Genetic-Based Vaccine Vectors


INTRODUCTION

The adoption and establishment of new vaccine platforms usually takes decades. The concept of delivering a recombinant virus whose payload is not the virus itself but rather the genetic instructions encoded within the viral genome originated in the 1980s with recombinant poxviral vaccine vectors. The approach mimicked live-attenuated vaccines (LAV), but provided a vision to replace the empiric and difficult strategy of viral attenuation through genetic deletion and insertion of target antigen transgenes for immune induction. Genetic vectors can be designed not to replicate or to replicate without the ability to revert to a pathogenic form. The inserted genetic segment is translated and transcribed into viral antigen by host-cell or virally driven machinery, thus mimicking properties immunologically associated with LAV and inducing both humoral and cellular immunity. Manufacturing and production systems can be standardized across vaccine platforms rather than being pathogen-based, thus accelerating production.

An important conceptual breakthrough occurred in the early 1990s with the description of pure nucleic acid vaccine approaches, that is, DNA vaccines and then RNA vaccines. Nucleic acid vaccines focus on antigen-insert genetic delivery, allowing the host cell to perform all required functions for transcription/translation, processing, and folding of vaccine antigens. They depend on delivery approaches such as lipid nanoparticle (LNP) formulations or physical delivery such as jet injection or electroporation for vaccine-construct entry into the host. They drive cellular and humoral immunity, providing advantages usually associated with live vector vaccines for immune protection. Genetic vaccines provide conceptual advantages in immune breadth over killed inactivated vaccine approaches, recombinant vaccine approaches, and polymer-based vaccine approaches.

The engineering of nonviral RNA- or DNA-based systems or viral recombinant systems to express foreign antigens in the host activates unique pathways driving effector immune functions and enhances immune pressure on a target pathogen. The resulting immunity includes the induction of memory responses, with the possibility of broad protective responses. A variety of systems, nonviral and viral, have been developed for these purposes (see Figs. 68.1 and 68.2 ).

Fig. 68.1, Representative genetic vaccine vector platforms. Genetic vaccines can be delivered using nucleic acid-based (DNA or RNA) (A) or using replication-defective viral vector-based (B) methods. Both DNA and RNA constructs can be delivered using lipid nanoparticles. 469 Moreover, Jet delivery and electroporation may be applied on intramuscular as well intradermal injections. 160 , 438 , 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486

Fig. 68.2, Representative vector platforms for gene-based vaccines that have advanced into clinical trials. Vaccination by gene delivery with replication-defective recombinant viral gene-based is shown above with its genetic organization and virus structure of the natural replication-competent virus.

In this chapter, we review gene-based vaccine approaches that have advanced through clinical trials, together with considerations of the individual vectors themselves and their influence on induction of effector arms of the immune system. Over the past few years, emerging and reemerging pathogens, immunotherapy studies, and the COVID-19 pandemic have provided dramatic field opportunities—along with a host of real-world challenges—for evaluating genetic platforms alongside traditional vaccine strategies. This has moved genetic vaccines from the periphery of vaccine tools to center-stage. Following the publication of the SARS-CoV-2 genome sequence in January 2020, , the development of multiple vaccine candidates using diverse platforms was undertaken on an unprecedented timescale, providing an unique window for platform evaluations and comparisons. Adenoviral-based vaccines from AstraZeneca, CanoSino, and Janssen, along with mRNA vaccines from Moderna and Pfizer, were among the first SARS-CoV-2 vaccines licensed and are making a substantial contribution to global protection against COVID-19 ( Fig. 68.3 ). The Pfizer product, Comirnaty, became the first FDA-approved COVID-19 vaccine after only 18 months’ development. An intradermally (ID) delivered DNA vaccine, Zydus-C, demonstrated efficacy in a trial performed during an outbreak of SARS-CoV-2 B.1.617.2/Delta variant of concern (VoC) and received emergency-use approval in India in August 2021. The speed of development, testing, and deployment has been unprecedented. Genetic vaccine approaches have challenged traditional thinking on vaccine development, from inception to regulatory evaluation and deployment. Here we review the important features of these newly proven and now central platforms that have brought us to this exciting point in their history.

Fig. 68.3, Number of country regulatory approvals for each COVID-19 vaccine. As of August 9, 2021, 22 vaccines were approved for use against SARS-CoV-2 globally, of which 10 are genetic vaccines. 489 Moreover, 75% of the approved vaccines used in 39 countries are genetic, by far the largest footprint. The size of the balls represent the number of country approvals as shown in the top part of the figure. The box on the bottom represents a zoom insert showing the vaccines that were approved in fewer countries, thus their smaller sphere-size, which is magnified for displaying the detail.

VIRAL VECTOR SYSTEMS

Advances in molecular virology have facilitated an understanding of the regulation of viral replication, gene expression, and molecular pathogenesis. At the same time, this understanding has enabled the development of novel viral vectors useful for vaccination. A variety of such vectors have now advanced to preclinical and clinical studies (see Fig. 68.1 ). Their ability to target antigen-presenting cells (APCs), the ease of developing packaging cell lines, the inherent immunogenicity of both the vector and insert, and other factors (see Table 68.1 ), all supported gene-based vectors as important tools for vaccine development.

TABLE 68.1
Advantages and Considerations of Genetic Vaccines
  • Plug and play systems allow for rapid speed of development

  • In vivo production of antigen mimics immune response aspects of live-attenuated vaccination

  • Potent immunogenicity in animal and human infectious diseases

  • Induction of both cellular and humoral immunity

  • Ease of analysis and development in the laboratory

  • Non-live, non-integrating, non-spreading, transient delivery

  • Rapid production capability for emerging infectious diseases (demonstrated for adenoviral and nucleic acid approaches)

  • Many potential prime-boost combinations

  • Favorable conceptual safety profile for non-replicating generic vaccines

  • No anti-vector serology allowing for repeat boosting and no limiting pre-existing serology (demonstrated for nucleic acid approaches)

  • No vector interference for multiple immunizations (demonstrated for nucleic acid approaches)

  • Some platforms do not require cold chain (DNA and some viral-vectored approaches)

Poxvirus Vectors for Immunization

Recombinant poxviral vectors initiated the field of genetic vaccines almost 40 years ago. The efficacy of vaccinia virus against smallpox represents one of the best examples of the impact of vaccination on infectious diseases. However, safety issues with using vaccinia strains against smallpox were of concern. Attenuated vaccinia viruses were reimagined as delivery vectors for gene-encoded antigens to drive immunity against specific pathogens in the laboratories of Enzo Paoletti and Bernie Fields in the early1980s. , Poxvirus vectors show thermostability, an ability to incorporate a large foreign transgene without compromising their replication capacity, and a lack of persistence or genomic integration, and important field impact through the successful eradication of smallpox.

One of the two major attenuated strains of poxvirus Modified Vaccinia Ankara (MVA) was developed by repeated passaging of the Ankara vaccinia strain on primary chicken embryo fibroblasts (CEFs), resulting in a safer better tolerated vaccine against smallpox. A second attenuated strain, the New York vaccinia strain (NYVAC), was developed through genetic modification including the deletion of 18 open reading frames associated with virulence and host range in the Copenhagen strain. , , , NYVAC has a block at an early stage of replication, although it is able to replicate productively in African green monkey kidney cells and primary CEFs.

ALVAC, derived from a plaque-purified virus isolated from an existing canarypox strain, canapox, does not replicate but does express inserted transgenes and has demonstrated immunogenicity in both animal studies and clinical trials. , , Additional avipox vectors including fowlpox and canarypox have been developed and studied in the clinic.

Polyvalent poxviral recombinants have been used to immunize experimental animals and have been studied in a variety of infectious disease models, including rabies, measles, simian immunodeficiency virus (SIV), canine distemper, respiratory syncytial virus (RSV), malaria, , and influenza. In addition, these vectors have been studied in a variety of human immunodeficiency virus 1 (HIV-1) challenge models in animals. Multiple clinical studies have also been conducted with vaccinia, NYVAC, , and ALVAC. , The RV144 Phase III study in Thailand evaluated ALVAC- EnvGag/Pol (clade B and AE) in combination with gp120 protein boosting. A subpopulation showed 31% reduction in the frequency of acquisition of HIV-1 infection among vaccinated heterosexual men and women when compared with placebo recipients. While a landmark study, the degree of efficacy was modest, and developing these vaccines has been challenging. For example, the NIH HIV Vaccine Trials Network created a copy study of the RV144 trial. However, this trial did not reach its efficacy endpoint putting a damper on the poxviral approach for HIV vaccine development. Difficulties with poxviral vectors include that the recombinant transgenes represent a small minority of gene products expressed in this otherwise large vector. Additionally, antivector immunity remains a concern with booster regimens.

An important poxviral vaccine was tested in a human malaria challenge model. The vaccine expressed a polyprotein insert consisting of a string of six pre-erythrocytic antigens designed from Plasmodium falciparum . The vaccines were safe; however, induced T-cell IFN-γ enzyme-linked immunosorbent spot (ELISPOT) responses were low and no vaccine efficacy was observed. A novel MVA vaccine expressing the antigen 85A of Mycobacterium tuberculosis was evaluated in a placebo-controlled Phase IIb trial in South Africa in infants. Those who had previously received bacille Calmette-Guérin (BCG) vaccination were randomized to receive either MVA85A or placebo ID and then followed for immune responses and protection against tuberculosis. The immunogenicity was modest and there was no efficacy against tuberculosis infection or disease.

Poxviral vectors have been important tools in multiple cancer immune therapies protocols with positive signals reported. Outcomes include activation of T cells, particularly CD4+ and some CD8+ responses induced against tumor antigen targets resulting in clinical regressions. Positive efficacy was reported in a Phase IIb study using a recombinant poxviral vector for treatment of prostate disease (Prostvac-V/F), which was developed by the National Cancer Institute as a possible treatment for men with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). The gene inserts include a sequence modified immune-stimulatory prostate-specific antigen (PSA) cassette along with additional gene cassettes for three human immunostimulatory molecules (known as TRICOM), leukocyte function-associated antigen-3, intercellular adhesion molecule-1, and B978-0-323-79058-1.1. The therapeutic immunization generated anti–PSA CD8+ T cell responses in up to 57% of vaccinees, but limited antibody responses. , , Bavarian Nordic in conjunction with the National Cancer Institute evaluated Prostvac with or without GM-CSF (n = 432 in each arm) in the PROSPECT double-blind, randomized, Phase III efficacy trial (NCT01322490) in patients with mCRPC. The outcome of median overall survival (OS) was 34.4 months in the Prostvac-V/F group and 33.2 months in the Prostvac-V/F plus GM-CSF group, and neither group showed an improvement versus the placebo arm. These studies suggest that improvement in poxviral transgene-driven immunity remains a focus. Multiple studies of poxviral vectors now focus on prime-boost approaches.

Replication-Defective Adenoviral Vectors

Recombinant adenoviral vector vaccines (AdVV) are an appealing vaccine platform due to their cost-effective, rapidly scalable production and their stability at traditional cold-chain storage temperatures, facilitating storage and transportation logistics for global vaccine distribution. , AdVV vectors can be genetically modified to deliver specific transgene antigens of ∼7.5 kb to replicating and quiescent host-cell populations but are unable to grow on their own and thus are replication-defective. The desired transgene is inserted into the virus genome in place of the E1 region under the control of a highly active promoter. Through receptor host-cell interactions AdVV transduce cells and synthesize their encoded gene products, permitting their processing and presentation to the immune system. The AdVV genome is expressed episomally within the host nucleus, and does not integrate into the host genome. AdVV induce humoral responses with broad effector functions, and Th1-skewed CD4+ and CD8+ T cells with diverse T-cell receptor repertoires. , Important developments with AdVV have led to an improved platform ( Fig. 68.4 ) that induces consistent immunogenicity and protective outcomes in a variety of animal models. ,

Fig. 68.4, Adenoviral vaccine production overview. Viral-vectored vaccines feature a viral vector, such as an adenovirus (top) or a poxvirus (bottom) as a nonreplicating antigenic carrier. To make an adenovirus, a specific serotype is selected (A) . This may be an adenovirus that infects human as well as other species, such as chimpanzee (not shown). The adenoviral vector is modified to make it replication-deficient (B) followed by recombination to insert the antigen of interest via transfer vector (C) . Recombination may be achieved through a variety of techniques such as recombinase-mediated insertion, a shuttle vector, or a helper adenovirus among others. After recombination, the vector is linearized and grown in a replication-competent cell line, such as PER.C6® or modified HEK-293T cells expressing the E1 gene of human adenoviruses (D) . 472 , 473 Similar to adenoviruses, poxvirus vectors start with a replication deficient poxvirus such as modified vaccinia Ankara (MVA) (E) . A shuttle vector containing the sequence for the antigen of interest is recombined with the parental genome and grown in specialized culture such as a chicken embryo fibroblast cell line (F) . Production of the recombined vector is followed by plaque purification (G). 470 , 471 At this stage, viral vectors are purified and undergo quality control steps (H) before formulation into a vaccine (I) .

Third-generation AdVV represents the majority of adenovirus vectors under investigation as vaccine candidates. These AdVV are rendered replication-defective by the removal of the E1 and E3 regions of the adenovirus genome, which mediate early lifecycle replication and viral-immune evasion, respectively. , Due to these deletions AdVV must be propagated in E1 trans-complementing recombinant cell lines such as HEK293 or PER.C6 TetR cells. ,

Cell Substrates for AdVV

Changes in regulatory requirements have been critical for advancing more efficient production methods, allowing for improved product development based on the adenoviral platform ( Fig. 68.4 ). The development of transformed cell lines for adenovector propagation, in contrast to the previous standard of avian leucosis-free primary CEFs, represented a major advance in viral vaccine production technology. These cell lines offer improved yields and stable production capacity. The development of these lines was exceptionally rigorous over many years in order to address important regulatory concerns regarding adventitious agents, tumorigenicity, and other safety and consistency-of-production issues.

Oversight and evaluation of the strengths and limitations of these cell substrates continue, based on guidelines created several years ago, , enabling improved characterization and availability of an increasing number of such lines. The PER.C6 TetR, HEK293, and GV11 cell lines, for example, have supported production of clinical-grade Ad5, Ad26, and simian adenoviruses, ChAd3, and ChAdOx1 for vaccines research in HIV-1, Ebolavirus, MERS-COV, Marburg virus, tuberculosis, malaria, and COVID-19 among others ( Fig. 68.4 ). ,

AdVV as Vaccine Candidates: An Historical Perspective Shaped by Recent Epidemics

More than 51 human serotypes in seven subfamilies (A to G) of adenoviruses are known. , The adenovirus 5 serotype (Ad5) is derived from the adenoviral C subfamily and is the most common and well-characterized adenovirus serotype. However, the relatively high seroprevalence of Ad5 neutralizing antibodies in certain human populations , , has suggested possible limitations to the global deployment of Ad5 vectors. Although pre-existing immunity has been shown to reduce the immunogenicity of Ad5 vaccines in mice, , rhesus monkeys, and potentially in humans, , it does not appear to completely block vaccine immunogenicity. Strategies to circumvent pre-existing immunity, such as engineering novel Ad5 vectors containing chimeric fiber and hexon virion proteins, chemically inhibiting antivector antibody binding with, for example, polyethylene glycol, or using oral–mucosal or nasal routes of administration, , , are areas of active investigation. Another approach to circumvent pre-existing immunity is to use rare human Ad serotypes (e.g., Ad26, Ad35, Ad11, Ad4, Ad7) or adenoviral vectors derived from other species such sheep, pigs, cows, and nonhuman primates (NHP; e.g., ChAdOx1, GRAd32, ChAd36, , SAdV23 94 ). In recent years, numerous human- and simian-derived AdVV have been developed as vaccine candidates for HIV-1, , Ebola, RSV, Middle East Respiratory Syndrome (MERS), and COVID-19. ,

Ad5 has been well characterized in a variety of animal models supporting its advancement in clinical studies. Ad5 vaccines and DNA-prime/Ad5-boost combinations conferred partial protection in rhesus macaques against multiple HIV isolates, including SHIV-89.6P, , SIVmac239, and SIVmac251. , , Accordingly, Phase I and Phase II clinical studies with replication-defective adenoviral vectors for HIV-1 were advanced. The STEP study, a large Phase III efficacy study, evaluated the effect of an Ad5 vaccine encoding Gag , Pol , and Nef genes of HIV-1 on prevention of infection or controlling viral load. Although the Ad5 vaccine induced both humoral and limited cellular immunity, no reduction in HIV-1 acquisition or improvement in long-term control of postinfection viremia was observed. Further analyses revealed that persons with specific human leukocyte antigen (HLA) types, as well as those who developed a CD8+ T-cell response to certain Gag and Nef HIV-1 epitopes, selected against viruses that contained the vaccine epitope in vivo. Additionally, there was an unexpected increase in HIV-1 infection in a subgroup of vaccinees who were both uncircumcised and immune to Ad5 before vaccination during the first 18 months postimmunization. A follow-up prime-boost study using 505 DNA-prime rAd5-boost had a similar outcome and ended early due to futility. However, the Ad5 serology insights gained from these studies spurred research into additional adenovirus serotypes and nonhuman adenoviral vector approaches.

Seroepidemiology analyses of Ad serotypes suggest that pre-existing immunity to Ad11, Ad35, and Ad26 is less common than observed with Ad5, thereby making these vectors attractive candidates for vaccine development. , A prime-boost regimen of Ad26 and Ad35 vaccine vectors expressing the Ebola virus glycoprotein confers protection in NHP models, irrespective of prior Ad5 immunity. Ad26.ZEBOV is an Ad26 AdVV expressing the Ebola virus glycoprotein that was developed by Janssen as part of the public health response to the 2014–2016 West African Ebola epidemic. Ad26.ZEBOV is administered as a prime dose followed by an 8-week booster dose of MVA-BN-Filo, an MVA vaccine encoding glycoproteins from Ebola virus, Sudan virus, Marburg virus, and Tai Forest virus nucleoprotein. , Ad26.ZEBOV/MVA-BN-Filo became the first AdVV-vaccine regimen to receive regulatory approval from the European Medicines Agency (EMA) in July 2020 and is a prospective tool to mitigate future Ebola outbreaks. Ad26.ZEBOV priming induced antibody and T-cell responses to Ebola virus glycoprotein 14 days postimmunization during early safety and immunogenicity studies. Substantial strengthening of humoral and cellular immune responses following the MVA-BN-Filo booster was maintained for 8–12 months across multiple Phase I studies. The safety profile of Ad26.ZEBOV prime and MVA-BN-Filo boost support licensure in adults , , and children. As conducting traditional randomized controlled efficacy studies of a prophylactic vaccine is only feasible with large outbreaks, manufacturers and regulators have relied on immunobridging analyses to infer the efficacy of Ad26.ZEBOV/MVA-BN-Filo against Ebola virus. Analyses using a fully lethal Ebola Kikwit NHP challenge model have estimated that antibody titers elicited by the prime-boost regimen should confer protection against mortality from Ebola virus. Janssen has also utilized the Ad26 vaccine platform to develop Ad26.RSV.preF, an AdVV expressing the prefusion spike protein of RSV. Ad26.RSV.preF induced Th1-skewed immunity and was protective against enhanced respiratory disease upon RSV challenge in murine models and during human challenge studies. Ad26.RSV.preF was 80% effective in protecting against lower respiratory tract illness in adults aged >65 years, supporting further clinical development.

In recent years, the utility of simian-derived adenoviruses as vaccine platforms for emerging infectious diseases (EID) has been demonstrated. Seroepidemiology studies have observed low levels of pre-existing neutralizing antibodies against chimpanzee adenoviral vectors in sub-Saharan Africa (1.7–18.7% for AdC6, AdC6, and AdC1 vs 55.8–95.8% for hAd5), the United States, and the United Kingdom. Simian-derived adenoviruses have shown promise as vaccine candidates for Ebola and MERS-CoV. A simian-derived replication-defective adenovirus (ChAd3) vaccine engineered to express the Ebola virus glycoprotein (ChAd3-EBO-Z) was immunogenic and induced protection against acute lethal challenges in rhesus macaques. When ChAd3-EBO-Z prime was boosted with a MVA vaccine also encoding the Ebola glycoprotein, more durable protection against lethal Ebola virus challenge was observed increasing from 5 weeks to 10 months. ChAd3-EBO-Z has been observed to be well tolerated and immunogenic in multiple clinical studies. Anti-Ebola virus glycoprotein antibody titers were detectable 1-month postimmunization, , , , , were largely maintained through 12 months, and were higher in individuals who received MVA boosts. , , , , ChAdOx1 is an AdVV vaccine platform developed by the University of Oxford that has been utilized to develop vaccines for EID. ChAdOx1 MERS is a simian-derived replication-defective adenovirus that expresses the spike glycoprotein of MERS-CoV. , Single-doses of ChAdOx1 MERS elicited neutralizing antibodies and cellular immune responses that conferred protection to humanized DPP4 mice and rhesus macaques from lethal MERS-CoV challenge, and reduced viral shedding and nasal discharge in Dromedary camels (i.e., the intermediate host of MERS-CoV). Single-dose ChAdOx1 MERS elicited humoral and cellular immune responses against MERS-CoV spike protein 14 days postimmunization and was well tolerated, and elucidation of the clinical efficacy of ChAdOx1 MERS is ongoing. The ChAdOx1 platform was used to develop AZD1222 (ChAdOx1 nCOV-19) to help mitigate the COVID-19 pandemic. Within the first year of deployment adenovirus-based COVID-19 vaccines derived from human adenoviruses (e.g., Ad26.COV.S, Ad5-nCoV, and Gam-COVID-Vac) and simian-derived adenoviruses (e.g., AZD1222) have demonstrated the utility of AdVV in combating an EID through their rapid deployment and development, as discussed next in context of the COVID-19 pandemic. , , ,

AdVV Platform Validation: Adenoviral Vectors as COVID-19 Vaccines

The rapid development of vaccines during the COVID-19 pandemic has yielded numerous adenovirus-vectored COVID-19 vaccine candidates. Four AdVV were among the first COVID-19 vaccines licensed for use, namely; AZD1222, Ad26.COV2.S, Ad5-nCoV, and Gam-COVID-Vac, a heterologous prime-boost combination of Ad26 and Ad5. Studies with other simian (e.g., GRAd32, ChAd36, , SAdV23 94 ) and human adenoviral vectors (e.g., human Ad49, single-cycle Ad6 154 ) are currently ongoing.

Characteristics of AdVV for COVID-19 Vaccines

The SARS-CoV-2 spike (S) glycoprotein is involved in angiotensin-converting enzyme (ACE) 2 receptor recognition, viral attachment, and entry into host cells. Functionally, the S glycoprotein is characterized by a short N-terminal signal peptide, an S1 domain that mediates host cell receptor binding via a receptor-binding domain (RBD), and an S2 domain that mediates virus membrane fusion. Due to its indispensable functions, first-wave adenovirus-based COVID-19 vaccine candidates were predominantly developed to target the S glycoprotein. , Other vaccine candidates such as AdCOVID specifically target the S glycoprotein RBD, while VXA-CoV2-1 158 and the hAd5 bivalent vaccine target both the SARS-CoV-2 nucleocapsid and S glycoprotein.

Once administered, adenovirus-vectored COVID-19 vaccines infect host cells and initiate production of the SARS-CoV-2 S glycoprotein (see Fig. 68.5 ). , , AZD1222 and species E adenovectors enter host cells via the widely expressed coxsackie and adenovirus receptor, while Ad26 and other species D adenovectors utilize CD46 for host-cell entry. Currently licensed adenovirus-vectored COVID-19 vaccines are administered via a deltoid intramuscular (IM) injection , , , , and other routes of administration are currently being explored, for example, intranasal and aerosolized. Preclinical evidence suggests that intranasal administration of AdVV can induce local respiratory tract mucosal immunity, reducing viral shedding, and conferring host protection from severe disease. , , A Phase I study demonstrated that two doses of intranasally administered Ad5-nCoV elicited comparable humoral and cellular immune responses to a single IM immunization.

Fig. 68.5, Schematic of immune response induced by adenovirus-based COVID-19 vaccines. 132 Species E adenovectors (e.g., AZD1222) enter host cells via the widely expressed coxsackie and adenovirus receptor (CAR), species D adenovectors (e.g., Ad26.COV.2) utilize CD46 for host cell entry. Upon host cell entry AdVV begin to express the SARS-CoV-2 S glycoprotein transgene. Host cells in turn release the SARS-CoV-2 spike protein, which is in turn processed by antigen presenting cells. S glycoprotein epitopes are presented to local and lymph node resident B and T cells to stimulate an adaptive immune response. Memory B and T cells are also induced conferring durable immunity.

Current adenovirus-vectored COVID-19 vaccine candidates have been licensed as either single-dose or two-dose homologous prime-boost regimens. , Preclinical studies of AZD1222, , , hAd5, and Ad26.COV2.S , demonstrated that although single-dose vaccination induces SARS-CoV-2 S glycoprotein-specific antibodies and cell-mediated immunity, a 4- or 8-week booster dose significantly increased titers of virus-neutralizing and RBD antibodies. Other adenovirus-based COVID-19 vaccines in development (e.g., Gam-COVID-Vac, Sad23LnCoV-S/Ad49L-nCoV-S 94 ) utilize a heterologous prime-boost strategy with two different AdVV in order to minimize potential vaccine attenuation by a host immune response to AdVV.

Immunogenicity and Reactogenicity of Adenovirus-Based COVID-19 Vaccines

The humoral and cell-mediated immune response to adenovirus-based COVID-19 vaccines has been extensively characterized in exploratory analyses of large clinical trials. A single dose of AZD1222 induced B-cell activation and proliferation and production of S glycoprotein IgA, IgM, IgG1, and IgG3 antibodies. , Subsequent systems serology analyses demonstrated that anti-S glycoprotein neutralizing antibody titers and Fc-mediated functional antibody responses (namely, antibody-dependent neutrophil/monocyte phagocytosis, natural killer cell activation, and complement activation) were substantially enhanced by a second dose of vaccine. Polyfunctional IFN-γ+, TNF-α+, Th1-CD4+, and CD8+ T cells against the SARS-CoV-2 S glycoprotein peptides were induced 14 days following single-dose immunization , and were increased 28 days postsecond immunization. T-cell receptor β sequences revealed a substantial breadth and depth of S glycoprotein epitopes for AZD1222-induced CD4+ and CD8+ T-cell responses.

Immunogenicity analyses from Phase I and Phase II studies of single-dose Ad5-nCoV immunization demonstrated that neutralizing antibodies and IFN-γ+, TNF-α+, Th1-CD4+, and CD8+ T cells were induced within 14–28 days of vaccination. , Exploratory Phase I immunogenicity analyses with Ad26.COV2.S , , illustrated that single-dose immunization elicited durable humoral responses, with a 1.8-fold reduction in neutralizing antibody titers, 8 months following immunization. S glycoprotein-specific IFN-γ+, CD4+, and CD8+ T-cell responses also showed durability and stability during this follow-up period. The immunogenicity of Gam-COVID-Vac was characterized in a Phase I/II study, wherein SARS-CoV-2 RBD-specific IgG antibody titers were observed 14 days following priming doses of either rAd26 or rAd5 and began to increase within 7 days of administering a booster dose of rAd5. S glycoprotein IFN-γ+, CD4+, and CD8+ T cells were induced following immunization, notably at 28 days following prime immunization and within 14 days of the rAd5 booster dose.

AZD1222, Ad26.COV2.S, Ad5-nCOV, and Gam-COVID-Vac were well tolerated throughout clinical development. , , , , , Injection-site tenderness, , , headache, , and fati-​gue , , were among the most frequently reported local and systematic reactions in Phase III studies. The overall reactogenicity profile has been shown to be more tolerable with lower doses of vaccine, , in individuals with increasing age, , and following a second dose. , ,

Efficacy of Adenovirus-Based COVID-19 Vaccines

The clinical profile observed in multiple Phase III studies has supported the licensure of several adenovirus-based COVID-19 vaccines that have demonstrated efficacy in preventing symptomatic SARS-CoV-2 illness (see Table 68.2 ), enabling the vaccination of billions of individuals globally within the first year of deployment. , Vaccine efficacy from adenovirus-based COVID-19 vaccines has been observed across diverse trial populations , , , including older adults, and those with comorbidities such as diabetes, cardiovascular, and respiratory disease. , , The efficacy of AZD1222 has been demonstrated in multiple randomized controlled clinical trials. , , An initial interim analysis pooling study in the United Kingdom, Brazil, and South Africa demonstrated an initial efficacy of 70.4% in preventing symptomatic SARS-CoV-2 infection and supported initial emergency use authorization in the United Kingdom. A primary analysis from a Phase III study in the United States, Peru, and Chile estimated an overall efficacy against symptomatic disease of 74.0%. Efficacy of AZD1222 at preventing infection with SARS-CoV-2 (as measured by nucleocapsid antibody seroconversion of all participants who tested positive for SARS-CoV-2 regardless of symptoms or severity) was estimated at 64.3%. Single-dose Ad26.COV2.S showed overall efficacy in preventing moderate-to-severe/critical infection of 66.9% at 14 days postimmunization and 66.1% at 28 days postimmunization. An interim analysis of a Phase III study of Gam-COVID-Vac—a heterologous 21-day prime-boost of Ad26 (prime) and Ad5 (boost)—demonstrated an efficacy of 91.6% against symptomatic infection 21 days postdose 1 (Ad26), that is, on the day of dose 2 (Ad5), AZD1222, , Ad26.COV.S, and Gam-COVID-Vac were highly protective against severe disease, hospitalization, and death during their respective Phase III studies.

TABLE 68.2
Efficacy in Phase III Clinical Studies and Real-World Effectiveness of Adenovirus-Based COVID-19 Vaccines , , , , , , , , , , , , , , , , , , , , , ,
Phase III Clinical Studies
Vaccine Studies N Efficacy Against
AZD1222 (ChAdOx1 nCov-19) ISRCTN89951424,
NCT04324606,
NCT04400838,
NCT04444674
11,636 * Symptomatic infection: Interim efficacy (Dec 2020)
Overall
SD/SD
LD/SD
70.4%; 95.8% CI: 54.8–80.6
62.1%; 95% CI: 41.0–75.7
90.0%; 95% CI: 67.4–97.0
17,178 * Symptomatic infection: Updated efficacy (Feb 2021)
Overall
Single-dose efficacy
≥12 weeks booster interval
<6 weeks booster interval
66.7%; 95% CI: 57.4–74.0
76.0%; 95% CI: 59.3–85.9
81.3%; 95% CI: 60.3–91.2
55.1%; 95% CI: 33.0–69.9
NCT04516746 32,459 Symptomatic infection:
Overall
Individuals aged >65 years
Preventing infection (via nucleocapsid seroconversion)
74.0%; 95% CI: 65.3–80.5
83.5%; 95% CI: 54.2–94.1
64.3%; 95% CI: 56.1–71.0
Ad5-nCoV NCT04526990 40,000 Ongoing Phase III study
Ad26.COV2.S NCT04505722 39,321 Moderate-to-severe–critical infection
14 days following immunization
28 days following immunization
66.9%; 95% CI: 59.0–73.4
66.1%; 95% CI: 55.0–74.8
Gam-COVID-Vac NCT04530396 21,977 Symptomatic infection
21 days following rAd26 immunization (day of receiving rAd5 dose)
91.6%; 95% CI: 85.6–95.2

Efficacy Against VoCs Observed in Phase III Studies
Vaccine Studies N SARS-CoV-2 Variant Efficacy Against
AZD1222 (ChAdOx1 nCov-19) NCT04400838 8534 Alpha Symptomatic infection Alpha
Symptomatic infection non-Alpha
70.4%; 95% CI: 43.6–84.5
81.5%; 95% CI: 67.9–89.4
NCT04444674 2021 Beta Symptomatic infection Beta 10.4%; 95% CI:−76.8–54.8
ISRCTN89951424 4772 Gamma
P2
B.1.1.28
B.1.1.33
Symptomatic infection Gamma
Symptomatic infection P.2
Symptomatic infection B.1.1.28
Symptomatic infection B.1.1.33
63.6%; 95% CI:-2.1–87.0
68.7%; 95% CI: 54.9–78.3
72.6%; 95% CI: 46.4–86.0
88.2%; 95% CI: 5.4–98.5
Ad26.COV2.S NCT04505722 2473 Beta Moderate-to-severe critical infection
14 days following immunization
28 days following immunization
52.0%; 95% CI: 30.3–67.4
64.0%; 95% CI: 41.2–78.7

Vaccine Effectiveness Against VoCs Observed in Real-World Studies
Vaccine Population N SARS-CoV-2 Variant Efficacy Against
AZD1222 (ChAdOx1 nCov-19) Individuals in the United Kingdom who were SARS-CoV-2 positive with Alpha or Delta VoCs § 19,109 Alpha Symptomatic infection
14 days postsecond immunization
74.5%; 95% CI: 68.4–79.4
Delta Symptomatic infection 14 days postsecond immunization 67.0%; 95% CI: 61.3–71.8

* Pooled interim analysis of safety and efficacy from four studies; vaccine efficacy was initially determined from 11,636 individuals from the COV002 (Phase II/III; United Kingdom), COV003 (Phase III; Brazil) studies. Updated efficacy was later determined using 17,178 individuals from all four studies.

95.8% CI used for primary efficacy analysis.

SD/SD and LD/SD seronegative efficacy cohorts only. CI, confidence interval; LD, low dose; N/A, not available; NS, not stated; SD, standard dose; VoC, variant of concern.

The rapid vaccination rollout in response to the pandemic has provided invaluable insights on the effectiveness of adenovirus-based COVID-19 vaccines. For example, multiple studies have estimated the effectiveness of two doses of AZD1222 at preventing symptomatic infection as 77–89%. Emerging data also suggest that vaccination may reduce disease transmission by 30–47%. , The emergence of new variants of SARS-CoV-2 possessing mutations in the S glycoprotein remains a focus of significant international attention following early observations of increased transmissibility with SARS-CoV-2 B.1.1.7/Alpha 182,186 and Delta, , reduced vaccine efficacy against mild-to-moderate disease with SARS-CoV-2 B.1.351/Beta, , and potential immune evasion mutations in B.1.1.28/Gamma variant/P.1 VoCs , (see Table 68.2 ). While investigations of vaccine effectiveness against VoC are ongoing, insights from real-world effectiveness studies suggest that existing COVID-19 vaccines may still confer adequate protection against severe disease, hospitalization, and death. The emergence of SARS-CoV-2 VoC emphasizes that ongoing vaccine development and vaccine re-engineering may be required for the duration of the pandemic.

Adeno-Associated Viruses

The adeno-associated viruses (AAVs) were defined initially as “helper” viruses that facilitated the propagation of wild-type adenovirus in cell culture. In contrast to the large genome sizes of rAd and vaccinia vectors, genome sizes of AAV are more limited, with an insert size of ∼5 kb. Like other replication-defective viruses, these particles can be produced in packaging lines that provide complementary structural proteins made constitutively by the cell rather than the virus. A variety of serotypes have been defined, and an HIV vaccine expressed in AAV2 has been evaluated in Phase I human studies, with poor immunogenicity results. Alternative serotypes, including AAV1, are currently under development and may be assessed both alone and in prime-boost combinations for efficacy in humans. A unique use of these vectors has also been described recently. Recombinant adeno-associated virus vectors have been studied as a platform to deliver recombinant antibody genes for direct in vivo production of antibodies. This strategy—vector immune prophylaxis—allows previously identified rare neutralizing antibodies to be engineered into a recombinant AAV, which, upon infection, results in in vivo antibody production of protective antibodies. Vector immune prophylaxis also allows for production of protective antibodies in vivo that have not been produced through vaccination. Studies have demonstrated the effectiveness of the delivery strategy in protecting NHPs against SIV, , humanized mice against HIV-1, , and mice and ferrets against influenza. , , This approach is undergoing clinical evaluation as a strategy to inhibit HIV-1 infection. Preclinically, it was observed that delivery to a subset of macaques could be associated with good vector take and IgG expression in animals. In one study, an individual NHP was examined in detail ; after receiving administration of AAV vector coding for a neutralizing anti-SIV antibody, a rhesus macaque showed expression levels of ≥200 µg/mL for several years, which conferred protection from SIVmac239 challenge. Delivery to the upper airway or nose using an AAV-IgG to prevent viral infection has been described in the case of influenza with success in animal models. Nucleic-acid–based vaccines are focusing on this approach as well. These tools could provide important options for immune protection or therapy in immunization poor responding populations.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here