Fact-checked by Grok 2 weeks ago

Immunomodulation

Immunomodulation is the therapeutic process of modifying the immune system's response, either by suppressing overactive immunity to reduce inflammation or by stimulating hypoactive immunity to enhance against pathogens and tumors, thereby restoring physiological balance and treating a range of diseases. This approach targets key components of both innate and adaptive immunity, including , T cells, B cells, natural killer cells, and macrophages, through mechanisms such as inhibition, blockade, or genetic editing. Immunosuppressive strategies often involve corticosteroids like dexamethasone or inhibitors such as to dampen proinflammatory signals like TNF-α and IL-6, while immunostimulatory methods utilize monoclonal antibodies, interferons, or mesenchymal stem cells (MSCs) to boost antitumor or antiviral activity. Historical developments trace back to the 1940s with early agents like sodium antimony gluconate for parasitic infections, evolving to modern biologics amid challenges like and . In clinical applications, immunomodulation is pivotal for managing autoimmune disorders, such as treated with TNF-α inhibitors like or with IFN-β, and for prevention using drugs like and mycophenolate. It also addresses infectious diseases, exemplified by targeting IL-6 in severe cases to mitigate storms, and enhances cancer therapies through CRISPR-edited T cells or CD40 agonists in ongoing trials. For conditions like or , MSCs offer regenerative potential by modulating macrophage polarization and reducing inflammation. Emerging strategies emphasize precision, with advancements in for targeted delivery in infections like and for selective , aiming to minimize side effects while improving efficacy in chronic inflammatory and neoplastic conditions.

Overview

Definition

Immunomodulation refers to the directed alteration of function to enhance, suppress, or regulate immune responses, encompassing both natural homeostatic processes and therapeutic interventions aimed at restoring balance in dysregulated immunity. This process involves modifying the activity of immune components to achieve therapeutic outcomes, such as attenuating excessive or bolstering defenses against pathogens, without completely eliminating immune capability. Central to immunomodulation are key immune cells, including T cells that orchestrate adaptive responses, B cells responsible for production, and macrophages that bridge innate and adaptive immunity through and . Cytokines play a pivotal role, with pro-inflammatory agents like (e.g., IL-1, IL-6) and interferons driving activation, while regulatory cytokines such as IL-10 promote suppression. Signaling pathways, notably the JAK-STAT pathway for cytokine-mediated and the NF-κB pathway for inflammatory gene regulation, integrate these signals to fine-tune immune activity. Unlike , which broadly inhibits immune function to prevent rejection or (e.g., via corticosteroids that dampen overall activity), or immunostimulation that solely amplifies responses (e.g., through microbial mimics), immunomodulation emphasizes balanced regulation to avoid extremes, targeting specific pathways for precise control. This distinction allows for tailored interventions that maintain protective immunity while addressing . The scope of immunomodulation spans physiological examples, such as pregnancy-induced tolerance where decidual macrophages and regulatory T cells express to suppress maternal anti-fetal responses, ensuring fetal survival despite paternal antigens. Similarly, vaccine adjuvants exemplify therapeutic modulation by activating innate receptors to enhance adaptive immunity, improving antigen-specific responses without systemic overactivation.

Types

Immunomodulation is broadly classified into three main types based on their impact on the : , which dampens overactive immune activity to prevent excessive or ; immunostimulation, which enhances weakened immune functions to combat or malignancies; and immune deviation, which redirects the nature of the , such as shifting from a pro-inflammatory Th1-dominated profile to an Th2-dominated one. These types can be further subdivided by their specificity and . Specific immunomodulation targets discrete immune pathways or components, such as agents that inhibit a single like TNF-α, allowing precise control over particular responses. In contrast, nonspecific immunomodulation exerts broad effects across multiple immune elements, as seen with corticosteroids that globally suppress . Regarding induction, active immunomodulation stimulates the host's own to generate a response, often through or signaling, whereas passive immunomodulation transfers pre-formed immune elements, such as exogenous antibodies, to confer immediate protection without endogenous activation. Classification criteria for immunomodulation types also encompass duration, target, and context. Duration differentiates acute immunomodulation, which provides short-term modulation to resolve immediate threats like acute , from chronic forms that sustain long-term adjustments for persistent conditions such as autoimmune disorders. Target criteria focus on whether modulation affects the , involving rapid, nonspecific defenses like macrophages and complement, or the , which relies on antigen-specific T and B cells for targeted responses. Context-based typing distinguishes physiological immunomodulation, which maintains in healthy states, from pathological applications that address disease-driven dysregulation. From an evolutionary standpoint, these immunomodulation types have developed to enhance survival by balancing immune vigilance against threats with self-tolerance to avoid auto-destruction. For instance, mechanisms, akin to a form of physiological or deviation, evolved in placental mammals to allow the maternal to accept the semi-allogeneic during development, preventing rejection while preserving defenses against pathogens—a process mediated by trophoblast-expressed molecules like and indoleamine 2,3-dioxygenase () that suppress T-cell activation at the feto-maternal interface. This evolutionary adaptation underscores how immunomodulation types prioritize and host preservation across generations.

Biological Mechanisms

Natural Processes

The body's immune system employs several endogenous mechanisms to maintain homeostasis and prevent excessive inflammation or autoimmunity through self-regulation. Central to these processes are regulatory T cells (Tregs), a subset of CD4+ T lymphocytes characterized by the expression of the transcription factor FOXP3, which is essential for their development and suppressive function. Tregs suppress effector immune responses by multiple means, including the upregulation of CTLA-4, which competes with CD28 for binding to CD80/CD86 on antigen-presenting cells, thereby inhibiting T cell activation and promoting anergy. Additionally, Tregs secrete anti-inflammatory cytokines such as IL-10, which dampens pro-inflammatory signaling in target cells like macrophages and dendritic cells, further enforcing peripheral tolerance. Cytokine networks play a pivotal role in balancing immune activation and suppression, with pro-inflammatory cytokines like IL-2 driving T cell proliferation and effector functions, while anti-inflammatory counterparts such as TGF-β induce tolerance by promoting Treg differentiation and inhibiting Th17 cell development. This dynamic equilibrium ensures that immune responses are appropriately scaled; for instance, TGF-β signaling through its receptors activates SMAD pathways that upregulate FOXP3 in naive T cells, fostering a tolerogenic environment. Disruptions in this balance, such as elevated IL-2 without sufficient TGF-β, can lead to unchecked inflammation, underscoring the network's role in fine-tuning responses to self-antigens. Certain anatomical sites exhibit immune privilege, where specialized barriers and local mechanisms actively suppress immune surveillance to protect vital tissues. In the eye, , and testis, expression of (FasL) on resident cells induces in infiltrating Fas-expressing lymphocytes, preventing inflammatory damage. Complement inhibition further contributes, as membrane-bound regulators like (DAF) and limit complement activation in these sites, reducing opsonization and lysis of self-tissues. For example, in the testis, Sertoli cells produce TGF-β and express FasL to create a tolerogenic niche, safeguarding from immune attack. Negative feedback loops provide additional layers of control, including activation-induced cell death (AICD), where repeated stimulation upregulates and FasL, triggering caspase-mediated to eliminate overactivated clones and maintain . Anergy induction complements this by rendering T cells unresponsive upon encounter without , often mediated by CTLA-4 signaling that halts IL-2 production. These mechanisms collectively prune excessive responses, as seen in AICD's role in resolving acute infections without chronic . Illustrative examples of these processes include oral in the gut, where commensal microbiota-derived antigens promote Treg expansion via TGF-β and IL-10, suppressing systemic responses to food and microbial antigens and preventing . Similarly, at the maternal-fetal interface, decidual Tregs and trophoblast-derived TGF-β establish , inhibiting maternal and T cell attacks on the semi-allogeneic through FasL expression and modulation. These site-specific regulations highlight how natural immunomodulation integrates cellular, molecular, and microbial elements for immune equilibrium.

Therapeutic Interventions

Therapeutic interventions in immunomodulation involve targeted strategies to externally regulate immune function, distinct from the body's intrinsic regulatory mechanisms. Pharmacological targets immune receptors to inhibit overactive pathways, such as through antagonists that prevent signaling cascades leading to excessive or . therapy leverages the administration of signaling molecules to either amplify or dampen immune responses, exploiting their role as orchestrators of cellular interactions within the . Cellular manipulation, exemplified by adoptive transfer techniques, entails the modification and reinfusion of immune cells to enhance their suppressive or stimulatory capabilities, thereby redirecting immune activity toward desired outcomes. These approaches often build on natural processes, such as enhancing (Treg) function to promote . Key techniques include strategies designed for immune deviation, such as tolerogenic vaccines that induce antigen-specific by promoting regulatory immune subsets rather than effector responses. Phototherapy employs controlled exposure to modulate immune cell activity, inducing in hyperactive cells or shifting profiles toward anti-inflammatory states. modulation through dietary interventions or alters gut microbial composition to influence systemic immunity, fostering a balanced microbial environment that supports immune via production and epithelial barrier reinforcement. These methods align with types of like suppression or deviation, aiming to recalibrate immune balance without broad . Delivery methods vary between , which affects widespread immune compartments for comprehensive modulation, and localized approaches, such as intra-articular injections, that confine effects to specific tissues to minimize off-target impacts. Timed dosing strategies synchronize interventions with circadian or inflammatory immune cycles, optimizing efficacy by aligning with peaks in immune cell trafficking or receptor expression; as of 2025, chronotherapy has shown promise in enhancing inhibitor outcomes in cancer by considering circadian rhythms. Integration with diagnostics enhances precision, using biomarkers such as profiles to monitor immune status and guide selection, ensuring therapies are tailored to individual inflammatory or tolerogenic needs. This biomarker-driven approach allows real-time adjustment, improving outcomes by correlating levels of pro- and cytokines with response patterns.

Immunomodulatory Agents

Pharmacological Agents

Pharmacological agents for immunomodulation primarily consist of small-molecule drugs that exert immunosuppressive or effects through targeted interference with immune , , or production. These agents are synthetic compounds designed to modulate the in a broad, non-specific manner, often as part of systemic therapeutic strategies for conditions involving dysregulated immunity. Key classes include corticosteroids, inhibitors, and antimetabolites, each acting on distinct pathways to suppress excessive immune activity. Corticosteroids, such as prednisone, are among the most widely used immunomodulatory agents, binding to glucocorticoid receptors to translocate into the nucleus and inhibit pro-inflammatory transcription factors like NF-κB, thereby reducing the expression of cytokines such as IL-1, IL-6, and TNF-α. This mechanism dampens T-cell activation and macrophage function, providing rapid anti-inflammatory effects. Calcineurin inhibitors, exemplified by cyclosporine, form complexes with cyclophilin to block the phosphatase activity of calcineurin, preventing dephosphorylation and nuclear translocation of NFAT, which in turn suppresses IL-2 gene transcription and T-cell proliferation. Antimetabolites like methotrexate inhibit dihydrofolate reductase, disrupting folate metabolism essential for DNA synthesis and cell division in rapidly proliferating lymphocytes, leading to reduced purine and pyrimidine production and subsequent immunosuppression. More targeted pharmacological agents include (JAK) inhibitors, such as , which competitively bind to the ATP-binding site of JAK enzymes, interrupting signaling through the JAK-STAT pathway and thereby attenuating pro-inflammatory responses driven by cytokines like IL-6 and IFN-γ. Similarly, like bind to FKBP12 to allosterically inhibit the mTORC1 complex, halting T-cell proliferation by blocking IL-2-induced signaling and promoting in immune cells. Non-steroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen, provide milder immunomodulation by inhibiting cyclooxygenase (COX) enzymes, particularly COX-2, to reduce prostaglandin synthesis that amplifies inflammation and immune cell recruitment. The of these agents influence their immune-modulating efficacy and require careful management to avoid toxicity. Corticosteroids like are well-absorbed orally with a of approximately 3-4 hours () and 2-4 hours (active metabolite prednisolone), exerting immune effects through genomic and non-genomic pathways, though their metabolism via can lead to interactions with inhibitors like , prolonging exposure. inhibitors such as cyclosporine exhibit variable oral (around 30%) due to extensive first-pass metabolism by in the gut and liver, with a of 8-10 hours, necessitating to maintain immunosuppressive levels without . Methotrexate has a triphasic (0.75, 2-3, and 8-10 hours), with renal excretion predominant, and its polyglutamated form accumulates in lymphocytes to sustain antifolate effects over weeks. JAK inhibitors like are rapidly absorbed with a of 3 hours and metabolized primarily by , allowing once- or twice-daily dosing for sustained blockade. mTOR inhibitors such as show nonlinear with a of 62 hours, also reliant on metabolism, which impacts dosing in hepatic impairment. The development of these agents traces back to mid-20th-century milestones, with corticosteroids like first introduced in the 1950s for treatment, dramatically alleviating symptoms in initial trials and earning the 1950 in Physiology or Medicine for their discoverers. Subsequent advancements in the 1980s brought inhibitors like cyclosporine, revolutionizing by enabling T-cell specific suppression, while methotrexate's immunosuppressive role evolved from its antineoplastic origins in the 1940s to routine use in autoimmune diseases by the 1960s. Targeted inhibitors like and emerged in the 1990s and 2000s, building on molecular insights into and pathways.

Biologic and Cellular Therapies

Biologic therapies encompass a range of engineered proteins designed to modulate immune responses with high specificity, including and fusion proteins. , such as rituximab, target specific on immune cells to induce targeted depletion or neutralization. Rituximab, a chimeric anti- , binds to the on the surface of B lymphocytes, leading to B-cell depletion primarily through (ADCC), (CDC), and direct induction. Similarly, , a chimeric against tumor necrosis factor-alpha (TNF-α), neutralizes soluble and membrane-bound TNF-α by with high , thereby inhibiting its pro-inflammatory signaling through TNF receptors. These antibodies achieve therapeutic effects by precisely interfering with immune pathways, minimizing off-target impacts compared to non-specific immunosuppressants. Fusion proteins represent another class of biologics that function as receptors to sequester . , a dimeric consisting of the extracellular domain of the human TNF receptor 2 (TNFR2) linked to the Fc portion of human IgG1, acts as a soluble receptor that binds and neutralizes both TNF-α and lymphotoxin-α (TNF-β), preventing their interaction with cell-surface receptors and thus dampening inflammatory cascades. This mechanism allows to broadly inhibit TNF-mediated immune responses in conditions like , with its dimeric structure enhancing binding for prolonged cytokine sequestration. Cellular therapies leverage modified cells to either stimulate or suppress immune functions, offering dynamic immunomodulation. Chimeric antigen receptor (CAR) T-cell therapy involves engineering patient-derived T cells to express synthetic CARs, which redirect them against specific tumor for enhanced cytotoxic activity. The CAR construct typically includes an extracellular (scFv) for antigen recognition, a , and intracellular signaling motifs (e.g., CD3ζ and costimulatory domains like or 4-1BB) that activate T-cell proliferation, release, and target cell upon antigen binding. In contrast, mesenchymal stem cells (MSCs) exert immunosuppressive effects primarily through , secreting factors such as , , and transforming growth factor-β that inhibit T-cell proliferation, promote regulatory T-cell expansion, and modulate polarization toward an anti-inflammatory phenotype. Production of these biologics and cellular therapies relies on advanced . Monoclonal antibodies are manufactured using technology, where antibody genes are cloned into expression vectors and transfected into host cells like Chinese hamster (CHO) cells for large-scale production in bioreactors, followed by purification via to yield high-purity therapeutics. For CAR-T cells, -Cas9 editing enables precise genomic modifications; the process involves isolating patient T cells, electroporating them with CRISPR ribonucleoprotein complexes to knock out endogenous genes (e.g., TCR or PD-1 for reduced alloreactivity and exhaustion), and inserting the CAR transgene via lentiviral transduction or CRISPR knock-in, culminating in expansion before reinfusion. The specificity of these agents is enhanced through techniques like and affinity maturation, which optimize target recognition. identifies the precise residues on an that interact with the antibody's , often using methods such as hydrogen-deuterium exchange or to refine binding interfaces. Affinity maturation, typically achieved via or yeast surface display libraries, involves iterative and selection to increase binding strength, as seen in variants where somatic hypermutation-like processes boost dissociation constants from micromolar to nanomolar ranges, ensuring selective immune modulation without . Checkpoint inhibitors exemplify the precision of biologic therapies in . Nivolumab, a fully IgG4 targeting programmed death-1 (PD-1), blocks the PD-1/ interaction that inhibits T-cell activation, thereby reinvigorating anti-tumor immunity; its binding kinetics feature a high (Kd ≈ 3 nM) to PD-1, with slow off-rates enabling sustained blockade on T cells. This targeted approach has transformed by harnessing endogenous immune surveillance, often in combination with other modalities for synergistic effects. As of 2024, subcutaneous formulations of nivolumab (Opdivo Qvantig) and (Tecentriq Hybreza) have been approved by the FDA, enabling faster administration outside traditional intravenous settings.

Clinical Applications

Autoimmune and Inflammatory Diseases

Immunomodulation plays a central role in managing autoimmune and inflammatory diseases, where dysregulated immune responses lead to self-tissue damage or chronic inflammation. By suppressing aberrant immune activity through targeted therapies, these approaches aim to restore balance, reduce symptoms, and achieve disease remission. Common strategies include disease-modifying antirheumatic drugs (DMARDs) and biologics that inhibit key immune pathways, such as signaling or B-cell function, thereby preventing ongoing . In rheumatoid arthritis (RA), methotrexate remains a cornerstone DMARD, inhibiting dihydrofolate reductase to suppress T-cell activation and inflammation. Clinical reviews indicate that methotrexate monotherapy achieves clinical remission in approximately 30-40% of early RA patients after 6-12 months, as measured by Disease Activity Score 28 (DAS28) criteria (DAS28 ≤ 2.6), though efficacy diminishes in inadequate responders, necessitating combination with biologics. For systemic lupus erythematosus (SLE), belimumab, a monoclonal antibody targeting B-lymphocyte stimulator (BLyS), reduces autoantibody production by inhibiting B-cell survival. Phase III trials demonstrated that belimumab plus standard therapy improved Systemic Lupus Erythematosus Responder Index (SRI) rates by 10-15% over placebo at 52 weeks, with sustained reductions in disease flares and steroid use.61354-2/abstract) Strategies for inducing often involve B-cell depletion, as seen with rituximab in , which targets to eliminate autoreactive B cells and promote regulatory immune responses. Fixed retreatment regimens with rituximab maintain long-term B-cell depletion in refractory patients, correlating with DAS28 remission in up to 50% of cases after 6-12 months, particularly when combined with . blockade addresses inflammatory cascades; for instance, , an IL-6 receptor inhibitor, neutralizes IL-6-driven pro-inflammatory signals in autoimmune conditions. In and systemic , induces remission in 40-60% of patients within 24 weeks, reducing levels and joint damage progression. Biologics like TNF inhibitors exemplify broader immunomodulatory agents used in these diseases, achieving DAS28 remission rates of 30-50% in biologic-naïve patients across meta-analyses. Personalized approaches enhance outcomes through HLA typing, which predicts drug responses and risks in autoimmune disorders; for example, specific HLA-DRB1 alleles associate with better efficacy or rituximab tolerance in cohorts. In (IBD), anti- therapies like selectively block α4β7 to prevent trafficking to gut tissues, sparing systemic immunity. The trials showed inducing clinical response in 40-50% of patients at week 6 and maintaining remission in 40% at 52 weeks, with similar efficacy in . Case studies highlight its utility in refractory IBD; for instance, a biologic-experienced patient achieved endoscopic remission after 14 weeks of , with sustained mucosal healing over 2 years despite prior anti-TNF failures, underscoring its gut-selective mechanism.

Cancer and Oncology

Immunomodulation in cancer and oncology primarily aims to enhance the body's anti-tumor immune response by overcoming mechanisms that tumors use to evade detection and destruction by the immune system. Key strategies include immune checkpoint inhibitors, which block inhibitory signals such as CTLA-4 and PD-1/PD-L1 pathways to unleash T-cell activity against tumors. For instance, ipilimumab, a monoclonal antibody targeting CTLA-4, has demonstrated objective response rates of 10-15% in patients with advanced melanoma in phase 3 trials, with approximately 20% of patients achieving long-term survival beyond three years. Additionally, oncolytic viruses represent another approach by selectively infecting and lysing tumor cells, thereby releasing tumor-associated antigens and damage-associated molecular patterns (DAMPs) that stimulate innate and adaptive immunity, converting "cold" tumors into immunogenic ones responsive to further therapies. Adoptive cell therapies, particularly tumor-infiltrating lymphocyte (TIL) therapy, involve harvesting TILs from a patient's tumor, expanding them using interleukin-2 (IL-2) for 2-3 weeks followed by a rapid expansion protocol with anti-CD3 and feeder cells, and reinfusing them after lymphodepleting to promote engraftment. In February 2024, the FDA approved lifileucel, an autologous TIL therapy, for unresectable or metastatic previously treated with PD-1 inhibitors and if BRAF V600 positive. Post-infusion persistence of TILs, often measured by detectable circulating TILs for months, correlates with durable objective responses in up to 30-50% of treated patients, with complete responses in approximately 5-20% in select studies, highlighting the importance of sustained antitumor activity. Combination therapies further amplify these effects; for example, PD-1 inhibitors like combined with platinum-based in the KEYNOTE-189 trial for non-small cell improved median overall survival from 10.6 months to 22.0 months ( 0.60, 95% CI 0.50-0.72) in the 5-year analysis, with benefits also seen in other trials such as KEYNOTE-966 (HR 0.83) for cancers. Biomarkers play a crucial role in patient selection for these immunomodulatory approaches. PD-L1 expression on tumor cells or immune infiltrates, assessed via immunohistochemistry, predicts higher response rates to PD-1/PD-L1 inhibitors, with patients exhibiting PD-L1 levels ≥1% showing objective response rates 10-20% greater than those with low expression in multiple solid tumors. Similarly, tumor mutational burden (TMB), quantified as mutations per megabase of DNA, serves as a proxy for neoantigen load, where high TMB (>10 mutations/Mb) is associated with improved outcomes to checkpoint inhibition across cancers like lung and melanoma, independent of PD-L1 status. Emerging immunomodulatory strategies include personalized neoantigen vaccines, which use tumor sequencing to identify patient-specific neoantigens for peptide-based , often combined with adjuvants or checkpoint inhibitors to elicit targeted T-cell responses. Clinical trials, such as those in , have reported neoantigen-specific T-cell induction in over 80% of vaccinated patients, with preliminary evidence of tumor regression in phase 1 studies up to 2025. These vaccines hold promise for addressing tumor heterogeneity and improving durability in immunotherapy-resistant settings.

Organ Transplantation and Infectious Diseases

In organ transplantation, immunomodulation plays a critical role in preventing allograft rejection by suppressing the recipient's to foreign tissues. Calcineurin inhibitors, such as , are cornerstone agents in maintenance regimens following , with target trough levels typically ranging from 5 to 15 ng/mL in the early posttransplant period to balance efficacy and toxicity. Induction therapy with basiliximab, a that blocks the interleukin-2 receptor (IL-2R) on activated T cells, is commonly administered to reduce the incidence of acute rejection episodes in the immediate posttransplant phase. Rejection in organ transplants manifests in distinct forms, with acute rejection primarily driven by T-cell mediated mechanisms involving direct and release, whereas chronic rejection is often antibody-mediated, characterized by progressive vascular and tissue damage from donor-specific antibodies. For ABO-incompatible kidney transplants, desensitization protocols are employed to mitigate hyperacute rejection risks, typically involving rituximab for B-cell depletion, to remove anti-ABO antibodies, and intravenous immunoglobulin to neutralize remaining antibodies, enabling successful engraftment in sensitized patients. In infectious diseases, immunomodulation enhances host defenses against pathogens through targeted immune enhancement. Vaccine adjuvants like AS01, used in the , stimulate innate immunity via agonism and saponin-based components, promoting the recruitment and activation of T follicular helper (Tfh) cells to boost production and long-term . Certain antivirals exhibit immunomodulatory properties; for instance, interferon-alpha therapies for chronic infection not only inhibit viral replication but also activate natural killer cells and enhance T-cell responses to facilitate viral clearance. A key challenge in transplantation is balancing to prevent rejection while mitigating infection risks, particularly opportunistic pathogens like (CMV). Posttransplant prophylaxis with , an oral , is routinely used alongside inhibitors to suppress CMV replication in at-risk recipients, reducing the incidence of symptomatic disease during periods of intensified . Clinical outcomes in reflect the efficacy of these immunomodulatory strategies, with one-year graft survival rates reaching approximately 90% in modern eras, further improved by (HLA) matching to minimize alloimmune responses—zero-mismatch grafts showing superior long-term durability compared to those with multiple mismatches.

Risks and Considerations

Adverse Effects

Immunomodulation therapies, while effective in managing immune-related disorders, carry significant risks of adverse effects due to their interference with immune . These effects can range from mild and reversible to severe and life-threatening, often necessitating careful selection and monitoring. The spectrum includes leading to heightened vulnerability to infections and malignancies, paradoxical immune overstimulation, autoimmune-like reactions, and chronic organ toxicities. Immunosuppression Risks
Immunosuppressive immunomodulatory agents increase susceptibility to opportunistic infections, such as (PJP) in solid organ transplant recipients, with incidence rates historically ranging from 5% to 15% in the pre-prophylaxis era. This risk is attributed to T-cell depletion and impaired pathogen clearance, particularly in the early post-transplant period. Additionally, chronic elevates the risk of malignancies, including (PTLD), with cumulative incidence of approximately 1% at five years in transplant patients.
Overstimulation Effects
Paradoxical immune activation can occur with immunostimulatory therapies, most notably (CRS) in chimeric antigen receptor T-cell (CAR-T) therapy. CRS is graded from I to IV based on symptom severity, with grade I involving mild fever and grade IV encompassing life-threatening and multiorgan failure; common symptoms include fever, , and . Incidence varies by patient population, affecting up to 70% of recipients overall, with severe (grade 3-4) cases occurring in 5-25% of patients treated with CAR-T cells.
Autoimmune-Like Reactions
Immunostimulants like checkpoint inhibitors can trigger immune-related adverse events (irAEs) resembling autoimmune conditions, such as and endocrinopathies. For instance, PD-1 inhibitors are associated with in 10-20% of patients, often presenting as or thyrotoxicosis due to autoimmune destruction of thyroid tissue. , another frequent irAE, manifests as and from immune-mediated mucosal , with incidence up to 10-15% in treated cohorts.
Long-Term Effects
Prolonged use of certain agents leads to organ-specific toxicities. Corticosteroids, commonly employed in immunomodulation, induce by suppressing activity and promoting , increasing fracture risk in up to 30-50% of long-term users. Calcineurin inhibitors, such as , cause in 76-94% of kidney transplant recipients, characterized by rising serum levels due to afferent arteriolar and tubular damage.
Meta-analyses of biologic immunomodulators report serious adverse event rates of 20-30% across indications like and , primarily driven by infections and infusion reactions. These risks underscore the need for balancing therapeutic benefits against potential harms in clinical decision-making. Risks of Emerging Therapies
Emerging immunomodulatory approaches, such as (MSC) therapies and CRISPR-edited T cells, introduce additional risks. MSCs may promote tumorigenesis or cause infusion-related reactions like fever and in up to 20% of cases, while CRISPR editing carries risks of off-target genetic modifications potentially leading to secondary malignancies or immune dysregulation.

Monitoring and Management

Effective of patients on immunomodulatory therapies requires systematic to identify adverse effects promptly and guide interventions. assessments, such as complete blood counts (), are essential for detecting , a common complication of immunosuppressive agents that heightens susceptibility; guidelines recommend weekly until stabilization in post-transplant settings. For cytomegalovirus () risk in transplant recipients, quantitative (PCR) assays on plasma enable early detection, with routine testing advised during the first year post-transplant to inform preemptive antiviral therapy. , including annual skin examinations and targeted or MRI scans, is recommended for malignancies like or skin cancers, particularly in solid organ transplant patients on long-term . Dose adjustments in immunomodulatory regimens rely on to balance efficacy and toxicity. levels in are routinely measured via high-performance liquid chromatography-tandem (HPLC-MS/MS) to maintain trough concentrations of 5-15 ng/mL, depending on the post-transplant phase, thereby minimizing and rejection risks. tapering follows structured protocols to restore hypothalamic-pituitary-adrenal axis function while preventing flares; a typical regimen reduces from 1 mg/kg/day to 5-10 mg/day over 4-8 weeks, with slower decrements thereafter based on clinical response. Supportive care strategies emphasize prophylaxis against opportunistic infections and targeted treatments for therapy-specific toxicities. Trimethoprim-sulfamethoxazole (TMP-SMX) at a single-strength daily dose serves as first-line prophylaxis for Pneumocystis jirovecii pneumonia in kidney transplant recipients, continued for 6-12 months or longer in high-risk cases to reduce incidence by over 90%. In managing cytokine release syndrome (CRS) from chimeric antigen receptor T-cell (CAR-T) therapies, tocilizumab at 8 mg/kg intravenously is administered for grade 2 or higher events, repeatable every 8 hours up to three doses, often combined with corticosteroids for refractory cases. Professional guidelines standardize these approaches across clinical contexts. The American Society of Transplantation () recommends serial monitoring and protocol biopsies for infectious complications in immunosuppression trials, integrating CMV PCR and assessments to optimize outcomes in organ recipients. For applications, immune-related adverse events (irAEs) are graded using the Common Terminology Criteria for Adverse Events (CTCAE) version 6.0, with algorithms directing hold, dose reduction, or discontinuation based on severity (e.g., grade 3 events prompt high-dose steroids). Patient education empowers adherence and early reporting of complications. Individuals are instructed to recognize signs like persistent fever, cough, or , and reaction symptoms such as chills, , dyspnea, or , prompting immediate medical contact to avert escalation. Pre-therapy against , pneumococcus, and is emphasized to enhance baseline immunity before immunomodulators impair responses, with updates ideally completed prior to treatment initiation.

History and Future Directions

Historical Development

The use of immunomodulatory substances dates back to ancient civilizations, where natural agents like were applied to wounds for their and immune-modulating properties, promoting by influencing local and . Sumerian texts from around 2100–2000 BC document honey's therapeutic role in wound care, a practice echoed in and for enhancing tissue repair through modulation and immune cell recruitment. A pivotal early milestone in targeted immunostimulation occurred in 1796 when developed the smallpox vaccine by inoculating a boy with material, demonstrating acquired immunity and laying the foundation for as a deliberate immune enhancement strategy. This breakthrough, published in 1798, marked the birth of by showing how controlled exposure could prevent severe disease without causing it. In the mid-20th century, the discovery of immunosuppressive agents transformed treatment for autoimmune conditions. In 1949, Philip S. Hench and colleagues at the administered to patients with (RA), achieving dramatic symptom relief by suppressing excessive immune responses, which earned Hench, Edward C. Kendall, and Tadeus Reichstein the 1950 in Physiology or Medicine. This introduction of glucocorticoids established pharmacological as a cornerstone for managing inflammatory diseases. The field advanced significantly in transplantation medicine during the 1960s, driven by foundational work on . Peter B. Medawar's experiments in the 1940s and 1950s demonstrated acquired immunological tolerance in fetal and neonatal animals, proving that the could be trained to accept foreign tissues without rejection; this earned Medawar and Frank Macfarlane Burnet the 1960 . Concurrently, , an derived from 6-mercaptopurine, was introduced in 1961 for clinical use in organ transplants, enabling the first successful unrelated donor kidney grafts when combined with corticosteroids. The brought a revolution in with cyclosporine, a inhibitor isolated from fungi and approved in 1983, which selectively blocked T-cell to prevent graft rejection. Prior to cyclosporine, one-year kidney graft survival rates hovered around 50–60%; post-introduction, they improved to approximately 80%, dramatically expanding solid viability. The advent of biologic therapies in the 1990s targeted specific immune pathways with monoclonal antibodies. Tumor necrosis factor (TNF) inhibitors emerged as a breakthrough for RA; infliximab, the first such agent, received FDA approval in 1999 for use with methotrexate, reducing joint damage by neutralizing pro-inflammatory cytokines and transforming disease management. In the 2010s, cancer immunotherapy advanced through immune checkpoint inhibitors, which unleash T-cell responses against tumors. Ipilimumab, a CTLA-4 blocker, was approved by the FDA in 2011 for metastatic melanoma, marking the first such therapy to improve survival in advanced disease. This innovation built on foundational discoveries by James P. Allison and Tasuku Honjo, who elucidated inhibitory checkpoints like CTLA-4 and PD-1, earning them the 2018 Nobel Prize in Physiology or Medicine. These historical developments have evolved into contemporary approaches, such as chimeric receptor T-cell (CAR-T) therapies, which engineer immune cells for targeted tumor destruction. Recent advancements in mRNA , building on post-COVID-19 platforms, have expanded into immunomodulators for autoimmune diseases, with ongoing clinical trials from 2023 to 2025 demonstrating potential in enhancing immune responses while minimizing flares in conditions like systemic . For instance, trials involving mRNA vaccines in immunosuppressed s with autoimmune disorders have shown improved and safety profiles, paving the way for targeted immunomodulation therapies. Parallel developments in microbiome engineering, particularly fecal transplantation (FMT) for (IBD), have been supported by 2024 meta-analyses indicating efficacy in inducing remission, especially in , with clinical response rates up to 40% in randomized trials. These analyses highlight FMT's role in restoring microbial balance, reducing through enhanced gut and regulatory T-cell activity. Nanotherapies represent a promising frontier, with preclinical studies in 2025 focusing on systems for targeted delivery, such as interleukin-10, to modulate immune responses while reducing systemic associated with traditional therapies. These bioresponsive nanoparticles enable site-specific release in inflamed tissues, improving therapeutic precision in autoimmune and inflammatory models by limiting off-target effects and enhancing . In , CRISPR-Cas9 applications for enhancing regulatory T-cells (Tregs) in transplantation have advanced to phase II trials by 2024, aiming to promote and prevent graft rejection through targeted modifications that boost Treg suppressive function. Such edits, including HLA class I/II alterations, have shown prolonged Treg stability in preclinical transplant models, offering a foundation for personalized . The integration of (AI) into immunomodulation is transforming predictive modeling, with algorithms analyzing genomic and multi-omics data to forecast patient responses and optimize personalized dosing in therapies for cancer and autoimmune diseases. For example, AI-driven models have achieved over 80% accuracy in predicting biologic therapy outcomes in by incorporating genetic risk factors and clinical variables, enabling tailored dosing adjustments to maximize efficacy and minimize adverse events. Emerging trends also address environmental influences, as 2025 reports underscore how and exacerbate immune dysregulation, with increased exposure linked to heightened allergic responses and chronic inflammation, necessitating novel immunomodulatory strategies. These insights highlight the need for into pollution-mitigating interventions to bolster innate and adaptive immunity in vulnerable populations.

References

  1. [1]
    Immunomodulation - an overview | ScienceDirect Topics
    Immunomodulation is a therapeutic approach to modify the immune response, either reducing hyperactivated or enhancing hypoactive immune systems.
  2. [2]
    Immunomodulation—a general review of the current state-of-the-art ...
    Modulation of a non-adequate, amplified immune response enables to attenuate the clinical course of a disease and restore homeostasis. The potential targets to ...
  3. [3]
    Immunomodulation - an overview | ScienceDirect Topics
    Immunomodulation is defined as the regulation and modulation of immunity, which can involve either reducing or enhancing the immune response. AI generated ...
  4. [4]
    The JAK/STAT signaling pathway: from bench to clinic - Nature
    Nov 26, 2021 · The JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and ...
  5. [5]
    NF-κB and JAK/STAT Signaling Pathways as Crucial Regulators of ...
    These pathways include the activation of kinases, cyclic AMP (cAMP), and key signaling molecules such as MAP (microtubule-associated protein), nuclear factor κ ...
  6. [6]
    The potential of immunomodulators in shaping the future of healthcare
    Sep 3, 2024 · Immunomodulators play a crucial role in modern medical practice, offering both stimulatory and suppressive effects to regulate and control the immune response.
  7. [7]
    Immunologic Regulation in Pregnancy: From Mechanism to ... - NIH
    Macrophages present within the decidual immune cells during pregnancy have the potential to regulate divergent demands: maintenance of immune tolerance toward ...
  8. [8]
    Vaccine adjuvants: mechanisms and platforms - Nature
    Jul 19, 2023 · Adjuvants enhance the adaptive immunity of vaccines by activating innate immune cells. The core concept is that adjuvants promote the generation ...
  9. [9]
    Immunomodulating Drugs - an overview | ScienceDirect Topics
    Immunomodulation means all the therapeutic interventions used for modification of immune system. Augmentation of immune response is necessary for the ...
  10. [10]
    Epitope-specific immunotherapy induces immune deviation ... - PNAS
    The objective of this work was to induce immune deviation by mucosal peptide-specific immunotherapy in rheumatoid arthritis (RA) patients.Sign Up For Pnas Alerts · Methods · Results And Discussion
  11. [11]
    Immunity Types | Vaccines & Immunizations - CDC
    Jul 30, 2024 · Immunity to a disease is achieved through the presence of antibodies to that disease in a person's system. There are two types of immunity: active and passive.
  12. [12]
    Immunomodulation—a general review of the current state-of-the-art ...
    In brief, there are various potential targets in immunomodulatory intervention which efficiently modulate the immune response at its many stages.Missing: immunostimulation | Show results with:immunostimulation
  13. [13]
    In brief: The innate and adaptive immune systems - NCBI - NIH
    Aug 14, 2023 · If the innate (general) immune system fails to destroy the germs, the adaptive (specialized) immune system takes over. The adaptive immune ...
  14. [14]
    Innate and adaptive immunity: specificities and signaling hierarchies ...
    Dec 20, 2004 · In general, innate immunity is considered a nonspecific response, whereas the adaptive immune system is thought of as being very specific.
  15. [15]
    An introduction to immunology and immunopathology
    Sep 12, 2018 · Innate and adaptive immunity are not mutually exclusive mechanisms of host defense, but rather are complementary, with defects in either system ...
  16. [16]
    Tolerance of the fetus by the maternal immune system
    The adaptive immune system of placental mammals has evolved to tolerate the fetus. Rejection of the fetus by adaptive immune responses is therefore a rare ...
  17. [17]
    Evolutionary-developmental perspectives on immune system ...
    This review identifies newer evolutionary concepts and processes related particularly to the human placenta, innate and adaptive immune systems involved in ...
  18. [18]
    Evolution of the immune system in humans from infancy to old age
    Dec 22, 2015 · This article reviews the development of the immune response through neonatal, infant and adult life, including pregnancy, ending with the decline in old age.
  19. [19]
    Mechanisms of regulatory T-cell suppression – a diverse arsenal for ...
    Here we review recent advances in the identification of distinct mechanisms of Treg action and of signals that enable cellular targets to escape regulation.Missing: paper | Show results with:paper
  20. [20]
    Regulatory T cells in homeostasis and disease - Nature
    Oct 14, 2025 · Tregs not only suppress T cells but also modulate B cells, natural killer (NK) cells, and nonlymphoid cells, such as those in adipose tissue.
  21. [21]
    The Foxp3+ regulatory T cell: a jack of all trades, master of regulation
    In contrast, in the absence of one of the suppressive mechanisms (such as CTLA-4), Treg cells rely more on alternative suppressive functions such as IL-10 and ...Missing: paper | Show results with:paper
  22. [22]
    Regulation of the Immune Response by TGF-β - PubMed Central - NIH
    Abstract. Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses.
  23. [23]
    Intricacies of TGF-β signaling in Treg and Th17 cell biology - Nature
    May 23, 2023 · Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance.
  24. [24]
    Th17 and regulatory T cells: rebalancing pro- and anti-inflammatory ...
    Mar 5, 2009 · Clearly, the balance between Tregs and Th17 cells is dependent on the localized cytokine milieu including levels of IL-2, IL-6 and TGF-β, or by ...
  25. [25]
    Ocular immune privilege: a review - ScienceDirect.com
    The eye is an “immune privileged” site, such as the brain, the testes, the placenta and the fetus (Barker and Billingham, 1977), probably to provide ...
  26. [26]
    The role of Fas ligand in immune privilege - PubMed
    This idea has recently been challenged, and here we review the evidence for and against a role for FasL in immune privilege.Missing: brain testis complement inhibition
  27. [27]
    Complementing Testicular Immune Regulation - PubMed Central - NIH
    Overall, the complement inhibitors function to keep complement activation in check to prevent detrimental inflammation and overactivation of the immune system.
  28. [28]
    Activation-induced cell death in T cells - PubMed - NIH
    This review addresses the phenomenon of activation-induced cell death (AICD) in T lymphocytes, in which activation through the T-cell receptor results in apopto
  29. [29]
    Mechanism of activation-induced cell death of T cells and regulation ...
    AICD in T cells by Fas/FasL-mediated apoptosis is triggered by the switch from life to death through several signaling molecules. The control or distribution of ...
  30. [30]
    Activation-induced Cell Death in T Cells and Autoimmunity - PubMed
    Activation-induced cell death (AICD), which results from the interaction between Fas and Fas ligand, is responsible for maintaining tolerance to self-antigen.
  31. [31]
    Gut microbiota-immune cell interactions from infancy to old age
    This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age.
  32. [32]
    Multi-Layered Mechanisms of Immunological Tolerance at ... - PubMed
    Jul 15, 2024 · This review outlines the multi-layered processes that establish fetomaternal tolerance, including the physical barrier of the placenta.Missing: oral gut microbiome
  33. [33]
    Mechanisms of Fetal T Cell Tolerance and Immune Regulation
    Fetal T cells with pro-inflammatory potential are born in a tolerogenic environment and are tightly controlled by both cell-intrinsic and -extrinsic mechanisms.
  34. [34]
    The history and advances in cancer immunotherapy - Nature
    Jul 1, 2020 · Cytokine therapies​​ Functioning as messengers to orchestrate cellular interactions and communications of the immune system, cytokines are ...
  35. [35]
    Engineering synthetic vaccines using cues from natural immunity
    Oct 23, 2013 · In this Review, we will describe recent advances in the development of biomaterials that show promise for promoting immunity or tolerance in ...
  36. [36]
    Material aid for vaccines - Nature
    May 23, 2018 · In addition, tolerogenic vaccines are envisioned to deal with immune responses against biologic drugs, autoimmune disorders and transplant ...Missing: review | Show results with:review
  37. [37]
    A systematic framework for understanding the microbiome in human ...
    Sep 23, 2024 · 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and ...
  38. [38]
    Evolving understanding of autoimmune mechanisms and new ...
    Oct 4, 2024 · This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases.
  39. [39]
    Immune interventions in COVID-19: a matter of time? - Nature
    Oct 28, 2021 · We aim to review the available data on the kinetics of the immune response in beta-coronaviruses infections, from animal models and longitudinal human studies.
  40. [40]
    Dictionary of immune responses to cytokines at single-cell resolution
    Dec 6, 2023 · Cytokine-based therapies and cytokine antagonists are used to treat a wide range of disorders, including cancer and autoimmunity. However, the ...
  41. [41]
    Targeting cytokine and chemokine signaling pathways for cancer ...
    Jul 22, 2024 · In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer.
  42. [42]
    Rituximab: mechanism of action - PMC - PubMed Central - NIH
    The chimeric anti-CD20 antibody rituximab induces apoptosis in B-cell chronic lymphocytic leukemia cells through a p38 mitogen activated protein-kinase- ...
  43. [43]
    Molecular mechanism of action of anti-tumor necrosis factor ...
    In TNF bioassays, infliximab, etanercept, adalimumab, certolizumab pegol and golimumab all neutralize sTNF[23]. However, using the method of surface plasmon ...Missing: seminal | Show results with:seminal
  44. [44]
    Full article: Etanercept – TNF receptor and IgG1 Fc fusion protein
    Mar 10, 2014 · This drug inhibits the binding of soluble TNF to cell-surface TNF receptors, functioning as a decoy receptor. Like the soluble forms of TNF ...1. Tnf Cytokine · 2. Tnf Blockers · 3. 1 Etanercept Is Effective...
  45. [45]
    CAR-T cell therapy for cancer: current challenges and future directions
    Jul 4, 2025 · This review begins with a comprehensive overview of CAR-T cell therapy for cancer, covering the structure of CAR-T cells and the history of their clinical ...
  46. [46]
    The secretion profile of mesenchymal stem cells and potential ...
    Mar 21, 2022 · An increasing body of evidence indicates that MSCs exert their therapeutic effects largely through their paracrine actions.
  47. [47]
    Production and purification of recombinant monoclonal antibodies ...
    Dec 16, 2022 · Here, we present a protocol for generating and purifying recombinant antibodies from human HEK293 suspension culture cells from a primary sequence.
  48. [48]
    Engineering the next-generation of CAR T-cells with CRISPR-Cas9 ...
    Mar 18, 2022 · This review evaluates several of the ongoing and future directions of combining next-generation CRISPR-Cas9 gene editing with synthetic biology to optimize CAR ...
  49. [49]
    Epitope Specific Affinity Maturation Improved Stability of Potent ... - NIH
    This study developed a competitive dual color FACS to maturate a matrix-metalloprotease 14 (MMP-14) inhibitory antibody.
  50. [50]
    Nivolumab, anti-programmed death-1 (PD-1) monoclonal antibody ...
    Nivolumab (Opdivo), an IgG4 immunoglobulin, is a PD-1 binding immune checkpoint inhibitor that has shown activity against a wide spectrum of advanced cancers.
  51. [51]
    Efficacy and toxicity of methotrexate (MTX) monotherapy ... - NCBI
    The review evaluated the efficacy and toxicity of methotrexate monotherapy compared to methotrexate in combination with other non-biological disease-modifying ...
  52. [52]
    Efficacy of synthetic and biological DMARDs: a systematic ... - PubMed
    Nov 11, 2022 · The efficacy of csDMARDs+ short-term glucocorticoids in early RA was confirmed and similar to bDMARD+MTX combination therapy. Interleukin-6 ...
  53. [53]
    Methotrexate treatment strategies for rheumatoid arthritis: a scoping ...
    Mar 5, 2024 · This scoping review provides evidence in support of optimising MTX treatment in terms of route and dose prior to concluding that MTX treatment in RA patients ...
  54. [54]
    Low rates of remission with methotrexate monotherapy in ... - NIH
    Jul 27, 2019 · With combination therapy, methotrexate + bDMARD or tsDMARD in early RA (<1 year), high remission rates of up to 86% can be achieved (table 1A,1B) ...
  55. [55]
    A phase III, randomized, placebo-controlled study of belimumab, a ...
    Conclusion: Belimumab plus standard therapy significantly improved SRI response rate, reduced SLE disease activity and severe flares, and was generally well ...
  56. [56]
    Induction of long-term B-cell depletion in refractory rheumatoid ...
    Fixed retreatment with rituximab induces long-term B-cell depletion in different tissue compartments of refractory rheumatoid arthritis patients. RA patients ...Missing: tolerance | Show results with:tolerance
  57. [57]
    Translating IL-6 biology into effective treatments - Nature
    Apr 23, 2020 · Two subsequent trials of tocilizumab in >150 children with sJIA confirmed extensive improvements in the signs and symptoms of disease following ...
  58. [58]
    Anti-interleukin-6 receptor antibody, tocilizumab, for the treatment of ...
    Dec 1, 2011 · Clinical trials have demonstrated the efficacy of tocilizumab, a humanized anti-IL-6 receptor antibody, for patients with rheumatoid arthritis, Castleman's ...
  59. [59]
    Comparative Efficacy (DAS28 Remission) of Targeted Immune ... - NIH
    This NMA demonstrated that tocilizumab is associated with a greater likelihood of remission (DAS28 < 2.6) at 12 and 24 weeks compared with most other TIMs.
  60. [60]
    Pharmacogenetics: A strategy for personalized medicine for ...
    Nov 30, 2017 · The immune system is dynamic and complex, which may cause some difficulty in predicting drug responses. Pharmacogenetic evaluation before the ...
  61. [61]
    Anti-Integrins for the Treatment of Inflammatory Bowel Disease
    Aug 24, 2021 · Vedolizumab (anti-α4β7) is approved for the treatment of ulcerative colitis (UC) and CD. It is the most widely used anti-integrin therapy in IBD ...
  62. [62]
    Vedolizumab in the treatment of inflammatory bowel disease - NIH
    The GEMINI registrational trials established the efficacy of vedolizumab for the induction and maintenance of remission in both CD and UC, with the most ...
  63. [63]
    Improved Survival with Ipilimumab in Patients with Metastatic ...
    Jun 5, 2010 · In a randomized, phase 3 study involving patients with metastatic melanoma, a significant improvement in progression-free survival and response ...
  64. [64]
    Oncolytic viruses for cancer immunotherapy
    Jun 29, 2020 · In this review, we discuss the use of oncolytic viruses in cancer immunotherapy treatments in general, with a particular focus on adenoviruses.
  65. [65]
    Immune correlates and mechanisms of TIL therapy efficacy
    Aug 29, 2025 · The TIL therapy process involves tumor excision, culturing tumor fragments with interleukin-2 (IL-2) for 2–3 weeks, followed by a rapid ...
  66. [66]
    Phase III KEYNOTE-789 Study of Pemetrexed and Platinum With or ...
    Aug 22, 2024 · Specifically, in the phase III KEYNOTE-189 study, pembrolizumab plus chemotherapy significantly improved overall survival (OS; hazard ratio [HR] ...
  67. [67]
    a pooled analysis of KEYNOTE-966 and TOPAZ-1 trails - PMC - NIH
    Jun 10, 2025 · PIC is associated with improved OS and PFS in advanced BTC compared to chemotherapy alone, though the elevated risk of irAEs calls for careful monitoring.
  68. [68]
    Predictive biomarkers for PD-1/PD-L1 checkpoint inhibitor response ...
    Feb 23, 2023 · The combined high TMB and PD-L1 expression was the strongest predictor of durable response. PD-L1 was also a stronger predictor in patients ...
  69. [69]
    Assessment of Tumor Mutational Burden and Outcomes in Patients ...
    May 2, 2023 · This cohort study evaluates tumor mutational burden and outcomes in patients with diverse advanced cancers treated with immunotherapy.
  70. [70]
    A neoantigen vaccine generates antitumour immunity in renal cell ...
    Feb 5, 2025 · Personalized cancer vaccines (PCVs) can generate circulating immune responses against predicted neoantigens1,2,3,4,5,6.
  71. [71]
    Tacrolimus Trough Levels After Month 3 as a Predictor of Acute ...
    In general, TAC trough levels of 8–15 ng/mL were targeted in months 0–3 post-transplant and targets of 5–10 ng/mL in months 4–6, as per the center's protocol.
  72. [72]
    Rabbit Antithymocyte Globulin versus Basiliximab in Renal ...
    Nov 9, 2006 · Induction therapy reduces the frequency of acute rejection and delayed graft function after transplantation. A rabbit antithymocyte ...
  73. [73]
    Acute Transplantation Rejection - StatPearls - NCBI Bookshelf - NIH
    [1] T-cell and antibody-mediated rejection are the primary mechanisms leading to acute transplantation rejection.
  74. [74]
    Laboratory approaches in desensitization for ABO-incompatible ...
    Sep 5, 2025 · Desensitization strategies include rituximab, plasmapheresis, intravenous immunoglobulin, bortezomib, and emerging agents such as tocilizumab ...
  75. [75]
    Transient pain and long-term gain: adjuvant dose directs immune ...
    Apr 15, 2025 · ... AS01, a lipid-based carrier designed to integrate multiple immunostimulatory molecules. AS01, used in the herpes zoster (shingles) vaccine ...
  76. [76]
    Treatment of Hepatitis C Infections with Interferon - NIH
    Recombinant IFN-αs presently are widely employed with some success in the treatment of chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infections.
  77. [77]
    Challenges for the improvement of valganciclovir prophylaxis in ...
    Jun 18, 2024 · Valganciclovir prophylaxis has lowered the incidence of CMV infection and its complications in immunosuppressed solid organ transplant recipients.
  78. [78]
    Diminishing Significance of HLA Matching in Kidney Transplantation
    Pooled censored and uncensored graft survival rates were 90.6% and 89.9% at 1 year, 85.8% and 84.5% at 2 years, and 80.7% and 78.6% at 3 years. HLA matching ...
  79. [79]
    Pneumocystis Jirovecii Pneumonia in a Kidney Transplant Recipient ...
    Jan 25, 2021 · In pre-chemoprophylaxis era the over-all incidence of P. jirovecii infection among SOT recipients varied in the range of 5–15% [5]. After the ...
  80. [80]
    Incidence, screening, and management of de novo malignancies in ...
    Aggressive immunosuppression leads to stronger immune impairment and increases the risk of developing PTLD with cumulative incidence of PTLD over five years in ...<|separator|>
  81. [81]
    Grading of cytokine release syndrome associated with the CAR T ...
    Mar 2, 2018 · We show that the Penn grading scale provides reproducible CRS grading that can be useful to guide therapy and that can be applied across clinical trials and ...
  82. [82]
    Kinetics and biomarkers of severe cytokine release syndrome after ...
    Nov 23, 2017 · CRS developed in 70% of patients, including 62.5% with grade 1 to 3 CRS (grade 1, 26%; grade 2, 32%; grade 3, 4.5%), 3.8% with grade 4, and 3.8% ...
  83. [83]
    Endocrinopathies from checkpoint inhibitors: Incidence, outcomes ...
    May 1, 2023 · In particular, endocrinopathies occur in roughly 10% of patients who receive immune checkpoint inhibitors. Hypopituitarism, type 1 diabetes ...Missing: colitis | Show results with:colitis
  84. [84]
    Adverse effects of immune-checkpoint inhibitors - Nature
    May 15, 2019 · The most common irAEs among patients in the nivolumab group were endocrinopathies (mostly thyroiditis), pneumonitis, hepatitis, diarrhoea and ...
  85. [85]
    Glucocorticoid-induced osteoporosis - PMC - NIH
    Apr 8, 2015 · Corticosteroid-induced osteoporosis is the most common form of secondary osteoporosis and the first cause in young people.
  86. [86]
    Risk factors for calcineurin inhibitor nephrotoxicity after renal tran
    Feb 28, 2018 · Nephrotoxicity secondary to CNIs is reported to occur in 76%–94% of the renal transplant recipients. CNI nephrotoxicity clinically presented as ...<|separator|>
  87. [87]
    Adverse effects of biologics: a network meta‐analysis and Cochrane ...
    Since important risks such as lymphomas, serious infections and tuberculosis (TB) reactivation may be more common to the biologics but occur in small numbers ...
  88. [88]
    [PDF] Leukopenia Management Guidelines - University of Cincinnati
    Mar 1, 2014 · • Monitor CBC and differential weekly until it recovers or stabilizes and do not adjust medication doses. If ANC is < 1500 and dropping. • ...
  89. [89]
    Utility of CMV-Specific Immune Monitoring for the Management ... - NIH
    We review recently published studies that utilize CMV-CMI, at various points before and after transplantation, to help predict risk and guide the management of ...Missing: biomarker CBC
  90. [90]
    Cancer Screening and Cancer Treatment in Kidney Transplant ...
    Oct 31, 2024 · In this review, we discuss up-to-date data on pretransplant screening and cancer-free wait time, as well as post-transplant cancer screening, prevention ...
  91. [91]
    Therapeutic Drug Monitoring of Tacrolimus in Kidney Transplantation
    Jan 19, 2020 · TAC is a drug with a narrow therapeutic window and TDM is necessary to keep the level within a therapeutic range to minimize the adverse effects.
  92. [92]
    The Glucocorticoid Taper: A Primer for the Clinicians - PMC
    The practice of tapering GC therapy allows for recovery of the HPA axis while minimising the risk of a disease flare-up or symptoms of AI.
  93. [93]
    Safety and efficacy of prophylaxis for Pneumocystis jirovecii ...
    Apr 5, 2019 · Trimethoprim-sulfamethoxazole (TMP-SMX) is the drug of choice for anti-Pneumocystis jirovecii pneumonia (PcP) prophylaxis in kidney transplant recipients (KTR).
  94. [94]
    Management of Immune-Related Adverse Events in Patients ...
    Nov 1, 2021 · The guideline offers expert guidance on the diagnosis, evaluation, and management of CRS, ICANS, and other potential but less common ...
  95. [95]
    American Society of Transplantation recommendations for ... - PubMed
    American Society of Transplantation recommendations for screening, monitoring and reporting of infectious complications in immunosuppression trials in ...
  96. [96]
    CTEP Trial Development and Conduct - NCI
    Jun 25, 2025 · Common Terminology Criteria for Adverse Events (CTCAE)​​ The current version of CTCAE is CTCAE v5. 0, released in 2017. CTEP expects to release ...LOI and Concept Development · Protocol and Informed... · CTCAE and CTEP Codes
  97. [97]
    Infusion or Immune Reactions | American Cancer Society
    Jun 4, 2024 · Infusion reactions are symptoms or side effects that can happen if your immune system overreacts to cancer treatment given through an IV (intravenously).
  98. [98]
    Vaccinations in patients with immune-mediated inflammatory diseases
    Vaccination status is best checked and updated before the start of immunomodulatory therapy: live vaccines are not contraindicated at that time and inactivated ...Imid And The Directly Linked... · Types Of Vaccines · Vaccine Safety: Infection...
  99. [99]
    The Benefits of Honey for Wound Healing and Skin Health
    May 13, 2025 · Honey has healed wounds since ancient times, with Sumerians first recognising its therapeutic potential around 2100-2000 BC.
  100. [100]
    Honey, Wound Repair and Regenerative Medicine - PMC - NIH
    May 8, 2018 · Honey has been known since ancient times as an agent that is able to encourage the repair of wounds, guaranteeing healing with little or no scar ...
  101. [101]
    A Brief History of Vaccination - World Health Organization (WHO)
    Dr Edward Jenner created the world's first successful vaccine. He found out that people infected with cowpox were immune to smallpox. © Credits. In May 1796 ...Missing: immunomodulation | Show results with:immunomodulation
  102. [102]
    Edward Jenner and the history of smallpox and vaccination - NIH
    Edward Jenner (Figure 1) is well known around the world for his innovative contribution to immunization and the ultimate eradication of smallpox.
  103. [103]
    Philip S. Hench – Facts - NobelPrize.org
    These became the basis for cortisone preparations that, with input from Kendall and Philip Hench, were used at the end of the 1940s to treat rheumatoid ...
  104. [104]
    The discovery of cortisone: a personal memory - PMC - NIH
    Fifty years ago this week, Philip Hench showed that “compound E” (cortisone) was capable of reversing the inflammation of rheumatoid arthritis.
  105. [105]
    Peter Medawar – Nobel Lecture - NobelPrize.org
    “Immunological tolerance” may be described as a state of indifference or non-reactivity towards a substance that would normally be expected to excite an ...
  106. [106]
    The Nobel Prize in Physiology or Medicine 1960 - NobelPrize.org
    ... Medawar "for discovery of acquired immunological tolerance". To cite this section. MLA style: The Nobel Prize in Physiology or Medicine 1960. NobelPrize.org.
  107. [107]
    Azathioprine - Immunotherapy in Transplantation
    Apr 16, 2010 · The introduction of azathioprine (AZA) in 1961 revolutionized immunosuppressive management and created a framework for the evolution of ...
  108. [108]
    A Second Transplant Revolution : A new generation of anti-rejection ...
    Jun 13, 1990 · For kidney transplants throughout the United States during the same period, the one-year survival rate leaped from 60% to 80%. But liver ...
  109. [109]
    Remicade (infliximab) FDA Approval History - Drugs.com
    Remicade FDA Approval History · Aug 24, 1998 - approved for Crohn's disease · Nov 10, 1999 - approved in combination with methotrexate for rheumatoid arthritis ...Missing: inhibitors | Show results with:inhibitors
  110. [110]
    Infliximab - StatPearls - NCBI Bookshelf
    Mar 21, 2024 · The United States Food and Drug Administration approved infliximab in 1998 for the following indications (with doses mentioned):. Crohn ...
  111. [111]
    FDA Approves YERVOY™ (ipilimumab) for the Treatment of Patients ...
    Mar 25, 2011 · YERVOY, which is a recombinant, human monoclonal antibody, is the first FDA-approved cancer immunotherapy that blocks the cytotoxic T- ...Missing: checkpoint | Show results with:checkpoint
  112. [112]
    The 2018 Nobel Prize in Physiology or Medicine - Press release
    Oct 1, 2018 · Allison and Honjo showed how different strategies for inhibiting the brakes on the immune system can be used in the treatment of cancer. The ...
  113. [113]
    Progress and prospects of mRNA-based drugs in pre-clinical and ...
    Nov 14, 2024 · Table 5 Ongoing and completed clinical trials of mRNA vaccines for the treatment of immunosuppressant patients/patients with autoimmune diseases.
  114. [114]
    Safety and efficacy of fecal microbiota transplantation (FMT) as a ...
    A recent systematic review and meta-analysis study conducted by Chehade N et al. concluded that FMT is an effective therapeutic option for inducing clinical ...
  115. [115]
    Higher alpha diversity and Lactobacillus blooms are associated with ...
    Aug 6, 2024 · Data from several studies, including recent meta-analyses, have demonstrated the efficacy of FMT in some patients with ulcerative colitis (UC)5, ...Results · Host Microbiome Response · Methods
  116. [116]
    Immunomodulatory effects of metal nanoparticles: current trends and ...
    Apr 4, 2025 · Immunomodulatory NPs (e.g., calcium phosphate) facilitate targeted cytokine delivery, such as IL-10, to regulate immune responses.
  117. [117]
    Bioresponsive engineered nanoparticles for immunomodulation - PMC
    Oct 30, 2025 · These nanoformulations not only improve the durability of the particles but also play a crucial role in targeted drug delivery. Utilising redox- ...
  118. [118]
    HLA matching or CRISPR editing of HLA class I/II enables ... - NIH
    Oct 13, 2025 · Significant progress towards harnessing the therapeutic potential of Tregs to dampen pathological immune activity has been achieved with several ...
  119. [119]
    CRISPR-Cas9 engineering of human T regulatory cells
    Here we outline the development of a process for the production of SRC-3 KO human Tregs (hTregs), which can subsequently be adapted for Current Good ...
  120. [120]
    Machine learning precision medicine: The 2025 Revolution - Lifebit
    Oct 31, 2025 · For rheumatoid arthritis patients, AI models now predict response to biologic therapies by integrating clinical and genomic data. This ...
  121. [121]
    Unlocking precision medicine: clinical applications of integrating ...
    Feb 7, 2025 · This comprehensive review explores the clinical applications of AI-driven analytics in unlocking personalized insights for patients with autoimmune rheumatic ...Missing: immunomodulation | Show results with:immunomodulation
  122. [122]
    Climate change, air pollution, pollen and health
    Jul 17, 2025 · Air pollution and climate change together worsen the health impacts of allergens like pollen and airborne biological particles, leading to increased ...
  123. [123]
    Effects of Climate Change on the Immune System: A Narrative Review
    Apr 18, 2025 · Climate change, particularly increased temperatures, significantly impacts immune system function, leading to both heightened inflammatory responses, and ...