Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2018 Dec 18;144(7):1664–1675. doi: 10.1002/ijc.31922

Decreased RORC expression and downstream signaling in HTLV‐1‐associated adult T‐cell lymphoma/leukemia uncovers an antiproliferative IL17 link: A potential target for immunotherapy?

Kritika Subramanian 1,2,, Tim Dierckx 2,, Ricardo Khouri 2,3, Soraya Maria Menezes 2, Huseini Kagdi 4, Graham P Taylor 4, Lourdes Farre 3, Achilea Bittencourt 5, Keisuke Kataoka 6,7, Seishi Ogawa 6, Johan Van Weyenbergh 2,
PMCID: PMC6590643  PMID: 30303535

Abstract

Retinoic acid‐related drugs have shown promising pre‐clinical activity in Adult T‐cell Leukemia/Lymphoma, but RORC signaling has not been explored. Therefore, we investigated transcriptome‐wide interactions of the RORC pathway in HTLV‐1 and ATL, using our own and publicly available gene expression data for ATL and other leukemias. Gene expression data from ATL patients were analyzed using WGCNA to determine gene modules and their correlation to clinical and molecular data. Both PBMCs and CD4+ T‐Cells exhibited decreased RORC expression in four different ATL cohorts. A small subset of RORChi ATL patients was identified with significantly lower pathognomonic CADM1 and HBZ levels but similar levels of other ATL markers (CD4/CD25/CCR4), hinting at a less aggressive ATL subtype. An age‐dependent decrease in RORC expression was found in HTLV‐1‐infected individuals, but not in healthy controls, suggesting an early molecular event predisposing to leukemogenesis. Genes upstream of RORC signaling were members of a proliferative gene module (containing proliferation markers PCNA/Ki67), whereas downstream members clustered in an anti‐proliferative gene module. IL17C transcripts showed the strongest negative correlation to PCNA in both ATL cohorts, which was replicated in two large cohorts of T‐ and B‐cell acute lymphoid leukemia (ALL). Finally, IL17C expression in purified CD4 + CCR4 + CD26‐CD7‐ “ATL‐like” cells from HTLV‐1‐infected individuals and ATL patients was negatively correlated with clonality, underscoring a possible antileukemic/antiproliferative role. In conclusion, decreased RORC expression and downstream signaling might represent an early event in ATL pathogenesis. An antiproliferative IL17C/PCNA link is shared between ATL, T‐ALL and B‐ALL, suggesting (immuno)therapeutic benefit of boosting RORC/IL17 signaling.

Keywords: leukemia, lymphoma, proliferation, Th17, retrovirus, inflammation, immunotherapy, carcinogenesis, PCNA, IL17C

Short abstract

What's new?

Drugs that affect the retinoic acid pathway are of interest for the treatment of adult T‐cell leukemia (ATL). Here, investigation of the role of retinoic acid‐related orphan receptor C (RORC), a regulator of the proinflammatory Th17/IL‐17 axis, reveals a prevailing occurrence of low RORC expression among ATL patients. By comparison, fewer patients exhibited a RORChi phenotype, which was associated with reduced levels of pathognomonic biomarkers CADM1 and HbZ, indicating a protective role for elevated RORC. An antiproliferative link was identified between RORC and IL17C. The data suggest that strategies to increase RORC/IL17C signaling could be important to improving ATL outcomes.


Abbreviations

AML

acute myeloid leukemia

ATL

adult T‐cell leukemia/lymphoma

ATRA

all‐trans retinoic acid

HTLV‐1

human T‐cell leukemia virus‐1

ID

infectious dermatitis

IL

interleukin

PCNA

proliferating cell nuclear antigen

RARɑr

retinoic acid receptor alpha

RORC

retinoic acid orphan receptor C

T‐ALL

T‐cell acute lymphoblastic leukemia

TBLV

type B leukomogenic virus

WGCNA

weighted gene correlation network analysis

Introduction

Human T‐lymphotropic virus‐1 (HTLV‐1) is a retrovirus with an estimated prevalence of 10–20 million worldwide.1 A recent return to the original name of Human T‐cell leukemia virus‐12 is in agreement with its exceptional oncogenicity.3 Although most HTLV‐1 infections are asymptomatic, 2–6% of HTLV‐1 infected individuals develop a CD4+CD25+ chemotherapy‐resistant and aggressive leukemia known as Adult T‐cell lymphoma/leukemia (ATL).4, 5, 6 ATL presents after a long latency period of the virus, commonly several decades.7 Patients therefore tend to be older individuals with an average age at diagnosis of 40 years in Central and South America and 60 years in Japan. Depending on the subtype (acute, lymphomatous, chronic, and smoldering), survival ranges from 4 months to over 5 years.8

HTLV‐1 has two viral oncoproteins: Tax and HBZ. Tax benefits cell survival in HTLV‐1 infected T‐cells by interacting with NFκB,9 a key player in immune regulation. However, Tax levels are undetectable in most ATL patients, either due to gene deletion or altered DNA methylation levels, whereas HBZ is expressed consistently in ATL.9 HBZ modulates Tax expression and induces CD4+ T‐cell proliferation.5 CADM1/TSLC1 is also consistently expressed in ATL cells, such that CADM1 staining overlaps with the CD4+CD25+ T‐cells in ATL and proviral sequences from these leukemic CD4+CADM1+ cells were consistently positive for the HBZ region.5 Thus, CADM1 is a sensitive biomarker for ATL and might be used to determine treatment efficacy.5, 9

ATL patients display an increased incidence of opportunistic infections,10 which could be attributed to a deregulation of the Th17 axis, as an intact Th17 response is necessary for the clearance of opportunistic infections.11, 12, 13, 14 IL‐17 and its upstream regulator IL‐6 were increased in long‐term cultured Tax+CD4+ T‐cell supernatant.15 IL‐17 mRNA was also found to be highly expressed in HTLV‐1 infected T‐cells and Tax‐expressing Jurkat cells.16 Therefore, we hypothesize Tax‐negative ATL cells are unlikely to express IL‐17. Induction of the Th17 axis via retinoic acid receptors (RARs) and RAR‐like orphan receptors (RORs) could potentially alleviate the increased opportunistic infection frequency caused by Th17 deregulation.

Retinoic acid blocks Th17 differentiation and stimulates regulatory T‐cell (Treg) production.17 Although HTLV‐1 proviral integration in the host genome showed greater enrichment of promotor sequence motifs binding p53 and STAT1 instead of the RORC locus,18 downstream effects of p53 and STAT1 downregulate RORC expression by suppressing the transcription factor STAT3.19 The relevance of RORC in leukemogenesis is further supported by the observed increased proliferation and apoptosis rates in mice deficient in the protein product of the RORC gene RORγ, leading to the development of T‐cell lymphoma20 and lymphoblastic lymphoma.21

Taken together, deregulation of the RORC/Th17 axis can provide an explanation to both the oncogenic persistence of ATL and to patient susceptibility to opportunistic infections. In our study, we generate a representative consensus gene set for the RORC pathway of the Th17 axis and proceed to a multicohort analysis of novel and existing data to test the biological significance of this pathway in ATL.

Results

Transcriptomic analysis of four independent cohorts reveals a RORClo ex vivo phenotype in ATL

Gene expression profiling of ex vivo primary cells from ATL patients showed decreased RORC normalized expression in all four independent cohorts, revealing a common RORClo phenotype (Fig. 1 ad). Japanese Cohort #3 (n = 73) and Caribbean Cohort (n = 38) had significant decreases in RORC expression of ATL patients (p < 0.0001 and p = 0.016 respectively). Japanese Cohorts #1 (n = 18) and #2 (n = 50) had borderline significant decreases in RORC expression of ATL patients (p = 0.083 and p = 0.10). HAM patients in the Caribbean Cohort did not have a significant change in RORC expression (p = 0.54), however asymptomatic HTLV‐1 infected individuals (AC) did display a significant decrease in RORC expression (p = 0.016) when compared to healthy controls. ACs in other cohorts were not found to have a significant change in RORC expression, relative to healthy controls. Thus, RORC expression, measured as normalized expression (Fig. 1) and percentile rank (Supporting Information Fig. S1), is consistently lower in ATL than in HC, but varies among cohorts for AC. Since RORC gene expression had been previously shown to decrease in AC.22 we performed a meta‐analysis of the fold changes in RORC expression in all four cohorts, using both normalized expression and percentile ranks. Normalized gene expression allows for a comparison of the fold change in absolute RORC levels across cohorts but does not consider the profound perturbation of the cellular transcriptome between healthy vs. leukemic CD4+ cells in AC and ATL patients, respectively. In contrast, percentile ranks are a measure of RORC expression relative to the overall transcriptome for each individual, which is more suitable for a comparison between divergent disease states. This was confirmed by two‐way ANOVA, analyzing cohorts and disease status (HC‐AC‐ATL) as separate variables. For fold‐change RORC normalized expression, cohort differences accounted for 21.2% of variation (p = 0.34) and disease status for 47.2% of variation (p = 0.06). For fold‐change RORC percentile ranks, only 1.3% of variation (p = 0.48) was explained by cohorts and 95.8% of variation (p < 0.0001) was explained by disease status. As shown in Figure 1 e, RORC normalized expression was significantly (p < 0.01) decreased in ATL patients, but not AC. Figure 1 f displays the median RORC percentile rank fold‐change, showing a significant 30% decrease in asymptomatic HTLV‐1‐infected individuals (p < 0.0001, vs. HC) and an even further (43%) decrease in ATL patients (p < 0.0001, vs. HC; p = 0.006 vs. AC). This two‐step decrease in RORC gene expression in ATL pathogenesis, first upon HTLV‐1 infection and next upon progression to malignant disease, prompted us to investigate the possible influence of age upon RORC expression.

Figure 1.

Figure 1

Normalized RORC expression levels for four independent cohorts consisting of ATL patients and healthy uninfected (HC) and/or HTLV‐1 infected healthy controls (AC) showed a consistent decreased expression in ATL (ad). (e) Meta‐analysis of RORC normalized expression fold‐change by disease status shows a significant decrease in ATL patients but not ACs. (f) Meta‐analysis of RORC percentile rank fold‐change by disease status shows a significant two‐step decrease for ACs and ATL. HCs = Healthy Controls, ACs = Asymptomatic Controls, ATL = Adult T‐cell Lymphoma/Leukemia Patients, HAM = HTLV‐1‐Associated Myelopathy patients. *p < 0.05, **p < 0.01, ****p < 0.0001. [Color figure can be viewed at wileyonlinelibrary.com]

RORC expression is not influenced by age in healthy controls but decreases with age in HTLV‐1 infected individuals

Since ATL usually occurs after several decades of HTLV‐1 infection5 and RORγt Tregs were shown to increase with age in mice,23 we investigated the effect of age upon RORC gene expression in healthy controls and HTLV‐1‐infected individuals from several cohorts. We found that RORC expression significantly decreased with age in HTLV‐1 infected individuals without ATL, either AC or HAM/TSP patients (r = −0.57, p = 0.0002, n = 30 from UK Cohort, Fig. 2 a). We observed a similar tendency of decreased RORC expression with age in our Brazilian cohort (r = −0.62), but this observation did not reach statistical significance levels (p = 0.10), most probably due to the small size of this ATL cohort (n = 8) (Fig. 2 b). Unfortunately, the age of ATL patients was not available for the larger Japanese cohort. Next, we examined paired CD4+ T‐cells (n = 293), CD8+ T‐cells (n = 283), and PBMC (n = 77) microarray results from a cohort of healthy controls with sufficient power to study the effects of age (Healthy Estonian Cohort, Table 1). We found that RORC expression levels did not significantly change with age in CD4+ T‐cells (r = 0.002, p = 0.45, Fig. 2 c), CD8+ T‐cells (r = 0.0001, p = 0.86, Fig. 2 d) or PBMC (r = 0.0001, p = 0.93), nor with gender (data not shown).

Figure 2.

Figure 2

HTLV‐1‐infected Individuals from UK Cohort (a) and ATL patients from Brazilian Cohort (b) showed a decrease in RORC expression as age increased. This decrease was absent in healthy controls, in either CD4+ cells (c), CD8+ cells‐(d) or PBMCs (not shown). [Color figure can be viewed at wileyonlinelibrary.com]

Table 1.

Transcriptomic (microarray and RNAseq) datasets used in RORC expression analyses

Data set Source Cell Type Disease Status Sample Size
GSE55851
Japanese Cohort #1
Kobayashi et al. (2014) CD4+ T‐cells HC 3
AC 6
ATL 12
EGAD1001411
Japanese Cohort #2
Kataoka et al. (2015) CD4+ T‐cells HC 3
PBMCs AC 3
ATL 44
GSE33615
Japanese Cohort #3
Yamagishi et al. (2012) CD4+ T‐cells HC 21
PBMCs ATL 52
GSE19080
Caribbean Cohort
Oliere et al. (2010) CD4+ T‐cells
(Immunoarray)
HC 8
AC 11
ATL 7
HAM 12
GSE85487
Brazilian Cohort
Dierckx et al. (2017) PBMCs* HC 5
ATL – Untreated 8
ATL ‐ IFN‐ɑ 6
ATL ‐ IFN‐β 6
GSE29312
UK Cohort
Tattermusch et al. (2012) Whole Blood HC 9
AC 20
HAM 10
GSE78840
Healthy Estonian Cohort
Kasela et al. (2017) CD4+ T‐cells HC 293
CD8+ T‐cells HC 283
PBMCs HC 77
ImmuCo Wang et al. (2015) CD4+ T‐cells HC 551
CD8+ T‐cells HC 149
Bone marrow Mononuclear Cells AML 814
Acute T‐cell Leukemia T‐ALL 138
Acute B‐cell Leukemia B‐ALL 300

HC = Healthy Control. AC = Asymptomatic HTLV‐1 Infected Control. ATL = Adult T‐cell Lymphoma/Leukemia patients. HAM = HTLV‐1 Associated Myopathy. AML = Acute Myeloid Leukemia. ALL = Acute Lymphoblastic Leukemia. *Short‐term in vitro cultured PBMCs.

A minor RORChi subgroup of ATL patients displays a unique CADM1loHBZlo phenotype

RORChi outliers (Rout Method,24 Q = 0.1%) were observed in the three Japanese cohorts, accounting for a total of 13 out of 108 ATL patients (12.04%), Therefore, we examined this phenotype more closely in the largest examined cohort (Japanese Cohort #2), where 7 outliers with a higher normalized RORC expression were identified (Fig. 1 b). The patients from this cohort were then split into two groups, according to their RORC levels, as shown in Figure 3. Interestingly, we noted that RORC expression was inversely associated with expression levels of pathognomonic, or unique disease identifying, ATL biomarkers CADM1 and HBZ. Thus, RORChi patients displayed significantly lower HBZ (p = 0.0061) and CADM1 (p = 0.045) levels, but similar expression levels of other ATL surface marker genes (CD4, CD25/IL2RA, CCR4) suggesting the RORChi subgroup might represent a distinct, possibly clinically relevant, molecular subgroup of ATL. The lower CADM1 and HBZ expression levels in RORChi patients may represent the decreased proliferation rate of chronic or less aggressive ATL subtypes. As shown in Supporting Information Figure S2, RORChi patients showed similar expression levels of other ATL driver genes (STAT3, PLCG1, NFKB1, RELA, FAS),25, 26 highlighting the specificity of the RORChiCADM1loHBZlo phenotype. Positive expression of IRF4 and c‐REL has been associated with resistance to IFN‐ɑ + AZT therapy in ATL patients.26, 27 Interestingly, IRF4 and c‐REL expression did not differ between RORChi and RORClo patients (Supporting Information Fig. S2). This finding suggests RORC expression is independent of IFN‐ɑ + AZT therapeutic resistance and offers an additional molecular target for patients failing this therapy.

Figure 3.

Figure 3

RORC expression levels of ATL patients from Japanese Cohort #2 separated into two groups: RORChi (Green) and RORClo (Red) show RORChi levels were associated with lower HBZ and CADM1 expression levels. RORChi (7 outliers) and RORClo groups were compared with expression levels for ATL driver/mutated genes. *p < 0.05 **p < 0.01 ****p < 0.0001. [Color figure can be viewed at wileyonlinelibrary.com]

Definition of a consensus RORC pathway and gene set and its relevance to ATL oncogenesis

To facilitate the molecular exploration of the RORChi phenotype, a RORC gene set was determined based on published literature findings, integrating the intrinsic oncogenic pathway for STAT3 activation, as defined by Yu et al.28 and RARA/RORC signaling summarized by Muranski and Restifo (2013).29 The consensus RORC pathway included IL6, IL23, IL21, IRF4, BATF, STAT1, STAT5, RARɑ, TGFβ, NFκB, SLC2A1 (GLUT1), BCL6, STAT3, FOXP3, SOCS1, RORC, and IL17A/F. Figure 4 a illustrates the interplay between these genes, as detailed in the legend. RNX1, T‐bet, RORA, and TGFB1R were not measured by the microarray used for the initial WGCNA analysis on the Brazilian cohort (pilot cohort) and therefore excluded from the gene set. To validate the biological significance of this manually compiled pathway, we applied STRING protein–protein interaction enrichment analysis, which confirmed highly significant interaction for the RORC consensus pathway (expected number of edges: 11, observed number of edges: 83, enrichment p < 10−16). As displayed in Figure 4 b, our compiled RORC pathway was significantly enriched for “Positive regulation of cytokine production” (p = 7.9 × 10−12), “Regulation of T‐helper cell differentiation” (p = 2.2 × 10−10), “Th17 immune response” (p = 1.9 × 10−9), “Jak‐STAT signaling pathway” (p = 5.1 × 10−9), “Pathways in cancer” (p = 1.2 × 10−8), “HTLV‐1 infection” (4.4 × 10−5) and “Viral carcinogenesis” (p = 0.0047), thus validating our approach.

Figure 4.

Figure 4

(a) Simplified figure depicting the roles of RORC consensus pathway members, adapted from Yu et al. (2009)36 and Muranski and Restifo (2013)37. The figure was produced using Servier Medical Art (http://www.servier.com) and edited using Inkscape software. (b) The RORC consensus pathway was validated using STRING protein–protein interaction, GO biological process and KEGG pathway enrichment analysis. Significant enrichment (genome‐wide FDR < 0.05) is shown for “Positive regulation of cytokine production” (red), “Regulation of T‐helper cell differentiation” (purple), “Th17 immune response” (green), “Jak‐STAT signaling pathway” (yellow), “Pathways in cancer” (magenta), “HTLV‐1 infection” (dark green) and “Viral carcinogenesis” (turquoise). [Color figure can be viewed at wileyonlinelibrary.com]

A modular approach reveals a link between the RORC consensus pathway, proliferation and leukemogenesis

Transcriptomic expression levels of RORC pathway members extracted from a UK HTLV‐1‐infected asymptomatic control dataset (UK Cohort; GSE29312) and ATL cohort (Japanese Cohort #2; EGAD1001411) showed that the majority were expressed at highly variable levels (Supporting Information Fig. S3). First, prominent STAT1 expression is in line with published findings in AC22, 29 and ATL.26, 30, 31 On the other hand, downstream members of the RORC pathway and particularly, IL17 family genes were either undetectable or poorly expressed.

WGCNA analysis of PBMCs from our pilot ATL cohort (n = 8, Brazil, Fig. 5 a) showed overlap of the RORC pathway with a gene module correlated to proliferation, containing bona fide proliferation markers PCNA (Proliferating Cell Nuclear Antigen) and MKI67 (the gene coding for Ki67 antigen, routinely used in flow cytometric quantification of proliferation). As shown in Figure 5 b, downstream pathway members RORC and the IL17 family, under‐expressed in ATL, were negatively correlated with the proliferative module and positively correlated with the anti‐proliferative module. Likewise, upstream and overexpressed gene members of the RORC pathway displayed the reverse trend. This resulted in a significant bifurcation in the RORC pathway, as shown by linear regression of correlation coefficients of member genes with proliferative and antiproliferative modules, respectively (Supporting Information Fig. S4, r = −0.97, p < 0.0001). Overall, WGCNA analysis suggested that inducing RORC and its downstream signaling, as well as blocking upstream pathway members may decrease the cell proliferation rate in ATL.

Figure 5.

Figure 5

Modular transcriptomic analysis of primary ATL cells reveals a strong association of the RORC/IL17 pathway with proliferation. (a) WGCNA findings for the Turquoise and Blue modules are shown for selected molecular and clinical correlations. R value and p values (in parenthesis) are shown *p < 0.05 **p < 0.01. (b) Most RORC pathway members were positively correlated with the turquoise (“antiproliferative”) gene module in primary ATL cells and negatively correlated with the blue (“proliferative”) modules, such that most downstream members were found to be associated with the antiproliferative module (Brazilian cohort, n = 8, left panel). WGCNA of Japanese Cohort #2 (right panel, n = 44) shows RORC has a positive correlation with CD4+ resting memory T‐cells (Royal Blue module, r = 0.47, p = 0.001). Yellow module is positively correlated with CD4+ memory active T‐cells (r = 0.39, p = 0.007), as well proliferation markers PCNA (r = 0.92, p = 10−15) and MKI67 (r = 0.87, p = 10−12). Brown module is positively correlated with CD4+ naïve T‐cells (r = 0.49, p = 0.0006). Genes which were validated in both ATL cohorts for proliferative modules are colored according to their R values. (c) ATL patients expressing IL17C (IL17pos, n = 17, Japanese Cohort #2) showed decreased PCNA expression as compared to patients with undetectable IL17C (IL17neg, n = 28), and IL17C levels were positively correlated with RORC levels. [Color figure can be viewed at wileyonlinelibrary.com]

To confirm and extend these findings on proliferation, we repeated the WGCNA in the larger cohort of ATL patients (n = 44, Japanese cohort #2). We additionally obtained in silico estimates of the relative size of 22 immune cell type populations using the CIBERSORT software.32 As shown in Figure 5 b, RORC was the only pathway member which was significantly and positively correlated (r = 0.42) with the presence of resting memory CD4+ T‐cells (p = 0.0041). Downstream pathway members IL17B (r = 0.62, p = 0.0000054) and IL17C (r = 0.42, p = 0.04) were positively correlated with the presence of naïve CD4+ T‐cells. STAT3 inducer NFκB subunits 1 and 2 were negatively correlated with naïve CD4+ T‐cells (p = 0.02 and p = 0.052 respectively) and resting memory CD4+ T‐cells (p‐0.000073 and p = 0.00084 respectively). Similar to the observations in the WGCNA of the pilot cohort, a reverse trend was also seen in the CIBERSORT analysis between upstream and downstream members of the RORC pathway and their correlation with naïve and activated memory CD4+ T‐cell fractions (Fig. 5 b). Together, the two WGCNA analyses, combined with CIBERSORT CD4+ subtype quantification, suggest a distinct change in proliferative pathways between upstream and downstream members of the RORC/IL17 pathway with opposite effects in activated memory vs. naïve and resting memory CD4+ T‐cells. Among downstream pathway members, IL17C showed the strongest antiproliferative gene module membership in both cohorts and was also more frequently detected than other IL17 family members (IL17A/B/D/F). Therefore, we classified ATL patients from the largest cohort (Japanese cohort #2) into IL17C expressing, (IL17Cpos, n = 17) and IL17C negative (IL17Cneg, n = 28). As shown in Figure 5 c, IL17C‐positive patients had significantly lower gene expression levels of proliferative marker PCNA (Mann–Whitney p = 0.022) and in those patients, IL17C was positively correlated to RORC gene expression (r = 0.54, p = 0.026), confirming the findings of our modular analysis.

Validation of IL17C as a potential “antileukemic” target in independent ATL, T‐ALL and B‐ALL cohorts

First, we analyzed IL17C expression in an independent UK cohort for which clonality analysis as well as clinical data (including therapeutic response) were available. RNAseq analysis of purified “ATL‐like” cells with a CD4 + CCR4 + CD26‐CD7‐ immunophenotype demonstrated that IL17C transcripts were detectable in all ATL patients, but at significantly lower levels, as compared to AC. As shown in Figure 6 a, IL17C transcripts were significantly decreased in both indolent and aggressive ATL (One‐way ANOVA, Bonferroni's post‐test p < 0.05). No difference in IL17C levels was observed between ATL clinical forms or with regard to therapeutic response (chemotherapy and IFN + AZT resistance, not shown). However, IL17C expression was negatively correlated to clonality (r = −0.72, p = 0.0086, n = 12) in AC (fraction of largest clone 0.02–0.34) and patients with ATL (fraction of largest clone 0.68–0.99) (Fig. 6 b), in support of our hypothesized antiproliferative/antileukemic role for IL17C. Of note, IL17A and IL17F transcripts were not expressed (data not shown) in “ATL‐like” cells, in agreement with Kagdi et al.,33 who demonstrated compartmentalized expression of most cytokines in nonleukemic cells.

Figure 6.

Figure 6

Validation of IL17C as a potential “antileukemic” target in independent ATL, T‐ALL and B‐ALL cohorts. ( a) In an independent UK cohort of HTLV‐1‐infected individuals, IL17C transcripts were significantly decreased in purified CD4 + CCR4 + CD26‐CD7‐ cells from both indolent and aggressive ATL patients, as compared to AC (One‐way ANOVA, Bonferroni's post‐test p < 0.05), with no difference in IL17C levels between ATL clinical forms. (b) IL17C expression was negatively correlated to clonality in AC and ATL patients (fraction of largest clone 0.02–0.34 and 0.68–0.99, respectively). A negative correlation between IL17C and PCNA transcript levels was replicated in (c) T‐cell Acute Lymphoid Leukemia (ALL) (n = 138) and B‐cell ALL cohorts (n = 300). [Color figure can be viewed at wileyonlinelibrary.com]

Second, to explore if the antiproliferative IL17C/PCNA link might be specific to ATL or shared with other leukemias, we analyzed two large cohorts of acute T‐ and B‐cell leukemia (T‐ALL, n = 138; B‐ALL, n = 300). Similar to ATL, we found a significant negative correlation between IL17C and PCNA expression levels in both T‐ALL (r = −0.24, p = 0.007) and B‐ALL (r = −0.28, p < 0.0001), as shown in Figure 6 c and d. Unfortunately, no clinical follow‐up data (survival or therapeutic response) are available for the T‐ALL and B‐ALL cohorts.

IFN‐ɑ, IFN‐β and ascorbic acid in vitro treatment differentially regulates RORC pathway members in primary ATL cells and HTLV‐1 transformed cell lines

We previously tested the effects of IFN‐ɑ and Ascorbic Acid (AA) on HTLV‐1‐infected transformed cell lines (MT2, MT4, C8166).34, 35, 36 Although both drugs have shown moderate success in decreasing HTLV‐1‐induced proliferation,34, 35, 36, 37 only the high‐dose AA affected the retinoic acid pathway, specifically the shared RORC/Th17 pathway. Reanalysis of our transcriptomic data showed that neither IFN‐ɑ nor high‐dose AA altered RORC expression levels (log fold‐change = 0.042, p = 0.59 and log fold‐change =0.069, p = 0.39, respectively). Ascorbic acid stimulated an increase in expression of a key gene in Th17 differentiation, IL23R (log fold‐change = 0.81, p = 0.000024), in support of its possible use in (combination) therapy for ATL. RARɑ expression was unchanged by IFN‐ɑ (log fold change = 0.003, p = 0.98) but decreased by ascorbic acid (log fold change = −0.28, p = 0.064). Interestingly, our in vitro data (Brazilian cohort) demonstrated that RARɑ levels are upregulated upon in vitro treatment of ATL PBMCs with IFN‐β (log fold‐change = 0.31, p = 0.017), but not IFN‐ɑ. IFN‐β also significantly modulated the expression levels of STAT1, IRF4, TGFB1R, IL23R, FOXP3, and IL6, while IFN‐ɑ significantly altered BCL6 only, as shown in Supporting Information Figure S5. This is in accord with our recently demonstrated differential anti‐proliferative and proapoptotic effect of both IFN subtypes.38

Discussion

Upon transcriptomic meta‐analysis of four different cohorts, we found a specific and consistent RORClo phenotype in primary ATL cells and to a lesser extent in HTLV‐1‐infected individuals, in contrast to healthy controls. In addition, HTLV‐1‐infected individuals displayed an age‐dependent decrease in RORC expression. The observed two‐step decrease of RORC in ACs and ATL patients might thus represent an early event in HTLV‐1‐driven leukemogenesis. We also identified a small subset (12.0%) of RORChi ATL patients with significantly lower pathognomonic CADM1 and HBZ levels but similar levels of other ATL markers (CD4, CD25 and CCR4), hinting at a less aggressive ATL subtype.

ATL pathogenesis develops over decades as is seen by patients presenting at least 20 years after HTLV‐1 infection; yet not all infected patients develop ATL. Observational studies suggest that ATL, at least in the Caribbean and Brazil, can be triggered by the pediatric cutaneous manifestation known as Infectious Dermatitis.39, 40, 41, 42 ID is a chronic, eczematous condition with scaly, crusted lesions often superimposed by Staphylococcus aureus or Streptococcus pyogenes infections.39, 40 Defects in the Th17 axis increase vulnerability to S. aureus and Candida albicans infections, whereas in vivo S. aureus primed memory Th17 cells inhibited IL‐17 production and increased IL‐10 production.41, 43 Of interest, two recent papers have demonstrated a role for IL‐10 as an unexpected proliferative trigger of infected CD4+ T‐cell clones and, possibly, leukemogenesis.44, 45 Corroborating these findings, IL10 was found to be a significant (r = 0.36, p = 0.013) member of the proliferative gene module, together with PCNA and MKI67, in our WGCNA analysis. In addition, RNAseq analysis of purified cells with a CD4 + CCR4 + CD26‐CD7‐ leukemic phenotype from an independent UK cohort revealed IL17C is expressed in “ATL‐like” cells. In agreement with our proposed protective role, IL17C was significantly and negatively correlated with clonality (Fig. 6 b). Therefore, our findings underscore an IL‐10 vs. RORC/IL‐17 antagonism in HTLV‐1‐associated pathologies and provide a possible molecular basis for the epidemiological link between ID and ATL, alteration of the RORC/Th17 axis, and subsequent progression to leukemogenesis.

Modular transcriptomic analysis in ATL shows a strong correlation of the RORC pathway with cell proliferation and possibly oncogenesis, which supports its therapeutic potential. WGCNA analysis combined with CIBERSORT suggested the involvement of RORC pathway members in the homeostasis of resting memory and naïve CD4+ T‐cells. Combining the RORClo observation in ATL cohorts with our WGCNA analysis, we find that decreased RORC expression is correlated with proliferation and ATL driver genes (STAT3, NF‐kB). Thus, inducing RORC and switching to a RORChi phenotype may convert ATL cells to a less aggressive subtype, suggested by the lower CADM1 and HBZ levels seen in the RORChi subset (Fig. 3). However, no overlap was found between the module memberships of RORC, HBZ, and Tax in the WGCNA of Japanese Cohort #2 (data not shown). In addition, no RORC gene module members were significantly correlated to HBZ or Tax transcript levels, suggesting decreased RORC levels and signaling in ATL are not a direct consequence of retroviral transcription. Therefore, we hypothesized the RORC/IL17 axis might be linked to proliferation in other (lymphoid) leukemias. Indeed, the strongest negative correlation observed in both ATL cohorts, between IL17C and proliferation marker PCNA, was replicated in two large cohorts of other acute lymphoid leukemias, namely T‐ALL and B‐ALL (Fig. 6 cd).

Thus, our data reveal a widely prevalent antagonistic regulation between Th17 cells, usually considered as proinflammatory, and leukemic cell proliferation. Regarding the clinical translation of these results, antitumor immunotherapy using Th17 cells has recently shown promising results in animal models. Adoptive cell therapy using ex vivo Th17 cell selection enhanced antitumor activity,44, 46 to a greater extent than Th1 cells and other CD4+ T‐cells.44, 46 In addition, inducing IL17 expression via RORC stimulation would also subsequently alter the host immune response to reduce the risk of opportunistic infections by increasing Th17 cell count.10, 11, 12, 13, 14

Although most often believed to antagonize IL17 production, IFN‐β can trigger and even exacerbate IL17 production, especially in Th17‐mediated inflammatory diseases.45, 47 This becomes problematic in cases of multiple sclerosis, where 30–50% of patients are resistant to IFN‐β therapy.45 However, this same exacerbation could be useful in ATL as a means of increasing Th17 cell production and decreasing proliferation of leukemic clones. IFN‐β significantly alters the expression of more RORC pathway members than IFN‐ɑ (Supporting Information Fig. S5), a common therapeutic adjuvant to zidovudine in ATL treatment. This finding, along with the observation that IFN‐β has superior anti‐proliferative and proapoptotic properties compared to IFN‐ɑ,38 makes IFN‐β a novel, valuable option for combination therapy in ATL.

Recently, immune checkpoint inhibitors have come to the forefront of anticancer immunotherapy.46, 47 Immunotherapeutic targeting of Programmed death ligand −1 (PD‐L1) can increase Th17 cell count, restoring IL‐17A protein levels in naïve T‐cells of patients with a loss‐of‐function STAT3 mutation.46, 47, 48 Conversely, inducing Th17 cell differentiation by RORy agonist LYC‐54143 simultaneously reduced PD‐1+ cell numbers and PD‐1 expression in vitro, and resulted in tumor growth inhibition in vivo in two murine models.49 In ATL, PDL1 gene amplifications have been associated with worse prognosis, especially in aggressive subtypes.50 For PDL1 transcript levels, we observed a trend for positive correlation to CD4+ cells (r = 0.66, p = 0.091) as well as proliferation (r = 0.66, p = 0.075) in our Brazilian ATL cohort (Subramanian et al. unpublished), in agreement with a deleterious role for PD‐L1. Again, combination immunotherapy by RORy agonists and PD‐L1 blockade might be a more effective option in ATL, similar to the superior response rate to dual therapy with PD‐1 and CTLA‐4 blocking antibodies in advanced melanoma, as compared to monotherapy.51

In conclusion, we describe a predominant RORClo phenotype observed in four cohorts of ATL patients and a minor RORChi molecular subgroup with significantly lower mRNA levels of pathognomonic ATL biomarkers CADM1 and HBZ mRNA levels. An age‐dependent decline in RORC level indicates a possible early event in HTLV‐1‐driven leukemogenesis, supported by modular transcriptomic analysis of ATL patients, revealing a strong negative correlation of the RORC/IL17 pathway with proliferation, which was shared with T‐ALL and B‐ALL patients. Thus, inducing RORC levels and/or downstream signaling might represent (immuno)therapeutic benefit in ATL and possibly other acute lymphoid leukemias, which awaits further testing in clinical settings.

Methods

In silico analysis

RORC expression levels were examined in publicly available transcriptomic data sets from patients with ATL, HTLV‐1 infected asymptomatic controls, and healthy controls. A total of 135 untreated ATL patients, 12 HAM patients, 40 asymptomatic controls (AC), and 242 healthy controls (HC) from the Gene Expression Omnibus datasets GSE55851, GSE33615, GSE19080, GSE85487, and the European Genome‐phenome archive EGAD1001411 dataset were used in our study (Table 1). EGAD1001411 initially contained 45 ATL patients, but one outlier with an overall strongly divergent transcriptome was removed. The effect of age on RORC expression was investigated in the Healthy Estonian Cohort for healthy controls (n = 293) and the UK Cohort for HTLV‐1 infected individuals (n = 30).

The Japanese Cohort #2 (EGAD1001411) RNA‐Seq data was quality‐ and adapter‐cleaned using trimmomatic52 and cutadapt53 and quantified with kallisto54 using an index built on the transcriptome obtained from the Genome Reference Consortium GRCh38, rel79. CIBERSORT was used to generate an in silico approximation of the relative composition of 22 immune cell types in the samples.32

To facilitate consistent analysis of both the microarray and RNA‐Seq data, the ensemble and/or Agilent IDs of the datasets were matched with corresponding Entrez IDs using the biomaRt package55, 56 in R. The Entrez IDs were verified with the associated GPL files on GEO where available. Considering transcriptomic analysis of the Caribbean Cohort was performed on a limited (nongenome‐wide) microarray platform, 2,134 Entrez IDs were common to all examined microarrays and comprised the list of genes examined in our study. To address the bias in the measurements inherent to each platform, we adapted the quantile discretization method proposed by Warnat et al.57 and transformed gene expression levels into percentile ranks among the surveyed 2,134 genes for the meta‐analysis. To further exclude the possibility of biasing our results, we refrain from making direct statistical comparisons of gene expression levels between datasets.

Published literature on RORC and RORγ, as cited and detailed in the results section, was used to develop a consensus molecular pathway, which was validated using STRING (version 10.5) protein–protein interaction enrichment analysis (www.string-db.org) using the whole genome as background.

Weighted Gene Correlation Network Analysis (WGCNA)58 clusters genes into modules according to their topological overlap measure which quantifies how many gene‐correlates were common to both members of each gene pair. To determine coherent gene modules and their correlation to clinical and molecular data, we performed WGCNA on each of the transcriptomic datasets from two independent ATL cohorts recently published by our group: in vitro gene expression data from short‐term cultured ATL patient PBMCs (n = 8, Brazilian Cohort) performed in parallel with lymphoproliferation, and ex vivo expression data from ATL patient PBMCs (n = 44) of Japanese Cohort #2.25 Module membership of the RORC gene set and the ATL signature genes were determined and correlated to demographic, clinical, and in vitro data.

In vitro analysis

Spontaneous lymphoproliferation of primary cells (PBMC) from ATL patients (n = 8, Brazilian Cohort) was measured by [3H]‐thymidine incorporation, as described previously.38

Statistical analysis

Statistical analysis was performed using GraphPad Prism 7.0. Differences in RORC gene expression were analyzed by Kruskall‐Wallis test for Japanese Cohorts #1 and #2, and the Caribbean Cohort. For Japanese Cohort #3, where ACs were not included, Mann–Whitney was used to compare HC and ATL patients. The false discovery rate two‐stage method of Benjamini, Krieger, and Yekutieli was used to correct for multiple comparisons. Spearman's Rho was used to correlate gene expression (either per gene or per WGCNA module using their eigengene expression) to demographic (age), clinical data (patient survival) and in vitro data (proliferation and apoptosis).

Supporting information

Supplementary Figure S1. RORC percentile ranks for four independent cohorts consisting of ATL patients and healthy uninfected (HC) and/or HTLV‐1 infected healthy controls (AC) showed a consistent decrease in expression in ATL (A‐D). HCs = Healthy Controls, ACs = Asymptomatic Controls, ATL = Adult T‐cell Lymphoma/Leukemia Patients, HAM = HTLV‐1‐Associated Myelopathy patients. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Supplementary Figure S2. Differential RORC expression of ATL patients from Japanese Cohort #2 is independent of ATL driver genes and genes involved in IFN/AZT response.

Supplementary Figure S3. RORC and downstream IL17 family genes are poorly expressed in both asymptomatic HTLV‐1 infected individuals (top) and ATL patients (bottom).

Supplementary Figure S4. RORC and downstream IL17 family genes cluster in an antiproliferative gene module. Using WGCNA analysis, a significant bifurcation in the RORC pathway is observed when correlation coefficients of member genes from the antiproliferative module (Turquoise) were regressed with the correlation coefficients from the proliferative (Blue) module (r = −0.97, p < 0.0001) in primary cells from ATL patients (Brazilian cohort).

Supplementary Figure S5: Differential effects of IFN‐β vs. IFN‐ɑ upon RORC pathway members in primary ATL cells. Blue represents genes which had significantly (p < 0.05) altered expression levels with IFN‐β treatment. BCL6 (Red) was the only gene member which was significantly altered by IFN‐ɑ treatment.

References

  • 1. Proietti FA, Carneiro‐Proietti ABF, Catalan‐Soares BC, et al. Global epidemiology of HTLV‐I infection and associated diseases. Oncogene 2005;24:6058–68. [DOI] [PubMed] [Google Scholar]
  • 2. Gallo RC, Willems L, Tagaya Y. Time to go Back to the original name. Front Microbiol 2017;8:1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Tagaya Y, Gallo RC. The exceptional Oncogenicity of HTLV‐1. Front Microbiol 2017;8:1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Arisawa K, Soda M, Endo S, et al. Evaluation of adult T‐cell leukemia/lymphoma incidence and its impact on non‐Hodgkin lymphoma incidence in southwestern Japan. Int J Cancer 2000;85:319–24. [DOI] [PubMed] [Google Scholar]
  • 5. Nakahata S, Saito Y, Marutsuka K, et al. Clinical significance of CADM1/TSLC1/IgSF4 expression in adult T‐cell leukemia/lymphoma. Leukemia 2012;26:1238–46. [DOI] [PubMed] [Google Scholar]
  • 6. Takatsuki K. Discovery of adult T‐cell leukemia. Retrovirology 2005;2:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Iwanaga M, Watanabe T, Yamaguchi K. Adult T‐cell leukemia: a review of epidemiological evidence. Front Microbiol 2012;3:322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Bittencourt AL, Vieira M d G, Brites CR, et al. Adult T‐cell leukemia/lymphoma in Bahia, Brazil: analysis of prognostic factors in a group of 70 patients. Am J Clin Pathol 2007;128:875–82. [DOI] [PubMed] [Google Scholar]
  • 9. Sasaki H, Nishikata I, Shiraga T, et al. Overexpression of a cell adhesion molecule, TSLC1, as a possible molecular marker for acute‐type adult T‐cell leukemia. Blood 2005;105:1204–13. [DOI] [PubMed] [Google Scholar]
  • 10. Sawada Y, Nakamura M, Kabashima‐Kubo R, et al. Defective epidermal innate immunity and resultant superficial Dermatophytosis in adult T‐cell leukemia/lymphoma. Clin Cancer Res 2012;18:3772–9. [DOI] [PubMed] [Google Scholar]
  • 11. Leal FE, Ndhlovu LC, Hasenkrug AM, et al. Expansion in CD39+ CD4+ Immunoregulatory T cells and rarity of Th17 cells in HTLV‐1 infected patients is associated with neurological complications. PLoS Negl Trop Dis 2013;7:e2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Conti HR, Peterson AC, Brane L, et al. Oral‐resident natural Th17 cells and γδ T cells control opportunistic Candida albicans infections. J Exp Med 2014;211:2075–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Tesmer LA, Lundy SK, Sarkar S, et al. Th17 cells in human disease. Immunol Rev 2008;223:87–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Curtis MM, Way SS. Interleukin‐17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology 2009;126:177–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Swaims AY, Khani F, Zhang Y, et al. Immune activation induces immortalization of HTLV‐1 LTR‐tax transgenic CD4+ T cells. Blood 2010;116:2994–3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Dodon MD. Tax protein of human T‐cell leukaemia virus type 1 induces interleukin 17 gene expression in T cells. J Gen Virol 2004;85:1921–32. [DOI] [PubMed] [Google Scholar]
  • 17. Elias KM, Laurence A, Davidson TS, et al. Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat‐3/Stat‐5 independent signaling pathway. Blood 2007;111:1013–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Bangham CRM, Cook LB, Melamed A. HTLV‐1 clonality in adult T‐cell leukaemia and non‐malignant HTLV‐1 infection. Semin Cancer Biol 2014;26:89–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lin J, Tang H, Jin X, et al. p53 regulates Stat3 phosphorylation and DNA binding activity in human prostate cancer cells expressing constitutively active Stat3. Oncogene 2002;21:3082–8. [DOI] [PubMed] [Google Scholar]
  • 20. Ueda E, Kurebayashi S, Sakaue M, et al. High incidence of T‐cell lymphomas in mice deficient in the retinoid‐related orphan receptor RORgamma. Cancer Res 2002;62:901–9. [PubMed] [Google Scholar]
  • 21. Liljevald M, Rehnberg M, Söderberg M, et al. Retinoid‐related orphan receptor γ (RORγ) adult induced knockout mice develop lymphoblastic lymphoma. Autoimmun Rev 2016;15:1062–70. [DOI] [PubMed] [Google Scholar]
  • 22. Tattermusch S, Skinner JA, Chaussabel D, et al. Systems biology approaches reveal a specific interferon‐inducible signature in HTLV‐1 associated myelopathy. PLoS Pathog 2012;8:e1002480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ohnmacht C, Park J‐H, Cording S, et al. The microbiota regulates type 2 immunity through ROR t+ T cells. Science 2015;349:989–93. [DOI] [PubMed] [Google Scholar]
  • 24. Motulsky HJ, Brown RE. Detecting outliers when fitting data with nonlinear regression – a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics 2006;7:1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kataoka K, Nagata Y, Kitanaka A, et al. Integrated molecular analysis of adult T cell leukemia/lymphoma. Nat Genet 2015;47:1304–15. [DOI] [PubMed] [Google Scholar]
  • 26. Farre L, Bittencourt AL, Silva‐Santos G, et al. Fas 670 promoter polymorphism is associated to susceptibility, clinical presentation, and survival in adult T cell leukemia. J Leukoc Biol 2007;83:220–2. [DOI] [PubMed] [Google Scholar]
  • 27. Ramos JC, Ruiz P Jr, Ratner L, et al. IRF‐4 and c‐Rel expression in antiviral resistant adult T‐cell leukemia/lymphoma. Blood 2006;109:3060–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 2009;9:798–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Muranski P, Restifo NP. Essentials of Th17 cell commitment and plasticity. Blood 2013;121:2402–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Khouri R, Silva‐Santos G, Farre L, et al. Superior antiviral and antiproliferative activity of IFN‐beta vs. IFN‐alpha in primary ATL cells occurs downstream of STAT1 signaling. Retrovirology 2014;11:O22. [Google Scholar]
  • 31. Khouri R, Silva‐Santos G, Dierckx T, et al. A genetic IFN/STAT1/FAS axis determines CD4 T stem cell memory levels and apoptosis in healthy controls and adult T‐cell leukemia patients. OncoImmunology 2018;7:e1426423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Newman AM, Liu CL, Green MR, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 2015;12:453–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kagdi H, Demontis MA, Ramos JC, et al. Switching and loss of cellular cytokine producing capacity characterize in vivo viral infection and malignant transformation in human T‐ lymphotropic virus type 1 infection. PLoS Pathog 2018;14:e1006861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Satou Y, Yasunaga J, Zhao T, et al. HTLV‐1 bZIP factor induces T‐cell lymphoma and systemic inflammation in vivo. PLoS Pathog 2011;7:e1001274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Mizuguchi M, Sasaki Y, Hara T, et al. Induction of cell death in growing human T‐cells and cell survival in resting cells in response to the human T‐cell leukemia virus type 1 tax. PLoS One 2016;11:e0148217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Moens B, Decanine D, Menezes SM, et al. Ascorbic acid has superior ex vivo Antiproliferative, cell death‐inducing and Immunomodulatory effects over IFN‐α in HTLV‐1‐associated myelopathy. PLoS Negl Trop Dis 2012;6:e1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Moens B, Pannecouque C, López G, et al. Simultaneous RNA quantification of human and retroviral genomes reveals intact interferon signaling in HTLV‐1‐infected CD4+ T cell lines. Virol J 2012;9:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Dierckx T, Khouri R, Menezes SM, et al. IFN‐β induces greater antiproliferative and proapoptotic effects and increased p53 signaling compared with IFN‐α in PBMCs of adult T‐cell leukemia/lymphoma patients. Blood Cancer J 2017;7:e519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Farre L, Paim De Oliveira MDF, Primo J, et al. Early sequential development of infective dermatitis, human T cell Lymphotropic virus type 1‐associated myelopathy, and adult T cell leukemia/lymphoma. Clin Infect Dis 2008;46:440–2. [DOI] [PubMed] [Google Scholar]
  • 40. Di Martino Ortiz B, Riveros R, Medina R, et al. Infective dermatitis in an adult patient with HTLV‐1. Am J Dermatopathol 2015;37:944–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. de Oliveira M d FSP, Fatal PL, Primo JRL, et al. Infective dermatitis associated with human T‐cell Lymphotropic virus type 1: evaluation of 42 cases observed in Bahia, Brazil. Clin Infect Dis 2012;54:1714–9. [DOI] [PubMed] [Google Scholar]
  • 42. Hanchard B, Lagrenade L, Carberry C, et al. Childhood infective dermatitis evolving into adult T‐cell leukaemia after 17 years. Lancet 1991;338:1593–4. [DOI] [PubMed] [Google Scholar]
  • 43. Zielinski CE, Mele F, Aschenbrenner D, et al. Pathogen‐induced human TH17 cells produce IFN‐γ or IL‐10 and are regulated by IL‐1β. Nature 2012;484:514–8. [DOI] [PubMed] [Google Scholar]
  • 44. Bowers JS, Nelson MH, Majchrzak K, et al. Th17 cells are refractory to senescence and retain robust antitumor activity after long‐term ex vivo expansion. JCI Insight 2017;2:e90772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Axtell RC, Raman C, Steinman L. Interferon‐β exacerbates Th17‐mediated inflammatory disease. Trends Immunol 2011;32:272–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Melero I, Berman DM, Aznar MA, et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer 2015;15:457–72. [DOI] [PubMed] [Google Scholar]
  • 47. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012;12:265–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Zhang H, Chen L, Cai S‐H, et al. Identification of TBK1 and IKKε, the non‐canonical IκB kinases, as crucial pro‐survival factors in HTLV‐1‐transformed T lymphocytes. Leuk Res 2016;46:37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Hu X, Liu X, Moisan J, et al. Synthetic RORγ agonists regulate multiple pathways to enhance antitumor immunity. OncoImmunology 2016;5:e1254854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Kataoka K, Iwanaga M, Yasunaga J‐I, et al. Prognostic relevance of integrated genetic profiling in adult T‐cell leukemia/lymphoma. Blood 2018;131:215–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus Ipilimumab in advanced melanoma. N Engl J Med 2013;369:122–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Martin M. Cutadapt removes adapter sequences from high‐throughput sequencing reads. EMBnet.journal 2011;17:10. [Google Scholar]
  • 54. Bray NL, Pimentel H, Melsted P, et al. Near‐optimal probabilistic RNA‐seq quantification. Nat Biotechnol 2016;34:525–7. [DOI] [PubMed] [Google Scholar]
  • 55. Durinck S, Moreau Y, Kasprzyk A, et al. BioMart and bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics 2005;21:3439–40. [DOI] [PubMed] [Google Scholar]
  • 56. Durinck S, Spellman PT, Birney E, et al. Mapping identifiers for the integration of genomic datasets with the R/bioconductor package biomaRt. Nat Protoc 2009;4:1184–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Warnat P, Eils R, Brors B. Cross‐platform analysis of cancer microarray data improves gene expression based classification of phenotypes. BMC Bioinformatics 2005;6:265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008;9:559. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Figure S1. RORC percentile ranks for four independent cohorts consisting of ATL patients and healthy uninfected (HC) and/or HTLV‐1 infected healthy controls (AC) showed a consistent decrease in expression in ATL (A‐D). HCs = Healthy Controls, ACs = Asymptomatic Controls, ATL = Adult T‐cell Lymphoma/Leukemia Patients, HAM = HTLV‐1‐Associated Myelopathy patients. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Supplementary Figure S2. Differential RORC expression of ATL patients from Japanese Cohort #2 is independent of ATL driver genes and genes involved in IFN/AZT response.

Supplementary Figure S3. RORC and downstream IL17 family genes are poorly expressed in both asymptomatic HTLV‐1 infected individuals (top) and ATL patients (bottom).

Supplementary Figure S4. RORC and downstream IL17 family genes cluster in an antiproliferative gene module. Using WGCNA analysis, a significant bifurcation in the RORC pathway is observed when correlation coefficients of member genes from the antiproliferative module (Turquoise) were regressed with the correlation coefficients from the proliferative (Blue) module (r = −0.97, p < 0.0001) in primary cells from ATL patients (Brazilian cohort).

Supplementary Figure S5: Differential effects of IFN‐β vs. IFN‐ɑ upon RORC pathway members in primary ATL cells. Blue represents genes which had significantly (p < 0.05) altered expression levels with IFN‐β treatment. BCL6 (Red) was the only gene member which was significantly altered by IFN‐ɑ treatment.


Articles from International Journal of Cancer are provided here courtesy of Wiley

RESOURCES