Next Article in Journal
Rapid Quantification of SARS-CoV-2 Neutralising Antibodies Using Time-Resolved Fluorescence Immunoassay
Next Article in Special Issue
Prone Positioning: A Safe and Effective Procedure in Pregnant Women Presenting with Severe Acute Respiratory Distress Syndrome
Previous Article in Journal
Exploring Critical Factors Associated with Completion of Childhood Immunisation in the Eastern Province of Saudi Arabia
Previous Article in Special Issue
Preclinical Toxicity and Immunogenicity of a COVID-19 Vaccine (ZF2001) in Cynomolgus Monkeys
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antibiotics Usage and Resistance among Patients with Severe Acute Respiratory Syndrome Coronavirus 2 in the Intensive Care Unit in Makkah, Saudi Arabia

1
Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
2
Molecular Genetics Department, King Faisal Hospital, Ministry of Health, Makkah 21955, Saudi Arabia
3
Diagnostic Microbiology Department, King Faisal Hospital, Ministry of Health, Makkah 21955, Saudi Arabia
4
Medical Genetics Department, King Faisal Hospital, Ministry of Health, Makkah 21955, Saudi Arabia
5
Pharmaceutical Department, King Faisal Hospital, Ministry of Health, Makkah 21955, Saudi Arabia
6
Health Administration, King Faisal Hospital, Ministry of Health, Makkah 21955, Saudi Arabia
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(12), 2148; https://doi.org/10.3390/vaccines10122148
Submission received: 24 October 2022 / Revised: 9 December 2022 / Accepted: 12 December 2022 / Published: 14 December 2022
(This article belongs to the Special Issue Intensive Care during COVID-19 Pandemic)

Abstract

:
Antibiotic resistance is a global health and development threat, especially during the Severe Acute Respiratory Syndrome Coronavirus 2 (COVID-19) pandemic. Therefore, the current study was conducted to describe antibiotic usage and resistance among patients with COVID-19 in the intensive care unit (ICU) in Makkah, Saudi Arabia. In this cross-sectional study, only patients with positive COVID-19 status (42 patients) admitted to the ICU at the King Faisal Hospital were selected using a census sampling method. The susceptibility test of bacteria was carried out according to the standard protocol. The identified strains were tested in-vitro against several antibiotics drugs. Statistical analysis was performed using SPSS version 24. A total of 42 patients were included, with a mean age of 59.35 ± 18 years. Of them, 38.1% were males, and 61.9% were females. 35.7% have blood group O +. For age and blood groups, statistically significant associations were found between males and females, with p-values = 0.037 and 0.031, respectively. A large percentage (42.7%) of the obtained samples contained Klebsiella Pneumoniae; all bacteria were multidrug-resistance bacteria. Furthermore, 76.2% of bacteria were resistant to Ampicillin, 66.7% were resistant to Ciprofloxacin, 64.3% were resistant to Levofloxacin, 57.1% were resistant to Imipenem, and 57.1% were resistant to Moxifloxacin. On the contrary, among the 40 examined antibiotics, the effective antibiotics were Daptomycin, Linezolid, Mupirocin, Synercid, Teicoplanin, Vancomycin, and Nitrofurantoin. Our study demonstrates that antibiotic resistance is highly prevalent among ICU patients with COVID-19 at the King Faisal Hospital. Additionally, all bacteria were multidrug-resistance bacteria. Therefore, this high prevalence should be seriously discussed and urgently considered.

1. Introduction

A challenge to global health and development is antimicrobial resistance (AMR), which includes antibiotic resistance [1]. One of the top 10 worldwide public health hazards to humanity, according to the World Health Organization (WHO), is AMR [2]. AMR mortality is predicted to surpass that of cancer and cardiovascular disease combined by 2050 [3]. Particularly, antibiotic resistance is brought on by drugs that are exclusively effective against specific bacterial parts. Since the medicine is highly selective, a change in these molecules will prevent or negate the drug’s destructive activity, resulting in antibiotic resistance [4]. In addition to changing the enzyme that antibiotics target, bacteria are also capable of using enzymes to change the antibiotic itself and thereby neutralize it [5]. As a result of drug resistance, antibiotics and other antimicrobial medicines become ineffective, and infections become increasingly difficult or impossible to treat [6]. In addition, misuse and overuse of antibiotics are the main drivers in the development of drug-resistant pathogens [7]. For instance, Saudi Arabia is one country where the public’s use of over-the-counter antibiotics without a prescription plays a significant role in how they behave [8]. Findings from a study conducted in Riyadh, Saudi Arabia, showed that antibiotics could readily be obtained without a prescription in 78% of pharmacies [9]. Without efficient antibiotics, even simple surgeries and regular treatments could become high-risk procedures, extending disease duration and, eventually, increasing the chance of premature death [10]. Additionally, antibiotic resistance has a significant financial impact on the economy. The prolonged disease causes death and incapacity, longer hospital admissions, the need for more expensive medications, and financial difficulties for those affected [11].
A recent study found that antibiotic-resistant bacteria cause 5 million indirect deaths and 1.3 million direct deaths annually. The projections were generated in 2019, before the COVID-19 pandemic that caused Severe Acute Respiratory Syndrome (SARS) aggravated the issue [12]. Unfortunately, those who are the most vulnerable to COVID-19 are also the most vulnerable to drug-resistant infections [13]. Although the world is struggling to control the COVID-19 pandemic, the development of AMR outbreaks should be considered [14]. Bacterial co-infection during viral infections is a significant cause of morbidity and mortality. However, the clinical evidence suggests that bacterial co-infection rates for COVID-19 patients are very low, but antibiotic prescribing remains high [15]. It was noted that about 72% of COVID-19 patients were treated with antibiotics even when not clinically indicated, and this heavy use of empiric antibiotics led to a high rate of AMR [16]. Furthermore, AMR is regarded as a significant factor in predicting patient outcomes and overall resource utilization following infections in ICU [17]. Some studies have shown that antibiotics have been administered to the majority of hospitalized COVID-19 patients and 80–100% of COVID-19 patients in the ICU [18,19]. Recently, studies showed a significant increase in AMR resulting from the COVID-19 pandemic [20,21]. ICUs are facing an emergency and spreading antibiotic-resistant bacterial strains all over the world. Moreover, some resistant bacterial strains have few treatment options [22]. Therefore, the current study was conducted to describe the antibiotics usage and resistance among patients with COVID-19 in the intensive care unit (ICU) at the King Faisal Hospital in Makkah, Saudi Arabia in Makkah, Saudi Arabia.

2. Methods

2.1. Study Design, Setting, and Period

This prospective cross-sectional study was conducted between 1 November 2020 and 31 January 2021 at the King Faisal Hospital in Makkah, Saudi Arabia, which includes 300 beds, serving 40 k patients monthly, more than 500,000 patients yearly, and the isolated ICU includes 30 beds.

2.2. Data Collection Procedure

Patients who were hospitalized in the ICU provided samples. Abscess, ascites fluid, blood, pleural fluid, nasal swabs, sputum, urine culture, and wound samples were taken from various sites. All samples were collected by hospital nurses using accepted techniques. Additionally, the Ministry of Health guideline was followed for blood-taking procedures, which calls for washing the skin with 2% chlorhexidine and a 70% isopropyl alcohol applicator for 30 s while employing a back-and-forth scrubbing motion. Additionally, two collections of blood cultures from patients were taken in order to rule out contamination. A reference lab received the materials for molecular identification.

2.3. Sample Size and Sampling

As all eligible patients were located during the study period, the census sampling approach was used to choose the study participants (between 1 November 2020 and 31 January 2021). All patients admitted to the King Faisal Hospital’s ICU during the study period, both genders, non-intubated patients who were spontaneously breathing, patients who were using antibiotics (narrow or broad spectrum) during the study period, patients of all ages with bacterial infections, and patients with a positive COVID-19 test were included in the study. Outpatients, patients with a negative COVID-19 test, and patients with other kinds of bacteria were all disqualified from the study. Each patient also provided one sample for a culture, and every single one of these cultures revealed the presence of germs.

2.4. Isolation and Identification of Pathogens

The ICU samples were handled by the King Faisal Hospital Microbiology Laboratory in accordance with the established protocols for bacterial isolation and identification. Blood culture bottles were incubated with Biomeieux BACT/ALERT. Once the machine indicated growth, the samples were cultured in blood, MacConkey, and chocolate agar. Grown-on blood, MacConkey, or CLED agar were urine sample samples. They spent the next five to seven days incubating. Preliminary identification of certain isolates was made using colony morphology, Gram stain, and typical rapid biochemical assays such as catalase, indole, and oxidase tests. According to the procedures of the hospital where the strain originated, gram-negative bacteria were found using Pos Breakpoint Combo Panel Type 50 in MicroScan (Beckman Coulter Inc., Brea, CA, USA) and gram-positive strains using Pos Breakpoint Combo Panel Type 28 in MicroScan (Beckman Coulter Inc., CA, USA).

2.5. Antibiotics Susceptibility Testing

The susceptibility test was conducted in accordance with the Clinical and Laboratory Standards Institute’s recommendations (CLSI). By utilizing the MicroScan automated microbiology technology, identified bacteria were evaluated in vitro against many types of antibiotic medications (Pos Breakpoint Combo 50 Panel). The following antibiotic agents were examined: Amikacin, Amoxicillin/Clavulanate, Sulbactam, Ampicillin, Aztreonam, Cefazolin, Cefepime, Cefotaxime, Cefoxitin, Ceftazidime, Ciprofloxacin, Cefuroxime, Colistin, Ertapenem, Gentamicin, Imipenem, Levofloxacin, Meropenem, Moxifloxacin, Piperacillin/Tazobactam, Tigecycline, Tobramycin, Trimethoprim/Sulfamethoxazole, Azithromycin, Clindamycin, Daptomycin, Erythromycin, Fosfomycin, Fusidic Acid, Linezolid, Mupirocin, Oxacillin, Penicillin, Rifampin, Synercid, Teicoplanin, Tetracycline, Vancomycin, Nitrofurantoin, and Norfloxacin. Quality control and maintenance were achieved according to the manufacturer’s guidelines.

2.6. Ethical Considerations

The College of Medicine at Umm Al Qura University’s Research and Ethical Committee gave its approval to the study protocol (HAPO-02-K-012-2021-08-713). Additionally, consent was obtained from the King Faisal Hospital. Conscious patients and the representatives of unconscious patients who agreed to take part in the study were required to complete a written informed consent form.

2.7. Statistical Analysis

Data analysis was done using the Statistical Package for Social Science (SPSS) version 24 (IBM Corp, Armonk, NY, USA). For continuous variables, data are expressed as means and standard deviation; for categorical variables, they are expressed as a percentage. The independent sample t-test was used to examine any variations in means. The prevalence of several categorical variables was compared using the chi-square test. A p-value of 0.05 or less was regarded as statistically significant.

3. Results

In the current study, out of 298 patients who were admitted to the ICU during the study period, only 42 patients with positive COVID-19 tests were included in the final analysis. Of them, 16 (38.1%) were males, and 26 (61.9%) were females (Figure 1). The mean age (years) for the study participants was 59.35 ± 18 (66.62 ± 15 for males and 54.88 ± 20 for females). The results revealed that 15 (35.7%), 9 (21.45), 9 (21.4%), 3 (7.1%), 2 (4.8%), 2 (4.8%), and 2 (4.8%) of them have blood group O+, O−, A+, A−, AB+, B+, and B− respectively. A large percentage of 12 (46.2%) female patients have blood group O +, while only 3 (18.8%) males have blood group O+. Concerning medical diagnosis of the patients, the results showed that 2 (4.8%) of the patients have an acute myocardial infarction, 4 (9.5%) have acute pain, 1 (2.4%) have chronic kidney disease, 1 (2.4%) have dyspnea, 4 (9.5%) have a heart attack, 4 (9.5%) have pneumonia, 3 (7.1%) have a stroke, 13 (30.9%) have sepsis, 1 (2.4%) have weakness, 7 (16.7%) have an unknown fever, and 2 (4.8%) have a viral infection.
In addition, the results demonstrated that 23 (54.8%) of the patients were discharged from the hospital after treatments, and 19 (45.25%) passed away. Furthermore, for age and blood groups, statistically significant associations were found between males and females, with p-values = 0.037 and 0.031, respectively (Table 1).
A total of 42 samples were collected from the patients. Of them, 12 (28.6%) were from blood, 10 (23.8%) from sputum, 9 (21.4%) from a urine culture, 6 (14.2%) from wounds, 2 (4.8%) from nasal swabs, 1 (2.4%) from an abscess, 1 (2.4%) from ascites fluid, and 1 (2.4%) from pleural fluid. Concerning the types of bacteria, the findings show that 18 (42.7%) of the samples contain Klebsiella Pneumoniae, 4 (9.5%) Methicillin Resistant Staphylococcus Aureus, 3 (7.1%) Acinetobacter Baumannii Complex/Hemolyticus, 3 (7.1%) Pseudomonas Aeruginosa, 2 (4.8%) Escherichia Coli, 2 (4.8%) Escherichia Coli ESBL, 2 (4.8%) Proteus Mirabilis, 2 (4.8%) Staphylococcus Aureus, 2 (4.8%) Staphylococcus Epidermidis, 2 (4.8%) Staphylococcus Hominis subspecies Hominis, 1 (2.4%) Staphylococcus Hemolyticus, and 1 (2.4%) contains Streptococcus agalactiae. No statistically significant association was found between both genders (Table 2).
Table 3 shows the types of bacteria by the sources of the samples. The results revealed that 18 (42.7%) of the samples contained Klebsiella Pneumoniae and were obtained from sputum n = 6 (60%), urine culture n = 4 (44.5%), blood n = 3 (25.1%), wound n = 2 (33.3%), body fluid n = 1 (100%), ascites fluid n = 1 (100%), and abscess culture n = 1 (100%). 1 (2.4%) of the samples contained Staphylococcus Hemolyticus, and 1 (2.4%) contained Streptococcus agalactiae were obtained from blood and urine cultures. In addition, 12 (28.6%) of the blood samples contain bacteria, 10 (23.8%) of the sputum samples contain bacteria, 9 (21.4%) of the urine cultures contain bacteria, and 6 (14.2%) of the wound cultures contain bacteria.
Moreover, Table 4 shows the resistance pattern to the most commonly used antibiotics by the types of bacteria among the study participants. The findings show that all bacteria in the current study, such as Acinetobacter Baumannii Complex/Hemolyticus, Escherichia Coli, Escherichia Coli ESBL, Klebsiella Pneumoniae, Methicillin Resistant Staphylococcus Aureus, Proteus Mirabilis, Pseudomonas Aeruginosa, Staphylococcus Aureus, Staphylococcus Epidermidis, Staphylococcus Hemolyticus, Staphylococcus Hominis subspecies Hominis, and Streptococcus agalactiae were multidrug-resistance bacteria. In addition, 32 (76.2%) of bacteria were resistant to Ampicillin, 28 (66.7%) were resistant to Ciprofloxacin, 27 (64.3%) were resistant to Levofloxacin, 24 (57.1%) were resistant to Imipenem, and 24 (57.1%) were resistant to Moxifloxacin.
On the contrary, among the 40 examined antibiotics (Amikacin, Amoxicillin/Clavulanate, Sulbactam, Ampicillin, Aztreonam, Cefazolin, Cefepime, Cefotaxime, Cefoxitin, Ceftazidime, Ciprofloxacin, Cefuroxime, Colistin, Ertapenem, Gentamicin, Imipenem, Levofloxacin, Meropenem, Moxifloxacin, Piperacillin/Tazobactam, Tigecycline, Tobramycin, Trimethoprim/Sulfamethoxazole, Azithromycin, Clindamycin, Daptomycin, Erythromycin, Fosfomycin, Fusidic Acid, Linezolid, Mupirocin, Oxacillin, Penicillin, Rifampin, Synercid, Teicoplanin, Tetracycline, Vancomycin, Nitrofurantoin, and Norfloxacin), the effective antibiotics against the included bacteria were Daptomycin, Linezolid, Mupirocin, Synercid, Teicoplanin, Vancomycin, and Nitrofurantoin.

4. Discussion

In this cross-sectional study, the susceptibility test of bacteria was carried out according to the standard protocol, and the identified strains were tested in-vitro against several antibiotics drugs among 42 patients with positive COVID-19 who were selected using a census sampling method and admitted to the ICU at the King Faisal Hospital. The current study included all patients with bacterial infections, all genders, non-intubated spontaneously breathing patients utilizing antibiotics (narrow or broad spectrum) during the study period, all ages, and positive COVID-19 test results who were admitted to the ICU during the study period.
Our study was not intended to investigate whether the interruption in hospital operations during the pandemic crisis or COVID-19, in general, is associated with the probability of MDRB acquisition. MDRB spreads by cross-transmission or environmental factors in the hospital context, with pressure from antimicrobial therapy selection favoring certain individuals [23]. Due to the frequent and complicated caring, which makes it easier for the contamination of health care personnel’s hands and, as a result, the spread of MDRB [23], cross-transmission occurs in the ICU environment in between 23% and 53% of patient encounters [24,25]. Key elements in preventing the spread of MDRB are the implementation of infection prevention and control measures and the supervision of their observance [26].
Since many earlier pandemics occurred before the period of antibiotic resistance, no information was available prior to the SARS-CoV-2 pandemic, including information on the spread of MDRB during the H1N1 pandemic [27]. Concerns concerning the spread of MDRB during the COVID-19 pandemic have been expressed by experts [28,29,30], and a report suggests a rise in bloodstream infection [31].
Our study showed that a large percentage (42.7%) of the obtained samples contained Klebsiella Pneumoniae, and all bacteria were multidrug-resistance bacteria highlighting the urgent need for the development of newer and more robust antimicrobial agents [32,33]. Additionally, 76.2% of bacteria were resistant to Ampicillin, 66.7% to Ciprofloxacin, 64.3% to Levofloxacin, 57.1% to Imipenem, and 57.1% to Moxifloxacin. The large rate of MDRB acquisition in COVID-19 patients is alarming, and this is in line with our concerns. Furthermore, it is important to remember that the following two elements ought to have lowered the MDRB acquisition rate: Patients with COVID-19 were admitted to ICUs that had been totally cleaned out and decontaminated, in contrast to normal ICU admission, which occurs in units where MDRB carriers are already present. A further effective defense against MDRB cross-transmission should have been the physical separation of COVID-19 patients.
The high rate of MDRB acquisition may be explained by a number of pandemic-related factors, including a lack of PPE [34], the ICU staff being overworked, the overcrowding of the ICU, and the reinforcement of less experienced staff, which reduced adherence to infection prevention and control measures [28,29,30,35]. The prevalence of MDRB was assessed in 2020 compared to the years 2017–2019, according to Aurilio C. et al. (2021); the prevalence of overall MDRB infection was 45.2% in 2017, 44.2% in 2018, 41.4% in 2019, 19.2% in 2020 in non–COVID–19 wards, and 29.3% in COVID–19 wards [36]. Although the purpose of our study was not to determine how each of these many systems functioned, we can make the following hypothesis to explain our results. First, there was a shortage of gowns, so we had to use the same gown on multiple patients. It is probable that gloves were not systematically removed at that time due to the difficulty of taking off PPE. This behavior was not observable in our investigation. A new four-bed ICU with two patients per room had to be created as a result of our study having to modify our standard single-room policy due to the spike in patients. A firm conclusion cannot be formed on this topic due to the small number of patients who were worried; however, staying in our four-bed ICU was not linked to MDRB acquisition. Third, we were unable to continue our audit of catheter dressing and hand hygiene practices as prevention and control measures.
A well-known factor linked to the acquisition of MDRBs is an increase in the use of antimicrobials, in addition to infection control measures [37]. Patients with COVID-19 were first thought to have a significant risk of bacterial co-infection and secondary nosocomial infections, similar to patients with other viral illnesses [38]. Additionally, even in cases when there is no bacterial infection, the early COVID-19 symptoms may encourage the start of antibiotic therapy [28]. Despite the possibility of confounding factors, our investigation demonstrated that, consistent with early findings [39], excessive antibiotic use was significantly related to a higher probability of MDRB acquisition in COVID-19 individuals. It is yet unclear if using broad-spectrum classes alone carries this risk or whether using any antibiotics at all.
Finally, throughout the initial wave, we avoided using immunosuppressive medications. The administration of dexamethasone or the other immunosuppressive medications under study may theoretically raise the likelihood of acquiring MDRB even further.
Many investigations were carried out in ICU settings, which quantitatively showed higher rates of AMR than in non-ICU settings. Despite being influenced by two cohort studies, it is not unusual for AMR to be found in substantially higher concentrations in ICU settings. Prior to COVID-19, patients admitted to ICU settings were more likely to contract infections; studies have shown that 20–50% of ICU hospitalizations were for nosocomial infections [40,41,42]. Higher co-infection rates, particularly those of a resistant nature, are not surprising given the COVID-19 pandemic, where patients are a priori given a combination of antimicrobial and immunosuppressive agents. This is especially true in patients who have been mechanically ventilated for extended periods of time. Furthermore, geographical differences that increase the risk of contracting AMR infections, particularly in low- and middle-income countries, high livestock and food product movement, poor clean water and sanitation facilities, as well as a lack of routine surveillance in these areas all contribute to the overall increase in AMR [43].
The misuse of antibiotics in hospitals may also be a contributing factor to antibiotic resistance; it is estimated that 25 to 50 percent of the antimicrobials prescribed in hospitals are unnecessary or inappropriate, directly affecting AMR [41]. Furthermore, the capacity of the pharmaceutical industry to release new antimicrobials onto the market is inferior to the capacity of microorganisms to develop resistance to previously susceptible drugs [42]. Since the start of the COVID-19 pandemic, scientists have cautioned against the risks of antibiotic abuse, despite clinical evidence with prior viral epidemics suggesting concerns of bacterial co-infection [43,44]. Significant antimicrobial use is anticipated at the hospital’s intensive care unit (ICU) due to the seriousness of the diseases treated and the number of interventions given to patients. Antimicrobial monitoring is therefore essential in the event of a pandemic to spot alarming indicators of abuse or overuse.
In some settings, surveillance programs, thorough testing with standardized protocols and reporting, as well as multimodal strategies emphasizing the strict use of antibiotics in conjunction with infection, prevention, and control practices, could improve antimicrobial stewardship, ultimately lowering mortality and morbidity, particularly in COVID-19 patients. Recent research suggests that these multimodal approaches can be very successful in preventing the spread of resistant microbes [44,45].
It has been determined which clinical and sociodemographic factors, such as health care settings, socioeconomic level, prior antibiotic use, and length of hospital stay, increase a patient’s chance of acquiring such co-infections. Particularly in the case of SARS-CoV-2, identifying individuals who are at higher risk of getting MDR or XDR infections beforehand may improve overall prognosis and outcomes. Additionally, some earlier studies [46,47,48,49] advocate the use of natural substances as fresh methods to combat AMR. We could better understand AMR during COVID-19 and in the future with prospective studies that combine well-designed microbiological investigations [50].
Our study showed that 35.7% have blood group O +. Similar findings were reported by Shesha and her colleagues in Saudi Arabia among non-ICU-admitted COVID-19 patients [51].
The main strength of our study was it is the first study that shows the antibiotics usage and resistance among patients with COVID-19 in the ICU at the King Faisal Hospital in Makkah, Saudi Arabia. The main limitation of this study is its cross-sectional design, which restricts the applicability of our findings. Additionally, there was no follow-up for the study participants because the study was cross-sectional. Additionally, all of the study isolates were obtained from inpatients, and it was impossible to distinguish between the precise amount of nosocomial and community-acquired bacteria. It is advised that these findings be confirmed by other multicenter trials with high sample sizes conducted over extended time periods.

5. Conclusions

In conclusion, our study demonstrates that antibiotic resistance is highly prevalent among ICU patients with COVID-19 at the King Faisal Hospital. Additionally, all of the obtained bacteria were multidrug-resistance bacteria. Furthermore, among the 40 examined antibiotics, the effective antibiotics were Daptomycin, Linezolid, Mupirocin, Synercid, Teicoplanin, Vancomycin, and Nitrofurantoin. Therefore, this high prevalence should be seriously discussed and urgently considered. Additionally, more creativity and funding are needed for operational research as well as for the development of novel antimicrobial drugs, vaccines, and diagnostic tools, particularly those that target dangerous gram-negative bacteria.

Author Contributions

Methodology, A.K., A.A. (Ahmed Alahmadi) and M.A.; Software, A.A. (Anwar Alsulami); Validation, S.A. and A.A. (Alaa Alkhotani); Formal analysis, I.A.; Investigation, A.K., F.B. and A.A. (Ahmed Alahmadi); Data curation, A.K., F.B., S.A. and M.A.; Writing—original draft, A.K. and F.B.; Writing—review & editing, S.A. and A.A. (Anwar Alsulami); Visualization, A.A. (Alaa Alkhotani) and I.A.; Project administration, A.K.; All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the biomedical Ethics Committee of Umm Al-Qura University (protocol code KKDB070821 and date of approval 22/08/2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy.

Acknowledgments

The author would like to thank the King Faisal Hospital in Makkah, Saudi Arabia, for facilitating the conduction of this study.

Conflicts of Interest

The author reported no potential conflict of interest.

References

  1. White, A.; Hughes, J.M. Critical importance of a one health approach to antimicrobial resistance. EcoHealth 2019, 16, 404–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. PLOS Medicine Editors. Call for Papers: PLOS Medicine Special Issue on Bacterial Antimicrobial Resistance—Surveillance and Prevention. PLoS Med. 2022, 19, e1004014. [Google Scholar]
  3. Subramanya, S.H.; Czyż, D.M.; Acharya, K.P.; Humphreys, H. The potential impact of the COVID-19 pandemic on antimicrobial resistance and antibiotic stewardship. Virusdisease 2021, 32, 330–337. [Google Scholar] [CrossRef] [PubMed]
  4. Hobson, C.; Chan, A.N.; Wright, G.D. The antibiotic resistome: A guide for the discovery of natural products as antimicrobial agents. Chem. Rev. 2021, 121, 3464–3494. [Google Scholar] [CrossRef] [PubMed]
  5. Munita, J.M.; Arias, C.A. Mechanisms of antibiotic resistance. Microbiol. Spectr. 2016, 4, 4-2. [Google Scholar] [CrossRef] [Green Version]
  6. Hughes, G.; Webber, M.A. Novel approaches to the treatment of bacterial biofilm infections. Br. J. Pharmacol. 2017, 174, 2237–2246. [Google Scholar] [CrossRef]
  7. Taleb, M.H.; Abou Elkhair, E.; Abed Timraz, R.; Bilbeisi, E.; Hassan, A.H. Prevalence of Antibiotics Resistance among Patients Undergoing Bronchoscopy in Chest Department at Al-Shifa medical complex in Gaza Strip, Palestine. Bull. Pharm. Sci. Assiut 2022, 45, 811–822. [Google Scholar] [CrossRef]
  8. Khojah, H.M. Over-the-counter sale of antibiotics during COVID-19 outbreak by community pharmacies in Saudi Arabia: A simulated client study. BMC Health Serv. Res. 2022, 22, 123. [Google Scholar] [CrossRef]
  9. Alhomoud, F.; Aljamea, Z.; Basalelah, L. “Antibiotics kill things very quickly”—Consumers’ perspectives on non-prescribed antibiotic use in Saudi Arabia. BMC Public Health 2018, 18, 1177. [Google Scholar] [CrossRef]
  10. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Aguilar, G.R.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  11. Gajdács, M.; Urbán, E.; Stájer, A.; Baráth, Z. Antimicrobial resistance in the context of the sustainable development goals: A brief review. Eur. J. Investig. Health Psychol. Educ. 2021, 11, 71–82. [Google Scholar] [CrossRef]
  12. Geta, K. Factors, impacts and possible solutions of antibiotic resistance. World Sci. News 2019, 138, 225–247. [Google Scholar]
  13. Meawed, T.E.; Ahmed, S.M.; Mowafy, S.M.; Samir, G.M.; Anis, R.H. Bacterial and fungal ventilator associated pneumonia in critically ill COVID-19 patients during the second wave. J. Infect. Public Health 2021, 14, 1375–1380. [Google Scholar] [CrossRef]
  14. Ansari, S.; Hays, J.P.; Kemp, A.; Okechukwu, R.; Murugaiyan, J.; Ekwanzala, M.D.; Ruiz Alvarez, M.J.; Paul-Satyaseela, M.; Iwu, C.D.; Balleste-Delpierre, C. The potential impact of the COVID-19 pandemic on global antimicrobial and biocide resistance: An AMR Insights global perspective. JAC Antimicrob. Resist. 2021, 3, dlab038. [Google Scholar] [CrossRef]
  15. Wang, L.; Amin, A.K.; Khanna, P.; Aali, A.; McGregor, A.; Bassett, P.; Gopal Rao, G. An observational cohort study of bacterial co-infectioncoinfection and implications for empirical antibiotic therapy in patients presenting with COVID-19 to hospitals in North West London. J. Antimicrob. Chemother. 2021, 76, 796–803. [Google Scholar] [CrossRef]
  16. Ghosh, S.; Bornman, C.; Zafer, M.M. Antimicrobial Resistance Threats in the emerging COVID-19 pandemic: Where do we stand? J. Infect. Public Health 2021, 14, 555–560. [Google Scholar] [CrossRef]
  17. Alhazzani, W.; Al-Suwaidan, F.A.; Al Aseri, Z.A.; Al Mutair, A.; Alghamdi, G.; Rabaan, A.A.; Algamdi, M.; Alohali, A.F.; Asiri, A.Y.; Alshahrani, M.S. The Saudi critical care society clinical practice guidelines on the management of COVID-19 patients in the intensive care unit. Saudi Crit. Care J. 2020, 4, 27. [Google Scholar] [CrossRef]
  18. Khamis, F.; Al-Zakwani, I.; Al Naamani, H.; Al Lawati, S.; Pandak, N.; Omar, M.B.; Al Bahrani, M.; Bulushi, Z.A.; Al Khalili, H.; Al Salmi, I. Clinical characteristics and outcomes of the first 63 adult patients hospitalized with COVID-19: An experience from Oman. J. Infect. Public Health 2020, 13, 906–913. [Google Scholar] [CrossRef]
  19. Bahçe, Y.G.; Acer, Ö.; Özüdoğru, O. Evaluation of bacterial agents isolated from endotracheal aspirate cultures of COVID-19 general intensive care patients and their antibiotic resistance profiles compared to pre-pandemic conditions. Microb. Pathog. 2022, 164, 105409. [Google Scholar] [CrossRef]
  20. Founou, R.C.; Blocker, A.J.; Noubom, M.; Tsayem, C.; Choukem, S.P.; Dongen, M.V.; Founou, L.L. The COVID-19 pandemic: A threat to antimicrobial resistance containment. Future Sci. OA 2021, 7, FSO736. [Google Scholar] [CrossRef]
  21. Lai, C.C.; Chen, S.Y.; Ko, W.C.; Hsueh, P.R. Increased antimicrobial resistance during the COVID-19 pandemic. Int. J. Antimicrob. Agents 2021, 57, 106324. [Google Scholar] [CrossRef] [PubMed]
  22. Fernández, J.; Bert, F.; Nicolas-Chanoine, M.-H. The challenges of multi-drug-resistance in hepatology. J. Hepatol. 2016, 65, 1043–1054. [Google Scholar] [CrossRef] [PubMed]
  23. Kernéis, S.; Lucet, J.-C. Controlling the diffusion of multidrug-resistant organisms in intensive care units. Proc. Semin. Respir. Crit. Care Med. 2019, 40, 558–568. [Google Scholar] [CrossRef] [PubMed]
  24. Grundmann, H.; Hahn, A.; Ehrenstein, B.; Geiger, K.; Just, H.; Daschner, F.D. Detection of cross-transmission of multiresistant Gram-negative bacilli and Staphylococcus aureus in adult intensive care units by routine typing of clinical isolates. Clin. Microbiol. Infect. 1999, 5, 355–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Chetchotisakd, P.; Phelps, C.L.; Hartstein, A.I. Assessment of bacterial cross-transmission as a cause of infections in patients in intensive care units. Clin. Infect. Dis. 1994, 18, 929–937. [Google Scholar] [CrossRef]
  26. Boyce, J.M.; Pittet, D. Guideline for hand hygiene in health-care settings: Recommendations of the Healthcare Infection Control Practices Advisory Committee and the HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force. Infect. Control. Hosp. Epidemiol. 2002, 23, S3–S40. [Google Scholar] [CrossRef] [Green Version]
  27. Osterholm, M.T. Preparing for the next pandemic. In Global Health; Routledge: London, UK, 2017; pp. 225–238. [Google Scholar]
  28. Nieuwlaat, R.; Mbuagbaw, L.; Mertz, D.; Burrows, L.L.; Bowdish, D.M.; Moja, L.; Wright, G.D.; Schünemann, H.J. Coronavirus disease 2019 and antimicrobial resistance: Parallel and interacting health emergencies. Clin. Infect. Dis. 2021, 72, 1657–1659. [Google Scholar] [CrossRef]
  29. Clancy, C.J.; Nguyen, M.H. Coronavirus disease 2019, superinfections, and antimicrobial development: What can we expect? Clin. Infect. Dis. 2020, 71, 2736–2743. [Google Scholar] [CrossRef]
  30. Rawson, T.M.; Moore, L.S.; Castro-Sanchez, E.; Charani, E.; Davies, F.; Satta, G.; Ellington, M.J.; Holmes, A.H. COVID-19 and the potential long-term impact on antimicrobial resistance. J. Antimicrob. Chemother. 2020, 75, 1681–1684. [Google Scholar] [CrossRef]
  31. Sturdy, A.; Basarab, M.; Cotter, M.; Hager, K.; Shakespeare, D.; Shah, N.; Randall, P.; Spray, D.; Arnold, A. Severe COVID-19 and healthcare-associated infections on the ICU: Time to remember the basics? J. Hosp. Infect. 2020, 105, 593–595. [Google Scholar] [CrossRef]
  32. Contou, D.; Claudinon, A.; Pajot, O.; Micaëlo, M.; Longuet Flandre, P.; Dubert, M.; Cally, R.; Logre, E.; Fraissé, M.; Mentec, H. Bacterial and viral co-infectionscoinfections in patients with severe SARS-CoV-2 pneumonia admitted to a French ICU. Ann. Intensive Care 2020, 10, 119. [Google Scholar] [CrossRef]
  33. Van Duin, D.; Barlow, G.; Nathwani, D. The impact of the COVID-19 pandemic on antimicrobial resistance: A debate. JAC Antimicrob. Resist. 2020, 2, dlaa053. [Google Scholar] [CrossRef]
  34. Ranney, M.L.; Griffeth, V.; Jha, A.K. Critical supply shortages—The need for ventilators and personal protective equipment during the COVID-19 pandemic. N. Engl. J. Med. 2020, 382, e41. [Google Scholar] [CrossRef]
  35. Murray, A.K. The novel coronavirus COVID-19 outbreak: Global implications for antimicrobial resistance. Front. Microbiol. 2020, 11, 1020. [Google Scholar] [CrossRef]
  36. Aurilio, C.; Sansone, P.; Paladini, A.; Barbarisi, M.; Coppolino, F.; Pota, V.; Pace, M.C. Multidrug resistence prevalence in COVID Area. Life 2021, 11, 601. [Google Scholar] [CrossRef]
  37. Chatzopoulou, M.; Reynolds, L. Role of antimicrobial restrictions in bacterial resistance control: A systematic literature review. J. Hosp. Infect. 2020, 104, 125–136. [Google Scholar] [CrossRef] [Green Version]
  38. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  39. Rawson, T.M.; Moore, L.S.; Zhu, N.; Ranganathan, N.; Skolimowska, K.; Gilchrist, M.; Satta, G.; Cooke, G.; Holmes, A. Bacterial and fungal co-infectioncoinfection in individuals with coronavirus: A rapid review to support COVID-19 antimicrobial prescribing. Clin. Infect. Dis. 2020, 71, 2459–2468. [Google Scholar]
  40. Ramadan, H.K.-A.; Mahmoud, M.A.; Aburahma, M.Z.; Elkhawaga, A.A.; El-Mokhtar, M.A.; Sayed, I.M.; Hosni, A.; Hassany, S.M.; Medhat, M.A. Predictors of severity and co-infectioncoinfection resistance profile in COVID-19 patients: First report from upper Egypt. Infect. Drug Resist. 2020, 13, 3409. [Google Scholar] [CrossRef]
  41. Vincent, J.-L.; Rello, J.; Marshall, J.; Silva, E.; Anzueto, A.; Martin, C.D.; Moreno, R.; Lipman, J.; Gomersall, C.; Sakr, Y. International study of the prevalence and outcomes of infection in intensive care units. JAMA 2009, 302, 2323–2329. [Google Scholar] [CrossRef] [Green Version]
  42. Vincent, J.-L.; Bihari, D.J.; Suter, P.M.; Bruining, H.A.; White, J.; Nicolas-Chanoin, M.-H.; Wolff, M.; Spencer, R.C.; Hemmer, M. The prevalence of nosocomial infection in intensive care units in Europe: Results of the European Prevalence of Infection in Intensive Care (EPIC) Study. JAMA 1995, 274, 639–644. [Google Scholar] [CrossRef] [PubMed]
  43. Hanberger, H.; Garcia-Rodriguez, J.-A.; Gobernado, M.; Goossens, H.; Nilsson, L.E.; Struelens, M.J. Antibiotic susceptibility among aerobic gram-negative bacilli in intensive care units in 5 European countries. JAMA 1999, 281, 67–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Frost, I.; Van Boeckel, T.P.; Pires, J.; Craig, J.; Laxminarayan, R. Global geographic trends in antimicrobial resistance: The role of international travel. J. Travel Med. 2019, 26, taz036. [Google Scholar] [CrossRef] [PubMed]
  45. Al-Maani, A.; Al Wahaibi, A.; Al-Zadjali, N.; Al-Sooti, J.; AlHinai, M.; Al Badawi, A.; Al Saidi, A.; AlZadjali, N.; Elshoubary, W.; Al-Harthi, K. The impact of the hand hygiene role model project on improving health-care workers’ compliance: A quasi-experimental observational study. J. Infect. Public Health 2022, 15, 324–330. [Google Scholar] [CrossRef] [PubMed]
  46. Surachat, K.; Deachamag, P.; Kantachote, D.; Wonglapsuwan, M.; Jeenkeawpiam, K.; Chukamnerd, A. In silico comparative genomics analysis of Lactiplantibacillus plantarum DW12, a potential gamma-aminobutyric acid (GABA)-producing strain. Microbiol. Res. 2021, 251, 126833. [Google Scholar] [CrossRef]
  47. Belguesmia, Y.; Spano, G.; Drider, D. Potentiating effects of leaderless enterocin DD14 in combination with methicillin on clinical methicillin-resistant Staphylococcus aureus S1 strain. Microbiol. Res. 2021, 252, 126864. [Google Scholar] [CrossRef]
  48. Hei, Y.; Zhang, H.; Tan, N.; Zhou, Y.; Wei, X.; Hu, C.; Liu, Y.; Wang, L.; Qi, J.; Gao, J.M. Antimicrobial activity and biosynthetic potential of cultivable actinomycetes associated with Lichen symbiosis from Qinghai-Tibet Plateau. Microbiol. Res. 2021, 244, 126652. [Google Scholar] [CrossRef]
  49. Huang, S.; Wang, S.; Li, Y.; Fang, M.; Kou, Z.; Chen, B.; Xu, L.; Bi, Z.; Xu, H.; Chi, X.; et al. Prevalence and transmission of mobilized colistin resistance (mcr-1) gene positive Escherichia coli in healthy rural residents in Shandong province, China. Microbiol. Res. 2021, 253, 126881. [Google Scholar] [CrossRef]
  50. Kariyawasam, R.M.; Julien, D.A.; Jelinski, D.C.; Larose, S.L.; Rennert-May, E.; Conly, J.M.; Dingle, T.C.; Chen, J.Z.; Tyrrell, G.J.; Ronksley, P.E. Antimicrobial resistance (AMR) in COVID-19 patients: A systematic review and meta-analysis (November 2019–June 2021). Antimicrob. Resist. Infect. Control. 2022, 11, 45. [Google Scholar] [CrossRef]
  51. Shesha, N.; Melebari, S.; Alghamdi, S.; Refaat, B.; Naffadi, H.; Alquthami, K. Associations of Clinical Factors and Blood Groups with the Severity of COVID-19 Infection in Makkah City, Saudi Arabia. Front. Cell. Infect. Microbiol. 2022, 12, 870096. [Google Scholar] [CrossRef]
Figure 1. Distribution of the study participants according to the results of the COVID-19 test and departments by gender.
Figure 1. Distribution of the study participants according to the results of the COVID-19 test and departments by gender.
Vaccines 10 02148 g001
Table 1. Characteristics and medical history variables of the study participants by gender.
Table 1. Characteristics and medical history variables of the study participants by gender.
VariablesTotal:
n = 42 (%)
Male:
n = 16 (%)
Female:
n = 26 (%)
p
Value
Age (years)
Mean ± SD59.35 ± 1866.62 ± 1554.88 ± 200.037
Blood group
A−2 (4.8)2 (100)0.0 (0.0)0.031
A+9 (21.4)2 (22.2)7 (77.8)
AB+2 (4.8)2 (100)0.0 (0.0)
B−2 (4.8)2 (100)0.0 (0.0)
B+3 (7.1)2 (66.7)1 (33.3)
O−9 (21.4)3 (33.3)6 (66.7)
O+15 (35.7)3 (20.0)12 (80.0)
Diagnosis
Acute myocardial infarction2 (4.8)2 (100)0.0 (0.0)0.085
Acute pain4 (9.5)0.0 (0.0)4 (100)
Chronic kidney disease1 (2.4)0.0 (0.0)1 (100)
Dyspnea1 (2.4)0.0 (0.0)1 (100)
Heart attack4 (9.5)3 (75.0)1 (25.0)
Pneumonia4 (9.5)1 (25.0)3 (75.0)
Stroke3 (7.1)3 (100)0.0 (0.0)
Sepsis13 (30.9)5 (38.5)8 (61.5)
Weakness1 (2.4)0.0 (0.0)1 (100)
Unknown fever7 (16.7)2 (28.6)5 (71.4)
Viral infection2 (4.8)0.0 (0.0)2 (100)
Outcome
Discharge23 (54.8)6 (26.1)17 (73.9)0.074
Passed away19 (45.2)10 (52.6)9 (47.4)
Data are expressed as means ± SD for continuous variables and as a percentage for categorical variables. The differences between means were tested by using the independent sample t-test. The chi-square test was used to examine differences in the prevalence of different categorical variables. A p-value less than 0.05 was considered statistically significant. SD, stander deviation.
Table 2. The source of samples and types of bacteria among the study participants by gender.
Table 2. The source of samples and types of bacteria among the study participants by gender.
VariablesTotal:
n = 42 (%)
Male:
n = 16 (%)
Female:
n = 26 (%)
p Value
Source of samples
Abscess1 (2.4)0.0 (0.0)1 (100)0.581
Ascites fluid1 (2.4)0.0 (0.0)1 (100)
Blood12 (28.6)7 (58.3)5 (41.7)
Pleural fluid1 (2.4)0.0 (0.0)1 (100)
Nasal swabs2 (4.8)1 (50.0)1 (50.0)
Sputum10 (23.8)3 (30.0)7 (70.0)
Urine culture9 (21.4)4 (44.4)5 (55.6)
Wound6 (14.2)1 (16.7)5 (83.3)
Types of bacteria
Acinetobacter Baumannii Complex/Hemolyticus3 (7.1)2 (66.7)1 (33.3)0.277
Escherichia coli2 (4.8)0.0 (0.0)2 (100)
Escherichia coli ESBL2 (4.8)1 (50.0)1 (50.0)
Klebsiella pneumoniae18 (42.7)5 (27.8)13 (72.2)
Methicillin-Resistant Staphylococcus Aureus4 (9.5)2 (50.0)2 (50.0)
Proteus Mirabilis2 (4.8)0.0 (0.0)2 (100)
Pseudomonas Aeruginosa3 (7.1)0.0 (0.0)3 (100)
Staphylococcus Aureus2 (4.8)1 (50.0)1 (50.0)
Staphylococcus Epidermidis2 (4.8)1 (50.0)1 (50.0)
Staphylococcus Hemolyticus1 (2.4)1 (100)0.0 (0.0)
Staphylococcus Hominis subspecies Hominis2 (4.8)2 (100)0.0 (0.0)
Streptococcus agalactiae1 (2.4)1 (100)0.0 (0.0)
In the case of categorical variables, data are presented as a percentage. The prevalence of several categorical variables was compared using the chi-square test. A p-value of 0.05 or less was regarded as statistically significant.
Table 3. Types of bacteria by the sources of the samples.
Table 3. Types of bacteria by the sources of the samples.
Types of BacteriaAbscessAscites
Fluid
BloodFluidNasal
Swap
SputumUrine
Culture
WoundTotal:
n (%)
Acinetobacter Baumannii Complex/Hemolyticus--1 (8.3)--1 (10)1 (11.1)-3 (7.1)
Escherichia coli------2 (22.2)-2 (4.8)
Escherichia coli ESBL--1 (8.3)--1 (10)--2 (4.8)
Klebsiella Pneumoniae1 (100)1 (100)3 (25.1)1 (100)-6 (60)4 (44.5)2 (33.3)18 (42.7)
Methicillin-Resistant Staphylococcus Aureus----2 (100)--2 (33.3)4 (9.5)
Proteus Mirabilis--1 (8.3)---1 (11.1)-2 (4.8)
Pseudomonas Aeruginosa-----2 (20)-1 (16.7)3 (7.1)
Staphylococcus Aureus--1 (8.3)----1 (16.7)2 (4.8)
Staphylococcus Epidermidis--2 (16.7)-----2 (4.8)
Staphylococcus Hemolyticus--1 (8.3)-----1 (2.4)
Staphylococcus Hominis subspecies Hominis--2 (16.7)-----2 (4.8)
Streptococcus agalactiae------1 (11.1)-1 (2.4)
Total:1 (2.4)1 (2.4)12 (28.6)1 (2.4)2 (4.8)10 (23.8)9 (21.4)6 (14.2)42 (100)
Data are expressed as a percentage of categorical variables.
Table 4. Resistance to the most commonly used antibiotics by the types of bacteria among the study participants.
Table 4. Resistance to the most commonly used antibiotics by the types of bacteria among the study participants.
Types of BacteriaAmikacinAmoxicillin/ClavulanateSulbactamAmpicillinAztreonamCefazolinCefepimeCefotaximeCefoxitinCeftazidimeCiprofloxacinCefuroximeColistinErtapenemGentamicinImipenemLevofloxacinMeropenemMoxifloxacinPiperacillin/TazobactamTigecyclineTobramycinTrimethoprim/SulfamethoxazoleAzithromycinClindamycinDaptomycinErythromycinFosfomycinFusidic AcidLinezolidMupirocinOxacillinPenicillinRifampinSynercidTeicoplaninTetracyclineVancomycinNitrofurantoinNorfloxacin
Acinetobacter Baumannii Complex/Hemolyticus: n = 33-33--33-33---3333---33-----------------
Escherichia Coli: n = 2----------------------------------------
Escherichia Coli ESBL: n = 2---2-22--212----1-2---------------------
Klebsiella Pneumoniae: n = 1814131417131141413131514-1314111413151321415-----------------
Methicillin Resistant Staphylococcus Aureus: n = 4-4-4------3----43-3------------44-------
Proteus Mirabilis: n = 2--------------------------------------12
Pseudomonas Aeruginosa: n = 3----1-------1---------------------------
Staphylococcus Aureus: n = 22--2------2----22------21-2-1--22-------
Staphylococcus Epidermidis: n = 222-2------2---222-2-------2----222------
Staphylococcus Hemolyticus: n = 11--1------1---111-1----11-111--11---1---
Staphylococcus Hominis subspecies Hominis: n = 2-1-1------1---111-1----11-1-1--11-------
Streptococcus Agalactiae: n = 1------------------------------------1---
Total: 42 (100%)22 (52.4%)20 (47.6%)17 (40.5%)32 (76.2%)14 (33.3%)3 (7.14%)19 (45.2%)17 (40.5%)13 (30.9%)18 (42.8%)28 (66.7%)16 (38.1%)1 (2.38%)13 (30.9%)21 (50.0%)24 (57.1%)27 (64.3%)16 (38.1%)24 (57.1%)13 (30.9%)2 (4.76%)17 (40.5%)18 (42.8%)4 (9.52%)3 (7.14%)0.0 (0.0%)6 (14.28%)1 (2.38%)3 (7.14%)0.0 (0.0%)0.0 (0.0%)10 (23.8%)10 (23.8%)2 (4.76%)0.0 (0.0%)0.0 (0.0%)2 (4.76%)0.0 (0.0%)1 (2.38%)2 (4.76%)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kabrah, A.; Bahwerth, F.; Alghamdi, S.; Alkhotani, A.; Alahmadi, A.; Alhuzali, M.; Aljerary, I.; Alsulami, A. Antibiotics Usage and Resistance among Patients with Severe Acute Respiratory Syndrome Coronavirus 2 in the Intensive Care Unit in Makkah, Saudi Arabia. Vaccines 2022, 10, 2148. https://doi.org/10.3390/vaccines10122148

AMA Style

Kabrah A, Bahwerth F, Alghamdi S, Alkhotani A, Alahmadi A, Alhuzali M, Aljerary I, Alsulami A. Antibiotics Usage and Resistance among Patients with Severe Acute Respiratory Syndrome Coronavirus 2 in the Intensive Care Unit in Makkah, Saudi Arabia. Vaccines. 2022; 10(12):2148. https://doi.org/10.3390/vaccines10122148

Chicago/Turabian Style

Kabrah, Ahmed, Fayez Bahwerth, Saad Alghamdi, Alaa Alkhotani, Ahmed Alahmadi, Mashari Alhuzali, Ibrahim Aljerary, and Anwar Alsulami. 2022. "Antibiotics Usage and Resistance among Patients with Severe Acute Respiratory Syndrome Coronavirus 2 in the Intensive Care Unit in Makkah, Saudi Arabia" Vaccines 10, no. 12: 2148. https://doi.org/10.3390/vaccines10122148

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop