A biomarker signature to predict complete response to itacitinib and corticosteroids in acute graft-versus-host disease

Michael Pratta, Sophie Paczesny, Gerard Socie, Natalie Barkey, Hao Liu, Sherry Owens, Michael C Arbushites, Mark A Schroeder, Michael D Howell, Michael Pratta, Sophie Paczesny, Gerard Socie, Natalie Barkey, Hao Liu, Sherry Owens, Michael C Arbushites, Mark A Schroeder, Michael D Howell

Abstract

A broad proteomic analysis was conducted to identify and evaluate candidate biomarkers potentially predictive of response to treatment with an oral selective Janus kinase 1 (JAK1) inhibitor, itacitinib, in acute graft-versus-host disease (GVHD). Plasma samples from 25 participants (identification cohort; NCT02614612) were used to identify novel biomarkers that were tested in a validation cohort from a placebo-controlled, randomised trial (n = 210; NCT03139604). The identification cohort received corticosteroids plus 200 or 300 mg itacitinib once daily. The validation cohort received corticosteroids plus 200 mg itacitinib once daily or placebo. A broad proteomic analysis was conducted using a proximity extension assay. Baseline and longitudinal comparisons were performed with unpaired t-test and one-way analysis of variance used to evaluate biomarker level changes. Seven candidate biomarkers were identified. Monocyte-chemotactic protein (MCP)3, pro-calcitonin/calcitonin (ProCALCA/CALCA), together with a previously identified prognostic acute GVHD biomarker, regenerating islet-derived protein (REG)3A, stratified complete responders from non-responders (participants with progressive disease) to itacitinib, but not placebo, potentially representing predictive biomarkers of itacitinib in acute GVHD. ProCALCA/CALCA, suppressor of tumorigenicity (ST)2, and tumour necrosis factor receptor (TNFR)1 were significantly reduced over time by itacitinib in responders, potentially representing response-to-treatment biomarkers. Novel biomarkers have the potential to identify patients with acute GVHD that may respond to itacitinib plus corticosteroid treatment (NCT02614612; NCT03139604).

Keywords: JAK inhibitors; acute graft-versus-host disease; biomarkers; itacitinib.

Conflict of interest statement

Sophie Paczesny has a patent, ‘Methods of detection of graft‐versus‐host disease’ (US 20130115232A1, WO 2013066369A3), licensed to Viracor‐IBT Laboratories, and received honoraria from Incyte, Genentech, and Omeros for advisory board meetings. Gerard Socie received lecture fees from Incyte Corporation and Novartis and honoraria from Novartis for advisory board meeting. Mark A. Schroeder received research funding paid to his institution by companies for which he is a Principal Investigator for a company‐sponsored research study including Genentech Inc., Incyte, Cellect Inc., Fortis, Seattle Genetics, Amgen, Celgene, PDB Incorporated, Genzyme Sanofi, and Janssen; served on advisory boards, and received honoraria and consultant fees from Amgen, Astellas, Dova Pharmaceuticals, FlatIron Inc., Incyte, Partners Therapeutics, Pfizer, and Sanofi Genzyme; and served on the speakers bureau and received honoraria as a consultant for AbbVie, Merck, and Takeda. Michael Pratta, Natalie Barkey, and Michael C. Arbushites are employees of Incyte Research Institute, Incyte Corporation. Hao Liu, Sherry Owens, and Michael D. Howell were employees of Incyte Corporation at the time these studies were undertaken.

© 2022 INCYTE Corporation. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Volcano plot representing differentially expressed proteins at baseline in plasma of CR compared with PD groups in identification cohort. Broad proteomic analysis of plasma samples was conducted by OLINK proteomics (Watertown, MA, USA) using a proximity extension assay as described by the manufacturer. The protein library consists of >1000 proteins. Data are presented as NPX in log2 scale. FC was calculated based on NPX values in CR (n = 10) and PD (n = 6). A negative FC represents proteins downregulated in CR relative to PD; a positive FC represents proteins upregulated in CR relative to PD. In the volcano plot shown, the x‐axis is the log2 transformed FC, calculated as log2(FC), and y‐axis is the negative log p value, calculated as –log10(raw p value). Identity of the novel predictive candidate biomarkers is shown in green, JAK/STAT‐related biomarkers are shown in red, and previously validated prognostic biomarkers are shown in blue. Identity of all other proteins illustrated is listed in Table S2. CALCA, calcitonin; CCL19, C‐C motif chemokine 19; CR, complete responder; CXCL10, C‐X‐C motif chemokine 10; FC, fold change; IL2RA, interleukin 2 receptor alpha chain; IL6, interleukin‐6; IL8, interleukin‐8; JAK, Janus kinase; MCP3, monocyte‐chemotactic protein 3; NPX, normalised protein expression; PD, progressive disease or death; PON3, paraoxonase 3; REG3A, regenerating islet‐derived protein; SCF, kit ligand; ST2, suppressor of tumorigenicity 2; STAT, signal transducer and activator of transcription; TNFR1, tumour necrosis factor receptor 1; TNFRSF6B, TNF receptor superfamily member 6b.
FIGURE 2
FIGURE 2
Baseline levels of novel predictive candidate biomarkers, JAK/STAT‐related biomarkers, and previously validated prognostic biomarkers in identification cohort. Baseline plasma samples from CR and PD participants (or participants who died) and from participants with an intermediate response, including VGPR and PR, were assayed for biomarker levels by proximal extension assay. Statistical analysis was by one‐way ANOVA. Significance was conferred when p < 0.05. The lines over particular cohorts indicate comparison of those cohorts with p values indicated. ANOVA, analysis of variance; CCL19, C‐C motif chemokine 19; CR, complete responder; CXCL10, C‐X‐C motif chemokine 10; IL2RA, interleukin 2 receptor alpha chain; IL6, interleukin 6; IL8, interleukin 8; JAK, Janus kinase; MCP3, monocyte‐chemotactic protein 3; nat, natural; PD, progressive disease; PON3, paraoxonase 3; ProCALCA/CALCA, pro‐calcitonin/calcitonin; PR, partial responder; REG3A, regenerating islet‐derived protein; SCF, kit ligand; ST2, suppressor of tumorigenicity 2; STAT, signal transducer and activator of transcription; TNFR1, tumour necrosis factor receptor 1; TNFRSF6B, TNF receptor superfamily member 6b; VGPR, very good partial responder; *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001; CR, blue; PD/death, black; VGPR/PR, red.
FIGURE 3
FIGURE 3
Comparison of day 28 treatment response between itacitinib versus placebo in validation cohort. Day 28 treatment responses from itacitinib (n = 101) and placebo (n = 109) treatment arms from the validation cohort are presented. PD includes participants whose disease progressed during treatment and/or who died before day 28. The ‘other’ category includes mixed responders, non‐responders, and unclassified participants. CR, complete responder; PD, progressive disease/death; VGPR/PR, very good partial responder/partial responder.
FIGURE 4
FIGURE 4
Baseline levels of candidate predictive biomarkers (A) ProCALCA/CALCA; (B) REG3A; (C) MCP3; (D) ST2 in the validation cohort. Baseline serum samples from itacitinib versus placebo arms of validation cohort were assayed for biomarker levels by proximal extension assay. Statistical analysis was by unpaired t‐test. Significance was conferred when p < 0.05. Itacitinib CR: Blue; Itacitinib PD: Black; placebo CR: Orange; placebo PD: Green. Open symbols represent participants with standard risk; closed symbols represent participants with high‐risk acute GVHD. CR, complete response; GVHD, graft‐versus‐host disease; MCP3, monocyte‐chemotactic protein 3; nat, natural; NS, not significant; PD, progressive disease/death; ProCALCA/CALCA, pro‐calcitonin/calcitonin; REG3A, regenerating islet‐derived protein; ST2, suppressor of tumorigenicity 2; *p < 0.05; **p < 0.01.
FIGURE 5
FIGURE 5
Longitudinal analysis of candidate biomarkers in responders from validation cohort. Serum levels of candidate biomarkers identified in the identification cohort were measured at baseline and at days 7, 14, and 28 (where available) from responders (CR, VGPR, PR) in the validation cohort, and mean ± SEM values are plotted from the itacitinib/corticosteroid combination (n = 74; red circles) versus corticosteroid alone (placebo n = 79; blue squares). Data are shown for each biomarker showing a significant difference between treatment arms. Data were compared at each time point by unpaired t‐test, and significant differences between treatment arms are shown (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). BL, baseline; CR, complete responder; MCP3, monocyte‐chemotactic protein 3; nat, natural; PR, partial responder; ProCALCA/CALCA, pro‐calcitonin/calcitonin; REG3A, regenerating islet‐derived protein; SEM, standard error of the mean; ST2, suppressor of tumorigenicity 2; TNFR1, tumour necrosis factor receptor 1; VGPR, very good partial responder.
FIGURE 6
FIGURE 6
Density plots comparing baseline levels of each candidate biomarker in the identification and validation cohorts. Baseline levels of each candidate biomarker are shown in density plots, where log2 of biomarker concentration was compared with the density of participants at a corresponding biomarker concentration. Baseline levels from identification cohort are shown in blue; baseline levels from the validation cohort are shown in yellow. The markers were classified according to novel predictive candidate biomarkers, JAK/STAT‐related biomarkers, and previously validated prognostic biomarkers. Statistical analysis was by unpaired t‐test. Significance was conferred when p < 0.05. The lines over particular cohorts indicate comparison of those cohorts with p values indicated. CCL19, C‐C motif chemokine 19; CXCL10, C‐X‐C motif chemokine 10; IL2RA, interleukin‐2 receptor alpha chain; IL6, interleukin 6; IL8, interleukin 8; JAK, Janus kinase; MCP3, monocyte‐chemotactic protein 3; NS, not significant; PD, progressive disease; PON3, paraoxonase 3; ProCALCA/CALCA, pro‐calcitonin/calcitonin; REG3A, regenerating islet‐derived protein; SCF, kit ligand; ST2, suppressor of tumorigenicity 2; STAT, signal transducer and activator of transcription; TNFR1, tumour necrosis factor receptor 1; TNFRSF6B, TNF receptor superfamily member 6b. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
FIGURE 7
FIGURE 7
Correlation between protein simple Ella and proximal extension assay in the detection of candidate biomarkers in samples from identification cohort. Plasma samples from CR and PD participants from the identification cohort were tested for (A) ProCALCA/CALCA; (B) REG3A; (C) MCP3; (D) ST2; and (E) TNFR1 by protein simple Ella (x‐axis) and by PEA (y‐axis). Correlation between the two assay platforms is shown for each biomarker. Statistical analysis was performed using Pearson correlation, and r and p values are shown. Significance was conferred when p < 0.05. CR, complete responder; MCP3, monocyte‐chemotactic protein 3; nat, natural; PD, progressive disease/death; PEA, proximal extension assay; ProCALCA/CALCA, pro‐calcitonin/calcitonin; REG3A, regenerating islet‐derived protein; ST2, suppressor of tumorigenicity 2; TNFR1, tumour necrosis factor receptor 1.

References

    1. Gooley TA, Chien JW, Pergam SA, Hingorani S, Sorror ML, Boeckh M, et al. Reduced mortality after allogeneic hematopoietic‐cell transplantation. N Engl J Med. 2010;363(22):2091–101.
    1. Vogelsang GB, Lee L, Bensen‐Kennedy DM. Pathogenesis and treatment of graft‐versus‐host disease after bone marrow transplant. Annu Rev Med. 2003;54:29–52.
    1. Kuykendall TD, Smoller BR. Lack of specificity in skin biopsy specimens to assess for acute graft‐versus‐host disease in initial 3 weeks after bone‐marrow transplantation. J Am Acad Dermatol. 2003;49(6):1081–5.
    1. Massi D, Fondi C, Nozzoli C, Benemei S, Lapi F, Albarello L, et al. The impact of histopathologic examination of graft‐versus‐host disease in the era of reduced‐intensity conditioning regimen: a study from the Gruppo Italiano Trapianto di Midollo Osseo. Hum Pathol. 2011;42(2):254–68.
    1. Martin PJ, Rizzo JD, Wingard JR, Ballen K, Curtin PT, Cutler C, et al. First‐ and second‐line systemic treatment of acute graft‐versus‐host disease: recommendations of the American Society of Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2012;18(8):1150–63.
    1. Garnett C, Apperley JF, Pavlu J. Treatment and management of graft‐versus‐host disease: improving response and survival. Ther Adv Hematol. 2013;4(6):366–78.
    1. MacMillan ML, Robin M, Harris AC, DeFor TE, Martin PJ, Alousi A, et al. A refined risk score for acute graft‐versus‐host disease that predicts response to initial therapy, survival, and transplant‐related mortality. Biol Blood Marrow Transplant. 2015;21(4):761–7.
    1. Westin JR, Saliba RM, De Lima M, Alousi A, Hosing C, Qazilbash MH, et al. Steroid‐refractory acute GVHD: predictors and outcomes. Adv Hematol. 2011;2011:601953.
    1. Arai S, Margolis J, Zahurak M, Anders V, Vogelsang GB. Poor outcome in steroid‐refractory graft‐versus‐host disease with antithymocyte globulin treatment. Biol Blood Marrow Transplant. 2002;8(3):155–60.
    1. Levine JE, Hogan WJ, Harris AC, Litzow MR, Efebera YA, Devine SM, et al. Improved accuracy of acute graft‐versus‐host disease staging among multiple centers. Best Pract Res Clin Haematol. 2014;27(3–4):283–7.
    1. Levine JE, Logan B, Wu J, Alousi AM, Ho V, Bolanos‐Meade J, et al. Graft‐versus‐host disease treatment: predictors of survival. Biol Blood Marrow Transplant. 2010;16(12):1693–9.
    1. Levine JE, Braun TM, Harris AC, Holler E, Taylor A, Miller H, et al. A prognostic score for acute graft‐versus‐host disease based on biomarkers: a multicentre study. Lancet Haematol. 2015;2(1):e21–9.
    1. Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, et al. A biomarker panel for acute graft‐versus‐host disease. Blood. 2009;113(2):273–8.
    1. Srinagesh HK, Levine JE, Ferrara JLM. Biomarkers in acute graft‐versus‐host disease: new insights. Ther Adv Hematol. 2019;10:1–8.
    1. Vander Lugt MT, Braun TM, Hanash S, Ritz J, Ho VT, Antin JH, et al. ST2 as a marker for risk of therapy‐resistant graft‐versus‐host disease and death. N Engl J Med. 2013;369(6):529–39.
    1. Major‐Monfried H, Renteria AS, Pawarode A, Reddy P, Ayuk F, Holler E, et al. MAGIC biomarkers predict long‐term outcomes for steroid‐resistant acute GVHD. Blood. 2018;131(25):2846–55.
    1. Srinagesh HK, Özbek U, Kapoor U, Ayuk F, Aziz M, Ben‐David K, et al. The MAGIC algorithm probability is a validated response biomarker of treatment of acute graft‐versus‐host disease. Blood Adv. 2019;3(23):4034–42.
    1. Pidala J, Hamadani M, Dawson P, Martens M, Alousi AM, Jagasia M, et al. Randomized multicenter trial of sirolimus vs prednisone as initial therapy for standard‐risk acute GVHD: the BMT CTN 1501 trial. Blood. 2020;135(2):97–107.
    1. Alousi AM, Weisdorf DJ, Logan BR, Bolanos‐Meade J, Carter S, Difronzo N, et al. Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acute graft‐versus‐host disease: a randomized phase 2 trial from the blood and marrow transplant clinical trials network. Blood. 2009;114(3):511–7.
    1. Cahn JY, Bordigoni P, Tiberghien P, Milpied N, Brion A, Widjenes J, et al. Treatment of acute graft‐versus‐host disease with methylprednisolone and cyclosporine with or without an anti‐interleukin‐2 receptor monoclonal antibody. A multicenter phase III study. Transplant. 1995;60(9):939–42.
    1. Couriel DR, Saliba R, de Lima M, Giralt S, Andersson B, Khouri I, et al. A phase III study of infliximab and corticosteroids for the initial treatment of acute graft‐versus‐host disease. Biol Blood Marrow Transplant. 2009;15(12):1555–62.
    1. Cragg L, Blazar BR, Defor T, Kolatker N, Miller W, Kersey J, et al. A randomized trial comparing prednisone with antithymocyte globulin/prednisone as an initial systemic therapy for moderately severe acute graft‐versus‐host disease. Biol Blood Marrow Transplant. 2000;6(4A):441–7.
    1. Lee SJ, Zahrieh D, Agura E, MacMillan ML, Maziarz RT, McCarthy PL Jr, et al. Effect of up‐front daclizumab when combined with steroids for the treatment of acute graft‐versus‐host disease: results of a randomized trial. Blood. 2004;104(5):1559–64.
    1. Levine JE, Paczesny S, Mineishi S, Braun T, Choi SW, Hutchinson RJ, et al. Etanercept plus methylprednisolone as initial therapy for acute graft‐versus‐host disease. Blood. 2008;111(4):2470–5.
    1. Khandelwal P, Emoto C, Fukuda T, Vinks AA, Neumeier L, Dandoy CE, et al. A prospective study of alemtuzumab as a second‐line agent for steroid‐refractory acute graft‐versus‐host disease in pediatric and young adult allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2016;22(12):2220–5.
    1. van Groningen LF, Liefferink AM, de Haan AF, Schaap NP, Donnelly JP, Blijlevens NM, et al. Combination therapy with inolimomab and etanercept for severe steroid‐refractory acute graft‐versus‐host disease. Biol Blood Marrow Transplant. 2016;22(1):179–82.
    1. Yalniz FF, Hefazi M, McCullough K, Litzow MR, Hogan WJ, Wolf R, et al. Safety and efficacy of infliximab therapy in the setting of steroid‐refractory acute graft‐versus‐host disease. Biol Blood Marrow Transplant. 2017;23(9):1478–84.
    1. Pidala J, Kim J, Anasetti C. Sirolimus as primary treatment of acute graft‐versus‐host disease following allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2009;15(7):881–5.
    1. Zeiser R, von Bubnoff N, Butler J, Mohty M, Niederwieser D, Or R, et al. Ruxolitinib for glucocorticoid‐refractory acute graft‐versus‐host disease. N Engl J Med. 2020;382(19):1800–10.
    1. Schroeder MA, Khoury HJ, Jagasia M, Ali H, Schiller GJ, Staser K, et al. A phase 1 trial of itacitinib, a selective JAK1 inhibitor, in patients with acute graft‐versus‐host disease. Blood Adv. 2020;4(8):1656–69.
    1. Zeiser R, Socie G, Schroeder MA, Abhyankar S, Pinho Vaz C, Kwon M, et al. Efficacy and safety of itacitinib or placebo in combination with corticosteroids for initial treatment of acute graft‐versus‐host disease (GRAVITAS‐301): a randomised, multicentre phase 3 study. Lancet Hematol. 2022;9(1):e14–25.
    1. Paczesny S. Post‐haematopoietic cell transplantation outcomes: why ST2 became a 'golden nugget' biomarker. Br J Haematol. 2020;192(6):951–67.
    1. Weissinger EM, Metzger J, Schleuning M, Schmid C, Messinger D, Beutel G, et al. A multicenter prospective, randomized, placebo‐controlled phase II/III trial for preemptive acute graft‐versus‐host disease therapy. Leukemia. 2021;35(6):1763–72.
    1. MacMillan ML, DeFor TE, Weisdorf DJ. The best endpoint for acute GVHD treatment trials. Blood. 2010;115(26):5412–7.
    1. Assarsson E, Lundberg M, Holmquist G, Björkesten J, Thorsen SB, Ekman D, et al. Homogenous 96‐plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS One. 2014;9(4):e95192.
    1. Cao J, Seegmiller J, Hanson NQ, Zaun C, Li D. A microfluidic multiplex proteomic immunoassay device for translational research. Clin Proteomics. 2015;12:28.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B Methodol. 1995;57(1):289–300.
    1. O'Neal WK, Anderson W, Basta PV, Carretta EE, Doerschuk CM, Barr RG, et al. Comparison of serum, EDTA plasma and P100 plasma for luminex‐based biomarker multiplex assays in patients with chronic obstructive pulmonary disease in the SPIROMICS study. J Transl Med. 2014;12:9.
    1. Newton Y, Rassekh SR, Deyell RJ, Shen Y, Jones MR, Dunham C, et al. Comparative RNA‐sequencing analysis benefits a pediatric patient with relapsed cancer. JCO precis. Oncologia. 2018;2:1–16.
    1. Xu Q, Fu R, Yin G, Liu X, Liu Y, Xiang M. Microarray‐based gene expression profiling reveals genes and pathways involved in the oncogenic function of REG3A on pancreatic cancer cells. Gene. 2016;578(2):263–73.

Source: PubMed

3
Tilaa