A subunit vaccine is a type of vaccine that contains one or more purified components, or antigens, from a pathogen—such as proteins, polysaccharides, or peptides—rather than the entire microorganism, designed to elicit a targeted immune response without the risk of causing infection.[1] These vaccines focus on the specific parts of the pathogen that the immune system recognizes as foreign, thereby stimulating the production of antibodies and T-cell responses to protect against the disease.[2] Unlike live-attenuated or inactivated vaccines, subunit vaccines avoid using whole pathogens, making them safer for individuals with compromised immune systems.[3]The development of subunit vaccines emerged in the early 20th century as an advancement over earlier vaccine types, with toxoid vaccines—such as those for tetanus and diphtheria, which use inactivated bacterial toxins as subunits—being among the first examples developed in the 1920s and licensed in the 1930s.[4][5] The breakthrough in recombinant DNA technology in the 1980s led to the first modern protein subunit vaccine, the hepatitis B vaccine (Recombivax HB), approved by the FDA in 1986, which uses yeast-produced viral surface antigens that self-assemble into virus-like particles.[6] Subsequent innovations included acellular pertussis vaccines in the 1990s, replacing whole-cell versions to reduce side effects,[7] and the human papillomavirus (HPV) vaccine Gardasil in 2006, utilizing virus-like particles derived from the L1 capsid protein.[8] More recently, protein subunit vaccines have been pivotal in responses to emerging threats, such as the Novavax COVID-19 vaccine authorized in 2021 and included in updated formulations as of 2025, which employs a recombinant spike protein stabilized in a prefusion conformation.[9][10]Subunit vaccines are typically produced through recombinant expression systems, such as bacteria, yeast, or mammalian cells, where the target antigen is genetically engineered, purified, and often combined with adjuvants to enhance immunogenicity.[11] This production method allows for precise control over the antigen, enabling scalability and consistency, though it may require multiple doses or boosters due to potentially weaker initial immune stimulation compared to live vaccines.[12] Common examples in routine immunization include the hepatitis B vaccine, acellular pertussis components in DTaP, and the shingles vaccine Shingrix, which uses a glycoprotein E antigen from varicella-zoster virus.[3]One key advantage of subunit vaccines is their high safety profile, as they contain no live or whole pathogens, minimizing risks of reversion to virulence or allergic reactions in sensitive populations.[13] They also offer stability for storage and transport, which is beneficial in resource-limited settings.[2] However, their immunogenicity can be limited, often necessitating adjuvants like aluminum salts or novel ones such as AS01 in Shingrix to boost responses, and they may not induce as broad cellular immunity as whole-organism vaccines.[14] Despite these challenges, ongoing research into advanced delivery systems, such as nanoparticles and mRNA-encoded subunits, continues to expand their efficacy against complex pathogens like HIV and malaria.[3]
Fundamentals
Definition and Principles
Subunit vaccines are a class of immunizations that incorporate purified antigenic components derived from a pathogen, such as proteins, polysaccharides, or peptides, rather than the entire microorganism. These components are selected specifically to provoke an immune response capable of conferring protection against the disease-causing agent without introducing the risk of infection from the whole pathogen.[2][13][1]The core principles of subunit vaccines revolve around targeting immunogenic epitopes—distinct molecular regions on the antigen that are recognized by the immune system—to elicit focused humoral immunity through antibody production and cellular immunity via T-cell activation. Unlike whole-pathogen approaches, subunit vaccines are non-replicating, which enhances their safety profile by eliminating the potential for pathogen revival or unintended replication in the host, making them suitable for immunocompromised individuals. Adjuvants are often incorporated to amplify these responses, compensating for the inherently lower immunogenicity of isolated subunits compared to intact organisms.[1][15][13]Subunit vaccines differ fundamentally from other vaccine modalities in their composition and mode of antigen presentation. In contrast to live-attenuated vaccines, which use weakened pathogens that replicate in the host to mimic natural infection, subunit vaccines pose no risk of reversion to virulence or disease causation. They also diverge from inactivated vaccines, which employ killed whole pathogens and may retain extraneous components that could trigger unwanted reactions, by focusing solely on purified antigens for a more targeted effect. Unlike nucleic acid vaccines, such as mRNA or DNA types, subunit vaccines deliver pre-formed antigens directly rather than instructing host cells to produce them, avoiding concerns related to genetic material integration or transient expression.[2][1][16]The concept of subunit vaccines was first conceptualized in the early 20th century as a safer alternative to whole-cell vaccines, with initial developments like bacterial toxoids in the 1920s paving the way for purified antigen strategies that gained prominence in the mid-20th century amid advances in immunology.[17][18]
Mechanism of Action
For protein and peptide subunit vaccine antigens, which are the most common type, these exogenous materials are taken up by antigen-presenting cells (APCs) such as dendritic cells and macrophages through endocytosis or phagocytosis.[19][20] Within APCs, these antigens undergo processing in endosomal compartments, where they are degraded into smaller peptides by proteases.[20] These peptides are then loaded onto major histocompatibility complex (MHC) class II molecules and presented on the cell surface to CD4+ T helper cells, initiating adaptive immune activation.[20] For MHC class I presentation, which is crucial for CD8+ cytotoxic T cell activation, subunit antigens often require cross-presentation pathways where exogenous material is routed to the cytosol for proteasomal degradation before peptide loading.[21]Polysaccharide antigens, in contrast, typically engage B cells directly in a T cell-independent manner, though conjugates link them to proteins for T cell involvement (see Types section for details).[22]This antigen presentation primarily elicits a humoral immune response through CD4+ T helper cell activation, which promotes B cell differentiation into plasma cells that secrete antigen-specific antibodies, such as IgG targeting pathogen surface proteins.[23] Subunit vaccines also induce cellular responses, including CD4+ T cell proliferation and cytokine production, but typically generate limited CD8+ T cell responses without additional enhancements, as the exogenous nature of antigens favors MHC II over MHC I pathways.[24] Adjuvants play a key role in overcoming this limitation by boosting immunogenicity; for instance, they promote cytokine release (e.g., IL-1β, IL-6) and APC maturation, enhancing antigen uptake and co-stimulatory molecule expression.[25] Aluminum-based adjuvants like alum specifically stimulate a Th2-biased response, favoring antibody production and IgG1/IgE class switching while modestly supporting CD4+ T cell help.[26][20]The activated immune cascade leads to the formation of immunological memory, with long-lived memory B cells and T cells persisting after initial vaccination to enable rapid recall responses upon pathogen re-exposure.[27] Memory B cells provide sustained antibody production, while memory CD4+ and limited CD8+ T cells contribute to quicker effector functions, ensuring durable protection.[27]Vaccine efficacy hinges on the selection of immunogenic epitopes that elicit broadly protective responses, such as neutralizing antibodies targeting conserved viral spike proteins to block host cell entry.[28] For example, SARS-CoV-2 protein subunit vaccines focusing on the receptor-binding domain of the spike protein generate potent neutralizing antibodies that inhibit viral attachment and fusion.[28]
Types
Protein Subunit Vaccines
Protein subunit vaccines consist of purified proteins or protein fragments derived from pathogenic organisms, serving as the primary antigens to induce protective immunity without introducing the whole pathogen. These antigens are commonly sourced from surface proteins that play key roles in pathogen-host interactions, such as viral glycoproteins or bacterial toxins. A prominent example is the recombinant spike protein from SARS-CoV-2, utilized in vaccines like NVX-CoV2373, which targets the virus's receptor-binding domain to elicit neutralizing antibodies. Similarly, the human papillomavirus (HPV) vaccine incorporates virus-like particles (VLPs) assembled from the L1 capsid protein, mimicking the native viral structure to prevent oncogenic infections.[29][30][31]Production of these vaccines relies on recombinant techniques, where the genetic sequence encoding the target protein is expressed in heterologous systems such as yeast, insect cells, or mammalian cells to yield high quantities of the antigen (detailed in the Production and Manufacturing section). This approach ensures the proteins fold into their native conformation, preserving critical structural features essential for immunogenicity. The hepatitis B surface antigen (HBsAg), a 22-nm lipoprotein particle, exemplifies this process; it is expressed in Saccharomyces cerevisiaeyeast and self-assembles into non-infectious VLPs that display conformational epitopes analogous to those on the virus. Acellular pertussis vaccines further illustrate this by incorporating purified bacterial proteins like pertussis toxin and filamentous hemagglutinin, detoxified and combined to broaden protection against Bordetella pertussis.[32][6][33]The hepatitis B vaccine marked a historic milestone as the first licensed protein subunit vaccine, approved in 1986 as Recombivax HB, transitioning from plasma-derived to recombinant production to eliminate risks of blood-borne contaminants. This vaccine elicits robust humoral responses by targeting conformational epitopes on HBsAg, leading to high-titer anti-HBs antibodies that confer long-term immunity in over 95% of healthy adults after three doses. In general, protein subunit vaccines stimulate B-cell activation and antibody production against these three-dimensional epitopes, which are crucial for viral neutralization and opsonization of bacterial components, though they may induce weaker T-cell responses compared to live vaccines.[34][35][32]Key advantages of protein subunit vaccines include their exceptional stability, allowing storage at refrigerator temperatures for extended periods—up to four years in some formulations—without loss of potency, which simplifies global distribution. Their well-defined composition also facilitates rigorous standardization and quality control during manufacturing, reducing batch-to-batch variability. However, a primary challenge is their inherently low immunogenicity due to the absence of pathogen-associated molecular patterns, often requiring adjuvants like aluminum salts or AS01 to amplify innate immune signaling and enhance both antibody and cellular responses. Despite this, their safety profile remains superior, avoiding risks of reversion to virulence seen in live vaccines.[36][6][37]
Polysaccharide Subunit Vaccines
Polysaccharide subunit vaccines are composed of purified capsular polysaccharides extracted from the outer layers of bacterial cell walls, particularly from encapsulated pathogens. These carbohydrates serve as key virulence factors by enabling bacteria to evade phagocytosis, and their isolation involves purification processes to remove impurities while preserving antigenic structure. A prominent example is the 23-valent pneumococcal polysaccharide vaccine (PPSV23, also known as Pneumovax 23), which includes 25 μg of purified capsular polysaccharide from each of 23 serotypes of Streptococcus pneumoniae, such as 1, 4, 6B, 14, and 19F, formulated in isotonic saline with phenol as a preservative.[38] Similarly, the quadrivalent meningococcal polysaccharide vaccine (MPSV4, formerly Menomune) contained 50 μg each of purified polysaccharides from serogroups A, C, Y, and W-135 of Neisseria meningitidis.[39]These vaccines induce a T-cell-independent immune response, primarily activating B cells through cross-linking of B-cell receptors without requiring T-cell assistance, leading to rapid production of antibodies. The resulting humoral response is dominated by IgM and IgG2 subclasses, which facilitate opsonization and complement activation for bacterial clearance, but it lacks class switching to other IgG subclasses or significant IgA production.[40][41] This T-independent type 2 mechanism does not generate robust immunological memory, resulting in short-lived plasma cells and minimal long-term protection upon re-exposure.[42]Polysaccharide subunit vaccines have been applied primarily against invasive diseases caused by encapsulated bacteria, targeting pathogens like S. pneumoniae that cause pneumonia, bacteremia, and meningitis in adults and older children. PPSV23, for instance, is recommended for immunocompetent adults aged 65 years and older, as well as those with certain risk factors, demonstrating efficacy in reducing invasive pneumococcal disease by 60–70% in healthy adults.[38] MPSV4 was used for outbreak control and travel-related protection against meningococcal disease, providing short-term immunity in adolescents and adults.[39] These vaccines are particularly valuable in resource-limited settings where conjugate alternatives may be unavailable.Despite their utility, polysaccharide subunit vaccines face significant challenges, including poor immunogenicity in infants and young children under 2 years due to immature marginal zone B cells and limited splenic function, often resulting in negligible antibody responses.[43] Booster doses typically fail to elicit anamnestic responses and may even induce hyporesponsiveness, diminishing efficacy over time, especially in the elderly or immunocompromised populations.[44] Age-related declines in immune competence further reduce their protective impact, with efficacy dropping to below 50% in high-risk children, highlighting the need for enhanced formulations to achieve broader T-cell involvement.[38]
Conjugate Vaccines
Conjugate vaccines are a type of subunit vaccine formed by the covalent chemical linkage of bacterial capsular polysaccharides to immunogenic carrier proteins, such as diphtheria toxoid (DT), tetanus toxoid (TT), or the non-toxic diphtheria toxin mutant CRM197. This conjugation process creates a hybrid antigen that combines the pathogen-specific B-cell epitopes from the polysaccharide with T-cell epitopes from the carrier protein, addressing the poor immunogenicity of polysaccharides alone, particularly in infants and young children whose immune systems respond inadequately to T-cell-independent antigens.[45][46]The primary mechanism of conjugate vaccines involves transforming the immune response from T-cell-independent, which typically elicits short-lived IgM antibodies without memory cell formation, to a T-cell-dependent response. This shift occurs because the carrier protein is processed by antigen-presenting cells, allowing T-helper cells to recognize its peptides and provide signals for B-cell activation, proliferation, and differentiation. As a result, conjugate vaccines promote immunoglobulin class switching to IgG subclasses, affinity maturation in germinal centers, and the generation of long-lived plasma cells and memory B cells, leading to sustained protection and booster responses upon re-exposure.[45][47][48]These vaccines have been pivotal in preventing invasive bacterial diseases, serving as the foundation for immunizations against Haemophilus influenzae type b (Hib), Neisseria meningitidis, and Streptococcus pneumoniae. For instance, Hib conjugate vaccines, which link the polyribosyl ribitol phosphate (PRP) polysaccharide to carriers like TT or outer membrane protein complex (OMPC), have virtually eliminated invasive Hib disease in vaccinated populations. Similarly, meningococcal conjugate vaccines target serogroups A, C, W, and Y, while pneumococcal conjugate vaccines like PCV13 cover 13 serotypes of pneumococcal capsular polysaccharides conjugated to CRM197, reducing invasive pneumococcal disease by over 90% in children under 5 years.[49][50][51]Conjugate vaccines emerged in the 1980s as a solution to the limitations of plain polysaccharide vaccines, which failed to induce protective immunity in children under 2 years due to immature immune responses; the first Hib conjugate vaccine was licensed in the United States in 1987. The World Health Organization now recommends their inclusion in routine childhood immunization programs worldwide, with Hib vaccine administration in a 3- or 4-dose schedule starting at 6 weeks of age, contributing to over 90% reduction in global Hib burden since widespread adoption. The selection of carrier protein is critical, as overuse of the same carrier (e.g., CRM197 in multiple vaccines) can lead to carrier-induced epitopic suppression, where pre-existing anti-carrier antibodies reduce responses to the polysaccharideantigen, potentially impacting overall vaccine efficacy and herd immunity benefits from reduced nasopharyngeal carriage.[52][53][45]
Peptide Subunit Vaccines
Peptide subunit vaccines utilize short synthetic peptide sequences, typically comprising 8 to 20 amino acids, designed to mimic specific immunogenic epitopes from pathogens, such as T-cell or B-cell epitopes.[54] These peptides can be linear or cyclic, with cyclic forms often enhancing stability and mimicking the conformational structure of native epitopes more effectively.[55] For instance, peptides derived from malaria parasite proteins, like those targeting T-cell epitopes in Plasmodium falciparum, have been synthesized to focus on precise antigenic regions without including extraneous pathogen material.[56]A key advantage of peptide subunit vaccines lies in their high specificity, which allows for targeted immune responses against defined epitopes, minimizing off-target effects and reducing the risk of allergic reactions associated with whole-pathogen vaccines.[57] Their chemical synthesis enables rapid production, high purity, and straightforward scalability, making them cost-effective and suitable for therapeutic applications, particularly in cancer immunotherapy where personalized epitope targeting is beneficial.[58] Additionally, these vaccines exhibit an excellent safety profile, with low toxicity and no risk of causing infection, as they lack replicative components.[59]Despite these benefits, peptide subunit vaccines face significant challenges, primarily their inherently poor immunogenicity, which often fails to elicit robust immune responses without enhancement.[54] This limitation arises from the small size of peptides, which hinders efficient uptake by antigen-presenting cells and proper processing for MHC presentation, frequently necessitating the co-administration of adjuvants or advanced delivery systems like nanoparticles to boost efficacy.[60] Conformational constraints also pose issues, as linear peptides may not fold correctly to replicate the three-dimensional structure of native epitopes, potentially reducing bindingaffinity to immune receptors.[61]In applications, peptide subunit vaccines have been explored experimentally for challenging infectious diseases, including HIV, where multi-epitope peptides target conserved viral regions to induce broad T-cell responses.[62] For Ebola, epitope-based designs have focused on glycoprotein peptides to elicit protective antibodies and cytotoxic T cells in preclinical models.[63]Malaria vaccine candidates, such as those using peptides from circumsporozoite protein epitopes, demonstrate potential for multi-epitope strategies that provide coverage against diverse parasite strains.[56] These approaches often incorporate multiple peptides to achieve broader immunogenicity.The evolution of peptide subunit vaccines has been markedly advanced since the 1990s through the development of peptide libraries, which enable high-throughput screening of immunogenic sequences, and computational design tools that predict epitope structures and interactions with MHC molecules.[61] Early efforts relied on empirical synthesis, but integration of bioinformatics and structural modeling has allowed for rational optimization, leading to more effective multi-epitope constructs and improved delivery formulations.[64] This progression has shifted peptide vaccines from basic prophylactic concepts toward sophisticated therapeutic platforms, particularly for chronic infections and cancers.[65]
Production and Manufacturing
Antigen Identification and Selection
Antigen identification and selection represent the foundational step in subunit vaccine development, where specific immunogenic components of a pathogen are chosen to elicit protective immunity without the risks associated with whole-pathogen vaccines. This process integrates empirical experimental techniques and computational tools to pinpoint antigens that can effectively stimulate B-cell and T-cell responses. Key methods include epitope mapping through structural biology approaches like X-ray crystallography, which resolves the atomic-level interactions between antigens and antibodies to identify neutralizing epitopes, as demonstrated in studies on viral surface proteins. Complementing this, enzyme-linked immunosorbent assays (ELISA) enable high-throughput screening of antibody binding to potential antigens, while animal challenge studies validate protective efficacy by assessing survival rates and immune correlates in vivo models. These methods collectively ensure the selection of antigens capable of inducing targeted, long-lasting immunity.Selection criteria emphasize antigens that are highly conserved across pathogen strains to minimize escape by variants, prominently exposed on the pathogen surface for accessibility to the immune system, and non-toxic to avoid adverse reactions in hosts. Additionally, candidates must steer clear of regions prone to immune evasion, such as hypervariable loops in viral envelopes that facilitate antigenic drift. For instance, surface exposure is prioritized using bioinformatics predictions of transmembrane domains and extracellular localization signals, which have proven effective in identifying vaccine targets for intracellular pathogens. Non-toxicity is assessed through sequence homology checks against host proteins to prevent autoimmunity, ensuring the antigen's safety profile aligns with regulatory standards.Computational tools accelerate this pipeline by predicting immunogenic epitopes from genomic data. The Immune Epitope Database (IEDB) serves as a primary resource for analyzing B-cell and T-cell epitopes, integrating experimental data to forecast binding affinities and immunogenicity scores for novel candidates. Reverse vaccinology, a genomics-driven approach, scans entire pathogen genomes to select surface-exposed proteins with adhesive properties and low similarity to human proteins, as pioneered in meningococcal vaccine development and extended to other bacteria and viruses. This method has identified over 600 potential antigens in Neisseria meningitidis, highlighting its efficiency in prioritizing candidates for empirical validation.Representative examples illustrate these principles in practice. For SARS-CoV-2 subunit vaccines, the spike protein was selected due to its surface exposure, role in host cell entry, and conservation in receptor-binding domains, enabling broad neutralization across variants in candidates like Novavax's NVX-CoV2373. In bacterial conjugate vaccines, capsular polysaccharides from Streptococcus pneumoniae were chosen for their immunogenicity and strain coverage, conjugated to carrier proteins to enhance T-cell help and overcome T-independent responses. These selections underscore the balance between specificity—targeting key pathogenic mechanisms—and breadth to counter strain diversity.A primary challenge in antigen selection lies in reconciling broad immunogenicity against diverse variants with sufficient specificity to avoid non-protective or reactogenic responses. Highly conserved antigens may elicit weaker responses due to immune tolerance, while variable ones risk obsolescence from mutations, as observed in evolving SARS-CoV-2 lineages where epitope escape reduces vaccine efficacy. Addressing this requires iterative refinement, combining multi-antigen formulations to expand coverage without diluting potency.
Recombinant Expression Systems
Recombinant expression systems are biotechnological platforms that utilize genetically engineered host organisms to produce subunit vaccine antigens, enabling scalable manufacturing of purified proteins or virus-like particles (VLPs). These systems involve inserting the gene encoding the target antigen into the host's genome or a vector, followed by expression under controlled conditions to yield high quantities of the recombinant protein. The choice of system depends on the antigen's structural requirements, such as the need for proper folding, disulfide bond formation, and post-translational modifications like glycosylation, which are critical for immunogenicity and stability.[6]Bacterial systems, particularly Escherichia coli, offer high yields and low production costs due to rapid growth and simple cultivation in inexpensive media. These prokaryotic hosts excel in expressing simple, non-glycosylated proteins but lack the machinery for eukaryotic post-translational modifications, often resulting in misfolded or insoluble proteins that require refolding steps. For instance, attempts to produce hepatitis B virus (HBV) surface antigen (HBsAg) in E. coli yield non-glycosylated particles with reduced immunogenicity compared to eukaryotic systems, highlighting limitations for glycoproteins. Despite these challenges, bacterial expression is suitable for peptide antigens or when modifications are unnecessary, achieving titers up to several grams per liter in optimized strains.[66][6]Yeast systems, such as Saccharomyces cerevisiae and Pichia pastoris, provide eukaryotic advantages including proper protein folding, secretion into the culture medium for easier purification, and basic glycosylation patterns, while maintaining relatively low costs and high expression levels. S. cerevisiae has been pivotal for the HBV vaccine, where recombinant HBsAg self-assembles into immunogenic VLPs with yields of 10-20 mg/L, demonstrating effective disulfide bond formation absent in bacteria. S. cerevisiae, which is utilized in the human papillomavirus (HPV) vaccine Gardasil, produces L1 capsid proteins that form VLPs mimicking native virions for robust antibody responses. These systems balance scalability with moderate post-translational capabilities, though their glycosylation differs from mammalian patterns, potentially affecting antigenicity.[67][68]Mammalian cell lines like Chinese hamster ovary (CHO) and human embryonic kidney 293 (HEK293) cells deliver the most authentic post-translational modifications, including complex N-linked glycosylation essential for proteins with intricate structures, ensuring native-like folding and immunogenicity. CHO cells, widely used for biopharmaceuticals, produce the herpes zoster subunit vaccine Shingrix's glycoprotein E (gE) antigen at titers exceeding 1 g/L in serum-free bioreactors, enabling high efficacy against shingles through proper sialylation and stability. HEK293 cells support transient expression for rapid prototyping, as seen in influenza hemagglutinin (HA) subunit production, where they facilitate scalable yields in suspension culture while replicating humanglycosylation for enhanced vaccine potency. However, these systems incur higher costs due to complex media and slower growth, limiting their use to antigens demanding precise modifications.[69][70][71]Insect cell systems employing baculovirus vectors, such as in Sf9 or High Five cells, enable rapid, high-level expression of complex eukaryotic proteins with post-translational modifications closer to mammalian profiles, including glycosylation and phosphorylation, at intermediate costs. The baculovirus expression vector system (BEVS) drives strong promoters for transient production, yielding up to 30 mg/L of influenza HA for the FluBlok vaccine, where the antigen's trimeric structure and sialic acid modifications elicit broad hemagglutination inhibition responses. This platform's versatility supports VLP formation and is advantageous for seasonal vaccines requiring quick adaptation, though potential for hyper-glycosylation necessitates optimization. Following expression in any system, the crude antigen proceeds to purification to achieve pharmaceutical-grade purity.[72][73]Selection of the expression system is guided by antigen complexity: prokaryotic hosts like E. coli suffice for simple, unmodified peptides, while eukaryotic systems—yeast for moderately complex antigens like HBV HBsAg, insect cells for viral glycoproteins like influenzaHA, and mammalian cells for highly demanding ones like shinglesgE—are chosen to ensure functional, immunogenic products without compromising yield or cost-effectiveness.[6][74]
Purification and Formulation
Following the production of subunit antigens through recombinant expression systems, purification involves downstream processing to isolate the target antigen from host cell components, ensuring high purity and removal of potential contaminants. Common techniques include affinity chromatography, which exploits specific interactions between the antigen and ligands such as monoclonal antibodies or tags (e.g., His-tag) to achieve purities often exceeding 95%, ion-exchange chromatography for separating based on charge differences, and ultrafiltration for concentration and size-based separation. These methods are sequentially applied to minimize impurities like host cell proteins (HCPs) and DNA, with regulatory guidelines requiring host cell DNA levels below 10 ng per dose to mitigate risks of oncogenicity or immunogenicity. [75][76][77]Once purified, antigens are formulated by integrating adjuvants to enhance immunogenicity, as subunit vaccines often require immune stimulation for robust responses. Aluminum-based adjuvants like alum promote Th2-biased responses by facilitating antigen uptake and sustained release, while advanced systems such as AS01—a liposome formulation containing monophosphoryl lipid A (MPL) and Quillaja saponaria 21 (QS-21)—induce a balanced Th1/Th2 profile and enable dose-sparing, reducing antigen requirements by up to 10-fold in some cases. Adjuvant selection depends on the target pathogen; for instance, alum is widely used in hepatitis B vaccines for antibody production, whereas AS01 improves T-cell responses in shingles vaccines. [78][20][79]Formulation further includes buffering to maintain pH stability (typically 6.5–7.5 for protein antigens), addition of stabilizers like sugars (e.g., sucrose) or surfactants to prevent aggregation, and lyophilization (freeze-drying) for long-term storage, which can extend shelf-life to 2–3 years at 2–8°C by removing water and preserving antigen structure. For multi-valent vaccines, such as those targeting multiple HPV types, purified antigens are mixed under controlled conditions to ensure compatibility and uniform potency without cross-interference. These steps prioritize thermal stability, with lyophilized forms often showing less than 10% potency loss after 6 months at elevated temperatures. [80][81][82]Quality control encompasses rigorous testing to verify product safety and efficacy, including sterility assays via membrane filtration or direct inoculation to confirm absence of viable microorganisms, and potency assessments using enzyme-linked immunosorbent assay (ELISA) to quantify antigen content against reference standards, ensuring levels meet predefined thresholds (e.g., ≥80% of labeled amount). Additional checks for purity, such as SDS-PAGE or HPLC, confirm removal of HCPs below 100 ppm, while endotoxin levels are limited to <5 EU/dose via Limulus amebocyte lysate testing. These assays are validated per ICH guidelines and performed on final lots to support batch release. [83][84][85]Subunit vaccines are predominantly formulated for injectable delivery, typically intramuscular or subcutaneous routes to optimize systemic immunity, with preservatives like thimerosal added for multi-dose vials. Emerging approaches include microneedle patches for painless, self-administered transdermal delivery, which have shown comparable immunogenicity to injections for influenza subunit vaccines by targeting skin-resident antigen-presenting cells. [86][87]
Advantages and Limitations
Advantages
Subunit vaccines offer a superior safety profile compared to live-attenuated vaccines, as they contain no viable pathogen components, eliminating the risk of reversion to virulence or incomplete inactivation during manufacturing or administration.[17] This inherent safety makes them particularly suitable for immunocompromised individuals, who may not tolerate vaccines with live elements due to the potential for adverse reactions.[88]The production of subunit vaccines leverages standardized recombinant DNA methods, enabling large-scale manufacturing that is both consistent and cost-effective relative to traditional vaccine platforms requiring pathogen cultivation.[68] These methods facilitate scalability by utilizing heterologous expression systems, such as bacterial, yeast, or mammalian cells, to produce antigens without the complexities of handling infectious agents.[89]By focusing exclusively on key immunogenic antigens, subunit vaccines elicit targeted immune responses that minimize extraneous reactions to non-essential pathogen components, thereby enhancing specificity and potentially simplifying pathways to regulatory approval through well-characterized, purified formulations.[90][1] This precision reduces the likelihood of off-target immune activation seen in whole-pathogen vaccines.[30]Some subunit vaccines exhibit thermostability, allowing storage at ambient temperatures without significant loss of potency, which streamlines logistics and distribution in low-resource settings where cold-chain infrastructure is limited.[91] For instance, certain recombinant protein formulations maintain efficacy after prolonged exposure to elevated temperatures, addressing a key barrier to vaccine access in developing regions.[92]Ethically, subunit vaccines avoid the need for propagating virulent pathogens during production, thereby reducing biosafety risks associated with high-containment facilities and minimizing environmental concerns related to pathogen handling.[68] This approach supports safer, more accessible vaccine development globally.[93]
Disadvantages
Subunit vaccines generally possess poor inherent immunogenicity, as they consist of isolated antigens that fail to replicate the full pathogen structure necessary for strong immune activation, often requiring adjuvants to boost responses and potentially leading to weaker cellular immunity compared to live-attenuated vaccines.[94][11] This limitation stems from the antigens' inability to mimic natural infection pathways, resulting in predominantly antibody-mediated responses rather than comprehensive T-cell activation.[36]Development of subunit vaccines is complex and resource-intensive, particularly in antigen selection and ensuring proper protein folding, which can lead to misfolded products with reduced efficacy and extend the timeline to licensure to 10-15 years due to iterative testing and optimization.[95][88] These challenges arise from the need to identify immunodominant epitopes that maintain conformational integrity during recombinant expression, often necessitating advanced bioinformatics and trial-and-error approaches.[96]Their limited breadth of protection poses another drawback, as subunit vaccines targeting specific antigens may not effectively counter antigenic drift in evolving pathogens, such as influenza viruses, where mismatches between vaccine strains and circulating variants reduce effectiveness.[97] Unlike whole-pathogen vaccines, they typically do not induce mucosal immunity, further restricting their utility against respiratory or enteric infections.[98]Production costs for subunit vaccines are high, primarily due to the reliance on recombinant systems like mammalian or insect cell cultures, which demand expensive media, bioreactors, and downstream purification processes compared to simpler inactivated vaccine manufacturing.[6] Yields can vary widely (e.g., 1-10 g/L in CHO cells), but scalability issues and quality control for complex proteins elevate overall expenses.[6]Regulatory requirements add further hurdles, mandating rigorous testing for purity, potency, and consistency to ensure the absence of contaminants and reliable immunogenicity, which prolongs approval and increases development burdens.[77][99] These standards, enforced by agencies like the FDA, emphasize lot-release assays for biologic activity, often requiring animal models or advanced analytics to verify each batch's performance.[83]
Safety Profile
Adverse Effects
Subunit vaccines are generally well-tolerated, with the most common adverse effects being mild and transient local reactions at the injection site, including pain, soreness, redness, and swelling, alongside systemic symptoms such as fatigue, headache, low-grade fever, and myalgia. These effects typically onset within hours to days post-vaccination and resolve spontaneously within 1-2 days without intervention. For instance, in clinical and post-marketing data for the recombinant hepatitis B vaccine, injection site pain affects 3% to 29% of recipients, while erythema and swelling occur in about 3%, and systemic symptoms like fever are reported in 1% to 6%. Similar profiles are seen with the human papillomavirus (HPV) vaccine, where pain, redness, or swelling at the site affects up to 90% of recipients, often accompanied by headache, fatigue, or nausea in 10-20% of cases.[34][100]Adjuvant use in subunit vaccines can amplify local reactogenicity, particularly through enhanced inflammation. The AS04 adjuvant, a combination of monophosphoryl lipid A (MPL) and aluminum hydroxide employed in certain HPV vaccines like Cervarix, is associated with more prolonged injection site pain and swelling compared to aluminum-only adjuvanted formulations, though these remain mild and self-limiting.[101] Aluminum-based adjuvants in protein subunit vaccines, such as those in hepatitis B formulations, may also contribute to transient arthralgia, myalgia, or headache in a subset of recipients, reflecting their role in stimulating innate immune responses. In contrast, non-adjuvanted polysaccharide subunit vaccines exhibit lower rates of these local inflammatory responses due to their reduced immunostimulatory potency.[102]Rare serious adverse events with subunit vaccines include hypersensitivity reactions such as anaphylaxis, occurring at an estimated rate of less than 1 per million doses across licensed products. For the hepatitis B vaccine, the incidence of anaphylaxis is approximately 1.1 per million doses, with a likely causal association in individuals sensitive to yeast-derived components. Post-licensure surveillance systems like the Vaccine Adverse Event Reporting System (VAERS) in the United States and global pharmacovigilance networks monitor these events, confirming that mild reactions occur in 10-20% of doses for vaccines like hepatitis B, while severe events remain exceedingly uncommon. Importantly, extensive reviews have found no established causal link between subunit vaccines and autoimmune disorders or Guillain-Barré syndrome in the majority of cases, with observed incidences aligning with background population rates.[103][104][105]
Contraindications and Precautions
Subunit vaccines, being non-live, have few absolute contraindications, primarily limited to individuals with a history of severe allergic reaction, such as anaphylaxis, to a vaccine component or following a previous dose of the same vaccine.[106] For yeast-derived subunit vaccines like the hepatitis B vaccine and certain HPV vaccines (e.g., Gardasil and Gardasil 9), a history of hypersensitivity to yeast is also an absolute contraindication.[34][107]Precautions warrant caution but do not preclude vaccination unless risks outweigh benefits. These include moderate or severe acute illness, with or without fever, where vaccination should be deferred until recovery to avoid confusing vaccine-related symptoms with those of the illness.[106]Pregnancy is not an absolute contraindication for most subunit vaccines; available data, including animal studies and human experience, show no evidence of risk. For example, the hepatitis B vaccine is considered safe during pregnancy with no adverse fetal outcomes reported in available studies.[108] However, HPV vaccines are not recommended during pregnancy due to insufficient safety data, and any inadvertently administered doses should be reported to vaccine manufacturers for monitoring.[107]In individuals with immunosuppression, subunit vaccines are generally safe to administer as they pose no risk of vaccine-derived infection, unlike live vaccines.[109] Nonetheless, immunocompromised patients may exhibit reduced immune responses, potentially necessitating higher doses, additional boosters, or serologic monitoring to confirm immunogenicity, as seen with hepatitis B vaccination in those with HIV or undergoing chemotherapy.[109]Drug interactions with subunit vaccines are minimal, and no specific spacing is required when co-administering with live vaccines, though general best practices recommend vaccinating during stable health periods.[106] For special populations, conjugate subunit vaccines, such as those for Haemophilus influenzae type b (Hib), are safe and routinely recommended for infants starting at 2 months of age.[106] In the elderly, subunit vaccines like the adjuvanted recombinant zoster vaccine (Shingrix) require precautions for those with acute illness but are otherwise indicated, often with booster considerations to maintain efficacy.[106]The Centers for Disease Control and Prevention (CDC) and World Health Organization (WHO) emphasize pre-vaccination screening for allergies and acute conditions, with providers trained to recognize and manage rare anaphylactic risks through immediate access to epinephrine.[106][53] WHO guidelines further stress that mild illnesses, such as low-grade fever or diarrhea, do not constitute contraindications for subunit vaccines, promoting broad access while prioritizing safety in vulnerable groups.[110]
Licensed Examples
Hepatitis B Vaccine
The recombinant hepatitis B subunit vaccine targets the hepatitis B surface antigen (HBsAg), a key viral protein expressed using recombinant DNA technology in yeast cells such as Saccharomyces cerevisiae. This approach produces noninfectious HBsAg particles that mimic the natural virus structure to elicit an immune response without the risks associated with live or whole-virus vaccines. The first such vaccine, Recombivax HB, was licensed by the U.S. Food and Drug Administration in 1986, marking a pivotal advancement in subunit vaccine technology by shifting from plasma-derived antigens to safer, scalable recombinant production.[111][112]The recommended immunization schedule for the hepatitis B subunit vaccine consists of three intramuscular doses at 0, 1, and 6 months, which induces protective anti-HBs antibodies in greater than 95% of healthy adults, conferring long-term seroprotection against infection. This regimen is highly effective in preventing both acute and chronic hepatitis B virus (HBV) disease, with seroprotection rates remaining above 90% for at least 20-30 years in most recipients. Universal infant immunization, initiated globally following World Health Organization recommendations, has dramatically reduced chronic HBV infections by approximately 90% in vaccinated birth cohorts, averting millions of cases of liver cirrhosis and hepatocellular carcinoma.[113][114]Available formulations include pediatric doses (typically 5 mcg HBsAg per 0.5 mL for infants and children) and adult doses (10 mcg HBsAg per 1 mL), allowing age-appropriate administration while maintaining equivalent immunogenicity. Combination vaccines, such as Twinrix, integrate the hepatitis B subunit antigen with inactivated hepatitis A virus components for dual protection in a single 3- or 4-dose series, particularly useful for travelers and high-risk adults. As of 2025, no major formulation changes have occurred for the core recombinant HBsAg vaccines, though vaccination strategies continue to evolve for high-risk groups, including expanded universal adult recommendations for ages 19-59 years and targeted catch-up for those 60 and older with risk factors like chronic liver disease.[111][115][116]
Human Papillomavirus (HPV) Vaccine
The human papillomavirus (HPV) vaccines represent a key application of subunit vaccine technology, utilizing virus-like particles (VLPs) derived from the L1 major capsid protein to mimic the structure of the viral capsid without incorporating viral DNA, thereby inducing a robust immune response against oncogenic and low-risk HPV types.[107] These VLPs self-assemble from recombinant L1 proteins expressed in systems such as yeast or insect cells, providing non-infectious antigens that target the primary causes of cervical and other HPV-related cancers.[117]Several variants of HPV VLP vaccines have been developed, differing in the number of HPV types covered and adjuvant composition. The bivalent vaccine, Cervarix, targets HPV types 16 and 18—responsible for approximately 70% of cervical cancers—and incorporates the AS04 adjuvant, which combines aluminum hydroxide with monophosphoryl lipid A to enhance T-cell mediated immunity and duration of protection.[118] In contrast, the quadrivalent formulation, originally licensed as Gardasil in 2006 by the U.S. Food and Drug Administration (FDA), protects against HPV types 6, 11, 16, and 18, addressing both high-risk oncogenic types and those causing genital warts.[119] An updated nonavalent version, Gardasil 9, approved by the FDA in December 2014, extends coverage to nine types (6, 11, 16, 18, 31, 33, 45, 52, and 58), potentially preventing up to 90% of HPV-related cervical cancers.[120]Clinical trials and real-world evidence demonstrate high efficacy of these vaccines, with protection exceeding 90% against persistent infection, precancerous lesions (such as cervical intraepithelial neoplasia grades 2 and 3), and vaccine-type cervical cancers when administered prior to HPV exposure.[121] For instance, the quadrivalent vaccine showed nearly 100% efficacy against HPV 16/18-related high-grade cervical lesions in phase III trials, while the nonavalent version achieved 96% efficacy against high-grade cervical disease caused by additional targeted types in HPV-naïve populations.[122] Bivalent Cervarix exhibited 92.9% efficacy against HPV 16/18-associated cervical intraepithelial neoplasia grade 2 or higher in per-protocol analyses.[123]The recommended vaccination schedule targets adolescents to maximize pre-exposure immunity, with a two-dose series (administered 6–12 months apart) for individuals initiating vaccination between ages 9 and 14, and a three-dose series (at 0, 1–2, and 6 months) for those starting at age 15 or older.[124] This approach ensures strong antibody responses, with seroconversion rates approaching 100% in clinical studies across variants.[121]By 2025, widespread HPV vaccination has significantly reduced the prevalence of vaccine-targeted HPV types and related disease burden in vaccinated populations, with U.S. data showing an 80% decrease in higher-grade cervical precancer incidence among women aged 20–24 screened during 2008–2022, reflecting herd immunity effects even among unvaccinated individuals.[125] Globally, similar trends have emerged in high-uptake regions, contributing to progress toward cervical cancer elimination goals set by the World Health Organization.[126]
Influenza Vaccine
Recombinant protein subunit vaccines represent a key advancement in influenza immunization, utilizing purified hemagglutinin (HA) proteins produced through recombinant DNA technology in insect cell systems, such as those used in Flublok. These vaccines target the HA surface glycoprotein, which facilitates viral attachment to host cells, and are formulated without the use of eggs, thereby avoiding potential adaptation issues or allergic reactions associated with traditional egg-based production.[127] Unlike whole-virus or split-virus formulations, subunit versions focus solely on HA to elicit a targeted immune response, with Flublok containing three times the standard HA dose per strain for enhanced immunogenicity.[128]The primary challenge in developing these vaccines lies in the annual antigenic drift of influenza viruses, necessitating biannual strain selection by the World Health Organization (WHO) based on global surveillance data to predict circulating variants. For the 2025-2026 northern hemisphere season, WHO recommends trivalent formulations incorporating an A(H1N1)pdm09-like virus, an A(H3N2) virus, and a B/Victoria lineage virus, reflecting the obsolescence of the B/Yamagata lineage and a shift away from quadrivalent vaccines. This process ensures vaccines match dominant strains, though mismatches can occur due to post-selection viral evolution.[129]Efficacy against culture-confirmed influenza in matched strains typically ranges from 40% to 60% in adults, with reduced protection—around 45%—observed in older adults aged 65 and above due to immunosenescence and lower antibody responses.[130] In the elderly, recombinant HA vaccines like Flublok demonstrate relative efficacy improvements of up to 30% compared to standard inactivated vaccines in those over 50.[131]The transition to recombinant subunit influenza vaccines accelerated in 2013 with the FDA approval of Flublok, marking the first fully recombinant option and providing a safer alternative for individuals with severe egg allergies by eliminating egg-derived components entirely. Clinical trials preceding approval showed 44.6% efficacy against influenza in adults, supporting its role in seasonal prophylaxis. By 2025, these vaccines remain a cornerstone of influenza prevention strategies, with ongoing efforts toward universal influenza vaccines—such as those targeting conserved viral epitopes—still in clinical development, while recombinant HA-based subunit formulations continue as the standard for annual updates.[132]
Other Notable Vaccines
Shingrix, a recombinant subunit vaccine targeting herpes zoster (shingles), consists of the glycoprotein E antigen derived from varicella-zoster virus combined with the AS01B adjuvant system.[133] Licensed by the U.S. Food and Drug Administration (FDA) in October 2017 for adults aged 50 years and older, it demonstrated vaccine efficacy of 97.2% against herpes zoster in individuals aged 50-59 years and 97.4% in those aged 60 years and older in pivotal phase 3 trials involving over 38,000 participants.[133] Additionally, it showed 89.3% efficacy against postherpetic neuralgia, a common complication of shingles, highlighting its role in reducing severe outcomes in older adults.[133] Real-world studies have confirmed sustained effectiveness, with vaccine effectiveness reaching 51.8% to 57.7% against herpes zoster in previously vaccinated adults as of 2025.[134]Novavax's COVID-19 vaccine, known as Nuvaxovid or NVX-CoV2373, is a protein nanoparticle subunit vaccine featuring the SARS-CoV-2 spike protein stabilized in prefusion conformation, adjuvanted with Matrix-M to enhance immune responses.[135] Initially authorized for emergency use by the FDA in 2021 and fully approved in May 2025 for the 2025-2026 formula targeting updated variants, it exhibited 90.4% efficacy against symptomatic COVID-19 caused by the original strain in a phase 3 trial of approximately 30,000 adults.[136][137] By 2025, formulations have been expanded to address variants like Omicron sublineages, with booster doses showing robust humoral responses and up to 80% effectiveness against hospitalization in real-world settings.[138] This vaccine represents a non-mRNA alternative, particularly valued for its stability and compatibility with existing cold-chain infrastructure.[139]Among bacterial subunit vaccines, Prevnar 13 (PCV13) is a 13-valent pneumococcal conjugate vaccine comprising purified capsular polysaccharides from Streptococcus pneumoniae serotypes conjugated to the CRM197 carrier protein, enabling T-cell dependent immune responses.[140] Licensed by the FDA in 2010 for children and extended to adults in 2011, it demonstrated approximately 90%-97% effectiveness against vaccine-type invasive pneumococcal disease in infants, based on post-licensure studies, and 45-46% efficacy against vaccine-type pneumococcal pneumonia in adults aged 65 and older in clinical studies.[141][50] Similarly, Menveo is a quadrivalent meningococcal conjugate vaccine targeting Neisseria meningitidis serogroups A, C, Y, and W-135, with polysaccharides linked to CRM197.[142] Approved by the FDA in 2010 for individuals aged 2 months through 55 years, it induced seroprotective antibody responses in 89-96% of infants after a two-dose series, correlating with high effectiveness against invasive meningococcal disease.[143][142]A notable advancement in respiratory vaccines is Arexvy, a subunit vaccine for respiratory syncytial virus (RSV) prevention in older adults, utilizing the prefusion-stabilized RSV F glycoprotein adjuvanted with AS01E.[144] Licensed by the FDA in May 2023 for individuals aged 60 years and older, it showed 82.6% efficacy against RSV-associated lower respiratory tract disease in the first season in a phase 3 trial of over 25,000 participants, with sustained protection of 62.9% over three seasons.[144][145] By 2025, it has been integrated into routine immunization recommendations for at-risk elderly populations, reducing hospitalizations by 60-65%.[146]Subunit vaccines are increasingly incorporated into combination formulations to simplify immunization schedules and enhance coverage, as seen in multivalent pneumococcal and meningococcal products that target multiple serotypes simultaneously.[147] Therapeutic applications are also expanding, with subunit designs explored for chronic conditions like cancer and infectious disease management, projecting a market growth to $86.8 billion by 2035 driven by adjuvanted and nanoparticle innovations.[148]
Historical Development
Early Discoveries
The development of subunit vaccines traces its roots to early 20th-century efforts to create safer alternatives to whole-pathogen vaccines by isolating specific protective components. In the 1920s, researchers developed toxoid vaccines, such as the diphtheria toxoid, by inactivating bacterial toxins with formalin to retain immunogenicity without toxicity; this approach, pioneered by Gaston Ramon and colleagues, marked an initial form of subunit vaccination and was widely used by the mid-1930s.[149] Similarly, in the 1930s, work by Oswald Avery and others at the Rockefeller Institute isolated pneumococcal capsular polysaccharides, demonstrating their ability to induce type-specific antibodies, which laid the groundwork for polysaccharide-based subunit vaccines tested in clinical trials during that decade and the 1940s.[150][151]The 1960s and 1970s saw pivotal advances with the emergence of recombinant DNA technology, enabling precise production of viral and bacterial antigens. Key milestones included the creation of the first recombinant DNA molecules in 1972 by Paul Berg and the development of cloning techniques by Stanley Cohen and Herbert Boyer in 1973, which opened pathways for genetically engineered subunit vaccines.[152] Concurrently, in the 1970s, the purification of hepatitis B surface antigen (HBsAg) from plasma of asymptomatic carriers provided the basis for the first plasma-derived subunit vaccine against hepatitis B, licensed in 1981 under the leadership of Maurice Hilleman at Merck & Co.[35][153] This vaccine was later transitioned to a recombinant form produced in yeast cells, approved in 1986, to eliminate risks associated with human plasma.[35]In the 1980s, early trials of conjugate subunit vaccines advanced the field, particularly for Haemophilus influenzae type b (Hib), where polysaccharides were linked to carrier proteins like diphtheria toxoid to enhance immunogenicity in infants; the first such vaccine, PRP-D, underwent clinical testing starting in 1986 and was licensed in 1987.[154] These developments highlighted the critical need for adjuvants, recognized since the 1920s with alum's use in toxoid and polysaccharide formulations to boost weak immune responses typical of purified subunit antigens.[155]The push for subunit vaccines during this era was largely driven by the hepatitis B epidemic, which caused significant global morbidity and mortality in the 1970s, and the subsequent AIDS crisis in the 1980s, which underscored the dangers of plasma-derived products due to HIV transmission risks and accelerated the adoption of recombinant technologies.[156][157]
Key Milestones and Evolution
The advent of recombinant DNA technology in the 1980s revolutionized subunit vaccine development, enabling the production of safer antigens without live or whole pathogens. In July 1986, the U.S. Food and Drug Administration (FDA) approved Recombivax HB, the first recombinant hepatitis B surface antigen vaccine developed by Merck using yeast cells, marking a pivotal shift from plasma-derived versions and addressing global concerns over blood-borne transmission. This was followed by advancements in conjugate vaccines for bacterial diseases; in December 1987, the FDA licensed ProHIBiT, the first Haemophilus influenzae type b (Hib) conjugate vaccine, which linked polysaccharideantigens to carrier proteins for enhanced immunogenicity in children under two years, dramatically reducing invasive Hib disease incidence. The 1990s saw further progress with virus-like particle (VLP) technology, culminating in early trials for human papillomavirus (HPV) vaccines, though full approvals came later.The 2000s brought innovations in adjuvants and broader applications. In 2007, the European Medicines Agency approved Cervarix, the first HPV vaccine using the AS04 adjuvant system (monophosphoryl lipid A plus aluminum hydroxide), which enhanced T-cell responses and provided longer-lasting protection against HPV-16/18 compared to aluminum-only formulations. The FDA followed with approval in 2009.[101] By 2013, recombinant technology extended to influenza with the FDA's approval of Flublok, a trivalent subunit vaccine produced in insect cells expressing hemagglutinin proteins, offering an egg-free alternative that improved production scalability during pandemics.The 2010s and 2020s highlighted subunit vaccines' resilience amid emerging threats. In October 2017, the FDA approved Shingrix, a recombinant zoster vaccine using glycoprotein E antigen with the AS01B adjuvant, demonstrating over 90% efficacy in preventing herpes zoster in adults aged 50 and older, surpassing live-attenuated options.[158] The COVID-19 pandemic accelerated subunit vaccine deployment; in July 2022, the FDA granted Emergency Use Authorization to Novavax's protein-based COVID-19 vaccine, utilizing nanoparticle-displayed spike protein with Matrix-M adjuvant, providing a non-mRNA option that maintained relevance despite mRNA vaccines' rapid rollout. Subunit approaches persisted due to their established safety profile and adaptability.Regulatory frameworks evolved to support global access. The FDA's fast-track designation, formalized under the 1997 FDA Modernization Act, expedited reviews for pandemic vaccines like Novavax's, allowing rolling submissions and priority evaluation to address unmet needs.[159] Similarly, the World Health Organization's prequalification program, expanded in the 2010s, vetted subunit vaccines such as recombinant hepatitis B and Hib conjugates for UN procurement, ensuring quality and affordability in low-income countries.Subunit vaccines have evolved from single-antigen formulations, like early hepatitis B, to multivalent designs targeting multiple strains or pathogens for comprehensive protection. Examples include the progression of HPV vaccines from bivalent (2007) to 9-valent Gardasil 9 (2014), covering additional oncogenic types.[160] By 2025, artificial intelligence integration in antigen design, using machine learning for epitope prediction and protein optimization, has accelerated development of next-generation multivalent subunit vaccines, reducing timelines from years to months.[161]
Research Directions
Current Challenges
One major challenge in subunit vaccine development is addressing immunogenicity gaps, particularly against rapidly evolving viral variants. The emergence of SARS-CoV-2 variants such as Omicron, characterized by over 30 mutations in the spike protein including the receptor-binding domain, has significantly reduced the neutralizing antibody efficacy of first-generation subunit vaccines targeting the receptor-binding domain, with significant reductions, often 4- to 30-fold compared to the original strain.[162] This immune escape, driven by the virus's high mutation rate of 0.8–2.38 × 10⁻³ nucleotide substitutions per site per year, complicates the design of broadly protective subunit vaccines, often requiring multivalent or mosaic formulations to elicit cross-reactive immunity.[163] Similarly, antigenic variability in other pathogens like influenza necessitates universal vaccine strategies, yet current subunit approaches struggle to provide durable protection across diverse strains without frequent reformulation.[163]Access and equity remain critical barriers, especially in low- and middle-income countries (LMICs), where high production costs and supply chain dependencies hinder widespread deployment. Despite the relative thermostability of subunit vaccines compared to mRNA platforms, cold-chain requirements for storage and transport exacerbate logistical challenges in resource-limited settings, contributing to uneven distribution and lower vaccination coverage.[164] The disproportionate disease burden in LMICs amplifies these inequities, as technical, manufacturing, and funding risks limit affordable access, with global vaccine importers like ASEAN nations relying heavily on external supplies.[164][165]Regulatory delays pose another hurdle, particularly in harmonizing global standards for novel adjuvants essential to enhancing subunit vaccine potency. New adjuvants must undergo rigorous evaluation for immunogenicity, toxicity, and duration of immune response, often facing scrutiny over insufficient manufacturing data or potential excessive immune activation, which prolongs approval timelines across jurisdictions.[166] This fragmented regulatory landscape, including varying requirements for safety assessments, impedes the rapid integration of innovative adjuvant systems into subunit platforms.[167]Lessons from the COVID-19 pandemic highlight tensions between development speed and safety, alongside persistent supply chain vulnerabilities. Subunit vaccines like Novavax experienced delays due to manufacturing scale-up issues and clinical trial hurdles, underscoring the need for balanced risk assessment to avoid compromising efficacy while accelerating responses.[168]Supply chain disruptions revealed dependencies on global infrastructure, with bottlenecks in raw materials and distribution amplifying inequities during crises.[169]Data gaps persist regarding long-term efficacy in diverse populations, with post-2020 surveillance revealing insufficient evidence on sustained protection across age groups, ethnicities, and comorbidities. Most trials focus on short-term immunogenicity in healthy adults, leaving uncertainties about waning immunity and real-world effectiveness in vulnerable cohorts like the elderly or immunocompromised.[170] Enhanced post-licensure monitoring is needed to address these voids and inform booster strategies.[170]
Emerging Technologies and Future Prospects
Advancements in adjuvant technology are enhancing the efficacy of subunit vaccines through next-generation immunostimulators and integrated delivery systems. Toll-like receptor (TLR) agonists, such as CpG oligodeoxynucleotides, activate TLR9 on antigen-presenting cells to promote a Th1-biased immune response, which is crucial for eliciting robust cellular immunity against intracellular pathogens in subunit formulations.[171] These adjuvants have demonstrated improved antibody titers and T-cell responses in preclinical models of viral subunit vaccines, addressing limitations in traditional alum-based systems that favor Th2 responses.[172] Self-adjuvanting nanoparticles further innovate by combining antigen presentation with built-in adjuvant activity, such as lipid or polymeric nanoparticles incorporating TLR ligands, which enhance antigen uptake and cross-presentation without requiring separate adjuvant components.[173] Clinical trials of these systems, including saponin-based Matrix-M™ for subunit antigens, have shown up to 10-fold increases in neutralizing antibody levels compared to non-adjuvanted controls.[174]Delivery innovations are expanding subunit vaccine accessibility and potency beyond injectable formats. mRNA-encoded subunit vaccines enable transient in vivo expression of specific antigens, leveraging mRNA's rapid manufacturability while maintaining the targeted immunogenicity of protein subunits.[175] Viral vectors, such as modified adenoviruses, facilitate the expression of subunit antigens like viral spikes, offering sustained antigen presentation and improved mucosal immunity in respiratory pathogen models.[175] Plant-based oral and edible vaccines represent a needle-free alternative, where subunit antigens are produced in transgenic plants (e.g., tobacco or rice) for ingestion, inducing systemic and mucosal responses; examples include the HIV-1 Tat protein expressed in tomatoes, which elicited immune responses including mucosal IgA in animal studies.[176]Computational design tools are accelerating subunit vaccine development by predicting optimal immunogens. Artificial intelligence and machine learning algorithms, such as deep learning models for epitope mapping, identify conserved B-cell and T-cell epitopes with high accuracy, reducing experimental screening needs; for instance, tools like DeepVac have designed multi-epitope constructs for emerging viruses with predicted immunogenicity scores exceeding 90%.[177] Structure-based vaccinology has been transformed by AlphaFold, which by 2025 enables precise prediction of antigen structures and epitope conformations, facilitating the rational design of stable subunit variants; AlphaFold3's integration of small-molecule interactions has aided in engineering coronavirus spike trimers for broader cross-reactivity.[178] These tools have shortened design timelines from years to months in recent subunit vaccine pipelines.[179]Future prospects for subunit vaccines include universal formulations targeting conserved epitopes to combat antigenic drift in viruses like influenza and coronaviruses. For influenza, recombinant protein subunits focusing on the hemagglutinin stem domain, such as COBRA-based constructs or ferritin nanoparticles, have demonstrated heterosubtypic protection in preclinical studies and early-phase trials (phase 1 as of 2025) in ferrets against diverse strains.[180] Similar approaches for coronaviruses emphasize nucleocapsid or receptor-binding domain subunits to elicit pan-sarbecovirus immunity, with preclinical data showing reduced viral loads in challenge models.[181] Therapeutic applications are promising, particularly peptide-based subunit vaccines for HIV, which target conserved Gag and Env epitopes to boost CD8+ T-cell responses in chronic infection, and for cancer, where neoantigen peptides personalized via AI prediction have induced tumor regression in phase 1 trials.[182]The COVID-19 pandemic has profoundly influenced subunit vaccine evolution, accelerating protein-based platforms through streamlined regulatory pathways and manufacturing scales. Novavax's adjuvanted spike protein subunit vaccine, whose original formulation demonstrated approximately 90% efficacy against symptomatic infection in clinical trials, with updated versions authorized for the 2024-2025 season based on immunogenicity against current variants, exemplifies this speedup.[183]Hybrid strategies combining subunit antigens with mRNA elements are in clinical trials, such as co-formulated protein-mRNA boosters for enhanced durability, with phase 2 data indicating synergistic humoral and cellular responses against variants.[181] These developments position subunit vaccines as versatile backbones for next-generation prophylactics and therapeutics.