Next Article in Journal
A Novel Recombinant Chicken-Derived H6N8 Subtype Avian Influenza Virus Caused Disease in Chickens and Mice
Previous Article in Journal
Arf GTPases Define BST-2-Independent Pathways for HIV-1 Assembly and Release
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Respiratory Viral Infection Prophylaxis and Treatment in the Transplant Population

1
Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, La Jolla, CA 92093, USA
2
Division of Infectious Diseases, Rady Children’s Hospital, San Diego, CA 92123, USA
3
Department of Pharmacy, University of California San Diego Health, La Jolla, CA 92093, USA
*
Author to whom correspondence should be addressed.
Viruses 2026, 18(1), 8; https://doi.org/10.3390/v18010008 (registering DOI)
Submission received: 14 November 2025 / Revised: 13 December 2025 / Accepted: 16 December 2025 / Published: 20 December 2025
(This article belongs to the Special Issue Opportunistic Viral Infections, 3rd Edition)

Abstract

Transplant patients experience high morbidity and mortality caused by respiratory viral infections (RVIs). In the past decade, numerous methods of prophylaxis and treatment have rapidly developed and continue to expand, with dozens of novel agents in preclinical and clinical trials. This includes recent scientific breakthroughs in virus structure, which have enabled the creation of respiratory syncytial virus (RSV) vaccines. While new vaccines, antivirals, monoclonal antibodies, and non-vaccine agents are becoming more available, their utility and safety in the transplant populations are often uncertain. This review summarizes the current landscape of RVIs in the transplant population, including approaches to pre- and post-exposure prophylaxis and treatment. We discuss the data behind vaccine timing, safety, and efficacy and current pre- and post-transplant recommendations, with a particular focus on influenza, SARS-CoV-2, and RSV. We also examine the potential benefits of antivirals, monoclonal antibodies, and novel agents used as prophylaxis, treatment, or adjuncts. While there remain many knowledge gaps, these new methods and ongoing advancements in RVI treatment and prevention promise to improve transplant patient outcomes.

1. Introduction and Epidemiology

Respiratory viral infections (RVIs) pose an unremitting risk to transplant recipients, an especially vulnerable population. Solid organ transplant (SOT) and hematopoietic stem cell transplant (HCT) recipients experience increased morbidity with RVIs, including an increased risk of acute and chronic graft dysfunction, severe lower respiratory tract disease, secondary bacterial infections, hospitalization, and mortality [1]. A retrospective study of allogeneic HCT recipients identified degree of immunosuppression as the most important factor in mortality rather than the properties of the different viruses themselves [2]. In addition to increased severity of disease, transplant recipients can exhibit prolonged viral shedding, which may lead to increased periods of contagiousness and risk of resistant variant emergence [3]. The timing of infection depends on virus seasonality, as depicted in Figure 1.
Decreasing vaccine rates and therefore decreased herd immunity to RVIs among the general population pose a significant risk to transplant patients [4]. Fortunately, the landscape of vaccines and novel RVI treatments is rapidly evolving. This review will discuss the current strategies for pre- and post-exposure prophylaxis and treatments available to transplant recipients, highlighting their efficacy, safety, and practical considerations in the immunocompromised population.
Figure 1. Seasonality of common respiratory viruses in temperate regions as included in this review [5,6,7].
Figure 1. Seasonality of common respiratory viruses in temperate regions as included in this review [5,6,7].
Viruses 18 00008 g001

2. Influenza

2.1. Background

Despite vaccination being available since 1945 to stymie seasonal pandemics, the influenza virus remains a major cause of RVIs, related to its tendency to mutate via processes of antigenic drift and shift. A multicenter prospective study (2010–2015) of SOT and HCT recipients with confirmed influenza A found that patients most commonly presented with cough, fever, and coryza, although fever was absent in 36.2% of cases [8]. Seasonal vaccination reduced the risk of severe disease (pneumonia and intensive care unit [ICU] admission) and early antiviral therapy within 48 h improved outcomes [8]. SOT recipients (SOTRs) have a higher risk of complications than the general population, including pneumonia in up to 22–49% of cases, increased ICU admissions, increased mortality, and risks of allograft dysfunction and acute rejection [3]. These complications underline the importance of vaccination and early presentation for appropriate treatment.

2.2. Vaccination

2.2.1. Available Vaccines

The inactivated influenza vaccine (IIV) is trivalent and directed towards two main types: influenza A (H1N1 and H3N2) and influenza B [9]. Annual vaccine strains are pre-selected in February in the Northern Hemisphere and in September in the Southern Hemisphere, based on those found in the global community.
Since its emergence in 1996, the highly pathogenic avian influenza (HPAI) A/H5N1 strain has remained a global public health concern. Ongoing research aims to develop effective countermeasures, including a universal influenza vaccine. Studies of split and adjuvanted whole-virus H5N1 vaccines have demonstrated safety and immunogenicity, while newer platforms such as virus-like particles (VLPs) and mRNA vaccines also show potential [10].
Although a live-attenuated influenza vaccine is available, it is contraindicated in immunosuppressed individuals. If administered, transplantation should be delayed for at least two weeks to minimize the risk of vaccine-derived viral replication [11].

2.2.2. Timing

It is recommended to administer the IIV on an annual basis preceding the influenza season. Two doses separated by four weeks are required in previously unvaccinated patients less than nine-years-old. Pre-transplant administration is preferred.
For SOTRs in the post-transplant period, the vaccine can be given as early as one month after transplant [11]. HCT recipients are recommended to receive the vaccine at six months post-transplant although earlier vaccination at three months may be considered during periods of high transmission [12]. For patients with severe graft versus host disease (GVHD) or lymphopenia, a booster dose at four weeks may be considered [12].

2.2.3. Efficacy and Safety

The immunogenicity of IIV in transplant recipients is consistently lower than in immunocompetent individuals- with seroprotection rates varying from 15–90% in SOTRs- and is variable depending on transplanted organ, level of immunosuppression, and vaccine composition [13]. Lung transplant recipients in particular were noted to have the lowest seropositive response rates [13].
The TRANSGRIPE 1–2 study demonstrated that a booster dose administered five weeks after the first dose in SOTRs was associated with higher seroconversion rate for H1N1 but not for other strains [14]. Recently, the STOP-FLU trial demonstrated improved vaccine response rate after administration of high dose (66%) or MF59-adjuvanted vaccine (60%) as compared to standard vaccine (42%) in SOTRs [15].
Despite reduced immunogenicity, the IIV is generally considered safe and has been associated with a decreased risk of complications including pneumonia, ICU admission, use of invasive ventilation, and death [3,8].

2.3. Pre- and Post-Exposure Prophylaxis

Pre-exposure prophylaxis with oseltamivir for 12 weeks may be an alternative for patients in whom the vaccine is not preferred or humoral response is expected to be diminished [3]. Post-exposure prophylaxis with oseltamivir for seven days is recommended in SOTRs who have had close contact with a patient with documented influenza [3].

2.4. Treatment

2.4.1. Neuraminidase Inhibitors

Neuraminidase inhibitors (NAIs)- including oseltamivir, zanamivir, and peramivir- block the release of newly formed influenza virions from infected host cells [16]. Oseltamivir (Tamiflu) remains the preferred agent in immunocompromised patients due to its favorable safety profile and oral formulation [16]. Zanamivir (Relenza), administered by inhalation, is avoided in patients with chronic respiratory disease because of post-licensure reports of bronchospasm [16]. Peramivir (Rapivab), given intravenously, offers an alternative for patients unable to tolerate oral or inhaled medications and has demonstrated comparable efficacy, though data on its safety and optimal duration in immunocompromised populations remain limited [17]. In a multicenter study of SOT and HCT recipients with influenza infection, 94.1% received oseltamivir, and early treatment within 48 h was associated with improved outcomes [8]. Standard-dose oseltamivir was better tolerated than high-dose therapy, though higher doses were linked to lower viral resistance rates [18].

2.4.2. Cap-Dependent Endonuclease Inhibitors

Cap-dependent endonuclease inhibitors (CDEIs) block a key step in viral mRNA synthesis. Baloxavir marboxil (Xofluza), approved for patients aged ≥5 years, is administered as a single dose and has shown efficacy comparable to oseltamivir with faster symptom resolution [18,19]. However, reduced viral susceptibility occurred in ~10% of cases, particularly with prolonged viral shedding, leading guidelines to discourage its routine use in immunocompromised patients [19]. Subsequent studies in high risk and immunocompromised individuals demonstrated similar efficacy to oseltamivir, and combination therapy with neuraminidase inhibitors was well tolerated but did not improve outcomes [8,20,21]. Novel CDEIs, including TG-1000, ZX-7101A, AL-794, and onradivir, show early promise but remain under investigation.

2.4.3. CD388

CD388 is a long-acting injectable drug Fc conjugate containing multiple neuraminidase inhibitor copies, designed to protect against all known seasonal and pandemic influenza strains regardless of immune status [22]. In the Phase 2b NAVIGATE trial, a single injection demonstrated up to 76% efficacy in preventing laboratory-confirmed influenza over a 24-week season [22]. Protection was consistent across influenza A and B strains, and the drug was well tolerated with no significant safety concerns or dose-limiting adverse effects [22]. These findings highlight CD388’s potential as a durable, non-vaccine preventive strategy for individuals with impaired immunity, including transplant recipients.
Please refer to Table 1 for a summary of prophylactic and therapeutic options for the prevention and management of influenza in transplant recipients.

3. SARS-CoV-2

3.1. Background

SARS-CoV-2 infection carries significant morbidity and mortality in immunocompromised patients, with organ transplant recipients experiencing a 38% higher risk of death compared to the general population [24]. With advancements in early detection, treatment, and vaccination as well as changes in virility between variants, mortality in SOTRs has decreased from 20–25% at the beginning of the pandemic to 8–10% by 2022 [25]. However, transplant patients remain especially at risk of severe disease even with less virulent strains, given higher viral mutation rates and prolonged shedding [26]. Studies have shown increased ICU admission, mechanical ventilation, and mortality rates in SOTRs due to SARS-CoV-2 relative to other respiratory viruses [27]. Risk factors for severe disease include lung transplantation, recent rituximab or corticosteroid use, older age, and chronic comorbidities [28]. Vaccination with three or more doses and early antiviral treatment within seven days of symptom onset is protective, though vaccine effectiveness against hospitalization in immunocompromised adults ≥65 years was modest at 40% [29]. As with influenza, early vaccination and prompt antiviral therapy remain critical.

3.2. Vaccination

3.2.1. Available Vaccines

Currently available vaccines include mRNA BNT162b2 (Cominarty, Pfizer-BioNTech—New York City, NY, USA), mRNA-1273 (Spikevax, Moderna—Cambridge, MA, USA), and the protein subunit vaccine NVX-CoV2373 (Novavax—Gaithersburg, MD, USA). The mRNA vaccines are preferred due to greater evidence of safety and efficacy in immunocompromised patients [30,31]. Normal dose vaccine is preferred; no improvement in immunogenicity was detected on using double dose in the RECOVAC trial [32].

3.2.2. Timing

Immunocompromised individuals require a three-dose primary SARS-CoV-2 vaccine series due to poor humoral responses, with only 20–40% of transplant recipients developing antibodies after two doses compared to nearly 100% in healthy individuals [24,33]. Multiple studies in SOTRs show improved immunogenicity after three doses, though a subset remains nonresponsive [31,33]. Both homologous (same vaccine type) and heterologous (different vaccine type) third doses enhance antibody and T-cell responses, particularly in kidney transplant recipients [34]. The Centers for Disease Control and Prevention (CDC) recommends a fourth dose at least two months after the primary series; this further increases seropositivity (from 29.4% to 55.6%) and enhances neutralization activity against most variants, though Omicron responses remain suboptimal [35,36].
Vaccination should ideally be completed pre-transplant, but due to peri-transplant immunosuppression, the series should be repeated post-transplant, beginning at least one month after SOT [24,30]. For HCT recipients, vaccination is recommended at six months post-transplant, or as early as three months during community outbreaks [12]. Booster timing varies by region (6–12 months), with longer intervals improving hybrid immunity but increasing interim infection risk [37].

3.2.3. Efficacy and Safety

Vaccine responses in immunocompromised patients are suboptimal but improve with additional doses, as shown in multiple meta-analyses [38]. Poor response is associated with older age, higher immunosuppression, and reduced renal function [33]. High-dose mycophenolate reduces humoral immunity in a dose-dependent manner [39]. While short-term mycophenolate interruption showed no benefit, studies suggest improved antibody responses when switching to everolimus or when mycophenolate is held for more than six weeks [32,40,41].
Despite low antibody titers, cross-protective cellular immunity may mitigate disease severity. Prior Omicron infection conferred T-cell–mediated protection against emerging variants such as JN.1 in SOTRs [42]. mRNA vaccines remain safe and well tolerated in both SOT and HCT recipients, with low rates of mild adverse effects [43].

3.3. Pre- and Post-Exposure Prophylaxis

Pemivibart (Pemgarda, Invivyd—New Haven, CT, USA) is a long-acting monoclonal IgG1 antibody authorized by the FDA for pre-exposure prophylaxis of SARS-CoV-2 in immunocompromised individuals aged ≥12 years [44,45]. While not a substitute for vaccination or treatment of active infection, it may be administered every three months; however, clinical benefit in this population remains uncertain [44].

3.4. Treatment

3.4.1. Remdesivir (Veklury, Gilead Sciences—Foster City, CA, USA)

Remdesivir is a nucleotide analogue inhibiting the RNA-dependent RNA polymerase in SARS-CoV-2. A large retrospective review of immunocompromised adults demonstrated improved mortality at 14 and 28 days, with an approximately 35% lower mortality risk in SOT and HCT recipients [46]. Early initiation within seven days of symptom onset in outpatient SOTRs is associated with decreased hospitalization rate, with an adjusted hazard ratio of 0.12 [47]. It is the preferred method of treatment in hospitalized immunocompromised individuals.

3.4.2. Nirmatrelvir-Ritonavir (Paxlovid, Pfizer—New York City, NY, USA)

Nirmatrelvir inhibits the SARS-CoV-2 main protease while ritonavir boosts the drug plasma levels via CYP3A inhibition [48]. Approved for early treatment of mild-to-moderate COVID-19 in high-risk patients, extended 10–15 day courses in immunocompromised hosts modestly reduced viral rebound without shortening viral shedding [49]. Use in transplant patients is limited by major drug-drug interactions with calcineurin and mTOR inhibitors [48].

3.4.3. Molnupiravir (Lagevrio, Merck & Co., Ltd.—Rahway, NJ, USA)

Molnupiravir, an oral nucleoside analogue, is authorized for mild-to-moderate SARS-CoV-2 infection in adults at risk for severe disease [50]. It is generally well tolerated and has been associated with reduced hospitalization rates among SOTRs, though efficacy appears lower than with remdesivir or nirmatrelvir-ritonavir [50]. Molnupiravir is considered teratogenic and the package insert recommends reliable contraception at least four days in females and for three months in males after completion of therapy [51].

3.4.4. Monoclonal Antibodies

Anti-SARS-CoV-2 monoclonal antibodies (mAbs) have been used effectively in SOTRs for pre-exposure and post-exposure prophylaxis and early treatment of mild-to-moderate infection. No evidence of rejection has been observed in observational studies. However, many mAbs have become less efficacious with the development of new variants [33].
Please refer to Table 2 for an overview of SARS-CoV-2 vaccines, monoclonal antibodies, and antiviral agents utilized for prevention and treatment in transplant recipients.

4. Respiratory Syncytial Virus (RSV)

4.1. Background

RSV causes substantial morbidity in transplant recipients, with high rates of lower respiratory tract disease, respiratory failure, graft dysfunction, and mortality. In lung transplant recipients, RSV infection is linked to a 29% incidence of new allograft dysfunction within three months [51]. Early vaccine development was hindered by enhanced respiratory disease (ERD) observed with a formalin-inactivated vaccine in the 1960s which increased the severity of illness caused by natural infection [52]. Recent advances in knowledge of the RSV pre-fusion (pre-F) protein structure and streamlined vaccine pathways following the SARS-CoV-2 pandemic have revitalized RSV prevention efforts, resulting in three approved adult vaccines and over thirty candidates in development [53].

4.2. Vaccination

4.2.1. Available Vaccines

Current RSV vaccines target the pre-F protein, including two protein subunit vaccines, Abrysvo (non-adjuvanted, Pfizer—New York City, NY, USA) and Arexvy (adjuvanted, GSK—London, UK), and one mRNA vaccine, mRESVIA (Moderna—Cambridge, MA, USA) [54]. Abrysvo is approved for adults ≥18 years and for use during pregnancy to protect infants [54]. Arexvy is indicated for at-risk adults ≥50 years or ≥60 years regardless of risk [54]. mRESVIA may be used in adults ≥18 years at high risk, including immunocompromised and transplant recipients, and in all adults ≥60 years [55].

4.2.2. Timing

A single dose of RSV vaccine is recommended prior to the RSV season, ideally before transplantation. Post-transplant vaccination lacks definitive guidance, though experts suggest delaying for three to six months [12]. Revaccination is not routinely advised but may be considered in non-responders [56]. Early data indicate that booster doses offer limited additional benefit in transplant recipients [56,57].

4.2.3. Efficacy and Safety

Ongoing studies continue to evaluate RSV vaccine efficacy and safety in transplant recipients. Vaccine effectiveness appears reduced compared to the general population (50–69% vs. 75%), with the lowest rates among stem cell transplant recipients (29–44%) [58]. Antibody persistence up to one year post-vaccination has been observed, particularly in those further from transplant, off immunosuppression, and with higher lymphocyte counts [59]. Despite modest seroconversion rates, robust CD4+ T-cell responses suggest potential clinical benefit [60,61]. The adjuvanted Arexvy vaccine may enhance immunogenicity compared to the non-adjuvanted Abrysvo vaccine [36]. There is limited data for the use of mRESVIA in the immunocompromised population and it is not currently recommended [56]. Mycophenolate use reduces vaccine response, though optimal management is undefined [61]. Safety data show no transplant-related rejection or serious immune complications, with a small risk of Guillain-Barré syndrome (~11.2 per million doses) with Abrysvo and Arexvy [58,61].

4.3. Pre-Exposure Prophylaxis

Palivizumab (Synagis, Sobi—Waltham, MA, USA), an RSV fusion protein-targeted mAb requiring monthly administration, was previously used off-label in high-risk adults but showed no clear clinical benefit in transplant populations [62,63]. It has been replaced by newer long-acting antibodies and is expected to be discontinued after 2025 [64].
Next-generation RSV mAbs incorporate Fc modifications that prolong half-life, enabling single-dose seasonal protection [53]. Nirsevimab (Beyfortus, SANOFI—Swiftwater, PA, USA) provides ~70% efficacy against RSV-related lower respiratory tract infection in infants, though adult dosing remains undefined [53]. Clesrovimab (Enflonsia, Merck & Co., Ltd.—Rahway, NJ, USA), a similar long-acting candidate, shows comparable duration and efficacy [53]. Both agents hold promise for immunocompromised and transplant patients, but clinical data in these groups are currently lacking. Although mAb-resistant RSV variants have been reported, they generally exhibit reduced viral fitness [53].
These emerging long-acting monoclonal antibodies represent an important step toward passive immunoprophylaxis for vulnerable populations and complement ongoing efforts to optimize vaccine-based prevention strategies in transplant recipients.

4.4. Treatment

4.4.1. Ribavirin

Ribavirin (multiple brand names and manufacturers), a nucleoside analog approved for treatment of RSV lower respiratory tract disease in high-risk and transplant populations, has shown variable benefit [3]. Early data in HCT recipients suggested reduced disease progression and mortality when combined with immunomodulators such as palivizumab, IVIG, or RSV-IVIG [3]. However, more recent systematic reviews in lung transplant recipients report conflicting efficacy, with no significant association between ribavirin use and reduced chronic lung allograft dysfunction [65].
Aerosolized ribavirin remains the preferred route due to higher bronchoalveolar concentrations compared with oral formulations [66]. While oral ribavirin may serve as a feasible alternative in resource-limited settings or when inhaled therapy is impractical, evidence supporting its efficacy remains limited [67]. It should be noted that ribavirin is highly teratogenic and is classified as Category X by the FDA [68].

4.4.2. Molnupiravir (Lagevrio, Merck & Co., Ltd.—Rahway, NJ, USA)

Similar to its use in SARS-CoV-2, Molnupiravir may have some activity against RSV however clinical data in immunocompromised patients is lacking. A recent study of healthy adults did not meet primary efficacy endpoints of quantitative viral culture however did show modest non-significant benefits with treatment including rapid improvement in symptoms [69].
Please refer to Table 3 for a summary of available RSV vaccines and monoclonal antibody products for prevention and management in transplant recipients.

5. Other Respiratory Viruses

Vaccines for rhinovirus/enterovirus, parainfluenza virus (PIV), human metapneumovirus (hMPV), adenovirus (HAdV), and non-COVID human coronaviruses (HCoV) are in early development. Cytomegalovirus (CMV), while not strictly a respiratory virus, is known to cause severe respiratory disease in transplant patients. High-risk and transplant populations remain underrepresented in trials, and progress is hindered by antigenic diversity, rapid mutation, limited correlates of protection, and weak commercial incentives for low-morbidity pathogens.

5.1. Cytomegalovirus (CMV)

Cytomegalovirus (CMV) is a common post-transplant opportunistic infection; CMV pneumonia remains a rare but severe complication with approximately 60% one-year mortality in HCT recipients despite advances in antiviral therapy [71]. Highest risk groups include seropositive HCT recipients and seronegative SOTRs receiving organs from seropositive donors [72]. Prevention relies on antiviral prophylaxis or preemptive therapy guided by CMV PCR surveillance [72]. Valganciclovir (Valcyte) and letermovir (Prevymix) are principal agents used for CMV prophylaxis, with valganciclovir standard for most SOTRs (3–6-month duration, up to 12 months in lung transplantation) and letermovir approved for allogeneic HCT and kidney transplant recipients, having demonstrated non-inferiority and fewer adverse effects [73]. In HCT recipients, preemptive therapy based on center-specific PCR thresholds is commonly employed [72]. CMV pneumonia is typically treated with intravenous ganciclovir, with adjunctive immunoglobulin sometimes used despite limited supporting evidence [74]. Alternative or combination therapies, including foscarnet (Foscavir) and cidofovir (Vistide) can be considered [74]. Although maribavir (Livtencity) is effective for refractory or resistant CMV infection, it is not preferred for invasive disease including pneumonia [75]. CMV-specific adoptive T-cell therapy is an emerging strategy for prophylaxis and treatment of refractory disease [76]. Vaccines remain under development, although none are currently approved [76].

5.2. Rhinovirus/Enterovirus

Vaccine development is limited by >150 rhinovirus serotypes [77]. Experimental mRNA platforms show cross-reactive immunity in animals [78]. Several antivirals including pleconaril, rupintrivir, and remdesivir, demonstrate in vitro activity but lack clinical evidence [79].

5.3. Human Metapneumovirus (hMPV)

No approved therapies exist. Early trials of combined RSV/hMPV/PIV-3 (Sanofi) and mRNA vaccines (Moderna) demonstrate safety and immunogenicity [77,80]. Investigational mAbs and T-cell-based therapies show promise in preclinical studies [81]. There is limited evidence for the efficacy of probenecid but further studies are needed [81].

5.4. Adenovirus (HAdV)

The AdV-4/7 vaccine, covering serotypes 4 and 7, two of the most prominent causing respiratory disease, provides durable protection but is restricted to military use [82]. Cidofovir remains standard therapy, with brincidofovir under investigation as a safer alternative [83].

5.5. Non-COVID Human Coronavirus (HCoV)

Multiple preclinical and early clinical vaccines are under investigation, though durable immunity remains a challenge [84]. No antiviral therapies are approved [84].

5.6. Non-RSV Paramyxoviridae (e.g., Parainfluenza [PIV])

Novel antivirals under study include GHP-88309 and DAS-181, the latter showing post hoc benefit in immunocompromised patients [85,86]. Ribavirin is sometimes used off-label for severe cases, though efficacy data are limited [87].

6. Future Directions

Transplant recipients remain underrepresented in antiviral and vaccine trials, limiting evidence and delaying guidance. While novel agents, some described in Table 4, are promising, few are tested in transplant or immunocompromised populations. Dedicated transplant-specific studies are needed to define optimal dosing, timing, and safety. Long-acting monoclonal antibodies and non-vaccine-based prophylaxis may overcome poor vaccine immunogenicity in this population. Continued viral surveillance and public vaccine advocacy remain essential amid evolving variants and declining vaccination rates.

7. Conclusions

Pre- and post-exposure prophylaxis are central to mitigating respiratory viral infection risk in transplant recipients. Pre- and post-exposure prophylaxis strategies are summarized in Table 5. Emerging long-acting and variant-targeted agents show promise but require further validation in transplant-specific populations. Vaccination remains foundational, complemented by antivirals and monoclonal antibodies.

Author Contributions

Conceptualization, N.L.; literature review, A.A.M.G.; original manuscript draft preparation, A.A.M.G.; original table draft preparation, V.C.; original figure preparation, A.A.M.G.; writing—critical reviewing and editing, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data sharing is not applicable (only appropriate if no new data is generated or the article describes entirely theoretical research). No new data were created or analyzed in this study.

Acknowledgments

We thank Victoria Hall (Transplant Infectious Diseases, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada) and Michael G. Ison (Respiratory Diseases Branch, DMID/NIAID/NIH) for their collegial engagement during the World Transplant Congress session, “Stopping the Inevitable? Approaches to Respiratory Viruses,” held on 3 August at 7:00 a.m., and for their thoughtful review of Nancy Law’s presentation titled “Pre-Exposure and Post-Exposure Prophylaxis for Respiratory Viruses.” Select data and references from this presentation were incorporated into the current manuscript to enhance its comprehensiveness and relevance.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
RVIRespiratory viral infection
SOTSolid organ transplant
HCTHematopoietic stem cell transplant
SOTRSolid organ transplant recipient
RSVRespiratory syncytial virus
mAbMonoclonal antibody

References

  1. Mombelli, M.; Lang, B.; Neofytos, D.; Aubert, J.; Benden, C.; Berger, C.; Boggian, K.; Egli, A.; Soccal, P.; Kaiser, L.; et al. Burden, epidemiology, and outcomes of microbiologically confirmed respiratory viral infections in solid organ transplant recipients: A nationwide, multi-season prospective cohort study. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2021, 21, 1789–1800. [Google Scholar] [CrossRef]
  2. Pérez, A.; Gómez, D.; Montoro, J.; Chorão, P.; Hernani, R.; Guerriero, M.; Villalba, M.; Albert, E.; Carbonell-Asins, J.A.; Hernández-Boluda, J.C.; et al. Are any specific respiratory viruses more severe than others in recipients of allogeneic stem cell transplantation? A focus on lower respiratory tract disease. Bone Marrow Transplant. 2024, 59, 1118–1126. [Google Scholar] [CrossRef]
  3. Manuel, O.; Estabrook, M.; American Society of Transplantation Infectious Diseases Community of Practice. RNA respiratory viral infections in solid organ transplant recipients: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin. Transplant. 2019, 33, e13511. [Google Scholar] [CrossRef]
  4. Nofzinger, T.B.; Huang, T.T.; Lingat, C.E.R.; Amonkar, G.M.; Edwards, E.E.; Yu, A.; Smith, A.D.; Gayed, N.; Gaddey, H. Vaccine fatigue and influenza vaccination trends across Pre-, Peri-, and Post-COVID-19 periods in the United States using epic’s cosmos database. PLoS ONE 2025, 20, e0326098. [Google Scholar] [CrossRef] [PubMed]
  5. Moriyama, M.; Hugentobler, W.J.; Iwasaki, A. Seasonality of Respiratory Viral Infections. Annu. Rev. Virol. 2020, 7, 83–101. [Google Scholar] [CrossRef]
  6. Wiemken, T.L.; Khan, F.; Puzniak, L.; Yang, W.; Simmering, J.; Polgreen, P.; Nguyen, J.L.; Jodar, L.; McLaughlin, J.M. Seasonal trends in COVID-19 cases, hospitalizations, and mortality in the United States and Europe. Sci. Rep. 2023, 13, 3886. [Google Scholar] [CrossRef]
  7. Giuliani, A. Created in BioRender. 2025. Available online: https://BioRender.com/cu1xygq (accessed on 10 December 2025).
  8. Kumar, D.; Ferreira, V.H.; Blumberg, E.; Silveira, F.; Cordero, E.; Perez-Romero, P.; Aydillo, T.; Danziger-Isakov, L.; Limaye, A.P.; Carratala, J.; et al. A 5-Year Prospective Multicenter Evaluation of Influenza Infection in Transplant Recipients. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2018, 67, 1322–1329. [Google Scholar] [CrossRef]
  9. Federal Drug Administration. Influenza Vaccine Composition for the 2025–2026 U.S. Influenza Season; Federal Drug Administration: Silver Spring, MD, USA, 2025.
  10. Faccin, F.C.; Perez, D.R. Pandemic preparedness through vaccine development for avian influenza viruses. Hum. Vaccines Immunother. 2024, 20, 2347019. [Google Scholar] [CrossRef]
  11. Danziger-Isakov, L.; Kumar, D.; AST ID Community of Practice. Vaccination of solid organ transplant candidates and recipients: Guidelines from the American society of transplantation infectious diseases community of practice. Clin. Transplant. 2019, 33, e13563. [Google Scholar] [CrossRef] [PubMed]
  12. Silva-Pinto, A.; Abreu, I.; Martins, A.; Bastos, J.; Araújo, J.; Pinto, R. Vaccination After Haematopoietic Stem Cell Transplant: A Review of the Literature and Proposed Vaccination Protocol. Vaccines 2024, 12, 1449. [Google Scholar] [CrossRef] [PubMed]
  13. Chong, P.P.; Handler, L.; Weber, D.J. A Systematic Review of Safety and Immunogenicity of Influenza Vaccination Strategies in Solid Organ Transplant Recipients. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2018, 66, 1802–1811. [Google Scholar] [CrossRef]
  14. Cordero, E.; Roca-Oporto, C.; Bulnes-Ramos, A.; Aydillo, T.; Gavaldà, J.; Moreno, A.; Torre-Cisneros, J.; Montejo, J.M.; Fortun, J.; Muñoz, P.; et al. Two Doses of Inactivated Influenza Vaccine Improve Immune Response in Solid Organ Transplant Recipients: Results of TRANSGRIPE 1–2, a Randomized Controlled Clinical Trial. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2017, 64, 829–838. [Google Scholar] [CrossRef]
  15. Mombelli, M.; Neofytos, D.; Huynh-Do, U.; Sánchez-Céspedes, J.; Stampf, S.; Golshayan, D.; Dahdal, S.; Stirnimann, G.; Schnyder, A.; Garzoni, C.; et al. Immunogenicity of High-Dose Versus MF59-Adjuvanted Versus Standard Influenza Vaccine in Solid Organ Transplant Recipients: The Swiss/Spanish Trial in Solid Organ Transplantation on Prevention of Influenza (STOP-FLU Trial). Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2024, 78, 48–56. [Google Scholar] [CrossRef]
  16. Moscona, A. Neuraminidase Inhibitors for Influenza. N. Engl. J. Med. 2005, 353, 1363–1373. [Google Scholar] [CrossRef]
  17. Uyeki, T.M.; Bernstein, H.H.; Bradley, J.S.; Englund, J.A.; File, T.M.; Fry, A.M.; Gravenstein, S.; Hayden, F.G.; Harper, S.A.; Hirshon, J.M.; et al. Clinical Practice Guidelines by the Infectious Diseases Society of America: 2018 Update on Diagnosis, Treatment, Chemoprophylaxis, and Institutional Outbreak Management of Seasonal Influenza. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2019, 68, e1–e47. [Google Scholar] [CrossRef] [PubMed]
  18. Mitha, E.; Krivan, G.; Jacobs, F.; Nagler, A.; Alrabaa, S.; Mykietiuk, A.; Kenwright, A.; Le Pogam, S.; Clinch, B.; Vareikiene, L. Safety, Resistance, and Efficacy Results from a Phase IIIb Study of Conventional- and Double-Dose Oseltamivir Regimens for Treatment of Influenza in Immunocompromised Patients. Infect. Dis. Ther. 2019, 8, 613–626. [Google Scholar] [CrossRef] [PubMed]
  19. Committee On Infectious Diseases. Recommendations for Prevention and Control of Influenza in Children 2022–2023. Pediatrics 2022, 150, e2022059275. [Google Scholar] [CrossRef]
  20. Ison, M.G.; Portsmouth, S.; Yoshida, Y.; Shishido, T.; Mitchener, M.; Tsuchiya, K.; Uehara, T.; Hayden, F.G. Early treatment with baloxavir marboxil in high-risk adolescent and adult outpatients with uncomplicated influenza (CAPSTONE-2): A randomised, placebo-controlled, phase 3 trial. Lancet. Infect. Dis. 2020, 20, 1204–1214. [Google Scholar] [CrossRef]
  21. Ringer, M.; Malinis, M.; McManus, D.; Davis, M.; Shah, S.; Trubin, P.; Topal, J.E.; Azar, M.M. Clinical outcomes of baloxavir versus oseltamivir in immunocompromised patients. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2024, 26, e14249. [Google Scholar] [CrossRef]
  22. Hayden, F.G.; Sugaya, N.; Hirotsu, N.; Lee, N.; de Jong, M.D.; Hurt, A.C.; Ishida, T.; Sekino, H.; Yamada, K.; Portsmouth, S.; et al. Baloxavir Marboxil for Uncomplicated Influenza in Adults and Adolescents. N. Engl. J. Med. 2018, 379, 913–923. [Google Scholar] [CrossRef] [PubMed]
  23. Cidara Therapeutics Announces Positive Topline Results from Its Phase 2b Navigate Trial Evaluating CD388, a Non-Vaccine Preventative of Seasonal Influenza (2025). Cidara Therepeutics. Available online: https://www.cidara.com/news/cidara-therapeutics-announces-positive-topline-results-from-its-phase-2b-navigate-trial-evaluating-cd388-a-non-vaccine-preventative-of-seasonal-influenza/ (accessed on 30 September 2025).
  24. Foroutan, F.; Rayner, D.; Oss, S.; Straccia, M.; de Vries, R.; Raju, S.; Ahmed, F.; Kingdon, J.; Bhagra, S.; Tarani, S.; et al. Clinical Practice Recommendations on the Effect of COVID-19 Vaccination Strategies on Outcomes in Solid Organ Transplant Recipients. Clin. Transplant. 2025, 39, e70100. [Google Scholar] [CrossRef]
  25. Khawaja, F.; Papanicolau, G.; Dadwal, S.; Pergam, S.A.; Wingard, J.R.; Boghdadly, Z.E.; Abidi, M.Z.; Waghmare, A.; Shahid, Z.; Michaels, L.; et al. Frequently Asked Questions on Coronavirus Disease 2019 Vaccination for Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T-Cell Recipients From the American Society for Transplantation and Cellular Therapy and the American Society of Hematology. Transplant. Cell. Ther. 2023, 29, 10–18. [Google Scholar] [CrossRef] [PubMed]
  26. Nimmo, A.; Gardiner, D.; Ushiro-Lumb, I.; Ravanan, R.; Forsythe, J.L.R. The Global Impact of COVID-19 on Solid Organ Transplantation: Two Years Into a Pandemic. Transplantation 2022, 106, 1312–1329. [Google Scholar] [CrossRef]
  27. Mendoza, M.A.; Motoa, G.; Raja, M.A.; Frattaroli, P.; Fernandez, A.; Anjan, S.; Courel, S.C.; Natori, A.; O’Brien, C.B.; Phancao, A.; et al. Difference between SARS-CoV-2, seasonal coronavirus, influenza, and respiratory syncytial virus infection in solid organ transplant recipients. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2023, 25, e13998. [Google Scholar] [CrossRef]
  28. Solera, J.T.; Árbol, B.G.; Mittal, A.; Hall, V.; Marinelli, T.; Bahinskaya, I.; Selzner, N.; McDonald, M.; Schiff, J.; Sidhu, A.; et al. Longitudinal outcomes of COVID-19 in solid organ transplant recipients from 2020 to 2023. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2024, 24, 1303–1316. [Google Scholar] [CrossRef]
  29. Ma, K.C.; Webber, A.; Lauring, A.S.; Bendall, E.; Papalambros, L.K.; Safdar, B.; Ginde, A.A.; Peltan, I.D.; Brown, S.M.; Gaglani, M.; et al. Effectiveness of 2024–2025 COVID-19 Vaccination Against COVID-19 Hospitalization and Severe In-Hospital Outcomes—IVY Network 2025, 26 Hospitals, September 1, 2024-April 30, 2025. medRxiv Prepr. Serv. Health Sci. 2025, 33, 4612. [Google Scholar] [CrossRef]
  30. Alshukairi, A.N.; Al-Qahtani, A.A.; Obeid, D.A.; Dada, A.; Almaghrabi, R.S.; Al-Abdulkareem, M.A.; Alahideb, B.M.; Alsanea, M.S.; Alsuwairi, F.A.; Alhamlan, F.S. Molecular Epidemiology of SARS-CoV-2 and Clinical Manifestations among Organ Transplant Recipients with COVID-19. Viruses 2023, 16, 25. [Google Scholar] [CrossRef]
  31. Figueroa, A.L.; Azzi, J.R.; Eghtesad, B.; Priddy, F.; Stolman, D.; Siangphoe, U.; Leony Lasso, I.; de Windt, E.; Girard, B.; Zhou, H.; et al. Safety and Immunogenicity of the mRNA-1273 Coronavirus Disease 2019 Vaccine in Solid Organ Transplant Recipients. J. Infect. Dis. 2024, 230, e591–e600. [Google Scholar] [CrossRef] [PubMed]
  32. Kho, M.M.L.; Messchendorp, A.L.; Frölke, S.C.; Imhof, C.; Koomen, V.J.; Malahe, S.R.K.; Vart, P.; Geers, D.; de Vries, R.D.; GeurtsvanKessel, C.H.; et al. Alternative strategies to increase the immunogenicity of COVID-19 vaccines in kidney transplant recipients not responding to two or three doses of an mRNA vaccine (RECOVAC): A randomised clinical trial. Lancet. Infect. Dis. 2023, 23, 307–319. [Google Scholar] [CrossRef]
  33. Vafea, M.T.; Haidar, G. COVID-19 Prevention in Solid Organ Transplant Recipients: Current State of the Evidence. Infect. Dis. Clin. N. Am. 2023, 37, 459–473. [Google Scholar] [CrossRef]
  34. Reindl-Schwaighofer, R.; Heinzel, A.; Mayrdorfer, M.; Jabbour, R.; Hofbauer, T.M.; Merrelaar, A.; Eder, M.; Regele, F.; Doberer, K.; Spechtl, P.; et al. Comparison of SARS-CoV-2 Antibody Response 4 Weeks After Homologous vs Heterologous Third Vaccine Dose in Kidney Transplant Recipients: A Randomized Clinical Trial. JAMA Intern. Med. 2022, 182, 165. [Google Scholar] [CrossRef]
  35. Osmanodja, B.; Ronicke, S.; Budde, K.; Jens, A.; Hammett, C.; Koch, N.; Seelow, E.; Waiser, J.; Zukunft, B.; Bachmann, F.; et al. Serological Response to Three, Four and Five Doses of SARS-CoV-2 Vaccine in Kidney Transplant Recipients. J. Clin. Med. 2022, 11, 2565. [Google Scholar] [CrossRef] [PubMed]
  36. 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]
  37. Hamm, S.R.; Loft, J.A.; Pérez-Alós, L.; Heftdal, L.D.; Hansen, C.B.; Møller, D.L.; Pries-Heje, M.M.; Hasselbalch, R.B.; Fogh, K.; Hald, A.; et al. The Impact of Time between Booster Doses on Humoral Immune Response in Solid Organ Transplant Recipients Vaccinated with BNT162b2 Vaccines. Viruses 2024, 16, 860. [Google Scholar] [CrossRef]
  38. 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]
  39. Sakai, A.; Morishita, T.; Suzumura, K.; Hanatate, F.; Yoshikawa, T.; Sasaki, N.; Lee, S.; Fujita, K.; Hara, T.; Araki, H.; et al. The Trajectory of the COVID-19 Vaccine Antibody Titers Over Time and the Association of Mycophenolate Mofetil in Solid Organ Transplant Recipients. Transplant. Proc. 2022, 54, 2638–2645. [Google Scholar] [CrossRef]
  40. de Boer, S.E.; Berger, S.P.; van Leer-Buter, C.C.; Kroesen, B.; van Baarle, D.; Sanders, J.F.; OPTIMIZE study group. Enhanced Humoral Immune Response After COVID-19 Vaccination in Elderly Kidney Transplant Recipients on Everolimus Versus Mycophenolate Mofetil-containing Immunosuppressive Regimens. Transplantation 2022, 106, 1615–1621. [Google Scholar] [CrossRef]
  41. Messchendorp, A.L.; Zaeck, L.M.; Bouwmans, P.; van den Broek, D.A.J.; Frölke, S.C.; Geers, D.; Imhof, C.; Malahe, S.R.K.; Schmitz, K.S.; Reinders, J.; et al. Replacing Mycophenolate Mofetil by Everolimus in Kidney Transplant Recipients to Increase Vaccine Immunogenicity: Results of a Randomized Controlled Trial. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2025, ciaf107. [Google Scholar] [CrossRef]
  42. Ferreira, V.H.; Keith, B.; Mavandadnejad, F.; Ferro, A.; Marocco, S.; Amidpour, G.; Kurtesi, A.; Qi, F.; Gingras, A.; Hall, V.G.; et al. Longitudinal Innate and Heterologous Adaptive Immune Responses to SARS-CoV-2 JN.1 in Transplant Recipients with Prior Omicron Infection: Limited Neutralization but Robust CD4+ T-Cell Activity. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2025, 27, e70067. [Google Scholar] [CrossRef] [PubMed]
  43. Albiol, N.; Aso, O.; Gómez-Pérez, L.; Triquell, M.; Roch, N.; Lázaro, E.; Esquirol, A.; González, I.; López-Contreras, J.; Sierra, J.; et al. mRNA-1273 SARS-CoV-2 vaccine safety and COVID-19 risk perception in recently transplanted allogeneic hematopoietic stem cell transplant recipients. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2022, 30, 9687–9690. [Google Scholar] [CrossRef]
  44. Bhimraj, A.; Falck-Ytter, Y.; Kim, A.Y.; Li, J.Z.; Baden, L.R.; Johnson, S.; Shafer, R.W.; Shoham, S.; Tebas, P.; Bedimo, R.; et al. 2024 Clinical Practice Guideline Update by the Infectious Diseases Society of America on the Management of COVID-19: Anti-SARS-CoV-2 Neutralizing Antibody Pemivibart for Pre-exposure Prophylaxis. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2024, ciae435. [Google Scholar] [CrossRef]
  45. INVIVYD. PEMGARDA Authorized for Emergency Use. Available online: https://pemgarda.com/?gad_source=1&gad_campaignid=21506626247 (accessed on 30 September 2025).
  46. Mozaffari, E.; Chandak, A.; Gottlieb, R.L.; Chima-Melton, C.; Berry, M.; Amin, A.N.; Sax, P.E.; Kalil, A.C. Remdesivir-Associated Survival Outcomes Among Immunocompromised Patients Hospitalized for COVID-19: Real-world Evidence from the Omicron-Dominant Era. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2024, 79, S149–S159. [Google Scholar] [CrossRef]
  47. Solera, J.T.; Árbol, B.G.; Bahinskaya, I.; Marks, N.; Humar, A.; Kumar, D. Short-course early outpatient remdesivir prevents severe disease due to COVID-19 in organ transplant recipients during the omicron BA.2 wave. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2023, 23, 78–83. [Google Scholar] [CrossRef] [PubMed]
  48. Lemaitre, F.; Budde, K.; Van Gelder, T.; Bergan, S.; Lawson, R.; Noceti, O.; Venkataramanan, R.; Elens, L.; Moes, D.J.A.R.; Hesselink, D.A.; et al. Therapeutic Drug Monitoring and Dosage Adjustments of Immunosuppressive Drugs When Combined with Nirmatrelvir/Ritonavir in Patients with COVID-19. Ther. Drug Monit. 2023, 45, 191–199. [Google Scholar] [CrossRef] [PubMed]
  49. Weinstein, E.; Paredes, R.; Gardner, A.; Almas, M.; Baniecki, M.L.; Guan, S.; Tudone, E.; Antonucci, S.; Gregg, K.; Garcia-Vidal, C.; et al. Extended nirmatrelvir-ritonavir treatment durations for immunocompromised patients with COVID-19 (EPIC-IC): A placebo-controlled, randomised, double-blind, phase 2 trial. Lancet Infect. Dis. 2025, 25, 1243–1253. [Google Scholar] [CrossRef]
  50. 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. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2022, 22, 2458–2463. [Google Scholar] [CrossRef]
  51. Testaert, H.; Bouet, M.; Valour, F.; Gigandon, A.; Lafon, M.; Philit, F.; Sénéchal, A.; Casalegno, J.; Blanchard, E.; Le Pavec, J.; et al. Incidence, management and outcome of respiratory syncytial virus infection in adult lung transplant recipients: A 9-year retrospective multicentre study. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2021, 27, 897–903. [Google Scholar] [CrossRef] [PubMed]
  52. Frenkel, L.D.; Gaur, S.; Bellanti, J.A. The third pandemic: The respiratory syncytial virus landscape and specific considerations for the allergist/immunologist. Allergy Asthma Proc. 2023, 44, 220–228. [Google Scholar] [CrossRef]
  53. Mazur, N.I.; Terstappen, J.; Baral, R.; Bardají, A.; Beutels, P.; Buchholz, U.J.; Cohen, C.; Crowe, J.E.; Cutland, C.L.; Eckert, L.; et al. Respiratory syncytial virus prevention within reach: The vaccine and monoclonal antibody landscape. Lancet Infect. Dis. 2023, 23, e2–e21. [Google Scholar] [CrossRef]
  54. Anastassopoulou, C.; Medić, S.; Ferous, S.; Boufidou, F.; Tsakris, A. Development, Current Status, and Remaining Challenges for Respiratory Syncytial Virus Vaccines. Vaccines 2025, 13, 97. [Google Scholar] [CrossRef]
  55. MRESVIA—Respiratory Syncytial Virus Vaccine Suspension (2025). DailyMed: Food and Drug Administration. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=e5c837e7-41e8-496a-9c85-6b0453b35948 (accessed on 30 September 2025).
  56. Trubin, P.; Azar, M.M.; Kotton, C.N. The respiratory syncytial virus vaccines are here: Implications for solid organ transplantation. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2024, 24, 897–904. [Google Scholar] [CrossRef]
  57. Almeida, N.C.; Parameswaran, L.; DeHaan, E.N.; Wyper, H.; Rahman, F.; Jiang, Q.; Li, W.; Patton, M.; Lino, M.M.; Majid-Mahomed, Z.; et al. Immunogenicity and Safety of the Bivalent Respiratory Syncytial Virus Prefusion F Subunit Vaccine in Immunocompromised or Renally Impaired Adults. Vaccines 2025, 13, 328. [Google Scholar] [CrossRef]
  58. Fry, S.E.; Terebuh, P.; Kaelber, D.C.; Xu, R.; Davis, P.B. Effectiveness and Safety of Respiratory Syncytial Virus Vaccine for US Adults Aged 60 Years or Older. JAMA Netw. Open 2025, 8, e258322. [Google Scholar] [CrossRef]
  59. Redjoul, R.; Robin, C.; Softic, L.; Ourghanlian, C.; Cabanne, L.; Beckerich, F.; Caillault, A.; Soulier, A.; Daumerie, P.; Fourati, S.; et al. Respiratory Syncytial Virus Vaccination in Allogeneic Hematopoietic Stem Cell Transplant Recipients. JAMA Netw. Open 2025, 8, e2533828. [Google Scholar] [CrossRef]
  60. Havlin, J.; Skotnicova, A.; Dvorackova, E.; Palavandishvili, N.; Smetanova, J.; Svorcova, M.; Vaculova, M.; Hubacek, P.; Fila, L.; Trojanek, M.; et al. Respiratory syncytial virus prefusion F3 vaccine in lung transplant recipients elicits CD4+ T cell response in all vaccinees. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2025, 25, 1452–1460. [Google Scholar] [CrossRef] [PubMed]
  61. Hall, V.G.; Alexander, A.A.; Mavandadnejad, F.; Kern-Smith, M.; Dang, X.; Kang, R.; Humar, S.; Winichakoon, P.; Johnstone, R.; Aversa, M.; et al. Safety and immunogenicity of adjuvanted respiratory syncytial virus vaccine in high-risk transplant recipients: An Interventional Cohort Study. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2025; ahead of print. https://doi.org/10.1016/j.cmi.2025.09.013. [Google Scholar]
  62. de Fontbrune, F.S.; Robin, M.; Porcher, R.; Scieux, C.; de Latour, R.P.; Ferry, C.; Rocha, V.; Boudjedir, K.; Devergie, A.; Bergeron, A.; et al. Palivizumab treatment of respiratory syncytial virus infection after allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2007, 45, 1019–1024. [Google Scholar] [CrossRef] [PubMed]
  63. Permpalung, N.; Mahoney, M.V.; McCoy, C.; Atsawarungruangkit, A.; Gold, H.S.; Levine, J.D.; Wong, M.T.; LaSalvia, M.T.; Alonso, C.D. Clinical characteristics and treatment outcomes among respiratory syncytial virus (RSV)-infected hematologic malignancy and hematopoietic stem cell transplant recipients receiving palivizumab. Leuk. Lymphoma 2019, 60, 85–91. [Google Scholar] [CrossRef] [PubMed]
  64. Ernst, D. RSV Prevention Therapy Palivizumab to Be Discontinued. MPR: Medical Professionals Reference. 13 August 2025. Available online: https://www.empr.com/news/rsv-prevention-therapy-palivizumab-to-be-discontinued/#xd_co_f=MjU4NDVkMTItODAzZi00OWUzLTljZjUtNmQyMTNiNTdmMjc4~" (accessed on 30 September 2025).
  65. De Zwart, A.; Riezebos-Brilman, A.; Lunter, G.; Vonk, J.; Glanville, A.R.; Gottlieb, J.; Permpalung, N.; Kerstjens, H.; Alffenaar, J.; Verschuuren, E. Respiratory Syncytial Virus, Human Metapneumovirus, and Parainfluenza Virus Infections in Lung Transplant Recipients: A Systematic Review of Outcomes and Treatment Strategies. Clin. Infect. Dis. 2022, 74, 2252–2260. [Google Scholar] [CrossRef]
  66. Mueller, S.W.; Kiser, T.H.; Morrisette, T.; Zamora, M.R.; Lyu, D.M.; Kiser, J.L. Ribavirin and cellular ribavirin-triphosphate concentrations in blood and bronchoalveolar lavage fluid in two lung transplant patients with respiratory syncytial virus. Transpl. Infect. Dis. 2021, 23, e13464. [Google Scholar] [CrossRef]
  67. Permpalung, N.; Thaniyavarn, T.; Saullo, J.L.; Arif, S.; Miller, R.A.; Reynolds, J.M.; Alexander, B.D. Oral and Inhaled Ribavirin Treatment for Respiratory Syncytial Virus Infection in Lung Transplant Recipients. Transplantation 2020, 104, 1280–1286. [Google Scholar] [CrossRef]
  68. Aronson, J.K. Meyler’s Side Effects of Drugs; Elsevier: Amsterdam, The Netherlands, 2016; pp. 115–129. [Google Scholar] [CrossRef]
  69. Cheng, M.H.; Mann, A.J.; Maas, B.M.; Zhao, T.; Bevan, M.; Schaeffer, A.K.; Liao, L.E.; Catchpole, A.P.; Hilbert, D.W.; Aubrey Stoch, S.; et al. A Phase 2a, Randomized, Placebo-Controlled Human Challenge Trial to Evaluate the Efficacy and Safety of Molnupiravir in Healthy Participants Inoculated with Respiratory Syncytial Virus. Pulm. Ther. 2025, 11, 285–304. [Google Scholar] [CrossRef]
  70. Department of Health and Human Services. Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection. Available online: https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/adult-adolescent-oi/guidelines-adult-adolescent-oi.pdf (accessed on 30 September 2025).
  71. Erard, V.; Guthrie, K.A.; Seo, S.; Smith, J.; Huang, M.; Chien, J.; Flowers, M.E.D. Reduced Mortality of Cytomegalovirus Pneumonia After Hematopoietic Cell Transplantation Due to Antiviral Therapy and Changes in Transplantation Practices. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2015, 61, 31–39. [Google Scholar] [CrossRef]
  72. Haidar, G.; Boeckh, M.; Singh, N. Cytomegalovirus Infection in Solid Organ and Hematopoietic Cell Transplantation: State of the Evidence. J. Infect. Dis. 2020, 221, S23–S31. [Google Scholar] [CrossRef]
  73. Stewart, A.G.; Kotton, C.N. What’s New: Updates on Cytomegalovirus in Solid Organ Transplantation. Transplantation 2024, 108, 884–897. [Google Scholar] [CrossRef]
  74. Ljungman, P.; de la Camara, R.; Robin, C.; Crocchiolo, R.; Einsele, H.; Hill, J.A.; Hubacek, P.; Navarro, D.; Cordonnier, C.; Ward, K.N.; et al. Guidelines for the management of cytomegalovirus infection in patients with haematological malignancies and after stem cell transplantation from the 2017 European Conference on Infections in Leukaemia (ECIL 7). Lancet Infect. Dis. 2019, 19, e260–e272. [Google Scholar] [CrossRef]
  75. Hardinger, K.L.; Brennan, D.C. Cytomegalovirus Treatment in Solid Organ Transplantation: An Update on Current Approaches. Ann. Pharmacother. 2024, 58, 1122–1133. [Google Scholar] [CrossRef] [PubMed]
  76. Heldman, M.R.; Boeckh, M.J.; Limaye, A. Current and Future Strategies for the Prevention and Treatment of Cytomegalovirus Infections in Transplantation. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2025, 81, 581–592. [Google Scholar] [CrossRef] [PubMed]
  77. Stobart, C.C.; Nosek, J.M.; Moore, M.L. Rhinovirus Biology, Antigenic Diversity, and Advancements in the Design of a Human Rhinovirus Vaccine. Front. Microbiol. 2017, 8, 2412. [Google Scholar] [CrossRef] [PubMed]
  78. Mamun, T.I.; Ali, A.; Hosen, N.; Rahman, J.; Islam, A.; Akib, G.; Zaman, K.; Rahman, M.; Hossain, F.M.A.; Ibenmoussa, S.; et al. Designing a multi-epitope vaccine candidate against human rhinovirus C utilizing immunoinformatics approach. Front. Immunol. 2025, 15, 1364129. [Google Scholar] [CrossRef]
  79. Ianevski, A.; Frøysa, I.T.; Lysvand, H.; Calitz, C.; Smura, T.; Schjelderup Nilsen, H.; Høyer, E.; Afset, J.E.; Sridhar, A.; Wolthers, K.C.; et al. The combination of pleconaril, rupintrivir, and remdesivir efficiently inhibits enterovirus infections in vitro, delaying the development of drug-resistant virus variants. Antivir. Res. 2024, 224, 105842. [Google Scholar] [CrossRef] [PubMed]
  80. Schnyder Ghamloush, S.; Essink, B.; Hu, B.; Kalidindi, S.; Morsy, L.; Egwuenu-Dumbuya, C.; Kapoor, A.; Girard, B.; Dhar, R.; Lackey, R.; et al. Safety and Immunogenicity of an mRNA-Based hMPV/PIV3 Combination Vaccine in Seropositive Children. Pediatrics 2024, 153, e2023064748. [Google Scholar] [CrossRef] [PubMed]
  81. Principi, N.; Fainardi, V.; Esposito, S. Human Metapneumovirus: A Narrative Review on Emerging Strategies for Prevention and Treatment. Viruses 2025, 17, 1140. [Google Scholar] [CrossRef]
  82. Kuschner, R.A.; Russell, K.L.; Abuja, M.; Bauer, K.M.; Faix, D.J.; Hait, H.; Henrick, J.; Jacobs, M.; Liss, A.; Lynch, J.A.; et al. A phase 3, randomized, double-blind, placebo-controlled study of the safety and efficacy of the live, oral adenovirus type 4 and type 7 vaccine, in U.S. military recruits. Vaccine 2013, 31, 2963–2971. [Google Scholar] [CrossRef] [PubMed]
  83. Narsana, N.; Ha, D.; Ho, D.Y. Treating Adenovirus Infection in Transplant Populations: Therapeutic Options Beyond Cidofovir? Viruses 2025, 17, 599. [Google Scholar] [CrossRef]
  84. Morens, D.M.; Taubenberger, J.K.; Fauci, A.S. Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses. Cell Host Microbe 2023, 31, 146–157. [Google Scholar] [CrossRef]
  85. Cox, R.M.; Sourimant, J.; Toots, M.; Yoon, J.; Ikegame, S.; Govindarajan, M.; Watkinson, R.E.; Thibault, P.; Makhsous, N.; Lin, M.J.; et al. Orally efficacious broad-spectrum allosteric inhibitor of paramyxovirus polymerase. Nat. Microbiol. 2020, 5, 1232–1246. [Google Scholar] [CrossRef]
  86. Chemaly, R.F.; Marty, F.M.; Wolfe, C.R.; Lawrence, S.J.; Dadwal, S.; Soave, R.; Farthin, J.; Hawley, S.; Montanez, P.; Hwang, J.; et al. DAS181 Treatment of Severe Lower Respiratory Tract Parainfluenza Virus Infection in Immunocompromised Patients: A Phase 2 Randomized, Placebo-Controlled Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2021, 73, e773–e781. [Google Scholar] [CrossRef]
  87. Chemaly, R.F.; Shah, D.; Boeckh, M.J. Management of respiratory viral infections in hematopoietic cell transplant recipients and patients with hematologic malignancies. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2014, 59, S344–S351. [Google Scholar] [CrossRef]
  88. Mortezavi, M.; Sloan, A.; Singh, R.S.P.; Chen, L.F.; Kim, J.H.; Shojaee, N.; Toussi, S.S.; Prybylski, J.; Baniecki, M.L.; Bergman, A.; et al. Virologic Response and Safety of Ibuzatrelvir, A Novel SARS-CoV-2 Antiviral, in Adults with COVID-19. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2025, 80, 673–680. [Google Scholar] [CrossRef]
  89. Furuya, C.; Yasuda, H.; Hiki, M.; Shirane, S.; Yamana, T.; Uchimara, A.; Inano, T.; Takaku, T.; Hamano, Y.; Ando, M. Case report: Ensitrelvir for treatment of persistent COVID-19 in lymphoma patients: A report of two cases. Front. Immunol. 2024, 15, 1287300. [Google Scholar] [CrossRef] [PubMed]
  90. Babusis, D.; Kim, C.; Yang, J.; Zhao, X.; Geng, G.; Ip, C.; Kozon, N.; Le, H.; Leung, J.; Pitts, J.; et al. Pharmacokinetics and Metabolism of Broad-Spectrum Antivirals Remdesivir and Obeldesivir with a Consideration to Metabolite GS-441524: Same, Similar, or Different? Viruses 2025, 17, 836. [Google Scholar] [CrossRef]
  91. Horga, A.; Kuritzkes, D.R.; Kowalczyk, J.J.; Pietropaolo, K.; Belanger, B.; Lin, K.; Perkins, K.; Hammond, J. Phase II study of bemnifosbuvir in high-risk participants in a hospital setting with moderate COVID-19. Future Virol. 2023, 18, 489–500. [Google Scholar] [CrossRef]
  92. Ewart, G.; Bobardt, M.; Bentzen, B.H.; Yan, Y.; Thomson, A.; Klumpp, K.; Becker, S.; Rosenkilde, M.M.; Miller, M.; Gallay, P. Post-infection treatment with the E protein inhibitor BIT225 reduces disease severity and increases survival of K18-hACE2 transgenic mice infected with a lethal dose of SARS-CoV-2. PLoS Pathog. 2023, 19, e1011328. [Google Scholar] [CrossRef]
  93. Ahmad, A.; Eze, K.; Noulin, N.; Horvathova, V.; Murray, B.; Baillet, M.; Grey, L.; Mori, J.; Adda, N. EDP-938, a Respiratory Syncytial Virus Inhibitor, in a Human Virus Challenge. N. Engl. J. Med. 2022, 386, 655–666. [Google Scholar] [CrossRef]
  94. Levene, R.E.; DeVincenzo, J.; Conery, A.L.; Ahmad, A.; Or, Y.S.; Rhodin, M.H.J. EDP-938 Has a High Barrier to Resistance in Healthy Adults Experimentally Infected with Respiratory Syncytial Virus. J. Infect. Dis. 2025, 231, e290–e298. [Google Scholar] [CrossRef]
  95. Sevendal, A.T.K.; Hurley, S.; Bartlett, A.W.; Rawlinson, W.; Walker, G.J. Systematic Review of the Efficacy and Safety of RSV-Specific Monoclonal Antibodies and Antivirals in Development. Rev. Med. Virol. 2024, 34, e2576. [Google Scholar] [CrossRef]
  96. Falsey, A.R.; Koval, C.; DeVincenzo, J.P.; Walsh, E.E. Compassionate use experience with high-titer respiratory syncytical virus (RSV) immunoglobulin in RSV-infected immunocompromised persons. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2017, 19, e12657. [Google Scholar] [CrossRef] [PubMed]
  97. Jalili, A.; Hajifathali, A.; Mohammadian, M.; Sankanian, G.; Sayahinouri, M.; Dehghani Ghorbi, M.; Roshandel, E.; Aghdami, N. Virus-Specific T Cells: Promising Adoptive T Cell Therapy Against Infectious Diseases Following Hematopoietic Stem Cell Transplantation. Adv. Pharm. Bull. 2023, 13, 469–482. [Google Scholar] [CrossRef] [PubMed]
Table 1. Influenza Vaccines and Antivirals in Transplant Recipients.
Table 1. Influenza Vaccines and Antivirals in Transplant Recipients.
AgentClass/MechanismDosing & ScheduleTimingClinical/EfficacySafety
Fluzone High-DoseHD-IIV3 (inactivated)60 µg hemagglutinin/0.5 mLAnnual; pre-Tx 1 preferred [15]; ≥1 month post-SOT [11], ≥3–6 month post-HCT [12]↑ Seroconversion vs. standard; ↓ pneumonia, ICU, mortality [8]Safe; mild AEs 2
FluadaIIV3 (adjuvanted)15 µg hemagglutinin/0.5 mLSame as aboveMF59 adjuvant enhances immune response [15]Well-tolerated
Oseltamivir (Tamiflu)Neuraminidase inhibitor75 mg PO daily ×12 weeks (pre-exp) or ×7 days (post-exp) [3]Pre-exposure if vaccine suboptimal [3]; post-exposure as treatment↓ Influenza complications; improved outcomes if given within 48 h [8]Safe; renally adjust
Baloxavir marboxil (Xofluza)Cap-dependent endonuclease inhibitorSingle PO dose, consider in combo with NAI [19]Early outpatient (<48 h) [19]Comparable efficacy to oseltamivir; ↑ resistance risk [22]Limited data in SOT
CD388Investigational neuraminidase inhibitor conjugateSingle injection (24 week efficacy) [23]Seasonal pre-exposure76% efficacy in healthy adults; non-vaccine option [23]Phase 2b; well tolerated [23]
1 “Tx”—transplant. 2 “AEs”—adverse effects.
Table 2. SARS-CoV-2 Vaccines, Monoclonal Antibodies, and Antivirals.
Table 2. SARS-CoV-2 Vaccines, Monoclonal Antibodies, and Antivirals.
AgentClass/MechanismDosing & ScheduleTimingClinical/EfficacySafety
mRNA-1273 (Spikevax)mRNA vaccine4-dose (0, 4 weeks, +4 weeks, 6 month booster)Pre-Tx 1 preferred but should also repeat post-Tx 1; ≥1 month post-SOT [28], ≥3 months post-HCT [24]20–40% seroconversion after 2 doses; improved ≥3 [31,33]No rejection/GVHD 2 [31,43]
mRNA BNT162b2 (Comirnaty)mRNA vaccine4-dose (0, 3 weeks, +4 weeks, 6 month booster)Same as aboveBoosters improve humoral response [38]Safe in SOT/HCT [24]
Pemivibart (Pemgarda)Long-acting mAb4500 mg IV q 3 monthsPre-Tx 1 preferred↓ Hospitalization; variant-dependent [44]EUA 3, limited data
Remdesivir (Veklury)RNA-polymerase inhibitor3–5 days IVEarly (<7 days) outpatient or inpatient [47]↓ Hospitalization/mortality [46,47]Safe; monitor renal function
Nirmatrelvir-Ritonavir (Paxlovid)Protease inhibitor combination5–15 days POEarly (<5 days) [49]↓ Viral load; monitor DDIs 4CYP3A interaction risk [48]
Molnupiravir (Lagevrio)Nucleoside analog5 days POEarly (<5 days) [50]↓ Hospitalization; less effective than remdesivir and nirmatrelvir-ritonavir [50]Well-tolerated
1 “Tx”—transplant. 2 “GVHD”—graft versus host disease. 3 “EUA”—emergency use authorization. 4 “DDIs”—drug drug interactions.
Table 3. RSV Vaccines and Monoclonal Antibodies.
Table 3. RSV Vaccines and Monoclonal Antibodies.
AgentClass/MechanismDosingTimingClinical/EfficacySafety
AbrysvoProtein subunit (non-adjuvanted) vaccineSingle pre-season dose [54]Pre-Tx 1 preferred; ≥3–6 months post-Tx 1 [12,61]50–69% VE 2; 1-year antibody persistence; Lower response in HCT (29–44%) [58,59]Rare GBS 3; no rejection [58,61]
ArexvyProtein subunit (adjuvanted) vaccineSame as aboveSame as above50–69% VE 2; improved CD4+ response; preferred for immunocompromised [61]Rare GBS 3; mild AEs 4 [58]
mRESVIAmRNA vaccineSame as above, not preferredSame as above, not preferredPotential in high-risk SOT [56]Safe in PLWH 5; awaiting SOT data [70]
NirsevimabLong-acting mAb (F-protein)Single IM pre-season, adult dose unknownNo adult data70% efficacy in infants; no adult data; potential SOT use [53]Safe in infants
1 “Tx”—transplant. 2 “VE”—vaccine efficacy. 3 “GBS”—Guillain-Barré Syndrome. 4 “AEs”—adverse effects. 5 “PLWH”—persons living with HIV.
Table 4. Novel Agents Under Investigation.
Table 4. Novel Agents Under Investigation.
VirusAgentMechanismStageFindingsStatus in Transplant
SARS-CoV-2IbuzatrelvirProtease inhibitorPhase 2↓ Viral load [88]No data
SARS-CoV-2EnsitrelvirProtease inhibitorPhase 3↓ Symptoms [89]Potential benefit in IC 1 [89]
SARS-CoV-2ObeldesivirRNA pol inhibitorPhase 3↓ Symptoms [90]No data
SARS-CoV-2BemnifosbuvirRNA pol inhibitorPhase 2↓ Viral load [91]No data
SARS-CoV-2BIT225Envelope protein inhibitorPhase 2Did not meet primary efficacy endpoint [92]Investigational
RSVRibavirinNucleoside analog (inhaled preferred)Approved↓ LRTI progression ± IVIG/mAb, ?effect in lung Tx 2 [3,66]Standard care
RSVMolnupiravirNucleoside analogApproved↓ Symptoms [69]No data
RSVEDP-938Nucleoprotein inhibitorPhase 2↓ Viral load/symptoms [93,94]No data
RSVALN-RSV01siRNA (inhaled)Phase 2↓ BOS 3 in lung Tx 2; safe [95]Promising adjunct
RSVRI-001 and RI-002RSV Ab-rich IVIGApprovedEarly use may improve outcomes in IC [96]Potential benefit in HCT, no SOT data [96]
Other RVIDAS-181Sialidase fusion protein (inhaled)Phase 2Benefit in select immunocompromised [86]Investigational
Other RVIT-cell adoptive therapySeropositive donor T cellsPhase 1–2↓ Viral load, improve survival [97]Investigational, promising in HCT
1 “IC”—immunocompromised. 2 “Tx”—transplant. 3 “BOS”—bronchiolitis obliterans syndrome.
Table 5. Comparative Pre-Exposure and Post-Exposure Prophylaxis Strategies.
Table 5. Comparative Pre-Exposure and Post-Exposure Prophylaxis Strategies.
VirusPre-ExposurePrEP 1 TimingPost-ExposurePEP 2 TimingNotes
InfluenzaAnnual IIV; oseltamivir ×12 weeksAnnual vaccine: ≥1 months post-SOT, ≥3–6 months post-HCTOseltamivir ×7 daysBest within 48 hVaccine cornerstone; antiviral bridge
SARS-CoV-2mRNA booster vaccines; Pemivibart q 3 monthsBoosters plus annual vaccine, timing same as aboveRemdesivir, nirmatrelvir-ritonavir, molnupiravirEarly within 7 daysVariant-dependent prophylaxis
RSVSingle vaccine; mAb Annual pre-seasonRibavirin ± IVIG/mAbEarlyAdjuvanted vaccines enhance immunity
Other RVITrial vaccines (hMPV, PIV)Vaccine-specificCidofovir, ribavirinCase-specificEncourage trial enrollment
1 “PrEP”—pre-exposure prophylaxis. 2 “PEP”—post-exposure prophylaxis.
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

Giuliani, A.A.M.; Chen, V.; Law, N. Respiratory Viral Infection Prophylaxis and Treatment in the Transplant Population. Viruses 2026, 18, 8. https://doi.org/10.3390/v18010008

AMA Style

Giuliani AAM, Chen V, Law N. Respiratory Viral Infection Prophylaxis and Treatment in the Transplant Population. Viruses. 2026; 18(1):8. https://doi.org/10.3390/v18010008

Chicago/Turabian Style

Giuliani, Adriana A. M., Victor Chen, and Nancy Law. 2026. "Respiratory Viral Infection Prophylaxis and Treatment in the Transplant Population" Viruses 18, no. 1: 8. https://doi.org/10.3390/v18010008

APA Style

Giuliani, A. A. M., Chen, V., & Law, N. (2026). Respiratory Viral Infection Prophylaxis and Treatment in the Transplant Population. Viruses, 18(1), 8. https://doi.org/10.3390/v18010008

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