Group B streptococcus in obstetrics: unsolved problems

Cover Page

Cite item

Abstract

For several decades, among all possible pathogens of neonatal infections, group B streptococcus has been one of the leading positions. Sepsis, meningitis, and pneumonia are among the most common clinical manifestations of neonatal infection associated with group B streptococcus. In this review, our goal was to analyze the literature demonstrating a worldwide approach to the prevention of vertical transmission of group B streptococcus from mother to child. When writing the review, scientific publications of foreign and domestic authors from the PubMed database were studied. The review considers the drugs of choice for intranatal antibiotic prophylaxis, and their pharmacodynamic, and pharmacokinetic features. The analysis details the problem of the growth of resistance of group B streptococcus to antibacterial drugs. The antimicrobial activity of lactoferrin was noted at a minimum inhibitory concentration of 500 μg/ml. The presented review also reflects the protective and therapeutic effects of oral intake of probiotics containing Lactobacillus acidophilus, Lactobacillus salivarius, Lactobacillus rhamnosus GR-1, and Lactobacillus reuteri RC-14 . Based on the analysis, it can be concluded that penicillin G and ampicillin have the most pronounced bactericidal effect against group B streptococcus. At the same time, the most common side effects of β-lactam penicillins include an allergic reaction with the possible development of anaphylactic shock. Given this, the antibiotics of the first-line reserve group include cefazolin, clindamycin, and vancomycin. At the same time, it is important to take into account the decrease in the therapeutic concentration of clindamycin with a change in the alpha-1-acid glycoprotein in the blood of the mother and fetus, the nephrotoxic effect of vancomycin and the cross-a llergic reaction of cefazolin with antibiotics of the penicillin group. A promising direction in solving the problem of group B streptococcus is the development of new strategies for the prevention of perinatal infection of the fetus and newborn based on a more detailed study of the effects of lactoferrin and probiotics.

Full Text

Introduction

Back in the 1970s, group B streptococcus became the leading cause of neonatal morbidity and mortality [1–3] with an intrauterine infection rate of 2–3 cases per 1000 live births [4] and a mortality rate of up to 50 % [3]. Depending on the timing of manifestation, two clinical forms of group B streptococcus (GBS) infection in newborns are distinguished: early (up to 7 days of life) and late (from 7 days to 3 months of life) [4, 5]. Clinical manifestations are more often represented by sepsis, meningitis, and pneumonia, and less often by lymphadenitis, conjunctivitis, and osteomyelitis [6–8].

Pharmacokinetics and pharmacodynamics of intranatal antibiotic prophylaxis

The widespread use of intranatal antibiotic prophylaxis has reduced the rate of early neonatal GBS infection from 1.7 to 0.22 ‰ [9]. At the same time, the resistance of S. agalactiea to antibacterial drugs has increased, which is a global problem in the public health system. In European and American countries, for intranatal antibiotic prophylaxis of vertical transmission of GBS from mother to child, penicillin G is recommended as a first-line drug according to the scheme: 5 million units starting dosage, followed by intravenous administration of 2.5 million units every 4 hours until the end of labor [10].

The effectiveness of the bactericidal properties of penicillin G according to the recommended scheme for the prevention of vertical transmission of GBS from mother to child was demonstrated in a clinical study by S. Scasso et al. (2015). Scientists using high-performance liquid chromatography determined the concentration of penicillin G in cord blood and amniotic fluid.

The results obtained made it possible to plot the concentration-time pharmacokinetic curve (Figure 1) [11]. The authors noted that the maximum inhibitory concentration of penicillin G in cord blood and amniotic fluid was reached after 195 minutes, which amounted to 5.6 μg/ml and 5.2 μg/ml, respectively. When comparing the results of rectovaginal cultures of a woman in labor and the concentration of penicillin G, it was found that 2 hours after the start of intravenous antibiotic prophylaxis, penicillin G inhibits the growth of GBS in 53 %, and after 4 hours the bactericidal effect is achieved in 88 % [11].

Fig. 1. Pharmacokinetic curve «concentration-time» of penicillin G  [11]

At the same time, penicillin G is not used as an antibiotic prophylaxis in childbirth in the Russian Federation. According to the clinical guidelines «Singleton birth, spontaneous delivery in the occipital presentation» dated July 6, 2021, women in labor with identified GBS in the urogenital tract are recommended to receive an initial dose of ampicillin 2000 mg intravenously, then 1000 mg every 4 hours until the end of labor [12]. Ampicillin, unlike penicillin G, has a wider spectrum of antimicrobial activity [13]. The effectiveness of the bactericidal properties of ampicillin was proven in a clinical study by A. Berardi et al (2017) [13]. Scientists using high-performance liquid chromatography evaluated the level of ampicillin concentration in umbilical cord blood depending on the duration of intranatal antibiotic prophylaxis. The achievement of the maximum inhibitory concentration of ampicillin in cord blood was noted 30 minutes after the start of the introduction of the starting dose of ampicillin 2000 mg. It has been proven that the bactericidal effect of the antibiotic persists in the serum of a newborn for 4–5 hours after birth [14]. Penicillin G and ampicillin have high clinical activity and low toxicity [13].

The presence of the β-lactam ring causes a strong bactericidal effect due to disruption of the synthesis of bacterial cell wall components [13]. At the same time, the most common side effects of β-lactam penicillins include an allergic reaction with the possible development of anaphylactic shock [15]. Other manifestations of an allergic reaction are skin peeling, itching, urticaria, rhinitis, conjunctivitis, Quincke’s edema, rarely — fever, arthralgia, eosinophilia, erythematous and maculopapular rash, erythema multiforme exudative, Stevens-J ohnson syndrome [15]. Because of this, when prescribing an antibiotic during childbirth to prevent vertical transmission of GBS from the mother, the child needs a thorough collection of an allergological history and close monitoring of the woman in labor in the first 30 minutes from the start of administration [13].

Prescribing second-line antibiotics to prevent mother-to-child transmission of GBS during childbirth is based on two principles: information about the presence of an allergic reaction to penicillin G/ampicillin and sensitivity of GBS to clindamycin [10]. First-line reserve group antibiotics include cefazolin, clindamycin, and vancomycin [12]. Dosages and frequency of drug administration are unanimously approved throughout the world and do not cause controversy. At the same time, many issues are the subject of discussion.

Cefazolin belongs to the first generation of cephalosporins [13]. It has been proven that cefazolin with a minimum inhibitory concentration (MIC90) of 0.5 µg/ml inhibits the growth of GBS > 90 % [16]. T. Mitchell et al. (2001) 1–2–4–6 hours before planned operative delivery, cefazolin 1.0 g was administered intravenously once and the concentration of cefazolin in the blood plasma of a pregnant woman, in the amniotic fluid and umbilical cord blood at the time of delivery was determined by high-performance liquid chromatography. It was noted that in all samples the concentration of cefazolin exceeded the MIC90, which amounted to 0.96 µg/ml in the blood plasma of a pregnant woman (95 % CI 0.89–1.0), in umbilical cord blood 0.96 µg/ml (95 % CI 0 .89–1.0) and the amniotic fluid 0.9 µg/ml (0.95 % CI 0.77–1.0) [16]. The presented data emphasize the bacteriostatic properties of cefazolin, manifested in the ability to inhibit the growth of GBS even after 6 hours from the moment of administration of the antibiotic. At the same time, there are conflicting data on the pharmacokinetics of cefazolin during pregnancy. In physiologically developing pregnancy, an increase in glomerular filtration rate is noted.

A. Philipson et al. (1987) noted an increase in the clearance of cefazolin by 57 % during pregnancy compared with non-pregnant patients [17].On the contrary, in a clinical study by J. Popovic et al.(2007), no statistically significant differences were found in the data obtained [18]. At the same time, it was shown that 70–95 % of cefazolin is excreted by the kidneys due to glomerular filtration, and renal clearance is directly proportional to the clearance of cefazolin [13], which corresponds to the data of A. Philipson et al. [17]. Limited data on the pharmacokinetics of cefazolin during pregnancy require a more detailed study.

Clindamycin has a bacteriostatic effect [18]. Antibiotic prophylaxis during childbirth can be prescribed according to the scheme: clindamycin 900 mg every 8 hours until the end of labor [10]. At the same time, C.D. Wear et al. argue that the therapeutic concentration of clindamycin in the blood of the fetus can be achieved only with repeated administration of the antibiotic at least 6 hours before birth [20]. Clindamycin binds primarily to alpha-1-acid glycoprotein [21]. Plasma protein binding is concentration dependent and ranges from 60 to 94 % at therapeutic serum concentrations [13]. Changes in the level of alpha-1-acid glycoprotein in the blood of the mother and fetus alter the pharmacokinetics of clindamycin, which may affect the effectiveness of the bacteriostatic action of antibiotic prophylaxis [21].

With GBS resistance to clindamycin or a high risk of developing an allergic reaction to β-lactam antibiotics, women with GBS are prescribed an antibiotic from the group of cyclic glycopeptides — vancomycin [10]. C.N. Onwuchuruba et al. (2014) determined the concentration of vancomycin in maternal and cord blood at various dosing regimens [22]. With intravenous antibiotic prophylaxis in childbirth according to the scheme vancomycin 1.0 g every 12 hours, the therapeutic concentration of vancomycin is set at 32 % in the blood of the woman in labor and 9 % in the umbilical cord blood. At a dosage of vancomycin 15 mg/kg every 12 hours, the therapeutic concentration was observed at 50 % in the blood of the woman in labor and 33 % in the umbilical cord blood. The dosing regimen of vancomycin 20 mg/kg every 8 hours proved to be the most optimal for the prevention of vertical transmission of GBS from mother to child, and the therapeutic concentration of the antibiotic was achieved in more than 80 % of cases [22].

C.V. Towers et al. in solidarity with the conclusions of C.N. Onwuchuruba et al. and also highlighted the efficacy of the proposed intravenous vancomycin regimen for intranatal antibiotic prophylaxis of GBS. In a clinical study by C.V. Towers et al., indicators of the maximum inhibitory concentration in the blood of a woman in labor and cord blood were established, which amounted to 44.4 μg/ml and 27.4 μg/ml [23]. It was noted that the results obtained were above the minimum inhibitory concentration of vancomycin (>1 μg/ml) for suppressing the growth of GBS, which again emphasizes the effectiveness of vancomycin [23].

Knowledge of the pharmacokinetic characteristics of antibiotics in the mother-p lacenta-fetus system plays a key role in the dosing schedule and frequency of antibiotic administration to suppress the growth of GBS. However, it is important to monitor the resistance of GBS to the above antibacterial drugs. If in a clinical study by Y. López et al. (2017) noted 100 % sensitivity of GBS to penicillin, ampicillin, and vancomycin [24], but the clinical work of S. Assefa et al. (2018) published the results of GBS resistance to penicillin, ampicillin, and vancomycin, which amounted to 19.5 %, 14.6 %, and 17 %, respectively [25]. A clinical study by M. Ábrók et al. (2019) in the period from 2012–2018. also highlights the growing resistance of GBS to antibacterial drugs. It was noted that the antibiotic resistance of GBS to erythromycin and clindamycin increased from 29.2 % to 39.7 % and from 30.2 % to

38.7 %, respectively [26]. Several of other works also demonstrate the resistance of GBS to macrolides and lincosamides [27, 28], which emphasizes the importance of determining the sensitivity of GBS to antibacterial drugs with the determination of the minimum inhibitory concentration.

Modern methods of prevention of group B streptococcus

Given the growing resistance of GBS to antibacterial drugs, it is necessary to develop new methods for the prevention of GBS in obstetrics. The effectiveness of antiseptic agents for the prevention of vertical transmission of GBS from mother to child was demonstrated in the clinical work of J.J. Hijona et al. (2018) [29]. The day before a «programmed birth», a woman with GBS colonization was vaginally injected with a tablet with the active ingredient dequalinium chloride 10 mg. It was noted that the number of women colonized with GBS on the day of delivery decreased by 57.21 %, which demonstrates the cost-effectiveness of the use in clinical practice [29]. At the same time, chlorhexidine proved to be an ineffective method for the prevention of GBS in pregnant women [30].

Many scientific studies demonstrate that lactoferrin can be used as a prebiotic for the prevention of neonatal GBS infections. Lactoferrin is a globular glycoprotein of the transferrin family [31]. It is produced in high concentrations in breast milk [32, 33]. It has been proven that lactoferrin at a concentration of 500 μg/ ml has antimicrobial activity and inhibits the growth of GBS [34]. Lactoferrin supplementation in late pregnancy may be a promising tool to improve pregnancy outcomes and neonatal GBS infections.

L. Hanson (2022) recommends taking oral probiotics as a prevention to reducing the growth of GBS in pregnant women. In a clinical study, pregnant women with GBS colonization from the 28th week of pregnancy took 1 capsule of daily by the mouth of the Floragen3 probiotic. It was noted that in 15.3 % of women at 36 weeks gestation, the GBS carrier status changed from positive to negative [35].

M. Ho et al. (2016) also studied the effectiveness of oral probiotics in pregnancy. Pregnant women with diagnosed GBS colonization at 35–37 weeks of gestation before delivery took 2 probiotic capsules daily before bedtime, containing Lactobacillus rhamnosus GR‑1 and Lactobacillus reuteri RC‑14 At the time of admission to the maternity hospital, 42.9 % of women were diagnosed with a negative recto-vaginal smear for GBS [36].

Y. Liu et al. (2020) also noted a decrease in the colonization of pregnant GBS against the background of oral intake of probiotics containing strains of Lactobacillus rhamnosus GR‑1 and Lactobacillus reuteri RC‑14 [37]. No less interesting data were obtained in a clinical study by V. Martin (2019). Pregnant women with GBS colonization from 26–38 weeks daily took a probiotic containing Lactobacillus salivarius by mouth 1 capsule daily. It was established that at 38 weeks of pregnancy, the rectal swab was negative in 72 % of cases, and the vaginal swab was negative in 68 % of cases [38].

Conclusion

To date, intrapartum antibiotic prophylaxis remains the only effective method for preventing mother-to-child transmission of GBS. The rise of antibiotic resistance is a global health problem. A promising direction in solving the problem of GBS is the development of new strategies for the prevention of perinatal infection of the fetus and newborn based on a more detailed study of the effects of lactoferrin and probiotics and the development of new technologies for the treatment of perinatal infections associated with GBS.

×

About the authors

Victor E. Radzinsky

Peoples’ Friendship University of Russia

Email: stetsyuko@list.ru
ORCID iD: 0000-0003-4956-0466
Moscow, Russian Federation

Olga K. Doronina

Peoples’ Friendship University of Russia

Email: stetsyuko@list.ru
ORCID iD: 0000-0002-4288-353X
Moscow, Russian Federation

Anton S. Olenev

Moscow Multidisciplinary Clinical Center «Kommunarka»

Email: stetsyuko@list.ru
ORCID iD: 0000-0001-9632-6731
Moscow, Russian Federation

Olga V. Stetsyuk

Moscow Multidisciplinary Clinical Center «Kommunarka»

Author for correspondence.
Email: stetsyuko@list.ru
ORCID iD: 0000-0001-8600-7112
Moscow, Russian Federation

References

  1. Phares CR, Lynfield R, Farley MM, Mohle-B oetani J, Harrison LH, Petit S, Craig AS, Schaffner W, Zansky SM, Gershman K, Stefonek KR, Albanese BA, Zell ER, Schrag SJ. Epidemiology of invasive group B streptococcal disease in the United States, 1999-2005. JAMA. 2008;299(17):2056-2065. doi.org/10.1001/jama.299.17.2056
  2. Baker CJ, Byington CL, Polin RA. Policy Statement - Recommendations for the prevention of perinatal group B streptococcal disease. Pediatrics. 2011;128(3):611-616. doi.org/10.1542/peds.2011-1466.
  3. Verani JR, McGee L, Schrag SJ. Prevention of perinatal group B streptococcal disease: Revised guidelines from CDC, 2010. MMWR Recomm Rep. 2010;59(10):1-36.
  4. Faro S, Brehm B, Smith F, Mouzoon M, Greisinger A, Wehmanen O, Turrentine MA. Screening for group B streptococcus: a private hospital’s experience. Infect Dis Obstet Gynecol. 2010;451096:1-4. doi: doi.org/10.1155/2010/451096
  5. Maisey HC, Doran KS, Nizet V. Recent advances in understanding the molecular basis of group B Streptococcus virulence. Expert Rev Mol Med. 2008;10:1-27. doi.org/10.1017/S1462399408000811
  6. Asghar, S, Khan, JА, Mahmood MS, Arshad MI. A Crosssectional Study of Group B Streptococcus-Associated Sepsis, Coinfections, and Antibiotic Susceptibility Profile in Neonates in Pakistan. Adv Neonatal Care. 2020;20(4):59-69. doi: 10.1097/anc.0000000000000701
  7. Joachim A, Matee MI, Massawe FA, Lyamuya EF. Maternal and neonatal colonisation of group B streptococcus at Muhimbili National Hospital in Dar es salaam, Tanzania: prevalence, risk factors and antimicrobial resistance. BMC Public Health. 2009;9:437-444. doi.org/10.1186/1471-2458-9-437
  8. Ezeonu I, Agbo M. Incidence and anti-microbial resistance profile of group B Streptococcus infection in pregnant women in Nsukka, Enugu state, Nigeria. Afr J Microbiol Res. 2014;8(1):91-95. doi.org/10.5897/AJMR12.2307
  9. Nanduri SA, Petit S, Smelser C, Apostol M, Alden NB, HarrisonLH, Lynfield R, VagnonePS, BurzlaffK, SpinaNL, Dufort EM, Schaffner W, ThomasAR, FarleyMM, JainJH, Pondo T, McGeeL, Beall BW, SchragSJ. Epidemiology of Invasive Early-O nset and Late- Onset Group B Streptococcal Disease in the United States, 2006 to 2015: Multistate Laboratory and Population- Based Surveillance. JAMA Pediatr. 2019;173(3):224-233. doi: 10.1001/jamapediatrics.2018.4826
  10. ACOG Committee Opinion number 797. Prevention of group B streptococcal early-o nset disease in newborns. Obstet Gynecol. 2020;135(2):51-72. doi: 10.1097/AOG.0000000000003668
  11. Scasso S, Laufer J, Rodriguez G, Alonso JG, Sosa CG. Vaginal group B streptococcus status during intrapartum antibiotic prophylaxis. Int J Gynaecol Obstes. 2015;129(1):9-12. doi.org/10.1016/j.ijgo.2014.10.018
  12. Russian Society of Obstetricians and Gynecologists. Clinical recommendations: Singleton birth, spontaneous delivery in the occipital presentation (normal delivery). Ministry of Health of the Russian Federation. 2021;1-66. (In Russian).
  13. Kukes VG, Sychev DA. Clinical pharmacology: textbook. GEOTAR-Media. 2017;5:1-1024. (In Russian).
  14. Berardi A, Pietrangiolillo Z, Reggiani ML, Bianco V, Gallesi D, Rossi K, FacchinettiF, Ferrari F. Are postnatal ampicillin levels actually related to the duration of intrapartum antibiotic prophylaxis prior to delivery? A pharmacokinetic study in 120 neonates. Arch Dis Child Fetal Neonatal Ed. 2018;103(2):152-156. doi.org/10.1136/archdischild-2016-312546
  15. Shenoy ES, Macy E, Rowe T, Blumenthal KG. Evaluation and management of penicillin allergy: a review. JAMA. 2019;321:188-199. doi.org/10.1001/jama.2018.19283
  16. Mitchell FT, Pearlman MD, Chapman RL, Bhatt-M ehta V, Faix RG. Maternal and transplacental pharmacokinetics of cefazolin. Obstet Gynecol. 2001;98(6):1075-1079. doi: 10.1016/s0029-7844(01)01629-5
  17. Philipson A, Stiernstedt G, Ehrnebo M. Comparison of the pharmacokinetics of cephradine and cefazolin in pregnant and non-pregnant women. Clin Pharmacokinet. 1987;12:136-144. doi. org/10.2165/00003088-198712020-00004
  18. Popovic J, Grujic Z, Sabo A. Influence of pregnancy on ceftriaxone, cefazolin and gentamicin pharmacokinetics in caesarean vs. non pregnant sectioned women. J Clin Pharm Ther. 2007;32:595-602. doi.org/10.1111/j.1365-2710.2007.00864.x
  19. Berg BR, Houseman JL, Steeg ZE, Le Bar WD, Newton DW. Antimicrobial Susceptibilities of Group B Streptococcus Isolates from Prenatal Screening Samples. Journal of Clinical Microbiology. 2014;52(9):3499-3500. doi: 10.1128/jcm.01781-14
  20. Wear CD, Towers CV, Brown MS, Weitz B, Porter S, Wolfe L. Transplacental passage of clindamycin from mother to neonate. J. Perinatol. 2016;36(11):960-961. doi.org/10.1038/jp.2016.122
  21. Muller AE, Mouton JW, Oostvogel PM, Dörr PJ, Voskuyl RA, DeJongh J, Steegers EA, Danhof M. Pharmacokinetics of clindamycin in pregnant women in the peripartum period. Antimicrob Agents Chemother. 2010;54(5):2175-2181. doi.org/10.1128/AAC.01017-09
  22. Onwuchuruba CN, Towers CV, Howard BC, Hennessy MD, Wolfe L, Brown MS. Transplacental passage of vancomycin from mother to neonate. Am J Obstet Gynecol. 2014;210(4):352-354. doi.org/10.1016/j.ajog.2014.01.019
  23. Towers CV, Weitz B. Transplacental passage of vancomycin. The Journal of Maternal- Fetal & Neonatal Medicine. 2017;31(8):1021 doi: 10.1080/14767058.2017.1306049
  24. López Y, Parra E, Cepas V, Sanfeliú I, Juncosa T, Andreu A, XercavinsM, PérezJ, SanzS, VergaraA, Bosch J, SotoSM. Serotype, virulence profile, antimicrobial resistance and macrolide-r esistance determinants in Streptococcus agalactiae isolates in pregnant women and neonates in Catalonia, Spain. Enferm Infecc Microbiol Clin. 2018;36(8):472-477. doi: 10.1016/j.eimc.2017.08.006
  25. Assefa S, Desta K, Lema T. Group B streptococci vaginal colonization and drug susceptibility pattern among pregnant women attending in selected public antenatal care centers in Addis Ababa, Ethiopia. BMC Pregnancy Childbirt. 2018;18(1):135-144. doi: 10.1186/s12884-018-1791-4
  26. Ábrók M, Tigyi P, Kostrzewa M, Burián, Deák J. Evaluation of the Results of Group B Streptococcus Screening by MALDI-TOF MS among Pregnant Women in a Hungarian Hospital. Pathogens. 2020;9(1):1-7. doi: 10.3390/pathogens9010001
  27. Ali MM., Woldeamanuel Y, Asrat D, Fenta DA, Beall B, Schrag S, McGee L. Features of Streptococcus agalactiae strains recovered from pregnant women and newborns attending different hospitals in Ethiopia. BMC Infect Dis. 2020;20(1):848-857. doi: 10.1186/s12879-020-05581-8
  28. Nascimento CS, Santos NFB., Ferreira RC, Taddei CR. Streptococcus agalactiae in pregnant women in Brazil: prevalence, serotypes, and antibiotic resistance. Braz J Microbiol. 2019;50(4):943-952. doi: 10.1007/s42770-019-00129-8
  29. Hijona JJ, Carballo AL, Sánchez MS, Dyachkova N, Expósito JF, Alcázar JL. Vaginal antiseptics reduce the risk of perinatal infection with group B streptococci.J Matern Fetal Neonatal Med. 2019;32(16):2741-2745. doi: 10.1080/14767058.2018.144919
  30. Ohlsson A, Shah VS, Stade BC. Vaginal chlorhexidine during labour to prevent early-onset neonatal group B streptococcal infection. Cochrane Database Syst Rev. 2014;12:1-51. doi.org/10.1002/14651858.CD003520.pub3
  31. Becker KW, Skaar EP. Metal limitation and toxicity at the interface between host and pathogen. FEMS Microbiol Rev. 2014;38:1235-1249. doi.org/10.1111/1574-6976.12087
  32. Manzoni P, Dall’Agnola A, Tomé D, Kaufman DA, Tavella E, Pieretto M, MessinaA, LucaD, Bellaiche M, MoscaA, PiloquetH, Simeoni U, Picaud JC, VecchioAD. Role of Lactoferrin in Neonates and Infants: An Update. Am J Perinatol. 2018;35(6):561-565. doi.org/10.1055/s-0038-1639359
  33. Rai D, Adelman AS, Zhuang W, Rai GP, Boettcher J, Lönnerdal B. Longitudinal changes in lactoferrin concentrations in human milk: a global systematic review. Crit Rev Food Sci Nutr. 2014;54:1539-1547. doi: 10.1080/10408398.2011.642422
  34. Lu J, Francis JD, Guevara MA, Moore RE, Chambers SA, Doster RS, Eastman AJ, RogersLM, NobleKN, Manning SD, Damo SM, Aronoff DM, TownsendSD, Gaddy JA. Antibacterial and Anti-biofilm Activity of the Human Breast Milk Glycoprotein Lactoferrin against Group B Streptococcus. Chembiochem. 2021;22(12):2124-2133. doi: 10.1002/cbic.202100016
  35. Hanson L, Vusse LV, Forqie M, Malloy E, Singh M, Scherer M, Kleber D, Dixon J, Hryckowian AJ, Safdar N. A randomized controlled trial of an oral probiotic to reduce antepartum group B Streptococcus colonization and gastrointestinal symptoms. Am J Obstet Gynecol MFM. 2023;5(1):100748. doi: 10.1016/j.ajogmf.2022.100748
  36. Ho M, Chang YY, Chang WC, Lin HC, Wang MH, Lin WC, Chiu TH. Oral Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 to reduce Group B Streptococcus colonization in pregnant women: A randomized controlled trial. Taiwan J Obstet Gynecol. 2016;55(4):515-518. doi: 10.1016/j.tjog.2016.06.003
  37. Liu Y, Huang Y, Cai W, LiD, Zheng W, Xiao Y, Liu Y, Zhao H, Pan S. Effect of oral Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 on vaginal Group B Streptococcus colonization and vaginal microbiome in late pregnancy. Nan Fang Yi Ke Da Xue Xue Bao. 2020;40(12):1753-1759. doi: 10.12122/j.issn.1673-4254.2020.12.09
  38. Martín V, Cárdenas N, Ocaña S, Marín M, Arroyo R, Beltrán D, BadiolaC, FernándezL, RodríguezJM. Rectal and Vaginal Eradication of Streptococcus agalactiae in Pregnant Women by Using Lactobacillus salivarius CECT 9145, A Target-s pecific Probiotic Strain. Nutrients. 2019;11(4):810-832. doi: 10.3390/nu11040810

Supplementary files

Supplementary Files
Action
1. Fig. 1. Pharmacokinetic curve «concentration-time» of penicillin G

Download (62KB)

Copyright (c) 2023 Radzinsky V.E., Doronina O.K., Olenev A.S., Stetsyuk O.V.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies