Vaccines – The Original Antigenic Sin
Abstract
Modern vaccines are widely credited with reducing infectious disease mortality, yet their long-term effects on immune function remain insufficiently examined. At the centre of this debate lies the polarisation of immune memory — particularly the skewing of T helper cell responses toward type 1 (Th1), type 2 (Th2), or type 17 (Th17) pathways. These profiles are not interchangeable, and once established, they define the trajectory of all future responses to that same antigen. This paper explores how aluminium-adjuvanted and mRNA vaccines increasingly imprint Th2/Th17-dominant immunity, in contrast to the durable, Th1-driven responses seen in historical live-virus vaccines like smallpox, and all wild viral infections.
Through a mechanistic analysis of adjuvant signalling, antigen persistence, and cytokine biasing, we show that vaccine-induced outcomes — including tolerance, IgG4 switching, persistent inflammation, and even antibody-dependent enhancement (ADE) — are not anomalies, but expected consequences of immune skewing by vaccine presentation.
The fundamental flaw in modern vaccine design is that it stimulates the wrong branch of the immune system for viral defence. Th2 responses are effective at dealing with extracellular parasites and allergens, but not with viruses that replicate inside human cells. It’s like sending the army to fight a naval war — the army may make some impact on land, but most of the battle takes place at sea. Worse still, the navy is disabled entirely. Your immune system is tricked into deploying the wrong force while the right one stands down. You’re left in a constant battle — but never able to win the war.
As the battle drags on, the immune system becomes exhausted. Its only remaining strategy is to try to coexist with the virus. Surveillance is suspended, and the virus is allowed to travel freely throughout the body, establishing footholds — like viral ghettos — where it can continue to replicate and cause tissue damage. These hidden, unresolved infections often go undetected but drive chronic inflammation and degeneration. Over time, this sets the stage for serious pathology — including cancer.
This is the core failure of vaccine-induced immune skewing: it trains the body to fight infections in the wrong way, and then locks that instruction into memory.
No vaccine using the current design paradigm — including aluminium-adjuvanted or mRNA-based formulations — can produce true antiviral immunity. Worse still, these vaccines retrain the immune system to respond incorrectly in future infections, committing immune memory to the Th2/Th17 profile when a Th1 response is required.
By contrast, the smallpox vaccine succeeded not because it reflects modern vaccine technology, but because it diverged from it. It used a live, replicating virus with no adjuvant, mimicking natural infection and producing robust Th1-driven, sterilising immunity. It should be recognised as the exception that proves the rule — not as a standard that modern vaccines can claim to emulate.
This paper argues that immunological misprogramming through inappropriate vaccine design undermines long-term health and immune resilience. It calls for a complete re-evaluation of vaccine metrics, shifting from short-term antibody titres toward durable, balanced immune education.
Introduction
The immune system is a highly adaptive learning system. It does not simply react; it remembers, categorises, and commits. Once a particular “learning environment” is established during a primary encounter with a pathogen or vaccine, that imprint determines how the body will respond to future exposures. This process, known as immune imprinting or functional polarisation, is deeply influenced by which T helper (Th) pathway is engaged at the start: Th1, Th2, or Th17 (McCarthy et al., 2021; Bancroft et al., 2016; Crotty, 2011).
What many clinicians and the public overlook is that this immune commitment is shaped at the very first point of contact, by the nature of the cells that detect the threat. If antigen-presenting cells like dendritic cells and macrophages dominate the frontline and are activated in a context of intracellular danger, they will release interleukin-12 (IL-12) and type I interferons, pushing naïve T cells toward a Th1 phenotype, ideal for clearing viruses, intracellular bacteria, and promoting cytotoxic immunity. This is the classic antiviral pathway (Seder, Darrah, and Roederer, 2008; Trinchieri, 2003).
By contrast, if basophils, mast cells, or eosinophils are the first to detect the antigen, especially in the absence of strong intracellular danger signals, they release IL-4 and IL-33, skewing the response toward Th2, which promotes antibody production, tissue repair, and, importantly, immune tolerance (Sokol et al., 2008; Perrigoue et al., 2009). Chronic antigen exposure, especially via non-natural routes (e.g., intramuscular injection with aluminium adjuvant), further entrenches these Th2 or Th17 profiles (Marrack, McKee, and Munks, 2009; Reed, Orr, and Fox, 2013).
This paper examines how different vaccine designs, particularly those using aluminium salts or encoding persistent mRNA antigens, subvert the natural antigen recognition sequence, bypassing mucosal barriers and favouring a non-cytotoxic immune imprint. We contrast this with historical success cases, such as the smallpox vaccine, which elicited a strong Th1 response via live dermal inoculation and viral replication (Fenner et al., 1987; Sanchez-Sampedro et al., 2015).
Understanding how these first cellular responders and their cytokine environments shape long-term memory is crucial for evaluating modern vaccine performance. When we force a Th2 or Th17 response in contexts where Th1 is required, the result will not be full protection but prolonged antigen presence, tolerance, and chronic inflammation (Shi et al., 2009; Katzman et al., 2011; van der Burg et al., 2021).
Understanding Th1, Th2, and Th17 Pathways
The adaptive immune system relies on T helper (Th) cell polarisation to mount appropriate responses to different classes of pathogens. Upon antigen presentation by dendritic cells, naive CD4⁺ T cells differentiate into one of several effector subsets, predominantly Th1, Th2, or Th17, depending on the cytokine environment, co-stimulatory signals, and the nature of the antigen (Zhu, and Paul, 2008).
- Th1 responses are driven by IL-12 and characterised by IFN-γ production. These responses activate cytotoxic T lymphocytes (CTLs) and macrophages to destroy intracellular pathogens, such as viruses and certain bacteria (Swain, McKinstry, and Strutt 2012; Szabo et al., 2003). Th1 cells also facilitate the development of long-lasting memory with the capacity for sterilising immunity, the complete elimination of a pathogen before it can replicate or cause symptoms (Seder, Darrah, and Roederer, 2008).
- Th2 responses, in contrast, are initiated in the presence of IL-4, leading to production of IL-4, IL-5, and IL-13. These pathways promote B cell class switching, particularly toward IgE and IgG4, and recruit eosinophils and mast cells to combat extracellular parasites. Th2 dominance is also central to allergic responses and can promote tolerance or immune exhaustion when persistently activated (Romagnani, 2000; Paul, and Zhu, 2010; Jeannin et al., 1998).
- Th17 responses, promoted by IL-6, IL-1β, and TGF-β, are involved in recruiting neutrophils and maintaining mucosal immunity. They play a crucial role in controlling fungal infections and extracellular bacteria, but excessive Th17 activity is associated with chronic inflammation and autoimmune disease (Harrington et al., 2005; Ouyang, Kolls, and Zheng 2008; Langrish et al., 2005).
Functional Commitment of Memory T Cells
One of the most critical insights in immunology is that memory T cells are lineage-committed. Once a naive T cell differentiates into a Th1, Th2, or Th17 cell, it retains that functional profile for life (Zhu, Yamane, and Paul, 2010a; Zhu, 2018). This means a memory T cell induced during a Th2-skewed response cannot later generate a cytotoxic Th1-type reaction upon re-exposure to the same antigen (Zhu and Paul, 2010b;).
This principle has profound implications for vaccination. A vaccine that primarily elicits a Th2 response, even if it produces neutralising antibodies, will fail to generate the robust cytotoxic memory needed to eliminate intracellular pathogens effectively. Instead, it may predispose the host to non-sterilising immunity, chronic antigen exposure, and repeated boosting cycles, ultimately reinforcing the same immune skew each time (Pulendran and Ahmed, 2011; Bungener et al., 2008)
This immunological imprinting, often called “original antigenic sin” in viral immunology, becomes especially problematic in the context of respiratory viruses, where Th1 cytotoxicity is required for full viral clearance (Kim et al., 1969; Halstead and O’Rourke, 1977; Reynolds et al., 2022a).
Smallpox and the Myth of Sterilising Immunity
The smallpox vaccine stands alone in the historical record as the only vaccine to confer consistent, long-term sterilising immunity at a population level. Unlike modern subunit or mRNA platforms, the smallpox vaccine used a live, replication-competent virus, vaccinia, which is genetically similar to the variola virus but sufficiently attenuated for use in humans. It did not contain aluminium-based adjuvants, nor did it require repeated boosters. The immune response it triggered was robustly Th1-dominant, involving widespread activation of cytotoxic T cells, NK cells, and high levels of IFN-γ and IL-2 — the classical hallmarks of intracellular pathogen defence (Moss, 2013; Demkowicz et al., 1992).
This vaccine induced strong and lasting memory in both CD4⁺ and CD8⁺ T cell compartments, enabling recipients to clear viral infection before replication could occur — the very definition of sterilising immunityand representing the idea of vaccines that we all believe in. Moreover, the replicating nature of the vaccine virus ensured endogenous antigen presentation via MHC class I pathways, a key requirement for effective cytotoxic T lymphocyte (CTL) priming (Tscharke et al., 2005; Miller et al, 2008). No modern vaccine attempts to replicate this model because of safety concerns and manufacturing limitations.
As a result, the smallpox vaccine is often misleadingly cited as a standard of vaccine efficacy. However, its success lies not in its representativeness, but in its exceptional divergence from modern vaccine design. Unlike today’s aluminium-adjuvanted or mRNA-based products, which bypass intracellular processing and focus on antibody production, the smallpox vaccine closely mimicked natural infection. Its use of a live virus without a strong Th2-skewing adjuvant enabled a balanced and robust Th1-mediated immune response, which no modern interventions can replicate (Simon et al., 2014; Kim et al., 2006; Sasaki et al., 2003).
Sterilising immunity — where the immune system prevents infection entirely — is the gold standard in vaccination and should be the only model pursued. Historically, this level of protection has only been reliably achieved using live-attenuated vaccines that closely mimic the behaviour and tissue tropism of the wild virus, enabling the immune system to mount a comprehensive and correctly balanced response (Slifka and Amanna, 2014; Plotkin, 2010). In contrast, inactivated or subunit vaccines fail to achieve sterilising immunity and require adjuvants to boost effectiveness (Cohet et al., 2019). However, adjuvants can distort the natural immune response, resulting in skewed immunity that fails to resolve infection and may promote long-term immune dysregulation (Pulendran and Ahmed, 2011; Reed, Orr, and Fox, 2013). Preserving the integrity of immune system architecture should take precedence over short-term antibody titres, particularly when long-term safety data are lacking (Liu et al., 2005).
Vaccine Pressure and Viral Mutation
Modern vaccines, by contrast, are designed to induce non-sterilising immunity, leaving individuals vulnerable to asymptomatic infection and transmission. The resulting immune memory is Th2-biased — a profile increasingly associated with chronic inflammation, immune tolerance dysfunction, and autoimmunity, especially with early-life or repeated exposures (Brewer et al., 1999; HogenEsch 2002; Lindblad 2004; Aimanianda et al., 2021; Berin and Screffler, 2008; Marrack, McKee, and Munks, 2009).
This partial immune pressure creates ideal conditions for viral evolution. When viruses are exposed to sub-neutralising antibody levels or poorly cytotoxic immune environments, they are pressured to mutate in ways that enhance their ability to evade immunity and persist — a phenomenon known as vaccine pressure (Muñoz-Alía et al., 2017; Geoghegan, and Holmes, 2018).
For example, the measles virus historically maintained an exceptionally stable genome (Langmuir et al, 1962), but after widespread vaccination, mutations began to accumulate in response to immune selection (Hadfield n.d.). Moore (2021) notes that “escape mutations” typically arise under selective pressure from antibodies that restrict but do not eliminate viral replication. Wang et al. (2021) demonstrated that vaccine-resistant mutations such as Y449S in SARS-CoV-2 emerged in direct correlation with rising vaccination rates in Europe and the US, disrupting neutralising antibodies while reducing infectivity, a form of immune evasion rather than viral enhancement (Wang et al., 2021).
Pouwels et al. (2021) further highlighted that waning vaccine-induced immunity, in combination with viral evolution, threatens long-term vaccine efficacy. This underscores the broader immunological risk: when vaccines train the immune system to tolerate rather than eliminate, we don't just fail to prevent disease, we cultivate its adaptation (Pouwels et al., 2021).
The use of non-sterilising vaccines should be avoided at all costs because they drive the target virus to mutate, to become more virulent (Read et al., 2015; Boots, 2015; Miller and Metcalf, 2019; Garcia-Beltran et al., 2021).
Th2-Skewed Immune Memory in Modern Vaccination
Modern vaccine formulations, particularly those using aluminium salts (commonly referred to as “alum”), are designed to elicit strong antibody responses. Alum has been used for nearly a century and remains the most widely used adjuvant in licensed human vaccines. Its mechanism of action is to stimulate the Th2 arm of the adaptive immune system, encouraging B cell differentiation and class switching to immunoglobulin subtypes such as IgG4 and IgE, rather than Th1-associated IgG1 or IgG3 cytotoxic T cell responses (Brewer et al., 1999; Marrack et al., 2009). Notably, this Th2-biased response occurs independently of the classical cytokine drivers IL-4 and IL-13 (Brewer et al., 1999).
While this response is sufficient to provide partial protection against many pathogens, it does not replicate the full immune architecture induced by natural infection or live-attenuated vaccines with no adjuvant. Importantly, Th2-biased immune memory is self-reinforcing: once initial priming is skewed toward Th2, subsequent exposures to the same antigen, whether through booster shots or natural infection, will continue to activate the same pathway (Bancroft et al., 2016; Bielinska et al., 2016; Kidd, 2003). As a result, this immune imprinting establishes a long-term bias, causing the host to respond to related pathogens with a Th2-skewed profile, even when later exposed to signals that would normally promote a Th1 response (Cao et al., 2021).
The preference for Th2 stimulation over Th1 is not benign. Th2-dominant responses are associated with humoral tolerance, chronic inflammation, and the inhibition of cell-mediated immunity, which is essential for clearing intracellular pathogens such as viruses (Bungener et al., 2008). Furthermore, in susceptible individuals, repetitive Th2 stimulation contributes to the development of immune pathologies, including IgE-mediated allergies, eosinophilic disorders, and IgG4-related disease (Finkelman et al., 1988; Umehara et al., 2012a). Immune cells also shift function in response to their chemical environment; for example, mast cells, typically known for their pro-inflammatory roles, can adopt anti-inflammatory and immunosuppressive behaviour in the presence of IgE (Galli, Grimbaldeston, and Tsai 2008).
It is highly improbable that repeated administration of aluminium-adjuvanted vaccines, particularly when started in infancy, would not contribute to a persistent Th2 bias in recipients, and regulatory IgG4 class switching in a portion of that population. It is the natural conclusion of events. Although manifestations may remain subclinical in many recipients, the underlying shift in immune architecture can silently increase vulnerability to immune evasion, chronic disease, and antibody-dependent enhancement (ADE) upon exposure to related pathogens later in life (Guimaraes et al., 2015; Bortolatto et al., 2015; Pillai 2023).
mRNA Vaccines and the Inescapable Shift Toward Th2 Immunity
Despite initial claims that mRNA vaccines elicit a Th1-skewed response, the documented induction of IgG4 class switching and even IgG4-related disease after repeated exposure demonstrates a definitive shift away from Th1 immunity. IgG4 cannot emerge from a sustained Th1-dominant environment; its production requires cytokine signals characteristic of Th2 and regulatory responses, including IL-4, IL-10, and TGF-β (Finkelman et al., 2005; Umehara et al., 2012a). This shift is not theoretical — it is evidenced by peer-reviewed studies documenting elevated IgG4 following mRNA vaccination (Irrgang et al., 2023; Gelderloos et al., 2024), as well as case reports of vaccine-induced IgG4-RD (Aochi, Uehara, and Yamamoto, 2023; Zhang et al, 2024). There is no known immunological pathway in which a persistent Th1 environment produces IgG4. Therefore, the presence of IgG4 alone is sufficient to prove that these vaccines have switched to a Th2-dominant profile. While population-level studies are still emerging, the well-established immunological pathways leading to IgG4 dominance and tolerance make this outcome a biologically predictable consequence of repeated mRNA vaccination (Valk et al., 2024; Irrgang et al., 2023).
IgG4 Class Switching and Immune Tolerance
When the immune system encounters persistent antigen exposure, whether through chronic infection, environmental allergens, or vaccination, it may adapt by dampening inflammatory responses to prevent tissue damage. One hallmark of this adaptive response is class switching to IgG4, a unique antibody subclass associated with immune tolerance, non-cytolytic behaviour, and suppressed effector functions. Unlike IgG1 or IgG3, IgG4 does not efficiently fix complement or trigger antibody-dependent cellular cytotoxicity (ADCC) (Oskam et al., 2023; Rispens and Huijbers, 2023; Crescioli et al., 2016).
Under conditions of repeated antigen exposure, IgG4 levels gradually rise as a tolerance mechanism. This phenomenon is well-established in allergen immunotherapy, where controlled exposure to allergens shifts the antibody profile from IgE to IgG4, signifying the induction of immune tolerance (Uversky et al., 2023; James et al., 2011).
A similar pattern is now being observed in recipients of repeated COVID-19 mRNA vaccines. Multiple studies confirm that successive dosing, particularly at short intervals with persistent spike protein expression, promotes IgG4 class switching (Irrgang et al., 2023; Buhre et al., 2023; Lasrado et al., 2024; Espino et al., 2024). Although initially interpreted as regulatory or non-inflammatory, the growing dominance of IgG4 signals a deeper concern: the immune system has become tolerant to a pathogenic antigen, the spike protein, when the goal should be robust clearance, not coexistence (Uversky et al., 2023; Martin Perez et al, 2025).
Importantly, this IgG4 bias is not unique to mRNA platforms. Chronic antigen exposure in a Th2- or Treg-skewed immune environment, such as that induced by aluminium-adjuvanted vaccines, can also drive IgG4 production (Brewer et al, 1999; Umehara et al., 2012b). It is therefore implausible that aluminium-based vaccines do not contribute to similar skewing, even if more gradually or sub-clinically.
While IgG4 is frequently described as “anti-inflammatory,” this comes at a cost. In contexts where antigen elimination is critical, such as viral infections or cancer, an IgG4-dominated response impairs immune efficacy and facilitates persistence or immune escape (Wang et al., 2020; Karagiannis etal., 2013; Topchyan, Lin, and Cui, 2023). The pathological potential of this regulatory antibody is exemplified in IgG4-related disease (IgG4-RD), a systemic fibroinflammatory condition that arises when this otherwise tolerogenic mechanism becomes dysregulated (Stone, Zen, and Deshpande, 2012; Aochi et al., 2023; Ohyama, 2013).
Immune Skewing and Chronic Disease
Persistent bias toward Th2 and regulatory T cell (Treg) responses gradually erodes the body’s capacity to mount effective cytotoxic and inflammatory reactions when necessary. This is especially concerning for cancer surveillance, viral clearance, and control of intracellular pathogens — all of which require strong Th1-type immunity. Vaccines that repeatedly stimulate Th2 or Treg pathways shift immune resources toward antibody production at the expense of cellular immunity, leaving the host vulnerable to chronic infection and immune escape (Rook, 2009; Brewer et al, 1999; Marrack, McKee, and Munks, 2009).
This is particularly troubling in the context of modern vaccination schedules, where infants and young children encounter dozens of antigens and adjuvants during critical windows of immune development. The early-life immune system is highly impressionable, and repeated activation of Th2 pathways fosters long-term immunological imprinting. This skewing contributes to the rising prevalence of allergic diseases, asthma, atopic dermatitis, and autoimmune conditions — all linked to dysregulated Th2 and Treg activity (Meng et al., 2016; Lyons-Weiler, 2020; de Planell-Saguer, Lovinsky-Desir, and Miller 2014; Sun et al., 2016).
When effector responses are persistently suppressed in favour of regulation, the result is a state of “immune exhaustion” or “immune paralysis.” This has been observed in chronic infections and persistent antigen exposure, and is now emerging in repeated mRNA vaccine recipients, where signs of a dysfunctional immune profile are being documented (Irrgang et al., 2023; Utzschneider et al., 2016; Buhre et al., 2023; McKinney and Smith, 2016; Benitez Fuentes et al., 2022).
Ultimately, skewing immune responses away from Th1 pathways undermines the body's ability to defend against real threats. This imbalance contributes to the growing epidemic of chronic disease in industrialised societies. The very tools intended to prime the immune system are, by design, disrupting it. This is reflected in increased healthcare visits and adverse outcomes following vaccination, as shown in several large observational studies (Aaby et al., 2018; Lyons-Weiler, 2020; Simpson et al., 2002; McFadden et al., 2015; Chen, Zhang, and Deng 2012).
The Role of IgA in Immune Skewing: From Mucosal Defence to Tolerance
Immunoglobulin A (IgA) is essential for mucosal immunity, particularly in the respiratory and gastrointestinal tracts. Secretory IgA (sIgA) neutralises pathogens and blocks epithelial adherence without provoking inflammation (Macpherson et al., 2008).
However, during chronic antigen exposure, such as repeated vaccination or persistent spike protein expression, IgA responses become dysregulated. This is mediated by the Th17–Treg axis, particularly in mucosal tissues (Cerutti et al., 2011; Cao et al., 2012; Traxinger et al., 2022).
Initially, Th17 cells, stimulated by IL-6 and TGF-β, promote mucosal defence and class switching to IgA (Ivanov et al., 2009; Hirota et al., 2013). But chronic stimulation shifts the balance toward Treg dominance (Akdis and Akdis 2014). Regulatory T cells release IL-10 and TGF-β, driving further IgA (especially IgA2) and IgG4 production (Cerutti et al., 2013; Izcue, Coombes, and Powrie, 2006; Taylor et al., 2006).
In this tolerogenic state, IgA is no longer protective. It coats pathogens in a way that blocks their detection by effector cells, a process known as “antigen blocking” (Wegman et al., 2021). This allows ongoing colonisation or viral shedding while masking true infection (Mantis, Rol, and Corthesy 2011).
This dysfunctional IgA response, coupled with IgG4 class switching, marks a suppressive immune state induced by vaccine-driven chronic antigen exposure. Rather than preventing infection, this form of “tolerance” facilitates pathogen persistence under the false appearance of immunity (Stacy et al., 2021)
Original Antigenic Sin and the Consequences of Th Skewing
Original Antigenic Sin (OAS) was first noted in influenza research: the immune system preferentially recalls memory responses from its first encounter with a virus, even against mutated strains (Francis, 1960). Traditionally considered in terms of epitope recognition, recent studies show that OAS also includes imprinting of the immune environment, particularly the dominant T-helper (Th) profile (Zang et al., 2019; Reynolds et al., 2022b).
When first exposure to a pathogen like SARS-CoV-2 occurs via intramuscular vaccination, the immune system is trained to mount a Th2-skewed response. This results in memory B cells biased toward IgG4 or IgE production, instead of Th1-driven cytotoxic activity more appropriate for viral defence (Tang et al., 2022; Irrgang et al., 2023; Espino et al., 2024).
Upon real viral exposure, the immune system recalls this biased profile, even if it is maladaptive. As a result, the host may:
- Fail to mount a robust cytotoxic T-cell response.
- Overproduce non-neutralising antibodies that form immune complexes without clearing the virus.
- In some cases, trigger antibody-dependent enhancement (ADE) or allow persistent asymptomatic infection and viral shedding.
This broader understanding of OAS suggests that immunological imprinting is not just about antigen targeting; it’s about the kind of immune response that becomes “hardwired.” While antigenic drift involves pathogen evolution, Th-skewed imprinting distorts host defence, potentially long-term and across related viruses (Park et al., 2022; Zang et al., 2019; Pusnik et al., 2024)
Antibody-Dependent Enhancement and COVID-19 Vaccination
Antibody-dependent enhancement (ADE) occurs when antibodies, instead of neutralising a pathogen, facilitate its entry into immune cells such as monocytes and macrophages, triggering hyperinflammation and worsening disease outcomes. This is not a rare anomaly. ADE is a well-documented risk whenever the immune system is primed incorrectly, particularly when Th2 and Th17 pathways dominate the response to viral infections. This skew is commonly induced by injected vaccines that bypass the mucosal immune system and employ adjuvants to boost antibody production (Ricke and Malone, 2020; Xu et al., 2021; Han et al., 2011; Shukla et al., 2020). Vaccine-induced Th17 cells retain their memory for years, and this polarisation can lead to suboptimal immune responses upon subsequent exposures, particularly when a cytotoxic Th1 response would be more appropriate (Lindenstrøm et al., 2012).
A Th2-dominant profile generates antibody-mediated immunity at the expense of the cytotoxic responses required to clear intracellular pathogens. IgG4 and IgE antibodies are characteristic of this skewed profile and are associated with poor viral clearance, allergic-type inflammation, and immune tolerance (Marchese et al., 2024; Portilho et al., 2024). Th2 activation drives the recruitment of eosinophils, mast cells, and basophils, which release IL-4, IL-5, and IL-13, cytokines that directly suppress Th1 and CD8+ cytotoxic T-cell activity (Guenova et al., 2013; van de Burg et al., 2023). This creates a permissive environment for viral replication while escalating inflammatory damage, the hallmark of ADE.
This mechanism is well-established in flaviviruses. Prior infection or vaccination against Dengue virus can enhance the severity of subsequent Zika virus exposure through heterologous ADE (Dejnirattisai et al., 2016).
Even live-virus vaccines, such as the measles-mumps-rubella (MMR) vaccine, tilt the immune response toward Th2, not because of aluminium (which they do not contain), but because they bypass the mucosal immune system and fail to fully replicate the tissue-tropic signalling of wild virus infection. Post-vaccination studies have shown elevated IgE levels, a signature of Th2 dominance (Imani and Kehoe, 2001). This recruits cells poorly suited for viral clearance while suppressing interferon-gamma and cytotoxic lymphocyte activity. Despite using replication-competent measles virus, these vaccines fail to recreate the sterilising immunity induced by natural infection. Instead, they produce “modified measles” — a milder clinical picture in vaccinated individuals who nonetheless carry and transmit the virus (Rosen et al., 2014; Damien et al., 1998). As a result, silent spread among the vaccinated has become the new norm. Measles outbreaks are now disproportionately reported in vaccinated populations (Avramovich et al., 2018; Poland, and Jacobson, 1994).
COVID-19 vaccination presents a more extreme case. The persistent antigen signal from mRNA vaccination, particularly with repeated dosing, drives class-switching toward IgG4 and entrenches a Th2-skewed profile (Lasrado et al., 2024; Perez et al., 2025). This suppresses cytotoxic lymphocytes and promotes immune tolerance, allowing for persistent viral reactivation, immune complex formation, and multi-system inflammation (Uversky et al., 2023). In this context, the vaccine not only fails to prevent disease but becomes a contributing factor to its chronicity and recurrence. The immune pathology observed after multiple mRNA doses, including organ damage, reactivation of latent viruses, and immune exhaustion, is a direct result of this maladaptive Th2/IgG4 imprinting (Erice et al., 2025; Marchese et al., 2024).
COVID-19 thus offers a clear proof-of-principle: when the wrong arm of the immune system is activated against a virus, the result is not protection, but enhanced disease (Hou, Kang and Kim, 2009; Uversky et al., 2023; Irrgang et al., 2022; Valk et al., 2024; Gelderloos et al., 2024). Additionally, repeated COVID-19 vaccination has been shown to trigger the reactivation of latent viral infections, compounding the individual’s total disease burden (Chen et al., 2023; Gringeri et al., 2022; Hertel et al., 2022).
Early warning signals regarding COVID-19 vaccine safety were evident within the first month of rollout (Gee et al., 2021), yet recognition of these concerns remained limited within the broader scientific community, a resistance noted by Lancaster (2024).
Discussion: The High Cost of Immune Misprogramming
The immune system is not a passive barrier; it is a dynamic, memory-driven network that learns, adapts, and responds based on its earliest instructions. The nature of these early encounters — whether through natural infection or vaccination — defines not just immediate protection, but the shape of all future responses. This is the essence of immune imprinting.
Understanding the distinct functions of the Th1, Th2, and Th17 pathways is critical to evaluating vaccine performance. Th1 responses generate cytotoxic memory capable of clearing intracellular pathogens, while Th2 and Th17 pathways promote antibody production, tissue repair, and mucosal defence. These pathways are mutually antagonistic: when one is activated, the others are suppressed. Once a naïve T cell commits to a lineage, it remains fixed — memory T cells cannot be reprogrammed later. This principle has profound implications for vaccination, as the type of response a vaccine elicits determines whether the body will resolve or tolerate a future infection.
The smallpox vaccine stands as a historical outlier, not a model of modern practice. It induced true sterilising immunity via a live, replication-competent virus, engaging the Th1 pathway and producing long-lasting cytotoxic memory. In contrast, today’s subunit, inactivated, and mRNA vaccines bypass mucosal immunity and intracellular replication. They rely on adjuvants or synthetic constructs that preferentially stimulate Th2 or Th17 responses — insufficient for complete viral clearance.
This immunological misalignment carries significant evolutionary consequences. Vaccines that induce partial, non-sterilising immunity apply selective pressure on viruses, encouraging mutations that evade immune detection. As demonstrated with SARS-CoV-2, vaccine-resistant mutations like Y449S have emerged in direct correlation with rising vaccination rates. This is not random drift but a predictable outcome of immune selection in a suboptimal response environment.
Aluminium-based adjuvants, used for nearly a century, are well-documented to drive Th2-dominant responses, even in the absence of IL-4 or IL-13. Repeated exposure reinforces this skewing and promotes class switching to IgG4 and IgE. This immune profile may offer transient protection but fails to recreate the coordinated, cytotoxic, and sterilising immunity induced by live-virus exposure. Over time, this imprinting becomes fixed, guiding all future responses along the same non-cytotoxic track.
mRNA vaccines exacerbate this issue. Despite early claims of a Th1 bias, repeated dosing has been shown to induce IgG4 class switching — a hallmark of Th2 and regulatory (Treg) environments. IgG4 cannot emerge from Th1 conditions. Its presence alone confirms a shift away from cytotoxic immunity. This shift, once initiated, is not reversible, even with subsequent Th1-oriented vaccination attempts.
IgG4 is not a neutral antibody. While often described as "anti-inflammatory," it promotes immune tolerance, impairs pathogen clearance, and facilitates persistence in contexts where resolution is required. In cases like COVID-19, where the spike protein persists through repeated mRNA exposure, this tolerance enables chronic infection, immune exhaustion, and potentially, the reactivation of latent viruses.
Beyond IgG4, chronic vaccine-driven antigen exposure also distorts mucosal immunity. The role of IgA, normally protective in mucosal surfaces, becomes suppressive when overproduced in a tolerogenic environment. Instead of neutralising pathogens, dysregulated IgA may mask them from immune detection — facilitating colonisation and spread under the illusion of protection.
All of this culminates in the phenomenon known as “original antigenic sin.” This concept, once limited to B-cell epitope recognition, now extends to the very type of immune response imprinted. When a viral vaccine teaches the immune system to respond with tolerance instead of clearance — to suppress instead of eliminate — that instruction becomes embedded. Even when the pathogen mutates, the immune system recalls the first response, not the one it needs.
This skewing does not only hinder viral clearance; it predisposes to a wide spectrum of chronic diseases. Persistent Th2 and Treg bias undermine cancer surveillance, promote allergic and autoimmune disorders, and contribute to the immune exhaustion increasingly documented in recipients of repeated vaccination. These outcomes are not coincidental — they are consistent with the immunological architecture imposed by modern vaccine design.
Antibody-dependent enhancement (ADE) is a final, severe manifestation of this misdirection. When non-neutralising antibodies facilitate viral entry into immune cells, the result is not immunity but intensified disease. This risk is amplified in a Th2/IgG4-dominant landscape, where cytotoxic responses are suppressed and viral clearance is impaired. The COVID-19 vaccination campaigns have demonstrated, at population scale, the dangers of programming the wrong immune pathway — reinforcing tolerance in a context that demands cytotoxic defence.
The immune system of a healthy human is a finely tuned, self-correcting mechanism that has evolved to learn from exposure, resolve infections, and adapt with minimal disruption to the host. This system is not designed to be micromanaged. As such, any external intervention — including vaccination — must operate within the framework of immune logic, not against it.
Current vaccine strategies often violate this principle. By promoting Th2-biased responses through adjuvants like aluminium or persistent mRNA exposure, they override the immune system’s natural preference for Th1-driven viral clearance. This artificial skewing can create a form of “immune paralysis” where tolerance, not resolution, becomes the default.
Vaccination is frequently framed as a public health imperative, but it must also meet the standard of first, do no harm. When early-life vaccination repeatedly activates Th2 pathways, the resulting immune imprint becomes permanent — shaping how that individual responds to pathogens for life. The rise in chronic immune disorders, allergies, and immune exhaustion observed in vaccinated populations should be interpreted in this light: not as random outcomes, but as consequences of long-term immune misdirection.
What is most troubling is not that this risk was unknown, but that it has been ignored. The literature has long documented the mutual exclusivity of Th1 and Th2 pathways, the permanence of immune memory imprinting, and the suppressive role of IgG4. Yet regulatory agencies and public health bodies continue to promote vaccine strategies that reinforce these patterns. If health professionals within national systems like the NHS understand these dynamics — and it is almost certain many do — then the silence must be attributed to systemic suppression, not scientific ignorance.
This calls for a profound realignment of priorities. Vaccine policy must be led by immunological outcomes, not pharmaceutical convenience or political narrative. Sterilising immunity — the gold standard — has been largely abandoned in favour of tolerising strategies that prioritise antibody levels over actual resolution. This has consequences not only for individual health but for population-level pathogen evolution.
We must now confront an uncomfortable truth: vaccines have the power not only to protect, but to miseducate. When the immune system is repeatedly trained to tolerate instead of resolve, we create the illusion of immunity while silently suppressing our true defences. The consequences are cumulative, persistent, and, in some cases, irreversible.
The failure to acknowledge immune misprogramming is not just a scientific oversight — it is a breach of ethical responsibility. It is time to re-evaluate not only the composition of vaccines but the structures of oversight that determine how such products are approved and deployed. Health, not ideology, must be our guiding principle.
References:
Aaby P, Mogensen SW, Rodrigues A, Benn CS. (2018). Evidence of Increase in Mortality After the Introduction of Diphtheria-Tetanus-Pertussis Vaccine to Children Aged 6-35 Months in Guinea-Bissau: A Time for Reflection? Front Public Health. Mar 19;6:79. doi: 10.3389/fpubh.2018.00079 https://pmc.ncbi.nlm.nih.gov/articles/PMC5868131/
Aimanianda, V., Hauser, C.A., Echtenacher, B. and Latgé, J.P., (2009). Novel cellular and molecular mechanisms of induction of immune responses by aluminum adjuvants. Trends in Immunology, 30(6), pp.287–295. https://pubmed.ncbi.nlm.nih.gov/19439372/
Akdis, C. A., & Akdis, M. (2014). Mechanisms of immune tolerance to allergens: role of IL-10 and Tregs. The Journal of clinical investigation, 124(11), 4678–4680. https://doi.org/10.1172/JCI78891
Aochi, S., Uehara, M. and Yamamoto, M., (2023). IgG4-related disease emerging after COVID-19 mRNA vaccination. Internal Medicine, 62(10), pp.1547–1551. https://pubmed.ncbi.nlm.nih.gov/36889713/
Avramovich, E., Indenbaum, V., Haber, M., Amitai, Z., Tsifanski, E., Farjun, S., Sarig, A., Bracha, A., Castillo, K., Markovich, M. P., & Galor, I. (2018). Measles Outbreak in a Highly Vaccinated Population - Israel, July-August 2017. MMWR. Morbidity and mortality weekly report, 67(42), 1186–1188. https://doi.org/10.15585/mmwr.mm6742a4
Bancroft, T., Dillon, M. B., da Silva Antunes, R., Paul, S., Peters, B., Crotty, S., Lindestam Arlehamn, C. S., & Sette, A. (2016). Th1 versus Th2 T cell polarization by whole-cell and acellular childhood pertussis vaccines persists upon re-immunization in adolescence and adulthood. Cellular immunology, 304-305, 35–43. https://doi.org/10.1016/j.cellimm.2016.05.002
Benitez Fuentes, J. D., Mohamed Mohamed, K., de Luna Aguilar, A., Jiménez García, C., Guevara-Hoyer, K., Fernandez-Arquero, M., Rodríguez de la Peña, M. A., Garciía Bravo, L., Jiménez Ortega, A. F., Flores Navarro, P., Bartolome Arcilla, J., Alonso Arenilla, B., Baos Muñoz, E., Delgado-Iribarren García-Campero, A., Montealegre Sanz, M., Sanchez-Ramon, S., & Perez Segura, P. (2022). Evidence of exhausted lymphocytes after the third anti-SARS-CoV-2 vaccine dose in cancer patients. Frontiers in oncology, 12, 975980. https://doi.org/10.3389/fonc.2022.975980
Berin, M.C. and Shreffler, W.G., 2008. Mechanisms of allergic sensitization: implications for food allergy. Journal of Allergy and Clinical Immunology, 121(6), pp.1319–1328. https://pubmed.ncbi.nlm.nih.gov/18539190/
Bielinska, A. U., O'Konek, J. J., Janczak, K. W., & Baker, J. R., Jr (2016). Immunomodulation of TH2 biased immunity with mucosal administration of nanoemulsion adjuvant. Vaccine, 34(34), 4017–4024. https://doi.org/10.1016/j.vaccine.2016.06.043
Boots M. (2015). The Need for Evolutionarily Rational Disease Interventions: Vaccination Can Select for Higher Virulence. PLoS biology, 13(8), e1002236. https://doi.org/10.1371/journal.pbio.1002236
Bortolatto, J., Mirotti, L., Rodriguez, D., Gomes, E., & Russo, M. (2015). Adsorption of Toll-Like Receptor 4 Agonist to Alum-Based Tetanus Toxoid Vaccine Dampens Pro-T Helper 2 Activities and Enhances Antibody Responses. Journal of immunology research, 2015, 280238. https://doi.org/10.1155/2015/280238
Brewer, J.M., Conacher, M., Satoskar, A., Bluethmann, H. and Alexander, J., 1999. Aluminium hydroxide adjuvant initiates strong antigen-specific Th2 responses in the absence of IL-4- or IL-13-mediated signaling. The Journal of Immunology, 163(12), pp.6448–6454. https://pubmed.ncbi.nlm.nih.gov/10586035/
Buhre, J.S., Pongracz, T., Künsting, I., Lixenfeld, A.S., Wang, W., Nouta, J., Lehrian, S., Schmelter, F., Lunding, H.B., Dühring, L. and Kern, C., (2023). mRNA vaccines against SARS-CoV-2 induce comparably low long-term IgG Fc galactosylation and sialylation levels but increasing long-term IgG4 responses compared to an adenovirus-based vaccine. Frontiers in Immunology, 13, p.1020844. https://pubmed.ncbi.nlm.nih.gov/36713457/
Bungener, L., Geeraedts, F., Ter Veer, W., Medema, J., Wilschut, J., & Huckriede, A. (2008). Alum boosts TH2-type antibody responses to whole-inactivated virus influenza vaccine in mice but does not confer superior protection. Vaccine, 26(19), 2350–2359. https://doi.org/10.1016/j.vaccine.2008.02.063
Cao, H., Yang, S., Wang, Y., Luan, N., Yin, X., Lin, K., & Liu, C. (2021). An Established Th2-Oriented Response to an Alum-Adjuvanted SARS-CoV-2 Subunit Vaccine Is Not Reversible by Sequential Immunization with Nucleic Acid-Adjuvanted Th1-Oriented Subunit Vaccines. Vaccines, 9(11), 1261. https://doi.org/10.3390/vaccines9111261
Cao, A. T., Yao, S., Gong, B., Elson, C. O., & Cong, Y. (2012). Th17 cells upregulate polymeric Ig receptor and intestinal IgA and contribute to intestinal homeostasis. Journal of immunology (Baltimore, Md. : 1950), 189(9), 4666–4673. https://doi.org/10.4049/jimmunol.1200955
Cerutti, A., Chen, K., & Chorny, A. (2011). Immunoglobulin responses at the mucosal interface. Annual review of immunology, 29, 273–293. https://doi.org/10.1146/annurev-immunol-031210-101317
Cerutti, A., Cols, M., & Puga, I. (2013). Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes. Nature reviews. Immunology, 13(2), 118–132. https://doi.org/10.1038/nri3383
Chen, J., Zhang, Y., & Deng, Z. (2012). Imbalanced shift of cytokine expression between T helper 1 and T helper 2 (Th1/Th2) in intestinal mucosa of patients with post-infectious irritable bowel syndrome. BMC gastroenterology, 12, 91. https://doi.org/10.1186/1471-230X-12-91
Chen, J., Li, F., Tian, J., Xie, X., Tang, Q., Chen, Y., & Ge, Y. (2023). Varicella zoster virus reactivation following COVID-19 vaccination in patients with autoimmune inflammatory rheumatic diseases: A cross-sectional Chinese study of 318 cases. Journal of medical virology, 95(1), e28307. https://doi.org/10.1002/jmv.28307
Cohet, C., van der Most, R., Bauchau, V., Bekkat-Berkani, R., Doherty, T.M., Schuind, A., Tavares Da Silva, F., Rappuoli, R., Garçon, N. and Innis, B.L., 2019. Safety of AS03-adjuvanted influenza vaccines: A review of the evidence. Vaccine, 37(23), pp.3006–3021. https://pubmed.ncbi.nlm.nih.gov/31031030/
Crescioli, S., Correa, I., Karagiannis, P., Davies, A. M., Sutton, B. J., Nestle, F. O., & Karagiannis, S. N. (2016). IgG4 Characteristics and Functions in Cancer Immunity. Current allergy and asthma reports, 16(1), 7. https://doi.org/10.1007/s11882-015-0580-7
Crotty, S., 2011. Follicular helper CD4 T cells (TFH). Annual Review of Immunology, 29, pp.621–663. https://pubmed.ncbi.nlm.nih.gov/21314428/
Damien, B., Huiss, S., Schneider, F., & Muller, C. P. (1998). Estimated susceptibility to asymptomatic secondary immune response against measles in late convalescent and vaccinated persons. Journal of medical virology, 56(1), 85–90. https://doi.org/10.1002/(sici)1096-9071(199809)56:1<85::aid-jmv14>3.0.co;2-v
de Planell-Saguer, M., Lovinsky-Desir, S., & Miller, R. L. (2014). Epigenetic regulation: the interface between prenatal and early-life exposure and asthma susceptibility. Environmental and molecular mutagenesis, 55(3), 231–243. https://doi.org/10.1002/em.21836
Dejnirattisai, W., Supasa, P., Wongwiwat, W., Rouvinski, A., Barba-Spaeth, G., Duangchinda, T., Sakuntabhai, A., Cao-Lormeau, V. M., Malasit, P., Rey, F. A., Mongkolsapaya, J., & Screaton, G. R. (2016). Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus. Nature immunology, 17(9), 1102–1108. https://doi.org/10.1038/ni.3515
Demkowicz, W.E., Littaua, R.A. and Ennis, F.A., 1992. Human cytotoxic T-cell memory: long-lived responses to vaccinia virus. Journal of Virology, 66(1), pp.716–721. https://pubmed.ncbi.nlm.nih.gov/8642697/
Erice, A., Nuño, N., Prieto, L., & Caballero, C. (2025). Immune Imprinting, Non-Durable Hybrid Immunity, and Hybrid Immune Damping Following SARS-CoV-2 Primary Vaccination with BNT162b2 and Boosting with mRNA-1273. Vaccines, 13(3), 310. https://doi.org/10.3390/vaccines13030310
Espino, A.M., Armina-Rodriguez, A., Alvarez, L., Ocasio-Malavé, C., Ramos-Nieves, R., Rodriguez Martinó, E.I., López-Marte, P., Torres, E.A. and Sariol, C.A., (2024). The anti-SARS-CoV-2 IgG1 and IgG3 antibody isotypes with limited neutralizing capacity against omicron elicited in a Latin population a switch toward IgG4 after multiple doses with the mRNA Pfizer–BioNTech vaccine. Viruses, 16(2), p.187. https://pubmed.ncbi.nlm.nih.gov/38399963/
Fenner, F., Henderson, D.A., Arita, I., Ježek, Z. and Ladnyi, I.D., 1987. Smallpox and its eradication. Geneva: World Health Organization. https://pmc.ncbi.nlm.nih.gov/articles/PMC2491071/pdf/bullwho00076-0026.pdf
Finkelman, F.D., Katona, I.M., Urban, J.F., Holmes, J., Ohara, J., Tung, A.S., Sample, J.V. and Paul, W.E., (1988). IL-4 is required to generate and sustain in vivo IgE responses. Journal of Immunology, 141(7), pp.2335–2341. https://pubmed.ncbi.nlm.nih.gov/2459206/
Finkelman, F.D., Hogan, S.P., Hershey, G.K., Rothenberg, M.E. and Wills-Karp, M., (2005). Importance of cytokines in murine allergic airway disease and human asthma. Journal of Immunology, 175(2), pp.1059-1066. https://pubmed.ncbi.nlm.nih.gov/20130218/
Francis, T., 1960. On the doctrine of original antigenic sin. Proceedings of the American Philosophical Society, 104(6), pp.572-578. https://www.jstor.org/stable/985534
Galli, S.J., Grimbaldeston, M. & Tsai, M. (2008). Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nature Reviews Immunology, 8(6), 478–486 https://pubmed.ncbi.nlm.nih.gov/18483499/
Gao, P., Kang, J., Johnson, Z., MacDonald, M., Andrews, J., Lin, X., Karas, A., Luo, L. and Yong, S., 2023. mRNA vaccination induces functional IgG4 antibodies. Nature Communications, 14(1), p.3451.
Garcia-Beltran, W.F., Lam, E.C., St Denis, K., Nitido, A.D., Garcia, Z.H., Hauser, B.M., Feldman, J., Pavlovic, M.N., Gregory, D.J., Poznansky, M.C. and Sigal, A., 2021. Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell, 184(9), pp.2372–2383.e9. https://pubmed.ncbi.nlm.nih.gov/33930298/
Gee, J., Marquez, P., Su, J., Calvert, G.M., Liu, R., Myers, T., Nair, N., Martin, S., Clark, T., Markowitz, L. and Shay, D.K., 2021. First month of COVID-19 vaccine safety monitoring—United States, December 14, 2020–January 13, 2021. MMWR. Morbidity and Mortality Weekly Report, 70(8), p.283. https://pubmed.ncbi.nlm.nih.gov/33630816/
Geoghegan, J.L. and Holmes, E.C., 2018. The phylogenomics of evolving virus virulence. Nature Reviews Genetics, 19(12), pp.756–769. https://pubmed.ncbi.nlm.nih.gov/30305704/
Gelderloos, A.T., Verheul, M.K., Middelhof, I., de Zeeuw-Brouwer, M.L., van Binnendijk, R.S., Buisman, A.M., and van Kasteren, P.B., (2024). Repeated COVID-19 mRNA vaccination results in IgG4 class switching and decreased NK cell activation by S1-specific antibodies in older adults. Immunity & Ageing, 21(1), p.63. https://pubmed.ncbi.nlm.nih.gov/39272189/
Gringeri, M., Battini, V., Cammarata, G., Mosini, G., Guarnieri, G., Leoni, C., Pozzi, M., Radice, S., Clementi, E., & Carnovale, C. (2022a). Herpes zoster and simplex reactivation following COVID-19 vaccination: new insights from a vaccine adverse event reporting system (VAERS) database analysis. Expert review of vaccines, 21(5), 675–684. https://doi.org/10.1080/14760584.2022.2044799
Guenova, E., Watanabe, R., Teague, J. E., Desimone, J. A., Jiang, Y., Dowlatshahi, M., Schlapbach, C., Schaekel, K., Rook, A. H., Tawa, M., Fisher, D. C., Kupper, T. S., & Clark, R. A. (2013). TH2 cytokines from malignant cells suppress TH1 responses and enforce a global TH2 bias in leukemic cutaneous T-cell lymphoma. Clinical cancer research: an official journal of the American Association for Cancer Research, 19(14), 3755–3763. https://doi.org/10.1158/1078-0432.CCR-12-3488
Guimarães, L. E., Baker, B., Perricone, C., & Shoenfeld, Y. (2015). Vaccines, adjuvants and autoimmunity. Pharmacological research, 100, 190–209.https://doi.org/10.1016/j.phrs.2015.08.003
Hadfield (n.d.) Real-time tracking of measles full genome virus evolution. Bioinformatics. Available at: https://nextstrain.org/measles/genome (Accessed: 07 May 2025).
Halstead, S.B. and O'Rourke, E.J., (1977). Antibody-enhanced dengue virus infection in primate leukocytes. Nature, 265(5596), pp.739–741. https://pubmed.ncbi.nlm.nih.gov/404559/
Harrington, L.E., Hatton, R.D., Mangan, P.R., Turner, H., Murphy, T.L., Murphy, K.M. and Weaver, C.T., 2005. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the Th1 and Th2 lineages. Nature Immunology, 6(11), pp.1123–1132. https://pubmed.ncbi.nlm.nih.gov/16200070/
Han, J. F., Cao, R. Y., Deng, Y. Q., Tian, X., Jiang, T., Qin, E. D., & Qin, C. F. (2011). Antibody dependent enhancement infection of enterovirus 71 in vitro and in vivo. Virology journal, 8, 106. https://doi.org/10.1186/1743-422X-8-106
Hertel, M., Heiland, M., Nahles, S., von Laffert, M., Mura, C., Bourne, P. E., Preissner, R., & Preissner, S. (2022). Real-world evidence from over one million COVID-19 vaccinations is consistent with reactivation of the varicella-zoster virus. Journal of the European Academy of Dermatology and Venereology: JEADV, 36(8), 1342–1348. https://doi.org/10.1111/jdv.18184
Hirota, K., Turner, J. E., Villa, M., Duarte, J. H., Demengeot, J., Steinmetz, O. M., & Stockinger, B. (2013). Plasticity of Th17 cells in Peyer's patches is responsible for the induction of T cell-dependent IgA responses. Nature immunology, 14(4), 372–379. https://doi.org/10.1038/ni.2552.
HogenEsch, H., 2002. Mechanism of immunopotentiation and safety of aluminum adjuvants. Frontiers in Bioscience, 7, pp.d1047–d1055. https://pubmed.ncbi.nlm.nih.gov/23335921/
Hou, W., Kang, H. S., & Kim, B. S. (2009). Th17 cells enhance viral persistence and inhibit T cell cytotoxicity in a model of chronic virus infection. The Journal of experimental medicine, 206(2), 313–328. https://doi.org/10.1084/jem.20082030
Imani, F., & Kehoe, K. E. (2001). Infection of human B lymphocytes with MMR vaccine induces IgE class switching. Clinical immunology (Orlando, Fla.), 100(3), 355–361. https://doi.org/10.1006/clim.2001.5073
Irrgang, P., Gerling, J., Kocher, K., Lapuente, D., Steininger, P., Habenicht, K., Wytopil, M., Beileke, S., Schäfer, S., Zhong, J. and Ssebyatika, G., (2023). Class switch toward noninflammatory, spike-specific IgG4 antibodies after repeated SARS-CoV-2 mRNA vaccination. Science Immunology, 8(79), p.eade2798. https://pubmed.ncbi.nlm.nih.gov/36548397/
Ivanov, I. I., Atarashi, K., Manel, N., Brodie, E. L., Shima, T., Karaoz, U., Wei, D., Goldfarb, K. C., Santee, C. A., Lynch, S. V., Tanoue, T., Imaoka, A., Itoh, K., Takeda, K., Umesaki, Y., Honda, K., & Littman, D. R. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell, 139(3), 485–498. https://doi.org/10.1016/j.cell.2009.09.033
Izcue, A., Coombes, J. L., & Powrie, F. (2006). Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunological reviews, 212, 256–271. https://doi.org/10.1111/j.0105-2896.2006.00423.x
James, L.K., Shamji, M.H., Walker, S.M., Wilson, D.R., Jacobson, M.R., Kimber, I., Till, S.J., Durham, S.R. and Francis, J.N., (2011). Long-term tolerance after allergen immunotherapy is accompanied by selective persistence of blocking antibodies. Journal of Allergy and Clinical Immunology, 127(2), pp.509–516.e5. https://pubmed.ncbi.nlm.nih.gov/21281875/
Jeannin, P., Lecoanet, S., Delneste, Y., Gauchat, J.F. and Bonnefoy, J.Y., 1998. IgE versus IgG4 production can be differentially regulated by IL-10. Journal of Immunology, 160(7), pp.3555–3561. https://pubmed.ncbi.nlm.nih.gov/9531318/
Karagiannis, P., Gilbert, A. E., Josephs, D. H., Ali, N., Dodev, T., Saul, L., Correa, I., Roberts, L., Beddowes, E., Koers, A., Hobbs, C., Ferreira, S., Geh, J. L., Healy, C., Harries, M., Acland, K. M., Blower, P. J., Mitchell, T., Fear, D. J., Spicer, J. F., … Karagiannis, S. N. (2013). IgG4 subclass antibodies impair antitumor immunity in melanoma. The Journal of clinical investigation, 123(4), 1457–1474. https://doi.org/10.1172/JCI65579
Katzman, S. D., Gallo, E., Hoyer, K. K., & Abbas, A. K. (2011). Differential requirements for Th1 and Th17 responses to a systemic self-antigen. Journal of immunology (Baltimore, Md. : 1950), 186(8), 4668–4673. https://doi.org/10.4049/jimmunol.1003786
Kidd, P., 2003. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Alternative Medicine Review, 8(3), pp.223–246. https://pubmed.ncbi.nlm.nih.gov/12946237/
Kim, H.W., Canchola, J.G., Brandt, C.D., Pyles, G., Chanock, R.M., Jensen, K. and Parrott, R.H., 1969. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. American Journal of Epidemiology, 89(4), pp.422–434. https://pubmed.ncbi.nlm.nih.gov/4305198/
Kim, S. H., Yeo, S. G., Cho, J. H., Kim, H. B., Kim, N. J., Oh, M. D., Choe, K. W., Jee, Y., & Cho, H. (2006). Cell-mediated immune responses to smallpox vaccination. Clinical and vaccine immunology: CVI, 13(10), 1172–1174. https://doi.org/10.1128/CVI.00181-06
Langmuir, A. D., Henderson, D. A., Serfling, R. E., & Sherman, I. L. (1962). The importance of measles as a health problem. American journal of public health and the nation's health, 52(2)Suppl(Suppl 2), 1–4. https://doi.org/10.2105/ajph.52.suppl_2.1
Langrish, C.L., Chen, Y., Blumenschein, W.M., Mattson, J., Basham, B., Sedgwick, J.D., McClanahan, T., Kastelein, R.A. and Cua, D.J., 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. Journal of Experimental Medicine, 201(2), pp.233–240. https://pubmed.ncbi.nlm.nih.gov/15657292/
Lataster, J., 2024. IgG4 switching after mRNA vaccines: immune tolerance or pathology? Australian Journal of General Practice, [online] Available at: https://www1.racgp.org.au/getattachment/abf6c70e-c2db-4035-8cf2-a60e3ae8d2c6/Letters.aspx
Lasrado, N., Collier, A.R.Y., Miller, J., Hachmann, N.P., Liu, J., Anand, T., Bondzie, E.A., Fisher, J.L., Mazurek, C.R., Patio, R.C. and Rodrigues, S.L., 2024. Waning immunity and IgG4 responses following bivalent mRNA boosting. Science Advances, 10(8), p.eadj9945. https://pubmed.ncbi.nlm.nih.gov/38394195/
Lindblad, E.B., 2004. Aluminium compounds for use in vaccines. Immunology and Cell Biology, 82(5), pp.497–505 https://pubmed.ncbi.nlm.nih.gov/15479435/
Lindenstrøm, T., Woodworth, J., Dietrich, J., Aagaard, C., Andersen, P., & Agger, E. M. (2012). Vaccine-induced th17 cells are maintained long-term postvaccination as a distinct and phenotypically stable memory subset. Infection and immunity, 80(10), 3533–3544. https://doi.org/10.1128/IAI.00550-12
Liu, L., Xu, Z., Fuhlbrigge, R.C., Pena-Cruz, V., Lieberman, J. and Kupper, T.S., 2005. Vaccinia virus induces strong immunoregulatory cytokine production in healthy human epidermal keratinocytes: a novel strategy for immune evasion. Journal of Virology, 79(12), pp.7363–7370. https://pubmed.ncbi.nlm.nih.gov/15919891/
Lyons-Weiler, J., 2020. Pathogenic priming likely contributes to serious and critical illness and mortality in COVID-19 via autoimmunity. International Journal of Vaccine Theory, Practice, and Research, 1(1), pp.12–18. https://pubmed.ncbi.nlm.nih.gov/32292901/
Macpherson, A. J., McCoy, K. D., Johansen, F. E., & Brandtzaeg, P. (2008). The immune geography of IgA induction and function. Mucosal immunology, 1(1), 11–22. https://doi.org/10.1038/mi.2007.6
Mantis, N. J., Rol, N., & Corthésy, B. (2011). Secretory IgA's complex roles in immunity and mucosal homeostasis in the gut. Mucosal immunology, 4(6), 603–611. https://doi.org/10.1038/mi.2011.41
Marchese, A. M., Fries, L., Beyhaghi, H., Vadivale, M., Zhu, M., Cloney-Clark, S., Plested, J. S., Chung, A. W., Dunkle, L. M., & Kalkeri, R. (2024). Mechanisms and implications of IgG4 responses to SARS-CoV-2 and other repeatedly administered vaccines. The Journal of infection, 89(6), 106317. https://doi.org/10.1016/j.jinf.2024.106317
Marrack, P., McKee, A.S. and Munks, M.W. (2009) Towards an understanding of the adjuvant action of aluminium. Nature Reviews Immunology, 9(4), pp.287–293. https://pubmed.ncbi.nlm.nih.gov/19247370/
Martín Pérez, C., Ruiz-Rius, S., Ramírez-Morros, A., Vidal, M., Opi, D. H., Santamaria, P., Blanco, J., Vidal-Alaball, J., Beeson, J. G., Molinos-Albert, L. M., Aguilar, R., Ruiz-Comellas, A., Moncunill, G., & Dobaño, C. (2025). Post-vaccination IgG4 and IgG2 class switch associates with increased risk of SARS-CoV-2 infections. The Journal of infection, 90(4), 106473. https://doi.org/10.1016/j.jinf.2025.106473
McCarthy, K. R., Von Holle, T. A., Sutherland, L. L., Oguin, T. H., 3rd, Sempowski, G. D., Harrison, S. C., & Moody, M. A. (2021). Differential immune imprinting by influenza virus vaccination and infection in nonhuman primates. Proceedings of the National Academy of Sciences of the United States of America, 118(23), e2026752118. https://doi.org/10.1073/pnas.2026752118
McFadden, J. P., Thyssen, J. P., Basketter, D. A., Puangpet, P., & Kimber, I. (2015). T helper cell 2 immune skewing in pregnancy/early life: chemical exposure and the development of atopic disease and allergy. The British journal of dermatology, 172(3), 584–591. https://doi.org/10.1111/bjd.13497
McKinney, E. F., & Smith, K. G. (2016). T-cell exhaustion: understanding the interface of chronic viral and autoinflammatory diseases. Immunology and cell biology, 94(10), 935–942. https://doi.org/10.1038/icb.2016.81
Meng, S. S., Gao, R., Yan, B. D., Ren, J., Wu, F., Chen, P., Zhang, J., Wang, L. F., Xiao, Y. M., & Liu, J. (2016). Maternal allergic disease history affects childhood allergy development through impairment of neonatal regulatory T-cells. Respiratory research, 17(1), 114. https://doi.org/10.1186/s12931-016-0430-8
Miller, I. F., & Metcalf, C. J. (2019). Vaccine-driven virulence evolution: consequences of unbalanced reductions in mortality and transmission and implications for pertussis vaccines. Journal of the Royal Society, Interface, 16(161), 20190642. https://doi.org/10.1098/rsif.2019.0642
Miller, J. D., van der Most, R. G., Akondy, R. S., Glidewell, J. T., Albott, S., Masopust, D., Murali-Krishna, K., Mahar, P. L., Edupuganti, S., Lalor, S., Germon, S., Del Rio, C., Mulligan, M. J., Staprans, S. I., Altman, J. D., Feinberg, M. B., & Ahmed, R. (2008). Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity, 28(5), 710–722. https://doi.org/10.1016/j.immuni.2008.02.020
Moore, J.P., 2021. Approaches for optimal use of different COVID-19 vaccines: issues of viral variation and vaccine efficacy. JAMA, 325(13), pp.1251–1252. https://pubmed.ncbi.nlm.nih.gov/33662101/
Moss, B., 2013. Poxvirus DNA replication. Cold Spring Harbor Perspectives in Biology, 5(9), a010199. https://pubmed.ncbi.nlm.nih.gov/23838441/
Muñoz-Alía, M.A., Nace, R.A., Zhang, L., Russell, S.J. and McFadden, G., 2021. Serotypic evolution of measles virus is constrained by multiple co-dominant B cell epitopes. Cell Reports, 35(3), p.108949. https://pubmed.ncbi.nlm.nih.gov/33948566/
Oskam, N., Damelang, T., Streutker, M., Ooijevaar-de Heer, P., Nouta, J., Koeleman, C., Van Coillie, J., Wuhrer, M., Vidarsson, G., & Rispens, T. (2023). Factors affecting IgG4-mediated complement activation. Frontiers in immunology, 14, 1087532. https://doi.org/10.3389/fimmu.2023.1087532
Ouyang, W., Kolls, J.K. and Zheng, Y., 2008. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity, 28(4), pp.454–467. https://pubmed.ncbi.nlm.nih.gov/18400188/
Ohyama, K., Koike, H., Iijima, M., Hashimoto, R., Tomita, M., Kawagashira, Y., Satou, A., Nakamura, S., & Sobue, G. (2013). IgG4-related neuropathy: a case report. JAMA neurology, 70(4), 502–505. https://doi.org/10.1001/jamaneurol.2013.658.
Park, Y. J., Pinto, D., Walls, A. C., Liu, Z., De Marco, A., Benigni, F., Zatta, F., Silacci-Fregni, C., Bassi, J., Sprouse, K. R., Addetia, A., Bowen, J. E., Stewart, C., Giurdanella, M., Saliba, C., Guarino, B., Schmid, M. A., Franko, N. M., Logue, J. K., Dang, H. V., … Veesler, D. (2022). Imprinted antibody responses against SARS-CoV-2 Omicron sublineages. Science (New York, N.Y.), 378(6620), 619–627. https://doi.org/10.1126/science.adc9127
Paul, W.E. and Zhu, J., 2010. How are T(H)2-type immune responses initiated and amplified?. Nature Reviews Immunology, 10(4), pp.225–235. https://pubmed.ncbi.nlm.nih.gov/20336151/
Martín Pérez, C., Ruiz-Rius, S., Ramírez-Morros, A., Vidal, M., Opi, D. H., Santamaria, P., Blanco, J., Vidal-Alaball, J., Beeson, J. G., Molinos-Albert, L. M., Aguilar, R., Ruiz-Comellas, A., Moncunill, G., & Dobaño, C. (2025). Post-vaccination IgG4 and IgG2 class switch associates with increased risk of SARS-CoV-2 infections. The Journal of infection, 90(4), 106473. https://doi.org/10.1016/j.jinf.2025.106473
Perrigoue, J.G., Saenz, S.A., Siracusa, M.C., Allenspach, E.J., Taylor, B.C., Giacomin, P.R., Nair, M.G., Du, Y., Zaph, C., van Rooijen, N., Comeau, M.R., Pearce, E.J., Goldschmidt, M., Artis, D. (2009) MHC class II–dependent basophil–CD4+ T cell interactions promote Th2 cytokine–dependent immunity. Nature Immunology, 10(7), pp.697–705. https://pubmed.ncbi.nlm.nih.gov/19465906/
Pillai, S., 2023. Is it bad, is it good, or is IgG4 just misunderstood? Science Immunology, 8(81), p.eadg7327. https://pubmed.ncbi.nlm.nih.gov/36749191/
Plotkin, S.A., 2010. Correlates of protection induced by vaccination. Clinical and Vaccine Immunology, 17(7), pp.1055–1065. https://pubmed.ncbi.nlm.nih.gov/20463105/
Poland, G. A., Jacobson, R. M. (1994). Failure to reach the goal of measles elimination. Apparent paradox of measles infections in immunized persons. Archives of internal medicine, 154(16), 1815–1820. https://pubmed.ncbi.nlm.nih.gov/8053748/
Portilho, A. I., Silva, V. O., Da Costa, H. H. M., Yamashiro, R., de Oliveira, I. P., de Campos, I. B., Prudencio, C. R., Matsuda, E. M., de Macedo Brígido, L. F., & De Gaspari, E. (2024). An unexpected IgE anti-receptor binding domain response following natural infection and different types of SARS-CoV-2 vaccines. Scientific reports, 14(1), 20003. https://doi.org/10.1038/s41598-024-71047-5
Pouwels, K.B., Pritchard, E., Matthews, P.C., Stoesser, N., Eyre, D.W., Vihta, K.D., House, T., Hay, J., Bell, I., Newton, J.N. and Farrar, J., 2021. Impact of Delta on viral burden and vaccine effectiveness against new SARS-CoV-2 infections in the UK. medRxiv. https://pubmed.ncbi.nlm.nih.gov/34650248/
Pulendran, B., Ahmed, R., 2011. Immunological mechanisms of vaccination. Nature Immunology, 12(6), pp.509–517. https://pubmed.ncbi.nlm.nih.gov/21739679/
Pulendran, B., S Arunachalam, P., & O'Hagan, D. T. (2021). Emerging concepts in the science of vaccine adjuvants. Nature reviews. Drug discovery, 20(6), 454–475. https://doi.org/10.1038/s41573-021-00163-y
Pušnik, J., Zorn, J., Monzon-Posadas, W. O., Peters, K., Osypchuk, E., Blaschke, S., & Streeck, H. (2024). Vaccination impairs de novo immune response to omicron breakthrough infection, a precondition for the original antigenic sin. Nature communications, 15(1), 3102. https://doi.org/10.1038/s41467-024-47451-w
Read, A. F., Baigent, S. J., Powers, C., Kgosana, L. B., Blackwell, L., Smith, L. P., Kennedy, D. A., Walkden-Brown, S. W., & Nair, V. K. (2015). Imperfect Vaccination Can Enhance the Transmission of Highly Virulent Pathogens. PLoS biology, 13(7), e1002198. https://doi.org/10.1371/journal.pbio.1002198
Reed, S.G., Orr, M.T. and Fox, C.B. (2013) Key roles of adjuvants in modern vaccines. Nature Medicine, 19(12), pp.1597–1608. https://pubmed.ncbi.nlm.nih.gov/24309663/
Reynolds, C.J., Pade, C., Gibbons, J.M., Otter, A.D., Lin, K.M., Munoz Sandoval, D., Pieper, F.P., Butler, D.K., Liu, S., Joy, G. and Cook, J.A., (2022a). Immune boosting by B.1.1.529 (Omicron) depends on previous SARS-CoV-2 exposure. Science, 377(6603), p.eabq1841. https://pubmed.ncbi.nlm.nih.gov/35699621/
Reynolds, C. J., Gibbons, J. M., Pade, C., Lin, K. M., Sandoval, D. M., Pieper, F., Butler, D. K., Liu, S., Otter, A. D., Joy, G., Menacho, K., Fontana, M., Smit, A., Kele, B., Cutino-Moguel, T., Maini, M. K., Noursadeghi, M., COVIDsortium Immune Correlates Network‡, Brooks, T., Semper, A., … Boyton, R. J. (2022b). Heterologous infection and vaccination shapes immunity against SARS-Cov-2 variants. Science (New York, N.Y.), 375(6577), 183–192. https://doi.org/10.1126/science.abm0811
Ricke, D. and Malone, R.W., 2020. Medical countermeasures analysis of 2019-nCoV and vaccine risks for antibody-dependent enhancement (ADE). Available at SSRN 3546070. https://papers.ssrn.com/sol3/papers.cfm?abstract_id=3546070
Rispens, T., & Huijbers, M. G. (2023). The unique properties of IgG4 and its roles in health and disease. Nature reviews. Immunology, 23(11), 763–778. https://doi.org/10.1038/s41577-023-00871-z
Romagnani, S., (2000). T-cell subsets (Th1 versus Th2). Annals of Allergy, Asthma & Immunology, 85(1), pp.9–21. https://pubmed.ncbi.nlm.nih.gov/10923599/
Rook, G.A.W., 2009. Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis. Immunology, 126(1), pp.3–11. https://pubmed.ncbi.nlm.nih.gov/19120493/
Rosen, J. B., Rota, J. S., Hickman, C. J., Sowers, S. B., Mercader, S., Rota, P. A., Bellini, W. J., Huang, A. J., Doll, M. K., Zucker, J. R., & Zimmerman, C. M. (2014). Outbreak of measles among persons with prior evidence of immunity, New York City, 2011. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, 58(9), 1205–1210. https://doi.org/10.1093/cid/ciu105
Sanchez-Sampedro, L., Perdiguero, B., Mejias-Perez, E., Garcia-Arriaza, J., Di Pilato, M., and Esteban, M., 2015. The evolution of poxvirus vaccines. Viruses, 7(4), pp.1726–1803. https://pubmed.ncbi.nlm.nih.gov/25853483/
Sasaki, K., Kato, M., Takahashi, T., Ochi, S., Ichinose, Y., Shiraki, K., Asano, Y., Iwanaga, M., & Tsuji, T. (2003). Live varicella vaccine polarizes the mucosal adjuvant action of cholera toxin or its B subunit on specific Th1-type helper T cells with a single nasal coadministration in mice. Journal of medical virology, 70(2), 329–335. https://doi.org/10.1002/jmv.10399
Seder, R.A., Darrah, P.A. and Roederer, M. (2008) T-cell quality in memory and protection: implications for vaccine design. Nature Reviews Immunology, 8(4), pp.247–258. https://pubmed.ncbi.nlm.nih.gov/18323851/
Shi, G., Ramaswamy, M., Vistica, B. P., Cox, C. A., Tan, C., Wawrousek, E. F., Siegel, R. M., & Gery, I. (2009). Unlike Th1, Th17 cells mediate sustained autoimmune inflammation and are highly resistant to restimulation-induced cell death. Journal of immunology (Baltimore, Md. : 1950), 183(11), 7547–7556. https://doi.org/10.4049/jimmunol.0900519
Shukla, R., Ramasamy, V., Shanmugam, R. K., Ahuja, R., & Khanna, N. (2020). Antibody-Dependent Enhancement: A Challenge for Developing a Safe Dengue Vaccine. Frontiers in cellular and infection microbiology, 10, 572681. https://doi.org/10.3389/fcimb.2020.572681
Simon, W.L., Salk, H.M., Ovsyannikova, I.G., Kennedy, R.B. and Poland, G.A., 2014. Cytokine production associated with smallpox vaccine responses. Immunotherapy, 6(7), pp.763–770. https://pubmed.ncbi.nlm.nih.gov/25428648/
Simpson, C. R., Anderson, W. J., Helms, P. J., Taylor, M. W., Watson, L., Prescott, G. J., Godden, D. J., & Barker, R. N. (2002). Coincidence of immune-mediated diseases driven by Th1 and Th2 subsets suggests a common aetiology. A population-based study using computerized general practice data. Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology, 32(1), 37–42. https://doi.org/10.1046/j.0022-0477.2001.01250.x
Slifka, M.K. and Amanna, I.J., 2014. How advances in immunology provide insight into improving vaccine efficacy. Vaccine, 32(25), pp.2948–2957. https://pubmed.ncbi.nlm.nih.gov/24709587/
Sokol, C.L., Barton, G.M., Farr, A.G. and Medzhitov, R. (2008) A mechanism for the initiation of allergen-induced T helper type 2 responses. Nature Immunology, 9(3), pp.310–318. https://pubmed.ncbi.nlm.nih.gov/18300366/
Stacey, H. D., Golubeva, D., Posca, A., Ang, J. C., Novakowski, K. E., Zahoor, M. A., Kaushic, C., Cairns, E., Bowdish, D. M. E., Mullarkey, C. E., & Miller, M. S. (2021). IgA potentiates NETosis in response to viral infection. Proceedings of the National Academy of Sciences of the United States of America, 118(27), e2101497118. https://doi.org/10.1073/pnas.2101497118
Stone, J.H., Zen, Y. and Deshpande, V., 2012. IgG4-related disease. New England Journal of Medicine, 366(6), pp.539–551. https://pubmed.ncbi.nlm.nih.gov/22316447/
Sun, R., Tang, X. Y., & Yang, Y. (2016). Immune imbalance of regulatory T/type 2 helper cells in the pathogenesis of allergic rhinitis in children. The Journal of laryngology and otology, 130(1), 89–94. https://doi.org/10.1017/S0022215115003096
Swain, S.L., McKinstry, K.K. and Strutt, T.M., 2012. Expanding roles for CD4⁺ T cells in immunity to viruses. Nature Reviews Immunology, 12(2), pp.136–148. https://pubmed.ncbi.nlm.nih.gov/22266691/
Szabo, S.J., Kim, S.T., Costa, G.L., Zhang, X., Fathman, C.G. and Glimcher, L.H., 2003. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell, 100(6), pp.655–669. https://pubmed.ncbi.nlm.nih.gov/10761931/.
Tang, J., Zeng, C., Cox, T. M., Li, C., Son, Y. M., Cheon, I. S., Wu, Y., Behl, S., Taylor, J. J., Chakaraborty, R., Johnson, A. J., Shiavo, D. N., Utz, J. P., Reisenauer, J. S., Midthun, D. E., Mullon, J. J., Edell, E. S., Alameh, M. G., Borish, L., Teague, W. G., … Sun, J. (2022). Respiratory mucosal immunity against SARS-CoV-2 after mRNA vaccination. Science immunology, 7(76), eadd4853. https://doi.org/10.1126/sciimmunol.add4853
Taylor, A., Verhagen, J., Blaser, K., Akdis, M., & Akdis, C. A. (2006). Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology, 117(4), 433–442. https://doi.org/10.1111/j.1365-2567.2006.02321.x
Topchyan, P., 2023. The role of CD4 T cell help in CD8 T cell differentiation and function during chronic infection and cancer. Frontiers in Immunology, 14, p.1224864. https://pubmed.ncbi.nlm.nih.gov/37970230/
Traxinger, B. R., Richert-Spuhler, L. E., & Lund, J. M. (2022). Mucosal tissue regulatory T cells are integral in balancing immunity and tolerance at portals of antigen entry. Mucosal immunology, 15(3), 398–407. https://doi.org/10.1038/s41385-021-00471-x
Trinchieri, G. (2003) Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nature Reviews Immunology, 3(2), pp.133–146. https://pubmed.ncbi.nlm.nih.gov/12563297/
Tscharke, D.C., Karupiah, G., Zhou, J., Palmore, T., Irvine, K.R., Haeryfar, S.M., Williams, S., Sidney, J., Sette, A., Bennink, J.R. and Yewdell, J.W., 2005. Identification of poxvirus CD8+ T cell determinants to enable rational design and characterization of smallpox vaccines. Journal of Experimental Medicine, 201(1), pp.95–104. https://pubmed.ncbi.nlm.nih.gov/15623576/
Umehara, H., Okazaki, K., Masaki, Y., Kawano, M., Yamamoto, M., Saeki, T., Matsui, S., Yoshino, T., Nakamura, S., Kawa, S. and Nagata, K., (2012a). Comprehensive diagnostic criteria for IgG4-related disease (IgG4-RD), 2011. Modern Rheumatology, 22(1), pp.21–30.https://pubmed.ncbi.nlm.nih.gov/22218969/
Umehara, H., Okazaki, K., Masaki, Y., Kawano, M., Yamamoto, M., Saeki, T., Matsui, S., Yoshino, T., Nakamura, S., Kawa, S. and Suzuki, S., (2012b). A novel clinical entity, IgG4-related disease (IgG4RD): general concept and details. Modern Rheumatology, 22(1), pp.1–14. https://pubmed.ncbi.nlm.nih.gov/21881964/
Utzschneider, D. T., Alfei, F., Roelli, P., Barras, D., Chennupati, V., Darbre, S., Delorenzi, M., Pinschewer, D. D., & Zehn, D. (2016). High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival. The Journal of experimental medicine, 213(9), 1819–1834. https://doi.org/10.1084/jem.20150598
Uversky, V. N., Redwan, E. M., Makis, W., & Rubio-Casillas, A. (2023). IgG4 Antibodies Induced by Repeated Vaccination May Generate Immune Tolerance to the SARS-CoV-2 Spike Protein. Vaccines, 11(5), 991. https://doi.org/10.3390/vaccines11050991
Valk, A. M., Keijser, J. B. D., van Dam, K. P. J., Stalman, E. W., Wieske, L., Steenhuis, M., Kummer, L. Y. L., Spuls, P. I., Bekkenk, M. W., Musters, A. H., Post, N. F., Bosma, A. L., Horváth, B., Hijnen, D. J., Schreurs, C. R. G., van Kempen, Z. L. E., Killestein, J., Volkers, A. G., Tas, S. W., Boekel, L., … T2B! Immunity against SARS‐CoV‐2 study group (2024). Suppressed IgG4 class switching in dupilumab- and TNF inhibitor-treated patients after mRNA vaccination. Allergy, 79(7), 1952–1961. https://doi.org/10.1111/all.16089
van der Burg, S.H., Arens, R., Ossendorp, F., van Hall, T. and Melief, C.J.M., 2021. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nature Reviews Cancer, 21(8), pp.550–567. https://pubmed.ncbi.nlm.nih.gov/26965076/
van der Ploeg, E. K., Krabbendam, L., Vroman, H., van Nimwegen, M., de Bruijn, M. J. W., de Boer, G. M., Bergen, I. M., Kool, M., Tramper-Standers, G. A., Braunstahl, G. J., Huylebroeck, D., Hendriks, R. W., & Stadhouders, R. (2023). Type-2 CD8+ T-cell formation relies on interleukin-33 and is linked to asthma exacerbations. Nature communications, 14(1), 5137. https://doi.org/10.1038/s41467-023-40820-x
Wang, H., Xu, Q., Zhao, C., Zhu, Z., Zhu, X., Zhou, J., Zhang, S., Yang, T., Zhang, B., Li, J., Yan, M., Liu, R., Ma, C., Quan, Y., Zhang, Y., Zhang, W., Geng, Y., Chen, C., Chen, S., Liu, D., … Gu, J. (2020). An immune evasion mechanism with IgG4 playing an essential role in cancer and implication for immunotherapy. Journal for immunotherapy of cancer, 8(2), e000661. https://doi.org/10.1136/jitc-2020-000661
Wang, R., Chen, J., & Wei, G. W. (2021). Mechanisms of SARS-CoV-2 Evolution Revealing Vaccine-Resistant Mutations in Europe and America. The journal of physical chemistry letters, 12(49), 11850–11857. https://doi.org/10.1021/acs.jpclett.1c03380
Wegman, A. D., Fang, H., Rothman, A. L., Thomas, S. J., Endy, T. P., McCracken, M. K., Currier, J. R., Friberg, H., Gromowski, G. D., & Waickman, A. T. (2021). Monomeric IgA Antagonizes IgG-Mediated Enhancement of DENV Infection. Frontiers in immunology, 12, 777672. https://doi.org/10.3389/fimmu.2021.777672
Xu, L., Ma, Z., Li, Y., Pang, Z., & Xiao, S. (2021). Antibody dependent enhancement: Unavoidable problems in vaccine development. Advances in immunology, 151, 99–133. https://doi.org/10.1016/bs.ai.2021.08.003
Zhang, A., Stacey, H. D., Mullarkey, C. E., & Miller, M. S. (2019). Original Antigenic Sin: How First Exposure Shapes Lifelong Anti-Influenza Virus Immune Responses. Journal of immunology (Baltimore, Md. : 1950), 202(2), 335–340. https://doi.org/10.4049/jimmunol.1801149
Zhang P, Wu, Q, Xu X, Chen M, (2024). A case of IgG4-related ophthalmic disease after SARS-CoV-2 vaccination. Frontiers in Immunology, 15, p.1303589. https://pubmed.ncbi.nlm.nih.gov/38455056/
Zhu, J. and Paul, W.E., 2008. CD4 T cells: fates, functions, and faults. Blood, 112(5), pp.1557–1569. https://pubmed.ncbi.nlm.nih.gov/18725574/
Zhu, J., Yamane, H. and Paul, W.E., 2010a. Differentiation of effector CD4 T cell populations. Annual Review of Immunology, 28, pp.445–489. https://pubmed.ncbi.nlm.nih.gov/20192806/
Zhu, J. and Paul, W.E., 2010b. Heterogeneity and plasticity of T helper cells. Cell Research, 20(1), pp.4–12. https://pubmed.ncbi.nlm.nih.gov/20010916/
Zhu J. (2018). T Helper Cell Differentiation, Heterogeneity, and Plasticity. Cold Spring Harbor perspectives in biology, 10(10), a030338. https://doi.org/10.1101/cshperspect.a030338