Interferon beta-1b is a recombinant form of the naturally occurring human cytokine interferon beta, produced using genetically engineered Escherichia coli bacteria, and is primarily indicated for the treatment of relapsing forms of multiple sclerosis (MS), including clinically isolated syndrome, relapsing-remitting MS, and active secondary progressive MS, to reduce the frequency of clinical exacerbations and delay disability progression.[1][2][3] Approved by the U.S. Food and Drug Administration (FDA) in 1993 under the brand name Betaseron, it was the first disease-modifying therapy for MS and is marketed as Extavia since 2009, both administered as a subcutaneous injection every other day at a dose of 0.25 mg.[4][5]The precise mechanism of action of interferon beta-1b in MS remains incompletely understood, but it is believed to exert immunomodulatory effects by binding to specific cell surface receptors, thereby inducing the expression of genes that promote anti-inflammatory pathways, suppress proinflammatory cytokine production, reduce T-cell activation and migration across the blood-brain barrier, and inhibit matrix metalloproteinases involved in demyelination.[6][7][8] Clinical trials, such as the pivotal 1993 phase III study, demonstrated that it reduces the annualized relapse rate by approximately 30% and decreases MRI lesion activity, supporting its role in altering the disease course.[1][2]Common side effects include flu-like symptoms (fever, chills, fatigue), injection-site reactions (pain, redness, swelling), and lymphopenia, while more serious risks encompass hepatotoxicity, depression, anaphylaxis, pulmonary arterial hypertension, and increased infection susceptibility, necessitating regular monitoring of liver function, blood counts, and mental health.[3][1][9][10] As a biologic therapy, interferon beta-1b has been a cornerstone of MS management for over three decades, though its use has evolved with the advent of oral and higher-efficacy alternatives, and it is contraindicated in patients with hypersensitivity to human albumin or interferon beta.[1][11]
Pharmacology
Mechanism of action
Interferon beta-1b is a recombinant form of human interferon beta, a cytokine belonging to the type I interferon family, produced in Escherichia coli with a serine substitution at position 17 to enhance stability.[12] It exerts its effects by binding to the heterodimeric interferon-alpha/beta receptor (IFNAR) complex, composed of IFNAR1 and IFNAR2 subunits, on the surface of various cell types including immune cells and endothelial cells.[12] This binding initiates a conformational change in the receptor, recruiting and activating Janus kinase 1 (JAK1) and tyrosine kinase 2 (TYK2).[8]Upon receptor activation, the JAK-STAT signaling pathway is engaged, where phosphorylated STAT1 and STAT2 form a complex with interferon regulatory factor 9 (IRF9) to create interferon-stimulated gene factor 3 (ISGF3), which translocates to the nucleus and induces transcription of hundreds of interferon-stimulated genes (ISGs).[12] These ISGs encode proteins with antiviral, antiproliferative, and immunomodulatory functions, such as MxA (an antiviral GTPase) and PKR (a protein kinase that inhibits viral replication).[13] The pathway's activation leads to pleiotropic biological activities that modulate immune responses without directly targeting viral pathogens in the context of multiple sclerosis therapy.[7]Key immunomodulatory effects of interferon beta-1b include enhancement of suppressor T-cell (Treg) activity, which promotes immune tolerance by inhibiting autoreactive T cells, and reduction of pro-inflammatory cytokine production, such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), while promoting anti-inflammatory cytokines like interleukin-10 (IL-10).[13][12] It also downregulates major histocompatibility complex (MHC) class II expression on antigen-presenting cells, thereby decreasing antigen presentation to T cells and limiting pro-inflammatory activation.[12] Additionally, interferon beta-1b upregulates MHC class I proteins on target cells, facilitating cytotoxic T-cell recognition and clearance of infected or abnormal cells while avoiding excessive inflammation.[14]In the central nervous system, interferon beta-1b inhibits blood-brain barrier (BBB) disruption by reducing the activity of matrix metalloproteinases (MMPs), particularly MMP-9, which degrade extracellular matrix components and facilitate immune cell entry.[12] This reduction restores the balance with tissue inhibitors of metalloproteinases (TIMPs), stabilizing the BBB integrity.[12] Furthermore, it decreases the migration of activated T cells into the CNS by downregulating adhesion molecules on endothelial cells, such as vascular cell adhesion molecule-1 (VCAM-1), thereby limiting inflammatory infiltration.[13]
Pharmacokinetics
Interferon beta-1b is administered by subcutaneous injection as the standard route. Following subcutaneous administration, the drug exhibits incomplete absorption, resulting in a bioavailability of approximately 50% based on studies with doses up to 0.5 mg.[15] Peak serum concentrations are achieved within 1 to 8 hours post-injection, with mean peak levels around 40 IU/mL at higher doses in healthy volunteers; however, at the recommended therapeutic dose of 0.25 mg, serum concentrations are often low or undetectable due to rapid clearance and a short elimination half-life.[15] The pharmacokinetics demonstrate a biphasic elimination profile, with an initial alpha phase half-life of approximately 0.25 to 0.5 hours and a terminal beta phase half-life of 2 to 3 hours, though values up to 5 hours have been reported in some intravenous and subcutaneous studies.[15][16]The drug is primarily distributed to the extracellular fluid, reflected in a steady-state volume of distribution ranging from 0.25 L/kg to 2.88 L/kg following intravenous administration.[15] Interferon beta-1b does not significantly cross the blood-brain barrier but produces its therapeutic effects through systemic immunomodulation. Metabolism occurs via proteolytic degradation and receptor-mediated endocytosis, with clearance mediated mainly through hepatic and renal routes at rates of 9.4 to 28.9 mL/min/kg; there is no significant involvement of cytochrome P450 enzymes in its metabolism.[15][2]Pharmacokinetics are not strictly dose-proportional at therapeutic levels, as serum levels remain low despite dose increases, and steady-state concentrations are not applicable given the every-other-day dosing schedule and lack of drug accumulation.[15]Absorption shows interpatient variability influenced by factors such as injection site and body mass, leading to irregular serum profiles. In patients with hepatic impairment, use with caution and monitor liver function due to the risk of hepatotoxicity, though specific pharmacokinetic data in this population are limited.[15] No major drug interactions have been identified that significantly alter the pharmacokinetics of interferon beta-1b.
Clinical applications
Indications
Interferon beta-1b is primarily indicated for the treatment of relapsing forms of multiple sclerosis (MS) to reduce the frequency of clinical exacerbations and delay the progression of physical disability.[1] This includes patients with relapsing-remitting MS (RRMS), where it serves as a disease-modifying therapy aimed at mitigating inflammatory activity and lesion development in the central nervous system.[1]It is also approved for the treatment of clinically isolated syndrome (CIS), defined as a first demyelinating event suggestive of MS, particularly in patients with MRI features consistent with the disease, to delay the conversion to clinically definite MS.[1] The European Medicines Agency (EMA) similarly authorizes its use in patients with a single severe demyelinating event requiring intravenous corticosteroids and at high risk of developing MS, after exclusion of alternative diagnoses.An extended indication covers secondary progressive MS (SPMS) with active disease, characterized by ongoing relapses, where interferon beta-1b has demonstrated efficacy in reducing relapse rates and slowing disability progression.[1] However, it is not indicated for primary progressive MS (PPMS), as clinical evidence does not support its use in this non-relapsing form.[1] Likewise, interferon beta-1b lacks approval for non-MS conditions, and off-label applications, such as in certain viral infections, are not endorsed by the FDA or EMA due to insufficient robust evidence.Patient selection typically focuses on individuals with early-stage relapsing MS, including CIS or RRMS, who have no contraindications such as hypersensitivity to the drug or decompensated liver disease, positioning it as a first-line disease-modifying therapy in appropriate cases.[1] Guidelines from organizations like the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) recommend its initiation in patients with active relapsing disease to optimize long-term outcomes.[17]
Efficacy
Interferon beta-1b demonstrated efficacy in reducing exacerbation rates in patients with relapsing-remitting multiple sclerosis (RRMS) in the pivotal randomized, double-blind, placebo-controlled trial conducted by the IFNB Multiple SclerosisStudy Group. In this study involving 372 ambulatory patients with RRMS, the annual exacerbation rate was 1.31 in the placebo group compared to 0.90 in the group receiving 8 million international units (MIU) of interferon beta-1b every other day over 2 years, representing approximately 31% reduction (p=0.0001).[18] Additionally, the treatment delayed the time to sustained progression in disability as measured by the Expanded Disability Status Scale (EDSS), with more patients in the high-dose group remaining exacerbation-free at 2 years (36 versus 18 in placebo; p=0.007).In clinically isolated syndrome (CIS) suggestive of multiple sclerosis, early initiation of interferon beta-1b reduced the risk of conversion to clinically definite multiple sclerosis (CDMS). The BENEFIT trial extension showed a 45% risk reduction in conversion to CDMS over 3 years compared to delayed treatment (hazard ratio 0.55; 95% CI 0.41-0.74; p=0.0001).[19] For established RRMS, meta-analyses of clinical trials confirm that interferon beta-1b achieves a 20-30% reduction in annualized relapse rate (ARR), alongside decreased MRI lesion activity and slowed disability progression based on EDSS scores.[20]In secondary progressive multiple sclerosis (SPMS), the European Betaferon trial demonstrated a modest delay in confirmed disability progression with interferon beta-1b. Among 718 patients, treatment delayed progression by 9-12 months over 2-3 years (odds ratio 0.65; 95% CI 0.52-0.83; p=0.0008), though no significant ARR reduction was observed in the subgroup with non-active SPMS. Long-term follow-up data from the original pivotal trial, extending up to 16 years, indicate sustained reduction in relapse frequency with early interferon beta-1b use but limited impact on ultimate disability accumulation.[21][22]Comparative efficacy analyses show that interferon beta-1b has similar effects to other beta-interferons in reducing ARR by approximately 30% in RRMS but is inferior to newer oral disease-modifying therapies, such as fingolimod or teriflunomide, which achieve greater ARR reductions (up to 50%). No head-to-head trials establish superiority among beta-interferons.[23]
Administration and dosing
Interferon beta-1b is administered via subcutaneous injection every other day at a standard dose of 0.25 mg (250 mcg or 8 million international units), with the initial injection typically performed under healthcare provider supervision to ensure proper technique.[15][24]To minimize flu-like symptoms, treatment initiation involves a titration schedule over six weeks, starting at 0.0625 mg every other day and increasing incrementally as follows:
Weeks
Dose (mg)
Volume (mL)
% of Recommended Dose
1–2
0.0625
0.25
25%
3–4
0.125
0.50
50%
5–6
0.1875
0.75
75%
7+
0.25
1.00
100%
This regimen allows gradual adaptation while reaching the full therapeutic dose.[15][24]Patients are trained for self-administration, rotating injection sites among the abdomen, buttocks, thighs, and upper arms to prevent skin reactions; aseptic technique is required, and needles or syringes should not be reused.[15]The lyophilized powder is reconstituted with 1.2 mL of provided diluent (0.54% sodium chloride solution) by gentle swirling—avoiding shaking—and used immediately or stored refrigerated at 2–8°C for up to 3 hours; it remains stable for 3 hours at room temperature post-reconstitution if needed. Unreconstituted vials should be stored at 2–30°C, with excursions permitted, and protected from light; do not freeze.[15]During initiation and ongoing therapy, regular monitoring includes blood tests for liver function, complete blood count (including differential and platelets), and thyroid function at 1, 3, and 6 months, then periodically, to detect potential abnormalities early.[15][25]No dose adjustments are typically required for renal impairment, though caution is advised in hepatic disease with close monitoring of liver function tests due to risk of injury. Use during pregnancy is not recommended, as animal studies indicate potential fetal harm at exposures ≥3 times the human dose, despite human observational data showing no increased risk of major birth defects; previous FDA classification was pregnancy category C.[15][26]
Adverse effects
Common side effects
Interferon beta-1b therapy is commonly associated with flu-like symptoms, affecting approximately 57% of patients in controlled clinical trials, including fever, chills, fatigue, myalgia, arthralgia, and headache, which typically occur within hours of injection and have a median duration of 7.5 days.[27] These symptoms often diminish in incidence over time, decreasing to about 10% after prolonged treatment, and can be managed effectively with premedication using analgesics or antipyretics such as acetaminophen or ibuprofen on injection days.[27][28]Injection site reactions are reported in up to 78% of patients, manifesting as inflammation (42%), pain (16%), hypersensitivity (4%), redness, swelling, or bruising, with rare instances of necrosis (4%).[27] These local reactions are generally mild and decrease in frequency with continued use; proper injection technique and rotation of sites (abdomen, arms, hips, thighs) help minimize severity and prevent complications.[27][29]Laboratory abnormalities frequently include leukopenia (18% vs. 6% placebo), lymphopenia, and elevated liver enzymes such as ALT (up to 12% with levels >5x baseline) and AST (4%), which are typically reversible upon dose reduction or discontinuation.[27] Regular monitoring of complete blood counts and liver function tests is recommended to detect these changes early.[27][28]Other common adverse effects encompass asthenia (53%), headache (50%), myalgia (23%), rash (21%), menstrual disorders, hair thinning, and mild depression, often derived from post-marketing surveillance data.[27][29] Dose titration, starting at lower doses and gradually increasing, can reduce the overall incidence of these effects while maintaining efficacy in multiple sclerosis patients.[27]
Serious adverse effects
Interferon beta-1b can cause hepatic injury, manifesting as severe elevations in liver enzymes or acute hepatitis, with elevations of SGPT greater than five times the upper limit of normal occurring in approximately 12% of patients compared to 4% on placebo, and severe cases including liver failure reported in post-marketing surveillance.[1] Such events, though infrequent (less than 1% for symptomatic severe injury), necessitate immediate discontinuation of the drug if jaundice, fatigue, or other signs of hepatic dysfunction appear, alongside regular monitoring of liver function tests.[1]Hematologic adverse effects include thrombocytopenia and severe neutropenia, with neutropenia reported in 13% of patients versus 5% on placebo, and leukopenia in 18% versus 6%.[1] These occur in less than 5% as severe events requiring intervention, prompting recommendations for monthly complete blood count monitoring, particularly in the initial treatment phase, to detect myelosuppression early.[1]Thrombocytopenia has been noted primarily in post-marketing reports.[1]Neuropsychiatric reactions encompass severe depression, suicidal ideation, and psychosis, with depression or suicide attempts observed in clinical trials at rates leading to three suicides and eight attempts among treated patients compared to one suicide and four attempts on placebo, equating to an approximate 1-5% incidence for severe mood disorders.[1] Pre-treatment screening for mood disorders is advised, and patients should be monitored closely, with discontinuation considered if severe symptoms emerge; psychosis has been reported post-marketing.[1]Hypersensitivity reactions such as anaphylaxis or angioedema are rare, occurring sporadically in post-marketing data, while neutralizing antibody development affects 30-45% of patients, potentially reducing efficacy but not typically linked to severe hypersensitivity.[1] Immediate discontinuation is required for anaphylactic events, with monitoring for allergic symptoms like dyspnea recommended.[1]Other serious risks include seizures, particularly in patients with a history of epilepsy, temporally associated with treatment but without established causality; increased susceptibility to infections due to immunomodulatory effects; and a potential association with malignancy, though no causal link has been confirmed in studies.[1] Patients with seizure history should report events promptly, and general infection vigilance is advised during administration.[1]Post-marketing surveillance has identified very rare events such as macular edema and pulmonary arterial hypertension, with frequency unknown and no definitive causal relationship established, requiring symptom reporting for ophthalmologic or cardiopulmonary changes.[1]
Contraindications and precautions
Interferon beta-1b is contraindicated in patients with a history of hypersensitivity to natural or recombinant interferon beta, human albumin, or mannitol.[27] It is also contraindicated in individuals with decompensated liver disease.[16]Relative precautions are advised for patients with a history of severe depression or suicide attempts, as the drug may exacerbate psychiatric symptoms; close monitoring is essential, and discontinuation should be considered if severe depression develops.[27] Caution is recommended in those with active infections due to the potential for leukopenia increasing infection risk, severe cardiac conditions such as congestive heart failure where worsening may occur, and during pregnancy owing to teratogenic risks observed in animal studies—effective contraception is advised for women of childbearing potential.[30][27]Drug interactions may include additive myelosuppression when combined with other immunosuppressants, such as corticosteroids, necessitating careful monitoring of blood counts.[31] Increased hepatotoxicity is possible with concomitant use of hepatotoxic drugs, so liver function should be assessed prior to initiation.[30]Monitoring requirements encompass baseline and periodic evaluations of liver function tests, complete blood counts, thyroid function, and neurologic status to detect potential complications early.[27]Discontinuation is warranted for persistent severe laboratory abnormalities, such as significant transaminase elevations or leukopenia, or for uncontrolled side effects like worsening depression or hepatic injury.[16]Interferon beta-1b is not approved for use in children under 18 years, as safety and efficacy have not been established in this population.[27] In elderly patients, data are limited, and use requires caution due to higher comorbidity risks, including cardiac and hepatic issues.[27]
History and development
Discovery and early research
Interferons were first discovered in 1957 by Alick Isaacs and Jean Lindenmann, who identified them as soluble proteins produced by virus-infected cells that confer resistance to viral infection in neighboring cells, marking a foundational advance in understanding innate antiviral immunity.[32] This discovery stemmed from studies on viral interference in chick embryo membranes, revealing interferons as non-antibody mediators of cellular protection against viruses.[33]In the 1960s, research differentiated types of interferons, with beta-interferon (IFN-β) specifically identified as the form produced by human fibroblasts in response to viral stimuli, distinguishing it from the leukocyte-derived alpha-interferon. By the 1980s, preclinical investigations demonstrated that type I interferons, including IFN-β, could suppress exacerbations in animal models of multiple sclerosis (MS), such as experimental autoimmune encephalomyelitis (EAE), by modulating immune responses in myelin-reactive T cells.[34] For instance, early studies showed that administration of type I interferons reduced disease severity in EAE through anti-inflammatory effects on central nervous systeminflammation.[35]The recombinant production of human IFN-β advanced in 1980 when David V. Goeddel and colleagues at Genentech cloned the human IFN-β gene and expressed it in Escherichia coli, enabling large-scale bacterial synthesis of the protein.[36] This prokaryotic expression system yielded IFN-β-1b, a non-glycosylated variant differing from the naturally glycosylated form produced in eukaryotic cells. The development of IFN-β-1b specifically involved collaboration between Berlex Laboratories and Cetus Corporation, which focused on optimizing the recombinant protein for therapeutic potential.[37]Preclinical studies of IFN-β-1b established its immunomodulatory properties, including reduced T-cell proliferation and enhanced suppressor T-cell activity in vitro, as well as antiviral efficacy against encephalomyocarditis virus in vivo.[38] Toxicity profiles were characterized in rodents and non-human primates, revealing dose-dependent flu-like symptoms and mild hepatic effects but overall tolerability at therapeutic levels.[39] The rationale for exploring IFN-β-1b in MS arose from the hypothesis that the disease involves immune-mediated demyelination driven by a Th1-dominant response, with type I interferons promoting a shift toward anti-inflammatory Th2 cytokines to restore immune balance.[40] This conceptual framework, supported by EAE observations, laid the groundwork for subsequent therapeutic evaluation.[12]
Pivotal clinical trials
The pivotal clinical trials for interferon beta-1b (IFNB-1b) primarily focused on its efficacy and safety in relapsing-remitting multiple sclerosis (RRMS), secondary progressive multiple sclerosis (SPMS), and clinically isolated syndrome (CIS). The landmark IFNB Multiple Sclerosis Study Group trial, conducted from 1988 to 1993, enrolled 372 ambulatory patients with RRMS in a multicenter, randomized, double-blind, placebo-controlled design.[18] Patients were assigned to receive placebo, 1.6 million international units (MIU) of IFNB-1b, or 8 MIU (equivalent to 250 mcg) subcutaneously every other day for up to 5 years, with a mean follow-up of 2 years.[18] The primary endpoint was the annual exacerbation rate, assessed alongside secondary measures including Expanded Disability Status Scale (EDSS) progression and magnetic resonance imaging (MRI) evaluations of lesion activity and burden.[18] In the 250 mcg dose group, IFNB-1b reduced the exacerbation rate by 29% compared to placebo (1.31 vs. 1.85 exacerbations per year; p=0.023), with fewer moderate-to-severe exacerbations and a significant decrease in new or enlarging MRI lesions (p<0.001).[18][41]Long-term follow-up of this cohort extended to 16 years in an open-label, observational study involving 88% of original participants, confirming sustained safety with no new serious risks but showing no significant difference in EDSS progression between early treatment and placebo groups, indicating plateaued long-term impact on disability.[42] For SPMS, the 1998 European multicenter trial randomized 718 patients to 250 mcg IFNB-1b or placebo subcutaneously every other day for up to 3 years in a double-blind, placebo-controlled design.[21] The primary endpoint was time to sustained EDSS progression by at least 1 point (or 0.5 points if baseline EDSS ≥6.0), confirmed after 6 months.[21] IFNB-1b delayed progression (hazard ratio 0.70; 95% CI 0.54-0.90; p=0.0058), with greater benefits in subgroups with active disease (recent relapses), including a 22% reduction in relapse rate and fewer active MRI lesions (p<0.0001), though no effect was observed in inactive SPMS.[21]The BENEFIT trial (2006-2007 core phase) evaluated early treatment in CIS, randomizing 468 patients with a first demyelinating event and at least two silent T2 lesions to immediate 250 mcg IFNB-1b every other day or placebo (delayed treatment after conversion to clinically definite MS [CDMS] or 24 months), in a double-blind design with 3-year follow-up.[43] Primary endpoints included time to CDMS (Poser criteria) and McDonald MS diagnosis, with secondary assessments of disability via EDSS and MRI outcomes.[43] Early IFNB-1b reduced the risk of CDMS by 45% (hazard ratio 0.55; 95% CI 0.38-0.80; p=0.0017) and McDonald MS by 36% (hazard ratio 0.64; 95% CI 0.47-0.86; p=0.003), alongside fewer new MRI lesions.[43] An open-label extension to 5 years (2009) and beyond (up to 11 years in 2016) sustained these benefits, with early treatment linked to lower EDSS progression and reduced brainatrophy.No major phase III trials for IFNB-1b have emerged since 2010, though real-world evidence from registries like MSBase has validated trial findings on relapse reduction and disability stabilization in diverse, long-term cohorts.[44] These pivotal studies faced limitations, including high dropout rates (up to 48% over 5 years in extensions, often due to injection-site reactions and flu-like symptoms) that may bias efficacy estimates toward completers.[45] Additionally, trial populations lacked ethnic and racial diversity, predominantly enrolling White patients from North America and Europe, potentially limiting generalizability to global MS demographics.[46]
Regulatory approvals
Interferon beta-1b received its initial regulatory approval from the U.S. Food and Drug Administration (FDA) on July 23, 1993, under the brand name Betaseron, developed by Chiron Corporation (later Berlex Laboratories), for the treatment of relapsing-remitting multiple sclerosis (RRMS) in ambulatory patients to reduce the frequency of clinical exacerbations; it was the first disease-modifying therapy approved for multiple sclerosis.[4][47][48]In Europe, the European Medicines Agency (EMA) granted marketing authorization for Betaferon on November 30, 1995, for the treatment of RRMS.[49] This approval was expanded in January 2006 to include secondary progressive multiple sclerosis (SPMS) with active disease, based on evidence from clinical trials demonstrating reduced disability progression.[50] Further expansion occurred in 2007 to cover clinically isolated syndrome (CIS) suggestive of multiple sclerosis, allowing early intervention to delay conversion to clinically definite MS.[51]Approvals followed in other regions, including Canada by Health Canada in December 1994 for RRMS, Australia by the Therapeutic Goods Administration in 1995 for RRMS, and Japan by the Ministry of Health, Labour and Welfare in January 2007 for relapsing forms of MS.[51][52] A pediatric investigation plan for interferon beta-1b was accepted by the EMA in 2013, but as of 2025, it remains unapproved for pediatric use due to insufficient data on safety and efficacy in children under 18 years.[49][53]Label expansions in the United States included updates in 1995 incorporating magnetic resonance imaging (MRI) data from pivotal trials, supporting its role in reducing the frequency and severity of MRI-detected lesions.[54] Post-2000 label revisions by the FDA emphasized monitoring for neutralizing antibodies, which can develop in up to 30-50% of patients and may reduce clinical efficacy, recommending periodic testing to guide treatment decisions.[30][55]In 2009, the FDA approved Extavia, a formulation of interferon beta-1b by Novartis Pharmaceuticals Corporation, on August 14, via an abbreviated pathway as a follow-on product to Betaseron, with identical indications for relapsing forms of MS including a first clinical episode.[56][5] The marketing authorisation for Extavia was withdrawn in the European Union on November 25, 2024, at the request of the marketing authorisation holder for commercial reasons.[57]As of 2025, no major new indications have been approved for interferon beta-1b globally, though product labels continue to carry warnings for severe hepatic injury (including rare cases of hepatic failure) and depression (with monitoring advised due to risks of suicidal ideation), updates to which were incorporated in the 2010s based on post-marketing surveillance.[58][59] It remains recommended in clinical guidelines, such as the American Academy of Neurology's 2018 practice guideline on disease-modifying therapies for adults with multiple sclerosis, as an option for reducing relapses in relapsing forms.[60]
Formulations and availability
Commercial products
Interferon beta-1b is commercially available primarily under two branded products in the United States: Betaseron, manufactured by Bayer HealthCare Pharmaceuticals, and Extavia, manufactured by Novartis Pharmaceuticals. Betaseron is supplied as a lyophilized powder containing 0.3 mg of interferon beta-1b per single-dose vial, which is reconstituted with 1.2 mL of 0.54% sodium chloridediluent to yield a 0.25 mg/mL solution for subcutaneous injection of 1 mL (0.25 mg dose) every other day.[61] The formulation includes human albumin and mannitol as stabilizers.[62] It is packaged in kits consisting of 15 vials, along with corresponding diluent syringes, vial adapters, and needles, and is compatible with the BETACONNECT auto-injector device for easier self-administration.[63] Unreconstituted Betaseron has a shelf life of up to 3 years when stored at room temperature (20°C to 25°C), with excursions permitted to 15°C to 30°C for up to 3 months; reconstituted solution must be used within 3 hours if refrigerated at 2°C to 8°C.[27]Extavia offers a formulation nearly identical to Betaseron and is approved by the FDA as a follow-on biologic demonstrating therapeutic equivalence.[64] It is provided as 0.3 mg of lyophilized interferon beta-1b powder per single-dose vial, reconstituted similarly with 1.2 mL of 0.54% sodium chloride to produce a 0.25 mg/mL solution for the standard 0.25 mg dose.[64] Packaging includes cartons of 15 blister units, each containing a vial, pre-filled diluentsyringe, vial adapter with 27-gauge needle, and alcohol prep pads.[64]Titration packs are available to support the recommended initiation schedule, starting at 0.0625 mg and escalating over 6 weeks to minimize side effects.[64] Like Betaseron, unreconstituted Extavia is stable for 3 years at 20°C to 25°C, with excursions to 15°C to 30°C allowed for up to 3 months, and reconstituted solution is usable within 3 hours under refrigeration.[64]Both products are supplied in single-use vials or kits to ensure sterility and ease of subcutaneous administration, with no major discontinued formulations reported as of 2025. The annual list price for either Betaseron or Extavia exceeds $70,000 USD in the United States as of 2025, though actual costs vary by insurance coverage.[65]Patient assistance programs, such as Bayer's BETAPLUS copay support and US Patient Assistance Foundation, as well as Novartis's offerings, provide financial aid including $0 copay options and free medication for eligible uninsured or underinsured patients.[66][67]
Biosimilars and generics
Extavia, approved by the U.S. Food and Drug Administration (FDA) in 2009, represents the first follow-on biologic to the original interferon beta-1b product Betaseron, demonstrating similarity in efficacy and safety through pharmacokinetic and pharmacodynamic studies.[68]Other biosimilars have emerged primarily in select markets, such as Ziferon in Iran for multiple sclerosis treatment and Ronbetal in Russia, reflecting limited global approvals due to the inherent complexity of biologic manufacturing processes.[69][70] No true small-molecule generics are possible for interferon beta-1b, as it is a complex recombinant protein requiring biological production methods.[71]Regulatory pathways for biosimilars include the FDA's 351(k) abbreviated licensure pathway, established under the 2010 Biologics Price Competition and Innovation Act, which mandates comparative analytical, nonclinical, and clinical studies to confirm similarity to the reference product.[72] The European Medicines Agency (EMA) has followed a similar framework since 2006, emphasizing totality of evidence for biosimilar approval, including immunogenicity assessments.In competitive markets like Iran and Latin America, biosimilars have reduced treatment costs by up to 30% compared to originators, enhancing access in low- and middle-income countries, though overall uptake remains constrained by the subcutaneous injection regimen and persistent high out-of-pocket expenses.[73][74]Key challenges include potential differences in immunogenicity, with biosimilars requiring rigorous post-approval monitoring for neutralizing antibody formation that could impact efficacy.[75] As of 2025, no new major biosimilars have received FDA or EMA approval, with research and development efforts increasingly shifting toward next-generation disease-modifying therapies for multiple sclerosis. Research continues for biosimilars in emerging markets, such as South-East Asia, though none meet FDA or EMA standards as of 2025.[72][76][77]