You are on page 1of 7

Cytokine 61 (2013) 885–891

Contents lists available at SciVerse ScienceDirect

Cytokine
journal homepage: www.journals.elsevier.com/cytokine

Cytokine profiles in acute myeloid leukemia patients at diagnosis: Survival is


inversely correlated with IL-6 and directly correlated with IL-10 levels
Beatriz Sanchez-Correa a, Juan M. Bergua b, Carmen Campos c, Inmaculada Gayoso c, Maria Jose Arcos b,
Helena Bañas b, Sara Morgado a, Javier G. Casado a,d, Rafael Solana c, Raquel Tarazona a,⇑
a
Immunology Unit, Department of Physiology, University of Extremadura, 10003 Caceres, Spain
b
Department of Hematology, Hospital San Pedro de Alcantara, Caceres, Spain
c
Immunology Unit, Instituto Maimonides para la Investigacion Biomedica de Cordoba (IMIBIC), University of Cordoba, Cordoba, Spain
d
Stem Cell Therapy Unit, Minimally Invasive Surgery Centre Jesus Uson, Caceres, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Background: Several evidences support the existence of cytokine deregulation in acute myeloid leukemia
Received 29 August 2012 (AML) patients that may be associated with pathogenesis, disease progression and patient survival.
Received in revised form 19 December 2012 Methods: In the present study, we analyzed plasma levels of pro- and anti-inflammatory cytokines in
Accepted 22 December 2012
AML patients and age-matched healthy donors. TNF-a, IL-6, IL-1b, IL-2, IFN-c, IL-17A, IL-12p70, IL-8,
Available online 26 January 2013
IL-10, IL-4 and IL-5 were analyzed using fluorescent bead-based technology and TGF-b by ELISA tech-
nique. Because age-associated differences in cytokine profiles have been described, patients and healthy
Keywords:
individuals were divided into two age groups: up to 65 years and over 65 years.
Acute myeloid Leukemia
Pro-inflammatory cytokines
Results: Our results showed that plasma TNF-a, IL-6 and IL-10 levels were higher in AML patients from
IL-6 both groups of age. IL-8 was increased in AML patients less than 65 years while the plasma concentra-
Survival tions of IL-4, IL-5 and IL-12p70 were significantly higher only in elderly AML patients compared with
IL-10 aged-matched healthy controls. Moreover, plasma levels of IL-6 and IL-10 were associated with patient
survival and event-free survival.
Conclusions: An aberrant production of the pro-inflammatory cytokines IL-6 and TNF-a and the anti-
inflammatory cytokine IL-10 is observed in AML patients. Low levels of IL-6 and high levels of IL-10 rep-
resent favorable prognostic factors for survival in AML patients. These results support the idea that cyto-
kine deregulation may be useful as a marker for predicting clinical evolution in AML patients.
Ó 2013 Elsevier Ltd. All rights reserved.

1. Introduction In AML patients, cytokines can be produced by both leukemic


blasts and cells of the immune system and the role of cytokines
Acute myeloid leukemia (AML) represents a group of clonal in the pathogenesis of acute leukemia has not been fully clarified
hematopoietic stem cell disorders in which both failure to differen- [9,10]. Thus, aberrant cytokine signaling is a feature of leukemia
tiate and overproliferation in the stem cell compartment result in that may contribute to proliferation, blast survival, resistance to
accumulation of non-functional myeloid cells termed myeloblasts chemotherapy and patients’ prognosis [10–14].
and loss of normal hematopoietic function. Considerable efforts At the tumor site, the presence of pro-inflammatory cytokines
have been spent in identifying prognostic markers that might pre- as IL-6 or IL1-b constitutes a hallmark of cancer-associated inflam-
dict clinical outcomes in AML patients and several characteristics mation. The major sources of IL-6 and IL-1 are activated monocytes
including older age, cytogenetics and performance status are com- or macrophages and their production is induced by TNF-a, a pleio-
monly used as predictors of survival [1,2]. Prognosis in AML pa- tropic cytokine mainly produced by macrophages but also by acti-
tients has also been related to the frequently observed defective vated NK cells, neutrophils, CD4 T helper 1 (Th1) cell subset and
function of their immune system that hampers the development CD8 T cells [15]. It has been demonstrated that these cytokines
of an effective response against leukemic blasts [3–8]. can be produced by AML blasts [16,17]. In the tumor microenviron-
Cytokines are secreted by different types of cells in response to ment, IL-1b promotes angiogenesis and tumor invasiveness [18]
a variety of stimuli such as tissue damage or infection and regulate and may suppress immune function by enhancing the accumula-
the immune response and other biologic processes. tion of myeloid-derived suppressor cells [19]. IL-1b may act as an
autocrine growth factor for AML and chronic myeloid leukemia
⇑ Corresponding author. Tel.: +34 927257000x51322; fax: +34 927257106. [20]. IL-6, a cytokine with pleiotropic inflammatory effects, has
E-mail address: rtarazon@unex.es (R. Tarazona).
demonstrated different effects (stimulatory, inhibitory or neutral)

1043-4666/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.cyto.2012.12.023
886 B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891

on the growth of leukemic blasts [14,21]. IL-6 may also exert anti- 2.3. Cytokine measurement
inflammatory biological activity by enhancing plasma IL-1 receptor
antagonist (IL1ra), IL-10 and cortisol in humans [22]. The concentrations of TNF-a, IL-6, IL-10, IL-1-b, IL-8, IL-5, IL-4,
Interactions between pro- and anti-inflammatory cytokines IL-2, IL-12, IFN-c and IL-17A in the plasma of AML patients and
regulate cytokine response. Thus, TNF-a and IL-1b can stimulate healthy controls were determined by using the commercially
their own and each other’s production and also IL-6 secretion available fluorescent bead immunoassay FlowCytomix human
[23,24]. This represents an important amplification loop of the Th1/Th2 11-plex kit (eBiosciences, Vienna, Austria). The cytokine
inflammatory response. On the other hand, IL-6 and TNF-a may in- measurement was performed following the instructions of man-
duce IL-10 secretion with potential to inhibit IL-6, TNF-a and IL-1b ufacturers. Four independent sets of experiments were per-
production. Changes in one of these cytokines lead to compensa- formed. Each Flowcytomix experiment included the kit’s
tory mechanisms that may alter the cytokine network [22,25]. standards run in triplicate and samples from AML patients and
Adverse clinical outcomes in elderly individuals have been asso- healthy controls. No significant variations were observed among
ciated with the increase of pro-inflammatory cytokines [26,27]. It the experiments.
has been described that the production of several cytokines in Standard curves were determined for each cytokine from a
the elderly is clearly influenced by health status and that normal range of 27.44–20,000 pg/mL. The lower limits of detection for all
IL-6 levels may be considered a good overall biomarker of healthy cytokines according to the manufacturer are between 0.5 and
aging [28–31]. 20.8 pg/mL. Samples were acquired in a FACScan flow cytometer
Here, we examined the plasma levels of 12 cytokines in 42 AML (BD Biosciences) and data were analyzed using FlowCytomix Pro
patients and 58 healthy adult volunteers, and analyzed the associ- 2.2 Software (eBiosciences). The standards used for determining
ations between the concentration of each cytokine, clinical charac- cytokine levels were measured in triplicate.
teristics, disease progression and survival of AML patients. To avoid TGF-b1 concentrations in the plasma were determined by en-
potential confounding effects from age-associated differences in zyme linked immunosorbent assay (ELISA) using a Human TGF-
cytokine profiles, we have analyzed plasma cytokines in AML pa- b1 kit (eBiosciences), according to the manufacturer’s instructions.
tients and healthy controls distributed in two groups according Standards were run in triplicate and samples were run in duplicate.
to their age. Four independent sets of experiments were performed. No signifi-
cant variations were observed among the experiments.

2.4. Statistical analysis


2. Materials and methods
Statistical analyses were performed using SPSS V.15 statistical
2.1. Patients and healthy donors software (SAS Institute, Cary, NC). The concentrations of each
cytokine were compared among the AML patients and healthy vol-
Forty-two AML patients (27 males, 15 females) with an age unteers using the Kruskal–Wallis test. The AML patients were di-
range of 21–86 years (median age, 62 ± 16 years) newly diagnosed vided into two groups according to the plasma level of each
at the Hospital San Pedro de Alcantara from January 2006 through cytokine; cut-off values for each cytokine were set using Receiver
April 2010 were included in this study. Patients with acute promy- Operating Characteristic (ROC) curves. The optimal cut-off point
elocytic leukemia were not included. Diagnoses were established for each cytokine was determined by the point of convergence of
according to the French–American–British (FAB) classification sys- sensitivity and specificity (i.e. by simultaneously maximizing the
tem. AML samples were collected at diagnosis (M0 = 10, M1 = 5, two). In our study, P 6 0.05 was considered as significant. Survival
M2 = 6, M4 = 4 and M5 = 17). analyses were performed by the Kaplan–Meier method, and
The control group was population-based and the cases were survival curves were compared using the log-rank and Breslow–
age-matched healthy volunteers. Fifty-eight healthy controls (33 Gehan–Wilcoxon tests. The starting time for survival was the date
males, 25 females) were included as controls. Samples were col- of blood collection. For survival analysis, all patients who were still
lected from control subjects only if the subjects had not had fever alive were censored at the date of last follow-up. Event free
within 1 week, were not receiving any medications, were not survival (EFS) was established as the interval of time between
known to be pregnant, and did not have a history of any chronic diagnosis and the relapse or death or documented failure of
disease. Due to the potential effects of age in the levels of cyto- treatment.
kines, we divided AML patients and healthy controls into two
groups: from 21 to 65 years of age (20 AML patients and 28 healthy
donors) and over 65 years (22 AML patients and 30 healthy 3. Results
donors).
The study was approved by the local Ethics Committee (2006/ 3.1. Plasma cytokine concentrations in AML patients and healthy adult
01) and samples collected after written informed consent in accor- volunteers
dance with the Declaration of Helsinki.
Concentrations of plasma cytokines in each group are shown in
Fig. 1. We have observed that the concentrations of TNF-a, IL-6 and
IL-10 were significantly higher in AML patients than in the healthy
2.2. Procedures of sample collection and processing volunteers in both groups of age (P = 0.003, P = 0.002 and P = 0.005,
respectively in the younger age group and P = 0.001, P = 0.04 and
Plasma was obtained by centrifugation of heparinized periphe- P = 0.009 in the elderly donors). TGF-b concentration was signifi-
ral blood and stored in aliquots at 80 °C until analysis. All the cantly lower in AML patients than in the healthy volunteers in both
analyses were performed on samples that had not been previously age groups (P = 0.004 and P = 0.0001).
thawed. The time from sample collection to processing was always IL-4, IL-5 and IL-12p70 levels were significantly higher in el-
lower than 24 months. Samples were kept in a freezer connected to derly AML patients compared to their aged-matched healthy vol-
an uninterruptible power supply to guarantee the maintenance of unteers (P = 0.048, P = 0.002 and P = 0.018 respectively). IL-8 level
optimum temperature. was higher in AML patients under 65 years when compared with
B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891 887

Fig. 1. Comparison of plasma cytokine concentrations in healthy donors and AML patients. The concentrations of cytokines in plasma samples obtained from 58 healthy
donors (28 6 65 years (d) and 30 (j) elderly) and 42 AML patients (20 patients 6 65 years (o) and 22 elderly (h) patients) are shown. The horizontal bars represent the mean
values. P 6 0.05, P 6 0.01 and P 6 0.001. HD: healthy donor.

their age matched healthy donors (P = 0.006). There were no 3.2. Correlations among the cytokines, FAB subtypes, blast and
significant differences between AML patients and healthy donors leukocyte counts
in the plasma concentrations of IL-1b, IL-17A, IFN-c and IL-2.
In relation to the effect of age in healthy donors and AML pa- There was no significant correlation between blast or leukocyte
tients we observed that IL-6 and IL-8 levels were higher in healthy counts with the plasma levels of all cytokines analyzed in our
elderly than in healthy donors under 65 years (P = 0.001 and study. The analysis of the cytokines involved in Th17 differentia-
P = 0.001 respectively). The only cytokine that showed statistically tion showed that higher IL-1b levels correlated with higher levels
significant age-associated differences in AML patients was IL-8 that of IL-17A (r = 0.91; P < 0.01). In contrast, IL-6 (r = 0.23) and TGF-b
was higher in elderly AML patients compared to AML patients less (r = 0.062) did not show any correlation with the concentration
than 65 years (P = 0.014). of IL-17A in AML patients.
888 B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891

Fig. 2. Kaplan–Meier survival curve for overall survival stratified by cytokine levels. We analyzed cytokine levels and overall survival in 42 AML patients. Kaplan–Meier
curves were performed according to IL-6 (a), IL-17A (b) and simultaneous detection of IL-6 and/or IL-17A (c). Statistical significance was calculated according to the log-rank
method. Values in parenthesis represent total number of patients/number of deaths.

No correlation was found between cytokine levels and FAB sub- patients compared to healthy donors and no correlation was de-
types that may be the consequence of the limited number of pa- tected between IL-6 and IL-1b levels. It has been also shown that
tients in each category. TNF-a, IL-1b, and IL-17 interact together to induce massive
amounts of IL-6 and this cytokine has demonstrated a role in mod-
ulating Th17/regulatory T cell balance [24]. In healthy elderly indi-
3.3. Association between plasma cytokine levels and survival among
viduals it has been reported that increased levels of TNF-a, IL-6 and
the AML patients
IL-1ra were associated with increased risk of death [26]. IL-6 has a
key role in the regulation of plasma levels of YKL-40, a glycoprotein
Overproduction of IL-17 has been associated with elevated pro-
that is elevated during inflammatory conditions [37] and high lev-
duction of pro-inflammatory mediators such as IL-6 [32]. In AML
els of YKL-40 have been correlated with short survival in AML pa-
patients IL-6 levels have been correlated positively with Th17 fre-
tients [38]. In AML patients IL-6 levels have been correlated
quencies suggesting a positive feedback loop [33].
positively with Th17 frequencies [33]. It has been suggested that
We have observed that patients with higher levels of IL-6 had
IL-17 acting on stromal cells sustains NF-kB-mediated IL-6 produc-
decreased survival compared with those patients with lower levels
tion, which in turn enhances Th17 differentiation in a positive
(log rank P = 0.05). Low levels of IL-6 were also associated with
feedback loop [32].
longer median event-free survival (EFS). The Kaplan–Meier curves
Pro-inflammatory cytokines as IL-1b and TNF-a can be counter-
and results of the log rank tests are shown in Fig. 2a. Plasma levels
balanced by the secretion of antagonist as IL-1ra or soluble TNF
of IL-17A were not associated with overall survival (Fig. 2b). Sur-
receptors. Another level of cytokine regulation is controlled by
vival analysis according to the plasma levels of IL-6 and IL-17A
anti-inflammatory cytokines such as IL-10 that shutdown the pro-
showed that AML patients who had both IL-6 and IL-17A elevated
duction of inflammatory cytokines [25]. This loop is tightly regu-
had a decreased survival (Fig. 2c) compared with patients with no
lated to govern the level of inflammation in response to a
elevation of these cytokines or with an increase of one of them (log
stimulus. Nevertheless, genetic variations (e.g. IL-10 promoter
rank P = 0.017). On the contrary, higher levels of IL-10 were associ-
polymorphisms) may explain some inter-individual differences
ated with longer survival and EFS (Table 1).
[25]. In accordance with previous reports [14], we have found that
Age was associated with complete remission (CR), median sur-
IL-10 levels are increased in AML patients. In our study patients
vival and median EFS as shown in Table 1. Regarding cytokine con-
were stratified according to age and the differences were statisti-
centrations, only IL-8 was associated with the achievement of CR.
cally significant in both groups of age when compared to age-
TNF-a, IL-1b, IL-2, IL-12, IFN-c, IL-5, IL-4 and TGF-b plasma levels
matched healthy donors. Here we also described an association
at diagnosis did not correlate with survival of AML patients.
of high IL-10 levels with median survival and median EFS. Kornb-
lau et al. [13] have previously shown that patients who attained
4. Discussion remission were more likely to have higher levels of IL-10. Con-
versely, Tsimberidou et al. [14] described no statistically significant
Cytokine deregulation has been postulated to play a role in the association of IL-10 levels with CR and disease progression. The
pathogenesis of several hematological malignances, including association of high levels of IL-10 with longer survival times can
AML, and plasma cytokine levels have been correlated with disease be explained by its capacity to inhibit granulocyte and granulo-
progression and survival [11–14,34]. Our results agree with previ- cyte-macrophage colony stimulating factors (G-CSF and GM-CSF)
ous authors who reported an increase of IL-6 in AML patients production and, consequently, blast proliferation [39]. IL-10 also
[33,35,36]. We have found that high levels of IL-6 were correlated inhibits IL-6, IL-1b and TNF-a, cytokines that have been associated
with lower survival and EFS. In addition, AML patients that con- with lower survival in different diseases [14,20,25].
comitantly exhibited high levels of IL-6 and IL-17A had lower sur- On the other hand, IL-10 displays strong immunosuppressive
vival. Recent findings suggest that more than one mechanism is effects inhibiting the proliferation of T cells and the production
involved in the promotion of abnormal IL-6 expression in cancer of IFN-c and IL-2 [40]. Thus, evidences for both tumor promoting
patients [20], whereas in some cases, high levels of IL-1b have been and anti-tumor function of IL-10 have been reported [41,40]. A bet-
found to be responsible of the induction of IL-6, in other patients ter understanding of the dual biological effects of IL-10 will be
no association has been observed between IL-6 and IL-1b. We have important for dissecting its role in pathology and may provide
not found statistically significant differences in IL-1b levels in AML new avenues for developing successful therapies against cancer.
B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891 889

Table 1
Treatment response, overall survival and event free survival according to cytokine levels.

No. CR (%) P Median survival (mo) P Median EFS (mo) P


Age (years)
665 (range 21–65; median 55) 20 75 0.004 26 <0.001 19.75 <0.001
>65 (range 66–86; median 74) 22 31 5.87 3.74
Cytogenetics (risk)
Favorable 9 88.9 0.06 31.1 0.004 22.67 0.014
Intermediate 10 60 14 11.66
Poor 19 42.1 8.7 5.77
WBC (103/lL)
<10 11 63.6 0.73 17.7 0.85 14.79 0.75
>10 27 55.5 14.5 9.9
Circulating blasts (%)
<50 14 54.3 0.5 17.1 0.9 13.74 0.36
>50 24 54.17 15.1 10.82
TNF-a, pg/ml
<10 10 50 0.71 16.3 0.45 14.1 0.29
>10 32 53.1 15.1 10.3
IL-6 (pg/ml)
<1 22 68.2 0.09 21.1 0.05 15.98 0.003
>1 20 35 8.37 5.57
IL-10 (pg/ml)
<10 11 72.7 0.14 10.6 0.014 8.45 0.023
>10 31 45.16 17.1 12.35
TGF-b (ng/ml)
<4 12 41.7 0.4 18.9 0.21 12.31 0.31
>4 21 23.8 8.9 6.99
IL-8 (pg/ml)
<9 8 87.5 0.014 18.5 0.16 13.36 0.51
>9 34 44.1 14.7 10.86
IL-1b (pg/ml)
<1 9 22.2 0.26 16.8 0.14 14.17 0.06
>1 33 60.6 14.9 10.31
IL-4 (pg/ml)
<5 11 54.5 1 17.7 0.62 15.22 0.32
>5 31 51.6 14.6 9.93
IL-5 (pg/ml)
<1.25 22 50 0.52 14.5 0.47 12.09 0.71
>1.25 20 55 16.4 10.47
IL-12p70 (pg/ml)
<1 25 56 1 14 0.24 10.67 0.14
>1 17 47 17.8 12.44
IFN-c (pg/ml)
<5 18 55.5 1 15.5 0.86 13.78 0.36
>5 24 50 15.3 9.34
IL-17A (pg/ml)
<4 21 57.1 1 15 0.65 12.47 0.79
>4 21 47.6 15.9 10.05
IL-2 (pg/ml)
<20 16 62.5 0.5 19.6 0.06 15.36 0.16
>20 26 46.1 12.7 8.69

CR indicates complete remission; EFS, event-free survival; mo, months; WBC, white blood cell count. Cut-off values were established using ROC curves.

In our study, the only cytokine associated with CR was IL-8. fied analysis according to age (patients aged 66–74 years versus
High plasma levels of IL-8 were associated with a lower percentage patients over 75 years) did not show statistically significant differ-
of patients achieving CR. Younger age at diagnosis was also associ- ences in the levels of cytokines (data not shown).
ated with CR. We have observed an increase in the levels of IL-8 in Although it has been reported that high serum TNF-a level is an
AML patients under 65 years. IL-8 is a pro-angiogenic mediator re- adverse prognostic factor for survival and event-free survival in pa-
leased by most leukemic blast populations that is involved in leu- tients with AML or high-risk MDS [14], we have not found statisti-
kemogenesis [34]. Previous reports have shown a high percentage cally significant correlations. TNF-a can be produced by a wide
of AML patients with elevated levels of IL-8 [13,35]. Hsu et al. [35] variety of tumor cells including solid tumors and hematological
observed that the level of IL-8 in patients with AML and myelodys- malignances. TNF-a has conflicting roles in cancer, as can act di-
plastic syndromes (MDSs) was high during diagnosis and de- rectly or indirectly as an autocrine tumor growth factor but may
creased to the level of healthy controls when patients were also induce apoptosis of tumor cells [42,43]. We have found that
under chemotherapy or in CR. It is interesting to note that we have mean levels of TNF-a were significantly higher in AML patients
found an age-associated increase in the levels of IL-8 in both than in the healthy volunteers in both age groups. An increase of
healthy donors and AML patients. In elderly AML patients, strati- TNF-a level was also observed in healthy elderly individuals
890 B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891

compared with younger donors as previously described [27,44]. sets suggest that patients with AML, in general, present a deregu-
Kornblau et al. [13] analyzed the expression of different cytokines lation in their immune system [3–5]. Several cytokines are
and chemokines profiles in AML and MDS showing that serum aberrantly produced in some AML patients that can be considered
TNF-a was higher in unfavorable and lower in favorable signatures. as indicators of tumor development and disease progression con-
On the contrary, in other study, the analysis of AML patients at tributing to proliferation, survival and resistance to chemotherapy
diagnosis or during relapsed disease showed that TNF-a was sig- of leukemic cells. Further research leading to identify the molecu-
nificantly elevated in chronic leukemia and acute lymphoblastic lar mechanisms underlying cytokine network deregulation are re-
leukemia but not in AML and MDS [45]. The discrepancies ob- quired and new approaches to modulate cytokine signaling should
served in the different studies are probably due to the existence be evaluated.
of high inter-individual variations as we observed in our study.
We have observed lower plasma levels of TGF-b1 in AML pa-
tients compared to healthy donors. TGF-b1 constitutes an impor- Disclosure statement
tant negative regulatory factor of both normal and neoplastic
hematopoietic progenitor cells [46,47]. Thus, our findings together The authors have nothing to disclose.
with those describing low expression of TGF-b1 high-affinity bind-
ing sites on AML cells suggest that AML cells may escape from the Acknowledgments
regulation by TGF-b1 [46]. IL-10 and TGF-b can be produced by
regulatory T cells, a subset of CD4 T cells that mediates immuno- This work was supported by Grants SAF2009-09711 to RT from
suppression and is increased in AML patients [6,48]. the Ministry of Science and Innovation of Spain, PRI09A029 and
The analysis of plasma levels of cytokines according to age re- Grants to INPATT research group from Junta de Extremadura
vealed some statistically significant differences restricted to the el- (GRU10104) and from University of Extremadura, cofinanced by
derly groups. Thus, IL-12p70, IL-4 and IL-5 levels were significantly European Regional Development Funds (FEDER), and Grants to RS
higher in elderly AML patients compared with healthy elderly indi- PS09/00723 from Spanish Ministry of Health and JA0292/07 from
viduals. IL-12 may exert potent anti-tumor activity through immu- Junta de Andalucia. We thank the technical assistance of J.J.
nostimulatory and antiangiogenic mechanisms [49,50]. Recently, it Gordillo.
has been reported that age is positively correlated with serum lev-
els of IL-12p70 [27]. A dual biological effect has been reported for
IL-4 in tumor immunity, whereas it can exhibit potent anti-tumor References
immunity, numerous reports have shown that IL-4 can also pro-
[1] Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, et al.
mote tumor growth [51]. In relation to IL-5, a previous report Diagnosis and management of acute myeloid leukemia in adults:
showed no statistically significant differences in AML and MDS pa- recommendations from an international expert panel, on behalf of the
tients compared to healthy donors [13]. The role of these cytokines European LeukemiaNet. Blood 2010;115:453–74.
[2] Estey EH. Acute myeloid leukemia: 2012 update on diagnosis, risk
in the prognosis of elderly patients with AML requires further stratification, and management. Am J Hematol 2012;87:89–99.
studies. [3] Fauriat C, Just-Landi S, Mallet F, Arnoulet C, Sainty D, Olive D, et al. Deficient
In our study, no statistically significant differences were ob- expression of NCR in NK cells from acute myeloid leukemia: evolution during
leukemia treatment and impact of leukemia cells in NCRdull phenotype
served in the plasma levels of IL-17A, IL-2 and IFN-c in AML pa- induction. Blood 2007;109:323–30.
tients compared with healthy donors. Our results are consistent [4] Sanchez-Correa B, Morgado S, Gayoso I, Bergua JM, Casado JG, Arcos MJ, et al.
with previous reports [13,52] and differ from those obtained by Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-
activating receptors and their ligands. Cancer Immunol Immunother
Wu et al. [33] that showed higher plasma levels of IL-17 in AML pa- 2011;60:1195–205.
tients compared with healthy donors. The high variability observed [5] Sanchez-Correa B, Gayoso I, Bergua JM, Casado JG, Morgado S, Solana R, et al.
in cytokine levels among AML patients could explain these Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia
patients. Immunol Cell Biol 2012;90:109–15.
discrepancies. [6] Shenghui Z, Yixiang H, Jianbo W, Kang Y, Laixi B, Yan Z, et al. Elevated
In the present study the limited number of patients makes dif- frequencies of CD4(+) CD25(+) CD127lo regulatory T cells is associated to poor
ficult broad generalizations and the potential confounding effect of prognosis in patients with acute myeloid leukemia. Int J Cancer
2011;129:1373–81.
age in the survival analysis cannot be excluded. It is difficult to dis-
[7] Le Dieu R, Taussig DC, Ramsay AG, Mitter R, Miraki-Moud F, Fatah R, et al.
tinguish between age-associated and disease-associated altera- Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis
tions in cytokine levels. Thus, the analysis of cytokines according have abnormal phenotype and genotype and form defective immune synapses
to age in healthy and AML patients showed that IL-6 and IL-8 are with AML blasts. Blood 2009;114:3909–16.
[8] Chan L, Hardwick NR, Guinn BA, Darling D, Gaken J, Galea-Lauri J, et al. An
increased in healthy elderly individuals compared with healthy immune edited tumour versus a tumour edited immune system: prospects for
controls up to 65 years. In AML patients only IL-8 was increased immune therapy of acute myeloid leukaemia. Cancer Immunol Immunother
in elderly patients compared with the younger group of patients 2006;55:1017–24.
[9] Panju AH, Danesh A, Minden MD, Kelvin DJ, Alibhai SM. Associations between
(665 years) supporting that the alterations observed in cytokine quality of life, fatigue, and cytokine levels in patients aged 50+ with acute
levels are associated to AML rather than to age. The results pre- myeloid leukemia. Support Care Cancer 2009;17:539–46.
sented confirm and extend previous findings by other research [10] Panteli KE, Hatzimichael EC, Bouranta PK, Katsaraki A, Seferiadis K, Stebbing J,
et al. Serum interleukin (IL)-1, IL-2, sIL-2Ra, IL-6 and thrombopoietin levels in
groups regarding cytokine profiles in AML patients and survival. patients with chronic myeloproliferative diseases. Brit J Haematol
Further studies are needed to clarify the clinical relevance of these 2005;130:709–15.
findings and the role of disturbed cytokine balance (e.g. IL-6, IL-17 [11] Van Etten RA. Aberrant cytokine signaling in leukemia. Oncogene
2007;26:6738–49.
and IL-10) in disease progression and patient survival. [12] Park HH, Kim M, Lee BH, Lim J, Kim Y, Lee EJ, et al. Intracellular IL-4, IL-10, and
In summary, plasma levels of several cytokines are associated IFN-gamma levels of leukemic cells and bone marrow T cells in acute
with disease progression and survival in AML patients. We have leukemia. Ann Clin Lab Sci 2006;36:7–15.
[13] Kornblau SM, McCue D, Singh N, Chen W, Estrov Z, Coombes KR. Recurrent
observed that CR was associated with age and IL-8 levels. In addi-
expression signatures of cytokines and chemokines are present and are
tion, older age, cytogenetic risk, low levels of IL-10 and high levels independently prognostic in acute myelogenous leukemia and
of IL-6 were found to be correlated with lower survival and EFS. myelodysplasia. Blood 2010;116:4251–61.
AML patients that concomitantly exhibited high levels of IL-6 [14] Tsimberidou AM, Estey E, Wen S, Pierce S, Kantarjian H, Albitar M, et al. The
prognostic significance of cytokine levels in newly diagnosed acute myeloid
and IL-17A had lower survival. The current findings together with leukemia and high-risk myelodysplastic syndromes. Cancer
previous reports showing alterations in different lymphocyte sub- 2008;113:1605–13.
B. Sanchez-Correa et al. / Cytokine 61 (2013) 885–891 891

[15] Moller B, Villiger PM. Inhibition of IL-1, IL-6, and TNF-alpha in immune- [34] Ryningen A, Wergeland L, Glenjen N, Gjertsen BT, Bruserud O. In vitro crosstalk
mediated inflammatory diseases. Springer Semin Immunopathol between fibroblasts and native human acute myelogenous leukemia (AML)
2006;27:391–408. blasts via local cytokine networks results in increased proliferation and
[16] Sugiyama H, Inoue K, Ogawa H, Yamagami T, Soma T, Miyake S, et al. The decreased apoptosis of AML cells as well as increased levels of proangiogenic
expression of IL-6 and its related genes in acute leukemia. Leuk Lymphoma Interleukin 8. Leuk Res 2005;29:185–96.
1996;21:49–52. [35] Hsu HC, Lee YM, Tsai WH, Jiang ML, Ho CH, Ho CK, et al. Circulating levels of
[17] Beauchemin V, Villeneuve L, Rodriguez-Cimadevilla JC, Rajotte D, Kenney JS, thrombopoietic and inflammatory cytokines in patients with acute
Clark SC, et al. Interleukin-6 production by the blast cells of acute myeloblastic myeloblastic leukemia and myelodysplastic syndrome. Oncology
leukemia: regulation by endogenous interleukin-1 and biological implications. 2002;63:64–9.
J Cell Physiol 1991;148:353–61. [36] El-Maksoud NA, Halla MR, El-Latif MM, Abdalla SH. Prognostic Impact of
[18] Apte RN, Dotan S, Elkabets M, White MR, Reich E, Carmi Y, et al. The elevated serum hyaluronic acid, ferritin and interleukin-6 in patients with
involvement of IL-1 in tumorigenesis, tumor invasiveness, metastasis and acute myeloid leukemia. J Am Sci 2010;6:532–41.
tumor-host interactions. Cancer Metastasis Rev 2006;25:387–408. [37] Nielsen AR, Plomgaard P, Krabbe KS, Johansen JS, Pedersen BK. IL-6, but not
[19] Elkabets M, Ribeiro VS, Dinarello CA, Ostrand-Rosenberg S, Di Santo JP, Apte TNF-alpha, increases plasma YKL-40 in human subjects. Cytokine
RN, et al. IL-1beta regulates a novel myeloid-derived suppressor cell subset 2011;55:152–5.
that impairs NK cell development and function. Eur J Immunol [38] Bergmann OJ, Johansen JS, Klausen TW, Mylin AK, Kristensen JS, Kjeldsen E,
2010;40:3347–57. et al. High serum concentration of YKL-40 is associated with short survival in
[20] Kurzrock R. Cytokine deregulation in cancer. Biomed Pharmacother patients with acute myeloid leukemia. Clin Cancer Res 2005;11:8644–52.
2001;55:543–7. [39] Asano Y, Shibata S, Kobayashi S, Okamura S, Niho Y. Interleukin-10 inhibits the
[21] Saily M, Koistinen P, Savolainen ER. The soluble form of interleukin-6 receptor autocrine growth of leukemic blast cells from patients with acute myeloblastic
modulates cell proliferation by acute myeloblastic leukemia blast cells. Ann leukemia. Int J Hematol 1997;66:445–50.
Hematol 1999;78:173–9. [40] Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response
[22] Steensberg A, Fischer CP, Keller C, Moller K, Pedersen BK. IL-6 enhances plasma against cancer: a counterpoint. J Leukoc Biol 2005;78:1043–51.
IL-1ra, IL-10, and cortisol in humans. Am J Physiol Endocrinol Metab [41] Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer
2003;285:E433–7. development and progression. Int J Biol Sci 2011;7:651–8.
[23] You M, Flick LM, Yu D, Feng GS. Modulation of the nuclear factor kappa B [42] Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress,
pathway by Shp-2 tyrosine phosphatase in mediating the induction of inflammation, and cancer: how are they linked? Free Radical Biol Med
interleukin (IL)-6 by IL-1 or tumor necrosis factor. J Exp Med 2010;49:1603–16.
2001;193:101–10. [43] Szlosarek P, Charles KA, Balkwill FR. Tumour necrosis factor-alpha as a tumour
[24] Kimura A, Kishimoto T. IL-6: regulator of Treg/Th17 balance. Eur J Immunol promoter. Eur J Cancer 2006;42:745–50.
2010;40:1830–5. [44] Bruunsgaard H, Pedersen BK. Age-related inflammatory cytokines and disease.
[25] Stenvinkel P, Ketteler M, Johnson RJ, Lindholm B, Pecoits-Filho R, Riella M, et al. Immunol Allergy Clin North Am 2003;23:15–39.
IL-10, IL-6, and TNF-alpha: central factors in the altered cytokine network of [45] Aguayo A, Kantarjian H, Manshouri T, Gidel C, Estey E, Thomas D, et al.
uremia–the good, the bad, and the ugly. Kidney Int 2005;67:1216–33. Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes.
[26] de Gonzalo-Calvo D, de Luxan-Delgado B, Martinez-Camblor P, Rodriguez- Blood 2000;96:2240–5.
Gonzalez S, Garcia-Macia M, Suarez FM, et al. Chronic inflammation as [46] Pasche B. Role of transforming growth factor beta in cancer. J Cell Physiol
predictor of 1-year hospitalization and mortality in elderly population. Eur J 2001;186:153–68.
Clin Invest 2012:10–2362. [47] Hu X, Cui D, Moscinski LC, Zhang X, Maccachero V, Zuckerman KS. TGFbeta
[27] Alvarez-Rodriguez L, Lopez-Hoyos M, Munoz-Cacho P, Martinez-Taboada VM. regulates the expression and activities of G2 checkpoint kinases in human
Aging is associated with circulating cytokine dysregulation. Cell Immunol myeloid leukemia cells. Cytokine 2007;37:155–62.
2012;273:124–32. [48] Szczepanski MJ, Szajnik M, Czystowska M, Mandapathil M, Strauss L, Welsh A,
[28] Larbi A, Franceschi C, Mazzatti D, Solana R, Wikby A, Pawelec G. Aging of the et al. Increased frequency and suppression by regulatory T cells in patients
immune system as a prognostic factor for human longevity. Physiology with acute myelogenous leukemia. Clin Cancer Res 2009;15:3325–32.
(Bethesda) 2008;23:64–74. [49] Colombo MP, Trinchieri G. Interleukin-12 in anti-tumor immunity and
[29] Palmeri M, Misiano G, Malaguarnera M, Forte GI, Vaccarino L, Milano S, et al. immunotherapy. Cytokine Growth Factor Rev 2002;13:155–68.
Cytokine serum profile in a group of sicilian nonagenarians. J Immunoassay [50] Ferretti E, Di CE, Cocco C, Ribatti D, Sorrentino C, Ognio E, et al. Direct
Immunochem 2012;33:82–90. inhibition of human acute myeloid leukemia cell growth by IL-12. Immunol
[30] Candore G, Caruso C, Colonna-Romano G. Inflammation, genetic background Lett 2010;133:99–105.
and longevity. Biogerontology 2010;11:565–73. [51] Li Z, Chen L, Qin Z. Paradoxical roles of IL-4 in tumor immunity. Cell Mol
[31] Naumova E, Ivanova M, Pawelec G. Immunogenetics of ageing. Int J Immunol 2009;6:415–22.
Immunogenet 2011;38:373–81. [52] Wrobel T, Mazur G, Jazwiec B, Kuliczkowski K. Interleukin-17 in acute myeloid
[32] Camporeale A, Poli V. IL-6, IL-17 and STAT3: a holy trinity in auto-immunity? leukemia. J Cell Mol Med 2003;7:472–4.
Front Biosci 2012;17:2306–26.
[33] Wu C, Wang S, Wang F, Chen Q, Peng S, Zhang Y, et al. Increased frequencies of
T helper type 17 cells in the peripheral blood of patients with acute myeloid
leukaemia. Clin Exp Immunol 2009;158:199–204.

You might also like