Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3580 2023-12-18 11:07:08 |
2 format change + 1 word(s) 3581 2023-12-19 03:16:09 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhgun, A.A. Industrial Production of Antibiotics in Fungi. Encyclopedia. Available online: https://encyclopedia.pub/entry/52866 (accessed on 02 May 2024).
Zhgun AA. Industrial Production of Antibiotics in Fungi. Encyclopedia. Available at: https://encyclopedia.pub/entry/52866. Accessed May 02, 2024.
Zhgun, Alexander A.. "Industrial Production of Antibiotics in Fungi" Encyclopedia, https://encyclopedia.pub/entry/52866 (accessed May 02, 2024).
Zhgun, A.A. (2023, December 18). Industrial Production of Antibiotics in Fungi. In Encyclopedia. https://encyclopedia.pub/entry/52866
Zhgun, Alexander A.. "Industrial Production of Antibiotics in Fungi." Encyclopedia. Web. 18 December, 2023.
Industrial Production of Antibiotics in Fungi
Edit

The natural fermentation of antibiotics, along with semi-synthetic and synthetic approaches, is one of the most important methods for their production. The majority of the antibiotic market comes from the fermentation of high-yielding (HY) fungal strains. These strains have been obtained since the 1950s from wild-type (WT) isolates as a result of classical strain improvement (CSI) programs primarily involving multi-round random mutagenesis and screening.

antibiotics secondary metabolites fermentation fungi biosynthetic gene clusters (BGCs)

1. Introduction

The discovery of antibiotics and their widespread introduction into medical practice has radically changed the life of mankind, since they made it possible to save human lives in situations that were previously hopeless for patients [1]. Since the 1900s, numerous attempts have been made to create drugs that satisfy Paul Ehrlich’s “magic bullet” (Zauberkugel, ger.) concept, where a compound selectively kills disease-causing microorganisms without harming the human body [2][3]. Ehrlich carried over the term “magic bullet” from Carl Maria von Weber’s popular 1821 opera Der Freischütz (The Freeshooter), which is based on the German legend of magic bullets that fly not according to the laws of ballistics, but according to the will of the shooter himself. The shooter buys seven magic bullets from the devil in exchange for a soul. For the first six bullets, he chooses the target himself, and the last one is controlled by the devil. Ehrlich obtained the first magic bullets, the anti-syphilis arsenical-based drugs salvarsan (arsphenamine, compound 606) and its less toxic analogue neosalvarsan (compound 914), against the spirochete Treponema pallidum by chemical synthesis [4][5] (Figure 1). Although these first organic antimicrobials (the first antibiotics), salvarsan and neosalvarsan, were commercialized in the early 1910s (by Hoechst AG), their structural formulas were only established a hundred years later, and the mechanism of action is still not clear [1][6]. The concept of the “magic bullet” formulated by Ehrlich and the introduction of the first chemically synthesized antimicrobial drugs into medical practice not only ushered in the era of chemotherapy, but also became a key event that marked the beginning of the evolution of antibiotics [4]. For 30 years, many laboratories around the world actively searched for similar magic bullets, until in the 1940s arsenical-based drugs were replaced as treatments for syphilis by less toxic penicillins, which were obtained as a result of natural fermentation of the fungus Penicillium chrysogenum (later reclassified as Penicillium rubens) [7]. Since then, the fermentation of natural fungal and bacterial strains has become, along with chemical synthesis, the most important industrial method for obtaining antibiotics [8].
Figure 1. Antibiotic Era timeline focused on classical strain improvement (CSI) programs for high-yielding (HY) antibiotic producers.
The revolution in the field of antimicrobials occurred after the realization that, among the numerous synthesized low molecular weight metabolites, some molds (filamentous fungi) and bacteria can produce compounds that effectively destroy other microorganisms [9][10][11]. These so-called natural products, or secondary metabolites (SMs), play an important ecological role, enabling a particular microorganism to occupy a certain ecological niche by inhibiting the growth of other microorganisms [12][13][14]. Since such compounds are directed against microorganisms that differ significantly at the molecular level from humans, some of them are able to selectively kill pathogenic microbes [15][16]. In other words, nature itself creates magic bullets, casting them in the furnaces of the evolution of microbiological communities [17][18][19][20][21]. The main task in the so-called Golden Age of antibiotics (the period from the beginning of the 1940s to the end of the 1960s) was treasure hunting, the search for magic bullets created by nature: natural antibiotics [1][22] (Figure 1). The first and most important discovery of such a bullet was the discovery by Fleming in 1928 of penicillin, which began to be widely introduced into pharmaceutical practice from 1942 [7].
Improved fungal strains produce key beta-lactam antibiotics (penicillins and cephalosporins), which account for nearly half of the antibiotic market [23][24][25] (Figure 2).
Figure 2. Antibiotic market share by drug class, according to the open access data from [23].

2. Fermentation of Beta-Lactam Antibiotics in Fungi

The fermented penicillin G (PenG) and cephalosporin C (CepC) are modified in vitro to create drugs used in medicine (PenG is also used unmodified) [25]. Obtaining PenG and CepC in improved fungal strains is more economical than in vitro production [26]. In fungi, the biosynthesis of these beta-lactams is under the control of a complex hierarchical regulatory system, modified in the process of strain improvement, and is accompanied by a series of biosynthetic reactions, with a significantly increased intensity in HY producers [27][28]. In these reactions at the first stage, LLD-ACV tripeptide δ-(L-α-Aminoadipoyl)-L-cysteinyl-D-valine is obtained as a result of non-ribosomal peptide synthesis by the enzyme PcbAB (EC: 6.3.2.26). This tripeptide is then cyclized to isopenicillin N (IPN) through a dioxygenase reaction catalyzed by PcbC (isopenicillin N-synthase (EC: 1.21.3.1)). The IPN core structure is then modified by tailoring the enzyme PenDE (IAT, isopenicillin-N N-acyltransferase [EC:2.3.1.164]) to produce penicillin G, or by sequential reactions of the tailoring enzymes to produce cephalosporin C (CefD1, isopenicillin N-CoA synthetase (EC: 5.1.1.17), CefD2 (isopenicillin N-CoA epimerase (EC: 5.1.1.17), CefEF (penicillin N expandase, EC: 1.14.20.1)/deacetoxycephalosporin C hydroxylase, EC: 1.14.11.26), and CefG (deacetylcephalosporin-C acetyltransferase (EC: 2.3. 1.175)).

2.1. Fermentation of P. chrysogenum for Penicillin G (PenG) Production

The main industrial producer of penicillin G (PenG, or benzylpenicillin, and “Peanut Butter Shot” in military slang) is P. chrysogenum (formerly known as Penicillium notatum), which belongs to the class Eurotiomycetes and the division Ascomycota [29][30][31]. Currently, a number of molds are known that are capable of producing PenG, for example from the genera Arthroderma, Aspergillus, Penicillium, and Trichophyton; however, only P. chrysogenum strains are used for the industrial production of this important antibiotic [32][33][34][35]. PenG is the first industrially produced natural antibiotic, which was accompanied by an innovative breakthrough in the development of deep fermentation [36]. From the early 1940s to the present day, PenG has been produced worldwide from the fermentation of improved strains of the P. chrysogenum NRRL 1951 initial isolate [37][38][39]. This strain was discovered in 1943 growing on a cantaloupe at the local market in Peoria (IL, USA) [40]. It turned out that P. chrysogenum NRRL 1951 produces more penicillin than the original Fleming isolate (which produces mostly the unstable and difficult to isolate penicillin F, named after its discoverer) and other P. chrysogenum industrial strains used at that time for commercial production of PenG (such as P. chrysogenum NRRL 1249-B21) [7][41][42]. NRRL 1951 was adapted for commercial use; CSI programs were carried out on its basis, which made it possible to increase the PenG yield by more than 1000 times [43][44][45][46] (Table 1). Seventy-seven years after its discovery, P. chrysogenum NRRL 1951 received the status of “State Microbe of Illinois” [40].
Table 1. Production level of the most important antibiotics from fungi of wild type (WT) and high-yielding (HY) strains derived from them.
Currently, the P. chrysogenum strains used for PenG production (both the original Fleming strain and the strains used to obtain improved industrial producers) have again been reclassified as P. rubens [61]. At the same time, the name P. chrysogenum was retained for a number of strains that are not used for the industrial production of penicillins and differ from P. rubens in some characteristics, such as their production of secondary metabolites [61]. However, in the vast majority of recent publications, it is still common to use the name P. chrysogenum for PenG producers, in order to avoid confusion [7][29]. PenG can be used as a final drug [62] or can serve as a raw material for the production of semisynthetic penicillin antibiotics (Figure 3a). To achieve this, at the first stage, penicillin nucleus or 6-aminopenicillanic acid (6-APA) is obtained from PenG as a result of the deacylation reaction in vitro by amidase enzymes of bacterial origin (penicillin G amidase, EC 3.5.1.11) [63][64]. Then, various semisynthetic penicillins are obtained by grafting different side chains onto 6-APA [65][66][67] (Figure 3a).
Figure 3. Industrial production of beta-lactam antibiotics. (a) Obtaining of penicillin drugs; both penicillin G (a direct fermentation product of Penicillium chrysogenum) and its semisynthetic derivatives are used in pharmaceutical industry. (b) Obtaining of cephalosporin drugs; cephalosporin C (a fermentation product of Acremonium chrysogenum) has low activity and is not used in the pharmaceutical industry, unlike its semisynthetic derivatives. Curly brackets indicate the stages occurring in vivo (stage I—fermentation of fungal strains) and in vitro (stage II—production of core structures and stage III—production of semisynthetic antibiotics). PGA—penicillin G amidase (EC 3.5.1.11); DAO—D-amino acid oxidase (EC 1.4.3.3); GA—glutaryl-7-aminocephalosporanic acid acylase (EC 3.5.1.93); 6-APA—6-aminopenicillanic acid, 7-ACA—7-aminocephalosporanic acid.

2.2. Fermentation of Penicillins Other Than PenG

Since the 1950s, intensive research has been carried out to produce PenG analogues with altered activity profiles by fermenting P. chrysogenum with various mono-substituted acetic acids [68]. As a result, industrial strains of P. chrysogenum, in addition to benzylpenicillin (PenG), also produce another pharmacologically important compound, phenoxymethylpenicillin (penicillin V, PenV) [47][69]. PenG is produced if phenylacetic acid is added to the culture medium as a side chain precursor, and PenV is synthesized if phenoxyacetic acid is added [64]. To effectively produce PenV in industrial strains, multi-round mutagenesis and selection are also carried out to reduce the level of by-products, such as para-hydroxypenicillin V [70]. PenV can be used either as a final drug or as a raw material to obtain semisynthetic penicillins through 6-APA (Figure 3). Currently, only two penicillins produced during fermentation are of industrial importance, namely PenG and PenV [68]. Other natural penicillins, such as PenF (Fleming’s penicillin), PenK, PenN, PenO, PenU1, PenU6, or PenX, are not currently used in medical practice.

2.3. Fermentation of A. chrysogenum for Cephalosporin C (CefC) Production

The exclusive industrial producer of CefC is A. chrysogenum (formerly known as Cephalosporium acremonium), which belongs to the class Sordariomycetes, division Ascomycota (Table 1) [44]. In contrast to numerous PenG producers, CefC production is found only in a few fungal species. In addition to A. chrysogenum, the biosynthesis of CefC was also found in Pochonia chlamydosporia, Kallichroma tethys, and Paecilomyces persicinus, where it is not used for industrial production [32][35]. This relatively rare distribution of the biosynthetic pathway for this beta-lactam antibiotic in fungi is apparently associated with the appearance of additional genes, the origin of which is discussed in [7][71][72]. In particular, for the biosynthesis of CefC, part of the additional genes is assembled into the so-called “late” beta-lactam BGC, which is located on a different chromosome relative to the localization of the “early” beta-lactam BGC [72][73]. Currently, in 2023, the A. chrysogenum have again been reclassified as Hapsidospora chrysogena [74]. Since Acremonium chrysogenum has been used for a long time to refer the CPC producer, the researchers use this name in current research to avoid confusion. Current industrial producers of CefC are derived from the Brotzu initial isolate A. chrysogenum ATCC 11550 as a result of CSI programs [75]. Italian pharmacologist Giuseppe Brotzu isolated A. chrysogenum ATCC 11550 in 1948 from seawater near Sardinia, Italy [76]. Industrial descendants of this strain produce several hundred times more CefC [44][48][49]. CefC itself, unlike PenG, has weak antibacterial activity and is not used as a final drug in medical practice (Figure 3b) [77]. The key role of cephalosporin C is associated with its use as a starting substance for the production of all pharmaceutically important cephalosporin antibiotics [78]. To achieve this, at the first stage, cephalosporin nucleus or 7-aminocephalosporanic acid (7-ACA) is obtained in vitro from CefC, either chemically or enzymatically (using D-amino acid oxidase (EC 1.4.3.3) and glutaryl-7-aminocephalosporanic acid acylase (EC 3.5.1.93)) [63][79][80]. The side groups of 7-ACA are then modified by various chemical or enzymatic methods to produce cephalosporin drugs [45][63][81][82].

3. Fermentation of Non-Beta-Lactam Antibiotics in Fungi

The fermentation of fungal strains is used to produce a number of non-beta-lactam antibiotics for medical use. Some of them, such as fusidic acid and griseofulvin, were introduced to the market during the Golden Age of antibiotics and are still used today [83][84]. Other antibiotics, such as pleuromutilins, were discovered in the early 1950s, but only entered the market as semi-synthetic derivatives in 2007 [85][86][87]. The emergence of new classes of antibiotics from fungi, which entered the market in the last 20–25 years, was one of the most important factors that made it possible to fill the innovative gap in the introduction of antibiotics that arose after the end of the Golden Era of antibiotics [88]. For example, relatively recently discovered antibiotics, from the classes echinocandins and enfumafungins, were introduced into medical practice at the beginning of the 21st century [89][90]. There are also antibiotics that were previously obtained using fermentation in fungal strains, but that have now been discontinued due to discovered toxicity. For example, fusafungine, which belongs to the enniatin class of antibiotics, was withdrawn from the market in 2016 due to toxicity [91]. Although this research focuses only on antibiotics produced by fungal fermentation, it should be noted that improved bacterial strains are also widely used for the industrial production of non-beta-lactam antibiotics [92][93][94]. For example, some major market antibiotics, such as macrolides and aminoglycosides, and some minor market antibiotics, such as tetracyclines and glycopeptides, were originally isolated from actinomycetes, especially from the genus Streptomyces (Figure 2) [92]. Bacteria are also a promising source for obtaining new antibiotics, for example, antimicrobial peptides [95]. On the other hand, there are vast classes of non-beta-lactam antibiotics, such as fluoroquinolones and sulfonamides, which are obtained by chemical synthesis [96][97].

3.1. Fusidanes

Steroid antibiotics from the fusidane class, such as fusidic acid, helvolic acid, and cephalosporin P1, are produced by various filamentous fungi [98]. Although these antibiotics have been known since the 1960s, they have drawn renewed attention, due to the fact that they have no cross-resistance to generally used antibiotics [50][99]. Fusidic acid, the key fusidane antibiotic, is obtained from the fermentation of improved strains of Fusidium coccineum [52]. The production level in the improvement process was increased by a hundred times (Table 1). Other fusidanes, helvolic acid from Aspergillus fumigatus and cephalosporin P1 from A. chrysogenum, do not currently have such extensive use in medicine (Table 2) [100][101].
Table 2. Some examples of antibiotic production by fungal strains.

3.2. Griseofulvin

Griseofulvin, an ascomycete polyketide metabolite that interferes with the polymerization of fungal microtubules, was marketed in 1959 [113]. It was first isolated in 1939 and is still widely used in fields ranging from medicine to agriculture [113][114]. The industrial fermentation of griseofulvin is carried out on the basis of improved strains of Penicillium griseofulvum (reclassified from Penicillium patulum) [53][115]. The production level is several tens of grams per liter; however, specific numbers and methods of classical improvement are described in the relevant patents and are successfully masked [56][113].

3.3. Pleuromutilins

Pleuromutilins are SMs of the terpene type [116]. Despite the fact that pleuromutilin, the first representative of this class of antibiotics, was discovered back in 1951, pleuromutilins only recently entered the market as a medicine for humans [85]; retapamulin, the first antibiotic of this class, which is approved for humans, began to be used in 2007 [117][118]. To obtain the semisynthetic antibiotic retapamulin, pleuromutilin is used, which is produced as a result of fermentation of Clitophilus scyphoides and other basidiomycetes [86][119]. In 2019, lefamulin, another semisynthetic antibiotic from the pleuromutilin class, was approved for systemic use in humans [91][120][121][122]. An important characteristic of pleuromutilins is the lack of cross-resistance with other antibiotics, such as the macrolides, which act on a similar target, the 50S subunit of the prokaryotic ribosome [123].

3.4. Echinocandins

Currently, more than 20 echinocandins, lipopeptide antibiotics, have been isolated from fungi, the fermentation of three of which—echinocandin B, pneumocandin B0, and FR901379—has been commercialized [124]. They are used as raw materials in semisynthetic synthesis to obtain anidulafungin, caspofungin, and micafungin, respectively, which were introduced into medical practice in the 2000s [89][125][126][127]. These compounds are also called “antifungal penicillins” because they act on the biosynthesis of the cell wall of fungi, just as penicillins destroy the cell wall of bacteria [128]. Echinocandin B was first isolated in 1974 from A. nidulans, becoming the first antibiotic belonging to the echinocandin class [129]. Currently, it is produced by improved strains of A. nidulans and is used as a raw material for the semisynthetic antibiotic anidulafungin, used in medical practice [130][131]. Industrial strains of A. nidulans produce approximately a thousand times more echinocandin B than WT strains [59]. Caspofungin, another semisynthetic lipopeptide widely used in medicine, is derived from pneumocandin B0, which is produced by fermentation of the ascomycete Glarea lozoyensis [132]. Wild-type strains of G. lozoyensis predominantly synthesize pneumocandin A0, approximately 10 times more than pneumocandin B0 [124]. As a result of CSI programs, HY producers of pneumocandin B0 were obtained, which in its structure is less similar to echinocandin B than pneumocandin A0 [58][133][134][135]. Now, industrial strains produce hundreds of times more pneumocandin B0 compared to WT strains [58]. One of the most important antibiotics of this class, semisynthetic micafungin, is derived from the natural product FR901379 (via the enzymatic synthesis of compound FR179642) [136][137]. Lipopeptide FR901379 is obtained as a result of the natural fermentation of the improved strains of the ascomycete Coleophoma empetri [138][139]. Recently, the industrial producer C. empetri MEFC009, using a genetic engineering approach, was able to further increase the yield of FR901379 [60][140].

3.5. Enfumafungins

Enfumafungin is a glycosylated triterpenoid that acts on the fungal cell wall [141]. This SM, like the echinocandins, targets β-1,3-glucan synthase, but appears to bind this multiprotein complex at a different site [142]. Due to this, there is a lack of cross-resistance between these two classes of antibiotics, and pathogenic fungi that have acquired resistance to echinocandins show sensitivity to enfumafungins [143][144]. Enfumafungin is obtained from the ascomycete Hormonema carpetanum [103]. In 2021, ibrexafungerp, a semi-synthetic derivative of enfumafungin, was introduced into medical practice, representing the first non-azole oral antifungal drug to be approved in the United States for the treatment of vaginal yeast infections [90][145] (Table 2).

3.6. Enniatins

Enniatins are cyclohexadepsipeptides that are produced by Fusarium, Verticillium, Halosarpheia, and several other fungal genera [146]. Currently, about thirty natural compounds of this class have been characterized [102]. The first enniatins were discovered more than 70 years ago [147]. Since then, the antibiotic fusafungine has been introduced into clinical practice [148]. It is a cocktail of enniatins resulting from the fermentation of Fusarium lateritium strains, due to partially the non-specific work of non-ribosomal multifunctional enzyme enniatin synthetase (NRPS) [102][149][150][151]. The European Medicines Agency, in 2016, recommended the withdrawal of fusafungine from the market due to possible rare but severe allergic reactions [91][152].

4. Fungal Secondary Metabolites for the Development of Novel Drugs

Fungi produce significant amounts of SMs with antimicrobial properties [153][154]. Only a few of them have been introduced into medical practice (Table 1 and Table 2) [84][153]. However, there are a significant number of compounds that have demonstrated antibiotic or other medically important activity [155]. A case in point is a literature survey showing that, among 1500 compounds isolated from fungi in the 1990s, more than half exhibited antibacterial, antifungal, or antitumor activity [156]. Modern approaches that combine advances in the fields of genetic engineering and bioinformatics have made it possible to significantly increase the number of fungal SMs that are potentially significant for human use [157][158]. Some of these compounds and their semisynthetic derivatives are currently in clinical trials at phase I or greater [153][159]. Thus, the semisynthetic beta-lactam antibiotics benapenem, sulopenem, and its prodrug sulopenem etzadroxil, which act on the cell wall of bacteria, are in phase-III clinical trials [159][160][161][162]. Semisynthetic beta-lactams sanfetrinem and its prodrug sanfetrinem cilexetil are in phase-II clinical trials [159][163][164]. Several fungal antibiotics are currently being tested in clinical trials for the treatment of other diseases, such as cancer. For example, wortmannin, an antifungal furanosteroid antibiotic isolated in 1975 from Talaromyces wortmannii (formerly Penicillium wortmannii) [165], was then also shown to have inhibitory activity against phosphatidylinositol 3-kinase [166]. Currently, a semisynthetic compound PX-866 has been developed based on wortmannin, which has entered phase II clinical trials for recurrent glioblastoma [167]. Fungal SMs are being tested in clinical trials for the treatment of a wide range of diseases [153]. For example, muscimol, an isoxazole from Amanita pantherina, is an ionotropic GABAA-R receptor agonist and was developed for the treatment of drug-resistant epilepsy (phase I) [168]. Psilocybin from hallucinogenic fungus Psilocybe mexicana binds to the 5-HT2A serotonergic receptor and is developed for treatment-resistant depression [169][170]. Antroquinonol was isolated in 2007 from Taiwanofungus camphoratus (formerly Antrodia camphorata), a parasitic fungus indigenous to Taiwan [171]. It inhibits isoprenyl transferase, an enzyme that is involved in the post-translational processing of Ras and Rho proteins, which leads to the inhibition of Ras and Rho signaling, and that has antiviral, anti-inflammatory, anti-fibrotic, anti-cancer, and anti-hyperlipidemia properties [172]. This ubiquinone derivative was entered into phase II clinical trials for treating non-small-cell lung cancer [173]. The introduction, in recent years, of new classes of antibiotics from fungi (and their semi-synthetic derivatives), such as echinocandins, enfumafungins, and pleuromutilins, indicates significant potential for the development of novel drugs based on fungal SMs.

References

  1. Aminov, R.I. A Brief History of the Antibiotic Era: Lessons Learned and Challenges for the Future. Front. Microbiol. 2010, 1, 134.
  2. Ehrlich, P. Experimental Researches on Specific Therapy: On Immunity with special Reference to the Relationship between Distribution and Action of Antigens: FIRST HARBEN LECTURE. In The Collected Papers of Paul Ehrlich; Elsevier: Amsterdam, The Netherlands, 1960; pp. 106–117.
  3. Bosch, F.; Rosich, L. The Contributions of Paul Ehrlich to Pharmacology: A Tribute on the Occasion of the Centenary of His Nobel Prize. Pharmacology 2008, 82, 171.
  4. Williams, K.J. The introduction of “chemotherapy” using arsphenamine—the first magic bullet. J. R. Soc. Med. 2009, 102, 343–348.
  5. Strebhardt, K.; Ullrich, A. Paul Ehrlich’s magic bullet concept: 100 years of progress. Nat. Rev. Cancer 2008, 8, 473–480.
  6. Lloyd, N.C.; Morgan, H.W.; Nicholson, B.K.; Ronimus, R.S. The composition of Ehrlich’s salvarsan: Resolution of a century-old debate. Angew. Chem. Int. Ed. Engl. 2005, 44, 941–944.
  7. Bayer, E.A.; Fierro, F.; Vaca, I.; Castillo, N.I.; Ovidio García-Rico, R.; Chávez, R. Penicillium chrysogenum, a Vintage Model with a Cutting-Edge Profile in Biotechnology. Microorganisms 2022, 10, 573.
  8. Sanchez, S.; Demain, A.L. Bioactive Products from Fungi. In Food Bioactives; Springer: Cham, Switzerland, 2017; p. 59.
  9. Hadacek, F.; Bachmann, G. Low-molecular-weight metabolite systems chemistry. Front. Environ. Sci. 2015, 3, 128574.
  10. Fouillaud, M.; Dufossé, L. Microbial Secondary Metabolism and Biotechnology. Microorganisms 2022, 10, 123.
  11. Pham, J.V.; Yilma, M.A.; Feliz, A.; Majid, M.T.; Maffetone, N.; Walker, J.R.; Kim, E.; Cho, H.J.; Reynolds, J.M.; Song, M.C.; et al. A review of the microbial production of bioactive natural products and biologics. Front. Microbiol. 2019, 10, 449147.
  12. Demain, A.L.; Fang, A. The natural functions of secondary metabolites. Adv. Biochem. Eng. Biotechnol. 2000, 69, 1–39.
  13. Maresca, M.; Muchagato Mauricio, E.; Brito, L.; Conrado, R.; Colombo Gomes, T.; Sales Calaço Roque, G.; Olívia De Souza, A. Overview of Bioactive Fungal Secondary Metabolites: Cytotoxic and Antimicrobial Compounds. Antibiotics 2022, 11, 1604.
  14. Keller, N.P. Translating biosynthetic gene clusters into fungal armor and weaponry. Nat. Chem. Biol. 2015, 11, 671–677.
  15. Gualerzi, C.O.; Brandi, L.; Fabbretti, A.; Pon, C.L. (Eds.) Antibiotics: Targets, Mechanisms and Resistance; Wiley: Hoboken, NJ, USA, 2013; Volume 1, ISBN 978-3-527-33305-9.
  16. Upmanyu, N.; Malviya, V.N. Antibiotics: Mechanisms of action and modern challenges. Microorg. Sustain. Environ. Health 2020, 1, 367–382.
  17. Ueda, K.; Beppu, T. Antibiotics in microbial coculture. J. Antibiot. 2016, 70, 361–365.
  18. Clardy, J.; Fischbach, M.A.; Currie, C.R. The natural history of antibiotics. Curr. Biol. 2009, 19, R437.
  19. Hutchings, M.; Truman, A.; Wilkinson, B. Antibiotics: Past, present and future. Curr. Opin. Microbiol. 2019, 51, 72–80.
  20. Moody, S.C. Microbial co-culture: Harnessing intermicrobial signaling for the production of novel antimicrobials. Future Microbiol. 2014, 9, 575–578.
  21. Zhang, C.; Straight, P.D. Antibiotic discovery through microbial interactions. Curr. Opin. Microbiol. 2019, 51, 64–71.
  22. Lyddiard, D.; Jones, G.L.; Greatrex, B.W. Keeping it simple: Lessons from the golden era of antibiotic discovery. FEMS Microbiol. Lett. 2016, 363, 84.
  23. Kbvresearch.com. Available online: https://www.kbvresearch.com/antibiotics-market/ (accessed on 17 November 2023).
  24. Latinoamericana, R.; Campos Muñiz, C.; Cuadra Zelaya, T.E.; Esquivel, G.R.; Fernández, F.J. Penicillin and cephalosporin production: A historical perspective. Rev. Latinoam. Microbiol. 2007, 49, 88–98.
  25. Pandey, N.; Cascella, M. Beta-Lactam Antibiotics, 1st ed.; StatPearls Publishing: St. Petersburg, FL, USA, 2023; Volume 1.
  26. De Rosa, M.; Verdino, A.; Soriente, A.; Marabotti, A. The Odd Couple(s): An Overview of Beta-Lactam Antibiotics Bearing More Than One Pharmacophoric Group. Int. J. Mol. Sci. 2021, 22, 617.
  27. Wu, M.; Crismaru, C.G.; Salo, O.; Bovenberg, R.A.L.; Driessena, A.J.M. Impact of Classical Strain Improvement of Penicillium rubens on Amino Acid Metabolism during β-Lactam Production. Appl. Environ. Microbiol. 2020, 86, e01561-19.
  28. Zhgun, A.; Dumina, M.; Valiakhmetov, A.; Eldarov, M. The critical role of plasma membrane H+-ATPase activity in cephalosporin C biosynthesis of Acremonium chrysogenum. PLoS ONE 2020, 15, e0238452.
  29. Martín, J.F. Insight into the Genome of Diverse Penicillium chrysogenum Strains: Specific Genes, Cluster Duplications and DNA Fragment Translocations. Int. J. Mol. Sci. 2020, 21, 3936.
  30. Why the Most Dreaded Injection is Called the “Peanut Butter” Shot. Available online: https://www.military.com/off-duty/2020/02/10/why-most-dreaded-injection-called-peanut-butter-shot.html (accessed on 13 September 2023).
  31. BICILLIN L-A-Penicillin g Benzathine Injection, Suspension. Available online: https://labeling.pfizer.com/showlabeling.aspx?id=691 (accessed on 13 September 2023).
  32. Elander, R.P. Strain Improvement and Preservation of β-Lactam-Producing Microorganisms. In Antibiotics: Containing the Beta-Lactam Structure; Demain, A.L., Solomon, N.A., Eds.; Springer: Berlin, Heidelberg, 1983; Volume 1, pp. 97–146.
  33. Laich, F.; Fierro, F.; Martín, J.F. Production of Penicillin by Fungi Growing on Food Products: Identification of a Complete Penicillin Gene Cluster in Penicillium griseofulvum and a Truncated Cluster in Penicillium verrucosum. Appl. Environ. Microbiol. 2002, 68, 1211.
  34. Aharonowitz, Y.; Cohen, G.; Martin, J.F. Penicillin and cephalosporin biosynthetic genes: Structure, organization, regulation, and evolution. Annu. Rev. Microbiol. 1992, 46, 461–495.
  35. Martín, J.F.; Liras, P. Transfer of Secondary Metabolite Gene Clusters: Assembly and Reorganization of the Β-Lactam Gene Cluster from Bacteria to Fungi and Arthropods. In Horizontal Gene Transfer: Breaking Borders between Living Kingdoms; Villa, T.G., Viñas, M., Eds.; Springer International Publishing: Cham, Switzerland, 2019; Volume 1, pp. 337–359. ISBN 9783030218621.
  36. Bud, R. Innovators, deep fermentation and antibiotics: Promoting applied science before and after the Second World War. Dynamis 2011, 31, 323–341.
  37. Sawant, A.M.; Vamkudoth, K.R. Biosynthetic process and strain improvement approaches for industrial penicillin production. Biotechnol. Lett. 2022, 44, 179–192.
  38. Weber, S.S.; Bovenberg, R.A.L.; Driessen, A.J.M. Biosynthetic concepts for the production of β-lactam antibiotics in Penicillium chrysogenum. Biotechnol. J. 2012, 7, 225–236.
  39. Newbert, R.W.; Barton, B.; Greaves, P.; Harper, J.; Turner, G. Analysis of a commercially improved Penicillium chrysogenum strain series: Involvement of recombinogenic regions in amplification and deletion of the penicillin biosynthesis gene cluster. J. Ind. Microbiol. Biotechnol. 1997, 19, 18–27.
  40. Penicillium Strain Named State Microbe of Illinois. Available online: https://www.ars.usda.gov/news-events/news/research-news/2021/penicillium-strain-named-state-microbe-of-illinois/ (accessed on 13 September 2023).
  41. Backus, M.P.; Stauffer, J.F. The Production and Selection of a Family of Strains in Penicillium chrysogenum. Mycologia 1955, 47, 429–463.
  42. Perlman, D. Some mycological aspects of penicillin production. Bot. Rev. 1950, 16, 449–523.
  43. Salo, O.V.; Ries, M.; Medema, M.H.; Lankhorst, P.P.; Vreeken, R.J.; Bovenberg, R.A.L.; Driessen, A.J.M. Genomic mutational analysis of the impact of the classical strain improvement program on β-lactam producing Penicillium chrysogenum. BMC Genom. 2015, 16, 937.
  44. Liu, L.; Chen, Z.; Liu, W.; Ke, X.; Tian, X.; Chu, J. Cephalosporin C biosynthesis and fermentation in Acremonium chrysogenum. Appl. Microbiol. Biotechnol. 2022, 106, 6413–6426.
  45. Elander, R.P. Industrial production of β-lactam antibiotics. Appl. Microbiol. Biotechnol. 2003, 61, 385–392.
  46. Wang, Z.; Wang, X.; Liu, C.; Hang, H.; Chu, J.; Guo, M.; Zhuang, Y. Method for Increasing Penicillin Fermentation Production Unit. China Patent CN113388658A, 14 September 2021.
  47. Jørgensen, H.; Nielsen, J.; Villadsen, J.; Møllgaard, H. Analysis of penicillin V biosynthesis during fed-batch cultivations with a high-yielding strain of Penicillium chrysogenum. Appl. Microbiol. Biotechnol. 1995, 43, 123–130.
  48. Duan, S.; Yuan, G.; Zhao, Y.; Ni, W.; Luo, H.; Shi, Z.; Liu, F. Simulation of computational fluid dynamics and comparison of cephalosporin C fermentation performance with different impeller combinations. Korean J. Chem. Eng. 2013, 30, 1097–1104.
  49. Yang, Y.; Xia, J.; Li, J.; Chu, J.; Li, L.; Wang, Y.; Zhuang, Y.; Zhang, S. A novel impeller configuration to improve fungal physiology performance and energy conservation for cephalosporin C production. J. Biotechnol. 2012, 161, 250–256.
  50. Godtfredsen, W.O.; Jahnsen, S.; Lorck, H.; Roholt, K.; Tybring, L. Fusidic acid: A new antibiotic. Nature 1962, 193, 987.
  51. Verbist, L. The antimicrobial activity of fusidic acid. J. Antimicrob. Chemother. 1990, 25, 1–5.
  52. Huang, W.W.; Ge, X.Y.; Huang, Y.; Chai, X.T.; Zhang, L.; Zhang, Y.X.; Deng, L.N.; Liu, C.Q.; Xu, H.; Gao, J. High-yield strain of fusidic acid obtained by atmospheric and room temperature plasma mutagenesis and the transcriptional changes involved in improving its production in fungus Fusidium coccineum. J. Appl. Microbiol. 2021, 130, 405–415.
  53. Aris, P.; Yan, L.; Wei, Y.; Chang, Y.; Shi, B.; Xia, X. Conservation of griseofulvin genes in the gsf gene cluster among fungal genomes. G3 Genes|Genomes|Genet. 2022, 12, jkab39.
  54. Venkata Dasu, V.; Panda, T.; Chidambaram, M. Determination of significant parameters for improved griseofulvin production in a batch bioreactor by Taguchi’s method. Process Biochem. 2003, 38, 877–880.
  55. K1yoshi, U.; Abe, S. Production of Griseofulvin by Some Strains of the Genus Penicillum. J. Antibiot. Ser. A 1961, 14, 215–220.
  56. Nigam, P.; Singh, D. METABOLIC PATHWAYS|Production of Secondary Metabolites—Fungi. Encycl. Food Microbiol. 1999, 2, 1319–1328.
  57. Song, P.; Yuan, K.; Qin, T.; Zhang, K.; Ji, X.; Ren, L.; Guan, R.; Wen, J.; Huang, H. Metabolomics profiling reveals the mechanism of increased pneumocandin B0 production by comparing mutant and parent strains. J. Ind. Microbiol. Biotechnol. 2018, 45, 767–780.
  58. Song, P.; Zhang, K.; Zhang, S.; Huang, B.Q.; Ji, X.J.; Ren, L.J.; Gao, S.; Wen, J.P.; Huang, H. Enhancement of Pneumocandin B0 Production in Glarea lozoyensis by Low-Temperature Adaptive Laboratory Evolution. Front. Microbiol. 2018, 9, 2788.
  59. Hu, Z.C.; Li, W.J.; Zou, S.P.; Niu, K.; Zheng, Y.G. Mutagenesis of echinocandin B overproducing Aspergillus nidulans capable of using starch as main carbon source. Prep. Biochem. Biotechnol. 2020, 50, 745–752.
  60. Men, P.; Zhou, Y.; Xie, L.; Zhang, X.; Zhang, W.; Huang, X.; Lu, X. Improving the production of the micafungin precursor FR901379 in an industrial production strain. Microb. Cell Fact. 2023, 22, 44.
  61. Houbraken, J.; Frisvad, J.C.; Samson, R.A. Fleming’s penicillin producing strain is not Penicillium chrysogenum but P. rubens. IMA Fungus Glob. Mycol. J. 2011, 2, 87.
  62. Torres, M.J.; Doña, I.; Fernández, T.D.; Bogas, G. Penicillin. In Cutaneous Drug Hypersensitivity: Clinical Features, Mechanisms, Diagnosis, and Management; Bircher, A.J., Maibach, H.I., Brockow, K., Barbaud, A., Eds.; Springer Nature Switzerland AG: Cham, Switzerland, 2022; Volume 1, pp. 169–176. ISBN 9783030827434.
  63. Srirangan, K.; Orr, V.; Akawi, L.; Westbrook, A.; Moo-Young, M.; Chou, C.P. Biotechnological advances on penicillin G acylase: Pharmaceutical implications, unique expression mechanism and production strategies. Biotechnol. Adv. 2013, 31, 1319–1332.
  64. Ball, A.P.; Gray, J.A.; Murdoch, J.M. The Natural Penicillins—Benzylpenicillin (Penicillin G) and Phenoxymethylpenicillin (Penicillin V). In Antibacterial Drugs Today; Springer: Dordrecht, The Netherlands, 1978; Volume 1, pp. 6–18.
  65. Perron, Y.G.; Minor, W.F.; Holdrege, C.T.; Gottstein, W.J.; Godfrey, J.C.; Crast, L.B.; Babel, R.B.; Cheney, L.C. Derivatives of 6-Aminopenicillanic Acid. I. Partially Synthetic Penicillins Prepared from α-Aryloxyalkanoic Acids. J. Am. Chem. Soc. 1960, 82, 3934–3938.
  66. Güngör, Ö.Ö.N.; Gürkan, P.; Özçelik, B.; Oyardi, Ö. Synthesis and antimicrobial activities of new higher amino acid Schiff base derivatives of 6-aminopenicillanic acid and 7-aminocephalosporanic acid. J. Mol. Struct. 2016, 1106, 181–191.
  67. Cheptea, C.; Zara, A.; Dimitriu, D.G.; Sunel, V.; Dorohoi, D.O.; Cigu, T.A. New Semisynthetic Penicillins Obtained by Coupling of the 6-Aminopenicillanic Acid with 5-Mercapto-1,2,4-triazoles-3,4-disubstituted. Int. J. Mol. Sci. 2023, 24, 24.
  68. Bruggink, A.; Roy, P.D. Industrial Synthesis of Semisynthetic Antibiotics. In Synthesis of β-Lactam Antibiotics; Bruggink, A., Ed.; Springer: Dordrecht, The Netherlands, 2001; Volume 1, pp. 12–54.
  69. Möller, J.; Niehoff, J.; Dors, M.; Hiddessen, R.; Schügerl, K. Influence of medium composition on penicillin V production in a stirred tank reactor. J. Biotechnol. 1992, 25, 245–259.
  70. Chang, L.T.; McGrory, E.L.; Elander, R.P.; Hook, D.J. Decreased production of para-hydroxypenicillin V in penicillin V fermentations. J. Ind. Microbiol. 1991, 7, 175–179.
  71. Gutiérrez, S.; Velasco, J.; Fernandez, F.J.; Martín, J.F. The cefG gene of Cephalosporium acremonium is linked to the cefEF gene and encodes a deacetylcephalosporin C acetyltransferase closely related to homoserine O-acetyltransferase. J. Bacteriol. 1992, 174, 3056.
  72. Gutiérrez, S.; Fierro, F.; Casqueiro, J.; Martín, J.F. Gene organization and plasticity of the beta-lactam genes in different filamentous fungi. Antonie Leeuwenhoek 1999, 75, 81–94.
  73. Dumina, M.V.; Zhgun, A.A.; Domracheva, A.G.; Novak, M.I.; El’darov, M.A. Chromosomal polymorphism of Acremonium chrysogenum strains producing cephalosporin C. Russ. J. Genet. 2012, 48, 778–784.
  74. Hou, L.W.; Giraldo, A.; Groenewald, J.W.; Rämä, T.; Summerbell, R.C.; Huang, G.Z.; Cai, L.; Crous, P.W. Redisposition of acremonium-like fungi in Hypocreales. Stud. Mycol. 2023, 105, 23–203.
  75. Newton, G.G.; Abraham, E.P. Isolation of cephalosporin C, a penicillin-like antibiotic containing D-alpha-aminoadipic acid. Biochem. J. 1956, 62, 651–658.
  76. Acremonium chrysogenum (Thirumalachar et Sukapure) Gams—11550|ATCC. Available online: https://www.atcc.org/products/11550 (accessed on 22 September 2023).
  77. Brakhage, A.A. (Ed.) Molecular Biotechnolgy of Fungal Beta-Lactam Antibiotics and Related Peptide Synthetases. In Advances in Biochemical Engineering/Biotechnology, 1st ed.; Springer: Berlin/Heidelberg, Germany, 2004; Volume 88, ISBN 978-3-540-22032-9.
  78. García-Estrada, C.; Martín, J.-F. Penicillins and Cephalosporins. In Comprehensive Biotechnology; Moo-Young, M., Ed.; Academic Press: Burlington, VT, USA, 2011; Volume 1, pp. 255–268. ISBN 978-0-08-088504-9.
  79. Ehl’darov, M.A.; Redo, V.A.; Zhgun, A.A.; Zejnalov, O.A.; Skrjabin, K.G. RU 231 68 C1 pETTvDAO2 RECOMBINANT PLASMID PROVIDING SYNTHESIS OF YEAST Trigonopsis variabilis D-AMINO ACID OXYDASE (DAO) IN Escherichia coli CELLS AND RECOMBINANT Escherichia coli STRAIN C41(DE3)/pETTvDAO2 AS PRODUCER OF DAO. Patent RU23168C1, 17 March 2006.
  80. Lee, S.K.; Park, S.W.; Kim, Y.I.; Chung, K.H.; Hong, S.I.; Kim, S.W. Immobilization of GL-7-ACA Acylase for the Production of 7-ACA. Korean J. Chem. Eng. 2002, 19, 261–266.
  81. Deaguero, A.L.; Blum, J.K.; Bommarius, A.S. Biocatalytic synthesis of β-lactam antibiotics. In Encyclopedia of Industrial Biotechnology: Bioprocess, Bioseparation, and Cell Technology; John Wiley Sons, Inc.: Hoboken, NJ, USA, 2010; pp. 1–32.
  82. Eldarov, M.A.; Sklyarenko, A.V.; Dumina, M.; Medvedeva, N.V.; Jgoun, A.A.; Satarova, J.E.; Sidorenko, A.I.; Emperian, A.S.; Yarotsky, S.V. Recombinant cephalosporin-acid synthesase: Optimisation of expression in E. coli cells, immobilisation and application for biocatalytic cefazolin synthesis. Biomed. Khim. 2015, 61, 646–651.
  83. Falagas, M.E.; Grammatikos, A.P.; Michalopoulos, A. Potential of old-generation antibiotics to address current need for new antibiotics. Expert Rev. Anti. Infect. Ther. 2008, 6, 593–600.
  84. Beekman, A.M.; Barrow, R.A.; Beekman, A.M.; Barrow, R.A. Fungal Metabolites as Pharmaceuticals. Aust. J. Chem. 2014, 67, 827–843.
  85. Kavanagh, F.; And, A.H.; Robbins, W.J. Antibiotic Substances from Basidiomycetes: VIII. Pleurotus Multilus (Fr.) Sacc. and Pleurotus Passeckerianus Pilat*. Proc. Natl. Acad. Sci. USA 1951, 37, 570.
  86. Hartley, A.J.; De Mattos-Shipley, K.; Collins, C.M.; Kilaru, S.; Foster, G.D.; Bailey, A.M. Investigating pleuromutilin-producing Clitopilus species and related basidiomycetes. FEMS Microbiol. Lett. 2009, 297, 24–30.
  87. Watkins, R.R.; File, T.M. Lefamulin: A Novel Semisynthetic Pleuromutilin Antibiotic for Community-acquired Bacterial Pneumonia. Clin. Infect. Dis. 2020, 71, 2757–2762.
  88. Fischbach, M.A.; Walsh, C.T. Antibiotics for Emerging Pathogens. Science 2009, 325, 1089.
  89. Szymański, M.; Chmielewska, S.; Czyżewska, U.; Malinowska, M.; Tylicki, A. Echinocandins—Structure, mechanism of action and use in antifungal therapy. J. Enzyme Inhib. Med. Chem. 2022, 37, 876.
  90. Mapook, A.; Hyde, K.D.; Hassan, K.; Kemkuignou, B.M.; Čmoková, A.; Surup, F.; Kuhnert, E.; Paomephan, P.; Cheng, T.; de Hoog, S.; et al. Ten decadal advances in fungal biology leading towards human well-being. Fungal Divers. 2022, 116, 547–614.
  91. European Medicines Agency—Human Medicines—Fusafungine Containing Medicinal Products for Oromucosal and Nasal Use. Available online: https://web.archive.org/web/20180620220021/http://www.ema.europa.eu/ema//index.jsp?curl=pages%2Fmedicines%2Fhuman%2Freferrals%2FFusafungine_for_oromucosal_and_nasal_use%2Fhuman_referral_prac_000054.jsp&mid=WC0b01ac05805c516f (accessed on 26 September 2023).
  92. Mast, Y.; Stegmann, E. Actinomycetes: The Antibiotics Producers. Antibiotics 2019, 8, 105.
  93. Taurino, C.; Frattini, L.; Marcone, G.L.; Gastaldo, L.; Marinelli, F. Actinoplanes teichomyceticus ATCC 31121 as a cell factory for producing teicoplanin. Microb. Cell Fact. 2011, 10, 82.
  94. Stapley, E.O.; Jackson, M.; Hernandez, S.; Zimmerman, S.B.; Currie, S.A.; Mochales, S.; Mata, J.M.; Woodruff, H.B.; Hendlin, D. Cephamycins, a New Family of β-Lactam Antibiotics I. Production by Actinomycetes, Including Streptomyces lactamdurans sp. Antimicrob. Agents Chemother. 1972, 2, 122.
  95. Browne, K.; Chakraborty, S.; Chen, R.; Willcox, M.D.P.; Black, D.S.; Walsh, W.R.; Kumar, N. A New Era of Antibiotics: The Clinical Potential of Antimicrobial Peptides. Int. J. Mol. Sci. 2020, 21, 7047.
  96. Naeem, A.; Badshah, S.L.; Muska, M.; Ahmad, N.; Khan, K. The Current Case of Quinolones: Synthetic Approaches and Antibacterial Activity. Molecules 2016, 21, 268.
  97. Ovung, A.; Bhattacharyya, J. Sulfonamide drugs: Structure, antibacterial property, toxicity, and biophysical interactions. Biophys. Rev. 2021, 13, 259.
  98. Binod, P.; Sindhu, R.; Pandey, A. Production of antibiotics by filamentous fungi. In Current Developments in Biotechnology and Bioengineering: Filamentous Fungi Biorefinery; Taherzadeh, M.J., Ferreira, J.A., Pandey, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2023; Volume 1, pp. 477–496. ISBN 9780323918725.
  99. Lv, J.M.; Hu, D.; Gao, H.; Kushiro, T.; Awakawa, T.; Chen, G.D.; Wang, C.X.; Abe, I.; Yao, X.S. Biosynthesis of helvolic acid and identification of an unusual C-4-demethylation process distinct from sterol biosynthesis. Nat. Commun. 2017, 8, 1644.
  100. Bills, G.F.; Platas, G.; Gams, W. Conspecificity of the cerulenin and helvolic acid producing ‘Cephalosporium caerulens’, and the hypocrealean fungus Sarocladium oryzae. Mycol. Res. 2004, 108, 1291–1300.
  101. Chou, T.S.; Eisenbraun, E.J.; Rapala, R.T. The chemistry of cephalosporin P1. Tetrahedron Lett. 1967, 8, 409–414.
  102. Sy-Cordero, A.A.; Pearce, C.J.; Oberlies, N.H. Revisiting the enniatins: A review of their isolation, biosynthesis, structure determination and biological activities. J. Antibiot. 2012, 65, 541–549.
  103. Pelaez, F.; Cabello, A.; Platas, G.; Díez, M.T.; Del Val, A.G.; Basilio, A.; Martán, I.; Vicente, F.; Bills, G.F.; Giacobbe, R.A.; et al. The discovery of enfumafungin, a novel antifungal compound produced by an endophytic Hormonema species biological activity and taxonomy of the producing organisms. Syst. Appl. Microbiol. 2000, 23, 333–343.
  104. Ali, H.; Ries, M.I.; Lankhorst, P.P.; Van Der Hoeven, R.A.M.; Schouten, O.L.; Noga, M.; Hankemeier, T.; Van Peij, N.N.M.E.; Bovenberg, R.A.L.; Vreeken, R.J.; et al. A non-canonical NRPS is involved in the synthesis of fungisporin and related hydrophobic cyclic tetrapeptides in Penicillium chrysogenum. PLoS ONE 2014, 9, e98212.
  105. Saleh, I.; Goktepe, I. The characteristics, occurrence, and toxicological effects of patulin. Food Chem. Toxicol. 2019, 129, 301–311.
  106. Saniewski, M.; Horbowicz, M.; Kanlayanarat, S. The biological activities of troponoids and their use in agriculture a review. J. Hortic. Res. 2014, 22, 5–19.
  107. Vicente, M.F.; Cabello, A.; Platas, G.; Basilio, A.; Díez, M.T.; Dreikorn, S.; Giacobbe, R.A.; Onishi, J.C.; Meinz, M.; Kurtz, M.B.; et al. Antimicrobial activity of ergokonin A from Trichoderma longibrachiatum. J. Appl. Microbiol. 2001, 91, 806–813.
  108. Prompanya, C.; Dethoup, T.; Bessa, L.J.; Pinto, M.M.M.; Gales, L.; Costa, P.M.; Silva, A.M.S.; Kijjoa, A. New Isocoumarin Derivatives and Meroterpenoids from the Marine Sponge-Associated Fungus Aspergillus similanensis sp. nov. KUFA 0013. Mar. Drugs 2014, 12, 5160.
  109. Chooi, Y.H.; Cacho, R.; Tang, Y. Identification of the viridicatumtoxin and griseofulvin gene clusters from Penicillium aethiopicum. Chem. Biol. 2010, 17, 483–494.
  110. Han, J.; Zhang, J.; Song, Z.; Zhu, G.; Liu, M.; Dai, H.; Hsiang, T.; Liu, X.; Zhang, L.; Quinn, R.J.; et al. Genome-based mining of new antimicrobial meroterpenoids from the phytopathogenic fungus Bipolaris sorokiniana strain 11134. Appl. Microbiol. Biotechnol. 2020, 104, 3835–3846.
  111. Yang, W.; Kim, W.S.; Fang, A.; Demain, A.L. Carbon and nitrogen source nutrition of fumagillin biosynthesis by Aspergillus fumigatus. Curr. Microbiol. 2003, 46, 275–279.
  112. Wu, L.; Zhang, L.; Li, X.; Lv, R.; Cao, W.; Gao, W.; Liu, J.; Xie, Z.; Liu, H. Effective production of kojic acid in engineered Aspergillus niger. Microb. Cell Fact. 2023, 22, 40.
  113. Aris, P.; Wei, Y.; Mohamadzadeh, M.; Xia, X. Griseofulvin: An Updated Overview of Old and Current Knowledge. Molecular 2022, 27, 7034.
  114. Oxford, A.E.; Raistrick, H.; Simonart, P. Studies in the biochemistry of micro-organisms: Griseofulvin, C(17)H(17)O(6)Cl, a metabolic product of Penicillium griseo-fulvum Dierckx. Biochem. J. 1939, 33, 240–248.
  115. Venkata Dasu, V.; Panda, T. Studies on production of griseofulvin. Bioprocess Eng. 1999, 21, 489–495.
  116. Zhgun, A.A. Fungal BGCs for Production of Secondary Metabolites: Main Types, Central Roles in Strain Improvement, and Regulation According to the Piano Principle. Int. J. Mol. Sci. 2023, 24, 11184.
  117. Drug and Biologic Approval and IND Activity Reports > NME Drug and New Biologic Approvals in 2007. Available online: https://wayback.archive-it.org/7993/20170406061415/https://www.fda.gov/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologicApprovalReports/ucm081690.htm (accessed on 26 September 2023).
  118. Jones, R.N.; Fritsche, T.R.; Sader, H.S.; Ross, J.E. Activity of Retapamulin (SB-275833), a Novel Pleuromutilin, against Selected Resistant Gram-Positive Cocci. Antimicrob. Agents Chemother. 2006, 50, 2583.
  119. Robert Daum, R.S.; Kar, S.; Kirkpatrick Retapamulin, P. Retapamulin. Nat. Rev. Drug Discov. 2007, 6, 865–866.
  120. Dillon, C.; Guarascio, A.J.; Covvey, J.R. Lefamulin: A promising new pleuromutilin antibiotic in the pipeline. Expert Rev. Anti. Infect. Ther. 2019, 17, 5–15.
  121. Wang, H.; Charles, C.V. A Review of Newly Approved Antibiotic Treatment for Community-Acquired Bacterial Pneumonia: Lefamulin. Sr. Care Pharm. 2020, 35, 349–354.
  122. Yuan, S.; Yu, B.; Liu, H.M. New drug approvals for 2019: Synthesis and clinical applications. Eur. J. Med. Chem. 2020, 205, 112667.
  123. Waites, K.B.; Crabb, D.M.; Duffy, L.B.; Jensen, J.S.; Liu, Y.; Paukner, S. In Vitro Activities of Lefamulin and Other Antimicrobial Agents against Macrolide-Susceptible and Macrolide-Resistant Mycoplasma pneumoniae from the United States, Europe, and China. Antimicrob. Agents Chemother. 2017, 61, 10-1128.
  124. Emri, T.; Majoros, L.; Tóth, V.; Pócsi, I. Echinocandins: Production and applications. Appl. Microbiol. Biotechnol. 2013, 97, 3267–3284.
  125. Denning, D.W. Echinocandins: A new class of antifungal. J. Antimicrob. Chemother. 2002, 49, 889–891.
  126. Debono, M.; Gordee, R.S. Antibiotics that inhibit fungal cell wall development. Annu. Rev. Microbiol. 1994, 48, 471–497.
  127. Yue, Q.; Chen, L.; Zhang, X.; Li, K.; Sun, J.; Liu, X.; An, Z.; Bills, G.F. Evolution of Chemical Diversity in Echinocandin Lipopeptide Antifungal Metabolites. Eukaryot. Cell 2015, 14, 698–718.
  128. Pharmacotherapy Update|New Antifungal Agents Additions to the Existing Armamentarium (Part 1). Available online: https://www.clevelandclinicmeded.com/medicalpubs/pharmacy/mayjune2003/antifungal.htm (accessed on 25 September 2023).
  129. Benz, F.; Knüsel, F.; Nüesch, J.; Treichler, H.; Voser, W.; Nyfeler, R.; Keller-Schierlein, W. Metabolites of microorganisms. 143. Echinocandin B, a novel polypeptide-antibiotic from Aspergillus nidulans var. echinulatus: Isolation and structural components. Helv. Chim. Acta 1974, 57, 2459–2477.
  130. Pfaller, M.A.; Boyken, L.; Hollis, R.J.; Messer, S.A.; Tendolkar, S.; Diekema, D.J. In Vitro Activities of Anidulafungin against More than 2,500 Clinical Isolates of Candida spp., Including 315 Isolates Resistant to Fluconazole. J. Clin. Microbiol. 2005, 43, 5425.
  131. Zida, A.; Bamba, S.; Yacouba, A.; Ouedraogo-Traore, R.; Guiguemdé, R.T. Anti-Candida albicans natural products, sources of new antifungal drugs: A review. J. Mycol. Med. 2017, 27, 1–19.
  132. Bouffard, F.A.; Zambias, R.A.; Dropinski, J.F.; Balkovec, J.M.; Hammond, M.L.; Abruzzo, G.K.; Bartizal, K.F.; Marrinan, J.A.; Kurtz, M.B.; McFadden, D.C.; et al. Synthesis and Antifungal Activity of Novel Cationic Pneumocandin Bo Derivatives. J. Med. Chem. 1994, 37, 222–225.
  133. Masurekar, P.S.; Fountoulakis, J.M.; Hallada, T.C.; Sosa, M.S.; Kaplan, L. Pneumocandins from Zalerion arboricola. II. Modification of product spectrum by mutation and medium manipulation. J. Antibiot. 1992, 45, 1867–1874.
  134. Tkacz, J.S.; Giacobbe, R.A.; Monaghan, R.L. Improvement in the titer of echinocandin-type antibiotics: A magnesium-limited medium supporting the biphasic production of pneumocandins A0 and B0. J. Ind. Microbiol. 1993, 11, 95–103.
  135. Qin, T.; Song, P.; Wang, X.; Ji, X.; Ren, L.; Huang, H. Protoplast mutant selection of Glarea Lozoyensis and statistical optimization of medium for pneumocandin B0 yield-up. Biosci. Biotechnol. Biochem. 2016, 80, 2241–2246.
  136. Fujie, A. Discovery of micafungin (FK463): A novel antifungal drug derived from a natural product lead. Pure Appl. Chem. 2007, 79, 603–614.
  137. Ueda, S.; Sakamoto, K.; Oohata, N.; Tsuboi, M.; Yamashita, M.; Hino, M.; Yamada, M.; Hashimoto, S. Screening and characterization of microorganisms with FR901379 acylase activity. J. Antibiot. 2010, 63, 65–70.
  138. Kanda, M.; Tsuboi, M.; Sakamoto, K.; Shimizu, S.; Yamashita, M.; Honda, H. Improvement of FR901379 production by mutant selection and medium optimization. J. Biosci. Bioeng. 2009, 107, 530–534.
  139. Kanda, M.; Yamamoto, E.; Hayashi, A.; Yabutani, T.; Yamashita, M.; Honda, H. Scale-up fermentation of echinocandin type antibiotic FR901379. J. Biosci. Bioeng. 2010, 109, 138–144.
  140. Men, P.; Wang, M.; Li, J.; Geng, C.; Huang, X.; Lu, X. Establishing an Efficient Genetic Manipulation System for Sulfated Echinocandin Producing Fungus Coleophoma empetri. Front. Microbiol. 2021, 12, 734780.
  141. Schwartz, R.E.; Smith, S.K.; Onishi, J.C.; Meinz, M.; Kurtz, M.; Giacobbe, R.A.; Wilson, K.E.; Liesch, J.; Zink, D.; Horn, W.; et al. Isolation and structural determination of enfumafungin, a triterpene glycoside antifungal agent that is a specific inhibitor of glucan synthesis. J. Am. Chem. Soc. 2000, 122, 4882–4886.
  142. Kuhnert, E.; Li, Y.; Lan, N.; Yue, Q.; Chen, L.; Cox, R.J.; An, Z.; Yokoyama, K.; Bills, G.F. Enfumafungin synthase represents a novel lineage of fungal triterpene cyclases. Environ. Microbiol. 2018, 20, 3325.
  143. Jiménez-Ortigosa, C.; Perez, W.B.; Angulo, D.; Borroto-Esoda, K.; Perlin, D.S. De Novo Acquisition of Resistance to SCY-078 in Candida glabrata Involves FKS Mutations That both Overlap and Are Distinct from Those Conferring Echinocandin Resistance. Antimicrob. Agents Chemother. 2017, 61, 10-1128.
  144. Pfaller, M.A.; Messer, S.A.; Rhomberg, P.R.; Borroto-Esoda, K.; Castanheira, M. Differential Activity of the Oral Glucan Synthase Inhibitor SCY-078 against Wild-Type and Echinocandin-Resistant Strains of Candida Species. Antimicrob. Agents Chemother. 2017, 61, 10-1128.
  145. SCYNEXIS Announces FDA Approval of BREXAFEMME® (Ibrexafungerp Tablets) as the First and Only Oral Non-Azole Treatment for Vaginal Yeast Infections: SCYNEXIS, Inc. (SCYX). Available online: https://web.archive.org/web/20211231040808/https://www.scynexis.com/news-media/press-releases/detail/240/scynexis-announces-fda-approval-of-brexafemme (accessed on 9 October 2023).
  146. Olleik, H.; Nicoletti, C.; Lafond, M.; Courvoisier-Dezord, E.; Xue, P.; Hijazi, A.; Baydoun, E.; Perrier, J.; Maresca, M. Comparative Structure–Activity Analysis of the Antimicrobial Activity, Cytotoxicity, and Mechanism of Action of the Fungal Cyclohexadepsipeptides Enniatins and Beauvericin. Toxins 2019, 11, 514.
  147. Gäumann, E.; Roth, S.; Ettlinger, L.; Plattner, P.A.; Nager, U. Enniatin, a new antibiotic that works against mycobacteria. Experientia 1947, 3, 202–203.
  148. Osman, M.F. A clinical assessment of locabiotal in the treatment of infections of the nose and throat. J. Int. Med. Res. 1977, 5, 139–146.
  149. Hornbogen, T.; Glinski, M.; Zocher, R. Biosynthesis of Depsipeptide Mycotoxins in Fusarium. Eur. J. Plant Pathol. 2002, 108, 713–718.
  150. Zocher, R.; Keller, U.; Kleinkauf, H. Enniatin synthetase, a novel type of multifunctional enzyme catalyzing depsipeptide synthesis in Fusarium oxysporum. Biochemistry 1982, 21, 43–48.
  151. Herrmann, M.; Zocher, R.; Haese, A. Enniatin Production by Fusarium Strains and Its Effect on Potato Tuber Tissue. Appl. Environ. Microbiol. 1996, 62, 393.
  152. Hedenmalm, K.; Kurz, X.; Morales, D. Effect of withdrawal of fusafungine from the market on prescribing of antibiotics and other alternative treatments in Germany: A pharmacovigilance impact study. Eur. J. Clin. Pharmacol. 2019, 75, 979–984.
  153. Prescott, T.A.K.; Hill, R.; Mas-Claret, E.; Gaya, E.; Burns, E. Fungal Drug Discovery for Chronic Disease: History, New Discoveries and New Approaches. Biomolecules 2023, 13, 986.
  154. Aly, A.H.; Debbab, A.; Proksch, P. Fifty years of drug discovery from fungi. Fungal Divers. 2011, 50, 3–19.
  155. Keller, N.P. Fungal secondary metabolism: Regulation, function and drug discovery. Nat. Rev. Microbiol. 2019, 17, 167–180.
  156. Pela´ez, F. Biological Activities of Fungal Metabolites. In Handbook of Industrial Mycology; An, Z., Ed.; CRC Press: Boca Raton, FL, USA, 2004; Volume 1, pp. 68–111. ISBN 9780429224560.
  157. Schueffler, A.; Anke, T. Fungal natural products in research and development. Nat. Prod. Rep. 2014, 31, 1425–1448.
  158. Demain, A.L.; Martens, E. Production of valuable compounds by molds and yeasts. J. Antibiot. 2016, 70, 347–360.
  159. Butler, M.S.; Henderson, I.R.; Capon, R.J.; Blaskovich, M.A.T. Antibiotics in the clinical pipeline as of December 2022. J. Antibiot. 2023, 76, 431–473.
  160. Yang, H.; Zhang, M.; Chen, Y.; Ren, H.; Zhang, H.; Yu, C.; Lu, J.; You, L.; Yu, J.; Liang, H.; et al. Pharmacokinetics of benapenem for injection in subjects with mild to moderate renal impairment. Eur. J. Clin. Pharmacol. 2022, 78, 1079–1086.
  161. Iterum Therapeutics Receives Complete Response Letter from U.S. Food and Drug Administration for Oral Sulopenem: Iterum Therapeutics plc (ITRM). Available online: https://www.iterumtx.com/news/press-releases/detail/73/iterum-therapeutics-receives-complete-response-letter-from (accessed on 30 November 2023).
  162. Minamimura, M.; Taniyama, Y.; Inoue, E.; Mitsuhashi, S. In vitro antibacterial activity and beta-lactamase stability of CP-70,429 a new penem antibiotic. Antimicrob. Agents Chemother. 1993, 37, 1547.
  163. Doern, G.V.; Pierce, G.; Brueggemann, A.B. In vitro activity of sanfetrinem (GV104326), a new trinem antimicrobial agent, versus Streptococcus pneumonia, Haemophilus influenzae, and Moraxella catarrhalis. Diagn. Microbiol. Infect. Dis. 1996, 26, 39–42.
  164. Sanfetrinem|Working Group for New TB Drugs. Available online: https://www.newtbdrugs.org/pipeline/compound/sanfetrinem (accessed on 30 November 2023).
  165. Brian, P.W.; Curtis, P.J.; Hemming, H.G.; Norris, G.L.F. Wortmannin, an antibiotic produced by Penicillium wortmanni. Trans. Br. Mycol. Soc. 1957, 40, 365-IN3.
  166. Karve, S.; Werner, M.E.; Sukumar, R.; Cummings, N.D.; Copp, J.A.; Wang, E.C.; Li, C.; Sethi, M.; Chen, R.C.; Pacold, M.E.; et al. Revival of the abandoned therapeutic wortmannin by nanoparticle drug delivery. Proc. Natl. Acad. Sci. USA 2012, 109, 8230–8235.
  167. Pitz, M.W.; Eisenhauer, E.A.; MacNeil, M.V.; Thiessen, B.; Easaw, J.C.; Macdonald, D.R.; Eisenstat, D.D.; Kakumanu, A.S.; Salim, M.; Chalchal, H.; et al. Phase II study of PX-866 in recurrent glioblastoma. Neuro. Oncol. 2015, 17, 1270.
  168. Heiss, J.D.; Argersinger, D.P.; Theodore, W.H.; Butman, J.A.; Sato, S.; Khan, O.I. Convection-Enhanced Delivery of Muscimol in Patients with Drug-Resistant Epilepsy. Neurosurgery 2019, 85, E4.
  169. Davis, A.K.; Barrett, F.S.; May, D.G.; Cosimano, M.P.; Sepeda, N.D.; Johnson, M.W.; Finan, P.H.; Griffiths, R.R. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry 2021, 78, 1.
  170. Goodwin, G.M.; Aaronson, S.T.; Alvarez, O.; Arden, P.C.; Baker, A.; Bennett, J.C.; Bird, C.; Blom, R.E.; Brennan, C.; Brusch, D.; et al. Single-Dose Psilocybin for a Treatment-Resistant Episode of Major Depression. N. Engl. J. Med. 2022, 387, 1637–1648.
  171. Lee, T.H.; Lee, C.K.; Tsou, W.L.; Liu, S.Y.; Kuo, M.T.; Wen, W.C. A new cytotoxic agent from solid-state fermented mycelium of Antrodia camphorata. Planta Med. 2007, 73, 1412–1415.
  172. Ho, C.L.; Wang, J.L.; Lee, C.C.; Cheng, H.Y.; Wen, W.C.; Cheng, H.H.Y.; Chen, M.C.M. Antroquinonol blocks Ras and Rho signaling via the inhibition of protein isoprenyltransferase activity in cancer cells. Biomed. Pharmacother. 2014, 68, 1007–1014.
  173. Beheshtirouy, S.; Khani, E.; Khiali, S.; Entezari-Maleki, T. Investigational antiviral drugs for the treatment of COVID-19 patients. Arch. Virol. 2022, 167, 751–805.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 195
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 19 Dec 2023
1000/1000