Abstract
The current epidemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is raising awareness of the need to act faster when dealing with new pathogens. Exposure to an emerging pathogen generates an antibody response that can be used for preventing and treating the infection. These antibodies might have a high specificity to a target, few side effects, and are useful in the absence of an effective vaccine for treating immunocompromised individuals. The approved antibodies against the receptor-binding domain (RBD) of the viral spike protein of SARS-CoV-2 (e.g., regdanvimab, bamlanivimab, etesevimab, and casirivimab/imdevimab) have been selected from the antibody repertoire of B cells from convalescent patients using flow cytometry, next-generation sequencing, and phage display. This encourages use of these techniques especially phage display, because it does not require expensive types of equipment and can be performed on the lab bench, thereby making it suitable for labs with limited resources. Also, the antibodies in blood samples from convalescent patients can be used to screen pre-made peptide libraries to identify epitopes for vaccine development. Different types of vaccines against SARS-CoV-2 have been developed, including inactivated virus vaccines, mRNA-based vaccines, non-replicating vector vaccines, and protein subunits. mRNA vaccines have numerous advantages over existing vaccines, such as efficacy, ease of manufacture, safety, and cost-effectiveness. Additionally, epitope vaccination may constitute an attractive strategy to induce high levels of antibodies against a pathogen and phages might be used as immunogenic carriers of such peptides. This is a point worth considering further, as phage-based vaccines have been shown to be safe in clinical trials and phages are easy to produce and tolerate high temperatures. In conclusion, identification of the antibody repertoire of recovering patients, and the epitopes they recognize, should be an attractive alternative option for developing therapeutic and prophylactic antibodies and vaccines against emerging pathogens.
Keywords Emerging pathogen, SARS-CoV-2, passive antibody therapy, peptide vaccine, phage display, therapeutic antibody
Introduction
Despite the impact of improved sanitation and the availability of antibiotics and vaccines, infectious diseases are still the leading cause of death worldwide. Each year, many new infections threaten the health of the local and world populations. Several factors that may influence the appearance of emerging and re-emerging infectious diseases have been described by Morse, including microbial adaptation, ecological and demographic factors, movement of people and goods, industry, and worsening public health services.1
Recently, the outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), known as coronavirus disease 2019 (COVID19), began in Wuhan and spread rapidly around the world. This outbreak resulted in more than 144,099,374 confirmed cases, with more than 3,061,912 deaths worldwide by 23 April 2021.2 Infection control focuses on quarantining infected persons and restricting the mobility of persons while vaccinating the world population until the threat disappears. Globalization and the movement of people are spreading new, emerging, and re-emerging pathogens worldwide. Therefore, the scientific community must be ready to put all its skills into the search for ways to act quickly to save lives. Advances in technology and knowledge in the life sciences make it possible to identify and isolate epitopes derived from a pathogen that has induced an immune response and the antibodies that have been generated during the infection. This article discusses the prospects for passive antibody therapy and active immunization, including peptide-based vaccines to combat pathogens, with a particular focus on emerging and re-emerging pathogens.
Methods
Literature search
An extensive literature research was conducted using keyword filters to select articles related to "therapeutic antibodies", "neutralizing antibodies", "peptide vaccine" in combination with "SARS-CoV-2", "emerging" or "re-emerging pathogen", and "infectious diseases". This research was carried out on articles published in the PubMed and Scopus databases for the English language from 1 January 2020 to 12 December 2020. Also, articles on the SARS-CoV2 2 were examined. Information about approved therapeutic antibodies was collected from the Antibody Society website,3 the incidence of COVID-19 cases was obtained from the World Health Organization (WHO) website,2 and information of the approved COVID-19 vaccines was obtained from the Regulatory Affairs Professionals Society (RAPS) website.4 The review process is graphically shown in Figure 1.
Clinical trials search
The entire database at ClinicalTrials.gov was searched on 10 January 2021 using the following search terms: condition or disease = "infectious disease"; other terms = "antibody" OR "therapeutic antibody" OR "monoclonal antibodies" OR "therapeutic peptide" OR "peptide vaccine". The number of clinical trials registered for therapeutic antibodies during the last ten years were counted for all diseases and infectious diseases. Tables were prepared that listed pathogen-specific antibodies, or therapeutic antibodies specific for human proteins, that were used for treatment of infectious diseases and were registered during 2020. Furthermore, a list was prepared of peptide vaccines that were used for the prevention of infectious diseases and that were registered in the last ten years.
Literature review
Emerging pathogens
In addition to SARS-CoV-2, several other emerging pathogens have been reported to infect humans during the last two decades,5 including human bocavirus,6 human coronavirus HKU1,7 human T-lymphotropic virus (HTLV) 3 and 4,8'9 human coronavirus NL63,10 SARS coronavirus,11 human metapneumovirus,12 Middle East respiratory syndrome coronavirus (MERS-CoV),13 and severe fever with thrombocytopenia syndrome (SFTS) virus.14 For example, the human bocavirus was identified for the first time in humans in 2005 in children with respiratory tract infections in Sweden. The human coronavirus HKU1 was isolated in January 2004 from a 71-year-old Chinese man who had been hospitalized with pneumonia and bronchiolitis. HTLV-3 and -4 were isolated in 2005 from two hunters of monkeys from the southern forests of Cameroon. The human coronavirus NL63 was isolated in 2004 from a 7-month-old child with bronchiolitis from the Netherlands. Cases of SARS coronavirus were first reported in China in 2002. The human metapneumovirus was isolated in 2001 from young children in the Netherlands. MERS-CoV was first reported in Saudi Arabia in 2012. SFTS was first clinically identified in a patient with several symptoms including fever and thrombocytopenia in China in 2009.
Some examples from before 2000 include the Nipah virus that appeared in 1998, Ebola in 1976, human immunodeficiency virus (HIV) in 1959, Marburg virus in 1967, and Lassa fever in 1969. Outbreaks of unknown infectious diseases often become a challenge due to lack of treatment and diagnosis, malnutrition, and the emergence of drug-resistant pathogens.
Antibody response
Exposure to an emerging pathogen, such as SARS-CoV-2, generates an antibody response that changes over time and between individuals. IgM antibodies against SARS-CoV-2 become detectable during the week following infection and are present for a month before progressively diminishing, while IgG antibodies are detectable within ten to 21 days up to a minimum of three months.15 The survival of individuals with severe SARS-CoV-2 infection can be attributed in part to a rapid and potent IgG class switching with high-affinity FcR binding capacities.16 Consequently, antibodies from patients who recover from COVID-19 or other emerging infections can provide valuable information about their neutralization and opsonization capabilities and the epitopes they recognize. The identification of these epitopes and the production of antibodies against them should be one of the main objectives of emerging pathogen research and this is what this review suggests.
Passive and active immunization
Convalescent plasma and therapeutic antibodies might be useful treatments for emergent pathogens when no other treatment is available, while vaccines are prophylactic and are designed to induce a specific host immune response against pathogens.
Convalescent plasma
Convalescent plasma therapy may be the most straightforward option for reducing symptoms and mortality in infected patients when vaccines or drugs are unavailable, but with varying degrees of success. The WHO gave priority to assessing treatment with convalescent whole blood or plasma derived from patients who recovered from Ebola virus disease during the outbreak in West Africa in 2014-2015. However, the report of Griensven et al. indicated that the convalescent plasma therapy did not induce a significant reduction of mortality among Ebola patients.17
On 23 August 2020, the Food and Drug Administration (FDA) issued an emergency use authorization for convalescent plasma as a potentially promising treatment of critically ill COVID-19 patients.18 They concluded that the convalescent plasma may be effective in reducing the severity or length of COVID-19 illness in some hospitalized patients. The advantages of convalescent plasma outweigh the potential risks in the absence of appropriate, approved and available alternative therapies. Since then, numerous studies have been published about how to apply this type of therapy to severe and critically ill COVID-19 patients.19-28 The FDA released a new evaluation that indicates that convalescent plasma no longer meets clear criteria of being "maybe effective".29 According to the new information available, it seems that high titer convalescent plasma should only be given at the beginning of the infection (within 3 days of symptom onset) and it is probably not beneficial after 8 days or more after the onset of symptoms. Therefore, convalescent plasma therapy should be considered with some discretion, but alternative approaches should also be explored.
Passive antibody therapy
Administration of purified antibodies may be an alternative to convalescent plasma transfusion. It has been speculated that patients with COVID19 could be treated with intravenous immunoglobulin (IVIg) or specific human monoclonal antibodies (mAbs) that inhibit SARS-CoV-2 binding to angiotensin-converting enzyme 2 (ACE-2) receptors and its entry into human cells.30'31 The receptor binding domain (RBD) of the spike glycoprotein (S) protein on the surface of SARS-CoV-2 is responsible for the attachment of the virus to ACE-2 to gain access to host cells.32 This interaction can be neutralized with antibodies that compete with ACE-2 for binding to RBD. Neutralizing antibody levels depends on the time after the onset of symptoms, age of the patient, and the severity of the disease. It is not clear what level of neutralizing antibodies is needed to ensure protection against infection and re-infection.
The number of new therapeutic antibodies entering clinical trials in all stages of development increased over time (a linear trend in phases 1 to 4) for all diseases including infectious diseases (Figure 2). Between January 2020 and December 2020, 429 trials were initiated for therapeutic antibodies against different diseases compared with 41 (9.6%) trials against infectious diseases. Among them, 23 antibodies targeted pathogens (Table 1), while the other 18 antibodies targeted human molecules (Table 2). Twenty-six clinical trials were evaluating antibodies against SARS-CoV-2, 6 focused on HIV, 2 on malaria, 1 on herpes simplex virus 1 (HSV-1), 1 on chikungunya virus, 1 on BK virus, 1 on rabies virus, 1 on dengue virus, 1 on Pseudomonas aeruginosa, and 1 on Clostridioides difficile. Moreover, a significant number of neutralizing antibodies have not been evaluated in clinical trials.
Jin and Simmons reviewed the available literature on neutralizing antibodies that block chikungunya virus from binding to host receptors thereby preventing membrane fusion and virus entry into cells.33 Chikungunya virus is a type of emerging mosquito-transmitted alphavirus that can cause acute or chronic polyarthritis and foot swelling, and is currently spreading rapidly from endemic regions of Africa and Asia to Europe and America.34 Corti et al. isolated LCA60, a potent neutralizing antibody derived from the memory B cells of an individual who had been infected with MERS-CoV. This antibody recognizes an epitope in the viral S protein and blocks virus binding to the host receptor CD26, reducing the disease and virus titers in a relevant mouse model.35,36
Right now, there is an increasing tendency to develop neutralizing antibodies as a result of COVID-19. Antibodies to treat COVID-19 are currently being tested in a number of clinical trials (See Tables 1 and 2). On 9 November 2020, FDA issued an emergency use authorization (EUA) to Eli Lilly and Company for the combination of antibodies bamlanivimab (LYCoV555) and etesevimab (LY-CoV016, also known as CB6 or JS016).37 These are antibodies produced by B cells from convalescent patients that target the SARS-CoV-2 S protein. Antigenspecific memory B cells were identified and isolated by flow cytometry and the VH and VL were amplified, cloned, and produced in an expression system. On 20 November 2020, Regeneron Pharmaceuticals obtained approval for the therapeutic application of the antibody cocktail casirivimab/ imdevimab (REGN10987+REGN10933),38 which contains both humanized mice antibodies and antibodies from blood samples from recovered COVID-19 patients.3 The antibody variable regions were determined by next-generation sequencing, and the repertoire for the heavy and light chain pairs was identified, cloned, and produced in an expression system. Recently, the European Medicines Agency (EMA) approved the use of the antibody regdanvimab (also known as CT-P59) for treating COVID-19.39 It functions by targeting the RBD of the viral S protein. Regdanvimab is a fully human immunoglobulin reformatted from a single-chain variable fragment that was selected from a phage display antibody library created by B cells from a convalescent patient.40 Approved therapeutic antibodies against SARS-CoV-2 are shown in Table 4.
Only few antibodies have been approved to treat infections in humans (e.g., ruxibacumab, obiltoxaximab, palivizumab, bezlotoxumab, ibalizumab, KamRAB, etc.) compared to the approved antibodies for other types of disease such as cancer. Raxibacumab and obiltoxaximab neutralize the binding of the anthrax exotoxins to host receptors and their application is for the treatment of Bacillus anthracis infections.41 Palivizumab is specific for the F-glycoprotein of the respiratory syncytial virus (RSV) and has been successfully used prophylactically for severe RSV disease especially in infants.42 Bezlotoxumab has been developed for the prevention of Clostridioides difficile infection recurrence, ibalizumab prescribed for HIV,43 and KamRAB for rabies infection.44 Approved therapeutic antibodies against different pathogens are shown in Table 5.
The increasing tendency to develop therapeutic antibodies against infectious diseases over the past ten years indicates that they have been accepted as an effective evolving treatment modality that may have a major clinical impact in the future.
Immune phage display libraries
Monoclonal antibodies can be generated by animal immunization and hybridoma techniques or by recombinant protein production.45 However, the identification and recovery of individual antigen-specific antibodies from the antibody repertoire from peripheral blood mononuclear cells (PBMCs) from a human blood donor can be done using flow cytometry,46 nextgeneration sequencing,47 or different in vitro display techniques (e.g., phage display,48 bacterial display,49 yeast display,50 or mammalian cell surface display51). Flow cytometry is a common method used for single B cell analysis and isolation and the next-generation sequencing enables simultaneous reading of up to several million variants of a single gene. However, the main disadvantage of these techniques is the high cost of their production equipment which makes them inaccessible to many labs, especially in developing countries. In contrast, phage display is a technique that only requires the types of equipment available in the majority of microbiology labs and can be done on the lab bench.
The genetic information encoding the selected antibodies from phage display libraries is immediately available which makes it easy to reclone the isolated antibodies for further manipulation and expression. In addition, it can be used against toxic antigens, with no need for animals, the selection process is fast, and with it human antibodies can be obtained. Some disadvantages of phage display are that it is a laborious process to construct a phage display library and to obtain a library with sufficient diversity to represent the antibody repertoire. Raxibacumab and regdanvimab are the only antibodies approved by the FDA and EMA against pathogens, that had been isolated from phage display libraries. Raxibacumab was selected from a naive library, also generated from healthy individuals. However, regdanvimab was isolated from a immune phage display library constructed from B cells from a convalescent COVID-19 patient. The immune libraries may be a better option for infectious diseases52 because they mimic a natural vaccination with a live pathogen in a human host. This is an attractive option to obtain antibodies against immunogenic epitopes since they can be obtained from the V-gene repertoire specific to an infection. Currently, the antibody foravirumab (CR4098), selected from a phage display library constructed from the B cell repertoire of rabies-vaccinated individuals,53 is in a phase II clinical trial. This indicates that phage display could be further exploited in finding therapeutic antibodies to treat infectious diseases as it has been exploited in oncology54 and autoimmune diseases.55
Developing immune or disease-specific antibody libraries helps understanding antibody reactions against specific disease antigens.
Several studies have used phage display to isolate human antibodies specific for bacterial, viral, and parasitic antigens. Recently, a set of fully human neutralizing antibodies was selected from phage display libraries generated from peripheral B cells collected from convalescent or severe COVID-19 patients. Several of these antibodies bind to distinct epitopes on the viral spike RBD and possess neutralizing activity for RBD binding to the human ACE2 receptor.56 In another study, panning a phage-displayed singledomain antibody library against RBD and S1 subunit identified neutralizing antibodies that could be promising candidates for therapy of COVID-19.57
Antibodies to other pathogens than SARSCoV-2 were identified through the use of immune libraries. For example, immune libraries against pentavalent botulinum toxoid (BoNT/A) revealed that single-chain variable fragments (scFvs) recognized a limited number of immunodominant epitopes exposed on the binding domain of BoNT/A. One group of these antibody fragments exhibited neutralizing activity.58 Another example is when an scFv phage library was generated from a patient infected with HIV-1 subtype B. Screening of this library with recombinant soluble gp140 envelope proteins led to the identification of scFv clones that specifically recognize a conserved pocket binding domain in gp41.59 Ekiert et al. identified antibodies from phage display libraries created from B cells that were collected from Turkish patients who survived H5N1 avian flu infection. These antibodies recognize conserved elements of the receptor-binding site on the hemagglutinin surface glycoprotein, enabling the neutralization of strains of multiple subtypes of influenza A viruses, including H1, H2, and H3.60 In a study of helminth parasite infections, scFv fragments against a specific filarial antigen (BmR1) were isolated from an immune library developed with the repertoire of antibodies from individuals who had been confirmed positive for filariasis infection.61
Active immunization with peptides
Active immunity is defined as the process of exposing an individual to an antigen to generate an adaptive immune response.62 Vaccines are generally considered the gold standard for infection control and, in normal situations, require many years to develop.
According to the WHO, as of 23 April 2021, 91 vaccine candidates were under clinical development to treat COVID-19, and 184 candidate vaccines were in pre-clinical development.63 More than 973 million vaccine doses have been administered worldwide, while 223 million people have been fully vaccinated by 23 April 23 2021.64
Currently, different types of vaccines against SARS-CoV-2 have been developed including inactivated virus vaccines (e.g., CoronaVac, BBIBP-CorV, etc.), mRNA-based vaccines (e.g., BNT162b2 and mRNA-1273), non-replicating vector vaccines (e.g., Convidicea, AZD1222, and Janssen), protein-based vaccines (e.g., ZF2001), and peptide vaccines (e.g., EpiVacCorona).65-67 Except for the inactivated virus vaccines, most of them target the surface glycoprotein (S-protein) of SARS-CoV-2 and were showed to have a minimal number of side effects, be risk-free, and efficient in phase III clinical trials. Approved vaccines against SARS-CoV-2 are shown in Table 6.
Since peptides can be selected using serum from infected patients, they should represent or mimic the immunogenic epitopes that triggered the immune response. Therefore, epitope vaccination may constitute an attractive strategy to induce high levels of antibodies against a pathogen.68 EpiVacCorona is a Russian SARSCoV-2 peptide vaccine approved in Belarus, Russia, and Turkmenistan. Two peptide vaccines are under development, UB-612 developed by the U.S.-based company Vaxxinity which is in phase 2/3 trial, and pVAC, developed by the University Hospital Tuebingen in Germany and is in phase 1 trial.4
Peptide vaccines have been applied to induce neutralizing antibodies in different mice models using epitopes of the varicella-zoster virus,69 Helicobacter pylori,70 H1N1 influenza virus,71 Ebola virus,72 Plasmodium falciparum,73 Nipah virus,74 and Tropheryma whipplei.75 There are 36 clinical trials evaluating peptide vaccines against different pathogens, primarily in phases I and II, of which only ten have begun in the past ten years (Table 3). Interestingly, only one study began in 2020 which was for SARS-CoV-2, and another is expected to begin during 2021 for Streptococcus pyogenes. The SARS-CoV-2 study is in phase I trial for evaluating a multi-peptide vaccine (CoVac-1) (ClinicalTrials.gov, identifier NCT04546841) that was developed based on identified multiple immunodominant human leukocyte antigen-D related (DR) T-cell epitopes in COVID-19 convalescent individuals.76 In addition, several other studies are underway to develop peptidebased vaccines for SARS-CoV-2.77
One advantage of peptide-based vaccines is that they may eliminate many side effects of allergic and auto-immune reactions associated with classical whole-organism vaccines.78 Recently, a melanoma peptide vaccine was tested for immunogenicity and safety in 47 participants by Slingluff and colleagues.79 The peptides were synthesized and purified in GMP conditions and the participants were vaccinated with them in an emulsion with incomplete Freund's adjuvant or/and polyICLC. They were found to be well tolerated, had clinical activity, and could elicit CD4+ T lymphocyte responses in most participants. Li et al. reviewed the progress and challenges of peptide vaccines,80 Kaumaya et al. reviewed the peptide vaccines in cancer therapy,81 and Di Natale et al. reviewed peptide-based vaccines for COVID-19.82 Synthetic peptides are easily synthesized, cost-effective, and relatively safe.78'83'84 One disadvantage of peptide-based vaccine is that peptides or a single epitope alone are not enough to induce a sufficiently high immune response, so they must be conjugated with biopolymers, encapsulated in nanoparticles85 or applied to carrier or adjuvants68 to improve the immune response. Virus-like particles can be an alternative to the nanoparticle delivery system of peptides including bacteriophages.86
The use of phages as immunogenic carriers of peptides could be an attractive vaccine approach. Roehnisch et al. used phage particles as immunological carriers in a clinical phase I/II trial to vaccinate with idiotype vaccine 15 patients with advanced multiple myeloma.87 They concluded that this approach might be a simple, time- and cost-efficient phage idiotype vaccination strategy. Therefore, this is an approach worthy of further consideration. Greenwood et al. showed that a peptide corresponding to Plasmodium falciparum surface protein displayed on phages was highly immunogenic, not requiring an adjuvant to induce an antibody response.88 Currently, a phase II clinical trial is evaluating phage vaccine SNS301 in patients with high-risk myelodysplastic syndromes and chronic myelomonocytic leukemia (ClinicalTrials.gov, identifier NCT04217720). Nonetheless, the potential for phage vaccination against pathogens, including emerging and re-emerging pathogens, needs to be further explored, in particular the behavior of phages in the human body and how they interact with cellular components.89 However, this is a promising way of vaccination because it can first select peptide displaying phages using convalescent plasma or convalescent antibodies, and then evaluate them as vaccine candidates.
Peptide libraries
Many phage display peptide libraries have been constructed and used extensively over the years to characterize epitopes.90'91 They allow us to select peptides that may share common sequence patterns or have similarities to proteins recorded in protein databases (e.g., NCBI) and thereby identify potential antigens from a pathogen. Screening random peptide libraries with antibodies from convalescent human serum from patients with SARS-CoV infections led to the identification of phages that were specifically bound to these antibodies. Phage derived peptides also show extensive sequence homology with structural proteins of SARS-CoV such as S and M-protein.92 Zamecnik et al. screened a programmable phage display library (HuCoV VirScan), containing overlapping peptide antigens from several coronavirus types (e.g., SARS-CoV-2), with COVID-19 patient sera. Peptide phages encoding the S (residues 783-839 and 1,124-1,178) and N proteins (residues 209265 and 142-208) were enriched during panning against human sera and used to develop serological tests for SARS-CoV-2 detection.93
Discussion
When an unknown pathogen appears and causes an outbreak, the B cells of the recovering patients become a valuable resource of the antibody repertoire that patients express during infections. This repertoire can be used in convalescent plasma transfusion or to screen for therapeutic antibodies and to identify immunogenic epitopes.
Passive antibody therapy can be an alternative to convalescent plasma therapy because the latter therapy may cause side effects, such as autoimmune reactions and its results can be unpredictable due to variability between lots. In recent years, the percent of approvals for antibodies has been increasing. Even so, there are only a few successful therapeutic monoclonal antibodies against infectious diseases that have reached the market compared with antibodies in other therapeutic areas.
An important challenge that therapeutic antibodies face is the strong competition with well-established treatments with antibiotics, vaccines, and anti-viral compounds. Laura DeFrancesco published an interesting article in which a group of experts commented on challenges with antibody therapy.94 There are only a few successful therapeutic antiviral antibodies compared to antibodies in other medical fields such as cancer. Expectations for antiviral antibody therapy seemed to be low because of the high cost of production in comparison to the profits. It is not a profitable market for pharmaceutical companies, as emerging infectious diseases are rare and are mainly affecting developing countries. However, the cost could be reduced by producing antibody fragments such as scFv, antigen-binding fragments (Fab), or nanobodies in prokaryotic systems.95 Different expression systems (e.g., mammalian, yeast, and prokaryotic) are suitable for both soluble and functionally active protein expression. Escherichia coli was found to be a faster and more cost-effective expression system than Pichia pastoris and Chinese hamster ovary (CHO).96 However, it has higher levels of impurities and breakdown products than the other two systems. Therefore, an additional downstream purification step would be required to remove these impurities to improve the quality of antibodies obtained by this system for therapeutic purposes.96 There are Fab fragments successfully generated in E. coli and approved by the FDA, such as certolizumab pegol (for treatment of Crohn's disease, rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis)97 and ranibizumab (anti-vascular endothelial growth factor).98 This encourages investment in prokaryotic production of Fabs or other forms of antibody therapies for the prevention and treatment of emerging or reemerging infectious diseases.
Even with a vaccine, antibodies could still be needed to treat people who do not respond significantly to vaccination (e.g., infants, elderly, and immunocompromised individuals). From a technical point of view, by Fc region engineering, the half-life of therapeutic antibodies can be increased and their functions such as antibodydependent cellular cytotoxicity and antibodydependent cellular phagocytosis can be controlled by modulating their recognition by effector cells of the immune system.99 It is interesting to note that smaller-sized antibodies such as Fabs, scFvs, and nanobodies that can be obtained by phage display can penetrate cells and target intracellular viral proteins.100 This means that passive antibody therapy with small antibodies could have valuable potential for targeting intracellular pathogens that escape the host immune response.
An alternative promising therapy for infectious diseases is gene therapy. The pathogen can be neutralized by supplying genes coding recombinant antibodies against the pathogen in a gene therapy vector, such as adeno-associated viral vectors (AVAs) to be expressed by the host's own cells (e.g., muscle cells).101 Currently, an antibody gene therapy is being evaluated in a phase I clinical trial (VRC 603), which is testing the safety of adeno-associated virus vector expressing an HIV-1 neutralizing antibody that was originally isolated from the blood of a person with HIV infection (VRC 603 trial is available on ClinicalTrials.gov using identifier NCT03374202).
Human B cell cloning might be the most common approach to identify neutralizing antibodies, and phage display is one of the techniques used to isolate the antibody repertoire from the convalescent B cells. Regdanvimab, an anti-SARS-CoV-2 antibody, is an example of phage display's effectiveness since it is a technique that can be used in any lab without requiring costly equipment, making it accessible to developing countries where the majority of emerging diseases occur and samples are easy to obtain. However, there are some drawbacks, such as the fact that it is a time-consuming process and there is a need to create a new library for each pathogen or disease. Another issue is the formation of artifacts during VH and VL combinations or the fact that certain bacterial clones grow faster than others. However, this can be avoided by continuously monitoring each step during library creation, for example by sequencing multiple clones and ensuring that the bacteria culture grows only for a short period of time.
Peptide vaccine is a promising alternative or complement to the already existing therapeutic approaches. However, more work is required to develop peptide vaccines before they can be used in clinical settings. The most difficult aspect of developing an epitope-based vaccine is determining the epitopes of interest with proper accuracy. Another challenge with peptide vaccine is the low immunogenicity which could be improved with nontoxic adjuvants. Another limitation is that multiple peptides may be required because a single peptide may not provide a sufficient immune response. Also, peptides that are susceptible to enzymatic digestion may need to be encapsulated and delivered with nanoparticles such as copolymer poly(lactic-co-glycolic acid) (PLGA).102'103 The safety of the delivery approach is of utmost concern when selecting strategies for peptide delivery. However, PLGA is an FDA-approved polymer for use in drug delivery, diagnostics, and other applications that have been in the market for many years.
mRNA vaccines represent a new type of vaccine and they have several advantages over other vaccines including safety, efficacy, speed of production, and cost-effectiveness.104 The mRNA enters the cells and is used to make viral antigen proteins from inside the cell, which involves natural, post-translational modifications. mRNA vaccine production does not require cell cultures and/ or fermentation-based manufacturing processes. They can be naturally degraded with no metabolic toxicity and mRNA vaccines do not enter the nucleus. Some of their disadvantages include the fact that they are not as stable as DNA and proteins and are susceptible to degradation. Also, there are few studies on mRNA vaccines for bacteria and parasites. A major disadvantage of mRNA vaccines is their instability at high temperatures, which makes them unavailable in remote locations. Therefore, phage-based vaccines are a strategy worth further consideration, as phages are stable at high temperatures.
Henry et al. reviewed studies of the filamentous bacteriophages' properties, as well as their various applications in research.105 Filamentous phage can be used to display polypeptides as fusion proteins with the major coat protein pVIII. In addition, each phage can display more than 2500 copies of pVIII, making it a good way to present peptides to the immune system. Phage-based vaccines are easy to develop and do not require expensive equipment, so they may be a useful tool for studying emerging and re-emerging pathogens in developing countries.
Conclusions
In conclusion, the antibody repertoire of recovering patients and the epitopes they recognized should be considered as a scientific priority for the development of therapeutic and prophylactic antibodies and vaccines against emerging pathogens. Several tools can be taken advantage of that can help us improve their efficacy and reduce their production costs, such as phage display, antibody engineering, and prokaryotic protein expression.
Conflicts of interest: none to declare.
Funding: None to declare.
Acknowledgments: Special thanks to David Wade for taking the time to read and revise the manuscript. Also, special thanks to the useful comments from the editor and reviewers at GERMS.
Ethics approval: Not required
Received: 16 January 2021; revised: 25 April 2021; accepted: 01 June 2021.
Please cite this article as: Palma M. Perspectives on passive antibody therapy and peptide-based vaccines against emerging pathogens like SARS-CoV-2. GERMS. 2021;11(2):287-305. doi: 10.18683/germs.2021.1264
*Corresponding author: Marco Palma, [email protected]
References
1. Morse SS. Factors in the emergence of infectious diseases. Emerg Infect Dis. 1995;1:7-15. https://doi.org/10.3201/eid0101.950102
2. World Health Organization. 2021. WHO coronavirus disease (COVID-19) dashboard. Accessed on: 23 April 2021. Available at: https: / / covid19.who.int/? gclid=EAIaI0obChMIxYXlu dnP6gIVVIXVCh1zu08iEAAYASAAEgK00fD BwE.
3. Antibody Society. 2021. COVID-19 biologics tracker - Clinical studies evaluating anti-SARS-CoV-2 monoclonal antibodies. Accessed on: 19 April 2021. Available at: https://www.antibodysociety.org/covid-19biologics-tracker/.
4. Regulatory Affairs Professionals Society. 2021. COVID19 vaccine tracker. Accessed on: 19 April 2021. Available at: https://www.raps.org/news-andarticles/news-articles/2020/3/covid-19-vaccine-tracker
5. Woolhouse ME, Howey R, Gaunt E, Reilly L, ChaseTopping M, Savill N. Temporal trends in the discovery of human viruses. Proc Biol Sci. 2008;275:2111-5. https://doi.org/10.1098/rspb.2008.0294
6. Allander T, Tammi MT, Eriksson M, Bjerkner A, Tiveljung-Lindell A, Andersson B. Cloning of a human parvovirus by molecular screening of respiratory tract samples. Proc Natl Acad Sci U S A. 2005;102:12891-6. https:/ / doi.org/10.1073/pnas.0504666102
7. Woo PCY, Lau SK, Chu CM, et al. Characterization and complete genome sequence of a novel coronavirus, coronavirus HKU1, from patients with pneumonia. J Virol. 2005;79:884-95. https://doi.org/10.1128/JVI.79.2.884-895.2005
8. Calattini S, Chevalier SA, Duprez R, et al. Discovery of a new human T-cell lymphotropic virus (HTLV-3) in Central Africa. Retrovirology. 2005;2:30. https://doi.org/10.1186/1742-4690-2-30
9. Wolfe ND, Heneine W, Carr JK, et al. Emergence of unique primate T-lymphotropic viruses among central African bushmeat hunters. Proc Natl Acad Sci U S A. 2005;102:7994-9. https://doi.org/10.1073/pnas.0501734102
10. van der Hoek L, Pyrc K, Berkhout B. Human coronavirus NL63, a new respiratory virus. FEMS Microbiol Rev. 2006;30:760-73. https://doi.org/10.1111/i.1574-6976.2006.00032.x
11. Rota PA, Oberste MS, Monroe SS, et la. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science. 2003;300:1394-9. https:/ / doi.org/10.1126/science.1085952
12. Hamelin M, Abed Y, Bo ivin G. Human metapneumovirus: a new player among respiratory viruses. Clin Infect Dis. 2004;38:983-90. https://doi.org/10.1086/382536
13. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME, Fouchier RAM. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367:1814-20. https://doi.org/10.1056/NEiMoa1211721
14. Yu XJ, Liang MF, Zhang SY, et al. Fever with thrombocytopenia associated with a novel bunyavirus in China. N Engl J Med. 2011;364:1523-32. https://doi.org/10.1056/NEiMoa1010095
15. Zhang G, Nie S, Zhang Z, Zhang Z. Longitudinal change of severe acute respiratory syndrome coronavirus 2 antibodies in patients with coronavirus disease 2019. J Infect Dis. 2020;222:183-8. https:/ / doi.org/10.1093/infdis/jiaa229
16. Zohar T, Loos C, Fischinger S, et al. Compromised humoral functional evolution tracks with SARS-CoV-2 mortality. Cell. 2020;183:1508-19.e12. https:/ / doi.org/10.1016/j.cell.2020.10.052
17. van Griensven J, Edwards T, de Lamballerie X, et al. Evaluation of convalescent plasma for ebola virus disease in Guinea. N Engl J Med. 2016;374:33-42. https://doi.org/10.1056/NEJMoa1511812
18. The United States Food and Drug Administration. 2020. FDA Issues emergency use authorization for convalescent plasma as potential promising COVID-19 treatment, another achievement in administration's fight against pandemic. Accessed on: 14 April 2021. Available at: https://www.fda.gov/news-events/pressannouncements/fda-issues-emergency-use-authorizationconvalescent-plasma-potential-promising-covid-19treatment.
19. Agarwal A, Mukherjee A, Kumar G, et al. Convalescent plasma in the management of moderate COVID-19 in adults in India: open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ. 2020;371:m3939. https://doi.org/10.1136/bmi.m3939
20. Simonovich VA, Burgos Pratx LD, Scibona P, et al. A randomized trial of convalescent plasma in COVID-19 severe pneumonia. N Engl J Med. 2021;384:619-29. https://doi.org/10.1056/NElMoa2031304
21. Libster R, Pérez Marc G, Wappner D, et al. Early hightiter plasma therapy to prevent severe COVID-19 in older adults. N Engl J Med. 2021;384:610-8. https://doi.org/10.1056/NEMoa2033700
22. Salazar E, Christensen PA, Graviss EA, et al. Significantly decreased mortality in a large cohort of coronavirus disease 2019 (COVID-19) patients transfused early with convalescent plasma containing high-titer anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein IgG. Am J Pathol. 2021;191:90-107. https:/ / doi.org/ 10.1016/i .aipath.2020.10.008
23. Shenoy AG, Hettinger AZ, Fernandez SJ, Blumenthal J, Baez V. Early mortality benefit with COVID-19 convalescent plasma: a matched control study. Br J Haematol. 2021;192:706-13. https://doi.org/10.1111/bih.17272
24. Alsharidah S, Ayed M, Ameen RM, et al. COVID-19 convalescent plasma treatment of moderate and severe cases of SARS-CoV-2 infection: a multicenter interventional study. Int J Infect Dis. 2021;103:439-46. https://doi.org/10.1016/i.iiid.2020.11.198
25. Rogers R, Shehadeh F, Mylona EK, et al. Convalescent plasma for patients with severe COVID-19: a matched cohort study. Clin Infect Dis. 2020:ciaa1548. https:/ / doi.org/10.1093/ cid/ ciaa1548
26. Joyner MJ, Carter RE, Senefeld JW, et al. Convalescent plasma antibody levels and the risk of death from COVID-19. N Engl J Med. 2021;384:1015-27. https://doi.org/10.1056/NEMoa2031893
27. Clark E, Guilpain P, Filip IL, et al. Convalescent plasma for persisting COVID-19 following therapeutic lymphocyte depletion: a report of rapid recovery. Br J Haematol. 2020;190:e154-6. https://doi.org/10.1111/bih.16981
28. Ferrari S, Caprioli C, Weber A, Rambaldi A, Lussana F. Convalescent hyperimmune plasma for chemoimmunotherapy induced immunodeficiency in COVID-19 patients with hematological malignancies. Leuk Lymphoma. 2021;62:1490-6. https://doi.org/10.1080/10428194.2021.1872070
29. The United States Food and Drug Administration. Clinical memorandum - COVID-19 convalescent plasma. Accessed on: 14 April 2021. Available at: https://www.fda.gov/media/141480/download.
30. Gasparyan AY, Misra DP, Yessirkepov M, Zimba O. Perspectives of immune therapy in coronavirus disease 2019. J Korean Med Sci. 2020;35:e176. https://doi.org/10.3346/ikms.2020.35.e176
31. Jawhara S. Could intravenous immunoglobulin collected from recovered coronavirus patients protect against COVID-19 and strengthen the immune system of new patients? Int J Mol Sci. 2020;21:2272. https:/ / doi.org/ 10.3390/iims21072272
32. Ju B, Zhang Q, Ge J, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature. 2020;584:115-9. https://doi.org/10.1038/s41586-020-2380-z
33. Jin J, Simmons G. Antiviral functions of monoclonal antibodies against chikungunya virus. Viruses. 2019;11:305. https://doi.org/10.3390/v11040305
34. Weaver SC, Charlier C, Vasilakis N, Lecuit M. Zika, chikungunya, and other emerging vector-borne viral diseases. Annu Rev Med. 2018;69:395-408. https:/ / doi.org/10.1146/annurev-med-050715-105122
35. Corti D, Zhao J, Pedotti M, et al. Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proc Natl Acad Sci U S A. 2015;112:10473-8. https://doi.org/10.1073/pnas.1510199112
36. de Wit E, Feldmann F, Horne E, et al. Prophylactic efficacy of a human monoclonal antibody against MERS-CoV in the common marmoset. Antiviral Res. 2019;163:70-4. https:/ / doi.org/ 10.1016/i .antiviral.2019.01.016
37. Shi R, Shan C, Duan X, et al. A human neutralizing antibody targets the receptor-binding site of SARSCoV-2. Nature. 2020;584:120-4. https://doi.org/10.1038/s41586-020-2381-y
38. Hansen J, Baum A, Pascal KE, et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science. 2020;369:1010-4. https:/ / doi.org/10.1126/science.abd0827
39. European Medicines Agency. 2021. EMA issues advice on use of regdanvimab for treating COVID-19. Accessed on: 11 April 2021. Available at: https://www.ema.europa.eu/en/news/ema-issuesadvice-use-regdanvimab-treating-covid-19.
40. Kim C, Ryu DK, Lee J, et al. A therapeutic neutralizing antibody targeting receptor binding domain of SARSCoV-2 spike protein. Nat Commun. 2021;12:288. https://doi.org/10.1038/s41467-020-20602-5
41. Wang-Lin SS, Balthasar JP. Pharmacokinetic and pharmacodynamic considerations for the use of monoclonal antibodies in the treatment of bacterial infections. Antibodies (Basel). 2018;7:5. https:/ / doi.org/10.3390/ antib7010005
42. Beeler JA, van Wyke Coelingh K. Neutralization epitopes of the F glycoprotein of respiratory syncytial virus: effect of mutation upon fusion function. J Virol. 1989;63:2941-50. https://doi.org/10.1128/ivi.63.7.2941-2950.1989
43. Lu RM, Hwang YC, Liu IJ, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci. 2020;27:1. https://doi.org/10.1186/s12929-019-0592-z
44. Matson MA, Schenker E, Stein M, Zamfirova V, Nguyen HB, Bergman GE. Safety and efficacy results of simulated post-exposure prophylaxis with human immune globulin (HRIG; KEDRAB) co-administered with active vaccine in healthy subjects: a comparative phase 2/3 trial. Hum Vaccin Immunother. 2020;16:452-9. https://doi.org/10.1080/21645515.2019.1656967
45. Cole SP. Monoclonal antibodies. Can Fam Physician. 1987;33:369-72.
46. Gross A, Schoendube J, Zimmermann S, Steeb M, Zengerle R, Koltay P. Technologies for single-cell isolation. Int J Mol Sci. 2015;16:16897-919. https://doi.org/10.3390/ijms160816897
47. Lushova AA, Biazrova MG, Prilipov AG, Sadykova GK, Kopylov TA, Filatov AV. Next-generation techniques for discovering human monoclonal antibodies. Mol Biol. 2017;51:782-7. https://doi.org/10.1134/S0026893317060103
48. Throsby M, van den Brink E, Jongeneelen M, et al. Heterosubtypic neutralizing monoclonal antibodies cross-protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS One. 2008;3:e3942. https:/ / doi.org/ 10.1371/journal.pone.0003942
49. Harvey BR, Georgiou G, Hayhurst A, Jeong KJ, Iverson BL, Rogers GK. Anchored periplasmic expression, a versatile technology for the isolation of high-affinity antibodies from Escherichia coii-expressed libraries. Proc Natl Acad Sci U S A. 2004;101:9193-8. https:/ / doi.org/10.1073/pnas.0400187101
50. Bowley DR, Labrijn AF, Zwick MB, Burton DR. Antigen selection from an HIV-1 immune antibody library displayed on yeast yields many novel antibodies compared to selection from the same library displayed on phage. Protein Eng Des Sel. 2007;20:81-90. https:/ / doi.org/10.1093/protein/gzl057
51. Zhou C, Shen WD. Mammalian cell surface display of full length IgG. Methods Mol Biol. 2012;907:293-302. https://doi.org/10.1007/978-1-61779-974-7 17
52. Aghebati-Maleki L, Bakhshinejad B, Baradaran B, et al. Phage display as a promising approach for vaccine development. J Biomed Sci. 2016;23:66. https://doi.org/10.1186/s 12929-016-0285-9
53. Bakker AB, Marissen WE, Kramer RA, et al. Novel human monoclonal antibody combination effectively neutralizing natural rabies virus variants and individual in vitro escape mutants. J Virol. 2005;79:9062-8. https://doi.org/10.1128/lVI.79.14.9062-9068.2005
54. Redman JM, Hill EM, AlDeghaither D, Weiner LM. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol. 2015;67(2 Pt A):28-45. https://doi.org/10.1016/i.molimm.2015.04.002
55. Palma M. Phage display identification of immunodominant epitopes and autoantibodies in autoimmune diseases. Curr Biosci. 2021;1:e02. https://doi.org/10.51959/cb.2021.v1n1.e02
56. Noy-Porat T, Makdasi E, Alcalay R, et al. A panel of human neutralizing mAbs targeting SARS-CoV-2 spike at multiple epitopes. Nat Commun. 2020;11:4303. https://doi.org/10.1038/s41467-020-18159-4
57. Wu Y, Li C, Xia S, et al. Identification of human singledomain antibodies against SARS-CoV-2. Cell Host Microbe. 2020;27:891-8.e5. https://doi.org/10.1016/i.chom.2020.04.023
58. Amersdorfer P, Wong C, Smith T, et al. Genetic and immunological comparison of anti-botulinum type A antibodies from immune and non-immune human phage libraries. Vaccine. 2002;20:1640-8. https://doi.org/10.1016/S0264-410X(01)00482-0
59. Trott M, Weiß S, Antoni S, et al. Functional characterization of two scFv-Fc antibodies from an HIV controller selected on soluble HIV-1 Env complexes: a neutralizing V3- and a trimer-specific gp41 antibody. PLoS One. 2014;9:e107089. https://doi.org/10.1371/iournal.pone.0097478
60. Ekiert DC, Kashyap AK, Steel J, et al. Crossneutralization of influenza A viruses mediated by a single antibody loop. Nature. 2012;489:526-32. https:/ / doi.org/10.1038/nature 11414
61. Rahumatullah A, Abdul Karim IZ, Noordin R, Lim TS. Antibody-based protective immunity against helminth infections: antibody phage display derived antibodies against BmR1 antigen. Int J Mol Sci. 2017; 18:2376. https://doi.org/10.3390/iims18112376
62. Grubbs H, Kahwaji CI. Physiology, active immunity. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2021.
63. World Health Organization. 2021. Draft landscape and tracker of COVID-19 candidate vaccines. Accessed on: 23 April 2021. Available at: https: //www .who.int/publications/m/item/draftlandscape-of-covid-19-candidate-vaccines.
64. OurWorldinData. 2021. Coronavirus (COVID-19) caccinations. Accessed on: 23 April 2021. Available from: https://ourworldindata.org/covid-vaccinations.
65. Dong Y, Dai T, Wei Y, Zhang L, Zheng M, Zhou F. A systematic review of SARS-CoV-2 vaccine candidates. Signal Transduct Target Ther. 2020;5:237. https://doi.org/10.1038/s41392-020-00352-y
66. Golob JL, Lugogo N, Lauring AS, Lok AS. SARS-CoV2 2 a triumph of science and collaboration. JCI Insight. 2021;6:149187. https:/ / doi.org/ 10.1172/ici.insight. 149187
67. Krammer F. SARS-CoV-2 vaccines in development. Nature. 2020;586:516-27. https://doi.org/10.1038/s41586-020-2798-3
68. Skwarczynski M, Toth I. Peptide-based synthetic vaccines. Chem Sci. 2016;7:842-54. https://doi.org/10.1039/C5SC03892H
69. Zhu R, Liu J, Chen C, et al. A highly conserved epitope-vaccine candidate against varicella-zoster virus induces neutralizing antibodies in mice. Vaccine. 2016;34:1589-96. https://doi.org/10.1016/i.vaccine.2016.02.007
70. Guo L, Yin R, Liu K, et al. Immunological features and efficacy of a multi-epitope vaccine CTB-UE against H. pylori in BALB/c mice model. Appl Microbiol Biotechnol. 2014;98:3495-507. https://doi.org/10.1007/s00253-013-5408-6
71. Wang J, Liu M, Ding N, et al. Vaccine based on antibody-dependent cell-mediated cytotoxicity epitope on the H1N1 influenza virus increases mortality in vaccinated mice. Biochem Biophys Res Commun. 2018;503:1874-9. https://doi.org/10.1016/i.bbrc.2018.07.129
72. Kadam A, Sasidharan S, Saudagar P. Computational design of a potential multi-epitope subunit vaccine using immunoinformatics to fight Ebola virus. Infect Genet Evol. 2020;85:104464. https:/ / doi.org/ 10.1016/j .meegid.2020.104464
73. Bemani P, Amirghofran Z, Mohammadi M. Designing a multi-epitope vaccine against blood-stage of Plasmodium falciparum by in silico approaches. J Mol Graph Model. 2020;99:107645. https:/ / doi.org/ 10.1016/j .imgm.2020.107645
74. Majee P, Jain N, Kumar A. Designing of a multiepitope vaccine candidate against Nipah virus by in silico approach: a putative prophylactic solution for the deadly virus. J Biomol Struct Dyn. 2021;39:1461-80. https://doi.org/10.1080/07391102.2020.1734088
75. Joshi A, Kaushik V. In-silico proteomic exploratory quest: crafting T-cell epitope vaccine against Whipple's disease. Int J Pept Res Ther. 2020:1-18. https://doi.org/10.1007/s10989-020-10077-9
76. Nelde A, Bilich T, Heitmann JS, et al. SARS-CoV-2derived peptides define heterologous and COVID-19induced T cell recognition. Nat Immunol. 2021;22:7485. https://doi.org/10.1038/s41590-020-00808-x
77. Kar T, Narsaria U, Basak S, et al. A candidate multiepitope vaccine against SARS-CoV-2. Sci Rep. 2020;10:10895. https://doi.org/10.1038/s41598-020-67749-1
78. Nevagi RJ, Toth I, Skwarczynski M. Peptide-based vaccines. In: Koutsopoulos S (ed). Peptide applications in biomedicine, biotechnology and bioengineering. Elsevier; 2018. pp: 327-58. https://doi.org/10.1016/B978-0-08-100736-5.00012-0
79. Slingluff CL Jr, Petroni GR, Chianese-Bullock KA, et al. Trial to evaluate the immunogenicity and safety of a melanoma helper peptide vaccine plus incomplete Freund's adjuvant, cyclophosphamide, and polyICLC (Mel63). J Immunother Cancer. 2021;9):e000934. https://doi.org/10.1136/jitc-2020-000934
80. Li W, Joshi MD, Singhania S, Ramsey KH, Murthy AK. Peptide vaccine: progress and challenges. Vaccines (Basel). 2014;2:515-36. https://doi.org/10.3390/vaccines2030515
81. Kaumaya PT. B-cell epitope peptide cancer vaccines: a new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol. 2020;16:1767-91. https://doi.org/10.2217/fon-2020-0224
82. Di Natale C, La Manna S, De Benedictis I, Brandi P, Marasco D. Perspectives in peptide-based vaccination strategies for syndrome coronavirus 2 pandemic. Front Pharmacol. 2020;11:578382. https://doi.org/10.3389/fphar.2020.578382
83. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18:1254-61. https://doi.org/10.1038/nm.2883
84. Rini BI, Stenzl A, Zdrojowy R, et al. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17:1599-611. https://doi.org/10.1016/S1470-2045(16)30408-9
85. Moynihan JS, Blair J, Coombes A, D'Mello F, Howard CR. Enhanced immunogenicity of a hepatitis B virus peptide vaccine using oligosaccharide ester derivative microparticles. Vaccine. 2002;20:1870-6. https://doi.org/10.1016/S0264-410X(01)00494-7
86. Rohovie MJ, Nagasawa M, Swartz JR. Virus-like particles: Next-generation nanoparticles for targeted therapeutic delivery. Bioeng Transl Med. 2017;2:43-57. https:/ / doi.org/10.1002/btm2.10049
87. Roehnisch T, Then C, Nagel W, et al. Phage idiotype vaccination: first phase I/II clinical trial in patients with multiple myeloma. J Transl Med. 2014; 12:119. https://doi.org/10.1186/1479-5876-12-119
88. Greenwood J, Willis AE, Perham RN. Multiple display of foreign peptides on a filamentous bacteriophage. J Mol Biol. 1991;220:821-7. https://doi.org/10.1016/0022-2836(91)90354-9
89. Sunderland KS, Yang M, Mao C. Phage-enabled nanomedicine: from probes to therapeutics in precision medicine. Angew Chem Int Ed Engl. 2017;56:1964-92. https://doi.org/10.1002/anie.201606181
90. Enshell-Seijffers D, Denisov D, Groisman B, et al. The mapping and reconstitution of a conformational discontinuous B-cell epitope of HIV-1. J Mol Biol. 2003;334:87-101. https:/ / doi.org/10.1016/i.imb.2003.09.002
91. Oleksiewicz MB, Bøtner A, Toft P, Normann P, Storgaard T. Epitope mapping porcine reproductive and respiratory syndrome virus by phage display: the nsp2 fragment of the replicase polyprotein contains a cluster of B-cell epitopes. J Virol. 2001;75:3277-90. https: //doi.org/ 10.1128/IVI. 75.7.3277-3290.2001
92. Yu H, Jiang LF, Fang DY, et al. Selection of SARScoronavirus-specific B cell epitopes by phage peptide library screening and evaluation of the immunological effect of epitope-based peptides on mice. Virology. 2007;359:264-74. https://doi.org/10.1016/i.virol.2006.09.016
93. Zamecnik CR, Rajan JV, Yamauchi KA, et al. ReScan, a multiplex diagnostic pipeline, pans human sera for SARS-CoV-2 antigens. Cell Rep Med. 2020;1:100123. https://doi.org/10.1101/2020.05.11.20092528
94. DeFrancesco L. COVID-19 antibodies on trial. Nat Biotechnol. 2021;39:246. https://doi.org/10.1038/s41587-020-0732-8
95. Both L, Banyard AC, van Dolleweerd C, Wright E, Ma JK, Fooks AR. Monoclonal antibodies for prophylactic and therapeutic use against viral infections. Vaccine. 2013;31:1553-9. https://doi.org/10.1016/j.vaccine.2013.01.025
96. Lebozec K, Jandrot-Perrus M, Avenard G, Favre-Bulle O, Billiald P. Quality and cost assessment of a recombinant antibody fragment produced from mammalian, yeast and prokaryotic host cells: a case study prior to pharmaceutical development. N Biotechnol. 2018;44:31-40. https://doi.org/10.1016/j.nbt.2018.04.006
97. Deeks ED. Certolizumab pegol: a review in inflammatory autoimmune diseases. BioDrugs. 2016;30:607-17. https://doi.org/10.1007/s40259-016-0197-y
98. Lien S, Lowman HB. Therapeutic anti-VEGF antibodies. In: Chernajovsky Y, Nissim A (eds). Therapeutic antibodies. Handbook of experimental pharmacology. Berlin: Springer-Verlag Berlin Heidelberg; 2008. pp: 131-50. https://doi.org/10.1007/978-3-540-73259-4 6
99. Bournazos S, Ravetch J V. Anti-retroviral antibody FcyR-mediated effector functions. Immunol Rev. 2017;275:285-95. https://doi.org/10.1111/imr.12482
100. Che Nordin MA, Teow SY. Review of current cellpenetrating antibody developments for HIV-1 therapy. Molecules. 2018;23:335. https:/ / doi.org/10.3390/ molecules23020335
101. Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358-78. https://doi.org/10.1038/s41573-019-0012-9
102. Panyam J, Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv Drug Deliv Rev. 2003;55:329-47. https://doi.org/10.1016/S0169-409X(02)00228-4
103. Lü JM, Wang X, Marin-Muller C, et al. Current advances in research and clinical applications of PLGAbased nanotechnology. Expert Rev Mol Diagn. 2009;9:325-41. https://doi.org/10.1586/erm.09.15
104. Wang Y, Zhang Z, Luo J, Han X, Wei Y, Wei X. mRNA vaccine: a potential therapeutic strategy. Mol Cancer. 2021;20:33. https://doi.org/10.1186/s12943-021-01311-z
105. Henry KA, Arbabi-Ghahroudi M, Scott JK. Beyond phage display: non-traditional applications of the filamentous bacteriophage as a vaccine carrier, therapeutic biologic, and bioconjugation scaffold. Front Microbiol. 2015;6:755. https://doi.org/10.3389/fmicb.2015.00755
106. Antibody society. 2021. Antibody therapeutics approved or in regulatory review in the EU or US. Accessed on: 22 April 2021. Available at: https://www.antibodvsocietv.org/resources/approvedantibodies/.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2021. This work is published under http://www.germs.ro/en/Pages/About-4 (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
The current epidemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is raising awareness of the need to act faster when dealing with new pathogens. Exposure to an emerging pathogen generates an antibody response that can be used for preventing and treating the infection. These antibodies might have a high specificity to a target, few side effects, and are useful in the absence of an effective vaccine for treating immunocompromised individuals. The approved antibodies against the receptor-binding domain (RBD) of the viral spike protein of SARS-CoV-2 (e.g., regdanvimab, bamlanivimab, etesevimab, and casirivimab/imdevimab) have been selected from the antibody repertoire of B cells from convalescent patients using flow cytometry, next-generation sequencing, and phage display. This encourages use of these techniques especially phage display, because it does not require expensive types of equipment and can be performed on the lab bench, thereby making it suitable for labs with limited resources. Also, the antibodies in blood samples from convalescent patients can be used to screen pre-made peptide libraries to identify epitopes for vaccine development. Different types of vaccines against SARS-CoV-2 have been developed, including inactivated virus vaccines, mRNA-based vaccines, non-replicating vector vaccines, and protein subunits. mRNA vaccines have numerous advantages over existing vaccines, such as efficacy, ease of manufacture, safety, and cost-effectiveness. Additionally, epitope vaccination may constitute an attractive strategy to induce high levels of antibodies against a pathogen and phages might be used as immunogenic carriers of such peptides. This is a point worth considering further, as phage-based vaccines have been shown to be safe in clinical trials and phages are easy to produce and tolerate high temperatures. In conclusion, identification of the antibody repertoire of recovering patients, and the epitopes they recognize, should be an attractive alternative option for developing therapeutic and prophylactic antibodies and vaccines against emerging pathogens.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details
1 PhD, Independent researcher, Calle San Jose, Torrevieja, 03181, Spain