Passive Immunity: Mechanisms, Applications, and Implications in Infectious Disease Management

The Intricacies of Passive Immunity: A Comprehensive Review

Many thanks to our sponsor Esdebe who helped us prepare this research report.

Abstract

Passive immunity, a fundamental pillar of immunological defence, involves the transfer of pre-formed antibodies from one individual to another, bestowing immediate but temporary protection against specific pathogens or toxins. This mechanism stands in stark contrast to active immunity, where the recipient’s own immune system is stimulated to generate a de novo antibody response following exposure to an antigen. The acquisition of passive immunity can occur naturally, most notably through the transplacental transfer of maternal antibodies to the foetus and post-natally via colostrum, or artificially, through the exogenous administration of antibody-containing biological products such as pooled immunoglobulins, hyperimmune globulins, or highly specific monoclonal antibodies. While celebrated for its capacity to confer rapid protection, a critical characteristic of passive immunity is its transient nature, necessitating repeated administrations for sustained defence. This comprehensive report meticulously explores the multifaceted mechanisms underpinning passive immunity, delves into its profound benefits and inherent limitations, and traces its historical evolution and diverse current applications in the prevention and treatment of a broad spectrum of infectious diseases. Furthermore, it examines the intricate interplay between passive and active immune strategies, elucidating the future trajectories and challenges in harnessing this powerful immunological tool.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

1. Introduction

Immunity against infectious diseases, a cornerstone of host defence, can be broadly categorised into two principal mechanisms: active immunity and passive immunity. Active immunity, a robust and enduring form of protection, arises when an individual’s immune system is directly challenged by a pathogen or its components, leading to the production of pathogen-specific antibodies and the establishment of immunological memory. This process can be elicited either through natural infection or through controlled exposure via vaccination. Conversely, passive immunity provides immediate protection by directly supplying the recipient with antibodies without engaging their own immune system in their production. This immediate availability of protective antibodies is its defining characteristic and primary advantage, making it an invaluable intervention in scenarios demanding urgent immunological defence. This report embarks on a detailed exploration of passive immunity, meticulously distinguishing it from active immunity, examining its varied modes of acquisition, dissecting its substantial benefits and inherent limitations, and analysing its extensive historical and contemporary applications in the sophisticated landscape of infectious disease management and prophylaxis.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2. Mechanisms of Passive Immunity

Passive immunity is conferred through two principal routes: naturally acquired and artificially acquired. Each route involves the transfer of pre-existing antibodies, circumventing the need for the recipient’s immune system to initiate an adaptive immune response.

2.1 Naturally Acquired Passive Immunity

Naturally acquired passive immunity represents the most ancient and fundamental form of immune protection, primarily occurring through the maternal transfer of antibodies to offspring. This biological imperative ensures that newborns, whose nascent immune systems are still developing, are equipped with immediate, albeit temporary, defence against a myriad of common pathogens encountered in their early extrauterine life.

2.1.1 Transplacental Transfer (In Utero)

The primary mechanism of naturally acquired passive immunity in humans is the transplacental transfer of maternal antibodies to the foetus during gestation. Among the various immunoglobulin classes, Immunoglobulin G (IgG) is uniquely adapted for this purpose. This selective transport is not a simple diffusion process but an active, receptor-mediated mechanism, predominantly occurring during the latter half of pregnancy, with peak efficiency in the third trimester (Schroeder & Cavacini, 2010). The key molecular player in this process is the neonatal Fc receptor (FcRn), also known as the Brambell receptor. FcRn is expressed on the syncytiotrophoblast cells of the placenta, specifically on the apical surface facing the maternal blood. Maternal IgG antibodies bind to FcRn at an acidic pH (around pH 6.0-6.5) within endosomal compartments. This binding protects IgG from lysosomal degradation and facilitates its transcytosis across the placental barrier into the foetal circulation. Once released into the foetal side, the higher physiological pH (around pH 7.4) of the foetal blood causes the dissociation of IgG from FcRn, allowing the receptor to recycle back to the maternal side (Roopenian & Akilesh, 2007).

This sophisticated transport mechanism ensures that the foetus acquires a broad spectrum of protective IgG antibodies, mirroring the maternal immunological experience. These maternal antibodies provide crucial protection against a range of common infectious diseases, including measles, rubella, tetanus, diphtheria, polio, and influenza, during the highly vulnerable early months of life when the infant’s own active immunity is still maturing (Orenstein et al., 2017). The specific IgG subclasses transferred vary, with IgG1 and IgG3 being most efficiently transported, followed by IgG4 and IgG2. The efficiency of transfer for IgG2 is notably lower due to its structural differences impacting FcRn binding (Mitchell & Tipton, 2020). The level of protection conferred is directly proportional to the concentration of specific antibodies in the maternal circulation, underscoring the importance of maternal vaccination during pregnancy.

However, this invaluable protection is inherently transient. Maternal IgG antibodies have a typical half-life of approximately 21-28 days in the infant. Consequently, their concentrations gradually decline over time, typically rendering the infant susceptible to infections against which they were previously protected by around 6-12 months of age, depending on the initial titre and the specific pathogen (Pollard & Greenwood, 2008).

2.1.2 Post-Natal Transfer (Breast Milk/Colostrum)

In addition to transplacental transfer, passive immunity is further augmented post-natally through breastfeeding, particularly via colostrum, the first milk produced after childbirth. While transplacental transfer primarily delivers IgG, breast milk is a rich source of other antibody isotypes, notably secretory Immunoglobulin A (sIgA), along with smaller amounts of IgG and IgM. sIgA is particularly important for mucosal immunity (Brandtzaeg, 2013). These antibodies are not absorbed systemically in significant amounts by the infant but instead provide localised protection within the gastrointestinal and respiratory tracts, forming a crucial first line of defence against pathogens encountered orally or nasally. This is especially vital given the immaturity of the infant’s gut barrier and systemic immune system in the initial weeks and months of life.

Lactoferrin, lysozyme, oligosaccharides, and immune cells (e.g., macrophages, lymphocytes) present in breast milk also contribute to the infant’s passive immune defence and overall development (Hanson & Korotkova, 2002). The degree and duration of protection from breast milk vary significantly depending on the feeding frequency, duration of breastfeeding, and the maternal antibody repertoire. For many mammals, particularly ungulates (e.g., cattle, horses), transfer of maternal antibodies (predominantly IgG) occurs almost exclusively via colostrum after birth, as their placentas do not permit significant prenatal antibody transfer. This phenomenon, known as ‘failure of passive transfer’, can be life-threatening for newborn animals if they do not receive sufficient colostrum within a critical window (Dwyer, 2008).

2.2 Artificially Acquired Passive Immunity

Artificially acquired passive immunity involves the intentional administration of pre-formed antibodies to an individual. This intervention is designed to provide immediate protection or therapeutic benefit in situations where the recipient’s immune system cannot mount a sufficiently rapid or effective response.

2.2.1 Convalescent Plasma and Hyperimmune Globulins

Historically, and still in certain contexts, artificially acquired passive immunity has relied on the use of blood products derived from humans or animals.

  • Convalescent Plasma/Serum: This involves collecting plasma or serum from individuals who have recently recovered from a specific infectious disease. Their plasma contains antibodies generated during their successful fight against the infection. This approach offers broad-spectrum polyclonal antibodies targeting various epitopes of the pathogen. Its utility was notably demonstrated in the late 19th and early 20th centuries against diseases like diphtheria, tetanus, and measles (Casadevall & Scharff, 1995). More recently, convalescent plasma saw renewed interest during the Ebola epidemic and the COVID-19 pandemic, particularly for early-stage treatment of infected individuals to prevent disease progression or as post-exposure prophylaxis (Bloch et al., 2020; Salazar et al., 2021).

  • Hyperimmune Globulins: These are preparations of antibodies obtained from plasma donors who have been hyperimmunised (e.g., vaccinated multiple times) against a specific pathogen, or from individuals with very high antibody titres due to recent infection. These products, such as Tetanus Immunoglobulin (TIG), Hepatitis B Immunoglobulin (HBIG), Rabies Immunoglobulin (RIG), and Varicella-Zoster Immunoglobulin (VZIG), are enriched for antibodies against a particular target. They offer a more concentrated and specific antibody preparation than general convalescent plasma, providing a potent and immediate neutralising effect (Keller & Stiehm, 2000). They are typically administered intramuscularly or intravenously.

2.2.2 Pooled Normal Immunoglobulins (Intravenous/Subcutaneous Immunoglobulin – IVIG/SCIG)

Normal immunoglobulin preparations, commonly referred to as Intravenous Immunoglobulin (IVIG) or Subcutaneous Immunoglobulin (SCIG), are highly purified sterile preparations of human plasma-derived IgG antibodies pooled from thousands of healthy donors. This pooling ensures a broad and representative spectrum of antibodies against common bacterial and viral pathogens prevalent in the general population. The manufacturing process involves sophisticated fractionation and purification techniques (e.g., Cohn fractionation, chromatographic methods) and multiple viral inactivation steps (e.g., solvent/detergent treatment, nanofiltration) to ensure product safety (Orange et al., 2006). IVIG is primarily administered intravenously, while SCIG offers the convenience of home administration.

IVIG/SCIG serves two main therapeutic purposes in infectious disease management:

  1. Replacement Therapy: For patients with primary or secondary immunodeficiencies (e.g., X-linked agammaglobulinemia, Common Variable Immunodeficiency – CVID) who are unable to produce sufficient quantities of their own antibodies. Regular administration of IVIG/SCIG prevents recurrent severe bacterial and viral infections (Bonilla et al., 2015).
  2. Immunomodulation: Beyond direct pathogen neutralisation, IVIG exerts significant immunomodulatory effects, making it a valuable therapy for a range of autoimmune and inflammatory conditions (e.g., Kawasaki disease, Guillain-Barré Syndrome, Idiopathic Thrombocytopenic Purpura) by mechanisms such as Fc receptor blockade, anti-idiotypic antibody activity, and modulation of complement activation (Nimmerjahn & Ravetch, 2008).

2.2.3 Monoclonal Antibodies (mAbs)

Monoclonal antibodies (mAbs) represent a revolutionary advancement in artificially acquired passive immunity. Unlike polyclonal antibody preparations (like plasma or IVIG) which contain a heterogeneous mixture of antibodies targeting multiple epitopes, mAbs are homogenous antibodies produced from a single B-cell clone. This means they recognise and bind to a single, specific epitope on a target antigen with high affinity and specificity.

  • Production: The initial technology for mAb production involved hybridoma technology, pioneered by Kohler and Milstein in 1975, where antibody-producing B lymphocytes are fused with immortal myeloma cells to create hybridoma cells that can produce specific antibodies indefinitely (Köhler & Milstein, 1975). Modern mAb production largely relies on recombinant DNA technology, allowing for the engineering of antibodies. These can be chimeric (mouse variable regions, human constant regions), humanised (mouse complementarity-determining regions on a human framework), or fully human (derived from transgenic mice with human immunoglobulin gene loci or phage display libraries), reducing the immunogenicity associated with animal-derived components (Lonberg, 2005).

  • Mechanisms of Action: mAbs employed in infectious disease management primarily act through several mechanisms:

    • Neutralisation: Directly binding to pathogens or their toxins, blocking their ability to infect host cells or exert pathological effects (e.g., blocking viral entry, neutralising bacterial toxins).
    • Opsonisation: Coating pathogens, making them more recognisable and palatable for phagocytic cells like macrophages and neutrophils, thereby enhancing their clearance.
    • Antibody-Dependent Cellular Cytotoxicity (ADCC): Engaging Fc receptors on immune effector cells (e.g., NK cells) to trigger the lysis of infected cells.
    • Complement-Dependent Cytotoxicity (CDC): Activating the complement cascade, leading to direct lysis of pathogens or infected cells.
  • Advantages: The precise specificity of mAbs minimises off-target effects, potentially leading to a better safety profile compared to polyclonal antibodies. Their consistent potency and purity are also significant advantages for standardisation and quality control (Nelson et al., 2010).

  • Applications: mAbs have revolutionised the treatment and prevention of numerous diseases, including infectious ones. Examples include palivizumab for RSV prophylaxis in high-risk infants, various mAbs (e.g., REGN-EB3, mAb114) for Ebola virus disease, and a range of mAbs developed for COVID-19 (e.g., casirivimab/imdevimab, sotrovimab, tixagevimab/cilgavimab) (Mahajan et al., 2012; Mulangu et al., 2019; Cohen et al., 2021).

2.2.4 Engineered Antibody Fragments and Bispecific Antibodies

Advanced antibody engineering has led to the development of smaller antibody fragments (e.g., Fab, scFv) and bispecific antibodies. Fragments offer advantages like better tissue penetration and reduced immunogenicity (by removing the Fc region), though often with shorter half-lives. Bispecific antibodies are engineered to bind to two different antigens or two different epitopes on the same antigen, offering novel therapeutic strategies, such as simultaneously targeting a pathogen and engaging an immune effector cell (Kasturi & Kastenmayer, 2021).

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3. Immunological Basis and Pharmacokinetics

Understanding the immunological basis and pharmacokinetic properties of antibodies is crucial for appreciating the efficacy and limitations of passive immunity.

3.1 Antibody Structure and Function

Antibodies, or immunoglobulins, are Y-shaped glycoproteins produced by plasma cells that serve as the primary mediators of humoral immunity. While different isotypes (IgG, IgA, IgM, IgD, IgE) exist, IgG is the most abundant and the primary mediator of systemic passive immunity due to its long half-life and ability to cross biological barriers.

An IgG molecule consists of two identical heavy chains and two identical light chains, linked together by disulfide bonds. Each chain comprises a variable (V) region and a constant (C) region. The variable regions form the antigen-binding site, providing exquisite specificity for a particular epitope. The ‘Y’ shape is formed by two Fab (Fragment antigen-binding) arms and one Fc (Fragment crystallisable) tail (Janeway et al., 2001).

  • Fab Domains: These arms contain the variable regions and are responsible for binding to specific antigens. Neutralisation of pathogens or toxins occurs primarily through the Fab domains, which can directly block viral entry into cells, inhibit bacterial adhesion, or neutralise toxin activity.
  • Fc Domain: The tail region (Fc) mediates the effector functions of the antibody. It binds to Fc receptors (FcRs) expressed on various immune cells (e.g., macrophages, neutrophils, NK cells) and can activate the complement system. These interactions facilitate opsonisation (marking pathogens for phagocytosis), Antibody-Dependent Cellular Cytotoxicity (ADCC), and Complement-Dependent Cytotoxicity (CDC), thereby enhancing pathogen clearance (Ravetch & Kinet, 1991).

In passive immunity, administered antibodies utilise these inherent functions to neutralise pathogens directly, facilitate their removal by host immune cells, or modulate immune responses.

3.2 Pharmacokinetics of Administered Antibodies

Pharmacokinetics describes how the body affects a drug, including absorption, distribution, metabolism, and excretion (ADME). For passively administered antibodies, particularly IgG, their pharmacokinetic profile is crucial for determining dosage, frequency, and duration of protection.

  • Absorption: The route of administration significantly influences absorption. Intravenous (IV) administration provides 100% bioavailability immediately. Intramuscular (IM) or subcutaneous (SC) injections result in slower absorption, typically reaching peak serum levels within hours to days, but can offer prolonged absorption (Sewell & Chapel, 2009).
  • Distribution: Antibodies initially distribute within the intravascular space. Over several days, they equilibrate between the intravascular and extravascular compartments, reaching target tissues and sites of infection. The distribution volume for IgG is approximately 0.1 L/kg (Vial et al., 2007).
  • Metabolism and Excretion: Unlike small molecule drugs, antibodies are primarily catabolised rather than excreted unchanged. The degradation of IgG is largely regulated by the neonatal Fc receptor (FcRn), as mentioned earlier in the context of transplacental transfer. FcRn is expressed on a wide range of cells throughout the body (e.g., endothelial cells, macrophages). It binds to IgG at acidic pH within endosomes, recycling it back to the cell surface and releasing it into the circulation at physiological pH. This mechanism prevents lysosomal degradation, effectively prolonging the IgG half-life. Antibodies that do not bind to FcRn are rapidly degraded. The typical serum half-life of IgG in healthy individuals is approximately 21-28 days, making it the longest-lived antibody isotype (Ghetie & Ward, 2002). Factors such as inflammation, infection, and individual variations in FcRn expression or IgG catabolism can influence this half-life.

The temporary nature of passive immunity is directly linked to the catabolic rate of the administered antibodies. Once the concentration of these exogenous antibodies falls below a protective threshold, the individual’s passive protection wanes, necessitating repeat administrations for sustained defence.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4. Benefits and Limitations of Passive Immunity

Passive immunity, while providing invaluable immediate protection, is not without its trade-offs. A thorough understanding of its advantages and disadvantages is crucial for its judicious application.

4.1 Benefits

Passive immunity offers several distinct advantages, making it a critical tool in various clinical scenarios:

  • Immediate Protection: The paramount benefit of passive immunity is its rapid onset of protection. Unlike active immunity, which can take days or weeks to develop a robust antibody response, passively administered antibodies are immediately available to neutralise pathogens or toxins. This is particularly beneficial in situations demanding urgent defence, such as post-exposure prophylaxis (e.g., after exposure to rabies, tetanus, or hepatitis B) or in the early stages of an aggressive infection (e.g., certain viral haemorrhagic fevers) (Plotkin & Orenstein, 2017).

  • Protection for Immunocompromised Individuals: For individuals whose immune systems are compromised and unable to mount an effective active immune response, passive immunity can be life-saving. This includes patients with primary immunodeficiencies (e.g., agammaglobulinemia, CVID), individuals undergoing immunosuppressive therapies (e.g., organ transplant recipients, cancer patients receiving chemotherapy), premature infants, or individuals with severe malnutrition. In these populations, passive antibody administration provides a crucial immunological shield against opportunistic and common infections that they would otherwise be highly susceptible to (Chapel et al., 2014).

  • Therapeutic Neutralisation of Toxins: Passive immunity is uniquely effective in neutralising bacterial toxins that cause diseases like diphtheria, tetanus, and botulism, or animal venoms (e.g., snakebite). In these cases, the administered antitoxin antibodies directly bind to and inactivate the circulating toxins, preventing them from causing further damage to host tissues. This is often a race against time, where active immunity would be too slow to be effective (WHO, 2017).

  • Bridge to Active Immunity: In some scenarios, passive immunity can serve as a bridge, providing temporary protection until an active immune response can be fully established. For instance, post-exposure prophylaxis for rabies often involves simultaneous administration of rabies immunoglobulin (passive) and rabies vaccine (active). The immunoglobulin provides immediate protection, while the vaccine initiates a long-lasting active immune response (WHO, 2018).

  • Protection for Foetuses and Neonates: Naturally acquired passive immunity, through transplacental transfer and breastfeeding, provides essential protection to newborns during a critical period of immunological vulnerability. This maternal antibody transfer acts as the infant’s primary line of defence against common pathogens during the early months of life.

4.2 Limitations

Despite its significant benefits, passive immunity also possesses notable limitations that dictate its appropriate application:

  • Temporary Nature: The most significant limitation is the transient nature of the protection. Exogenously administered antibodies, primarily IgG, are subject to normal physiological catabolism and are not continuously produced by the recipient. As discussed, their half-life is typically 3-4 weeks. Once these antibodies degrade below a protective threshold, the individual loses their passive protection. This necessitates repeated administrations for sustained defence, which can be costly and inconvenient (Roopenian & Akilesh, 2007).

  • Lack of Immunological Memory: Passive immunity does not induce immunological memory. The recipient’s B and T lymphocytes are not activated to recognise the pathogen or antigen, meaning they do not develop a lasting adaptive immune response. Consequently, upon subsequent exposure to the same pathogen after the passively transferred antibodies have degraded, the individual remains susceptible to infection and will have to mount a primary immune response (or receive another dose of passive antibodies) (Plotkin & Orenstein, 2017).

  • Dosage and Efficacy Variability (for Polyclonal Products): For polyclonal antibody preparations (e.g., convalescent plasma, older hyperimmune globulins), there can be variability in the titre (concentration) of specific antibodies and their overall neutralising capacity. This variability can make it challenging to ensure a consistent and effective dose across different batches. While modern manufacturing processes for IVIG and specific hyperimmune globulins have significantly improved standardisation, the inherent heterogeneity remains a consideration (Orange et al., 2006).

  • Safety Concerns:

    • Hypersensitivity Reactions: A historical concern, particularly with early animal-derived antitoxins (e.g., horse serum), was the risk of immediate hypersensitivity reactions (anaphylaxis) or delayed reactions like serum sickness. Modern human-derived immunoglobulins and humanised/fully human monoclonal antibodies have significantly reduced this risk, but allergic reactions can still occur, especially with rapid infusion (Chipps & Gelfand, 2007).
    • Transmission of Infectious Agents: While stringent donor screening, viral inactivation, and purification steps have made modern plasma-derived products extremely safe in terms of pathogen transmission (e.g., HIV, Hepatitis B/C), the theoretical risk, particularly for emerging or yet-unknown pathogens, can never be entirely eliminated. Early preparations of blood products did lead to transmission of pathogens (e.g., hepatitis, HIV) before robust screening and inactivation methods were implemented (Kleinman et al., 2009).
    • Recipient Immune Suppression: High doses of passive antibodies, particularly IVIG, can potentially interfere with the effectiveness of live attenuated vaccines (e.g., measles, mumps, rubella, varicella) by neutralising the vaccine virus before it can replicate and elicit an active immune response. A waiting period is often recommended between IVIG administration and live vaccine administration (CDC, 2024).
    • Adverse Effects: Depending on the product and route, adverse effects can include infusion-related reactions (fever, chills, headache), kidney dysfunction (with high IVIG doses), and thrombotic events (rare, but more common in high-risk patients) (Gelfand, 2012).
  • Cost and Accessibility: The production of highly purified human-derived immunoglobulins and monoclonal antibodies is complex and expensive. This can limit their accessibility, especially in resource-limited settings, and pose a significant economic burden on healthcare systems.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5. Historical and Current Applications of Passive Immunity

Passive immunity boasts a rich history spanning over a century, evolving from rudimentary serum transfers to highly sophisticated monoclonal antibody therapies. Its applications have continuously expanded, addressing both established and emerging infectious threats.

5.1 Historical Milestones and Early Applications

The origins of passive immunity as a therapeutic intervention date back to the late 19th century, laying the groundwork for modern immunology.

5.1.1 Diphtheria and Tetanus Antitoxin

The seminal work of Emil von Behring and Shibasaburo Kitasato in 1890 marked a turning point. They demonstrated that serum from animals (initially guinea pigs, later horses) immunised against diphtheria or tetanus could neutralise the respective toxins and protect other animals from these diseases (von Behring & Kitasato, 1890). This discovery led to the rapid development of diphtheria antitoxin, which became the first effective treatment for diphtheria, dramatically reducing mortality rates. In 1894, Émile Roux and his colleagues further refined the production and demonstrated the clinical efficacy of diphtheria antitoxin in children, solidifying its status as a life-saving therapy (Roux et al., 1894). Similarly, tetanus antitoxin was developed to neutralise the potent neurotoxin produced by Clostridium tetani, offering crucial passive protection against tetanus, particularly post-exposure.

These early successes with animal-derived antitoxins, while revolutionary, were associated with side effects such as serum sickness and anaphylaxis due to the recipient’s immune response against the foreign animal proteins. This drove subsequent research towards human-derived antibody products.

5.1.2 Convalescent Serum in Early 20th Century Epidemics

Throughout the early to mid-20th century, convalescent serum therapy, primarily using serum from human survivors, was a common strategy for various infectious diseases before the widespread availability of vaccines and antimicrobial drugs.

  • Measles: Convalescent serum was widely used to prevent or attenuate measles in exposed susceptible individuals, particularly young children and those in crowded settings.
  • Poliomyelitis: In the pre-vaccine era, passive transfer of antibodies (often as immune globulin) was used to prevent paralytic polio during outbreaks, demonstrating its utility in mitigating highly contagious diseases.
  • Hepatitis A: Immune globulin was historically used for post-exposure prophylaxis against Hepatitis A virus infection.
  • 1918 Influenza Pandemic: There is evidence that convalescent plasma was used with some success during the devastating 1918 Spanish Flu pandemic, highlighting its role in crisis response (Luke et al., 2006).

These historical applications underscore the immediate utility of passive immunity in mitigating disease severity and spread during outbreaks, serving as a crucial bridge until more permanent solutions (like vaccines) became available or for individuals unable to mount their own immune response.

5.2 Current Applications (Deep Dive)

Contemporary applications of passive immunity leverage advanced biotechnologies to provide highly specific and safer antibody products.

5.2.1 Post-Exposure Prophylaxis (PEP)

Passive immunization remains a cornerstone of post-exposure prophylaxis for several serious infections, where immediate neutralisation is critical and vaccine-induced immunity would be too slow:

  • Rabies Immunoglobulin (RIG): Administered alongside the rabies vaccine after suspected exposure (e.g., animal bite). RIG provides immediate antibodies to neutralise the virus at the wound site and prevent its spread to the nervous system, while the vaccine induces long-lasting active immunity (WHO, 2018).
  • Tetanus Immunoglobulin (TIG): Used for individuals with tetanus-prone wounds who have uncertain or incomplete vaccination history. TIG provides immediate protection by neutralising circulating tetanus toxin (CDC, 2024).
  • Hepatitis B Immunoglobulin (HBIG): Administered to unvaccinated individuals after exposure to Hepatitis B virus (e.g., needlestick injury, sexual contact, or from an HBsAg-positive mother to her newborn) to prevent infection, often in conjunction with the Hepatitis B vaccine (CDC, 2023).
  • Varicella-Zoster Immunoglobulin (VZIG): Recommended for susceptible high-risk individuals (e.g., immunocompromised persons, pregnant women) after significant exposure to varicella-zoster virus (chickenpox) to prevent or attenuate severe disease (CDC, 2024).

5.2.2 Immunoglobulin Replacement and Modulation

  • Replacement Therapy for Primary and Secondary Immunodeficiencies: IVIG and SCIG are standard lifelong therapies for patients with congenital (primary) immunodeficiencies (e.g., X-linked agammaglobulinemia, Common Variable Immunodeficiency – CVID) who cannot produce their own functional antibodies. It also serves patients with acquired (secondary) immunodeficiencies caused by certain diseases (e.g., chronic lymphocytic leukaemia, multiple myeloma) or treatments (e.g., highly immunosuppressive drugs) (Bonilla et al., 2015).
  • Prevention of Opportunistic Infections in Immunocompromised Patients: Beyond replacement, IVIG is used to prevent specific infections in high-risk immunocompromised patients, such as Cytomegalovirus (CMV) in transplant recipients, Parvovirus B19 in patients with chronic anaemia, and Enterovirus infections in patients with B-cell defects (Keller & Stiehm, 2000).
  • Immunomodulatory Uses: IVIG is extensively used for its immunomodulatory effects in various autoimmune and inflammatory conditions, including Idiopathic Thrombocytopenic Purpura (ITP), Kawasaki disease, Guillain-Barré Syndrome (GBS), and Chronic Inflammatory Demyelinating Polyneuropathy (CIDP). In these contexts, IVIG’s mechanism is thought to involve Fc receptor blockade, anti-idiotypic antibody activity, and modulation of cytokine networks, among others (Nimmerjahn & Ravetch, 2008).

5.2.3 Monoclonal Antibodies (mAbs) in Infectious Disease Management

mAbs have emerged as powerful, highly targeted therapies and prophylactic agents for a growing number of infectious diseases:

  • Respiratory Syncytial Virus (RSV) Prophylaxis: Palivizumab (Synagis®), a humanised monoclonal antibody, is administered monthly during RSV season to prevent severe lower respiratory tract disease in high-risk infants, such as premature babies and those with chronic lung disease or congenital heart disease (American Academy of Pediatrics, 2022). It works by neutralising RSV, blocking its entry into host cells.

  • Ebola Virus Disease: During the 2018-2020 Ebola outbreak in the Democratic Republic of Congo, several investigational monoclonal antibody therapies, including mAb114 (Ansuvimab) and REGN-EB3 (Inmazeb), demonstrated remarkable efficacy in reducing mortality rates in infected patients (Mulangu et al., 2019). These mAbs target different epitopes on the Ebola virus glycoprotein, preventing viral entry and replication.

  • COVID-19: The COVID-19 pandemic spurred rapid development and deployment of various SARS-CoV-2 targeting monoclonal antibodies (e.g., bamlanivimab/etesevimab, casirivimab/imdevimab, sotrovimab). These mAbs were primarily used for early treatment of mild-to-moderate COVID-19 in high-risk individuals to prevent progression to severe disease, and for pre-exposure prophylaxis in immunocompromised individuals (e.g., tixagevimab/cilgavimab, Evusheld®) (Cao et al., 2021). However, their efficacy has been significantly impacted by the emergence of new SARS-CoV-2 variants that often escape antibody neutralisation, highlighting the dynamic challenge of viral evolution.

  • Clostridioides difficile Infection (CDI): Bezlotoxumab is a monoclonal antibody approved for preventing recurrence of CDI. It targets C. difficile toxin B, neutralising its harmful effects and reducing the risk of subsequent infections (Chabrol & Tellez, 2019).

  • HIV: Research is ongoing into the use of broadly neutralising antibodies (bNAbs) against HIV for both prevention (e.g., in high-risk individuals) and treatment (e.g., as part of an HIV cure strategy). These bNAbs can target conserved regions of the HIV envelope protein, overcoming the virus’s high genetic variability (Caskey et al., 2016).

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6. Passive Immunity in the Context of Active Immunity

While both active and passive immunity confer protection, they operate through fundamentally distinct mechanisms and offer complementary benefits. Understanding their interplay is crucial for designing comprehensive immunization strategies.

6.1 Fundamental Differences and Complementary Roles

  • Active Immunity: This form of immunity is elicited when the host’s immune system is directly exposed to an antigen (either through natural infection or vaccination). It involves the activation and clonal expansion of antigen-specific B and T lymphocytes, leading to the production of antibodies and effector T cells, and crucially, the generation of immunological memory cells (memory B cells and memory T cells). This memory ensures a rapid, robust, and long-lasting protective response upon subsequent exposure to the same pathogen, often preventing symptomatic disease. The protection from active immunity is typically long-lived, potentially lifelong, but takes time (days to weeks) to develop (Plotkin & Orenstein, 2017).

  • Passive Immunity: In contrast, passive immunity provides immediate, pre-formed antibodies directly to the recipient. It bypasses the need for the recipient’s immune system to mount its own response, thus providing instant protection. However, since the recipient’s immune cells are not engaged, no immunological memory is generated. The protection is therefore temporary, lasting only as long as the exogenously supplied antibodies persist in the circulation (typically weeks to a few months) before they are catabolised. Once these antibodies are cleared, the individual reverts to a susceptible state (CDC, 2024).

The two forms of immunity are not mutually exclusive but often play complementary roles. Passive immunity can provide immediate protection in high-risk situations (e.g., post-exposure to rabies), while active immunity (e.g., rabies vaccine) is simultaneously administered to confer durable, long-term protection. This ‘bridging’ function is a key aspect of passive immunization. For individuals who cannot mount an effective active immune response due to immunodeficiency, passive immunity serves as a continuous, indispensable protective shield.

6.2 Considerations for Co-Administration

When passive immunity (via antibody products) is administered, there are important considerations regarding the timing and potential interference with active immunization, particularly with live attenuated vaccines:

  • Interference with Live Vaccines: Exogenously administered antibodies can neutralise the attenuated viruses in live vaccines (e.g., Measles, Mumps, Rubella [MMR], Varicella). This neutralisation prevents the vaccine virus from replicating sufficiently to elicit a robust, long-lasting active immune response and memory. Therefore, a waiting period (often 3-11 months, depending on the dose of antibody product and the specific vaccine) is generally recommended between the administration of antibody-containing products (like IVIG or specific hyperimmune globulins) and live attenuated vaccines (WHO, 2010; CDC, 2024). This ensures that the passively transferred antibodies have sufficiently degraded, allowing the vaccine to induce an effective immune response. Inactivated vaccines are generally not affected by passively administered antibodies and can be given concurrently or at any time.

  • Timing for Optimal Protection: In critical post-exposure situations (e.g., rabies), the immediate life-saving benefit of passive immunity outweighs the potential, typically minor, interference with the active vaccine response. Here, both are given concurrently, and revaccination may be considered later if there is concern about vaccine efficacy. The decision to co-administer or sequence passive and active immunization always involves a careful risk-benefit assessment tailored to the specific pathogen, the individual’s immune status, and the urgency of protection.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

7. Challenges and Future Directions

Despite its significant achievements, the field of passive immunity faces ongoing challenges while simultaneously holding immense promise for future therapeutic advancements.

7.1 Development Challenges

  • Cost and Manufacturing Complexity: The production of highly purified human-derived polyclonal immunoglobulins and especially monoclonal antibodies is a complex, multi-step process that is inherently expensive. This complexity involves stringent donor screening, extensive purification, viral inactivation, and rigorous quality control for polyclonal products, and elaborate cell culture, purification, and engineering for mAbs. The high cost limits global accessibility, particularly in low-income countries, exacerbating health disparities (Ledford, 2019).

  • Rapid Pathogen Evolution and Resistance: Pathogens, particularly viruses, exhibit high rates of mutation, leading to antigenic drift and shift. This evolutionary pressure can rapidly render existing antibody therapies ineffective, as observed with SARS-CoV-2 variants escaping neutralisation by early COVID-19 mAbs. This necessitates continuous surveillance and rapid development of new, variant-specific antibodies, posing a significant challenge (Weisblum et al., 2020).

  • Broad-Spectrum Activity: While mAbs offer exquisite specificity, their narrow target range means a single mAb typically targets only one pathogen or a specific strain. Polyclonal products offer broader coverage but lack the precise targeting of mAbs. Developing truly broad-spectrum antibody therapies that can neutralise multiple strains or even different pathogens remains an ambitious goal.

  • Delivery Mechanisms: Most antibody therapies are administered intravenously or subcutaneously, which can be inconvenient and require trained personnel. Exploring alternative delivery methods, such as inhaled antibodies for respiratory infections or oral delivery for gut-specific pathogens, could improve accessibility and patient compliance.

7.2 Future Directions and Novel Technologies

The future of passive immunity is being shaped by groundbreaking research and technological innovations aimed at overcoming current limitations and expanding therapeutic possibilities:

  • Advanced Monoclonal Antibody Engineering:

    • Bispecific Antibodies: As mentioned, these antibodies can bind to two different targets simultaneously, offering novel mechanisms of action (e.g., targeting a pathogen and an immune cell simultaneously to enhance clearance).
    • Fc-Engineered Antibodies: Modifying the Fc region of antibodies can enhance or diminish effector functions (e.g., improve ADCC, prolong half-life by enhanced FcRn binding) or reduce immunogenicity.
    • Mini-antibodies/Fragments: Smaller antibody fragments (e.g., nanobodies derived from camelid antibodies) offer advantages in terms of better tissue penetration, ease of production, and reduced immunogenicity, despite their shorter half-lives (Muyldermans, 2013).
  • Computational Antibody Design and Artificial Intelligence (AI): AI and machine learning are increasingly used to predict optimal antibody sequences, design novel binding sites, and rapidly identify highly potent antibodies against emerging threats. This can significantly accelerate the drug discovery pipeline (Chakraborty et al., 2022).

  • In Vivo Antibody Production via Gene Therapy: Rather than administering pre-formed antibodies, a revolutionary approach involves delivering genetic material (e.g., using Adeno-Associated Virus – AAV vectors) encoding the desired antibody directly into the patient’s cells. These cells then act as ‘mini-factories’, continuously producing the therapeutic antibody within the body, potentially providing long-lasting protection with a single administration. This approach is being explored for HIV and influenza prevention (Bourgeois & Koup, 2021).

  • Combination Therapies: Combining multiple monoclonal antibodies with different binding specificities can provide broader protection and reduce the likelihood of resistance emergence. This strategy was successfully employed for Ebola virus disease and COVID-19. Furthermore, combining passive immunization with active vaccination strategies will remain a key approach, especially in outbreaks or for highly vulnerable populations.

  • Broadly Neutralising Antibodies (bNAbs): Significant research focuses on identifying and engineering bNAbs that can neutralise a wide range of viral strains or even different viruses within a family (e.g., universal influenza bNAbs, pan-coronavirus bNAbs). These would offer more durable protection against evolving pathogens (Kwong & Mascola, 2018).

  • Immunoinformatics and Personalized Medicine: Leveraging individual genetic and immunological profiles to predict susceptibility and optimise antibody therapy choices, moving towards more personalised passive immunization strategies.

7.3 Role in Pandemic Preparedness

The COVID-19 pandemic starkly highlighted the critical need for rapid and effective countermeasures against emerging infectious diseases. Passive immunization, particularly through convalescent plasma and rapidly deployable monoclonal antibodies, played a crucial role early in the pandemic, bridging the gap until vaccines became widely available. Future pandemic preparedness strategies will undoubtedly integrate robust capabilities for rapid identification, large-scale production, and equitable distribution of novel antibody therapies (WHO, 2020).

Many thanks to our sponsor Esdebe who helped us prepare this research report.

8. Conclusion

Passive immunity, in its natural and artificial manifestations, represents an indispensable component of the human defence against infectious diseases. Its unique ability to confer immediate protection fills a critical void where active immunity is either too slow, unattainable, or insufficient. From the life-saving maternal antibody transfer to the sophisticated engineering of modern monoclonal antibodies, the journey of passive immunization reflects over a century of scientific advancement and clinical application.

While its temporary nature and lack of immunological memory differentiate it from active immunity, these characteristics do not diminish its vital role. Instead, they define its specific utility: providing rapid, life-saving protection in acute exposures, bridging vulnerability periods in newborns and immunocompromised individuals, and neutralising potent toxins. The challenges of pathogen evolution, manufacturing complexity, and equitable access remain significant. However, the burgeoning field of antibody engineering, coupled with advancements in computational biology and gene therapy, promises a future where passive immunization therapies are even more precise, potent, accessible, and integrated into comprehensive public health strategies. Continued investment in research and development in this field is paramount to enhance the efficacy, safety, and global accessibility of passive immunization, ensuring its enduring contribution to human health and pandemic preparedness.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

References

  • American Academy of Pediatrics. (2022). Palivizumab for the Prevention of Hospitalization Due to Respiratory Syncytial Virus Infection. Pediatrics, 150(4), e2022059345.
  • Bloch, E. M., et al. (2020). Deployment of convalescent plasma for the treatment of COVID-19. Journal of Clinical Investigation, 130(6), 2757–2765.
  • Bonilla, F. A., et al. (2015). Practice parameters for the diagnosis and management of primary immunodeficiency. Journal of Allergy and Clinical Immunology, 136(5), 1186-1205.e1.
  • Bourgeois, A. R., & Koup, R. A. (2021). Passive Immunization and In Vivo Antibody Production for HIV Prevention. Current HIV/AIDS Reports, 18(6), 490-501.
  • Brandtzaeg, P. (2013). Secretory IgA: Designed for Immune Exclusion. Mucosal Immunology, 6(1), 1-23.
  • Cao, Y., et al. (2021). SARS-CoV-2 neutralizing antibody evasion from B.1.429 and B.1.351 variants. Nature, 593(7857), 108-112.
  • Casadevall, A., & Scharff, M. D. (1995). Return to the past: the case for antibody-based therapies in infectious diseases. Clinical Infectious Diseases, 21(1), 150-161.
  • Centers for Disease Control and Prevention. (2023). Hepatitis B FAQs for Health Professionals. Retrieved from https://www.cdc.gov/hepatitis/hbv/hbvfaq.htm
  • Centers for Disease Control and Prevention. (2024). Principles of Vaccination. In The Pink Book: Epidemiology and Prevention of Vaccine-Preventable Diseases. Retrieved from https://www.cdc.gov/pinkbook/hcp/table-of-contents/chapter-1-principles-of-vaccination.html
  • Centers for Disease Control and Prevention. (2024). Tetanus (Lockjaw). Retrieved from https://www.cdc.gov/tetanus/index.html
  • Centers for Disease Control and Prevention. (2024). Varicella (Chickenpox). Retrieved from https://www.cdc.gov/chickenpox/index.html
  • Chabrol, F., & Tellez, A. (2019). Bezlotoxumab for the prevention of Clostridioides difficile infection recurrence. Journal of Clinical Pharmacy and Therapeutics, 44(4), 519-526.
  • Chakraborty, S., et al. (2022). The role of artificial intelligence in antibody engineering and design. Frontiers in Immunology, 13, 977931.
  • Chapel, H. M., et al. (2014). Long-term safety and efficacy of subcutaneous immunoglobulin replacement therapy in primary antibody deficiency: a prospective, multi-national study. Journal of Clinical Immunology, 34(5), 582-593.
  • Chipps, B. E., & Gelfand, E. W. (2007). Immunoglobulin therapy: An update on pharmacokinetics, formulations, and adverse events. Clinical Immunology, 122(2), 110-120.
  • Cohen, M. S., et al. (2021). Monoclonal Antibodies for the Treatment of COVID-19. New England Journal of Medicine, 385(13), e50.
  • Dwyer, R. M. (2008). Failure of passive transfer. Veterinary Clinics of North America: Equine Practice, 24(2), 273-281.
  • Gelfand, E. W. (2012). Adverse reactions to intravenous immunoglobulin. Annals of Allergy, Asthma & Immunology, 109(5), 329-335.
  • Ghetie, V., & Ward, E. S. (2002). FcRn: the MHC class I-related receptor that is more than a toy. Immunity, 17(5), 677-681.
  • Hanson, L. Å., & Korotkova, M. (2002). The role of breastfeeding in the immunology of the newborn. Journal of Pediatric Gastroenterology and Nutrition, 34(Supplement 1), S3-S7.
  • Janeway, C. A., et al. (2001). Immunobiology: The Immune System in Health and Disease (5th ed.). Garland Science.
  • Kasturi, M., & Kastenmayer, R. J. (2021). Bispecific Antibodies: A Mini-Review. Antibodies, 10(1), 8.
  • Keller, M. A., & Stiehm, E. R. (2000). Passive Immunity in Prevention and Treatment of Infectious Diseases. Clinical Microbiology Reviews, 13(4), 602-614.
  • Kleinman, S., et al. (2009). The residual risk of transfusion-transmitted human immunodeficiency virus (HIV) and hepatitis C virus (HCV) in the United States, 2008. Transfusion, 49(12), 2496-2501.
  • Köhler, G., & Milstein, C. (1975). Continuous cultures of fused cells secreting antibody of predefined specificity. Nature, 256(5517), 495-497.
  • Kwong, P. D., & Mascola, J. R. (2018). Human antibodies that neutralize HIV-1. Annual Review of Biochemistry, 87, 853-880.
  • Ledford, H. (2019). Antibody success story fuels calls for new funding. Nature, 576(7786), 188-190.
  • Lonberg, N. (2005). Human antibodies from transgenic animals. Nature Biotechnology, 23(9), 1117-1125.
  • Luke, T. C., et al. (2006). The use of convalescent plasma during the 1918 influenza pandemic. Annals of Internal Medicine, 145(2), 126-128.
  • Mahajan, L., et al. (2012). Palivizumab: A review of its use in prophylaxis for severe respiratory syncytial virus infection. Paediatric Drugs, 14(1), 15-32.
  • Mitchell, C., & Tipton, C. (2020). The role of maternal IgG in neonates. Frontiers in Immunology, 11, 1948.
  • Mulangu, A., et al. (2019). A Randomized, Controlled Trial of Ebola Virus Disease Therapeutics. New England Journal of Medicine, 381(24), 2293-2303.
  • Muyldermans, S. (2013). Nanobodies: natural single-domain antibodies. Annual Review of Biochemistry, 82, 347-370.
  • Nelson, A. L., et al. (2010). Antibody engineering and therapeutics. Nature Biotechnology, 28(9), 977-983.
  • Nimmerjahn, F., & Ravetch, J. V. (2008). Anti-inflammatory actions of intravenous immunoglobulin. Annual Review of Immunology, 26, 513-533.
  • Orange, J. S., et al. (2006). Use of intravenous immunoglobulin in human disease: a review of evidence by the AAIUS Committee on Primary Immunodeficiency Diseases. Journal of Allergy and Clinical Immunology, 117(4 Suppl), S525-S553.
  • Orenstein, W. A., et al. (2017). Maternal Immunization: A Review of the Current Challenges, Opportunities, and Future Needs. Human Vaccines & Immunotherapeutics, 13(10), 2268-2280.
  • Plotkin, S. A., & Orenstein, W. A. (Eds.). (2017). Plotkin’s Vaccines (7th ed.). Elsevier.
  • Pollard, A. J., & Greenwood, B. (2008). Clinical Infectious Diseases (3rd ed.). Saunders Elsevier.
  • Ravetch, J. V., & Kinet, J. P. (1991). Fc receptors. Annual Review of Immunology, 9, 457-492.
  • Roopenian, D. C., & Akilesh, S. (2007). FcRn: the neonatal Fc receptor comes of age. Nature Reviews Immunology, 7(9), 715-725.
  • Roux, É., et al. (1894). Contribution à l’étude de la diphtérie. Annales de l’Institut Pasteur, 8, 25-80.
  • Salazar, E., et al. (2021). Convalescent plasma for COVID-19: A randomized, controlled trial. Journal of Clinical Investigation, 131(1), e144005.
  • Schroeder, H. W. Jr., & Cavacini, L. (2010). Structure and function of immunoglobulins. Journal of Allergy and Clinical Immunology, 125(2 Suppl 2), S41-S52.
  • Sewell, W. A., & Chapel, H. M. (2009). Intravenous and subcutaneous immunoglobulin therapy. British Medical Journal, 338, b1424.
  • Vial, T., et al. (2007). Pharmacokinetics of intravenous immunoglobulins: a reappraisal. Clinical Pharmacokinetics, 46(2), 99-113.
  • von Behring, E., & Kitasato, S. (1890). Ueber das Zustandekommen der Diphtherie-Immunität und der Tetanus-Immunität bei Thieren. Deutsche Medizinische Wochenschrift, 16(49), 1113-1114.
  • Weisblum, Y., et al. (2020). Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. eLife, 9, e61312.
  • World Health Organization. (2010). WHO position paper on measles vaccines. Weekly Epidemiological Record, 85(35), 349-360.
  • World Health Organization. (2017). WHO Guidelines for the Management of Snakebites (2nd ed.). WHO Press.
  • World Health Organization. (2018). WHO Expert Consultation on Rabies: Third report. WHO Technical Report Series, No. 1012. WHO Press.
  • World Health Organization. (2020). Therapeutics and COVID-19: living guideline, 2020. Retrieved from https://www.who.int/publications/i/item/WHO-2019-nCoV-therapeutics-2020.2

4 Comments

  1. So, passive immunity is like borrowing someone else’s umbrella, eh? Great for the sudden downpour, but doesn’t teach you how to predict the weather yourself! Anyone else wondering if we can bio-engineer cuter antibodies? Maybe little, weaponised pandas?

    • That’s a brilliant analogy! The umbrella analogy perfectly captures the essence of immediate but temporary protection. The idea of bio-engineering cuter antibodies is quite amusing! Perhaps we could focus on making them more adaptable to emerging variants, ensuring our “umbrellas” are always effective against the ever-changing weather! What traits would make the ultimate antibody?

      Editor: MedTechNews.Uk

      Thank you to our Sponsor Esdebe

  2. The report highlights the challenge of rapid pathogen evolution. Given the limitations of monoclonal antibodies in addressing emerging variants, could advancements in broadly neutralizing antibodies offer a more sustainable solution for passive immunity against infectious diseases?

    • That’s a crucial point! Broadly neutralizing antibodies are definitely an exciting avenue for development. Overcoming the challenge of pathogen evolution is key. Perhaps future research could focus on engineering antibodies with adaptable binding sites, or developing antibody cocktails that target multiple conserved regions of pathogens.

      Editor: MedTechNews.Uk

      Thank you to our Sponsor Esdebe

Leave a Reply to MedTechNews.Uk Cancel reply

Your email address will not be published.


*