journal/jkac246] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/15/2022] [Indexed: 11/18/2022]
Abstract
Antifungal resistance in pathogenic fungi is a growing global health concern. Nonpathogenic laboratory strains of Saccharomyces cerevisiae are an important model for studying mechanisms of antifungal resistance that are relevant to understanding the same processes in pathogenic fungi. We have developed a series of laboratory modules in which high school students used experimental evolution to study antifungal resistance by isolating azole-resistant S. cerevisiae mutants and examining the genetic basis of resistance. We have sequenced 99 clones from these experiments and found that all possessed mutations previously shown to impact azole resistance, validating our approach. We additionally found recurrent mutations in an mRNA degradation pathway and an uncharacterized mitochondrial protein (Csf1) that have possible mechanistic connections to azole resistance. The scale of replication in this initiative allowed us to identify candidate epistatic interactions, as evidenced by pairs of mutations that occur in the same clone more frequently than expected by chance (positive epistasis) or less frequently (negative epistasis). We validated one of these pairs, a negative epistatic interaction between gain-of-function mutations in the multidrug resistance transcription factors Pdr1 and Pdr3. This high school-university collaboration can serve as a model for involving members of the broader public in the scientific process to make meaningful discoveries in biomedical research.
Collapse
Affiliation(s)
- Matthew Bryce Taylor
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Program in Biology, Loras College, Dubuque, IA 52001, USA
| | | | - Alexa R Warwick
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48824, USA
| | - Renee C Geck
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Josephine M Boyer
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - yEvo Students
- Westridge School, Pasadena, CA 91105, USA
- Moscow High School, Moscow, ID 83843, USA
| | - Margaux Walson
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Christopher R L Large
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- UW Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Angela Shang-Mei Hickey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Present address: Department of Genetics, Stanford University, Biomedical Innovations Building, Palo Alto, CA 94304, USA
| | - Paul A Rowley
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Maitreya J Dunham
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
45
|
Carradori S, Ammazzalorso A, De Filippis B, Şahin AF, Akdemir A, Orekhova A, Bonincontro G, Simonetti G. Azole-Based Compounds That Are Active against Candida Biofilm: In Vitro , In Vivo and In Silico Studies. Antibiotics (Basel) 2022; 11:antibiotics11101375. [PMID: 36290033 PMCID: PMC9598150 DOI: 10.3390/antibiotics11101375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/19/2022] Open
Abstract
Fungal pathogens, including Candida spp., Aspergillus spp. and dermatophytes, cause more than a billion human infections every year. A large library of imidazole- and triazole-based compounds were in vitro screened for their antifungal activity against C. albicans, C. glabrata, C. krusei, A. fumigatus and dermatophytes, such as Microsporum gypseum, Trichophyton rubrum and Trichophyton mentagrophytes. The imidazole carbamate 12 emerged as the most active compound, showing a valuable antifungal activity against C. glabrata (MIC 1−16 μg/mL) and C. krusei (MIC 4−24 μg/mL). No activity against A. fumigatus or the dermatophytes was observed among all the tested compounds. The compound 12 inhibited the formation of C. albicans, C. glabrata and C. krusei biofilms and reduced the mature Candida biofilm. In the Galleria mellonella larvae, 12 showed a significant reduction in the Candida infection, together with a lack of toxicity at the concentration used to activate its antifungal activity. Moreover, the in silico prediction of the putative targets revealed that the concurrent presence of the imidazole core, the carbamate and the p-chlorophenyl is important for providing a strong affinity for lanosterol 14α-demethylase (CgCYP51a1) and the fungal carbonic anhydrase (CgNce103), the S-enantiomer being more productive in these interactions.
Collapse
Affiliation(s)
- Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
- Computer-Aided Drug Discovery Laboratory, Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Alessandra Ammazzalorso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence:
| | - Barbara De Filippis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Ahmet Fatih Şahin
- Department of Drug Discovery and Development, Institute of Health Sciences, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Atilla Akdemir
- Computer-Aided Drug Discovery Laboratory, Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
- Department of Drug Discovery and Development, Institute of Health Sciences, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Anastasia Orekhova
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Graziana Bonincontro
- Department of Environmental Biology, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Giovanna Simonetti
- Department of Environmental Biology, “Sapienza” University of Rome, 00185 Rome, Italy
| |
Collapse
|
46
|
Devasia J, Nizam A, V. L. V. Azole-Based Antibacterial Agents: A Review on Multistep Synthesis Strategies and Biology. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.1938615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jyothis Devasia
- Department of Chemistry, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Aatika Nizam
- Department of Chemistry, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Vasantha V. L.
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| |
Collapse
|
47
|
Antifungal Encapsulated into Ligand-Functionalized Nanoparticles with High Specificity for Macrophages. Pharmaceutics 2022; 14:pharmaceutics14091932. [PMID: 36145686 PMCID: PMC9501281 DOI: 10.3390/pharmaceutics14091932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/28/2022] Open
Abstract
Infectious diseases caused by intracellular microorganisms such as Histoplasma capsulatum represent a significant challenge worldwide. Drug encapsulation into functionalized nanoparticles (NPs) is a valuable alternative to improving drug solubility and bioavailability, preventing undesirable interactions and drug degradation, and reaching the specific therapeutic target with lower doses. This work reports on Itraconazole (ITZ) encapsulated into core-shell-like polymeric NPs and functionalized with anti-F4/80 antibodies for their targeted and controlled release into macrophages. Uptake assay on co-culture showed significant differences between the uptake of functionalized and bare NPs, higher with functionalized NPs. In vitro assays showed that F4/80-NPs with 0.007 µg/mL of encapsulated ITZ eliminated the H. capsulatum fungus in co-culture with macrophages effectively compared to the bare NPs, without any cytotoxic effect on macrophages after 24 h interaction. Furthermore, encapsulated ITZ modulated the gene expression of anti and pro-inflammatory cytokines (IL-1, INF-Y, IL-6 and IL-10) on macrophages. Additionally, the anti-F4/80 antibody-coating enhanced natural and adequate antifungal response in the cells, exerting a synergistic effect that prevented the growth of the fungus at the intracellular level. Functionalized NPs can potentially improve macrophage-targeted therapy, increasing NPs endocytosis and intracellular drug concentration.
Collapse
|
48
|
de Jesus Antunes N, Coombes G, Francisco da Cunha K, de Lima Moreira F, Pilon AC, Lopes NP, Luiz da Costa J, Kipper K, Couchman L, Johnston A, De Nucci G. In vitro metabolism of the new antifungal dapaconazole using liver microsomes. Drug Metab Pharmacokinet 2022; 47:100475. [DOI: 10.1016/j.dmpk.2022.100475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/26/2022] [Accepted: 09/08/2022] [Indexed: 12/01/2022]
|
49
|
Design, synthesis, and biological evaluation of selenium-containing small molecule compounds based on the dual mechanism of fungal CYP51 inhibition and fungal ROS generation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Multicenter Registry of Patients Receiving Systemic Mold-Active Triazoles for the Management of Invasive Fungal Infections. Infect Dis Ther 2022; 11:1609-1629. [PMID: 35716251 PMCID: PMC9334502 DOI: 10.1007/s40121-022-00661-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/19/2022] [Indexed: 11/11/2022] Open
Abstract
Introduction ‘Real-world’ data for mold-active triazoles (MATs) in the treatment of invasive fungal infections (IFIs) are lacking. This study evaluated usage of MATs in a disease registry for the management of IFIs. Methods Data were collected for this multicenter, observational, prospective study from 55 US centers, between March 2017 and April 2020. Eligible patients received isavuconazole, posaconazole, or voriconazole as MAT monotherapy (one MAT) or multiple/sequenced MAT therapy (more than one MAT) for prophylaxis or treatment. Patients were enrolled within 60 days of MAT initiation. The primary objective was to characterize patients receiving a MAT and their patterns of therapy. The full analysis set (FAS) included eligible patients for the relevant enrollment protocol, and the safety analysis set (SAF) included patients who received ≥ 1 MAT dose. Results Overall, 2009 patients were enrolled in the SAF. The FAS comprised 1993 patients (510 isavuconazole; 540 posaconazole; 491 voriconazole; 452 multiple/sequenced MAT therapies); 816 and 1177 received treatment and prophylaxis at study index/enrollment, respectively. Around half (57.8%) of patients were male, and median age was 59 years. Among patients with IFIs during the study, the most common pathogens were Aspergillus fumigatus in the isavuconazole (18.2% [10/55]) and voriconazole (25.5% [12/47]) groups and Candida glabrata in the posaconazole group (20.9% [9/43]); the lungs were the most common infection site (58.2% [166/285]). Most patients were maintained on MAT monotherapy (77.3% [1541/1993]), and 79.4% (1520/1915) completed their MAT therapies. A complete/partial clinical response was reported in 59.1% (591/1001) of patients with a clinical response assessment. Breakthrough IFIs were reported in 7.1% (73/1030) of prophylaxis patients. Adverse drug reactions (ADRs) were reported in 14.7% (296/2009) of patients (3.9% [20/514] isavuconazole; 11.3% [62/547] posaconazole; 14.2% [70/494] voriconazole). Conclusions In this ‘real-world’ study, most patients remained on their initial therapy and completed their MAT therapy. Over half of patients receiving MATs for IFIs had a successful response, and most receiving prophylaxis did not develop breakthrough IFIs. ADRs were uncommon. Supplementary Information The online version contains supplementary material available at 10.1007/s40121-022-00661-5.
Collapse
|