Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Viruses ; 15(5)2023 05 11.
Article in English | MEDLINE | ID: covidwho-20243425

ABSTRACT

Antibody-dependent enhancement of infection (ADE) is clinically relevant to Dengue virus (DENV) infection and poses a major risk to the application of monoclonal antibody (mAb)-based therapeutics against related flaviviruses such as the Zika virus (ZIKV). Here, we tested a two-tier approach for selecting non-cross-reactive mAbs combined with modulating Fc glycosylation as a strategy to doubly secure the elimination of ADE while preserving Fc effector functions. To this end, we selected a ZIKV-specific mAb (ZV54) and generated three ZV54 variants using Chinese hamster ovary cells and wild-type (WT) and glycoengineered ΔXF Nicotiana benthamiana plants as production hosts (ZV54CHO, ZV54WT, and ZV54ΔXF). The three ZV54 variants shared an identical polypeptide backbone, but each exhibited a distinct Fc N-glycosylation profile. All three ZV54 variants showed similar neutralization potency against ZIKV but no ADE activity for DENV infection, validating the importance of selecting the virus/serotype-specific mAbs for avoiding ADE by related flaviviruses. For ZIKV infection, however, ZV54CHO and ZV54ΔXF showed significant ADE activity while ZV54WT completely forwent ADE, suggesting that Fc glycan modulation may yield mAb glycoforms that abrogate ADE even for homologous viruses. In contrast to the current strategies for Fc mutations that abrogate all effector functions along with ADE, our approach allowed the preservation of effector functions as all ZV54 glycovariants retained antibody-dependent cellular cytotoxicity (ADCC) against the ZIKV-infected cells. Furthermore, the ADE-free ZV54WT demonstrated in vivo efficacy in a ZIKV-infection mouse model. Collectively, our study provides further support for the hypothesis that antibody-viral surface antigen and Fc-mediated host cell interactions are both prerequisites for ADE, and that a dual-approach strategy, as shown herein, contributes to the development of highly safe and efficacious anti-ZIKV mAb therapeutics. Our findings may be impactful to other ADE-prone viruses, including SARS-CoV-2.


Subject(s)
COVID-19 , Dengue Virus , Dengue , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Mice , Cricetinae , Zika Virus/genetics , CHO Cells , Dengue Virus/genetics , Cricetulus , SARS-CoV-2 , Antibodies, Viral , Antibodies, Monoclonal/therapeutic use , Cross Reactions , Antibodies, Neutralizing/therapeutic use
2.
Am J Transl Res ; 15(5): 3040-3059, 2023.
Article in English | MEDLINE | ID: covidwho-20233431

ABSTRACT

Recent data have revealed various effector functions of FcγRs in immune responses against challenges with SARS-CoV-2 virus. FcγRs act as a bridge between antibody specificity and effector cells. In many cases, IgG/FcγR interactions generate cell-mediated immune protection from infection via ADCP or ADCC. These responses are beneficial, as they may participate in virus elimination and persist longer than neutralizing anti-Spike antibodies. In contrast, these interactions may sometimes prove beneficial to the virus by enhancing viral uptake into phagocytic cells via ADE and causing excessive inflammation. Here, we summarize key features of FcγRs, discuss effector functions, clinical relevance, and factors influencing FcγR-mediated immune responses in COVID-19 and vaccine responses, and consider IVIg and kinase inhibitors for targeting FcγRs signaling in COVID-19.

3.
Cureus ; 14(11): e31877, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2307222

ABSTRACT

The COVID-19 pandemic caused by SARS-CoV-2 spread across many countries between 2020 and 2022. The similarities in clinical presentation with other endemic diseases pose a challenge to physicians in effectively diagnosing and treating the infection. Approximately 129 nations have a risk of dengue infection, and more than 100 of those are endemic to dengue. During the COVID-19 pandemic, the number of dengue cases decreased in many countries owing to the isolation measures followed. However, the common clinical presentation between them has led to misdiagnosis. Both COVID-19 and dengue fever cause a surge in pro-inflammatory cytokines and chemokines, thus sharing a common pathophysiology. False positive serological test results also posed difficulty differentiating between COVID-19 and dengue fever. This review aims to compare the clinical features, pathophysiology, and immune response between dengue and COVID-19, to benefit public health management during the pandemic.

4.
Microorganisms ; 11(4)2023 Apr 13.
Article in English | MEDLINE | ID: covidwho-2300703

ABSTRACT

Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies produced in the body after infection or vaccination may enhance subsequent viral infections in vitro and in vivo. Although rare, symptoms of viral diseases are also enhanced by ADE following infection or vaccination in vivo. This is thought to be due to the production of antibodies with low neutralizing activity that bind to the virus and facilitate viral entry, or antigen-antibody complexes that cause airway inflammation, or a predominance of T-helper 2 cells among the immune system cells which leads to excessive eosinophilic tissue infiltration. Notably, ADE of infection and ADE of disease are different phenomena that overlap. In this article, we will describe the three types of ADE: (1) Fc receptor (FcR)-dependent ADE of infection in macrophages, (2) FcR-independent ADE of infection in other cells, and (3) FcR-dependent ADE of cytokine production in macrophages. We will describe their relationship to vaccination and natural infection, and discuss the possible involvement of ADE phenomena in COVID-19 pathogenesis.

6.
Vaccines (Basel) ; 11(4)2023 Mar 31.
Article in English | MEDLINE | ID: covidwho-2306114

ABSTRACT

Antibody-dependent enhancement (ADE) can increase the rates and severity of infection with various viruses, including coronaviruses, such as MERS. Some in vitro studies on COVID-19 have suggested that prior immunization enhances SARS-CoV-2 infection, but preclinical and clinical studies have demonstrated the contrary. We studied a cohort of COVID-19 patients and a cohort of vaccinated individuals with a heterologous (Moderna/Pfizer) or homologous (Pfizer/Pfizer) vaccination scheme. The dependence on IgG or IgA of ADE of infection was evaluated on the serum samples from these subjects (twenty-six vaccinated individuals and twenty-one PCR-positive SARS-CoV-2-infected patients) using an in vitro model with CD16- or CD89-expressing cells and the Delta (B.1.617.2 lineage) and Omicron (B.1.1.529 lineage) variants of SARS-CoV-2. Sera from COVID-19 patients did not show ADE of infection with any of the tested viral variants. Some serum samples from vaccinated individuals displayed a mild IgA-ADE effect with Omicron after the second dose of the vaccine, but this effect was abolished after the completion of the full vaccination scheme. In this study, FcγRIIIa- and FcαRI-dependent ADE of SARS-CoV-2 infection after prior immunization, which might increase the risk of severe disease in a second natural infection, was not observed.

7.
Vaccine ; 41(15): 2427-2429, 2023 04 06.
Article in English | MEDLINE | ID: covidwho-2263857

ABSTRACT

A potential risk associated with vaccines for COVID-19 is antibody-dependent disease enhancement (ADE) in which vaccine induced antibody mediated immune responses may lead to enhanced SARS CoV- 2 acquisition or increased disease severity. Though ADE has not been clinically demonstrated with any of the COVID-19 vaccines so far, when neutralizing antibodies are suboptimal, the severity of COVID-19 has been reported to be greater. ADE is presumed to occur via abnormal macrophages induced by the vaccine based immune response by antibody-mediated virus uptake into Fc gamma receptor IIa (FcγRIIa) or by the formation of Fc-mediated excessive antibody effector functions. Beta-glucans which are naturally occurring polysaccharides known for unique immunomodulation by capability to interact with macrophages, eliciting a specific beneficial immune-response and enhancing all arms of the immune system, importantly without over-activation are suggested as safer nutritional supplement-based vaccine adjuvants for COVID-19.


Subject(s)
COVID-19 Vaccines , COVID-19 , beta-Glucans , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Immunoglobulin Fc Fragments , SARS-CoV-2 , Vaccination
8.
Nonlinear Dyn ; : 1-16, 2022 Oct 06.
Article in English | MEDLINE | ID: covidwho-2235811

ABSTRACT

The advent and swift global spread of the novel coronavirus (COVID-19) transmitted by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused massive deaths and economic devastation worldwide. Antibody-dependent enhancement (ADE) is a common phenomenon in virology that directly affects the effectiveness of the vaccine, and there is no fully effective vaccine for diseases. In order to study the potential role of ADE on SARS-CoV-2 infection, we establish the SARS-CoV-2 infection dynamics model with ADE. The basic reproduction number is computed. We prove that when R 0 < 1 , the infection-free equilibrium is globally asymptotically stable, and the system is uniformly persistent when R 0 > 1 . We carry out the sensitivity analysis by the partial rank correlation coefficients and the extended version of the Fourier amplitude sensitivity test. Numerical simulations are implemented to illustrate the theoretical results. The potential impact of ADE on SARS-CoV-2 infection is also assessed. Our results show that ADE may accelerate SARS-CoV-2 infection. Furthermore, our findings suggest that increasing antibody titers can have the ability to control SARS-CoV-2 infection with ADE, but enhancing the neutralizing power of antibodies may be ineffective to control SARS-CoV-2 infection with ADE. Our study presumably contributes to a better understanding of the dynamics of SARS-CoV-2 infection with ADE.

9.
Front Immunol ; 13: 1050478, 2022.
Article in English | MEDLINE | ID: covidwho-2198891

ABSTRACT

Our review summarizes the evidence that COVID-19 can be complicated by SARS-CoV-2 infection of immune cells. This evidence is widespread and accumulating at an increasing rate. Research teams from around the world, studying primary and established cell cultures, animal models, and analyzing autopsy material from COVID-19 deceased patients, are seeing the same thing, namely that some immune cells are infected or capable of being infected with the virus. Human cells most vulnerable to infection include both professional phagocytes, such as monocytes, macrophages, and dendritic cells, as well as nonprofessional phagocytes, such as B-cells. Convincing evidence has accumulated to suggest that the virus can infect monocytes and macrophages, while data on infection of dendritic cells and B-cells are still scarce. Viral infection of immune cells can occur directly through cell receptors, but it can also be mediated or enhanced by antibodies through the Fc gamma receptors of phagocytic cells. Antibody-dependent enhancement (ADE) most likely occurs during the primary encounter with the pathogen through the first COVID-19 infection rather than during the second encounter, which is characteristic of ADE caused by other viruses. Highly fucosylated antibodies of vaccinees seems to be incapable of causing ADE, whereas afucosylated antibodies of persons with acute primary infection or convalescents are capable. SARS-CoV-2 entry into immune cells can lead to an abortive infection followed by host cell pyroptosis, and a massive inflammatory cascade. This scenario has the most experimental evidence. Other scenarios are also possible, for which the evidence base is not yet as extensive, namely productive infection of immune cells or trans-infection of other non-immune permissive cells. The chance of a latent infection cannot be ruled out either.


Subject(s)
COVID-19 , Animals , Humans , SARS-CoV-2 , Antibodies, Viral , Virus Internalization , Phagocytes
10.
Front Immunol ; 13: 1008285, 2022.
Article in English | MEDLINE | ID: covidwho-2154728

ABSTRACT

Since immune system and internal environment in vivo are large and complex, the interpretation of the observed immune effect from the perspective of a single immune cell or antibody seems a little feeble. Many studies have shown that specific antibodies against " former" viruses have a reduced ability to neutralize "new" mutant strains. However, there is no comprehensive and clear view of whether there will be Antibody-dependent enhancement (ADE). We review the latest relevant studies, hoping to explain the ADE of SARS-CoV-2 infection sometimes observed in some patients.


Subject(s)
Antibody-Dependent Enhancement , COVID-19 , Humans , SARS-CoV-2 , Antibodies, Viral
11.
Mol Immunol ; 152: 172-182, 2022 12.
Article in English | MEDLINE | ID: covidwho-2105610

ABSTRACT

Antibody-dependent enhancement (ADE) has been associated with severe disease outcomes in several viral infections, including respiratory infections. In vitro and in vivo studies showed that antibody-response to SARS-CoV and MERS-CoV could exacerbate infection via ADE. Recently in SARS CoV-2, the in vitro studies and structural analysis shows a risk of disease severity via ADE. This phenomenon is partially attributed to non-neutralizing antibodies or antibodies at sub-neutralizing levels. These antibodies result in antigen-antibody complexes' deposition and propagation of a chronic inflammatory process that destroys affected tissues. Further, antigen-antibody complexes may enhance the internalization of the virus into cells through the Fc gamma receptor (FcγR) and lead to further virus replication. Thus, ADE occur via two mechanisms; 1. Antibody mediated replication and 2. Enhanced immune activation. Antibody-mediated effector functions are mainly driven by complement activation, and the first complement in the cascade is complement 1q (C1q) which binds to the virus-antibody complex. Reports say that deficiency in circulating plasma levels of C1q, an independent predictor of mortality in high-risk patients, including diabetes, is associated with severe viral infections. Complement mediated ADE is reported in several viral infections such as dengue, West Nile virus, measles, RSV, Human immunodeficiency virus (HIV), and Ebola virus. This review discusses ADE in viral infections and the in vitro evidence of ADE in coronaviruses. We outline the mechanisms of ADE, emphasizing the role of complements, especially C1q in the outcome of the enhanced disease.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome , Humans , Antibody-Dependent Enhancement , Complement C1q , Antigen-Antibody Complex , Antibodies, Viral
12.
BMC Infect Dis ; 22(1): 742, 2022 Sep 19.
Article in English | MEDLINE | ID: covidwho-2038669

ABSTRACT

Coronavirus disease 2019 (COVID-19) continues to constitute an international public health emergency. Vaccination is a prospective approach to control this pandemic. However, apprehension about the safety of vaccines is a major obstacle to vaccination. Amongst health professionals, one evident concern is the risk of antibody-dependent enhancement (ADE), which may increase the severity of COVID-19. To explore whether ADE occurs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and increase confidence in the safety of vaccination, we conducted a meta-analysis to investigate the relationship between post-immune infection and disease severity from a population perspective. Databases, including PubMed, EMBASE, Chinese National Knowledge Infrastructure, SinoMed, Scopus, Science Direct, and Cochrane Library, were searched for articles on SARS-CoV-2 reinfection published until 25 October 2021. The papers were reviewed for methodological quality, and a random effects model was used to analyse the results. Heterogeneity was assessed using the I2 statistic. Publication bias was evaluated using a funnel plot and Egger's test. Eleven studies were included in the final meta-analysis. The pooled results indicated that initial infection and vaccination were protective factors against severe COVID-19 during post-immune infection (OR = 0.55, 95%CI = 0.31-0.98). A subgroup (post-immune infection after natural infection or vaccination) analysis showed similar results. Primary SARS-CoV-2 infection and vaccination provide adequate protection against severe clinical symptoms after post-immune infection. This finding demonstrates that SARS-CoV-2 may not trigger ADE at the population level.


Subject(s)
COVID-19 , Vaccines , Antibodies, Viral , Antibody-Dependent Enhancement , COVID-19/prevention & control , Humans , SARS-CoV-2 , Vaccination
13.
Biomed Eng Adv ; 4: 100054, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-2031157

ABSTRACT

With severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as an emergent human virus since December 2019, the world population is susceptible to coronavirus disease 2019 (COVID-19). SARS-CoV-2 has higher transmissibility than the previous coronaviruses, associated by the ribonucleic acid (RNA) virus nature with high mutation rate, caused SARS-CoV-2 variants to arise while circulating worldwide. Neutralizing antibodies are identified as immediate and direct-acting therapeutic against COVID-19. Single-domain antibodies (sdAbs), as small biomolecules with non-complex structure and intrinsic stability, can acquire antigen-binding capabilities comparable to conventional antibodies, which serve as an attractive neutralizing solution. SARS-CoV-2 spike protein attaches to human angiotensin-converting enzyme 2 (ACE2) receptor on lung epithelial cells to initiate viral infection, serves as potential therapeutic target. sdAbs have shown broad neutralization towards SARS-CoV-2 with various mutations, effectively stop and prevent infection while efficiently block mutational escape. In addition, sdAbs can be developed into multivalent antibodies or inhaled biotherapeutics against COVID-19.

14.
Cell Rep ; 40(7): 111214, 2022 08 16.
Article in English | MEDLINE | ID: covidwho-1966424

ABSTRACT

Vaccine-associated enhanced respiratory disease (VAERD) is a severe complication for some respiratory infections. To investigate the potential for VAERD induction in coronavirus disease 2019 (COVID-19), we evaluate two vaccine leads utilizing a severe hamster infection model: a T helper type 1 (TH1)-biased measles vaccine-derived candidate and a TH2-biased alum-adjuvanted, non-stabilized spike protein. The measles virus (MeV)-derived vaccine protects the animals, but the protein lead induces VAERD, which can be alleviated by dexamethasone treatment. Bulk transcriptomic analysis reveals that our protein vaccine prepares enhanced host gene dysregulation in the lung, exclusively up-regulating mRNAs encoding the eosinophil attractant CCL-11, TH2-driving interleukin (IL)-19, or TH2 cytokines IL-4, IL-5, and IL-13. Single-cell RNA sequencing (scRNA-seq) identifies lung macrophages or lymphoid cells as sources, respectively. Our findings imply that VAERD is caused by the concerted action of hyperstimulated macrophages and TH2 cytokine-secreting lymphoid cells and potentially links VAERD to antibody-dependent enhancement (ADE). In summary, we identify the cytokine drivers and cellular contributors that mediate VAERD after TH2-biased vaccination.


Subject(s)
COVID-19 , Vaccines , Animals , Antibodies, Viral , Cricetinae , Cytokines/metabolism , Immunization , Lung/pathology , Mice , Mice, Inbred BALB C , Th1 Cells , Th2 Cells , Vaccination
15.
Front Immunol ; 13: 889196, 2022.
Article in English | MEDLINE | ID: covidwho-1957157

ABSTRACT

The dynamics of host-virus interactions, and impairment of the host's immune surveillance by dengue virus (DENV) serotypes largely remain ambiguous. Several experimental and preclinical studies have demonstrated how the virus brings about severe disease by activating immune cells and other key elements of the inflammatory cascade. Plasmablasts are activated during primary and secondary infections, and play a determinative role in severe dengue. The cross-reactivity of DENV immune responses with other flaviviruses can have implications both for cross-protection and severity of disease. The consequences of a cross-reactivity between DENV and anti-SARS-CoV-2 responses are highly relevant in endemic areas. Here, we review the latest progress in the understanding of dengue immunopathogenesis and provide suggestions to the development of target strategies against dengue.


Subject(s)
COVID-19 , Dengue Virus , Dengue , Antibodies, Viral , Antibody-Dependent Enhancement , Humans
16.
J Indian Inst Sci ; 102(2): 671-687, 2022.
Article in English | MEDLINE | ID: covidwho-1943765

ABSTRACT

Antibody-dependent enhancement (ADE) is an alternative route of viral entry in the susceptible host cell. In this process, antiviral antibodies enhance the entry access of virus in the cells via interaction with the complement or Fc receptors leading to the worsening of infection. SARS-CoV-2 variants pose a general concern for the efficacy of neutralizing antibodies that may fail to neutralize infection, raising the possibility of a more severe form of COVID-19. Data from various studies on respiratory viruses raise the speculation that antibodies elicited against SARS-CoV-2 and during COVID-19 recovery could potentially exacerbate the infection through ADE at sub-neutralizing concentrations; this may contribute to disease pathogenesis. It is, therefore, of utmost importance to study the effectiveness of the anti-SARS-CoV-2 antibodies in COVID-19-infected subjects. Theoretically, ADE remains a general concern for the efficacy of antibodies elicited during infection, most notably in convalescent plasma therapy and in response to vaccines where it could be counterproductive.

17.
Cell Rep ; 39(9): 110904, 2022 05 31.
Article in English | MEDLINE | ID: covidwho-1926270

ABSTRACT

Despite SARS-CoV-2 being a "novel" virus, early detection of anti-spike IgG in severe COVID-19 patients may be caused by the amplification of humoral memory responses against seasonal coronaviruses. Here, we examine this phenomenon by characterizing anti-spike IgG responses in non-hospitalized convalescent individuals across a spectrum of COVID-19 severity. We observe that disease severity positively correlates with anti-spike IgG levels, IgG cross-reactivity against other betacoronaviruses (ß-CoVs), and FcγR activation. Analysis of IgG targeting ß-CoV-conserved and non-conserved immunodominant epitopes within the SARS-CoV-2 spike protein revealed epitope-specific relationships: IgG targeting the conserved heptad repeat (HR) 2 region significantly correlates with milder disease, while targeting the conserved S2'FP region correlates with more severe disease. Furthermore, a lower HR2-to-S2'FP IgG-binding ratio correlates with greater disease severity, with ICU-hospitalized COVID-19 patients showing the lowest HR2/S2'FP ratios. These findings suggest that HR2/S2'FP IgG profiles may predict disease severity and offer insight into protective versus deleterious humoral recall responses.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Immunoglobulin G , Seasons , Spike Glycoprotein, Coronavirus
18.
Aims Allergy and Immunology ; 6(2):64-70, 2022.
Article in English | Web of Science | ID: covidwho-1896955

ABSTRACT

The SARS-CoV-2 virus causes the COVID-19 disease associated with over 6.2 million deaths globally. Multiple early indicators raised the potential risk of the SARS-CoV-2 virus infecting monocytes and macrophages via Fc-receptor antibody binding based on closely related beta coronaviruses. Antibody Fc-receptor infection of phagocytic monocytes and macrophages is one type of antibody dependent enhancement of disease. Increased COVID-19 severity correlated with early high antibody responses on initial infection for unvaccinated adults. Clinical evidence suggests that for moderate antibody titer levels, antibodies binding to SARS-CoV-2 may contribute to viral spread, cytokine dysregulation, and enhanced COVID-19 disease severity. Primary immune responses appear to have too low of antibody titer to significantly contribute to Fc-receptor uptake by monocytes and macrophages for COVID-19 patients. Very high antibody titers created by SARS-CoV-2 vaccines also appear to inhibit Fc-receptor uptake and infection of monocytes and macrophages;this inhibition appears to decrease as antibody titer levels decrease. Cross reactive antibodies to other coronaviruses or moderate levels of SARS-CoV-2 antibodies may be contributing to antibody dependent enhancement of disease in critical COVID-19 patients.

19.
Clin Epidemiol Glob Health ; 14: 100966, 2022.
Article in English | MEDLINE | ID: covidwho-1797102

ABSTRACT

The COVID-19 outbreak sparked by SARS-CoV-2, begat significant rates of malady worldwide, where children with an abnormal post-COVID ailment called the Multisystem Inflammatory Syndrome (MIS-C), were reported by April 2020. Here we have reviewed the clinical characteristics of the pediatric patients and the prognosis currently being utilized. A vivid comparison of MIS-C with other clinical conditions has been done. We have addressed the probable etiology and fundamental machinery of the inflammatory reactions, which drive organ failure. The involvement of androgen receptors portrays the likelihood of asymptomatic illness in children below adolescence, contributing to the concept of antibody-dependent enhancement.

20.
Alergologia Polska-Polish Journal of Allergology ; 9(1):14-32, 2022.
Article in Polish | Web of Science | ID: covidwho-1761031

ABSTRACT

Publication indicates that antiviral therapies are definitely more harmful and less effective as well as less accessible than vaccinations. Antiviral therapies are generally not effective in phase II and III of the disease. The use of antiviral therapies in all patients would doom them to many side effects, and only 10% of patients go from phase I to phase II and require such treatment. The chance of curing COVID-19 patients in phase II and III of the disease is rapidly declining. Vaccinations, on the other hand, are very effective. Confirmed AEFI are rare and intensive research is underway to elucidate their etiopathogenesis. From a biotechnological point of view, vaccination leads to the production of CAR-T-like immunotherapy or monoclonal antibodies, that work from the 1st day of infection, not from the first day of symptom onset, when patients can start some pharmacological therapy. In the case of COVID-19, a clear advantage of vaccination over pharmacological therapy has emerged. Nevertheless, because vaccination may not be effective for all patients, working on therapy should also be intensively carried out. Proper pharmacotherapy administration depends on the quick cooperation of diagnosticians with doctors. It also depends on the development of tests to select therapies and determine in whom a disease such as COVID-19 will change from phase I to phase II. Vaccinated patients will probably respond better to treatments.

SELECTION OF CITATIONS
SEARCH DETAIL