The TNF receptor family member OX40 promotes activation and proliferation of T cells, which fuels efforts to modulate this immune checkpoint to reinforce antitumor immunity. Besides T cells, NK cells are a second cytotoxic lymphocyte subset that contributes to antitumor immunity, particularly in leukemia. Accordingly, these cells are being clinically evaluated for cancer treatment through multiple approaches, such as adoptive transfer of ex vivo expanded polyclonal NK cells (pNKC). Here, we analyzed whether and how OX40 and its ligand (OX40L) influence NK-cell function and antileukemia reactivity. We report that OX40 is expressed on leukemic blasts in a substantial percentage of patients with acute myeloid leukemia (AML) and that OX40 can, after stimulation with agonistic OX40 antibodies, mediate proliferation and release of cytokines that act as growth and survival factors for the leukemic cells. We also demonstrate that pNKC differentially express OX40L, depending on the protocol used for their generation. OX40L signaling promoted NK-cell activation, cytokine production, and cytotoxicity, and disruption of OX40–OX40L interaction impaired pNKC reactivity against primary AML cells. Together, our data implicate OX40/OX40L in disease pathophysiology of AML and in NK-cell immunosurveillance. Our findings indicate that effects of the OX40–OX40L receptor–ligand system in other immune cell subsets and also malignant cells should be taken into account when developing OX40-targeted approaches for cancer immunotherapy. Cancer Immunol Res; 6(2); 209–21. ©2018 AACR.

Modulation of immune checkpoints has become a mainstay in oncological treatment. Besides already approved approaches that block inhibitory molecules like CTLA-4 or PD-1, agonistic antibodies that trigger activating receptors on T cells are being developed. One such receptor is OX40 (1–4). This member of the TNF receptor (TNFR) superfamily is upregulated on effector T cells after activation and promotes their differentiation, proliferation, expansion, and long-term survival while inhibiting the suppressive activity of regulatory T cells (5, 6). In patients with cancer, the frequency of tumor-infiltrating OX40+ T cells correlates with survival. Moreover, application of OX40 agonists, alone or in combination with other checkpoint modulators, stimulates the cytolytic activity of T cells and causes tumor regression in preclinical models (7–11). First evidence from early clinical trials, most of which are in progress, indicates that OX40 stimulation is also effective in patients with cancer (3, 12).

Besides T cells, which are a component of adaptive immunity, NK cells, as the major cytotoxic lymphocyte subset of the innate immune system, also play an important role in tumor immunosurveillance, particularly in hematologic malignancies. This is supported by observations that NK-cell counts and activity are reduced in patients with leukemia and that activity levels of autologous NK cells are associated with survival of patients with leukemia (13–15). The role of NK cells in acute myeloid leukemia (AML) is highlighted by studies on haploidentical stem cell transplantation (SCT), in which the resulting KIR mismatch seems to be associated with a pronounced graft-versus-leukemia reaction and improved clinical outcome (16). Besides their role in SCT, multiple approaches presently aim to utilize adoptive transfer of allogeneic/KIR-mismatched NK cells for cancer treatment (17).

Beyond KIR, signals mediated by multiple other activating and inhibitory receptors determine whether NK cells do or do not respond to tumor cells. This comprises various members of the TNF/TNFR family which influence NK-cell reactivity upon interaction with their counterparts expressed, e.g., on leukemic cells (18, 19). OX40L is upregulated on NK cells following activation (20), and its counterpart OX40 is expressed by T-cell–derived leukemia cells (21). However, the influence of the OX40/OX40L system on NK function or its role in AML is so far unknown. Here, we show that AML cells express OX40, and exposure to an agonistic OX40 antibody (mAb) promotes leukemia cell proliferation and release of cytokines that influence growth and survival of the malignant cells (22, 23). We found that OX40L is differentially expressed on polyclonal NK cells (pNKC) generated for adoptive transfer depending on the particular protocol utilized, and that OX40L (reverse) signaling alters NK-cell function, including their reactivity to AML cells.

Cells

Peripheral blood mononuclear cells (PBMC) and bone marrow (BM) cells of patients and healthy donors were isolated by density gradient centrifugation after informed consent in accordance with the Helsinki protocol. The study was conducted according to the guidelines of the local ethics committee.

Isolation of highly pure (purity >95%) NK cells from pNKC and AML cells from patient PBMC was performed by immunomagnetic separation using the NK-cell isolation kit and negative selection using microbeads CD3, CD14, CD19, and CD56 from Miltenyi Biotec according to the manufacturer's instructions.

pNKC were generated according to standard protocols by incubating nonplastic-adherent PBMC with irradiated RPMI8866 (pNKC-8866) or K562-mb15-41BBL (pNKC-SJ) feeder cells obtained from DSMZ and St. Jude's Children's Research Hospital as previously described (24, 25). Functional experiments were performed when purity of NK cells (CD56+CD3) was above 90% as determined by flow cytometry. In addition, K562 cells (received from DSMZ) were transfected using the vector pcDNA3 containing the open reading frame of human 4-1BBL (K562-4-1BBL) or empty vector as control (K562-mock) as described previously (25).

U937 cells (obtained from DSMZ) were transfected using the vector pcDNA3 containing the open-reading frame of human OX40 (U937-OX40) or empty vector as control (mock) and cultured as described previously (25).

The OX40:Fas reporter cells (Jurkat-JOM2) and their use in cytotoxic assays were previously described (26).

RPMI8866, K562-mb15-41BBL, K562,U937, and NK-92 (DSMZ) were obtained in 2005, 2004, 2003, and 2004, respectively. Authenticity was routinely determined by validating the respective immunophenotype described by the provider using flow cytometry after thawing, and cell lines were cultured for a maximum of 2 months prior to use in experiments. Contamination with mycoplasma was excluded by routine testing of all cultures every 3 months.

Reagents

OX40 mAb BerAct35, OX40L mAb ANC10G1, and mouse IgG1 isotype control were from Ancell Corporation and BD Biosciences, respectively. OX40L mAb lk-1 and 11C3.1 were from BD Biosciences and Biolegend, respectively. All fluorescence conjugates were from BD Biosciences, secondary goat anti–mouse-PE was from Dako (BIOZOL). Fusion proteins consisting of human OX40 with a murine (OX40–Fc) and human (OX40–huFc) Fc-part were from R&D Systems, and Ancell, respectively. RhIL2 was from ImmunoTools.

In addition, antibodies against human OX40 were raised by immunization of C57BL/6 mice by repeated injection of 20 × 106 OX40-transfected CHO cells. Then, spleen cells were fused with SP2/0-Ag14 cells, and hybridoma cells secreting OX40 mAbs were cloned by limiting dilution. mAbs were purified from hybridoma supernatants using Protein A Agarose columns (GE Healthcare). F(ab′)2 fragments were generated using previously described standard protocols (27).

Flow cytometry

Analysis of OX40 and OX40L surface expression was performed using specific mAb or isotype control followed by anti–mouse-PE using a BD FACSCanto II.

Leukemic cells in patient samples were selected by FSC/SSC and employing the surface markers CD33/CD34/CD14/CD117 based on the individual immunophenotype defined upon routine diagnosis. Specific fluorescence indices (SFI) were calculated by dividing median fluorescences obtained with specific mAb by median fluorescences obtained with isotype control. Expression was considered positive in case of SFI ≥ 1.5. Intracellular staining was performed using the Fixation/Permeabilization Solution Kit with BD GolgiStop from BD Biosciences according to manufacturer's instructions.

PCR analysis

OX40 primers were 5′-TGTAACCTCAGAAGTGGGAGTG-3′ and 5′-GGTCCCTGTCCTCACAGATTG-3′. 18S rRNA primers were 5′-CGGCTACCACATCCAAGGAA-3′ and 5′-GCTGGAATTACCGCGGCT-3′. OX40L primers were 5′-CTGCTCCTGTGCTTCACCTAC-3′ and 5′-TCCAGGGAGGTATTGTCAGTG-3′. GAPDH primers were 5′-AGCCACATCGCTCAGACAC-3′ and 5′-GCCCAATACGACCAAATCC-3′. Reverse transcriptase–polymerase chain reaction (RT-PCR) was performed as described previously (18).

For quantitative PCR, total RNA was isolated using the High Pure RNA Isolation Kit and transcribed into cDNA using qScript XLT cDNA SuperMix (Quanta Biosciences) according to the manufacturer's instructions. Amplification of OX40 cDNA was performed using PerfeCTa SYBR Green FastMix (Quanta Biosciences) on a LightCycler 480 instrument. Primer assays (QuantiTect Primer Assay) for OX40 and 18S ribosomal RNA were used according to the manufacturer's instructions. Relative mRNA expression was calculated by the ΔΔ cycle-threshold (Ct) method.

Cytotoxicity assays

Cytotoxicity of NK cells against primary leukemia cells and U937-transfectants was determined by 51chromium release assays after 4 or 24 hours as previously described (28).

Measurement of cytokines and metabolic activity

Cytokine determination was performed by ELISA according to the manufacturer's instructions using OptEIA sets from BD Pharmingen or DuoSet ELISA development systems from R&D Systems. Metabolic activity was measured using the cell proliferation reagent WST-1 set (Roche) according to the manufacturer's instructions.

OX40L is differentially expressed on pNKC

As OX40L can be upregulated on NK cells upon activation and ex vivo-preactivated pNKC are being evaluated for cancer treatment (17, 20), we characterized OX40L expression in pNKC generated according to two differing standard protocols [pNKC-8866 (25) and pNKC-SJ (24)]. RT-PCR revealed similar OX40L mRNA levels in both pNKC preparations (Fig. 1A). Next, we used commercially available OX40L mAb to study surface expression using NK-92 cells, which do not express OX40L mRNA (Fig. 1A), as negative control and OX40–Fc fusion protein to ascertain specificity. Comparative FACS analyses revealed that only mAb ANC10G1 can be used to determine OX40L expression. The other two mAbs (but not OX40–Fc) unspecifically bound to NK-92 cells (Fig. 1B). Although resting NK cells of healthy donors were never found positive for surface OX40L and pNKC-8866 displayed only low or no relevant levels, pNKC-SJ displayed substantial and significantly higher OX40L surface expression. This difference between pNKC-8866 and pNKC-SJ cells held true for preparations generated with PBMC of the same or independent donors despite the considerable variation among individual donors (Fig. 1C; P < 0.05, Mann–Whitney test). Expression of OX40L in pNKC-SJ peaked after 4 to 6 days of culture and declined thereafter, but OX40L still was profoundly expressed on day 8 and later, the time these cells usually are used for functional experiments and clinical application by us and others (29). We occasionally observed upregulation of OX40L early during culture of pNKC-8866, but expression was always lower than in pNKC-SJ and OX40L was never detectable after day 9 (Fig. 1D). In line with previous findings (20), activation of NK cells with IL2 alone was not sufficient for induction of OX40L. Upregulation of OX40L expression was dependent on 4-1BB stimulation as revealed by coculture experiments involving K562-41BBL and mock transfectants and transwell settings (Fig. 1E).

Figure 1.

OX40L expression on human NK cells. pNKC were generated using standard protocols (comprising medium containing 25 U/mL IL2) with either RPMI 8866 (pNKC-8866) or K562-mb15-41BBL (pNKC-SJ) as feeder cells. NK cells within PBMC of healthy donors (HD-NKC, selected by counterstaining for CD56+CD3) and NK-92 cells served as controls. A, OX40L mRNA expression in pNKC-8866 and pNKC-SJ generated with PBMC of three different donors and NK-92 cells was determined on day 8 by RT-PCR of equal mRNA levels with 18S rRNA or GAPDH serving as control. B, OX40L surface expression was analyzed on day 8 by FACS using OX40–Fc or the anti-OX40L clones ANC10G1, ik-1, 11C3.1 (shaded peaks) and their respective controls (open peaks) followed, in case of unlabeled antibodies, by secondary PE-conjugates. C, SFI levels of OX40L surface expression as obtained by analysis of pNKC-SJ, pNKC-8866, and HD-NKC (obtained from seven independent healthy donors each) on day 7 or 8 using mAb ANC10G1 as described above. D, SFI levels of OX40L surface expression on NK cells were determined at the indicated time points of coculture of PBMC with RPMI8866 or K562-mb15-41BBL as described in C. E, To unravel the molecular mechanism involved in OX40L upregulation, PBMC of healthy donors were cultured and analyzed by FACS as described above on day 7 after culture with K562-mb15-41BBL [separated by a transwell insert (TW) to prevent cell contact where indicated], K562-mock or K562-4-1BBL as indicated. Data of one representative experiment of a total of three with similar results are shown. *, Statistically significant differences, P < 0.05.

Figure 1.

OX40L expression on human NK cells. pNKC were generated using standard protocols (comprising medium containing 25 U/mL IL2) with either RPMI 8866 (pNKC-8866) or K562-mb15-41BBL (pNKC-SJ) as feeder cells. NK cells within PBMC of healthy donors (HD-NKC, selected by counterstaining for CD56+CD3) and NK-92 cells served as controls. A, OX40L mRNA expression in pNKC-8866 and pNKC-SJ generated with PBMC of three different donors and NK-92 cells was determined on day 8 by RT-PCR of equal mRNA levels with 18S rRNA or GAPDH serving as control. B, OX40L surface expression was analyzed on day 8 by FACS using OX40–Fc or the anti-OX40L clones ANC10G1, ik-1, 11C3.1 (shaded peaks) and their respective controls (open peaks) followed, in case of unlabeled antibodies, by secondary PE-conjugates. C, SFI levels of OX40L surface expression as obtained by analysis of pNKC-SJ, pNKC-8866, and HD-NKC (obtained from seven independent healthy donors each) on day 7 or 8 using mAb ANC10G1 as described above. D, SFI levels of OX40L surface expression on NK cells were determined at the indicated time points of coculture of PBMC with RPMI8866 or K562-mb15-41BBL as described in C. E, To unravel the molecular mechanism involved in OX40L upregulation, PBMC of healthy donors were cultured and analyzed by FACS as described above on day 7 after culture with K562-mb15-41BBL [separated by a transwell insert (TW) to prevent cell contact where indicated], K562-mock or K562-4-1BBL as indicated. Data of one representative experiment of a total of three with similar results are shown. *, Statistically significant differences, P < 0.05.

Close modal

Signaling via OX40L modulates NK-cell reactivity

In addition to acting as a ligand for OX40, OX40L can, like other TNF family members, itself transduce signals into the ligand-bearing cell (30). To determine whether and how such reverse signaling via OX40L affects NK cells, we cultured pNKC on immobilized OX40–Fc or Fc-control to allow for OX40L crosslinking in the absence of a second, OX40-expressing (target) cell population. FACS analysis of the activation markers CD69 and NKp44 revealed a significant (both P < 0.05; Mann–Whitney test) upregulation on pNKC-SJ following OX40L signaling, whereas the OX40L pNKC-8866 were not affected (Fig. 2A). In addition, triggering OX40L also led to a significant (P < 0.001; Mann–Whitney test) induction of IFNγ release by OX40L+ but not OX40L pNKC, which again confirmed that signals were mediated via OX40L. The effect of OX40L signaling was observed both in the absence and presence of IL2, the latter serving to mimic an augmented state, which indicates that OX40L signaling may further enhance the activity of activated NK cells (Fig. 2B).

Figure 2.

OX40L stimulates reactivity of human NK cells. A and B, OX40L pNKC-8866 and OX40L+ pNKC-SJ were cultured alone, on immobilized OX40–Fc or isotype control for 24 hours. Where indicated, 25 U/mL IL2 was added during culture. Exemplary results (left) and combined data of at least 6 experiments with pNKC-SJ of independent donors after setting results with pNKC alone to 1 for normalization to account for donor variability (right) are shown. Horizontal bars, the mean of the results within each culture condition. A, Expression of CD69 and NKp44 was analyzed by FACS of CD56+CD3 pNKC-8866 (top two panels) or pNKC-SJ (bottom two panels) using specific fluorescence conjugates and isotype control. B, IFNγ levels in culture supernatants were determined by ELISA. C, Expression of OX40 on transfectants (U937-OX40) and mock controls (U937-mock) was ascertained by FACS (left; shaded peaks, OX40 mAb; open peaks, isotype control) prior to use in 4-hour 51chromium release assays with pNKC-SJ (right). Results of one experiment out of five with similar results are shown. D, Top, U937-OX40 and U937-mock transfectants were analyzed by FACS using the OX40-mAbs M-OX17 and M-OX2 followed by secondary PE-conjugate. Shaded peaks, specific mAb; open peaks, isotype control. Bottom, The OX40 mAbs M-OX17 and M-OX2 or isotype control (10 μg/mL each) were preincubated with OX40–huFc or huFc-control (both at 2 μg/mL) for 1 hour. Then, pNKC-SJ cells were incubated with pretreated OX40–huFc/huFc-control followed by anti-human PE-conjugate and FACS analysis. Cross-competition and thus blocking properties of the antibodies were identified by the reduction of OX40–huFc binding to pNKC-SJ; dark grey peaks, OX40–huFc preincubated with isotype control; light grey peaks, OX40–huFc preincubated with the OX40mAb; dashed line, huFc-control. E, U937-mock (left) or U937-OX40 (right) cells were incubated with pNKC-SJ in the presence or absence of blocking M-OX2 F(ab)2-fragments or isotype control (both at 2 μg/mL) and cytotoxicity was evaluated by 4-hour 51chromium release assays. Data represent means of triplicates with SD, and one representative experiment of a total of at least 3 with similar results is shown. *, Statistically significant differences; P < 0.05.

Figure 2.

OX40L stimulates reactivity of human NK cells. A and B, OX40L pNKC-8866 and OX40L+ pNKC-SJ were cultured alone, on immobilized OX40–Fc or isotype control for 24 hours. Where indicated, 25 U/mL IL2 was added during culture. Exemplary results (left) and combined data of at least 6 experiments with pNKC-SJ of independent donors after setting results with pNKC alone to 1 for normalization to account for donor variability (right) are shown. Horizontal bars, the mean of the results within each culture condition. A, Expression of CD69 and NKp44 was analyzed by FACS of CD56+CD3 pNKC-8866 (top two panels) or pNKC-SJ (bottom two panels) using specific fluorescence conjugates and isotype control. B, IFNγ levels in culture supernatants were determined by ELISA. C, Expression of OX40 on transfectants (U937-OX40) and mock controls (U937-mock) was ascertained by FACS (left; shaded peaks, OX40 mAb; open peaks, isotype control) prior to use in 4-hour 51chromium release assays with pNKC-SJ (right). Results of one experiment out of five with similar results are shown. D, Top, U937-OX40 and U937-mock transfectants were analyzed by FACS using the OX40-mAbs M-OX17 and M-OX2 followed by secondary PE-conjugate. Shaded peaks, specific mAb; open peaks, isotype control. Bottom, The OX40 mAbs M-OX17 and M-OX2 or isotype control (10 μg/mL each) were preincubated with OX40–huFc or huFc-control (both at 2 μg/mL) for 1 hour. Then, pNKC-SJ cells were incubated with pretreated OX40–huFc/huFc-control followed by anti-human PE-conjugate and FACS analysis. Cross-competition and thus blocking properties of the antibodies were identified by the reduction of OX40–huFc binding to pNKC-SJ; dark grey peaks, OX40–huFc preincubated with isotype control; light grey peaks, OX40–huFc preincubated with the OX40mAb; dashed line, huFc-control. E, U937-mock (left) or U937-OX40 (right) cells were incubated with pNKC-SJ in the presence or absence of blocking M-OX2 F(ab)2-fragments or isotype control (both at 2 μg/mL) and cytotoxicity was evaluated by 4-hour 51chromium release assays. Data represent means of triplicates with SD, and one representative experiment of a total of at least 3 with similar results is shown. *, Statistically significant differences; P < 0.05.

Close modal

To determine whether OX40L also affects NK lysis, we transfected U937 cells to express high levels of OX40 (U937-OX40) and generated mock-transfectants (U937-mock) as control. When the transfectants were used in cytotoxicity assays, we observed significantly (P < 0.05, Student t test) higher lysis rates for the OX40+ targets (Fig. 2C). Next, we disrupted receptor–ligand engagement in this experimental setting to confirm that OX40–OX40L interaction enhances NK lysis of target cells. We used for this purpose a blocking OX40 mAb (instead of using OX40L mAb) to avoid binding to and induction of signaling in NK cells. In addition, to exclude Fc-mediated effects/ADCC after mAb binding to OX40 on target cells, we used F(ab′)2-fragments. As no OX40 mAb with distinct blocking capacity was commercially available, we generated mouse OX40 mAb as described in the methods section and, after production and definition of specificity, used these in cross-competition experiments using OX40–huFc. Both of our mAb clones M-OX2 and M-OX17 bound to the U937-OX40 transfectants, but only M-OX2 disrupted OX40–OX40L interaction, as revealed by reduced binding of OX40–huFc to OX40L on pNKC-SJ (Fig. 2D). We then produced F(ab′)2-fragments of M-OX2 according to standard protocols (27) and used these in cytotoxicity assays with pNKC-SJ and U937-transfectants. Although no effect on NK lysis of mock-transfectants was observed, blocking OX40 significantly (P < 0.05, Student t test) decreased the per se higher cytotoxicity observed with the OX40-transfectants, which confirmed the stimulatory effect of OX40–OX40L interaction on NK-cell reactivity (Fig. 2E).

Expression of OX40 on AML cells

We used FACS analysis to study whether OX40 is expressed on the surface of leukemic cells. We used a total of 111 different AML patient samples and also CD34+ progenitor cells obtained from peripheral blood and BM of healthy donors. Leukemic blasts within PBMC were selected as described in the Materials and Methods section. The clinical characteristics of each patient and individual SFI levels are given in Table 1. Although no surface expression was observed on healthy CD34+ cells, their malignant counterparts displayed relevant OX40 expression in a substantial proportion of AML cases [SFI ≥ 1.5, n = 60 (54%); SFI ≥ 2.0, n = 41 (37%); Fig. 3A and B]. CD34+ cells from patients with chronic myeloid leukemia (CML, n = 10) and myelodysplastic syndrome (MDS, n = 6) showed no relevant OX40 expression (Supplementary Fig. S1). In AML, OX40 expression was significantly associated with the t(15;17) translocation (PML/RARA) and FLT3-ITD mutation (both P < 0.05, Mann–Whitney-U test), whereas no association with other genetic abnormalities, risk according to the ELN classification, FAB classification or disease etiology (i.e., secondary AML from MDS) was observed (Table 2). OX40 expression by AML cells was also confirmed on mRNA level using RT-PCR. Amplicons of OX40 were detected in all 8 investigated samples of patients with at least 80% blast count (Fig. 3C). This also comprised samples of three patients without detectable surface expression on leukemic cells. Very low or no OX40 mRNA was detected in CD34-enriched BM cells of healthy donors (Fig. 3D). Quantitative PCR revealed significantly (P < 0.05, Mann–Whitney test) lower mRNA levels in healthy CD34+ BM-cell samples and surface-negative compared with surface-positive AML samples (Fig. 3E). Next, we cultured primary AML cells in the presence of OX40L, G-CSF, GM-CSF, IFNγ, TNF, IL6, IL8, and IL10 and then analyzed OX40 expression by FACS using an (noncompeting) OX40 mAb. We found that TNF significantly induced OX40 expression, whereas other factors had no effect (Fig. 3F).

Table 1.

Patient characteristics and levels of cell-surface OX40

UPNOX40FABAgeSexPBB (%) Diff.KaryoWBC (G/L)Hb (g/dL)Plt (G/L)ELNNCCNMRCEtiology
1.4 M0 78 84 45,XY, −7,+13,-21 [28], 46,XY [2] 85 6.3 20 Adverse Poor Poor pAML 
M0 46 97 46, XY 60.5 39 Favorable Favorable Better pAML 
3.5 M0 49 83 46, XY, t(1;20)(p32;p13),del(12)(p12p13)[20] 29.2 12.9 39 Intermediate II Intermediate Intermediate sAML 
2.4 M1 50 93 46, XX 270 18 Intermediate I Intermediate Poor pAML 
5.1 M1 40 100 complex 81.3 10.8 51 Adverse Poor Poor pAML 
1.4 M1 27 95 46, XY 135.1 8.1 20 Favorable Favorable Better pAML 
1.3 M1 58 93 46,XY 23.9 8.2 42 Intermediate I Intermediate Intermediate n.a. 
M1 83 88 n.d. 128.9 7.5 15 n.a. n.a. n.a. pAML 
2.6 M1 67 78 complex 11.1 8.9 48 Adverse Poor Poor pAML 
10 2.3 M1 76 76 n.d. 15.6 10.7 46 n.a. n.a. n.a. pAML 
11 1.3 M1 76 64 Complex 12180 7,3 134 Adverse Poor Poor tAML 
12 1.1 M1 61 63 46,XY[25] 59.4 13 69 Intermediate I Intermediate Intermediate tAML 
13 1.5 M1 68 86 46,XY 84.1 6.7 332 Intermediate I Poor Poor pAML 
14 1.5 M1 75 84 46, XX 150 10.8 174 Favorable Favorable Better pAML 
15 3.8 M1 80 93 n.d. 34.9 11.2 28 n.a. n.a. n.a. pAML 
16 1.5 M1 72 80 46,XX [20] 25.8 7.8 50.0 Intermediate I Intermediate Intermediate pAML 
17 2.8 M1 64 94 n.d. 222 9.2 44 n.a. n.a. Poor pAML 
18 1.9 M1 63 60 46,XX, del(9)(q22) [5]/46,XX [25] 11.3 11.3 292 Favorable Intermediate Intermediate pAML 
19 1.4 M1 50 98 46,XY 19.7 10.4 55 Favorable Favorable Better pAML 
20 1.4 M1 67 99 n.d. 67.7 9.8 23 n.a. n.a. n.a. n.a. 
21 1.5 M1 62 77 46,XY 45 7.1 270 Intermediate I Poor Poor pAML 
22 1.1 M1 50 89 46,XX, del(9)(q13,q22)[4]/46,XX [16] 13.8 8.8 25 Intemediate II Intermediate Intermediate pAML 
23 M1 44 75 46, XY 10.7 7.9 234 Intemediate I Poor Poor pAML 
24 2.2 M1 82 81 46, XX 130 10.1 127 Intermediate I Intermediate Intermediate pAML 
25 2.7 M1 47 98 n.d. 56.3 9.4 60 n.a. n.a. n.a. pAML 
26 0.9 M1 76 81 46,XX[20], 10.7 7.4 145 Favorable Favorable Better pAML 
27 6.9 M1 47 91 47,XX, +8 27.6 10.0 26 Intermediate II Intermediate Intermediate sAML 
28 1.1 M1 62 89 n.a. 18.3 12.3 18 n.a. n.a. Poor pAML 
29 1.4 M1 52 98 46,XY 153.3 9.2 23 Intermediate I Poor Poor pAML 
30 3.8 M1 56 87 46, X, t(X;12)(p11;p13)[14];46, XX [7] 4.14 8.3 111 Intermediate II Intermediate Intermediate pAML 
31 0.9 M1 68 93 n.d. 42.5 9.8 80 n.a. n.a. n.a. pAML 
32 1.7 M1 21 95 46,XX [16], 46,XX, del(9)(q13q22) [4] 84 7.1 30 Intermediate II Intermediate Poor pAML 
33 4.5 M1 41 92 46,XX 68.7 8.1 55 Intermediate I Poor Poor pAML 
34 1.1 M1 34 75 46, XY 33.3 10.2 11 Favorable Favorable Better pAML 
35 1.1 M1 38 60 46, XX 26 9.6 153 Intermediate I Poor Poor pAML 
36 1.2 M2 84 79 n.d. 115.2 7.8 107 n.a. n.a. n.a. pAML 
37 2.3 M2 30 53 46, XY 24.5 7.4 54 Intermediate I Poor Poor pAML 
38 1.1 M2 54 71 46,XX,t(8;21)(q22;q22)[21] 8.9 40 Favorable Favorable Better tAML 
39 1.2 M2 38 28 46, XY 8.8 8.7 52 Intermediate I Intermediate Intermediate sAML 
40 1.1 M2 72 97 Complex 56.9 8.9 30 Adverse Poor Poor sAML 
41 1.4 M2 56 63 46, XY 8.8 9.7 311 Favorable Favorable Better pAML 
42 1.2 M2 81 96 n.d. 10 10 46 n.a. n.a. n.a. sAML 
43 1.2 M2 66 80 n.d. 27.8 10.1 49 Favorable Better Better pAML 
44 2.3 M2 29 70 46,XY 9.6 8.5 17 Intermediate I Poor Poor tAML 
45 1.3 M2 73 83 46,X,inv(X)(p22.3q13) [14]; 46,XX [6] 6.3 95 Intermediate II Intermediate Intermediate pAML 
46 1.8 M2 73 83 46,XY 33.8 8.1 80 Intermediate I Poor Poor pAML 
47 1.4 M2 68 96 46,XY 85.5 9.5 146 Intermediate II Intermediate Intermediate pAML 
48 1.6 M2 72 81 46, XY, del(17)(q11q21) [6]/47,XY,+8 [3] 46, XY [16] 78.9 6,4 47 Intermediate II Intermediate  pAML 
49 1.4 M2 51 43 46,XX,t(2;2)(q21;q23)[21] 35.3 9.3 51 Intermediate II Intermediate Better pAML 
50 1.2 M2 48 87 46,XY 36.9 7.1 66 Favorable Favorable Better pAML 
51 1.8 M2 63 78 46,XY 180 30 Favorable Favorable Better pAML 
52 1.5 M2 64 82 47, XY+8[16]/47,XY,+8,del(9)(q22)[1]/46,XY,del(9)(q22)[1]/46,XY[7] 338.5 8.1 19 Intermediate II Intermediate Intermediate pAML 
53 0.4 M2 58 92 46,XX 62 7.4 25 Intermediate I Poor Poor sAML 
54 1.1 M2 78 99 45, XY, −7[7], 46, XY[15] 26 9.7 36 Adverse Poor  sAML 
55 6.4 M2 68 84 47,XX,+11[23] 165.5 3.8 222 Intermediate II Intermediate Poor pAML 
56 1.6 M2 71 94 46, XX 105.4 10.4 83 Intermediate I Poor Poor pAML 
57 1.7 M2 58 20 Complex 39.1 9.3 23 Adverse Poor Poor pAML 
58 1.3 M2 23 69 47,XX, +21 [2]/46,XX [23] 50 9.9 135 Intermediate II Intermediate Intermediate pAML 
59 1.1 M2 71 81 47, XX +11 26.1 9.4 10 Intermediate II Intermediate Intermediate sAML 
60 1.3 M2 45 80 Complex 17.1 9.0 51 Favorable Favorable Better pAML 
61 2.5 M2 44 94 46,XY 138.5 10.3 57 Intermediate I Poor Poor pAML 
62 1.1 M2 45 94 47, XY, +8 44.4 8.7 11 Intermediate II Intermediate Poor pAML 
63 2.2 M2 45 94 Complex 44.4 8.7 11 Adverse Poor Poor pAML 
64 1.3 M2 68 93 47, XY, +11 110.9 27 Intermediate II Intermediate Intermediate pAML 
65 0.9 M2 70 20 47,XX,+8,del(17)(p11-12)[5]/46,XX[20] 13,6 182 Adverse Poor Poor sAML 
66 5.7 M3 46 51 Complex 23.1 13.3 25 n.a. n.a. n.a. pAML 
67 1.1 M3 67 94 46,XX 99 9.9 128 Intermediate II Intermediate Intermediate sAML 
68 14.3 M3 65 40 46, XY, t(15;17)(q22;q12)[20] 6.9 8.5 17 Favorable Favorable Better pAML 
69 5.2 M3 58 96 46,XX,t(15;17)(q22;q12)[20]. 42.1 8.4 17 Favorable Favorable Better pAML 
70 M4 70 71 45, XX, -7 282.3 5.7 68 Adverse Poor Poor sAML 
71 1.8 M4 49 72 46,Y,t(X;17)(p11;p1?1),add(21)(q22)[13] 61.9 5.8 18 Adverse Poor Poor pAML 
72 2.9 M4 71 93 47, XY, + 11 (6) 79.7 9.9 40 Intermediate II Intermediate Intermediate pAML 
73 1.4 M4 55 72 46,XY 17.2 8.6 162 Favorable Favorable Better pAML 
74 9.7 M4 61 70 46, XY 85.4 8.5 12 Intermediate I Poor Poor pAML 
75 6.1 M4 76 94 46, XX, t (9;11) (q22; q23) [12]/52, XXX, +3, +6, +8 t (t;11) (q22; q23) +12, +13, +18 [13] 141 12 70 Adverse Poor Poor tAML 
76 7.3 M4 43 86 46, XX 63.5 9.2 178 Favorable Favorable Better pAML 
77 1.3 M4 41 73 Complex 112.7 8.5 30 Adverse Poor Poor pAML 
78 M4 63 96 46,XY 92.3 10.5 431 Intermediate I Poor Poor pAML 
79 2.2 M4 73 85 46,XX (25) 52.6 7.5 48 Intermediate I Poor Poor tAML 
80 1.2 M4 71 75 46, XY,del(20)(q11)[6];47,idem,+11[14]; 161.1 8.6 61 Intermediate II Intermediate Intermediate sAML 
81 1.6 M4 83 95 46,XX add(14)(p11)[8],/46, XX[5] 155.8 11.6 144 Intermediate II Intermediate Intermediate pAML 
82 2.3 M4 78 39 51,XX,+6,+9,+9,+11,+13[20] 8.8 9.2 146 Adverse Poor Poor pAML 
83 2.7 M4 64 22 46,XY 62.4 7.5 40 Intermediate I Poor Poor n.a. 
84 1.5 M4 36 78 46, XY 207.4 6.1 55 Intermediate I Poor Poor pAML 
85 M4 68 82 46, XY 148.7 9.1 134 Intermediate I Poor Poor pAML 
86 4.4 M4 67 75 46,XY,?dup(9)(p13p22)c 64.4 9.1 33 Intermediate II Intermediate Intermediate pAML 
87 0.6 M4 72 81 46,XX 26.1 8.2 70 Intermediate I Intermediate Interemdiate pAML 
88 M4 54 46 46, XX 17.2 10.6 167 Intermediate I Poor Poor pAML 
89 M4 57 68 n.d. 334 9.4 293 n.a. n.a. n.a. pAML 
90 2.2 M4 57 29 46,XX 17.5 8.0 117 Intermediate I Intermediate Intermediate pAML 
91 3.3 M4 56 64 46, XX, inv(16)(p13.1q22)[11] 56.6 10.9 99 Favorable Favorable Better pAML 
92 0.8 M5 70 90 46,XY 190 7.1 65 Intermediate I Intermediate Intermediate pAML 
93 1.6 M5 70 46, XY 73.6 13.2 25 Favorable Favorable Better pAML 
94 1.6 M5 26 18 46, XX 51 6,2 17 Favorable Favorable Better pAML 
95 7.3 M5 73 97 47, XY,+8[2]; 46,XY[23] 78.9 6.4 47 Intermediate II Intermediate Poor tAML 
96 28.5 M5 69 80 n.d. 275 7.1 47 n.a. n.a. Poor pAML 
97 1.3 M5 65 91 n.d. 394.2 7.9 189 n.a. n.a. n.a. sAML 
98 1.7 M5 69 11 n.d. 22.1 8.7 53 n.a. n.a. n.a. pAML 
99 1.1 M5 24 90 n.d. n.d. n.d. n.d. n.a. n.a. n.a. pAML 
100 1.3 M5 78 49 Complex 11.7 13 Adverse Poor Poor pAML 
101 1.3 M5 74 >90 46,XY 239 5.7 122 Intermediate I Poor Poor pAML 
102 9.5 M5 52 46, XX 15.7 7.2 21 Favorable Favorable Better pAML 
103 1.2 M5 54 89 46, XY, del(9)(q13q22)[8]/48, XY[16] 109.4 8.2 78 Intermediate II Intermediate Intermediate pAML 
104 4.6 M5 72 40 46,XY 14.5 5.3 28 Intermediate I Intermediate Intermediate sAML 
105 31.6 M5 82 35 n.d. 61.3 11.7 110 n.a. n.a. n.a. pAML 
106 1.5 M5 23 83 48,XY,+8,+13[17]/46,XY[3] 145.1 7.6 34 Intermediate II Intermediate Poor pAML 
107 1.3 M5 58 70 46,XX 0.6 9.1 42 Intermediate I Poor Poor pAML 
108 0.7 M5 45 80 46,XX 248.1 7.3 46 Favorable Favorable Better pAML 
109 18.7 M5 53 85 46,XY 105.6 8.1 35 Intermediate I Poor Poor pAML 
110 M5 50 46, XY 27.3 11.3 85 Intermediate I Poor Poor pAML 
111 0.9 M5 54 96 46,XY 70.3 8.1 139 Favorable Favorable Better pAML 
UPNOX40FABAgeSexPBB (%) Diff.KaryoWBC (G/L)Hb (g/dL)Plt (G/L)ELNNCCNMRCEtiology
1.4 M0 78 84 45,XY, −7,+13,-21 [28], 46,XY [2] 85 6.3 20 Adverse Poor Poor pAML 
M0 46 97 46, XY 60.5 39 Favorable Favorable Better pAML 
3.5 M0 49 83 46, XY, t(1;20)(p32;p13),del(12)(p12p13)[20] 29.2 12.9 39 Intermediate II Intermediate Intermediate sAML 
2.4 M1 50 93 46, XX 270 18 Intermediate I Intermediate Poor pAML 
5.1 M1 40 100 complex 81.3 10.8 51 Adverse Poor Poor pAML 
1.4 M1 27 95 46, XY 135.1 8.1 20 Favorable Favorable Better pAML 
1.3 M1 58 93 46,XY 23.9 8.2 42 Intermediate I Intermediate Intermediate n.a. 
M1 83 88 n.d. 128.9 7.5 15 n.a. n.a. n.a. pAML 
2.6 M1 67 78 complex 11.1 8.9 48 Adverse Poor Poor pAML 
10 2.3 M1 76 76 n.d. 15.6 10.7 46 n.a. n.a. n.a. pAML 
11 1.3 M1 76 64 Complex 12180 7,3 134 Adverse Poor Poor tAML 
12 1.1 M1 61 63 46,XY[25] 59.4 13 69 Intermediate I Intermediate Intermediate tAML 
13 1.5 M1 68 86 46,XY 84.1 6.7 332 Intermediate I Poor Poor pAML 
14 1.5 M1 75 84 46, XX 150 10.8 174 Favorable Favorable Better pAML 
15 3.8 M1 80 93 n.d. 34.9 11.2 28 n.a. n.a. n.a. pAML 
16 1.5 M1 72 80 46,XX [20] 25.8 7.8 50.0 Intermediate I Intermediate Intermediate pAML 
17 2.8 M1 64 94 n.d. 222 9.2 44 n.a. n.a. Poor pAML 
18 1.9 M1 63 60 46,XX, del(9)(q22) [5]/46,XX [25] 11.3 11.3 292 Favorable Intermediate Intermediate pAML 
19 1.4 M1 50 98 46,XY 19.7 10.4 55 Favorable Favorable Better pAML 
20 1.4 M1 67 99 n.d. 67.7 9.8 23 n.a. n.a. n.a. n.a. 
21 1.5 M1 62 77 46,XY 45 7.1 270 Intermediate I Poor Poor pAML 
22 1.1 M1 50 89 46,XX, del(9)(q13,q22)[4]/46,XX [16] 13.8 8.8 25 Intemediate II Intermediate Intermediate pAML 
23 M1 44 75 46, XY 10.7 7.9 234 Intemediate I Poor Poor pAML 
24 2.2 M1 82 81 46, XX 130 10.1 127 Intermediate I Intermediate Intermediate pAML 
25 2.7 M1 47 98 n.d. 56.3 9.4 60 n.a. n.a. n.a. pAML 
26 0.9 M1 76 81 46,XX[20], 10.7 7.4 145 Favorable Favorable Better pAML 
27 6.9 M1 47 91 47,XX, +8 27.6 10.0 26 Intermediate II Intermediate Intermediate sAML 
28 1.1 M1 62 89 n.a. 18.3 12.3 18 n.a. n.a. Poor pAML 
29 1.4 M1 52 98 46,XY 153.3 9.2 23 Intermediate I Poor Poor pAML 
30 3.8 M1 56 87 46, X, t(X;12)(p11;p13)[14];46, XX [7] 4.14 8.3 111 Intermediate II Intermediate Intermediate pAML 
31 0.9 M1 68 93 n.d. 42.5 9.8 80 n.a. n.a. n.a. pAML 
32 1.7 M1 21 95 46,XX [16], 46,XX, del(9)(q13q22) [4] 84 7.1 30 Intermediate II Intermediate Poor pAML 
33 4.5 M1 41 92 46,XX 68.7 8.1 55 Intermediate I Poor Poor pAML 
34 1.1 M1 34 75 46, XY 33.3 10.2 11 Favorable Favorable Better pAML 
35 1.1 M1 38 60 46, XX 26 9.6 153 Intermediate I Poor Poor pAML 
36 1.2 M2 84 79 n.d. 115.2 7.8 107 n.a. n.a. n.a. pAML 
37 2.3 M2 30 53 46, XY 24.5 7.4 54 Intermediate I Poor Poor pAML 
38 1.1 M2 54 71 46,XX,t(8;21)(q22;q22)[21] 8.9 40 Favorable Favorable Better tAML 
39 1.2 M2 38 28 46, XY 8.8 8.7 52 Intermediate I Intermediate Intermediate sAML 
40 1.1 M2 72 97 Complex 56.9 8.9 30 Adverse Poor Poor sAML 
41 1.4 M2 56 63 46, XY 8.8 9.7 311 Favorable Favorable Better pAML 
42 1.2 M2 81 96 n.d. 10 10 46 n.a. n.a. n.a. sAML 
43 1.2 M2 66 80 n.d. 27.8 10.1 49 Favorable Better Better pAML 
44 2.3 M2 29 70 46,XY 9.6 8.5 17 Intermediate I Poor Poor tAML 
45 1.3 M2 73 83 46,X,inv(X)(p22.3q13) [14]; 46,XX [6] 6.3 95 Intermediate II Intermediate Intermediate pAML 
46 1.8 M2 73 83 46,XY 33.8 8.1 80 Intermediate I Poor Poor pAML 
47 1.4 M2 68 96 46,XY 85.5 9.5 146 Intermediate II Intermediate Intermediate pAML 
48 1.6 M2 72 81 46, XY, del(17)(q11q21) [6]/47,XY,+8 [3] 46, XY [16] 78.9 6,4 47 Intermediate II Intermediate  pAML 
49 1.4 M2 51 43 46,XX,t(2;2)(q21;q23)[21] 35.3 9.3 51 Intermediate II Intermediate Better pAML 
50 1.2 M2 48 87 46,XY 36.9 7.1 66 Favorable Favorable Better pAML 
51 1.8 M2 63 78 46,XY 180 30 Favorable Favorable Better pAML 
52 1.5 M2 64 82 47, XY+8[16]/47,XY,+8,del(9)(q22)[1]/46,XY,del(9)(q22)[1]/46,XY[7] 338.5 8.1 19 Intermediate II Intermediate Intermediate pAML 
53 0.4 M2 58 92 46,XX 62 7.4 25 Intermediate I Poor Poor sAML 
54 1.1 M2 78 99 45, XY, −7[7], 46, XY[15] 26 9.7 36 Adverse Poor  sAML 
55 6.4 M2 68 84 47,XX,+11[23] 165.5 3.8 222 Intermediate II Intermediate Poor pAML 
56 1.6 M2 71 94 46, XX 105.4 10.4 83 Intermediate I Poor Poor pAML 
57 1.7 M2 58 20 Complex 39.1 9.3 23 Adverse Poor Poor pAML 
58 1.3 M2 23 69 47,XX, +21 [2]/46,XX [23] 50 9.9 135 Intermediate II Intermediate Intermediate pAML 
59 1.1 M2 71 81 47, XX +11 26.1 9.4 10 Intermediate II Intermediate Intermediate sAML 
60 1.3 M2 45 80 Complex 17.1 9.0 51 Favorable Favorable Better pAML 
61 2.5 M2 44 94 46,XY 138.5 10.3 57 Intermediate I Poor Poor pAML 
62 1.1 M2 45 94 47, XY, +8 44.4 8.7 11 Intermediate II Intermediate Poor pAML 
63 2.2 M2 45 94 Complex 44.4 8.7 11 Adverse Poor Poor pAML 
64 1.3 M2 68 93 47, XY, +11 110.9 27 Intermediate II Intermediate Intermediate pAML 
65 0.9 M2 70 20 47,XX,+8,del(17)(p11-12)[5]/46,XX[20] 13,6 182 Adverse Poor Poor sAML 
66 5.7 M3 46 51 Complex 23.1 13.3 25 n.a. n.a. n.a. pAML 
67 1.1 M3 67 94 46,XX 99 9.9 128 Intermediate II Intermediate Intermediate sAML 
68 14.3 M3 65 40 46, XY, t(15;17)(q22;q12)[20] 6.9 8.5 17 Favorable Favorable Better pAML 
69 5.2 M3 58 96 46,XX,t(15;17)(q22;q12)[20]. 42.1 8.4 17 Favorable Favorable Better pAML 
70 M4 70 71 45, XX, -7 282.3 5.7 68 Adverse Poor Poor sAML 
71 1.8 M4 49 72 46,Y,t(X;17)(p11;p1?1),add(21)(q22)[13] 61.9 5.8 18 Adverse Poor Poor pAML 
72 2.9 M4 71 93 47, XY, + 11 (6) 79.7 9.9 40 Intermediate II Intermediate Intermediate pAML 
73 1.4 M4 55 72 46,XY 17.2 8.6 162 Favorable Favorable Better pAML 
74 9.7 M4 61 70 46, XY 85.4 8.5 12 Intermediate I Poor Poor pAML 
75 6.1 M4 76 94 46, XX, t (9;11) (q22; q23) [12]/52, XXX, +3, +6, +8 t (t;11) (q22; q23) +12, +13, +18 [13] 141 12 70 Adverse Poor Poor tAML 
76 7.3 M4 43 86 46, XX 63.5 9.2 178 Favorable Favorable Better pAML 
77 1.3 M4 41 73 Complex 112.7 8.5 30 Adverse Poor Poor pAML 
78 M4 63 96 46,XY 92.3 10.5 431 Intermediate I Poor Poor pAML 
79 2.2 M4 73 85 46,XX (25) 52.6 7.5 48 Intermediate I Poor Poor tAML 
80 1.2 M4 71 75 46, XY,del(20)(q11)[6];47,idem,+11[14]; 161.1 8.6 61 Intermediate II Intermediate Intermediate sAML 
81 1.6 M4 83 95 46,XX add(14)(p11)[8],/46, XX[5] 155.8 11.6 144 Intermediate II Intermediate Intermediate pAML 
82 2.3 M4 78 39 51,XX,+6,+9,+9,+11,+13[20] 8.8 9.2 146 Adverse Poor Poor pAML 
83 2.7 M4 64 22 46,XY 62.4 7.5 40 Intermediate I Poor Poor n.a. 
84 1.5 M4 36 78 46, XY 207.4 6.1 55 Intermediate I Poor Poor pAML 
85 M4 68 82 46, XY 148.7 9.1 134 Intermediate I Poor Poor pAML 
86 4.4 M4 67 75 46,XY,?dup(9)(p13p22)c 64.4 9.1 33 Intermediate II Intermediate Intermediate pAML 
87 0.6 M4 72 81 46,XX 26.1 8.2 70 Intermediate I Intermediate Interemdiate pAML 
88 M4 54 46 46, XX 17.2 10.6 167 Intermediate I Poor Poor pAML 
89 M4 57 68 n.d. 334 9.4 293 n.a. n.a. n.a. pAML 
90 2.2 M4 57 29 46,XX 17.5 8.0 117 Intermediate I Intermediate Intermediate pAML 
91 3.3 M4 56 64 46, XX, inv(16)(p13.1q22)[11] 56.6 10.9 99 Favorable Favorable Better pAML 
92 0.8 M5 70 90 46,XY 190 7.1 65 Intermediate I Intermediate Intermediate pAML 
93 1.6 M5 70 46, XY 73.6 13.2 25 Favorable Favorable Better pAML 
94 1.6 M5 26 18 46, XX 51 6,2 17 Favorable Favorable Better pAML 
95 7.3 M5 73 97 47, XY,+8[2]; 46,XY[23] 78.9 6.4 47 Intermediate II Intermediate Poor tAML 
96 28.5 M5 69 80 n.d. 275 7.1 47 n.a. n.a. Poor pAML 
97 1.3 M5 65 91 n.d. 394.2 7.9 189 n.a. n.a. n.a. sAML 
98 1.7 M5 69 11 n.d. 22.1 8.7 53 n.a. n.a. n.a. pAML 
99 1.1 M5 24 90 n.d. n.d. n.d. n.d. n.a. n.a. n.a. pAML 
100 1.3 M5 78 49 Complex 11.7 13 Adverse Poor Poor pAML 
101 1.3 M5 74 >90 46,XY 239 5.7 122 Intermediate I Poor Poor pAML 
102 9.5 M5 52 46, XX 15.7 7.2 21 Favorable Favorable Better pAML 
103 1.2 M5 54 89 46, XY, del(9)(q13q22)[8]/48, XY[16] 109.4 8.2 78 Intermediate II Intermediate Intermediate pAML 
104 4.6 M5 72 40 46,XY 14.5 5.3 28 Intermediate I Intermediate Intermediate sAML 
105 31.6 M5 82 35 n.d. 61.3 11.7 110 n.a. n.a. n.a. pAML 
106 1.5 M5 23 83 48,XY,+8,+13[17]/46,XY[3] 145.1 7.6 34 Intermediate II Intermediate Poor pAML 
107 1.3 M5 58 70 46,XX 0.6 9.1 42 Intermediate I Poor Poor pAML 
108 0.7 M5 45 80 46,XX 248.1 7.3 46 Favorable Favorable Better pAML 
109 18.7 M5 53 85 46,XY 105.6 8.1 35 Intermediate I Poor Poor pAML 
110 M5 50 46, XY 27.3 11.3 85 Intermediate I Poor Poor pAML 
111 0.9 M5 54 96 46,XY 70.3 8.1 139 Favorable Favorable Better pAML 

Abbreviations: UPN, uniform patient number; OX40 SFI, specific fluorescence index; FAB, French–American–British classification; F, female; M, male; PBB, peripheral blood blasts among nucleated cells; WBC, white blood count; Hb, hemoglobin; Plt, platelets; ELN, European LeukemiaNet; NCCN, National Comprehensive Cancer Network; MRC, Medical Research Council; Etiology: pAML, de novo AML; tAML, therapy-related AML; sAML, secondary AML evolving from MDS/MPS; n.d., not determined; n.a., not available.

Figure 3.

OX40 is expressed on AML cells. A and B, PBMC from patients with AML (top) and CD34+ BM and peripheral blood cells of healthy donors (middle and bottom) were analyzed by FACS using the OX40 mAb BerAct35 (shaded peaks) and isotype control (open peaks) followed by anti-mouse PE. Exemplary (A) and combined (SFI levels, B) results with AML cells of different FAB types are shown. C and D, PBMC of patients with AML with >90% blast count (C) and CD34-enriched BM cells from healthy donors (D) were analyzed by RT-PCR for OX40 mRNA; 18S rRNA served as control. E, Quantitative real-time PCR analysis of OX40 mRNA expression in PBMC samples of different OX40 surface-positive and -negative AML patients (both n = 8) and CD34-enriched BM cells from 7 different healthy donors. F, PBMC of 7 patients with AML were exposed to sOX40L [2 μg/mL], G-CSF, GM-CSF, IFNγ, TNF, IL6, IL8, and IL10 [all 100 ng/mL] for 24 hours followed by FACS analysis with a noncompeting OX40 mAb (M-OX17). *, Statistically significant differences, P < 0.05.

Figure 3.

OX40 is expressed on AML cells. A and B, PBMC from patients with AML (top) and CD34+ BM and peripheral blood cells of healthy donors (middle and bottom) were analyzed by FACS using the OX40 mAb BerAct35 (shaded peaks) and isotype control (open peaks) followed by anti-mouse PE. Exemplary (A) and combined (SFI levels, B) results with AML cells of different FAB types are shown. C and D, PBMC of patients with AML with >90% blast count (C) and CD34-enriched BM cells from healthy donors (D) were analyzed by RT-PCR for OX40 mRNA; 18S rRNA served as control. E, Quantitative real-time PCR analysis of OX40 mRNA expression in PBMC samples of different OX40 surface-positive and -negative AML patients (both n = 8) and CD34-enriched BM cells from 7 different healthy donors. F, PBMC of 7 patients with AML were exposed to sOX40L [2 μg/mL], G-CSF, GM-CSF, IFNγ, TNF, IL6, IL8, and IL10 [all 100 ng/mL] for 24 hours followed by FACS analysis with a noncompeting OX40 mAb (M-OX17). *, Statistically significant differences, P < 0.05.

Close modal
Table 2.

Association of OX40 expression with genetic landscape and clinical parameters

CharacteristicAnalyzed patientsP value two-sidedaCox regressionCorrelationb
PML/RARA t(15;17) 96 <0.05   
RUNX1/RUNX1T1 t(8;21) 94 n.s.   
CBFB/MYH11 inv(16) 93 n.s.   
Subtype of FLT3 mutation     
FLT3-ITD 84 <0.05   
FLT3-TKD 83 n.s.   
NPM1 84 n.s.   
CEBPA 55 n.s.   
MLL-PTD 78 n.s.   
ELN 90 n.s.   
ELN 2017 88 n.s.   
Survival 105  n.s.  
CR 63 n.s.   
Etiology 108 n.s.   
pAML 87    
sAML 14    
tAML    
Relapse  n.s.   
FAB 105 n.s.   
Age at diagnosis 110   n.s. 
Sex 110   n.s. 
PBB (%) 109   n.s. 
WBC 108   n.s 
Plt (G/L) 108   n.s. 
HB (g/dL) 108   n.s. 
CRP (mg/dL) 108   n.s. 
CharacteristicAnalyzed patientsP value two-sidedaCox regressionCorrelationb
PML/RARA t(15;17) 96 <0.05   
RUNX1/RUNX1T1 t(8;21) 94 n.s.   
CBFB/MYH11 inv(16) 93 n.s.   
Subtype of FLT3 mutation     
FLT3-ITD 84 <0.05   
FLT3-TKD 83 n.s.   
NPM1 84 n.s.   
CEBPA 55 n.s.   
MLL-PTD 78 n.s.   
ELN 90 n.s.   
ELN 2017 88 n.s.   
Survival 105  n.s.  
CR 63 n.s.   
Etiology 108 n.s.   
pAML 87    
sAML 14    
tAML    
Relapse  n.s.   
FAB 105 n.s.   
Age at diagnosis 110   n.s. 
Sex 110   n.s. 
PBB (%) 109   n.s. 
WBC 108   n.s 
Plt (G/L) 108   n.s. 
HB (g/dL) 108   n.s. 
CRP (mg/dL) 108   n.s. 

Abbreviations: ELN, European LeukemiaNet classification; ELN 2017, modified European LeukimiaNet classification; CR, complete remission; pAML, de novo AML; tAML, therapy-related AML; sAML, secondary AML evolving from MDS/MPS; FAB, French–American–British classification; PBB, peripheral blood blasts among nucleated cells; WBC, white blood count; Plt, platelets; HB, hemoglobin; CRP, C-reactive protein; n.s., not significant.

aAll P values are two-sided and were calculated either with Mann–Whitney or Kruskal–Wallis test or, for categorial variables, with the χ2 tests.

bCorrelation Spearman r.

OX40 can induce cytokine release and promote proliferation of AML cells

To elucidate the role of OX40 on AML cells, we characterized our OX40 mAb using reporter cell assays in which Jurkat-JOM2 cells expressing a human OX40–Fas chimeric receptor are killed upon engagement of the OX40 portion of the receptor (26). OX40 mAb clone M-OX17 killed these reporter cells in a dose-dependent manner, indicating that it has agonist activity (Fig. 4A). The same mAb also stimulated a robust IL8 production in U937-OX40 transfectants, but not in mock controls, which further confirmed its stimulatory property (Fig. 4B).

Figure 4.

OX40 induces cytokine release and metabolic activity of AML cells. A, OX40: Fas reporter cells (Jurkat-JOM2) were exposed to the indicated concentrations of the anti-OX40 antibody M-OX17 or isotype control and metabolic activity was measured by WST-1 assays. Representative data from one out of four experiments with similar results are shown. B, U937-OX40 and U937-mock transfectants were cultured alone, on immobilized OX40 mAb (M-OX17) or isotype control for 24 hours. Then, IL8 in culture supernatants was measured by ELISA. Representative data from one out of four experiments with similar results are shown. C–F, AML patient cells were cultured alone, on immobilized M-OX17 mAb or isotype control for 6 hours (TNF and IL8) or 24 hours (IL10 and IL6). Then (C) levels of the indicated cytokines in culture supernatants were determined by ELISA. Data from OX40+ patients (n = 19; UPN 3, 4, 5, 15, 27, 30, 33, 52, 55, 61, 70, 72, 76, 81, 83, 86, 94, 96, 106) are shown. Horizontal bars, the mean of the results in each culture condition. D, The results obtained in C were analyzed with regard to release of specific cytokine combinations. Positive response (+) was defined as >2-fold increase of each individual cytokine upon OX40 signaling. The percentage of samples responding with the indicated cytokine pattern is depicted. E, Intracellular cytokine levels were analyzed by FACS using specific mAbs and isotype control after 12 hours. AML cells within PBMC were selected as CD3/CD19/CD33+. Numbers in top right quadrants indicate the percentage of IL8+ and TNF+ AML cells. F, Metabolic activity was measured by WST-1 assays after 24 hours. Data of OX40+ patients (n = 15; UPN 3, 10, 15, 21, 25, 27, 33, 46, 56, 61, 71, 72, 81, 91, 106) are shown. Horizontal bars represent the mean of the results in each culture condition. *, Statistically significant differences, P < 0.05.

Figure 4.

OX40 induces cytokine release and metabolic activity of AML cells. A, OX40: Fas reporter cells (Jurkat-JOM2) were exposed to the indicated concentrations of the anti-OX40 antibody M-OX17 or isotype control and metabolic activity was measured by WST-1 assays. Representative data from one out of four experiments with similar results are shown. B, U937-OX40 and U937-mock transfectants were cultured alone, on immobilized OX40 mAb (M-OX17) or isotype control for 24 hours. Then, IL8 in culture supernatants was measured by ELISA. Representative data from one out of four experiments with similar results are shown. C–F, AML patient cells were cultured alone, on immobilized M-OX17 mAb or isotype control for 6 hours (TNF and IL8) or 24 hours (IL10 and IL6). Then (C) levels of the indicated cytokines in culture supernatants were determined by ELISA. Data from OX40+ patients (n = 19; UPN 3, 4, 5, 15, 27, 30, 33, 52, 55, 61, 70, 72, 76, 81, 83, 86, 94, 96, 106) are shown. Horizontal bars, the mean of the results in each culture condition. D, The results obtained in C were analyzed with regard to release of specific cytokine combinations. Positive response (+) was defined as >2-fold increase of each individual cytokine upon OX40 signaling. The percentage of samples responding with the indicated cytokine pattern is depicted. E, Intracellular cytokine levels were analyzed by FACS using specific mAbs and isotype control after 12 hours. AML cells within PBMC were selected as CD3/CD19/CD33+. Numbers in top right quadrants indicate the percentage of IL8+ and TNF+ AML cells. F, Metabolic activity was measured by WST-1 assays after 24 hours. Data of OX40+ patients (n = 15; UPN 3, 10, 15, 21, 25, 27, 33, 46, 56, 61, 71, 72, 81, 91, 106) are shown. Horizontal bars represent the mean of the results in each culture condition. *, Statistically significant differences, P < 0.05.

Close modal

Next, we asked whether OX40 on leukemic cells of patients with AML was functional. We observed no association of constitutive OX40 expression with the basal release of the cytokines TNF, IL10, IL6, and IL8, metabolic activity/proliferation of leukemic cells or apoptosis/death in vitro (Supplementary Fig. S2). However, M-OX17-induced OX40 signaling resulted in a significant (all P < 0.05, Wilcoxon signed rank test) induction of TNF, IL10, IL6, and IL8 by OX40+ AML cells (both in cases with de novo and secondary AML from MDS; Fig. 4C; Supplementary Fig. S3A). We observed inter-individual differences concerning the cytokine release of AML cells upon OX40 signaling: None of the 19 investigated samples released all four cytokines. Release of TNF, IL10, IL6, and IL8 was observed with 15, 10, 15, and 3 of the 19 samples, respectively (Fig. 4D). Analysis of intracellular IL8 and TNF levels by FACS and gating for CD33+/CD19/CD3 cells served to ascertain that in fact the leukemic cells among patient-PBMC produced the respective cytokines upon OX40 stimulation (Fig. 4E). WST-1 assays then revealed that OX40 signaling can also significantly (P < 0.05, Wilcoxon signed rank test) enhance AML-cell proliferation/viability: in OX40+ AML patient samples, 8 (53%) and 5 (33%) out of 15 investigated patients responded to OX40 stimulation with a 1.5-fold and 2-fold increase, respectively (Fig. 4F; Supplementary Fig. S3B).

OX40–OX40L interaction enhances NK-cell cytotoxicity in response to AML cells

Finally, we analyzed the outcome of OX40–OX40L interaction for NK-cell antileukemia reactivity. As a first step, we studied the frequency and activation state of NK cells within patients with AML with different OX40 expression levels on leukemic blasts, which, however, did not reveal a correlation (Supplementary Fig. S4). This can be attributed to the fact that NK-cell reactivity is influenced by a multitude of activating and inhibitory receptors as well as cytokines and other immune cell subsets far beyond the OX40/OX40L molecule system (17). Accordingly, we next aimed to delineate the role of this molecule system by employing our blocking OX40–F(ab′)2-fragments in long-term (24 hours) chromium release assays with OX40L+ pNKC-SJ of 9 different healthy donors and OX40+ primary AML cells from 8 different patients (at least 80% blast count). Blocking OX40 significantly (P < 0.0001, Wilcoxon signed rank test) decreased lysis of primary AML cells, whereas the isotype control had no relevant effect (Fig. 5A and B). To further exclude an influence of other immune effector cells remaining in AML patient samples or the pNKC preparations, we conducted lysis assays using primary AML samples after MACS-depletion of CD3+/CD14+/CD19+/CD56+ cells as targets with purified NK cells obtained by MACS-isolation from bulk pNKC-SJ as effectors. Again, we found that blocking OX40–OX40L interaction reduced AML cell lysis, which confirmed the stimulatory role of this molecule system in AML-NK-cell interaction (Fig. 5C).

Figure 5.

OX40–OX40L interaction enhances NK-cell reactivity against AML cells. A and B, OX40L+ pNKC-SJ were cultured with PBMC of the indicated OX40-expressing AML patients with more than 80% blast count in the presence or absence of blocking OX40-F(ab′)2 or isotype control and cytotoxicity was evaluated by 24-hour 51chromium release assays. In A, three representative results are depicted, B shows data of 17 independent experiments at an E:T ratio of 40:1. C, Lysis was analyzed as described above using primary AML cells after MACS depletion of CD3+/CD14+/CD19+/CD56+ cells contained in the samples as targets and NK cells isolated by MACS isolation from bulk pNKC-SJ as effectors. Three representative results from 9 experiments with similar results are shown. *, Statistically significant differences, P < 0.05.

Figure 5.

OX40–OX40L interaction enhances NK-cell reactivity against AML cells. A and B, OX40L+ pNKC-SJ were cultured with PBMC of the indicated OX40-expressing AML patients with more than 80% blast count in the presence or absence of blocking OX40-F(ab′)2 or isotype control and cytotoxicity was evaluated by 24-hour 51chromium release assays. In A, three representative results are depicted, B shows data of 17 independent experiments at an E:T ratio of 40:1. C, Lysis was analyzed as described above using primary AML cells after MACS depletion of CD3+/CD14+/CD19+/CD56+ cells contained in the samples as targets and NK cells isolated by MACS isolation from bulk pNKC-SJ as effectors. Three representative results from 9 experiments with similar results are shown. *, Statistically significant differences, P < 0.05.

Close modal

The therapeutic inhibition of immune checkpoints to reinforce antitumor immunity of T cells has become a mainstay of cancer treatment. However, many patients benefit only temporarily or not at all from checkpoint blockers that target CTLA-4 and PD-1/PD-L1 (2). Additional strategies are being developed to better exploit the immune system's potential to combat malignant disease. Such approaches include, among others, therapeutic stimulation of activating immune receptors on T cells as well as ex vivo manipulation/expansion and subsequent transfer of cytotoxic lymphocytes such as chimeric antigen receptor (CAR) T cells (31) or pNKC (29).

The TNFR family member OX40 is an activating receptor that can reinforce T-cell antitumor reactivity in the sense of a “stimulatory” immune checkpoint. Preclinical studies with agonistic mAb revealed that the ability of OX40 to stimulate T cells is comparable with CTLA-4 blockade (12). Due to its ability to sustain T-cell proliferation/survival, OX40 is also used in the costimulatory signaling domain of CAR (32, 33). The latter confines the effects of OX40 activation to the transfected T cells, whereas systemic application of agonistic mAb may also affect other cellular components of the immune system and the many non-immune cells that express OX40 (4–6). Here, we used FACS analysis to study 111 samples from patients with AML. We found that leukemic cells from a large proportion of patients (but not CD34+ cells of healthy donors or patients with MDS or CML) express OX40 on the cell surface. OX40 mRNA expression was also observed in AML samples without expression of the protein on the cell surface. Although contamination with OX40-expressing healthy cells may have influenced the respective PCR results, we consider it more likely that posttranscriptional or posttranslational mechanisms blocked expression of transcribed OX40 mRNA. In addition, as for other TNF/TNFR members, protein expressed on the cell surface may be shed and released in soluble form. Indeed, soluble OX40 is found in sera of patients with malignant and autoimmune diseases (34–37). Our finding that OX40 expression can be upregulated by exposure of AML cells to TNF indicates that environmental stimuli in the micromilieu may affect OX40 expression via these or other yet unidentified mechanisms.

Functional analyses using newly generated mAb with defined specificity and agonistic properties revealed that OX40 signaling can induce the release of cytokines that act as autocrine/paracrine growth and survival factors in AML and are associated with development and progression of the disease (22, 23). OX40 signaling did not always induce release of the same cytokines. Rather, we found distinct patterns of cytokine release upon OX40 signaling: TNF and IL6 were released in more than 70% of the investigated cases, IL10 released in about 50% of cases, and IL8 released in <20% of cases. In none of the AML samples were all four cytokines released, although all investigated OX40+ AML patient samples responded to OX40 signaling by release of at least one cytokine. OX40 may thus (variably) contribute to the cytokine milieu associated with AML. As with T cells, OX40 signaling enhanced viability/metabolic activity of the leukemic cells in a substantial proportion of the AML cases. It seems thus possible that OX40 confers a survival benefit for leukemic cells that is enacted through interaction with OX40L-bearing immune or bystander cells. This is in line with evidence regarding the role of the immune and stromal microenvironment in malignancies in general, which also holds true for AML (38). Moreover, these findings concur with our hypothesis regarding therapeutic application of “untargeted” agonistic OX40 mAb. Other investigators have reported on OX40 expression on cancer cells of various origins other than AML, although without analyzing functionality (39). Another layer of complexity is added by the issue of whether mAb-binding to OX40 affects interaction with cells that express its cognate ligand. OX40–OX40L interaction can lead to transduction of bidirectional signals, this is into the receptor- and the ligand-bearing cell, a characteristic feature of many ligands of the TNF family (30, 40). Besides healthy tissues like endothelial cells, antigen-presenting cells—including B cells and monocytes/dendritic cells—express OX40L, and various cellular functions of these cells are affected by OX40L "reverse signaling" (30, 41–43), that may also occur upon their interaction with OX40-expressing AML cells.

Zingoni and colleagues reported that NK cells, which play an important role in immune surveillance, particularly of leukemia (44), also express OX40L following activation and stimulate OX40-expressing T cells via this receptor–ligand system (20). Despite the multiple approaches presently used to evaluate the clinical efficacy of ex vivo expanded/activated NK cells upon adoptive transfer (17), the expression and function of OX40L by such cell preparations has so far not been analyzed. When we studied OX40L expression on pNKC generated as described in the work of Zingoni and colleagues (; pNKC-8866; ref. 20) or generated according to a protocol used for large-scale clinical grade expansion (pNKC-SJ; refs. 24, 29, 45), we observed no or very low OX40L expression on the first which are generated by culture with RPMI8866 feeder cells in the presence of IL2. In contrast, OX40L was highly expressed on pNKC-SJ that are expanded in the presence of K562 cells transfected with 4-1BBL and membrane-bound IL15 as well as soluble IL2. In line with the findings of Zingoni and colleagues (20), cytokine activation of NK cells alone was not sufficient to induce OX40L expression, and additional expression of 4-1BBL as revealed by analyses involving transfectants and transwell settings was required. In a clinical study in which patients with pediatric solid tumors received IL15/4-1BBL-activated NK cells after allogeneic SCT, acute GVHD was observed despite the fact that the T-cell dose in the grafts was below the threshold usually required for GVHD in this setting (46). Although the underlying mechanism is unknown, we speculate that the ability of NK cells to stimulate T cells via OX40–OX40L interaction may have contributed to the result. This hypothesis is supported by observations in mouse models of GVHD, where OX40 signals in effector T cells play a crucial pathophysiological role (47–49).

When we analyzed how OX40L reverse signaling affected NK cells, we found that OX40L-induced activation and production of IFNγ, a cytokine which participates in cancer elimination by inhibiting cellular proliferation and angiogenesis, promoting apoptosis, and stimulating the adaptive immune system (50). Analyses with OX40 transfectants revealed that OX40–OX40L interaction also enhanced NK lysis of target cells. This was further confirmed in experiments using F(ab′)2-fragments of a blocking OX40-antibody, which reduced NK-cell killing. Independent experiments (n = 17) with primary OX40-expressing AML cells and OX40L+ pNKC of different patients/donors also revealed that disruption of OX40–OX40L interaction reduces target cell lysis despite donor variability. The latter might be due to different OX40 and OX40L expression (or differences in other immunoregulatory molecules that influence NK function) on AML cells and pNKC, respectively. Thus, even though the pathways and mechanisms that mediate OX40L signaling in NK cells remain unclear, our results demonstrate that OX40L enhances the reactivity of NK cells.

Here, we have studied the expression of OX40 in AML and its involvement in disease pathophysiology. In addition, we report on the consequences of reverse signaling via its counterpart OX40L for NK cells including their antileukemia reactivity. We were not able to demonstrate expression of OX40L on NK cells of patients with AML directly ex vivo, possibly because all OX40L antibodies available for FACS analysis compete with OX40 for OX40L binding. NK-cell–expressed OX40L could thus be masked by soluble OX40 (36, 37). OX40L can be released by shedding, which might be enhanced after binding to its AML-expressed counterpart. So far, the reason for our failure to detect OX40L on NK cells in our ex vivo analyses remains unclear. In addition, it remains to be determined whether and how OX40/OX40L interactions affect AML cells or NK cells in an autocrine/paracrine manner, as the latter themselves can express OX40.

The potential consequences of bidirectional signaling through OX40 and OX40L should be considered when using adoptive transfer of ex vivo expanded pNKC. This bidirectional signaling may also affect therapeutic strategies targeted at checkpoint stimulation by agonistic OX40 mAbs, as OX40 can affect multiple cell types beyond T cells including malignant cells. The effects of OX40 agonists may be more complex than anticipated: the treatment may also influence (i) cellular properties of the malignant cells, (ii) their interaction with the microenvironment, (iii) other OX40-expressing immune cells (including NK cells), and, in case of blocking properties of the applied mAb, (iv) reactivity of other OX40L-expressing immune cells, including NK-cell immunosurveillance. Thus, additional work is warranted to unravel the role of the OX40/OX40L molecule system and to exploit the potential of OX40 stimulation for cancer immunotherapy.

No potential conflicts of interest were disclosed.

Conception and design: T. Nuebling, C.E. Schumacher, H.R. Salih

Development of methodology: T. Nuebling, C.E. Schumacher, M. Hofmann, B.J. Schmiedel, P. Schneider

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Nuebling, C.E. Schumacher, M. Hofmann, I. Hagelstein, S. Maurer, B. Federmann, K. Rothfelder, M. Roerden, D. Dörfel

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T. Nuebling, C.E. Schumacher, I. Hagelstein, B.J. Schmiedel, S. Maurer, K. Rothfelder, M. Roerden, D. Dörfel, H.R. Salih

Writing, review, and/or revision of the manuscript: T. Nuebling, C.E. Schumacher, I. Hagelstein, B.J. Schmiedel, D. Dörfel, P. Schneider, H.R. Salih

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.E. Schumacher, I. Hagelstein, S. Maurer, M. Roerden, D. Dörfel, G. Jung

Study supervision: T. Nuebling, H.R. Salih

This study was supported by grants from the Deutsche Forschungsgemeinschaft (NU341/1-1, SA1360/7-3, SFB685, project A07) and Deutsche Krebshilfe (projects 111828 and 111134). P. Schneider is supported by grants from the Swiss National Science Foundation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sharma
P
,
Allison
JP
. 
Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential
.
Cell
2015
;
161
:
205
14
.
2.
Postow
MA
,
Callahan
MK
,
Wolchok
JD
. 
Immune checkpoint blockade in cancer therapy
.
J Clin Oncol
2015
;
33
:
1974
82
.
3.
Linch
SN
,
McNamara
MJ
,
Redmond
WL
. 
OX40 agonists and combination immunotherapy: putting the pedal to the metal
.
Front Oncol
2015
;
5
:
34
.
4.
Sugamura
K
,
Ishii
N
,
Weinberg
AD
. 
Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40
.
Nat Rev Immunol
2004
;
4
:
420
31
.
5.
Croft
M
. 
Control of immunity by the TNFR-related molecule OX40 (CD134)
.
Annu Rev Immunol
2010
;
28
:
57
78
.
6.
Ito
T
,
Wang
YH
,
Duramad
O
,
Hanabuchi
S
,
Perng
OA
,
Gilliet
M
, et al
OX40 ligand shuts down IL-10-producing regulatory T cells
.
Proc Natl Acad Sci USA
2006
;
103
:
13138
43
.
7.
Piconese
S
,
Valzasina
B
,
Colombo
MP
. 
OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection
.
J Exp Med
2008
;
205
:
825
39
.
8.
Ali
SA
,
Ahmad
M
,
Lynam
J
,
McLean
CS
,
Entwisle
C
,
Loudon
P
, et al
Anti-tumour therapeutic efficacy of OX40L in murine tumour model
.
Vaccine
2004
;
22
:
3585
94
.
9.
Lee
SJ
,
Myers
L
,
Muralimohan
G
,
Dai
J
,
Qiao
Y
,
Li
Z
, et al
4–1BB and OX40 dual costimulation synergistically stimulate primary specific CD8 T cells for robust effector function
.
J Immunol
2004
;
173
:
3002
12
.
10.
Ladanyi
A
,
Somlai
B
,
Gilde
K
,
Fejos
Z
,
Gaudi
I
,
Timar
J
. 
T-cell activation marker expression on tumor-infiltrating lymphocytes as prognostic factor in cutaneous malignant melanoma
.
Clin Cancer Res
2004
;
10
:
521
30
.
11.
Petty
JK
,
He
K
,
Corless
CL
,
Vetto
JT
,
Weinberg
AD
. 
Survival in human colorectal cancer correlates with expression of the T-cell costimulatory molecule OX-40 (CD134)
.
Am J Surg
2002
;
183
:
512
8
.
12.
Curti
BD
,
Kovacsovics-Bankowski
M
,
Morris
N
,
Walker
E
,
Chisholm
L
,
Floyd
K
, et al
OX40 is a potent immune-stimulating target in late-stage cancer patients
.
Cancer Res
2013
;
73
:
7189
98
.
13.
Pierson
BA
,
Miller
JS
. 
CD56+bright and CD56+dim natural killer cells in patients with chronic myelogenous leukemia progressively decrease in number, respond less to stimuli that recruit clonogenic natural killer cells, and exhibit decreased proliferation on a per cell basis
.
Blood
1996
;
88
:
2279
87
.
14.
Lowdell
MW
,
Craston
R
,
Samuel
D
,
Wood
ME
,
O'Neill
E
,
Saha
V
, et al
Evidence that continued remission in patients treated for acute leukaemia is dependent upon autologous natural killer cells
.
Br J Haematol
2002
;
117
:
821
7
.
15.
Tajima
F
,
Kawatani
T
,
Endo
A
,
Kawasaki
H
. 
Natural killer cell activity and cytokine production as prognostic factors in adult acute leukemia
.
Leukemia
1996
;
10
:
478
82
.
16.
Ruggeri
L
,
Capanni
M
,
Urbani
E
,
Perruccio
K
,
Shlomchik
WD
,
Tosti
A
, et al
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants
.
Science
2002
;
295
:
2097
2100
.
17.
Handgretinger
R
,
Lang
P
,
Andre
MC
. 
Exploitation of natural killer cells for the treatment of acute leukemia
.
Blood
2016
;
127
:
3341
9
.
18.
Schmiedel
BJ
,
Nuebling
T
,
Steinbacher
J
,
Malinovska
A
,
Wende
CM
,
Azuma
M
, et al
Receptor activator for NF-kappaB ligand in acute myeloid leukemia: expression, function, and modulation of NK cell immunosurveillance
.
J Immunol
2013
;
190
:
821
31
.
19.
Liu
B
,
Li
Z
,
Mahesh
SP
,
Pantanelli
S
,
Hwang
FS
,
Siu
WO
, et al
Glucocorticoid-induced tumor necrosis factor receptor negatively regulates activation of human primary natural killer (NK) cells by blocking proliferative signals and increasing NK cell apoptosis
.
J Biol Chem
2008
;
283
:
8202
10
.
20.
Zingoni
A
,
Sornasse
T
,
Cocks
BG
,
Tanaka
Y
,
Santoni
A
,
Lanier
LL
. 
Cross-talk between activated human NK cells and CD4+ T cells via OX40–OX40 ligand interactions
.
J Immunol
2004
;
173
:
3716
24
.
21.
Imura
A
,
Hori
T
,
Imada
K
,
Ishikawa
T
,
Tanaka
Y
,
Maeda
M
, et al
The human OX40/gp34 system directly mediates adhesion of activated T cells to vascular endothelial cells
.
J Exp Med
1996
;
183
:
2185
95
.
22.
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 leukemia and high-risk myelodysplastic syndromes
.
Cancer
2008
;
113
:
1605
13
.
23.
Kornblau
SM
,
McCue
D
,
Singh
N
,
Chen
W
,
Estrov
Z
,
Coombes
KR
. 
Recurrent expression signatures of cytokines and chemokines are present and are independently prognostic in acute myelogenous leukemia and myelodysplasia
.
Blood
2010
;
116
:
4251
61
.
24.
Fujisaki
H
,
Kakuda
H
,
Shimasaki
N
,
Imai
C
,
Ma
J
,
Lockey
T
, et al
Expansion of highly cytotoxic human natural killer cells for cancer cell therapy
.
Cancer Res
2009
;
69
:
4010
17
.
25.
Baltz
KM
,
Krusch
M
,
Bringmann
A
,
Brossart
P
,
Mayer
F
,
Kloss
M
, et al
Cancer immunoediting by GITR (glucocorticoid-induced TNF-related protein) ligand in humans: NK cell/tumor cell interactions
.
FASEB J
2007
;
21
:
2442
54
.
26.
Schneider
P
,
Willen
L
,
Smulski
CR
. 
Tools and techniques to study ligand-receptor interactions and receptor activation by TNF superfamily members
.
Methods Enzymol
2014
;
545
:
103
25
.
27.
Jung
G
,
Freimann
U
,
Von Marschall
Z
,
Reisfeld
RA
,
Wilmanns
W
. 
Target cell-induced T cell activation with bi- and trispecific antibody fragments
.
Eur J Immunol
1991
;
21
:
2431
35
.
28.
Cemerlic
D
,
Dadey
B
,
Han
T
,
Vaickus
L
. 
Cytokine influence on killing of fresh chronic lymphocytic leukemia cells by human leukocytes
.
Blood
1991
;
77
:
2707
15
.
29.
Szmania
S
,
Lapteva
N
,
Garg
T
,
Greenway
A
,
Lingo
J
,
Nair
B
, et al
Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients
.
J Immunother
2015
;
38
:
24
36
.
30.
Eissner
G
,
Kolch
W
,
Scheurich
P
. 
Ligands working as receptors: reverse signaling by members of the TNF superfamily enhance the plasticity of the immune system
.
Cytokine Growth Factor Rev
2004
;
15
:
353
66
.
31.
Sadelain
M
,
Riviere
I
,
Brentjens
R
. 
Targeting tumours with genetically enhanced T lymphocytes
.
Nat Rev Cancer
2003
;
3
:
35
45
.
32.
Hombach
AA
,
Abken
H
. 
Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling
.
Int J Cancer
2011
;
129
:
2935
44
.
33.
Finney
HM
,
Akbar
AN
,
Lawson
AD
. 
Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain
.
J Immunol
2004
;
172
:
104
13
.
34.
Schmiedel
BJ
,
Scheible
CA
,
Nuebling
T
,
Kopp
HG
,
Wirths
S
,
Azuma
M
, et al
RANKL expression, function, and therapeutic targeting in multiple myeloma and chronic lymphocytic leukemia
.
Cancer Res
2013
;
73
:
683
94
.
35.
Lum
L
,
Wong
BR
,
Josien
R
,
Becherer
JD
,
Erdjument-Bromage
H
,
Schlöndorff
J
, et al
Evidence for a role of a tumor necrosis factor-alpha (TNF-alpha)-converting enzyme-like protease in shedding of TRANCE, a TNF family member involved in osteoclastogenesis and dendritic cell survival
.
J Biol Chem
1999
;
274
:
13613
8
.
36.
Taylor
L
,
Schwarz
H
. 
Identification of a soluble OX40 isoform: development of a specific and quantitative immunoassay
.
J Immunol Methods
2001
;
255
:
67
72
.
37.
Komura
K
,
Yoshizaki
A
,
Kodera
M
,
Iwata
Y
,
Ogawa
F
,
Shimizu
K
, et al
Increased serum soluble OX40 in patients with systemic sclerosis
.
J Rheumatol
2008
;
35
:
2359
62
.
38.
Tripodo
C
,
Burocchi
A
,
Piccaluga
PP
,
Chiodoni
C
,
Portararo
P
,
Cappetti
B
, et al
Persistent immune stimulation exacerbates genetically driven myeloproliferative disorders via stromal remodeling
.
Cancer Res
2017
;
77
:
3685
99
.
39.
Xie
F
,
Wang
Q
,
Chen
Y
,
Gu
Y
,
Shi
Q
,
Ge
Y
, et al
Characterization and application of two novel monoclonal antibodies against human OX40: costimulation of T cells and expression on tumor as well as normal gland tissues
.
Tissue Antigens
2006
;
67
:
307
17
.
40.
Locksley
RM
,
Killeen
N
,
Lenardo
MJ
. 
The TNF and TNF receptor superfamilies: integrating mammalian biology
.
Cell
2001
;
104
:
487
501
.
41.
Stuber
E
,
Neurath
M
,
Calderhead
D
,
Fell
HF
,
Strober
W
. 
cross-linking of Ox40 Ligand, A Member of the Tnf/Ngf cytokine family, induces proliferation and differentiation in murine splenic B-Cells
.
Immunity
1995
;
2
:
507
21
.
42.
Ohshima
Y
,
Tanaka
Y
,
Tozawa
H
,
Takahashi
Y
,
Maliszewski
C
,
Delespesse
G
. 
Expression and function of OX40 ligand on human dendritic cells
.
J Immunol
1997
;
159
:
3838
48
.
43.
Kotani
A
,
Hori
T
,
Matsumura
Y
,
Uchiyama
T
. 
Signaling of gp34 (OX40 ligand) induces vascular endothelial cells to produce a CC chemokine RANTES/CCL5
.
Immunol Lett
2002
;
84
:
1
7
.
44.
Vivier
E
,
Raulet
DH
,
Moretta
A
,
Caligiuri
MA
,
Zitvogel
L
,
Lanier
LL
, et al
Innate or adaptive immunity? The example of natural killer cells
.
Science
2011
;
331
:
44
49
.
45.
Cho
D
,
Shook
DR
,
Shimasaki
N
,
Chang
YH
,
Fujisaki
H
,
Campana
D
. 
Cytotoxicity of activated natural killer cells against pediatric solid tumors
.
Clin Cancer Res
2010
;
16
:
3901
9
.
46.
Shah
NN
,
Baird
K
,
Delbrook
CP
,
Fleisher
TA
,
Kohler
ME
,
Rampertaap
S
, et al
Acute GVHD in patients receiving IL-15/4–1BBL activated NK cells following T-cell-depleted stem cell transplantation
.
Blood
2015
;
125
:
784
92
.
47.
Stuber
E
,
Von Freier
A
,
Marinescu
D
,
Folsch
UR
. 
Involvement of OX40–OX40L interactions in the intestinal manifestations of the murine acute graft-versus-host disease
.
Gastroenterology
1998
;
115
:
1205
15
.
48.
Tsukada
N
,
Akiba
H
,
Kobata
T
,
Aizawa
Y
,
Yagita
H
,
Okumura
K
. 
Blockade of CD134 (OX40)-CD134L interaction ameliorates lethal acute graft-versus-host disease in a murine model of allogeneic bone marrow transplantation
.
Blood
2000
;
95
:
2434
9
.
49.
Blazar
BR
,
Sharpe
AH
,
Chen
AI
,
Panoskaltsis-Mortari
A
,
Lees
C
,
Akiba
H
, et al
Ligation of OX40 (CD134) regulates graft-versus-host disease (GVHD) and graft rejection in allogeneic bone marrow transplant recipients
.
Blood
2003
;
101
:
3741
8
.
50.
Dunn
GP
,
Koebel
CM
,
Schreiber
RD
. 
Interferons, immunity and cancer immunoediting
.
Nat Rev Immunol
2006
;
6
:
836
48
.