What is the role of microbial biotechnology and genetic engineering in medicine?
Corresponding Author
Fernando Santos-Beneit
Institute of Sustainable Processes, Valladolid, Spain
Department of Chemical Engineering and Environmental Technology, School of Industrial Engineering, University of Valladolid, Valladolid, Spain
Correspondence Fernando Santos-Beneit, Institute of Sustainable Processes, Dr. Mergelina s/n, Valladolid 47011, Spain.
Email: [email protected]
Contribution: Conceptualization, Writing - original draft, Writing - review & editing
Search for more papers by this authorCorresponding Author
Fernando Santos-Beneit
Institute of Sustainable Processes, Valladolid, Spain
Department of Chemical Engineering and Environmental Technology, School of Industrial Engineering, University of Valladolid, Valladolid, Spain
Correspondence Fernando Santos-Beneit, Institute of Sustainable Processes, Dr. Mergelina s/n, Valladolid 47011, Spain.
Email: [email protected]
Contribution: Conceptualization, Writing - original draft, Writing - review & editing
Search for more papers by this authorAbstract
Microbial products are essential for developing various therapeutic agents, including antibiotics, anticancer drugs, vaccines, and therapeutic enzymes. Genetic engineering techniques, functional genomics, and synthetic biology unlock previously uncharacterized natural products. This review highlights major advances in microbial biotechnology, focusing on gene-based technologies for medical applications.
CONFLICT OF INTEREST STATEMENT
None declared.
REFERENCES
- Acunzo, M., Romano, G., Nigita, G., Veneziano, D., Fattore, L., Laganà, A., Zanesi, N., Fadda, P., Fassan, M., Rizzotto, L., Kladney, R., Coppola, V., & Croce, C. M. (2017). Selective targeting of point-mutated KRAS through artificial microRNAs. Proceedings of the National Academy of Sciences, 114(21), E4203–E4212. https://doi.org/10.1073/pnas.1620562114
- Afrin, H., Geetha Bai, R., Kumar, R., Ahmad, S. S., Agarwal, S. K., & Nurunnabi, M. (2023). Oral delivery of RNAi for cancer therapy. Cancer and Metastasis Reviews, 42(3), 699–724. https://doi.org/10.1007/s10555-023-10099-x
- Afzal, S., Yadav, A. K., Poonia, A. K., Choure, K., Yadav, A. N., & Pandey, A. (2023). Antimicrobial therapeutics isolated from algal source: Retrospect and prospect. Biologia, 78(2), 291–305. https://doi.org/10.1007/s11756-022-01207-3
- Agarwal, S., Mohamed, M. S., Mizuki, T., Maekawa, T., & Sakthi Kumar, D. (2019). Chlorotoxin modified morusin-PLGA nanoparticles for targeted glioblastoma therapy. Journal of Materials Chemistry B, 7(39), 5896–5919. https://doi.org/10.1039/c9tb01131e
- Aggarwal, V., Bala, E., Kumar, P., Raizada, P., Singh, P., & Verma, P. K. (2023). Natural products as potential therapeutic agents for SARS-CoV-2: A medicinal chemistry perspective. Current Topics in Medicinal Chemistry, 23(17), 1664–1698. https://doi.org/10.2174/1568026623666230327125918
- Ahmad, S. U., Li, B., Sun, J., Arbab, S., Dong, Z., Cheng, F., Zhou, X., Mahfuz, S., & Zhang, J. (2021). Recent advances in microencapsulation of drugs for veterinary applications. Journal of Veterinary Pharmacology and Therapeutics, 44(3), 298–312. https://doi.org/10.1111/jvp.12946
- Ali, S. O., Yu, X. Q., Robbie, G. J., Wu, Y., Shoemaker, K., Yu, L., DiGiandomenico, A., Keller, A. E., Anude, C., Hernandez-Illas, M., Bellamy, T., Falloon, J., Dubovsky, F., & Jafri, H. S. (2019). Phase 1 study of MEDI3902, an investigational anti-Pseudomonas aeruginosa PcrV and Psl bispecific human monoclonal antibody, in healthy adults. Clinical Microbiology and Infection, 25(5), 629.e1–629.e6. https://doi.org/10.1016/j.cmi.2018.08.004
- Allahyari, E., Velaei, K., Sanaat, Z., Jalilzadeh, N., Mehdizadeh, A., & Rahmati, M. (2023). RNA interference: Promising approach for breast cancer diagnosis and treatment. Cell Biology International, 47(5), 833–847. https://doi.org/10.1002/cbin.11979
- Aminov, R. (2017). History of antimicrobial drug discovery: Major classes and health impact. Biochemical Pharmacology, 133, 4–19. https://doi.org/10.1016/j.bcp.2016.10.001
- Andreu, J. M., Huecas, S., Araújo-Bazán, L., Vázquez-Villa, H., & Martín-Fontecha, M. (2022). The search for antibacterial inhibitors targeting cell division protein FtsZ at its nucleotide and allosteric binding sites. Biomedicines, 10(8), 1825. https://doi.org/10.3390/biomedicines10081825
- Andrews, N. P., Boeckman, J. X., Manning, C. F., Nguyen, J. T., Bechtold, H., Dumitras, C., Gong, B., Nguyen, K., van der List, D., Murray, K. D., Engebrecht, J., & Trimmer, J. S. (2019). A toolbox of IgG subclass-switched recombinant monoclonal antibodies for enhanced multiplex immunolabeling of brain. eLife, 8, e43322. https://doi.org/10.7554/eLife.43322
- Ashe, W. D. Jr., & Van Reken, D. E. (1977). 5-fluorocytosine: A brief review. Clinical Pediatrics, 16(4), 364–366. https://doi.org/10.1177/000992287701600412
- Asma, S. T., Acaroz, U., Imre, K., Morar, A., Shah, S. R. A., Hussain, S. Z., Arslan-Acaroz, D., Demirbas, H., Hajrulai-Musliu, Z., Istanbullugil, F. R., Soleimanzadeh, A., Morozov, D., Zhu, K., Herman, V., Ayad, A., Athanassiou, C., & Ince, S. (2022). Natural products/bioactive compounds as a source of anticancer drugs. Cancers, 14(24), 6203. https://doi.org/10.3390/cancers14246203
- Atanasov, A. G., Zotchev, S. B., Dirsch, V. M., & Supuran, C. T. (2021). Natural products in drug discovery: Advances and opportunities. Nature Reviews Drug Discovery, 20(3), 200–216. https://doi.org/10.1038/s41573-020-00114-z
- Baginski, M., & Czub, J. (2009). Amphotericin B and its new derivatives—Mode of action. Current Drug Metabolism, 10(5), 459–469. https://doi.org/10.2174/138920009788898019
- Bahrami, Y., Bouk, S., Kakaei, E., & Taheri, M. (2022). Natural products from actinobacteria as a potential source of new therapies against colorectal cancer: A review. Frontiers in Pharmacology, 13, 929161. https://doi.org/10.3389/fphar.2022.929161
- Barberán, J., Restrepo, R., & Cardinal-Fernández, P. (2021). Community-acquired pneumonia: Similarities and differences between European and American guidelines—A narrative review. Revista Española de Quimioterapia, 34(2), 72–80. https://doi.org/10.37201/req/114.2020
- Barrangou, R., Fremaux, C., Deveau, H., Richards, M., Boyaval, P., Moineau, S., Romero, D. A., & Horvath, P. (2007). CRISPR provides acquired resistance against viruses in prokaryotes. Science, 315(5819), 1709–1712. https://doi.org/10.1126/science.1138140
- Basak, P., Sachdeva, N., & Dayal, D. (2021). Can BCG vaccine protect against COVID-19 via trained immunity and tolerogenesis? BioEssays, 43(3), e2000200. https://doi.org/10.1002/bies.202000200
- Basu, K., Green, E. M., Cheng, Y., & Craik, C. S. (2019). Why recombinant antibodies—Benefits and applications. Current Opinion in Biotechnology, 60, 153–158. https://doi.org/10.1016/j.copbio.2019.01.012
- Batra, P., Deo, V., Mathur, P., & Gupta, A. K. (2017). Cotrimoxazole, a wonder drug in the era of multiresistance: Case report and review of literature. Journal of Laboratory Physicians, 9(3), 210–213. https://doi.org/10.4103/0974-2727.208261
- Bellini, A., Finocchietti, M., Rosa, A. C., Nordio, M., Ferroni, E., Massari, M., Spila Alegiani, S., Masiero, L., Bedeschi, G., Cardillo, M., Lucenteforte, E., Piccolo, G., Leoni, O., Pierobon, S., Ledda, S., Garau, D., Davoli, M., Addis, A., Belleudi, V., & CESIT study group. (2024). Effectiveness and safety of immunosuppressive regimens used as maintenance therapy in kidney transplantation: The CESIT study. PLoS One, 19(1), e0295205. https://doi.org/10.1371/journal.pone.0295205
- Bhatia, S. K., Otari, S. V., Jeon, J. M., Gurav, R., Choi, Y. K., Bhatia, R. K., Pugazhendhi, A., Kumar, V., Rajesh Banu, J., Yoon, J. J., Choi, K. Y., & Yang, Y. H. (2021). Biowaste-to-bioplastic (polyhydroxyalkanoates): Conversion technologies, strategies, challenges, and perspective. Bioresource Technology, 326, 124733. https://doi.org/10.1016/j.biortech.2021.124733
- Blasi, J., Chapman, E. R., Link, E., Binz, T., Yamasaki, S., Camilli, P. D., Südhof, T. C., Niemann, H., & Jahn, R. (1993). Botulinum neurotoxin A selectively cleaves the synaptic protein SNAP-25. Nature, 365(6442), 160–163. https://doi.org/10.1038/365160a0
- Blin, K., Shaw, S., Augustijn, H. E., Reitz, Z. L., Biermann, F., Alanjary, M., Fetter, A., Terlouw, B. R., Metcalf, W. W., Helfrich, E. J. N., van Wezel, G. P., Medema, M. H., & Weber, T. (2023). antiSMASH 7.0: New and improved predictions for detection, regulation, chemical structures and visualisation. Nucleic Acids Research, 51(W1), W46–W50. https://doi.org/10.1093/nar/gkad344
- Bobba, S., & Khader, S. A. (2023). Rifampicin drug resistance and host immunity in tuberculosis: More than meets the eye. Trends in Immunology, 44(9), 712–723. https://doi.org/10.1016/j.it.2023.07.003
- Borba-Santos, L. P., Rollin-Pinheiro, R., da Silva Fontes, Y., Dos Santos, G. M. P., de Sousa Araújo, G. R., Rodrigues, A. M., Guimarães, A. J., de Souza, W., Frases, S., Ferreira-Pereira, A., Barreto-Bergter, E., & Rozental, S. (2022). Screening of pandemic response box library reveals the high activity of olorofim against pathogenic sporothrix species. Journal of Fungi, 8(10), 1004. https://doi.org/10.3390/jof8101004
- Bordel, S., van Spanning, R. J. M., & Santos-Beneit, F. (2021). Imaging and modelling of poly(3-hydroxybutyrate) synthesis in Paracoccus denitrificans. AMB Express, 11(1), 113. https://doi.org/10.1186/s13568-021-01273-x
- Bormann, C., Huhn, W., & Zähner, H. (1985). Metabolic products of microorganisms. 228. New nikkomycins produced by mutants of Streptomyces tendae. The Journal of Antibiotics, 38(1), 9–16. https://doi.org/10.7164/antibiotics.38.9
- Boutin, C. A., & Luong, M. L. (2024). Update on therapeutic approaches for invasive fungal infections in adults. Therapeutic Advances in Infectious Disease, 11, 20499361231224980. https://doi.org/10.1177/20499361231224980
- Bron, P. A., & Kleerebezem, M. (2018). Lactic acid bacteria for delivery of endogenous or engineered therapeutic molecules. Frontiers in Microbiology, 9, 1821. https://doi.org/10.3389/fmicb.2018.01821
- Bruell, D., Bruns, C. J., Yezhelyev, M., Huhn, M., Müller, J., Ischenko, I., Fischer, R., Finnern, R., Jauch, K. W., & Barth, S. (2005). Recombinant anti-EGFR immunotoxin 425(scFv)-ETA' demonstrates anti-tumor activity against disseminated human pancreatic cancer in nude mice. International Journal of Molecular Medicine, 15(2), 305–313.
- Butler, M. S., Henderson, I. R., Capon, R. J., & Blaskovich, M. A. T. (2023). Antibiotics in the clinical pipeline as of December 2022. The Journal of Antibiotics, 76(8), 431–473. https://doi.org/10.1038/s41429-023-00629-8
- Caffrey, P., Lynch, S., Flood, E., Finnan, S., & Oliynyk, M. (2001). Amphotericin biosynthesis in Streptomyces nodosus: Deductions from analysis of polyketide synthase and late genes. Chemistry & Biology, 8(7), 713–723. https://doi.org/10.1016/s1074-5521(01)00046-1
- Carascal, M. B., Dela Cruz-Papa, D. M., Remenyi, R., Cruz, M. C. B., & Destura, R. V. (2022). Phage revolution against multidrug-resistant clinical pathogens in Southeast Asia. Frontiers in Microbiology, 13, 820572. https://doi.org/10.3389/fmicb.2022.820572
- Cen, Y. K., He, N. Y., Zhou, W. Y., Liu, Z. Q., & Zheng, Y. G. (2024). Development of a yeast cell based method for efficient screening of high yield tacrolimus production strain. 3 Biotech, 14(1), 26. https://doi.org/10.1007/s13205-023-03870-y
- Challis, G. L., & Hopwood, D. A. (2003). Synergy and contingency as driving forces for the evolution of multiple secondary metabolite production by Streptomyces species. Proceedings of the National Academy of Sciences, 100(Suppl 2), 14555–14561. https://doi.org/10.1073/pnas.1934677100
- Chiang, A. J., & Hasty, J. (2023). Design of synthetic bacterial biosensors. Current Opinion in Microbiology, 76, 102380. https://doi.org/10.1016/j.mib.2023.102380
- Chien, T., Doshi, A., & Danino, T. (2017). Advances in bacterial cancer therapies using synthetic biology. Current Opinion in Systems Biology, 5, 1–8. https://doi.org/10.1016/j.coisb.2017.05.009
- Cho, S. W., Kim, S., Kim, J. M., & Kim, J. S. (2013). Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nature Biotechnology, 31(3), 230–232. https://doi.org/10.1038/nbt.2507
- Chopra, B., Dhingra, A. K., Dhar, K. L., & Nepali, K. (2021). Emerging role of terpenoids for the treatment of cancer: A review. Mini-Reviews in Medicinal Chemistry, 21(16), 2300–2336. https://doi.org/10.2174/1389557521666210112143024
- Christmann, J., Cao, P., Becker, J., Desiderato, C. K., Goldbeck, O., Riedel, C. U., Kohlstedt, M., & Wittmann, C. (2023). High-efficiency production of the antimicrobial peptide pediocin PA-1 in metabolically engineered Corynebacterium glutamicum using a microaerobic process at acidic pH and elevated levels of bivalent calcium ions. Microbial Cell Factories, 22(1), 41. https://doi.org/10.1186/s12934-023-02044-y
- Claessen, D., & Errington, J. (2019). Cell wall deficiency as a coping strategy for stress. Trends in Microbiology, 27(12), 1025–1033. https://doi.org/10.1016/j.tim.2019.07.008
- Clavell, L. A., Gelber, R. D., Cohen, H. J., Hitchcock-Bryan, S., Cassady, J. R., Tarbell, N. J., Blattner, S. R., Tantravahi, R., Leavitt, P., & Sallan, S. E. (1986). Four-agent induction and intensive asparaginase therapy for treatment of childhood acute lymphoblastic leukemia. New England Journal of Medicine, 315(11), 657–663. https://doi.org/10.1056/NEJM198609113151101
- Costa, A. F., Silva, L. C., & Amaral, A. C. (2021). Farnesol: An approach on biofilms and nanotechnology. Medical Mycology, 59(10), 958–969. https://doi.org/10.1093/mmy/myab020
- Cotas, J., Lomartire, S., Gonçalves, A. M. M., & Pereira, L. (2024). From ocean to medicine: Harnessing seaweed's potential for drug development. International Journal of Molecular Sciences, 25(2), 797. https://doi.org/10.3390/ijms25020797
- Crofts, T. S., Gasparrini, A. J., & Dantas, G. (2017). Next-generation approaches to understand and combat the antibiotic resistome. Nature Reviews Microbiology, 15(7), 422–434. https://doi.org/10.1038/nrmicro.2017.28
- Curry, S., Kaufhold, R. M., Monslow, M. A., Zhang, Y., McGuinness, D., Kim, E., Nawrocki, D. K., McHugh, P. M., Briggs, M. L., Smith, W. J., He, J., Joyce, J. G., & Skinner, J. M. (2023). Preclinical evaluation of an investigational 21-valent pneumococcal conjugate vaccine, V116, in adult-rhesus monkey, rabbit, and mouse models. Vaccine, 41(4), 903–913. https://doi.org/10.1016/j.vaccine.2022.12.017
- Davies, J. (2011). How to discover new antibiotics: Harvesting the parvome. Current Opinion in Chemical Biology, 15(1), 5–10. https://doi.org/10.1016/j.cbpa.2010.11.001
- Demain, A. L., & Martens, E. (2017). Production of valuable compounds by molds and yeasts. The Journal of Antibiotics, 70(4), 347–360. https://doi.org/10.1038/ja.2016.121
- Dickstein, R., Wu, N., Cowan, B., Dunshee, C., Franks, M., Wolk, F., Belkoff, L., Castellucc, S., Holzbeierlein, J., Kulkarn, G., Weizer, A., Lamm, D., Ali, S., Epstein, J., Adams, G. P., Youssoufian, H., & Kassouf, W. (2018). VISTA, Phase 3 Trial of Vicineum, an EpCAM-Targeted Pseudomonas Exotoxin, in BCG-Unresponsive Non-Muscle Invasive Bladder Cancer. Global Congress on Bladder Cancer.
- Dijkmans, A. C., Zacarías, N. V. O., Burggraaf, J., Mouton, J. W., Wilms, E., van Nieuwkoop, C., Touw, D. J., Stevens, J., & Kamerling, I. M. C. (2017). Fosfomycin: Pharmacological, clinical and future perspectives. Antibiotics (USSR), 6(4), 24. https://doi.org/10.3390/antibiotics6040024
- Dyer, C., Hutt, L. P., Burky, R., & Joshi, L. T. (2019). Biocide resistance and transmission of clostridium difficile spores spiked onto clinical surfaces from an American Health Care Facility. Applied and Environmental Microbiology, 85(17), e01090. https://doi.org/10.1128/AEM.01090-19
- Efferth, T., & Oesch, F. (2021). Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology. Seminars in Cancer Biology, 68, 143–163. https://doi.org/10.1016/j.semcancer.2019.12.010
- Errington, J. (2013). L-form bacteria, cell walls and the origins of life. Open Biology, 3(1), 120143. https://doi.org/10.1098/rsob.120143
- Errington, J. (2017). Cell wall-deficient, L-form bacteria in the 21st century: A personal perspective. Biochemical Society Transactions, 45(2), 287–295. https://doi.org/10.1042/BST20160435
- Errington, J., Mickiewicz, K., Kawai, Y., & Wu, L. J. (2016). L-form bacteria, chronic diseases and the origins of life. Philosophical Transactions of the Royal Society, B: Biological Sciences, 371(1707), 20150494. https://doi.org/10.1098/rstb.2015.0494
- Espinosa-Viñals, C., García-Rivera, D., Rodríguez Noda, L., Amador Gómez, A., Nicot, M., Valle, O., Núñez, J. F., Martin, Y., Santana, D., Valdés, Y., & Vérez Bencomo, V. (2017). Influence of the co-administration of heptavalent conjugate vaccine PCV7-TT on the immunological response elicited by VA-MENGOC-BC® and Heberpenta®-L in rabbits. Immunological Investigations, 46(4), 395–408. https://doi.org/10.1080/08820139.2017.1288238
- Everard, A., Matamoros, S., Geurts, L., Delzenne, N. M., & Cani, P. D. (2014). Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. mBio, 5(3), e01011–e01014. https://doi.org/10.1128/mBio.01011-14
- Fernández-Martínez, L. T., Santos-Beneit, F., & Martín, J. F. (2012). Is PhoR-PhoP partner fidelity strict? PhoR is required for the activation of the pho regulon in Streptomyces coelicolor. Molecular Genetics and Genomics, 287(7), 565–573. https://doi.org/10.1007/s00438-012-0698-4
- Fogg, P. C. M., Haley, J. A., Stark, W. M., & Smith, M. C. M. (2017). Genome integration and excision by a new streptomyces bacteriophage, ϕJoe. Applied and Environmental Microbiology, 83(5), e02767. https://doi.org/10.1128/AEM.02767-16
- Gabarin, A., Yarmolinsky, L., Budovsky, A., Khalfin, B., & Ben-Shabat, S. (2023). Cannabis as a source of approved drugs: A new look at an old problem. Molecules, 28(23), 7686. https://doi.org/10.3390/molecules28237686
- Gabbianelli, R., Shahar, E., de Simone, G., Rucci, C., Bordoni, L., Feliziani, G., Zhao, F., Ferrati, M., Maggi, F., Spinozzi, E., & Mahajna, J. (2023). Plant-derived epi-nutraceuticals as potential broad-spectrum anti-viral agents. Nutrients, 15(22), 4719. https://doi.org/10.3390/nu15224719
- García-Depraect, O., Bordel, S., Lebrero, R., Santos-Beneit, F., Börner, R. A., Börner, T., & Muñoz, R. (2021). Inspired by nature: Microbial production, degradation and valorization of biodegradable bioplastics for life-cycle-engineered products. Biotechnology Advances, 53, 107772. https://doi.org/10.1016/j.biotechadv.2021.107772
- Gardner, N., & Champagne, C. P. (2005). Production of Propionibacterium shermanii biomass and vitamin B12 on spent media. Journal of Applied Microbiology, 99(5), 1236–1245. https://doi.org/10.1111/j.1365-2672.2005.02696.x
- Giraud, E. L., de Lijster, B., Krens, S. D., Desar, I. M. E., Boerrigter, E., & van Erp, N. P. (2023). Dose recommendations for anticancer drugs in patients with renal or hepatic impairment: An update. The Lancet Oncology, 24(6), e229. https://doi.org/10.1016/S1470-2045(23)00216-4
- Girigoswami, A., Adhikesavan, H., Mudenkattil, S., Devi, S., & Girigoswami, K. (2023). Role of cerium oxide nanoparticles and doxorubicin in improving cancer management: A mini review. Current Pharmaceutical Design, 29(33), 2640–2654. https://doi.org/10.2174/0113816128270290231029161741
- Goeddel, D. V., Kleid, D. G., Bolivar, F., Heyneker, H. L., Yansura, D. G., Crea, R., Hirose, T., Kraszewski, A., Itakura, K., & Riggs, A. D. (1979). Expression in Escherichia coli of chemically synthesized genes for human insulin. Proceedings of the National Academy of Sciences, 76(1), 106–110. https://doi.org/10.1073/pnas.76.1.106
- Gonzales-Luna, A. J., Carlson, T. J., & Garey, K. W. (2023a). Review article: Safety of live biotherapeutic products used for the prevention of clostridioides difficile infection recurrence. Clinical Infectious Diseases, 77(Suppl 6), S487–S496. https://doi.org/10.1093/cid/ciad642
- Gonzales-Luna, A. J., Carlson, T. J., & Garey, K. W. (2023b). Gut microbiota changes associated with Clostridioides difficile infection and its various treatment strategies. Gut Microbes, 15(1), 2223345. https://doi.org/10.1080/19490976.2023.2223345
- Haddad, E. M., McAlister, V. C., Renouf, E., Malthaner, R., Kjaer, M. S., & Gluud, L. L. (2006). Cyclosporin versus tacrolimus for liver transplanted patients. Cochrane Database of Systematic Reviews, 2006(4), CD005161. https://doi.org/10.1002/14651858.CD005161
- Hall-Stoodley, L., Costerton, J. W., & Stoodley, P. (2004). Bacterial biofilms: From the natural environment to infectious diseases. Nature Reviews Microbiology, 2(2), 95–108. https://doi.org/10.1038/nrmicro821
- Han, M., Lei, W., Liang, J., Li, H., Hou, M., & Gao, Z. (2024). The single-cell modification strategies for probiotics delivery in inflammatory bowel disease: A review. Carbohydrate Polymers, 324, 121472. https://doi.org/10.1016/j.carbpol.2023.121472
- Harimoto, T., Hahn, J., Chen, Y. Y., Im, J., Zhang, J., Hou, N., Li, F., Coker, C., Gray, K., Harr, N., Chowdhury, S., Pu, K., Nimura, C., Arpaia, N., Leong, K. W., & Danino, T. (2022). A programmable encapsulation system improves delivery of therapeutic bacteria in mice. Nature Biotechnology, 40(8), 1259–1269. https://doi.org/10.1038/s41587-022-01244-y
- Henrique, I. M., Sacerdoti, F., Ferreira, R. L., Henrique, C., Amaral, M. M., Piazza, R. M. F., & Luz, D. (2022). Therapeutic antibodies against shiga toxins: Trends and perspectives. Frontiers in Cellular and Infection Microbiology, 12, 825856. https://doi.org/10.3389/fcimb.2022.825856
- Herr, H. W. (2012). Intravesical Bacillus Calmette-Guérin outcomes in patients with bladder cancer and asymptomatic bacteriuria. Journal of Urology, 187(2), 435–437. https://doi.org/10.1016/j.juro.2011.10.032
- Hertweck, C. (2009). The biosynthetic logic of polyketide diversity. Angewandte Chemie International Edition, 48(26), 4688–4716. https://doi.org/10.1002/anie.200806121
- Hervin, V., Roy, V., & Agrofoglio, L. A. (2023). Antibiotics and antibiotic resistance-Mur ligases as an antibacterial target. Molecules, 28(24), 8076. https://doi.org/10.3390/molecules28248076
- Hoang Trung Chau, T., Hoang Anh Mai, D., Ngoc Pham, D., Thi Quynh Le, H., & Yeol Lee, E. (2020). Developments of riboswitches and toehold switches for molecular detection-biosensing and molecular diagnostics. International Journal of Molecular Sciences, 21(9), 3192. https://doi.org/10.3390/ijms21093192
- Holmes, K. V., Doller, E. W., & Sturman, L. S. (1981). Tunicamycin resistant glycosylation of a coronavirus glycoprotein: Demonstration of a novel type of viral glycoprotein. Virology, 115(2), 334–344. https://doi.org/10.1016/0042-6822(81)90115-x
- Houšť, J., Spížek, J., & Havlíček, V. (2020). Antifungal drugs. Metabolites, 10(3), 106. https://doi.org/10.3390/metabo10030106
- Howell, L. M., & Forbes, N. S. (2022). Bacteria-based immune therapies for cancer treatment. Seminars in Cancer Biology, 86(Pt 2), 1163–1178. https://doi.org/10.1016/j.semcancer.2021.09.006
- Huan, Y., Kong, Q., Mou, H., & Yi, H. (2020). Antimicrobial peptides: Cassification, design, application and research progress in multiple fields. Frontiers in Microbiology, 11, 582779. https://doi.org/10.3389/fmicb.2020.582779
- Huang, Y., Yang, Y., Liu, G., & Xu, M. (2023). New clinical application prospects of artemisinin and its derivatives: A scoping review. Infectious Diseases of Poverty, 12(1), 115. https://doi.org/10.1186/s40249-023-01152-6
- Hui Xian, T., Parasuraman, S., Ravichandran, M., & Prabhakaran, G. (2022). Dual-use vaccine for diarrhoeal diseases: Cross-protective immunogenicity of a cold-chain-free, live-attenuated, oral cholera vaccine against Enterotoxigenic Escherichia coli (ETEC) challenge in BALB/c mice. Vaccines, 10(12), 2161. https://doi.org/10.3390/vaccines10122161
- Hyoung, K. J., Hajam, I. A., & Lee, J. H. (2017). A consensus-hemagglutinin-based vaccine delivered by an attenuated Salmonella mutant protects chickens against heterologous H7N1 influenza virus. Oncotarget, 8(24), 38780–38792. https://doi.org/10.18632/oncotarget.16353
- Jawalagatti, V., Kirthika, P., & Lee, J. H. (2022). Oral mRNA vaccines against infectious diseases—A bacterial perspective [invited]. Frontiers in Immunology, 13, 884862. https://doi.org/10.3389/fimmu.2022.884862
- Jawale, C. V., Chaudhari, A. A., Jeon, B. W., Nandre, R. M., & Lee, J. H. (2012). Characterization of a novel inactivated Salmonella enterica serovar Enteritidis vaccine candidate generated using a modified cI857/λ PR/gene E expression system. Infection and Immunity, 80(4), 1502–1509. https://doi.org/10.1128/IAI.06264-11
- Ji, S., An, F., Zhang, T., Lou, M., Guo, J., Liu, K., Zhu, Y., Wu, J., & Wu, R. (2024). Antimicrobial peptides: An alternative to traditional antibiotics. European Journal of Medicinal Chemistry, 265, 116072. https://doi.org/10.1016/j.ejmech.2023.116072
- Jia, Q., & Horwitz, M. A. (2018). Live attenuated tularemia vaccines for protection against respiratory challenge with virulent F. tularensis subsp. tularensis. Frontiers in Cellular and Infection Microbiology, 8, 154. https://doi.org/10.3389/fcimb.2018.00154
- Jospe-Kaufman, M., Ben-Zeev, E., Mottola, A., Dukhovny, A., Berman, J., Carmeli, S., & Fridman, M. (2024). Reshaping echinocandin antifungal drugs to circumvent glucan synthase point-mutation-mediated resistance. Angewandte Chemie International Edition, 63, e202314728. https://doi.org/10.1002/anie.202314728
- Kaletta, C., & Entian, K. D. (1989). Nisin, a peptide antibiotic: cloning and sequencing of the nisA gene and posttranslational processing of its peptide product. Journal of Bacteriology, 171(3), 1597–1601. https://doi.org/10.1128/jb.171.3.1597-1601.1989
- Kaplon, H., Crescioli, S., Chenoweth, A., Visweswaraiah, J., & Reichert, J. M. (2023). Antibodies to watch in 2023. mAbs, 15(1), 2153410. https://doi.org/10.1080/19420862.2022.2153410
- Kaufmann, S. H. E. (2023). Vaccine development against tuberculosis before and after Covid-19. Frontiers in Immunology, 14, 1273938. https://doi.org/10.3389/fimmu.2023.1273938
- Kelley, E. J., Henson, S. N., Rahee, F., Boyle, A. S., Engelbrektson, A. L., Nelson, G. A., Mead, H. L., Anderson, N. L., Razavi, M., Yip, R., Ladner, J. T., Scriba, T. J., & Altin, J. A. (2023). Virome-wide detection of natural infection events and the associated antibody dynamics using longitudinal highly-multiplexed serology. Nature Communications, 14(1), 1783. https://doi.org/10.1038/s41467-023-37378-z
- Kifayat, S., Yele, V., Ashames, A., Sigalapalli, D. K., Bhandare, R. R., Shaik, A. B., Nasipireddy, V., & Sanapalli, B. K. R. (2023). Filamentous temperature sensitive mutant Z: A putative target to combat antibacterial resistance. RSC Advances, 13(17), 11368–11384. https://doi.org/10.1039/d3ra00013c
- Kim, J. H., Hajam, I. A., & Lee, J. H. (2018). Oral immunization with a novel attenuated Salmonella typhimurium encoding influenza HA, M2e and NA antigens protects chickens against H7N9 infection. Veterinary Research, 49(1), 12. https://doi.org/10.1186/s13567-018-0509-y
- Köhler, G., & Milstein, C. (1975). Continuous cultures of fused cells secreting antibody of predefined specificity. Nature, 256(5517), 495–497. https://doi.org/10.1038/256495a0
- Koon, H. W., Su, B., Xu, C., Mussatto, C. C., Tran, D. H. N., Lee, E. C., Ortiz, C., Wang, J., Lee, J. E., Ho, S., Chen, X., Kelly, C. P., & Pothoulakis, C. (2016). Probiotic Saccharomyces boulardii CNCM I-745 prevents outbreak-associated Clostridium difficile-associated cecal inflammation in hamsters. American Journal of Physiology-Gastrointestinal and Liver Physiology, 311(4), G610–G623. https://doi.org/10.1152/ajpgi.00150.2016
- Kosec, G., Goranovič, D., Mrak, P., Fujs, Š., Kuščer, E., Horvat, J., Kopitar, G., & Petković, H. (2012). Novel chemobiosynthetic approach for exclusive production of FK506. Metabolic Engineering, 14(1), 39–46. https://doi.org/10.1016/j.ymben.2011.11.003
- Kroemer, G., Chan, T. A., Eggermont, A. M. M., & Galluzzi, L. (2023). Immunosurveillance in clinical cancer management. CA: A Cancer Journal for Clinicians, 74, 187–202. https://doi.org/10.3322/caac.21818
- Kryl, M., Sedláček, V., & Kučera, I. (2023). Structural insight into catalysis by the flavin-dependent NADH oxidase (Pden_5119) of Paracoccus denitrificans. International Journal of Molecular Sciences, 24(4), 3732. https://doi.org/10.3390/ijms24043732
- Kumar, V., Yasmeen, N., Pandey, A., Ahmad Chaudhary, A., Alawam, A. S., Ahmad Rudayni, H., Islam, A., Lakhawat, S. S., Sharma, P. K., & Shahid, M. (2023). Antibiotic adjuvants: Synergistic tool to combat multi-drug resistant pathogens. Frontiers in Cellular and Infection Microbiology, 13, 1293633. https://doi.org/10.3389/fcimb.2023.1293633
- Lakowitz, A., Godard, T., Biedendieck, R., & Krull, R. (2018). Mini review: Recombinant production of tailored bio-pharmaceuticals in different Bacillus strains and future perspectives. European Journal of Pharmaceutics and Biopharmaceutics, 126, 27–39. https://doi.org/10.1016/j.ejpb.2017.06.008
- Lamparelli, E. P., Marino, M., Szychlinska, M. A., Della Rocca, N., Ciardulli, M. C., Scala, P., D'Auria, R., Testa, A., Viggiano, A., Cappello, F., Meccariello, R., Della Porta, G., & Santoro, A. (2023). The other side of plastics: Bioplastic-based nanoparticles for drug delivery systems in the brain. Pharmaceutics, 15(11), 2549. https://doi.org/10.3390/pharmaceutics15112549
- Law, J. W. F., Law, L. N. S., Letchumanan, V., Tan, L. T. H., Wong, S. H., Chan, K. G., Ab Mutalib, N. S., & Lee, L. H. (2020). Anticancer drug discovery from microbial sources: The unique mangrove streptomycetes. Molecules, 25(22), 5365. https://doi.org/10.3390/molecules25225365
- Le, P. H., Philippeaux, S., Mccollins, T., Besong, C., Kellar, A., Klapper, V. G., De Witt, A. S., Drinkard, J., Ahmadzadeh, S., Shekoohi, S., Varrassi, G., & Kaye, A. (2023). Pathogenesis, clinical considerations, and treatments: A narrative review on leprosy. Cureus, 15(12), e49954. https://doi.org/10.7759/cureus.49954
- Leader, B., Baca, Q. J., & Golan, D. E. (2008). Protein therapeutics: A summary and pharmacological classification. Nature Reviews Drug Discovery, 7(1), 21–39. https://doi.org/10.1038/nrd2399
- Lee, M. H., & Kim, B. J. (2022). COVID-19 vaccine development based on recombinant viral and bacterial vector systems: Combinatorial effect of adaptive and trained immunity. Journal of Microbiology, 60(3), 321–334. https://doi.org/10.1007/s12275-022-1621-2
- Lehar, S. M., Pillow, T., Xu, M., Staben, L., Kajihara, K. K., Vandlen, R., DePalatis, L., Raab, H., Hazenbos, W. L., Hiroshi Morisaki, J., Kim, J., Park, S., Darwish, M., Lee, B. C., Hernandez, H., Loyet, K. M., Lupardus, P., Fong, R., Yan, D., … Mariathasan, S. (2015). Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature, 527(7578), 323–328. https://doi.org/10.1038/nature16057
- Leschner, S., & Weiss, S. (2010). Salmonella-allies in the fight against cancer. Journal of Molecular Medicine, 88(8), 763–773. https://doi.org/10.1007/s00109-010-0636-z
- Li, L., Wang, T., Zhong, Y., Li, R., Deng, W., Xiao, X., Xu, Y., Zhang, J., Hu, X., & Wang, Y. (2024). A review of nanomaterials for biosensing applications. Journal of Materials Chemistry B, 12, 1168–1193. https://doi.org/10.1039/d3tb02648e
- Li, S., Jiang, W., Zheng, C., Shao, D., Liu, Y., Huang, S., Han, J., Ding, J., Tao, Y., & Li, M. (2020). Oral delivery of bacteria: Basic principles and biomedical applications. Journal of Controlled Release, 327, 801–833. https://doi.org/10.1016/j.jconrel.2020.09.011
- Lomovskaya, N., Otten, S. L., Doi-Katayama, Y., Fonstein, L., Liu, X. C., Takatsu, T., Inventi-Solari, A., Filippini, S., Torti, F., Colombo, A. L., & Hutchinson, C. R. (1999). Doxorubicin overproduction in Streptomyces peucetius: Cloning and characterization of the dnrU ketoreductase and dnrV genes and the doxA cytochrome P-450 hydroxylase gene. Journal of Bacteriology, 181(1), 305–318. https://doi.org/10.1128/JB.181.1.305-318.1999
- Lopes, R., & Prasad, M. K. (2024). Beyond the promise: Evaluating and mitigating off-target effects in CRISPR gene editing for safer therapeutics. Frontiers in Bioengineering and Biotechnology, 11, 1339189. https://doi.org/10.3389/fbioe.2023.1339189
- Lopez-Beltran, A., Cookson, M. S., Guercio, B. J., & Cheng, L. (2024). Advances in diagnosis and treatment of bladder cancer. BMJ, 384, e076743. https://doi.org/10.1136/bmj-2023-076743
- Lo Re, D., Zhou, Y., Mucha, J., Jones, L. F., Leahy, L., Santocanale, C., Krol, M., & Murphy, P. V. (2015). Synthesis of migrastatin analogues as inhibitors of tumour cell migration: Exploring structural change in and on the macrocyclic ring. Chemistry – A European Journal, 21(50), 18109–18121. https://doi.org/10.1002/chem.201502861
- Luo, P., Ming, Z., & Yang, Z. (2022). A critical role for CARD9 in intestinal microbiota modulation and colorectal malignancies. Frontiers in Bioscience-Landmark, 27(12), 320. https://doi.org/10.31083/j.fbl2712320
- Ma, W. W., & Adjei, A. A. (2009). Novel agents on the horizon for cancer therapy. CA: A Cancer Journal for Clinicians, 59(2), 111–137. https://doi.org/10.3322/caac.20003
- Mahata, B., Cabrera, A., Brenner, D. A., Guerra-Resendez, R. S., Li, J., Goell, J., Wang, K., Guo, Y., Escobar, M., Parthasarathy, A. K., Szadowski, H., Bedford, G., Reed, D. R., Kim, S., & Hilton, I. B. (2023). Compact engineered human mechanosensitive transactivation modules enable potent and versatile synthetic transcriptional control. Nature Methods, 20(11), 1716–1728. https://doi.org/10.1038/s41592-023-02036-1
- Mahato, R. K., Bhattacharya, S., Khullar, N., Sidhu, I. S., Reddy, P. H., Bhatti, G. K., & Bhatti, J. S. (2024). Targeting long non-coding RNAs in cancer therapy using CRISPR-Cas9 technology: A novel paradigm for precision oncology. Journal of Biotechnology, 379, 98–119. https://doi.org/10.1016/j.jbiotec.2023.12.003
- Maillard, J. Y., & Pascoe, M. (2024). Disinfectants and antiseptics: Mechanisms of action and resistance. Nature Reviews Microbiology, 22(1), 4–17. https://doi.org/10.1038/s41579-023-00958-3
- Makary, M. A., Kaczmarski, K., & Nachman, K. (2018). A call for doctors to recommend antibiotic-free foods: Agricultural antibiotics and the public health crisis of antimicrobial resistance. The Journal of Antibiotics, 71(8), 685–687. https://doi.org/10.1038/s41429-018-0062-y
- Maksimenko, O., Malinovskaya, J., Shipulo, E., Osipova, N., Razzhivina, V., Arantseva, D., Yarovaya, O., Mostovaya, U., Khalansky, A., Fedoseeva, V., Alekseeva, A., Vanchugova, L., Gorshkova, M., Kovalenko, E., Balabanyan, V., Melnikov, P., Baklaushev, V., Chekhonin, V., Kreuter, J., … Gelperina, S. (2019). Doxorubicin-loaded PLGA nanoparticles for the chemotherapy of glioblastoma: Towards the pharmaceutical development. International Journal of Pharmaceutics, 572, 118733. https://doi.org/10.1016/j.ijpharm.2019.118733
- Malico, A. A., Nichols, L., & Williams, G. J. (2020). Synthetic biology enabling access to designer polyketides. Current Opinion in Chemical Biology, 58, 45–53. https://doi.org/10.1016/j.cbpa.2020.06.003
- Malla, R., Padmaraju, V., & Kundrapu, D. B. (2022). Tumor-associated macrophages: Potential target of natural compounds for management of breast cancer. Life Sciences, 301, 120572. https://doi.org/10.1016/j.lfs.2022.120572
- Martín, J. F., Sola-Landa, A., Santos-Beneit, F., Fernández-Martínez, L. T., Prieto, C., & Rodríguez-García, A. (2011). Cross-talk of global nutritional regulators in the control of primary and secondary metabolism in Streptomyces. Microbial Biotechnology, 4(2), 165–174. https://doi.org/10.1111/j.1751-7915.2010.00235.x
- Martin-Killias, P., Stefan, N., Rothschild, S., Plückthun, A., & Zangemeister-Wittke, U. (2011). A novel fusion toxin derived from an EpCAM-specific designed ankyrin repeat protein has potent antitumor activity. Clinical Cancer Research, 17(1), 100–110. https://doi.org/10.1158/1078-0432.CCR-10-1303
- Martín-Martín, S., Rodríguez-García, A., Santos-Beneit, F., Franco-Domínguez, E., Sola-Landa, A., & Martín, J. F. (2017). Self-control of the PHO regulon: The PhoP-dependent protein PhoU controls negatively expression of genes of PHO regulon in Streptomyces coelicolor. The Journal of Antibiotics, 71, 113–122. https://doi.org/10.1038/ja.2017.130
- Mathieu, C., Martens, P. J., & Vangoitsenhoven, R. (2021). One hundred years of insulin therapy. Nature Reviews Endocrinology, 17(12), 715–725. https://doi.org/10.1038/s41574-021-00542-w
- May, R. C., & Plasterk, R. H. (2005). RNA interference spreading in C. elegans. Methods in Enzymology, 392, 308–315. https://doi.org/10.1016/S0076-6879(04)92018-6
- Mba, I. E., & Nweze, E. I. (2022). Antimicrobial peptides therapy: An emerging alternative for treating drug-resistant bacteria. The Yale Journal of Biology and Medicine, 95(4), 445–463.
- McCallum, G., & Tropini, C. (2023). The gut microbiota and its biogeography. Nature Reviews Microbiology, 22, 105–118. https://doi.org/10.1038/s41579-023-00969-0
- McCarty, J., Bedell, L., De Lame, P. A., Cassie, D., Lock, M., Bennett, S., & Haney, D. (2022). Update on CVD 103-HgR single-dose, live oral cholera vaccine. Expert Review of Vaccines, 21(1), 9–23. https://doi.org/10.1080/14760584.2022.2003709
- Meier-Kriesche, H. U., Li, S., Gruessner, R. W. G., Fung, J. J., Bustami, R. T., Barr, M. L., & Leichtman, A. B. (2006). Immunosuppression: Evolution in practice and trends, 1994–2004. American Journal of Transplantation, 6(5 Pt 2), 1111–1131. https://doi.org/10.1111/j.1600-6143.2006.01270.x
- Meltzer, P. S., & Helman, L. J. (2021). New horizons in the treatment of osteosarcoma. New England Journal of Medicine, 385(22), 2066–2076. https://doi.org/10.1056/NEJMra2103423
- Menzella, H. G., Reid, R., Carney, J. R., Chandran, S. S., Reisinger, S. J., Patel, K. G., Hopwood, D. A., & Santi, D. V. (2005). Combinatorial polyketide biosynthesis by de novo design and rearrangement of modular polyketide synthase genes. Nature Biotechnology, 23(9), 1171–1176. https://doi.org/10.1038/nbt1128
- Miao, Z. W., Wang, Z., Zheng, S. L., Wang, S. N., & Miao, C. Y. (2024). Anti-stroke biologics: From recombinant proteins to stem cells and organoids. Stroke and Vascular Neurology, svn-2023-002883. https://doi.org/10.1136/svn-2023-002883
10.1136/svn-2023-002883 Google Scholar
- Miethke, M., Pieroni, M., Weber, T., Brönstrup, M., Hammann, P., Halby, L., Arimondo, P. B., Glaser, P., Aigle, B., Bode, H. B., Moreira, R., Li, Y., Luzhetskyy, A., Medema, M. H., Pernodet, J. L., Stadler, M., Tormo, J. R., Genilloud, O., Truman, A. W., … Müller, R. (2021). Towards the sustainable discovery and development of new antibiotics. Nature Reviews Chemistry, 5, 726–749. https://doi.org/10.1038/s41570-021-00313-1
- Mihaylova, N. M., Manoylov, I. K., Nikolova, M. H., Prechl, J., & Tchorbanov, A. I. (2024). DNA and protein-generated chimeric molecules for delivery of influenza viral epitopes in mouse and humanized NSG transfer models. Human Vaccines & Immunotherapeutics, 20(1), 2292381. https://doi.org/10.1080/21645515.2023.2292381
- Mills, H., Acquah, R., Tang, N., Cheung, L., Klenk, S., Glassen, R., Pirson, M., Albert, A., Hoang, D. T., & Van, T. N. (2022). The use of bacteria in cancer treatment: A review from the perspective of cellular microbiology. Emergency Medicine International, 2022, 1–6. https://doi.org/10.1155/2022/8127137
- Mirhoseini, A., Amani, J., & Nazarian, S. (2018). Review on pathogenicity mechanism of enterotoxigenic Escherichia coli and vaccines against it. Microbial Pathogenesis, 117, 162–169. https://doi.org/10.1016/j.micpath.2018.02.032
- Mitra, S., & Tomar, P. C. (2021). Hybridoma technology; advancements, clinical significance, and future aspects. Journal of Genetic Engineering and Biotechnology, 19(1), 159. https://doi.org/10.1186/s43141-021-00264-6
- Montané, X., Kowalczyk, O., Reig-Vano, B., Bajek, A., Roszkowski, K., Tomczyk, R., Pawliszak, W., Giamberini, M., Mocek-Płóciniak, A., & Tylkowski, B. (2020). Current perspectives of the applications of polyphenols and flavonoids in cancer therapy. Molecules, 25(15), 3342. https://doi.org/10.3390/molecules25153342
- Moore, S. J., Lai, H. E., Li, J., & Freemont, P. S. (2023). Streptomyces cell-free systems for natural product discovery and engineering. Natural Product Reports, 40(2), 228–236. https://doi.org/10.1039/d2np00057a
- Moreau Bachelard, C., Coquan, E., du Rusquec, P., Paoletti, X., & Le Tourneau, C. (2021). Risks and benefits of anticancer drugs in advanced cancer patients: A systematic review and meta-analysis. EClinicalMedicine, 40, 101130. https://doi.org/10.1016/j.eclinm.2021.101130
- Mullis, K. B., & Faloona, F. A. (1987). Specific synthesis of DNA in vitro via a polymerase-catalyzed chain reaction. Methods in Enzymology, 155, 335–350. https://doi.org/10.1016/0076-6879(87)55023-6
- Murray, P. R., Rosenthal, K., & Pfaller, M. A. (2021). Medical microbiology ( 9th ed). Elsevier.
- Mutlu, E. A., Keshavarzian, A., Losurdo, J., Swanson, G., Siewe, B., Forsyth, C., French, A., Demarais, P., Sun, Y., Koenig, L., Cox, S., Engen, P., Chakradeo, P., Abbasi, R., Gorenz, A., Burns, C., & Landay, A. (2014). A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects. PLoS Pathogens, 10(2), e1003829. https://doi.org/10.1371/journal.ppat.1003829
- Nelson, A. W. (1910). Ehrlich's cure for syphilis with hata's dioxydiamidoarsenobenzol. Texas Medical Journal, 26(3), 102–106.
- Newman, D. J., & Cragg, G. M. (2020). Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. Journal of Natural Products, 83(3), 770–803. https://doi.org/10.1021/acs.jnatprod.9b01285
- Nicolaou, K. C., & Rigol, S. (2018). A brief history of antibiotics and select advances in their synthesis. The Journal of Antibiotics, 71(2), 153–184. https://doi.org/10.1038/ja.2017.62
- Nie, X., Wang, D., Pan, Y., Lü, P., & Yang, Y. (2024). Discovery, classification and application of the CPISPR-Cas13 system. Technology and Health Care, 32(2), 525–544. https://doi.org/10.3233/THC-230258
- Nishijima, T., Gatanaga, H., & Oka, S. (2019). Tenofovir nephrotoxicity among Asians living with HIV: Review of the literature. Global Health & Medicine, 1(2), 88–94. https://doi.org/10.35772/ghm.2019.01021
- Nova, P., Gomes, A. M., & Costa-Pinto, A. R. (2023). It comes from the sea: Macroalgae-derived bioactive compounds with anti-cancer potential. Critical Reviews in Biotechnology, 1–15. https://doi.org/10.1080/07388551.2023.2174068
10.1080/07388551.2023.2174068 Google Scholar
- Di Nunno, V., Gatto, L., Tosoni, A., Bartolini, S., & Franceschi, E. (2023). Implications of BRAF V600E mutation in gliomas: Molecular considerations, prognostic value and treatment evolution. Frontiers in Oncology, 12, 1067252. https://doi.org/10.3389/fonc.2022.1067252
- Oates, J. A., Wood, A. J. J., Jankovic, J., & Brin, M. F. (1991). Therapeutic uses of botulinum toxin. New England Journal of Medicine, 324(17), 1186–1194. https://doi.org/10.1056/NEJM199104253241707
- Ohnishi, Y., Tamura, Y., Yoshida, M., Tokunaga, K., & Hohjoh, H. (2008). Enhancement of allele discrimination by introduction of nucleotide mismatches into siRNA in allele-specific gene silencing by RNAi. PLoS One, 3(5), e2248. https://doi.org/10.1371/journal.pone.0002248
- Ordóñez-Robles, M., Santos-Beneit, F., Albillos, S. M., Liras, P., Martín, J. F., & Rodríguez-García, A. (2017). Streptomyces tsukubaensis as a new model for carbon repression: Transcriptomic response to tacrolimus repressing carbon sources. Applied Microbiology and Biotechnology, 101(22), 8181–8195. https://doi.org/10.1007/s00253-017-8545-5
- Ordóñez-Robles, M., Santos-Beneit, F., & Martín, J. (2018). Unraveling nutritional regulation of tacrolimus biosynthesis in Streptomyces tsukubaensis through omic approaches. Antibiotics (USSR), 7(2), 39. https://doi.org/10.3390/antibiotics7020039
10.3390/antibiotics7020039 Google Scholar
- Oxford, A. E., Raistrick, H., & Simonart, P. (1939). Studies in the biochemistry of micro-organisms. Biochemical Journal, 33(2), 240–248. https://doi.org/10.1042/bj0330240
- Pai, L., Patil, S., Liu, S., & Wen, F. (2023). A growing battlefield in the war against biofilm-induced antimicrobial resistance: Insights from reviews on antibiotic resistance. Frontiers in Cellular and Infection Microbiology, 13, 1327069. https://doi.org/10.3389/fcimb.2023.1327069
- Pang, Y., Zhang, H., Wen, H., Wan, H., Wu, H., Chen, Y., Li, S., Zhang, L., Sun, X., Li, B., & Liu, X. (2022). Yeast probiotic and yeast products in enhancing livestock feeds utilization and performance: An overview. Journal of Fungi, 8(11), 1191. https://doi.org/10.3390/jof8111191
- Di Paolo, C., Willuda, J., Kubetzko, S., Lauffer, I., Tschudi, D., Waibel, R., Plückthun, A., Stahel, R. A., & Zangemeister-Wittke, U. (2003). A recombinant immunotoxin derived from a humanized epithelial cell adhesion molecule-specific single-chain antibody fragment has potent and selective antitumor activity. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, 9(7), 2837–2848.
- dos Passos, G. R., Adoni, T., Mendes, M. F., & Sato, D. K. (2023). Reshaping neuroimmunology: Diagnosis and treatment in the era of precision medicine. Arquivos de Neuro-psiquiatria, 81(12), 1125–1133. https://doi.org/10.1055/s-0043-1777752
- Patil, A. D., Kasabe, P. J., & Dandge, P. B. (2022). Pharmaceutical and nutraceutical potential of natural bioactive pigment: Astaxanthin. Natural Products and Bioprospecting, 12(1), 25. https://doi.org/10.1007/s13659-022-00347-y
- Peck, M., Rothenberg, M. E., Deng, R., Lewin-Koh, N., She, G., Kamath, A. V., Carrasco-Triguero, M., Saad, O., Castro, A., Teufel, L., Dickerson, D. S., Leonardelli, M., & Tavel, J. A. (2019). A phase 1, randomized, single-ascending-dose study to investigate the safety, tolerability, and pharmacokinetics of DSTA4637S, an anti-Staphylococcus aureus thiomab antibody-antibiotic conjugate, in healthy volunteers. Antimicrobial Agents and Chemotherapy, 63(6), e02588. https://doi.org/10.1128/AAC.02588-18
- Poria, R., Kala, D., Nagraik, R., Dhir, Y., Dhir, S., Singh, B., Kaushik, N. K., Noorani, M. S., Kaushal, A., & Gupta, S. (2024). Vaccine development: Current trends and technologies. Life Sciences, 336, 122331. https://doi.org/10.1016/j.lfs.2023.122331
- PRAC. (2021). Penicillins toxicity and recommendations. https://www.ema.europa.eu/en/documents/prac-recommendation/prac-recommendations-signals-adopted-27-30-september-2021-prac-meeting_en.pdf
- Qi, L. S., Larson, M. H., Gilbert, L. A., Doudna, J. A., Weissman, J. S., Arkin, A. P., & Lim, W. A. (2013). Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell, 152(5), 1173–1183. https://doi.org/10.1016/j.cell.2013.02.022
- Qi, Y. K., Tang, X., Wei, N. N., Pang, C. J., Du, S. S., & Wang, K. (2022). Discovery, synthesis, and optimization of teixobactin, a novel antibiotic without detectable bacterial resistance. Journal of Peptide Science, 28(11), e3428. https://doi.org/10.1002/psc.3428
- Qin, R., You, F. M., Zhao, Q., Xie, X., Peng, C., Zhan, G., & Han, B. (2022). Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: From molecular mechanisms to potential therapeutic targets. Journal of Hematology & Oncology, 15(1), 133. https://doi.org/10.1186/s13045-022-01350-z
- Ramírez-Rendon, D., Passari, A. K., Ruiz-Villafán, B., Rodríguez-Sanoja, R., Sánchez, S., & Demain, A. L. (2022). Impact of novel microbial secondary metabolites on the pharma industry. Applied Microbiology and Biotechnology, 106(5–6), 1855–1878. https://doi.org/10.1007/s00253-022-11821-5
- Randall, J. R., & Davies, B. W. (2021). Mining for novel antibiotics. Current Opinion in Microbiology, 63, 66–69. https://doi.org/10.1016/j.mib.2021.06.001
- Rangel-Torres, B. E., García-Montoya, I. A., Jiménez-Vega, F., & Rodríguez-Tadeo, A. (2022). Efecto de los prebióticos, probióticos y simbióticos sobre marcadores moleculares de inflamación en la obesidad [Effect of prebiotics, probiotics, and symbiotics on molecular markers of inflammation in obesity]. Revista Espanola de Salud Publica [in Spanish], 96, e202212090.
- Rao, D. B., Sane, P. G., & Georgiev, E. L. (1975). Collagenase in the treatment of dermal and decubitus ulcers. Journal of the American Geriatrics Society, 23(1), 22–30. https://doi.org/10.1111/j.1532-5415.1975.tb00376.x
- Ribeiro, R., Costa, L., Pinto, E., Sousa, E., & Fernandes, C. (2023). Therapeutic potential of marine-derived cyclic peptides as antiparasitic agents. Marine Drugs, 21(12), 609. https://doi.org/10.3390/md21120609
- Rodríguez-Cendal, A. I., Gómez-Seoane, I., de Toro-Santos, F. J., Fuentes-Boquete, I. M., Señarís-Rodríguez, J., & Díaz-Prado, S. M. (2023). Biomedical applications of the biopolymer poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV): Drug encapsulation and scaffold fabrication. International Journal of Molecular Sciences, 24(14), 11674. https://doi.org/10.3390/ijms241411674
- Roohvand, F., Ehsani, P., Abdollahpour-Alitappeh, M., Shokri, M., & Kossari, N. (2020). Biomedical applications of yeasts—A patent view, part two: Era of humanized yeasts and expanded applications. Expert Opinion on Therapeutic Patents, 30(8), 609–631. https://doi.org/10.1080/13543776.2020.1781816
- Roohvand, F., Shokri, M., Abdollahpour-Alitappeh, M., & Ehsani, P. (2017). Biomedical applications of yeast—A patent view, part one: Yeasts as workhorses for the production of therapeutics and vaccines. Expert Opinion on Therapeutic Patents, 27(8), 929–951. https://doi.org/10.1080/13543776.2017.1339789
- Równicki, M., Lasek, R., Trylska, J., & Bartosik, D. (2020). Targeting type II toxin-antitoxin systems as antibacterial strategies. Toxins, 12(9), 568. https://doi.org/10.3390/toxins12090568
- Saeed, U., Insaf, R. A., Piracha, Z. Z., Tariq, M. N., Sohail, A., Abbasi, U. A., Fida Rana, M. S., Gilani, S. S., Noor, S., Noor, E., Waheed, Y., Wahid, M., Najmi, M. H., & Fazal, I. (2023). Crisis averted: A world united against the menace of multiple drug-resistant superbugs -pioneering anti-AMR vaccines, RNA interference, nanomedicine, CRISPR-based antimicrobials, bacteriophage therapies, and clinical artificial intelligence strategies to safeguard global antimicrobial arsenal. Frontiers in Microbiology, 14, 1270018. https://doi.org/10.3389/fmicb.2023.1270018
- Santos-Beneit, F. (2015). The Pho regulon: A huge regulatory network in bacteria. Frontiers in Microbiology, 6, 402. https://doi.org/10.3389/fmicb.2015.00402
- Santos-Beneit, F. (2018). Genome sequencing analysis of Streptomyces coelicolor mutants that overcome the phosphate-depending vancomycin lethal effect. BMC Genomics, 19(1), 457. https://doi.org/10.1186/s12864-018-4838-z
- Santos-Beneit, F. (2021). Expression of the Mycobacterium tuberculosis RipA cell wall hydrolase in Streptomyces coelicolor hampers vancomycin resistance. Journal of Global Antimicrobial Resistance, 27, 115–117. https://doi.org/10.1016/j.jgar.2021.08.013
- Santos-Beneit, F., Ceniceros, A., Nikolaou, A., Salas, J. A., & Gutierrez-Merino, J. (2022). Identification of antimicrobial compounds in two streptomyces sp. strains isolated from beehives. Frontiers in Microbiology, 13, 742168. https://doi.org/10.3389/fmicb.2022.742168
- Santos-Beneit, F., Fernández-Martínez, L. T., Rodríguez-García, A., Martín-Martín, S., Ordóñez-Robles, M., Yagüe, P., Manteca, A., & Martín, J. F. (2014). Transcriptional response to vancomycin in a highly vancomycin-resistant Streptomyces coelicolor mutant. Future Microbiology, 9(5), 603–622. https://doi.org/10.2217/fmb.14.21
- Santos-Beneit, F., & Martín, J. F. (2013). Vancomycin resistance in Streptomyces coelicolor is phosphate-dependent but is not mediated by the PhoP regulator. Journal of Global Antimicrobial Resistance, 1(2), 109–113. https://doi.org/10.1016/j.jgar.2013.03.003
- Santos-Beneit, F., Ordóñez-Robles, M., & Martín, J. F. (2017). Glycopeptide resistance: Links with inorganic phosphate metabolism and cell envelope stress. Biochemical Pharmacology, 133, 74–85. https://doi.org/10.1016/j.bcp.2016.11.017
- Santos-Beneit, F., Raškevičius, V., Skeberdis, V. A., & Bordel, S. (2021). A metabolic modeling approach reveals promising therapeutic targets and antiviral drugs to combat COVID-19. Scientific Reports, 11(1), 11982. https://doi.org/10.1038/s41598-021-91526-3
- Santos-Beneit, F., Roberts, D. M., Cantlay, S., McCormick, J. R., & Errington, J. (2017). A mechanism for FtsZ-independent proliferation in Streptomyces. Nature Communications, 8(1), 1378. https://doi.org/10.1038/s41467-017-01596-z
- Santos-Beneit, F., Rodríguez-García, A., & Martín, J. F. (2011). Complex transcriptional control of the antibiotic regulator afsS in Streptomyces: PhoP and AfsR are overlapping, competitive activators. Journal of Bacteriology, 193(9), 2242–2251. https://doi.org/10.1128/JB.01462-10
- Santos-Beneit, F., Rodríguez-García, A., Sola-Landa, A., & Martín, J. F. (2009). Cross-talk between two global regulators in Streptomyces: PhoP and AfsR interact in the control of afsS, pstS and phoRP transcription. Molecular Microbiology, 72(1), 53–68. https://doi.org/10.1111/j.1365-2958.2009.06624.x
- Sato, A., Hatta, Y., Imai, C., Oshima, K., Okamoto, Y., Deguchi, T., Hashii, Y., Fukushima, T., Hori, T., Kiyokawa, N., Kato, M., Saito, S., Anami, K., Sakamoto, T., Kosaka, Y., Suenobu, S., Imamura, T., Kada, A., Saito, A. M., … Horibe, K. (2023). Nelarabine, intensive L-asparaginase, and protracted intrathecal therapy for newly diagnosed T-cell acute lymphoblastic leukaemia in children and young adults (ALL-T11): A nationwide, multicenter, phase 2 trial including randomisation in the very high-risk group. The Lancet. Haematology, 10(6), 419. https://doi.org/10.1016/S2352-3026(23)00072-8
- El Sayed, K. A. (2000). Natural products as antiviral agents. Studies in Natural Products Chemistry, 24, 473–572. https://doi.org/10.1016/S1572-5995(00)80051-4
- Schiavo, G. G., Benfenati, F., Poulain, B., Rossetto, O., de Laureto, P. P., DasGupta, B. R., & Montecucco, C. (1992). Tetanus and botulinum-B neurotoxins block neurotransmitter release by proteolytic cleavage of synaptobrevin. Nature, 359(6398), 832–835. https://doi.org/10.1038/359832a0
- Schnell, N., Entian, K. D., Schneider, U., Götz, F., Zähner, H., Kellner, R., & Jung, G. (1988). Prepeptide sequence of epidermin, a ribosomally synthesized antibiotic with four sulphide-rings. Nature, 333(6170), 276–278. https://doi.org/10.1038/333276a0
- Sen, S., & Mansell, T. J. (2020). Yeasts as probiotics: Mechanisms, outcomes, and future potential. Fungal Genetics and Biology, 137, 103333. https://doi.org/10.1016/j.fgb.2020.103333
- Sengupta, S., Kaufmann, A., & Chandra, T. S. (2012). Development of fluorescent reporter tagged RIB gene cassettes for replicative transformation, early expression, and enhanced riboflavin production in Eremothecium ashbyi. Fungal Biology, 116(10), 1042–1051. https://doi.org/10.1016/j.funbio.2012.07.008
- Shafer, R. H., Formica, J. V., Delfini, C., Brown, S. C., & Mirau, P. A. (1982). Biosynthesis and characterization of [15N]actinomycin D and conformational analysis by nitrogen-15 nuclear magnetic resonance. Biochemistry, 21(25), 6496–6503. https://doi.org/10.1021/bi00268a027
- Shah, J. S., Burrascano, J. J., & Ramasamy, R. (2023). Recombinant protein immunoblots for differential diagnosis of tick-borne relapsing fever and Lyme disease. Journal of Vector Borne Diseases, 60(4), 353–364. https://doi.org/10.4103/0972-9062.383641
- Sherpa, R. T., Reese, C. J., & Montazeri Aliabadi, H. (2015). Application of iChip to grow “Uncultivable” microorganisms and its impact on antibiotic discovery. Journal of Pharmacy & Pharmaceutical Sciences, 18(3), 303–315. https://doi.org/10.18433/j30894
- Siegel, S. E., Stock, W., Johnson, R. H., Advani, A., Muffly, L., Douer, D., Reed, D., Lewis, M., Freyer, D. R., Shah, B., Luger, S., Hayes-Lattin, B., Jaboin, J. J., Coccia, P. F., DeAngelo, D. J., Seibel, N., & Bleyer, A. (2018). Pediatric-inspired treatment regimens for adolescents and young adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: A review. JAMA Oncology, 4(5), 725–734. https://doi.org/10.1001/jamaoncol.2017.5305
- Silva, D. F., Melo, A. L. P., Uchôa, A. F. C., Pereira, G. M. A., Alves, A. E. F., Vasconcellos, M. C., Xavier-Júnior, F. H., & Passos, M. F. (2023). Biomedical approach of nanotechnology and biological risks: A mini-review. International Journal of Molecular Sciences, 24(23), 16719. https://doi.org/10.3390/ijms242316719
- Smits, W. K., Lyras, D., Lacy, D. B., Wilcox, M. H., & Kuijper, E. J. (2016). Clostridium difficile infection. Nature Reviews Disease Primers, 2, 16020. https://doi.org/10.1038/nrdp.2016.20
- Song, D., Yang, X., Chen, Y., Hu, P., Zhang, Y., Zhang, Y., Liang, N., Xie, J., Qiao, L., Deng, G., Chen, F., & Zhang, J. (2023). Advances in anti-tumor based on various anaerobic bacteria and their derivatives as drug vehicles. Frontiers in Bioengineering and Biotechnology, 11, 1286502. https://doi.org/10.3389/fbioe.2023.1286502
- Song, F., Bian, Y., Liu, J., Li, Z., Zhao, L., Fang, J., Lai, Y., & Zhou, M. (2021). Indole alkaloids, synthetic dimers and hybrids with potential in vivo anticancer activity. Current Topics in Medicinal Chemistry, 21(5), 377–403. https://doi.org/10.2174/1568026620666200908162311
- Strecker, J., Ladha, A., Gardner, Z., Schmid-Burgk, J. L., Makarova, K. S., Koonin, E. V., & Zhang, F. (2019). RNA-guided DNA insertion with CRISPR-associated transposases. Science, 365(6448), 48–53. https://doi.org/10.1126/science.aax9181
- Suigo, L., Margolin, W., Ulzurrun, E., Hrast Rambaher, M., Zanotto, C., Sebastián-Pérez, V., Campillo, N. E., Straniero, V., & Valoti, E. (2023). Benzodioxane-benzamides as FtsZ inhibitors: Effects of Linker's functionalization on Gram-positive antimicrobial activity. Antibiotics (USSR), 12(12), 1712. https://doi.org/10.3390/antibiotics12121712
- Survase, S. A., Kagliwal, L. D., Annapure, U. S., & Singhal, R. S. (2011). Cyclosporin—A review on fermentative production, downstream processing and pharmacological applications. Biotechnology Advances, 29(4), 418–435. https://doi.org/10.1016/j.biotechadv.2011.03.004
- Suvarna, V. (2023). Ivermectin: A critical review on characteristics, properties, and analytical methods. Journal of AOAC International, 106(3), 534–557. https://doi.org/10.1093/jaoacint/qsad031
- Taliento, C., Restaino, S., Scutiero, G., Arcieri, M., Bernardi, G., Martinello, R., Driul, L., Perrone, A. M., Fagotti, A., Scambia, G., Greco, P., & Vizzielli, G. (2023). Pressurized intraperitoneal aerosol chemotherapy (PIPAC) with cisplatin and doxorubicin in patients with ovarian cancer: A systematic review. European Journal of Surgical Oncology, 49(12), 107250. https://doi.org/10.1016/j.ejso.2023.107250
- Teachey, D. T., & Pui, C. H. (2019). Comparative features and outcomes between paediatric T-cell and B-cell acute lymphoblastic leukaemia. The Lancet Oncology, 20(3), e142–e154. https://doi.org/10.1016/S1470-2045(19)30031-2
- Thakur, R., Shishodia, S. K., Sharma, A., Chauhan, A., Kaur, S., & Shankar, J. (2024). Accelerating the understanding of Aspergillus terreus: Epidemiology, physiology, immunology and advances. Current Research in Microbial Sciences, 6, 100220. https://doi.org/10.1016/j.crmicr.2024.100220
- Timmons, L., & Fire, A. (1998). Specific interference by ingested dsRNA. Nature, 395(6705), 854. https://doi.org/10.1038/27579
- Tindall, K. R., & Kunkel, T. A. (1988). Fidelity of DNA synthesis by the Thermus aquaticus DNA polymerase. Biochemistry, 27(16), 6008–6013. https://doi.org/10.1021/bi00416a027
- Tomasz, M. (1995). Mitomycin C: Small, fast and deadly (but very selective). Chemistry & Biology, 2(9), 575–579. https://doi.org/10.1016/1074-5521(95)90120-5
- Tran, N. H., Rahman, M. Z., He, L., Xin, L., Shan, B., & Li, M. (2016). Complete de novo assembly of monoclonal antibody sequences. Scientific Reports, 6, 31730. https://doi.org/10.1038/srep31730
- Tu, Y., Tan, L., Tao, H., Li, Y., & Liu, H. (2023). CETSA and thermal proteome profiling strategies for target identification and drug discovery of natural products. Phytomedicine, 116, 154862. https://doi.org/10.1016/j.phymed.2023
- Uğurlu, Ö., Man, E., Gök, O., Ülker, G., Soytürk, H., Özyurt, C., & Evran, S. (2024). A review of aptamer-conjugated nanomaterials for analytical sample preparation: Classification according to the utilized nanomaterials. Analytica Chimica Acta, 1287, 342001. https://doi.org/10.1016/j.aca.2023.342001
- Umezawa, H., Maeda, K., Takeuchi, T., & Okami, Y. (1966). New antibiotics, bleomycin A and B. The Journal of Antibiotics, 19(5), 200–209.
- Usman, S. S., Uba, A. I., & Christina, E. (2023). Bacteriophage genome engineering for phage therapy to combat bacterial antimicrobial resistance as an alternative to antibiotics. Molecular Biology Reports, 50(8), 7055–7067. https://doi.org/10.1007/s11033-023-08557-4
- Ustick, L. J., Larkin, A. A., Garcia, C. A., Garcia, N. S., Brock, M. L., Lee, J. A., Wiseman, N. A., Moore, J. K., & Martiny, A. C. (2021). Metagenomic analysis reveals global-scale patterns of ocean nutrient limitation. Science, 372(6539), 287–291. https://doi.org/10.1126/science.abe6301
- Velásquez, P. A., Hernandez, J., Galeano, E., Hincapié-García, J., Rugeles, M., & Zapata-Builes, W. (2024). Effectiveness of drug repurposing and natural products against SARS-CoV-2: A comprehensive review. Clinical Pharmacology: Advances and Applications, 16, 1–25. https://doi.org/10.2147/CPAA.S429064
- Vivier, E., Rebuffet, L., Narni-Mancinelli, E., Cornen, S., Igarashi, R. Y., & Fantin, V. R. (2024). Natural killer cell therapies. Nature, 626(8000), 727–736. https://doi.org/10.1038/s41586-023-06945-1
- Waitayangkoon, P., Moon, S. J., Tirupur Ponnusamy, J. J., Zeng, L., Driban, J., & McAlindon, T. (2023). Long-term safety profiles of macrolides and tetracyclines: A systematic review and meta-analysis. The Journal of Clinical Pharmacology, 64, 164–177. https://doi.org/10.1002/jcph.2358
- Walker, A. W., & Hoyles, L. (2023). Human microbiome myths and misconceptions. Nature Microbiology, 8(8), 1392–1396. https://doi.org/10.1038/s41564-023-01426-7
- Wang, G. (2023). The antimicrobial peptide database is 20 years old: Recent developments and future directions. Protein Science, 32(10), e4778. https://doi.org/10.1002/pro.4778
- Wang, H., Lottenberg, R., & Boyle, M. D. P. (1995). Analysis of the interaction of group A streptococci with fibrinogen, streptokinase and plasminogen. Microbial Pathogenesis, 18(3), 153–166. https://doi.org/10.1016/s0882-4010(95)90013-6
- Wang, Y., Zhang, K., Zhao, Y., Li, Y., Su, W., & Li, S. (2023). Construction and applications of mammalian cell-based DNA-encoded peptide/protein libraries. ACS Synthetic Biology, 12(7), 1874–1888. https://doi.org/10.1021/acssynbio.3c00043
- Watve, M., Tickoo, R., Jog, M., & Bhole, B. (2001). How many antibiotics are produced by the genus Streptomyces? Archives of Microbiology, 176(5), 386–390. https://doi.org/10.1007/s002030100345
- Wei, D., & Tan, Y. (2023). Research progress on medical devices of polyhydroxyalkanoate in orthopedics. Zhongguo xiu fu chong jian wai ke za zhi = Zhongguo xiufu chongjian waike zazhi = Chinese Journal of Reparative and Reconstructive Surgery [in Chinese], 37(8), 909–917. https://doi.org/10.7507/1002-1892.202211018
- Wheeler, A. H. (1997). Therapeutic uses of botulinum toxin. American Family Physician, 55(2), 541–545.
- Woolhouse, M., Scott, F., Hudson, Z., Howey, R., & Chase-Topping, M. (2012). Human viruses: Discovery and emergence. Philosophical Transactions of the Royal Society, B: Biological Sciences, 367(1604), 2864–2871. https://doi.org/10.1098/rstb.2011.0354
- Worley, M. V., & Estrada, S. J. (2014). Bedaquiline: A novel antitubercular agent for the treatment of multidrug-resistant tuberculosis. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy, 34(11), 1187–1197. https://doi.org/10.1002/phar.1482
- Xiang, S., Fruehauf, J., & Li, C. J. (2006). Short hairpin RNA-expressing bacteria elicit RNA interference in mammals. Nature Biotechnology, 24(6), 697–702. https://doi.org/10.1038/nbt1211
- Xuan, J., Feng, W., Wang, J., Wang, R., Zhang, B., Bo, L., Chen, Z. S., Yang, H., & Sun, L. (2023). Antimicrobial peptides for combating drug-resistant bacterial infections. Drug Resistance Updates, 68, 100954. https://doi.org/10.1016/j.drup.2023.100954
- Yahsi, B., Palaz, F., & Dincer, P. (2024). Applications of CRISPR epigenome editors in tumor immunology and autoimmunity. ACS Synthetic Biology, 13(2), 413–427. https://doi.org/10.1021/acssynbio.3c00524
- Yang, D., Eun, H., & Prabowo, C. P. S. (2023). Metabolic engineering and synthetic biology approaches for the heterologous production of aromatic polyketides. International Journal of Molecular Sciences, 24(10), 8923. https://doi.org/10.3390/ijms24108923
- Yang, Y., Jiang, X., Lai, H., & Zhang, X. (2022). Smart bacteria-responsive drug delivery systems in medical implants. Journal of Functional Biomaterials, 13(4), 173. https://doi.org/10.3390/jfb13040173
- Yuan, Y., Shi, C., & Zhao, H. (2023). Machine learning-enabled genome mining and bioactivity prediction of natural products. ACS Synthetic Biology, 12(9), 2650–2662. https://doi.org/10.1021/acssynbio.3c00234
- Yuzawa, S., Backman, T. W. H., Keasling, J. D., & Katz, L. (2018). Synthetic biology of polyketide synthases. Journal of Industrial Microbiology and Biotechnology, 45(7), 621–633. https://doi.org/10.1007/s10295-018-2021-9
- Zahedipour, F., Zahedipour, F., Zamani, P., Jaafari, M. R., & Sahebkar, A. (2024). Harnessing CRISPR technology for viral therapeutics and vaccines: From preclinical studies to clinical applications. Virus Research, 341, 199314. https://doi.org/10.1016/j.virusres.2024.199314
- Zampaloni, C., Mattei, P., Bleicher, K., Winther, L., Thäte, C., Bucher, C., Adam, J. M., Alanine, A., Amrein, K. E., Baidin, V., Bieniossek, C., Bissantz, C., Boess, F., Cantrill, C., Clairfeuille, T., Dey, F., Di Giorgio, P., du Castel, P., Dylus, D., … Bradley, K. A. (2024). A novel antibiotic class targeting the lipopolysaccharide transporter. Nature, 625(7995), 566–571. https://doi.org/10.1038/s41586-023-06873-0
- Zawada, J. F., Burgenson, D., Yin, G., Hallam, T. J., Swartz, J. R., & Kiss, R. D. (2022). Cell-free technologies for biopharmaceutical research and production. Current Opinion in Biotechnology, 76, 102719. https://doi.org/10.1016/j.copbio.2022.102719
- Zhang, B., Li, A., Zuo, F., Yu, R., Zeng, Z., Ma, H., & Chen, S. (2016). Recombinant Lactococcus lactis NZ9000 secretes a bioactive kisspeptin that inhibits proliferation and migration of human colon carcinoma HT-29 cells. Microbial Cell Factories, 15(1), 102. https://doi.org/10.1186/s12934-016-0506-7
- Zhao, J., Shakir, Y., Deng, Y., & Zhang, Y. (2023). Use of modified ichip for the cultivation of thermo-tolerant microorganisms from the hot spring. BMC Microbiology, 23(1), 56. https://doi.org/10.1186/s12866-023-02803-2
- Zhou, F., & Zhang, D. (2023). Recent advance in the development of tuberculosis vaccines in clinical trials and virus-like particle-based vaccine candidates. Frontiers in Immunology, 14, 1238649. https://doi.org/10.3389/fimmu.2023
- Zhou, J., Ren, X., Wang, X., Li, Z., & J Xian, C. (2023). Recent advances and challenges of the use of the CRISPR/Cas system as a non-nucleic acid molecular diagnostic. Heliyon, 9(12), e22767. https://doi.org/10.1016/j.heliyon.2023.e22767
- Zhu, D., Mengyue, M., Qimuge, A., Bilige, B., Baiyin, T., Temuqile, T., Chen, S., Borjigen, S., Baigude, H., & Yang, D. (2022). Oral delivery of SARS-CoV-2 DNA vaccines using attenuated salmonella typhimurium as a carrier in rat. Molecular Genetics, Microbiology and Virology, 37(3), 159–166. https://doi.org/10.3103/S0891416822030107
- Zhu, S., Su, Y., Shams, S., Feng, Y., Tong, Y., & Zheng, G. (2019). Lassomycin and lariatin lasso peptides as suitable antibiotics for combating mycobacterial infections: Current state of biosynthesis and perspectives for production. Applied Microbiology and Biotechnology, 103(10), 3931–3940. https://doi.org/10.1007/s00253-019-09771-6