Multiple myeloma is a plasma cell malignancy that is still largely incurable, despite considerable progress in recent years. NF-κB is a well-established therapeutic target in multiple myeloma, but none of the currently available treatment options offer direct, specific pharmacologic targeting of NF-κB transcriptional activity. Thus, we designed a novel direct NF-κB inhibitor (IT848) as a drug candidate with strong potential for clinical translation and conducted comprehensive in vitro and in vivo mechanistic studies in multiple myeloma cell lines, primary multiple myeloma cells, xenograft models, and immunocompetent mouse models of multiple myeloma. Here, we show that IT848 inhibits NF-κB activity through inhibition of DNA binding of all five NF-κB subunits. IT848 treatment of multiple myeloma cell lines and patient samples inhibited proliferation and induced caspase-dependent and independent apoptosis. In addition to direct NF-κB inhibitory effects, IT848 treatment altered the redox homeostasis of multiple myeloma cells through depletion of the reduced glutathione pool, selectively inducing oxidative stress in multiple myeloma but not in healthy cells. Multiple myeloma xenograft studies confirmed the efficacy of IT848 as single agent and in combination with bortezomib. Furthermore, IT848 significantly improved survival when combined with programmed death protein 1 inhibition, and correlative immune studies revealed that this clinical benefit was associated with suppression of regulatory T-cell infiltration of the bone marrow microenvironment. In conclusion, IT848 is a potent direct NF-κB inhibitor and inducer of oxidative stress specifically in tumor cells, displaying significant activity against multiple myeloma cells in vitro and in vivo, both as monotherapy as well as in combination with bortezomib or immune checkpoint blockade.

Multiple myeloma is the second most common hematologic malignancy, with an incidence of about 32,000 new cases and almost 13,000 deaths per year in the United States. Current first-line therapies include proteasome inhibitors, immunomodulators such as lenalidomide, alkylating agents, monoclonal antibodies such as daratumumab, and steroids. Several phase III trials have established the use of high-dose chemotherapy followed by autologous stem cell transplantation as standard of care in eligible patients (1). Second-generation proteasome inhibitors as well as third-generation immunomodulators have contributed to improving outcomes in the previous decade (2), and B-cell maturation antigen (BCMA)-targeted agents have shown promise in relapsed or refractory multiple myeloma (3). However, almost all patients eventually become refractory to currently available therapies. Therefore, there is an urgent need for novel agents with enhanced efficacy in this setting.

The NF-κB family of transcription factors is found in all nucleated cells. This pathway plays an important role in B-cell proliferation, T-cell activation, regulation of inflammatory responses, as well as the pathogenesis of cancer (4). The NF-κB family comprises of 5 structurally related monomeric subunits: Rel A, Rel B, c-Rel, p50, and p52. The combinatorial association of these subunits produces transcription factors with dimerization and DNA binding domains. In latent form, NF-κB is bound to an inhibitory protein (IκB). When stimulated extracellularly (via ligands such as TNFα, IL6, RANKL, etc.), NF-κB dimers translocate into the nuclei and mediate the expression of a plethora of immune/ stress response genes and pro-survival factors (4). NF-κB is constitutively active in multiple myeloma cells (5, 6) promoting cell proliferation, survival and resistance to chemotherapy (7). Consequently, NF-κB is an attractive therapeutic target in multiple myeloma, with both canonical and noncanonical NF-κB pathways contributing to the total NF-κB activity. NF-κB also regulates transcriptional signatures involved in inflammation and immunity, including the programmed death-ligand 1 (PD-L1)/programmed death protein 1 (PD-1) axis (8). Inhibition of NF-κB DNA binding in multiple myeloma might therefore not only counteract upstream activating mutations but also modulate the immunosuppressive tumor microenvironment (TME). PD-L1/PD-1 interaction results in suppression of T cell–mediated cytotoxicity (9), promoting an immunosuppressive milieu in TME (10). Disruption of the PD-L1/PD-1 axis by monoclonal antibody therapy has been highly effective in several solid tumors but has shown only limited activity against multiple myeloma in initial trials (11). This has led to the additional investigation of protocols combining immune checkpoint blockade with lenalidomide or dexamethasone (12, 13). After initial setbacks, immunotherapeutic agents have now emerged as an attractive option for the treatment of multiple myeloma, but additional preclinical and clinical investigations are warranted to establish safe and effective treatment regimens (14) such as the combination of NF-κB inhibition with immune checkpoint blockade. Here, we introduce IT848, a small molecule inhibitor of NF-κB DNA binding with drug-like physicochemical properties, as a candidate for future clinical development of an NF-κB inhibitor drug. We show that IT848 combines NF-κB inhibition with dysregulation of the redox homeostasis in multiple myeloma cells as well as favorable immunomodulatory properties, translating into significant in vitro and in vivo efficacy against multiple myeloma.

Small molecule NF-κB inhibitor compound

We previously identified thiohydantoin (IT603) and naphthalenethiobarbiturate (IT901) derivatives as conformation-disrupting direct NF-κB inhibitors. After additional structure activity relationship analysis and pharmacokinetics (PK) studies, the novel small molecule IT848 [9-chloro-8-(hexyloxy)-2H-chromeno(2,3-d)pyrimidine-2,4(3H)-dione], molecular weight = 348.78 g/mol was selected as our drug candidate. The IT848 synthesis method is described in detail in our patent (see example # 21, Table 1 of ref. 15).

Mouse models of multiple myeloma, in vivo imaging, and NF-κB inhibitor treatment

NOD/SCID/IL2Rγ(null) (NSG) mice were obtained from the Jackson Laboratory for xenograft experiments of multiple myeloma. MM.1S human multiple myeloma cells were transduced with dtomato-firefly luciferase and 5×106 cells were administered to NSG mice via tail vein injection. C57Bl/KaLwRij mice were originally obtained from Envigo (Horst, the Netherlands) and maintained at the Center for Discovery and Innovation. All animal studies were conducted in accordance with Institutional Animal Care and Use Committee (IACUC) standards and with approval of the IACUC of the CDI (protocol#291).

Mice used for these experiments were 6 to 9 weeks old. For the study in immunocompetent mice, 5TGM1 mouse multiple myeloma cells (16) were transduced with dtomato-firefly luciferase and 1.5×106 cells were administered to C57Bl/KaLwRij mice via tail vein injection. Bone marrow engraftment of multiple myeloma cells was confirmed by in vivo bioluminescence imaging (BLI) prior to initiation of treatment, and multiple myeloma progression was monitored at the indicated time points by in vivo BLI. Briefly, mice were anesthetized using isoflurane inhalant anesthetic and administered 3-mg/kg firefly luciferin intraperitoneally and imaged using the IVIS Lumina X5 optical imaging platform. We superimposed pseudocolor images showing the whole-body distribution of bioluminescent signal intensity on grayscale photographs and quantified total flux (photons s−1) for individual mice using Living Image 4.7.3 software (Perkin Elmer, Waltham, MA). In addition, coelenterazine was used to detect changes in regional reactive oxygen species production in vivo in response to IT848 administration (17). For in vivo efficacy studies, IT848 was dissolved in PBS with 30% (w/w) cyclodextrin and 5% PEG 1000 to prepare a 1-mg/mL solution. Mice received either empty vehicle solution or 10-mg/kg body weight of IT848 solution via intraperitoneal injection on either 3 days/week for 4 weeks or on 5 days/week for 2 weeks.

Cell lines and primary cells

Vendor-authenticated human multiple myeloma cell lines U266, MM.1S, and NCI-H929 were originally obtained from the ATCC (Manassas, VA) in 2018. The human diffuse large B-cell lymphoma (DLBCL)-derived cell lines U2932, HBL1, and TMD8 were kindly provided by Dr. Anas Younes (Memorial Sloan Kettering Cancer Center) in 2013 and authenticated before using them by the MD Anderson Characterized Cell Lines Core Facility. These cells were cultured in RPMI1640 medium supplemented with 10% to 20% heat-inactivated FBS (GIBCO BRL, Gaithersburg, MD), 1% l-glutamine, and penicillin–streptomycin in a humid environment of 5% CO2 at 37°C. The 5TGM1 cell line was kindly provided by Dr. Rena Feinman (Center for Discovery and Innovation, Nutley, NJ). Vendor-authenticated NIH 3T3 cells were purchased from ATCC in 2019.

A vendor-authenticated NF-κB/Jurkat/GFP transcriptional reporter cell line (18) was purchased from System Biosciences (Mountain View, CA) in 2015. Cells were stimulated with human TNFα (10 ng/mL; Gemini Bio Products, West Sacramento, CA) and incubated in the presence of IT848 or empty vehicle. NF-κB activity (GFP fluorescence intensity) was analyzed by flow cytometry. Vendor-authenticated ARE/HepG2/Luciferase reporter cells and Nuclear factor of activated T-cells (NFAT)/Jurkat/luciferase transcriptional reporter cells were purchased from BPS Bioscience (San Diego, CA) in 2017. ARE/Nrf2 and NFAT transcriptional activity was determined using the One-Step Luciferase Assay System (BPS Biosciences, San Diego, CA). Luminescence was measured with a multimodal plate reader (Infinite 200 pro from Tecan, Switzerland). Human Epstein-Barr Virus (EBV)-transformed B lymphoblastoid cells were generated from peripheral blood mononuclear cell (PBMC) of a healthy donor and kindly provided by Richard O'Reilly (Memorial Sloan Kettering Cancer Center, NY) in 2018. All cell lines were used for experiments within 3 weeks of thawing (up to 4 passages) and were annually tested for Mycoplasma negativity using a Mycoplasma PCR Kit (MycoAlert Kit, Lonza).

Human PBMCs were purchased from United States Biological (Salem, MA). Human multiple myeloma patient cells were collected after informed written consent was obtained, using Institutional Review Board protocol Pro00004259 at Hackensack Meridian Health–John Theurer Cancer Center. All studies of patient materials were conducted in accordance with the Declaration of Helsinki.

MM.1S and 5TGM1 cells were transduced using 3rd generation lentiviral vector with bi-cistronic expression of dtomato and luciferase (pUltra-Chili-Luc; Addgene #48688). Transduced cells were expanded and individual clones/population with high dtomato expression were sorted using a BD FACSMelody. Bioluminescence signal intensity of single clones was determined, and the brightest clones were used for subsequent experiments.

RNA sequencing

RNA was purified using an AllPrep DNA/RNA extraction kit (Qiagen, Hilden, Germany) according to the manufacturer's instructions. RNA quantity was measured with a Nanodrop spectrophotometer (DeNovix, Wilmington, DE). RNA was submitted to Genewiz (South Plainfield, NJ) for cDNA library creation and sequencing. RNA sequencing (RNA-seq) was performed in triplicate for each treatment condition on an Illumina Hi-Seq 2×150 bp configuration, single index, per lane with estimated data output as ∼350 M raw paired-end reads per lane (19). Initial sample quality control involved assessing RNA integrity and library size by Agilent Tapestation and concentration by Qubit assay, and final quantitation by qPCR. Differential expression analysis between sets of conditions was performed using the DESeq2 pipeline of Basepair analysis platform (Basepair, New York, NY). Gene set enrichment analysis was performed as previously described (20) and a visualization of the canonical NF-κB pathway from the KEGG database is presented.

Data availability statement

The data generated in this study are available within the article and its Supplementary Data files. The RNA-seq data generated in this study are publicly available in Gene Expression Omnibus at GSE200385.

IT848 is a potent inhibitor of NF-κB DNA binding

We previously described a novel small molecule compound mediating anticancer properties by combining inhibition of NF-κB transcriptional activity with the capacity to modulate the redox balance of B-cell lymphoma cells (21). After additional structure activity relationship analysis and in vivo drug metabolism and PK testing of our lead molecules, IT848 emerged as the most suitable candidate for drug development (Fig. 1A). In silico molecular docking analysis (22) identified IT848 binding sites on all NF-κB subunits (Fig. 1B), and homology modeling indicated that IT848 interacts with thiol groups of cysteines of the Rel/NF-κB subunits. IT848 elicited strong and dose-dependent inhibition of global NF-κB transcriptional activity both in lymphoma (Jurkat, TMD8) and myeloma (U266) cell lines or primary cells (Epstein–Barr virus—transformed B lymphoblastoid cells; Fig. 1C and D). Moreover, we confirmed dose-dependent target-specific in vivo activity of IT848 (Supplementary Fig. S1). Western blot analysis of NF-κB complex proteins [IKΚBeta, IκBalpha, interferon regulatory factor 4 (IRF4)] further validated IT848 as a potent NF-κB inhibitor with consistent, inhibitory effects across several DLBCL and multiple myeloma cell lines (Fig. 1E). Analysis of the NF-κB target gene and pro-tumorigenic cytokine IL6 (23, 24) in representative DLBCL and multiple myeloma cell lines by RT-qPCR (Fig. 1F) and enzyme-linked immunosorbent assay (ELISA; Fig. 1G) demonstrated IT848 mediated supression of IL6 expression, an effect that might not only be useful therapeutically, but also as a biomarker for IT848 activity in multiple myeloma cells. To further elucidate the mechanism of action of IT848 and confirm its capacity to act as intended as a direct NF-κB inhibitor, we assessed DNA binding of individual NF-κB subunits in response to IT848 treatment. We used MicroScale Thermophoresis to characterize binding affinities of IT848 to c-Rel and p50, and to analyze DNA binding of c-Rel and p50 following IT848 treatment. We found that IT848 did not bind to DNA, but it showed binding affinity to both c-Rel and p50 in the low μmo/L range (Kd of 3.9 and 7.0, respectively). In addition, IT848 treatment disrupted DNA binding of c-Rel and decreased p50 DNA binding 30 to 100-fold (Supplementary Fig. S2). In line with this, NF-κB DNA binding ELISA of nuclear extracts from IT848 treated representative DLBCL and multiple myeloma cell lines showed decreased DNA binding of all five NF-κB subunits (although with cell type-specific variability; Fig. 1H). We next assessed the specificity of IT848 and found that inhibition of transcriptional activity was specific to NF-κB but spared unrelated transcription factors that were tested as controls (NFAT and Nrf2; Fig. 1I). Taken together, our findings identify IT848 as a potent and specific small molecule NF-κB inhibitor that decreases the affinity of NF-κB subunits to their DNA binding sites and disrupts downstream biological events.

Figure 1.

IT848 inhibits NF-κB transcriptional activity as a result of decreased DNA binding of NF-κB subunits. A, Chemical structure of IT848. B, Pose view interaction of IT848 (red) with NF-κB subunits (cyan): (A) RelB, (B) p52, (C) RelA, (D) p50, and (E) c-Rel. The white regions represent NF-κB residues which exhibit nonpolar interactions with IT848, and the yellow regions represent NF-κB residues which exhibit polar interactions with IT848. C, Jurkat/GFP/NF-κB transcriptional reporter cells were stimulated with TNFα and treated for 22 hours in the presence of empty vehicle or 1, 3, 6 μmol/L of IT848. NF-κB transcriptional activity (GFP median fluorescence intensity) was analyzed at 8 and 22 hours by flow cytometry. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown. D, U266 multiple myeloma cells, TMD8 DLBCL cells, and an EBV-transformed B lymphoblastic cell line (BLCL) were engineered to constitutively express Renilla luciferase as well as NF-κB–driven firefly luciferase. Cells were incubated for 24 hours in the presence of empty vehicle or 2, 4, and 6 μmol/L of IT848. NF-κB transcriptional activity was analyzed by dual luciferase assay. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown. E, DLBCL and multiple myeloma cell lines were cultured in the presence of IT848 (2, 4, or 6 μmol/L) or DMSO for 12 hours. NF-κB activity was analyzed by Western blot of the NF-κB pathway signaling transduction molecules phospho-IKKb, phospho-IκBa, and IRF. Beta Actin was included as an internal control. F, TMD8 and U266 cells were treated with IT848 (4 μmol/L) or empty vehicle for 4 hours, followed by RNA extraction and qRT-PCR of IL6. Mean and SEM of relative gene expression is presented: *, P < 0.001; **, P = 0.002. One of three independent experiments is shown. G, TMD8 and U266 cells were incubated for 24 hours in the presence of empty vehicle or IT-848 (4 and 6 μmol/L). IL6 concentrations in the culture media were analyzed after 24 hours by ELISA. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline expression): *, P < 0.01. H, MM.1S and U266 human multiple myeloma and HBL-1 human DLBCL cells were treated with IT848 (4 μmol/L) or empty vehicle for 4 hours, followed by nuclear extraction and analysis of DNA binding of all five NF-κB subunits by ELISA. Mean and SEM of normalized values (percentages of vehicle-treated baseline DNA binding) are presented. One of two independent experiments is shown. I, Jurkat/GFP/NF-κB transcriptional reporter cells were stimulated with TNFα, Jurkat/Luciferase/NFAT transcriptional reporter cells were stimulated with PMA/ionomycin, and HepG2/Luciferase/Nrf2-antioxidant response element (ARE) transcriptional reporter cells were treated with tert-butylhydroquinone. The respective cell lines were incubated for 12 hours in the presence of empty vehicle or 1, 3, 6, and 10 μmol/L of IT848. NF-κB transcriptional activity was analyzed by flow cytometry, NFAT and Nrf2 transcriptional activities were analyzed by luminescence measurement. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown.

Figure 1.

IT848 inhibits NF-κB transcriptional activity as a result of decreased DNA binding of NF-κB subunits. A, Chemical structure of IT848. B, Pose view interaction of IT848 (red) with NF-κB subunits (cyan): (A) RelB, (B) p52, (C) RelA, (D) p50, and (E) c-Rel. The white regions represent NF-κB residues which exhibit nonpolar interactions with IT848, and the yellow regions represent NF-κB residues which exhibit polar interactions with IT848. C, Jurkat/GFP/NF-κB transcriptional reporter cells were stimulated with TNFα and treated for 22 hours in the presence of empty vehicle or 1, 3, 6 μmol/L of IT848. NF-κB transcriptional activity (GFP median fluorescence intensity) was analyzed at 8 and 22 hours by flow cytometry. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown. D, U266 multiple myeloma cells, TMD8 DLBCL cells, and an EBV-transformed B lymphoblastic cell line (BLCL) were engineered to constitutively express Renilla luciferase as well as NF-κB–driven firefly luciferase. Cells were incubated for 24 hours in the presence of empty vehicle or 2, 4, and 6 μmol/L of IT848. NF-κB transcriptional activity was analyzed by dual luciferase assay. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown. E, DLBCL and multiple myeloma cell lines were cultured in the presence of IT848 (2, 4, or 6 μmol/L) or DMSO for 12 hours. NF-κB activity was analyzed by Western blot of the NF-κB pathway signaling transduction molecules phospho-IKKb, phospho-IκBa, and IRF. Beta Actin was included as an internal control. F, TMD8 and U266 cells were treated with IT848 (4 μmol/L) or empty vehicle for 4 hours, followed by RNA extraction and qRT-PCR of IL6. Mean and SEM of relative gene expression is presented: *, P < 0.001; **, P = 0.002. One of three independent experiments is shown. G, TMD8 and U266 cells were incubated for 24 hours in the presence of empty vehicle or IT-848 (4 and 6 μmol/L). IL6 concentrations in the culture media were analyzed after 24 hours by ELISA. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline expression): *, P < 0.01. H, MM.1S and U266 human multiple myeloma and HBL-1 human DLBCL cells were treated with IT848 (4 μmol/L) or empty vehicle for 4 hours, followed by nuclear extraction and analysis of DNA binding of all five NF-κB subunits by ELISA. Mean and SEM of normalized values (percentages of vehicle-treated baseline DNA binding) are presented. One of two independent experiments is shown. I, Jurkat/GFP/NF-κB transcriptional reporter cells were stimulated with TNFα, Jurkat/Luciferase/NFAT transcriptional reporter cells were stimulated with PMA/ionomycin, and HepG2/Luciferase/Nrf2-antioxidant response element (ARE) transcriptional reporter cells were treated with tert-butylhydroquinone. The respective cell lines were incubated for 12 hours in the presence of empty vehicle or 1, 3, 6, and 10 μmol/L of IT848. NF-κB transcriptional activity was analyzed by flow cytometry, NFAT and Nrf2 transcriptional activities were analyzed by luminescence measurement. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline activity). One of two independent experiments is shown.

Close modal

IT848 inhibits proliferation and induces apoptosis in multiple myeloma cells

NF-κB is known to be constitutively active in all multiple myeloma cells (5, 6, 25). Indeed, NF-κB DNA binding analysis of two multiple myeloma cell lines (MM.1S and U266) confirmed constitutive activity of all five NF-κB subunits (Supplementary Fig. S3). To assess the anti-myeloma activity of IT848 we treated MM.1S, U266, and NCI-H929 cells with increasing doses of the compound. This resulted in a dose-dependent decrease in cell numbers over a 72-hour period as measured by luciferase assay (Fig. 2A) and direct cell counting (Fig. 2B). Importantly, PBMCs of several healthy control subjects were not negatively affected by IT848 treatment except for a 10 percent decrease in metabolic viability in the presence of the highest tested concentration (6 μmol/L; Fig. 2C). To further evaluate the mechanism of action associated with the cytotoxic effects of IT848, we examined its impact on cell-cycle phase distribution after 24 hours of treatment. The percentages of IT848-treated cells in S phase were increased while the frequency of cells in G2–M phase was lowered in a dose-dependent manner (Fig. 2D). This suggests that IT848 effectively affects cell-cycle checkpoints leading to S phase arrest and inhibition of cell division, which was confirmed by concordant decreased levels of Ki-67 expression, a nuclear protein present in proliferating cells (Fig. 2E). Again, normal PBMCs were insensitive to the anti-proliferative effect of IT848 (Fig. 2E). Furthermore, Annexin-V and 7-AAD staining revealed that IT848 induced a shift from early apoptosis (Annexin V+, 7-AAD−) to multiple myeloma cell death (Annexin V+, 7-AAD+) after 24 hours of treatment (Fig. 2F). Therefore, the IT848-induced decrease in cell numbers is likely due to a dual effect, including inhibition of proliferation and induction of apoptosis. IT848 treatment resulted in a dose-dependent increase in caspase activity in multiple myeloma cells (using a kit detecting activity of caspase 1, 3, 4, 5, 6, 7, 8 and 9; Fig. 2G). Furthermore, IT848 treatment enhanced nuclear translocation of apoptosis-inducing factor (AIF), a factor involved in a caspase-independent pathway of apoptosis (Fig. 2H; refs. 26–28). AIF is a mitochondrial proapoptotic factor that is released from mitochondria upon decrease of the mitochondrial membrane potential and subsequent increased permeability of the mitochondrial membrane (26, 27). We found that while low-dose IT848 treatment (2 and 4 μmol/L) did not alter the mitochondrial membrane potential of multiple myeloma cells, higher doses triggered a loss of the mitochondrial membrane potential (Fig. 2I), consistent with bioenergetic stress and release of proapoptotic factors such as AIF.

Figure 2.

IT848 inhibits growth and induces apoptosis of multiple myeloma cells. MM.1S, U266 and NCI-H929 cells and normal PBMCs were incubated in the presence of IT848 or empty vehicle (control) for 72 hours (AC) or 24 hours (DF). A, Cell numbers after 0, 24, 48, 72 hours were quantified by luciferase assay. Mean and SEM are presented. B, Cell numbers after 0, 24, 48, 72 hours were quantified by manual cell counting. Mean and SEM are presented. C, metabolic viability of PBMC (n = 4) was measured by MTS assay. Mean and SEM of normalized values (percentages of vehicle-treated baseline activity) are presented. D, Cell cycle analysis of live MM.1S cells treated with IT848 (4 μmol/L) or DMSO for 24 hours. Percentage of cells differentially stained with Hoechst 33342 and Ki-67 were quantified to show DNA content distribution/changes of affected S and G2–M phases. Representative dot plots as well as mean and SEM are presented. E, proliferation of the indicated cell lines and normal PBMC was analyzed by Ki-67 assay. Representative histograms and mean and SEM are presented. F, Apoptosis of MM.1S and U266 cells was analyzed by Annexin V/7AAD staining. Mean and SEM are presented. G, MM.1S, U266 and NCI-H929 cells were incubated in the presence of 2, 4, and 6 μmol/L of IT848 or empty vehicle (control) for 24 hours. Activity of caspase 1, 3, 4, 5, 6, 7, 8, and 9 was quantified using a caspase activity detection kit as described in Materials and Methods. Mean and SEM of normalized data (percentage of control) are presented. H, U266 and NCI-H929 cells were incubated in the presence of 4 μmol/L of IT848 or empty vehicle (control) for 24 hours and stained with DAPI and a monoclonal antibody for microscopic detection of AIF. Representative fluorescence microscopy images are shown (blue, DNA; green, AIF; for more experimental details see Materials and Methods). I, MM.1S, U266, and NCI-H929 cells were incubated in the presence of 2, 4, and 6 μmol/L of IT848 or empty vehicle (control) for 24 hours. The mitochondrial membrane potential was quantified using a mitochondrial transmembrane potential detection kit as described in Materials and Methods. Mean and SEM of normalized data (percentage of control) are presented.

Figure 2.

IT848 inhibits growth and induces apoptosis of multiple myeloma cells. MM.1S, U266 and NCI-H929 cells and normal PBMCs were incubated in the presence of IT848 or empty vehicle (control) for 72 hours (AC) or 24 hours (DF). A, Cell numbers after 0, 24, 48, 72 hours were quantified by luciferase assay. Mean and SEM are presented. B, Cell numbers after 0, 24, 48, 72 hours were quantified by manual cell counting. Mean and SEM are presented. C, metabolic viability of PBMC (n = 4) was measured by MTS assay. Mean and SEM of normalized values (percentages of vehicle-treated baseline activity) are presented. D, Cell cycle analysis of live MM.1S cells treated with IT848 (4 μmol/L) or DMSO for 24 hours. Percentage of cells differentially stained with Hoechst 33342 and Ki-67 were quantified to show DNA content distribution/changes of affected S and G2–M phases. Representative dot plots as well as mean and SEM are presented. E, proliferation of the indicated cell lines and normal PBMC was analyzed by Ki-67 assay. Representative histograms and mean and SEM are presented. F, Apoptosis of MM.1S and U266 cells was analyzed by Annexin V/7AAD staining. Mean and SEM are presented. G, MM.1S, U266 and NCI-H929 cells were incubated in the presence of 2, 4, and 6 μmol/L of IT848 or empty vehicle (control) for 24 hours. Activity of caspase 1, 3, 4, 5, 6, 7, 8, and 9 was quantified using a caspase activity detection kit as described in Materials and Methods. Mean and SEM of normalized data (percentage of control) are presented. H, U266 and NCI-H929 cells were incubated in the presence of 4 μmol/L of IT848 or empty vehicle (control) for 24 hours and stained with DAPI and a monoclonal antibody for microscopic detection of AIF. Representative fluorescence microscopy images are shown (blue, DNA; green, AIF; for more experimental details see Materials and Methods). I, MM.1S, U266, and NCI-H929 cells were incubated in the presence of 2, 4, and 6 μmol/L of IT848 or empty vehicle (control) for 24 hours. The mitochondrial membrane potential was quantified using a mitochondrial transmembrane potential detection kit as described in Materials and Methods. Mean and SEM of normalized data (percentage of control) are presented.

Close modal

In contrast to antibody-based targeted therapy, chemical approaches using small molecules are expected to be less specific and off-target effects are common. Therefore, to broadly assess the effects of IT848 on cellular processes in multiple myeloma cells we performed RNA-seq of U266 and MM.1S cells treated with IT848 for 6 hours (Fig. 3A and B). This timepoint was chosen because significant cell death has not occurred yet and it therefore allows to assess the impact of IT848 treatment on the transcriptome of multiple myeloma cells. As expected, IT848 treatment of MM.1S cells resulted in downregulation of a variety of NF-κB target genes (Supplementary Table S1). However, we also found highly significant upregulations of reactive oxygen species (ROS) and apoptosis pathway genes, including heme oxygenase 1 (HMOX1) and oxidative stress-induced growth inhibitor 1 (OSGIN1), following exposure to IT848 (Fig. 3A and B). GSEA revealed that oxidative stress and apoptosis signatures were among the most significantly enriched gene sets in myeloma cells treated with IT848 (Fig. 3C; Supplementary Fig. S4). We validated RNA-seq findings regarding HMOX1 and OSGIN1 using RT-qPCR (Fig. 4A and B). Remarkably, we showed that the effect of IT848 on oxidative stress was selective for multiple myeloma cells while only minimal changes were observed in normal PBMCs (Fig. 4A and B). Specifically, IT848 treatment induced an increase of cellular superoxide (Fig. 4C) but not of hydrogen peroxide levels. Glutathione (GSH) is one of the major ROS-scavenger systems protecting cells from oxidative stress. We therefore compared GSH levels in MM.1S and U266 cells with and without IT848 treatment and found up to 70% depletion of the GSH pool with increasing drug concentrations (Fig. 4D). Control cells (splenocytes and NIH-3T3 cells) showed a negligible inhibition pattern, reinforcing our previous observation that myeloma cells are selectively sensitive to oxidative stress induction by IT848. Repletion of GSH partially reversed the anti-myeloma effect of IT848 in assays monitoring cell growth (Fig. 4E) and apoptosis (Supplementary Fig. S5), validating GSH depletion in cancer cells as a critical component of the mechanism of action of our drug. For in vivo confirmation of its pro-oxidant properties, we tested IT848-mediated ROS induction in NGS mice inoculated with MM.1S cells. Administration of the light emitting molecule coelenterazine resulted in enhanced chemiluminescent detection of superoxide anion (29) in the treatment group, suggesting that IT848 treatment can achieve in vivo modulation of the cellular redox homeostasis (Fig. 4F).

Figure 3.

IT848 treatment induces distinctive gene expression patterns in multiple myeloma cells. U266 and MM.1S cells were treated with IT848 (4 μmol/L) or DMSO for 6 hours and gene expression was analyzed by RNA-seq (n = 3). A+B. Volcano plot (left) and principal component analysis (right) showing significantly upregulated genes (P < 0.05) and a clearly distinguishable pattern in IT848 versus vehicle treated U266 cells (A) and MM.1S cells (B) as determined by RNA-seq. Statistical significance was determined by linear modelling and Bayesian statistics after correcting for multiple testing using the Benjamini–Hochberg procedure. C, GSEA of MM.1S cells. Enrichment plots of categories that are enriched in genes correlated with IT848 treatment and representative genes from each category are shown. Statistical significance was determined by Wald test with Benjamini-Hochberg's multiple-comparison correction.

Figure 3.

IT848 treatment induces distinctive gene expression patterns in multiple myeloma cells. U266 and MM.1S cells were treated with IT848 (4 μmol/L) or DMSO for 6 hours and gene expression was analyzed by RNA-seq (n = 3). A+B. Volcano plot (left) and principal component analysis (right) showing significantly upregulated genes (P < 0.05) and a clearly distinguishable pattern in IT848 versus vehicle treated U266 cells (A) and MM.1S cells (B) as determined by RNA-seq. Statistical significance was determined by linear modelling and Bayesian statistics after correcting for multiple testing using the Benjamini–Hochberg procedure. C, GSEA of MM.1S cells. Enrichment plots of categories that are enriched in genes correlated with IT848 treatment and representative genes from each category are shown. Statistical significance was determined by Wald test with Benjamini-Hochberg's multiple-comparison correction.

Close modal
Figure 4.

IT848 mediates oxidative stress selectively in multiple myeloma cells. A+B, Myeloma cell lines U266 and MM.1S and human PBMC (as control) were treated with IT-848 (4 μmol/L) or control vehicle for 6 hours. HMOX1 and OSGIN1 gene expression levels by qPCR are shown in A and B, respectively. C, U266 and MM.1S cells were further analyzed by Cellular Superoxide Detection Assay Kit to monitor real time superoxide production in live cells post–IT-848 treatment and normalized to the vehicle treatment. Cells were treated with Tert-Butyl hydroperoxide (TBHP, 200 μmol/L) as a positive control to generate high levels of oxidative stress. D, Myeloma cell lines (U266 and MM.1S) and control cells (splenocytes and NIH3T3 cells) were cultured in presence of IT848 (1, 2, 4, and 6 μmol/L) or empty vehicle for 24 hours. Cellular levels of reduced GSH levels in response to oxidative stress caused by IT-848 treatment were measured using ThiolTracker Violet GSH detection reagent as described in Materials and Methods. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline levels). E, MM.1S cells were cultured with empty vehicle, IT848 (4 μmol/L), l-glutathione reduced (1 mmol/L GSH), and a combination of IT848 and GSH and luciferase assay was performed over a 72-hour period to demonstrate the antioxidant effect of GSH repletion on IT848 action and anti-myeloma potency. Values represent mean ± SEM (n = 3, technical replicates) at each timepoint. One of two independent experiments is presented. F, Coelenterazine administration in vivo results in chemiluminescent detection of superoxide anion (n = 3 per group). Changes in regional reactive oxygen species production (localization and magnitude) in response to IT848 and vehicle administration over 4.5-hour period are presented. All data are representative of three independent experiments.

Figure 4.

IT848 mediates oxidative stress selectively in multiple myeloma cells. A+B, Myeloma cell lines U266 and MM.1S and human PBMC (as control) were treated with IT-848 (4 μmol/L) or control vehicle for 6 hours. HMOX1 and OSGIN1 gene expression levels by qPCR are shown in A and B, respectively. C, U266 and MM.1S cells were further analyzed by Cellular Superoxide Detection Assay Kit to monitor real time superoxide production in live cells post–IT-848 treatment and normalized to the vehicle treatment. Cells were treated with Tert-Butyl hydroperoxide (TBHP, 200 μmol/L) as a positive control to generate high levels of oxidative stress. D, Myeloma cell lines (U266 and MM.1S) and control cells (splenocytes and NIH3T3 cells) were cultured in presence of IT848 (1, 2, 4, and 6 μmol/L) or empty vehicle for 24 hours. Cellular levels of reduced GSH levels in response to oxidative stress caused by IT-848 treatment were measured using ThiolTracker Violet GSH detection reagent as described in Materials and Methods. Mean and SEM of normalized values are presented (percentages of vehicle-treated baseline levels). E, MM.1S cells were cultured with empty vehicle, IT848 (4 μmol/L), l-glutathione reduced (1 mmol/L GSH), and a combination of IT848 and GSH and luciferase assay was performed over a 72-hour period to demonstrate the antioxidant effect of GSH repletion on IT848 action and anti-myeloma potency. Values represent mean ± SEM (n = 3, technical replicates) at each timepoint. One of two independent experiments is presented. F, Coelenterazine administration in vivo results in chemiluminescent detection of superoxide anion (n = 3 per group). Changes in regional reactive oxygen species production (localization and magnitude) in response to IT848 and vehicle administration over 4.5-hour period are presented. All data are representative of three independent experiments.

Close modal

IT848 is effective against multiple myeloma xenografts and patient cells

Next, we investigated the PK of IT848 by analyzing its whole blood or plasma levels following oral, intraperitoneal, or intravenous administration (Supplementary Fig. S6). We found that the maximum plasma concentration (cmax) and half-life (t1/2) of IT848 following all three routes of administration was sufficient to achieve sustained plasma concentrations equivalent to the 4 to 6 μmol/L range, the therapeutic window associated with in vitro efficacy (see Fig. 2A and B). In addition, toxicology studies demonstrated the absence of organ toxicities during a 12-day course of IT848 treatment.

For in vivo efficacy testing of IT848 we first utilized a xenograft model of multiple myeloma and included the proteasome inhibitor bortezomib (30, 31). We chose bortezomib as a reference drug because it is a well-established anti-myeloma agent with NF-κB inhibitory properties but a different mechanism of action than IT848, hypothesizing that combination therapy should be safe and may producde additive effects. Mice were treated with either vehicle (control), bortezomib monotherapy, IT848 monotherapy, or the combination of the two. Monitoring disease progression by in vivo BLI, we observed significant efficacy with both bortezomib (P = 0.008) and IT848 monotherapy (P < 0.001; Fig. 5A and B). Notably, IT848 showed superior tumor growth inhibition compared to bortezomib (P = 0.032), and combination treatment resulted in improved efficacy when compared with either monotherapy (P < 0.001, P = 0.029). RNA-seq comparing MM.1S cells treated with bortezomib alone versus bortezomib plus IT848 (Supplementary Fig. S7) revealed a more significant downregulation of seven NF-κB related genes with combination treatment, including the NF-κB essential modulator (NEMO; P = 0.02), which is a validated target for molecular cancer therapy (32–34).

Figure 5.

IT848 is effective as monotherapy, and it provides additive effects in combination with bortezomib. NSG mice received 2.5×106 luciferase expressing MM.1S cells intravenously. After 10 days engraftment was confirmed and mice were assigned to the following groups: 1. empty vehicle I.P. 3x/week x 4 weeks; 2. bortezomib 0.5 mg/kg I.P. 2x/week x 4 weeks; 3. IT848 10 mg/kg I.P. 3x/week x 4 weeks; 4. Bortezomib + IT848×4 weeks. A, Multiple myeloma progression was analyzed by in vivo BLI at the indicated time points. Pseudocolor images superimposed on conventional photographs are presented. One of two independent experiments is shown. B, Mean and SEM of bioluminescence intensities are presented (n = 7–10). The AUC was used to summarize intensity values for each subject and a permutation test was performed to determine of there was a significant difference between groups. One of two independent experiments is shown. C, Cause-specific survival (death from disease) is presented. Differences between groups were analyzed by Log-rank test. Combined data from two independent experiments are presented (n = 10–20). D+E, CD138-selected cells from patients with newly diagnosed multiple myeloma (n = 2) and plasma cell leukemia (n = 1) were incubated in the presence of IT848 or empty vehicle (control) for 24 hours. D, apoptosis of patient cells was analyzed by Annexin V/7AAD staining. Representative FACS plots and mean and SEM are presented. E, proliferation of patient cells was analyzed by Ki-67 assay. Mean and SEM are presented.

Figure 5.

IT848 is effective as monotherapy, and it provides additive effects in combination with bortezomib. NSG mice received 2.5×106 luciferase expressing MM.1S cells intravenously. After 10 days engraftment was confirmed and mice were assigned to the following groups: 1. empty vehicle I.P. 3x/week x 4 weeks; 2. bortezomib 0.5 mg/kg I.P. 2x/week x 4 weeks; 3. IT848 10 mg/kg I.P. 3x/week x 4 weeks; 4. Bortezomib + IT848×4 weeks. A, Multiple myeloma progression was analyzed by in vivo BLI at the indicated time points. Pseudocolor images superimposed on conventional photographs are presented. One of two independent experiments is shown. B, Mean and SEM of bioluminescence intensities are presented (n = 7–10). The AUC was used to summarize intensity values for each subject and a permutation test was performed to determine of there was a significant difference between groups. One of two independent experiments is shown. C, Cause-specific survival (death from disease) is presented. Differences between groups were analyzed by Log-rank test. Combined data from two independent experiments are presented (n = 10–20). D+E, CD138-selected cells from patients with newly diagnosed multiple myeloma (n = 2) and plasma cell leukemia (n = 1) were incubated in the presence of IT848 or empty vehicle (control) for 24 hours. D, apoptosis of patient cells was analyzed by Annexin V/7AAD staining. Representative FACS plots and mean and SEM are presented. E, proliferation of patient cells was analyzed by Ki-67 assay. Mean and SEM are presented.

Close modal

However, frequent intraperitoneal administration, which is a clincially irrelevant drug devivery route but commonly used in preclinical animal models, of IT848 for more than two weeks was associated with considerable toxicity (such as development of ascites and diarrhea) and concordant treatment-associated mortality in the range of 20 to 30 percent (Fig. 5A; Supplementary Fig. S8). Analysis of cause-specific survival revealed that despite this mouse model-specific drawback, the compound still mediated therapeutically beneficial in vivo efficacy resulting in significantly improved disease-specific survival (Fig. 5C). These observations suggest the need for the development of more clinically relevant better tolerated treatment protocols based on intravenous or oral delivery routes. Importantly, we were recently able to demonstrate that less frequent dosing of intravenously delivered IT848 was efficacious in the absence of toxicity (Supplementary Fig. S9), and additional studies with the goal to refine and optimize our protocols are currently underway.

Encouraged by the effectiveness of IT848 against multiple myeloma cell lines both in vitro and in vivo we sought to assess the response of primary multiple myeloma cells to IT848. We therefore performed apoptosis and proliferation assays with multiple myeloma samples derived from 2 patients with newly diagnosed multiple myeloma and 1 patient with highly aggressive advanced disease (plasma cell leukemia). These assays confirmed that IT848 induced cell death (Fig. 5D) and inhibited proliferation in a dose-dependent manner (Fig. 5E) not just in multiple myeloma cell lines but also primary multiple myeloma cells.

IT848 can be harnessed to overcome immunosuppression in the multiple myeloma microenvironment

Finally, given the known immunomodulatory effects of NF-κB inhibition, we investigated whether IT848 treatment could be exploited to maximize immunotherapeutic approaches in multiple myeloma. We previously demonstrated that small molecules inhibiting NF-κB DNA binding are active against alloimmunity (acute graft-versus-host disease) without compromising T cell–mediated protective immunity (21, 35). Furthermore, we recently discovered that our drug candidate IT848 was active against autoimmunity (rheumatoid arthritis model, Supplementary Fig. S10) and did not reduce the cytotoxic activity of CAR T cells (Supplementary Fig. S11). Because NF-κB regulates PD-L1 expression in B-cell lymphomas (8), we assessed PD-L1 levels in lymphoma and multiple myeloma cells treated with IT848 and found that IT848 treatment resulted in the downregulation of PD-L1 expression in both cell types (Supplementary Fig. S12), suggesting that IT848 might be a promising candidate for combination therapy with PD-1 blockade in B-cell malignancies. In addition to suppression of the PD-1/PD-L1 axis, NF-κB inhibition might enhance anti-myeloma immunity by modulating either myeloid-derived suppressor cells (MDSC) or regulatory T cells (Treg) in the bone marrow (BM) microenvironment in the setting of multiple myeloma (36–38), providing an additional rationale for combining IT848 with an immune checkpoint inhibitor strategy. To test our hypothesis we investigated the efficacy of IT848/anti–PD-1 combination both in vitro and in vivo in the immunocompetent mouse model 5TGM1 (16). In vitro coculture experiments of BM mononuclear cells and 5TGM1 cells in the presence and absence of IT848, anti–PD-1, or their combination, revealed that only combination treatment resulted in significant anti-myeloma activity (Fig. 6A). Furthermore, PD-1 expression was decreased on all T cells of the combination treatment group (Fig. 6B). While this decrease in PD-1 expression also included Tregs (Supplementary Fig. S13A), analysis of Ki-67 expression by PD-1–positive Tregs showed no evidence of Treg hyperproliferation (Supplementary Fig. S13A), a phenomenon that has been associated with cancer progression following anti–PD-1 therapy (39). Combination treatment also resulted in increased CD25 expression by cytotoxic T lymphocytes (CTL; Fig. 6B) and an increase in the frequency of activated CTLs (CD25+IFNγ+; Fig. 6B). We also analyzed immunosuppressive, Treg-promoting growth factors and cytokines such as TGFβ and IL10 (40). However, intracellular cytokine analysis of M-MDSCs and PMN-MDSCs showed overall no significant alterations in TGFβ and IL10 expression in any of the tested conditions (Supplementary Fig. S13B). We next compared the same conditions (control, IT848 monotherapy, anti–PD-1 monotherapy, and combination therapy) in the 5TGM1 model in vivo. We found that a 14-day treatment course of IT848/anti–PD-1 reduced multiple myeloma progression and improved progression free survival (Fig. 6C and D). Amongst immunosuppressive cell populations in the BM of multiple myeloma bearing mice, the number of Tregs (but not of MDSCs) was significantly decreased in mice receiving IT848/anti–PD-1 combination treatment (Fig. 6E, Supplementary Fig. S14), and CD25 expression of Tregs was lowest in Tregs of the combination therapy group (Fig. 6E), suggestive of decreased Treg-mediated immunosuppression of the TME. Collectively, our findings suggest that the observed therapeutic effect of IT848/anti–PD-1 combination therapy was at least partially due to modulation of the immunosuppressive BM microenvironment.

Figure 6.

Modulation of the BM microenvironment by IT848 can be exploited for combination therapies. A+B: 100,000 dtomato-expressing 5TGM1 cells were cultured with 900,000 BM mononuclear cells from C57BL/6 mice and treated with empty vehicle (control), IT848 (4 μmol/L), anti–PD-1 antibody (10 ug/mL), or IT848 + anti–PD-1 antibody. After 72 hours of coculture cells were analyzed by multiparameter flow cytometry. Mean and SEM of one of three independent experiments are presented (n = 5). A, left: dtomato+ percentage of CD45+ live cells; right: absolute number of live 5TGM1 cells. B, left: PD-1 median fluorescence intensity (MFI) of CD3+ Tells; center: CD25 MFI of CD8+ T cells; left: cells were gated on CD3+CD8+ T cells and analyzed for CD25+ IFNγ+ cells. C–F, C57Bl/KaLwRij mice received 1.5×106 luciferase expressing 5TGM1 cells intravenously. After 14 days engraftment was confirmed and mice were assigned to the following groups: 1. Empty vehicle I.P. 5x/week x 2 weeks; 2. IT848 10 mg/kg I.P. 5x/week x 2 weeks; 3. anti–PD-1 200ug I.P. 3x/week x 2 weeks; and 4. IT848 + anti–PD-1 combination therapy x 2 weeks. C, Multiple myeloma progression was analyzed by in vivo BLI. Mean and SEM of bioluminescence intensities are presented (n = 5). D, Progression-free survival is presented. Differences between groups were analyzed by Log-rank test. C+ D, One of two independent experiments is shown (n = 5). E, BM was harvested on day 14 and analyzed for the presence of Tregs. Left: Treg (CD45+CD3+CD4+CD25+FoxP3+) frequency of CD3+CD4+ T cells; Middle: Treg frequency of CD45+ BM cells; right: CD25 MFI of CD3+CD4+FoxP3+ T cells. Mean and SEM are presented. Combined data from three independent experiments are shown (n = 7–16). F, Schematic diagram of IT848-mediated effects on multiple myeloma cells and microenvironment. Three major mechanisms contributing to anti-myeloma activity of IT848 are depicted: Inhibition of NF-κB DNA binding in multiple myeloma cell nuclei, oxidative stress induced apoptosis of multiple myeloma cells and enhancement of immune checkpoint blockade by depletion of bone marrow Treg pool.

Figure 6.

Modulation of the BM microenvironment by IT848 can be exploited for combination therapies. A+B: 100,000 dtomato-expressing 5TGM1 cells were cultured with 900,000 BM mononuclear cells from C57BL/6 mice and treated with empty vehicle (control), IT848 (4 μmol/L), anti–PD-1 antibody (10 ug/mL), or IT848 + anti–PD-1 antibody. After 72 hours of coculture cells were analyzed by multiparameter flow cytometry. Mean and SEM of one of three independent experiments are presented (n = 5). A, left: dtomato+ percentage of CD45+ live cells; right: absolute number of live 5TGM1 cells. B, left: PD-1 median fluorescence intensity (MFI) of CD3+ Tells; center: CD25 MFI of CD8+ T cells; left: cells were gated on CD3+CD8+ T cells and analyzed for CD25+ IFNγ+ cells. C–F, C57Bl/KaLwRij mice received 1.5×106 luciferase expressing 5TGM1 cells intravenously. After 14 days engraftment was confirmed and mice were assigned to the following groups: 1. Empty vehicle I.P. 5x/week x 2 weeks; 2. IT848 10 mg/kg I.P. 5x/week x 2 weeks; 3. anti–PD-1 200ug I.P. 3x/week x 2 weeks; and 4. IT848 + anti–PD-1 combination therapy x 2 weeks. C, Multiple myeloma progression was analyzed by in vivo BLI. Mean and SEM of bioluminescence intensities are presented (n = 5). D, Progression-free survival is presented. Differences between groups were analyzed by Log-rank test. C+ D, One of two independent experiments is shown (n = 5). E, BM was harvested on day 14 and analyzed for the presence of Tregs. Left: Treg (CD45+CD3+CD4+CD25+FoxP3+) frequency of CD3+CD4+ T cells; Middle: Treg frequency of CD45+ BM cells; right: CD25 MFI of CD3+CD4+FoxP3+ T cells. Mean and SEM are presented. Combined data from three independent experiments are shown (n = 7–16). F, Schematic diagram of IT848-mediated effects on multiple myeloma cells and microenvironment. Three major mechanisms contributing to anti-myeloma activity of IT848 are depicted: Inhibition of NF-κB DNA binding in multiple myeloma cell nuclei, oxidative stress induced apoptosis of multiple myeloma cells and enhancement of immune checkpoint blockade by depletion of bone marrow Treg pool.

Close modal

Even though constitutive NF-κB activity is crucial to the survival and progression of multiple myeloma cells and plays a role in the development of resistance to cancer therapeutics, targeting NF-κB safely remains a challenging balancing act. In addition, the crosstalk between myeloma cells and the immune components of the TME within the BM further complicates the therapeutic design (41). As a result, even though hundreds of NF-κB inhibitor compounds have been designed over the years, to date there are no approved agents that directly inhibit NF-κB transactivation (42). However, multiple indirect strategies to target NF-κB, including agents that were not originally conceived as NF-κB inhibitors, have yielded some significant advances. Examples include immunomodulatory drugs (IMiD) such as lenalidomide that suppresses IκB kinase (IKK) activity (via TNFα, IL1β inhibition; ref. 43), and proteasome inhibitors such as bortezomib and carfilzomib that target IκBα degradation (30). These classes of drugs have been used in multiple myeloma and B-cell lymphoma but lack specificity, have off-target effects, and dose-limiting toxicities (31, 44). Inhibitors of a central regulator of NF-κB activation, the IKK complex (45), on the other hand, are highly NF-κB–specific, but their efficacy in clinical trials has been underwhelming (46). As far as pharmaceutical inhibition of NF-κB DNA binding is concerned, a limited number of molecules (DHMEQ, SEMBL, PBS-1086) derived from the natural compound epoxyquinomicin C have been researched in the past, but the poor bioavailability and PK restricted their use and potential for clinical translation (47–49). On the other hand, our synthetic compound IT848 is characterized by drug-like physicochemical properties. IT848 inhibits NF-κB transcriptional activity by reducing the ability of NF-κB subunits that migrated to the nucleus to bind to DNA, thereby interfering with both canonical and noncanonical NF-κB signaling. Importantly, during healthy steady state conditions, NF-κB is kept in an inactive state in the cytoplasm and as such, in the absence of NF-κB activating stimuli, healthy cells are not expected to be negatively affected by pharmaceuticals specifically preventing DNA binding of NF-κB. Although small molecules alone are unlikely to induce durable responses in aggressive cancers such as multiple myeloma, we demonstrated significant in vivo efficacy of IT848 monotherapy. The above-mentioned adverse events following prolonged courses of clinically irrelevant intraperitoneal administration of IT848 in mice (e.g., diarrhea, ascites) do not occur with intravenous or oral administrations; both administration routes are viable options, given the favorable PK profile of IT848 and its efficacy in recently used both oral and intravenous regimens in mouse models of T-cell lymphoma (Supplementary Fig. S9) and rheumatoid arthritis (Supplementary Fig. S10).

We found an additional mechanism of action of IT848 that exploits the heightened ROS production in cancer cells, a consequence of redox homeostasis imbalance, oncogenic stimulation, increased metabolic activity, and mitochondrial malfunction (50, 51). We observed a dose-dependent increase in superoxide levels and a decrease of GSH in multiple myeloma cell lines but not in healthy control cells where basal ROS levels are minimal. A previous study found a direct correlation between GSH depletion and ROS-mediated apoptosis of cancer cells (52). Notably, NF-κB activity is required for cellular GSH production (53), and GSH depletion mediated specifically by NF-κB inhibition has been proposed as a therapeutic strategy for cancer treatment (53). Our findings were further reiterated by RNA-seq and qPCR data supporting a transcriptional upregulation of oxidative stress signatures and genes involved in oxidative stress-induced apoptosis. Collectively, our data indicate that therapeutic manipulation by IT848 can preferentially kill myeloma cells via NF-κB/ROS-mediated mechanisms utilizing both caspase-dependent and independent apoptosis pathways. Induction of cancer cell–selective oxidative stress by IT848 is closely related to GSH depletion and inadequate upregulation of oxidative stress response genes, which can both be attributed to the NF-κB inhibitory effects of the molecule. This suggests that IT848 has the potential to be effective against a wide range of cancer cells and its clinical application may not have to be limited to cancer cell types that rely on constitutive NF-κB activity for their survival.

We showed that IT848 induces prominent anti-myeloma activity against cell lines, primary cells, and in a xenograft model, where combining IT848 with bortezomib enhanced antitumor efficacy. However, because tumor modeling in the absence of a functional immune system limits clinical relevance (especially when studying approaches that have the potential to alter immune responses), we additionally explored the effect of IT848 in combination with PD-1 blockade in an immunocompetent mouse model of multiple myeloma. It has been shown that malignant plasma cells are able to induce T-cell anergy via PD-L1/PD-1 interaction (54), and PD-L1 expression in multiple myeloma cells correlates with BCMA signaling, which activates the canonical NF-κB pathway (55). We found that single agent treatment with either anti–PD-1 checkpoint blockade or IT848 was not effective, but combination therapy significantly decreased infiltration of the BM by Tregs and multiple myeloma cells, and it improved survival. In vitro testing of this combination therapy confirmed that combination of direct NF-κB inhibition with immune checkpoint blockade induces a shift of the multiple myeloma microenvironment toward enhanced cytotoxicity. MDSC populations were not quantitatively affected by IT848 or anti–PD-1 treatment, and there was no evidence of a role of MDSCs in promoting Treg activity through cytokine release. However, we found that IT848/anti–PD-1 combination treatment resulted in significant downregulation of the IL2 receptor alpha chain (CD25), which is an NF-κB target gene (56), in Tregs, suggesting that decreased responsiveness to the Treg growth factor IL2 may be a contributing factor for the depletion of BM resident Tregs (57). In addition, IT848 likely suppresses thymic Treg development, given that NF-κB regulates FoxP3 expression (58). Finally, it is well established that immune checkpoint blockade can be associated with autoreactive T cell–mediated adverse events such as colitis or hepatitis. Our previous findings in GVHD (35) and rheumatoid arthritis models suggest that IT848 treatment might ameliorate autoimmune-related adverse events associated with immune checkpoint blockade (59) without compromising antitumor activity of T cells (21, 35), providing an additional rationale for the development of IT848 combination therapies with immune checkpoint blockade. Taken together, these cellular and molecular interactions suggest that while high-dose IT848 displays antitumor activity in the absence of a functional immune system, harnessing the immunomodulary effects of IT848 for cancer immunotherapy promises synergistic therapeutic effects at lower doses in the absence of toxicities. Studies designed to further validate the latter concept are currently underway.

In conclusion, our study shows that the small molecule IT848 is a novel and highly specific type of NF-κB inhibitor and cancer cell–selective oxidative stress inducer with significant anti-myeloma activity against both cell lines and primary cells. IT848 demonstrated in vivo efficacy as single agent and in combination therapies designed to overcome immunosuppression and drug resistance in the TME. IT848 has a favorable PK profile and we found no toxicities associated with clinically relevant administration routes. Our data therefore provide a strong preclinical rationale for the clinical development of IT848 in B-cell malignancies.

I. Colorado reports grants from IMS during the conduct of the study. R. Feinman reports grants from International Myeloma Society during the conduct of the study. D.S. Siegel reports other support from BMS, Takeda, GSK, Merck, Janssen, Amgen, Karyopharm, IMF, Celularity, Neximmune; and other support from Sanofi outside the submitted work. J.L. Zakrzewski reports grants from ImmuneTarget Inc., Leukemia & Lymphoma Society; and grants from International Myeloma Society during the conduct of the study. No disclosures were reported by the other authors.

M. Bariana: Data curation, formal analysis, validation, investigation, visualization, methodology, writing–original draft. E. Cassella: Data curation, formal analysis, methodology. J. Rateshwar: Data curation, formal analysis, methodology. S. Ouk: Resources, data curation, investigation. H.-C. Liou: Resources, data curation, investigation. C. Heller: Data curation, investigation. I. Colorado: Investigation. R. Feinman: Resources, supervision, investigation, methodology. A. Makhdoom: Data curation, investigation. D.S. Siegel: Resources, project administration. G. Heller: Formal analysis, methodology. A. Tuckett: Data curation, formal analysis, investigation. P. Mondello: Data curation, formal analysis, supervision, investigation, methodology, writing–review and editing. J.L. Zakrzewski: Conceptualization, resources, formal analysis, supervision, funding acquisition, validation, project administration, writing–review and editing.

This study was supported by a Sponsored Research Agreement from ImmuneTarget Inc. to J.L. Zakrzewski. J.L. Zakrzewski also received grant support from the NCI (NCI 1R37CA250661-01A1), the Leukemia and Lymphoma Society (grant # 6465-15), the American Society of Hematology, the International Myeloma Society, Hyundai Hope on Wheels, the HMH MSK Immunology Collaboration, and the HMH Foundation/Tackle Kids Cancer. IT848 was provided by Immunetarget, Inc.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

1.
Smith
D
,
Yong
K
.
Multiple myeloma
.
BMJ
2013
;
346
:
f3863
.
2.
Highsmith
KN
,
Chen
SE
,
Horowitz
S
.
Carfilzomib and pomalidomide: recent advances in the treatment of multiple myeloma
.
Pharmacotherapy
2014
;
34
:
927
40
.
3.
Yu
B
,
Jiang
T
,
Liu
D
.
BCMA-targeted immunotherapy for multiple myeloma
.
J Hematol Oncol
2020
;
13
:
125
.
4.
Taniguchi
K
,
Karin
M
.
NF-κB, inflammation, immunity, and cancer: coming of age
.
Nat Rev Immunol
2018
;
18
:
309
24
.
5.
Demchenko
YN
,
Kuehl
WM
.
A critical role for the NFkB pathway in multiple myeloma
.
Oncotarget
2010
;
1
:
59
68
.
6.
Feinman
R
,
Koury
J
,
Thames
M
,
Barlogie
B
,
Epstein
J
,
Siegel
DS
.
Role of NF-κB in the rescue of multiple myeloma cells from glucocorticoid-induced apoptosis by Bcl-2
.
Blood
1999
;
93
:
3044
52
.
7.
Godwin
P
,
Baird
AM
,
Heavey
S
,
Barr
MP
,
O'Byrne
KJ
,
Gately
K
.
Targeting nuclear factor-kappa B to overcome resistance to chemotherapy
.
Front Oncol
2013
;
3
:
120
.
8.
Vincent-Fabert
C
,
Roland
L
,
Zimber-Strobl
U
,
Feuillard
J
,
Faumont
N
.
Preclinical blocking of PD-L1 molecule, which expression is down regulated by NF-κB, JAK1/JAK2, and BTK inhibitors, induces regression of activated B-cell lymphoma
.
Cell Commun Signal
2019
;
17
:
89
.
9.
Freeman
GJ
,
Long
AJ
,
Iwai
Y
,
Bourque
K
,
Chernova
T
,
Nishimura
H
, et al
.
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation
.
J Exp Med
2000
;
192
:
1027
34
.
10.
Paiva
B
,
Azpilikueta
A
,
Puig
N
,
Ocio
EM
,
Sharma
R
,
Oyajobi
BO
, et al
.
PD-L1/PD-1 presence in the tumor microenvironment and activity of PD-1 blockade in multiple myeloma
.
Leukemia
2015
;
29
:
2110
3
.
11.
Ribrag
V
,
Avigan
DE
,
Green
DJ
,
Wise-Draper
T
,
Posada
JG
,
Vij
R
, et al
.
Phase 1b trial of pembrolizumab monotherapy for relapsed/refractory multiple myeloma: KEYNOTE-013
.
Br J Haematol
2019
;
186
:
e41
e4
.
12.
Mateos
MV
,
Blacklock
H
,
Schjesvold
F
,
Oriol
A
,
Simpson
D
,
George
A
, et al
.
Pembrolizumab plus pomalidomide and dexamethasone for patients with relapsed or refractory multiple myeloma (KEYNOTE-183): a randomized, open-label, phase III trial
.
Lancet Haematol
2019
;
6
:
e459
e69
.
13.
Badros
A
,
Hyjek
E
,
Ma
N
,
Lesokhin
A
,
Dogan
A
,
Rapoport
AP
, et al
.
Pembrolizumab, pomalidomide, and low-dose dexamethasone for relapsed/refractory multiple myeloma
.
Blood
2017
;
130
:
1189
97
.
14.
Costello
C
.
The future of checkpoint inhibition in multiple myeloma?
Lancet Haematol
2019
;
6
:
e439
-e40.
15.
Ouk
S
,
Liou
H-c
.
Methods of treating cancer with small molecule NF-kB inhibitors
.
US Patent App. 16/020,688
;
2019
.
16.
Garrett
I
,
Dallas
S
,
Radl
J
,
Mundy
GR
.
A murine model of human myeloma bone disease
.
Bone
1997
;
20
:
515
20
.
17.
Bronsart
L
,
Nguyen
L
,
Habtezion
A
,
Contag
C
.
Reactive oxygen species imaging in a mouse model of inflammatory bowel disease
.
Mol Imaging Biol
2016
;
18
:
473
8
.
18.
Egan
LJ
,
Toruner
M
.
NF-kappaB signaling: pros and cons of altering NF-kappaB as a therapeutic approach
.
Ann NY Acad Sci
2006
;
1072
:
114
22
.
19.
Li
B
,
Dewey
CN
.
RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome
.
BMC Bioinf
2011
;
12
:
1
16
.
20.
Kakoki
M
,
Tsai
Y-S
,
Kim
H-S
,
Hatada
S
,
Ciavatta
DJ
,
Takahashi
N
, et al
.
Altering the expression in mice of genes by modifying their 3′ regions
.
Dev Cell
2004
;
6
:
597
606
.
21.
Shono
Y
,
Tuckett
AZ
,
Liou
HC
,
Doubrovina
E
,
Derenzini
E
,
Ouk
S
, et al
.
Characterization of a c-Rel inhibitor that mediates anticancer properties in hematologic malignancies by blocking NF-κB–controlled oxidative stress responses
.
Cancer Res
2016
;
76
:
377
89
.
22.
Meng
XY
,
Zhang
HX
,
Mezei
M
,
Cui
M
.
Molecular docking: a powerful approach for structure-based drug discovery
.
Curr Comput Aided Drug Des
2011
;
7
:
146
57
.
23.
Gadó
K
,
Domján
G
,
Hegyesi
H
,
Falus
A
.
Role of interleukin 6 in the pathogenesis of multiple myeloma
.
Cell Biol Int
2000
,
24
:
195
209
.
24.
Matthes
T
,
Manfroi
B
,
Huard
B
.
Revisiting IL6 antagonism in multiple myeloma
.
Crit Rev Oncol Hematol
2016
;
105
:
1
4
.
25.
Demchenko
YN
,
Glebov
OK
,
Zingone
A
,
Keats
JJ
,
Bergsagel
PL
,
Kuehl
WM
.
Classical and/or alternative NF-kappaB pathway activation in multiple myeloma
.
Blood
2010
;
115
:
3541
52
.
26.
Jo
WS
,
Jeong
MH
,
Jin
YH
,
Jang
JY
,
Nam
BH
,
Son
SH
, et al
.
Loss of mitochondrial membrane potential and caspase activation enhance apoptosis in irradiated K562 cells treated with herbimycin A
.
Int J Radiat Biol
2005
;
81
:
531
43
.
27.
Candé
C
,
Cohen
I
,
Daugas
E
,
Ravagnan
L
,
Larochette
N
,
Zamzami
N
, et al
.
Apoptosis-inducing factor (AIF): a novel caspase-independent death effector released from mitochondria
.
Biochimie
2002
;
84
:
215
22
.
28.
Liu
W
,
Ji
Y
,
Sun
Y
,
Si
L
,
Fu
J
,
Hayashi
T
, et al
.
Estrogen receptors participate in silibinin-caused nuclear translocation of apoptosis-inducing factor in human breast cancer MCF-7 cells
.
Arch Biochem Biophys
2020
;
689
:
108458
.
29.
Bronsart
L
,
Nguyen
L
,
Habtezion
A
,
Contag
C
.
Reactive oxygen species imaging in a mouse model of inflammatory bowel disease
.
Mol Imaging Biol
2016
;
18
:
473
8
.
30.
Chen
D
,
Frezza
M
,
Schmitt
S
,
Kanwar
J
,
Dou
Q
.
Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives
.
Curr Cancer Drug Targets
2011
;
11
:
239
53
.
31.
Obeng
EA
,
Carlson
LM
,
Gutman
DM
,
Harrington
WJ
,
Lee
KP
,
Boise
LH
.
Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells
.
Blood
2006
;
107
:
4907
16
.
32.
Scata
KA
,
El-Deiry
WS
.
Taming NEMO to slay cancer cells
.
Cancer Biol Ther
2006
;
5
:
1096
7
.
33.
Rhodes
CA
,
Dougherty
PG
,
Cooper
JK
,
Qian
Z
,
Lindert
S
,
Wang
Q-E
, et al
.
Cell-permeable bicyclic peptidyl inhibitors against NEMO-IκB kinase interaction directly from a combinatorial library
.
J Am Chem Soc
2018
;
140
:
12102
10
.
34.
Yu
Z
,
Gao
J
,
Zhang
X
,
Peng
Y
,
Wei
W
,
Xu
J
, et al
.
Characterization of a small molecule inhibitor targeting NEMO/IKKβ to suppress colorectal cancer growth
.
Signal Transduct Target Ther
2022
;
7
:
1
13
.
35.
Shono
Y
,
Tuckett
AZ
,
Ouk
S
,
Liou
HC
,
Altan-Bonnet
G
,
Tsai
JJ
, et al
.
A small molecule c-Rel inhibitor reduces alloactivation of T cells without compromising antitumor activity
.
Cancer Discov
2014
;
4
:
578
91
.
36.
Malek
E
,
de Lima
M
,
Letterio
JJ
,
Kim
BG
,
Finke
JH
,
Driscoll
JJ
, et al
.
Myeloid-derived suppressor cells: the green light for myeloma immune escape
.
Blood Rev
2016
;
30
:
341
8
.
37.
Serafini
P
,
Mgebroff
S
,
Noonan
K
,
Borrello
I
.
Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells
.
Cancer Res
2008
;
68
:
5439
49
.
38.
Condamine
T
,
Gabrilovich
DI
.
Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function
.
Trends Immunol
2011
;
32
:
19
25
.
39.
Kamada
T
,
Togashi
Y
,
Tay
C
,
Ha
D
,
Sasaki
A
,
Nakamura
Y
, et al
.
PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer
.
Proc Natl Acad Sci USA
2019
;
116
:
9999
10008
.
40.
Taylor
A
,
Verhagen
J
,
Blaser
K
,
Akdis
M
,
Akdis
CA
.
Mechanisms of immune suppression by interleukin 10 and transforming growth factor-beta: the role of T regulatory cells
.
Immunology
2006
;
117
:
433
42
.
41.
Wong
AH-H
,
Shin
EM
,
Tergaonkar
V
,
Chng
WJ
.
Targeting NF-κB signaling for multiple myeloma
.
Cancers
2020
;
12
:
2203
.
42.
Gilmore
T
,
Herscovitch
M
.
Inhibitors of NF-κB signaling: 785 and counting
.
Oncogene
2006
;
25
:
6887
99
.
43.
Keifer
JA
,
Guttridge
DC
,
Ashburner
BP
,
Baldwin
AS
Jr
.
Inhibition of NF-κB activity by thalidomide through suppression of IκB kinase activity
.
J Biol Chem
2001
;
276
:
22382
7
.
44.
Vrábel
D
,
Pour
L
,
Ševčíková
S
.
The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma
.
Blood Rev
2019
;
34
:
56
66
.
45.
Israël
A
.
The IKK complex, a central regulator of NF-kappaB activation
.
Cold Spring Harb Perspect Biol
2010
;
2
:
a000158
.
46.
Grothe
K
,
Flechsenhar
K
,
Paehler
T
,
Ritzeler
O
,
Beninga
J
,
Saas
J
, et al
.
IκB kinase inhibition as a potential treatment of osteoarthritis–results of a clinical proof-of-concept study
.
Osteoarthritis Cartilage
2017
;
25
:
46
52
.
47.
Sidthipong
K
,
Ma
J
,
Yu
WL
,
Wang
YF
,
Kobayashi
S
,
Kishino
S
, et al
.
Rational design, synthesis and in vitro evaluation of novel exo-methylene butyrolactone salicyloylamide as NF-κB inhibitor
.
Bioorg Med Chem Lett
2017
;
27
:
562
6
.
48.
Fabre
C
,
Mimura
N
,
Bobb
K
,
Gorgun
G
,
Cirstea
DD
,
Hu
Y
, et al
.
Dual inhibition of canonical and noncanonical NF-KB pathway demonstrates significant antitumor activities in multiple myeloma (MM)
.
Clin Cancer Res
2012
;
18
:
4669
81
.
49.
Lin
Y
,
Ukaji
T
,
Koide
N
,
Umezawa
K
.
Inhibition of late and early phases of cancer metastasis by the NF-κB inhibitor DHMEQ derived from microbial bioactive metabolite epoxyquinomicin: a review
.
Int J Mol Sci
2018
;
19
:
729
.
50.
Pelicano
H
,
Carney
D
,
Huang
P
.
ROS stress in cancer cells and therapeutic implications
.
Drug Resist Updat
2004
;
7
:
97
110
.
51.
Trachootham
D
,
Alexandre
J
,
Huang
P
.
Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?
Nat Rev Drug Discov
2009
;
8
:
579
91
.
52.
Tang
H
,
Li
C
,
Zhang
Y
,
Zheng
H
,
Cheng
Y
,
Zhu
J
, et al
.
Targeted manganese doped silica nano GSH-cleaner for treatment of liver cancer by destroying the intracellular redox homeostasis
.
Theranostics
2020
;
10
:
9865
87
.
53.
Meng
Q
,
Peng
Z
,
Chen
L
,
Si
J
,
Dong
Z
,
Xia
Y
.
Nuclear Factor-κB modulates cellular glutathione and prevents oxidative stress in cancer cells
.
Cancer Lett
2010
;
299
:
45
53
.
54.
Rosenblatt
J
,
Avigan
D
.
Targeting the PD-1/PD-L1 axis in multiple myeloma: a dream or a reality?
Blood
2017
;
129
:
275
9
.
55.
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
.
56.
Ballard
DW
,
Böhnlein
E
,
Lowenthal
JW
,
Wano
Y
,
Franza
BR
,
Greene
WC
.
HTLV-I tax induces cellular proteins that activate the kappa B element in the IL2 receptor alpha gene
.
Science
1988
;
241
:
1652
5
.
57.
Chinen
T
,
Kannan
AK
,
Levine
AG
,
Fan
X
,
Klein
U
,
Zheng
Y
, et al
.
An essential role for the IL2 receptor in T(reg) cell function
.
Nat Immunol
2016
;
17
:
1322
33
.
58.
Long
M
,
Park
SG
,
Strickland
I
,
Hayden
MS
,
Ghosh
S
.
Nuclear factor-kappaB modulates regulatory T
-
cell development by directly regulating expression of Foxp3 transcription factor
.
Immunity
2009
;
31
:
921
31
.
59.
Weinmann
SC
,
Pisetsky
DS
.
Mechanisms of immune-related adverse events during the treatment of cancer with immune checkpoint inhibitors
.
Rheumatology
2019
;
58
:
vii59
67
.