Artificial Intelligence in Combating Antimicrobial Resistance: Comparison
Please note this is a comparison between Version 2 by Jessie Wu and Version 3 by Jessie Wu.

Artificial intelligence (AI) is a branch of science and engineering that focuses on the computational understanding of intelligent behavior. Many human professions, including clinical diagnosis and prognosis, are greatly useful from AI. TAntimicrobial resistance (AMR) is among the rising incidence of antimicrobial resistance most critical challenges facing Pakistan and the rest of the world. The rising incidence of AMR has become a significant issue, and authorities must take measures to combat the overuse and incorrect use of antibiotics in order to combat rising resistance rates. The widespread use of antibiotics in clinical practice has not only resulted in drug resistance but has also increased the threat of superresistant bacteria emergence. As AMR rises, clinicians find it more difficult to treat many bacterial infections in a timely manner, and therapy becomes prohibitively costly for patients. To combat the rise in AMR rates, it is critical to implement an institutional antibiotic stewardship program that monitors correct antibiotic use, controls antibiotics, and generates antibiograms. Furthermore, these types of tools may aid in the treatment of patients in the event of a medical emergency in which a physician is unable to wait for bacterial culture results. AI’s applications in healthcare might be unlimited, reducing the time it takes to discover new antimicrobial drugs, improving diagnostic and treatment accuracy, and lowering expenses at the same time. The majority of suggested AI solutions for AMR are meant to supplement rather than replace a doctor’s prescription or opinion, but rather to serve as a valuable tool for making their work easier. When it comes to infectious diseases, AI has the potential to be a game-changer in the battle against antibiotic resistance.  Finally, when selecting antibiotic therapy for infections, data from local antibiotic stewardship programs are critical to ensuring that these bacteria are treated quickly and effectively. Furthermore, organizations such as the World Health Organization (WHO) have underlined the necessity of selecting the appropriate antibiotic and treating for the shortest time feasible to minimize the spread of resistant and invasive resistant bacterial strains.

  • AI
  • AMR
  • AST

1. Introduction

The widespread use of antibiotics in clinical practice has not only resulted in drug resistance but has also increased the threat of super-resistant bacteria emergence. Pakistan is one of the countries that have a high rate of antimicrobial resistance (AMR) and little healthcare expertise and assistance to tackle it, which raises questions about high AMR rates. Alexander Fleming’s discovery of penicillin in 1928 marked the beginning of the modern age of antibiotics [1]. Since then, antibiotics have saved the lives of many individuals suffering from bacterial and fungal infections. However, the widespread use of antibiotics in clinical practice has resulted in drug resistance, in addition to increasing the threat of super-resistant bacteria emergence [2]. Antimicrobial resistance (AMR) is anticipated to cause around 10 million deaths per year by 2050, and the economic impact of AMR is expected to approach USD 100 billion during the same period [3]. It is imperative that required efforts to implement new regulations and revive research efforts to manage the AMR epidemic are carried out to address this crisis [4].
Due to the recent AMR emergence, the world is in desperate need of some relief, and to this end, the Food and Drug Administration of the United States has proposed regulations that would specify the types, quantities, and frequencies of adequate antibiotic use [5]. A complete prohibition on the use of antibiotics in cattle feed was recommended by the European Union in 2006 [6]. Japanese and Chinese policymakers, in contrast to their counterparts in Europe and the United States, have concentrated on proposals that are more compelling in nature. In 2016, the Chinese government announced the National Action Plan to Contain Antimicrobial Resistance (NAPACAR) [7]. However, despite increased awareness of antimicrobial resistance (AMR), the general situation is deteriorating, and people must continue to create antimicrobial peptides (AMPs), antibiotic combinations, and monitoring systems to effectively control AMR [8].
Artificial intelligence (AI) has demonstrated substantial competence in the field of AMR control in recent years. For example, artificial intelligence applications based on sequencing have been used to explore AMR [9]. Furthermore, the collection of clinical data for the development of clinical decision support systems could assist clinicians in monitoring trends in antimicrobial resistance to promote antibiotics’ sensible applications [10]. Additionally, artificial intelligence applications are commonly used in the development of new antibiotics and the exploration of synergistic medication combinations [11]. Interestingly, most past publications on AMR have also been written from the standpoint of structural and molecular mechanisms [12]. The schematic diagram of the possible use of AI and the dataflow integration is shown in Figure 1.
Figure 1. Schematic diagram of dataflow integration.

Antibiotic Resistance; the Current Scenario

Antibiotics are medications that are utilized to both prevent and treat infections caused by bacteria and fungi in animals (significantly humans). Antibiotic resistance arises when bacteria alter their genetic makeup in response to the usage of antibiotics [13]. Antibiotic-resistant bacteria are the main causative agents of antibiotic resistance. It is possible for these germs to infect higher-order animals, and the diseases resulting from such infections are more difficult to tackle, compared with those resulting from nonresistant bacterial infections [14]. longer hospital stays, higher medical expenses, and an increased mortality rate are all associated with this phenomenon [15].
Studies show that majority of infections exhibit strong resistance to routinely used medicines; in addition, researchers are discovering gaps and breaches in surveillance and methodical data collection [16]. Based on these data, it is urged that surveillance practices must be formalized, and specific efforts must be taken to prevent AMR in the region. Furthermore, the world needs to change the way it consumes antibiotics. Without a change in public behavior, medical expeditions to find newer antibiotics will not be fruitful. The adoption of new behaviors should include measures to minimize the transmission of infectious diseases, such as immunization, hand washing, and excellent food hygiene [17].
Increasing numbers of antibiotic-resistant microorganisms are being discovered in hospitals and the general surroundings. Therefore, it is imperative to formulate new antibiotics to combat these increasing cases, but development has been slow in this area. Historically, most antibiotics have been derived from a few numbers of molecular scaffolds, with their viability extended by cycles of synthetic tailoring and optimization [18]. Considering the escalation of multidrug resistance in the most-recent generation of pathogens, the identification of novel scaffolds is a top priority. New techniques of scaffold discovery and identification are gaining traction, such as mining untapped microbial pockets for natural compounds, building screens to avoid rehashing old scaffolds, and reclaiming synthetic molecular catalogs as antibiotics [19].
Due to the development of high-throughput gene sequencing, researchers have a potent tool for profiling the complete DNA complement, which includes ARGs and DNA extracts taken from a variety of environmental sources [20]. Using this type of metagenomics technique, for example, ARGs have now been identified in many environmental samples such as soil, cattle dung, wastewater treatment plants, compost, water, and other potentially contaminated habitats [21].

2. Artificial Intelligence against Antibiotic Resistance

Antibiotic resistance (AMR) is, unfortunately, a result of antibiotic misuse. As AMR drastically reduces antibiotic therapeutic efficacy, it is critical that researchers follow its emergence and dissemination [22]. Currently, two approaches for diagnosing AMR are commonly utilized. One is called the whole-genome sequencing for antimicrobial susceptibility testing (WGS-AST) and the other one is antibiotic susceptibility testing (AST). The latter is the traditional approach for quantifying antimicrobial resistance levels, but it is not efficient, nor does it explain the mechanism of antimicrobial resistance [23]. It is possible to diagnose AMR with high accuracy and consistency using WGS-AST; however, to extract information properly, large, and high-dimensional datasets are required [12]. As a result, artificial intelligence technologies are being used to improve upon existing methodologies in the previously discussed ways [9]Table 1 shows the application of AI in efforts to control high AMR rates with their advantages and disadvantages.
In the computer sciences field, AI has a dynamic part to play in human intelligence-stimulation systems and its research. The processes including speech recognition, visual perception, natural language processing, and decision making according to perceived data are stimulated using the technology [9][11]. The metadata from available health records and developments in processing performance are critical factors in the growth of AI systems. These two aspects are inextricably linked to complicated mathematical algorithms including neural networks (NN) and machine/deep learning, which are inextricably linked to elements such as health records and breakthroughs in computer performance [24]. This is especially true with the development of deep neural network designs, where the sophistication (commonly known as the number of factors the networks must learn) has skyrocketed in the previous decade [25].
ML is a part of artificial intelligence that goes through a change in its results when dealing with a large capacity of data. While specialized systems are related to the expertise of humans, likely to the human brain working. This characteristic makes it independent of man’s specialties [26]. NNs, then, are numerical informatics estimation models dependent on the working of organic neural organizations (human or creature) and, ultimately, models comprising interconnections of data that can perceive a dataset’s fundamental connections. A DNN is designed using a few layers (generally above five) of handling units that permit researchers to further develop forecasts from the information, thus finding how to comprehend them autonomously [27].
A significant advantage of NNs is that their display improves dynamically as the quantity of the dataset grows [28], allowing them to adapt to shifting information sources [11]. Currently, there are a plethora of elements associated with a patient’s consideration and clinical history that complicate patient administration. According to a new distribution, multiple times more clinical data than a person would have the choice to read in their lifetime would be provided within the present year [9]. Artificial intelligence, via naturally dealing with this massive amount of information, can assume a progressive part in supporting clinical dynamics. Nevertheless, even today, most specialists do not comprehend the convenience of AI and continue to settle on choices dependent on close-to-home insight and therapy rules [12]. This audit is to demonstrate the possible contribution of AI in battling the developing marvel of AMR, with specific pediatric patients at the center. The focus was placed on the utilization of artificial intelligence for pediatric infections in settled countries [9]. A difference between the latest AI-based diagnosis methods for AST versus the gold standards methods is shown in Figure 2.
Figure 2. Gold standards method for AST (disc diffusion) v/s automation methods.

Assistance Strategies of Artificial Intelligence in Antimicrobial Resistance

Early detection of infectious diseases, the differentiation between infectious and noninfectious pathologies, and correct therapy of consequences are all important aspects of combating antibiotic resistance. In this global issue, AI can play a very vital role. The preparation of antibiograms and then the development of personalized machine learning (ML)-based AMR prediction models could be very useful AI techniques for high-peak risk infectious bugs and their trends in the susceptibility patterns [9]. Using this strategy, Yelin et al. conducted a study to examine a 10-year longitudinal dataset of over 0.7 million community-acquired UTIs and identified a significant association between AMR and demographic characteristics, previous history of urine cultures, and the previous history of using the antibiotics by the patients. After examinations, they developed an ML-based AMR prediction model and described the high potential bugs for UTIs and their AMR patterns [29]. The description of the use of deep sequencing AI models is shown in Figure 3.
Figure 3. Deep antibiotic-resistant gene-sequencing model.

3. Artificial Intelligence vs. Antibiotic Stewardship Program

Many publications prove the importance of an antimicrobial stewardship program (ASP). The data clearly demonstrate the significant impact an ASP has in reducing the overuse of antibiotics and reducing the collateral damage that often results from the overutilization of antimicrobials [30][31][32]. The data also indicate that an ASP leads to an infection prevention strategy [33]. The increasing risk of antimicrobial resistance due to inappropriate prescribing habits has led to devastating consequences [34]. The CDC estimated that, in the US alone, 28 lakh individuals contract an antibiotic-resistant infection, and more than 35 thousand individual annual mortality [35].
In addition, the Centers for Disease Control and Prevention also documented 223,900 incidents of Clostridioides difficile in the year 2017 and around 13,000 people were reported to be deceased [35]. As a result, the Centers for Medicare and Medicaid Services (CMS) now needed hospitals and nursing homes to implement ASPs [36]. However, this requirement is not found in the outpatient setting. Recently, the Joint Commission, an independent body that provides accreditation and certification to healthcare institutions, has made it a requirement to have ASPs in ambulatory medical institutions that are acknowledged by the joint Commission and prescribe antibiotics routinely [37]. However, this initiative has not been adopted by other agencies. Based on research, most studies confirming the benefits of an ASP have focused exclusively on hospital settings, thereby limiting the quantity of available data regarding the benefits of an ASP in outpatient settings. Outpatient ASPs may be difficult to implement due to many obstacles [35].
This includes limited resources, such as limited personnel with expertise in infectious disease and antimicrobials, the inability to track data across multiple electronic health systems and pharmacies, as well as limited finances, support, and infrastructure [38]. Additional obstacles may include time constraints for otherwise no reimbursable tasks. Clinicians are unlikely to dedicate time to implementing an ASP if it does not generate revenue or could incur additional costs for their practice [39]. A typical ASP program may involve pharmacists, ID physicians, educational programs for providers and patients, and mechanisms in place for interventions, tracking, and reporting data. Current CMS- and relative value unit (RVU)-based payment models are tied to the number of patient visits or procedures, and therefore, dedicating time to nonpatient-specific endeavors and nonprocedural may inadvertently affect clinicians’ bottom line [22]. The CDC postulates that a minimum of 30% of prescribed antibiotics in outpatient cases are unnecessary [40]. It has published core elements to promote outpatient ASPs. However, most outpatient facilities do not have the means in place to implement these measures [41].
In addition, since medical practices have not been incentivized to participate in outpatient ASPs, this undertaking has fallen by the wayside. According to the National the Infection Prevention Strategy Ambulatory Medical Care Survey in 2016, more than 60% of patient visits in the United States were to practices with five or fewer practitioners, and only 3% of patient visits were to institutions associated with medical or academic health centers [41]. Lastly, 89.7% of patient visits were to facilities categorized as private practices. Despite the CDC’s figures on the overuse of antibiotics in outpatient settings, there have been no uniformly adapted programs in place to address the need for an ASP in outpatient settings [38]. It is, therefore, imperative that the principles of an ASP are applied to both the inpatient and outpatient settings, to truly have a significant impact on reducing the threat of antimicrobial resistance [40]. Furthermore, an ASP in the outpatient setting must be designed in a way that can be adapted by a variety of institutions with ease and efficiency, regardless of the facilities’ finances, endorsements, and resources available [3].

References

  1. Ahmed, I.; Rabbi, M.B.; Sultana, S. Antibiotic resistance in Bangladesh: A systematic review. Int. J. Infect. Dis. 2019, 80, 54–61.
  2. Bilal, H.; Khan, M.N.; Rehman, T.; Hameed, M.F.; Yang, X. Antibiotic resistance in Pakistan: A systematic review of past decade. BMC Infect. Dis. 2021, 21, 244.
  3. Bullens, M.; de Cerqueira Melo, A.; Raziq, S.; Lee, J.; Khalid, G.; Khan, S.; Zada, A.; Wailly, Y.; Zeshan, S.; Saad, N. Antibiotic resistance in patients with urinary tract infections in Pakistan. Public Health Action 2022, 12, 48–52.
  4. Hormozi, S.F.; Vasei, N.; Aminianfar, M.; Darvishi, M.; Saeedi, A.A. Antibiotic resistance in patients suffering from nosocomial infections in Besat Hospital. Eur. J. Transl. Myol. 2018, 28, 7594.
  5. Susmita, R.C.; Zubayed, A.; Krishna, R.; Abdullah, A.N.; Rashid, M.H.; Kamol, C.M. Emerging threats of antibiotic resistance in Salmonella typhi and Salmonella paratyphi A among enteric fever cases of Dhaka, Bangladesh. Afr. J. Bacteriol. Res. 2022, 14, 8–15.
  6. Martino, F.; Tijet, N.; Melano, R.; Petroni, A.; Heinz, E.; De Belder, D.; Faccone, D.; Rapoport, M.; Biondi, E.; Rodrigo, V. Isolation of five Enterobacteriaceae species harbouring bla NDM-1 and mcr-1 plasmids from a single paediatric patient. PLoS ONE 2019, 14, e0221960.
  7. Gemert, T.V. On the Influence of Dataset Characteristics on Classifier Performance. Bachelor’s Thesis, Utrecht University, Utrecht, The Netherlands, 2017.
  8. Ahmed, N.; Zeshan, B.; Naveed, M.; Afzal, M.; Mohamed, M. Antibiotic resistance profile in relation to virulence genes fimH, hlyA and usp of uropathogenic E. coli isolates in Lahore, Pakistan. Trop. Biomed. 2019, 36, 559–568.
  9. Lv, J.; Deng, S.; Zhang, L. A review of artificial intelligence applications for antimicrobial resistance. Biosaf. Health 2021, 3, 22–31.
  10. Lau, H.J.; Lim, C.H.; Foo, S.C.; Tan, H.S. The role of artificial intelligence in the battle against antimicrobial-resistant bacteria. Curr. Genet. 2021, 67, 421–429.
  11. Raisch, S.; Krakowski, S. Artificial intelligence and management: The automation–augmentation paradox. Acad. Manag. Rev. 2021, 46, 192–210.
  12. Song, L.; Gildea, D.; Zhang, Y.; Wang, Z.; Su, J. Semantic neural machine translation using AMR. Trans. Assoc. Comput. Linguist. 2019, 7, 19–31.
  13. Ahmed, N.; Khan, M.; Saleem, W.; Karobari, M.I.; Mohamed, R.N.; Heboyan, A.; Rabaan, A.A.; Mutair, A.A.; Alhumaid, S.; Alsadiq, S.A. Evaluation of bi-lateral co-infections and antibiotic resistance rates among COVID-19 patients. Antibiotics 2022, 11, 276.
  14. Zhen, X.; Lundborg, C.S.; Sun, X.; Hu, X.; Dong, H. Economic burden of antibiotic resistance in ESKAPE organisms: A systematic review. Antimicrob. Resist. Infect. Control 2019, 8, 137.
  15. Sattar, H.; Toleman, M.; Nahid, F.; Zahra, R. Co-existence of bla NDM-1 and bla KPC-2 in clinical isolates of Klebsiella pneumoniae from Pakistan. J. Chemother. 2016, 28, 346–349.
  16. Williams, M.A.; Wyner, S.N. Antimicrobial Resistance: Facing the Rise of a Global Threat; American Public Health Association: Washington, DC, USA, 2019.
  17. Hadjadj, L.; Syed, M.A.; Bushra, J.; Abbasi, S.A.; Rolain, J.-M. Emergence of Vancomycin-resistant Enterococcus faecium ST 80 in Pakistan. Surg. Infect. 2019, 20, 524–525.
  18. Dantas, G.; Sommer, M.O.; Oluwasegun, R.D.; Church, G.M. Bacteria subsisting on antibiotics. Science 2008, 320, 100–103.
  19. Sohail, M.; Khurshid, M.; Saleem, H.G.M.; Javed, H.; Khan, A.A. Characteristics and antibiotic resistance of urinary tract pathogens isolated from Punjab, Pakistan. Jundishapur J. Microbiol. 2015, 8, e19272.
  20. Fasih, N.; Zafar, A.; Khan, E.; Jabeen, K.; Hasan, R. Clonal dissemination of vanA positive Enterococcus species in tertiary care hospitals in Karachi, Pakistan. J. Pak. Med. Assoc. 2010, 60, 805.
  21. Miller, J.R.; Dunham, S.; Mochalkin, I.; Banotai, C.; Bowman, M.; Buist, S.; Dunkle, B.; Hanna, D.; Harwood, H.J.; Huband, M.D. A class of selective antibacterials derived from a protein kinase inhibitor pharmacophore. Proc. Natl. Acad. Sci. USA 2009, 106, 1737–1742.
  22. Arango-Argoty, G.; Garner, E.; Pruden, A.; Heath, L.S.; Vikesland, P.; Zhang, L. DeepARG: A deep learning approach for predicting antibiotic resistance genes from metagenomic data. Microbiome 2018, 6, 23.
  23. Cánovas-Segura, B.; Campos, M.; Morales, A.; Juarez, J.M.; Palacios, F. Development of a clinical decision support system for antibiotic management in a hospital environment. Prog. Artif. Intell. 2016, 5, 181–197.
  24. Ahmed, N.; Ali, Z.; Riaz, M.; Zeshan, B.; Wattoo, J.I.; Aslam, M.N. Evaluation of antibiotic resistance and virulence genes among clinical isolates of Pseudomonas aeruginosa from cancer patients. Asian Pac. J. Cancer Prev. APJCP 2020, 21, 1333.
  25. Zafar, A.; Hasan, R.; Nizami, S.Q.; von Seidlein, L.; Soofi, S.; Ahsan, T.; Chandio, S.; Habib, A.; Bhutto, N.; Siddiqui, F.J. Frequency of isolation of various subtypes and antimicrobial resistance of Shigella from urban slums of Karachi, Pakistan. Int. J. Infect. Dis. 2009, 13, 668–672.
  26. Nava Lara, R.A.; Aguilera-Mendoza, L.; Brizuela, C.A.; Peña, A.; Del Rio, G. Heterologous machine learning for the identification of antimicrobial activity in Human-Targeted drugs. Molecules 2019, 24, 1258.
  27. van Belkum, A.; Burnham, C.-A.D.; Rossen, J.W.; Mallard, F.; Rochas, O.; Dunne, W.M. Innovative and rapid antimicrobial susceptibility testing systems. Nat. Rev. Microbiol. 2020, 18, 299–311.
  28. Gajdács, M.; Paulik, E.; Szabó, A. Knowledge, attitude and practice of community pharmacists regarding antibiotic use and infectious diseases: A cross-sectional survey in Hungary (KAPPhA-HU). Antibiotics 2020, 9, 41.
  29. Yelin, I.; Snitser, O.; Novich, G.; Katz, R.; Tal, O.; Parizade, M.; Chodick, G.; Koren, G.; Shalev, V.; Kishony, R. Personal clinical history predicts antibiotic resistance of urinary tract infections. Nat. Med. 2019, 25, 1143–1152.
  30. Hameed, F.; Khan, M.A.; Muhammad, H.; Sarwar, T.; Bilal, H.; Rehman, T.U. Plasmid-mediated mcr-1 gene in Acinetobacter baumannii and Pseudomonas aeruginosa: First report from Pakistan. Rev. Soc. Bras. Med. Trop. 2019, 52, e20190237.
  31. Pal, C.; Bengtsson-Palme, J.; Kristiansson, E.; Larsson, D.J. The structure and diversity of human, animal and environmental resistomes. Microbiome 2016, 4, 1–15.
  32. Ahmed, N.; Khalid, H.; Mushtaq, M.; Basha, S.; Rabaan, A.A.; Garout, M.; Halwani, M.A.; Al Mutair, A.; Alhumaid, S.; Al Alawi, Z.; et al. The Molecular Characterization of Virulence Determinants and Antibiotic Resistance Patterns in Human Bacterial Uropathogens. Antibiotics 2022, 11, 516.
  33. Zahra, N.; Zeshan, B.; Qadri, M.M.A.; Ishaq, M.; Afzal, M.; Ahmed, N. Phenotypic and Genotypic Evaluation of Antibiotic Resistance of Acinetobacter baumannii Bacteria Isolated from Surgical Intensive Care Unit Patients in Pakistan. Jundishapur J. Microbiol. 2021, 14, 104922.
  34. Zeshan, B.; Karobari, M.I.; Afzal, N.; Siddiq, A.; Basha, S.; Basheer, S.N.; Peeran, S.W.; Mustafa, M.; Daud, N.H.A.; Ahmed, N. The usage of antibiotics by COVID-19 patients with comorbidities: The risk of increased antimicrobial resistance. Antibiotics 2021, 11, 35.
  35. Saleem, Z.; Godman, B.; Azhar, F.; Kalungia, A.C.; Fadare, J.; Opanga, S.; Markovic-Pekovic, V.; Hoxha, I.; Saeed, A.; Al-Gethamy, M. Progress on the national action plan of Pakistan on antimicrobial resistance (AMR): A narrative review and the implications. Expert Rev. Anti-Infect. Ther. 2022, 20, 71–93.
  36. Oluwafemi, R.; Olawale, I.; Alagbe, J. Recent trends in the utilization of medicinal plants as growth promoters in poultry nutrition—A review. Res. Agric. Vet. Sci. 2020, 4, 5–11.
  37. Rodríguez-González, A.; Zanin, M.; Menasalvas-Ruiz, E. Public health and epidemiology informatics: Can artificial intelligence help future global challenges? An overview of antimicrobial resistance and impact of climate change in disease epidemiology. Yearb. Med. Inform. 2019, 28, 224–231.
  38. Feretzakis, G.; Sakagianni, A.; Loupelis, E.; Kalles, D.; Skarmoutsou, N.; Martsoukou, M.; Christopoulos, C.; Lada, M.; Petropoulou, S.; Velentza, A. Machine Learning for Antibiotic Resistance Prediction: A Prototype Using Off-the-Shelf Techniques and Entry-Level Data to Guide Empiric Antimicrobial Therapy. Healthc. Inform. Res. 2021, 27, 214–221.
  39. Cartelle Gestal, M.; Dedloff, M.R.; Torres-Sangiao, E. Computational health engineering applied to model infectious diseases and antimicrobial resistance spread. Appl. Sci. 2019, 9, 2486.
  40. Anahtar, M.N.; Yang, J.H.; Kanjilal, S. Applications of machine learning to the problem of antimicrobial resistance: An emerging model for translational research. J. Clin. Microbiol. 2021, 59, e01260-20.
  41. Agnello, S.; Brand, M.; Chellat, M.F.; Gazzola, S.; Riedl, R. A structural view on medicinal chemistry strategies against drug resistance. Angew. Chem. Int. Ed. 2019, 58, 3300–3345.
More
ScholarVision Creations