We previously showed that the interaction of programmed death-ligand 1 (PD-L1) on multiple myeloma (MM) cells with PD-1 not only inhibits tumor-specific cytotoxic T-lymphocyte activity via the PD-1 signaling pathway but also induces drug resistance via PD-L1–mediated reverse signals. We here examined the regulation of PD-L1 expression by immunomodulatory drugs (IMiDs) and antimyeloma effects of the anti–PD-L1 antibody durvalumab in combination with IMiDs. IMiDs induced PD-L1 expression on IMiD-insensitive MM cells and plasma cells from patients newly diagnosed with MM. Gene-expression profiling analysis demonstrated that not only PD-L1, but also a proliferation-inducing ligand (APRIL), was enhanced by IMiDs. PD-L1 induction by IMiDs was suppressed by using the APRIL inhibitor recombinant B-cell maturation antigen (BCMA)-Ig, the antibody against BCMA, or an MEK/ERK inhibitor in in vitro and in vivo assays. In addition, its induction was abrogated in cereblon (CRBN)-knockdown MM cells, whereas PD-L1 expression was increased and strongly induced by IMiDs in Ikaros-knockdown cells. These results demonstrated that PD-L1 upregulation by IMiDs on IMiD-insensitive MM cells was induced by (i) the BCMA–APRIL pathway via IMiD-mediated induction of APRIL and (ii) Ikaros degradation mediated by CRBN, which plays a role in inhibiting PD-L1 expression. Furthermore, T-cell inhibition induced by PD-L1–upregulated cells was effectively recovered after combination treatment with durvalumab and IMiDs. PD-L1 upregulation by IMiDs on MM cells might promote aggressive myeloma behaviors and immune escape in the bone marrow microenvironment.

The programmed cell death 1 (PD-1; also known as CD279)–programmed death-ligand 1 (PD-L1; also known as CD274 and B7-H1) pathway (PD-1/PD-L1 signaling pathway) plays a key role in immune suppression in the tumor microenvironment (1–4). PD-L1 expressed on various tumor cells interacts with PD-1 on tumor-infiltrating T cells, inhibits functional T-cell activation through the inhibitory signal of T-cell receptor signaling, and reduces adaptive immune responses (5–7). Its blockade promotes endogenous adaptive anti-immunity, and PD-1/PD-L1–targeted therapy has shown clinical success in several types of cancers, including melanoma, non–small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, and microsatellite instability–high cancers (8, 9).

Multiple myeloma (MM) is an incurable hematologic malignancy characterized by expansion of abnormal plasma cells (PCs) caused by multistep genetic and bone marrow (BM) microenvironmental changes (10, 11). PD-L1 expression levels on PCs from patients with MM are markedly upregulated compared with cells from monoclonal gammopathy of undetermined significance (MGUS) patients and healthy volunteers (12, 13). On the other hand, high levels of PD-1 are expressed on CD4+ and CD8+ T-cell subsets from the peripheral blood (PB) and BM of patients with MM (14, 15). The PD-1/PD-L1 interaction in MM attenuates the production of MM-specific cytotoxic T lymphocytes (12). Furthermore, our previous study showed that PD-L1 on MM cells is associated with aggressive myeloma behaviors including cell proliferation capacity and drug resistance, and the interaction of PD-L1 with PD-1 molecules induces drug resistance in MM cells by antiapoptotic responses through the Akt signaling pathway (12, 16). Thus, PD-L1 on MM cells is associated with not only immune dysfunction in the BM microenvironment but also disease progression in patients with MM.

Recently, the availability of immunomodulatory drugs (IMiDs), such as thalidomide (THAL), lenalidomide (LEN), and pomalidomide (POM), has dramatically prolonged the overall survival rates of MM patients. IMiDs directly bind to cereblon (CRBN), which is part of an E3–ubiquitin ligase complex, and lead to the ubiquitination and subsequent degradation of Ikaros (encoded by Ikaros family zinc finger protein 1; IKZF1) and Aiolos (encoded by IKZF3; refs. 17–20). Ikaros and Aiolos are the key regulators of both transcriptional factors and repressors in hematopoietic cells, and their degradation mediated by IMiDs leads to anti-MM effects through downregulation of interferon regulatory factor 4 (IRF4) and c-Myc (19, 21). However, many patients with MM develop IMiD resistance over time, necessitating long-term survival to overcome it. Hence, it is crucial to elucidate the mechanism of this resistance.

In this study, we investigated the regulation of PD-L1 expression by IMiDs and antimyeloma effects of the anti–PD-L1 antibody durvalumab in combination with IMiDs.

Human MM cell lines

Human MM cell lines were cultured in RPMI-1640 medium (Wako Chemical Industries) containing 10% fetal bovine serum, 100 U/mL of penicillin, and 100 mg/mL of streptomycin (Thermo Fisher Scientific) at 37°C under 5% CO2. KMS-18, KMS-27, and KMS-28PE cell lines were kindly provided by Dr Takemi Otsuki in 2004 (Kawasaki Medical School, Okayama, Japan). U266, RPIM 8226 (obtained in 2005), and MM.1S cells (obtained in 2010) were obtained from the American Type Culture Collection, and OPM2 cells were purchased from DSMZ in 2010. The human MM cell line MOSTI-1 was established using BM samples from LEN/bortezomib double-refractory MM patients in our laboratory (obtained in 2012; ref. 16). Cell characteristics and phenotypes of these MM cell lines are shown in Supplementary Table S1. PD-L1–transfected KMS28-PE (PD-L1.KMS-28PE) cells were established, as described previously (12). Mycoplasma was tested and was negative in all cells using the e-Myco Mycoplasma PCR Detection Kit (iNtRON Biotechnology). These cell lines were not examined by short tandem repeat analysis. The cells were cultured to passages 3 to 5, frozen in the cell-freezing medium TC-protector (DS Pharma Biomedical), and used within 20 passages in all experiments.

Human samples

BM samples were obtained from patients with newly diagnosed MM and MGUS for diagnostic purposes at Nippon Medical School after informed written consent was obtained according to the Institutional Review Board–approved protocol (Supplementary Table S2). The diagnoses were made according to International Myeloma Working Group criteria (22). PB samples were obtained from healthy adult volunteers. Mononuclear cells (MNCs) were separated from BM and PB samples with Histopaque (Sigma-Aldrich) density centrifugation.

Flow cytometry analysis

Cells were suspended in 50 μL of fluorescence-activated cell sorter (FACS) buffer (PBS with 5% bovine serum albumin and 10 mmol/L sodium azide) and blocked with Human TruStain FcX (BioLegend). Then, the cells were stained with flow cytometry (FCM) antibodies for 30 minutes. Data acquisition was performed in an LSRFortess X-20 flow cytometer with FACSDiva software version 8.0.1 and analyzed using FlowJo software (BD Biosciences). PCs were identified as CD138-positive and CD38-strong positive cells. Monocytes and lymphocytes were gated using the CD45-gating method, and the gated cells were confirmed by CD14 and CD3 positivity, respectively.

mRNA expression analysis

Total RNA was extracted from CD138+ PCs isolated from BM MNCs of MM patients and MM cell lines. The cDNA from total RNA was synthesized, and then quantification of mRNA using real-time PCR was performed (12, 16). Primer sequences are shown in Supplementary Table S3.

Western blotting

Cells were lysed using RIPA lysis buffer (Merck Millipore) plus protease inhibitor and phosphatase inhibitor (Roche Diagnostics). Twenty micrograms of protein was electrophoresed on an SDS–12.5% polyacrylamide gel and then transferred to a PVDF membrane. The target protein was detected using the ECL Prime Western Blotting Detection System (GE Healthcare) combined with LuminoGraph I (ATTO Corporation). The chemiluminescence of targeted protein was quantified using ImageJ version 1.50e software (NIH).

Apoptosis and cell viability assay

MM cells were exposed to LEN and POM for 3 days and then the cells were stained with annexin-FITC (BioLegend) and propidium iodide (PI; Wako Chemical Industries) for the analysis using FCM (23). For assessing drug sensitivity to LEN and POM, 2 × 104 MM cells in 100 μL of complete medium were seeded in 96-well plates and then treated with LEN or POM (0–500 μmol/L) for 3 days. Following exposure to IMiDs, cell viability was determined in the 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium monosodium salt (WST-8) assay using the Cell Counting Kit-8 (CCK-8; Dojindo; ref. 16). The IC50 is the concentration of drug that inhibits cell proliferation by 50% of that of the untreated controls.

shRNA-mediated knockdown and overexpression

We used the lentiviral short-hairpin RNA (shRNA) expression vector pRSI9-U6-(sh)-UbiC-TagRFP-2A-Puro vector (Addgene) for knockdown experiments and the lentiviral expression vector pLV-EF1a-IRES-Hygro (Addgene) for overexpression experiments. The targeted sequences of the shRNAs are shown in Supplementary Table S3. Ikaros cDNA fragments (isoform 1) were amplified from human MM cell line–derived cDNA using the primer shown in Supplementary Table S3. shRNA DNA fragments and Ikaros cDNA were inserted into each vector. These plasmids and ViraPower Lentiviral packaging mix (Thermo Fisher Scientific) were cotransfected into 293FT cells. The supernatant containing lentivirus 2 days posttransfection was harvested, and subsequently the MM cells were transduced with lentiviral supernatant. To establish stable cell lines, the cells were cultured with complete medium containing 1 μg/mL of puromycin (Sigma-Aldrich) or 200 μg/mL of hygromycin (Wako Chemical Industries) 48 hours after transduction. The empty vectors were used as the controls.

Xenograft mouse model

Seven-week-old NOD/Shi-scid, IL2RγKOJic (NOG) female mice (CLEA Japan Inc.) were used for the in vivo assays. NOG mice were inoculated subcutaneously in the right flank with 2 × 106 MM.1S cells in 100 μL of PBS. Tumor sizes were measured with calipers every 3 to 4 days, and the volumes were calculated using the formula α2 × β × 0.5, where α is the shortest diameter, and β is the diameter perpendicular to α. After the tumor volume reached 1,000 mm3, mice were administered 25 mg/kg of LEN orally daily for 12 days. The animal study protocols were approved by the Institutional Animal Ethical Committee of Nippon Medical School.

Statistical analysis

The Student t and Mann–Whitney U tests were used to analyze the data. P values of less than 0.05 were considered to represent statistically significant differences. Statistical analyses were performed using the GraphPad Prism 8 (GraphPad Software) and SPSS version 23 software (SPSS. Inc.).

Reagents and other conventional techniques are described in the Supplementary Information.

Upregulation of PD-L1 expression in MM cells by LEN and POM

We first assessed the impact of IMiDs on cell-surface PD-L1 expression in MM cells after their coculture. Of the IMiDs, LEN and POM, but not THAL, enhanced cell-surface expression of PD-L1 in MOSTI-1 cells in a concentration-dependent manner (Fig. 1A). Its induction by LEN and POM in MOSTI-1 cells peaked at 3–4 days (Fig. 1B). In plasma from patients who received 25 mg LEN or 4 mg POM, the maximum concentration (Cmax) was approximately 2 and 0.2 μmol/L, respectively, and the Cmax of POM was reported to be 10-fold higher than that of LEN (24). Thus, we set the concentration of LEN and POM at 10 and 1 μmol/L, respectively, and the treatment period at 3 days for the in vitro assay. In addition to MOSTI-1 cells, the mRNA expression of PD-L1 in U266, MM1.S, KMS-18, and KMS-27 cells was also upregulated by POM (Fig. 1C). The cytotoxicity of these MM cell lines was hardly affected by POM, except for RPMI 8226 cells (Fig. 1D). POM strongly promoted the apoptosis of RPMI 8226 cells, although their PD-L1 expression was not increased by POM (Fig. 1C and D). MOSTI-1, U266, MM1.S, KMS-18, and KMS-27 cell lines showed lower sensitivity to LEN and POM than RPMI 8226 cells, and the IC50 values for POM were 322, 252, 250, 217, 224, and 39 μmol/L, respectively (Supplementary Fig. S1). On the other hand, the 50% inhibitory activity of LEN was not reached in MOSTI-1, U266, MM.1S, KMS-18, and KMS-27 cell lines (Supplementary Fig. S1). The PD-L1 induction levels in these MM cell lines were significantly correlated with the IC50 values for POM (r = 0.813, P = 0.0491; Fig. 1E). Furthermore, CRBN expression in MOSTI-1, U266, KMS-18, and KMS-27 cells was lower than in MM.1S and RPMI 8226 cells, and IC50 values for POM tended to be negatively correlated with CRBN expression levels (r = −0.887, P = 0.0147; Supplementary Fig. S2). Although PD-L1 induction levels by POM in MOSTI-1 cells were the same as in the other MM cell lines, the level of PD-L1 expression in MOSTI-1 was markedly higher than in other MM cell lines because PD-L1 gene amplification caused cell-surface overexpression in MOSTI-1 cells (Fig. 1F; Supplementary Fig. S3). Moreover, the expression of PD-L2, which is located in close proximity to the PD-L1 gene locus, was scarcely induced by LEN and POM (Supplementary Fig. S4).

Figure 1.

PD-L1 upregulation in MM cell lines and primary PCs from patients with MM by LEN and POM. A, Expression of cell-surface PD-L1 on MOSTI-1 cells as shown by FCM analysis after 3-day cultivation with thalidomide (THAL), lenalidomide (LEN), and pomalidomide (POM). Relative mean fluorescence intensity (MFI) is the ratio between the MFI of antibody staining and that of control IgG staining. Data represent the fold difference of relative MFI compared with the data for untreated cells. B, Time course of cell-surface PD-L1 expression on MOSTI-1 and U266 cells in response to 10 μmol/L LEN or 1 μmol/L POM. PD-L1 mRNA expression (C) and the percentage of apoptotic cells (D) after 3-day cultivation with POM in MOSTI-1, U266, MM1.S, KMS-18, KMS-27, and RPMI 8226 cells. E, Correlation between PD-L1 induction by POM and IC50 of POM in these MM cells. The x-axis represents the fold change of gene expression at 1 μmol/L of POM and the y-axis the IC50 of POM shown in Supplementary Fig. S1B. F, Histogram of PD-L1 surface expression on the MM cell lines after 3-day cultivation with 1 μmol/L of POM. G, Fluorescence density plots of gated CD38+CD138+ PCs, CD14+ monocytes, and CD3+ T cells showing PD-L1 expression when BM NMCs isolated from MM and MGUS patients were cultured with 10 μmol/L LEN or 1 μmol/L POM for 3 days. H, Cell-surface PD-L1 expression quantified on gated populations of PCs, monocytes, and lymphocytes from patients with MM (n = 5) after 3-day cultivation with LEN or POM. Data represent relative MFI, which is the ratio of the MFI of antibody staining to MFI of control IgG staining. N.S., not significant. Data in A–E are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Figure 1.

PD-L1 upregulation in MM cell lines and primary PCs from patients with MM by LEN and POM. A, Expression of cell-surface PD-L1 on MOSTI-1 cells as shown by FCM analysis after 3-day cultivation with thalidomide (THAL), lenalidomide (LEN), and pomalidomide (POM). Relative mean fluorescence intensity (MFI) is the ratio between the MFI of antibody staining and that of control IgG staining. Data represent the fold difference of relative MFI compared with the data for untreated cells. B, Time course of cell-surface PD-L1 expression on MOSTI-1 and U266 cells in response to 10 μmol/L LEN or 1 μmol/L POM. PD-L1 mRNA expression (C) and the percentage of apoptotic cells (D) after 3-day cultivation with POM in MOSTI-1, U266, MM1.S, KMS-18, KMS-27, and RPMI 8226 cells. E, Correlation between PD-L1 induction by POM and IC50 of POM in these MM cells. The x-axis represents the fold change of gene expression at 1 μmol/L of POM and the y-axis the IC50 of POM shown in Supplementary Fig. S1B. F, Histogram of PD-L1 surface expression on the MM cell lines after 3-day cultivation with 1 μmol/L of POM. G, Fluorescence density plots of gated CD38+CD138+ PCs, CD14+ monocytes, and CD3+ T cells showing PD-L1 expression when BM NMCs isolated from MM and MGUS patients were cultured with 10 μmol/L LEN or 1 μmol/L POM for 3 days. H, Cell-surface PD-L1 expression quantified on gated populations of PCs, monocytes, and lymphocytes from patients with MM (n = 5) after 3-day cultivation with LEN or POM. Data represent relative MFI, which is the ratio of the MFI of antibody staining to MFI of control IgG staining. N.S., not significant. Data in A–E are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Close modal

We next investigated whether PD-L1 expression on primary PCs from five patients newly diagnosed with MM who had not received chemotherapy, was also induced by LEN and POM. The percentage of CD138+CD38+ PCs in BM MNCs from MM patients was 24.34% ± 14.8% [mean ± standard deviation (SD); range, 10.8%–49.2%] in this study (Supplementary Table S2). CD138+CD38+ PCs and monocytes expressed high levels of PD-L1, but lymphocytes did not (Fig. 1G). Its expression on PCs, but not on monocytes and lymphocytes, obtained from patients with MM was significantly enhanced by LEN and POM (Fig. 1G and H). On the other hand, PD-L1 expression on PCs from patient with MGUS was low and not upregulated by LEN or POM (Fig. 1G).

PD-L1 induction by LEN and POM via the APRIL–BCMA pathway

Next, to clarify the mechanism of PD-L1 induction by LEN and POM, we performed gene and microRNA (miRNA) expression profiling analysis in the MM cell lines MOSTI-1 and U266, with and without POM coculture. In gene-expression microarray analysis, a total of 404 genes with differential expression showing more than a twofold difference after coculture with POM in both MOSTI-1 and U266 cells (366 upregulated and 38 downregulated genes) were identified, as described in Supplementary Table S4. The 366 upregulated genes were defined in seven categories by gene ontology (GO) molecular function enrichment analysis (Fig. 2A). Because both U266 and MOSTI-1 cells were HLA-DR–positive cell lines, MHC class II–antigen-presentation pathway-related genes were upregulated by POM treatment (Fig. 2A). Furthermore, the category of receptor ligand activity contained 17 genes, among which we focused on APRIL [also known as tumor necrosis factor ligand superfamily member 13 (TNFSF13)], which is related to MM tumor growth (Fig. 2B). On the other hand, we could not identify the miRNA associated with PD-L1 upregulation by LEN and POM by human miRNA array.

Figure 2.

PD-L1 induction by LEN and POM via the APRIL–BCMA pathway. A, 366 upregulated genes (fold change > 2.0; Supplementary Table S4) were analyzed using GO molecular function enrichment analysis. B, Heatmapping of 17 genes including “receptor ligand activity” of A (red bar). C, APRIL mRNA expression after 3-day cultivation with POM in MOSTI-1, U266, MM1.S, KMS-18, KMS-27, and RPMI 8226 cells. mRNA (D) and protein (E) expression of PD-L1 in MOSTI-1 cells after 3-day cultivation with 10 μmol/L LEN, 1 μmol/L POM, or 200 ng/mL recombinant human (rh) APRIL. F, mRNA expression of APRIL in primary CD38+CD138+ PCs from patients with MM. MOSTI-1 cells were cultured with 1 μmol/L POM and TACI-Ig or BCMA-Ig (G), and anti-TACI or anti-BCMA antibody (H) for 3 days. Then PD-L1 expression was analyzed using FCM. I, MOSTI-1 cells treated with the following signal transduction inhibitors were cultured with 1 μmol/L POM and 20 μmol/L U0126 (MEK1/2 inhibitor), 20 μmol/L LY294002 (PI3K/AKT inhibitor), 500 nmol/L STAT3 inhibitor V, 25 μmol/L AG490 (JAK2 inhibitor), or 50 μmol/L PDTC (NF-κB inhibitor). Data represent the fold difference of relative MFI compared with the data for untreated cells. Data in C–I are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Figure 2.

PD-L1 induction by LEN and POM via the APRIL–BCMA pathway. A, 366 upregulated genes (fold change > 2.0; Supplementary Table S4) were analyzed using GO molecular function enrichment analysis. B, Heatmapping of 17 genes including “receptor ligand activity” of A (red bar). C, APRIL mRNA expression after 3-day cultivation with POM in MOSTI-1, U266, MM1.S, KMS-18, KMS-27, and RPMI 8226 cells. mRNA (D) and protein (E) expression of PD-L1 in MOSTI-1 cells after 3-day cultivation with 10 μmol/L LEN, 1 μmol/L POM, or 200 ng/mL recombinant human (rh) APRIL. F, mRNA expression of APRIL in primary CD38+CD138+ PCs from patients with MM. MOSTI-1 cells were cultured with 1 μmol/L POM and TACI-Ig or BCMA-Ig (G), and anti-TACI or anti-BCMA antibody (H) for 3 days. Then PD-L1 expression was analyzed using FCM. I, MOSTI-1 cells treated with the following signal transduction inhibitors were cultured with 1 μmol/L POM and 20 μmol/L U0126 (MEK1/2 inhibitor), 20 μmol/L LY294002 (PI3K/AKT inhibitor), 500 nmol/L STAT3 inhibitor V, 25 μmol/L AG490 (JAK2 inhibitor), or 50 μmol/L PDTC (NF-κB inhibitor). Data represent the fold difference of relative MFI compared with the data for untreated cells. Data in C–I are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Close modal

APRIL is a protein of the tumor necrosis factor (TNF) superfamily recognized by the cell-surface receptor transmembrane activator and CAML interactor (TACI) and B-cell maturation antigen (BCMA; ref. 25). The gene expression of APRIL showed 2.3-fold and 5.2-fold changes with POM treatment in U266 and MOSTI-1 cells, respectively (Supplementary Table S4). We confirmed that the expression of mRNA was increased in MOSTI-1, U266, and MM1.S cells cocultured with POM, but not increased in KMS-18, KMS-27, and RPMI 8226 cells (Fig. 2C). The cell-surface expression of BCMA and TACI was not changed after POM coculture (Supplementary Fig. S5A and S5B). When MM cell lines and primary PCs from newly diagnosed MM patients were cultured with recombinant human APRIL for 3 days, PD-L1 was induced by APRIL as well as by LEN and POM (Fig. 2D–F). Its induction on MOSTI-1 cells was suppressed by both TACI-Ig and BCMA-Ig (Fig. 2G), by the antibody against BCMA but not that against TACI (Fig. 2H) and, furthermore, by the MEK/ERK inhibitor U0126 (Fig. 2I). Gene set enrichment analysis (GSEA) showed that the MAPK/ERK signal cascade was significantly upregulated in MM cells treated with POM (Supplementary Fig. S6).

To confirm the in vitro assay data, we treated MM.1S xenograft‐bearing NOG mice with 25 mg/kg LEN orally daily for 12 days (Fig. 3A). The cell-surface PD-L1 in MM.1S cells was increased in a concentration-dependent manner by POM in vitro (Fig. 1C). The mRNA and cell-surface PD-L1 expression levels in MM.1S cells engrafted in NOG mice were significantly upregulated by LEN treatment in comparison with controls (Fig. 3B and C). Similarly, the APRIL expression level in LEN-treated mice was markedly higher than that in controls (Fig. 3D and E). Next, while receiving LEN, MM.1S xenograft-bearing NOG mice were administered 100 μg anti-BCMA antibody intraperitoneally every 4 days (Fig. 3F). The upregulation of PD-L1 in MM.1S cells in NOG mice treated with LEN was suppressed by anti-BCMA antibody (Fig. 3G). However, the tumor size did not differ among the three treatment groups (Supplementary Fig. S7). The results indicated that the PD-L1 induction in MM cells by LEN treatment might be associated with the APRIL–BCMA pathway.

Figure 3.

PD-L1 induction by LEN in the in vivo assay. A, NOG mice were inoculated subcutaneously (s.c.) with MM.1S cells and then received 25 mg/kg LEN orally (p.o.) daily for 12 days after tumor volume reached 1,000 mm3. mRNA (B) and cell-surface (C) expression of PD-L1 in MM.1S cells engrafted in LEN-treated NOG mice. Right: histogram plot of PD-L1 expression in MM.1S cells analyzed using FCM; left: percentages of PD-L1–expressing MM.1S cells (filled area, staining with PE-conjugated antibody to PD-L1; solid line, staining with isotype-matched control Ig). mRNA (D) and protein (E) expression of APRIL in MM.1S cells engrafted in LEN-treated NOG mice. Numerals above the bands of APRIL indicate the relative intensity of each protein normalized to the signal intensity of β-actin. The control group received oral vehicle (20% DMSO). F, Mice treated with LEN were intraperitoneally (i.p.) administered 100-μg anti-BCMA antibody every 4 days. G, Cell-surface expression of PD-L1 in MM.1S cells engrafted in LEN- and LEN + anti-BCMA–treated NOG mice. Right: histogram plot of PD-L1 expression in MM.1S cells analyzed using FCM; left: percentages of PD-L1–expressing MM.1S cells.

Figure 3.

PD-L1 induction by LEN in the in vivo assay. A, NOG mice were inoculated subcutaneously (s.c.) with MM.1S cells and then received 25 mg/kg LEN orally (p.o.) daily for 12 days after tumor volume reached 1,000 mm3. mRNA (B) and cell-surface (C) expression of PD-L1 in MM.1S cells engrafted in LEN-treated NOG mice. Right: histogram plot of PD-L1 expression in MM.1S cells analyzed using FCM; left: percentages of PD-L1–expressing MM.1S cells (filled area, staining with PE-conjugated antibody to PD-L1; solid line, staining with isotype-matched control Ig). mRNA (D) and protein (E) expression of APRIL in MM.1S cells engrafted in LEN-treated NOG mice. Numerals above the bands of APRIL indicate the relative intensity of each protein normalized to the signal intensity of β-actin. The control group received oral vehicle (20% DMSO). F, Mice treated with LEN were intraperitoneally (i.p.) administered 100-μg anti-BCMA antibody every 4 days. G, Cell-surface expression of PD-L1 in MM.1S cells engrafted in LEN- and LEN + anti-BCMA–treated NOG mice. Right: histogram plot of PD-L1 expression in MM.1S cells analyzed using FCM; left: percentages of PD-L1–expressing MM.1S cells.

Close modal

PD-L1 induction by LEN and POM via Ikaros degradation through cereblon

In some MM cell lines, APRIL was induced by POM, while in others it was not (Fig. 2C). Furthermore, PD-L1 induction by POM was not completely suppressed by the APRIL inhibitors and antibody against the APRIL receptors (Fig. 2G and H). Thus, we attempted to confirm whether PD-L1 induction by LEN/POM was directly associated with the degradation of Ikaros/Aiolos meditated via cereblon (CRBN; Fig. 4A). We constructed two pGL3-promoter vectors carrying the PD-L1 promoter regions, −353 to +1 and −1,013 to +1, and investigated their activity using a luciferase reporter assay in APRIL- and BCMA-negative HeLa cells. The luciferase activity of the −353-bp promoter induced was 2.2 ± 0.6- and 3.5 ± 0.7-fold higher following LEN and POM treatment, respectively, compared with that in untreated controls (Fig. 4B). Similarly, the highly inducible activity of the −1,013-bp promoter was conserved in response to LEN (2.6 ± 0.4-fold change) and POM (3.3 ± 0.5-fold change; Fig. 4B). Next, to clarify the participation of CRBN, Ikaros, and Aiolos, we performed the knockdown of these proteins in MM cells. Upregulation of PD-L1 by LEN and POM was not seen in stable CRBN knockdown MOSTI-1 cells (Fig. 4C). On the other hand, the knockdown of Ikaros increased expression levels of PD-L1 protein and mRNA in MOSTI-1 cells, and PD-L1 upregulation was markedly enhanced by LEN and POM (Fig. 4D). PD-L1 expression was conversely downregulated in Ikaros-overexpressing MOSTI-1 cells (Fig. 4E). However, the expression of PD-L1 protein in Aiolos-knockdown cells after culture with LEN or POM was the same as in controls, although the induction levels of mRNA expression in knockdown cells were lower than in controls (Fig. 4F). In other MM cell lines, CRBN-knockout MM cells decreased PD-L1 induction, whereas Ikaros-knockdown MM cells increased PD-L1 expression (Supplementary Fig. S8). Analysis of Ikaros ChIPseq data from the human GM12878 lymphoblastoid cell line demonstrated one Ikaros-binding peak in the PD-L1 promoter region, suggesting that Ikaros might directly bind to the upstream promoter region (−1 to −353; Supplementary Fig. S9A). To confirm this, we constructed a pGL3-promoter vector containing a nonbinding mutation at the Ikaros-binding site of the −353-bp promoter region for the luciferase reporter assay. The promoter activity of the mutant type was upregulated in comparison with the wild-type in the absence of LEN and POM (Fig. 4G). Therefore, Ikaros might function as a transcriptional repressor of the PD-L1 gene, and PD-L1 might be directly induced by LEN and POM via CRBN-mediated Ikaros degradation as well as via the APRIL–BCMA pathway.

Figure 4.

PD-L1 induction by LEN and POM via Ikaros degradation through CRBN. A, Ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via E3 ubiquitin ligase complex including cereblon (CRBN) by binding with IMiDs. In the absence of LEN and POM, Ikaros/Aiolos binds to CRBN and then is modified with ubiquitination, causing it to be degraded by the 26S proteasome. It is hypothesized that Ikaros or Aiolos suppresses the transcription of the PD-L1 gene. B, Promoter constructs of the PD-L1 promoter region and PD-L1 promoter activity. Activation of the PD-L1 promoter was determined by luciferase activity in HeLa cells transiently cotransfected with pGL3 vector containing PD-L1 promoter regions and Renilla luciferase plasmid (pRL) as the control vector. Data are presented as fold changes compared with the activity of each type of untreated cell. Luminescence of the pGL3 promoter vector was normalized to the Renilla luciferase luminescence from the pRL control vector. After treatment with 10 μmol/L LEN or 1 μmol/L POM for 3 days, expression levels of PD-L1 in protein (upper photographs) and mRNA (lower graphs) in stable CRBN knockdown MOSTI-1 cells (CRBN.sh; C), transient Ikaros-knockdown MOSTI-1 cells (Ikaros.sh; D) and transient Aiolos-knockdown MOSTI-1 cells (Aiolos.sh; F) were analyzed by Western blot analysis and real-time PCR, respectively. MOSTI-1 cells transfected with empty vector were used as controls (Ctl.sh). E, PD-L1 expression in Ikaros-overexpressing MOSTI-1 cells was detected by Western blotting. MOSTI-1 cells transfected with empty pLV-EF1a-IRES-Hygro vector were used as controls. G, Promoter assay for wild-type (WT) and mutant of the Ikaros family (IKZF) binding sites in the PD-L1 gene. Data in B–F and G are expressed as mean ± standard deviation of duplicate experiments. *, P < 0.05; **, P < 0.01.

Figure 4.

PD-L1 induction by LEN and POM via Ikaros degradation through CRBN. A, Ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via E3 ubiquitin ligase complex including cereblon (CRBN) by binding with IMiDs. In the absence of LEN and POM, Ikaros/Aiolos binds to CRBN and then is modified with ubiquitination, causing it to be degraded by the 26S proteasome. It is hypothesized that Ikaros or Aiolos suppresses the transcription of the PD-L1 gene. B, Promoter constructs of the PD-L1 promoter region and PD-L1 promoter activity. Activation of the PD-L1 promoter was determined by luciferase activity in HeLa cells transiently cotransfected with pGL3 vector containing PD-L1 promoter regions and Renilla luciferase plasmid (pRL) as the control vector. Data are presented as fold changes compared with the activity of each type of untreated cell. Luminescence of the pGL3 promoter vector was normalized to the Renilla luciferase luminescence from the pRL control vector. After treatment with 10 μmol/L LEN or 1 μmol/L POM for 3 days, expression levels of PD-L1 in protein (upper photographs) and mRNA (lower graphs) in stable CRBN knockdown MOSTI-1 cells (CRBN.sh; C), transient Ikaros-knockdown MOSTI-1 cells (Ikaros.sh; D) and transient Aiolos-knockdown MOSTI-1 cells (Aiolos.sh; F) were analyzed by Western blot analysis and real-time PCR, respectively. MOSTI-1 cells transfected with empty vector were used as controls (Ctl.sh). E, PD-L1 expression in Ikaros-overexpressing MOSTI-1 cells was detected by Western blotting. MOSTI-1 cells transfected with empty pLV-EF1a-IRES-Hygro vector were used as controls. G, Promoter assay for wild-type (WT) and mutant of the Ikaros family (IKZF) binding sites in the PD-L1 gene. Data in B–F and G are expressed as mean ± standard deviation of duplicate experiments. *, P < 0.05; **, P < 0.01.

Close modal

Durvalumab recovers inhibition of T-cell proliferation by PD-L1 upregulation on MM cells induced by LEN and POM

When untreated or POM-pretreated U266 sells were cocultured with T cells, we ascertained the impact of each cell type on T-cell activity. POM-pretreated U266 cells significantly suppressed T-cell proliferation and IFNγ production from T cells as compared with untreated U266 cells at the effector:target (E:T) ratios of 1:0.25 and 1:0.5 (Fig. 5A and B). Therefore, PD-L1 upregulation on MM cells robustly induced T-cell suppression. However, untreated U266 cells also showed strongly suppressed T-cell activation identical to POM-treated U266 cells at the E:T ratio of 1:1, and therefore the difference in immune suppression between the two cell types was difficult to determine. In this study, it appeared that T-cell suppression may be associated with cell–cell contact signaling such as the PD-1–PD-L1 pathway but not with other mechanisms because immunosuppressive soluble factors were not generated from irradiated MM cells.

Figure 5.

T-cell inhibition by cocultivation with POM-pretreated U266 cells was recovered after combination treatment with durvalumab (DUR) and POM. Cell proliferation of CD8+ T cells (A) and IFNγ production from T cells (B) under varying effector:tumor cell (E:T) ratios after cocultivation with untreated or POM-pretreated U266 (U266 and U266POM, respectively). Cell proliferation was determined by measuring dilution of CFSE using FCM. C, T-cell assay in combination treatment with 20 μg/mL durvalumab and 1 μmol/L POM. After U266 cells were pretreated with POM for 4 days (U266POM), CFSE-labeled CD3+ T cells and U266POM at a 1:0.5 ratio were cocultured with durvalumab in the presence or absence of POM. T-cell activity was assessed by T-cell proliferation of CD8+ T cells (D) and IFNγ production from T cells (E) on days 4 and 1, respectively. Representative CFSE histograms of CD8+ T cells are shown in Fig. 2D (left). The bar graph in Fig. 2D (right) shows the percentage of cells divided into CD8+ T cells. F, Representative flow plots of CD45RO and CD25 expression, which are activation markers in CD8+ T cells (left). The right graph shows the percentage of double-positive CD45RO and CD25 in CD8+ T cells. Data are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Figure 5.

T-cell inhibition by cocultivation with POM-pretreated U266 cells was recovered after combination treatment with durvalumab (DUR) and POM. Cell proliferation of CD8+ T cells (A) and IFNγ production from T cells (B) under varying effector:tumor cell (E:T) ratios after cocultivation with untreated or POM-pretreated U266 (U266 and U266POM, respectively). Cell proliferation was determined by measuring dilution of CFSE using FCM. C, T-cell assay in combination treatment with 20 μg/mL durvalumab and 1 μmol/L POM. After U266 cells were pretreated with POM for 4 days (U266POM), CFSE-labeled CD3+ T cells and U266POM at a 1:0.5 ratio were cocultured with durvalumab in the presence or absence of POM. T-cell activity was assessed by T-cell proliferation of CD8+ T cells (D) and IFNγ production from T cells (E) on days 4 and 1, respectively. Representative CFSE histograms of CD8+ T cells are shown in Fig. 2D (left). The bar graph in Fig. 2D (right) shows the percentage of cells divided into CD8+ T cells. F, Representative flow plots of CD45RO and CD25 expression, which are activation markers in CD8+ T cells (left). The right graph shows the percentage of double-positive CD45RO and CD25 in CD8+ T cells. Data are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01.

Close modal

We next questioned whether PD-L1 upregulation on MM cells by LEN and POM could inhibit T-cell immune responses and whether durvalumab (MED14736), a human immunoglobulin G1 (IgG1) kappa monoclonal antibody that binds to PD-L1 (26), could induce T-cell activation in combination with LEN or POM. To resolve these questions, we examined T-cell activation after cocultivating POM-pretreated MM cells in the presence of durvalumab and POM (Fig. 5C). When T cells were cocultured with POM-pretreated U266 cells, the cell proliferation of CD8+ T cells was inhibited, whereas this T-cell suppression was not prevented by durvalumab alone (Fig. 5D). However, its suppression was recovered with the combination of durvalumab and POM during cocultivation of POM-treated U266 cells with T cells (Fig. 5D). Furthermore, the IFNγ production from T cells and population of both CD25+CD45RO+ double-positive CD8+ T cells (activated T cells) cocultured with POM-treated U266 cells were significantly increased after combined POM and durvalumab treatment (Fig. 5E and F). Similarly, the inhibition of CD8+ T-cell proliferation by cocultivation with LEN-treated U266 cells was disarmed by the combination of durvalumab with LEN (Supplementary Fig. S10).

Durvalumab combined with POM and bortezomib induces MM cell apoptosis by blockade of PD-1−PD-L1 signaling

Previously, we demonstrated that the binding of PD-1 with PD-L1 can induce drug resistance through Akt activation in MM cells (16). Thus, we investigated whether the ligation of PD-1-Fc with PD-L1 upregulated by IMiDs could increase phosphorylation of Akt in MM cells. The amount of PD-1-Fc bound was increased by POM, although the binding was completely blocked by durvalumab (Fig. 6A). The binding of PD-1-Fc to PD-L1 on MM cells decreased the percentage of apoptotic cells induced by bortezomib, similar to our previous findings, although durvalumab overcame the drug resistance induced by PD-1−PD-L1 signaling (Fig. 6B). Expression levels of Akt phosphorylation did not increase with PD-L1 upregulation (Fig. 6C). Consistent with those results, the number of bortezomib-induced apoptotic cells did not decrease when POM-treated MM cells, which had increased PD-L1 expression, were cultured with PD-1-Fc. However, the POM/bortezomib combination increased the number of apoptotic cells in the presence of durvalumab through the blockade of PD-1−PD-L1 signaling (Fig. 6D).

Figure 6.

Blocking effect of durvalumab (DUR) on anti-MM agent resistance induced by PD-1−PD-L1 signaling. A, Binding of PD-L1–expressing MM cells with PD-1-Fc beads was inhibited by DUR. PD-L1+ MOSTI-1 cells were incubated with/without POM in the presence/absence of DUR, and then PD-1−PD-L1 interaction was detected by FITC-conjugated PD-1-Fc using FCM. B, Percentages of apoptotic cells induced by bortezomib (BOR) were analyzed by Annexin V staining using FCM after MM cells were cultured with/without BOR in the presence/absence of PD-1Fc. C, Western blotting analysis of PD-L1, phosphorylated Akt (Phosho-Akt), Akt, and β-actin in MOSTI-1 3 days after treatment with 10 μmol/L LEN or 1 μmol/L POM. D, POM-treated/untreated MM cells were cultured with/without BOR in the presence/absence of PD-1Fc, and then apoptotic cells were analyzed by Annexin V staining using FCM. Data in A–C are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01. E, Two mechanisms of PD-L1 upregulation by IMiDs in the MM microenvironment. First, APRIL expression in MM cells is increased by IMiDs, and subsequently PD-L1 on MM cells is induced via MAP/ERK signal activation through the BCMA–APRIL interaction. Second, PD-L1 on MM cells is upregulated via Ikaros degradation through IMiD binding to CRBN. Ikaros may act as a repressor of PD-L1 expression. PD-L1–expressing MM cells not only inhibit T-cell proliferation but also induce tumor growth and drug resistance through reverse PD-1−PD-L1 signaling.

Figure 6.

Blocking effect of durvalumab (DUR) on anti-MM agent resistance induced by PD-1−PD-L1 signaling. A, Binding of PD-L1–expressing MM cells with PD-1-Fc beads was inhibited by DUR. PD-L1+ MOSTI-1 cells were incubated with/without POM in the presence/absence of DUR, and then PD-1−PD-L1 interaction was detected by FITC-conjugated PD-1-Fc using FCM. B, Percentages of apoptotic cells induced by bortezomib (BOR) were analyzed by Annexin V staining using FCM after MM cells were cultured with/without BOR in the presence/absence of PD-1Fc. C, Western blotting analysis of PD-L1, phosphorylated Akt (Phosho-Akt), Akt, and β-actin in MOSTI-1 3 days after treatment with 10 μmol/L LEN or 1 μmol/L POM. D, POM-treated/untreated MM cells were cultured with/without BOR in the presence/absence of PD-1Fc, and then apoptotic cells were analyzed by Annexin V staining using FCM. Data in A–C are expressed as mean ± standard deviation of triplicate experiments. *, P < 0.05; **, P < 0.01. E, Two mechanisms of PD-L1 upregulation by IMiDs in the MM microenvironment. First, APRIL expression in MM cells is increased by IMiDs, and subsequently PD-L1 on MM cells is induced via MAP/ERK signal activation through the BCMA–APRIL interaction. Second, PD-L1 on MM cells is upregulated via Ikaros degradation through IMiD binding to CRBN. Ikaros may act as a repressor of PD-L1 expression. PD-L1–expressing MM cells not only inhibit T-cell proliferation but also induce tumor growth and drug resistance through reverse PD-1−PD-L1 signaling.

Close modal

PD-L1 on MM cells inhibits the effector function of immune cells such as T and NK cells in patients with MM (1, 3). In the present study, we showed that PD-L1 expression on IMiD-insensitive MM cells and PCs from newly diagnosed MM patients is upregulated by LEN and POM via ERK activation induced by the APRIL–BCMA pathway and Ikaros degradation through CRBN (Fig. 6E). Furthermore, T-cell suppression in MM cells after cultivation with LEN/POM and drug resistance via PD-L1–PD-1 interaction is disarmed by durvalumab. Our findings suggest that PD-L1 upregulation by IMiDs might induce aggressive myeloma behaviors and tumoral immune escape in MM patients.

Several researchers reported that PD-L1 in the MM cell line RPMI 8226 and in CD138+ PCs in the BM from relapsed/refractory MM patients was downregulated after cultivation with LEN (27, 28). However, our results showed that LEN and POM upregulated PD-L1 on MOSTI-1, U266, MM1.S, KMS-18, and KMS-27 cells but not on RPMI 8226 cells. Iwasa and colleagues also reported that PD-L1 on the MM cell lines MM.1S, KMS-11, and OPM-2 was increased by LEN and POM (29). There was no difference in cell characteristics and phenotypes among these MM cell lines (Supplementary Table S1). In particular, the MOSTI-1 cell line was established from BM PCs with LEN/bortezomib double-refractory MM patients. Its IC50 value of IMiDs and the rate of PD-L1 induction by IMiDs were higher than those in other MM cell lines. Furthermore, the IC50 values of POM were negatively correlated with CRBN expression. Lopez-Girona and colleagues demonstrated that the proliferation of RPMI 8226 cells was strongly inhibited by LEN in comparison with other MM cell lines including U266 cells (30). They further reported that CRBN knockdown in U266 cells induced resistance to the antiproliferative effects of LEN (31). CRBN reduction decreased the antiproliferative potency of IMiDs mediated via induction of the cell-cycle progression inhibitor p21 and the suppression of IRF4, a gene critical for myeloma cell growth (31). Therefore, because CRBN expression was higher in RPMI 8226 cells than in other MM cell lines, RPMI 8226 cells might be IMiD sensitive. Thus, it is likely that the phenomenon of PD-L1 upregulation by IMiDs may be observed in IMiD-insensitive MM cells because it is difficult to induce apoptosis via IMiDs in these MM cell lines except for RPMI 8226. On the other hand, PD-L1 expression on PCs from newly diagnosed MM patients was increased by LEN and POM. Two of five patients with MM received LEN-based therapy and were refractory to Rd (LEN and dexamethasone; Supplementary Table S2). In the future, we need to analyze the relationship of PD-L1 upregulation by IMiDs with disease progression in patients with refractory MM using a large sample size.

One mechanism of its induction by IMiDs was associated with ERK signaling transmitted through the BCMA–APRIL pathway. A recent report has demonstrated that BCMA-overexpressing RPMI8226 cells implanted into severe combined immunodeficiency mice enhance not only MM tumor growth but also PD-L1 expression, and PD-L1 upregulation by recombinant APRIL on MM cell lines was suppressed by antagonistic anti-APRIL monoclonal antibody in vitro (32). Furthermore, APRIL, secreted by osteoclasts and myeloid cells, increased PD-L1 expression on MM cell lines via the BCMA receptor in the MEK/ERK pathway (33). However, our data showed that PD-L1 expression on MM cells was induced by autocrine or paracrine secretion of APRIL enhanced by LEN/POM from MM cells. Although PD-L1 enhancement by IMiDs in the in vitro and in vivo assays was inhibited by anti-BCMA antibody, APRIL was not detected in cell culture supernatants of IMiD-treated MM cells and serum of MM.1S xenograft‐bearing NOG mice treated with IMiDs. The reason for this may be that the APRIL concentration in the samples was too low or because upon release it immediately binds to BCMA on MM cells in the tumor microenvironment. The promoter activity of the APRIL gene is regulated by specificity protein 1 (Sp1) and nuclear factor kappa B (NF-κB; ref. 34). In GSEA, the NF-κB signaling pathway-related gene sets were not enriched in POM-treated MM cells. Furthermore, APRIL expression was increased in CRBN-knockdown cells, but was not upregulated by IMiDs in CRBN-, Ikaros-, and Aiolos-knockdown cells (Supplementary Fig. S11). These results suggest that APRIL induction by IMiDs might not be associated with CRBN, Ikaros, and Aiolos degradation. In this study, the APRIL gene was only upregulated by IMiDs in some MM cell lines, and therefore the APRIL–BCMA pathway may be involved in some subsets of MM cells (Fig. 2C). Thus, the mechanism of APRIL induction by IMiDs remains unclear. The BCMA–APRIL signaling pathway has been shown to promote cell growth, survival, and immunosuppression (25). Similarly, PD-L1–positive MM cells have a greater proliferative capacity and drug resistance in comparison with negative cells (12, 16). Therefore, BCMA/APRIL blockade might be effective in suppressing not only PD-L1 expression but also antimyeloma activity.

In addition to the APRIL–BCMA pathway, PD-L1 was upregulated directly by the CRBN-dependent degradation of Ikaros but not of Aiolos. PD-L1 expression was strongly induced in Ikaros-knockdown cells by IMiDs, whereas it was downregulated by Ikaros overexpression. These results suggest that Ikaros acts as a suppressor of PD-L1 gene expression. The upstream promoter region of PD-L1 (−1 to −353 bp) contains one Ikaros-binding element (AGGAA), and its site is included in the sequence of the NF-κB binding site (Supplementary Fig. S9B). PD-L1 expression is upregulated by the MyD88 and TRAF6 adaptor proteins stimulated by Toll-like receptor ligands, and its signaling activates transcranial factor NF-κB (13, 35). Ikaros may inhibit the binding of NF-κB to the promoter of PD-L1, and therefore the degradation of Ikaros mediated by IMiDs disarms its PD-L1 suppression. Our data suggest that PD-L1 upregulation by IMiDs on IMiD-insensitive MM cells might promote immune escape in the BM microenvironment even if IMiDs augment both the adaptive and innate immune systems.

PD-1/PD-L1 blockade alone dramatically improves the status of some patients with non–small cell lung, prostate and renal cell cancer, and melanoma (36, 37). Anti–PD-1 antibodies such as nivolumab and pembrolizumab bind to PD-1–expressing T and natural killer cells and reverse their dysfunction (38). On the other hand, anti–PD-L1 antibodies including durvalumab, avelumab, and atezolizumab bind to several types of tumor cells, dendritic cells, and myeloid-derived suppressor cells expressing PD-L1 and reverse the inhibition of T-cell proliferation (38). De Sousa Linhares and colleagues reported that the functional and binding assay results after therapeutic PD-L1 antibody administration are superior to those with PD-1 antibodies (39). In clinical trials of anti–PD-1 inhibitors in MM, although almost no MM patients showed a response to treatment with nivolumab alone (NCT01592370; ref. 40), the combination of pembrolizumab with IMiDs and dexamethasone yielded a more than 50% objective response rate including complete remission, very good partial response, and partial response in patients with relapsed/refractory MM (NCT02036502 and NCT02289222; refs. 41, 42). Clinical trials of durvalumab in combination with POM and dexamethasone in relapsed/refractory MM are in progress (NCT02616640 and NCT02807454; ref. 43). In our study, T-cell suppression induced by PD-L1 upregulation was recovered after coculture with durvalumab combined with IMiDs but not after that with durvalumab alone. LEN enhances the PD-1/PD-L1 blockade-induced anti-MM immune response in comparison with blockade alone (27). These data suggest that IMiDs may be necessary for the functional recovery of tumoral T-cell activation because their dysfunction is induced by not only the PD-1/PD-L1 pathway but also by other mechanisms.

Our study demonstrated that IMiDs enhance PD-L1 expression on MM cells as well as APRIL production, and one mechanism may involve the APRIL–BCMA interaction (Fig. 6E). Another mechanism may be directly involved in PD-L1 upregulation by Ikaros degradation through CRBN, resulting in AKT signaling via reverse PD-1−PD-L1 signaling (Fig. 6E). Interestingly, these results suggested that the induction of PD-L1 expression by IMiDs might be associated with disease progression in IMiD-insensitive MM patients with the acquisition of aggressive myeloma behaviors and tumor immune evasion. However, because the number of patients with MM was small in this study, in the future, we must investigate PD-L1 expression on PCs from patients with MM before and after IMiD-based treatment, or PD-L1 induction in responders and nonresponders to IMiDs. Furthermore, T-cell inhibition induced by PD-L1–upregulated MM cells was recovered after combination treatment with durvalumab and IMiDs, suggesting that this combination therapy may be a reasonable treatment for patients with relapsed/refractory MM. Our findings provide new insight into the mechanism of disease progression in patients with IMiD-insensitive MM.

J. Yamamoto reports affiliation with an endowed course from Celgene Corporation and personal fees (honoraria) from Celgene Corporation outside the submitted work. H. Handa reports receipt of an international joint research grant from Celgene/BMS, USA. H. Tamura reports grants and personal fees from Celgene during the conduct of the study; grants and personal fees from Ono Pharmaceutical Co., Ltd.; personal fees from Janssen Pharmaceutical K.K.; Takeda Pharmaceutical Co., Ltd.; and Sanofi outside the submitted work. No disclosures were reported by the other authors.

M. Ishibashi: Conceptualization, data curation, software, formal analysis, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing, analysis and interpretation of data. J. Yamamoto: Resources, methodology, analysis and interpretation of data. T. Ito: Resources, methodology, analysis and interpretation of data. H. Handa: Resources, methodology, analysis and interpretation of data. M. Sunakawa: Resources. K. Inokuchi: Resources. R. Morita: Resources, methodology, writing–review and editing. H. Tamura: Conceptualization, resources, supervision, funding acquisition, project administration, writing–review and editing, analysis and interpretation of data.

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.
Chen
DS
,
Mellman
I
. 
Oncology meets immunology: the cancer-immunity cycle
.
Immunity
2013
;
39
:
1
10
.
2.
Sharpe
AH
,
Pauken
KE
. 
The diverse functions of the PD1 inhibitory pathway
.
Nat Rev Immunol
2018
;
18
:
153
67
.
3.
Dong
H
,
Strome
SE
,
Salomao
DR
,
Tamura
H
,
Hirano
F
,
Flies
DB
, et al
Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion
.
Nat Med
2002
;
8
:
793
800
.
4.
Chen
L
,
Han
X
. 
Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future
.
J Clin Invest
2015
;
125
:
3384
91
.
5.
Chen
L
,
Flies
DB
. 
Molecular mechanisms of T cell co-stimulation and co-inhibition
.
Nat Rev Immunol
2013
;
13
:
227
42
.
6.
Iwai
Y
,
Ishida
M
,
Tanaka
Y
,
Okazaki
T
,
Honjo
T
,
Minato
N
. 
Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade
.
Proc Natl Acad Sci U S A
2002
;
99
:
12293
7
.
7.
Wang
SF
,
Fouquet
S
,
Chapon
M
,
Salmon
H
,
Regnier
F
,
Labroquere
K
, et al
Early T cell signalling is reversibly altered in PD-1+ T lymphocytes infiltrating human tumors
.
PLoS One
2011
;
6
:
e17621
.
8.
Siu
LL
,
Ivy
SP
,
Dixon
EL
,
Gravell
AE
,
Reeves
SA
,
Rosner
GL
. 
Challenges and opportunities in adapting clinical trial design for immunotherapies
.
Clin Cancer Res
2017
;
23
:
4950
8
.
9.
Dudley
JC
,
Lin
MT
,
Le
DT
,
Eshleman
JR
. 
Microsatellite instability as a biomarker for PD-1 blockade
.
Clin Cancer Res
2016
;
22
:
813
20
.
10.
Palumbo
A
,
Anderson
K
. 
Multiple myeloma
.
N Engl J Med
2011
;
364
:
1046
60
.
11.
Prideaux
SM
,
Conway O'Brien
E
,
Chevassut
TJ
. 
The genetic architecture of multiple myeloma
.
Adv Hematol
2014
;
2014
:
864058
.
12.
Tamura
H
,
Ishibashi
M
,
Yamashita
T
,
Tanosaki
S
,
Okuyama
N
,
Kondo
A
, et al
Marrow stromal cells induce B7-H1 expression on myeloma cells, generating aggressive characteristics in multiple myeloma
.
Leukemia
2013
;
27
:
464
72
.
13.
Liu
J
,
Hamrouni
A
,
Wolowiec
D
,
Coiteux
V
,
Kuliczkowski
K
,
Hetuin
D
, et al
Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway
.
Blood
2007
;
110
:
296
304
.
14.
Zelle-Rieser
C
,
Thangavadivel
S
,
Biedermann
R
,
Brunner
A
,
Stoitzner
P
,
Willenbacher
E
, et al
T cells in multiple myeloma display features of exhaustion and senescence at the tumor site
.
J Hematol Oncol
2016
;
9
:
116
.
15.
Sponaas
AM
,
Yang
R
,
Rustad
EH
,
Standal
T
,
Thoresen
AS
,
D Vo
C
, et al
PD1 is expressed on exhausted T cells as well as virus specific memory CD8+ T cells in the bone marrow of myeloma patients
.
Oncotarget
2018
;
9
:
32024
35
.
16.
Ishibashi
M
,
Tamura
H
,
Sunakawa
M
,
Kondo-Onodera
A
,
Okuyama
N
,
Hamada
Y
, et al
Myeloma drug resistance induced by binding of myeloma B7-H1 (PD-L1) to PD-1
.
Cancer Immunol Res
2016
;
4
:
779
88
.
17.
John
LB
,
Ward
AC
. 
The Ikaros gene family: transcriptional regulators of hematopoiesis and immunity
.
Mol Immunol
2011
;
48
:
1272
8
.
18.
Yoshida
T
,
Georgopoulos
K
. 
Ikaros fingers on lymphocyte differentiation
.
Int J Hematol
2014
;
100
:
220
9
.
19.
Kronke
J
,
Udeshi
ND
,
Narla
A
,
Grauman
P
,
Hurst
SN
,
McConkey
M
, et al
Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells
.
Science
2014
;
343
:
301
5
.
20.
Powell
MD
,
Read
KA
,
Sreekumar
BK
,
Oestreich
KJ
. 
Ikaros zinc finger transcription factors: regulators of cytokine signaling pathways and CD4(+) T helper cell differentiation
.
Front Immunol
2019
;
10
:
1299
.
21.
Bartlett
JB
,
Dredge
K
,
Dalgleish
AG
. 
The evolution of thalidomide and its IMiD derivatives as anticancer agents
.
Nat Rev Cancer
2004
;
4
:
314
22
.
22.
Kyle
RA
,
Rajkumar
SV
. 
Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma
.
Leukemia
2009
;
23
:
3
9
.
23.
Ishibashi
M
,
Takahashi
R
,
Tsubota
A
,
Sasaki
M
,
Handa
H
,
Imai
Y
, et al
SLAMF3-mediated signaling via ERK pathway activation promotes aggressive phenotypic behaviors in multiple myeloma
.
Mol Cancer Res
2020
;
18
:
632
43
.
24.
Davies
F
,
Baz
R
. 
Lenalidomide mode of action: linking bench and clinical findings
.
Blood Rev
2010
;
24
:
S13
9
.
25.
Cho
SF
,
Anderson
KC
,
Tai
YT
. 
Targeting B cell maturation antigen (BCMA) in multiple myeloma: potential uses of BCMA-based immunotherapy
.
Front Immunol
2018
;
9
:
1821
.
26.
Stewart
R
,
Morrow
M
,
Hammond
SA
,
Mulgrew
K
,
Marcus
D
,
Poon
E
, et al
Identification and characterization of MEDI4736, an antagonistic anti-PD-L1 monoclonal antibody
.
Cancer Immunol Res
2015
;
3
:
1052
62
.
27.
Gorgun
G
,
Samur
MK
,
Cowens
KB
,
Paula
S
,
Bianchi
G
,
Anderson
JE
, et al
Lenalidomide enhances immune checkpoint blockade-induced immune response in multiple myeloma
.
Clin Cancer Res
2015
;
21
:
4607
18
.
28.
Benson
DM
 Jr
,
Bakan
CE
,
Mishra
A
,
Hofmeister
CC
,
Efebera
Y
,
Becknell
B
, et al
The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody
.
Blood
2010
;
116
:
2286
94
.
29.
Iwasa
M
,
Harada
T
,
Oda
A
,
Bat-Erdene
A
,
Teramachi
J
,
Tenshin
H
, et al
PD-L1 upregulation in myeloma cells by panobinostat in combination with interferon-gamma
.
Oncotarget
2019
;
10
:
1903
17
.
30.
Lopez-Girona
A
,
Heintel
D
,
Zhang
LH
,
Mendy
D
,
Gaidarova
S
,
Brady
H
, et al
Lenalidomide downregulates the cell survival factor, interferon regulatory factor-4, providing a potential mechanistic link for predicting response
.
Br J Haematol
2011
;
154
:
325
36
.
31.
Lopez-Girona
A
,
Mendy
D
,
Ito
T
,
Miller
K
,
Gandhi
AK
,
Kang
J
, et al
Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide
.
Leukemia
2012
;
26
:
2326
35
.
32.
Tai
YT
,
Acharya
C
,
An
G
,
Moschetta
M
,
Zhong
MY
,
Feng
X
, et al
APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment
.
Blood
2016
;
127
:
3225
36
.
33.
An
G
,
Acharya
C
,
Feng
X
,
Wen
K
,
Zhong
M
,
Zhang
L
, et al
Osteoclasts promote immune suppressive microenvironment in multiple myeloma: therapeutic implication
.
Blood
2016
;
128
:
1590
603
.
34.
Xu
J
,
Ding
WF
,
Shao
KK
,
Wang
XD
,
Wang
GH
,
Li
HQ
, et al
Transcription of promoter from the human APRIL gene regulated by Sp1 and NF-kB
.
Neoplasma
2012
;
59
:
341
7
.
35.
Wang
JQ
,
Jeelall
YS
,
Ferguson
LL
,
Horikawa
K
. 
Toll-like receptors and cancer: MYD88 mutation and inflammation
.
Front Immunol
2014
;
5
:
367
.
36.
Xu-Monette
ZY
,
Zhang
M
,
Li
J
,
Young
KH
. 
PD-1/PD-L1 blockade: have we found the key to unleash the antitumor immune response?
Front Immunol
2017
;
8
:
1597
.
37.
Akinleye
A
,
Rasool
Z
. 
Immune checkpoint inhibitors of PD-L1 as cancer therapeutics
.
J Hematol Oncol
2019
;
12
:
92
.
38.
Oliva
S
,
Troia
R
,
D'Agostino
M
,
Boccadoro
M
,
Gay
F
. 
Promises and pitfalls in the use of PD-1/PD-L1 inhibitors in multiple myeloma
.
Front Immunol
2018
;
9
:
2749
.
39.
De Sousa Linhares
A
,
Battin
C
,
Jutz
S
,
Leitner
J
,
Hafner
C
,
Tobias
J
, et al
Therapeutic PD-L1 antibodies are more effective than PD-1 antibodies in blocking PD-1/PD-L1 signaling
.
Sci Rep
2019
;
9
:
11472
.
40.
Lesokhin
AM
,
Ansell
SM
,
Armand
P
,
Scott
EC
,
Halwani
A
,
Gutierrez
M
, et al
Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study
.
J Clin Oncol
2016
;
34
:
2698
704
.
41.
Neri
P
,
Bahlis
NJ
,
Lonial
S
. 
New strategies in multiple myeloma: immunotherapy as a novel approach to treat patients with multiple myeloma
.
Clin Cancer Res
2016
;
22
:
5959
65
.
42.
Jelinek
T
,
Paiva
B
,
Hajek
R
. 
Update on PD-1/PD-L1 inhibitors in multiple myeloma
.
Front Immunol
2018
;
9
:
2431
.
43.
Tremblay-LeMay
R
,
Rastgoo
N
,
Chang
H
. 
Modulating PD-L1 expression in multiple myeloma: an alternative strategy to target the PD-1/PD-L1 pathway
.
J Hematol Oncol
2018
;
11
:
46
.

Supplementary data