2,861
Views
8
CrossRef citations to date
0
Altmetric
Review

Applications and challenges of multivalent recombinant vaccines

Pages 457-461 | Received 20 Oct 2012, Accepted 02 Nov 2012, Published online: 18 Dec 2012

Abstract

The exceptional discoveries of antigen/gene delivery systems have allowed the development of novel prophylactic and therapeutic vaccine candidates. The vaccine candidates employ various antigen-delivery systems, particularly recombinant viral vectors. Recombinant viral vectors are experimental vaccines similar to DNA vaccines, but they use attenuated viruses or bacterium as a carrier “vector” to introduce microbial DNA to cells of the body. They closely mimic a natural infection and therefore can efficiently stimulate the immune system. Although such recombinant vectors may face extensive preclinical testing and will possibly have to meet stringent regulatory requirements, some of these vectors (e.g. measles virus vectors) may benefit from the profound industrial and clinical experience of the parent vaccine. Most notably, novel vaccines based on live attenuated viruses combine the induction of broad, strong and persistent immune responses with acceptable safety profiles. We assess certain technologies in light of their use against human immunodeficiency virus (HIV).

Introduction

A vaccine works by presenting pieces of the pathogen, virus or bacteria, to the body's immune system that causes an immune reaction. Immunogens or antigens “prime” the immune system so that when it is exposed to the real pathogen, the body’s immune system can efficiently attack and neutralize the incoming pathogen.Citation1

Almost all current vaccines work through the induction of humoral antibodies, IgG and IgA in serum and mucosa. These Antibodies neutralize “functionally” the invasion of a microbe and, thus, prevents infection at a particular level of antibody in the plasma or mucosa. Whereas the neutralizing antibodies can maintain protection, cell-mediated immunity operates as another correlate of protection against a disease.Citation2

While it was feasible to generate vaccines for some of the world’s deadliest diseases (e.g. small pox and measles), no one have been able to find a vaccine against AIDS, caused by HIV.Citation3 There are a number of scientific challenges that can be addressed in order to gain more prospects towards successful HIV-vaccine clinical trials.Citation4 One reason is that scientists still need to know more about the type of immune response(s) to prevent HIV infection and control of HIV replication. Moreover, methods to induce HIV-specific mucosal immunity, optimization of HIV envelope immunogens that induce sterilizing immunity, methods to optimize antiviral T-cell and NK cell activity, and to determine the mechanism of protection of an efficacious vaccine in the nonhuman primate model need extensive optimization. Additionally, research focused on the interaction between HIV and the human immune system may also address many of these unanswered questions and can provide insight for the design and development of more effective vaccine candidates.Citation3,Citation4

Vaccine Approaches and Technologies

A variety of viruses have been investigated for their ability to express heterologous antigens derived from certain pathogens to induce stronger and longer-lasting humoral and cellular immune responses.Citation5,Citation6 Extensive experience has been gathered using subunit vaccine formulation,Citation7 DNA, viral and bacterial vectors (). Importantly, candidate vaccines should be safe, induce humoral and cellular immune responses against the transgene, and should provide long-lasting protection.Citation8

Table 1. Vaccine Designs and strategies in light of their use against HIV

DNA vaccines

DNA vaccines are circular or linear plasmids that encode pieces of viral genes of interest. After the vaccine is injected into patients the plasmid is taken up by cells at the site of injection and viral genes are expressed into proteins.Citation9 For gene expression to occur a stretch of genetic code called the promoter is necessary. The viral proteins are degraded into small peptide fragments, which are then presented by MHC class I and class II molecules on the cell surface where T cells recognizing these complexes to generate an immune response.

DNA vaccines represent a favored vaccine strategy because they are safe, stable, easy to engineer and produce and immune responses generated pose no interference against later boost immunizations. However, DNA vaccines need to be improved significantly (e.g. by adding molecular adjuvants) as they do not induce sufficient levels of immune responses.Citation9

In addition, optimization of both the coding sequence and the gene regulatory elements may be necessary for high levels of gene expression. Moreover, novel delivery technologies may be necessary [e.g. particle-mediated delivery into antigen presenting cells using, viral or bacterial vectors, lipid polymers and gold microparticles, or virus like particles (VLP)]Citation10-Citation12 and physical delivery methods such as needle-free devices such as epidermal patches.

Live vectors

Live recombinant vector vaccines are constructed by inserting HIV or simian immunodeficiency virus genes into genomes of live, infectious, but non-disease-causing forms of viruses or bacteria such as vaccinia virusCitation13 or Bacille Calmette-Guérin (BCG).Citation14 One can think of these viral and bacterial vectors as backpacks that shuttle the cargo of their own genes as well as “foreign” genes into cells.

Viral vectors

Viruses have evolved sophisticated structures and mechanisms to infect cells, and hence, they serve as efficient delivery vectors of various antigens,Citation15 namely the HIV genes.Citation15-Citation17 Scientists use viruses other than HIV as delivery vehicles or vectors to transport and express HIV genes. The modular nature of viral genomes allows scientists to insert foreign genes and sometimes replace the vector’s own genes with the desired HIV or other genes.Citation18-Citation20 Such a recombinant virus is safe and cannot cause HIV infection. HIV proteins generated from the recombinant genes inside the cell are either secreted or displayed on the cell surface and presented to the immune system in the same way that proteins from a virus-infected cell would be.Citation19,Citation20,Citation22

For a virus to be considered a delivery vehicle, it must have the capacity to accommodate large pieces of foreign genes, maintain genetic stability, be feasible for large-scale manufacturing and most, importantly, not cause disease or be toxic. Viruses used as vaccine vectors can be engineered to retain their replication capacity or be rendered non-replicating, e.g. measles viruses (MV) or Adenoviruses (Adv), respectively. In the replication-defective kind, one or more gene(s) that play a role in virus replication are removed or mutated, and the resultant virus will not reproduce itself as it can only infect one cell.Citation6 Replicating viral vectors infect and reproduce in cells using the cell’s machinery.Citation16,Citation21 The new copies of the virus generated can subsequently infect other cells but because it has been manipulated it cannot cause disease. Several replication-incompetent viral vectors have been tested in clinical trials for HIV vaccines, including Adenovirus and adeno-associated virus, alphavirus and poxvirus.Citation18 Replication competent viral vectors like measles virus, vesicular stomatitis and rabies viruses, have been tested with varying levels of success in preclinical studies in animals.Citation16,Citation19,Citation21 Some vaccine developers are focusing on the use of replicating vectors in clinical trials, as they are more fruitful in animal studies in activating elements of innate immunity, the first responders to an invading organism, and are more likely to induce stronger cellular and mucosal immune responses and antibodies. These effects are achieved at much lower doses than the non-replicating vectors.Citation16,Citation17

Live attenuated viral vectors induce strong, long-lasting immune responses against the expressed proteins, including antibody and T cell responses in the blood, and many generate immune responses at mucosal surfaces, depending on their cell targets and sites of replication.Citation24 Importantly, immune responses can be generated to the vector as well as to the incorporated immunogens. Ironically, immune responses to the vector components could limit the effectiveness of subsequent vaccinations using the same vector.Citation24

Gatekeepers for the Processing of Recombinant Multivalent Vaccine

In order to generate a recombinant viral vector a series of gate-keepers must be set before the advancement of the vector to clinical testing. Among the major challenges are: (1) the stability of the insert and protein expression; (2) The use of properly defined vector backbone (e.g., from a clinically approved original vaccine) and the manufacturing process; (3) Efficacy and safety of the recombinant vector.

Stability of the vector and insert

Among the major challenges in making viral vector-based vaccine candidates, particularly the live attenuated viral vectors, relates to the chemistry, or compatibility between the vector backbone and the insert; for example (1) some vectors are simply incompatible with the insert, due to certain nucleotide sequences that render the vector inefficient; (2) The length of the insert may not be optimal and its configuration is not suitable to the viral vector, thus the vector may simply reject the insert; (3) Consequently, the virus acting as the vector may introduce mutations into the inserted gene (the HIV genes) that may prevent production of the complete protein once inside the body. These changes can ultimately impact the generation of a good immune response after vaccination. It is, therefore, necessary to analyze the stability of these vectors during the early stages of vaccine development, in cell culture and in animal model. The stability of the recombinant vector particles are tested by subjecting them to a series of stress tests, forcing them to more than 10 rounds of amplification in cell culture that determines whether they are stable enough to express foreign proteins (e.g., the HIV proteins) before clinical trials.Citation25 This has been successfully established for MV vectors and other members of paramyxoviridae.Citation16,Citation22

Origin of the vector backbone

The cloning of the vector backbones has not been solely performed for clinical use. Often, replication deficient and subsequently live viral vectors were engineered in academic labs not fully conform to industrial standards and quality control systems. The stepwise development of such vectors resulted, sometimes, in the loss of authenticityCitation8 and thus it became difficult to relate efficacy of such vectors with the authentic vaccine. The first live attenuated MV vector, for example, originated from an attenuated “lab strain” whose efficacy in clinical trials may become obscure irrespective of its success in preclinical tests.Citation16 Therefore, it became necessary and urgent to develop vectors with the same genetic properties and manufacturing procedure as that of the authentic vaccine strain,Citation26 especially if a “parental” vaccine strain is available. The substantial knowledge gathered in the last decade about the replication machinery of different viruses, specifically RNA viruses and the generation of defective interfering particles (DIs), shed the light on the possible errors that such RNA viruses could carry. RNA polymerases are known to be much more error-prone than DNA polymerases due to the lack of proof-reading; thus, the error-rate in RNA viruses is on average about three orders of magnitude higher than in DNA viruses.Citation16 The so-called quasispecies concept for RNA viruses have been explored.Citation16 Therefore, it is necessary to set quality control procedures to assure fitness and suitability of an expression vector for clinical development. shows a comparative study between an attenuated MV “lab strain”, the authentic vaccines and the cloned vector from an authentic-parental vaccine. This example shows that the “lab strain” induced immune responses by an order of magnitude less than the authentic vaccine and the cloned counterpart (MVb) (). Moreover, the growth kinetics showed that the authentic vaccines and the cloned vector (MVb) were superior to that of the “lab strain” ().

Figure 1. Comparison of the cloned MV vaccine with various commercially available MV vaccine strains and a “lab-strain.” (A) Transgenic mice expressing human CD46 were immunized i.m. with 1 × 104 pfu of an authentic cloned vaccine (rMVb), a Moraten vaccine (MVbv), Edmonston Zagreb vaccine (MVEZII) and a “lab strain” MVtag. Measles end point titers are shown on a logarithmic scale. (B) Growth kinetic analyses. Comparison of the propagation kinetics of the standard MVbv, cloned MVb and the MVEZII compared to the “lab strain” MVtag. Sub-confluent Cells were infected with the designated viruses and incubated at 31°C, media were collected every day up to 6 days. The shed viruses were titrated by plaque assays.

Figure 1. Comparison of the cloned MV vaccine with various commercially available MV vaccine strains and a “lab-strain.” (A) Transgenic mice expressing human CD46 were immunized i.m. with 1 × 104 pfu of an authentic cloned vaccine (rMVb), a Moraten vaccine (MVbv), Edmonston Zagreb vaccine (MVEZII) and a “lab strain” MVtag. Measles end point titers are shown on a logarithmic scale. (B) Growth kinetic analyses. Comparison of the propagation kinetics of the standard MVbv, cloned MVb and the MVEZII compared to the “lab strain” MVtag. Sub-confluent Cells were infected with the designated viruses and incubated at 31°C, media were collected every day up to 6 days. The shed viruses were titrated by plaque assays.

Efficacy and safety

Generally, killed vaccines or subunit vaccines are not necessarily safer than replication-competent vaccines as drastically shown for killed MV vaccine which proved not only to lack efficacy, but even exacerbated the effects of subsequent wild-type MV infection. Thus, there is no general increasing gradient of safety when one compares replication competent versus replication restricted versus dead vaccines. Any component delivered to humans must be planned, developed and evaluated with equal care, without a priori bias based on theoretical general and illusive classifications of danger for health.

What are the consequences of recombinant vaccines to be replication-competent? Replication-deficient vectors such as most DNA viruses in use, and RNA virus vectors based on alpha-viruses are broadly considered to be generally safer. However, replication-deficiency entails major drawback. To be efficiently immunogenic, higher doses (many orders of magnitude in comparison with replicating vehicles) have to be delivered. In contrast, attenuated live vaccines, like MV, are delivered at low doses and are highly efficacious due to systemic spread and preferential infection of professional antigen-presenting cells and lymphoid tissues. In the case of MV, replication competence appears only as an advantage both in terms of safety and efficacy. Due to the extremely good record of safety, MV vaccination is recommended even for immunocompromized patients, e.g. HIV.Citation16 Importantly, cell targeting of MV recombinants is not altered by transgenes.Citation27 Clinical trials with any recombinant vaccine candidate based on MV are only in the planning stage; thus, practical success is still to be awaited in view of the severe and costly hurdles imposed by regulatory agencies.

Discussion

Although recent progress in genetic engineering technique has enabled us to develop live attenuated mutants of several viruses and bacteria as potential vaccine vectors for antigen delivery, certain issues remain answered. The application routes of the vaccines, the quality of the immune responses and whether results in transgenic mice and nonhuman primates are representatives for an efficacious vaccine.

The limitations of the animal models to evaluate AIDS vaccine are now more defined, especially since the choice of challenge viruses affected the outcome of vaccine studies in nonhuman primates.Citation28 Through the study of infection with simian immunodeficiency virus (SIV) - a monkey virus that is similar but not identical to HIV - researchers have uncovered several clues about how the virus is transmitted following infection and disease progression or pathogenesis.Citation29 There is also much to be learned from the study of SIV infection in species of nonhuman primates that can successfully control SIV infection and not develop the monkey equivalent of AIDS.

Before a vaccine candidate can be tested in humans, it is first evaluated extensively in laboratory tests, cell culture and animal models. Animal models help scientists gain important insights into the diseases and how to prevent them and to determine if a candidate vaccine is safe to administer in people.

The industrial partners, involved in the manufacturing and production of clinical lots, have contributed substantially to the understanding of the vector intrinsic requirement, vector genetic’s background, the cell substrate necessary for an efficient growth and the maintenance of stable protein expression upon multiple rounds of amplification. Not only the genetic structure of the insert is the sole determinant for an efficient and stable protein expression but the genetic stability of the vector backbone determines vector fitness and the subsequent stable amplification of the progeny; especially the RNA live viral vectors (e.g. mononegavirales).

The pre-existing immunity to most of the used vectors is a major concern, especially since multiple immunization doses are used in clinical trials. There are various studies that propose heterologous prime-boost scenarios, e.g. use of two different vectors carrying the same insert,Citation30 or heterologous immunization routes e.g. intramuscular-intranasal.Citation24 Scientists have, also, begun to shed light on analyzing data from various HIV vaccine trials conducted, where some (Thailand, RV144) have shown modest success in preventing HIV infection.Citation31 These analyses are providing some hint as to what type of immune responses may be needed and will help inform future clinical trial design.

Abbreviations:
MV=

measles virus

rMV=

recombinant measles viruses

HIV=

human immunodeficiency virus

VLP=

virus like particles

Acknowledgements

This study was supported by NIH grant AI-46007 to H.Y.N. I would like to thank all members of my Group who contributed to the success of MV vector. During the preparation of this review, H.Y.N. was a part-time faculty member at the Lebanese American University.

Disclosure of Potential Conflicts of Interest

The contents of this review can be used or disclosed for commercial purposes after a written consent from the author.

References

  • Plotkin SA. Vaccines: past, present and future. Nat Med 2005; 11:Suppl S5 - 11; http://dx.doi.org/10.1038/nm1209; PMID: 15812490
  • Plotkin SA, Gilbert PB. Nomenclature for immune correlates of protection after vaccination. Clin Infect Dis 2012; 54:1615 - 7; http://dx.doi.org/10.1093/cid/cis238; PMID: 22437237
  • Girard MP, Plotkin SA. HIV vaccine development at the turn of the 21st century. Curr Opin HIV AIDS 2012; 7:4 - 9; http://dx.doi.org/10.1097/COH.0b013e32834ddc96; PMID: 22156840
  • Johnston MI, Fauci AS. An HIV vaccine--challenges and prospects. N Engl J Med 2008; 359:888 - 90; http://dx.doi.org/10.1056/NEJMp0806162; PMID: 18753644
  • Reyes-Sandoval A, Harty JT, Todryk SM. Viral vector vaccines make memory T cells against malaria. Immunology 2007; 121:158 - 65; http://dx.doi.org/10.1111/j.1365-2567.2006.02552.x; PMID: 17462077
  • Havenga M, Vogels R, Zuijdgeest D, Radosevic K, Mueller S, Sieuwerts M, et al. Novel replication-incompetent adenoviral B-group vectors: high vector stability and yield in PER.C6 cells. J Gen Virol 2006; 87:2135 - 43; http://dx.doi.org/10.1099/vir.0.81956-0; PMID: 16847108
  • Glück R, Burri KG, Metcalfe I. Adjuvant and antigen delivery properties of virosomes. Curr Drug Deliv 2005; 2:395 - 400; http://dx.doi.org/10.2174/156720105774370302; PMID: 16305442
  • Tangy F, Naim HY. Live attenuated measles vaccine as a potential multivalent pediatric vaccination vector. Viral Immunol 2005; 18:317 - 26; http://dx.doi.org/10.1089/vim.2005.18.317; PMID: 16035943
  • Li L, Saade F, Petrovsky N. The future of human DNA vaccines. J Biotechnol 2012; 162:171 - 82; http://dx.doi.org/10.1016/j.jbiotec.2012.08.012; PMID: 22981627
  • Zeltins A. Construction and Characterization of Virus-Like Particles. Rev Mol Biotechnol 2012; In Press
  • Girard MP, Osmanov SK, Kieny MP. A review of vaccine research and development: the human immunodeficiency virus (HIV). Vaccine 2006; 24:4062 - 81; http://dx.doi.org/10.1016/j.vaccine.2006.02.031; PMID: 16530298
  • Kim YC, Quan FS, Compans RW, Kang SM, Prausnitz MR. Formulation of microneedles coated with influenza virus-like particle vaccine. AAPS PharmSciTech 2010; 11:1193 - 201; http://dx.doi.org/10.1208/s12249-010-9471-3; PMID: 20676947
  • Elena Gómez C, Perdiguero B, Garcia-Arriaza J, Esteban M. Poxvirus vectors as HIV/AIDS vaccines in humans. Hum Vaccin Immunother 2012; 8:1192 - 207; http://dx.doi.org/10.4161/hv.20778; PMID: 22906946
  • Lynch A, Meyers AE, Williamson AL, Rybicki EP. Stability studies of HIV-1 Pr55gag virus-like particles made in insect cells after storage in various formulation media. Virol J 2012; 9:210; http://dx.doi.org/10.1186/1743-422X-9-210; PMID: 22988963
  • Sauter SL, Rahman A, Muralidhar G. Non-replicating viral vector-based AIDS vaccines: interplay between viral vectors and the immune system. Curr HIV Res 2005; 3:157 - 81; http://dx.doi.org/10.2174/1570162053506900; PMID: 15853721
  • Billeter MA, Naim HY, Udem SA. Reverse genetics of measles virus and resulting multivalent recombinant vaccines: applications of recombinant measles viruses. Curr Top Microbiol Immunol 2009; 329:129 - 62; http://dx.doi.org/10.1007/978-3-540-70523-9_7; PMID: 19198565
  • Liniger M, Zuniga A, Naim HY. Use of viral vectors for the development of vaccines. Expert Rev Vaccines 2007; 6:255 - 66; http://dx.doi.org/10.1586/14760584.6.2.255; PMID: 17408374
  • Spielhofer P, Bächi T, Fehr T, Christiansen G, Cattaneo R, Kaelin K, et al. Chimeric measles viruses with a foreign envelope. J Virol 1998; 72:2150 - 9; PMID: 9499071
  • Mebatsion T, Conzelmann KK. Specific infection of CD4+ target cells by recombinant rabies virus pseudotypes carrying the HIV-1 envelope spike protein. Proc Natl Acad Sci U S A 1996; 93:11366 - 70; http://dx.doi.org/10.1073/pnas.93.21.11366; PMID: 8876141
  • Mebatsion T, Finke S, Weiland F, Conzelmann KKA. A CXCR4/CD4 pseudotype rhabdovirus that selectively infects HIV-1 envelope protein-expressing cells. Cell 1997; 90:841 - 7; http://dx.doi.org/10.1016/S0092-8674(00)80349-9; PMID: 9298896
  • Schnell MJ. Viral vectors as potential HIV-1 vaccines. FEMS Microbiol Lett 2001; 200:123 - 9; http://dx.doi.org/10.1111/j.1574-6968.2001.tb10703.x; PMID: 11425463
  • Zuniga A, Wang Z, Liniger M, Hangartner L, Caballero M, Pavlovic J, et al. Attenuated measles virus as a vaccine vector. Vaccine 2007; 25:2974 - 83; http://dx.doi.org/10.1016/j.vaccine.2007.01.064; PMID: 17303293
  • Ye L, Wen Z, Dong K, Wang X, Bu Z, Zhang H, et al. Induction of HIV neutralizing antibodies against the MPER of the HIV envelope protein by HA/gp41 chimeric protein-based DNA and VLP vaccines. PLoS One 2011; 6:e14813; http://dx.doi.org/10.1371/journal.pone.0014813; PMID: 21625584
  • Knuchel MC, Marty RR, Azzouz Morin TN, Ilter O, Zuniga A, Naim HY. Relevance of a pre-existing measles immunity prior immunization with a recombinant measles vector. Hum Vaccin Immunother 2012; In Press
  • Wang Z, Hangartner L, Cornu TI, Martin LR, Zuniga A, Billeter MA, et al. Recombinant measles viruses expressing heterologous antigens of mumps and simian immunodeficiency viruses. Vaccine 2001; 19:2329 - 36; http://dx.doi.org/10.1016/S0264-410X(00)00523-5; PMID: 11257357
  • Zuniga A, Liniger M, Azzouz Morin TN, Marty RR, Wiegand M, Ilter O, et al. Sequence and immunogenicity of a clinically approved novel measles virus vaccine vector. Hum Vaccin Immunother 2012; In Press
  • Liniger M, Zuniga A, Morin TN, Combardiere B, Marty R, Wiegand M, et al. Recombinant measles viruses expressing single or multiple antigens of human immunodeficiency virus (HIV-1) induce cellular and humoral immune responses. Vaccine 2009; 27:3299 - 305; http://dx.doi.org/10.1016/j.vaccine.2009.01.057; PMID: 19200842
  • Lee EM, Chung HK, Livesay J, Suschak J, Finke L, Hudacik L, et al. Molecular methods for evaluation of virological status of nonhuman primates challenged with simian immunodeficiency or simian-human immunodeficiency viruses. J Virol Methods 2010; 163:287 - 94; http://dx.doi.org/10.1016/j.jviromet.2009.10.012; PMID: 19878696
  • Watkins DI. HIV vaccine development. Top HIV Med 2010; 18:35 - 6; PMID: 20516522
  • Bolton DL, Santra S, Swett-Tapia C, Custers J, Song K, Balachandran H, et al. Priming T-cell responses with recombinant measles vaccine vector in a heterologous prime-boost setting in non-human primates. Vaccine 2012; 30:5991 - 8; http://dx.doi.org/10.1016/j.vaccine.2012.06.029; PMID: 22732429
  • McEnery R, Kresge KJ. First Evidence of Efficacy from Large-Scale HIV Vaccine Trial Results from a trial in Thailand show that the combination of two vaccine candidates provides some protection against HIV infection. VAX, 2009; Vol. 07, No. 09

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.