Abstract
Despite substantial recent advances in treatment, multiple myeloma (MM) remains an incurable disease, with a shortage of treatment options for patients with high-risk disease, warranting the need for novel therapeutic targets and treatment approaches. Threonine and tyrosine kinase (TTK), also known as monopolar spindle 1 (MPS1), is a kinase essential for the mitotic spindle checkpoint whose expression correlates to unfavorable prognosis in several cancers. Here, we report the importance of TTK in MM, and the effects of the TTK inhibitor OSU-13. Elevated TTK expression correlated with amplification/ gain of 1q21 and decreased overall and event-free survival in MM. Treatment with OSU-13 inhibited TTK activity efficiently and selectively at a similar concentration range to other TTK inhibitor clinical candidates. OSU-13 reduced proliferation and viability of primary human MM cells and cell lines, especially those with high 1q21 copy numbers, and triggered apoptosis through caspase 3 and 7 activation. In addition, OSU-13 induced DNA damage and severe defects in chromosome alignment and segregation, generating aneuploidy. In vivo, OSU-13 decreased tumor growth in mice with NCI-H929 xenografts. Collectively, our findings reveal that inhibiting TTK with OSU-13 is a potential therapeutic strategy for MM, particularly for a subset of high-risk patients with poor outcome.
Introduction
Multiple myeloma (MM) is a plasma cell disorder that accounts for more than 10% of hematologic cancers.1 Despite the recent improvement in overall survival of patients with MM due to novel treatment options,2,3 MM is still a mostly incurable cancer,4 warranting the need for novel therapeutic targets, especially for patients with high-risk disease. The presence of genetic alterations is an important hallmark of cancer.5,6 In MM, structural and numerical genetic abnormalities are usually associated with disease development and progression.7 Aneuploidy is present in about 70% of MM cases.8 In addition, deletions, duplications, or translocations are very common events in MM,9,10 some of which - t(4;14)(p16;q32), t(14;16)(q32;q23), and del(17p) - are considered poor prognostic indicators.11 Possible mechanisms leading to genomic instability in MM include elevated homologous recombination activity, which causes an increased mutation rate and accumulation of genetic variation,12 and centrosome amplification, which is frequent in MM and has been associated with high-risk disease and poor prognosis.13
Adequate chromosome segregation is essential for genomic stability and relies on a group of proteins known as the spindle assembly checkpoint (SAC).14 The SAC blocks cell cycle progression until the chromosomes are correctly attached to the spindle microtubules.15 Failure of this process can cause uneven chromosomal segregation and aneuploidy, resulting in chromosomal instability.16 Manipulation of SAC proteins leads to tumor formation in animal models.17,18 Accordingly, drugs that interfere with SAC, such as inhibitors of Aurora kinases, Polo-like kinases, CENP-E, and threonine and tyrosine kinase (TTK) have been investigated or are being tested in clinical trials for several cancers including MM.19-21 TTK, also known as monopolar spindle 1 (MPS1), participates in the regulation of the DNA damage checkpoint response,22 centrosome duplication,23 and mitosis arrest until proper chromosome alignment, playing an essential role in SAC.24 High levels of TTK expression correlate to unfavorable prognosis in several cancers,25-27 and TTK inhibition shows potential utility for the treatment of glioma,28 melanoma,29 and colon,30 breast,31,32 lung,32 ovarian,29,32 and cervical33 cancers. Consequently, many clinical trials have tested TTK inhibitors - either as monotherapy or combinations - for the treatment of solid tumors, especially breast cancer. However, to date, there have been no clinical trials with TTK inhibitors involving patients with any kind of hematologic malignancies.
In 2017, the TTK/MPS1 inhibitor OSU-13 was identified as a potential therapy for breast cancer.31 Here, we provide new data establishing the relevance of TTK expression in MM prognosis and perform the first comprehensive study using a TTK/MPS1 inhibitor as a therapeutic strategy for a hematologic malignancy.
Methods
OSU-13 drug
OSU-13 was provided by the Drug Development Institute of the Ohio State University and stored at -20°C at 10 mM in dimethyl sulfoxide (DMSO).
Prognostic survival analysis
Overall survival and event-free survival outcome studies were performed in 769 patients using the GSE2658 dataset. The median value of TTK levels was used as a cut-off to define patients with low (n=386) or high (n=383) TTK expression. Prognostic value of TTK was evaluated by the Kaplan-Meier curve, obtained from www.canevolve.org. TTK expression analysis in high-risk MM with specific genetic alterations (17p del, t(4;14), t(11;14), t(8;14), and 1q21 gain) was performed in the MMRF CoMMpass database.
Viability, apoptosis and necroptosis assays
For viability measurement, cells were plated at 100,000 cells/mL in medium containing DMSO or 0.5 mM OSU-13 and incubated for 24, 48, and 72 hours (h) at 37°C and 5% CO2. Afterwards, cells were stained with Zombie-aqua (Life Technologies; Carlsbad, CA, USA) according to manufacturer’s instructions. Cells were then analyzed using an Attune Nxt cytometer (Invitrogen; Waltham, MA, USA), and FlowJo software (FlowJo LLC; Ashland, OR, USA).
For apoptosis and necroptosis inhibition assays, OPM-2 cells (250,000 cells/mL) were pre-incubated for 1 h at 37oC in medium containing 100 mM of the general caspase inhibitor Z-VAD-FMK (Sigma-Aldrich; St Louis, MO, USA) or the necroptosis inhibitor necrostatin-1s (Cell Signaling Inc.; Danvers, MA, USA). Then, cells were washed and incubated for 72 h in medium containing DMSO or 1 mM OSU-13, stained with Zombie-aqua (Life Technologies), and cell viability was assessed as previously described.
Fluorescence microscopy of chromosome segregation
OPM-2 cells (1x106) were synchronized at G1/S by double thymidine block. Cells were incubated for 16 h in medium containing 2 mM thymidine (Sigma-Aldrich), released for 8 h in fresh medium, and incubated in 2 mM thymidine for additional 16 h. Next, cells were washed, and cell cycle release was induced with 24 mM 2’-deoxycytidine (Sigma-Aldrich) plus DMSO or 1 mM OSU-13 for 9 h. Cells were resuspended in 100 mL phosphate-buffered saline (PBS), immobilized onto a microscope slide using a Cytospin (Thermo Fisher Scientific; Waltham, MA, USA), mounted with Prolong Glass Antifade Mountant with NucBlue (Thermo Fisher Scientific), and imaged using a Nikon DM5000 B microscope (Tokyo, Japan) equipped with fluorescence optics with a Leica X63 oil immersion objective (Wetzlar, Germany). Images were analyzed using ImageJ (NIH; Bethesda, MD, USA), and the percentage of anaphase and telophase cells with and without lagging chromosomes was manually calculated.
Statistical considerations
Unless specified otherwise, all data are presented as mean values ± standard deviation (SD) from independent experiments. Student’s t test was used to evaluate differences between conditions with P value <0.05 considered to be statistically significant. *P<0.05, **P<0.01, ***P<0.001 and ****P<0.0001. The correlation between sensitivity to OSU-13 and 1q21 copy number in the cell lines was analyzed using the non-parametric Spearman’s rank correlation coefficient.
Additional methods
Cell lines, primary cell experiments, small interfering RNA (siRNA) knockdown, real-time polymerase chain reaction (PCR), NanoBRET™, kinase profiling, co-crystallization, cell viability and proliferation assays, caspases 3 and 7 activity assay, western blotting, TUNEL, cell cycle analysis, metaphase chromosome spread, and studies in murine model are described in the Online Supplementary Appendix. Experiments involving human subject samples were conducted with the approval of The Ohio State University Institutional Review Board (IRB 2023C0065).
Results
TTK expression correlates with multiple myeloma prognosis and plays a role in the survival of multiple myeloma cell lines
High TTK expression correlates to unfavorable prognosis in several cancers.25-28,34 In order to determine the relevance of TTK expression in MM prognosis, we examined the GSE2658 dataset and the CoMMpass database, analyzing the association of TTK expression with clinical outcome and chromosomal alterations in MM patients (n=769). We found that elevated TTK expression correlated with decreased overall (P=0.0001) and event-free (P<0.0001) survival (Figure 1A, B). Furthermore, TTK expression was higher in patients with amplification or gain of 1q21, a genetic alteration found in high-risk MM (P<0.0001) (Figure 1C). No correlation was seen between TTK expression and other high-risk genetic alterations, such as deletion of 17p or t(4;14), t(8;14), or t(11;14) translocations (Online Supplementary Figure S1).
We also compared TTK expression levels in eight human MM cell lines and primary CD138+ plasma cells isolated from bone marrow (BM) of four recently diagnosed, untreated MM patients (Figure 1D). MM cell lines exhibited higher TTK expression than primary CD138+ cells. MM.1S showed the highest expression (16.2-fold higher than CD138+ cells), whereas L363 had the lowest expression (3.2-fold increase). In order to investigate the role of TTK in MM cell lines, we specifically knocked down TTK expression in OPM-2 and NCI-H929 using Alexa Fluor 647-conjugated non-targeting scrambled or TTK-specific siRNA (Online Supplementary Figure S2). Streptolysin was used to facilitate siRNA uptake, and cell viability was evaluated using Zombie Aqua staining. Knock-down of TTK was confirmed by quantitative PCR (data not shown). Our results exhibited a significant decrease in viability in TTK-siRNA+ cells compared to cells transfected with non-targeting scrambled siRNA in both cell lines, with a more pronounced effect in NCI-H929. Following treatment with 20 units (U) of streptolysin, 51±5.8% of TTK-siRNA+ NCI-H929 cells were dead (Zombie+), in contrast to only 16±6.2% of scrambled siRNA+ cells (P<0.001). These findings highlight the critical role of TTK in MM cell survival and suggest that targeting TTK could serve as a potential therapeutic strategy for MM.
OSU-13 efficiently and selectively inhibits TTK
In order to analyze the interaction between TTK and its novel small molecule inhibitor OSU-13, their co-crystal structure was obtained at a 2.37 Å resolution (Figure 2A). We determined that OSU-13 binds to the ATP-binding pocket, forming multiple interactions with the protein kinase hinge region, P-loop, and A-loop. OSU-13 inserts between Val539, Ala551 and Ile531 on one side and Leu654, Ile663 and Ile586 on the other, as well as the gatekeeper residue Met602 thiol deep in the pocket. The pyrrolopyrimidine bicyclic scaffold donates a H-bond to the Glu603 main chain carbonyl oxygen and accepts a H-bond from the Gly605 amide nitrogen. Additionally, OSU-13 forms a H-bond with the Gly605 main chain carbonyl oxygen and establishes van der Waals contacts with the ribose-binding pocket and the solvent-exposed channel.
OSU-13’s activity was initially measured by a TTK target engagement assay (NanoBRET™). We compared its relative inhibition of TTK-NanoLuc binding to a fluorescent tracer with other reported TTK inhibitors. OSU-13 showed inhibition within the same concentration range as other compounds, with a half maximal effective concentration (EC50) =10 nM, nearly 10-fold lower than the clinical candidate CFI-402257 (EC50=94.3 nM) (Figure 2B). We also verified that OSU-13 inhibits both mouse and human TTK at physiologic (1 mM) and low (10 μM) ATP concentrations, demonstrating activity comparable to the clinical candidate BOS-172722 (Figure 2C, D). Additionally, OSU-13 showed high stability, with a half-life exceeding 90 minutes in human hepatocyte stability assays.
In order to evaluate its selectivity, OSU-13 was profiled against a panel of human kinases in a cell-free kinase activity inhibition assay (Online Supplementary Table S1). Among the 399 kinases tested, only seven showed >80% inhibition when incubated with 1 mM OSU-13. Of these, only LRRK2 exhibited a half maximal inhibitory concentration (IC50) in a similar concentration range to TTK/MPS1 (7.5 nM vs. 4.3 nM), whereas the other kinases (ALK, IR, LTK, INSRR, and FAK) had IC50 values at least 36-fold higher than TTK/MPS1 (Table 1). Importantly, OSU-13 did not significantly inhibit other mitosis-related kinases, such as Aurora kinase, PLK, or Cyclin-dependent kinase family members.
Overall, these results indicate that OSU-13 is a biologically stable molecule that selectively and efficiently inhibits TTK kinase activity, comparable to other TTK/MPS1 inhibitor clinical candidates.
OSU-13 selectively decreases proliferation and viability of multiple myeloma cells
We investigated the effects of OSU-13 on primary cells derived from the bone marrow of a MM patient and the peripheral blood mononuclear cells (PBMC) of a plasma cell leukemia (PCL) patient. Compared to DMSO, OSU-13 treatment for 72 h reduced viable CD138+ plasma cells from MM and PCL patients by 44% and 18%, respectively (Figure 3A). Only a small effect was observed in viable B cells (reduced by 37% in MM and 6% in PCL) and natural killer (NK) cells (reduced by 11% in MM and 1% in PCL). No significant effect was observed in other lymphocyte populations (Online Supplementary Figure S3A). Similarly, PBMC from three healthy donors showed only a 22% decrease in viable B cells in one of the three donors after OSU-13 treatment (Online Supplementary Figure S3B). These findings suggest that OSU-13 displays relative selectivity for plasma cells, with mild effects on B-cell lineage lymphocytes and no substantial adverse effects on other PBMC populations. In order to study OSU-13’s effects on human MM cell lines, we evaluated proliferation and viability. Cells were treated with various concentrations of OSU-13 or DMSO, and cell division was monitored using a dye dilution experiment (Figure 3B). OSU-13 decreased proliferation of OPM-2 and NCI-H929 cell lines in a dose-dependent manner. At 5 mM OSU-13, proliferation decreased to 54±10% and 49±4% compared to DMSO control in OPM-2 and NCI-H929, respectively (P<0.01). RPMI-8226 and U266 showed a less pronounced effect on proliferation (data not shown).
The cytotoxic potential of OSU-13 was evaluated via MTS assay in nine human MM cell lines with different 1q21 copy numbers (Table 2). L363 was the most sensitive (IC50=132±1 nM) cell line, whereas RPMI-8226 was the least sensitive (IC50=10,448±1,184 nM) to OSU-13. Further analysis revealed a significant inverse correlation (rs=-0.76; P=0.017) between the number of 1q21 copies and the in vitro IC50 of OSU-13. MM cell lines with a higher number of 1q21 copies exhibited increased sensitivity to OSU-13. This finding suggests that OSU-13, through TTK inhibition, is particularly effective in a subgroup of high-risk MM patients with a poor prognosis.
Next, we assessed viability of NCI-H929 and OPM-2 cells using Zombie staining (Figure 3C). DMSO-treated cells showed an increase in viable cell numbers over time, while OSU-13-treated cells remained constant. After 72 h, the quantity of viable DMSO-treated cells was 5-fold and 3.6-fold higher (P<0.05) than OSU-13-treated cells in OPM-2 and NCI-H929, respectively (Figure 3C, left). The percentage of dead cells (Zombie+) peaked after 72 h, reaching 35±4.5% of OPM-2 (P<0.01) and 21±6.3% of NCI-H929 cells (P<0.05). Meanwhile, DMSO-treated cells did not show increase in Zombie+ cells over time (Figure 3C, right). Moreover, some dead cells turned into debris, resulting in a smaller Zombie+ population than expected.
OSU-13 induces apoptosis in human multiple myeloma cell lines through caspase activation
In order to investigate whether OSU-13-induced cell death was apoptotic, caspase 3 and 7 activity was assessed in NCI-H929 cells at different time points after treatment with varying concentrations of OSU-13. Caspase 3 and 7 activation started at 24 h with 0.1 mM (P<0.05) and peaked after 72 h with 0.5 mM OSU-13 (P<0.0001) (Figure 3D). Accordingly, western blot analysis of cell lysates showed increased levels of cleaved caspases 3 and 7 in OPM-2 and NCI-H929 cells after 72 h of OSU-13 treatment (Figure 3E), and pretreatment with the pan-caspase inhibitor Z-VAD-FMK decreased OSU-13-mediated cell death by 20±6.7% in OPM-2 cells (Figure 3F). In contrast, pretreatment with the necroptosis inhibitor Necrostatin-1s had no measurable effect on cell death, and there was no increase in the autophagy marker LC3B following OSU-13 treatment (Online Supplementary Figure S4A, B, respectively).
Collectively, these results indicate that OSU-13 triggers cell death in MM cell lines through caspase activation, and that apoptosis - but not necroptosis or autophagy - is partially responsible for this OSU-13-mediated cell death.
OSU-13 induces DNA damage in multiple myeloma cell lines
Apoptosis has been reported to induce DNA fragmentation. In order to measure internucleosomal DNA degradation, we treated OPM-2 and NCI-H929 cells with various concentrations of OSU-13 for 48 h and performed TUNEL analysis. DNA damage increased in a dose-dependent manner in both cell lines, as indicated by the correlation between OSU-13 concentration and percentage of TUNEL+ cells (Figure 3G, H). In OPM-2 cells, TUNEL positivity rates were 22±5.4%, 25±3.6% (P<0.05), and 43±3.1% (P<0.05) with 2.5 mM, 5 mM, and 10 mM OSU-13 treatment, respectively, compared to DMSO control (TUNEL+ =7±0.4%). In NCI-H929, TUNEL positivity increased with 2.5 mM OSU-13 treatment (P<0.05) (Figure 3H). Western blot analysis confirmed the presence of DNA damage, showing increased levels of p-H2AX and cleaved PARP1 after 72 hours of OSU-13 treatment (Figure 3I).
OSU-13 interferes in the cell cycle and ploidy of multiple myeloma cell lines
TTK/MPS1 inhibitors affect the cell cycle in a variety of cancer cells.30,33,35 In our study, treatment with OSU-13 induced significant alterations in DNA content in OPM-2 cells, with an increase in tetraploid (4N) and hyperploid (>4N) cells (Figure 4A). Accordingly, flow cytometric analysis revealed a substantial increase in the proportion of OPM-2 cells in the G2 phase (4N) after OSU-13 treatment, with a 1.9-fold increase compared to DMSO-treated cells (P<0.05). Additionally, there was a higher proportion of cells in the >G2 phase (>4N), although this difference was not statistically significant (Figure 4B).
This increase in the hyperploid population was confirmed by fluorescence microscopy of OPM-2 chromosome spreads (Figure 4C, D). There was a higher quantity of chromosomes (Figure 4C), with a significant increase in cells with more than 91 chromosomes (13.2±2.9% in OSU-13 vs. 0.9±0.5% in DMSO; P<0.01). In addition, cells with 81-90 chromosomes also increased (9.7±0.8% in OSU-13 vs. 4.4±2.7% in DMSO; P<0.05) (Figure 4D). Similar effects were observed in NCI-H929 cells (Online Supplementary Figure S5A-C).
Chromosome segregation was analyzed by microscopy to understand the mechanism behind this increase in the hyper-ploid population. OPM-2 cells treated with OSU-13 exhibited severe defects in chromosome alignment and segregation (Figure 4E), with 86.6±7.7% of the cells (P<0.01) presenting lagging chromosomes during anaphase (Figure 4F).
OSU-13 has in vivo effect against multiple myeloma in mice
The therapeutic potential of OSU-13 was evaluated in an NCI-H929 subcutaneous MM xenograft model in immunodeficient CB.17 SCID mice (Figure 5). Mice were treated daily per oral gavage starting 14 days after tumor implantation (day -14). Vehicle control or 10 mg/kg dose of OSU-13 were administered for 21 days, with a dosing holiday on days 14-17 due to unexpected weight loss in the OSU-13-treat-ed group (Figure 5A). Treatment with OSU-13 produced a significant delay in tumor growth (Figure 5B). On average, vehicle-treated mice reached the endpoint tumor volume (≥2,000 mm3) by day 18, whereas OSU-13-treated mice only achieved this size on day 25. At day 18, 50% of vehicle-treated mice reached the threshold, compared to only 10% of OSU-13-treated mice (P<0.05). In addition, the average tumor volume in the OSU-13-treated group was half of the vehicle-treated group (1,084 mm3 vs. 1,980 mm3). Treatment ended on day 21, justifying the increased tumor growth after this date. Altogether, treatment with OSU-13 resulted in a 22% tumor growth delay (P<0.05) and showed superior efficacy compared to the clinical candidate CFI-402257 in a parallel experiment (data not shown).
In another MM xenograft model using NOD SCID mice (Figure 5C), OSU-13 treatment also led to a significant delay in tumor growth. By the end of the treatment period (day 14), the average tumor volume in the OSU-13-treated group was 453 mm3, significantly lower than the control group (681 mm3; P<0.05) (Figure 5D). In addition, western blot analysis of tumor lysates showed increased expression of the apoptosis markers cleaved-caspase 3 and p-H2AX in the OSU-13-treated group (Figure 5E, F). These findings demonstrate that OSU-13 effectively reduces MM tumor burden and induces apoptosis in MM cells in vivo.
Discussion
TTK/Mps1 plays an essential role in SAC,24 and high levels of TTK expression correlate to unfavorable prognosis in several cancers.25,28,34 Accordingly, TTK inhibition has been explored in solid tumors as a therapeutic strategy to halt cell cycle arrest and induce genomic instability.28,30,33 However, the impact of TTK expression and TTK/MPS1 inhibition in hematologic cancers remains poorly understood.36,37 Here, we show that elevated TTK expression correlates with reduced overall and event-free survival along with amplification or gain of 1q21, found in high-risk MM. Indeed, TTK expression is significantly increased in high-risk compared to low-risk MM,36 and TTK is included in the MM Kinome Index, a gene expression profile risk score that predicts poor prognosis in MM.36
We report the first comprehensive analysis of a TTK/MPS1 inhibitor, OSU-13, in MM. Co-crystal structures reveal its binding to the ATP-binding pocket of TTK similarly to other TTK/MPS1 inhibitors,33,38 suggesting a conserved mechanism of inhibition. OSU-13 is stable and selectively inhibits TTK, showing comparable activity to the TTK/MPS1 inhibitor clinical candidates CFI-402257 and BOS-172722, which reinforces its clinical potential.
OSU-13 exhibited similar effects in MM as previously observed in breast cancer31 or with other TTK inhibitors in several cancers.29,30,32,33
First, OSU-13 showed cytotoxicity and anti-proliferative activity across MM cell lines. Sensitivity to OSU-13 varied among cell lines and correlated with 1q21 copy number. Nonetheless, sensitivity to TTK/MPS1 inhibitors seems to be multifactorial as observed in other cancer types,29,35 and identifying additional predictive biomarkers would further define patients who might benefit from TTK/MPS1 inhibitor-based therapies, expanding the potential applications of these treatments.
Second, OSU-13 induced apoptosis in MM cells, evidenced by caspase 3 and 7 activation and DNA damage, indicated by PARP1 cleavage, phosphorylation of H2AX, and TUNEL assay. However, partial rescue by the caspase inhibitor Z-VAD-FMK suggested involvement of caspase-independent mechanisms in OSU-13-induced cell death. Unlike the TTK/MPS1 inhibitor reversine that triggers autophagy by increasing LC3-B and Beclin 1 via the AKT pathway in cholangiocarcinoma cells,39 it seems that OSU-13 does not induce autophagy or necroptosis in MM cell lines.
Third, OSU-13 triggered abnormal mitosis, leading to chromosome missegregation, aneuploidy, and elevated chromosome numbers. Comparable effects have been reported in hematologic cancers with the TTK/MPS1 inhibitor AZ3146, inducing chromosome instability and DNA damage in MM36 and acute myeloid leukemia.37
Lastly, OSU-13 achieved significant tumor growth delay in NCI-H929 xenografts in two immunodeficient mouse models, despite a 3-day treatment interruption and a dose reduction due to weight loss. In a breast cancer mouse model, OSU-13 previously reduced tumor growth without affecting body weight.31 However, the aforementioned study used athymic nude mice, potentially less sensitive to OSU-13 than the CB.17 SCID and NOD SCID mice we used.
OSU-13 showed modest efficacy in our experiment as a single agent. Combination strategies for TTK/MPS1 inhibitors have been explored in various cancer models. Combination with the anti-programmed cell death 1 (PD-1) antibody was more effective than monotherapy in a colon cancer model.35 TTK inhibition also sensitized cells to paclitaxel treatment in colon,33 breast, and lung cancers.32 Accordingly, clinical trials with CFI-402257 are currently investigating combination therapies with paclitaxel and fulvestrant for breast cancer treatment.
Standard-of-care therapies for MM involve combinations of different agents to enhance response and prevent resistance. However, the combination strategies used for TTK inhibitors in solid tumors are unlikely to succeed in MM. Clinical trials with PD-1/PD-L1 inhibitors have shown limited results in MM - probably due to the immunosuppressive environment in MM - and have been halted due to severe adverse effects.40 Similarly, studies with the albumin-bound formulation Nab-paclitaxel41 did not achieve sustained responses in MM.42
In acute myeloid leukemia, the TTK inhibitor AZ3146 induced robust upregulation of the interferon gene,37 which has both direct anti-cancer effects43,44 and immune-activation potential.45-47 It would be interesting to investigate whether TTK inhibition triggers similar immunological effects in MM, potentially enhancing the effect of immunomodulatory drugs. In addition, MM cells may be sensitized to TTK inhibition by DNA-damaging agents such as proteasome inhibitors, alkylating agents, or experimental drugs that induce DNA double-strand breaks.48-50 Indeed, our initial findings suggest synergistic effects of OSU-13 with melphalan, a DNA-damaging agent. Identifying optimal combination strategies for OSU-13 will provide maximum efficacy and tolerability in MM treatment, providing new treatment options for relapsed/refractory MM patients.
In summary, our findings emphasize the importance of TTK in MM prognosis and show that OSU-13-mediated TTK inhibition induces cell death in MM cell lines and reduces tumor growth in vivo. We propose that inhibition of TTK using OSU-13 is an effective approach for treating multiple myeloma, particularly in a significant subgroup of high-risk patients with poor prognosis. Importantly, this is the first time that TTK inhibition has been comprehensively explored as a potential therapeutic strategy for a hematologic malignancy.
Footnotes
- Received January 27, 2023
- Accepted July 20, 2023
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
LVGL, BM-L, FC, TH and GH conceptualized and designed experiments, validated and analyzed data. LVGL, BM-L, TH and FC performed experiments. LVGL, TH and FC designed and created figures for the manuscript. LVGL wrote the manuscript, and FC, TH, GH and EM reviewed and edited the manuscript. DB advised on experimental design and data analysis, supervised, and provided resources for the study.
Data-sharing statement
The data are available from the corresponding author upon request.
Funding
This work was supported by OSU divisional funds (to DB and FC), the Pelotonia Foundation (to FC and BM), the Paula and Rodger Riney Foundation (to DB and FC), the Elsa U. Pardee Foundation (to FC), the International Myeloma Society (to FC), the National Cancer Institute (1K08CA263476-01A1 to FC) and the OSU Comprehensive Cancer Center Drug Development Institute.
Acknowledgments
The authors would like to thank the OSU Comprehensive Cancer Center Leukemia Tissue Bank, the OSUCCC support (core) grant (CCSG) 2P30CA016058-45, Dr. Jian-Qiu Wu and the OSU Molecular Genetics Department for the use of the fluorescence and confocal microscopes, and the OSU Comprehensive Cancer Center Drug Development Institute for providing the OSU-13 drug and collaborating with experiments and discussions.
References
- Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019; 69(1):7-34. https://doi.org/10.3322/caac.21551PubMedGoogle Scholar
- Attal M, Lauwers-Cances V, Hulin C. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017; 376(14):1311-1320. https://doi.org/10.1056/NEJMoa1611750PubMedPubMed CentralGoogle Scholar
- Mikkilineni L, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood. 2017; 130(24):2594-2602. https://doi.org/10.1182/blood-2017-06-793869PubMedPubMed CentralGoogle Scholar
- Robak P, Drozdz I, Szemraj J, Robak T. Drug resistance in multiple myeloma. Cancer Treat Rev. 2018; 70:199-208. https://doi.org/10.1016/j.ctrv.2018.09.001PubMedGoogle Scholar
- Bharadwaj R, Yu H. The spindle checkpoint, aneuploidy, and cancer. Oncogene. 2004; 23(11):2016-2027. https://doi.org/10.1038/sj.onc.1207374PubMedGoogle Scholar
- Kops GJ, Weaver BA, Cleveland DW. On the road to cancer: aneuploidy and the mitotic checkpoint. Nat Rev Cancer. 2005; 5(10):773-785. https://doi.org/10.1038/nrc1714PubMedGoogle Scholar
- Neuse CJ, Lomas OC, Schliemann C. Genome instability in multiple myeloma. Leukemia. 2020; 34(11):2887-2897. https://doi.org/10.1038/s41375-020-0921-yPubMedGoogle Scholar
- Drach J, Schuster J, Nowotny H. Multiple-myeloma - high-incidence of chromosomal aneuploidy as detected by interphase fluorescence in-situ hybridization. Cancer Res. 1995; 55(17):3854-3859. Google Scholar
- Bergsagel PL, Kuehl WM. Chromosome translocations in multiple myeloma. Oncogene. 2001; 20(40):5611-5622. https://doi.org/10.1038/sj.onc.1204641PubMedGoogle Scholar
- Bergsagel PL, Kuehl WM. Molecular pathogenesis and a consequent classification of multiple myeloma. J Clin Oncol. 2005; 23(26):6333-6338. https://doi.org/10.1200/JCO.2005.05.021PubMedGoogle Scholar
- Sawyer JR. The prognostic significance of cytogenetics and molecular profiling in multiple myeloma. Cancer Genet. 2011; 204(1):3-12. https://doi.org/10.1016/j.cancergencyto.2010.11.002PubMedGoogle Scholar
- Shammas MA, Shmookler Reis RJ, Koley H, Batchu RB, Li C, Munshi NC. Dysfunctional homologous recombination mediates genomic instability and progression in myeloma. Blood. 2009; 113(10):2290-2297. https://doi.org/10.1182/blood-2007-05-089193PubMedPubMed CentralGoogle Scholar
- Chng WJ, Fonseca R. Centrosomes and myeloma; aneuploidy and proliferation. Environ Mol Mutagen. 2009; 50(8):697-707. https://doi.org/10.1002/em.20528PubMedPubMed CentralGoogle Scholar
- Lara-Gonzalez P, Westhorpe FG, Taylor SS. The spindle assembly checkpoint. Curr Biol. 2012; 22(22):R966-980. https://doi.org/10.1016/j.cub.2012.10.006PubMedGoogle Scholar
- Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 2007; 8(5):379-393. https://doi.org/10.1038/nrm2163PubMedGoogle Scholar
- Thompson SL, Bakhoum SF, Compton DA. Mechanisms of chromosomal instability. Curr Biol. 2010; 20(6):R285-295. https://doi.org/10.1016/j.cub.2010.01.034PubMedPubMed CentralGoogle Scholar
- Diaz-Rodriguez E, Sotillo R, Schvartzman JM, Benezra R. Hec1 overexpression hyperactivates the mitotic checkpoint and induces tumor formation in vivo. Proc Natl Acad Sci U S A. 2008; 105(43):16719-16724. https://doi.org/10.1073/pnas.0803504105PubMedPubMed CentralGoogle Scholar
- Sotillo R, Hernando E, Diaz-Rodriguez E. Mad2 overexpression promotes aneuploidy and tumorigenesis in mice. Cancer Cell. 2007; 11(1):9-23. https://doi.org/10.1016/j.ccr.2006.10.019PubMedPubMed CentralGoogle Scholar
- Borisa AC, Bhatt HG. A comprehensive review on aurora kinase: small molecule inhibitors and clinical trial studies. Eur J Med Chem. 2017; 140:1-19. https://doi.org/10.1016/j.ejmech.2017.08.045PubMedGoogle Scholar
- Talati C, Griffiths EA, Wetzler M, Wang ES. Polo-like kinase inhibitors in hematologic malignancies. Crit Rev Oncol Hematol. 2016; 98:200-210. https://doi.org/10.1016/j.critrevonc.2015.10.013PubMedGoogle Scholar
- Chung V, Heath EI, Schelman WR. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein E (CENP-E), in patients with refractory cancer. Cancer Chemother Pharmacol. 2012; 69(3):733-741. https://doi.org/10.1007/s00280-011-1756-zPubMedGoogle Scholar
- Wei JH, Chou YF, Ou YH. TTK/hMps1 participates in the regulation of DNA damage checkpoint response by phosphorylating CHK2 on threonine 68. J Biol Chem. 2005; 280(9):7748-7757. https://doi.org/10.1074/jbc.M410152200PubMedGoogle Scholar
- Fisk HA, Mattison CP, Winey M. Human Mps1 protein kinase is required for centrosome duplication and normal mitotic progression. Proc Natl Acad Sci U S A. 2003; 100(25):14875-14880. https://doi.org/10.1073/pnas.2434156100PubMedPubMed CentralGoogle Scholar
- Liu X, Winey M. The MPS1 family of protein kinases. Annu Rev Biochem. 2012; 81:561-585. https://doi.org/10.1146/annurev-biochem-061611-090435PubMedPubMed CentralGoogle Scholar
- Slee RB, Grimes BR, Bansal R. Selective inhibition of pancreatic ductal adenocarcinoma cell growth by the mitotic MPS1 kinase inhibitor NMS-P715. Mol Cancer Ther. 2014; 13(2):307-315. https://doi.org/10.1158/1535-7163.MCT-13-0324PubMedPubMed CentralGoogle Scholar
- Zhang L, Jiang B, Zhu N. Mitotic checkpoint kinase Mps1/ TTK predicts prognosis of colon cancer patients and regulates tumor proliferation and differentiation via PKCalpha/ERK1/2 and PI3K/Akt pathway. Med Oncol. 2019; 37(1):5. https://doi.org/10.1007/s12032-019-1320-yPubMedGoogle Scholar
- Al-Ejeh F, Simpson PT, Saunus JM. Meta-analysis of the global gene expression profile of triple-negative breast cancer identifies genes for the prognostication and treatment of aggressive breast cancer. Oncogenesis. 2014; 3:e124. https://doi.org/10.1038/oncsis.2014.41PubMedPubMed CentralGoogle Scholar
- Tannous BA, Kerami M, Van der Stoop PM. Effects of the selective MPS1 inhibitor MPS1-IN-3 on glioblastoma sensitivity to antimitotic drugs. J Natl Cancer Inst. 2013; 105(17):1322-1331. https://doi.org/10.1093/jnci/djt168PubMedPubMed CentralGoogle Scholar
- Colombo R, Caldarelli M, Mennecozzi M. Targeting the mitotic checkpoint for cancer therapy with NMS-P715, an inhibitor of MPS1 kinase. Cancer Res. 2010; 70(24):10255-10264. https://doi.org/10.1158/0008-5472.CAN-10-2101PubMedGoogle Scholar
- Tardif KD, Rogers A, Cassiano J. Characterization of the cellular and antitumor effects of MPI-0479605, a small-molecule inhibitor of the mitotic kinase Mps1. Mol Cancer Ther. 2011; 10(12):2267-2275. https://doi.org/10.1158/1535-7163.MCT-11-0453PubMedGoogle Scholar
- Sugimoto Y, Sawant DB, Fisk HA. Novel pyrrolopyrimidines as Mps1/TTK kinase inhibitors for breast cancer. Bioorg Med Chem. 2017; 25(7):2156-2166. https://doi.org/10.1016/j.bmc.2017.02.030PubMedGoogle Scholar
- Wengner AM, Siemeister G, Koppitz M. Novel Mps1 kinase inhibitors with potent antitumor activity. Mol Cancer Ther. 2016; 15(4):583-592. https://doi.org/10.1158/1535-7163.MCT-15-0500PubMedGoogle Scholar
- Jemaa M, Galluzzi L, Kepp O. Characterization of novel MPS1 inhibitors with preclinical anticancer activity. Cell Death Differ. 2013; 20(11):1532-1545. https://doi.org/10.1038/cdd.2013.105PubMedPubMed CentralGoogle Scholar
- Choi M, Min YH, Pyo J, Lee CW, Jang CY, Kim JE. TC Mps1 12, a novel Mps1 inhibitor, suppresses the growth of hepatocellular carcinoma cells via the accumulation of chromosomal instability. Br J Pharmacol. 2017; 174(12):1810-1825. https://doi.org/10.1111/bph.13782PubMedPubMed CentralGoogle Scholar
- Mason JM, Wei X, Fletcher GC. Functional characterization of CFI-402257, a potent and selective Mps1/TTK kinase inhibitor, for the treatment of cancer. Proc Natl Acad Sci U S A. 2017; 114(12):3127-3132. https://doi.org/10.1073/pnas.1700234114PubMedPubMed CentralGoogle Scholar
- de Boussac H, Bruyer A, Jourdan M. Kinome expression profiling to target new therapeutic avenues in multiple myeloma. Haematologica. 2020; 105(3):784-795. https://doi.org/10.3324/haematol.2018.208306PubMedPubMed CentralGoogle Scholar
- Jin N, Lera RF, Yan RE. Chromosomal instability upregulates interferon in acute myeloid leukemia. Genes Chromosomes Cancer. 2020; 59(11):627-638. https://doi.org/10.1002/gcc.22880PubMedPubMed CentralGoogle Scholar
- Kwiatkowski N, Jelluma N, Filippakopoulos P. Small-molecule kinase inhibitors provide insight into Mps1 cell cycle function. Nat Chem Biol. 2010; 6(5):359-368. https://doi.org/10.1038/nchembio.345PubMedPubMed CentralGoogle Scholar
- Prajumwongs P, Waenphimai O, Vaeteewoottacharn K, Wongkham S, Sawanyawisuth K. Reversine, a selective MPS1 inhibitor, induced autophagic cell death via diminished glucose uptake and ATP production in cholangiocarcinoma cells. Peer J. 2021; 9:e10637. https://doi.org/10.7717/peerj.10637PubMedPubMed CentralGoogle Scholar
- 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. https://doi.org/10.3389/fimmu.2018.02749PubMedPubMed CentralGoogle Scholar
- Desai N, Trieu V, Yao Z. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res. 2006; 12(4):1317-1324. https://doi.org/10.1158/1078-0432.CCR-05-1634PubMedGoogle Scholar
- Jain T, Dueck AC, Kosiorek HE. Phase II trial of nab-paclitaxel in patients with relapsed or refractory multiple myeloma. Am J Hematol. 2016; 91(12):E504-E505. https://doi.org/10.1002/ajh.24548PubMedGoogle Scholar
- Takaoka A, Hayakawa S, Yanai H. Integration of interferonalpha/beta signalling to p53 responses in tumour suppression and antiviral defence. Nature. 2003; 424(6948):516-523. https://doi.org/10.1038/nature01850PubMedGoogle Scholar
- Thyrell L, Erickson S, Zhivotovsky B. Mechanisms of interferon-alpha induced apoptosis in malignant cells. Oncogene. 2002; 21(8):1251-1262. https://doi.org/10.1038/sj.onc.1205179PubMedGoogle Scholar
- Prchal M, Pilz A, Simma O. Type I interferons as mediators of immune adjuvants for T- and B cell-dependent acquired immunity. Vaccine. 2009; 27(Suppl 6):G17-20. https://doi.org/10.1016/j.vaccine.2009.10.016PubMedGoogle Scholar
- Tough DF, Kamath AT. Interferon with dendritic cells?. Nat Immunol. 2001; 2(12):1098-1100. https://doi.org/10.1038/ni1201-1098PubMedGoogle Scholar
- Tudor D, Riffault S, Carrat C, Lefevre F, Bernoin M, Charley B. Type I IFN modulates the immune response induced by DNA vaccination to pseudorabies virus glycoprotein C. Virology. 2001; 286(1):197-205. https://doi.org/10.1006/viro.2001.0957PubMedGoogle Scholar
- Bergsagel DE, Sprague CC, Austin C, Griffith KM. Evaluation of new chemotherapeutic agents in the treatment of multiple myeloma. IV. L-Phenylalanine mustard (NSC-8806). Cancer Chemother Rep. 1962; 21:87-99. Google Scholar
- Cottini F, Hideshima T, Suzuki R. Synthetic lethal approaches exploiting DNA damage in aggressive myeloma. Cancer Discov. 2015; 5(9):972-987. https://doi.org/10.1158/2159-8290.CD-14-0943PubMedPubMed CentralGoogle Scholar
- Neri P, Ren L, Gratton K. Bortezomib-induced “BRCAness” sensitizes multiple myeloma cells to PARP inhibitors. Blood. 2011; 118(24):6368-6379. https://doi.org/10.1182/blood-2011-06-363911PubMedPubMed CentralGoogle Scholar
- Hanamura I, Stewart JP, Huang Y. Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood. 2006; 108(5):1724-1732. https://doi.org/10.1182/blood-2006-03-009910PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.