Phase 1 double-blind randomized safety trial of the Janus kinase inhibitor tofacitinib in systemic lupus erythematosus

Sarfaraz A Hasni, Sarthak Gupta, Michael Davis, Elaine Poncio, Yenealem Temesgen-Oyelakin, Philip M Carlucci, Xinghao Wang, Mohammad Naqi, Martin P Playford, Rishi R Goel, Xiaobai Li, Ann J Biehl, Isabel Ochoa-Navas, Zerai Manna, Yinghui Shi, Donald Thomas, Jinguo Chen, Angélique Biancotto, Richard Apps, Foo Cheung, Yuri Kotliarov, Ashley L Babyak, Huizhi Zhou, Rongye Shi, Katie Stagliano, Wanxia Li Tsai, Laura Vian, Nathalia Gazaniga, Valentina Giudice, Shajia Lu, Stephen R Brooks, Meggan MacKay, Peter Gregersen, Nehal N Mehta, Alan T Remaley, Betty Diamond, John J O'Shea, Massimo Gadina, Mariana J Kaplan, Sarfaraz A Hasni, Sarthak Gupta, Michael Davis, Elaine Poncio, Yenealem Temesgen-Oyelakin, Philip M Carlucci, Xinghao Wang, Mohammad Naqi, Martin P Playford, Rishi R Goel, Xiaobai Li, Ann J Biehl, Isabel Ochoa-Navas, Zerai Manna, Yinghui Shi, Donald Thomas, Jinguo Chen, Angélique Biancotto, Richard Apps, Foo Cheung, Yuri Kotliarov, Ashley L Babyak, Huizhi Zhou, Rongye Shi, Katie Stagliano, Wanxia Li Tsai, Laura Vian, Nathalia Gazaniga, Valentina Giudice, Shajia Lu, Stephen R Brooks, Meggan MacKay, Peter Gregersen, Nehal N Mehta, Alan T Remaley, Betty Diamond, John J O'Shea, Massimo Gadina, Mariana J Kaplan

Abstract

Increased risk of premature cardiovascular disease (CVD) is well recognized in systemic lupus erythematosus (SLE). Aberrant type I-Interferon (IFN)-neutrophil interactions contribute to this enhanced CVD risk. In lupus animal models, the Janus kinase (JAK) inhibitor tofacitinib improves clinical features, immune dysregulation and vascular dysfunction. We conducted a randomized, double-blind, placebo-controlled clinical trial of tofacitinib in SLE subjects (ClinicalTrials.gov NCT02535689). In this study, 30 subjects are randomized to tofacitinib (5 mg twice daily) or placebo in 2:1 block. The primary outcome of this study is safety and tolerability of tofacitinib. The secondary outcomes include clinical response and mechanistic studies. The tofacitinib is found to be safe in SLE meeting study's primary endpoint. We also show that tofacitinib improves cardiometabolic and immunologic parameters associated with the premature atherosclerosis in SLE. Tofacitinib improves high-density lipoprotein cholesterol levels (p = 0.0006, CI 95%: 4.12, 13.32) and particle number (p = 0.0008, CI 95%: 1.58, 5.33); lecithin: cholesterol acyltransferase concentration (p = 0.024, CI 95%: 1.1, -26.5), cholesterol efflux capacity (p = 0.08, CI 95%: -0.01, 0.24), improvements in arterial stiffness and endothelium-dependent vasorelaxation and decrease in type I IFN gene signature, low-density granulocytes and circulating NETs. Some of these improvements are more robust in subjects with STAT4 risk allele.

Conflict of interest statement

The NIH and J.J.O.S. have a patent related to JAK inhibitors and receive royalties. The NIH and J.J.O.S. have had a collaborative agreement and development award (CRADA) with Pfizer that pertains to JAK inhibition and tofacitinib. The NIH and J.J.O.S. have an ongoing CRADA for new JAK inhibitors. The remaining authors declare no competing interests.

Figures

Fig. 1. Tofacitinib inhibits STAT1 phosphorylation in…
Fig. 1. Tofacitinib inhibits STAT1 phosphorylation in CD4 + T cells.
a Significant inhibition of pSTAT1 in patients on tofacitinib at day 56 (P = 0.023) with the return to baseline at day 84 n = 20 biologically independent samples. Data are presented as mean values + /− SEM. A mixed linear model for repeated measures was used. b No significant change noted in patients on placebo. n = 10 biologically independent samples. Data are presented as mean values + /− SEM. A mixed linear model for repeated measures was used. c Tofacitinib decreases the type I IFN gene signature, gene expression in peripheral blood by Nanostring: Heatmap showing interferon-stimulated gene expression in peripheral blood by nanostring: Log2 mean fold change in expression of interferon-stimulated genes from baseline to day 56 and 84 in subjects on tofacitinib (n = 20 biologically independent samples) and placebo (n = 10 biologically independent samples). Source data are provided as a Source Data file. Results shown as fold change in stimulated vs. unstimulated cell population. A mixed linear model for repeated measures was used. Unpaired two-tailed, t test were used where appropriate. No adjustments were made for multiple comparisons.
Fig. 2. Circulating LDGs and NETs are…
Fig. 2. Circulating LDGs and NETs are modulated by tofacitinib.
a Results represent changes in percentage of circulating LDGs: Subjects treated with tofacitinib vs. placebo. Significant decrease in LDGs in tofacitinib group P = 0.048 at day 56 and P = 0.014 at day 84 using unpaired t test. b Changes in absolute neutrophil counts. c Circulating NET levels at baseline: Individuals positive for STAT4 risk allele (each subject represented by closed circles) have enhanced circulating NET levels (assessed by human neutrophil elastase (HNE)-dsDNA complexes) then subjects who are STAT4 risk allele negative (each subject represented by open circles) P = 0.02 using unpaired t test. d Changes in circulating NET levels during the study: Patients who are STAT4 risk allele positive and receive tofacitinib display significant decrease in circulating NET complexes P = 0.037 using unpaired t test. Source data are provided as a Source Data file. All data represent mean ± SEM, *P < 0.05, and are based on tofacitinib n = 20, placebo n = 10. Unpaired two-tailed, t test were used for all results. No adjustments were made for multiple comparisons.
Fig. 3. Tofacitinib modulates HDL levels and…
Fig. 3. Tofacitinib modulates HDL levels and function in SLE.
a Percent change in serum HDL-C at day 56 compared to day 1 based on STAT4 risk allele status (each circle and square represent individual subject): Results represent *P = 0.037 for the difference between STAT4 risk allele-positive subjects on tofacitinib vs placebo and **P = 0.002 STAT4 risk allele-negative subjects on tofacitinib vs placebo. Unpaired t test was used. b Percent change in Lecithin–cholesterol acyltransferase (LCAT) concentration at day 56 compared to day 1 based on STAT4 risk allele status (each circle and square represent individual subject): Results represent *P = 0.033 for the difference between STAT4 risk allele-positive subjects on tofacitinib vs placebo and *P = 0.044 for the difference between STAT4 risk allele-positive subjects on tofacitinib vs STAT4 risk allele-negative subjects on tofacitinib. Kruskal–Wallis test (unpaired, nonparametric) was used. Source data are provided as a Source Data file. All results represent mean ± SEM, *P < 0.05 **P < 0.01, and are based on tofacitinib n = 20, placebo n = 10. Two-tailed tests were used where appropriate. No adjustments were made for multiple comparisons.
Fig. 4. Tofacitinib improves arterial stiffness and…
Fig. 4. Tofacitinib improves arterial stiffness and endothelial dysfunction in association with STAT4 risk allele status.
a Right cardioankle vascular index (CAVI): Results represent changes in CAVI in subjects on tofacitinib vs placebo in STAT4 risk allele-positive and -negative subjects during the trial. b Pulse wave velocity (PWV): Results represent changes in PWV in subjects on tofacitinib vs placebo in STAT4 risk allele-positive and -negative subjects during the trial. c Reactive hyperemia index (RHI): Results represent changes in RHI in subjects on tofacitinib vs placebo in STAT4 risk allele-positive and -negative subjects during the trial. Source data are provided as a Source Data file. All results represent mean ± SEM and are based on tofacitinib n = 20, placebo n = 10. The paired t test, Mann–Whitney U, or ANOVA were used for comparison where appropriate based on normality of distribution. Two-tailed tests were used where appropriate. No adjustments were made for multiple comparisons.

References

    1. Tsokos GC. Systemic lupus erythematosus. N. Engl. J. Med. 2011;365:2110–2121.
    1. Weidenbusch M, Kulkarni OP, Anders HJ. The innate immune system in human systemic lupus erythematosus. Clin. Sci. 2017;131:625–34..
    1. Manzi S, et al. Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham Study. Am. J. Epidemiol. 1997;145:408–415.
    1. Giannelou M, Mavragani CP. Cardiovascular disease in systemic lupus erythematosus: a comprehensive update. J. Autoimmun. 2017;82:1–12.
    1. Sánchez P, Toro-Trujillo E, Muñoz-Velandia OM, García AA, Fernández-Ávila DG. Therapeutic Impact of Statins on the Lipid Profile and Cardiovascular Risk in Patients With Systemic Lupus Erythematosus: Systematic Review of the Literature and a Meta-analysis. Reumatol Clin. 2019;15:e86–e91.
    1. Liu Y, Kaplan MJ. Cardiovascular disease in systemic lupus erythematosus: an update. Curr. Opin. Rheumatol. 2018;30:441–448.
    1. Carlucci PM, et al. Neutrophil subsets and their gene signature associate with vascular inflammation and coronary atherosclerosis in lupus. JCI Insight. 2018;3:e99276.
    1. Goel, R. R. & Kaplan, M. J. Deadliest catch: neutrophil extracellular traps in autoimmunity. Curr. Opin. Rheumatol.2, 64–70 (2019).
    1. O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N. Engl. J. Med. 2013;368:161–170.
    1. Dean GS, Tyrrell-Price J, Crawley E, Isenberg DA. Cytokines and systemic lupus erythematosus. Ann. Rheum. Dis. 2000;59:243–251.
    1. Alunno, A., Padjen, I., Fanouriakis, A. & Boumpas, D. T. Pathogenic and therapeutic relevance of JAK/STAT signaling in systemic lupus erythematosus: integration of distinct inflammatory pathways and the prospect of their inhibition with an oral agent. Cells8, 898 (2019).
    1. Furumoto Y, et al. Tofacitinib ameliorates murine lupus and its associated vascular dysfunction. Arthritis Rheumatol. 2017;69:148–160.
    1. You, H. et al. Successful treatment of arthritis and rash with tofacitinib in systemic lupus erythematosus: the experience from a single centre. Ann. Rheum. Dis.78, 1441–1443 (2019).
    1. Wallace DJ, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2018;392:222–31..
    1. Taylor KE, et al. Specificity of the STAT4 genetic association for severe disease manifestations of systemic lupus erythematosus. PLoS Genet. 2008;4:e1000084.
    1. Svenungsson E, et al. A STAT4 risk allele is associated with ischaemic cerebrovascular events and anti-phospholipid antibodies in systemic lupus erythematosus. Ann. Rheum. Dis. 2010;69:834–840.
    1. Kariuki SN, et al. Cutting edge: autoimmune disease risk variant of STAT4 confers increased sensitivity to IFN-alpha in lupus patients in vivo. J. Immunol. 2009;182:34–38.
    1. Furie R, et al. Anifrolumab, an anti-interferon-alpha receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol. 2017;69:376–86..
    1. Futosi K, Fodor S, Mocsai A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int. Immunopharmacol. 2013;17:638–650.
    1. Taraborelli M, et al. Endothelial dysfunction in early systemic lupus erythematosus patients and controls without previous cardiovascular events. Arthritis care Res. 2018;70:1277–1283.
    1. Tziomalos K, et al. Arterial stiffness and peripheral arterial disease in patients with systemic lupus erythematosus. Rheumatol. Int. 2017;37:293–298.
    1. Wohlfahrt P, et al. Reference values of cardio-ankle vascular index in a random sample of a white population. J. Hypertens. 2017;35:2238–44.
    1. Mendoza-Pinto C, et al. Endothelial dysfunction and arterial stiffness in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Atherosclerosis. 2020;297:55–63.
    1. Bruce IN. ‘Not only…but also’: factors that contribute to accelerated atherosclerosis and premature coronary heart disease in systemic lupus erythematosus. Rheumatology. 2005;44:1492–1502.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–31.
    1. Knight JS, et al. Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis. Circulation Res. 2014;114:947–956.
    1. Knight JS, et al. Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. J. Clin. Investig. 2013;123:2981–2993.
    1. Thacker SG, et al. Type I interferons modulate vascular function, repair, thrombosis, and plaque progression in murine models of lupus and atherosclerosis. Arthritis Rheum. 2012;64:2975–2985.
    1. Carmona-Rivera C, Zhao W, Yalavarthi S, Kaplan MJ. Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrix metalloproteinase-2. Ann. Rheum. Dis. 2015;74:1417–1424.
    1. Chang HH, Dwivedi N, Nicholas AP, Ho IC. The W620 polymorphism in PTPN22 disrupts its interaction with peptidylarginine deiminase type 4 and enhances citrullination and NETosis. Arthritis Rheumatol. 2015;67:2323–2334.
    1. Odqvist L, et al. Genetic variations in A20 DUB domain provide a genetic link to citrullination and neutrophil extracellular traps in systemic lupus erythematosus. Ann. Rheum. Dis. 2019;78:1363–70..
    1. Garcia-Romo GS, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 2011;3:73ra20.
    1. Morand EF, et al. Trial of anifrolumab in active systemic lupus erythematosus. N. Engl. J. Med. 2020;382:211–21.
    1. Furie R, et al. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus erythematosus. J. Clin. Investig. 2019;129:1359–71.
    1. Purmalek MM, et al. Association of lipoprotein subfractions and glycoprotein acetylation with coronary plaque burden in SLE. Lupus Sci. Med. 2019;6:e000332.
    1. McMahon M, et al. Dysfunctional proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheum. 2009;60:2428–2437.
    1. Smith CK, et al. Lupus high-density lipoprotein induces proinflammatory responses in macrophages by binding lectin-like oxidised low-density lipoprotein receptor 1 and failing to promote activating transcription factor 3 activity. Ann. Rheum. Dis. 2017;76:602–11.
    1. Yang X, Wan M, Cheng Z, Wang Z, Wu Q. Tofacitinib inhibits ox-LDL-induced adhesion of THP-1 monocytes to endothelial cells. Artif. Cells Nanomed. Biotechnol. 2019;47:2775–82.
    1. Taghavie-Moghadam PL, et al. STAT4 regulates the CD8(+) regulatory T cell/T follicular helper cell axis and promotes atherogenesis in insulin-resistant Ldlr(−/−) mice. J. Immunol. 2017;199:3453–65.
    1. Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40:1725.
    1. Pincus T, Swearingen C, Wolfe F. Toward a multidimensional Health Assessment Questionnaire (MDHAQ): assessment of advanced activities of daily living and psychological status in the patient-friendly health assessment questionnaire format. Arthritis Rheum. 1999;42:2220–2230.
    1. Gompertz P, Harwood R, Ebrahim S, Dickinson E. Validating the SF-36. BMJ (Clin. Res. Ed.) 1992;305:645–646.
    1. Gladman DD, Ibanez D, Urowitz MB. Systemic lupus erythematosus disease activity index 2000. J. Rheumatol. 2002;29:288–91.
    1. Isenberg DA, et al. An assessment of disease flare in patients with systemic lupus erythematosus: a comparison of BILAG 2004 and the flare version of SELENA. Ann. Rheum. Dis. 2011;70:54–59.
    1. Aranow C. A pilot study to determine the optimal timing of the Physician Global Assessment (PGA) in patients with systemic lupus erythematosus. Immunol. Res. 2015;63:167–169.
    1. Villaverde V, et al. Activity indices in rheumatoid arthritis. J. Rheumatol. 2000;27:2576–2581.
    1. Tsai, W. L. et al. High throughput pSTAT signaling profiling by fluorescent cell barcoding and computational analysis. J. Immunol. Methods477, 112667 (2020).
    1. Denny MF, et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J. Immunol. 2010;184:3284–3297.
    1. Lood C, et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 2016;22:146–153.
    1. Matthews DR, et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–419.
    1. Mehta NN, et al. Abnormal lipoprotein particles and cholesterol efflux capacity in patients with psoriasis. Atherosclerosis. 2012;224:218–221.
    1. Hasni S, et al. Safety and tolerability of omalizumab: a randomized clinical trial of humanized anti-IgE monoclonal antibody in systemic lupus erythematosus. Arthritis Rheumatol. 2019;71:1135–1140.
    1. Hartmann, S. et al. A clinical population pharmacokinetic/pharmacodynamic model for BIIB059, a monoclonal antibody for the treatment of systemic and cutaneous lupus erythematosus. J. Pharmacokinet. Pharmacodyn.47, 255–266 (2020).
    1. Tsai WL, et al. High throughput pSTAT signaling profiling by fluorescent cell barcoding and computational analysis. J. Immunol. Methods. 2020;477:112667.

Source: PubMed

3
Tilaa