JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies

Gina A Montealegre Sanchez, Adam Reinhardt, Suzanne Ramsey, Helmut Wittkowski, Philip J Hashkes, Yackov Berkun, Susanne Schalm, Sara Murias, Jason A Dare, Diane Brown, Deborah L Stone, Ling Gao, Thomas Klausmeier, Dirk Foell, Adriana A de Jesus, Dawn C Chapelle, Hanna Kim, Samantha Dill, Robert A Colbert, Laura Failla, Bahar Kost, Michelle O'Brien, James C Reynolds, Les R Folio, Katherine R Calvo, Scott M Paul, Nargues Weir, Alessandra Brofferio, Ariane Soldatos, Angelique Biancotto, Edward W Cowen, John J Digiovanna, Massimo Gadina, Andrew J Lipton, Colleen Hadigan, Steven M Holland, Joseph Fontana, Ahmad S Alawad, Rebecca J Brown, Kristina I Rother, Theo Heller, Kristina M Brooks, Parag Kumar, Stephen R Brooks, Meryl Waldman, Harsharan K Singh, Volker Nickeleit, Maria Silk, Apurva Prakash, Jonathan M Janes, Seza Ozen, Paul G Wakim, Paul A Brogan, William L Macias, Raphaela Goldbach-Mansky, Gina A Montealegre Sanchez, Adam Reinhardt, Suzanne Ramsey, Helmut Wittkowski, Philip J Hashkes, Yackov Berkun, Susanne Schalm, Sara Murias, Jason A Dare, Diane Brown, Deborah L Stone, Ling Gao, Thomas Klausmeier, Dirk Foell, Adriana A de Jesus, Dawn C Chapelle, Hanna Kim, Samantha Dill, Robert A Colbert, Laura Failla, Bahar Kost, Michelle O'Brien, James C Reynolds, Les R Folio, Katherine R Calvo, Scott M Paul, Nargues Weir, Alessandra Brofferio, Ariane Soldatos, Angelique Biancotto, Edward W Cowen, John J Digiovanna, Massimo Gadina, Andrew J Lipton, Colleen Hadigan, Steven M Holland, Joseph Fontana, Ahmad S Alawad, Rebecca J Brown, Kristina I Rother, Theo Heller, Kristina M Brooks, Parag Kumar, Stephen R Brooks, Meryl Waldman, Harsharan K Singh, Volker Nickeleit, Maria Silk, Apurva Prakash, Jonathan M Janes, Seza Ozen, Paul G Wakim, Paul A Brogan, William L Macias, Raphaela Goldbach-Mansky

Abstract

Background: Monogenic IFN-mediated autoinflammatory diseases present in infancy with systemic inflammation, an IFN response gene signature, inflammatory organ damage, and high mortality. We used the JAK inhibitor baricitinib, with IFN-blocking activity in vitro, to ameliorate disease.

Methods: Between October 2011 and February 2017, 10 patients with CANDLE (chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperatures), 4 patients with SAVI (stimulator of IFN genes-associated [STING-associated] vasculopathy with onset in infancy), and 4 patients with other interferonopathies were enrolled in an expanded access program. The patients underwent dose escalation, and the benefit was assessed by reductions in daily disease symptoms and corticosteroid requirement. Quality of life, organ inflammation, changes in IFN-induced biomarkers, and safety were longitudinally assessed.

Results: Eighteen patients were treated for a mean duration of 3.0 years (1.5-4.9 years). The median daily symptom score decreased from 1.3 (interquartile range [IQR], 0.93-1.78) to 0.25 (IQR, 0.1-0.63) (P < 0.0001). In 14 patients receiving corticosteroids at baseline, daily prednisone doses decreased from 0.44 mg/kg/day (IQR, 0.31-1.09) to 0.11 mg/kg/day (IQR, 0.02-0.24) (P < 0.01), and 5 of 10 patients with CANDLE achieved lasting clinical remission. The patients' quality of life and height and bone mineral density Z-scores significantly improved, and their IFN biomarkers decreased. Three patients, two of whom had genetically undefined conditions, discontinued treatment because of lack of efficacy, and one CANDLE patient discontinued treatment because of BK viremia and azotemia. The most common adverse events were upper respiratory infections, gastroenteritis, and BK viruria and viremia.

Conclusion: Upon baricitinib treatment, clinical manifestations and inflammatory and IFN biomarkers improved in patients with the monogenic interferonopathies CANDLE, SAVI, and other interferonopathies. Monitoring safety and efficacy is important in benefit-risk assessment.

Trial registration: ClinicalTrials.gov NCT01724580 and NCT02974595.

Funding: This research was supported by the Intramural Research Program of the NIH, NIAID, and NIAMS. Baricitinib was provided by Eli Lilly and Company, which is the sponsor of the expanded access program for this drug.

Keywords: Immunology; Innate immunity; Monogenic diseases; Therapeutics; Translation.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1. Expanded access program overview and…
Figure 1. Expanded access program overview and effect of baricitinib treatment on clinical outcomes.
(A) Expanded access program overview. Phase 1: Time before the first baricitinib dose. Phase 2: Period of dose escalation, including the time between the first baricitinib dose and achievement of an optimal dose regimen. Phase 3: Time on optimal baricitinib doses, excluding the last 90 days prior to the final visit. Phase 4: Ninety days prior to the final visit, with analysis of primary data (daily diaries, steroid doses, and biomarkers of IFN signaling). The program is ongoing. #The number of days in each phase is reported as the mean ± SD. For phases 2 and 3, patients O1 and O3 were not included in the calculation. Both patients discontinued treatment because of a lack of efficacy and/or osteonecrosis after only 77 and 56 days on optimal doses, respectively. (B) Effect of baricitinib treatment on clinical outcomes. To confirm trends in longitudinally collected data, the diary scores and corticosteroid doses were fitted to a repeated-measures model with “phase” as a categorical independent variable. Least-squares means with 95% CIs for each phase were assessed. *P < 0.05 and **P < 0.001 (both unadjusted) by 2-sided paired Student’s t test.
Figure 2. Self-reported and physician’s global assessments…
Figure 2. Self-reported and physician’s global assessments by disease subgroup.
Parent’s or patient’s overall assessment of pain and health (Pt. global) and the physician’s assessment (MD global) were measured using a visual analog scale (VAS), in which a value of 100 mm indicates the worst possible measure for the condition assessed by the test. Quality of life (PedsQL) was measured using a standardized age-matched test that ranged from 0% to 100%, with higher percentages indicating improvement. Data are presented by disease, with CANDLE in red, SAVI in blue, and other interferonopathies in green. Only the 2 patients who stayed in the study are shown. Darker shades indicate pretreatment, and lighter shades indicate the last included visit on baricitinib treatment. **P < 0.05 and ***P < 0.001 (both unadjusted) by 2-sided paired Student’s t test.
Figure 3. Improvement in clinical disease manifestations…
Figure 3. Improvement in clinical disease manifestations in CANDLE and SAVI patients treated with baricitinib.
(AD) Images of 2 patients with CANDLE who achieved the remission criteria (C2 and C10, respectively) are shown. Pretreatment images of the face show typical distribution of facial panniculitis with periorbital swelling and erythema as well as lipodystrophy affecting temporal regions and areas above and below the zygomatic bone. Lip swelling is also evident. Post-treatment images show complete resolution of areas of panniculitis on the face and neck. (E and F) Images of 2 of the 4 patients with SAVI are shown. Images of the lower leg of a SAVI patient (S3) show extensive eschar formation overlying infected, nonhealing ulcers on the left lower leg. After treatment, the ulcers healed, with complete reepithelialization. (G and H) Images of the right palmar surface of the hand of a patient with SAVI (S4) show chronic cutaneous vasculitis that resulted in partial amputation of the second and third fingers and complete loss of the fourth and fifth fingers. On baricitinib treatment, a significant improvement in cutaneous vasculitis resulted in preservation of the fingers without further tissue loss.
Figure 4. Improvement in longitudinal growth and…
Figure 4. Improvement in longitudinal growth and hematologic parameters.
(A) Clinically significant improvement was seen in height Z-scores and the percentiles of patients with growth potential (n = 13) when comparing data from before baricitinib treatment with data from the last visit. Mean height Z-scores improved from –4.03 ± 2.64 to –3.19 ± 2.33, with catch-up growth observed in 9 patients whose improvement translated into a mean height percentile increase from the 1.4th percentile to the 7.2nd percentile. (B) Photos of a patient with CANDLE (C8) with stunted growth since 2 years of age and a severe delay in bone age (chronological age of 14.3 years vs. bone age of 2 years). Within 30 months of treatment, her linear height increased from 90 cm to 106.8 cm, and her bone age improved from 2 years to 7.8 years. (C) Signs of bone marrow immunosuppression improved in all patients but 2 (C1 and O3), with increases in platelet counts, ALCs, and hemoglobin (Hgb) levels. Patient C1 continues to have persistent lymphopenia (ALC of 0.5), and patient O3 (discontinued from the program because of a poor response to treatment and osteonecrosis) had lower hemoglobin and platelet counts at the time of his last visit. This patient had multiple comorbidities including upper gastrointestinal bleeding, esophageal varices, IgA nephropathy, and idiopathic thrombocytopenia. **P < 0.05 (unadjusted) by paired Student’s t tests were used for both calculations; a 1-sided t test was used for height (in A) and paired Student’s t tests were used for all calculations, including the two asterisks (in C).
Figure 5. Assessment of conventional inflammatory parameters…
Figure 5. Assessment of conventional inflammatory parameters (CRP) and IFN biomarkers (serum IP-10 levels and 25-gene IFN score) with baricitinib treatment.
(A) CRP levels dropped most significantly in CANDLE patients, with CRP levels returning to normal in 5 of 10 of these patients. The patients with other interferonopathies (O2 and O4) who stayed in the program had improved CRP levels. The 2 patients who discontinued the program because of a lack of efficacy had no improvement and are circled. **P < 0.05 (unadjusted by paired 2-sided Student’s t-test). Data represent the mean ± SD. (B) The 25-gene IFN score was graphed for the baseline score and the IFN score obtained at the last included visit only. Colors indicate data by disease, with CANDLE in red, SAVI in blue, and other interferonopathies in green. Statistical data obtained by paired 2-sided Student’s t-test. The IFN score normalized in 5 of 10 patients with CANDLE who achieved the remission criteria. (C and D) Longitudinally assessed serum IP-10 levels and 25-gene IFN score measurements were fitted to a repeated-measures model with “treatment phase” as a categorical independent variable. Least-squares means (LSM) of serum IP-10 and IFN response gene score with 95% CIs for each phase are graphed. *P < 0.05 (unadjusted) by paired 2-sided Student’s t-test.

References

    1. de Jesus AA, Canna SW, Liu Y, Goldbach-Mansky R. Molecular mechanisms in genetically defined autoinflammatory diseases: disorders of amplified danger signaling. Annu Rev Immunol. 2015;33:823–874. doi: 10.1146/annurev-immunol-032414-112227.
    1. Kim H, Sanchez GA, Goldbach-Mansky R. Insights from Mendelian Interferonopathies: Comparison of CANDLE, SAVI with AGS, Monogenic Lupus. J Mol Med. 2016;94(10):1111–1127. doi: 10.1007/s00109-016-1465-5.
    1. Crow YJ. Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci. 2011;1238:91–98. doi: 10.1111/j.1749-6632.2011.06220.x.
    1. Agarwal AK, et al. PSMB8 encoding the β5i proteasome subunit is mutated in joint contractures, muscle atrophy, microcytic anemia, and panniculitis-induced lipodystrophy syndrome. Am J Hum Genet. 2010;87(6):866–872. doi: 10.1016/j.ajhg.2010.10.031.
    1. Arima K, et al. Proteasome assembly defect due to a proteasome subunit beta type 8 (PSMB8) mutation causes the autoinflammatory disorder, Nakajo-Nishimura syndrome. Proc Natl Acad Sci U S A. 2011;108(36):14914–14919. doi: 10.1073/pnas.1106015108.
    1. Kitamura A, et al. A mutation in the immunoproteasome subunit PSMB8 causes autoinflammation and lipodystrophy in humans. J Clin Invest. 2011;121(10):4150–4160. doi: 10.1172/JCI58414.
    1. Liu Y, et al. Mutations in proteasome subunit β type 8 cause chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature with evidence of genetic and phenotypic heterogeneity. Arthritis Rheum. 2012;64(3):895–907. doi: 10.1002/art.33368.
    1. Brehm A, et al. Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest. 2015;125(11):4196–4211. doi: 10.1172/JCI81260.
    1. Liu Y, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371(6):507–518. doi: 10.1056/NEJMoa1312625.
    1. Jeremiah N, et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. J Clin Invest. 2014;124(12):5516–5520. doi: 10.1172/JCI79100.
    1. Melki I, et al. Disease-associated mutations identify a novel region in human STING necessary for the control of type I interferon signaling. J Allergy Clin Immunol. 2017;140(2):543–552.e5. doi: 10.1016/j.jaci.2016.10.031.
    1. Kim H, et al. Development of a validated interferon score using NanoString technology. J Interferon Cytokine Res. 2018;38(4):171–185. doi: 10.1089/jir.2017.0127.
    1. Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293.
    1. Igaz P, Tóth S, Falus A. Biological and clinical significance of the JAK-STAT pathway; lessons from knockout mice. Inflamm Res. 2001;50(9):435–441.
    1. Fridman JS, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol. 2010;184(9):5298–5307. doi: 10.4049/jimmunol.0902819.
    1. Genovese MC, et al. Baricitinib in Patients with Refractory Rheumatoid Arthritis. N Engl J Med. 2016;374(13):1243–1252. doi: 10.1056/NEJMoa1507247.
    1. König N, et al. Familial chilblain lupus due to a gain-of-function mutation in STING. Ann Rheum Dis. 2017;76(2):468–472. doi: 10.1136/annrheumdis-2016-209841.
    1. Frémond ML, et al. Efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in the treatment of vasculopathy associated with TMEM173-activating mutations in 3 children. J Allergy Clin Immunol. 2016;138(6):1752–1755. doi: 10.1016/j.jaci.2016.07.015.
    1. Ramesh V, et al. Intracerebral large artery disease in Aicardi-Goutières syndrome implicates SAMHD1 in vascular homeostasis. Dev Med Child Neurol. 2010;52(8):725–732. doi: 10.1111/j.1469-8749.2010.03727.x.
    1. Argetsinger LS, et al. Identification of JAK2 as a growth hormone receptor-associated tyrosine kinase. Cell. 1993;74(2):237–244. doi: 10.1016/0092-8674(93)90415-M.
    1. Metcalf D, et al. Gigantism in mice lacking suppressor of cytokine signalling-2. Nature. 2000;405(6790):1069–1073. doi: 10.1038/35016611.
    1. Otoda T, et al. Proteasome dysfunction mediates obesity-induced endoplasmic reticulum stress and insulin resistance in the liver. Diabetes. 2013;62(3):811–824. doi: 10.2337/db11-1652.
    1. Tanimoto A, Murata Y, Wang KY, Tsutsui M, Kohno K, Sasaguri Y. Monocyte chemoattractant protein-1 expression is enhanced by granulocyte-macrophage colony-stimulating factor via Jak2-Stat5 signaling and inhibited by atorvastatin in human monocytic U937 cells. J Biol Chem. 2008;283(8):4643–4651. doi: 10.1074/jbc.M708853200.
    1. Thorens B, Mermod JJ, Vassalli P. Phagocytosis and inflammatory stimuli induce GM-CSF mRNA in macrophages through posttranscriptional regulation. Cell. 1987;48(4):671–679. doi: 10.1016/0092-8674(87)90245-5.
    1. Lendemans S, Rani M, Selbach C, Kreuzfelder E, Schade FU, Flohé S. GM-CSF priming of human monocytes is dependent on ERK1/2 activation. J Endotoxin Res. 2006;12(1):10–20.
    1. Qian BZ, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475(7355):222–225. doi: 10.1038/nature10138.
    1. Xu LL, Warren MK, Rose WL, Gong W, Wang JM. Human recombinant monocyte chemotactic protein and other C-C chemokines bind and induce directional migration of dendritic cells in vitro. J Leukoc Biol. 1996;60(3):365–371. doi: 10.1002/jlb.60.3.365.
    1. Ivashkiv LB. Epigenetic regulation of macrophage polarization and function. Trends Immunol. 2013;34(5):216–223. doi: 10.1016/j.it.2012.11.001.
    1. Mostafavi S, et al. Parsing the interferon transcriptional network and its disease associations. Cell. 2016;164(3):564–578. doi: 10.1016/j.cell.2015.12.032.
    1. Keystone EC, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis. 2015;74(2):333–340. doi: 10.1136/annrheumdis-2014-206478.
    1. Taylor PC, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med. 2017;376(7):652–662. doi: 10.1056/NEJMoa1608345.
    1. Knowles WA. Discovery and epidemiology of the human polyomaviruses BK virus (BKV) and JC virus (JCV) Adv Exp Med Biol. 2006;577:19–45. doi: 10.1007/0-387-32957-9_2.
    1. Rinaldo CH, Tylden GD, Sharma BN. The human polyomavirus BK (BKPyV): virological background and clinical implications. APMIS. 2013;121(8):728–745. doi: 10.1111/apm.12134.
    1. Polo C, Pérez JL, Mielnichuck A, Fedele CG, Niubò J, Tenorio A. Prevalence and patterns of polyomavirus urinary excretion in immunocompetent adults and children. Clin Microbiol Infect. 2004;10(7):640–644. doi: 10.1111/j.1469-0691.2004.00882.x.
    1. Rodero MP, Crow YJ. Type I interferon-mediated monogenic autoinflammation: The type I interferonopathies, a conceptual overview. J Exp Med. 2016;213(12):2527–2538. doi: 10.1084/jem.20161596.
    1. Kim H, et al. Pharmacokinetics, pharmacodynamics proposed dosing of the oral JAK1 JAK2 inhibitor baricitinib in pediatric young adult CANDLE SAVI Patients. Clin Pharmacol Ther. doi: 10.1002/cpt. [published online ahead of print November 14, 2017]. .
    1. Dixon WT. Simple proton spectroscopic imaging. Radiology. 1984;153(1):189–194. doi: 10.1148/radiology.153.1.6089263.
    1. Hamlett A, Ryan L, Serrano-Trespalacios P, Wolfinger R. Mixed models for assessing correlation in the presence of replication. J Air Waste Manag Assoc. 2003;53(4):442–450. doi: 10.1080/10473289.2003.10466174.
    1. Ford ES, Ajani UA, Mokdad AH, National Health Nutrition Examination The metabolic syndrome and concentrations of C-reactive protein among U.S. youth. Diabetes Care. 2005;28(4):878–881. doi: 10.2337/diacare.28.4.878.

Source: PubMed

3
Se inscrever