Next Article in Journal
The Non-Muscle-Splitting Mini-Incision Donor Nephrectomy Remains a Feasible Technique in the Laparoscopic Era of Living Kidney Donation
Previous Article in Journal
Calcified Granulomatous Pneumocystis Jirovecii Pneumonia in a Toddler with Severe Combined Immunodeficiency—A Case Report
Previous Article in Special Issue
COVID-19 and Kidney Transplantation: Epidemiology, Histopathological Presentation, Clinical Presentation and Outcomes, and Therapeutic Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

What Is New in Prophylaxis and Treatment of COVID-19 in Renal Transplant Patients? A Report from an ESOT Meeting on the Topic

by
Maurizio Salvadori
Department of Renal Transplantation, Careggi University Hospital, Viale Pieraccini 18, 50139 Florence, Italy
Transplantology 2022, 3(4), 288-306; https://doi.org/10.3390/transplantology3040030
Submission received: 6 September 2022 / Revised: 20 October 2022 / Accepted: 26 October 2022 / Published: 2 November 2022
(This article belongs to the Special Issue COVID-19 in Kidney Transplantation)

Abstract

:
I should highlight that this manuscript is not a formal review on the topic, but a report from an ESOT meeting held on 22 June 2022. The assumption of immunosuppressants exposes kidney transplant recipients to the risk of infections, including COVID-19 infection. A transplant patient having COVID-19 infection raises several questions, including whether the immunosuppressive therapy should be reduced with the consequent risk of favoring acute rejections. Patient vaccination before transplantation is probably the gold standard to avoid the risk of COVID-19 infection after transplantation. In the case of transplant patients, three measures may be undertaken: vaccination, use of monoclonal antibodies and use of therapeutic antiviral small molecules. Concerning vaccination, it is still debated which one is the best and how many doses should be administered, particularly considering the new variants of the virus. The onset of virus variants has stimulated researchers to find new active vaccines. In addition, not all transplant patients develop antibodies. An alternative prophylactic measure to be principally used for patients that do not develop antibodies after vaccination is the use of monoclonal antibodies. These drugs may be administered as prophylaxis or in the early stage of the disease. Finally, the small antiviral molecules may be used again as prophylaxis or treatment. Their major drawbacks are their interference with immunosuppressive drugs and the fact that some of them cannot be administered to patients with low eGFR.

1. Introduction

Kidney transplant (KTx) recipients affected by COVID-19 infection present several challenges principally concerning prophylaxis and therapy.
SARS-CoV-2 has a great impact on immunocompromised individuals with multiple comorbid conditions, as is common in KTx recipients.
Severe disease in immunocompromised individuals may reflect the inability to mount an effective immune response, even after vaccination [1].
The assessment of which individuals will benefit from monoclonal antibodies and small molecular therapeutics is complicated by an incomplete understanding of the thresholds for a protective immunity.
Moreover, in transplant patients, a discordance exists between cellular and immune responses.
In addition, in the immunodominant spike (S) protein, 5016 different amino acid replacements or substitutions have been identified, and multiple deletions may be present. As variants emerged, natural antibodies, therapeutic monoclonal antibodies and some vaccine-elicited antibodies have become less effective in preventing disease progression [2].
Overall, initially, the mutation rates were thought to be rather low, but it was later well recognized that spike protein mutations by altered membrane fusion of virus and host cells led to either altered pathogenicity and human-to-human spread, altered susceptibility to vaccine-induced immunity and an altered response to monoclonal and small-molecule therapeutics.
Additionally, multiple studies identified several variables associated with a poor humoral immune response, including older age, high-dose assumption of corticosteroids in the last 12 months, triple immunosuppression and the use of mycophenolate mofetil and belatacept.
On 16 June 2022, the European Society for Organ Transplantation held a meeting on the Prevention and Treatments of COVID-19 in Solid Organ Transplant Recipients.
This study is conducted to convey the main findings of the meeting, taking into account several papers on the topic and that were mentioned in the meeting. In addition, several guidelines, position statements or guidelines of international relevance have been considered [3,4,5,6].
In this study, we will examine new approved therapies with particular reference to:
(a)
Vaccination;
(b)
Monoclonal antibodies, examining pre-exposure prophylaxis with tixagevimab and cilgavimab (Eurisheld) and other monoclonal and polyclonal antibody products;
(c)
Small antiviral proteins, such as Nirmatrelvir/ritonavir, Molnupivar and Remdesivir (RMD) with particular concern toward their interaction with immunosuppressive agents.

2. Vaccination

In normal conditions, after vaccination, the immune response includes neutralizing antibodies that inhibit the binding of the virus to the receptor and T cell responses that are detectable either after vaccination or natural infection. Antibodies have a main function in preventing infection; T cells and antibodies both contribute to the prevention of severe disease.
Two wide studies documented firstly the efficacy and safety of two mRNA SARS-CoV-2 vaccines in healthy subjects.
In one study, 43,548 participants underwent randomization to receive BNT162b2 or a placebo [7]. The conclusion of the study was that a two-dose regimen of BNT 162b2 conferred 95% protection against COVID-19. A different study evaluated the safety and efficacy of the mRNA-1273 vaccine. This vaccine is a lipid nanoparticle-encapsulated mRNA that encodes the full-length spike protein of the SARS-CoV-2 virus. The study was conducted in 99 centers in the United States involving 30,420 subjects assigned 1:1 to receive either the vaccine or a placebo [8]. The mRNA-1273 vaccine had an efficacy of 94.1% in preventing COVID-19.
Both studies were conducted in non-transplanted and healthy subjects and against the Delta variant of the virus.
In a different study, Hamm et al. [9] evaluated the decline in the antibody concentration 6 months after two doses of the SARS-CoV-2 BNT162b2 vaccine in 200 solid organ transplant (SOT) recipients and in 200 matched healthy controls. The decline of both cellular and humoral responses was higher in the transplant patients with respect to the healthy subjects, and risk factors for the decline were older age, treatment with mycophenolate and treatment with corticosteroids. The decline was higher in KTX recipients with respect to other SOT recipients.
Another study [10] conducted a systematic review and a meta-analysis to compare the seroconversion rate with two doses of BNT162b2 versus mRNA-1273 in SOT recipients. The conclusion of the study was that, in SOT recipients, a higher conversion rate was observed after vaccination with mRNA-1273 compared with BNT162b2 (Figure 1). The authors concluded that further studies are needed to verify whether this difference remains after a third dose of vaccination.
A study by Liefeldt [11] examined the predictors of the serological response to SARS-CoV-2 vaccination in kidney transplant patients. The study found that predictors of a weak response were the age at vaccination, time after kidney transplantation, immunosuppression, estimated glomerular filtration rate (eGFR) at vaccination and lymphocyte count at vaccination (Table 1a). The time after transplantation had an important role in the immune response, because, in the early period, the higher immunosuppression and the higher incidence of rejection may prevent or reduce the immune response. The use of mycophenolate further impaired the response to vaccination and the authors hypothesized that erythrocyte IMPDH activity could be used to monitor mycophenolate treatment (Table 1b).
The main message of Liefeldt’s study is that, in addition to risk factors found by other authors, their study highlights the relevance of the MPA treatment.
Another study by Balsby et al. [12], in SOT patients, found that the antibody response after a third dose of BNT162b2 was improved, but the overall response was still low, with a significant ratio of non-responders. The predictors of a poor response were similar to those found in other studies. The KTx recipients were confirmed to be the lower responders (Figure 2).
A study by Hod et al. [13] in 99 KTx patients documented the relevance of a third booster dose of BNT162b2. The study reported that a third dose increased both the level of neutralizing antibodies and receptor binding domain (RBD) antibodies in KTx patients. The results of both responders and non-responders are shown in Table 2.
The study found that, after a third BNT162b2 booster dose, the humoral response assessed by both RBD IgG and neutralizing antibodies (NAs) significantly increased.
Protective values of RBD IgG and neutralizing antibodies, their interrelationship and factors influencing their levels.
In an initial study by Dimeglio et al. [14], 8758 healthy people were studied and followed over time. Approximately half of them received one or two doses of a vaccine. Regarding NAs, a titer of 64 to 128 afforded 94% protection and a titer of 256 provided full protection. Considering the RBD IgG, a concentration between 141 and 1700 binding antibody units (BAU/mL) provided 89.3% protection and full protection was provided by BAU/mL > 1700.
The study referred to healthy people and to wild-type or delta SARS-CoV-2.
In a further study, Dimeglio et al. [15] considered the protective values after the emergence of Omicron BA.1 and BA.2. They analyzed 259 healthy subjects after vaccination. A NA titer of 64 to 128 afforded 78.4% protection and 94% protection against the delta variant. Levels of BAU/mL as high as 20,000 provided only 87.7% protection.
A study by Suntronwong et al. [16] found a strong correlation between the binding and neutralizing antibodies after a third vaccination dose.
Almost all of the studies documented a decline over time in the antibody levels and, as expected, transplant patients had significantly lower levels, even after vaccination.
A recent systematic review and meta-analysis [17] documented again the immunogenicity and risk factors associated with a poor humoral immune response to any SARS-CoV-2 vaccines in SOT recipients. Overall, 112 studies were included in the meta-analysis with 11,713 SOT recipients. The antibody responses both for anti-spike antibodies and for neutralizing antibodies were higher according to the number of vaccines. The factors principally associated with a poor antibody response were older age, deceased donor, antimetabolite use, recent rituximab use and recent antithymocyte globulin exposure. The authors suggested that more effort is needed to modulate the risk factors associated with reduced humoral responses among recipients of SOT.
Several studies [18,19,20] documented the low immunization rate that occurs in several subjects receiving two doses of the mRNA-1273 SARS-CoV-2 vaccine. Among these were kidney transplant recipients, as highlighted by the study of Benotmane et al. [18] on 205 KT recipients that developed an immunization rate as low as 48%. Other non-transplant patients with a weak humoral immune response were older patients. The weak response occurred both after naïve COVID-19 infection and after BNT162b2 vaccination [19]. The study also suggested that, in these patients, vaccination after infection may be useful as it maintains a higher antibody titer for a long period.
Yang et al. [20] highlighted the relevance of the factor “age” in the response to vaccination in non-transplant patients. The authors studied the antibody response in 3648 adult patients, and their analysis found that a distinct antibody response characterized different age groups after two doses of the vaccine. The study suggested that age-targeted strategies for disease screening and management, as well as vaccine development, may be warranted.
A recent study by Mazzoni et al. [21] on non-transplant patients documented that SARS-CoV-2 infection and vaccination triggered long-living B cells and CD4 lymphocytes. Either in patients vaccinated after recovering from a COVID-19 infection and in patients vaccinated naïve, there was a significant decrease in all antibody levels, even if the decline was more pronounced in naïve individuals. The decline was detectable 8 months after vaccination or COVID-19 infection. Memory cells are still detectable after 8 months. The decrease in humoral immunity may account for reinfection. A third (booster) dose restores the humoral activity in vaccinated subjects, while the need of a booster dose is still an object of discussion for previously infected patients. The results of this study confirmed those of previous studies also performed in non-transplant patients, which documented similar data [22,23,24,25].
Interesting data were also documented by the already-cited study of Hamm [9]. Previous studies had already documented a reduced humoral response after two vaccine doses in SOT recipients [26,27,28,29]. The Hamm study evaluated the anti-receptor binding domain (RBD) immunoglobulins after two doses of BNT162b2 in SOT recipients versus controls and confirmed the reduced immunological response in SOT recipients. In addition, the response was weaker in KTx recipients than in those receiving other types of transplants.
In conclusion, in SOT recipients, two doses of vaccination confer to the patients low immunogenicity [30].
A randomized trial of a third dose of the mRNA-1273 vaccine in transplant recipients was conducted by Hall et al. [31]. The study documented an increase in anti-RBD antibodies, an increase in neutralizing antibodies and an increase in polyfunctional CD4 T cells after a third dose in SOT recipients.
The beneficial effect of three vaccine doses in SOT recipients was documented by the study of Kamar et al. [32], who highlighted a relevant increase in anti-SARS-CoV-2 antibodies after three doses of the mRNA vaccine BNT162b2 (Figure 3).
In a different study, Kamar et al. [33] documented the efficacy of three vaccine doses (BNT162b2) in 850 SOT recipients. The study evaluated the anti-SARS-CoV-2 spike protein and neutralizing antibodies at 1 and 3 months after three doses. The study reported that only two-thirds of SOT recipients developed antibodies, while one-third remained unprotected. This study highlights the importance of immune monitoring to optimize vaccination in SOT recipients.
As 30% of SOT recipients do not develop protecting antibodies, several studies evaluated whether a fourth dose was effective in producing protective antibodies. The first study was conducted in France and documented an increase in the anti-SARS-CoV-2 antibody concentrations in SOT patients after four doses of the mRNABNT162b2 vaccine. Similarly, there was an increase in SARS-CoV-2-reactive IFN-gamma-producing cells [34].
A different study [35] documented, in 71 KTx patients, a relevant and protective increase after the fourth dose of an mRNA-1273 vaccine in a phase 4 study (NCT04801667).
However, in this study, as in another [36], patients with advanced age and with deceased donors may not have developed a protective immune response.
Other studies documented the beneficial effect of a fourth vaccine dose. In a case series of 92 KTx recipients [37], a significant increase in IgG antibodies against the spike protein was reported.
Benotmane et al. [38] documented, in 67 KTx recipients, that a fourth dose of the mRNA-1273 SARS-CoV-2 vaccine improved serum neutralization against the Delta variant. In a different study, Masset et al. [39] highlighted the need for a fourth SARS-CoV-2 mRNA vaccine in strictly seronegative KTx recipients. After the fourth dose, 50% of patients developed protective antibody levels. Factors influencing the positive response were low steroid use, less lymphopenia and a longer time between the third and the fourth doses.
The identification of the Omicron variants changed the picture described due to the number of alterations in the spike glycoproteins that lead to antibody evasion and to a reduced immune response to vaccination.
The study of Iketani et al. [40], in non-transplant patients, documented that, after two or three doses, the levels of neutralizing antibodies were significantly lower in the case of omicron variants than against delta variants.
The study of Shen [41] collected other studies [42,43,44,45] and documented that, after two mRNA vaccine doses, there was minimal antibody-neutralizing level against omicron in non-transplant patients. Robust neutralization of omicron developed after a third mRNA vaccine dose. The magnitude of omicron neutralization after a third (booster) dose was comparable to the delta neutralization after two doses. A fourth dose (second booster) had the potential to further improve the magnitude and durability of the immune response.
In Israel, Bar On et al. [46] selected 1,252,331 subjects aged 60 years and older. Half of the subjects received three vaccine doses and half received four doses. The rates of SARS-CoV-2 infection and severe COVID-19 were lower after a fourth dose of the BNT162b2 vaccine than after only three doses.
These data were confirmed by another similar study [47], even if this study highlighted higher protection in people older than 70 years.
Other studies [48] highlighted that the clinical characteristics of the subjects affected by the omicron variant were milder than those of subjects affected by previous variants. In any case, full vaccination is required for effective protection against the development of clinical severity.
In the case of transplant patients, several studies [49,50,51] reported a suboptimal antibody response against SARS-CoV-2 omicron variants with respect to the wild type (WT) and the delta variant after a third dose of an mRNA vaccine (Figure 4). In a different study [52], in transplant patients, a fourth dose of a COVID-19 vaccine did not induce neutralization of the omicron variant among SOT recipients with a suboptimal vaccine response.
An overall review of the effects of different doses of vaccine on the serological response is shown in Table 3. The review is the result of a study by Sakuraba et al. [53], who collected other epidemiological studies [54,55].
In conclusion, we may state that:
(a)
The immune responses to COVID-19 mRNA vaccines include both B and T cells;
(b)
The immune responses in SOT recipients are inferior to those obtained in both healthy controls and SOT candidates;
(c)
Risk factors for non-responses include short duration from TX, treatment for acute rejection and use of Mycophenolate;
(d)
Natural infection, and third and fourth doses improve the immune response;
(e)
A third dose reduces the risk of severe COVID-19;
(f)
A fourth dose may provide protection against the omicron variant;
(g)
Vaccination of all SOT candidates and SOT recipients is a priority.

3. Monoclonal Antibodies

Monoclonal antibodies may be used either for COVID-19 prophylaxis or for the early treatment of COVID-19 infection. We have already described that, despite receiving three or four doses of a vaccine, not all patients are protected, particularly patients with reduced immunocompetence, such as transplant patients.
There are two strategies to protect patients with a weak immune response to a third dose or with no response at all.
We may proceed with an additional vaccine dose or with the aid of pre-exposition monoclonal antibodies.
REGEN-COV is a combination of two neutralizing monoclonal antibodies, casirivimab and imdevimab, that bind to the receptor-binding domain of the spike protein [56,57].
A trial with the use of REGEN-COV administered subcutaneously was conducted in 112 sites [58]. A total of 1505 participants neither having received a transplant nor being on a waiting list for transplantation and without any evidence of previous or ongoing infection were assigned to receive REGEN-COV or a placebo. At the follow-up, 7.8% of participants in the placebo group developed symptomatic infection versus 1.5% in the REGEN-COV group, with a significant difference (p < 0.001). This study documented the efficacy and safety of REGEN-COV. Almost simultaneously, another study [59] highlighted the efficacy of a different combination of monoclonal antibodies (bamlanivimab–etesevimab), as documented by the study of Dougan et al. [60]. All these studies were conducted in healthy, non-immunocompromised subjects.
After a recommendation of the France Authorities for Health [61], Dimeglio et al. [62] afforded the possibility to SOT recipients who were not responders or weak responders to receive casirivimab and indevimab in two distinct doses. Out of 478 patients, 182 received treatment while 296 remained untreated for different reasons. In the follow-up period of 60 days, no SARS-CoV-2 infection was verified in the treated group versus 4.4% infection in the non-treated group. In a different study, Ducloux et al. [63] obtained similar results with a combination of monoclonal antibodies in KTx recipients. Out of 119 KTx recipients who did not develop protective antibodies after vaccination, 88 were treated versus 31 not treated. No COVID-19 infection developed in monoclonal treated patients, while 16% of infections occurred in non-treated patients. The authors conclude that treatment with monoclonal antibodies conferred protection in immunocompromised patients.
This state-of-the-art changed with the advent of the Omicron variants.
Kamar et al. [64] reported an Omicron breakthrough infection in a KTx patient administered pre-exposition casirivimab and imdevimab monoclonal antibodies. The infection occurred despite a high concentration of anti-S antibodies that usually conferred 100% protection against non-Omicron variants. This highlights that very high anti-S antibodies are required to prevent Omicron infection. In the study of Planas et al. [65], the authors examined Omicron’s sensitivity to nine monoclonal antibodies that have been clinically approved or studied in trials [66]. The authors found that Omicron was completely or partially resistant to all monoclonal antibodies tested. Previous studies have already documented the reduced sensitivity of Omicron to monoclonal antibodies [67,68]. The study of Planas [65] documented a considerable escape of SARS-CoV-2 Omicron from antibody neutralization. All these studies were conducted in non-transplant patients.
A different approach involves the use of a different monoclonal antibody combination (AZD7442) composed of tixagevimab and cilgavimab. Tixagevimab and cilgavimab bind to distinct epitopes of the SARS-CoV-2 spike protein receptor binding site, neutralizing the virus [69,70,71]. In an ongoing phase 3 trial involving 5197 participants randomized to receive either AZD7442 or a placebo, the safety and efficacy of AZD7442 was documented in healthy subjects [72] (PROVENT trial, NCT04625725). The study was conducted in non-transplant patients, but at risk of SARS-CoV-2 infection.
The association of tixagevimab and cilgavimab has been named Evushled. A study by Bertrand et al. [73] in KTx recipients compared Evusheld-treated patients with vaccinated and protected patients and with patients receiving the association of casirivimab and imdevimab. Patients treated with Evusheld had similar outcomes to vaccinated patients, while patients treated with casirivimab–imdevimab exhibited higher infection rates, principally due to the Omicron variants.
However, another relevant study on prophylaxis induced by Evusheld in KTx recipients [74] documented that less than 10% of patients treated with Evusheld were able to neutralize the Omicron BA.1 variant when administered a dose of 300 mg. Therefore, the Food and Drug Administration (FDA) recommended the revision of Evusheld dosing [75]. Overall, the Omicron variant represents a problem in the use of monoclonal antibodies as a prophylactic measure. Indeed, in the study of Iketani [40], BA.2 exhibited marked resistance to 17 of the neutralizing monoclonal antibodies tested.
The question of whether an increased dosage of Evusheld could achieve adequate protection without collateral effects remains open.
A different way to use monoclonal antibodies against SARS-CoV-2 is to administer the antibodies not as a prophylaxis, but as an early treatment in patients already affected by COVID-19 infection. A study by Mazzotta et al. [76], in line with other previous studies [77,78], compared the effectiveness of casirivimab/imdevimab with that of bamlanivimab/etesevimab as an early treatment in non-hospitalized and non-transplant patients with COVID-19. Worsening of the infection occurred in 6.3% of patients treated with bamlanivimab/etesevimab versus 2% of patients treated with casirivimab/imdevimab, with a significant difference. The main limitations of the study were that it was conducted in non-immunocompromised patients and in patients not affected by the Omicron variant. Similar limitations were present in a study that aimed to evaluate the early treatment of COVID-19 with the SARS-CoV-2-neutralizing antibody sotrovimab [79]. Sotrovimab, also known as VIR-7831, is an engineered human monoclonal antibody acting against SARS-CoV-2 and other sarbecoviruses [80]. In a phase 3 study, 583 patients were enrolled from 37 sites and admitted to receive sotrovimab or a placebo. All patients had symptomatic COVID-19 infection and were not affected by the Omicron variant. The effectiveness of sotrovimab was documented by a study that demonstrated no progression of the disease in the treated group.
In a different study [81], 51 patients with a large prevalence of immunocompromised or transplanted subjects were successfully treated with sotrovimab. The median SARS-CoV-2 nucleoprotein (NP) viral load decreased from 7.1 log10 copies/mL before sotrovimab infusion to 5.1 log10 copies/mL 7 days post-infusion. No sotrovimab-resistant spike mutations were detected before infusion, but 53% of these patients had acquired sotrovimab-resistant mutations 7 to 21 days post-treatment. Previously, in vitro studies had shown that sotrovimab could trigger a resistant spike protein due to mutations at positions 340 and 337 [82]. All these data call for a close monitoring of patients treated with monoclonal antibodies for the possibility of the emergence of mutations and resistance to treatment.
A relevant study on the efficacy of monoclonal antibodies in KTx recipients was conducted in France [83]. The antibodies (either bamlanivimab alone, bamlanivimab/etesevimab or casirivimab + imdevimab) were administered to 80 KTx recipients affected by COVID-19 infection and compared with 155 controls. COVID-19-related hospitalization, 30-day admission to an intensive care unit (ICU) and death within 30 days were the endpoints. The early administration of monoclonal antibodies was beneficial, and the overall effects are presented in Table 4. These data demonstrate the efficacy of monoclonal antibodies administered to KTx recipients with a mild COVID-19 form and highlight that monoclonal antibody administration for SOT recipients with a weak vaccine response should be considered. In addition, the study confirmed the results of previous studies conducted on transplant patients [84,85,86,87] who used different associations of monoclonal antibodies.
Another study evaluated the association of monoclonal antibodies with remdesivir (RMD) as an early treatment in immune-compromised patients with unsatisfactory responses to vaccination [88]. The study was conducted either in transplant or non-transplanted patients. The conclusions of the study were that the association of RMD and mAbs did not cause relevant adverse effects while resulting in good outcomes for the disease.
A more recent study on the effectiveness of early administration of sotrovimab was conducted on 498 high-risk, non-immunocompromised and immunocompromised patients affected by the B.1.1.529 Omicron variant [89,90]. The study documented the efficacy of sotrovimab in preventing hospitalization and mortality in both immunocompromised and non-immunocompromised COVID-19 patients affected by the Omicron variant.
In an interesting study, sotrovimab administration in vaccinated and not vaccinated KTx recipients with SARS-CoV-2 infection due to the Delta and the Omicron BA.1 surges were evaluated [91]. Surprisingly, the outcomes were similar, despite Omicron’s mildness [92]. This fact could be ascribed either to Omicron’s immune evasion or to the inadequate immune response of KTx recipients.
Again for sotrovimab, the time of its administration after the onset of COVID-19 symptoms is critical. In a study on KTx recipients, Villanego et al. [93] compared 46 patients who received sotrovimab <5 days after the onset of symptoms with 36 patients who were treated >5 days. The results of the COVID-19 outcomes were critical, and are shown in Table 5.
In conclusion, according to the data available, early exposition to monoclonal antibodies indicates that the association of casirivimab and imdesimab is effective for treating the Delta variant. Sotrovimab had no efficacy against the Omicron variants BA.1 and BA.2. The association of tixagevimab and cilgavimab is under evaluation for the treatment of the Omicron variant BA.2.
The main characteristics of the monoclonal antibodies are shown in Table 6.

4. Direct-Acting Small-Molecule SARS-CoV-2 Antivirals

These compounds do not target the variable spike protein, but target either the viral RNA-dependent RNA polymerase (RdRp) or the viral main protease (Mpro) [94]. Our study will consider three products: Remdesivir, which was the first antiviral approved to treat COVID-19; Molnupivar, an inhibitor of the viral RdRp [95]; and Nirmatrelvir, which is an irreversible inhibitor of SARS-CoV-2 Mpro. This compound, when co-formulated with ritonavir, has been named Paxlovid [96]. In vitro and preliminary in vivo studies [86] documented that these compounds maintained their activity against all SARS-CoV-2 variants of concern (VOCs), Omicron included.
These findings were confirmed by other studies [97], confirming the effectiveness of antiviral therapies against highly transmittable variants of SARS-CoV-2.
The same antiviral treatments for COVID-19 are recommended by an update on the treatment for COVID-19 recently published by the British Medical Journal [98].
A wide, recent study [99] confirmed, based on 40,776 non-transplant patients affected by COVID-19, the effectiveness of early molnupivar or nirmatrelvir–ritonavir treatment in hospitalized patients affected by the Omicron variant BA.2. Though this was a retrospective study, according to the authors, it was the first study to document, from a large number of patients, the efficacy of the antivirals when administered within 2 days of admission to the hospital. An overview of these antiviral molecules is shown in Table 7.
The use of the antivirals in SOT patients and, in particular, in KTx recipients faces two major problems: the efficacy and safety in immunocompromised patients and the interactions with immunosuppressants.
A study [100] evaluated the early outcomes and renal functioning following antiviral treatments after KTx in patients affected by COVID-19. Ten of them received generic antivirals and eight received RDV. A proportion of 24% of the patients had acute kidney injury (AKI) at admission. Upon discharge, more patients treated with RDV exhibited a decrease in the eGFR, but the majority of patients returned to the baseline eGFR within one month after discharge. Due to the beneficial effects of antivirals, the authors concluded that these drugs appeared to be safe, without affecting renal function. A different study from Northern Italy [101] evaluated the effect of early RDV administration to prevent severe COVID-19 in SOT recipients. Seven out of twenty-four patients received pre-emptive RDV with a 3-day course. There was a significant reduction in the hospitalization rate in outpatient SOT recipients in contrast to the clinical worsening of the already-hospitalized SOT recipients. The authors highlighted the efficacy of RDV when administered as a pre-emptive drug before hospitalization. In a different retrospective cohort study [102], 38 hospitalized KTx recipients received 5-day RDV treatment versus 127 who received a standard-of-care treatment. Mortality was significantly reduced (39% vs. 83%) and the eGFR values improved at discharge.
In a different study [103], Radcliffe et al. evaluated the outpatient COVID-19 therapies in SOT recipients during the Omicron surge.
In this study, 122 patients with SOT and with COVID-19 diagnosed as outpatients were evaluated. Forty-nine patients received molnupivar and were compared with twenty-four patients who received the monoclonal antibody sotrovimab as outpatients and with forty-eight patients who received the standard-of-care therapy. The results of the study indicated that outpatient therapies are important in the management of mild to moderate COVID-19 in SOT recipients. According to the study, patients receiving molnupivar and sotrovimab had a reduced hospitalization rate and death, also when affected by the Omicron surge.
A similar study evaluated nirmatrelvir/ritonavir, sotrovimab and no SARS-CoV-2-specific treatment [104]. The results were, overall, the same and support the use of SARS-CoV-2-specific agents in the treatment of SOT recipients with COVID-19 infection.
Among the antiviral agents, Remdesivir and Nilmatrelvir/Ritonavir (Paxlovid) seem to be the most effective, but a major problem of RMD is the need for intravenous administration. A major problem with the use of Paxlovid is a severe drug–drug interaction due to the CYP3A inhibition by ritonavir, which can strongly increase the blood concentrations of calcineurin inhibitors (CNIs) [105]. Several studies documented the dangerous interaction between Paxlovid and CNIs. A study by Salerno et al. [106] documented the risk of reaching supratherapeutic TAC concentrations after restarting the intake of the drugs. According to the authors, physicians should carefully re-introduce CNIs after the completion of a Paxlovid course. Prikis et al. [107] reported a case study on Paxlovid and TAC interaction in a KTx recipient. Wang et al. [108] also reported a dangerous interaction between Paxlovid and TAC in four SARS-CoV-2-infected KTx recipients. A recommendation on how to manage the interactions was published by Lange et al. [109]. The study suggested interrupting CNIs or mammalian target of rapamycin (mTOR) inhibitors during the Paxlovid course and to start taking the immunosuppressants again 2–3 days after the end of the Paxlovid course. Monitoring the drug levels was also highly recommended. Other recommendations are to not use RMD or Paxlovid with an eGFR of less than 30 mL/min.
Paxlovid interacts with several immunosuppressants. A useful guideline on how to manage these drugs was described by the Guidelines of the French Society of Pharmacology and Therapeutics [110], as shown in Table 8.

5. Conclusions

The best prophylactic measure is to administer all transplant candidates the full cycle of the SARS-CoV-2 vaccine before transplantation.
A two-dose vaccination is not adequate to protect all SOT recipients, and a third or a fourth dose is recommended [53,54,55].
To evaluate the level of protection against severe COVID-19, the titer of anti-spike IgG may be useful. The absence of any detectable antibody indicates the lack of effective protection and indicates that SOT recipients need additional protection. Such patients need an additional booster vaccine dose, possibly against the dominant virus variant circulating [33,34].
The administration of the vaccine should be avoided within the first 3 months after transplantation or in patients recently treated with lymphocyte-depleting therapies [11]. In such cases, is better to postpone the vaccination.
Immunosuppressive drugs limit the immune response after vaccination, but the reduction of the immunosuppressant may cause rejection. However, in SOT recipients with the absence of antibodies in response to vaccination, over one year from transplantation and with stable graft function, a reduction of the immunosuppressive drugs may be evaluated under strict medical control, as recommended by several guidelines and position statements, including the ESOT recommendation [3].
Patients without an antibody response after 3–4 doses should be treated with prophylaxis with monoclonal antibodies [61,62]. The risk of potential resistance of new variants against the administered antibodies should be considered [74,75].
Antiviral molecules may be administered to patients with COVID-19 infection with concern regarding drug–drug interference with immunosuppressive drugs [105,107].
RMD should not be administered to patients with eGFR less than 30/mL/min [109].

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Conflicts of Interest

Maurizio Salvadori does not have any conflict of interest in relation to the manuscript.

References

  1. Fishman, J.A. The Immunocompromised Transplant Recipient and SARS-CoV-2 Infection. J. Am. Soc. Nephrol. 2020, 31, 1147–1149. [Google Scholar] [CrossRef]
  2. Fishman, J.A.; Roberts, M.B.; Zhang, E.W.; Kumar, D.; Hirsch, H.H.; Maggiore, U. Case 29-2020: A 66-Year-Old Man with Fever and Shortness of Breath after Liver Transplantation. N. Engl. J. Med. 2020, 383, 1168–1180. [Google Scholar] [CrossRef]
  3. Charmetant, X.; Thanaut, O.; Grossi, P.A.; Cillo, U.; Aguado, J.M.; Cervera, C.; Potena, L. Position Statement on Measures to Reduce the Risk of COVID-19 in Solid Organ Transplant Recipients by Use of Vaccination, Immune Testing, and Public Health Policies; European Society for Organ Transplantation: Padova, Italy, 2022. [Google Scholar]
  4. National Institutes of Health (NIH): COVID-19 Treatment Guidelines. Special Considerations in Solid Organ Transplant, Hematopoietic Stem Cell Transplant, and Cellular Therapy Candidates, Donors and Recipients. 2022. Available online: https://www.covid19treatmentguidelines.nih.gov/special-populations/transplant/ (accessed on 1 September 2022).
  5. National Health Service (NHS) Blood and Transplant Organ and Tissue Donation and Transplantation (OTDT) and British Toracic Society (BTS): Joint SARSCoV-2 Vaccination in Adult Solid Organ and Islet Transplant Wait-Listed Patients and Adult Living Donor Transplant Recipients. 2021. Available online: https://www.google.com.hk/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&cad=rja&uact=8&ved=2ahUKEwiK48n0t477AhUn-3MBHQLZCiMQFnoECBQQAQ&url=https%3A%2F%2Fbts.org.uk%2Fwp-content%2Fuploads%2F2021%2F01%2FDAT3911.pdf&usg=AOvVaw1pMQd7fSR1RCsJsECd9nhr (accessed on 1 September 2022).
  6. The Transplantation Society (TTS): Guidance on Coronavirus Disease 2019 (COVID-19) for Transplant Clinicians (2020). Available online: https://tts.org/tid-about/tid-officers-and-council?id=749 (accessed on 1 September 2022).
  7. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. C4591001 Clinical Trial Group. Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  8. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef]
  9. Hamm, S.R.; Møller, D.L.; Pérez-Alós, L.; Hansen, C.B.; Pries-Heje, M.M.; Heftdal, L.D.; Hasselbalch, R.B.; Fogh, K.; Madsen, J.R.; Almagro Armenteros, J.J.; et al. Decline in Antibody Concentration 6 Months After Two Doses of SARS-CoV-2 BNT162b2 Vaccine in Solid Organ Transplant Recipients and Healthy Controls. Front. Immunol. 2022, 13, 832501. [Google Scholar] [CrossRef]
  10. Verleye, A.; Wijtvliet, V.; Abrams, S.; Hellemans, R.; Bougrea, R.; Massart, A.; Pipeleers, L.; Wissing, K.M.; Ariën, K.K.; De Winter, B.Y.; et al. Seroconversion rate after primary vaccination with two doses of BNT162b2 versus mRNA-1273 in solid organ transplant recipients: A systematic review and meta-analysis. Nephrol. Dial. Transplant. 2022, 37, 1566–1575. [Google Scholar] [CrossRef]
  11. Liefeldt, L.; Glander, P.; Klotsche, J.; Straub-Hohenbleicher, H.; Budde, K.; Eberspächer, B.; Friedersdorff, F.; Halleck, F.; Hambach, P.; Hofmann, J.; et al. Predictors of Serological Response to SARS-CoV-2 Vaccination in Kidney Transplant Patients: Baseline Characteristics, Immunosuppression, and the Role of IMPDH Monitoring. J. Clin. Med. 2022, 11, 1697. [Google Scholar] [CrossRef]
  12. Balsby, D.; Nilsson, A.C.; Möller, S.; Lindvig, S.O.; Davidsen, J.R.; Abazi, R.; Poulsen, M.K.; Holden, I.K.; Justesen, U.S.; Bistrup, C.; et al. Determinants of Antibody Response to a Third SARS-CoV-2 mRNA Vaccine Dose in Solid Organ Transplant Recipients: Results from the Prospective Cohort Study COVAC-Tx. Vaccines 2022, 10, 565. [Google Scholar] [CrossRef]
  13. Hod, T.; Ben-David, A.; Olmer, L.; Scott, N.; Ghinea, R.; Mor, E.; Levy, I.; Indenbaum, V.; Lustig, Y.; Grossman, E.; et al. BNT162b2 Third Booster Dose Significantly Increases the Humoral Response Assessed by Both RBD IgG and Neutralizing Antibodies in Renal Transplant Recipients. Transpl. Int. 2022, 35, 10239. [Google Scholar] [CrossRef]
  14. Dimeglio, C.; Herin, F.; Martin-Blondel, G.; Miedougé, M.; Izopet, J. Antibody titers and protection against a SARS-CoV-2 infection. J. Infect. 2022, 84, 257–259. [Google Scholar] [CrossRef]
  15. Dimeglio, C.; Migueres, M.; Bouzid, N.; Chapuy-Regaud, S.; Gernigon, C.; Da-Silva, I.; Porcheron, M.; Martin-Blondel, G.; Herin, F.; Izopet, J. Antibody Titers and Protection against Omicron (BA.1 and BA.2) SARS-CoV-2 Infection. Vaccines 2022, 10, 1548. [Google Scholar] [CrossRef]
  16. Suntronwong, N.; Assawakosri, S.; Kanokudom, S.; Yorsaeng, R.; Auphimai, C.; Thongmee, T.; Vichaiwattana, P.; Duangchinda, T.; Chantima, W.; Pakchotanon, P.; et al. Strong Correlations between the Binding Antibodies against Wild-Type and Neutralizing Antibodies against Omicron BA.1 and BA.2 Variants of SARS-CoV-2 in Individuals Following Booster (Third-Dose) Vaccination. Diagnostics 2022, 12, 1781. [Google Scholar] [CrossRef]
  17. Manothummetha, K.; Chuleerarux, N.; Sanguankeo, A.; Kates, O.S.; Hirankarn, N.; Thongkam, A.; Dioverti-Prono, M.V.; Torvorapanit, P.; Langsiri, N.; Worasilchai, N.; et al. Immunogenicity and Risk Factors Associated with Poor Humoral Immune Response of SARS-CoV-2 Vaccines in Recipients of Solid Organ Transplant: A Systematic Review and Meta-Analysis. JAMA Netw. Open. 2022, 5, e226822. [Google Scholar] [CrossRef]
  18. Benotmane, I.; Gautier-Vargas, G.; Cognard, N.; Olagne, J.; Heibel, F.; Braun-Parvez, L.; Martzloff, J.; Perrin, P.; Moulin, B.; Fafi-Kremer, S.; et al. Low immunization rates among kidney transplant recipients who received 2 doses of the mRNA-1273 SARS-CoV-2 vaccine. Kidney Int. 2021, 99, 1498–1500. [Google Scholar] [CrossRef]
  19. Meyer, M.; Constancias, F.; Worth, C.; Meyer, A.; Muller, M.; Boussuge, A.; Kaltenbach, G.; Schmitt, E.; Chayer, S.; Velay, A.; et al. Humoral immune response after COVID-19 infection or BNT162b2 vaccine among older adults: Evolution over time and protective thresholds. Geroscience 2022, 44, 1229–1240. [Google Scholar] [CrossRef]
  20. Yang, H.S.; Costa, V.; Racine-Brzostek, S.E.; Acker, K.P.; Yee, J.; Chen, Z.; Karbaschi, M.; Zuk, R.; Rand, S.; Sukhu, A.; et al. Association of Age With SARS-CoV-2 Antibody Response. JAMA Netw. Open. 2021, 4, e214302. [Google Scholar] [CrossRef]
  21. Mazzoni, A.; Vanni, A.; Spinicci, M.; Lamacchia, G.; Kiros, S.T.; Rocca, A.; Capone, M.; Di Lauria, N.; Salvati, L.; Carnasciali, A.; et al. SARS-CoV-2 infection and vaccination trigger long-lived B and CD4+ T lymphocytes with implications for booster strategies. J. Clin. Investig. 2022, 132, e157990. [Google Scholar] [CrossRef]
  22. Dan, J.M.; Mateus, J.; Kato, Y.; Hastie, K.M.; Yu, E.D.; Faliti, C.E.; Grifoni, A.; Ramirez, S.I.; Haupt, S.; Frazier, A.; et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021, 371, eabf4063. [Google Scholar] [CrossRef]
  23. Mazzoni, A.; Di Lauria, N.; Maggi, L.; Salvati, L.; Vanni, A.; Capone, M.; Lamacchia, G.; Mantengoli, E.; Spinicci, M.; Zammarchi, L.; et al. First-dose mRNA vaccination is sufficient to reactivate immunological memory to SARS-CoV-2 in subjects who have recovered from COVID-19. J. Clin. Investig. 2021, 131, e149150. [Google Scholar] [CrossRef]
  24. Mazzoni, A.; Maggi, L.; Capone, M.; Spinicci, M.; Salvati, L.; Colao, M.G.; Vanni, A.; Kiros, S.T.; Mencarini, J.; Zammarchi, L.; et al. Cell-mediated and humoral adaptive immune responses to SARS-CoV-2 are lower in asymptomatic than symptomatic COVID-19 patients. Eur. J. Immunol. 2020, 50, 2013–2024. [Google Scholar] [CrossRef]
  25. Mazzoni, A.; Maggi, L.; Capone, M.; Vanni, A.; Spinicci, M.; Salvati, L.; Tekle Kiros, S.; Semeraro, R.; Pengue, L.; Colao, M.G.; et al. Heterogeneous magnitude of immunological memory to SARS-CoV-2 in recovered individuals. Clin. Transl. Immunol. 2021, 10, e1281. [Google Scholar] [CrossRef] [PubMed]
  26. Buehrle, D.J.; Sutton, R.R.; McCann, E.L.; Lucas, A.E. A Review of Treatment and Prevention of Coronavirus Disease 2019 among Solid Organ Transplant Recipients. Viruses 2021, 13, 1706. [Google Scholar] [CrossRef] [PubMed]
  27. Dęborska-Materkowska, D.; Kamińska, D. The Immunology of SARS-CoV-2 Infection and Vaccines in Solid Organ Transplant Recipients. Viruses 2021, 13, 1879. [Google Scholar] [CrossRef]
  28. Rozen-Zvi, B.; Yahav, D.; Agur, T.; Zingerman, B.; Ben-Zvi, H.; Atamna, A.; Tau, N.; Mashraki, T.; Nesher, E.; Rahamimov, R. Antibody response to SARS-CoV-2 mRNA vaccine among kidney transplant recipients: A prospective cohort study. Clin. Microbiol. Infect. 2021, 27, 1173.e1–1173.e4. [Google Scholar] [CrossRef] [PubMed]
  29. Marion, O.; Del Bello, A.; Abravanel, F.; Couat, C.; Faguer, S.; Esposito, L.; Hebral, A.L.; Izopet, J.; Kamar, N. Safety and Immunogenicity of Anti-SARS-CoV-2 Messenger RNA Vaccines in Recipients of Solid Organ Transplants. Ann. Int. Med. 2021, 174, 1336–1338. [Google Scholar] [CrossRef] [PubMed]
  30. Boyarsky, B.J.; Werbel, W.A.; Avery, R.K.; Tobian, A.A.R.; Massie, A.B.; Segev, D.L.; Garonzik-Wang, J.M. Antibody Response to 2-Dose SARS-CoV-2 mRNA Vaccine Series in Solid Organ Transplant Recipients. JAMA 2021, 325, 2204–2206. [Google Scholar] [CrossRef]
  31. Hall, V.G.; Ferreira, V.H.; Ku, T.; Ierullo, M.; Majchrzak-Kita, B.; Chaparro, C.; Selzner, N.; Schiff, J.; McDonald, M.; Tomlinson, G.; et al. Randomized Trial of a Third Dose of mRNA-1273 Vaccine in Transplant Recipients. N. Engl. J. Med. 2021, 385, 1244–1246. [Google Scholar] [CrossRef]
  32. Kamar, N.; Abravanel, F.; Marion, O.; Couat, C.; Izopet, J.; Del Bello, A. Three Doses of an mRNA COVID-19 Vaccine in Solid-Organ Transplant Recipients. N. Engl. J. Med. 2021, 385, 661–662. [Google Scholar] [CrossRef]
  33. Kamar, N.; Abravanel, F.; Marion, O.; Esposito, L.; Hebral, A.L.; Médrano, C.; Guitard, J.; Lavayssière, L.; Cointault, O.; Nogier, M.B.; et al. Anti-SARS-CoV-2 spike protein and neutralizing antibodies at 1 and 3 months after three doses of SARS-CoV-2 vaccine in a large cohort of solid organ transplant patients. Am. J. Transplant. 2022, 22, 1467–1474. [Google Scholar] [CrossRef]
  34. Kamar, N.; Abravanel, F.; Marion, O.; Romieu-Mourez, R.; Couat, C.; Del Bello, A.; Izopet, J. Assessment of 4 Doses of SARS-CoV-2 Messenger RNA-Based Vaccine in Recipients of a Solid Organ Transplant. JAMA Netw. Open. 2021, 4, e2136030. [Google Scholar] [CrossRef]
  35. Cristelli, M.P.; Nakamura, M.R.; Viana, L.A.; Tedesco-Silva, H.; Medina-Pestana, J. The Fourth Dose of CoronaVac Vaccine Results in a Small Increase of Seroconversion and Antibody Values Among Kidney Transplant Recipients. Transplantation, 2022; online ahead of print. [Google Scholar] [CrossRef]
  36. Mitchell, J.; Kim, J.; Alejo, J.L.; Chiang, T.P.; Karaba, A.H.; Blankson, J.N.; Aytenfisu, T.Y.; Chang, A.; Abedon, A.T.; Avery, R.K.; et al. Antibody Response to a Fourth Dose of SARS-CoV-2 Vaccine in Solid Organ Transplant Recipients: An Update. Transplantation 2022, 106, e338–e340. [Google Scholar] [CrossRef]
  37. Caillard, S.; Thaunat, O.; Benotmane, I.; Masset, C.; Blancho, G. Antibody Response to a Fourth Messenger RNA COVID-19 Vaccine Dose in Kidney Transplant Recipients: A Case Series. Ann. Int. Med. 2022, 175, 455–456. [Google Scholar] [CrossRef] [PubMed]
  38. Benotmane, I.; Bruel, T.; Planas, D.; Fafi-Kremer, S.; Schwartz, O.; Caillard, S. A fourth dose of the mRNA-1273 SARS-CoV-2 vaccine improves serum neutralization against the Delta variant in kidney transplant recipients. Kidney Int. 2022, 101, 1073–1076. [Google Scholar] [CrossRef] [PubMed]
  39. Masset, C.; Benotmane, I.; Dantal, J.; Garandeau, C.; Gauthier-Vargas, G.; Cantarovich, D.; Meurette, A.; Giral, M.; Caillard, S.; Blancho, G. A fourth SARS-CoV-2 mRNA vaccine in strictly seronegative kidney transplant recipients. Kidney Int. 2022, 10, 825–826. [Google Scholar] [CrossRef] [PubMed]
  40. Iketani, S.; Liu, L.; Guo, Y.; Liu, L.; Chan, J.F.; Huang, Y.; Wang, M.; Luo, Y.; Yu, J.; Chu, H.; et al. Antibody evasion properties of SARS-CoV-2 Omicron sublineages. Nature 2022, 604, 553–556. [Google Scholar] [CrossRef] [PubMed]
  41. Shen, X. Boosting immunity to Omicron. Nat. Med. 2022, 28, 445–446. [Google Scholar] [CrossRef]
  42. Pérez-Then, E.; Lucas, C.; Monteiro, V.S.; Miric, M.; Brache, V.; Cochon, L.; Vogels, C.B.F.; Malik, A.A.; De la Cruz, E.; Jorge, A.; et al. Neutralizing antibodies against the SARS-CoV-2 Delta and Omicron variants following heterologous CoronaVac plus BNT162b2 booster vaccination. Nat. Med. 2022, 28, 481–485. [Google Scholar] [CrossRef]
  43. Cheng, S.M.S.; Mok, C.K.P.; Leung, Y.W.Y.; Ng, S.S.; Chan, K.C.K.; Ko, F.W.; Chen, C.; Yiu, K.; Lam, B.H.S.; Lau, E.H.Y.; et al. Neutralizing antibodies against the SARS-CoV-2 Omicron variant BA.1 following homologous and heterologous CoronaVac or BNT162b2 vaccination. Nat. Med. 2022, 28, 486–489. [Google Scholar] [CrossRef]
  44. Gruell, H.; Vanshylla, K.; Tober-Lau, P.; Hillus, D.; Schommers, P.; Lehmann, C.; Kurth, F.; Sander, L.E.; Klein, F. mRNA booster immunization elicits potent neutralizing serum activity against the SARS-CoV-2 Omicron variant. Nat. Med. 2022, 28, 477–480. [Google Scholar] [CrossRef]
  45. VanBlargan, L.A.; Errico, J.M.; Halfmann, P.J.; Zost, S.J.; Crowe, J.E., Jr.; Purcell, L.A.; Kawaoka, Y.; Corti, D.; Fremont, D.H.; Diamond, M.S. An infectious SARS-CoV-2 B.1.1.529 Omicron virus escapes neutralization by therapeutic monoclonal antibodies. Nat. Med. 2022, 28, 490–495. [Google Scholar] [CrossRef] [PubMed]
  46. Bar-On, Y.M.; Goldberg, Y.; Mandel, M.; Bodenheimer, O.; Amir, O.; Freedman, L.; Alroy-Preis, S.; Ash, N.; Huppert, A.; Milo, R. Protection by a Fourth Dose of BNT162b2 against Omicron in Israel. N. Engl. J. Med. 2022, 386, 1712–1720. [Google Scholar] [CrossRef] [PubMed]
  47. Wohl, A.; Leibowitz, R. Protection by a Fourth Dose of BNT162b2 against Omicron in Israel. N. Engl. J. Med. 2022, 386, 2441. [Google Scholar]
  48. Zhang, J.; Chen, N.; Zhao, D.; Zhang, J.; Hu, Z.; Tao, Z. Clinical Characteristics of COVID-19 Patients Infected by the Omicron Variant of SARS-CoV-2. Front. Med. 2022, 9, 912367. [Google Scholar] [CrossRef] [PubMed]
  49. Al Jurdi, A.; Gassen, R.B.; Borges, T.J.; Lape, I.T.; Morena, L.; Efe, O.; Solhjou, Z.; El Fekih, R.; Deban, C.; Bohan, B.; et al. Suboptimal antibody response against SARS-CoV-2 Omicron variant after third dose of mRNA vaccine in kidney transplant recipients. Kidney Int. 2022, 101, 1282–1286. [Google Scholar] [CrossRef]
  50. Dejnirattisai, W.; Shaw, R.H.; Supasa, P.; Liu, C.; Stuart, A.S.; Pollard, A.J.; Liu, X.; Lambe, T.; Crook, D.; Stuart, D.I.; et al. Reduced neutralisation of SARS-CoV-2 omicron B.1.1.529 variant by post-immunisation serum. Lancet 2022, 399, 234–236. [Google Scholar] [CrossRef]
  51. Karaba, A.H.; Zhu, X.; Liang, T.; Wang, K.H.; Rittenhouse, A.G.; Akinde, O.; Eby, Y.; Ruff, J.E.; Blankson, J.N.; Abedon, A.T.; et al. A third dose of SARS-CoV-2 vaccine increases neutralizing antibodies against variants of concern in solid organ transplant recipients. Am. J. Transplant. 2022, 22, 1253–1260. [Google Scholar] [CrossRef]
  52. Karaba, A.H.; Johnston, T.S.; Aytenfisu, T.Y.; Akinde, O.; Eby, Y.; Ruff, J.E.; Abedon, A.T.; Alejo, J.L.; Blankson, J.N.; Cox, A.L.; et al. A Fourth Dose of COVID-19 Vaccine Does Not Induce Neutralization of the Omicron Variant Among Solid Organ Transplant Recipients with Suboptimal Vaccine Response. Transplantation 2022, 106, 1440–1444. [Google Scholar] [CrossRef]
  53. Sakuraba, A.; Luna, A.; Micic, D. A Systematic Review and Meta-Analysis of Serologic Response following Coronavirus Disease 2019 (COVID-19) Vaccination in Solid Organ Transplant Recipients. Viruses 2022, 14, 1822. [Google Scholar] [CrossRef]
  54. Massa, F.; Cremoni, M.; Gérard, A.; Grabsi, H.; Rogier, L.; Blois, M.; Couzin, C.; Hassen, N.B.; Rouleau, M.; Barbosa, S.; et al. Safety and cross-variant immunogenicity of a three-dose COVID-19 mRNA vaccine regimen in kidney transplant recipients. EBioMedicine 2021, 73, 103679. [Google Scholar] [CrossRef]
  55. Masset, C.; Kerleau, C.; Garandeau, C.; Ville, S.; Cantarovich, D.; Hourmant, M.; Kervella, D.; Houzet, A.; Guillot-Gueguen, C.; Guihard, I.; et al. A third injection of the BNT162b2 mRNA COVID-19 vaccine in kidney transplant recipients improves the humoral immune response. Kidney Int. 2021, 100, 1132–1135. [Google Scholar] [CrossRef] [PubMed]
  56. Baum, A.; Fulton, B.O.; Wloga, E.; Copin, R.; Pascal, K.E.; Russo, V.; Giordano, S.; Lanza, K.; Negron, N.; Ni, M.; et al. Antibody cocktail to SARS-CoV-2 spike protein prevents rapid mutational escape seen with individual antibodies. Science 2020, 369, 1014–1018. [Google Scholar] [CrossRef] [PubMed]
  57. Copin, R.; Baum, A.; Wloga, E.; Pascal, K.E.; Giordano, S.; Fulton, B.O.; Zhou, A.; Negron, N.; Lanza, K.; Chan, N.; et al. The monoclonal antibody combination REGEN-COV protects against SARS-CoV-2 mutational escape in preclinical and human studies. Cell 2021, 184, 3949–3961. [Google Scholar] [CrossRef] [PubMed]
  58. O’Brien, M.P.; Forleo-Neto, E.; Musser, B.J.; Isa, F.; Chan, K.C.; Sarkar, N.; Bar, K.J.; Barnabas, R.V.; Barouch, D.H.; Cohen, M.S.; et al. COVID-19 Phase 3 Prevention Trial Team. Subcutaneous REGEN-COV Antibody Combination to Prevent COVID-19. N. Engl. J. Med. 2021, 385, 1184–1195. [Google Scholar] [CrossRef]
  59. Merison, T.; Goldman, A.; Bomze, D. Subcutaneous REGEN-COV Antibody Combination to Prevent COVID-19. N. Engl. J. Med. 2021, 385, e70. [Google Scholar]
  60. Dougan, M.; Nirula, A.; Azizad, M.; Mocherla, B.; Gottlieb, R.L.; Chen, P.; Hebert, C.; Perry, R.; Boscia, J.; Heller, B.; et al. Bamlanivimab plus Etesevimab in Mild or Moderate COVID-19. N. Engl. J. Med. 2021, 385, 1382–1392. [Google Scholar] [CrossRef]
  61. National French Health Authority. RONAPREVE (Casirivimab-Imdevimab) (Prophylaxie Pré-Exposition de L’infection à SARS-CoV-2). Available online: https://www.has-sante.fr/jcms/p_3281544/fr/ronapreve-casirivimab-imdevimab-prophylaxie-pre-expositionj-de-l-infectiona-sars-cov-2 (accessed on 6 August 2022).
  62. Dimeglio, C.; Del Bello, A.; Chapuy-Regaud, S.; Esposito, L.; Danet, C.; Couat, C.; Izopet, J.; Kamar, N. Casirivimab-imdevimab to Prevent SARS-CoV-2 Infections in Solid Organ Transplant Recipients. Transplantation 2022, 106, e275–e276. [Google Scholar] [CrossRef]
  63. Ducloux, D.; Courivaud, C. REGEN-Cov antibody combination to prevent COVID-19 infection in kidney transplant recipient without detectable antibody response to optimal vaccine scheme. Kidney Int. 2022, 101, 645–646. [Google Scholar] [CrossRef]
  64. Kamar, N.; Gouin, A.; Izopet, J. Omicron breakthrough infection in a kidney-transplant patient given pre-exposition casirivimab and imdevimab monoclonal antibodies. Transpl. Infect. Dis. 2022, 24, e13803. [Google Scholar] [CrossRef]
  65. Planas, D.; Saunders, N.; Maes, P.; Guivel-Benhassine, F.; Planchais, C.; Buchrieser, J.; Bolland, W.H.; Porrot, F.; Staropoli, I.; Lemoine, F.; et al. Considerable escape of SARS-CoV-2 Omicron to antibody neutralization. Nature 2022, 602, 671–675. [Google Scholar] [CrossRef]
  66. Taylor, P.C.; Adams, A.C.; Hufford, M.M.; de la Torre, I.; Winthrop, K.; Gottlieb, R.L. Neutralizing monoclonal antibodies for treatment of COVID-19. Nat. Rev. Immunol. 2021, 21, 382–393. [Google Scholar] [CrossRef] [PubMed]
  67. Cao, Y.; Wang, J.; Jian, F.; Xiao, T.; Song, W.; Yisimayi, A.; Huang, W.; Li, Q.; Wang, P.; An, R.; et al. Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies. Nature 2022, 602, 657–663. [Google Scholar] [CrossRef] [PubMed]
  68. Cameroni, E.; Bowen, J.E.; Rosen, L.E.; Saliba, C.; Zepeda, S.K.; Culap, K.; Pinto, D.; VanBlargan, L.A.; De Marco, A.; di Iulio, J.; et al. Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift. Nature 2022, 602, 664–670. [Google Scholar] [CrossRef]
  69. Loo, Y.M.; McTamney, P.M.; Arends, R.H.; Abram, M.E.; Aksyuk, A.A.; Diallo, S.; Flores, D.J.; Kelly, E.J.; Ren, K.; Roque, R.; et al. The SARS-CoV-2 monoclonal antibody combination, AZD7442, is protective in nonhuman primates and has an extended half-life in humans. Sci. Transl. Med. 2022, 14, eabl8124. [Google Scholar] [CrossRef] [PubMed]
  70. Zost, S.J.; Gilchuk, P.; Case, J.B.; Binshtein, E.; Chen, R.E.; Nkolola, J.P.; Schäfer, A.; Reidy, J.X.; Trivette, A.; Nargi, R.S.; et al. Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature 2020, 584, 443–449. [Google Scholar] [CrossRef] [PubMed]
  71. Dong, J.; Zost, S.J.; Greaney, A.J.; Starr, T.N.; Dingens, A.S.; Chen, E.C.; Chen, R.E.; Case, J.B.; Sutton, R.E.; Gilchuk, P.; et al. Genetic and structural basis for SARS-CoV-2 variant neutralization by a two-antibody cocktail. Nat. Microbiol. 2021, 6, 1233–1244. [Google Scholar] [CrossRef] [PubMed]
  72. Levin, M.J.; Ustianowski, A.; De Wit, S.; Launay, O.; Avila, M.; Templeton, A.; Yuan, Y.; Seegobin, S.; Ellery, A.; Levinson, D.J.; et al. Intramuscular AZD7442 (Tixagevimab-Cilgavimab) for Prevention of COVID-19. N. Engl. J. Med. 2022, 386, 2188–2200. [Google Scholar] [CrossRef]
  73. Bertrand, D.; Laurent, C.; Lemée, V.; Lebourg, L.; Hanoy, M.; Le Roy, F.; Nezam, D.; Pruteanu, D.; Grange, S.; de Nattes, T.; et al. Efficacy of anti-SARS-CoV-2 monoclonal antibody prophylaxis and vaccination on the Omicron variant of COVID-19 in kidney transplant recipients. Kidney Int. 2022, 102, 440–442. [Google Scholar] [CrossRef]
  74. Benotmane, I.; Velay, A.; Gautier-Vargas, G.; Olagne, J.; Thaunat, O.; Fafi-Kremer, S.; Caillard, S. Pre-exposure prophylaxis with 300 mg Evusheld elicits limited neutralizing activity against the Omicron variant. Kidney Int. 2022, 102, 442–444. [Google Scholar] [CrossRef]
  75. US Food & Drug Administration. FDA Authorizes Revisions to Evusheld Dosing. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/fda-authorizes-revisions-evusheld-dosing (accessed on 24 May 2002).
  76. Mazzotta, V.; Cozzi-Lepri, A.; Colavita, F.; Lanini, S.; Rosati, S.; Lalle, E.; Mastrorosa, I.; Cimaglia, C.; Vergori, A.; Bevilacqua, N.; et al. Emulation of a Target Trial from Observational Data to Compare Effectiveness of Casirivimab/Imdevimab and Bamlanivimab/Etesevimab for Early Treatment of Non-Hospitalized Patients With COVID-19. Front. Immunol. 2022, 13, 868020. [Google Scholar] [CrossRef]
  77. Gottlieb, R.L.; Nirula, A.; Chen, P.; Boscia, J.; Heller, B.; Morris, J.; Huhn, G.; Cardona, J.; Mocherla, B.; Stosor, V.; et al. Effect of Bamlanivimab as Monotherapy or in Combination With Etesevimab on Viral Load in Patients With Mild to Moderate COVID-19: A Randomized Clinical Trial. JAMA 2021, 325, 632–644. [Google Scholar] [CrossRef] [PubMed]
  78. Dougan, M.; Azizad, M.; Mocherla, B.; Gottlieb, R.L.; Chen, P.; Hebert, C.; Perry, R.; Boscia, J.; Heller, B.; Morris, J.; et al. A Randomized, Placebo-Controlled Clinical Trial of Bamlanivimab and Etesevimab Together in High-Risk Ambulatory Patients With COVID-19 and Validation of the Prognostic Value of Persistently High Viral Load. Clin. Infect. Dis. 2022, 75, e440–e449. [Google Scholar] [CrossRef] [PubMed]
  79. Gupta, A.; Gonzalez-Rojas, Y.; Juarez, E.; Crespo Casal, M.; Moya, J.; Falci, D.R.; Sarkis, E.; Solis, J.; Zheng, H.; Scott, N.; et al. Early Treatment for COVID-19 with SARS-CoV-2 Neutralizing Antibody Sotrovimab. N. Engl. J. Med. 2021, 385, 1941–1950. [Google Scholar] [CrossRef]
  80. Pinto, D.; Park, Y.J.; Beltramello, M.; Walls, A.C.; Tortorici, M.A.; Bianchi, S.; Jaconi, S.; Culap, K.; Zatta, F.; De Marco, A.; et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature 2020, 583, 290–295. [Google Scholar] [CrossRef] [PubMed]
  81. Vellas, C.; Trémeaux, P.; Del Bello, A.; Latour, J.; Jeanne, N.; Ranger, N.; Danet, C.; Martin-Blondel, G.; Delobel, P.; Kamar, N.; et al. Resistance mutations in SARS-CoV-2 omicron variant in patients treated with sotrovimab. Clin. Microbiol. Infect. 2022, 28, 1297–1299. [Google Scholar] [CrossRef]
  82. Rockett, R.; Basile, K.; Maddocks, S.; Fong, W.; Agius, J.E.; Johnson-Mackinnon, J.; Arnott, A.; Chandra, S.; Gall, M.; Draper, J.; et al. Resistance Mutations in SARS-CoV-2 Delta Variant after Sotrovimab Use. N. Engl. J. Med. 2022, 386, 1477–1479. [Google Scholar] [CrossRef]
  83. Gueguen, J.; Colosio, C.; Del Bello, A.; Scemla, A.; N’Guyen, Y.; Rouzaud, C.; Carvalho-Schneider, C.; Gautier Vargas, G.; Tremolières, P.; Eddine, A.J.; et al. Early Administration of Anti-SARS-CoV-2 Monoclonal Antibodies Prevents Severe COVID-19 in Kidney Transplant Patients. Kidney Int. Rep. 2022, 7, 1241–1247. [Google Scholar] [CrossRef]
  84. Dhand, A.; Lobo, S.A.; Wolfe, K.; Feola, N.; Lee, L.; Nog, R.; Chen, D.; Glicklich, D.; Diflo, T.; Nabors, C. Casirivimab-imdevimab for Treatment of COVID-19 in Solid Organ Transplant Recipients: An Early Experience. Transplantation 2021, 105, e68–e69. [Google Scholar] [CrossRef]
  85. Dhand, A.; Lobo, S.A.; Wolfe, K.; Feola, N.; Nabors, C. Bamlanivimab for treatment of COVID-19 in solid organ transplant recipients: Early single-center experience. Clin. Transplant. 2021, 35, e14245. [Google Scholar] [CrossRef]
  86. Kutzler, H.L.; Kuzaro, H.A.; Serrano, O.K.; Feingold, A.; Morgan, G.; Cheema, F. Initial experience of bamlanivimab monotherapy use in solid organ transplant recipients. Transpl. Infect. Dis. 2021, 23, e13662. [Google Scholar] [CrossRef]
  87. Vellas, C.; Del Bello, A.; Debard, A.; Steinmeyer, Z.; Tribaudeau, L.; Ranger, N.; Jeanne, N.; Martin-Blondel, G.; Delobel, P.; Kamar, N.; et al. Influence of treatment with neutralizing monoclonal antibodies on the SARS-CoV-2 nasopharyngeal load and quasispecies. Clin. Microbiol. Infect. 2022, 28, 139.e5–139.e8. [Google Scholar] [CrossRef] [PubMed]
  88. Biscarini, S.; Villa, S.; Genovese, C.; Tomasello, M.; Tonizzo, A.; Fava, M.; Iannotti, N.; Bolis, M.; Mariani, B.; Valzano, A.G.; et al. Safety Profile and Outcomes of Early COVID-19 Treatments in Immunocompromised Patients: A Single-Centre Cohort Study. Biomedicines 2022, 10, 2002. [Google Scholar] [CrossRef] [PubMed]
  89. Birk, N.K.; Jain, S.; Massoud, L.; Ramesh, D.; Monday, L.; Muma, B.; Williams, J.; Alangaden, G.; Ramesh, M. Real-world Experience of Sotrovimab in High-risk, Immunocompromised COVID-19 Patients. Open Forum. Infect. Dis. 2022, 9, ofac282. [Google Scholar] [CrossRef] [PubMed]
  90. Centers for Disease Control and Prevention. COVID Data Trecker. 2022. Available online: https://covid.cdc.gov/covid-data-tracker (accessed on 21 July 2022).
  91. Vathsala, A.; Somani, J.; Ross D’Costa, M.; Lum, L.; Wong, E.T.; Sran, H.K. Comparative outcomes after early Sotrovimab administration in vaccinated and unvaccinated kidney transplant recipients with SARS-CoV-2 infection during the Delta and Omicron BA.1 surges. Transpl. Infect. Dis. 2022, e13930. [Google Scholar] [CrossRef]
  92. Lauring, A.S.; Tenforde, M.W.; Chappell, J.D.; Gaglani, M.; Ginde, A.A.; McNeal, T.; Ghamande, S.; Douin, D.J.; Talbot, H.K.; Casey, J.D.; et al. Influenza and Other Viruses in the Acutely Ill (IVY) Network. Clinical severity of, and effectiveness of mRNA vaccines against, COVID-19 from omicron, delta, and alpha SARS-CoV-2 variants in the United States: Prospective observational study. BMJ 2022, 376, e069761. [Google Scholar] [CrossRef]
  93. Villanego, F.; Mazuecos, A.; Cubillo, B.; Merino, M.J.; Poveda, I.; Saura, I.M.; Segurado, Ó.; Cruzado, L.; Eady, M.; Zárraga, S.; et al. Treatment with sotrovimab for SARS-CoV-2 infection in a cohort of high-risk kidney transplant recipients. Clin. Kidney J. 2022, 15, 1847–1855. [Google Scholar] [CrossRef]
  94. Vangeel, L.; Chiu, W.; De Jonghe, S.; Maes, P.; Slechten, B.; Raymenants, J.; André, E.; Leyssen, P.; Neyts, J.; Jochmans, D. Remdesivir, Molnupiravir and Nirmatrelvir remain active against SARS-CoV-2 Omicron and other variants of concern. Antivir. Res. 2022, 198, 105252. [Google Scholar] [CrossRef]
  95. Painter, W.P.; Holman, W.; Bush, J.A.; Almazedi, F.; Malik, H.; Eraut, N.C.J.E.; Morin, M.J.; Szewczyk, L.J.; Painter, G.R. Human Safety, Tolerability, and Pharmacokinetics of Molnupiravir, a Novel Broad-Spectrum Oral Antiviral Agent with Activity Against SARS-CoV-2. Antimicrob. Agents Chemother. 2021, 65, e02428-20. [Google Scholar] [CrossRef]
  96. 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 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science 2021, 374, 1586–1593. [Google Scholar] [CrossRef]
  97. Schöning, V.; Kern, C.; Chaccour, C.; Hammann, F. Effectiveness of Antiviral Therapy in Highly-Transmissible Variants of SARS-CoV-2: A Modeling and Simulation Study. Front. Pharmacol. 2022, 13, 816429. [Google Scholar] [CrossRef]
  98. WHO. Update to living WHO guideline on drugs for COVID-19. BMJ 2022, 378, o1713. [Google Scholar]
  99. 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; in press. [Google Scholar] [CrossRef]
  100. Elec, A.D.; Oltean, M.; Goldis, P.; Cismaru, C.; Lupse, M.; Muntean, A.; Elec, F.I. COVID-19 after kidney transplantation: Early outcomes and renal function following antiviral treatment. Int. J. Infect. Dis. 2021, 104, 426–432. [Google Scholar] [CrossRef] [PubMed]
  101. Colaneri, M.; Amarasinghe, N.; Rezzonico, L.; Pieri, T.C.; Segalini, E.; Sambo, M.; Roda, S.; Meloni, F.; Gregorini, M.; Rampino, T.; et al. Early remdesivir to prevent severe COVID-19 in recipients of solid organ transplant: A real-life study from Northern Italy. Int. J. Infect. Dis. 2022, 121, 157–160. [Google Scholar] [CrossRef] [PubMed]
  102. Elec, F.; Magnusson, J.; Elec, A.; Muntean, A.; Antal, O.; Moisoiu, T.; Cismaru, C.; Lupse, M.; Oltean, M. COVID-19 and kidney transplantation: The impact of remdesivir on renal function and outcome—A retrospective cohort study. Int. J. Infect. Dis. 2022, 118, 247–253. [Google Scholar] [CrossRef]
  103. Radcliffe, C.; Palacios, C.F.; Azar, M.M.; Cohen, E.; Malinis, M. Real-world experience with available, outpatient COVID-19 therapies in solid organ transplant recipients during the omicron surge. Am. J. Transplant. 2022; online ahead of print. [Google Scholar] [CrossRef]
  104. Hedvat, J.; Lange, N.W.; Salerno, D.M.; DeFilippis, E.M.; Kovac, D.; Corbo, H.; Chen, J.K.; Choe, J.Y.; Lee, J.H.; Anamisis, A.; et al. COVID-19 therapeutics and outcomes among solid organ transplant recipients during the Omicron BA.1 era. Am. J. Transplant. 2022; online ahead of print. [Google Scholar] [CrossRef]
  105. Gandolfini, I.; Crespo, M.; Hellemans, R.; Maggiore, U.; Mariat, C.; Mjoen, G.; Oniscu, G.C.; Peruzzi, L.; Sever, M.S.; Watschinger, B.; et al. Issues regarding COVID-19 in kidney transplantation in the era of the omicron variant: A commentary by the era descartes working group. Nephrol. Dial. Transplant. 2022, 37, 1824–1829. [Google Scholar] [CrossRef]
  106. Salerno, D.M.; Jennings, D.L.; Lange, N.W.; Kovac, D.B.; Shertel, T.; Chen, J.K.; Hedvat, J.; Scheffert, J.; Brown, R.S., Jr.; Pereira, M.R. Early clinical experience with nirmatrelvir/ritonavir for the treatment of COVID-19 in solid organ transplant recipients. Am. J. Transplant. 2022, 22, 2083–2088. [Google Scholar] [CrossRef]
  107. Prikis, M.; Cameron, A. Paxlovid (Nirmatelvir/Ritonavir) and Tacrolimus Drug-Drug Interaction in a Kidney Transplant Patient with SARS-2-CoV infection: A Case Report. Transplant. Proc. 2022, 54, 1557–1560. [Google Scholar] [CrossRef]
  108. Wang, A.X.; Koff, A.; Hao, D.; Tuznik, N.M.; Huang, Y. Effect of nirmatrelvir/ritonavir on calcineurin inhibitor levels: Early experience in four SARS-CoV-2 infected kidney transplant recipients. Am. J. Transplant. 2022, 22, 2117–2119. [Google Scholar] [CrossRef] [PubMed]
  109. Lange, N.W.; Salerno, D.M.; Jennings, D.L.; Choe, J.; Hedvat, J.; Kovac, D.B.; Scheffert, J.; Shertel, T.; Ratner, L.E.; Brown, R.S., Jr.; et al. Nirmatrelvir/ritonavir use: Managing clinically significant drug-drug interactions with transplant immunosuppressants. Am. J. Transplant. 2022, 22, 1925–1926. [Google Scholar] [CrossRef] [PubMed]
  110. Lemaitre, F.; Grégoire, M.; Monchaud, C.; Bouchet, S.; Saint-Salvi, B.; Polard, E.; SFPT Therapeutic Drug Monitoring and Treatment Personalization Group (STP-PT) of the French Society of Pharmacology and Therapeutics (SFPT); French Pharmacovigilance Network (CRPV); ANRS-MIE AC-43 Clinical Pharmacology Committee, Joint Working Group; SFPT Therapeutic Drug Monitoring and Treatment Personalization Group (STP-PT) of the French Society of Pharmacology and Therapeutics (SFPT); et al. Management of drug-drug interactions with nirmatrelvir/ritonavir in patients treated for COVID-19: Guidelines from the French Society of Pharmacology and Therapeutics (SFPT). Therapie 2022, 77, 509–521. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Relative seroconversion rate of BNT162b2 compared with mRNA-1273 = 79.5%.
Figure 1. Relative seroconversion rate of BNT162b2 compared with mRNA-1273 = 79.5%.
Transplantology 03 00030 g001
Figure 2. Anti-SARS-CoV-2 antibody titers according to the type of transplantation.
Figure 2. Anti-SARS-CoV-2 antibody titers according to the type of transplantation.
Transplantology 03 00030 g002
Figure 3. Prevalence of anti-SARS-CoV-2 antibodies after different doses of the mRNABNT162b2 vaccine.
Figure 3. Prevalence of anti-SARS-CoV-2 antibodies after different doses of the mRNABNT162b2 vaccine.
Transplantology 03 00030 g003
Figure 4. Proportion of KTx recipients with a positive neutralization response before and after the third vaccine dose.
Figure 4. Proportion of KTx recipients with a positive neutralization response before and after the third vaccine dose.
Transplantology 03 00030 g004
Table 1. (a) Multivariate analysis of factors associated with the serological response to vaccination after kidney transplantation according to the study of Liefeldt et al. [11]; (b) multivariate analysis of factors associated with the serological response to vaccination after kidney transplantation in MPA-treated patients after KTx transplantation.
Table 1. (a) Multivariate analysis of factors associated with the serological response to vaccination after kidney transplantation according to the study of Liefeldt et al. [11]; (b) multivariate analysis of factors associated with the serological response to vaccination after kidney transplantation in MPA-treated patients after KTx transplantation.
Multivariate Analysis
FactorsOR95% CIp Value
(a)
Age at 2nd vaccination0.980.96; 1.000.039
Time after kidney TX1.061.02; 1.100.001
TAC + MPA + Steroid0.150.08; 0.28<0.001
CyA + MPA + Steroid0.510.27; 0.960.038
TAC/CyA + Steroid4.111.71; 9.900.002
eGFR at vaccination1.031.02; 1.04<0.001
Lymphocyte count at vaccination1.121.06; 1.18<0.001
CNI trough levels at vaccination0.940.90; 1.000.036
(b)
Female0.410.20; 0.830.013
Age at 2nd vaccination0.960.94; 0.990.002
Time after kidney TX1.071.01; 1.130.031
TAC MPA Steroids0.430.20; 0.950.036
CyA MPA Steroids1.300.57–2.980.534
eGFR at vaccination1.031.01; 1.050.014
Lymphocyte count at vaccination1.060.99; 1.140.077
CNI trough levels at vaccination0.920.84; 1.000.040
MPA dose at vaccination0.720.59; 0.870.001
IMPDH activity0.340.25; 0.46<0.001
Abbreviations: OR, odds ratio; CI, confidence interval; TAC, Tacrolimus; MPA, Mycophenolate acid; CyA, Cyclosporine; eGFR, estimated glomerular filtration rate; CNI, calcineurine; IMPDH, inosine monophosphate dehydrogenase.
Table 2. Univariate analysis of immune status before the third vaccine vs. post-third-vaccine in RTR according to the study of Hod et al. [13].
Table 2. Univariate analysis of immune status before the third vaccine vs. post-third-vaccine in RTR according to the study of Hod et al. [13].
Before 3rd VaccinePost-3rd-Vaccinep Value
All cohort
IgG-RBD GMT (95% CI)0.79 (0.65–0.96)3.08 (2.76–3.45)<0.0001
NA GMT (95% CI)17.46 (12.38–24.62)362.2 (220.7–594.6)<0.0001
Positive responders
N (%)32 (32.3)85 (85.9)<0.0001
=35IgG-RBD GMT (95% CI)2.53 (2.07–3.11)3.57 (3.28–3.88)<0.0001
NA GMT (95% CI)89.12 (53.03–149.8)689.9 (456.3–1043)<0.0001
Negative responders
N (%)67 (67.7)14 (14.14)<0.0001
IgG-RBD GMT (95% CI)0.45 (0.39–052)1.28 (0.87–1.86)<0.0001
NA GMT (95% CI)8.01 (5.92–10.84)7.25 (2.42–21.71)0.85
Abbreviations: CI, confidence interval; GMT, geometric mean titer; NA, neutralizing antibodies; RBD, receptor binding domain.
Table 3. Serological response after different vaccine doses in KTx recipients according to the studies of Sakuraba et al., Massa et al. and Masset et al. [50,51,52].
Table 3. Serological response after different vaccine doses in KTx recipients according to the studies of Sakuraba et al., Massa et al. and Masset et al. [50,51,52].
KidneyStatisticsTransplantControl
A Serological response compared with controls after one dose of vaccine
KidneyOdds ratioLower limitUpper limitp Value
0.00630.00250.0160<0.00120/29989/96
B Serological response compared with controls after two doses of vaccine
0.00630.00250.0159<0.001208/655271/273
C Serological response compared with controls after three doses of vaccine
0.6690.6010.732<0.001132/197
Table 4. Outcomes at 1 month of kidney transplant recipients treated or not with anti-SARS-CoV-2 monoclonal antibodies [80].
Table 4. Outcomes at 1 month of kidney transplant recipients treated or not with anti-SARS-CoV-2 monoclonal antibodies [80].
OutcomesMoAb GroupControl Groupp Value
Severe COVID-19, n (%)3 (3.8)30 (19.4)0.001
Admission to ICU, n (%)2 (2.5)24 (15.5)0.002
Need for mechanical ventilation, n (%)0 (0.0)18 (11.6)<0.001
Death, n (%)1 (1.25)18 (11.6)0.005
Abbreviation: ICU, intensive care unit.
Table 5. Outcome comparison between kidney transplant recipients treated with sotrovimab early in the onset of symptoms and those treated late [90].
Table 5. Outcome comparison between kidney transplant recipients treated with sotrovimab early in the onset of symptoms and those treated late [90].
Variable<5 Days
(n = 46)
5 Days
(n = 36)
p-Value
Ventilator support, n (%)1 (2.2)13 (36.1) <0.001
ICU admission, n (%)1 (2.2)9 (25)0.002
Death, n (%)1 (2.2)6 (16.7)0.02
Table 6. Characteristics of monoclonal antibodies for the treatment of COVID-19. 2022.
Table 6. Characteristics of monoclonal antibodies for the treatment of COVID-19. 2022.
Class and AgentDosingPlace in TherapyDrug-Drug InteractionsAdverse EffectsSpecial Consideration for TX Recipients
Monoclonal antibodiesBemlanivimab 700 mg plus etesevimab 1400 mg IV as a single doseNIH Guidelines
# Casirivimab plus imdevimab or sotrovimab recommended for outpatients with mild to moderate COVID-19
# Recommend against use of bamlanivimab plus etesevimab
# Recommend against use in hospitalized patients outside a clinical trial
May decrease the effects of COVID-19 vaccination; postpone administration of COVID-19 vaccine until at least 90 days after treatmentHypersensitivity
Pruritis
Injection site reactions
Fever
# Authorized under FDA EUA
# Administer in healthcare settings
# Monitor patient for at least 1 h post-administration
# Use of bamlanivimab alone and bamlanivimab plus etesevimab is not recommended due to decreased susceptibility of SARS-CoV-2 variants
Casirivimab 600 mg plus imdevimab 600 mg IV/SQ as a single doseWHO Guidelines
# Class not addressed
Sotrovimab 500 mg IV as a single doseIDSA Guidelines
# Recommended for ambulatory patients with mild to moderate COVID-19 at high risk
Table 7. Overview of SARS-CoV-2 antivirals.
Table 7. Overview of SARS-CoV-2 antivirals.
Nirmatrelvir/Ritonavir (Paxlovid)Molnupiravir (Lagevrio)Remdesivir (Veklury)
PopulationAge > 12 years and >40 kg. Mild to moderate COVID-19 and high risk of progression to hospitalization or death. EMA and FDA approvedAge > 18 years. Mild to moderate COVID-19 and high risk of progression or death. EMA and FDA approvedAge > 12 years and >40 kg requiring supplemental oxygen. EMA and FDA approved (in and out of hospital)
Efficacy (high-risk population)NNT (number needed to treat) = 18 (all cause hospitalization or death)NNT = 35 (all cause hospitalization or death) when administered within the first 5 days of symptom onsetNNT = 22 (all cause hospitalization or death)
Drug interactionsSerious concern (ritonavir strongly inhibits CYP3A4)NegligibleMonitor when co-administered with strong CYP3A4 inducers/inhibitors
Common side-effectsDysgeusia and diarrheaDiarrhea, nausea, anemia and potentially mutagenicityBradycardia, drug-induced liver injury and acute kidney injury
Renal/hepatic impairmentDose adjustment with moderate renal impairmentNo dose adjustment requiredNot recommended if eGFR < 30 mL/min
PregnancyContraindicatedContraindicatedReassuring data
Activity vs. variantsAll known variantsAll known variantsAll known variants
Table 8. Guidelines of the French Society of Pharmacology and Therapeutics (SFPT).
Table 8. Guidelines of the French Society of Pharmacology and Therapeutics (SFPT).
Immunosuppressive DrugNature and Magnitude of the EffectTherapeutic Strategy
TacrolimusIncrease in tacrolimus exposure by 40-foldAdminister 1/8 of the usual daily dose (DD) on day 1, then stop. Administer 1/2 of the DD on day 6, then 3/4 on day 7 and restart usual DD on day 8.
Alternative for low immunological risk: start nirmatrelvir/ritonavir 12 h after the last intake of tacrolimus and restart tacrolimus at usual DD 24 h after the last antiviral dose. TDM if possible
CyclosporineIncrease in cyclosporine exposure by 8-foldAdminister 1/5 of the usual DD every day of nirmatrelvir/ritonavir treatment. Administer 1/2 of the DD on day 6, then 3/4 on day 7 and restart usual DD on day 8. TDM if possible
Everolimus/SirolimusIncrease in everolimus and sirolimus exposure by 15- and 11-fold, respectivelyAdminister 1/8 of the usual DD on day 1, day 3 and day 5. Usual DD can be restarted on day-7. TDM if possible
Mycophenolate mofetilWeak interaction expected. Possible decrease in mycophenolic acid exposureThe dosage can be maintained
Abbreviation: TDM, therapeutic dose monitoring.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Salvadori, M. What Is New in Prophylaxis and Treatment of COVID-19 in Renal Transplant Patients? A Report from an ESOT Meeting on the Topic. Transplantology 2022, 3, 288-306. https://doi.org/10.3390/transplantology3040030

AMA Style

Salvadori M. What Is New in Prophylaxis and Treatment of COVID-19 in Renal Transplant Patients? A Report from an ESOT Meeting on the Topic. Transplantology. 2022; 3(4):288-306. https://doi.org/10.3390/transplantology3040030

Chicago/Turabian Style

Salvadori, Maurizio. 2022. "What Is New in Prophylaxis and Treatment of COVID-19 in Renal Transplant Patients? A Report from an ESOT Meeting on the Topic" Transplantology 3, no. 4: 288-306. https://doi.org/10.3390/transplantology3040030

Article Metrics

Back to TopTop