Next Article in Journal
Perioperative Coronavirus Disease 2019 Infection and Its Impact on Postoperative Outcomes: Pulmonary Complications and Mortality Based on Korean National Health Insurance Data
Previous Article in Journal
Selective IgM Hypogammaglobulinemia and Multiple Sclerosis Treated with Natalizumab and Ofatumumab: A Case Report
Previous Article in Special Issue
Impact of COVID-19 on Pediatric Inflammatory Bowel Diseases—From Expectations to Reality
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course

1
Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
2
Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
3
Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Voli Square, 1, 46001 Ternopil, Ukraine
4
Department of Biochemistry and Pharmacology, Uzhhorod National University, 88000 Uzhhorod, Ukraine
5
Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2025, 15(4), 156; https://doi.org/10.3390/jpm15040156
Submission received: 10 March 2025 / Revised: 11 April 2025 / Accepted: 12 April 2025 / Published: 17 April 2025
(This article belongs to the Special Issue Personalized Medicine in Post-COVID-19 Era)

Abstract

:
Background: This study investigated the role of genetic polymorphisms in IFNAR2, OAS1, OAS3, and ACE2 as predictors of Paxlovid treatment response, specifically examining their influence on the clinical course and laboratory parameters of COVID-19 patients. Methods: We analyzed the impact of polymorphisms in genes associated with the interferon pathway (IFNAR2 rs2236757), antiviral response (OAS1 rs10774671, OAS3 rs10735079), and viral entry (ACE2 rs2074192) in individuals treated with Paxlovid. Results: Our findings suggest that genetic variations in these genes may modulate the immune response and coagulation pathways in the context of Paxlovid treatment during COVID-19 infection. Specifically, the IFNAR2 rs2236757 G allele was associated with alterations in inflammatory and coagulation markers, while polymorphisms in OAS1 and OAS3 influenced coagulation parameters. Furthermore, specific genotypes were linked to changes in clinical parameters such as oxygen saturation, leukocyte count, and liver function markers in Paxlovid-treated patients. Conclusions: These results highlight the potential of considering genetic factors in understanding individual responses to COVID-19 treatment with Paxlovid and informing future personalized approaches.

1. Introduction

Since the onset of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, a wide range of antiviral therapeutic strategies have been investigated to combat COVID-19 [1,2,3,4].
Paxlovid, a combination therapy comprising nirmatrelvir and ritonavir, has garnered significant attention as a potential treatment option [5,6,7,8]. Nirmatrelvir, a specific inhibitor targeting the SARS-CoV-2 main protease, is synergistically enhanced by ritonavir, a cytochrome P450 3A4 inhibitor [9,10,11,12,13]. Several studies have demonstrated the efficacy of Paxlovid in reducing COVID-19-related mortality and hospitalization rates [14,15,16,17]. In particular, a randomized controlled trial reported an 89% reduction in the risk of hospitalization and death within 28 days when Paxlovid was administered within 3 days of symptom onset [18].
The limited availability of data on adverse events associated with the nirmatrelvir-ritonavir combination hinders a comprehensive understanding of its safety profile [19,20,21,22,23]. Clinical studies have documented dysgeusia, characterized by alterations in taste perception, and diarrhea as common side effects [15,24,25,26,27]. A comprehensive analysis by Li et al. [28] further elucidated the spectrum of adverse events associated with nirmatrelvir-ritonavir administration. The most commonly reported adverse events were mild to moderate in severity and included dysgeusia, diarrhea, nausea, headache, pyrexia, vomiting, and malaise [29,30,31].
Differences in drug absorption, distribution, metabolism, and excretion can lead to varying drug concentrations in different individuals [32,33,34]. For instance, genetic polymorphisms in drug-metabolizing enzymes can affect the rate at which a drug is metabolized, influencing its efficacy and potential side effects [34,35,36,37]. Drugs primarily metabolized by a single enzyme and with a broad therapeutic index may exhibit significant pharmacokinetic variability due to pharmacogenetic variants [38,39,40]. However, given their wide therapeutic window, these genetic differences may not necessarily translate into clinically relevant drug efficacy or toxicity variations [41,42,43,44]. For instance, drug interactions or underlying diseases that inhibit one metabolic pathway and genetic variations impairing a second pathway can contribute to atypical drug responses [45,46,47]. Certain medications, originally prescribed for other conditions, exhibit pleiotropic effects that contribute to their efficacy in the treatment of COVID-19 [48,49,50,51]. These drugs, beyond their primary therapeutic action, engage additional mechanisms that influence the pathogenesis of COVID-19, thereby enhancing clinical outcomes in affected patients. [52,53,54]. Evidence suggests that certain medications, typically used for other conditions, may reduce the severity of COVID-19 outcomes in individuals with metabolic disorders, such as type 2 diabetes, by mitigating complications related to hyperglycemia [55,56,57]. The repurposing of existing drugs for diverse diseases is highly valuable, as it utilizes established pharmacological insights to expedite the discovery of effective treatments [58,59,60,61].
Genetic polymorphisms have been implicated in both the susceptibility to and severity of COVID-19, affecting various biological pathways relevant to the disease [62,63,64,65]. The angiotensin-converting enzyme 2 (ACE2) receptor is the primary cellular receptor for SARS-CoV-2 entry [62,66,67]. The intronic variant rs2074192 has been implicated in changes to the secondary structure of ACE2 mRNA, potentially disrupting the equilibrium between ACE2 transcription and translation [64,68]. This imbalance may consequently affect the binding affinity of SARS-CoV-2 to angiotensin receptors [63,69,70]. Notably, the rs2074192 polymorphism in the ACE2 gene has been implicated in COVID-19 severity, particularly in adult populations [64,71,72].
Furthermore, genetic variations affecting immune response pathways, such as those involving interferon and cytokine signaling, have been associated with differential susceptibility to infection and disease severity [73,74,75,76]. Variations in interferon genes or their receptors have been linked to increased susceptibility or more severe clinical outcomes [65,77,78]. The IFNAR2 rs2236757 variant, in particular, has been strongly associated with increased disease severity [65,79,80]. Furthermore, polymorphisms in antiviral 2′,5′-oligoadenylate synthetase (OAS) enzymes, which are essential for the immune response against SARS-CoV-2, have also been implicated in COVID-19 severity [65,81,82]. SNPs such as rs10774671 in OAS1 [83] and rs10735079 in OAS3 [65] may be associated with more severe clinical outcomes following COVID-19 infection [84,85].
As Paxlovid has become a standard treatment for COVID-19, further research is essential to fully understand its impact on clinical and laboratory parameters [86,87,88]. The presence of specific single nucleotide polymorphisms and their potential effects on treatment outcomes adds to the complexity of managing COVID-19 patients [89,90]. These genetic polymorphisms have been studied as predictors of COVID-19 severity. Considering that these patients are indicated for treatment against COVID-19, investigating the combined effect of these polymorphisms and treatment response is crucial. This study examines their association with treatment and clinical and laboratory parameters for the first time.

2. Materials and Methods

2.1. Sample Collection

This study included 72 adults of European ancestry (Ukrainian ethnicity) aged 23 to 86 years who tested positive for SARS-CoV-2 and were subsequently hospitalized between October 2022 and May 2023. Confirmation of SARS-CoV-2 infection was achieved through real-time polymerase chain reaction (RT-PCR) analysis of nasopharyngeal swab samples. Study participants were enrolled from Ternopil City Community Hospital No. 1. Informed consent was obtained prior to the collection of blood samples, which were subsequently stored at −80 °C for subsequent analysis. All research procedures were conducted in strict adherence to the ethical principles outlined in the Declaration of Helsinki. Prior to the commencement of any study activities, the research protocol (No. 74, dated 13 October 2023) underwent rigorous ethical review and received formal approval from the Ethics Committee of the I. Horbachevsky Ternopil National Medical University.
Participants were included if they had a confirmed COVID-19 diagnosis requiring hospitalization, no history of chronic diseases, and no antibiotic or probiotic use within the past three months.
All study participants adhered to the standard treatment protocol for COVID-19 as outlined by national guidelines. This regimen included symptomatic relief: antipyretic therapy with paracetamol or ibuprofen; respiratory support: mucolytic and expectorant agents, such as Ambroxol, and non-invasive oxygen therapy as needed; thromboembolic prophylaxis: anticoagulant therapy with low-molecular-weight heparins, like enoxaparin, at a dose of 40 mg or 4000 IU anti-Xa; antimicrobial therapy: prophylactic antimicrobial treatment for potential co-infections, consisting of amoxicillin/clavulanate combined with macrolides (azithromycin or clarithromycin) or cephalosporins of the second or third generation; and immunomodulation: corticosteroid therapy with intravenous dexamethasone at a dose of 0.15 mg/kg daily (8–16 mg) for 7–10 days. The use of corticosteroids and antibiotics did not differ between the groups.
Twenty-three out of seventy-three patients received nirmatrelvir–ritonavir (Paxlovid) in accordance with Food and Drug Administration (FDA) recommendations, consisting of an oral dose of 300/100 mg twice daily for 5 days [91].

2.2. Laboratory and Clinical Data

A comprehensive laboratory workup was performed, including assessment of oxygen saturation, complete blood count (CBC) with differential, erythrocyte sedimentation rate (ESR), coagulation profile (platelet count, hematocrit, INR, PT), quick pro-thrombin time (QTP), activated partial thromboplastin time (APTT), fibrinogen, total bilirubin, alanine aminotransferase (ALT), aspartate aminotransferase (AST), serum creatinine, gamma-glutamyl transferase (GGT), total protein, albumin, alkaline phosphatase (ALP), C-reactive protein (CRP), and blood glucose.

2.3. Identifying Genetic Polymorphisms

Venous blood samples were collected, and genomic DNA was extracted using a commercial kit, the Thermo Scientific™ GeneJET™ Whole Blood Genomic DNA Purification Mini Kit Cat. No. K0781. Polymorphisms in the ACE2 rs2074192, IFNAR2 rs2236757, OAS1 rs10774671, and OAS3 rs10735079 genes were analyzed using real-time PCR. PCR amplification and melting curve analysis were performed using TaqMan assays under optimized conditions. The CFX96™ Real-Time PCR Basic Software was utilized for genotyping analysis based on the melting curve.

2.4. Statistical Analysis

Descriptive statistics were utilized to delineate the demographic characteristics and clinical outcomes of the study population. Due to non-normal data distribution, non-parametric tests were employed for comparisons. The Mann–Whitney U test, Kruskal–Wallis test, Dunn’s multiple comparison test, and Wilcoxon matched pairs test were utilized for appropriate comparisons.
Repeated measures ANOVA was employed to investigate the influence of specific factors on a continuous variable. Post hoc comparisons with Bonferroni correction were subsequently conducted to identify significant differences between groups. In instances where the normality assumption was not sufficiently met, data are displayed as medians and interquartile range (IQR).
A two-tailed alpha level of 0.05 was used to establish statistical significance. Analyses were conducted using GraphPad Prism (version 8.4.3) and IBM SPSS Statistics (version 25).

2.5. Power Analysis

To assess the statistical power of our study, we employed the G*power 3.1.9.7 software (https://www.psychologie.hhu.de/arbeitsgruppen/allgemeine-psychologie-und-arbeitspsychologie/gpower(accessed on 2 April 2025). To analyze the impact of genetic variations and Paxlovid treatment on the trajectory of laboratory parameters from hospitalization to discharge, we utilized repeated measures ANOVA. Our study design involved two groups and two measurement points.
Assuming a medium effect size of 0.3, an α-error probability of 0.05, a total sample size of 72, and a correlation among repeated measures of 0.5, our analysis indicates a statistical power of 82%.

3. Results

3.1. Baseline Patient Parameters

Of the 72 enrolled patients, 23 were assigned to the Paxlovid treatment group (52.17% male; median age of 64 years, IQR 46–71) and 49 to the standard treatment group (63.26% male; median age of 66 years, IQR 51–72). No significant differences were observed between the two groups in terms of demographic characteristics, peripheral oxygen saturation, oxygen therapy requirements, COVID-19 severity, or body mass index, as detailed in Table 1. The median time to hospital discharge was shorter in the Paxlovid treatment group compared with the standard treatment group (9 days, IQR 7–11 vs. 11 days, IQR 9–14, p = 0.001).
The genotype distributions for the ACE2 rs2074192, IFNAR2 rs2236757, OAS1 rs10774671, and OAS3 rs10735079 polymorphisms were found to be in Hardy-Weinberg equilibrium (HWE) in both the Paxlovid and standard treatment groups (p > 0.05). Detailed genotype frequency data are presented in Table 2.

3.2. Alleles and Clinical Dynamics

To examine the potential interaction between hospital stay (time) and genetic factors (specific SNPs), repeated measures ANOVA was conducted. This analysis assessed both within-subject effects (interaction between time and genetic factors) and between-subject effects (main effects of genetic factors). Effect size was calculated using partial eta-squared (η2p) to quantify the influence of each factor on the continuous variable. The results of this analysis are presented in Table 3.
Repeated measures ANOVA revealed a significant interaction effect between time and the presence of the IFNAR2 rs2236757 G allele on band neutrophil levels (F = 5.051, p = 0.028, η2p = 0.067), as well as a significant between-subjects effect (F = 7.632, p = 0.007, η2p = 0.098). Additionally, a significant interaction effect between the presence of the IFNAR2 rs2236757 G allele and time was observed for platelet count (F = 5.977, p = 0.017, η2p = 0.079), segmented neutrophils (F = 5.688, p = 0.020, η2p = 0.075), and fibrinogen levels (F = 5.101, p = 0.027, η2p = 0.068). Significant between-subjects effects were also found for platelet count (F = 4.468, p = 0.038, η2p = 0.060) and fibrinogen levels (F = 6.263, p = 0.015, η2p = 0.082).
Patients carrying the IFNAR2 rs2236757 A allele exhibited a significant interaction effect between the presence of this allele and time on APTT levels (F = 6.236, p = 0.015, η2p = 0.082) and albumin levels (F = 6.258, p = 0.015, η2p = 0.082). Meanwhile, the between-subjects effect was not significant for APTT (F = 0.172, p = 0.453, η2p = 0.002) or albumin (F = 0.783, p = 0.379, η2p = 0.011).
Similarly, significant interactions were observed between the presence of the A allele and time for both OAS3 and OAS1 polymorphisms on QPT levels (F = 5.380, p = 0.023, η2p = 0.071 for both OAS3 and OAS1), with non-significant between-subjects effects (F = 0.570, p = 0.453, η2p = 0.008 for both OAS3 and OAS1). Additionally, significant interactions were found for fibrinogen levels (F = 4.128, p = 0.046, η2p = 0.056 for OAS3 and F = 4.452, p = 0.038, η2p = 0.060 for OAS1), with non-significant between-subjects effects (F = 1.951, p = 0.167, η2p = 0.027 for OAS3 and F = 2.748, p = 0.102, η2p = 0.038 for OAS1).
Paxlovid treatment did not interact significantly with any laboratory outcomes.
Figure 1 and Figure 2 provide a detailed visualization of the dynamic changes in laboratory parameters over the course of hospitalization, stratified by the presence of specific genetic polymorphisms.
Mean values and standard deviations for these parameters are presented in Table 4, while pairwise comparisons of mean differences are detailed in Table 5.
Patients lacking the IFNAR2 rs2236757 G allele at admission exhibited higher band neutrophil (MD = 10.022, 95% CI: 2.402, 17.642) and fibrinogen levels (MD = 1.908, 95% CI: 0.362, 3.454) compared with those with the G allele. These patients continued to show higher band neutrophil (MD = 12.429, 95% CI: 4.766, 20.091) and fibrinogen levels (MD = 1.643, 95% CI: 0.092, 3.194) and lower platelet count (MD = −85.000, 95% CI: −149.063, −20.937) compared with those without the G allele at discharge. Additionally, these patients exhibited higher band neutrophil (MD = 15.776, 95% CI: 8.156, 23.396) and fibrinogen levels (MD = 2.193, 95% CI: 0.647, 3.739) compared with those with the G allele at discharge.
The presence of the IFNAR2 rs2236757 G allele at admission was associated with lower platelet count (MD = −122.569, 95% CI: −215.494, −29.645) compared with those without the G allele at discharge, higher band neutrophil levels (MD = 5.754, 95% CI: 3.239, 8.268) and lower platelet count (MD = −24.292, 95% CI: −45.316, −3.269) compared with those with the G allele at discharge, and higher platelet count (MD = 98.277, 95% CI: 5.352, 191.201) compared with those without the G allele at discharge.
Patients lacking the IFNAR2 rs2236757 A allele at admission exhibited a higher activated partial thromboplastin time (APTT) level (MD = 4.129, 95% CI: 1.897, 6.361) compared with those without the A allele at discharge. Conversely, the presence of the A allele at admission was associated with a higher albumin level (MD = 6.293, 95% CI: 2.592, 9.993) compared with those with the A allele at discharge.
Patients carrying the G allele in both OAS3 rs10735079 and OAS1 rs10774671 polymorphisms at admission showed higher fibrinogen levels compared with those with the A allele at discharge (MD = 0.725, 95% CI: 0.088, 1.362 for OAS3 and MD = 0.755, 95% CI: 0.103, 1.406 for OAS1).

3.3. Genetic Determinants of Laboratory Parameter Differences

A comparative analysis of clinical and laboratory outcomes at hospital discharge in patients treated with Paxlovid is presented in Table 6.
Patients carrying the IFNAR2 rs2236757 G allele exhibited significantly lower band neutrophil counts (2%, IQR 2–4 vs. 5.5%, IQR 4.25–6.75, p = 0.001) compared with those without the G allele (Figure 3A).
Patients with the OAS3 rs10735079 G allele demonstrated elevated levels of leukocytes (10.4 × 109/L, IQR 7.59–14.2 vs. 6.59 × 109/L, IQR 4.56–8.29, p = 0.015), monocytes (7%, IQR 5–11 vs. 4%, IQR 3–7.25, p = 0.019), and hematocrit (41%, IQR 36.2–44 vs. 34.2%, IQR 30.5–37.3, p = 0.018) compared with those without the G allele (Figure 1B–D). Additionally, patients with the OAS1 rs10774671 G allele exhibited higher leukocyte (10.1 × 109/L, IQR 6.74–14 vs. 6.19 × 109/L, IQR 4.40–8.34, p = 0.023) and hematocrit (40.2%, IQR 35.2–43.5 vs. 35%, IQR 30.2–37.4, p = 0.040) levels compared with those without the G allele (Figure 3C,D).
We further compared patients with different SNP genotypes (Figure 4). Patients with the IFNAR2 rs2236757 AA genotype exhibited higher band neutrophil counts compared with those with the AG genotype (5.5% IQR 4.25–6.75 vs. 3%, 2–4, p = 0.042) and the GG genotype (5.5% IQR 4.25–6.75 vs. 2%, 1.25–3.5, p = 0.011) (Figure 4A).
Patients with the OAS1 rs10774671 AA genotype demonstrated lower leukocyte levels compared with those with the AG genotype (6.59 × 109/L, IQR 4.56–8.29 vs. 10.59 × 109/L, IQR 8.4–14.9, p = 0.037) (Figure 2B). Patients with the OAS3 rs10735079 genotype exhibited significant differences in monocyte levels between genotypes as assessed by the Kruskal–Wallis test (p = 0.039). However, no significant differences were observed when analyzed using Dunn’s multiple comparisons test (Figure 4C).

3.4. Alleles, Genotypes, and Clinical Outcomes

To investigate the potential impact of specific alleles on laboratory outcome variability during hospitalization in COVID-19 patients receiving Paxlovid treatment, a comparative analysis was conducted (Table 7).
At discharge, patients with the IFNAR2 rs2236757 G allele exhibited higher SpO2 (98%, IQR 97–98 vs. 96%, IQR 92–97, p = 0.019), segmented neutrophil counts (66%, IQR 52–78 vs. 55%, IQR 46–75, p = 0.029), and AST (30.8 mmol/L, IQR 23.3–94.1 vs. 22.6 mmol/L, IQR 16.6–25.8, p = 0.014) levels. Conversely, they displayed lower eosinophil counts (1%, IQR 0–1 vs. 1%, IQR 1–2, p = 0.048), hematocrit (37%, IQR 32–42.7 vs. 40%, IQR 34.2–45, p = 0.040), APTT (29.8 s, IQR 25–33.7 vs. 33.2 s, IQR 29.4–37, p = 0.025), and total bilirubin (11.2 mmol/L, IQR 10.5–13.5 vs. 13.7 mmol/L, IQR 10.8–19.1, p = 0.029) levels. Patients with the IFNAR2 rs2236757 A allele demonstrated higher platelet counts (215 × 109/L, IQR 166–244 vs. 173 × 109/L, IQR 142–204, p = 0.041) and lower ESR (5 mm/h, IQR 4–6 vs. 7 mm/h, IQR 4–11, p = 0.021), creatinine (92 mmol/L, IQR 84–109 vs. 103 mmol/L, IQR 95–117, p = 0.044), and albumin (43 g/L, IQR 37–51 vs. 50 g/L, IQR 45–57, p = 0.023) levels.
Patients with the ACE2 rs2074192 C allele exhibited higher SpO2 (98%, IQR 97–98 vs. 96%, IQR 93.5–98, p = 0.019), segmented neutrophil counts (66%, IQR 51.3–75.8 vs. 57%, IQR 46–71.3, p = 0.016), and AST (32.3 mmol/L, IQR 24.2–81.2 vs. 22.4 mmol/L, IQR 17.2–26.3, p = 0.004) levels along with lower APTT (29.8 s, IQR 25.5–34.9 vs. 33.3 s, IQR 29.7–37.1, p = 0.027) and fibrinogen (3.33 g/L, IQR 2.76–3.99 vs. 3.99 g/L, IQR 3.55–4.94, p = 0.017) levels. In contrast, patients with the ACE2 rs2074192 T allele displayed lower total bilirubin (10.8 mmol/L, IQR 10.2–12 vs. 12.9 mmol/L, IQR 10.7–17.8, p = 0.028) and ALP (136 mmol/L, IQR 94.3–148 vs. 148 mmol/L, IQR 125–165, p = 0.025) levels.
Patients with the OAS3 rs10735079 A allele exhibited higher segmented neutrophil counts (70.5%, IQR 53.3–77.3 vs. 61%, IQR 47.5–73.8, p = 0.027) and AST (30.1 mmol/L, IQR 24.5–67.4 vs. 22.7 mmol/L, IQR 19.4–27.3, p = 0.006) levels but lower hematocrit (37.2%, IQR 31.5–42 vs. 38.6%, IQR 34.3–43.5, p = 0.021), APTT (29.8 s, IQR 26.9–35.2 vs. 34.2 s, IQR 30–37.2, p = 0.033), and ALP (113 mmol/L, IQR 93.8–144 vs. 136 mmol/L, IQR 115–152, p = 0.025) levels. Conversely, patients with the OAS3 rs10735079 G allele displayed lower eosinophil count (1%, IQR 0–1 vs. 1%, IQR 1–2.5, p = 0.046), fibrinogen (3.33 g/L, IQR 2.11–3.88 vs. 3.99 g/L, IQR 3.55–4.1, p = 0.031), and total bilirubin (10.7 mmol/L, IQR 10.3–13.2 vs. 12.9 mmol/L, IQR 11.1–20.6, p = 0.021) levels.
Patients with the OAS1 rs10774671 A allele exhibited higher segmented neutrophil counts (70.5%, IQR 53.3–77.3 vs. 47.5%, IQR 61–73.8, p = 0.027) and AST (30.1 mmol/L, IQR 24.5–67.4 vs. 22.7 mmol/L, IQR 19.4–27.3, p = 0.006) levels and lower hematocrit (37.2%, IQR 31.5–42 vs. 38.6%, IQR 34.3–43.5, p = 0.021) and ALP (113 mmol/L, IQR 93.8–144 vs. 136 mmol/L, IQR 115–152, p = 0.025) levels. Patients with the OAS1 rs10774671 G allele showed lower eosinophil count (1%, IQR 0–1 vs. 1%, IQR 1–2.25, p = 0.035) and fibrinogen (3.33 g/L, IQR 2.27–3.99 vs. 3.99 g/L, IQR 3.55–4.26, p = 0.046) and total bilirubin (10.9 mmol/L, IQR 10.4–13.7 vs. 13.3 mmol/L, IQR 11.2–22, p = 0.014) levels.

4. Discussion

IFNAR2, a transmembrane receptor, is a component of the type I interferon (IFN) receptor complex, recognizing IFN-α and IFN-β [85,92]. The binding of IFN-I to IFNAR initiates a signaling cascade, leading to the expression of interferon-stimulated genes (ISGs) with antiviral, antiproliferative, and immunomodulatory functions [93]. One critical ISG is the RNA-activated protein kinase (PKR). Additionally, IFN activates the oligoadenylate synthetase (OAS) family proteins (OAS1, 2, and 3), which catalyze the synthesis of 2′-5′ oligoadenylate (2′-5′A). Subsequently, 2′-5′A activates RNase L, resulting in viral RNA degradation [93]. Genetic variations within IFNAR2, OAS1, and OAS3 could potentially disrupt this signaling pathway, leading to decreased protein abundance, impaired receptor internalization, or altered ligand interactions, thereby exacerbating the severity of COVID-19 [65,77,83].
All four SNPs (IFNAR2, OAS1, OAS2, and ACE2) investigated are non-synonymous SNPs (nsSNPs), resulting in alterations to the amino acid sequence of the encoded protein. The variants rs2074192, rs2236757, and rs10735079 are located within introns, whereas rs10774671 is a splice acceptor variant. Although our study focused on a Ukrainian population, extrapolating these findings to other ethnicities necessitates further investigation. To this end, we compared our data with publicly available allele frequencies for the European population from Ensembl.org (https://www.ensembl.org/). No significant discrepancies in allele distribution were identified between the two populations.
Nirmatrelvir, an orally administered protease inhibitor, binds to the catalytic dyad of Mpro via its nitrile moiety [94,95,96]. Ritonavir, a tripeptide, inhibits HIV protease by binding to its active site [95,97]. Nirmatrelvir demonstrates potent inhibitory activity against Mpro in all seven human coronavirus genotypes, including alpha-coronaviruses (HCoV-NL63 and HCoV-229E) and beta-coronaviruses (MERS-CoV, SARS-CoV-1, SARS-CoV-2, HCoV-OC43, and HCoV-HKU1) [97,98].
The findings from this study provide valuable insights into the role of genetic polymorphisms in shaping the clinical outcomes of COVID-19 patients treated with Paxlovid. These results align with, and in some cases extend, findings from previous studies that have explored the impact of host genetics on disease progression and treatment response in COVID-19 [65,99,100].
The association between the IFNAR2 rs2236757 G allele and altered inflammatory and coagulation markers observed in our study is consistent with previous research indicating that polymorphisms in the interferon receptor pathway play a significant role in modulating immune responses [101,102]. Similarly, our findings suggest that the G allele may contribute to an elevated inflammatory state, which could exacerbate disease severity [103,104]. However, while our study identified significant changes in hematological parameters, such as neutrophils and fibrinogen, further studies are needed to explore whether these variations directly affect the clinical outcomes of Paxlovid treatment.
The role of OAS1 and OAS3 polymorphisms in influencing immune and coagulation responses, as observed in this study, is also supported by prior research on type I interferons [105,106,107]. Polymorphisms in these genes have been shown to impact the production of interferons and subsequent antiviral immunity, which is crucial in the early stages of infection [108,109,110]. For example, researches demonstrated that OAS1 and OAS3 polymorphisms influence the innate immune response and the body’s ability to mount an effective defense against SARS-CoV-2 [111,112,113]. Our study builds on this by suggesting that these polymorphisms also affect coagulation pathways, potentially influencing the severity of complications in COVID-19 patients.
In accordance with Hardy–Weinberg equilibrium principles, the observed allele frequencies in our sample were consistent with those reported for the general population. While the statistical power of the current sample size was sufficient to discern associations between the investigated alleles and the clinical and laboratory parameters, future research endeavors should consider increasing the sample size to adequately address the potential influence of additional covariates, such as age, sex, and comorbidity, as well as their potential interactions.
The relationship between immune markers and the genetic variants we studied, particularly the influence of OAS1 and OAS3 alleles on leukocyte, monocyte, and hematocrit levels, is also in line with previous findings. For example, studies highlighted the crucial role of inflammatory biomarkers such as leukocyte counts and neutrophil-to-lymphocyte ratio in predicting COVID-19 severity [114,115,116]. The elevation of these markers in individuals with specific OAS3 and OAS1 alleles in our study suggests a potential link between genetic susceptibility and heightened immune activation, which may influence clinical outcomes and treatment efficacy [117,118,119]. ACE2 remains a critical receptor for SARS-CoV-2 entry; its genetic variation may not play as central a role in influencing the clinical effects of antiviral treatments like Paxlovid, at least in the context of the immune and coagulation parameters studied here [120,121,122,123].
When comparing patients at hospitalization and discharge, we identified statistically significant differences in certain outcomes based on the studied polymorphisms (Figure 5).
Regarding oxygen saturation, we observed increases in patients with the IFNAR2 rs2236757 G allele and ACE2 rs2074192 C allele. This suggests a potential link between this variant and a more robust initial antiviral response, possibly leading to better oxygenation. This observation aligns with the broader understanding of type I interferons’ crucial role in controlling viral replication and modulating the immune response in viral infections, including SARS-CoV-2 (125,126).
Additionally, patients with the IFNAR2 rs2236757 G allele and OAS3 rs10735079 and OAS1 rs1077467 A alleles exhibited elevated segmented neutrophil counts and AST levels and decreased hematocrit. This combination of findings could indicate a more pronounced inflammatory response in these individuals. This constellation of findings could reflect a heightened inflammatory state, potentially driven by an overactive innate immune response. Elevated AST, a marker of liver injury, may indicate systemic inflammation or direct viral effects on the liver. The observed decrease in hematocrit, potentially indicative of anemia, could be a consequence of chronic inflammation or other disease-related factors.
The G allele of the three interferon-related SNPs was associated with lower eosinophil and total bilirubin levels. The reduction in eosinophils might reflect a suppression of Th2-mediated immune responses, potentially influenced by the interferon signaling pathways. Lower bilirubin levels, while potentially multifactorial, could be related to altered liver function or reduced hemolysis.
Fibrinogen and ALP levels were decreased in patients with the OAS3 rs10735079 and OAS1 rs1077467 G and A alleles, respectively, and with the ACE2 rs2074192 C and T alleles. Other changes linked to the IFNAR2 rs2236757 A allele included increased platelet count and decreased creatinine, albumin, and ESR levels, while the G allele demonstrated a decreased APTT level. Patients with the ACE2 rs2074192 C allele exhibited elevated segmented neutrophils and AST levels and decreased APTT, and the T allele was associated with lower total bilirubin levels. These findings are in agreement with previous research, which has highlighted the influence of genetic polymorphisms in interferon-related genes (e.g., IFNAR2, OAS3, OAS1) on immune response modulation and disease outcomes in COVID-19 [124,125,126,127,128].
In our prior investigation, we presented the findings of a study assessing the efficacy of Paxlovid treatment in patients diagnosed with COVID-19 [17]. Notably, the administration of this antiviral agent was associated with a statistically significant reduction in the median duration of hospital stay (9 days vs. 11 days, p < 0.05). Furthermore, a positive correlation was observed between Paxlovid treatment and SpO2. The presence of comorbid conditions, including MAFLD, obesity, and chronic obstructive pulmonary disease (COPD), was documented in the patient cohort. Statistical analysis indicated that patient age and gender did not exert a significant influence on the length of hospitalization, whereas T2DM, the severity of the COVID-19 infection, and the presence of pneumonia were identified as factors significantly impacting the duration of hospital stay.
In the present investigation, the primary objective was to elucidate the impact of specific genetic polymorphisms on the dynamic profile of clinical and laboratory parameters in individuals diagnosed with COVID-19, with a particular emphasis on those receiving Paxlovid. Nevertheless, subsequent research endeavors should prioritize the expansion of the study cohort to concurrently evaluate the interplay between the presence of defined alleles and therapeutic intervention as determinants influencing the dynamics of laboratory parameters.

5. Limitations

The present study is subject to several limitations. The sample size of 72 participants may limit the statistical power of the study, especially for subgroup analyses and the detection of smaller effect sizes. Nevertheless, the Power Analysis we conducted demonstrated that the sample size is sufficient for reliable statistical conclusions. The study population was predominantly of Ukrainian ethnicity, which may limit the generalizability of the findings to other populations with different genetic backgrounds. This study was conducted at a single center, which may introduce potential biases related to patient selection and treatment protocols. The study did not include long-term follow-up of patients, which could provide valuable information on the long-term consequences of COVID-19 and the potential impact of genetic factors on post-acute COVID-19 syndrome. The study did not account for all potential confounding factors, such as socioeconomic status, comorbidities, and lifestyle factors, which could influence disease progression and laboratory parameters. The scope of this study was circumscribed to a specific set of four SNPs. These SNPs were selected based on prior evidence from the scientific literature demonstrating their association with increased severity of COVID-19 infection.

6. Conclusions

This study highlights the significant impact of genetic variations in IFNAR2, OAS1, OAS3, and ACE2 on the clinical and laboratory outcomes of COVID-19 patients receiving Paxlovid treatment. The findings suggest that these SNPs may influence the immune response, liver function, and coagulation parameters.
Polymorphisms in the interferon-related genes IFNAR2, OAS1, and OAS3 were associated with variations in inflammatory and coagulation markers. Notably, the IFNAR2 rs2236757 G allele was linked to alterations in band neutrophil, platelet, and fibrinogen levels. OAS1 and OAS3 polymorphisms influenced coagulation parameters such as QPT and fibrinogen. These findings highlight the potential role of these genes in modulating the immune response and coagulation pathways during COVID-19 infection.
Further research is necessary to elucidate the precise mechanisms underlying these associations and to explore the potential therapeutic implications of targeting these genetic markers for personalized treatment strategies in COVID-19 patients.

Author Contributions

Conceptualization and writing—original draft preparation, M.B. and O.K.; writing—review and editing, P.P., I.H. and I.K.; supervision, O.K. and V.O.; project administration, O.K.; visualization, I.K.; funding acquisition, M.B. and I.H. All authors have read and agreed to the published version of the manuscript.

Funding

RECOOP Grant #36—CSMC Senior Scientists (RCSS) “Comprehensive Analysis of Genetic Predictors for MAFLD Development in Patients with COVID-19”.

Institutional Review Board Statement

This study was conducted in accordance with the ethical principles of the Declaration of Helsinki. The research protocol was reviewed and approved by the Ethics Committee of the I. Horbachevsky Ternopil National Medical University (protocol number 74, dated 13 October 2023).

Informed Consent Statement

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

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sharma, A.; Tiwari, S.; Deb, M.K.; Marty, J.L. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2): A global pandemic and treatment strategies. Int. J. Antimicrob. Agents 2020, 56, 106054. [Google Scholar] [CrossRef] [PubMed]
  2. Li, G.; Hilgenfeld, R.; Whitley, R.; De Clercq, E. Therapeutic strategies for COVID-19: Progress and lessons learned. Nat. Rev. Drug Discov. 2023, 22, 449–475. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef] [PubMed]
  4. Patel, M.; Chaubey, A.K.; Pittman, C.U.; Mlsna, T., Jr.; Mohan, D. Coronavirus (SARS-CoV-2) in the environment: Occurrence, persistence, analysis in aquatic systems and possible management. Sci. Total Environ. 2021, 765, 142698. [Google Scholar] [CrossRef]
  5. Najjar-Debbiny, R.; Gronich, N.; Weber, G.; Khoury, J.; Amar, M.; Stein, N.; Goldstein, L.H.; Saliba, W. Effectiveness of Paxlovid in Reducing Severe Coronavirus Disease 2019 and Mortality in High-Risk Patients. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 76, e342–e349. [Google Scholar] [CrossRef]
  6. Reis, S.; Metzendorf, M.I.; Kuehn, R.; Popp, M.; Gagyor, I.; Kranke, P.; Meybohm, P.; Skoetz, N.; Weibel, S. Nirmatrelvir combined with ritonavir for preventing and treating COVID-19. Cochrane Database Syst. Rev. 2022, 9, Cd015395. [Google Scholar] [CrossRef]
  7. Bege, M.; Borbás, A. The Design, Synthesis and Mechanism of Action of Paxlovid, a Protease Inhibitor Drug Combination for the Treatment of COVID-19. Pharmaceutics 2024, 16, 217. [Google Scholar] [CrossRef]
  8. Zatovkaňuková, P.; Veselý, D.; Slíva, J. Evaluating Drug Interaction Risks: Nirmatrelvir & Ritonavir Combination (PAXLOVID®) with Concomitant Medications in Real-World Clinical Settings. Pathogens 2024, 13, 1055. [Google Scholar] [CrossRef]
  9. Moon, C.; Porges, E.; Roberts, A.; Bacon, J. A combination of nirmatrelvir and ombitasvir boosts inhibition of SARS-CoV-2 replication. Antivir. Res. 2024, 225, 105859. [Google Scholar] [CrossRef]
  10. Kronenberger, T.; Laufer, S.A.; Pillaiyar, T. COVID-19 therapeutics: Small-molecule drug development targeting SARS-CoV-2 main protease. Drug Discov. Today 2023, 28, 103579. [Google Scholar] [CrossRef]
  11. Pagliano, P.; Spera, A.; Sellitto, C.; Scarpati, G.; Folliero, V.; Piazza, O.; Franci, G.; Conti, V.; Ascione , T. Preclinical discovery and development of nirmatrelvir/ritonavir combinational therapy for the treatment of COVID-19 and the lessons learned from SARS-COV-2 variants. Expert Opin. Drug Discov. 2023, 18, 1–11. [Google Scholar] [CrossRef] [PubMed]
  12. Blair, H.A. Nirmatrelvir plus ritonavir in COVID-19: A profile of its use. Drugs Ther. Perspect. Ration. Drug Sel. Use 2023, 39, 41–47. [Google Scholar] [CrossRef] [PubMed]
  13. Ton, A.T.; Pandey, M.; Smith, J.R.; Ban, F.; Fernandez, M.; Cherkasov, A. Targeting SARS-CoV-2 papain-like protease in the postvaccine era. Trends Pharmacol. Sci. 2022, 43, 906–919. [Google Scholar] [CrossRef] [PubMed]
  14. Wong, C.K.H.; Au, I.C.H.; Lau, K.T.K.; Lau, E.H.Y.; Cowling, B.J.; Leung, G.M. Real-world effectiveness of early molnupiravir or nirmatrelvir-ritonavir in hospitalised patients with COVID-19 without supplemental oxygen requirement on admission during Hong Kong’s omicron BA.2 wave: A retrospective cohort study. Lancet Infect. Dis. 2022, 22, 1681–1693. [Google Scholar] [CrossRef]
  15. Hammond, J.; Leister-Tebbe, H.; Gardner, A.; Abreu, P.; Bao, W.; Wisemandle, W.; Baniecki, M.; Hendrick, V.M.; Damle, B.; Simón-Campos, A.; et al. Oral Nirmatrelvir for High-Risk, Nonhospitalized Adults with Covid-19. N. Engl. J. Med. 2022, 386, 1397–1408. [Google Scholar] [CrossRef]
  16. Yip, T.C.; Lui, G.C.; Lai, M.S.; Wong, V.W.; Tse, Y.K.; Ma, B.H.; Hui, E.; Leung, M.K.W.; Chan, H.L.; Hui, D.S.; et al. Impact of the Use of Oral Antiviral Agents on the Risk of Hospitalization in Community Coronavirus Disease 2019 Patients (COVID-19). Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 76, e26–e33. [Google Scholar] [CrossRef]
  17. Buchynskyi, M.; Oksenych, V.; Kamyshna, I.; Kamyshnyi, O. Exploring Paxlovid Efficacy in COVID-19 Patients with MAFLD: Insights from a Single-Center Prospective Cohort Study. Viruses 2024, 16, 112. [Google Scholar] [CrossRef]
  18. Mahase, E. Covid-19: Pfizer’s paxlovid is 89% effective in patients at risk of serious illness, company reports. BMJ (Clin. Res. ed.) 2021, 375, n2713. [Google Scholar] [CrossRef]
  19. Akinosoglou, K.-A.; Schinas, G.; Gogos, C. Oral Antiviral Treatment for COVID-19: A Comprehensive Review on Nirmatrelvir/Ritonavir. Viruses 2022, 14, 2540. [Google Scholar] [CrossRef]
  20. Fitero, A.; Negrut, N.; Popa, A.; John, H.T.; Ferician, A.C.; Manole, F.; Marian, P. Integrated Analysis of Remdesivir and Paxlovid in COVID-19 Patients: A Personalized Approach to High-Risk Individuals for Severe Evolution. J. Clin. Med. 2024, 13, 6670. [Google Scholar] [CrossRef]
  21. Lim, S.; Tignanelli, C.; Hoertel, N.; Boulware, D.; Usher, M. Prevalence of Medical Contraindications to Nirmatrelvir/Ritonavir in a Cohort of Hospitalized and Nonhospitalized Patients With COVID-19. Open Forum Infect. Dis. 2022, 9, ofac389. [Google Scholar] [CrossRef] [PubMed]
  22. González-Gómez, Á.; Caro-Teller, J.M.; González-Barrios, I.; Castro-Frontiñán, A.; Rodríguez-Quesada, P.P.; Ferrari-Piquero, J.M. Safety profile of nirmatrelvir-ritonavir: Evidence of adverse events due to drug-drug interactions. Farm. Hosp. 2024, 48, 70–74. [Google Scholar] [CrossRef]
  23. Kwok, W.C.; Tam, T.C.C.; Ho, J.C.M.; Lam, D.C.L.; Ip, M.S.; Ho, P.L. Real-World Effectiveness Study of Nirmatrelvir-Ritonavir or Molnupiravir in Hospitalized Unvaccinated Patients with Chronic Respiratory Diseases and Moderate COVID-19 at Presentation. Int. J. Chronic Obstr. Pulm. Dis. 2024, 19, 77–86. [Google Scholar] [CrossRef]
  24. Saravolatz, L.D.; Depcinski, S.; Sharma, M. Molnupiravir and Nirmatrelvir-Ritonavir: Oral Coronavirus Disease 2019 Antiviral Drugs. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 76, 165–171. [Google Scholar] [CrossRef]
  25. Lamb, Y.N. Nirmatrelvir Plus Ritonavir: First Approval. Drugs 2022, 82, 585–591. [Google Scholar] [CrossRef]
  26. Cvancara, D.J.; Baertsch, H.C.; Lehmann, A.E.; Humphreys, I.M.; Farrell, N.F.; Marshall, T.B.; Bhatt,, N.K.; Abuzeid, W.M.; Jafari, A. Postmarketing Reporting of Paxlovid-Related Dysgeusia: A Real-World Pharmacovigilance Study. Otolaryngol.—Head Neck Surg. Off. J. Am. Acad. Otolaryngol.-Head Neck Surg. 2023, 169, 55–61. [Google Scholar] [CrossRef]
  27. Caronia, L.; Xi, R.; Margolskee, R.F.; Jiang, P. Paxlovid mouth likely is mediated by activation of the TAS2R1 bitter receptor by nirmatrelvir. Biochem. Biophys. Res. Commun. 2023, 682, 138–140. [Google Scholar] [CrossRef]
  28. Li, M.; Zhang, Q.S.; Liu, X.L.; Wang, H.L.; Liu, W. Adverse Events Associated with Nirmatrelvir/Ritonavir: A Pharmacovigilance Analysis Based on FAERS. Pharmaceuticals 2022, 15, 1455. [Google Scholar] [CrossRef]
  29. Sayed Ahmed, H.A.; Mohamed, S.F.; Mostafa, M.; Elotla, S.F.; Shah, A.; Shah, J.; Fouad, A.M. Psychometric evaluation of the Arabic version of the 5-item Problem Areas in Diabetes (AR-PAID-5) scale. BMC Prim. Care 2022, 23, 148. [Google Scholar] [CrossRef]
  30. Liang, Y.; Ma, L.; Wang, Y.; Zheng, J.; Su, L.; Lyu, J. Adverse events associated with molnupiravir: A real-world disproportionality analysis in food and drug administration adverse event reporting system. Front. Pharmacol. 2023, 14, 1253799. [Google Scholar] [CrossRef]
  31. Pérez-Iruela, J.A.; Pastor-Fructuoso, P.; De Gracia-Rodríguez, C.; Soler-Vigil, M.; Gómez-Martínez, M.D.V. Adverse reactions to radiopharmaceuticals. Farm. Hosp. 2021, 45, 142–149. [Google Scholar] [PubMed]
  32. Doogue, M.P.; Polasek, T.M. The ABCD of clinical pharmacokinetics. Ther. Adv. Drug Saf. 2013, 4, 5–7. [Google Scholar] [CrossRef] [PubMed]
  33. Nwabufo, C.K.; Bendayan, R. Pharmacokinetic considerations to optimize clinical outcomes for COVID-19 drugs. Trends Pharmacol. Sci. 2022, 43, 1041–1054. [Google Scholar] [CrossRef]
  34. Pilla Reddy, V.; El-Khateeb, E.; Jo, H.; Giovino, N.; Lythgoe, E.; Sharma, S.; Tang, W.; Jamei, M.; Rastomi-Hodjegan, A. Pharmacokinetics under the COVID-19 storm. Br. J. Clin. Pharmacol. 2023, 89, 158–186. [Google Scholar] [CrossRef]
  35. McColl, E.R.; Croyle, M.A.; Zamboni, W.C.; Honer, W.G.; Heise, M.; Piquette-Miller, M.; Goralski, K.B. COVID-19 Vaccines and the Virus: Impact on Drug Metabolism and Pharmacokinetics. Drug Metab. Dispos. Biol. Fate Chem. 2023, 51, 130–141. [Google Scholar] [CrossRef]
  36. Dzhuryak, V.; Sydorchuk, L.; Sydorchuk, A.; Kamyshnyi, O.; Kshanovska, A.; Levytska, S.; Knut, R.; Sheremet, M.; Ivashchuk, S.; Petrynych, O.; et al. The cytochrome 11B2 aldosterone synthase gene CYP11B2 (RS1799998) polymorphism associates with chronic kidney disease in hypertensive patients. Biointerface Res. Appl. Chem. 2020, 10, 5406–5411. [Google Scholar] [CrossRef]
  37. Kamyshna, I.I.; Pavlovych, L.B.; Maslyanko, V.A.; Kamyshnyi, A.M. Analysis of the transcriptional activity of genes of neuropeptides and their receptors in the blood of patients with thyroid pathology. J. Med. Life 2021, 14, 243–249. [Google Scholar] [CrossRef]
  38. Roden, D.M.; McLeod, H.L.; Relling, M.V.; Williams, M.S.; Mensah, G.A.; Peterson, J.F.; Van Driest, S.L. Pharmacogenomics. Lancet 2019, 394, 521–532. [Google Scholar] [CrossRef]
  39. Ahmed, S.; Zhou, Z.; Zhou, J.; Chen, S.Q. Pharmacogenomics of Drug Metabolizing Enzymes and Transporters: Relevance to Precision Medicine. Genom. Proteom. Bioinform. 2016, 14, 298–313. [Google Scholar] [CrossRef]
  40. Babayeva, M.; Loewy, Z. Repurposing Drugs for COVID-19: Pharmacokinetics and Pharmacogenomics of Chloroquine and Hydroxychloroquine. Pharmgenom. Pers. Med. 2020, 13, 531–542. [Google Scholar] [CrossRef]
  41. Pinto, N.; Dolan, M.E. Clinically relevant genetic variations in drug metabolizing enzymes. Curr. Drug Metab. 2011, 12, 487–497. [Google Scholar] [CrossRef] [PubMed]
  42. Madian, A.G.; Wheeler, H.E.; Jones, R.B.; Dolan, M.E. Relating human genetic variation to variation in drug responses. Trends Genet. TIG 2012, 28, 487–495. [Google Scholar] [CrossRef] [PubMed]
  43. Halabitska, I.; Oksenych, V.; Kamyshnyi, O. Exploring the Efficacy of Alpha-Lipoic Acid in Comorbid Osteoarthritis and Type 2 Diabetes Mellitus. Nutrients 2024, 16, 3349. [Google Scholar] [CrossRef] [PubMed]
  44. Halabitska, I.; Babinets, L. Different consequences of the treatment of osteoarthritis in gastrointestinal comorbidity with exocrine pancreatic insufficiency. Fam. Med. Prim. Care Rev. 2021, 23, 422–428. [Google Scholar] [CrossRef]
  45. Repchuk, Y.; Sydorchuk, L.P.; Sydorchuk, A.R.; Fedonyuk, L.Y.; Kamyshnyi, O.; Korovenkova, O.; Plehutsa, I.M.; Dzhuryak, V.S.; Myshkovskii, Y.M.; Iftoda, O.M.; et al. Linkage of blood pressure, obesity and diabetes mellitus with angiotensinogen gene (AGT 704T>C/rs699) polymorphism in hypertensive patients. Bratisl. Lekárske listy 2021, 122, 715–720. [Google Scholar] [CrossRef]
  46. Carli, M.; Kolachalam, S.; Longoni, B.; Pintaudi, A.; Baldini, M.; Aringhieri, S.; Fasciani, I.; Annibale, P.; Maggio, R.; Scarselli, M. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals 2021, 14, 238. [Google Scholar] [CrossRef]
  47. Halabitska, I.; Babinets, L.; Oksenych, V.; Kamyshnyi, O. Diabetes and Osteoarthritis: Exploring the Interactions and Therapeutic Implications of Insulin, Metformin, and GLP-1-Based Interventions. Biomedicines 2024, 12, 1630. [Google Scholar] [CrossRef]
  48. Weinberg Sibony, R.; Segev, O.; Dor, S.; Raz, I. Drug Therapies for Diabetes. Int. J. Mol. Sci. 2023, 24, 17147. [Google Scholar] [CrossRef]
  49. Pavlo, P.; Kamyshna, I.; Kamyshnyi, A. Effects of metformin on the gut microbiota: A systematic review. Mol. Metab. 2023, 77, 101805. [Google Scholar] [CrossRef]
  50. Petakh, P.; Griga, V.; Mohammed, I.B.; Loshak, K.; Poliak, I.; Kamyshnyiy, A. Effects of Metformin, Insulin on Hematological Parameters of COVID-19 Patients with Type 2 Diabetes. Med. Arch. 2022, 76, 329–332. [Google Scholar] [CrossRef]
  51. Bailey, C.J. Metformin: Therapeutic profile in the treatment of type 2 diabetes. Diabetes Obes. Metab. 2024, 26 (Suppl. S3), 3–19. [Google Scholar] [CrossRef] [PubMed]
  52. Petakh, P.; Kamyshna, I.; Oksenych, V.; Kainov, D.; Kamyshnyi, A. Metformin Therapy Changes Gut Microbiota Alpha-Diversity in COVID-19 Patients with Type 2 Diabetes: The Role of SARS-CoV-2 Variants and Antibiotic Treatment. Pharmaceuticals 2023, 16, 904. [Google Scholar] [CrossRef]
  53. Petakh, P.; Kamyshna, I.; Kamyshnyi, A. Gene expression of protein kinase AMP-activated catalytic subunit alpha 1 (PRKAA1), solute carrier family 2 member 1 (SLC2A1) and mechanistic target of rapamycin (MTOR) in metformin-treated type 2 diabetes patients with COVID-19: Impact on inflammation markers. Inflammopharmacology 2024, 32, 885–891. [Google Scholar]
  54. Rommasi, F.; Nasiri, M.J.; Mirsaeidi, M. Immunomodulatory agents for COVID-19 treatment: Possible mechanism of action and immunopathology features. Mol. Cell. Biochem. 2022, 477, 711–726. [Google Scholar] [CrossRef]
  55. Zemlyak, O.S.; Babinets, L.S.; Halabitska, I.M. The Role of Endotoxicosis And Inflammation In Deepening The Pancreatic Functional Insufficiency In Chronic Pancreatitis In Combination With Type 2 Diabetes. Pol. Merkur. Lek. Organ Pol. Tow. Lek. 2023, 51, 207–215. [Google Scholar] [CrossRef]
  56. Redkva, O.V.; Babinets, L.S.; Halabitska, I.M. Evaluation of Parameters of Actual Typical Pathogenetic Syndromes In Comorbidity Of Type 2 Diabetes Mellitus And Chronic Pancreatitis. Wiad. Lek. (1960) 2021, 74, 2557–2559. [Google Scholar] [CrossRef]
  57. Chan, L.E.; Casiraghi, E.; Laraway, B.; Coleman, B.; Blau, H.; Zaman, A.; Harris, N.; Wilkins, K.; Gargano, M.; Valentini, G.; et al. Metformin is associated with reduced COVID-19 severity in patients with prediabetes. Diabetes Res. Clin. Pract. 2022, 194, 110157. [Google Scholar] [CrossRef]
  58. Halabitska, I.; Petakh, P.; Kamyshna, I.; Oksenych, V.; Kainov, D.E.; Kamyshnyi, O. The interplay of gut microbiota, obesity, and depression: Insights and interventions. Cell. Mol. Life Sci. CMLS 2024, 81, 443. [Google Scholar] [CrossRef]
  59. Hua, Y.; Dai, X.; Xu, Y.; Xing, G.; Liu, H.; Lu, T.; Chen, Y.; Zhang, Y. Drug repositioning: Progress and challenges in drug discovery for various diseases. Eur. J. Med. Chem. 2022, 234, 114239. [Google Scholar] [CrossRef]
  60. Low, Z.Y.; Farouk, I.A.; Lal, S.K. Drug Repositioning: New Approaches and Future Prospects for Life-Debilitating Diseases and the COVID-19 Pandemic Outbreak. Viruses 2020, 12, 1058. [Google Scholar] [CrossRef]
  61. Khan, S.; Agnihotri, J.; Patil, S.; Khan, N. Drug repurposing: A futuristic approach in drug discovery. J. Pharm. Biol. Sci. 2023, 11, 66–69. [Google Scholar] [CrossRef]
  62. Harrison, A.G.; Lin, T.; Wang, P. Mechanisms of SARS-CoV-2 Transmission and Pathogenesis. Trends Immunol. 2020, 41, 1100–1115. [Google Scholar] [CrossRef] [PubMed]
  63. Pouladi, N.; Abdolahi, S. Investigating the ACE2 polymorphisms in COVID-19 susceptibility: An in silico analysis. Mol. Genet. Genom. Med. 2021, 9, e1672. [Google Scholar] [CrossRef] [PubMed]
  64. Sienko, J.; Marczak, I.; Kotowski, M.; Bogacz, A.; Tejchman, K.; Sienko, M.; Kotfis, K. Association of ACE2 Gene Variants with the Severity of COVID-19 Disease-A Prospective Observational Study. Int. J. Environ. Res. Public Health 2022, 19, 12622. [Google Scholar] [CrossRef]
  65. Pairo-Castineira, E.; Clohisey, S.; Klaric, L.; Bretherick, A.D.; Rawlik, K.; Pasko, D.; Walker, S.; Parkinson, N.; Fourman, M.H.; Russell, C.D.; et al. Genetic mechanisms of critical illness in COVID-19. Nature 2021, 591, 92–98. [Google Scholar] [CrossRef]
  66. Adli, A.; Rahimi, M.; Khodaie, R.; Hashemzaei, N.; Hosseini, S.M. Role of genetic variants and host polymorphisms on COVID-19: From viral entrance mechanisms to immunological reactions. J. Med. Virol. 2022, 94, 1846–1865. [Google Scholar] [CrossRef]
  67. Chen, F.; Zhang, Y.; Li, X.; Li, W.; Liu, X.; Xue, X. The Impact of ACE2 Polymorphisms on COVID-19 Disease: Susceptibility, Severity, and Therapy. Front. Cell. Infect. Microbiol. 2021, 11, 753721. [Google Scholar] [CrossRef]
  68. Devaux, C.A.; Camoin-Jau, L. An update on angiotensin-converting enzyme 2 structure/functions, polymorphism, and duplicitous nature in the pathophysiology of coronavirus disease 2019: Implications for vascular and coagulation disease associated with severe acute respiratory syndrome coronavirus infection. Front. Microbiol. 2022, 13, 1042200. [Google Scholar]
  69. Costa, L.B.; Perez, L.G.; Palmeira, V.A.; Macedo, E.C.T.; Ribeiro, V.T.; Lanza, K.; Simões E Silva, A.C. Insights on SARS-CoV-2 Molecular Interactions With the Renin-Angiotensin System. Front. Cell Dev. Biol. 2020, 8, 559841. [Google Scholar] [CrossRef]
  70. Delpino, M.V.; Quarleri, J. SARS-CoV-2 Pathogenesis: Imbalance in the Renin-Angiotensin System Favors Lung Fibrosis. Front. Cell. Infect. Microbiol. 2020, 10, 340. [Google Scholar] [CrossRef]
  71. Hamet, P.; Pausova, Z.; Attaoua, R.; Hishmih, C.; Haloui, M.; Shin, J.; Paus, T.; Abrahamowicz, M.; Gaudet, D.; Santucci, L.; et al. SARS-CoV-2 Receptor ACE2 Gene Is Associated with Hypertension and Severity of COVID 19: Interaction with Sex, Obesity, and Smoking. Am. J. Hypertens. 2021, 34, 367–376. [Google Scholar] [CrossRef] [PubMed]
  72. Martínez-Gómez, L.E.; Herrera-López, B.; Martinez-Armenta, C.; Ortega-Peña, S.; Camacho-Rea, M.D.C.; Suarez-Ahedo, C.; Vázquez-Cárdenas, P.; Vargas-Alarcón, G.; Rojas-Velasco, G.; Fragoso, J.M.; et al. ACE and ACE2 Gene Variants Are Associated With Severe Outcomes of COVID-19 in Men. Front. Immunol. 2022, 13, 812940. [Google Scholar] [CrossRef] [PubMed]
  73. Forbester, J.L.; Humphreys, I.R. Genetic influences on viral-induced cytokine responses in the lung. Mucosal Immunol. 2021, 14, 14–25. [Google Scholar] [CrossRef]
  74. Ovsyannikova, I.G.; Haralambieva, I.H.; Crooke, S.N.; Poland, G.A.; Kennedy, R.B. The role of host genetics in the immune response to SARS-CoV-2 and COVID-19 susceptibility and severity. Immunol. Rev. 2020, 296, 205–219. [Google Scholar] [CrossRef]
  75. Buchynskyi, M.; Oksenych, V.; Kamyshna, I.; Budarna, O.; Halabitska, I.; Petakh, P.; Kamyshnyi, O. Genomic insight into COVID-19 severity in MAFLD patients: A single-center prospective cohort study. Front. Genet. 2024, 15, 1460318. [Google Scholar] [CrossRef]
  76. Buchynskyi, M.; Oksenych, V.; Kamyshna, I.; Vorobets, I.; Halabitska, I.; Kamyshnyi, O. Modulatory Roles of AHR, FFAR2, FXR, and TGR5 Gene Expression in Metabolic-Associated Fatty Liver Disease and COVID-19 Outcomes. Viruses 2024, 16, 985. [Google Scholar] [CrossRef]
  77. Fricke-Galindo, I.; Martínez-Morales, A.; Chávez-Galán, L.; Ocaña-Guzmán, R.; Buendía-Roldán, I.; Pérez-Rubio, G.; Hernández-Zenteno, R.J.; Verónica-Aguilar, A.; Alarcón-Dionet, A.; Aguilar-Duran, H.; et al. IFNAR2 relevance in the clinical outcome of individuals with severe COVID-19. Front. Immunol. 2022, 13, 949413. [Google Scholar] [CrossRef]
  78. Dieter, C.; de Almeida Brondani, L.; Lemos, N.E.; Schaeffer, A.F.; Zanotto, C.; Ramos, D.T.; Girardi, E.; Pellenz, F.M.; Camargo, J.L.; Moresco, K.S.; et al. Polymorphisms in ACE1, TMPRSS2, IFIH1, IFNAR2, and TYK2 Genes Are Associated with Worse Clinical Outcomes in COVID-19. Genes 2022, 14, 29. [Google Scholar] [CrossRef]
  79. Hamidah, B.; Pakpahan, C.; Wulandari, L.; Tinduh, D.; Wibawa, T.; Prakoeswa, C.R.S.; Oceandy, D. Expression of interferon-stimulated genes, but not polymorphisms in the interferon α/β receptor 2 gene, is associated with coronavirus disease 2019 mortality. Heliyon 2024, 10, e39002. [Google Scholar] [CrossRef]
  80. López-Bielma, M.F.; Falfán-Valencia, R.; Abarca-Rojano, E.; Pérez-Rubio, G. Participation of Single-Nucleotide Variants in IFNAR1 and IFNAR2 in the Immune Response against SARS-CoV-2 Infection: A Systematic Review. Pathogens 2023, 12, 1320. [Google Scholar] [CrossRef]
  81. Gokul, A.; Arumugam, T.; Ramsuran, V. Genetic Ethnic Differences in Human 2′-5′-Oligoadenylate Synthetase and Disease Associations: A Systematic Review. Genes 2023, 14, 527. [Google Scholar] [CrossRef] [PubMed]
  82. Bonnevie-Nielsen, V.; Field, L.L.; Lu, S.; Zheng, D.J.; Li, M.; Martensen, P.M.; Nielsen, T.B.; Beck-Nielsen, H.; Lau, Y.L.; Pociot, F. Variation in antiviral 2′,5′-oligoadenylate synthetase (2′5′AS) enzyme activity is controlled by a single-nucleotide polymorphism at a splice-acceptor site in the OAS1 gene. Am. J. Hum. Genet. 2005, 76, 623–633. [Google Scholar] [CrossRef] [PubMed]
  83. Banday, A.R.; Stanifer, M.L.; Florez-Vargas, O.; Onabajo, O.O.; Papenberg, B.W.; Zahoor, M.A.; Mirabello, L.; Ring, T.G.; Lee, C.H.; Albert, P.S.; et al. Genetic regulation of OAS1 nonsense-mediated decay underlies association with COVID-19 hospitalization in patients of European and African ancestries. Nat. Genet. 2022, 54, 1103–1116. [Google Scholar] [CrossRef] [PubMed]
  84. Kozak, K.; Pavlyshyn, H.; Kamyshnyi, O.; Shevchuk, O.; Korda, M.; Vari, S.G. The Relationship between COVID-19 Severity in Children and Immunoregulatory Gene Polymorphism. Viruses 2023, 15, 2093. [Google Scholar] [CrossRef]
  85. Kamyshnyi, A.; Koval, H.; Kobevko, O.; Buchynskyi, M.; Oksenych, V.; Kainov, D.; Lyubomirskaya, K.; Kamyshna, I.; Potters, G.; Moshynets, O. Therapeutic Effectiveness of Interferon-α2b against COVID-19 with Community-Acquired Pneumonia: The Ukrainian Experience. Int. J. Mol. Sci. 2023, 24, 6887. [Google Scholar] [CrossRef]
  86. Du, Z.; Wang, L.; Bai, Y.; Liu, Y.; Lau, E.H.Y.; Galvani, A.P.; Krug, R.M.; Cowling, B.J.; Meyers, L.A. A retrospective cohort study of Paxlovid efficacy depending on treatment time in hospitalized COVID-19 patients. eLife 2024, 13, 89801. [Google Scholar] [CrossRef]
  87. Bai, Y.; Du, Z.; Wang, L.; Lau, E.; Fung, I.; Holme, P.; Cowling, B.J.; Galvani, A.P.; Krug, R.M.; Meyers, L.A. Public Health Impact of Paxlovid as Treatment for COVID-19, United States. Emerg. Infect. Dis. 2024, 30, 262–269. [Google Scholar] [CrossRef]
  88. Rahmah, L.; Abarikwu, S.O.; Arero, A.G.; Essouma, M.; Jibril, A.T.; Fal, A.; Flisiak, R.; Makuku, R.; Marquez, L.; Mohamed, K.; et al. Oral antiviral treatments for COVID-19: Opportunities and challenges. Pharmacol. Rep. PR 2022, 74, 1255–1278. [Google Scholar] [CrossRef]
  89. Balzanelli, M.G.; Distratis, P.; Lazzaro, R.; Pham, V.H.; Tran, T.C.; Dipalma, G.; Bianco, A.; Serlenga, E.M.; Aityan, S.K.; Pierangeli, V.; et al. Analysis of Gene Single Nucleotide Polymorphisms in COVID-19 Disease Highlighting the Susceptibility and the Severity towards the Infection. Diagnostics 2022, 12, 2824. [Google Scholar] [CrossRef]
  90. Grimaudo, S.; Amodio, E.; Pipitone, R.M.; Maida, C.M.; Pizzo, S.; Prestileo, T.; Tramuto, F.; Sardina, D.; Vitale, F.; Casuccio, A.; et al. PNPLA3 and TLL-1 Polymorphisms as Potential Predictors of Disease Severity in Patients With COVID-19. Front. Cell Dev. Biol. 2021, 9, 627914. [Google Scholar] [CrossRef]
  91. Imran, L.; Zubair, R.; Mughal, S.; Shakeel, R. Ritonavir-boosted Nirmatrelvir and COVID-19 outcomes in the age of Omicron variant. Ann. Med. Surg. (2012) 2023, 85, 313–315. [Google Scholar] [CrossRef] [PubMed]
  92. Buchynskyi, M.; Kamyshna, I.; Lyubomirskaya, K.; Moshynets, O.; Kobyliak, N.; Oksenych, V.; Kamyshnyi, A. Efficacy of interferon alpha for the treatment of hospitalized patients with COVID-19: A meta-analysis. Front. Immunol. 2023, 14, 1069894. [Google Scholar] [CrossRef] [PubMed]
  93. Sodeifian, F.; Nikfarjam, M.; Kian, N.; Mohamed, K.; Rezaei, N. The role of type I interferon in the treatment of COVID-19. J. Med. Virol. 2022, 94, 63–81. [Google Scholar] [CrossRef]
  94. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. Inhibition of the main protease of SARS-CoV-2 (M(pro)) by repurposing/designing drug-like substances and utilizing nature’s toolbox of bioactive compounds. Comput. Struct. Biotechnol. J. 2022, 20, 1306–1344. [Google Scholar] [CrossRef]
  95. Citarella, A.; Dimasi, A.; Moi, D.; Passarella, D.; Scala, A.; Piperno, A.; Micale, N. Recent Advances in SARS-CoV-2 Main Protease Inhibitors: From Nirmatrelvir to Future Perspectives. Biomolecules 2023, 13, 1339. [Google Scholar] [CrossRef]
  96. Joyce, R.P.; Hu, V.W.; Wang, J. The history, mechanism, and perspectives of nirmatrelvir (PF-07321332): An orally bioavailable main protease inhibitor used in combination with ritonavir to reduce COVID-19-related hospitalizations. Med. Chem. Res. Int. J. Rapid Commun. Des. Mech. Action Biol. Act. Agents 2022, 31, 1637–1646. [Google Scholar] [CrossRef]
  97. Owen, D.R.; Allerton, C.M.N.; Anderson, A.S.; Aschenbrenner, L.; Avery, M.; Berritt, S.; Boras, B.; Cardin, R.D.; Carlo, A.; Coffman, K.J.; et al. An oral SARS-CoV-2 M(pro) inhibitor clinical candidate for the treatment of COVID-19. Science 2021, 374, 1586–1593. [Google Scholar] [CrossRef]
  98. Hashemian, S.M.R.; Sheida, A.; Taghizadieh, M.; Memar, M.Y.; Hamblin, M.R.; Bannazadeh Baghi, H.; Sadri Nahand, J.; Asemi, Z.; Mirzaei, H. Paxlovid (Nirmatrelvir/Ritonavir): A new approach to Covid-19 therapy? Biomed. Pharmacother. 2023, 162, 114367. [Google Scholar] [CrossRef]
  99. Pairo-Castineira, E.; Rawlik, K.; Bretherick, A.D.; Qi, T.; Wu, Y.; Nassiri, I.; McConkey, G.A.; Zechner, M.; Klaric, L.; Griffiths, F.; et al. GWAS and meta-analysis identifies 49 genetic variants underlying critical COVID-19. Nature 2023, 617, 764–768. [Google Scholar] [CrossRef]
  100. Fakhkhari, M.; Caidi, H.; Sadki, K. HLA alleles associated with COVID-19 susceptibility and severity in different populations: A systematic review. Egypt. J. Med. Hum. Genet. 2023, 24, 1–9. [Google Scholar] [CrossRef] [PubMed]
  101. Abdelhafez, M.; Nasereddin, A.; Shamma, O.A.; Abed, R.; Sinnokrot, R.; Marof, O.; Heif, T.; Erekat, Z.; Al-Jawabreh, A.; Ereqat, S. Association of IFNAR2 rs2236757 and OAS3 rs10735079 Polymorphisms with Susceptibility to COVID-19 Infection and Severity in Palestine. Interdiscip. Perspect. Infect. Dis. 2023, 2023, 9551163. [Google Scholar] [CrossRef] [PubMed]
  102. Jalkanen, J.; Khan, S.; Elima, K.; Huttunen, T.; Wang, N.; Hollmén, M.; Elo, L.L.; Jalkanen, S. Polymorphism in interferon alpha/beta receptor contributes to glucocorticoid response and outcome of ARDS and COVID-19. Crit. Care 2023, 27, 112. [Google Scholar] [CrossRef] [PubMed]
  103. Paranga, T.G.; Pavel-Tanasa, M.; Constantinescu, D.; Plesca, C.E.; Petrovici, C.; Miftode, I.L.; Moscalu, M.; Cianga, P.; Miftode, E.G. Comparison of C-reactive protein with distinct hyperinflammatory biomarkers in association with COVID-19 severity, mortality and SARS-CoV-2 variants. Front. Immunol. 2023, 14, 1213246. [Google Scholar] [CrossRef]
  104. Sagris, M.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Tsioufis, K.; Tousoulis, D. Genetic Predisposition and Inflammatory Inhibitors in COVID-19: Where Do We Stand? Biomedicines 2022, 10, 242. [Google Scholar] [CrossRef]
  105. Leisching, G.; Wiid, I.; Baker, B. The Association of OASL and Type I Interferons in the Pathogenesis and Survival of Intracellular Replicating Bacterial Species. Front. Cell. Infect. Microbiol. 2017, 7, 196. [Google Scholar] [CrossRef]
  106. Lee, W.B.; Choi, W.Y.; Lee, D.H.; Shim, H.; Kim-Ha, J.; Kim, Y.J. OAS1 and OAS3 negatively regulate the expression of chemokines and interferon-responsive genes in human macrophages. BMB Rep. 2019, 52, 133–138. [Google Scholar] [CrossRef]
  107. Bader El Din, N.; Moustafa, R.; Ghaleb, E.; El-Shenawy, R.; Agwa, M.; Helmy, N.; El‑Shiekh, M.; Yousif, A.; Mahfouz, M.; Seif, A.; et al. Association of OAS1 gene polymorphism with the severity of COVID-19 infection. World Acad. Sci. J. 2024, 6, 1–9. [Google Scholar] [CrossRef]
  108. Córdova-Dávalos, L.E.; Hernández-Mercado, A.; Barrón-García, C.B.; Rojas-Martínez, A.; Jiménez, M.; Salinas, E.; Cervantes-García, D. Impact of genetic polymorphisms related to innate immune response on respiratory syncytial virus infection in children. Virus Genes 2022, 58, 501–514. [Google Scholar] [CrossRef]
  109. Mihaescu, G.; Chifiriuc, M.C.; Filip, R.; Bleotu, C.; Ditu, L.M.; Constantin, M.; Cristian, R.E.; Grigore, R.; Bertesteanu, S.V.; Bertesteanu, G.; et al. Role of interferons in the antiviral battle: From virus-host crosstalk to prophylactic and therapeutic potential in SARS-CoV-2 infection. Front. Immunol. 2023, 14, 1273604. [Google Scholar] [CrossRef]
  110. Garcia-Del-Barco, D.; Risco-Acevedo, D.; Berlanga-Acosta, J.; Martos-Benítez, F.D.; Guillén-Nieto, G. Revisiting Pleiotropic Effects of Type I Interferons: Rationale for Its Prophylactic and Therapeutic Use Against SARS-CoV-2. Front. Immunol. 2021, 12, 655528. [Google Scholar] [CrossRef] [PubMed]
  111. Aliyari, S.R.; Quanquin, N.; Pernet, O.; Zhang, S.; Wang, L.; Cheng, G. The Evolutionary Dance between Innate Host Antiviral Pathways and SARS-CoV-2. Pathogens 2022, 11, 538. [Google Scholar] [CrossRef] [PubMed]
  112. Kaur, R.; Tada, T.; Landau, N.R. Restriction of SARS-CoV-2 replication by receptor transporter protein 4 (RTP4). mBio 2023, 14, e0109023. [Google Scholar] [CrossRef] [PubMed]
  113. Awady, M.; Anany, M.; Esmat, G.; Zayed, N.; Tabll, A.; Helmy, A.; Zayady, A.R.; Abdalla, M.S.; Sharada, H.M.; El Raziky, M.; et al. Single nucleotide polymorphism at exon 7 splice acceptor site of OAS1 gene determines response of hepatitis C virus patients to interferon therapy. J. Gastroenterol. Hepatol. 2010, 26, 843–850. [Google Scholar] [CrossRef]
  114. Méndez Rodríguez, M.L.; Ponciano-Gómez, A.; Campos-Aguilar, M.; Tapia-Sánchez, W.D.; Duarte-Martínez, C.L.; Romero-Herrera, J.S.; Olivas-Quintero, S.; Saucedo-Campos, A.D.; Méndez-Cruz, A.R.; Jimenez-Flores, R.; et al. Neutrophil-to-Lymphocyte Ratio and Cytokine Profiling as Predictors of Disease Severity and Survival in Unvaccinated COVID-19 Patients. Vaccines 2024, 12, 861. [Google Scholar] [CrossRef]
  115. Farah, R.; Khamisy-Farah, R.; Dziedzic, K.; Skrzypek, E.; Pruc, M.; Bragazzi, N.L.; Szarpak, L.; Jomah, M. The role of the neutrophil-lymphocyte ratio in predicting poor outcomes in COVID-19 patients. Cardiol. J. 2024, 31, 374–380. [Google Scholar] [CrossRef]
  116. Lagunas-Rangel, F. Neutrophil-to-Lymphocyte ratio and Lymphocyte-to-C-reactive protein ratio in patients with severe coronavirus disease 2019 (COVID-19): A meta-analysis. J. Med. Virol. 2020, 92, 1733–1734. [Google Scholar] [CrossRef]
  117. Magusali, N.; Graham, A.; Piers, T.; Panichnantakul, P.; Yaman, Ü.; Shoai, M.; Reynolds, R.H.; Botia, J.A.; Brookes, K.J.; Guetta-Baranes, T.; et al. Genetic variability associated with OAS1 expression in myeloid cells increases the risk of Alzheimer’s disease and severe COVID-19 outcomes. bioRxiv 2021. [Google Scholar] [CrossRef]
  118. Gunasekara, S.; Tamil Selvan, M.; Murphy, C.L.; Shatnawi, S.; Cowan, S.; More, S.; Ritchey, J.; Miller, C.A.; Rudd, J.M. Characterization of Neutrophil Functional Responses to SARS-CoV-2 Infection in a Translational Feline Model for COVID-19. Int. J. Mol. Sci. 2024, 25, 10054. [Google Scholar] [CrossRef]
  119. Odon, V.; Fros, J.J.; Goonawardane, N.; Dietrich, I.; Ibrahim, A.; Alshaikhahmed, K.; Nguyen, D.; Simmonds, P. The role of ZAP and OAS3/RNAseL pathways in the attenuation of an RNA virus with elevated frequencies of CpG and UpA dinucleotides. Nucleic Acids Res. 2019, 47, 8061–8083. [Google Scholar] [CrossRef]
  120. Datta, P.K.; Liu, F.; Fischer, T.; Rappaport, J.; Qin, X. SARS-CoV-2 pandemic and research gaps: Understanding SARS-CoV-2 interaction with the ACE2 receptor and implications for therapy. Theranostics 2020, 10, 7448–7464. [Google Scholar] [CrossRef] [PubMed]
  121. Zamorano Cuervo, N.; Grandvaux, N. ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. eLife 2020, 9, e61390. [Google Scholar] [CrossRef]
  122. Luo, D.; Bai, M.; Zhang, W.; Wang, J. The possible mechanism and research progress of ACE2 involved in cardiovascular injury caused by COVID-19: A review. Front. Cardiovasc. Med. 2024, 11, 1409723. [Google Scholar] [CrossRef]
  123. Bhimraj, A.; Morgan, R.L.; Shumaker, A.H.; Lavergne, V.; Baden, L.; Cheng, V.C.; Edwards, K.M.; Gandhi, R.; Muller, W.J.; O’Horo, J.C.; et al. Infectious Diseases Society of America Guidelines on the Treatment and Management of Patients with COVID-19. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2020, 78, e83–e102. [Google Scholar]
  124. Montenegro, A.F.L.; Clementino, M.A.F.; Yaochite, J.N.U. Type I interferon pathway genetic variants in severe COVID-19. Virus Res. 2024, 342, 199339. [Google Scholar] [CrossRef]
  125. Schönfelder, K.; Breuckmann, K.; Elsner, C.; Dittmer, U.; Fistera, D.; Herbstreit, F.; Risse, J.; Schmidt, K.; Sutharsan, S.; Taube, C.; et al. The influence of IFITM3 polymorphisms on susceptibility to SARS-CoV-2 infection and severity of COVID-19. Cytokine 2021, 142, 155492. [Google Scholar] [CrossRef]
  126. Paim, A.A.O.; Lopes-Ribeiro, Á.; Daian, E.S.D.S.O.; Andrade, L.A.F.; Moraes, T.F.S.; Barbosa-Stancioli, E.F.; da Fonseca, F.G.; Coelho-Dos-Reis, J.G. Will a little change do you good? A putative role of polymorphisms in COVID-19. Immunol. Lett. 2021, 235, 9–14. [Google Scholar] [CrossRef]
  127. van der Made, C.I.; Netea, M.G.; van der Veerdonk, F.L.; Hoischen, A. Clinical implications of host genetic variation and susceptibility to severe or critical COVID-19. Genome Med. 2022, 14, 96. [Google Scholar] [CrossRef]
  128. Ji, X.S.; Chen, B.; Ze, B.; Zhou, W.H. Human genetic basis of severe or critical illness in COVID-19. Front. Cell. Infect. Microbiol. 2022, 12, 963239. [Google Scholar] [CrossRef]
Figure 1. Repeated measures ANOVA demonstrates the impact of the presence of the IFNAR2 rs2236757 G and A alleles on the temporal dynamics of band neutrophil (A), segmented neutrophil (B), platelet count (C), APTT (D), and albumin (E) levels. Data are presented as mean ± standard error (SE).
Figure 1. Repeated measures ANOVA demonstrates the impact of the presence of the IFNAR2 rs2236757 G and A alleles on the temporal dynamics of band neutrophil (A), segmented neutrophil (B), platelet count (C), APTT (D), and albumin (E) levels. Data are presented as mean ± standard error (SE).
Jpm 15 00156 g001
Figure 2. Repeated measures ANOVA demonstrates the impact of the OAS3 rs10735079 and OAS1 rs10774671 A allele on the temporal changes in quick plasma clotting time (QPT) levels (A,C) and the OAS3 rs10735079 and OAS1 rs10774671 G allele on fibrinogen levels (B,D). Data are presented as mean ± standard error (SE).
Figure 2. Repeated measures ANOVA demonstrates the impact of the OAS3 rs10735079 and OAS1 rs10774671 A allele on the temporal changes in quick plasma clotting time (QPT) levels (A,C) and the OAS3 rs10735079 and OAS1 rs10774671 G allele on fibrinogen levels (B,D). Data are presented as mean ± standard error (SE).
Jpm 15 00156 g002
Figure 3. Comparison of median clinical and laboratory findings at hospital discharge in COVID-19 patients treated with Paxlovid, stratified by the presence of the G allele. Panel (A): IF-NAR2 rs2236757—band neutrophil levels. Panel (B): OAS3 rs10735079—monocyte levels. Panels (C,D): OAS3 rs10735079 and OAS1 rs10774671—leukocyte and hematocrit levels. Median values with interquartile ranges (IQRs) are reported. Statistical significance was determined using the Wilcoxon matched pairs test.
Figure 3. Comparison of median clinical and laboratory findings at hospital discharge in COVID-19 patients treated with Paxlovid, stratified by the presence of the G allele. Panel (A): IF-NAR2 rs2236757—band neutrophil levels. Panel (B): OAS3 rs10735079—monocyte levels. Panels (C,D): OAS3 rs10735079 and OAS1 rs10774671—leukocyte and hematocrit levels. Median values with interquartile ranges (IQRs) are reported. Statistical significance was determined using the Wilcoxon matched pairs test.
Jpm 15 00156 g003
Figure 4. A Kruskal–Wallis test with Dunn’s multiple comparisons post hoc analysis was used to compare the median laboratory findings at discharge among patients with different genotypes of IFNAR2 rs2236757, OAS1 rs10774671, and OAS3 rs10735079. (A) shows the segmented neutrophil levels across the AA, AG, and GG genotypes of the IFNAR2 rs2236757 polymorphism. (B) shows the leukocyte levels across the AA, AG, and GG genotypes of the OAS3 rs10735079 polymorphism. (C) shows the monocyte levels across the AA, AG, and GG genotypes of the OAS1 rs10774671 polymorphism.
Figure 4. A Kruskal–Wallis test with Dunn’s multiple comparisons post hoc analysis was used to compare the median laboratory findings at discharge among patients with different genotypes of IFNAR2 rs2236757, OAS1 rs10774671, and OAS3 rs10735079. (A) shows the segmented neutrophil levels across the AA, AG, and GG genotypes of the IFNAR2 rs2236757 polymorphism. (B) shows the leukocyte levels across the AA, AG, and GG genotypes of the OAS3 rs10735079 polymorphism. (C) shows the monocyte levels across the AA, AG, and GG genotypes of the OAS1 rs10774671 polymorphism.
Jpm 15 00156 g004
Figure 5. A Venn diagram illustrates changes in blood parameters between admission and dis-charge, categorized by SPPs’ (IFNAR2 rs2236757, OAS3 rs10735079, OAS1 rs1077467, and ACE2 rs2074192) presence of different alleles.
Figure 5. A Venn diagram illustrates changes in blood parameters between admission and dis-charge, categorized by SPPs’ (IFNAR2 rs2236757, OAS3 rs10735079, OAS1 rs1077467, and ACE2 rs2074192) presence of different alleles.
Jpm 15 00156 g005
Table 1. Initial Characteristics.
Table 1. Initial Characteristics.
Paxlovid Treatment (n = 23)Standard Treatment (n = 49)p-Value a
Age, median (IQR) b64 (46–71)66 (51–72)0.721
Male, No. (%)12 (52.17%)31 (63.26%)0.339
BMI, kg/m226.5 (2.09–29.7)26.1 (23.84–30.97)0.717
Duration of hospital stay, days9 (7–11)11 (9–14)0.001
COVID-19 severity (moderate/severe/critical), n18/4/124/21/40.062
Need for oxygen supply, n (%)5 (1.17%)15 (30.61%)0.575
a Fisher exact, chi-square, or Mann–Whitney U test, as appropriate; b data are presented as medians (interquartile range). Abbreviation: IQR—interquartile range.
Table 2. Assessment of Hardy–Weinberg equilibrium.
Table 2. Assessment of Hardy–Weinberg equilibrium.
Paxlovid Treatment
GenotypeACE2 rs2074192GenotypeIFNAR2 rs2236757OAS3 rs10735079OAS1 rs10774671
ExpectedObservedExpectedObservedExpectedObservedExpectedObserved
CC14.8815AA3.9249.14199.7810
CT7.247AG11.151110.721110.4310
TT0.881GG7.9283.1432.783
χ2 = 0.025; p = 0.874χ2 = 0.004; p = 0.949χ2 = 0.039; p = 0.841χ2 = 0.016; p = 0.899
Standard Treatment
CC26.4525AA4.29319.612021.5523
CT19.1022AG20.422322.782221.5919
TT3.452GG24.29236.6175.567
χ2 = 1.128; p = 0.569χ2 = 0.783; p = 0.676χ2 = 0.057; p = 0.972χ2 = 0.853; p = 0.653
Deviations from Hardy–Weinberg equilibrium (HWE) can signal inbreeding, population stratification, genotyping errors, or genetic associations in affected individuals. HWE is typically assessed using a chi-square goodness-of-fit test.
Table 3. Repeated measures ANOVA with between- and within-subject effects.
Table 3. Repeated measures ANOVA with between- and within-subject effects.
IFNAR2 rs2236757
Repeated MeasuresWithin-Subject EffectBetween-Subject Effect
InteractionFp-Valueη2pFp-Valueη2p
Band neutrophils, %Allele G × Time5.051p = 0.0280.0677.632p = 0.0070.098
Segmented neutrophils, %5.688p = 0.0200.0750.264p = 0.6090.004
Platelet count5.977p = 0.0170.0794.468p = 0.0380.060
Fibrinogen, g/L5.101p = 0.0270.0686.263p = 0.0150.082
APPT, s.Allele A × Time6.236p = 0.0150.0820.172p = 0.6790.002
Albumin, g/L6.258p = 0.0150.0820.783p = 0.3790.011
OAS3 rs10735079
QPT, %Allele A × Time5.380p = 0.0230.0710.570p = 0.4530.008
Fibrinogen, g/LAllele G × Time4.128p = 0.0460.0561.951p = 0.1670.027
OAS1 rs10774671
QPT, %Allele A × Time5.380p = 0.0230.0710.570p = 0.4530.008
Fibrinogen, g/LAllele G × Time4.452p = 0.0380.0602.748p = 0.1020.038
Inter-group differences can be modeled as a between-subjects factor, while intra-group variations resulting from repeated measures can be modeled as a within-subjects factor.
Table 4. Descriptive statistics for interaction effects.
Table 4. Descriptive statistics for interaction effects.
Band Neutrophils, %IFNAR2 rs2236757 Allele GMeanSDSE
AdmissionNo allele G19.71419.5767.399
Allele G9.6926.6850.829
DischargeNo allele G7.2866.6262.504
Allele G3.9385.1660.641
Segmented Neutrophils, %
AdmissionNo allele G52.42922.1508.372
Allele G61.76912.4271.541
DischargeNo allele G67.42911.2974.270
Allele G62.55413.1041.625
Platelet Count
AdmissionNo allele G253.000173.30565.503
Allele G215.43166.9378.302
DischargeNo allele G338.000169.05163.895
Allele G239.72380.1809.945
Fibrinogen, g/L
AdmissionNo allele G5.8962.8091.062
Allele G3.9881.5110.187
DischargeNo allele G4.2531.6210.613
Allele G3.7031.1470.142
APPT, s.IFNAR2 rs2236757 Allele AMeanSDSE
AdmissionNo allele A34.1775.4590.980
Allele A33.2684.5220.706
DischargeNo allele A30.0485.4570.980
Allele A31.8605.1160.799
Albumin, g/L
AdmissionNo allele A46.8399.2561.662
Allele A47.78010.3981.624
DischargeNo allele A45.7428.0001.437
Allele A41.4887.9161.236
QPT, %OAS3 rs10735079 Allele AMeanSDSE
AdmissionNo allele A93.32016.1945.121
Allele A90.67618.4902.348
DischargeNo allele A82.00024.1197.627
Allele A93.18218.4532.344
Fibrinogen, g/L
AdmissionNo allele G3.7101.2370.226
Allele G4.5041.9820.306
DischargeNo allele G3.7240.9430.172
Allele G3.7791.3610.210
QPT, %OAS1 rs10774671 Allele AMeanSDSE
AdmissionNo allele A93.32016.1945.121
Allele A90.67618.4902.348
DischargeNo allele A82.00024.1197.627
Allele A93.18218.4532.344
Fibrinogen, g/L
AdmissionNo allele G3.6861.2470.220
Allele G4.5631.9910.315
DischargeNo allele G3.6910.9050.160
Allele G3.8081.3980.221
Data are presented as mean ± standard deviation (SD) and standard error (SE) of the mean.
Table 5. Post hoc tests with Bonferroni correction.
Table 5. Post hoc tests with Bonferroni correction.
Band Neutrophils, %
95% CI for Mean Difference
IFNAR2 rs2236757 Allele G×TimeMean DifferenceLowerUpperSEtp bonf.
No allele G, AdmissionAllele G, Admission10.0222.40217.6422.8393.5300.004
No allele G, Discharge12.4294.76620.0912.8224.404<0.001
Allele G, Discharge15.7768.15623.3962.8395.557<0.001
Allele G, AdmissionNo allele G, Discharge2.407−5.21410.0272.8390.8481.000
Allele G, Discharge5.7543.2398.2680.9266.213<0.001
No allele G, DischargeAllele, G, Discharge3.347−4.27310.9672.8391.1791.000
Segmented Neutrophils, %
No allele G, AdmissionAllele G, Admission−9.341−23.4744.7935.271−1.7720.473
No allele G, Discharge−15.000−30.3780.3785.664−2.6490.060
Allele G, Discharge−10.125−24.2594.0085.271−1.9210.342
Allele G, AdmissionNo allele G, Discharge−5.659−19.7938.4745.271−1.0741.000
Allele G, Discharge−0.785−5.8314.2621.859−0.4221.000
No allele G, DischargeAllele, G, Discharge4.875−9.25919.0085.2710.9251.000
Platelet Count
No allele G, AdmissionAllele G, Admission37.569−55.355130.49434.4481.0911.000
No allele G, Discharge−85.000−149.063−20.93723.593−3.6030.004
Allele G, Discharge13.277−79.648106.20134.4480.3851.000
Allele G, AdmissionNo allele G, Discharge−122.569−215.494−29.64534.448−3.5580.004
Allele G, Discharge−24.292−45.316−3.2697.743−3.1380.015
No allele G, DischargeAllele, G, Discharge98.2775.352191.20134.4482.8530.032
Fibrinogen, g/L
No allele G, AdmissionAllele G, Admission1.9080.3623.4540.5763.3140.007
No allele G, Discharge1.6430.0923.1940.5712.8760.032
Allele G, Discharge2.1930.6473.7390.5763.8090.001
Allele G, AdmissionNo allele G, Discharge−0.265−1.8111.2800.576−0.4601.000
Allele G, Discharge0.285−0.2240.7940.1871.5210.796
No allele G, DischargeAllele, G, Discharge0.550−0.9952.0960.5760.9561.000
APPT, s
95% CI for Mean Difference
IFNAR2 rs2236757 Allele A × TimeMean DifferenceLowerUpperSEtp bonf.
No allele A, AdmissionAllele A, Admission0.909−2.3614.1791.2160.7481.000
No allele A, Discharge4.1291.8976.3610.8225.022<0.001
Allele A, Discharge2.317−0.9535.5881.2161.9060.356
Allele A, AdmissionNo allele A, Discharge3.220−0.0506.4901.2162.6490.056
Allele A, Discharge1.408−0.5333.3490.7151.9700.317
No allele A, DischargeAllele, A, Discharge−1.812−5.0821.4591.216−1.4900.835
Albumin, g/L
No allele A, AdmissionAllele A, Admission−0.942−6.6934.8092.140−0.4401.000
No allele A, Discharge1.097−3.1595.3531.5670.7001.000
Allele A, Discharge5.351−0.40011.1022.1402.5000.083
Allele A, AdmissionNo allele A, Discharge2.039−3.7137.7902.1400.9521.000
Allele A, Discharge6.2932.5929.9931.3634.617<0.001
No allele A, DischargeAllele, A, Discharge4.254−1.49710.0052.1401.9880.296
QPT, %
OAS3 rs10735079 Allele A × TimeMean DifferenceLowerUpperSEtp bonf.
No allele A, AdmissionAllele A, Admission2.644−14.53619.8246.3900.4141.000
No allele A, Discharge11.320−3.69926.3395.5312.0460.267
Allele A, Discharge0.138−17.04217.3186.3900.0221.000
Allele A, AdmissionNo allele A, Discharge8.676−8.50425.8566.3901.3581.000
Allele A, Discharge−2.506−8.5383.5262.221−1.1281.000
No allele A, DischargeAllele, A, Discharge−11.182−28.3625.9986.390−1.7500.498
Fibrinogen, g/L
OAS3 rs10735079 Allele G × TimeMean DifferenceLowerUpperSEtp bonf.
No allele G, AdmissionAllele G, Admission−0.794−1.7440.1570.354−2.2420.161
No allele G, Discharge−0.014−0.7680.7400.278−0.0501.000
Allele G, Discharge−0.069−1.0190.8820.354−0.1941.000
Allele G, AdmissionNo allele G, Discharge0.780−0.1711.7310.3542.2030.178
Allele G, Discharge0.7250.0881.3620.2353.0890.017
No allele G, DischargeAllele, G, Discharge−0.055−1.0060.8960.354−0.1551.000
QPT, %
OAS1 rs10774671 Allele A × TimeMean DifferenceLowerUpperSEtp bonf.
No allele A, AdmissionAllele A, Admission2.644−14.53619.8246.3900.4141.000
No allele A, Discharge11.320−3.69926.3395.5312.0460.267
Allele A, Discharge0.138−17.04217.3186.3900.0221.000
Allele A, AdmissionNo allele A, Discharge8.676−8.50425.8566.3901.3581.000
Allele A, Discharge−2.506−8.5383.5262.221−1.1281.000
No allele A, DischargeAllele, A, Discharge−11.182−28.3625.9986.390−1.7500.498
Fibrinogen, g/L
OAS1 rs10774671 Allele G × TimeMean DifferenceLowerUpperSEtp bonf.
No allele G, AdmissionAllele G, Admission−0.877−1.8160.0620.350−2.5070.081
No allele G, Discharge−0.005−0.7330.7240.268−0.0181.000
Allele G, Discharge−0.122−1.0610.8170.350−0.3491.000
Allele G, AdmissionNo allele G, Discharge0.872−0.0671.8110.3502.4940.084
Allele G, Discharge0.7550.1031.4060.2403.1450.015
No allele G, DischargeAllele, G, Discharge−0.117−1.0560.8220.350−0.3351.000
Table 6. Impact of SNP alleles on clinical and laboratory outcomes at discharge in Paxlovid-treated COVID-19 patients.
Table 6. Impact of SNP alleles on clinical and laboratory outcomes at discharge in Paxlovid-treated COVID-19 patients.
IFNAR2 rs2236757
No Allele G (n = 4)Allele G (n = 19)p-Value a
Band neutrophils, % (IQR)5.5 (4.25–6.75)2 (2–4)p = 0.001
OAS3 rs10735079
No Allele G (n = 10)Allele G (n = 13)p-Value
Leukocytes, 109/L6.59 (4.56–8.29)10.4 (7.59–14.2)p = 0.015
Monocytes, %4 (3–7.25)7 (5–11)p = 0.019
Hematocrit, %34.2 (30.5–37.3)41 (36.2–44)p = 0.018
OAS1 rs10774671
No Allele G (n = 9)Allele G (n = 14)p-Value
Leukocytes, 109/L6.19 (4.40–8.34)10.1 (6.74–14)p = 0.023
Hematocrit, %35 (30.2–37.4)40.2 (35.2–43.5)p = 0.040
a Mann–Whitney test; statistically significant findings are denoted in bold.
Table 7. Impact of genetic variation on clinical course and laboratory parameters during Paxlovid treatment for hospitalized COVID-19 patients.
Table 7. Impact of genetic variation on clinical course and laboratory parameters during Paxlovid treatment for hospitalized COVID-19 patients.
IFNAR2 rs2236757
Allele A (n = 15)
Allele G (n = 19)
AdmissionDischargep-Value a
SpO2, %, median (IQR)Allele A96 (94–98)98 (97–98)p = 0.151
Allele G96 (92–97)98 (97–98)p = 0.019
Segmented neutrophils, %Allele A55 (46–75)66 (48–74)p = 0.059
Allele G61 (47–70)66 (52–78)p = 0.029
Eosinophils, %Allele A1 (0–2)1 (0–1)p = 0.169
Allele G1 (1–2)1 (0–1)p = 0.048
ESR, mm/hAllele A7 (4–11)5 (4–6)p = 0.021
Allele G5 (4–10)4 (4–5)p = 0.371
Platelet count, 109/LAllele A173 (142–204)215 (166–244)p = 0.041
Allele G193 (165–231)220 (169–262)p = 0.064
Hematocrit, %Allele A37.2 (34–44)36.6 (30.8–41)p = 0.132
Allele G40 (34.2–45)37 (32–42.7)p = 0.040
APTT, sAllele A33.2 (29.4–35.3)32.8 (24.6–35.8)p = 0.177
Allele G33.2 (29.4–37)29.8 (25–33.7)p = 0.035
Total bilirubin, mmol/LAllele A13.4 (11.1–19.1)11.2 (10.7–14.1)p = 0.128
Allele G13.7 (10.8–19.1)11.2 (10.5–13.5)p = 0.029
AST, mmol/LAllele A23.3 (19–27.8)25.5 (23.3–67.4)p = 0.112
Allele G22.2 (16.6–25.8)30.8 (23.3–94.1)p = 0.014
Creatinine, mmol/LAllele A103 (95–117)92 (84–109)p = 0.044
Allele G96 (80–117)98 (86–109)p = 0.825
Albumin, g/LAllele A50 (45–57)43 (37–51)p = 0.023
Allele G50 (45–56)46 (42–51)p = 0.159
ACE 2 rs2074192
Allele C (n = 22)
Allele T (n = 8)
AdmissionDischargep-Value a
SpO2, %, median (IQR)Allele C96 (93.5–98)98 (97–98)p = 0.019
Allele T96 (92–97.8)97 (97–98)p = 0.102
Segmented neutrophils, %Allele C57 (46–71.3)66 (51.3–75.8)p = 0.016
Allele T57 (44.8–65.5)68.5 (52.5–73.8)p = 0.078
APTT, sAllele C33.3 (29.7–37.1)29.8 (25.5–34.9)p = 0.027
Allele T32.8 (29.6–35.1)29.8 (25.2–34.5)p = 0.093
Fibrinogen, g/LAllele C3.99 (3.55–4.94)3.33 (2.76–3.99)p = 0.017
Allele T3.63 (3.55–4.33)3.83 (1.75–3.99)p = 0.611
Total, mmol/LAllele C12.7 (10.8–15.9)11.1 (10.6–13.7)p = 0.112
Allele T12.9 (10.7–17.8)10.8 (10.2–12)p = 0.028
AST, mmol/LAllele C22.4 (17.2–26.3)32.3 (24.2–81.2)p = 0.004
Allele T21.9 (14.9–28.1)29.1 (21.3–81.1)p = 0.327
ALP, mmol/LAllele C140 (116–1600122 (95.3–147)p = 0.077
Allele T148 (125–165)136 (94.3–148)p = 0.025
OAS3 rs10735079
Allele A (n = 20)
Allele G (n = 13)
AdmissionDischargep-Value a
Segmented neutrophils, %Allele A61 (47.5–73.8)70.5 (53.3–77.3)p = 0.027
Allele G55 (46–69.5)63 (48.5–75)p = 0.307
Eosinophils, %Allele A1 (0.25–2)1 (0–1)p = 0.134
Allele G1 (1–2.5)1 (0–1)p = 0.046
Hematocrit, %Allele A38.6 (34.3–43.5)37.2 (31.5–42)p = 0.021
Allele G42 (35.3–48.2)41 (36.2–44)p = 0.916
APTT, sAllele A34.2 (30–37.2)29.8 (26.9–35.2)p = 0.033
Allele G33.4 (29.9–37.1)29.1 (24.7–34.8)p = 0.066
Fibrinogen, g/LAllele A3.99 (3.55–)5.053.63 (2.92–3.99)p = 0.064
Allele G3.99 (3.55–4.1)3.33 (2.11–3.88)p = 0.031
Total bilirubin, mmol/LAllele A13.2 (10.7–16.7)11.1 (10.6–14.3)p = 0.070
Allele G12.9 (11.1–20.6)10.7 (10.3–13.2)p = 0.021
AST, mmol/LAllele A22.7 (19.4–27.3)30.1 (24.5–67.4)p = 0.006
Allele G22.2 (18.6–26.8)26.5 (23.9–96.3)p = 0.116
ALP, mmol/LAllele A136 (115–152)113 (93.8–144)p = 0.025
Allele G138 (121–156)127 (94–151)p = 0.196
OAS1 rs10774671
Allele A (n = 20)
Allele G (n = 14)
AdmissionDischargep-Value a
Segmented neutrophils, %Allele A47.5 (61–73.8)70.5 (53.3–77.3)p = 0.027
Allele G57 (46–69.3)63 (48.8–75)p = 0.183
Eosinophils, %Allele A1 (0.25–2)1 (0–1)p = 0.134
Allele G1 (1–2.25)1 (0–1)p = 0.032
Hematocrit, %Allele A38.6 (34.3–43.5)37.2 (31.5–42)p = 0.021
Allele G41 (35.8–47.7)40.2 (35.2–43.5)p = 0.638
Fibrinogen, g/LAllele A3.99 (3.55–55)3.63 (2.92–3.99)p = 0.064
Allele G3.99 (3.55–4.26)3.33 (2.27–3.99)p = 0.046
Total bilirubin, mmol/LAllele A13.2 (10.7–16.7)11.1 (10.6–14.3)p = 0.170
Allele G13.3 (11.2–22)10.9 (10.4–13.7)p = 0.014
AST, mmol/LAllele A22.7 (19.4–27.3)30.1 (24.5–67.4)p = 0.006
Allele G22.4 (19.5–26.3)26 (24.2–95.2)p = 0.096
ALP, mmol/LAllele A136 (115–152)113 (93.8–144)p = 0.025
Allele G137 (120–155)125 (95–148)p = 0.158
a Wilcoxon matched pairs test; statistically significant findings are denoted in bold.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Buchynskyi, M.; Kamyshna, I.; Halabitska, I.; Petakh, P.; Oksenych, V.; Kamyshnyi, O. Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course. J. Pers. Med. 2025, 15, 156. https://doi.org/10.3390/jpm15040156

AMA Style

Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Oksenych V, Kamyshnyi O. Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course. Journal of Personalized Medicine. 2025; 15(4):156. https://doi.org/10.3390/jpm15040156

Chicago/Turabian Style

Buchynskyi, Mykhailo, Iryna Kamyshna, Iryna Halabitska, Pavlo Petakh, Valentyn Oksenych, and Oleksandr Kamyshnyi. 2025. "Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course" Journal of Personalized Medicine 15, no. 4: 156. https://doi.org/10.3390/jpm15040156

APA Style

Buchynskyi, M., Kamyshna, I., Halabitska, I., Petakh, P., Oksenych, V., & Kamyshnyi, O. (2025). Genetic Predictors of Paxlovid Treatment Response: The Role of IFNAR2, OAS1, OAS3, and ACE2 in COVID-19 Clinical Course. Journal of Personalized Medicine, 15(4), 156. https://doi.org/10.3390/jpm15040156

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