AMG-811 is an investigational human monoclonal antibody of the IgG1 isotype, developed by Amgen Inc..[1] It was engineered to selectively target and neutralize human interferon-gamma (IFNγ), a pivotal cytokine implicated in the complex pathogenesis of various autoimmune and inflammatory diseases.[2] IFNγ is a pleiotropic cytokine known for its potent pro-inflammatory and immunomodulatory functions, including the activation of critical immune cells such as B cells, T cells, and macrophages. Its dysregulation and overexpression have been documented in several autoimmune conditions, particularly in affected tissues, positioning IFNγ as a rational therapeutic target for intervention.[2] The development of AMG-811 represented an effort to modulate these IFNγ-driven pathological processes.
The selection of a human IgG1 isotype for AMG-811 is a deliberate choice in antibody engineering. Beyond simple neutralization of IFNγ, IgG1 antibodies possess effector functions such as Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) and Complement-Dependent Cytotoxicity (CDC). While the provided information emphasizes IFNγ neutralization as the primary mechanism [2], the potential for these effector functions could have implications for both desired therapeutic effects (e.g., depletion of IFNγ-producing cells) and undesired side effects. This aspect, though not explicitly detailed as a primary mechanism of action, is an inherent characteristic of the chosen antibody format.
The development of AMG-811 was ultimately discontinued during Phase 1.[1] This early-stage termination across multiple investigated indications suggests fundamental challenges were encountered. These challenges likely related to a consistent lack of convincing efficacy signals across different disease contexts or a safety/tolerability profile that, upon fuller evaluation or in specific subpopulations, was deemed non-viable for further development, even if initial, limited cohort reports described it as "well-tolerated."
Table 1: Summary of AMG-811 Basic Information
Feature | Details | Data Source(s) |
---|---|---|
Drug Name | AMG-811 | |
Drug Type | Monoclonal antibody | 1 |
Synonyms | AMG 811 | 1 |
Originator | Amgen, Inc. | 1 |
Target | Interferon-gamma (IFNγ) | 1 |
Mechanism of Action | IFNγ inhibitor | 1 |
Highest Development Phase | Discontinued Phase 1 | 1 |
Key Investigated (Inactive) Indications | Psoriasis, Discoid Lupus Erythematosus, Systemic Lupus Erythematosus +/- Glomerulonephritis (Lupus Nephritis) | 1 |
AMG-811 was designed to specifically target human Interferon-gamma (IFNγ).[1] IFNγ is a cytokine with a well-established, multifaceted role in the immune system, contributing to both host defense and, when dysregulated, to the pathology of autoimmune diseases. Its involvement in conditions like Discoid Lupus Erythematosus (DLE) and Systemic Lupus Erythematosus (SLE) is supported by evidence of elevated IFNγ messenger RNA (mRNA) and IFNγ receptor expression in affected DLE skin [2], and increased systemic levels of IFNγ or IFNγ-inducible markers (like CXCL10, neopterin, HLA class II) in SLE patients, which often correlate with disease activity and flares.[3] This provided a strong biological rationale for targeting IFNγ.
AMG-811 functions as a selective inhibitor of human IFNγ.[1] By binding with high affinity to IFNγ, it sterically hinders the cytokine from engaging with its cell surface receptors. This blockade prevents the initiation of downstream intracellular signaling cascades that would normally mediate IFNγ's diverse biological effects, including the activation of immune cells and the expression of numerous pro-inflammatory genes.[2]
The clinical trials for AMG-811 incorporated several biomarkers to assess target engagement and biological activity:
The consistent demonstration of pharmacodynamic activity, evidenced by reductions in IGBS and CXCL10 in SLE patients without LN and in DLE patients, confirms that AMG-811 successfully engaged its target, IFNγ, and modulated its downstream signaling pathways. However, the subsequent failure to achieve clinical efficacy (detailed in Section 4) creates a critical disconnect. This strongly implies that either IFNγ, while biologically active and modulated, is not the sole or dominant pathological driver in the clinical manifestations of these diseases; the extent or duration of IFNγ inhibition achieved was insufficient to overcome the disease process; or the IFNγ-driven biomarkers, while indicative of target engagement, are not robust surrogate endpoints for clinical improvement in these complex, heterogeneous autoimmune conditions.
The significantly diminished and non-sustained pharmacodynamic effect of AMG-811 in SLE patients with active lupus nephritis, even at higher doses and with potentially slightly higher systemic drug exposure [3], is a pivotal observation. This discrepancy suggests that factors within the inflamed renal microenvironment—such as very high local concentrations of IFNγ, impaired drug penetration into the kidney tissue, altered drug catabolism locally, or a fundamentally different role/regulation of the IFNγ pathway in LN—may limit the efficacy of systemically administered IFNγ inhibitors in this severe disease manifestation. This has major implications for treating organ-specific manifestations of systemic diseases with biologic therapies.
The development and utilization of sophisticated gene expression signatures, such as the IGBS and the keratinocyte IFNγ RNA score, as pharmacodynamic markers in these early-phase trials represents an advanced approach to assessing target engagement and biological response. Even though AMG-811 ultimately failed, the data generated from these molecular signatures provide valuable insights into the activity of the IFNγ pathway in human disease and its responsiveness to therapeutic intervention, contributing to the broader knowledge base for future drug development in autoimmunity.
AMG-811 was systematically evaluated by Amgen across a spectrum of autoimmune conditions where IFNγ was hypothesized to play a significant pathogenic role. These primarily encompassed Immune System Diseases, Skin and Musculoskeletal Diseases, and Urogenital Diseases.[1] Specific indications explored in clinical trials included:
The clinical development of AMG-811 did not progress beyond Phase 1. The program was ultimately discontinued.[1]
The strategic selection of Psoriasis, DLE, and SLE as initial target indications for an anti-IFNγ therapeutic agent is consistent with the prevailing understanding of IFNγ's role in T-cell mediated inflammation (relevant to psoriasis and DLE) and broader autoantibody-driven systemic autoimmunity (relevant to SLE). The consistent failure to demonstrate convincing efficacy across these mechanistically related but clinically distinct conditions suggests that IFNγ, while clearly implicated preclinically and by biomarker data, may not be a universally dominant or effectively "druggable" checkpoint for these diseases with AMG-811 as a monotherapy.
The discontinuation of the entire AMG-811 program during Phase 1, despite some positive pharmacodynamic biomarker responses, underscores the high attrition rates in early-phase clinical development, particularly for novel mechanisms in complex autoimmune diseases. It highlights the critical challenge of translating biomarker modulation into tangible, clinically meaningful patient outcomes.
Table 2: Overview of Key AMG-811 Clinical Trials
ClinicalTrials.gov ID | Phase | Indication(s) | Brief Study Design Summary | Key Stated Objectives (from sources) | Reported Status (Clarified) | Data Source(s) |
---|---|---|---|---|---|---|
NCT00818948 | Ib | SLE (with and without Glomerulonephritis/LN) | Randomized, double-blind, placebo-controlled, multiple ascending dose escalation | Evaluate safety, tolerability, PK, PD (CXCL10, IGBS, IL-18, MCP-1), preliminary clinical activity (SELENA-SLEDAI, proteinuria, anti-dsDNA, C3, C4) | Completed (Development discontinued due to lack of efficacy) | 1 |
NCT01164917 | I | Discoid Lupus Erythematosus (DLE) | Randomized, double-blind, placebo-controlled, 2-period crossover, single dose | Estimate effect on CLASI-A; secondary: PD biomarkers (IGBS, keratinocyte IFNγ score, CXCL10), CLASI-D, global assessments, SLEDAI-2K | Completed (Development for DLE terminated due to lack of benefit) | 1 |
NCT01510951 | 1 | Psoriasis (moderate to severe) | Randomized, double-blind, placebo-controlled, single-dose | Evaluate safety, tolerability, PK, PD, clinical effects | Completed (Development discontinued) | 1 |
This pivotal Phase Ib study (NCT00818948) was a multicenter, randomized, double-blind, placebo-controlled, multiple ascending dose escalation trial. It was structured with two distinct parts: Part A enrolled SLE subjects without active lupus nephritis (LN), while Part B focused on the more severe cohort of SLE subjects with active LN.[3] The primary objective was to evaluate the safety and tolerability profile of multiple subcutaneous doses of AMG 811. Secondary objectives were comprehensive, encompassing the assessment of pharmacokinetics (PK), a suite of pharmacodynamic (PD) markers (including serum concentrations of CXCL10, IL-18, MCP-1, and changes in the blood-based IFNγ Blockade Signature - IGBS), and preliminary evaluations of clinical activity using established instruments like SELENA-SLEDAI scores, 24-hour urine protein for LN patients, anti-dsDNA antibody titers, and C3 and C4 complement levels.[3]
AMG 811 (IgG1 anti-IFNγ mAb) or placebo was administered subcutaneously every four weeks for three doses. SLE subjects without LN received 6 mg, 20 mg, or 60 mg of AMG 811, while those with LN received higher doses of 20 mg, 60 mg, or 120 mg.[3]
Pharmacokinetics (PK) Profile:
Following subcutaneous administration, the median tmax ranged from 4.0 to 8.1 days.3 Minimal drug accumulation was observed with the q4w dosing. The mean terminal t1/2 after the third dose was 12 to 24 days.3 Exposure (Cmax and AUC) increased approximately dose-proportionally in SLE subjects without LN (6-60 mg). In subjects with LN, exposure increased slightly greater than dose-proportionally (20-120 mg).3 AMG 811 exposures appeared slightly higher (1.2 to 1.8-fold) in subjects with LN at the 60 mg dose compared to those without LN, but overall PK profiles were considered similar.3 Importantly, no subjects developed anti-AMG 811 antibodies.3
Efficacy Results (Clinical and Biomarker):
In SLE subjects without LN, AMG 811 treatment resulted in dose-related and sustained reductions in serum CXCL10 protein levels and the blood-based IGBS compared with placebo, persisting for over 85 days.3 However, in SLE subjects with LN, these biomarker reductions were notably less pronounced and were not sustained, even at the 120 mg dose.3 This differential PD response is a significant finding. AMG 811 did not produce significant effects on serum IL-18 or MCP-1 levels.3 The available information does not detail positive clinical efficacy outcomes (e.g., on SELENA-SLEDAI, proteinuria), and the drug's discontinuation, along with statements like "failed to establish benefit" 4, implies these were not compelling.
Safety and Tolerability:
Most subjects reported at least one TEAE, mostly grade 2 or 3.3 No meaningful imbalances in AE incidence were observed between AMG 811 and placebo groups overall.3
Serious Adverse Events (SAEs) were reported:
The DLE program centered on a Phase I, multicenter, randomized, double-blind, placebo-controlled, 2-period crossover study (NCT01164917).[2] The primary objective was to estimate the effect size of AMG 811 on the CLASI-A score. Secondary objectives included assessing changes in PD biomarkers (IGBS in serum/skin, keratinocyte IFNγ RNA score in skin, serum CXCL10), CLASI-D score, physician's and patient's global assessments, and SLEDAI-2K (for patients with underlying SLE).[2] A single subcutaneous dose of AMG 811 (180 mg) or placebo was administered in a crossover fashion.[2]
Efficacy Results:
No statistically significant difference was found in median changes in CLASI-A scores between AMG 811 and placebo over the first 85 days.2 No significant clinical benefits were observed in CLASI-D, physician's assessment, or patient's self-assessment of skin disease.2 Despite this lack of clinical efficacy, AMG 811 showed pharmacologic activity, reducing IFNγ-associated biomarkers (IGBS, CXCL10).2 This highlights the disconnect between target engagement and clinical outcome.
Safety and Tolerability:
AMG 811 was generally well tolerated.2 Common AEs (>1 patient in AMG 811 group) were arthralgia, headache, hypertriglyceridemia, and upper respiratory tract infections.2
Three patients (20%) in the AMG 811 group reported SAEs: one case of Legionella pneumonia (treatment-related) and another patient experienced gastroenteritis, migraine (treatment-related), splenic infarction, and viral gastroenteritis.2 No deaths or withdrawals due to AEs were reported.2
Reasons for Lack of Clinical Benefit/Discontinuation of Development for DLE:
Study authors suggested the single 180 mg SC dose might not have provided sufficient coverage or skin penetration.2 Other pathogenic pathways (e.g., type I IFN) may need concurrent inhibition, or the heterogeneity of DLE and its pathogenesis might explain the lack of efficacy from single pathway blockade.2
This Phase 1 study (NCT01510951) was a randomized, double-blind, placebo-controlled, single-dose trial designed to evaluate the safety, tolerability, PK, PD, and clinical effects of AMG 811 in subjects with moderate to severe psoriasis.[1] The trial was completed and conducted in Australia and New Zealand.[1] Detailed results regarding efficacy or specific safety findings are not available in the provided information beyond its completion and the drug's overall discontinuation. The absence of progression to later phases suggests it also failed to show compelling benefit in psoriasis.
A striking and consistent pattern emerges when comparing the SLE and DLE trial outcomes: AMG-811 reliably demonstrated pharmacodynamic effects on IFNγ-related biomarkers (IGBS, CXCL10) in both systemic circulation and, where measured, in target tissues like skin. However, this clear evidence of target engagement did not translate into clinically meaningful improvements in validated disease activity scores (SELENA-SLEDAI in SLE, CLASI in DLE). This recurring theme is the central narrative of AMG-811's clinical development and strongly suggests that IFNγ, while an active component of the inflammatory milieu in these diseases, may not be a critical, rate-limiting checkpoint whose blockade alone can reverse or significantly ameliorate established disease. The drug was performing its intended molecular action (target engagement), but if the disease itself did not improve, it implies that the target's role is either less critical than initially hypothesized, other pathways compensate for its blockade, or the disease pathology is too advanced or complex to be reversed by modulating this single cytokine. This is a common challenge when translating preclinical rationale to clinical success in multifaceted autoimmune diseases.
The observation of less pronounced and non-sustained biomarker modulation in SLE patients with active LN [3] compared to those without LN, despite similar or even slightly higher drug exposure, points towards tissue-specific factors or disease severity influencing drug efficacy at the target site. If systemic drug levels are adequate or even higher, but local target modulation is poor, it suggests a barrier at the tissue level. Lupus nephritis is characterized by intense local inflammation and immune complex deposition, which could create a challenging environment for antibody therapeutics to effectively engage their targets. This has implications for developing therapies for organ-specific manifestations of systemic autoimmune diseases.
The DLE trial's crossover design [2] with a single dose, while common for early PK/PD assessment, might have been insufficient to truly assess clinical efficacy for a chronic skin condition that may require sustained pathway modulation for visible improvement. The authors of the DLE study themselves suggest the single dose might not have provided adequate coverage.[2] Chronic inflammatory skin diseases often require weeks to months of continuous treatment to show clinical changes. A single dose, even if it modulates biomarkers transiently, may not be enough to alter the underlying disease course. This highlights a potential limitation in the DLE trial design for assessing clinical efficacy, though the consistent lack of efficacy in the multiple-dose SLE trial tempers this consideration.
There is a subtle but important distinction in how the DLE trial (NCT01164917) status is reported. One source lists it as "Terminated" [1], which could imply premature stoppage. However, detailed trial reports [2] indicate the study was completed as planned but did not show benefit, leading to the termination of further development for DLE. "Terminated trial" can imply premature stoppage due to safety or futility, whereas "completed trial with negative results leading to program termination" represents a different scenario, which appears to be the case here.
Across the Phase 1/1b studies in DLE and SLE, AMG 811 was generally described by investigators as well-tolerated.[2] This assessment is typical for early-phase trials with limited patient numbers and duration of exposure.
While generally well-tolerated, several SAEs were reported:
The characterization of AMG-811 as "well-tolerated" in these early-phase studies needs to be interpreted with caution. While there were no overwhelming acute toxicity signals leading to immediate trial halts, the emergence of SAEs like Legionella pneumonia (treatment-related in DLE) [2], PRES, and salmonella gastroenteritis (in the SLE with LN cohort) [3] are not trivial. In the absence of a strong efficacy signal, such SAEs would significantly skew the risk/benefit analysis negatively.
Table 3: Summary of Key Adverse Events Reported for AMG-811 (Across DLE and SLE Trials)
Adverse Event Category | Specific Event Type | Trial(s) Reported In (NCT ID) | Frequency/Severity Notes (if available) | Investigator-Assessed Relationship to AMG-811 | Data Source(s) |
---|---|---|---|---|---|
Common AEs | Arthralgia, Headache, Hypertriglyceridemia, Upper respiratory tract infections | DLE (NCT01164917) | Reported in >1 patient in AMG 811 group | Not specified for each | 2 |
Common AEs | General AEs | SLE (NCT00818948) | Majority Grade 2 or 3; no meaningful imbalances vs placebo | Not specified for each | 3 |
SAEs | Legionella pneumonia | DLE (NCT01164917) | 1 patient | Treatment-related | 2 |
SAEs | Gastroenteritis | DLE (NCT01164917) | 1 patient | Not specified | 2 |
SAEs | Migraine | DLE (NCT01164917) | 1 patient | Treatment-related | 2 |
SAEs | Splenic infarction | DLE (NCT01164917) | 1 patient | Not specified | 2 |
SAEs | Viral gastroenteritis | DLE (NCT01164917) | 1 patient | Not specified | 2 |
SAEs | Subarachnoid haemorrhage | SLE without LN (NCT00818948) | 1 patient in AMG 811 group | Not specified | 3 |
SAEs | Impetigo | SLE without LN (NCT00818948) | 1 patient in AMG 811 group | Not specified | 3 |
SAEs | Migraine | SLE without LN (NCT00818948) | 1 patient in AMG 811 group | Not specified | 3 |
SAEs | Neutropenia | SLE with LN (NCT00818948) | Reported in AMG 811 group | Not specified | 3 |
SAEs | Posterior Reversible Encephalopathy Syndrome (PRES) | SLE with LN (NCT00818948) | Reported in AMG 811 group; occurred in context of LN | Not specified | 3 |
SAEs | Salmonella gastroenteritis | SLE with LN (NCT00818948) | Reported in AMG 811 group; occurred in context of LN | Not specified | 3 |
SAEs | Worsening of Lupus Nephritis | SLE with LN (NCT00818948) | Reported in AMG 811 group | Not specified | 3 |
SAEs | Pancreatitis, Acute Renal Failure | SLE with LN (NCT00818948) | Reported in AMG 811 group | Not specified | 3 |
The primary and overriding reason for the discontinuation of the AMG-811 development program was its failure to establish clinically meaningful benefit or efficacy in the investigated indications of SLE and DLE.[2] This was despite clear evidence of pharmacodynamic activity (i.e., target engagement and modulation of IFNγ-related biomarkers). Drug development follows a logical progression: positive pharmacodynamic effects are a necessary but not sufficient condition for advancement. The absence of clinical efficacy, despite hitting the target, breaks this progression and typically leads to discontinuation unless a clear reason (e.g., incorrect dose, easily identifiable non-responsive patient population) can be found and addressed. The broad discontinuation of AMG-811 suggests this was not a simple fix.
Specific to DLE, contributing factors to the lack of efficacy were hypothesized to include potentially insufficient drug dosage or skin penetration from a single administration, the complex multifactorial nature of DLE pathogenesis where IFNγ blockade alone is inadequate, and the inherent heterogeneity of the disease.[2] The potential importance of other pathways, such as Type I IFN signaling, was also highlighted.[2] The LUPUS 2019 reference succinctly captures this by stating that "a preliminary study with anti-IFN-gamma mAb (AMG811) failed to establish benefit".[4]
The failure of AMG-811, despite a strong biological rationale and evidence of target engagement, underscores the complexity of autoimmune diseases and the challenge of translating preclinical findings and biomarker modulation into clinical success. It also highlights the importance of considering pathway redundancy and disease heterogeneity in therapeutic development. This outcome likely influenced subsequent research strategies for IFN pathway modulation in autoimmunity.
AMG-811, a human IgG1 monoclonal antibody developed by Amgen to selectively neutralize IFNγ, underwent early-phase clinical investigation for SLE (with and without nephritis), DLE, and Psoriasis.[1] Across these studies, AMG-811 consistently demonstrated target engagement, evidenced by modulation of IFNγ-related pharmacodynamic biomarkers like the IGBS and serum CXCL10.[2] However, this biological activity did not translate into clinically meaningful improvements in disease-specific outcome measures. Consequently, the development program for AMG-811 was discontinued at Phase 1.[1]
The key learning from the AMG-811 program is the significant disconnect between biomarker activity and clinical efficacy. This suggests that IFNγ blockade alone, as achieved by AMG-811, is insufficient to substantially alter the course of these complex autoimmune diseases. The failure highlights the intricate and often redundant nature of inflammatory pathways in conditions like DLE and SLE, where other mediators (e.g., Type I interferons) likely play crucial roles.[2] Differential pharmacodynamic responses in SLE patients with and without LN also point to disease heterogeneity and tissue-specific complexities in targeting IFNγ.[3] While generally termed "well-tolerated," some SAEs, particularly in vulnerable LN patients [3], would have contributed to a negative risk/benefit profile without efficacy.
The AMG-811 experience serves as a valuable case study in the challenges of translating preclinical rationale into clinical success in immunology. It underscores the need for robust preclinical models that better predict human responses and for adaptive clinical trial designs. While AMG-811 was discontinued for the indications studied, the data raise further questions: Could different dosing regimens or combination therapies have yielded different results? Are there other, perhaps more IFNγ-dominant, conditions where such an agent might be effective? The negative outcome of AMG-811, particularly when viewed alongside developments like the success of anifrolumab (targeting Type I IFN receptor) in SLE, likely contributed to a strategic recalibration in targeting IFN pathways, steering efforts towards mechanisms with a higher probability of clinical validation. Such failures, while disappointing for a specific drug, are crucial for advancing scientific understanding and redirecting research efforts.
Published at: May 12, 2025
This report is continuously updated as new research emerges.