Skip to main content
Erschienen in: Clinical and Translational Medicine 1/2019

Open Access 01.12.2019 | Case report

The use of leukocytes’ secretome to individually target biological therapy in autoimmune arthritis: a case report

verfasst von: Patrice E. Poubelle, Nathalie Pagé, Marie-Pier Longchamps, Natalia Sampaio Moura, David B. Beck, Ivona Aksentijevich, Philippe A. Tessier, Martin Pelletier

Erschienen in: Clinical and Translational Medicine | Ausgabe 1/2019

Abstract

Background

Biological agents have allowed remarkable improvement in controlling autoimmune arthropathies, although none of the numerous biologics readily available represent a universal treatment standard. Moreover, classical and genetic predictors are currently unsatisfactory to predict individual response to a biologic, and the best treatment selection is still based on a trial-and-error approach. Here, we report a clinical case demonstrating the usefulness of examining the leukocytes’ secretome of patients. We set up and standardized a protocol that examines a patient’s immune responses to establish the secretome of the blood mononuclear leukocytes and personalize the biotherapy.

Case presentation

A 24-year-old woman was diagnosed with active early rheumatoid arthritis. The initial treatment regimen (prednisone, methotrexate, hydroxychloroquine, naproxen) was inefficient, as well as the anti-TNF adalimumab. The diagnosis was revised as possible rheumatoid arthritis-like psoriatic arthritis and adalimumab was replaced by abatacept (IgG1 Fc-CTLA-4) to no avail. Five years later, abatacept was replaced by the anti-IL-12/IL-23 ustekinumab with no objective control over the symptoms. The patient was thus enrolled in a prospective study based on the quantification of cytokines secreted by peripheral blood leukocytes stimulated with well-known immune activators of pattern recognition receptors and cytokine signalling. The results of this study revealed that plasma concentrations of cytokines were similar between the patient and healthy donors. In comparison to leukocytes from healthy donors, the patient’s secretome showed a unique overproduction of IL-6. The anti-IL-6 receptor tocilizumab was, therefore, administered with a rapid improvement of her active psoriatic arthritis that remained dependent on low prednisone dosage. Clinical parameters progressively returned to normal levels and her quality of life was greatly improved, despite the major delay to begin the present personalized treatment.

Conclusions

An efficient way to effectively treat patients with complex autoimmune arthropathies, and avoid irreversible disability, is to know their leukocytes’ secretome to identify abnormally secreted cytokines and personalize their biotherapy, as exemplified by this case report.
Abkürzungen
ANA
antinuclear antibody
Anti-CCP
anti-cyclic citrullinated peptide
DAS28
disease activity score of 28 joints
DAS28-CRP
DAS28 using the C-reactive protein
GWAS
genome-wide association study
IL
interleukin
PsoA
psoriatic arthritis
RA
rheumatoid arthritis
RF
rheumatoid factor
SNP
single nucleotide polymorphism
TNF
tumor necrosis factor

Background

Autoimmune arthropathies are treated with biologics that allowed remarkable advancement in the control of disease alteration [1]. However, some patients do not respond to the first biologic, or even to the others successively given with or without methotrexate and studies to identify predictors of response to biologic therapy are still limited [2]. The classical predictors such as the presence of auto-antibodies [the rheumatoid factor (RF) and the anti-cyclic citrullinated peptide (anti-CCP)], genotypes encoding the shared epitope HLA-DRB1 gene, smoking and/or periodontitis are largely insufficient to foresee the patient response to a biologic [3]. Genetic predictors represent an ongoing field of research and bear the potential to contribute to the development of a precision medicine approach in the management of autoimmune arthropathies [4, 5]. Nonetheless, the identification of genetic markers of disease outcome and response to treatment is still at its infancy and has been somewhat disappointing so far [6].
The most fruitful findings to treat autoimmune arthropathies remain the characterization of immune mediators involved in the disease. This is greatly supported by the numerous biologics readily available to treat most of the autoimmune diseases, including biotherapies targeting specific immune cells, such as the cytotoxic T-lymphocyte-associated protein-4 (CTLA-4, also known as CD152) or the B-lymphocyte antigen CD20, or secreted mediators like the pro-inflammatory cytokines tumor necrosis factor (TNF), interleukin (IL)-1, IL-6, IL-12/IL-23 and IL-17 [7]. Although a range of treatment options can be addressed with biologics, none of them are universally effective and the best treatment selection is still based on a trial-and-error approach, where the most suitable one is determined when a drug reduces disease activity or remission is identified [8]. Considering the severity of these life-threatening diseases and the high cost of biologics, the best treatment option should target, from the start, the patients’ own pattern of cytokines [9].
Here, we report a clinical case demonstrating the usefulness of examining the leukocytes’ secretome of patients. We set up and standardized a protocol that investigates the immune responses of the patients to establish the secretome of their blood mononuclear leukocytes. The results were used to personalize the biotherapy of a patient suffering from an autoimmune arthropathy, providing insights on how to tailor the best treatment option and therefore avoid definitive disability and loss of quality of life.

Case presentation

A 24-year-old woman was examined for the first time 3 months after the onset of symmetrical polyarthritis with major synovitis of 2nd, 3rd, 4th metacarpophalangeal joints of both hands, wrists, elbows, knees, ankles, forefeet, without any spinal signs. The disease activity score of 28 joints (DAS28) and DAS28 using the C-reactive protein (DAS28-CRP) were 8.09 and 7.75, respectively. Increased ferritin and thrombocytosis in the absence of detectable levels of RF, anti-CCP and antinuclear antibody (ANA) were also noticeable. Her liver function tests and lipid panel were normal and no bone erosion was visible by X-rays. She was diagnosed with active early rheumatoid arthritis (RA) (Table 1).
Table 1
Patient information and diagnostics summary
Time since onset of disease
3 months
1 year
2 years
5 years
10 years
11 years
Clinical parameters
 DAS28
8.09
8.37
7.94
6.87
7.61
3.76
 DAS28-CRP
7.75
7.98
7.55
6.63
7.44
4.34
 Bone erosions at X-rays
None
None
Yes
Yes
Yes
Yes
Biological parameters
 ESR (mm, first hour; Nl < 20)
63
68
37
8
24
5
 CRP (mg/L; Nl < 10)
56
85
25
1
18
7
 Ferritin (μg/mL; Nl < 200)
347
314
202
141
165
Not evaluated
 Platelet count (Nl < 400 billion/L)
469
390
348
225
247
263
 RF
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
 Anti-CCP
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
 ANA
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
Undetectable
 Synovial fluid analysis
 
8.93 billion WBC/L
21.15 billion WBC/L
120.8 billion WBC/L
  
  
99% neutrophils
99% neutrophils
83% neutrophils
  
Diagnosis
Active early RA
Active early RA
Possible RA-like PsoA
RA-like PsoA with bone erosions
RA-like PsoA with bone erosions
RA-like PsoA with bone erosions
Treatment prescribed
Prednisone
Methotrexate
Hydroxychloroquine
Naproxen
Initial treatment + adalimumab (anti-TNF)
Stop adalimumab; Initial treatment + abatacept (IgG1 Fc-CTLA-4)
Initial treatment + abatacept (IgG1 Fc-CTLA-4)
Stop abatacept; Initial treatment + ustekinumab (anti-IL-12/IL-23)
Initial treatment + tocilizumab
Notes
HLA-B27 absent
Treatment at visit: diclofenac
Sterile synovial fluid was removed by joint aspiration
Sterile synovial fluid was removed by joint aspiration
Mother and maternal grandmother have psoriasis
An aunt and her son have inflammatory spondylarthritis
Appearance of psoriasis skin lesions
Sterile synovial fluid was removed by joint aspiration
Ustekinimab (anti-IL-12/IL-23) and secukinimab (anti-IL-17) were not authorized at the time
No improvement after 6 months treatment with ustekinumab
Stop ustekinumab
Enrollement in study to quantify cytokines in plasma and secreted by activated PBMCs
Tocilizumab (anti-IL-6) begun
 
ANA antinuclear antibody, anti-CCP anti-cyclic citrullinated peptide, CRP C-reactive protein, DAS28 disease activity score of 28 joints, ESR erythrocyte sedimentation rate, Nl normal level, PsoA psoriatic arthritis, RA rheumatoid arthritis, RF rheumatoid factor, WBC white blood cells
Initial treatments with prednisone, methotrexate, hydroxychloroquine and naproxen were without efficacy. The anti-TNF adalimumab was added to the treatment regimen for 2 years. After only mild improvement, she experienced a progressive flare-up of polyarthritis and a loss of treatment efficacy. Two years after the onset of the disease, wrist and tarsal (right and left) demineralization, as well as bone erosions of ulnar styloids (right and left), appeared. Erythrocyte sedimentation rate (ESR), CRP and ferritin were persistently increased while RF and anti-CCP remained undetectable. The diagnosis was revised as possible RA-like psoriatic arthritis (PsoA), especially as her mother has skin psoriasis. Bone lesions were increased rapidly, in particular at both wrists. Adalimumab was replaced by abatacept (IgG1 Fc-CTLA-4) with a mild effect on arthritis. Five years after disease onset, psoriatic skin lesions appeared, and diagnosis of cutaneous psoriasis was confirmed by a dermatologist. The final diagnosis was aggressive RA-like PsoA with bone erosions, without RF and anti-CCP. Abatacept was replaced by the anti-interleukin (IL)-12/IL-23 ustekinumab with an increase of prednisone dosage. A mild relieve of polysynovitis was noted, which was dependent on prednisone. Reduction of prednisone led to a major flare-up of polysynovitis associated with asthenia, and after 3 months of ustekinumab administration, no objective effect on the patient’s symptoms was noted.
The patient was enrolled in a prospective study based on the quantification of cytokines secreted by peripheral blood leukocytes. Blood (50 mL) of the patient as well as of healthy donors was drawn after informed consent was obtained. Plasma was collected following centrifugation (400×g for 10 min) of anti-coagulated blood and stored at − 80 °C for further protein quantification. Peripheral blood mononuclear cells (PBMCs) were obtained following centrifugation (600×g for 20 min) of the cellular fraction of blood over density gradient medium (Lymphocyte separation medium, density 1.077–1.080 g/mL; Wisent Bioproducts Inc., St-Bruno, Québec, Canada). Density gradient-purified PBMCs were stimulated with well-known immune activators of pattern recognition receptors and cytokine signalling for 24 h at 1 × 106/mL in RPMI 1640 (Wisent Bioproducts Inc., St-Bruno, Québec, Canada) supplemented with 10% fetal bovine serum (VWR Life Science Seradigm, Mississauga, Ontario, Canada) and 1% primocin (InvivoGen, San Diego, California, USA) in the absence (control) or presence of plate-bound mouse anti-human CD3 (1 μg/mL, clone OKT3) + anti-human CD28 (10 μg/mL, clone 9.3), lipopolysaccharides (LPS—100 ng/mL; 45 nM, InvivoGen, San Diego, California, USA) + adenosine triphosphate (ATP—1 mM; added for the last 30 min, Sigma-Aldrich Canada Co., Oakville, Ontario, Canada), L18-muramyl dipeptide (L18-MDP—1 μg/mL; 1.32 μM, InvivoGen, San Diego, California, USA), Poly(deoxyadenylate–thymidylate) [Poly(dA:dT)—1 μg/mL; 1.57 μM, InvivoGen, San Diego, California, USA], anisomycin (20 μM, Millipore (Canada) Ltd, Etobicoke, Ontario, Canada) or pro-inflammatory cytokines IL-1β (100 ng/mL; 5.85 nM, PeproTech US, Rocky Hill, New Jersey, USA), TNF (100 ng/mL; 5.85 nM, STEMCELL Technologies Canada Inc., Vancouver, British Columbia, Canada), IL-6 (100 ng/mL; 3.83 nM, PeproTech US, Rocky Hill, New Jersey, USA) and IFN-γ (100 U/mL; 0.30 nM, PeproTech US, Rocky Hill, New Jersey, USA) to respectively activate T cells, the NLRP3, NOD2, AIM2 and pyrin inflammasomes as well as cytokine signalling. Following stimulation, cell supernatants were collected and stored at − 80 °C for further protein quantification. Proteins involved in inflammation (IL-1α, IL-1β, IL-6, IL-9, IL-15, IL-17A, IL-18, IL-21, IL-31, TNF, LT-α, IFN-γ), immunoregulation (IL-1RA, IL-4, IL-7, IL-10, IL-12, IL-13, IL-22, IL-23, IL-27, IFN-α), chemotaxis (CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CCL5/RANTES, CCL11/Eotaxin, CXCL1/GROα, CXCL8/IL-8, CXCL10/IP-10, CXCL12/SDF-1α) and cellular growth (IL-2, IL-5, GM-CSF) were quantified by multiplex analyses in plasma and cell supernatants using Luminex technology according to the manufacturers’ instructions (Cytokine & Chemokine 34-Plex Human ProcartaPlex™ Panel 1A, Thermo Fisher Scientific Inc., Burlington, Ontario, Canada).
The results of this study revealed that plasma concentrations of cytokines were similar between the patient and healthy donors (Fig. 1 and data not shown). In comparison to leukocytes from healthy donors, the patient’s secretome showed a unique overproduction of IL-6 in response to multiple stimuli, including the inflammasome activators LPS + ATP, MDP and poly(dA:dT), as well as the pro-inflammatory cytokines IL-1β, TNF and IFN-γ, to levels (up to 133,000 pg/mL) at least twice the ones produced by healthy donors’ cells (Fig. 2). This overproduction of IL-6 occurred without substantial increase of pro-inflammatory cytokines such as TNF, IL-12 and IL-23, which correlates with the inefficacy of the anti-TNF adalimumab and the anti-IL-12/IL-23 ustekinumab treatments. No substantial differences were observed for members of the IL-1 cytokine family (IL-1α, IL-1β, IL-18), as well as IFN-γ and IL-17. Increased secretion of the IL-1 receptor antagonist (IL-1RA) was shown upon stimulation with the AIM2 inflammasome activator poly(dA:dT) as well as IFN-γ, suggesting that the patient’s leukocytes can synthesize high amounts of IL-1RA to neutralize the production of IL-1. Of note, the stimulation of the patient’s T cells using a combination of anti-CD3 and anti-CD28 also led to increased production of IL-6, as well as IL-23, IL-1RA and IL-17, but not to the levels observed for IL-6 (Fig. 2). Finally, no substantial differences were observed between the patient and the healthy donors regarding the production of IL-2, IL-4, IL-5, IL-7, IL-9, IL-10, IL-13, IL-15, IL-21, IL-22, IL-27, IL-31, LT-α, IFN-α, CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CCL5/RANTES, CCL11/Eotaxin, CXCL1/GROα, CXCL8/IL-8, CXCL10/IP-10 and CXCL12/SDF-1α by unstimulated or stimulated PBMCs (data not shown).
Thus, considering the unique major overproduction of IL-6 by the patient’s leukocytes, the anti-IL-6 receptor tocilizumab was administered with a rapid improvement of her active PsoA that remained dependent on low prednisone dosage. DAS28 and DAS28-CRP were greatly improved at 3.76 and 4.34, respectively. ESR, CRP and ferritin were progressively normalized. Her quality of life was greatly improved with, in particular, a progressive reduction of asthenia. Have the patient been enrolled in this prospective study sooner, her refractory PsoA would have probably benefited from the personalized treatment without the current associated irreversible destructive arthritis and partial functional handicap, especially at both wrists.

Discussion

The immunological disease continuum, that includes all autoinflammatory and autoimmune diseases, as proposed by McGonagle & McDermott [10], allows to better understand the complexity of immune factors that can be associated with the numerous immunological disorders. In this regard, the present case report can possibly be related to polygenic autoinflammatory diseases and mixed pattern diseases, both conceptual classification in which psoriatic arthritis can be found. The investigation of the patient’s secretome, as we report here, remains non-exhaustive, with a major focus on the biologics currently available to treat autoimmune arthropathies such as RA and PsoA (Table 2). This focus could easily be extended to other factors depending on new biologics, as well as other stimuli to activate relevant immune signalling as the field progresses further. Nonetheless, the targeted secretome allows deciphering whether a factor could be largely produced over others to be able to adapt more precisely the treatment. This case report exemplifies the personalized treatment option based on the patient’s medical history and the determination of the secretome of the patient’s blood leukocytes in response to immune activators that revealed a unique overproduction of the IL-6 cytokine. In fact, the overproduction of IL-6 in the present case report of a refractory PsoA was in line with reports of mediators from synovial inflammation of PsoA where anti-IL-6 biologics were effective treatment choices [1115].
Table 2
Biotherapies and small molecule inhibitors against secreted mediators or immune cells and their approval for rheumatoid arthritis and psoriatic arthritis
Target
Agent
Structure
Mechanism of action
Approval for RA
Approval for PsoA
FDA
Health Canada
FDA
Health Canada
TNF
Etanercept
Fusion protein Fc IgG1-TNFR2
Decoy receptor
Nov 1998
Dec 2000
Jan 2002
Mar 2004
Infliximab
Chimeric mAb to TNF
Antagonist
Nov 1999
Sep 2001
May 2005
Jun 2006
Adalimumab
Humanized mAb to TNF
Jan 2003
Sep 2004
Oct 2005
Jun 2006
Golimumab
Human mAb to TNF
Apr 2009
Apr 2009
Oct 2017
Apr 2009
Certolizumab pegol
Pegylated Fab’ fragment of humanized mAb to TNF
May 2009
Sep 2009
Sep 2013
Jan 2014
IL-1
Anakinra
Recombinant human IL-1 receptor
Receptor antagonist
Nov 2001
Jun 2002
Canakinumab
Human mAb to IL-1β
Antagonist
Rilonacept
Fusion protein Fc IgG1 linked to ligand-binding domains of IL-1R1 and IL-1RAcP
    
CTLA-4
Abatacept
Fusion protein Fc IgG1-CTLA-4
Binds CD80 and CD86 on APCs; prevents co-stimulation of T cells
Dec 2005
Jun 2006
Jul 2017
Mar 2018
CD20
Rituximab
Chimeric mAb to CD20
Binds CD20 on B cells; triggers cell death
Feb 2006
Jun 2006
Ocrelizumab
Humanized mAb to CD20
Ofatunumab
Human mAb to CD20
Obinutuzumab
Humanized mAb to CD20 with a glycoengineered Fc
IL-6
Tocilizumab
Humanized mAb to IL-6 receptor
Receptor antagonist
Jan 2010
May 2010
Sarilumab
Human mAb to IL-6 receptor
May 2017
Feb 2017
IL-12, IL-23
Ustekinumab
Human mAb to IL-12 and IL-23 p40 subunit
Antagonist
Sep 2013
Jan 2014
IL-17
Secukinumab
Human mAb to IL-17A
Antagonist
Jan 2016
Apr 2016
Ixekizumab
Human mAb to IL-17A
Dec 2017
Jun 2018
Brodalumab
Human mAb to IL-17 receptor A
Receptor antagonist
Janus kinase 1–3
Tofacitinib
Small molecule
Intracellular inhibitor; blocks activity of cytokines (IL-2, -4, -6, -7, -9, -10, -12, -15, -21, -23, IFN-α, -β, -γ)
Nov 2012
Apr 2014
Dec 2017
Oct 2018
Another consideration is at the level of a unique overproduction of IL-6 in the present case of refractory PsoA, which could indicate a predominant autoinflammatory pattern over the autoimmune part of PsoA. In this regard, genomic DNA samples were isolated from the patient’s peripheral blood, and whole exome sequencing and data analysis were performed on the proband as previously described [16]. Publicly-available databases (ExAC, 1000 genomes, dbSNP, NHLBI GO Exome Sequencing Project and ClinSeq), as well as an in-house exome database, were used to filter for rare candidate variants. We did not identify any pathogenic mutations in known disease-causing genes. We analyzed the sequencing data for novel causes of immune dysregulation accounting for different modes of inheritance. Ideally, whole exome sequencing should be performed in trios to help with filtering a long list of candidate variants that are typically found when doing exome sequencing in singleton cases. Nonetheless, we did not identify any strong candidate variants to explain the phenotype. We also performed Sanger sequencing to analyze specific non-coding polymorphisms in the IL6 promoter and 5′UTR that could potentially account for some of the patient’s phenotype since the clinical spectrum of PsoA, more precisely the active peripheral arthritis, has been linked to IL6 (-174G/C) polymorphism [17]. Coding regions as well as flanking 5′ untranslated regions of the IL6 gene (RefSeq: NM_000600.5) were amplified by AmpliTaq Gold Fast PCR Master Mix (Thermo Fisher Scientific Inc., Waltham, Massachusetts, USA) and sequenced on SeqStudio Genetic Analyzer (Applied Biosystems Inc., Foster City, California, USA). We found that the patient had reference alleles at -597G, -572G, -473A8/T12 and -174G [18]. We finally analyzed the data for common low-penetrance coding variants associated with psoriatic or rheumatoid arthritis. We found that the patient is a heterozygous carrier for the single nucleotide polymorphism (SNP) in TRAF3IP2 rs33980500 (NM_001164281.2:c.28G>A; p.Asp10Asn; NM_147200.2:c.55G>A, NP_671733.2;p.Asp19Asn), which was identified by two genome-wide association studies (GWAS). The susceptibility allele rs33980500 has been shown to cause altered TRAF6 binding and thereby affect multiple immune pathways [19, 20]. Because TRAF3IP2 binding motifs have been associated with multiple TRAF proteins, the inhibition of the TRAF6 pathway could upregulate the TRAF2/5 pathway, leading to an enhanced immune response. Interestingly, studies showed that, in the TRAF6-independent pathway, TRAF2 and TRAF5 transduce the IL-17 signals to stabilize mRNA transcripts of chemokines and cytokines, such as CXCL1/GROα and IL-6 [21, 22]. Although these results did not identify a high penetrance genetic cause, they did identify a risk allele in TRAF3IP2, which in combination with additional environmental and genetic factors may help to explain this patient’s phenotype and suggest a further link between the PsoA, IL-6 production and TRAF3IP2/TRAFs signalling (Fig. 3).
While we examined the secretome of blood cells, one limitation of this study is that many other cell types in autoimmune arthritis are known to secrete cytokines, such as synoviocytes, fibroblasts, chondrocytes, osteoblasts/osteoclasts and epithelial cells (the latter ones in psoriatic arthritis for example), and that these cells, through their cytokine secretion, could contribute to the local pathology [2326]. The cytokine pattern could evidently be different from the ones produced by the blood cells. However, the use of these cells often requires invasive techniques to obtain them and their purification, for subsequent activation, is more labour-intensive than blood cells [27]. It is also useful to stress that the presence of a SNP in TRAF3IP2 rs33980500, as shown in this case report, should be present in all cell types using this signalling pathway [28, 29].

Conclusion

As a corollary and conclusion, an efficient way to effectively treat patients with complex autoimmune arthropathies, and to avoid irreversible disability, is to determine their blood leukocytes’ secretome to identify abnormally secreted cytokines to personalize their biotherapy. Another concluding detail should also be noted as the finding of an abnormal patients’ secretome can justify pursuing a causal investigation such as a genetic evaluation, as exemplified by this case report.

Authors’ contributions

PEP, the rheumatologist responsible for clinically following the patient, analyzed and interpreted the results and wrote the manuscript. NP obtained participants’ consents, purified and stimulated blood cells and performed multiplex assays. MPL performed multiplex assays. NSM helped prepare samples for whole exome and Sanger sequencing. DBB and IA analyzed and interpreted the sequencing data. PAT designed the study, analyzed and interpreted the results. MP designed the study, analyzed and interpreted the results and wrote the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We acknowledge Julie-Christine Lévesque from the Bioimaging platform of the Infectious Disease Research Centre, funded by an equipment and infrastructure grant from the Canadian Foundation for Innovation (CFI), for her technical assistance with the multiplex assays. We would like to thank Michel J. Tremblay (Centre de recherche du CHU de Québec-Université Laval) for generously providing the anti-CD3 and anti-CD28 antibodies as well as Ingrid Saba for reviewing the manuscript.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding authors on request.
A consent for publication form was signed by the patient. All authors approved the manuscript for submission.
The institutional review board of the CHU de Québec-Université Laval Research Center approved the study (2016–2519), and volunteers signed consent forms.

Funding

This research was sponsored by operating grants from La Fondation du Grand défi Pierre Lavoie, Rare Disease Foundation and the BC Children’s Hospital Foundation. The publication fees were sponsored by a grant (# 3623) from La Fondation du CHU de Québec. MP is a Junior 2 scholar from the Fonds de recherche du Québec-Santé (FRQS). The funding agencies had no role in the design and conduct of the study, collection, management, analysis, and interpretation of the data, the decision to submit for publication or preparation, review, or approval of the manuscript for publication.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Littlejohn EA, Monrad SU (2018) Early diagnosis and treatment of rheumatoid arthritis. Prim Care 45(2):237–255CrossRefPubMed Littlejohn EA, Monrad SU (2018) Early diagnosis and treatment of rheumatoid arthritis. Prim Care 45(2):237–255CrossRefPubMed
2.
Zurück zum Zitat Hyrich KL, Watson KD, Silman AJ, Symmons DP, British Society for Rheumatology Biologics R (2006) Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford) 45(12):1558–1565CrossRef Hyrich KL, Watson KD, Silman AJ, Symmons DP, British Society for Rheumatology Biologics R (2006) Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford) 45(12):1558–1565CrossRef
3.
Zurück zum Zitat Wijbrandts CA, Tak PP (2017) Prediction of response to targeted treatment in rheumatoid arthritis. Mayo Clin Proc. 92(7):1129–1143CrossRefPubMed Wijbrandts CA, Tak PP (2017) Prediction of response to targeted treatment in rheumatoid arthritis. Mayo Clin Proc. 92(7):1129–1143CrossRefPubMed
4.
Zurück zum Zitat Cherlin S, Plant D, Taylor JC, Colombo M, Spiliopoulou A, Tzanis E et al (2018) Prediction of treatment response in rheumatoid arthritis patients using genome-wide SNP data. Genet Epidemiol 42(8):754–771CrossRefPubMedPubMedCentral Cherlin S, Plant D, Taylor JC, Colombo M, Spiliopoulou A, Tzanis E et al (2018) Prediction of treatment response in rheumatoid arthritis patients using genome-wide SNP data. Genet Epidemiol 42(8):754–771CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Bluett J, Barton A (2017) Precision medicine in rheumatoid arthritis. Rheum Dis Clin North Am 43(3):377–387CrossRefPubMed Bluett J, Barton A (2017) Precision medicine in rheumatoid arthritis. Rheum Dis Clin North Am 43(3):377–387CrossRefPubMed
6.
7.
Zurück zum Zitat Her M, Kavanaugh A (2016) Alterations in immune function with biologic therapies for autoimmune disease. J Allergy Clin Immunol. 137(1):19–27CrossRefPubMed Her M, Kavanaugh A (2016) Alterations in immune function with biologic therapies for autoimmune disease. J Allergy Clin Immunol. 137(1):19–27CrossRefPubMed
8.
Zurück zum Zitat Smolen JS, Landewe R, Breedveld FC, Buch M, Burmester G, Dougados M et al (2014) EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 73(3):492–509CrossRefPubMed Smolen JS, Landewe R, Breedveld FC, Buch M, Burmester G, Dougados M et al (2014) EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 73(3):492–509CrossRefPubMed
9.
Zurück zum Zitat Turner MD, Nedjai B, Hurst T, Pennington DJ (2014) Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 1843(11):2563–2582CrossRefPubMed Turner MD, Nedjai B, Hurst T, Pennington DJ (2014) Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 1843(11):2563–2582CrossRefPubMed
11.
Zurück zum Zitat Alenius GM, Eriksson C, Rantapaa Dahlqvist S (2009) Interleukin-6 and soluble interleukin-2 receptor alpha-markers of inflammation in patients with psoriatic arthritis? Clin Exp Rheumatol 27(1):120–123PubMed Alenius GM, Eriksson C, Rantapaa Dahlqvist S (2009) Interleukin-6 and soluble interleukin-2 receptor alpha-markers of inflammation in patients with psoriatic arthritis? Clin Exp Rheumatol 27(1):120–123PubMed
12.
Zurück zum Zitat Costa L, Caso F, Cantarini L, Del Puente A, Scarpa R, Atteno M (2014) Efficacy of tocilizumab in a patient with refractory psoriatic arthritis. Clin Rheumatol 33(9):1355–1357CrossRefPubMed Costa L, Caso F, Cantarini L, Del Puente A, Scarpa R, Atteno M (2014) Efficacy of tocilizumab in a patient with refractory psoriatic arthritis. Clin Rheumatol 33(9):1355–1357CrossRefPubMed
13.
Zurück zum Zitat FitzGerald O (2016) Spondyloarthropathies: IL-6 blockade in psoriatic arthritis—a new therapeutic option? Nat Rev Rheumatol 12(6):318–319CrossRefPubMed FitzGerald O (2016) Spondyloarthropathies: IL-6 blockade in psoriatic arthritis—a new therapeutic option? Nat Rev Rheumatol 12(6):318–319CrossRefPubMed
14.
Zurück zum Zitat Mease PJ, Gottlieb AB, Berman A, Drescher E, Xing J, Wong R et al (2016) The efficacy and safety of clazakizumab, an anti-interleukin-6 monoclonal antibody, in a phase IIb study of adults with active psoriatic arthritis. Arthritis Rheumatol. 68(9):2163–2173CrossRefPubMed Mease PJ, Gottlieb AB, Berman A, Drescher E, Xing J, Wong R et al (2016) The efficacy and safety of clazakizumab, an anti-interleukin-6 monoclonal antibody, in a phase IIb study of adults with active psoriatic arthritis. Arthritis Rheumatol. 68(9):2163–2173CrossRefPubMed
15.
Zurück zum Zitat van Kuijk AW, Reinders-Blankert P, Smeets TJ, Dijkmans BA, Tak AA (2006) Detailed analysis of the cell infiltrate and the expression of mediators of synovial inflammation and joint destruction in the synovium of patients with psoriatic arthritis: implications for treatment. Ann Rheum Dis. 65(12):1551–1557CrossRefPubMedPubMedCentral van Kuijk AW, Reinders-Blankert P, Smeets TJ, Dijkmans BA, Tak AA (2006) Detailed analysis of the cell infiltrate and the expression of mediators of synovial inflammation and joint destruction in the synovium of patients with psoriatic arthritis: implications for treatment. Ann Rheum Dis. 65(12):1551–1557CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y et al (2016) Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet 48(1):67–73CrossRefPubMed Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y et al (2016) Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet 48(1):67–73CrossRefPubMed
17.
Zurück zum Zitat Cubino N, Montilla C, Usategui-Martin R, Cieza-Borrela C, Carranco T, Calero-Paniagua I et al (2016) Association of IL1Beta (-511 A/C) and IL6 (− 174 G>C) polymorphisms with higher disease activity and clinical pattern of psoriatic arthritis. Clin Rheumatol 35(7):1789–1794CrossRefPubMed Cubino N, Montilla C, Usategui-Martin R, Cieza-Borrela C, Carranco T, Calero-Paniagua I et al (2016) Association of IL1Beta (-511 A/C) and IL6 (− 174 G>C) polymorphisms with higher disease activity and clinical pattern of psoriatic arthritis. Clin Rheumatol 35(7):1789–1794CrossRefPubMed
18.
Zurück zum Zitat Terry CF, Loukaci V, Green FR (2000) Cooperative influence of genetic polymorphisms on interleukin 6 transcriptional regulation. J Biol Chem 275(24):18138–18144CrossRefPubMed Terry CF, Loukaci V, Green FR (2000) Cooperative influence of genetic polymorphisms on interleukin 6 transcriptional regulation. J Biol Chem 275(24):18138–18144CrossRefPubMed
19.
Zurück zum Zitat Ellinghaus E, Ellinghaus D, Stuart PE, Nair RP, Debrus S, Raelson JV et al (2010) Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet 42(11):991–995CrossRefPubMedPubMedCentral Ellinghaus E, Ellinghaus D, Stuart PE, Nair RP, Debrus S, Raelson JV et al (2010) Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet 42(11):991–995CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Huffmeier U, Uebe S, Ekici AB, Bowes J, Giardina E, Korendowych E et al (2010) Common variants at TRAF3IP2 are associated with susceptibility to psoriatic arthritis and psoriasis. Nat Genet 42(11):996–999CrossRefPubMedPubMedCentral Huffmeier U, Uebe S, Ekici AB, Bowes J, Giardina E, Korendowych E et al (2010) Common variants at TRAF3IP2 are associated with susceptibility to psoriatic arthritis and psoriasis. Nat Genet 42(11):996–999CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Bulek K, Liu C, Swaidani S, Wang L, Page RC, Gulen MF et al (2011) The inducible kinase IKKi is required for IL-17-dependent signaling associated with neutrophilia and pulmonary inflammation. Nat Immunol 12(9):844–852CrossRefPubMedPubMedCentral Bulek K, Liu C, Swaidani S, Wang L, Page RC, Gulen MF et al (2011) The inducible kinase IKKi is required for IL-17-dependent signaling associated with neutrophilia and pulmonary inflammation. Nat Immunol 12(9):844–852CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Sun D, Novotny M, Bulek K, Liu C, Li X, Hamilton T (2011) Treatment with IL-17 prolongs the half-life of chemokine CXCL1 mRNA via the adaptor TRAF5 and the splicing-regulatory factor SF2 (ASF). Nat Immunol 12(9):853–860CrossRefPubMedPubMedCentral Sun D, Novotny M, Bulek K, Liu C, Li X, Hamilton T (2011) Treatment with IL-17 prolongs the half-life of chemokine CXCL1 mRNA via the adaptor TRAF5 and the splicing-regulatory factor SF2 (ASF). Nat Immunol 12(9):853–860CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Ganesan R, Rasool M (2017) Fibroblast-like synoviocytes-dependent effector molecules as a critical mediator for rheumatoid arthritis: current status and future directions. Int Rev Immunol 36(1):20–30CrossRefPubMed Ganesan R, Rasool M (2017) Fibroblast-like synoviocytes-dependent effector molecules as a critical mediator for rheumatoid arthritis: current status and future directions. Int Rev Immunol 36(1):20–30CrossRefPubMed
24.
Zurück zum Zitat Danis J, Janovak L, Guban B, Goblos A, Szabo K, Kemeny L et al (2018) Differential inflammatory-response kinetics of human keratinocytes upon cytosolic RNA- and DNA-fragment induction. Int J Mol Sci 19(3):774CrossRefPubMedCentral Danis J, Janovak L, Guban B, Goblos A, Szabo K, Kemeny L et al (2018) Differential inflammatory-response kinetics of human keratinocytes upon cytosolic RNA- and DNA-fragment induction. Int J Mol Sci 19(3):774CrossRefPubMedCentral
26.
Zurück zum Zitat Veale DJ, Fearon U (2018) The pathogenesis of psoriatic arthritis. Lancet 391(10136):2273–2284CrossRefPubMed Veale DJ, Fearon U (2018) The pathogenesis of psoriatic arthritis. Lancet 391(10136):2273–2284CrossRefPubMed
27.
Zurück zum Zitat Amos PJ, Cagavi Bozkulak E, Qyang Y (2012) Methods of cell purification: a critical juncture for laboratory research and translational science. Cells Tissues Organs. 195(1–2):26–40CrossRefPubMed Amos PJ, Cagavi Bozkulak E, Qyang Y (2012) Methods of cell purification: a critical juncture for laboratory research and translational science. Cells Tissues Organs. 195(1–2):26–40CrossRefPubMed
28.
Zurück zum Zitat Matsushima Y, Kikkawa Y, Takada T, Matsuoka K, Seki Y, Yoshida H et al (2010) An atopic dermatitis-like skin disease with hyper-IgE-emia develops in mice carrying a spontaneous recessive point mutation in the Traf3ip2 (Act1/CIKS) gene. J Immunol. 185(4):2340–2349CrossRefPubMed Matsushima Y, Kikkawa Y, Takada T, Matsuoka K, Seki Y, Yoshida H et al (2010) An atopic dermatitis-like skin disease with hyper-IgE-emia develops in mice carrying a spontaneous recessive point mutation in the Traf3ip2 (Act1/CIKS) gene. J Immunol. 185(4):2340–2349CrossRefPubMed
29.
Zurück zum Zitat Das NA, Carpenter AJ, Yoshida T, Kumar SA, Gautam S, Mostany R et al (2018) TRAF3IP2 mediates TWEAK/TWEAKR-induced pro-fibrotic responses in cultured cardiac fibroblasts and the heart. J Mol Cell Cardiol 121:107–123CrossRefPubMed Das NA, Carpenter AJ, Yoshida T, Kumar SA, Gautam S, Mostany R et al (2018) TRAF3IP2 mediates TWEAK/TWEAKR-induced pro-fibrotic responses in cultured cardiac fibroblasts and the heart. J Mol Cell Cardiol 121:107–123CrossRefPubMed
Metadaten
Titel
The use of leukocytes’ secretome to individually target biological therapy in autoimmune arthritis: a case report
verfasst von
Patrice E. Poubelle
Nathalie Pagé
Marie-Pier Longchamps
Natalia Sampaio Moura
David B. Beck
Ivona Aksentijevich
Philippe A. Tessier
Martin Pelletier
Publikationsdatum
01.12.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
Clinical and Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 2001-1326
DOI
https://doi.org/10.1186/s40169-019-0236-7

Weitere Artikel der Ausgabe 1/2019

Clinical and Translational Medicine 1/2019 Zur Ausgabe