Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Oct 1.
Published in final edited form as: Expert Opin Biol Ther. 2016 Aug 8;16(10):1303–1309. doi: 10.1080/14712598.2016.1217988

Drug Evaluation

Room for more IL-6 blockade? Sarilumab for the treatment of rheumatoid arthritis

Rayford R June 1, Nancy J Olsen 2
PMCID: PMC5090261  NIHMSID: NIHMS816743  PMID: 27464017

Abstract

Introduction

Rheumatoid arthritis (RA) treatment has been revolutionized by the development of highly efficacious biotherapeutics. However, a significant subset of RA patients has persistently active disease and ongoing erosive joint damage despite the available therapies. Sarilumab targets interleukin-6, one of the main cytokines mediating inflammation in RA. Positive results with sarilumab in RA clinical trials support the licensing application currently under review with the US Food and Drug Administration.

Areas covered

The rationale for IL-6 targeting in RA, the pharmacologic properties of sarilumab, and the clinical trial results are reviewed focusing on the pending application for the RA indication. Comparisons with other IL-6 targeting biologics as well as additional potential therapeutic directions are discussed.

Expert opinion

Sarilumab is a highly active therapeutic in patients with RA. While pharmacologic data demonstrate that sarilumab has a higher affinity than tocilizumab for the target receptor, available clinical results suggest that efficacy and adverse event profiles are similar to this other IL-6 blocker, which is currently approved for the treatment of RA. Whether there are other distinct differences or advantages of sarilumab that will support the approval and successful marketing of this drug, over existing therapies, remains to be determined.

Keywords: Rheumatoid arthritis, Interleukin-6, Monoclonal antibody, Sarilumab, Tocilizumab

1.Introduction

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by synovial inflammation causing a symmetrical, polyarticular arthritis1. RA has a worldwide prevalence of approximately 1%, incidence rises with age, women are disproportionately affected and the most common age of onset is between 35–50 years2. RA is associated with long term disability, with 28% of RA patients less than age 65 considering themselves disabled after 15 years of disease3. Furthermore, as a systemic disease, RA patients have increased mortality, which in male RA patients is estimated as 7 years less than expected4.

Treatment of RA revolves around three classes of medications: non-biologic disease modifying anti-rheumatic drugs (DMARDs), synthetic small molecule inhibitors, and biologic therapies5. Cornerstone DMARD therapy is low dose methotrexate (MTX), with efficacy reported initially in 1985, and increased use has been associated with improved functional outcomes68. While MTX can be effective as monotherapy9, up to 50% of patients discontinue MTX at one year10. Targeted inhibition of Tumor Necrosis Factor-α (TNF) was shown in combination therapy with MTX to be effective in 199911 and soon became standard of care in MTX incomplete responders (MTX-IR)5. However, TNF-inhibition is only effective in 20–50% of RA patients at 6 months of therapy12. Inhibition of other cytokines, IL-113 and IL-614, proved effective in combination with MTX and now 9 biologic therapies plus one small molecule inhibitor (Table 1) are available5, 15.

Table 1.

Crowded market: biologics/synthetic small molecule inhibitors FDA approved or pending for treatment of RA5759

Class Drug Approval year or Phase
TNF alpha
inhibitors
Etanercept
Infliximab
Adalimumab
Certolizumab
Golimumab
1998
1999
2002
2009
2009
Anti-B cell
Directed
Rituximab 2006
Co-stimulatory
blockade
Abatacept 2005
IL-1 inhibitor Anakinra 2001
Kinase
Directed
Tofacitinib
Baricitinib
ABT 494
Filgotinib
2012
Phase III
Phase III
Phase III
IL-6 targeted Tocilizumab
Sarilumab
Sirukumab
2010
Phase III
Phase III

Despite these advances7, only 5–10% of RA patients are in remission and remission is rarely sustained16. Serum IL-6 levels are the most predominant cytokine in RA and correlate with disease activity12. It is therefore an attractive therapeutic target. Sarilumab (SAR) is a fully human monoclonal antibody directed against the IL-6 receptor-α and has higher affinity for the target molecule than the only approved and available inhibitor of IL-6, tocilizumab(TCZ)17. Based on favorable clinical trial results (Table 2), SAR is currently awaiting FDA approval for use in RA. This review focuses on the pharmacology, clinical efficacy, and expected uses of SAR in RA.

Table 2.

Sarilumab Clinical Trials

Phase Trial Study Population Efficacy Outcomes
I Studies 801, 802, 803 Active RA on
methotrexate
None evaluated.
II ALIGN Active Ankylosing
Spondylitis despite
conventional treatment
No clinically significant
responses.
II MOBILITY Part A Active RA despite
treatment with
methotrexate
Significant ACR20
response at 12 weeks
III MOBILITY Part B Active RA despite
treatment with
methotrexate
Sustained clinical
efficacy including
improved radiographic
scores.

2.Market Overview

In the United States, RA has an economic burden of up to 41.6 billion dollars18. Specialty drugs account for over 50% of the cost of care in RA and the medico-economics landscape is changing with biosimilar development19. Despite the availability of these new therapeutics, a large unmet need remains in the treatment of RA, with about half of patients still failing to achieve minimal disease activity scores18.

RA drug development focuses on four populations of RA patients: MTX IR, monotherapy in patients who are either MTX naïve, MTX-intolerant or in whom it is contraindicated, non-MTX DMARD incomplete responders, and finally, the most difficult treatment group, TNF-inhibitor non-responders. While combination non-biologic DMARD therapy, coined “triple therapy” (MTX, hydroxychloroquine and sulfasalazine), has been shown equivalent to MTX with added TNF inhibition11, 20, 21, clinical practice has not seen a significantly increased use of triple therapy22. Practical application of therapies such as payer mix, out of pocket costs and route of administration all affect clinical use and market share18, 23. Pharmaceutical companies aim to meet patient preferences, as seen by the further development of abatacept and TCZ, originally approved for IV administration, into subcutaneous formulations5.

Large comparative effectiveness trials between the many biologic therapies are not been available to guide many therapy choices, but recent randomized and pragmatic clinical trials have begun to clarify the picture. The tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA) trial was a randomized controlled double-blind design in 326 severe RA patients, comparing monotherapy of IL-6 versus TNF inhibition24. TCZ monotherapy compared to adalimumab showed improved 24 week DAS28 scores. The AMPLE trial, abatacept versus adalimumab comparison in biologic-naïve RA subjects with background methotrexate, compared T cell costimulation blockade with abatacept versus TNF inhibition with adalimumab in 646 RA subjects and showed similar outcomes between the two biologics25. Similarly, in an open-label, randomized non-inferiority designed trial in 295 RA patients with inadequate response to DMARDs, TNF inhibition versus rituximab for patients with rheumatoid arthritis who require biological treatment (ORBIT), rituximab was non-inferior to TNF inhibition for 12 month outcome measures26. Recommendations for first biologics therapies suggest equivalence between TNF and non-TNF biologics5, 15. However, despite guidelines and recommendations, TNF inhibitors still remain first line biologics in DMARD failure, which is largely driven in the United States by the reimbursement system. However, even TNF inhibitors have inadequate responses in 20–40% of patients27, 28. Fortunately, additional therapies have been developed, increasing the options (Table 1). Biomarkers predictive of responses to available drugs will be needed to improve efficient matching of the most appropriate therapy to an individual patient.

2.1 IL-6 Inhibition with Tocilizumab

Tocilizumab (TCZ) is a recombinant humanized anti-IL-6R monoclonal antibody that prevents IL-6 from binding soluble and membrane bound forms of the IL-6R29. TCZ was FDA approved in 2010 for the treatment of moderate to severe RA with inadequate response to one or more DMARDs30. It is available for intravenous administration every 4 weeks and also as a subcutaneous dosing once weekly. In the CHARISMA study, intravenous TCZ demonstrated efficacy in MTX-IR RA patients, with 61% and 68% ACR 20 responses at 4 and 8 mg/kg, respectively14. The phase III TCZ trials showed efficacy in multiple RA patient populations: for MTX-IRs31, 32(Table 3), as monotherapy versus MTX33, in traditional DMARD inadequate responders34, and in patients refractory to TNF inhibitors27. Side effect profiles showed increases in total and LDL cholesterol, transaminitis, thrombocytopenia, neutropenia, intestinal perforation and infections.

Table 3.

Comparison of phase III studies of sarilumab and tocilizumab in MTX-IRs: ACR response rates at 24 weeks

Placebo 150 mg 200 mg
Sarilumab
MOBILITY Study, part B42
Genovese et al.
ACR 20 33% 58% 66%
ACR 50 17% 37% 46%
ACR 70 7% 20% 25%
Placebo 4 mg/kg 8 mg/kg
Tocilizumab
OPTION Study29
Smolen et al.
ACR 20 26% 48% 59%
ACR 50 11% 32% 11%
ACR 70 2% 12% 22%
Placebo 4 mg/kg 8 mg/kg
Tocilizumab
LITHE Study28
Kremer et al.
ACR 20 27% 51% 56%
ACR 50 10% 25% 32%
ACR 70 2% 11% 13%

3. Introduction to the compound

Many therapies derived from biologic molecules are now available for the treatment of RA. Most of these block the actions of tumor necrosis factor (TNF) alpha, and TCZ is directed at the IL-6 receptor29. Sarilumab (SAR), highlighted in this review, is one of at least five biologic therapies in development targeting IL-6 (Table 4)35,17. It has been co-developed by Sanofi and Regeneron for the treatment of RA. The United States FDA has accepted for review a licensing application for use of SAR in the treatment of moderate to severe RA. A decision is expected in the last quarter of 2016.

Table 4.

IL-6 targeted biologics in advanced human development for RA

IL-6 Agent Industry association Target Phase
Tocilizumab Genentech / Roche IL-6R and soluble IL-6R
inhibitor
IV
Sarilummab Sanofi / Regeneron IL-6R and soluble IL-6R
inhibitor
III
Sirukinumab GSK / Janssen IL-6 blockade II
Clazakizumab Alder Biopharmaceuticals IL-6 blockade II
Olokizumab UCB/ R-Pharm IL-6 blockade II
ALX-0061 Ablynx IL-6 blockade II

4.Pharmacologic properties

4.1. Pharmacodynamic properties

SAR binds to both the membrane-bound and soluble forms of the IL-6 receptor (CD126). This receptor is expressed on mature T cells, activated B cells, neutrophils, macrophage lineage cells, hepatocytes and myeloma cells. Activation occurs when IL-6 is first bound by IL-6Rα, and this complex is then presented to the gp130 molecule that is present on many cell types36. Internalization of the activation complex initiates signaling through the Jak/STAT pathway35. Soluble forms of IL-6Ralpha that lack the transmembrane domain are formed through cell surface cleavage or alternative splicing. These circulating molecules can also activate gp130-bearing cells through a trans-signaling mechanism, so successful IL-6 receptor therapeutics need to block both membrane and soluble forms35. IL-6 is a major driver of C-reactive protein synthesis, and levels were decreased by up to 90% in Phase I and II studies37, 38,39. Blockade of the IL-6R with TCZ has been shown to reduce numbers of memory B cell subsets in treated patients, and to increase regulatory T cells while not changing Th17 T cell subset frequency40,41; comparable data with SAR have not been reported. Concentrations of IL-6 were transiently increased after initiation of SAR in Phase I studies and in the first 12 weeks of treatment in Phase II39; TCZ has a similar effect, which is thought to be secondary to blockade of the sIL-6R causing decreased consumption of IL-642.

4.2 Pharmacokinetic properties

SAR is a fully human monoclonal IgG1 antibody that was developed in mice engineered to produce human antibodies43. The affinity of SAR for the human IL-6 receptor is approximately 20-fold greater than TCZ, and it is about 4-fold more potent than TCZ in a luciferase reporter assay17. The dissociation constant (Kd) of SAR for the target receptor is 12.8 pM, which is about 55 times lower than that of TCZ, consistent with the higher binding affinity35. Clearance of SAR is biphasic and target-mediated with a dose dependent effect and non-linear clearance38. Trough serum levels of functional and bound SAR are measurable throughout a 2 week dosing interval. Serum drug levels show dose-related increases in treated patients through at least 12 weeks39. Metabolism likely follows the same pathways as other fully human monoclonal IgG1 antibodies such as secukinumab, which has a half-life of approximately 27 days44. Antidrug antibodies were found in up to 16.7% of patients in the phase III SAR study but immunogenicity was not associated with hypersensitivity reactions45. Associations with drug levels are not known.

5.0 Drug Efficacy

5.1 Rheumatoid Arthritis

Phase I: Three phase I randomized double-blind, placebo controlled trials of SAR in RA were carried out in a total of 83 active treatment and 24 placebo patients using ascending doses of 50 mg, 100mg, 150 mg, and 200 mg37. A dose dependent reduction in levels of acute phase reactants was observed, with a greater than 90% reduction in hsCRP and serum amyloid A after a single 200 mg dose of SAR.

Phase II: Safety and efficacy of SAR was evaluated in Monoclonal antiBody to IL-6Rα In RA patients: A pivotal Trial with X-raY(MOBILITY)39. Part A was a Phase II multicenter, randomized, double-blind placebo controlled study in 306 RA patients46 with active disease despite a stable dose of methotrexate (10–25 mg/week, mean dose of 16.6 mg/week). Patients were randomized to placebo or one of five subcutaneous SAR dosing regimens: 100 mg Q2W, 150 mg Q2W, 200 mg Q2W, 100 mg qW and 150 mg QW. Patients with a history of non-response to anti-TNF therapy were excluded47. Significant ACR20 responses compared to placebo were seen after 12 weeks of treatment across all doses of 150 mg q2 weeks or greater. DAS28 CRP responses showed a dose response relationship with SAR and the highest remission response, defined as DAS28 CRP< 2.6, was seen in the 150 mg q week regimen. Greater than 60% of the SAR 150 mg Q week and higher doses achieved ACR 20%, and 17% of the SAR 200mg Q2W dose group achieved ACR 70 response; 20–30% of subjects achieved DAS28CRP <2.6 by week 12.

Phase III:Press reports indicate that 2500 RA patients have been enrolled in seven phase III studies submitted with the FDA application. However, the only phase III data published and available for review is from the MOBILITY study. As noted, this was a seamless design Phase II/III study in MTX-IR RA patients with erosions or CCP positivity. It offered as an extension to Phase II a 52 week double blind placebo controlled treatment period. Patients were allocated to one of three arms: placebo (n=398), SAR 150 mg every 2 weeks (n=400) and SAR 200 mg every 2 weeks(n=399)45. Results in all domains, disease response, function and radiographic scores, were favorable for SAR. A high placebo response was seen with an ACR20 response of more than 33%, but even higher ACR20 responses of 58% and 66% were seen in the 150 and 200 mg groups. All three primary endpoints for the trial were met at 24 weeks and were sustained at 52 weeks. ACR70 major clinical response and maintenance for 24 weeks was seen in 3% of placebo versus 12.8% and 14.8% of the 150 mg and 200 mg groups respectively, with 34.1% of the 200 mg group achieving week 24 DAS28-CRP remission. Functional assessment measured using the Health Assessment Questionnaire (HAQ) showed statistically significant improvement throughout the 52 week study period. Structural changes assessed with the Sharp-van der Heijde score (SHS) showed improvement compared to placebo, with a 2.78 placebo change versus 0.9 for the 150 mg and 0.25 for the 200 mg dose at week 52. While this was a statistically significant change, SHS scores greater than 4.6 are thought to be clinically significant to impact therapy48. However, given the relatively short time of evaluation, the trend and dose response imply a clinically significant effect. Looked at another way, 39% of the placebo group had no radiographic progression at 1 year versus 48–56% of the SAR treated patients. Across the board, considering signs and symptoms, functional response and structural damage, SAR showed significant efficacy in MTX-IR RA patients.

The phase III TARGET study, presented at the ACR 2015 annual scientific meeting, evaluated SAR in combination with non biologic DMARDs versus placebo in 546 active RA patients with inadequate response or intolerant of 1 or more TNF inhibitors49. In this randomized placebo controlled study, SAR in doses of 150 mg and 200 mg resulted in week 24 ACR 20 responses of 56% and 61% versus 34% in placebo. ACR70 responses of 20% and 16% were also seen in the 150 mg and 200mg groups with improvement in physical function. In both MTX-IR and TNF non-responders, SAR demonstrated efficacy in improving the signs, symptoms, and outcomes in patients with moderate to severe RA.

A comparison of American College of Rheumatology (ACR) response rates for SAR from the MOBILITY trial with TCZ in MTX-IR patients shows comparable results between the two medications (Table 3). Both can be given subcutaneously, with TCZ requiring weekly injections as opposed to every 2 week injections for SAR.

5.2 Other indications

Nine SAR trials with non-RA indications have been registered on clinicaltrials.gov. Most are in non-infectious uveitis and have not been reported. The ALIGN study in patients with AS was a randomized, multicenter, double blind parallel group placebo controlled trial of 100/150/200 mg every 2 weeks or 100/150 mg weekly for 12 weeks in 300 patients. The primary endpoint, ASAS20 in SAR versus placebo was not met, and further development for AS is not ongoing.

6.0 Drug administration/dosage

SAR is administered as a subcutaneous injection. In the Phase 2, Part A trial of the MOBILITY study, doses of 100, 150 and 200 mg were given weekly or at 2 week intervals39. The 150 and 200 mg q 2 week groups both showed similar efficacy to the weekly dosing groups. Some trends in safety measures favored the q 2 week dosing groups. Given these data and the fact that every other week dosing has greater convenience for the patient, the Phase 3 Part B trial of MOBILITY used 150 and 200 mg, each dosed at 2 week intervals. The dose or doses that have been requested for licensing approval in the FDA application have not been disclosed.

7.0 Safety and Tolerability

In the phase I studies, a total of 83 patients were evaluated for SAR safety and tolerability50. Four patients withdrew due to adverse events, 3 in the SAR group and 1 in placebo; reasons for withdrawal were RA flare, upper respiratory tract infection, and ALT elevation. In the 16 week follow up period, 1 patient each in placebo and SAR groups had ALT elevation of 3–5× upper limit of normal (ULN) and 1 patient in SAR had >5× ULN. Five patients in the SAR group had transient neutropenia (0.5–1.0 × 10^3/uL). No dose limiting toxicities were observed.

In the MOBILITY Part A (phase II) trial of 306 patients with active RA and concurrent stable methotrexate, dose-related neutropenia was observed. ALT increased to 3× upper limit of normal in 4% and mean cholesterol was elevated in 10–20% at 12 weeks. Treatment emergent adverse events (TEAE) were more frequent in the SAR100 mg qW dosing group, with infection and neutropenia as the primary reasons for treatment discontinuation. While there were increased TEAE in SAR vs placebo, there was no dose trend to the serious adverse events, and no serious infections.

In Mobility part B (phase III) trial in which 1282 patients were included in the safety analysis, adverse events (AE) and serious adverse events (SAE) were more common in SAR (11.3% in the 200 mg group vs. 5.4% placebo 5.4%; Table 5). Events leading to drug discontinuation occurred in 13.9% of the SAR 200 mg dose group. Infections were the most common AE and SAE. One patient in the SAR 150 mg group who was on concomitant aspirin and had a history of gastroesophageal reflux disease had a perforation of a duodenal ulcer and died. Given reported gastrointestinal perforations with TCZ, this event might be considered treatment related. Injection site reactions were reported in approximately 10% of the SAR treated patients. Eight malignancies occurred, with a higher number in the SAR treated patients; no lymphomas were reported. Neutropenia in the range of 0.5–1.0 × 109/liter was observed in 5.1% of the SAR 150 mg and 7.8% in the 200 mg groups. Neutropenia occurred in a dose dependent manner with 9.3% and 14.4% in the 150 mg and 200 mg SAR groups respectively. Overall, grade 3 or 4 neutropenia normalized either during continued treatment or with discontinuation in 24/26 (92%) in the 150 mg SAR dose versus 29/36 (81%) in the 200 mg SAR dose; 9/62 (15%) subjects had grade 3 or 4 neutropenia that never normalized during the study. Neutropenia led to treatment discontinuation in 2.1% of patients in 150 mg and 2.4% in 200 mg SAR, but the grade of neutropenia was not associated with infection. ALT greater than 3× ULN occurred in 9.5% of the 150 mg SAR group versus 2.1% of placebo. Increase in serum cholesterol occurred in 43% of the SAR group versus 18% of placebo. Antidrug antibodies were found in 16.7% of the SAR 200 mg dose, but were not associated with hypersensitivity, drug discontinuation, or lack of efficacy.

Table 5.

Sarilumab treatment-emergent adverse events in phase III

Event type Description Prevalence (%)
Low/High/Placebo
Comments
Infections:

  Serious

  Opportunistic
Infection most
common emergent
adverse event


2.6 / 4.0 / 2.3

0.7 / 0.9 / 0.5
No cases of TB

One fungal bronchitis

H. Zoster
Abnormal liver
function tests
ALT more than 3×
ULN
9.5/ 8.0 / 2.1 24 patients were
discontinued due to
elevations
Elevated total
cholesterol
From < 240 mg/dl to
≥ 240 mg/dl
36.8 / 43.0 / 18.3 No cardiac events
Decreased
neutrophil
counts
0.5 – <1.0 × 109/L

< 0.5 × 109/L
5.1 / 7.8 / 0.0

0.9 / 0.7 / 0.0
No relationship of low
neutrophil count to
infections.

Overall, adverse events appeared to be dose dependent across the studies. SAE were rare, but comparable with what is observed with other biologics and tofacitinib51, 52. Adverse events with SAR are similar to those with TCZ.

8.0 Conclusion

Phase II and III clinical trials have established that SAR is an effective treatment for patients with moderate to severe RA who have had an incomplete response to MTX. The efficacy and safety profiles are very similar to the available IL6 targeting drug TCZ, and include significant decreases in progression of structural damage. SAR was not effective in AS, which is also similar to findings with TCZ53. This is despite the fact that circulating IL-6 levels correlate with disease features in AS, and is another reminder that predictions based on biologic data do not always translate into therapeutic effects54. Infections were the most common adverse events with SAR, similar to findings with TNF blockade, while neutropenia, transaminitis and elevated cholesterol are more specific for the IL6 pathway.

9.0 Expert Opinion

Sarilumab is an efficacious drug for the treatment of RA patients with at least moderately active disease who have had an incomplete response to MTX. In addition to controlling symptoms, it has an impressive effect on structural damage. Despite pharmacologic differences showing that SAR has greater target affinity than TCZ, efficacy outcomes are similar. Similar to the differences between two TNF inhibitors adalimumab and etanercept, SAR offers patients and clinicians an additional subcutaneous IL-6 inhibitor to TCZ of SC dosing every 2 weeks as opposed to every week. SAR is likely to obtain FDA approval, as it meets the agency guidelines for efficacy, and safety is not significantly different from TCZ55. The field of RA therapeutics is crowded, but there is still a need for new effective therapies that improve sustained remission. The bigger question is not if SAR is effective, but where SAR will fit into treatment algorithms12. Moreover, with 4 other IL-6 blocking agents being tested; IL-6 may become the next TNF in RA, highlighting the need for more comparative effectiveness trials between IL-6 and TNF based therapeutics12, 35.

This equilibrium of TNF-first could be disrupted, however, by availability of new data from head-to-head trials of IL-6 and TNF-targeting therapeutics. The SARIL-RA-MONARCH trial comparing SAR and adalimumab monotherapy has been completed, and press releases claim that SAR had significantly better efficacy, but these data have not yet been presented in abstract or published form. The Hoffman-La Roche ENTRACTE trial, which compares cardiovascular outcomes in RA patients at high risk of cardiovascular disease treated for up to 5 years with either etanercept or TCZ, may produce data about whether these treatments have different risk profiles. If TCZ does not show significantly more adverse outcomes, this may lay to rest the concerns about raising cholesterol levels in a population that is already at risk for cardiovascular disease. Basically, differences in either efficacy or adverse outcomes could tip the balance one way or the other between IL-6 and TNF blockade as first-line strategies in MTX-IRs. The other dimension is whether treatment failures with one class can be rescued by treatment with the other, and if so, which drugs work best. A Phase II study comparing the humanized anti-IL6 antibody olokizumab to TCZ in TNF incomplete responders showed that both had efficacy56. This at least suggests that the principle of targeting IL-6 in patients without good responses to TNF blockers may be valid. But to improve individual treatment, biomarkers that predict the likelihood of a favorable response to a given therapeutic are needed.

The bottom line is that while sarilumab likely will win FDA approval, finding its niche in the panoply of available RA therapeutics may be a challenge.

Drug Summary Box.

Drug Name Sarilumab
Phase Pre-Registration.
Indication Adults with active moderate to severe rheumatoid
arthritis and incomplete response to methotrexate.
Pharmacology description Fully human monoclonal IgG1 antibody that binds
specifically to the interleukin-6 receptor alpha
Route of Administration Subcutaneous injection 150 or 200 mg every 2
weeks
Pivotal Trials MOBILITY39, 45

Acknowledgments

Funding:

RR June has received National Institutes of Health grant support via grants UL1-TR00012 and K12-TR000126. NJ Olsen has received lab funding from Mallinckrodt Pharmaceuticals.

NJ Olsen has been a site investigator for Mallinckrodt Pharmaceuticals, Genentech/Roche, Horizon, Aurinia Pharmaceuticals Inc., Invion Limited and Resolve Therapeutics.

Abbreviation List

ACR

American College of Rheumatology

AS

Ankylosing Spondylitis

DMARD

Disease Modifying Anti-Rheumatic Drug

EULAR

European League Against Rheumatism

HAQ

Health Assessment Questionnaire

MTX

methotrexate

MTX-IR

methotrexate incomplete responders

RA

Rheumatoid Arthritis

SAE

serious adverse events

SAR

Sarilumab

SHS

Sharp-van der Heijde score

TCZ

Tocilizumab

TEAE

Treatment adverse events

TNF

Tumor necrosis factor

ULN

upper limit of normal range

Footnotes

Declaration of Interest

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Contributor Information

Rayford R. June, Division of Rheumatology, Department of Medicine, Penn State MS Hershey Medical Center, Hershey PA 17033.

Nancy J. Olsen, Division of Rheumatology, Department of Medicine, Penn State MS Hershey Medical Center, Hershey PA 17033.

Bibliography

  • 1.McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011 Dec 8;365(23):2205–2219. doi: 10.1056/NEJMra1004965. [DOI] [PubMed] [Google Scholar]
  • 2.Gibofsky A. Epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis: A Synopsis. Am J Manag Care. 2014 May;20(7 Suppl):S128–S135. [PubMed] [Google Scholar]
  • 3.Wolfe F, Michaud K, Pincus T. A composite disease activity scale for clinical practice, observational studies, and clinical trials: the patient activity scale (PAS/PAS-II) J Rheumatol. 2005 Dec;32(12):2410–2415. [PubMed] [Google Scholar]
  • 4.Gabriel SE. Why do people with rheumatoid arthritis still die prematurely? Ann Rheum Dis. 2008 Dec;67(Suppl 3):iii30–iii34. doi: 10.1136/ard.2008.098038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Singh JA, Saag KG, Bridges SL, et al. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2016 Jan;68(1):1–26. doi: 10.1002/art.39480. [DOI] [PubMed] [Google Scholar]
  • 6.Weinblatt ME, Coblyn JS, Fox DA, et al. Efficacy of low-dose methotrexate in rheumatoid arthritis. N Engl J Med. 1985 Mar;312(13):818–822. doi: 10.1056/NEJM198503283121303. [DOI] [PubMed] [Google Scholar]
  • 7.Kievit W, Fransen J, de Waal Malefijt MC, et al. Treatment changes and improved outcomes in RA: an overview of a large inception cohort from 1989 to 2009. Rheumatology (Oxford) 2013 Aug;52(8):1500–1508. doi: 10.1093/rheumatology/ket166. [DOI] [PubMed] [Google Scholar]
  • 8.Pincus T, Gibson KA, Castrejón I. Update on methotrexate as the anchor drug for rheumatoid arthritis. Bull Hosp Jt Dis. 2013;201371(Suppl 1):S9–S19. [PubMed] [Google Scholar]
  • 9.Pincus T. RAPID3, an index of only 3 patient self-report core data set measures, but not ESR, recognizes incomplete responses to methotrexate in usual care of patients with rheumatoid arthritis. Bull Hosp Jt Dis (2013) 2013;71(2):117–120. [PubMed] [Google Scholar]
  • 10.Curtis JR, Zhang J, Xie F, et al. Use of oral and subcutaneous methotrexate in rheumatoid arthritis patients in the United States. Arthritis Care Res (Hoboken) 2014 Nov;66(11):1604–1611. doi: 10.1002/acr.22383. [DOI] [PubMed] [Google Scholar]
  • 11.Moreland LW, O'Dell JR, Paulus HE, et al. A randomized comparative effectiveness study of oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid arthritis: the treatment of Early Aggressive Rheumatoid Arthritis Trial. Arthritis Rheum. 2012 Sep;64(9):2824–2835. doi: 10.1002/art.34498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Tanaka Y, Martin Mola E. IL-6 targeting compared to TNF targeting in rheumatoid arthritis: studies of olokizumab, sarilumab and sirukumab. Ann Rheum Dis. 2014 Sep;73(9):1595–1597. doi: 10.1136/annrheumdis-2013-205002. * This is a balanced overview of therapeutics that target IL-6 which compares their similarities and differences, and considers whether an evidence-based rationale for chosing initial treatments and for switching agents when one fails might be developed.
  • 13.Cohen SB, Moreland LW, Cush JJ, et al. A multicentre, double blind, randomised, placebo controlled trial of anakinra (Kineret), a recombinant interleukin 1 receptor antagonist, in patients with rheumatoid arthritis treated with background methotrexate. Ann Rheum Dis. 2004 Sep;63(9):1062–1068. doi: 10.1136/ard.2003.016014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Maini RN, Taylor PC, Szechinski J, et al. Double-blind randomized controlled clinical trial of the interleukin-6 receptor antagonist, tocilizumab, in European patients with rheumatoid arthritis who had an incomplete response to methotrexate. Arthritis Rheum. 2006 Sep;54(9):2817–2829. doi: 10.1002/art.22033. [DOI] [PubMed] [Google Scholar]
  • 15. Smolen JS, Landewé R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis. 2014 Mar;73(3):492–509. doi: 10.1136/annrheumdis-2013-204573. ** These updated guidelines suggest that available biologic DMARDs including, but not limited to, TNF and IL6 blockers, have similar efficacy and that if one fails, any other biologic DMARD may be tried.
  • 16.Shahouri SH, Michaud K, Mikuls TR, et al. Remission of rheumatoid arthritis in clinical practice: application of the American College of Rheumatology/European League Against Rheumatism 2011 remission criteria. Arthritis Rheum. 2011 Nov;63(11):3204–3215. doi: 10.1002/art.30524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rafique A, Martin J, Blome M, et al. Evaluation of the binding kinetics and functional bioassay activity of sarilumab and tocilizumab to the human IL-6 receptor alpha. Ann Rheum Dis. 2013:A797. [Google Scholar]
  • 18. Taylor PC, Moore A, Vasilescu R, et al. A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: a current perspective. Rheumatol Int. 2016 May;36(5):685–695. doi: 10.1007/s00296-015-3415-x. * The authors of this literature review emphasize that there are unmet needs in RA treatment, including domains that are not usually included in pivotal trials, such as fatigue.
  • 19.Gleason PP, Alexander GC, Starner CI, et al. Health plan utilization and costs of specialty drugs within 4 chronic conditions. J Manag Care Pharm. 2013 Sep;19(7):542–548. doi: 10.18553/jmcp.2013.19.7.542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.O'Dell JR, Mikuls TR, Taylor TH, et al. Therapies for active rheumatoid arthritis after methotrexate failure. N Engl J Med. 2013 Jul 25;369(4):307–318. doi: 10.1056/NEJMoa1303006. [DOI] [PubMed] [Google Scholar]
  • 21.van Vollenhoven RF, Geborek P, Forslind K, et al. Conventional combination treatment versus biological treatment in methotrexate-refractory early rheumatoid arthritis: 2 year follow-up of the randomised, non-blinded, parallel-group Swefot trial. Lancet. 2012 May;379(9827):1712–1720. doi: 10.1016/S0140-6736(12)60027-0. [DOI] [PubMed] [Google Scholar]
  • 22.Sparks JA, Krumme AA, Shrank WH, et al. Intensification to triple therapy non-biologic disease-modifying antirheumatic drugs for rheumatoid arthritis in the United States from 2009 to 2014. Arthritis Rheumatol. 2016 Feb; doi: 10.1002/art.39617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hopson S, Saverno K, Liu LZ, et al. Impact of Out-of-Pocket Costs on Prescription Fills Among New Initiators of Biologic Therapies for Rheumatoid Arthritis. J Manag Care Spec Pharm. 2016 Feb;22(2):122–130. doi: 10.18553/jmcp.2016.14261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gabay C, Emery P, van Vollenhoven R, et al. Tocilizumab monotherapy versus adalimumab monotherapy for treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind, controlled phase 4 trial. Lancet. 2013 May;381(9877):1541–1550. doi: 10.1016/S0140-6736(13)60250-0. [DOI] [PubMed] [Google Scholar]
  • 25.Schiff M, Weinblatt ME, Valente R, et al. Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: two-year efficacy and safety findings from AMPLE trial. Ann Rheum Dis. 2014 Jan;73(1):86–94. doi: 10.1136/annrheumdis-2013-203843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Porter D, van Melckebeke J, Dale J, et al. Tumour necrosis factor inhibition versus rituximab for patients with rheumatoid arthritis who require biological treatment (ORBIT): an open-label, randomised controlled, non-inferiority, trial. Lancet. 2016 May; doi: 10.1016/S0140-6736(16)00380-9. [DOI] [PubMed] [Google Scholar]
  • 27.Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann Rheum Dis. 2008 Nov;67(11):1516–1523. doi: 10.1136/ard.2008.092932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vander Cruyssen B, Van Looy S, Wyns B, et al. Four-year follow-up of infliximab therapy in rheumatoid arthritis patients with long-standing refractory disease: attrition and long-term evolution of disease activity. Arthritis Res Ther. 2006;8(4):R112. doi: 10.1186/ar2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mihara M, Kasutani K, Okazaki M, et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol. 2005 Nov;5(12):1731–1740. doi: 10.1016/j.intimp.2005.05.010. [DOI] [PubMed] [Google Scholar]
  • 30.Tocilizumab Full Prescribing Information. [cited 2016 April 14th];2016 Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/125276lbl.pdf.
  • 31.Kremer JM, Blanco R, Brzosko M, et al. Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with inadequate responses to methotrexate: results from the double-blind treatment phase of a randomized placebo-controlled trial of tocilizumab safety and prevention of structural joint damage at one year. Arthritis Rheum. 2011 Mar;63(3):609–621. doi: 10.1002/art.30158. [DOI] [PubMed] [Google Scholar]
  • 32.Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008 Mar;371(9617):987–997. doi: 10.1016/S0140-6736(08)60453-5. [DOI] [PubMed] [Google Scholar]
  • 33.Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010 Jan;69(1):88–96. doi: 10.1136/ard.2008.105197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Genovese MC, McKay JD, Nasonov EL, et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 2008 Oct;58(10):2968–2980. doi: 10.1002/art.23940. [DOI] [PubMed] [Google Scholar]
  • 35.Kim GW, Lee NR, Pi RH, et al. IL-6 inhibitors for treatment of rheumatoid arthritis: past, present, and future. Arch Pharm Res. 2015;38(5):575–584. doi: 10.1007/s12272-015-0569-8. [DOI] [PubMed] [Google Scholar]
  • 36.Boulanger MJ, Chow DC, Brevnova EE, Garcia KC. Hexameric structure and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 complex. Science. 2003 Jun;300(5628):2101–2104. doi: 10.1126/science.1083901. [DOI] [PubMed] [Google Scholar]
  • 37.Radin AR, Mellis SJ, Jasson M, et al. REGN88/SAR153191, A fully-human interleukin-6 receptor monoclonal antibody, reduces acute phase reactants in patients with rheumatoid arthritis: Preliminary observations from Phase 1 studies. Arthritis Rheum: Blackwell. 2010:1121. [Google Scholar]
  • 38.Belomestnov P, Hamilton J, DiCioccio AT, et al. Sarilumab, a subcutaneously-administered, fully human monoclonal antibody inhibitor of the IL-6 Receptor: Pharmacokinetic profile and its relationship to changes in pharmacodynamic markers in patients with rheumatoid arthritis. Arthritis Rheum. 2012:S576. [Google Scholar]
  • 39. Huizinga TW, Fleischmann RM, Jasson M, et al. Sarilumab, a fully human monoclonal antibody against IL-6Rα in patients with rheumatoid arthritis and an inadequate response to methotrexate: efficacy and safety results from the randomised SARIL-RA-MOBILITY Part A trial. Ann Rheum Dis. 2014 Sep;73(9):1626–1634. doi: 10.1136/annrheumdis-2013-204405. * This Phase II dose-ranging study demonstrated positive clinical effects that supported continuation of sarilumab development into Phase III.
  • 40. Thiolat A, Semerano L, Pers YM, et al. Interleukin-6 receptor blockade enhances CD39+ regulatory T cell development in rheumatoid arthritis and in experimental arthritis. Arthritis Rheumatol. 2014 Feb;66(2):273–283. doi: 10.1002/art.38246. ** Data generated in a mouse model as well as from RA patients indicate that anti-IL-6R treatment is associated with increased functionally-active T regulatory cells, suggesting this as a possible mechanism contributing to therapeutic efficacy.
  • 41.Roll P, Muhammad K, Schumann M, et al. In vivo effects of the anti-interleukin-6 receptor inhibitor tocilizumab on the B cell compartment. Arthritis Rheum. 2011 May;63(5):1255–1264. doi: 10.1002/art.30242. [DOI] [PubMed] [Google Scholar]
  • 42.Nishimoto N, Terao K, Mima T, et al. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008 Nov;112(10):3959–3964. doi: 10.1182/blood-2008-05-155846. [DOI] [PubMed] [Google Scholar]
  • 43.Scott CT. Mice with a human touch. Nat Biotechnol. 2007 Oct;25(10):1075–1077. doi: 10.1038/nbt1007-1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rothstein B, Gottlieb A. Secukinumab for treating plaque psoriasis. Expert Opin Biol Ther. 2016 Jan;16(1):119–128. doi: 10.1517/14712598.2016.1121986. [DOI] [PubMed] [Google Scholar]
  • 45. Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab Plus Methotrexate in Patients With Active Rheumatoid Arthritis and Inadequate Response to Methotrexate: Results of a Phase III Study. Arthritis Rheumatol. 2015 Jun;67(6):1424–1437. doi: 10.1002/art.39093. ** This is a large Phase III trial showing efficacy in combination with methotrexate, including radiographic outcomes.
  • 46.Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988 Mar;31(3):315–324. doi: 10.1002/art.1780310302. [DOI] [PubMed] [Google Scholar]
  • 47.Felson DT, Anderson JJ, Boers M, et al. American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum. 1995 Jun;38(6):727–735. doi: 10.1002/art.1780380602. [DOI] [PubMed] [Google Scholar]
  • 48.Bruynesteyn K, van der Heijde D, Boers M, et al. Determination of the minimal clinically important difference in rheumatoid arthritis joint damage of the Sharp/van der Heijde and Larsen/Scott scoring methods by clinical experts and comparison with the smallest detectable difference. Arthritis Rheum. 2002 Apr;46(4):913–920. doi: 10.1002/art.10190. [DOI] [PubMed] [Google Scholar]
  • 49.Fleischmann R, Castelar-Pinheiro G, Brzezicki J, et al. Efficacy and Safety of Sarilumab in Combination with Csdmards in Patients with Active Rheumatoid Arthritis Who Were Indadequate Responders or Intolerant of Anti-TNF Therapy: Results from a Phase 3 Study [abstract] Arthritis Rheumatology. 2015 [Google Scholar]
  • 50. Sieper J, Braun J, Kay J, et al. Sarilumab for the treatment of ankylosing spondylitis: results of a Phase II, randomised, double-blind, placebo-controlled study (ALIGN) Ann Rheum Dis. 2015 Jun;74(6):1051–1057. doi: 10.1136/annrheumdis-2013-204963. * The results in patients with Ankylosing Spondylitis reported in this placebo-controlled, dose-ranging trial were not positive and further development of Sarilumab for this indication was not pursued.
  • 51.Cohen S, Radominski SC, Gomez-Reino JJ, et al. Analysis of infections and all-cause mortality in phase II, phase III, and long-term extension studies of tofacitinib in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014 Nov;66(11):2924–2937. doi: 10.1002/art.38779. [DOI] [PubMed] [Google Scholar]
  • 52.Singh JA, Wells GA, Christensen R, et al. Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev. 2011;(2):CD008794. doi: 10.1002/14651858.CD008794.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chen C, Zhang X, Xiao L, Ma X. Comparative Effectiveness of Biologic Therapy Regimens for Ankylosing Spondylitis: A Systematic Review and a Network Meta-Analysis. Medicine (Baltimore) 2016 Mar;95(11):e3060. doi: 10.1097/MD.0000000000003060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Strand V, Kimberly R, Isaacs JD. Biologic therapies in rheumatology: lessons learned, future directions. Nat Rev Drug Discov. 2007 Jan;6(1):75–92. doi: 10.1038/nrd2196. [DOI] [PubMed] [Google Scholar]
  • 55.Emery P, Rondon J, Garg A, et al. Safety and tolerability of subcutaneous sarilumab compared to intravenous tocilizumab in patients with A. Arthritis Rheum. 2015 doi: 10.1093/rheumatology/key361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Genovese MC, Fleischmann R, Furst D, et al. Efficacy and safety of olokizumab in patients with rheumatoid arthritis with an inadequate response to TNF inhibitor therapy: outcomes of a randomised Phase IIb study. Ann Rheum Dis. 2014 Sep;73(9):1607–1615. doi: 10.1136/annrheumdis-2013-204760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Reichert JM. Antibodies to watch in 2016. MAbs 2016. 2016 Feb-Mar;8(2):197–204. doi: 10.1080/19420862.2015.1125583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Reichert JM. Antibodies to watch in 2015. MAbs. 2015;7(1):1–8. doi: 10.4161/19420862.2015.988944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Drugs@ FDA: FDA Approved Drug Products. [cited 2016 April 8th];2016 Available from: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm.

RESOURCES