リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Studies on the antibacterial activity of a novel fluoroquinolone, OPS-2071, against enteropathogenic bacteria」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Studies on the antibacterial activity of a novel fluoroquinolone, OPS-2071, against enteropathogenic bacteria

岡, 大輔 北海道大学

2022.12.26

概要

Intestinal infections are diseases caused by the growth of pathogens in the intestines, causing diarrhea and other symptoms and most frequently resulting from consuming contaminated food or water. Although bacteria, viruses, and protozoa are the causative pathogens, this thesis focuses only on bacteria as the target pathogens. Intestinal infections are common in developing countries where inadequate sanitation is widespread. It is one of the most serious diseases, with 1.7 million deaths reported annually worldwide, especially in children and the elderly over 70 years of age 70) . The number of patients who have died from intestinal infections is still too high, even though economic development has significantly reduced it over the past decades by improving access to treatment (Figure 1) 2) .

Drug-resistant bacteria are becoming a therapeutic threat in intestinal infections. Drugresistant bacteria, such as typhoidal/non-typhoidal Salmonella, Shigella spp, Campylobacter spp, Vibrio spp, and diarrheal Escherichia coli, have been reported as a problem in their treatment 40, 62, 70) . The spread of Salmonella with ESBL (extended spectrum beta-lactamase), azithromycinresistant Shigella, and ciprofloxacin resistance among a wide range of enteric infectious bacteria has reduced the effectiveness of antimicrobial agents, resulting in a depletion of therapeutic agents 70) . Antibiotic resistance is associated with the widespread use and misuse of antibiotics in humans and agriculture. In developing countries, in addition to the widespread of infectious diseases, the abuse and misuse of antibiotics through the purchase without prescriptions, and the lack of restrictions on antibiotic use in agriculture further contribute to the emergence of resistant bacteria13, 49) .

Enteric infections are also an important public health problem in developed countries; the Centers for Disease Control and Prevention (CDC) reported on the threat of antibiotic resistance in the United States in 2019, listing 18 antimicrobial-resistant bacteria and fungi including intestinal infectious bacteria, such as Clostridioides difficile, Campylobacter, typhoidal/nontyphoidal Salmonella, and Shigella 9) . Resistance to therapeutic agents against these organisms is rapidly increasing, and if this trend continues, treatment options may be lost for patients who need treatment. For instance, Campylobacter spp, one of the most common intestinal infections in the developed world, is estimated at 1.5 million cases per year in the U.S. and an annual medical cost of $2.7 million. Ciprofloxacin and azithromycin are used for treatment, but their drug resistance is increasing every year. Ciprofloxacin-resistant strains have increased to 28% as of 2017 9) .

C. difficile is also one of the important pathogens that cause infections in the intestinal tract. C. difficile is a Gram-positive anaerobic rod that is resistant to many clinically used antimicrobial agents and is known to cause antimicrobial-associated diarrhea caused by disruption of the intestinal flora due to antimicrobial treatment, which leads to microbial substitution, followed by an increase in C. difficile (Figure 2) 12) . C. difficile is not becoming increasingly resistant to therapeutic agents, but in addition to being inherently resistant to almost all clinically used antimicrobials, it has an estimated 12,800 deaths and 2,230,900 hospitalizations in the U.S. in 2017 and $1 billion in annual health care costs, making it a major problem 9) . The risk factors include antimicrobial therapy, prolonged hospitalization, use of immunosuppressants or proton pump inhibitors, and being over 65 years of age; C. difficile is known as one of the most important nosocomial organisms. Vancomycin, metronidazole, and fidaxomicin are therapeutic drugs for C. difficile Infection (CDI), metronidazole, however, is considered less effective than the other two drugs. The therapeutic challenge of CDI is its frequent recurrence after treatment. Although treatment with antimicrobial agents is reported to be highly effective, exceeding 80%, recurrence occurs in 10-25% of patients after treatment, and further recurrence is reported in up to 65% of patients with recurrent disease 21) . C. difficile is a spore-forming bacterium and is thought to recur when bacteria remaining in the intestine as spores re-grow after treatment. Spores are resistant to heat, acid, and antibiotics. CDI is caused by inoculation with the spores, and the growth of C. difficile is normally inhibited by the intestinal flora. When the flora is disrupted by the administration of antimicrobial agents, C. difficile begins to proliferate, causing damage to the intestinal tract by two toxins called enterotoxin A and cytotoxin B, the pathogenic agents of CDI (Figure 2) 12) . The C. difficile BI/NAP1/027 strain is reported as a hypervirulent strain producing C. difficile transferase toxin (CDT; or binary toxin), with strong sporulation ability and increased production of enterotoxin A and cytotoxin B, which are said to be involved in the severe disease. Epidemics of this highly virulent strain have been reported in North America and are considered a new threat to C. difficile. Fidaxomicin has been available as a therapeutic agent since 2011 and showed lower results than vancomycin for relapse (25% vancomycin, 15% fidaxomicin), but this inhibition was not confirmed for the BI/NAP/027 strain. Another treatment option, Bezlotoxumab (a monoclonal antibody that binds to C. difficile cytotoxin B), was approved by the FDA in 2016. It has shown efficacy in reducing relapse, but its use has been limited by its high cost and potential side effects. Fecal microbiota transplantation (FMT) is also being investigated as a new treatment option for CDI. Since disruption of intestinal flora is responsible for the development of CDI, FMT, which transplants normal intestinal flora, is a promising treatment for CDI 36) . In fact, the therapeutic effect in combination with conventional antimicrobial agents has shown the lowest recurrence compared to any other therapy and has been reported as a promising treatment, but the therapeutic process has not yet been established, and more time is needed before it can be widely offered as a treatment. These facts have increased the need for further therapeutic agents for intestinal infections.

An important aspect in the development of a therapeutic drug is not only its therapeutic efficacy but also considerations regarding the risk of the emergence of bacterial resistance. In the long history of antibiotic use, the emergence of resistant bacteria seems inevitable. However, the emergence of resistant bacteria must be prevented as much as possible by their proper use. Pharmacokinetic/pharmacodynamic (PK/PD) modeling is an important concept for its proper use (Figure 3) 4) . The PD parameter is sometimes the minimum inhibitory concentration (MIC), an indicator of antimicrobial activity, particularly MIC90, which is the MIC value widely evaluated in clinical isolates, or breakpoint, which is considered the clinically treatable MIC. However, these are only concentrations at which antimicrobial activity is observed, and they do not take into account whether they prevent the emergence of resistant bacteria. In contrast, mutant prevention concentration (MPC) is a parameter for inhibiting the emergence of resistant bacteria and is defined as the concentration that prevents the emergence of resistant bacteria. On the other hand, concentrations below the MPC but above the MIC are defined as a mutant selection window (MSW) because they inhibit the growth of susceptible bacteria but not of resistant bacteria, and thus have a high risk of selectively increasing the number of resistant bacteria, and treatment should avoid this MSW as much as possible 8) . Unfortunately, MPC is not currently widely used as an indicator of PD, as standardized test methods such as the Clinical and Laboratory Standards Institute (CLSI) method, like MIC, have not yet been defined and have not been widely evaluated through clinical isolates. The variability among strains has not been adequately studied. Nevertheless, the emergence of resistant bacteria is a major challenge common to the entire world, and this concept will become even more important in the future for the long use of antimicrobial agents.

OPS-2071 is a novel quinolone compound synthesized by Otsuka Pharmaceutical Co., Ltd. that targets intestinal infections (Figure 5) 54) . Quinolone antibiotic is one of the most important classes of antibiotics due to their wide spectrum and potent antimicrobial activity, and their favorable pharmacokinetics 20) . In particular, ciprofloxacin, a second-generation quinolone antibacterial agent, was launched in the late 1980s and has still been widely used to date including for intestinal infections. Despite reports of bacterial resistance, it remains a therapeutically important antimicrobial agent, listed by the WHO as an essential drug and an extremely important antibiotic, and one of the most commonly prescribed drugs in the world 79) . Although ciprofloxacin is also recommended as a treatment for intestinal infections, its longstanding use has led to increasing reports of quinolone-resistant strains, particularly in developing countries.

Quinolones target DNA gyrase and topoisomerase, which are essential for bacteria, and are known to work bactericidal by inhibiting these enzymes. There are two major mechanisms of quinolone resistance: one is direct mutations of the target molecule that reduces the binding affinity of the quinolone antimicrobial agent, and the other is mutations to induce inhibition of the uptake of quinolone antimicrobial agents into the bacteria or an accelerated efflux of the drug out of the bacteria. The combination of these two mechanisms is known to induce highly resistant bacteria, but the resistance mechanisms that induce inhibition of drug uptake or promotion of drug efflux do not induce highly resistant bacteria and do not lead to clinically significant resistance 20, 38) . In contrast, direct mutations to target molecules are known to induce high levels of resistance on their own. The region where resistance mutations are introduced is called the quinolone resistance determination region (QRDR), and similar mutations have been reported in many strains of different species 20) . Evaluation against these resistance mutations is useful in terms of predicting antibacterial activity against resistant strains and differentiating them from existing quinolone antimicrobials.

In my study, the potential use of OPS-2071 for various intestinal infections as a therapeutic drug was evaluated. Various aspects of the drug need to be tested in order to evaluate its potential as a therapeutic agent. In vitro antibacterial activity, in vivo pharmacokinetics, and in vivo therapeutic efficacy are essential for predicting therapeutic efficacy. In addition to estimating therapeutic efficacy, it is also important to consider the risk of the emergence of resistant strains, since the problem is that antimicrobial therapy may become ineffective due to the acquisition of resistance after an initial strong therapeutic effect. In addition, the evaluation of inhibitory activity against the target molecules is useful information for characterizing the drug.

This thesis is composed of two chapters and a conclusion. In CHAPTER I, to evaluate its potential as an antimicrobial agent against C. difficile infection, the in vitro activity, in vivo PK profile, and in vivo efficacy were examined. In addition, the frequency of spontaneous resistance and mutant prevention concentration was evaluated as the evaluation of the risk of the emergence of drug resistance. CHAPTER II, to see if OPS-2071 is effective against a wide range of intestinal infections, we evaluated the in vitro antibacterial activity and mechanism of action of OPS-2071 against a wide range of enteric infection-causing bacteria, excluding C. difficile, as well as the risk of emergence of drug-resistant strains.

この論文で使われている画像

関連論文

参考文献

1. Andersson MI, MacGowan AP. Development of the quinolones. J Antimicrob Chemother 51, Suppl 1, 1-11, 2003.

2. Anonymous. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 1736-1788, 2018.

3. Asha NJ, Tompkins D, Wilcox MH. Comparative analysis of prevalence, risk factors, and molecular epidemiology of antibiotic-associated diarrhea due to Clostridium difficile, Clostridium perfringens, and Staphylococcus aureus. J Clin Microbiol 44, 2785-2791, 2006.

4. Asín-Prieto E, Rodríguez-Gascón A, Isla A. Applications of the pharmacokinetic/pharmacodynamic (PK/PD) analysis of antimicrobial agents. J Infect Chemother 21, 319-329, 2015.

5. Bauer M, Notermans D, van Benthem BB, Brazier J, Wilcox M, Rupnik M, Monnet D, van Dessel J, Kuijper E. Clostridium difficile infection in Europe: a hospital-based survey. The Lancet 377, 63-73, 2011.

6. Bilverstone TW, Minton NP, Kuehne SA. Phosphorylation and functionality of CdtR in Clostridium. Anaerobe 58, 103-109, 2019.

7. Borenfreund E, Puerner JA. Toxicity determined in vitro by morphological alterations and neutral red absorption. Toxicol Lett 24, 119-124, 1985.

8. Cantón R, Morosini MI. Emergence and spread of antibiotic resistance following exposure to antibiotics. FEMS Microbiol Rev 35, 977-991, 2011.

9. Centers for Disease Control and Prevention. Antibiotic resistance threats in the United States, 2019. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threatsreport-508.pdf. 2019.

10. Centers for Disease Control and Prevention. National Antimicrobial Resistance Monitoring System for Enteric Bacteria (NARMS). https://www.cdc.gov/narms/antibiotics-tested.html. 2019.

11. Changkwanyeun R, Yamaguchi T, Kongsoi S, Changkaew K, Yokoyama K, Kim H, Suthienkul O, Usui M, Tamura Y, Nakajima C, Suzuki Y. Impact of mutations in DNA gyrase genes on quinolone resistance in Campylobacter jejuni. Drug Test Anal 8, 1071- 1076, 2016.

12. Chilton CH, Pickering DS, Freeman J. Microbiologic factors affecting Clostridium difficile recurrence. Clin Microbiol Infect 24, 476-482, 2018.

13. Christaki E, Marcou M, Tofarides A. Antimicrobial Resistance in Bacteria: Mechanisms, Evolution, and Persistence. J Mol Evol 88, 26-40, 2020.

14. Clark CL, Kosowska-Shick K, Ednie LM, Appelbaum PC. Capability of 11 antipneumococcal antibiotics to select for resistance by multistep and single-step methodologies. Antimicrob Agents Chemother 51, 4196-4201, 2007.

15. Clinical and Laboratory Standards Institute. Methods for Antimicrobial dilution and disk susceptibility testing of infrequently isolated or fastidious bacteria; Approved Guideline– Second Edition, 2010.

16. Clinical and Laboratory Standards Institute. Methods for antimicrobial susceptibility testing of anaerobic bacteria; approved standard - Eight Edition, CLSI document M11- A8. Clinical and Laboratory Standards Institute, Wayne, PA, 2012.

17. Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Ninth Edition, 2012.

18. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial disk and dilution susceptibility tests for bacteria isolated from animals; Approved Standard– Third Edition, 2008.

19. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing; twenty-second informational supplement, CLSI document M100- S22, 2012.

20. Correia S, Poeta P, Hébraud M, Capelo JL, Igrejas G. Mechanisms of quinolone action and resistance: where do we stand? J Med Microbiol 66, 551-559, 2017.

21. Czepiel J, Dróżdż M, Pituch H, Kuijper EJ, Perucki W, Mielimonka A, Goldman S, Wultańska D, Garlicki A, Biesiada G. Clostridium difficile infection: review. Eur J Clin Microbiol Infect Dis 38, 1211-1221, 2019.

22. D'Aoust J, Battat R, Bessissow T. Management of inflammatory bowel disease with Clostridium difficile infection. World J Gastroenterol 23, 4986-5003, 2017.

23. DeAlmeida MN, Heffernan H, Dervan A, Bakker S, Freeman JT, Bhally H, Taylor SL, Riley TV, Roberts SA. Severe Clostridium difficile infection in New Zealand associated with an emerging strain, PCR-ribotype 244. N Z Med J 126, 9-14, 2013.

24. Deshpande A, Pasupuleti V, Thota P, Pant C, Rolston DD, Hernandez AV, Donskey CJ, Fraser TG. Risk factors for recurrent Clostridium difficile infection: a systematic review and meta-analysis. Infect Control Hosp Epidemiol 36, 452-460, 2015.

25. Douros A, Grabowski K, Stahlmann R. Safety issues and drug-drug interactions with commonly used quinolones. Expert Opin Drug Metab Toxicol 11, 25-39, 2015.

26. Dridi L, Tankovic J, Burghoffer B, Barbut F, Petit JC. gyrA and gyrB mutations are implicated in cross-resistance to Ciprofloxacin and moxifloxacin in Clostridium difficile. Antimicrob Agents Chemother 46, 3418-3421, 2002.

27. EUCAST. Breakpoint tables for interpretation of MICs and zone diameters, version 12.0, 2022, https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_1 2.0_Breakpoint_Tables.pdf., 2022.

28. Fatima R, Aziz M. The Hypervirulent Strain of Clostridium Difficile: NAP1/B1/027 - A Brief Overview. Cureus 11, e3977, 2019.

29. Fischer S, Uckert AK, Landenberger M, Papatheodorou P, Hoffmann-Richter C, Mittler AK, Ziener U, Hagele M, Schwan C, Muller M, Kleger A, Benz R, Popoff MR, Aktories K, Barth H. Human peptide alpha-defensin-1 interferes with Clostridioides difficile toxins TcdA, TcdB, and CDT. FASEB J 34, 6244-6261, 2020.

30. Fisher LM, Pan XS. Methods to assay inhibitors of DNA gyrase and topoisomerase IV activities. Methods Mol Med 142, 11-23, 2008.

31. Gianvecchio C, Lozano NA, Henderson C, Kalhori P, Bullivant A, Valencia A, Su L, Bello G, Wong M, Cook E, Fuller L, Neal JB, 3rd, Yeh PJ. Variation in Mutant Prevention Concentrations. Front Microbiol 10, 42, 2019.

32. Gil F, Lagos-Moraga S, Calderón-Romero P, Pizarro-Guajardo M, Paredes-Sabja D. Updates on Clostridium difficile spore biology. Anaerobe 45, 3-9, 2017.

33. González-Torralba A, García-Esteban C, Alós JI. Enteropathogens and antibiotics. Enferm Infecc Microbiol Clin 36, 47-54, 2018.

34. Guery B, Galperine T, Barbut F. Clostridioides difficile: diagnosis and treatments. Bmj 366, l4609, 2019.

35. Gupta A, Saha S, Khanna S. Therapies to modulate gut microbiota: Past, present and future. World J Gastroenterol 26, 777-788, 2020.

36. Haber SL, Raney CRK, Larson TL, Lau JP. Fecal microbiota transplantation for recurrent Clostridioides difficile infection. Am J Health Syst Pharm 76, 935-942, 2019.

37. Hooper DC. Mechanisms of action and resistance of older and newer fluoroquinolones. Clin Infect Dis 31 Suppl 2, S24-28, 2000.

38. Hooper DC, Jacoby GA. Mechanisms of drug resistance: quinolone resistance. Ann N Y Acad Sci 1354, 12-31, 2015.

39. Issa M, Vijayapal A, Graham MB, Beaulieu DB, Otterson MF, Lundeen S, Skaros S, Weber LR, Komorowski RA, Knox JF, Emmons J, Bajaj JS, Binion DG. Impact of Clostridium difficile on inflammatory bowel disease. Clin Gastroenterol Hepatol 5, 345- 351, 2007.

40. Klemm EJ, Shakoor S, Page AJ, Qamar FN, Judge K, Saeed DK, Wong VK, Dallman TJ, Nair S, Baker S, Shaheen G, Qureshi S, Yousafzai MT, Saleem MK, Hasan Z, Dougan G, Hasan R. Emergence of an Extensively Drug-Resistant Salmonella enterica Serovar Typhi Clone Harboring a Promiscuous Plasmid Encoding Resistance to Fluoroquinolones and Third-Generation Cephalosporins. mBio 9, e00105-00118, 2018.

41. Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, Faruque AS, Zaidi AK, Saha D, Alonso PL, Tamboura B, Sanogo D, Onwuchekwa U, Manna B, Ramamurthy T, Kanungo S, Ochieng JB, Omore R, Oundo JO, Hossain A, Das SK, Ahmed S, Qureshi S, Quadri F, Adegbola RA, Antonio M, Hossain MJ, Akinsola A, Mandomando I, Nhampossa T, Acácio S, Biswas K, O'Reilly CE, Mintz ED, Berkeley LY, Muhsen K, Sommerfelt H, Robins-Browne RM, Levine MM. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-control study. Lancet 382, 209-222, 2013.

42. Kumar M, Mathur T, Barman TK, Ramkumar G, Bhati A, Shukla G, Kalia V, Pandya M, Raj VS, Upadhyay DJ, Vaishnavi C, Chakrabarti A, Das B, Bhatnagar PK. In vitro and in 68 vivo activities of the novel Ketolide RBx 14255 against Clostridium difficile. Antimicrob Agents Chemother 56, 5986-5989, 2012.

43. Leffler DA, Lamont JT. Clostridium difficile Infection. N Engl J Med 373, 287-288, 2015.

44. Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, Farley MM, Holzbauer SM, Meek JI, Phipps EC, Wilson LE, Winston LG, Cohen JA, Limbago BM, Fridkin SK, Gerding DN, McDonald LC. Burden of Clostridium difficile infection in the United States. N Engl J Med 372, 825-834, 2015.

45. Levitus M, Rewane A, Perera TB. Vancomycin-Resistant Enterococci (VRE). StatPearls. Treasure Island (FL): StatPearls Publishing LLC., 2020.

46. Lim SK, Stuart RL, Mackin KE, Carter GP, Kotsanas D, Francis MJ, Easton M, Dimovski K, Elliott B, Riley TV, Hogg G, Paul E, Korman TM, Seemann T, Stinear TP, Lyras D, Jenkin GA. Emergence of a ribotype 244 strain of Clostridium difficile associated with severe disease and related to the epidemic ribotype 027 strain. Clin Infect Dis 58, 1723-1730, 2014.

47. Loo VG, Poirier L, Miller MA, Oughton M, Libman MD, Michaud S, Bourgault AM, Nguyen T, Frenette C, Kelly M, Vibien A, Brassard P, Fenn S, Dewar K, Hudson TJ, Horn R, Rene P, Monczak Y, Dascal A. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 353, 2442-2449, 2005.

48. Lopez Y, Tato M, Gargallo-Viola D, Canton R, Vila J, Zsolt I. Mutant prevention concentration of ozenoxacin for quinolone-susceptible or -resistant Staphylococcus aureus and Staphylococcus epidermidis. PLoS One 14, e0223326, 2019.

49. Manyi-Loh C, Mamphweli S, Meyer E, Okoh A. Antibiotic Use in Agriculture and Its Consequential Resistance in Environmental Sources: Potential Public Health Implications. Molecules 23, 2018.

50. Mathur T, Barman TK, Kumar M, Singh D, Kumar R, Khera MK, Yamada M, Inoue SI, Upadhyay DJ, Masuda N. In Vitro and In Vivo Activities of DS-2969b, a Novel GyrB Inhibitor, against Clostridium difficile. Antimicrob Agents Chemother 62, 2018.

51. McDonald LC, Gerding DN, Johnson S, Bakken JS, Carroll KC, Coffin SE, Dubberke ER, Garey KW, Gould CV, Kelly C, Loo V, ShakleeSammons J, Sandora TJ, Wilcox 69 MH. Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: 2017 Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis 66, 987-994, 2018.

52. McDonald LC, Killgore GE, Thompson A, Owens RC, Jr., Kazakova SV, Sambol SP, Johnson S, Gerding DN. An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med 353, 2433-2441, 2005.

53. Navalkele BD, Chopra T. Bezlotoxumab: an emerging monoclonal antibody therapy for prevention of recurrent Clostridium difficile infection. Biologics 12, 11-21, 2018.

54. Oka D, Yamaya N, Kuno T, Asakawa Y, Shiragiku T, Chen L, Xue J, Mamuti A, Ye F, Sun J, Ohguro K, Miyamoto H, Uematsu Y, Inagaki K, Cheng JF, Matsumoto M. In Vitro and In Vivo Antibacterial Activities of a Novel Quinolone Compound, OPS-2071, against Clostridioides difficile. Antimicrob Agents Chemother 65, e01170-01120, 2021.

55. Parkhill J, Wren BW, Mungall K, Ketley JM, Churcher C, Basham D, Chillingworth T, Davies RM, Feltwell T, Holroyd S, Jagels K, Karlyshev AV, Moule S, Pallen MJ, Penn CW, Quail MA, Rajandream MA, Rutherford KM, van Vliet AH, Whitehead S, Barrell BG. The genome sequence of the food-borne pathogen Campylobacter jejuni reveals hypervariable sequences. Nature 403, 665-668, 2000.

56. Payot S, Bolla JM, Corcoran D, Fanning S, Mégraud F, Zhang Q. Mechanisms of fluoroquinolone and macrolide resistance in Campylobacter spp. Microbes Infect 8, 1967- 1971, 2006.

57. Pepin J, Saheb N, Coulombe MA, Alary ME, Corriveau MP, Authier S, Leblanc M, Rivard G, Bettez M, Primeau V, Nguyen M, Jacob CE, Lanthier L. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis 41, 1254-1260, 2005.

58. Petri WA, Jr., Miller M, Binder HJ, Levine MM, Dillingham R, Guerrant RL. Enteric infections, diarrhea, and their impact on function and development. J Clin Invest 118, 1277-1290, 2008.

59. Pham TDM, Ziora ZM, Blaskovich MAT. Quinolone antibiotics. Medchemcomm 10, 1719-1739, 2019.

60. Pulse M, Weiss W, Kers J, DeFusco A, Park J, Handfield M. Pharmacological, Toxicological, and Dose Range Assessment of OG716, a Novel Lantibiotic for the Treatment of Clostridium difficile -Associated Infection. Antimicrobial Agents and Chemotherapy 63, e01904-01918, 2019.

61. Rodemann JF, Dubberke ER, Reske KA, Seo DH, Stone CD. Incidence of Clostridium difficile infection in inflammatory bowel disease. Clin Gastroenterol Hepatol 5, 339-344, 2007.

62. Rousham EK, Unicomb L, Islam MA. Human, animal and environmental contributors to antibiotic resistance in low-resource settings: integrating behavioural, epidemiological and One Health approaches. Proceedings of the Royal Society B: Biological Sciences 285, 20180332, 2018.

63. Roychoudhury S, Catrenich CE, McIntosh EJ, McKeever HD, Makin KM, Koenigs PM, Ledoussal B. Quinolone resistance in Staphylococci: activities of new nonfluorinated quinolones against molecular targets in whole cells and clinical isolates. Antimicrob Agents Chemother 45, 1115-1120, 2001.

64. Saidel-Odes L, Borer A, Odes S. Clostridium difficile infection in patients with inflammatory bowel disease. Ann Gastroenterol 24, 263-270, 2011.

65. Sato M, Fujii K, Takagi H, Shibuya I, Oka D, Yamaya N, Hagita H, Matsumoto M, Inagaki K. Antibacterial and Immunosuppressive Effects of OPS-2071, a Candidate Therapy for Inflammatory Bowel Disease. Dig Dis Sci 67, 3993-4007, 2022.

66. Sattar A, Thommes P, Payne L, Warn P, Vickers RJ. SMT19969 for Clostridium difficile infection (CDI): in vivo efficacy compared with fidaxomicin and vancomycin in the hamster model of CDI. J Antimicrob Chemother 70, 1757-1762, 2015.

67. Schiaffino F, Colston JM, Paredes-Olortegui M, François R, Pisanic N, Burga R, Peñataro-Yori P, Kosek MN. Antibiotic Resistance of Campylobacter Species in a Pediatric Cohort Study. Antimicrob Agents Chemother 63, e01911-01918, 2019.

68. Science Council of Japan. Guidelines for proper conduct of animal experiments. http://www.scj.go.jp/en/animal/, 2006.

69. Sears P, Crook DW, Louie TJ, Miller MA, Weiss K. Fidaxomicin attains high fecal concentrations with minimal plasma concentrations following oral administration in patients with Clostridium difficile infection. Clin Infect Dis 55 Suppl 2, S116-120, 2012.

70. Shakoor S, Platts-Mills JA, Hasan R. Antibiotic-Resistant Enteric Infections. Infect Dis Clin North Am 33, 1105-1123, 2019.

71. Shane AL, Mody RK, Crump JA, Tarr PI, Steiner TS, Kotloff K, Langley JM, Wanke C, Warren CA, Cheng AC, Cantey J, Pickering LK. 2017 Infectious Diseases Society of America Clinical Practice Guidelines for the Diagnosis and Management of Infectious Diarrhea. Clin Infect Dis 65, 1963-1973, 2017.

72. Singh S, Nautiyal A. Aortic Dissection and Aortic Aneurysms Associated with Fluoroquinolones: A Systematic Review and Meta-Analysis. Am J Med 130, 1449- 1457.e1449, 2017.

73. Sproston EL, Wimalarathna HML, Sheppard SK. Trends in fluoroquinolone resistance in Campylobacter. Microb Genom 4, e000198, 2018.

74. Swanson RN, Hardy DJ, Shipkowitz NL, Hanson CW, Ramer NC, Fernandes PB, Clement JJ. In vitro and in vivo evaluation of tiacumicins B and C against Clostridium difficile. Antimicrobial agents and chemotherapy 35, 1108-1111, 1991.

75. Tariq R, Law CCY, Khanna S, Murthy S, McCurdy JD. The Impact of Clostridium difficile Infection on Mortality in Patients With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. J Clin Gastroenterol 53, 127-133, 2019.

76. Vogelman BS, Craig WA. Postantibiotic effects. J Antimicrob Chemother 15 Suppl A, 37-46, 1985.

77. Walker AS, Eyre D, Wyllie D, Dingle K, Griffiths D, Shine B, Oakley S, O'Connor L, Finney J, Vaughan A, Crook D, Wilcox M, Peto TA, Infectionsin Oxford shire Research D. Relationship between bacterial strain type, host biomarkers, and mortality in Clostridium difficile infection. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America 56, 1589-1600, 2013.

78. Wieczorek K, Osek J. Antimicrobial resistance mechanisms among Campylobacter. Biomed Res Int 2013, 340605, 2013.

79. World Health Organization. Clinically important antimicrobials for human medicine. WHO report. 2018.

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る