Abstract
Human immunodeficiency virus (HIV) poses a major health problem around the globe, resulting in hundred-thousands of deaths from AIDS and over a million new infections annually. Although the standard treatment of HIV infection, antiretroviral therapy, has proven effective in preventing HIV transmission, it is unsuitable for worldwide use due to its substantial costs and frequent adverse effects. Besides promoting HIV/AIDS awareness through education, there is hardly an alternative for inhibiting the spread of the disease. One promising approach is the development of an HIV vaccine. Unfortunately, the high variability of envelope proteins from HIV subtypes, their frequency of mutation and the lack of fully understanding the mechanisms of protection against the virus constitute an obstacle for vaccine development. Efforts for developing successful anti-HIV vaccines have been underway for decades now, with little success. Lately, significant progress has been made in adopting the novel mRNA vaccine approach as an anti-HIV strategy. mRNA vaccines received a great thrust during the COVID-19 pandemic. Now, several mRNA-based HIV vaccines are undergoing clinical trials to evaluate their safety and efficacy. This review offers an overview of the pathogenesis and treatment of HIV / AIDS, previous efforts of HIV vaccine development and introduces mRNA vaccines as a promising and potential game changing platform for HIV vaccination.
Keywords: HIV, AIDS, antiretroviral therapy, vaccine, bNAbs, mRNA.
SUMMARY
1. Introduction
2. HIV
2.1 Introduction
2.2 HIV replication cycle
2.3 Transmission and pathogenesis
2.4 AIDS
2.5 Current treatment and prevention methods
3. HIV vaccines
4. mRNA-based vaccines
4.1 What do mRNA vaccines consist of?
4.2 How do they work?
4.3 Approaches of mRNA vaccines
5. mRNA-based vaccines for HIV
6. Benefits and limitations of the anti-HIV mRNA-based vaccines
7. Conclusion
1. Introduction
Acquired immunodeficiency syndrome (AIDS), the disease resulting from infection with the human immunodeficiency virus (HIV), is the second most fatal infectious disease, leading to numerous deaths annually and posing a major health problem, especially in Sub-Saharan African countries1. According to the World Health Organization (WHO), 680.000 people died from AIDS and 1,5 million people got infected with HIV in 20202. In the US, approximately 1,2 million people are currently living with HIV/AIDS and approximately 34.800 people acquired the virus in 20193.
HIV is a virus of the retroviridae family that attacks the immune system, leading to an increased susceptibility of the organism to diseases and thus, causes AIDS. Although treatment consisting of lifelong administration of antiretroviral medication is effective in prolonging patients’ freedom of symptoms in the latent period and in impeding transmission, the infection with HIV cannot be cured4-6. Furthermore, antiretroviral therapy implies great costs7,8 and adherence of the patient to the therapy plan is essential. In order to prevent further spread of HIV / AIDS infection, it has been the focus of research to find an efficient HIV vaccine.
However, due to the complex viral features of HIV, this is a very challenging task. The international renown of mRNA-based vaccines during the COVID-19 pandemic encourages the use of this novel platform for developing and delivering complex vaccination formulas.
This article provides an overview over HIV infection and introduces mRNA vaccines and their potential in HIV vaccine development.
2. HIV
2.1 Introduction
Human immunodeficiency virus (HIV) is the cause of AIDS. Currently, two types of HIV can be distinguished: HIV-1 and HIV-29. HIV-1 can be found worldwide, whereas HIV-2 is the predominant type in Western Africa10. The virus measures 100 nm in diameter and is made up of an RNA-genome surrounded by a capsule9,10. The RNA is single-stranded and consists of two identical copies that - amongst others - contain the genes gag, pol and env, that encode important viral proteins and are typical for retroviruses10. While the gag gene codes for structural core and matrix proteins (p24, p7, p6, p17), the env gene encodes the glycoproteins gp120 and gp41 found in the viral envelope that are used to bind to the CD4 receptor of the host cell9. Moreover, the pol gene carries the information for different enzymes needed for the HIV replication cycle: reverse transcriptase, integrase and HIV protease (Figure 1).
Figure 1. HIV structure.
HIV contains an RNA genome and the enzymes reverse transcriptase, integrase and HIV protease, that are all surrounded by a capsid, matrix proteins and an envelope. Acquired from Alamy (alamy.de website11).
2.2 HIV replication cycle
The virus uses seven steps to replicate:
2.2.1 Binding and entry
With the help of the envelope protein gp120, HIV can attach itself to cells expressing the CD4 receptor on their surface, namely immune cells, such as the T-helper-lymphocytes in the blood or progenitors in the bone marrow9,12. This induces a change in conformation of gp120, revealing an HIV-coreceptor recognizing chemokines9. These are small molecules involved in stimulating the migration of leukocytes during inflammation processes. While there are several types of chemokine receptors, in most cases CXCR4 and CXCR5 function as coreceptors for HIV12. When the viral protein gp120 is simultaneously bound to the CD4 and chemokine receptor, cell entry is attained through gp41-mediated membrane fusion9.
2.2.2 Uncoating
This word describes the release of a viral genome into a host cell’s cytoplasm.
2.2.3 Reverse transcription
Once being inside the host, retroviruses face several problems that they solve in a very interesting way. One of them is that the viral and host genome are incompatible as they differ in the nucleic acid used, making integration of the viral into the host genome impossible. This is where the viral enzymes come into play: reverse transcriptase possesses the ability to convert RNA into complementary DNA (cDNA)13. This process takes place in the cytoplasm and begins with the synthesis of an RNA/DNA hybrid double helix13. Later, the RNA strand is degraded through ribonuclease H activity and is replaced with the second complementary DNA strand13. The cDNA can also be referred to as provirus13.
2.2.4 Integration of the provirus
Another aspect refers to the integration of the viral genome into the host genome. The viral cDNA must first be transported into the host nucleus and integrated into the host genome in order to transcribe an mRNA strand the host cell’s ribosomes can read to produce the proteins needed for viral replication. The enzyme responsible for these purposes is called integrase14. Firstly, it removes several nucleotides from the 3’ ends of the cDNA, thus leaving sticky ends of the provirus (3’ processing reaction)14. In this state the provirus can enter the nucleus. Here, integrase cuts the host DNA and connects viral DNA 3’-OH groups with DNA 5’-phosphates (strand transfer)14. This leaves a gap between the viral 5’ end and the host DNA, which is repaired by the host cell’s machinery14. Once it is integrated into the host genome, the provirus can remain dormant for years before becoming activated15.
2.2.5 Synthesis of viral proteins
If the infected host cell is activated, the provirus can be transcribed into mRNA15. In an initial phase, regulatory proteins are synthesized from the mRNA, namely Tat and Rev, which enhance transcription of HIV-genes9. Hence, longer RNA transcripts can be produced in the next phase, that encode the information for structural HIV proteins and are synthesized in the cytoplasm using the host cell machinery9. Interestingly, larger proteins are often produced in a first step, just to be divided into separate viral particles9. For example, a synthesized 160 kD precursor molecule is later cleaved into the pol and gag proteins by HIV-specific protease9.
2.2.6 Assembly
During assembly, a new virion is formed by enclosing two viral mRNA strands and the enzymes needed for replication within the HIV capsid made up of HIV core proteins9.
2.2.7 Budding
The virion can now cross the plasma membrane towards the extracellular space, receiving a lipid envelope in the process16.
The newly released HIV-viruses can continue infecting other CD4+ cells, namely T-helper cells, monocytes, macrophages and dendritic cells9.
Replication steps are detailed in Figure 2.
Figure 2. HIV replication cycle.
After attaching its g120 to the CD4+ receptor and chemokine coreceptor of a host cell (1), viral entry and uncoating takes place (2). The viral enzyme reverse transcriptase turns the viral RNA genome into a cDNA strand (3) which can enter the nucleus in order to be integrated into the DNA-genome of the host cell by action of the viral enzyme integrase (4). Now, biosynthesis of the viral genome can take place creating HIV-RNA strands and viral proteins that can assemble (5) and leave the cell by budding (6) in a next step. Acquired from istockphoto.com17.
2.3 Transmission and pathogenesis
Following transmission of HIV, the course of the disease can be divided into three stages: acute phase, chronic/asymptomatic phase and terminal pahse/AIDS.
HIV transmission can occur in different ways. The most common transmission events are sexual intercourse, exchange of body fluids such as blood, e.g. by sharing needles or syringes for drug injection, and perinatal transmission18. According to the UNAIDS report from 2020, 65% of HIV infections worldwide were observed in key populations that are relatively often exposed to the above-mentioned transmission mechanisms, such as sex workers and drug addicts19. Hence, HIV infection necessitates direct contact of infected blood cells from the HIV-infected person with skin damage or abrasions existent in the host person9. Whether HIV is transmitted or not depends on the contagiousness of the HIV subtype, its concentration in the infectious fluid transmitted, the infection route and in the susceptibility of the host person9,20. Furthermore, studies suggest that the prevalence of other infectious diseases in the host that are sexually transmitted, e.g. herpes simplex types 2 and 1 producing genital ulcers, bacterial vaginosis etc, increases the risk of HIV infection12,21.
2.3.1 Acute phase
After acquiring HIV, the virus starts its replication in the mucosa and submucosa, which causes an innate and adaptive immune response20. This is characterized by cytokine and chemokine secretion, and propagation of the infection to dendritic cells and CD4+ lymphocytes that can carry HIV to the lymphatic tissue12. Infected immune cells are either eliminated or they persist as viral reservoirs, meaning that they carry the dormant provirus in their genome until reactivation of the virus occurs9,22. Hence, viral load inside the body dramatically increases, whereas CD4+ lymphocyte numbers significantly drop during the early acute phase12. HIV becomes detectable in the blood 1 - 3 weeks after infection, once the amount of free viral RNA and p24 antigen, an HIV core protein, have sufficiently increased20. This event marks the end of the eclipse phase20 (see Figure 3). HIV copies in the blood peak at about 4 weeks after infection with over 1 million copies per ml23. However, viral load quickly regresses due to the host’s immune response, which is accompanied by seroconversion, describing the onset of antibody production, and a partial recovery in CD4+ cell number12. Seroconversion usually takes place within 3 to 5 weeks after infection and can be detected by analyzing antibodies in blood samples24. At the end of the acute phase, a setpoint in viral and CD4+ cells is reached24. Possible symptoms during this period resemble those of an influenza infection and include headache, fever and rashes24. Studies have found correlations between the severity of symptoms during acute phase infection and of the prognosis of HIV patients9,25.
Figure 3. The course of HIV infection.
The diagram shows the changes of CD4 cell count (red) and plasma viral load (blue, amount of HIV detectable in blood) per milliliter in the course of HIV disease. Three stages can be distinguished.
1. The acute phase starts with the eclipse phase, where the virus cannot yet be detected. After this threshold is surpassed, characteristics of this phase are a peak in plasma viral load, while CD4+ cell numbers drop and then partially recover.
2. In the asymptomatic phase, the body relatively manages to keep a set point of viral load with ongoing immune reactions and CD4+ cell depletion.
3. The terminal phase, also called AIDS, is defined by a CD4+ cell drop below 200 cells per µL blood, clinical symptoms become apparent and opportunistic infections lead to a fatal outcome.
Reproduced from the reference30(distributed under the “Creative Commons CC0 License”)
2.3.2 Chronic, asymptomatic phase (clinical latency)
The second phase of HIV infection is characterized by an asymptomatic period in which HIV continuously multiplies and stimulates the humoral and cell-mediated immune response accompanied by inflammation, while gradually depleting the organism of its CD4+ immune cells12. On the one hand, antibodies against HIV antigens produced by B-cells aim to specifically bind and neutralize free HI viruses, as well as to mark HIV for Antibody-dependent Cellular Cytotoxicity (ADCC), a process in which the antibody-tagged virus can be recognized by natural killer cells and other immune cells26. On the other hand, viral antigens that are being expressed on major histocompatibility complex (MHC) proteins of infected CD4+ cells can also be detected by cytotoxic T-cells, triggering the release of perforins and granzymes and thus, the induction of apoptosis in the infected CD4+ cell27. Along comes the secretion of cytokines, mediators of inflammation, further increasing immune activation21. However, all of these measures do not achieve HIV eradication because of the remaining viral reservoirs and high mutation rate21. As the disease progresses, the lymphatic tissue gets damaged, and the CD4+ lymphocyte number progressively declines9. The duration of this phase extends from 2-3 years to over a decade depending on the HIV subtype, i.e. the severity of escape mutations, and the vulnerability of the host21. In conclusion, the immune system’s attempt to control the viral infection results in an immune system exhaustion and dysfunction.
2.3.3 AIDS
The last phase is acquired immunodeficiency syndrome (AIDS), which is characterized by the proceeding viral replication and a drop in CD4+ lymphocyte number below 200 CD4+ T cells per µL blood9,24,28 (normal: 500 - 1500 cells / µL)28. At this stage, the immune system has already been too severely compromised to maintain protective function from pathogens, thus, making the organism vulnerable to opportunistic infections with bacteria, other viruses, fungi, parasites or cancer which can quickly become lethal9,28.
2.3.4 Elite controllers
HIV-infected people that are not treated with ART but nevertheless manage to maintain undetectable viral loads for one year or more are referred to as Elite controllers29. These people are very rare and account for less than 1% of the HIV-infected. Differences in MHC proteins and other receptors of various immune cells and their response to infection have been investigated, but the underlying mechanisms that explain the virological control are still unknown. The phases of HIV infection are detailed in Figure 3.
2.4 Current treatment and prevention methods
Antiretroviral therapy (ART)
HIV patients are treated with drug combinations that target different steps of the HIV replication process. This method has proved to be highly effective being able to slow down the progression of the disease, but often comes along with side effects and requires the patient’s strict and life-long adherence to the therapy plan to avoid mutations resulting in irreversible drug resistance of the virus31. It should be pointed out that HIV / AIDS can only be suppressed, not cured, due to the escape of the dormant provirus from the immune system. Nevertheless, viral load can be reduced to a great extent using this therapy, limiting the risk of mutation and prolonging life expectancy32.
Currently, five classes of ART medication are being used.
· entry inhibitors32
These substances prevent attachment of the virus to a host cell by binding to HIV envelope proteins.
· nucleoside reverse transcription inhibitors (NRTIs)32
As soon as these substances get integrated into the cDNA during synthesis by the enzyme reverse transcriptase, they lead to chain termination.
· non-nucleoside reverse transcription inhibitors (NNRTIs)32
These drugs use a specific binding site of the enzyme reverse transcriptase and inactivate it through non-competitive inhibition.
· integrase strand transfer inhibitors (INSTIs)
These substances hinder the integration of the cDNA into the host genome by binding to the enzyme integrase.
· protease inhibitors (PIs)33
By blocking the HIV protease, these substances impede viral assembly.
The usage of ART is not limited to treating infected individuals but has also proven to be effective in preventing HIV transmission in the context of pre- and post-exposure prophylaxis5,12. Despite this, the application of ART as a prevention method is complicated and impractical because of the problem of establishing broad availability to the drug and the substantial costs that arise5. That’s why a lot of hope relies in the development of a potent vaccine to ensure protection from HIV among the public.
3. HIV vaccines
Although a lot of research has been conducted to develop an HIV vaccine, for years none have proven to be effective because of many challenging features of HIV. Those include the huge variability of HIV antigens in the viral envelope, which may differ up to 35%, and the frequent escape mutations occuring in the virus34,35. During the last years, research has focused on broadly neutralizing antibodies (bNAbs) as the key substance, and recently, vaccines using the nucleic acid mRNA are getting more and more attention.
3.1 Broadly neutralizing antibodies (bNAbs)-based vaccines
In the course of the disease, 20 - 30 % of individuals naturally produce bNAbs 2 - 4 years post-infection5. HIV bNAbs are antibodies that are able to effectively neutralize commonly circulating HIV subtypes by binding to highly conserved areas of the virus36. More precisely, these antibodies bind to antigens on the HIV Env protein, a protein needed for attachment of the virus to the host cell’s CD4 receptor5,36. Env consists of the glycoprotein gp120, interacting with the CD4 receptor, and gp41, anchoring Env in the viral envelope5,36. Simultaneously, with their constant region, bNAbs are able to bind to the Fc receptor on immune cells, also stimulating inflammatory processes and cytotoxicity37. Thus, the goal is to induce bNAbs against target epitopes of HIV in the infected organism in order to enhance effective immunity. Many approaches have extensively been studied. Here, we briefly present three of them that have shown potential and have progressed in the last years.
3.1.1 Env-resembling molecule
One idea is the injection of a molecule that structurally resembles the Env protein, into the host organism to elicit an immune response with bNAb-production. A major challenge of this approach is the difficulty in reproducing the hidden nature of conserved epitopes of Env. Namely, such epitopes are mostly masked by glycans and its accessibility seems to vary depending on the state the flexible Env currently is in34,38. The challenge resides in finding a simultaneously stable and immunogenic substance39. After extensive research, some advances have been made. For instance, BG505.SOSIP.664 gp140 is such a protein mimicking clade A Env protein that has shown first success and is currently undergoing phase 1 of clinical trials (NCT03699241)39-41.
3.1.2 Nanoparticles for germline-targeting
Another idea is the usage of HIV nanoparticles for germline-targeting39,42. This means that nanoparticles designed to be similar to the site, Env gp120 binds to the CD4 receptor with, aim to activate rare B-cell precursors that undergo the complex affinity maturation in order to synthesize bNAbs5. Preclinical studies of the nanoparticle eOD-GT8 provided promising results, i.e. bNAb production from B-cell precursors of the class VRC01 in mice5,39. Also, recently the results of the phase 1 clinical trial (NCT03547245) revealed that VRC01-bNAbs could be found in 97% of the vaccinated participants43,44.
3.1.3 B-cell lineage vaccine
An alternative method is the design of a B-cell lineage vaccine. This method involves multiple injections, starting with an initial form of Env in the first injection followed by gradually evolved Env proteins in the next injections, thus mimicking its evolutionary progression which triggers B-cells into production of bNAbs39. In addition, this approach is in phase 1 of clinical trials (NCT03220724)45.
The most common epitopes for bNAbs on the HIV-1 envelope glycoprotein Env are shown in Figure 4.
Figure 4. Epitopes for bNAbs on the HIV-1 envelope glycoprotein Env.
The picture shows the most common epitopes on the surface of an Env trimer, that are recognized by different bNAb classes which are specified in the boxes on the right. The CD4-binding site is marked red, V1/V2 blue, V3/Asn322 glycan patch green, gp120/gp41-interface brown and Membrane Proximal External Region (MPER) is represented by the dark grey box. Other surface areas of Env are colored in grey: light grey representing parts belonging to gp120, and dark grey representing parts belonging to gp41. Adapted from the references46,47(distributed under the “Creative Commons Attribution (CC-BY) license”).
4. mRNA-based vaccines
Public interest in mRNA-based vaccines to prevent infectious diseases arose greatly during the COVID-19 pandemic. Indeed, this type of vaccine has shown to be highly effective against SARS-CoV-2 in the clinical trials and helped in containing morbidity and mortality by immunizing vaccinated people around the world48-51. Other advantages of this vaccine design method include the safety of the use of mRNA vaccines, the feasibility of large-scale production, and the high adaptability of these vaccines to combat various diseases.
4.1 What do mRNA vaccines consist of?
mRNA-based vaccines consist of a single-stranded mRNA encoding a specific protein of the viral particle that the immune system should be educated to recognize52,53. UTR sequences, a 5’ cap, a 3’ poly(A) tail flank the coding region and nucleosides within the mRNA are modified in order to delay degradation in the cytoplasm, decrease immunogenicity and enhance translation52-54. In vivo delivery of the mRNA can be achieved by transfection of the designed mRNA into antigen presenting cells (APCs), e.g. dendritic cells, that display the target antigen to the immune system when transferred back in vivo55. Newer methods such as the SARS-CoV-2 mRNA vaccines encapsulate the mRNA in lipid nanoparticles (LNPs) that contain cationic ionizable lipids that aid LNP-uptake into the cell52-54.
4.2 How do they work?
After delivery into a host cell, the mRNA gets translated into the viral protein using the host cell’s translation machinery54,55. Thus, a normal body cell starts producing viral proteins that can be displayed on MHC proteins on the cellular surface, where the viral protein can be detected as an antigen by the organism’s immune cells54. This triggers an immune response resulting in antibody production, cell-mediated apoptosis of infected body cells and the production of memory cells that remain even after the recovery process54. The mRNA strand gets broken down by nucleases and has a rather short half-life54 (see Figure 5).
Figure 5. Working mechanism of mRNA-based vaccines.
An mRNA strand encoding the viral antigen is delivered in a LNP to facilitate cellular uptake. One inside the cell, the mRNA gets translated by the host cell’s replication machinery, so the viral protein the mRNA codes for gets produced. The viral antigen can then be displayed on MHC proteins on the cellular surface or can get secreted into the extracellular matrix, where the viral antigen is accessible for recognition by immune cells. An immune response is generated. Acquired from istockphoto.com56.
4.3 Approaches of mRNA vaccines
There are different approaches to mRNA vaccination depending on whether the delivered mRNA replicates in the host cell or not.
· Self-amplifying mRNA vaccines (saRNA)
Besides the gene for the target antigen, this type of vaccine additionally contains genes that encode an RNA-dependent RNA polymerase57. These genes are obtained from RNA viruses, mostly alphaviruses57,58. Thus, saRNAs are able to multiply their mRNA in the host cell which results in an increased production of the encoded immunogen57. Although already very small doses of vaccine have shown to produce a strong and potent immune response, problems regarding delivery of the large saRNA size and higher risk of reactogenicity have to be faced57,58.
· Non-replicating mRNA vaccines
Currently, this type of vaccine is used for the containment of the SARS-CoV-2 pandemic54. It contains the information for synthesis of the antigen of interest as described above. In comparison to saRNA, non-replicating mRNA vaccines cannot multiply in the host cell57. Hence, the generated immune response is weaker but still potent54,57.
· mRNA vaccines for cancer
mRNA vaccines cannot only be used for infectious diseases, but have great potential in combating cancers by teaching the immune system which tumor antigens it has to look for54,58. Due to the mutations that occur when cells become cancerous, cancer cells often display so-called neoantigens with neoepitopes58. When analyzing tumors, such neoepitopes can be used to design an mRNA vaccine58. The aim is to activate an immune response in order to eliminate cancerous cells58. At the moment, there are many studies exploring the use of mRNA vaccines in various cancers58.
5. mRNA-based vaccines for HIV
Several studies have been conducted investigating different mRNA vaccine approaches against HIV and research is ongoing34,59-68. In Table 1, some of the recent promising results are presented. The first phase 1 clinical trials have been launched to evaluate the safety and immunogenicity of different HIV mRNA vaccine formulas (Table 2).
Table 1. Recent studies investigating different HIV-mRNA vaccine approaches.
STUDY | YEAR | DESCRIPTION |
---|---|---|
Pardi et al.64 | 2017 | In their study, Pardi et al. injected mice with LNP-encapsulated nucleoside-modified mRNA encoding the light and heavy chain of VRC01, a bNAb against HIV. VRC01 could be detected in the blood and generated immunity against intravenously injected HIV-1 viruses. |
Pardi et al.65 | 2019 | The same group later used a nucleoside-modified and purified mRNA-LNP vaccine that encoded a clade C Env protein. This vaccine was injected intradermally into rabbits and rhesus macaques and caused the production of antibodies against gp120, while ADCC was also observed. |
Bogers et al.66 | 2015 | This group encoded a clade C Env protein into a saRNA, used cationic nanoemulsion (CNE) for delivery and injected the formula into rhesus macaques. Activation of the immune system by the synthesized viral particles resulted in strong humoral and cell-mediated immune responses, already from a low dosis. |
Movo et al.67 | 2019 | They induced a saRNA containing tHIVconsvX, a mix of six highly conserved HIV immunogens, into mice. Results indicated strong CD4+ and CD8+ immune responses that lasted 22 weeks after injection. |
Zhang et al.34,68 | 2021 | Combining several strategies, this recent study used an mRNA-based vaccine. Firstly, experiments were conducted on mice, injecting them with an mRNA vaccine encoding the Env and Gag protein. After endogenous protein synthesis in the host cell, these proteins assemble to build virus-like proteins (VLPs) that are immunogenic but not infectious as they lack the viral genome encoding the instructions for replication. An important advance in this mRNA vaccine is the similarity to natural Env protein and the quantity of the Env proteins generated on the VLPs that induced antibody production in the mice. Secondly, macaques were used to further investigate the results of this env-gag VLP mRNA vaccine. In a first step, the macaques were primed with an mRNA encoding clade B Env protein that has been modified in a way to be easily recognized by the immune system. Then, the animals received multiple env-gag boosters over one year, each consisting of gag-mRNA and a changing env-mRNA. In total, four different env-mRNAs from three HIV-1 clades were used, because antibody production should be directed towards eliciting antibodies (bNAbs) against the more conserved areas of the Env protein. The vaccine induced production of neutralizing antibodies and T-cell responses, while causing only mild adverse effects in the macaques, e.g. appetite loss. Thirdly, the protection of immunized animals compared to a control group was assessed from week 60 by 13 weekly injections of simian-human immunodeficiency virus (SHIV) into the rectum. Out of 7 animals, 2 remained uninfected, whereas the others showed an infection delay of on average 5 weeks compared to the control group. Thus, the risk of infection with SHIV per exposure was reduced by 79% in the vaccinated rhesus macaques. |
Table 2. HIV-mRNA vaccines currently undergoing clinical trial.
Clinical trial identifiers | Vaccine | Study Sponsor | Collaborators | Status | Participants |
---|---|---|---|---|---|
NCT05217641 HVTN30259,60 | BG505 MD39.3 mRNA, BG505 MD39.3 gp151 mRNA, BG505 MD39.3 gp151 CD4KO mRNA | National Institute of Allergy and Infectious Diseases (NIAID) | Scripps Research Institute; Bill and Melinda Gates Foundation; Cambridge University; ModernaTX, Inc.; 11 study sites in the US | Phase 1 | 108 estimated participants healthy, HIV-negative ages 18 - 55 |
NCT05001373 IAVIG00261,62 | eOD-GT8 60mer mRNA Vaccine (mRNA-1644) Core-g28v2 60mer mRNA Vaccine (mRNA-1644v2-Core) | International AIDS Vaccine Initiative (IAVI) | ModernaTX, Inc.; Scripps Research Institute; 4 study sites (in Atlanta, Seattle, San Antonio) | Phase 1 | 56 estimated participants healthy, HIV-negative ages 18 - 50 |
One clinical trial has been launched by a division of the US National Institute of Health (NIH)59,60, and another clinical trial is funded by the National Institute of Allergy and Infectious Diseases (IAVI)61,62, representing some of the cases where the mRNA technology is for the first time employed as an anti-HIV strategy. The mRNA HIV vaccines in both studies have been developed in cooperation with Moderna (Cambridge, MA, USA).
The HVTN302 (NCT05217641) study is based on previous work of Steichen et al.63. This study group designed HIV Env trimers with germline-targeting mutations that induced priming PGT121-antibody responses in mice. They further proposed a vaccine containing BG505 MD39.3 variants administered in booster injections to enhance PGT121-specific-bNAb production by guiding the affinity maturation of precursor B cells. Currently, this approach is being tested in the clinical trial HVTN302 using mRNA-based delivery59,60. Healthy participants are injected with one of the three mRNA vaccines intramuscularly at doses of either 100µg or 250µg and will receive a booster vaccination at month two and six. Participants' general and immune response to the vaccine will be evaluated in blood and lymph-node fine-needle aspiration probes. The higher dose will only be tested if the lower dose raises no safety concerns.
Building on the promising results of the eOD-GT8 60mer vaccine (NCT03547245), the clinical trial IAVIG002 aims to assess the safety and efficacy of the eOD-GT8 nanoparticles for germline-targeting using an mRNA platform43,61,62. Healthy participants will be injected with mRNA-1644 first and mRNA-1644v2-Core sequentially to stimulate precursor B cells into producing bNAbs. In other arms of the study, participants will be injected with mRNA-1644 or mRNA-1644v2-Core only.
This article focuses on mRNA vaccine technologies for active immunization against HIV. Nevertheless, it is worth mentioning that mRNA-based vaccines are currently also being tested in HIV-infected patients with the goal of passive immunization. Therefore, dendritic cell (DC) mRNA vaccines are mainly being used55. To do so, autologous DCs are either transfected with antigen-encoding mRNA ex vivo and then reinjected intradermally into the patient or the DCs are being targeted with the mRNA in situ by intranodal injection55. Several clinical trials have already been conducted investigating these strategies and others are ongoing55.
6. Benefits and limitations of the anti-HIV mRNA-based vaccines
mRNA-based vaccines in general and anti-HIV mRNA-based vaccines in particular have several significant benefits compared with conventional vaccines, in terms of safety, efficacy, production and applications (see Table 3). As limitations, mRNA-based vaccines are based on a relatively new technology and the adverse effects on long term are yet to be determined.
Table 3. Benefits and Limitations of anti-HIV mRNA-based vaccines.
BENEFITS & LIMITATIONS | COMMENTS |
---|---|
Relatively new technology | Although research for mRNA-based vaccine development started decades ago, the worldwide use of mRNA vaccines has only begun recently, in the search for a potent anti-SARS-CoV-2 vaccine54,69. Adapting the mRNA platform to act against HIV is a very promising approach55. However, besides the major challenges encountered trying to find a safe and potent HIV-immunogen to be encoded within the mRNA, new anti-HIV vaccines require thorough evaluation to ensure they fulfill their function and are safe to use in the population55. |
Efficacy | Studies as well as the COVID-19 pandemic have confirmed the effectiveness of mRNA vaccines48,49,54,70. The mRNA intake is facilitated by LNPs, which enables body cells to produce the viral antigen, so a strong immune response can be generated and provides the organism immunity54. However, the complex characteristics of HIV pose a great challenge to potent HIV vaccine design and it still remains to be determined whether the current approaches reach their intended effect55. |
Safety | mRNA vaccines are not infectious and there is no ground for concerns that it could integrate into the host genome54,58,70. Due to its short in vivo half-life the mRNA strand is degraded quickly and is thus less likely to cause any side effects because of immunogenicity54,70. However, concerns about safety of the anti-HIV vaccine may arise regarding the immune system’s reaction to the Env protein encoded in the mRNA sequence and the possible short- and long-term adverse effects55. |
Production | Compared with traditional forms of vaccination, less resources are needed for mRNA vaccine production. Firstly, this refers to the time needed: no viruses need to be grown in order to obtain the needed viral particle54,70. Secondly, material can be saved because tissues for viral growth become redundant54. Thirdly, costs can be saved, with the exception of costs that incur for distribution and storage of mRNA vaccine vials at low temperatures54. |
Distribution and Storage | The downside of mRNA-based vaccines lies in the challenge of maintaining the low temperatures required for mRNA-vaccine storage during vaccine transport and distribution. In addition to the special cooling equipment, a well-organized logistic network is needed to ensure quality control following the guidelines for temperatures and shelf-life during shipping and storage54,71. Thus, the risk of faults occurring during transport might pose an obstacle, especially in low-income countries72. |
Potential Applications | mRNA vaccines are highly adaptable, as the same platform can be used to fight various diseases. Once the viral genome is known and a section encoding a specific protein is selected, vaccine production can start at a large scale54,58. This feature makes mRNA vaccines a very potent instrument in containing emerging pandemics54,70,73. |
7. Conclusion
To present, there are no effective agents that can be used for HIV prevention. Currently, the only method to contain the pandemic is through education by raising awareness about the disease. During the last years, a lot of research has been conducted for developing a potent HIV vaccine. Major challenges include huge variability of HIV antigens in the viral envelope and the frequent escape mutations occuring in the virus. Because of their ability to bind more conserved epitopes of the Env protein of HIV, bNAbs provide a useful source for immunization efforts. However, directing B-cell-precursors into producing bNAbs is difficult because their maturation process is complex and targeting those conserved epitopes proved to be complicated due to the hidden nature of those epitopes within the Env antigen.
Since the beginning of the SARS-CoV-2 pandemic, the usage of mRNA to deliver instructions for protein synthesis of target proteins into host cells has been greatly promoted. mRNA vaccines are safe and efficient, but also allow for large-scale manufacturing to facilitate accessibility to the vaccine worldwide. One of the greatest advantages, however, is the possibility of adapting mRNA vaccines to fight various diseases, simply by replacing the sequence in the mRNA used to code for a different target protein.
Thus, this type of vaccine is appealing for HIV vaccine research. Indeed, there have been several studies using mRNA-based vaccines to pursue and combine different strategies to induce bNAb production in the host cells that could be the basis for HIV immunity. Results were very promising and the first clinical trials evaluating mRNA-based HIV vaccines are ongoing, but a lot of research is still needed to assess and further improve the HIV-mRNA vaccination. Hence, until we will find a potent HIV vaccine that can change the course of this pandemic, obstacles still have to be overcome. Still, the most recent findings are encouraging and call for more research to pursue this goal of finding a vaccine against HIV.
KEY POINTS
◊ Human immunodeficiency virus (HIV), the cause of AIDS, poses a serious health issue around the world
◊ Until now, most approaches have focused on the use of bNAbs for development of an HIV vaccine. Lately, mRNA-based HIV vaccines have emerged as a promising method
◊ HIV presents a difficult target for vaccine production because of its high variability in envelope protein structure and mutation rate
◊ Currently, the first mRNA-based HIV vaccines are being tested in clinical trials
Acknowledgments
OB is funded by a grant of the Romanian Ministry of Education and Research, CNCS - UEFISCDI, project number PN-III-P4-ID-PCE-2020-2027, within PNCDI III. AF thanks to the Albert-Ludwigs-Universitat Freiburg for support. Authors would like to acknowledge the funding from Ministry of Research, Innovation and Digitization in Romania, under Program 1 – The Improvement of the National System of Research and Development, Subprogram 1.2 – Institutional Excellence – Projects of Excellence Funding in RDI, Contract No. 31PFE/30.12.2021. AF designed, organized the review, analyzed and summarized the data and wrote the manuscript. OB supervised the work and contributed to the design, organization and writing of the manuscript.
Footnotes
Conflict of interests: The authors declare no conflicts of interest.
Abbreviations: Acquired Immunodeficiency Syndrome (AIDS); Human Immunodeficiency Virus (HIV); World Health Organization (WHO); Complementary DNA (cDNA); Antibody-dependent Cellular Cytotoxicity (ADCC); Major Histocompatibility Complex (MHC); Antiretroviral therapy (ART); Nucleoside reverse transcription inhibitor (NRTI); Non-nucleoside reverse transcription inhibitor (NNRTI); Integrase strand transfer inhibitor (INSTI); Protease inhibitor (PI); Broadly neutralizing antibody (bNAb); Antigen presenting cell (APC); Lipid nanoparticles (LNP); Self-amplifying mRNA vaccine (saRNA); National Institute of Health (NIH); Cationic nanoemulsion (CNE); Virus-like protein (VLP); Simian-human immunodeficiency virus (SHIV); National Institute of Allergy and Infectious Diseases (NIAID); International AIDS Vaccine Initiative (IAVI).
DISCOVERIES is a peer-reviewed, open access, online, multidisciplinary and integrative journal, publishing high impact and innovative manuscripts from all areas related to MEDICINE, BIOLOGY and CHEMISTRY
References
- 1.Roser Max, Ritchie Hannah HIV / AIDS. . Our World in Data. 2018. https://ourworldindata.org/hiv-aids https://ourworldindata.org/hiv-aids
- 2.HIV. World Health Organization; Accessed on March 30, 2022. 2022. https://www.who.int/news-room/fact-sheets/detail/hiv-aids https://www.who.int/news-room/fact-sheets/detail/hiv-aids
- 3.U.S. Statistics. HIV.gov; Accessed on March 30, 2022. 2022. https://www.hiv.gov/hiv-basics/overview/data-and-trends/statistics https://www.hiv.gov/hiv-basics/overview/data-and-trends/statistics
- 4.HIV/AIDS epidemiology, pathogenesis, prevention, and treatment. Simon Viviana, Ho David D, Abdool Karim Quarraisha. Lancet (London, England) 2006;368(9534):489–504. doi: 10.1016/S0140-6736(06)69157-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Progress in HIV vaccine development. Hsu Denise C, O'Connell Robert J. Human vaccines and immunotherapeutics. 2017;13(5):1018–1030. doi: 10.1080/21645515.2016.1276138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Prevention of HIV-1 infection with early antiretroviral therapy. Cohen Myron S, Chen Ying Q, McCauley Marybeth, Gamble Theresa, Hosseinipour Mina C, Kumarasamy Nagalingeswaran, Hakim James G, Kumwenda Johnstone, Grinsztejn Beatriz, Pilotto Jose H S, Godbole Sheela V, Mehendale Sanjay, Chariyalertsak Suwat, Santos Breno R, Mayer Kenneth H, Hoffman Irving F, Eshleman Susan H, Piwowar-Manning Estelle, Wang Lei, Makhema Joseph, Mills Lisa A, de Bruyn Guy, Sanne Ian, Eron Joseph, Gallant Joel, Havlir Diane, Swindells Susan, Ribaudo Heather, Elharrar Vanessa, Burns David, Taha Taha E, Nielsen-Saines Karin, Celentano David, Essex Max, Fleming Thomas R. The New England journal of medicine. 2011;365(6):493–505. doi: 10.1056/NEJMoa1105243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pattern, Determinants, and Impact of HIV Spending on Care and Treatment in 38 High-Burden Low- and Middle-Income Countries. Granich Reuben, Gupta Somya, Montaner Julio, Williams Brian, Zuniga José M. Journal of the International Association of Providers of AIDS Care. 2016;15(2):91–100. doi: 10.1177/2325957415623261. [DOI] [PubMed] [Google Scholar]
- 8.Clinical Treatment Options Infectious Diseases: Update on PrEP Implementation, Adherence, and Advances in Delivery. Doblecki-Lewis Susanne, Cohen Stephanie, Liu Albert. Current treatment options in infectious diseases. 2015;7(2):101–112. doi: 10.1007/s40506-015-0046-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.HIV virology and pathogenetic mechanisms of infection: a brief overview. Fanales-Belasio Emanuele, Raimondo Mariangela, Suligoi Barbara, Buttò Stefano. Annali dell'Istituto superiore di sanita. 2010;46(1):5–14. doi: 10.4415/ANN_10_01_02. [DOI] [PubMed] [Google Scholar]
- 10.Pebody Roger. HIV-1 and HIV-2. AIDSMap.com; Accessed on March 30, 2022. 2021. https://www.aidsmap.com/about-hiv/hiv-1-and-hiv-2 https://www.aidsmap.com/about-hiv/hiv-1-and-hiv-2
- 11.Structure of the human immunodeficiency virus. YAY Media AS, Alamy Vector; Accessed on April 08, 2022. 2022.
- 12.HIV infection: epidemiology, pathogenesis, treatment, and prevention. Maartens Gary, Celum Connie, Lewin Sharon R. Lancet (London, England) 2014;384(9939):258–71. doi: 10.1016/S0140-6736(14)60164-1. [DOI] [PubMed] [Google Scholar]
- 13.HIV-1 reverse transcription. Hu Wei-Shau, Hughes Stephen H. Cold Spring Harbor perspectives in medicine. 2012;2(10) doi: 10.1101/cshperspect.a006882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Multifaceted HIV integrase functionalities and therapeutic strategies for their inhibition. Engelman Alan N. The Journal of biological chemistry. 2019;294(41):15137–15157. doi: 10.1074/jbc.REV119.006901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.What Integration Sites Tell Us about HIV Persistence. Hughes Stephen H, Coffin John M. Cell host and microbe. 2016;19(5):588–98. doi: 10.1016/j.chom.2016.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.HIV-1 assembly, budding, and maturation. Sundquist Wesley I, Kräusslich Hans-Georg. Cold Spring Harbor perspectives in medicine. 2012;2(7):a006924. doi: 10.1101/cshperspect.a006924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.HIV Viral Life Cycle. iStock - istockphoto.com; Accessed on April 08, 2022. 2022. https://www.istockphoto.com/ro/vector/ciclul-de-replicare-hiv-gm509233595-46172088 https://www.istockphoto.com/ro/vector/ciclul-de-replicare-hiv-gm509233595-46172088
- 18.Ways HIV can be transmitted. Centers for Disease Control and Transmission (CDC); Accessed on March 30, 2022. 2022. https://www.cdc.gov/hiv/basics/hiv-transmission/ways-people-get-hiv.html https://www.cdc.gov/hiv/basics/hiv-transmission/ways-people-get-hiv.html
- 19.Global AIDS Update. UNAIDS; Accessed on March 30, 2022. 2021. https://www.unaids.org/en/resources/presscentre/pressreleaseandstatementarchive/2021/july/20210714_global-aids-update https://www.unaids.org/en/resources/presscentre/pressreleaseandstatementarchive/2021/july/20210714_global-aids-update
- 20.Acute HIV-1 Infection. Cohen Myron S, Shaw George M, McMichael Andrew J, Haynes Barton F. The New England journal of medicine. 2011;364(20):1943–54. doi: 10.1056/NEJMra1011874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Virus-Host Gene Interactions Define HIV-1 Disease Progression. Monaco Daniela C, Ende Zachary, Hunter Eric. Current topics in microbiology and immunology. 2017;407:31–63. doi: 10.1007/82_2017_33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cellular reservoirs of HIV-1 and their role in viral persistence. Alexaki Aikaterini, Liu Yujie, Wigdahl Brian. Current HIV research. 2008;6(5):388–400. doi: 10.2174/157016208785861195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Viral dynamics of acute HIV-1 infection. Little S J, McLean A R, Spina C A, Richman D D, Havlir D V. The Journal of experimental medicine. 1999;190(6):841–50. doi: 10.1084/jem.190.6.841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Diagnosis of Human Immunodeficiency Virus Infection. Parekh Bharat S, Ou Chin-Yih, Fonjungo Peter N, Kalou Mireille B, Rottinghaus Erin, Puren Adrian, Alexander Heather, Hurlston Cox Mackenzie, Nkengasong John N. Clinical microbiology reviews. 2019;32(1) doi: 10.1128/CMR.00064-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Severity and prognosis of acute human immunodeficiency virus type 1 illness: a dose-response relationship. Vanhems P, Lambert J, Cooper D A, Perrin L, Carr A, Hirschel B, Vizzard J, Kinloch-de Loës S, Allard R. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 1998;26(2):323–9. doi: 10.1086/516289. [DOI] [PubMed] [Google Scholar]
- 26.Antibody-dependent cellular cytotoxicity in HIV infection. Forthal Donald N, Finzi Andrés. AIDS (London, England) 2018;32(17):2439–2451. doi: 10.1097/QAD.0000000000002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pathogenesis of human immunodeficiency virus infection. Levy J A. Microbiological Reviews. 1993;57(1):183-289. doi: 10.1128/mr.57.1.183-289.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Garcia Sofia A. Battistini, Guzman Nilmarie. Acquired Immune Deficiency Syndrome CD4+ Count. StatPearls; 2021. [PubMed] [Google Scholar]
- 29.HIV-1 elite controllers: an immunovirological review and clinical perspectives. Gebara Nour Y., El Kamari Vanessa, Rizk Nesrine. Journal of Virus Eradication. 2019;5(3):163-166. doi: 10.1016/S2055-6640(20)30046-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.File:Hiv-timecourse copy.svg. https://commons.wikimedia.org/w/index.php?curid=15383502 Wikimedia Commons; Accessed on April 08, 2022. 2022 [Google Scholar]
- 31.Antiretroviral nephrotoxicities. Atta Mohamed G, Deray Gilbert, Lucas Gregory M. Seminars in nephrology. 2008;28(6):563–75. doi: 10.1016/j.semnephrol.2008.08.009. [DOI] [PubMed] [Google Scholar]
- 32.Clinical Pharmacology in HIV Therapy. Atta Mohamed G, De Seigneux Sophie, Lucas Gregory M. Clinical journal of the American Society of Nephrology : CJASN. 2019;14(3):435–444. doi: 10.2215/CJN.02240218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.HIV protease inhibitors: a review of molecular selectivity and toxicity. Lv Zhengtong, Chu Yuan, Wang Yong. HIV/AIDS (Auckland, N.Z.) 2015;7:95–104. doi: 10.2147/HIV.S79956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.A multiclade env-gag VLP mRNA vaccine elicits tier-2 HIV-1-neutralizing antibodies and reduces the risk of heterologous SHIV infection in macaques. Zhang Peng, Narayanan Elisabeth, Liu Qingbo, Tsybovsky Yaroslav, Boswell Kristin, Ding Shilei, Hu Zonghui, Follmann Dean, Lin Yin, Miao Huiyi, Schmeisser Hana, Rogers Denise, Falcone Samantha, Elbashir Sayda M, Presnyak Vladimir, Bahl Kapil, Prabhakaran Madhu, Chen Xuejun, Sarfo Edward K, Ambrozak David R, Gautam Rajeev, Martin Malcom A, Swerczek Joanna, Herbert Richard, Weiss Deborah, Misamore Johnathan, Ciaramella Giuseppe, Himansu Sunny, Stewart-Jones Guillaume, McDermott Adrian, Koup Richard A, Mascola John R, Finzi Andrés, Carfi Andrea, Fauci Anthony S, Lusso Paolo. Nature medicine. 2021;27(12):2234–2245. doi: 10.1038/s41591-021-01574-5. [DOI] [PubMed] [Google Scholar]
- 35.Evolutionary and immunological implications of contemporary HIV-1 variation. Korber B, Gaschen B, Yusim K, Thakallapally R, Kesmir C, Detours V. British medical bulletin. 2001;58:19–42. doi: 10.1093/bmb/58.1.19. [DOI] [PubMed] [Google Scholar]
- 36.To bnAb or Not to bnAb: Defining Broadly Neutralising Antibodies Against HIV-1. Griffith Sarah A, McCoy Laura E. Frontiers in immunology. 2021;12:708227. doi: 10.3389/fimmu.2021.708227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.CD8 Effector T Cells Function Synergistically With Broadly Neutralizing Antibodies to Enhance Suppression of HIV Infection. Veenhuis Rebecca T, Garliss Caroline C, Bailey Justin R, Blankson Joel N. Frontiers in immunology. 2021;12:708355. doi: 10.3389/fimmu.2021.708355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Passive and active antibody studies in primates to inform HIV vaccines. Hessell Ann J, Malherbe Delphine C, Haigwood Nancy L. Expert review of vaccines. 2018;17(2):127–144. doi: 10.1080/14760584.2018.1425619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Innovations in HIV-1 Vaccine Design. Jones Letitia D, Moody M Anthony, Thompson Amelia B. Clinical therapeutics. 2020;42(3):499–514. doi: 10.1016/j.clinthera.2020.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.HIV-1 VACCINES. HIV-1 neutralizing antibodies induced by native-like envelope trimers. Sanders Rogier W, van Gils Marit J, Derking Ronald, Sok Devin, Ketas Thomas J, Burger Judith A, Ozorowski Gabriel, Cupo Albert, Simonich Cassandra, Goo Leslie, Arendt Heather, Kim Helen J, Lee Jeong Hyun, Pugach Pavel, Williams Melissa, Debnath Gargi, Moldt Brian, van Breemen Mariëlle J, Isik Gözde, Medina-Ramírez Max, Back Jaap Willem, Koff Wayne C, Julien Jean-Philippe, Rakasz Eva G, Seaman Michael S, Guttman Miklos, Lee Kelly K, Klasse Per Johan, LaBranche Celia, Schief William R, Wilson Ian A, Overbaugh Julie, Burton Dennis R, Ward Andrew B, Montefiori David C, Dean Hansi, Moore John P. Science (New York, N.Y.) 2015;349(6244):aac4223. doi: 10.1126/science.aac4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.A Randomized, Double-blinded, Placebo-controlled, Dose-escalation Phase 1 Clinical Trial to Evaluate the Safety and Immunogenicity of Recombinant HIV Envelope Protein BG505 SOSIP.664 gp140 Vaccine, Adjuvanted, in Healthy, HIV-1 Uninfected Adults. https://clinicaltrials.gov/ct2/show/NCT03699241 ClinicalTrials.gov; Accessed on March 30, 2022. 2022;ClinicalTrials.gov Identifier: NCT03699241 [Google Scholar]
- 42.Rational HIV Immunogen Design to Target Specific Germline B Cell Receptors. Jardine J., Julien J.-P., Menis S., Ota T., Kalyuzhniy O., McGuire A., Sok D., Huang P.-S., MacPherson S., Jones M., Nieusma T., Mathison J., Baker D., Ward A. B., Burton D. R., Stamatatos L., Nemazee D., Wilson I. A., Schief W. R. Science. 2013;340(6133):711-716. doi: 10.1126/science.1234150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.A Phase I Trial to Evaluate the Safety and Immunogenicity of eOD-GT8 60mer Vaccine, Adjuvanted. https://clinicaltrials.gov/ct2/show/NCT03547245 ClinicalTrials.gov; Accessed on March 30, 2022. 2022;ClinicalTrials.gov Identifier: NCT03547245 [Google Scholar]
- 44.First-in-human clinical trial confirms novel HIV vaccine approach. Iavi.org; Accessed on March 30, 2022. 2021. https://www.iavi.org/news-resources/press-releases/2021/first-in-human-clinical-trial-confirms-novel-hiv-vaccine-approach-developed-by-iavi-and-scripps-research https://www.iavi.org/news-resources/press-releases/2021/first-in-human-clinical-trial-confirms-novel-hiv-vaccine-approach-developed-by-iavi-and-scripps-research
- 45.Evaluating the Safety and Immunogenicity of EnvSeq-1 and CH505 M5 gp120 Envs Adjuvanted With GLA-SE in Healthy, HIV-Uninfected Adults. https://clinicaltrials.gov/ct2/show/NCT03220724 ClinicalTrials.gov; Accessed on March 30, 2022. 2022;ClinicalTrials.gov Identifier: NCT03220724 [Google Scholar]
- 46.Antiviral Therapy by HIV-1 Broadly Neutralizing and Inhibitory Antibodies. Zhang Zhiqing, Li Shaowei, Gu Ying, Xia Ningshao. International journal of molecular sciences. 2016;17(11) doi: 10.3390/ijms17111901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Attribution 4.0 International (CC BY 4.0) Creative Commons; Accessed on April 08, 2022. 2022. https://creativecommons.org/licenses/by/4.0/ https://creativecommons.org/licenses/by/4.0/
- 48.Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. Polack Fernando P, Thomas Stephen J, Kitchin Nicholas, Absalon Judith, Gurtman Alejandra, Lockhart Stephen, Perez John L, Pérez Marc Gonzalo, Moreira Edson D, Zerbini Cristiano, Bailey Ruth, Swanson Kena A, Roychoudhury Satrajit, Koury Kenneth, Li Ping, Kalina Warren V, Cooper David, Frenck Robert W, Hammitt Laura L, Türeci Özlem, Nell Haylene, Schaefer Axel, Ünal Serhat, Tresnan Dina B, Mather Susan, Dormitzer Philip R, Şahin Uğur, Jansen Kathrin U, Gruber William C. The New England journal of medicine. 2020;383(27):2603–2615. doi: 10.1056/NEJMoa2034577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.mRNA Vaccines in the COVID-19 Pandemic and Beyond. Hogan Michael J, Pardi Norbert. Annual review of medicine. 2022;73:17–39. doi: 10.1146/annurev-med-042420-112725. [DOI] [PubMed] [Google Scholar]
- 50.COVID-19 and the Cardiovascular System: How the First PostModern Pandemic ‘Weakened’ our Hearts. Towhid Syeda Tasneem, Rakhi Nadira Naznin, Arefin ASM Shamsul, Saha Otun, Mamun Sumaiya, Moniruzzaman Mohammad, Rahaman Md. Mizanur. Discoveries Reports. 2020;3:e15. [Google Scholar]
- 51.SARS-CoV-2 and COVID-19: A Threat to Global Health. Saha Otun, Rakhi Nadira Naznin, Sultana Afroza, Rahman Md. Mahbubur, Rahaman Md. Mizanur. Discoveries Reports. 2020;3:e13. [Google Scholar]
- 52.Engineering of the current nucleoside-modified mRNA-LNP vaccines against SARS-CoV-2. Granados-Riveron Javier T, Aquino-Jarquin Guillermo. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie. 2021;142:111953. doi: 10.1016/j.biopha.2021.111953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.The Development of mRNA Vaccines for Infectious Diseases: Recent Updates. Nitika , Wei Jiao, Hui Ai-Min. Infection and drug resistance. 2021;14:5271–5285. doi: 10.2147/IDR.S341694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.First COVID-19 Vaccines Receiving the US FDA and EMA Emergency Use Authorization. Fortner Andra, Schumacher David. Discoveries. 2021;9(1):e122. doi: 10.15190/d.2021.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.HIV and Messenger RNA Vaccine. Khalid Khizer, Padda Jaskamal, Khedr Anwar, Ismail Dina, Zubair Ujala, Al-Ewaidat Ola A, Padda Sandeep, Cooper Ayden Charlene, Jean-Charles Gutteridge. Cureus. 2021 doi: 10.7759/cureus.16197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ilustrație de stoc Mecanismul de acțiune - vaccinul ARN mesager. iStock.com; Accessed on April 08, 2022. 2022. https://www.istockphoto.com/ro/vector/mecanismul-de-ac%C8%9Biune-vaccinul-arn-mesager-gm1307885458-398025519 https://www.istockphoto.com/ro/vector/mecanismul-de-ac%C8%9Biune-vaccinul-arn-mesager-gm1307885458-398025519
- 57.HIV mRNA Vaccines-Progress and Future Paths. Mu Zekun, Haynes Barton F, Cain Derek W. Vaccines. 2021;9(2) doi: 10.3390/vaccines9020134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.mRNA vaccines - a new era in vaccinology. Pardi Norbert, Hogan Michael J, Porter Frederick W, Weissman Drew. Nature reviews. Drug discovery. 2018;17(4):261–279. doi: 10.1038/nrd.2017.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.A Clinical Trial to Evaluate the Safety and Immunogenicity of BG505 MD39.3, BG505 MD39.3 gp151, and BG505 MD39.3 gp151 CD4KO HIV Trimer mRNA Vaccines in Healthy, HIV-uninfected Adult Participants. ClinicalTrials.gov; Accessed on March 30, 2022. 2023. https://clinicaltrials.gov/ct2/show/study/NCT05217641 https://clinicaltrials.gov/ct2/show/study/NCT05217641
- 60.NIH launches clinical trial of three mRNA HIV vaccines. NIH; Accessed on Apr 07, 2022. 2022. https://www.nih.gov/news-events/news-releases/nih-launches-clinical-trial-three-mrna-hiv-vaccines https://www.nih.gov/news-events/news-releases/nih-launches-clinical-trial-three-mrna-hiv-vaccines
- 61.IAVI and Moderna launch trial of HIV vaccine antigens delivered through mRNA technology. Iavi.com; Accessed on March 30, 2022. 2022. https://www.iavi.org/news-resources/press-releases/2022/iavi-and-moderna-launch-trial-of-mrna-hiv-vaccine-antigens https://www.iavi.org/news-resources/press-releases/2022/iavi-and-moderna-launch-trial-of-mrna-hiv-vaccine-antigens
- 62.A Phase 1 Study to Evaluate the Safety and Immunogenicity of eOD-GT8 60mer mRNA Vaccine (mRNA-1644) and Core-g28v2 60mer mRNA Vaccine (mRNA-1644v2-Core) https://clinicaltrials.gov/ct2/show/study/NCT05001373 ClinicalTrials.gov (accessed on Apr 07, 2022). 2022;ClinicalTrials.gov Identifier: NCT05001373 [Google Scholar]
- 63.HIV Vaccine Design to Target Germline Precursors of Glycan-Dependent Broadly Neutralizing Antibodies. Steichen Jon M, Kulp Daniel W, Tokatlian Talar, Escolano Amelia, Dosenovic Pia, Stanfield Robyn L, McCoy Laura E, Ozorowski Gabriel, Hu Xiaozhen, Kalyuzhniy Oleksandr, Briney Bryan, Schiffner Torben, Garces Fernando, Freund Natalia T, Gitlin Alexander D, Menis Sergey, Georgeson Erik, Kubitz Michael, Adachi Yumiko, Jones Meaghan, Mutafyan Andrew A, Yun Dong Soo, Mayer Christian T, Ward Andrew B, Burton Dennis R, Wilson Ian A, Irvine Darrell J, Nussenzweig Michel C, Schief William R. Immunity. 2016;45(3):483–496. doi: 10.1016/j.immuni.2016.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Pardi Norbert, Secreto Anthony J, Shan Xiaochuan, Debonera Fotini, Glover Joshua, Yi Yanjie, Muramatsu Hiromi, Ni Houping, Mui Barbara L, Tam Ying K, Shaheen Farida, Collman Ronald G, Karikó Katalin, Danet-Desnoyers Gwenn A, Madden Thomas D, Hope Michael J, Weissman Drew. Nature communications. 2017;8:14630. doi: 10.1038/ncomms14630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Characterization of HIV-1 Nucleoside-Modified mRNA Vaccines in Rabbits and Rhesus Macaques. Pardi Norbert, LaBranche Celia C., Ferrari Guido, Cain Derek W., Tombácz István, Parks Robert J., Muramatsu Hiromi, Mui Barbara L., Tam Ying K., Karikó Katalin, Polacino Patricia, Barbosa Christopher J., Madden Thomas D., Hope Michael J., Haynes Barton F., Montefiori David C., Hu Shiu-Lok, Weissman Drew. Molecular Therapy - Nucleic Acids. 2019;15:36-47. doi: 10.1016/j.omtn.2019.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Potent Immune Responses in Rhesus Macaques Induced by Nonviral Delivery of a Self-amplifying RNA Vaccine Expressing HIV Type 1 Envelope With a Cationic Nanoemulsion. Bogers W. M., Oostermeijer H., Mooij P., Koopman G., Verschoor E. J., Davis D., Ulmer J. B., Brito L. A., Cu Y., Banerjee K., Otten G. R., Burke B., Dey A., Heeney J. L., Shen X., Tomaras G. D., Labranche C., Montefiori D. C., Liao H.-X., Haynes B., Geall A. J., Barnett S. W. Journal of Infectious Diseases. 2015;211(6):947-955. doi: 10.1093/infdis/jiu522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Efficient Induction of T Cells against Conserved HIV-1 Regions by Mosaic Vaccines Delivered as Self-Amplifying mRNA. Moyo Nathifa, Vogel Annette B., Buus Søren, Erbar Stephanie, Wee Edmund G., Sahin Ugur, Hanke Tomáš. Molecular Therapy - Methods and Clinical Development. 2019;12:32-46. doi: 10.1016/j.omtm.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Experimental mRNA HIV Vaccine Safe, Shows Promise in Animals. NIH - National Institute of Allergy and Infectious Diseases; Accessed on Apr 07, 2022. 2021. https://www.niaid.nih.gov/news-events/experimental-mrna-hiv-vaccine-safe-shows-promise-animals https://www.niaid.nih.gov/news-events/experimental-mrna-hiv-vaccine-safe-shows-promise-animals
- 69.Why are mRNA vaccines so exciting? Harvard Health Publishing; Accessed on Apr 07, 2022. 2021. https://www.health.harvard.edu/blog/why-are-mrna-vaccines-so-exciting-2020121021599 https://www.health.harvard.edu/blog/why-are-mrna-vaccines-so-exciting-2020121021599
- 70.RNA vaccines: an introduction. University of Cambridge - PHG Foundation; Accessed on Apr 07, 2022. 2018. https://www.phgfoundation.org/briefing/rna-vaccines https://www.phgfoundation.org/briefing/rna-vaccines
- 71.Addressing the Cold Reality of mRNA Vaccine Stability. Crommelin Daan J A, Anchordoquy Thomas J, Volkin David B, Jiskoot Wim, Mastrobattista Enrico. Journal of pharmaceutical sciences. 2021;110(3):997–1001. doi: 10.1016/j.xphs.2020.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Stability Modelling of mRNA Vaccine Quality Based on Temperature Monitoring throughout the Distribution Chain. Kis Zoltán. Pharmaceutics. 2022;14(2) doi: 10.3390/pharmaceutics14020430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Drug Repurposing for Prevention and Treatment of COVID-19: A Clinical Landscape. Hossain Md. Shahadat, Hami Ithmam, Sawrav Md. Sad Salabi, Rabbi Md. Fazley, Saha Otun, Bahadur Newaz Mohammed, Rahaman Md. Mizanur. Discoveries. 2020;8(4):e121. doi: 10.15190/d.2020.18. [DOI] [PMC free article] [PubMed] [Google Scholar]