In Vivo PET Imaging of the Activated Immune Environment in a Small Animal Model of Inflammatory Arthritis

Benjamin L Franc, Sam Goth, John MacKenzie, Xiaojuan Li, Joseph Blecha, Tina Lam, Salma Jivan, Randall A Hawkins, Henry VanBrocklin, Benjamin L Franc, Sam Goth, John MacKenzie, Xiaojuan Li, Joseph Blecha, Tina Lam, Salma Jivan, Randall A Hawkins, Henry VanBrocklin

Abstract

Background: Evolving immune-mediated therapeutic strategies for rheumatoid arthritis (RA) may benefit from an improved understanding of the complex role that T-cell activation plays in RA. This study assessed the potential of fluorine-18-labeled 9-β-d-arabinofuranosylguanine ([18F]F-AraG) positron emission tomography (PET) imaging to report immune activation in vivo in an adjuvant-induced arthritis (AIA) small animal model.

Methods: Using positron emission tomography-computed tomography imaging, uptake of [18F]F-AraG in the paws of mice affected by arthritis at 6 (acute) and 20 (chronic) days following AIA induction in a single paw was assessed and compared to uptake in contralateral control paws. Fractions of T cells and B cells demonstrating markers of activation at the 2 time points were determined by flow cytometry.

Results: Differential uptake of [18F]F-AraG was demonstrated on imaging of the affected joint when compared to control at both acute and chronic time points with corresponding changes in markers of T-cell activation observed on flow cytometry.

Conclusion: [18F]F-AraG may serve as an imaging biomarker of T-cell activation in inflammatory arthritis. Further development of this technique is warranted and could offer a tool to explore the temporal link between activated T cells and RA as well as to monitor immune-mediated therapies for RA in clinical trials.

Keywords: animal models of disease; molecular imaging of inflammation.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article. Two of the authors (S.G., T.L.) are employed by CellSight.

Figures

Figure 1.
Figure 1.
Coronal images of representative subjects at 6 days (top) and 20 days (bottom) following initial model preparation. For both time points: (A) PET image, (B) fused PET-CT image, (C) fused PET-CT image rescaled to demonstrate difference in uptake between paws. A = arthritic paw (adjuvant injection); C = control paw (saline injection). PET indicates positron emission tomography; PET-CT, positron emission tomography–computed tomography.
Figure 2.
Figure 2.
Average percentage injected dose per volume in paws on PET images in control and affected animals and ratio of signals (affected/control) measured on days 6 and 20. PET indicates positron emission tomography.
Figure 3.
Figure 3.
Summary of cell surface marker expression at 6 and 20 days. A, Percentages of T-cell populations expressing markers of activation including CD25 (only CD4+ cells tested), CD44 (only CD4+ cells tested at 6 days), and CD69 (both CD4+ and CD8+ cells populations tested). B, Percentages of T-cell populations expressing CD62 L marker of resting state (only CD4+ cells tested at 20 days). C, Percentage of CD4+ B-cell populations expressing markers of activation including CD19 and CD80. (*P < .05; **P < .01).
Figure 3.
Figure 3.
Summary of cell surface marker expression at 6 and 20 days. A, Percentages of T-cell populations expressing markers of activation including CD25 (only CD4+ cells tested), CD44 (only CD4+ cells tested at 6 days), and CD69 (both CD4+ and CD8+ cells populations tested). B, Percentages of T-cell populations expressing CD62 L marker of resting state (only CD4+ cells tested at 20 days). C, Percentage of CD4+ B-cell populations expressing markers of activation including CD19 and CD80. (*P < .05; **P < .01).
Figure 4.
Figure 4.
Ki67 staining at day 6 demonstrates no significant difference in mean fluorescence of CD4+ cells in affected versus control paws, but greater nuclear activity in CD8+ cells (A), *P ≤ .02. Examples of Ki67 flow cytometry data from single mouse showing fluorescence of CD4+ (B) and CD8+ (C) cells from affected (red) versus control (blue) paws.
Figure 5.
Figure 5.
Hematoxylin–eosin stain (×200) demonstrating inflammatory cell infiltrate in cartilage of affected (A) versus control (B) paws at day 6. B = bone; DS = dorsal surface; I = inflammation; M = muscle; T = tendon.

References

    1. Ostrov BE. Immunotherapeutic biologic agents in autoimmune and autoinflammatory diseases. Immunol invest. 2015;44(8):777–802.
    1. Li Y, Shen Y, Hohensinner P, et al. Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis. Immunity. 2016;45(4):903–916.
    1. Ren CF, Zhao YX, Hou CF, et al. Expression of soluble programmed death-1 protein in peripheral blood regulatory T cells and its effects on rheumatoid arthritis progression. Mol Med Rep. 2017;15(1):460–466.
    1. Mellado M, Martinez-Munoz L, Cascio G, Lucas P, Pablos JL, Rodriguez-Frade JM. T cell migration in rheumatoid arthritis. Front Immunol. 2015;6:384.
    1. Ahmed MS, Bae YS. Dendritic cell-based immunotherapy for rheumatoid arthritis: from bench to bedside. Immune Netw. 2016;16(1):44–51.
    1. McInnes IB, al-Mughales J, Field M, et al. The role of interleukin-15 in T-cell migration and activation in rheumatoid arthritis. Nat Med. 1996;2(2):175–182.
    1. Lebre MC, Vieira PL, Tang MW, et al. Synovial IL-21/TNF-producing CD4+ T cells induce joint destruction in rheumatoid arthritis by inducing matrix metalloproteinase production by fibroblast-like synoviocytes. J Leukoc Biol. 2017;101(3):775–783.
    1. Berard M, Tough DF. Qualitative differences between naive and memory T cells. Immunology. 2002;106(2):127–138.
    1. Roeleveld DM, Koenders MI. The role of the Th17 cytokines IL-17 and IL-22 in rheumatoid arthritis pathogenesis and developments in cytokine immunotherapy. Cytokine. 2015;74(1):101–107.
    1. Cope AP. T cells in rheumatoid arthritis. Arthritis Res Ther. 2008;10(suppl 1):S1.
    1. Semerano L, Minichiello E, Bessis N, Boissier MC. Novel immunotherapeutic avenues for rheumatoid arthritis. Trends Mol Med. 2016;22(3):214–229.
    1. Brown PM, Pratt AG, Isaacs JD. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nat Rev Rheumatol. 2016;12(12):731–742.
    1. Cohen MD, Keystone E. Rituximab for rheumatoid arthritis. Rheumatol Ther. 2015;2(2):99–111.
    1. Lavielle M, Mulleman D, Goupille P, et al. Repeated decrease of CD4+ T-cell counts in patients with rheumatoid arthritis over multiple cycles of rituximab treatment. Arthritis Res Ther. 2016;18(1):253.
    1. Huang X, Wu H, Lu Q. The mechanisms and applications of T cell vaccination for autoimmune diseases: a comprehensive review. Clin Rev Allergy Immunol. 2014;47(2):219–233.
    1. Rosenthal KS, Mikecz K, Steiner HL, III, et al. Rheumatoid arthritis vaccine therapies: perspectives and lessons from therapeutic ligand epitope antigen presentation system vaccines for models of rheumatoid arthritis. Expert Rev Vaccines. 2015;14(6):891–908.
    1. Kremer JM, Westhovens R, Leon M, et al. Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. N Engl J Med. 2003;349(20):1907–1915.
    1. Deeks ED. Certolizumab Pegol: a review in inflammatory autoimmune diseases. BioDrugs. 2016;30(6):607–617.
    1. Aspalter RM, Eibl MM, Wolf HM. Regulation of TCR-mediated T cell activation by TNF-RII. J Leukoc Biol. 2003;74(4):572–582.
    1. Aspalter RM, Eibl MM, Wolf HM. Defective T-cell activation caused by impairment of the TNF receptor 2 costimulatory pathway in common variable immunodeficiency. J Allergy Clin Immunol. 2007;120(5):1193–1200.
    1. Aspalter RM, Wolf HM, Eibl MM. Chronic TNF-alpha exposure impairs TCR-signaling via TNF-RII but not TNF-RI. Cell Immunol. 2005;237(1):55–67.
    1. Monaco C, Nanchahal J, Taylor P, Feldmann M. Anti-TNF therapy: past, present and future. Int Immunol. 2015;27(1):55–62.
    1. Alfonso-Cristancho R, Armstrong N, Arjunji R, et al. Comparative effectiveness of biologics for the management of rheumatoid arthritis: systematic review and network meta-analysis. Clin Rheumatol. 2017;36(1):25–34.
    1. 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.
    1. Mitoma H, Horiuchi T, Tsukamoto H, Ueda N. Molecular mechanisms of action of anti-TNF-α agents—comparison among therapeutic TNF-α antagonists [published online August 24, 2016]. Cytokine. pii:S1043–4666(16) 30468–30469.
    1. Hodge JA, Kawabata TT, Krishnaswami S, et al. The mechanism of action of tofacitinib—an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 2016;34(2):318–328.
    1. England CG, Ehlerding EB, Hernandez R, et al. Preclinical pharmacokinetics and biodistribution studies of 89Zr-labeled pembrolizumab. J Nucl Med. 2017;58(1):162–168.
    1. Hettich M, Braun F, Bartholoma MD, Schirmbeck R, Niedermann G. High-resolution PET imaging with therapeutic antibody-based PD-1/PD-L1 checkpoint tracers. Theranostics. 2016;6(10):1629–1640.
    1. Li A, Wu Y, Linnoila J, et al. Surface biotinylation of cytotoxic T lymphocytes for in vivo tracking of tumor immunotherapy in murine models. Cancer Immunol Immunother. 2016;65(12):1545–1554.
    1. Mall S, Yusufi N, Wagner R, et al. Immuno-PET imaging of engineered human T cells in tumors. Cancer Res. 2016;76(14):4113–4123.
    1. Shao X, Wang X, English SJ, et al. Imaging of carrageenan-induced local inflammation and adjuvant-induced systemic arthritis with [(11)C]PBR28 PET. Nucl Med Biol. 2013;40(7):906–911.
    1. Vedvyas Y, Shevlin E, Zaman M, et al. Longitudinal PET imaging demonstrates biphasic CAR T cell responses in survivors. JCI Insight. 2016;1(19):e90064.
    1. Yaghoubi SS, Creusot RJ, Ray P, Fathman CG, Gambhir SS. Multimodality imaging of T-cell hybridoma trafficking in collagen-induced arthritic mice: image-based estimation of the number of cells accumulating in mouse paws. J Biomed Opt. 2007;12(6):064025.
    1. Keu KV, Witney TH, Yaghoubi S, et al. Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Sci Transl Med. 2017;9(373). pii: eaag2196.
    1. Larimer BM, Wehrenberg-Klee E, Caraballo A, Mahmood U. Quantitative CD3 PET imaging predicts tumor growth response to anti-CTLA-4 therapy. J Nucl Med. 2016;57(10):1607–1611.
    1. Namavari M, Chang YF, Kusler B, Yaghoubi S, Mitchell BS, Gambhir SS. Synthesis of 2’-deoxy-2’-[18F]fluoro-9-β-D-arabinofuranosylguanine: a novel agent for imaging T-cell activation with PET. Mol Imaging Biol. 2011;13(5):812–818.
    1. Ronald JA, Kim BS, Gowrishankar G, et al. A PET imaging strategy to visualize activated T cells in acute graft-versus-host disease elicited by allogenic hematopoietic cell transplant. Cancer Res. 2017. In press.
    1. Billiau A, Matthys P. Modes of action of Freund’s adjuvants in experimental models of autoimmune diseases. J Leukoc Biol. 2001;70(6):849–860.
    1. Caruso A, Licenziati S, Corulli M, et al. Flow cytometric analysis of activation markers on stimulated T cells and their correlation with cell proliferation. Cytometry. 1997;27(1):71–76.
    1. Obar JJ, Molloy MJ, Jellison ER, et al. CD4+ T cell regulation of CD25 expression controls development of short-lived effector CD8+ T cells in primary and secondary responses. Proc Natl Acad Sci U S A. 2010;107(1):193–198.
    1. Tao JH, Cheng M, Tang JP, Liu Q, Pan F, Li XP. Foxp3, regulatory T cell, and autoimmune diseases. Inflammation. 2017;40(1):328–339.
    1. Leung K. 1-(2’-Deoxy-2’-[18F]Fluoroarabinofuranosyl)Cytosine In: Molecular Imaging and Contrast Agent Database (MICAD). Bethesda, MD: National Center for Biotechnology Information (US); 2004.
    1. Lai NS, Koo M, Yu CL, Lu MC. Immunopathogenesis of systemic lupus erythematosus and rheumatoid arthritis: the role of aberrant expression of non-coding RNAs in T cells. Clin Exp Immunol. 2017;187(3):327–336.
    1. Rogier R, Koenders MI, Abdollahi-Roodsaz S. Toll-like receptor mediated modulation of T cell response by commensal intestinal microbiota as a trigger for autoimmune arthritis. J Immunol Res. 2015;2015:527696.
    1. Ali AM, Vino S. Genetic markers as therapeutic target in rheumatoid arthritis: a game changer in clinical therapy. Rheumatol Int. 2016;36(11):1601–1607.
    1. Quaresma JA, Yoshikawa GT, Koyama RV, Dias GA, Fujihara S, Fuzii HT. HTLV-1, immune response and autoimmunity. Viruses. 2015;8(1). pii: E5.
    1. Merlo LM, Mandik-Nayak L. IDO2: a pathogenic mediator of inflammatory autoimmunity. Clin Med Insights Pathol. 2016;9(suppl 1):21–28.

Source: PubMed

3
Subscribe