7,678
Views
60
CrossRef citations to date
0
Altmetric
Reports

Secukinumab, a novel anti–IL-17A antibody, shows low immunogenicity potential in human in vitro assays comparable to other marketed biotherapeutics with low clinical immunogenicity

, &
Pages 536-550 | Received 10 Jul 2015, Accepted 22 Dec 2015, Published online: 04 Mar 2016

References

  • Martin DA, Towne JE, Kricorian G, Klekotka P, Gudjonsson JE, Krueger JG, Russell CB. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol 2013; 133:17-26; PMID:22673731; http://dx.doi.org/10.1038/jid.2012.194
  • Lowes MA, Kikuchi T, Fuentes-Duculan J, Cardinale I, Zaba LC, Haider AS, Bowman EP, Krueger JG. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. J Invest Dermatol 2008; 128:1207-11; PMID:18200064; http://dx.doi.org/10.1038/sj.jid.5701213
  • Johansen C, Usher P, Kjellerup RB, Lundsgaard D, Iverson L, Kragballe K. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br J Dermatol 2009; 160:319-24; PMID:19016708; http://dx.doi.org/10.1111/j.1365-2133.2008.08902.x
  • Lin AM, Rubin CJ, Khandpur R, Wang JY, Riblett M, Yalavarthi S, Villaneuva EC, Shah P, Kaplan MJ, Bruce AT. Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J Immunol 2011; 187:490-500; PMID:21606249; http://dx.doi.org/10.4049/jimmunol.1100123
  • Res PC, Piskin G, deBoer OJ, van der Loos CM, Teeling P, Bos JD, Teunissen MB. Overrepresentation of IL-17A and IL-22 producing CD8 T cells in lesional skin suggest their involvement in the pathogenesis of psoriasis. PLoS One 2010; 5:e14108; PMID:21124836; http://dx.doi.org/10.1371/journal.pone.0014108
  • Ortega C, Fernandez AS, Carrillo JM, Romero P, Molina IJ, Moreno JC, Santamaria M. IL-17-producing CD8+T lymphocytes from psoriasis skin plaques are cytotoxic effector cells that secrete Th17-related cytokines. J Leukoc Biol 2009; 86:435-43; PMID:19487306; http://dx.doi.org/10.1189/JLB.0109046
  • Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, Puig L, Nakagawa H, Spelman L, Sigurgeirsson B, Rivas E, et al. ERASURE Study Group; FIXTURE Study Group. Secukinumab in plaque psoriasis – results of two phase 3 trials. N Engl J Med 2014; 371:326-38; PMID:25007392; http://dx.doi.org/10.1056/NEJMoa1314258
  • Blauvelt A, Prinz JC, Gottlieb AB, Kingo K, Sofen H, Ruer-Mulard M, Singh V, Pathan R, Papavassilis C, Cooper S; FEATURE Study Group. Secukinumab administration by pre-filled syringe: efficacy, safety, and usability results from a randomized controlled trial in psoriasis (FEATURE). Br J Dermatol 2015; 172:484-93; PMID:25132411; http://dx.doi.org/10.1111/bjd.13348
  • Paul C, Lacour JP, Tedremets L, Kreutzer K, Jazayeri S, Adams S, Guindon C, You R, Papavassilis C. JUNCTURE Study Group. Efficacy, safety and usability of secukinumab administration by autoinjector/pen in psoriasis: a randomized, controlled trial (JUNCTURE). J Eur Acad Dermatol Venereol 2015; 29:1082-90; PMID:25243910; http://dx.doi.org/10.1111/jdv.12751
  • Mrowietz U, Leonardi CL, Girolomoni G, Toth D, Morita A, Balki SA, Szepietowski JC, Regnault P, Thurston H, Papavassilis C, SCULPTURE Study Group. Secukinumab retreatment-as-needed vs. fixed-interval maintenance regimen for moderate to severe plaque psoriasis: a randomized, double-blind, noninferiority trial (SCULPTURE). J Am Acad Dermatol 2015 Jul; 73(1):277–36.e1; http://dx.doi.org/10.1016/j.jaad.2015.04.011. Epub 2015 May 14.
  • Thaçi D, Humeniuk J, Frambach Y, Bissonnette R, Goodman JJ, Shevade S, Gong Y, Papavassilis C. STATURE Study Group. Secukinumab in psoriasis: randomized, controlled phase 3 trial results assessing the potential to improve treatment response in partial responders (STATURE). Br J Dermatol 2015 Sep; 173(3):777–87; http://dx.doi.org/10.1111/bjd.13814. Epub 2015 May 16.
  • Mease PJ, McInnes IB, Kirkham B, Kavanaugh A, Rahman P, van der Heijde D, Landewé R, Nash P, Pricop L, Yuan J, et al. Secukinumab interleukin-17A inhibition in patients with psoriatic arthritis. N Engl J Med 2015; 373:1329-39; PMID:26422723; http://dx.doi.org/10.1056/NEJMoa1412679
  • McInnes IB, Mease PJ, Kirkham B, Kavanaugh A, Ritchlin CT, Rahman P, van der Heijde D, Landewé R, Conaghan PG, Gottlieb AB, et al. Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015; 386:1137-46; PMID:26135703; http://dx.doi.org/10.1016/S0140-6736(15)61134-5
  • Baeten D, Braun J, Baraliakos X, Sieper J, Dougados M, Emery P, Deodhar AA, Porter B, Martin R, Mpofu S, et al. Secukinumab, a monoclonal antibody to interleukin-17A, significantly improves signs and symptoms of active ankylosing spondylitis: results of a 52-week phase 3 randomized placebo-controlled trial with intravenous loading and subcutaneous maintenance dosing. Arthritis Rheumatol 2014; 66:S360.
  • Sieper J, Braun J, Baraliakos X, Baeten DL, Dougados M, Emery P, Deodhar AA, Porter B, Andersson M, Mpofu S, et al. Secukinumab, a monoclonal antibody to interleukin-17A, significantly improves signs and symptoms of active ankylosing spondylitis: results of a phase 3, randomized, placebo-controlled trial with subcutaneous loading and maintenance dosing. Arthritis Rheumatol 2014; 66:S232; http://dx.doi.org/10.1002/art.38604
  • Mease PJ, McInnes I, Kirkham B, Kavanaugh A, Rahman P, van der Heijde D, Landewé R, Nash P, Pricop L, Yuan J, et al. Secukinumab, a human anti–interleukin-17A monoclonal antibody, improves active psoriatic arthritis and inhibits radiographic progression: efficacy and safety data from a phase 3 randomized, multicenter, double-blind, placebo-controlled study. Arthritis Rheumatol 2014; 66:S423.
  • Xue L, Hickling T, Song R, Nowak J, Rup B. Contribution of enhanced engagement of antigen presentation machinery to the clinical immunogenicity of a human interleukin (IL)-21 receptor-blocking therapeutic antibody. Clin Exp Immunol. 2016 jan; 183(1):102–13; http://dx.doi.org/10.1111/cei.12711
  • Purcell RT, Lockey RF. Immunologic responses to therapeutic biologic agents. J Invest Allergol Clin Immunol 2008; 18(5):335-42
  • Leach MW, Rottman JB, Hock MB, Finco D, Rojko JL, Beyer JC. Immunogenicity/hypersensitivity of biologics. Toxicol Pathol 2014; 42:293-300; PMID:24240973; http://dx.doi.org/10.1177/0192623313510987
  • Descotes J. Immunotoxicity of monoclonal antibodies. mAbs 2009; 1:104-10; PMID:20061816; http://dx.doi.org/10.4161/mabs.1.2.7909
  • De Simone C, Amerio P, Amoruso G, Bardazzi F, Campanati A, Conti A, Gisondi P, Gualdi G, Guarneri C, Leoni L, et al. Immunogenicity of anti-TNFα therapy in psoriasis: a clinical issue. Expert Opin Biol Ther 2013; 13:1673-82; PMID:24107126; http://dx.doi.org/10.1517/14712598.2013.848194
  • Jullien D, Prinz JC, Nestle FO. Immunogenicity of biotherapy used in psoriasis: the science behind the scenes. J Invest Dermatol 2014; 135:31-8; PMID:25120005; http://dx.doi.org/10.1038/jid.2014.295
  • Nelson AL, Dhimolea E, Reichert JM. Development trends for human monoclonal antibody therapeutics. Nat Rev Drug Discov 2010; 9:767-74; PMID:20811384; http://dx.doi.org/10.1038/nrd3229
  • De Groot AS, Scott DW. Immunogenicity of protein therapeutics. Trend Immunol 2007; 28:482-90; http://dx.doi.org/10.1016/j.it.2007.07.011
  • Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies: residual immunogenicity residue in the CDR regions. mAbs 2010; 2:256-65; PMID:20400861; http://dx.doi.org/10.4161/mabs.2.3.11641
  • van Schouwenburg PA, Kruithof S, Votsmeier C, van Schie K, Hart MH, de Jong RN, van Buren EE, van Ham M, Aarden L, Wolbink G, et al. Functional analysis of the anti-adalimumab response using patient-derived monoclonal antibodies. J Biol Chem 2014; 289:34482-8; PMID:25326381; http://dx.doi.org/10.1074/jbc.M114.615500
  • Malucchi S, Bertolotto A. Clinical aspects of immunogenicity to biotherapeutics. In: van de Weert M, Moller EH, eds. Immunogenicity of Biopharmaceuticals. New York, NY: Springer Science-Business Media, LLC. 2008.
  • Rombach-Riegraf V, Karle AC, Wolf B, Sorde L, Koepke S, Gottlieb S, Krieg J, Djidja MC, Baban A, Spindeldreher S, et al. Aggregation of human recombinant monoclonal antibodies influences the capacity of dendritic cells to stimulate adaptive T-cell responses in vitro. PLoS One 2014; 9:e86322; PMID:24466023; http://dx.doi.org/10.1371/journal.pone.0086322
  • Joubert MK, Hokum M, Eakin C, Zhou L, Deshpande M, Baker MP, Goletz TJ, Kerwin BA, Chirmule N, Narhi LO, Jawa V. Highly aggregated antibody therapeutics can enhance the in vitro innate and late-stage T-cell immune responses. J Biol Chem 2012; 287:25266-79; PMID:22584577; http://dx.doi.org/10.1074/jbc.M111.330902
  • Ratanji KD, Derrick JP, Dearman RJ, Kimber I. Immunogenicity of therapeutic proteins: influence of aggregation. J Immunotoxicol 2014; 11:99-109; PMID:23919460; http://dx.doi.org/10.3109/1547691X.2013.821564
  • Van Walle I, Gansemans Y, Parren PW, Stas P, Lasters I. Immunogenicity screening in protein drug development. Expert Opin Biol Ther 2007; 7:405-18; PMID:17309332; http://dx.doi.org/10.1517/14712598.7.3.405
  • Vyas JM, Van der Veen AG, Ploegh HL. The known unknowns of antigen processing and presentation. Nat Rev Immunol 2008; 8:607-18; PMID:18641646; http://dx.doi.org/10.1038/nri2368
  • Chicz RM, Urban RG, Lane WS, Gorga JC, Stern LJ, Vignali DA, Strominger JL. Predominant naturally processed peptides bound to HLA-DR1 are derived from MHC-related molecules and are heterogeneous in size. Nature 1992; 358:764-8; PMID:1380674; http://dx.doi.org/10.1038/358764a0
  • Weber CA, Mehta PJ, Ardito M, Moise L, Martin B, De Groot AS. T cell epitope: friend or foe? Immunogenicity of biologics in context. Adv Drug Deliv Rev 2009; 30:965-76; http://dx.doi.org/10.1016/j.addr.2009.07.001
  • Jawa V, Cousens LP, Awwad M, Wakshull E, Kropshofer H, De Groot AS. T-cell dependent immunogenicity of protein therapeutics: preclinical assessment and mitigation. Clin Immunol 2013; 149:534-55; PMID:24263283; http://dx.doi.org/10.1016/j.clim.2013.09.006
  • Mehra NK. Histocompatibility antigens. Encyclopedia of Life Sciences 2001. Available at: http://web.udl.es/usuaris/e4650869/docencia/GenClin/content/recursos_classe_(pdf)/revisionsPDF/HLA.pdf. Accessed 18 June 2015.
  • Shiina T, Hosomichi K, Inoko H, Kulski JK. The HLA genomic loci map: expression, interaction, diversity and disease. J Hum Genet 2009; 54:15-39; PMID:19158813; http://dx.doi.org/10.1038/jhg.2008.5
  • Oseroff C, Sidney J, Vita R, Tripple V, McKinney DM, Southwood S, Brodie TM, Sallusto F, Grey H, Alam R, et al. T cell responses to known allergen proteins are differently polarized and account for a variable fraction of total response to allergen extracts. J Immunol 2012; 189:1800-11; PMID:22786768; http://dx.doi.org/10.4049/jimmunol.1200850
  • McKinney DM, Southwood S, Hinz D, Oseroff C, Arlehamn CS, Schulten V, Taplitz R, Broide D, Hanekom WA, Scriba TJ, et al. A strategy to determine HLA class II restriction broadly covering the DR, DP, and DQ allelic variants most commonly expressed in the general population. Immunogenetics 2013; 65:357-70; PMID:23392739; http://dx.doi.org/10.1007/s00251-013-0684-y
  • De Groot AS, Tassone R, Cousens LP, Terry F, Martin R, Ardito M, Martin W. In silico prediction of HLA-DP and -DQ epitope content is poorly correlated with clinical immunogenicity of therapeutic proteins, manuscript in preparation, Abstract presented at the American Association of Pharmaceutical Scientists (AAPS) National Biotechnology Conference (NBC), San Diego, CA, USA. May 20-22, 2013. (http://www.epivax.com/wp-content/uploads/2013/04/EpiVax_AAPS_DPDQ_Poster_16May13_Final.pdf).
  • Hsu L, Snodgrass BT, Armstrong AW. Antidrug antibodies in psoriasis: a systematic review. Br J Dermatol 2014; 170:261-73; PMID:24117166; http://dx.doi.org/10.1111/bjd.12654
  • Bito T, Nishikawa R, Hatakeyama M, Kikusawa A, Kanki H, Nagai H, Sarayami Y, Ikeda T, Yoshizaki H, Seto H, et al. Influence of neutralizing antibodies to adalimumab and infliximab on the treatment of psoriasis. Br J Dermatol 2014; 170:922-9; PMID:24329764; http://dx.doi.org/10.1111/bjd.12791
  • Carrascosa JM. Immunogenicity in biologic therapy: implications for dermatology. Actas Dermosifiliogr 2013; 104:471-9; PMID:23622932; http://dx.doi.org/10.1016/j.ad.2013.02.005
  • Farhangian ME, Feldman SR. Immunogenicity of biologic treatments for psoriasis: therapeutic consequences and the potential value of concomitant methotrexate. Am J Clin Dermatol 2015 Aug; 16(4):285-94. http://dx.doi.org/ 10.1007/s40257–015–0131–y
  • Anthony DD, Milkovich KA, Zhang W, Rodriguez B, Yonkers NL, Tary-Lehmann M, Lehmann PV. Dissecting the T cell response: proliferation assays vs. cytokine signatures by ELISPOT. Cells 2012; 1:127-40; PMID:24710419; http://dx.doi.org/10.3390/cells1020127
  • Kropshofer H, Spindeldreher S, Röhn TA, Platania N, Grygar C, Daniel N, Wölpl A, Langen H, Horejsi V, Vogt AB. Tetraspan microdomains distinct from lipid rafts enrich select peptide-MHC class II complexes. Nat Immunol 2002; 3:61-8; PMID:11743588; http://dx.doi.org/10.1038/ni750
  • Röhn TA, Reitz A, Paschen A, Nguyen XD, Schadendorf D, Voight AB, Kropshofer H. A novel strategy for the discovery of MHC class II-restricted tumor antigens: identification of a melanotransferrin helper T-cell epitope. Cancer Res 2005; 65:10068-78; PMID:16267033; http://dx.doi.org/10.1158/0008-5472.CAN-05-1973
  • Kropshofer H, Singer T. Overview of cell-based tools for pre-clinical assessment of immunogenicity of biotherapeutics. J Immunotoxicol 2006; 3:131-6; PMID:18958693; http://dx.doi.org/10.1080/15476910600845625
  • Rup B, Pallardy M, Sikkema D, Albert T, Allez M, Broet P, Carini C, Creeke P, Davidson J, De Vries N, et al. ABIRISK Consortium. Standardizing terms, definitions and concepts for describing and interpreting unwanted immunogenicity of biopharmaceuticals: recommendations of the innovative medicines initiative ABIRISK consortium. Clin Exp Immunol 2015 May 8; 181(3):385-400; http://dx.doi.org/10.1111/cei.12652. [Epub ahead of print].
  • Shankar G, Arkin S, Cocea L, Devanarayan V, Kirshner S, Kromminga A, Quarmby V, Richards S, Schneider CK, Subramanyam M, Swanson S, et al. American Association of Scientists. Assessment and reporting of the clinical immunogenicity of therapeutic proteins and peptides-harmonized terminology and tactical recommendations. AAPS J 2014; 16:658-73; PMID:24764037; http://dx.doi.org/10.1208/s12248-014-9599-2
  • Tovey MG, Legrand J, Lallemand C. Overcoming immunogenicity associated with the use of biopharmaceuticals. Expert Rev Clin Pharmacol 2011; 4:623-631; PMID:22114889; http://dx.doi.org/10.1586/ecp.11.39
  • Bendtzen K. Immunogenicity of Anti-TNF-α Biotherapies: I. Individualized medicine based on immunopharmacological evidence. Front Immunol 2015 Apr 8; 6:152; http://dx.doi.org/10.3389/fimmu.2015.00152
  • Jaber A, Baker M. Assessment of the immunogenicity of different interferon β-1a formulations using ex vivo T-cell assays. J Pharm Biomed Anal 2007; 43:1256-61; PMID:17118612; http://dx.doi.org/10.1016/j.jpba.2006.10.023
  • Sauerborn M. The Immunogenicity of Therapeutic Antibodies. Handbook of Therapeutic Antibodies: Wiley-VCH Verlag GmbH & Co. KGaA. 2014; pp. 665-680.
  • Wullner D, Zhou L, Bramhall E, Kuck A, Goletz TJ, Swanson S, Chirmule N, Jawa V. Considerations for optimization and validation of an in vitro PBMC derived T cell assay for immunogenicity prediction of biotherapeutics. Clin Immunol 2010; 137: 5-14; PMID:20708973; http://dx.doi.org/10.1016/j.clim.2010.06.018
  • Kim A, Sadegh-Nasseri S. Determinants of immunodominance for CD4 T cells. Curr Opin Immunol 2015; 34:9-15; PMID:25576665; http://dx.doi.org/10.1016/j.coi.2014.12.005
  • Karle AC, Oostingh GJ, Mutschlechner S, Ferreira F, Lackner P, Bohle B, Fischer GF, Vogt AB, Duschl A. Nitration of the pollen allergen bet v 1.0101 enhances the presentation of bet v 1-derived peptides by HLA-DR on human dendritic cells. PLoS One 2012; 7(2):e31483; PMID:22348091; http://dx.doi.org/10.1371/journal.pone.0031483
  • Scott AM, Lee FT, Jones R, Hopkins W, MacGregor D, Cebon JS, Hannah A, Chong G, Papenfuss A, Rigopoulos A, et al. A phase I trial of humanized monoclonal antibody A33 in patients with colorectal carcinoma: biodistribution, pharmacokinetics, and quantitative tumor uptake. Clin Cancer Res 2005; 11:4810-7; PMID:16000578; http://dx.doi.org/10.1158/1078-0432.CCR-04-2329
  • Carrasquillo JA, Pandit-Taskar N, O'Donoghue JA, Humm JL, Zanzonico P, Smith-Jones PM, Divgi CR, Pryma DA, Ruan S, Kemeny NE et al. 124I-huA33 antibody PET of colorectal cancer. J Nucl Med 2011; 52:1173-80; PMID:21764796; http://dx.doi.org/10.2967/jnumed.110.086165
  • Ting YT, Temme S, Koch N, McLellan AD. A new monoclonal antibody recognizing a linear determinant on the HL-DRα chain N-terminus. Hybridoma 2009; 28:423-9; PMID:20025501; http://dx.doi.org/10.1089/hyb.2009.0050
  • Hsu L, Armstrong AW. Anti-drug antibodies in psoriasis: a critical evaluation of clinical significance and impact on treatment response. Expert Rev Clin Immunol 2013; 9:949-58; PMID:24128157; http://dx.doi.org/10.1586/1744666X.2013.836060
  • Krieckaert CL, Bartelds GM, Lerns WF, Wolbink GJ. The effect of immunomodulators on the immunogenicity of TNF-blocking therapeutic monoclonal antibodies: a review. Arthritis Res Ther 2010; 12:217-22; PMID:21029481; http://dx.doi.org/10.1186/ar3147
  • Mazilu D, Opris D, Gainaru C, Iliuta M, Apetrei N, Luca G, Borangiu A, Gudu T, Peltea A, Gorseanu L, et al. Monitoring drug and antidrug levels: a rational approach in rheumatoid arthritis patients treated with biologic agents who experience inadequate response while being on a stable biologic treatment. Biomed Res Int 2014:702701; http://dx.doi.org/10.1155/2014/702701
  • de Vries MK, Wolbink GJ, Stapel SO, de Vrieze H, van Denderen JC, Dijkmans BA, Aarden LA, van der Horst-Bruinsma IE. Decreased clinical response to infliximab in ankylosing spondylitis is correlated with anti-infliximab formation. Ann Rheum Dis 2007; 66:1252-4; PMID:17472991; http://dx.doi.org/10.1136/ard.2007.072397
  • Zisapel M, Zisman D, Madar-Balakirski N, Arad U, Padova H, Matz H, Maman-Sarvagyl H, Kaufman I, Paran D, Feld J, et al. Prevalence of TNF-α blocker immunogenicity in psoriatic arthritis. J Rheumatol 2015; 42:73-8; PMID:25399390
  • Krieckaert CL, Jamnitski A, Nurmohamed MT, Kostense PJ, Boers M, Wolbink G. Comparison of long-term clinical outcome with etanercept treatment and adalimumab treatment of rheumatoid arthritis with respect to immunogenicity. Arthritis Rheum 2012; 64:3850-5; PMID:22933315; http://dx.doi.org/10.1002/art.34680
  • de Vries MK, Brouwer E, van der Horst-Bruinsma IE, Spoorenberg A, van Denderen JC, Jamnitski A, Nurmohamed MT, Dijkmans BA, Aarden LA, Wolbink GJ. Decreased clinical response to adalimumab in ankylosing spondylitis is associated with antibody formation. Ann Rheum Dis 2009; 68:1787-8; PMID:19822712; http://dx.doi.org/10.1136/ard.2009.109702
  • Paramarta JE, Baeten DL. Adalimumab serum levels and antidrug antibodies towards adalimumab in peripheral spondyloarthritis: no association with clinical response to treatment or with disease relapse upon treatment discontinuation. Arthritis Res Ther 2014; 16:R160; PMID:25074046; http://dx.doi.org/10.1186/ar4675
  • van Kujik AW, de Groot M, Stapel SO, Dijkmans BA, Wolbink GJ, Tak PP. Relationship between the clinical response to adalimumab treatment and serum levels of adalimumab and anti-adalimumab antibodies in patients with psoriatic arthritis. Ann Rheum Dis 2010; 69:624-5; PMID:20223840; http://dx.doi.org/10.1136/ard.2009.108787
  • van Vollenhoven RF, Emery P, Bingham CO 3rd, Keystone EC, Fleischmann R, Furst DE, Macey K, Sweetser M, Kelman A, Rao R. Longterm safety of patients receiving rituximab in rheumatoid arthritis clinical trials. J Rheumatol 2010; 37:558-67; PMID:20110520; http://dx.doi.org/10.3899/jrheum.090856
  • Mok CC. Rituximab for the treatment of rheumatoid arthritis: an update. Drug Des Devel Ther 2013; 4:87-100; http://dx.doi.org/10.2147/DDDT.S41645
  • Ritchlin C, Rahman P, Kavanaugh A, McInnes IB, Puig L, Li S, Wang Y, Shen YK, Doyle DK, Mendelsohn AM, et al. PSUMMIT 2 Study Group. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumor necrosis factor therapy: 6-month and 1-year results of the phase 3, multicentre, double-blind, placebo-controlled, randomised PSUMMIT 2 trial. Ann Rheum Dis 2014; 73:990-9; PMID:24482301; http://dx.doi.org/10.1136/annrheumdis-2013-204655
  • Sandborn WJ, Gasink C, Gao LL, Blank MA, Johanns J, Guzzo C, Sands BE, Hanauer SB, Targan S, Rutgeerts P, et al. CERTIFI Study Group. Ustekinumab induction and maintenance therapy in refractory Crohn's disease. N Engl J Med 2012; 367:1519-28; PMID:23075178; http://dx.doi.org/10.1056/NEJMoa1203572
  • Emi Aikawa N, de Carvalho JF, Artur Alemida Silva C, Bonfa E. Immunogenicity of anti-TNF-α agents in autoimmune diseases. Clin Rev Allergy Immunol 2010; 38:82-9; PMID:19565360; http://dx.doi.org/10.1007/s12016-009-8140-3
  • Genovese MC, Durez P, Richards HB, Supronik J, Dokoupilova E, Mazurov V, Aelion JA, Lee SH, Codding CE, Kellner H, et al. Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study. Ann Rheum Dis 2013; 72:863-9; PMID:22730366; http://dx.doi.org/10.1136/annrheumdis-2012-201601