Abstract
This study describes the characterization of a novel kinase inhibitor, ON123300, which inhibits CDK4/6 and PI3K-δ and exhibits potent activity against mantle cell lymphomas (MCLs) both in vitro and in vivo. We examined the effects of PD0332991 and ON 123300 on cell cycle progression, modulation of the Rb and PI3K/AKT pathways, and the induction of apoptosis in MCL cell lines and patient-derived samples. When Granta 519 and Z138C cells were incubated with PD0332991 and ON123300, both compounds were equally efficient in their ability to inhibit the phosphorylation of Rb family proteins. However, only ON123300 inhibited the phosphorylation of proteins associated with the PI3K/AKT pathway. Cells treated with PD0332991 rapidly accumulated in the G0/G1 phase of cell cycle as a function of increasing concentration. Although ON123300-treated cells arrested similarly at lower concentrations, higher concentrations resulted in the induction of apoptosis, which was not observed in PD 0332991-treated samples. Mouse xenograft assays also showed a strong inhibition of MCL tumor growth in ON123300-treated animals. Finally, treatment of ibrutinib-sensitive and resistant patient-derived MCLs with ON123300 also triggered apoptosis and inhibition of the Rb and PI3K/AKT pathways, suggesting that this compound might be an effective agent in MCL, including ibrutinib-resistant forms of the disease.
Keywords: mantle cell lymphoma, CYCLIN D, CDK4, RB, PI3K, targeted therapy
Introduction
Mantle cell lymphoma (MCL) is a well-defined and aggressive B-cell non-Hodgkin’s lymphoma that is genetically characterized by the t(11;14)(q13;q32) chromosomal translocation which results in constitutive over-expression of CYCLIN D11–3. CYCLIN D1 over-expression is considered to be a hallmark of MCL, and is closely correlated with the proliferative rate of these malignant cells4. Because CYCLIN D1 over-expression has been shown to be the most common feature of MCL, PD0332991 (palbococlib), a selective CDK4/6 inhibitor5, has been tested in Phase I/II clinical trials as a targeted therapy for MCL. Unfortunately, as a single agent, PD0332991 was reported to be cytostatic and hence, resulted in only stabilization of the disease with very few objective clinical responses6, 7. Despite these shortcomings, this agent has been found to sensitize MCL cells to other cytotoxic drugs due to the fact that MCLs frequently exhibit lesions in other signaling pathways including those mediated by the B-cell receptor (BCR), phosphatidylinositol 3 kinase (PI3K), WNT, NOTCH and TGF-β8, 9 Of these, dysregulated PI3K signaling is one of the most widely studied and represents an attractive druggable target.
Class I PI3Ks are the major activators of the AKT serine-threonine kinase and are sub-divided into class IA and IB kinases10. Of the three class IA subtypes, PI3K-α, PI3K-β, and PI3K-δ, both the α and δ isoforms play critical roles in immune signaling of normal cells. While PI3k-δ is lineage restricted, expressed at high levels in B-lymphocytes and transmits signals from the BCR, PI3K-α plays a ligand-independent role in this process. Furthermore, both isoforms are mediators of agonist-independent, constitutive BCR signaling that occurs in MCL11–13. Studies have shown that MCLs constitutively activate the PI3K-AKT-mammalian target of rapamycin (mTOR) pathway and that this signaling module plays a key role in the aggressive blastoid variant of the disease13–15. Furthermore, in the case of refractory disease, both PI3K-α and PI3K-δ are also involved in ligand-independent activation of this pathway13. Ibrutinib (PCI-32765, Imbruvica®), an inhibitor of the Burton's tyrosine kinase (BTK), has been remarkably successful in the treatment of chronic lymphocytic leukemia (CLL) and has recently been approved for the treatment of certain refractory or relapsed MCL cases. Notably, this drug effectively induces complete remission in approximately 21% of these patients16 and is thought to do so, in part, by indirectly inhibiting PI3K-δ mediated signals. As a result, several PI3K-δ inhibitors are now in various stages of clinical trials for the treatment of MCL and other B-cell malignancies17, 18. In spite of ibrutinib’s efficacy, acquired resistance to the drug is beginning to emerge in the clinic. Whole exome sequencing has revealed the presence of a single point mutation in the BTK ibrutinib binding site, C481S, which results in enhanced BTK and AKT activities in ibrutinib-treated patients with relapsed MCL19. Furthermore, subsets of patients that exhibit primary resistance or respond temporarily to the drug underscore the need to overcome alternate mechanisms of ibrutinib-resistance19.
Owing to the near complete dysfunction of the CYCLIN D1/CDK4/Rb pathway and activation of the PI3K/AKT pro-survival pathway in most MCLs, recent studies have focused on combination therapies targeting both signaling modules. These studies have shown that treatment of MCL cells with PD0332991 induces G1 arrest of cells and sensitizes them to selective PI3K-δ inhibitors, such as GS-1101 (idelalisib, CAL-101)20. It is hypothesized that cells that are blocked in G1 by CDK4/6 inhibition allows for sustained inhibition of PI3K-δ that is unlikely to be overcome by compensation of other PI3K isoforms19, 20.
Towards the goal of inhibiting both CYCLIN D/CDK4/Rb and PI3Kδ in MCL, we recently developed a dual inhibitor of both pathways, ON12330021, and examined its effects on the growth and apoptosis of MCL cell lines. Our studies show that unlike PD0332991, treatment with ON123300 was cytotoxic and induced apoptosis of MCL cell lines and patient-derived MCLs. Our studies also show that ON123300 was effective in reducing the tumor burden in mouse xenograft models of MCL in the absence of adverse side effects.
Materials/Subjects and Methods
MCL Patient Samples
Primary human lymphoma samples were obtained either as part of standard excisional biopsy or from peripheral blood samples from patients at the Icahn School of Medicine at Mount Sinai with informed and written consent for tissue banking following a protocol approved by the Institutional Review Board (MSSM IRB#11-1669) and in accordance with the Declaration of Helsinki. Specimens were processed to viable, sterile single-cell suspensions. Briefly, lymph node tissue was diced and forced through a metal sieve in a laminar flow hood into RPMI medium. Peripheral blood mononuclear cells or disaggregated mantle cell lymphoma biopsy cells were pelleted by low-speed centrifugation, resuspended in media composed of 90% fetal bovine serum (FBS) and 10% DMSO (Sigma), frozen slowly in the vapor phase of liquid nitrogen in multiple cryovials, and stored in liquid nitrogen.
Cell Lines
Cell lines were obtained from ATCC and cultured in Dulbecco’s Modified Eagle Medium (DMEM) or RPMI (Life Technologies) supplemented with 10% FBS and penicillin-streptomycin at 37°C under humidified conditions and 5% CO2. All cell lines tested negative for mycoplasma. To determine GI50 values, cells were seeded at a density of 2.5×103 cells-0.1ml−1 per well of a 96-well plate. The compounds were added 24 hrs post-plating at the indicated concentrations. Cell counts were determined from duplicate wells 96 hrs post-treatment. The total number of viable cells was determined using the Cell Titer Blue assay in conjunction with the GloMax plate reader (Promega).
Reagents and Antibodies
TNF-a was purchased from R&D Systems (catalog #210-TA/CF). ON123300 was obtained from Onconova Therapeutics, Inc. PD0332991, U0126 and QNZ were purchased from Selleckchem. Antibodies directed against PARP (catalog #9542), CASPASES 3 (catalog #9665), 7 (catalog #9492) and 9 (catalog #9502), phospho-RbSer780 (catalog #9307), Rb (catalog #9309), phospho-mTORSer2481 (catalog #2974), mTOR (catalog #4517, phospho-4EBP1 (catalog #9459), 4EBP1 (catalog #9644), phospho-S6RB (catalog #2211), S6RB (catalog #2317), phospho-FOXO1 (catalog #9461), phospho-ERK (catalog #4370), ERK (catalog #9107), phospho-MEK (catalog #9154), MEK (catalog #4694), phospho-NF-κB p65Ser536 (catalog #3033), and phospho-RbSer807/811 (catalog #9308) were obtained from Cell Signaling Technologies. GAPDH (catalog #SC-47724), AKT (catalog #SC-1618), p130 (catalog #SC-317), CDK4 (catalog #SC-260), CDK2 (catalog #SC-163), CYCLIN D1 (catalog #SC-8396), NF-κB p65 (catalog #sc-8008) and p21-specific (catalog #SC-756) antisera were purchased from Santa Cruz Biotechnology. The phospho-AKTSer473 antibody was purchased from R&D Systems (catalog #AF-887). Phospho-p130 antiserum was obtained from Abcam (catalog #AB68136).
Kinase Assays and Western Blot Analysis
Kinase assay were performed as described earlier using recombinant proteins21. For whole cell extracts derived from cell lines, cells were grown as indicated and lysed in NP-40 cell lysis buffer (20mM Tris, pH 7.4/ 250nM NaCl/ 5nM EDTA/ 50mM NaF/ 1nM Na3VO4/ 1% NP40/ 0.02% NaN3) (Invitrogen). Patient derived samples were incubated with amonium chloride to remove red blood cells and the remaining cells lysed in RIPA buffer (25mM Tris-HCl, pH 7.5/ 150mM NaCl/ 1mM EDTA/ 1mM EGTA/ 1% NP40/ 1% SDS/ 2.5mM sodium pyrophosphate/ 1nM β-glycerophosphate/ 1mM Na3VO4/ 1mM leupeptin). Proteins were resolved by SDS-PAGE, transferred to nitrocellulose membranes and subjected to Western blot analysis using the indicated antibodies. Proteins were visualized using an Odyssey imaging system (LI-COR Biosciences).
Flow Cytometric Analysis
To determine cell cycle distribution, ethanol fixed cells were washed with phosphate buffered saline (PBS), resuspended in PBS and incubated with RNase and propidium iodide at 37°C for 20 min. DNA content was measured using a FACS Calibur (BD Biosciences) and the data analyzed using FlowJo (Treestar). Annexin V staining of patient-derived material was performed using the FITC-Annexin V Apoptosis Detection Kit I (BD Biosciences). All data were acquired using a LSRFortessa (BD Biosciences) and analyzed using Cytobank (https://www.cytobank.org/).
Animal Studies
To determine the maximum tolerated dose of ON123300, female CD-1 mice (10–12 weeks of age) were injected intraperitonially (ip) with a single dose of 100mg/kg or 200 mg/kg of ON123300. Signs of toxicity and body weights were monitored and recorded over the next 7 days. In a second study, 200mg/kg ON123300 was administered via ip injection for 5 consecutive days. Signs of toxicity and body weights were monitored and recorded over the next 10 days. n=3 per group.
ON123300 plasma level concentrations were determined using female CD-1 mice (10–12 weeks of age). Mice were treated with ON123300 (50mg/kg) dissolved in water via ip injection or oral gavage (po). Mice were euthanized at 10 minutes, 30 minutes, 1 hour, 2 hours, 4 hours and 8 hours post-administration. Blood was collected by heart puncture into heparinized tubes containing 10µl of 0.5 M EDTA and plasma was obtained following centrifugation for 10 minutes at 10,000 rpm. Plasma samples were analyzed by liquid chromatography/mass spectrometry (LC/MS) (Agilent 1200 HPLC /Agilent Triple Quad). Area Under the Curve (AUC) was calculated according to the trapezoidal rule: (1/2)(C1+C2)(t1−t2). n=2 per time point.
For unblinded tumor xenograft assays, 1×107 Z138C cells were implanted subcutaneously into female nu/nu mice (12 weeks of age). Tumors were allowed to reach 100mm3 in size before the animals were randomly assigned to treatment groups. Five mice per group were treated with ON123300 (100mg/kg) or vehicle (water) daily or every alternate day in two independent experiments. Tumor volumes and bodyweights were recorded as indicated. Tumor volumes were calculated using the following equation: (4/3)((L+W)/4)3.
All experiments were performed under protocols approved by the Icahn School of Medicine at Mount Sinai’s Institutional Animal Care and Use Committee according to federal and institutional guidelines and regulations. Animals were purchased from Charles River Laboratories.
Statistical Analysis
Statistical analysis was performed using a standard, unpaired, two-tailed Student’s t test. Data are graphed as mean ± SD. Results were considered significant as p≤0.05.
Results
Derivation of a small molecule inhibitor of MCL growth
We recently synthesized a large number (approximately 3,000) of novel ATP mimetics, some of which contained the basic backbone shown in Fig. 1A and exhibited potent CDK4 inhibitory activity21. These compounds were tested in in vitro tissue culture growth inhibitory (GI50) assays against a panel of cell clines representing a variety of tumor types. This screen revealed that one of these compounds, ON0123300, exhibited 10–100-fold higher cytotoxicities against MCL cell lines as compared to those of other tumors (Table 1). Subsequent testing of this compound by Reaction Biology Corp (Malvern, PA) for activity against a series of 285 functional kinases revealed that ON123300 is a multi-kinase inhibitor21, with highest activity against CDK4 and ARK5 (Table 2), two kinases intimately associated with growth, survival and metastasis of human tumor cells22, 23. In addition to these kinases, this compound was shown to inhibit PI3K-δ, with an IC50 of 144nM (Table 2). The kinase inhibition profile of ON123300 along with the synthetic procedures used to synthesize ON123300 have been published earlier21.
Figure 1. ON123300 is a multi-kinase inhibitor.
(A) Structure of ON123300. (B) Calculation of IC50 values against CDK4/CYCLIN D1 and (C) PI-3 Kinase δ by ON123300. Recombinant proteins were incubated with the indicated concentrations of ON123300 and subjected to in vitro kinase assays as described in the methods section. Values obtained were plotted as a function of log drug concentration. IC50 values were determined by plotting sigmoidal non-linear regression curves with a variable slope.
Table 1.
ON123300 GI50 values in human tumor cell lines.
| Cell Line | Tumor Type | GI50 (µM) |
|---|---|---|
| Z138C | MCL | 0.025 |
| GRANTA-519 | MCL | 0.035 |
| DLD-1 | Colorectal | 0.100 |
| SW480 | Colorectal | 0.100 |
| COLO-205 | Colorectal | 0.200 |
| HCT-15 | Colorectal (MDR high) | 0.400 |
| MIA-PaCa-2 | Pancreatic | 0.250 |
| SNU-5 | Gastric | 0.200 |
| SNU-398 | Gastric | 0.300 |
| MDA-MB-231 | Breast (Triple Negative) | 0.200 |
| MDA-MB-157 | Breast (Triple Negative) | 0.250 |
| BT-474 | Breast (ER+HER2+) | 0.250 |
| SK-BR-3 | Breast (HER2+) | 0.600 |
| MCF-7 | Breast (ER+) | 0.150 |
| BT-20 | Breast | 0.100 |
Table2.
Kinase inhibition profile of ON123300.
| Kinase |
ON123300 IC50 (nM) |
PD0332991 IC50 (nM) |
|---|---|---|
| CDK4/CYCLIN D1 | 3.87 | 5.36 |
| CDK6/CYCLIN D1 | 9.82 | 3.76 |
| ARK5 | 4.95 | >5,000 |
| FGFR1 | 26.0 | >10,000 |
| PDGFRβ | 26.0 | >10,000 |
| PI3K-δ | 144.0 | >10,000 |
Inhibition by CDK4 kinase activity by ON123300
To confirm the observation that CDK4 and PI3K-δ are targets of ON123300, we independently tested its activity in in vitro kinase assays using recombinant CDK4 and PI3K-δ (Fig. 1B & C). Our results showed that ON123300 is a potent inhibitor of CDK4, with an IC50 of 3.8nM, with little inhibitory activity against CDKs 1,2,5 and 8 (data not shown). As a positive control, we used PD0332991, a commercially available CDK4 inhibitor, which is highly selective towards CDK4 and CDK6 and is currently in various phases of clinical trials5, 7. Kinase inhibition assays showed that PD0332991 showed similar inhibition of CDK4 with an IC50 of 5.36nM (Table 2). However, when these assays were performed using PI3K-δ, ON123300 inhibited the kinase with an IC50 of 144nM while PD0332991 failed to show any inhibitory activity (Figure 1C and Table 2).
Effect of ON123300 and PD0332991 on cell cycle progression and induction of apoptosis of MCL cells
We next examined the effect of ON123300 and PD0332991 treatment on the cell cycle progression of MCL cell lines. For these studies, Z138C cells were cultured in the presence of increasing concentrations of ON123300 or PD0332991 for 24 hours. The cells were then harvested and subjected to propidium iodide staining and flow cytometric analysis. Figure 2A shows the distribution of cells in various phases of the cell cycle. DMSO-treated cells served as negative controls. In the absence of drug, the majority of cells were in the G1 phase of the cell cycle with smaller percentages of the population in the S and G2 phases (Fig. 2A). As expected, a rapid accumulation of the cells in the G1 phase of the cell cycle was evident following treatment with PD0332991 (Fig. 2A), with little or no accumulation of cells in the sub-G1 phase. While cells treated with ON123300 also accumulated in the G1 phase at lower concentrations (0.1–1.0µM), at higher concentrations of the compound, a large proportion of the cells progressed through the S and G2/M phases of the cell cycle and eventually accumulated in the sub-G1 phase, suggesting an induction of apoptosis (Fig. 2A).
Figure 2. Modulation of the cell cycle and induction of apoptosis in ON123300-treated MCL cell lines.
(A) Z138C cells were treated with vehicle (DMSO) or increasing concentrations of ON123300 or PD0332991 for 24 hours, stained with propidium iodide and subjected to flow cytometric analysis. (B) Granta and (C) Z138C cells were treated with vehicle (V) or increasing concentrations of ON123300 or PD0332991 for 24 hours. Whole cell extracts were subjected to western blot analysis using antibodies directed against the indicated proteins. GAPDH is shown as a loading control. Images representative of 4 independent experiments are shown.
To determine whether higher concentrations of ON123300 induced apoptosis in MCL cells, the levels of caspases 3, 7 and 9 as well as PARP cleavage were determined by Western blot analysis. The results presented in Figure 2B and C show that while there is no evidence of apoptosis in PD0332991-treated cells, we could readily see PARP cleavage in cells treated with ON123300 at concentrations greater than or equal to 2.5µM for 24 hrs. We also observed decreases in the levels of full-length forms of caspases 3, 7 and 9, which are cleaved into their active forms upon the onset of apoptotic cell death.
Because ON123300 induced growth arrest and cell death over a 96 hr period at concentrations of 25–50nM, we repeated these studies using lower concentrations of the compound. These studies showed that incubation of the Granta 519 and Z138C cell lines with PD0332991 for 72–96 hrs did not show any PARP cleavage, while similar incubation of these cells with identical concentrations of ON123300 readily resulted in PARP cleavage (data not shown). These results confirm previous studies5, 20, 24 and suggest that while PD0332991 induces effective growth arrest of MCL cell lines, it is not accompanied by apoptosis. On the other hand, while ON123300 triggers growth arrest at lower concentrations in a manner similar to PD0332991, at higher concentrations, this compound induces apoptosis of MCL cell lines.
Status of cell cycle regulatory proteins in ON123300-treated cells
It is now well established that the Retinoblastoma family of proteins (pRb, Rb2/p107 and p130) are primary targets of CDK47. It is therefore highly likely that phosphorylation of Rb family proteins is inhibited due to inhibition of CDK4 in MCL cells treated with PD0332991 or ON123300. To determine the effects of PD0332991 and ON123300 on Rb and its related proteins, Granta 519 and Z138C cells were treated with increasing concentrations of ON123300 or PD0332991 for 24 hours. Whole cell extracts were subjected to western blot analysis using antibodies directed against Rb and p130 and their phosphorylated forms as well as CDK4, CDK2 and p21. The results of this study, presented in figure 3, showed that both compounds (ON123300 as well as PD0332991) inhibited the phosphorylation of pRb and p130 in a dose- dependent manner. Although the levels of CYCLIN D1 declined as a function of ON0123300 concentration, these results are in agreement with previous studies showing that inhibition of PI3K/AKT/mTOR signaling leads to loss of CYCLIN D1 expression14, 15. p107 levels were extremely low in both cell lines, making it difficult to determine the effects of both ON0123300 and PD0332991 on its phosphorylation status (data not shown). While the levels of CDK4 and CDK2 were unaffected in cells treated with ON123300 and PD0332991, we consistently observed loss of p21 levels in ON123300 treated cells, which was not readily seen in cells treated with PD0332991, which appears to be a consequence of PI3K pathway inhibition described below. These results suggest that both PD0332991 as well as ON123300 inactivate the Rb pathway in the two MCL cell lines studied here with equal efficiency.
Figure 3. Inhibition of the CDK4/CYCLIN D/Rb pathway by ON123300.
(A) Granta 519 and (B) Z138C cells were treated with increasing concentrations of ON123300 or PD0332991 for 24 hours. Western blot analysis was performed as described in Fig. 2. Images representative of 4 independent experiments are shown.
Effect of ON123300 on the PI3K/AKT/mTOR pathway
Because ON123300 inhibits PI3K-δ the phosphorylation status of members of PI3K pathway in cells treated with ON123300 and PD0332991. As predicted, there was no discernable change in the phosphorylation status of AKT, mTOR, 4EBP1 and S6 ribosomal protein (S6RB) in PD0332991-treated Granta519 and Z138C cells (Figure 4). However, we readily detected inhibition of AKT Ser473 phosphorylation, which is mediated by members of the PI3K family25, 26, in ON123300-treated cells. Inhibition of AKT phosphorylation subsequently resulted in an inhibition of mTOR phosphorylation at Ser2481, a site that is phosphorylated by AKT25. Similarly, phosphorylation of downstream AKT/mTOR targets, 4EBP1 and S6RB27, 28, were inhibited in ON123300-treated cells. In addition, we also noted inhibition of FOXO1 phosphorylation, which is also known to be a target of mTOR29, in cells treated with ON123300 (Figure 4). Dephosphorylation of FOXO1 positively regulates the transcriptional activity of this growth suppressor protein, which has been shown to activate apoptotic pathways and cell cycle arrest30. Together, these studies provide a molecular basis for the observed induction of apoptotic pathways seen in cells treated with ON123300.
Figure 4. Inhibition of PI3K-mediated signaling by ON123300.
(A) Granta 519 and (B) Z138C cells were treated with increasing concentrations of ON123300 or PD0332991 for 24 hours and subjected to western blot analysis as described in Fig. 2. Images representative of 4 independent experiments are shown.
Effect of ON123300 on the MAP kinase and NF-kB Pathways
Because ON123300 is a multi-kinase inhibitor, it is possible that some of its anti-tumor activity might be associated with its ability to inhibit MAPK and/or NF-kB pathways. To test this possibility, we examined the phosphorylation status of MEK and ERK1 & 2 kinases whose activation is mediated by growth factor receptors. These studies, presented in supplemental Fig. 1, show that neither ON123300 nor PD0332991 has any effect on the phosphorylation status of MEK or ERK kinases, whereas U0126, a known MEK inhibitor that served as a positive control, potently inhibited MEK and ERK phosphorylation in Granta 519 and Z138 cell lines.
We next examined the effects of ON123300 and PD0332991 on the phosphorylation status of p65 (NF-kB), a recently identified druggable target in MCL31, in exponentially growing Z138 cells and observed no change in the phosphorylation status of this protein (Supplemental Figure 2A). As a positive control, we used HeLa cells that were treated with 5ng/ml of TNF-α which are known to activate the NF-kB pathway in response to this chemokine. Following treatment with ON123300 or PD0332991, cells were incubated with TNF-α for 5 min and the level of p65 NF-kB phosphorylation examined by Western blot analysis. These studies, presented in supplemental figure 2B, show that ON123300 and PD0332991 had little or no effect on the phosphorylation status of p65, while QNZ (EVP4593), a known inhibitor of the NF-kB pathway induced strong inhibition of p65 phosphorylation.
Effect of ON123300 on primary patient-derived MCL viability
To test the effects of ON123300 on primary tumor samples, we collected peripheral blood by apheresis from MCL patients with leukemic disease. Two samples, one sensitive to ibrutinib (MEM1), and a second sample from a patient with ibrutinib-resistant MCL (MEM2), were used in these studies. The cells were treated with increasing doses of ON123300 and the level of apoptosis measured using Annexin V staining. Figure 5A shows the increase in Annexin V positive cells of both samples following exposure to ON123300 for 24 hrs. While the onset of apoptosis in both the ibrutinib-sensitive and resistant samples was observed in cells treated with as little as 500nM of the compound, nearly 100% of cells stained Annexin V+ in the presence of 5µM ON123300 (Fig 5A).
Figure 5. Effect of ON123300 on primary patient-derived MCLs.
(A) Induction of apoptosis in ON123300-treated ibrutinib-sensitive (MEM1) and resistant (MEM2) MCLs. Cells were treated with the indicated concentrations of ON123300 for 18 hrs. The level of Annexin V staining was measured using flow cytometric analysis. (B) Inhibition of the CDK4/Rb and AKT pathways in ON123300-treated ibrutinib-sensitive (MEM1) and resistant (MEM2) primary MCLs. Cells were treated with increasing concentrations of ON123300 for 18 hrs., and western blot analysis was performed as described in Fig. 2.
We also analyzed the levels of Rb and AKT phosphorylation in these samples. Figure 5B shows that both primary MCL samples exhibited reductions in the levels of Rb and AKT phosphorylation. Furthermore, the phosphorylation of S6RB was also inhibited, suggesting inhibition of the PI3K-AKT-mTOR pathway (Fig 5B)28. These studies are in agreement with those presented for the Granta 519 and Z138C MCL cell lines and confirm that ON123300 effectively blocks both the CDK4/Rb and AKT/PI3K pathways that are critical for MCL growth and survival.
Safety Pharmacology and in vivo efficacy
We next performed studies to determine the maximum tolerated dose of ON123300 in mice. Three CD-1 mice were injected intraperitonially (ip) with 100 or 200 mg/kg of ON123300. Signs of toxicity and body weights were monitored and recorded over the next 7 days. No signs of toxicity or weight loss were observed with a survival rate of 100%. A second study, whereby 200mg/kg ON123300 was administered for 5 consecutive days, showed that this dosing regimen did not exhibit any signs of toxicity and had a survival rate of 100% for at least 10 days post-administration (data not shown). We also performed LC/MS to determine the pharmacokinetics of ON123300 in the plasma. These studies show that the area under the curve (AUC) values were 7.18 and 2.78µg/ml/hr in mice that received the drug via ip injection and oral gavage (po), respectively, which reflect concentrations of 16.71 (ip) and 6.47µM/ml/hr (po) in the plasma. Because the plasma concentration of ON123300 was higher in animals that received the compound via ip injection, we used this route of administration for subsequent mouse xenograft assays.
To determine the in vivo efficacy of ON123300, we used a nude mouse model of mantle cell lymphoma where Z138 cells were injected into nude mice and allowed to grow to approximately 100mm3 in size. Mice with tumor xenografts were randomly assigned into 2 treatment groups (n=5 per group) and treated daily with either water (vehicle) or ON123300 (100mg/kg) on a daily basis. Body weights and tumor volumes from two independent studies were recorded and plotted as a function of time (Figure 6). Figure 6B shows a representative image of tumors in control and ON123300-treated mouse cohorts on day 14 of experiment #2. As seen here and in figure 6A, treatment with ON123300 significantly inhibited (p<0.05) tumor growth over a 2-week period, suggesting that this compound is a potent inhibitor of MCL proliferation in vivo. In addition, daily treatment with ON123300 did not cause a significant reduction in the total body weights of ON123300-treated animals (Fig 6C). (The increase in the body weights of the untreated animals is likely attributable to the rapid growth of tumors). These results, in addition to those obtained in vitro, suggest that ON123300 is an effective therapeutic agent in MCL with minimal associated toxicity.
Figure 6. Effect of ON123300 on the growth of MCL cells in a mouse xenograft model.
(A) Mean tumor volumes of control- and ON123300-treated mouse cohorts, n=5 per group, in two independent experiments. p<0.05 for both experiments (B) Representative image of tumors in control and ON123300-treated mice. White arrows mark tumors in the flanks. (C) Body weights of control- and ON123300-treated mouse cohorts. Error bars represent ±SD.
Discussion
The pathogenesis of MCL is characterized by cell cycle dysregulation, with CYCLIN D1 overexpression being a hallmark of this disease. Cyclin D1 complexes with its catalytic partners, cyclin-dependent kinases (CDKs) 4 and 6, driving retinoblastoma (Rb) protein phosphorylation and G1 cell-cycle progression32. Initial studies with PD0332991 (palbococlib), a selective small molecule inhibitor of CYCLIN D- associated CDK4/6 inhibitor activity5, demonstrated potent inhibition of Rb phosphorylation and G0/G1 cell cycle arrest in the absence of any associated apoptosis24. In accordance, the drug was cytostatic in a Phase I/II clinical trial for MCL and resulted in disease stabilization with an 18% overall response rate6. Although PI3K signaling is activated in MCL, in a Phase I trial, idelalisib (GS-1101), a selective PI3K-δ inhibitor, was also modestly effective as a single agent in patients with refractory disease33. In spite of these outcomes, PD0332991 has been shown to sensitize MCL cell lines to PI3K-δ inhibition, in part, by enhancing the effect of the loss/reduction of AKT phosphorylation when the cells are arrested in the G1 phase17, 19, 20.
In this communication, we examined the effects of ON123300, a dual inhibitor of CDK4 and PI3K-δ on MCL cell growth and compared its activity with PD0332991, a highly specific CDK4/6 inhibitor. Our results show that while both inhibitors induce G1 arrest of MCLs, only ON123300 elicits an apoptotic response from these tumor cells. The G1 arrest mediated by the two compounds studied here is clearly due to inhibition of phosphorylation of members of the Rb family, a direct result of CDK4 and CDK6 inhibition by both compounds. Our results also show that in contrast to PD0332991, ON123300 acts as a potent inhibitor of the PI3K/AKT/mTOR pathway, leading to the activation of apoptotic pathways, as assayed by caspase activation and PARP cleavage. Our results also show that ON123300 treatment results in inhibition of FOXO1 phosphorylation, a target of mTOR (Figure 4). Dephosphorylation of FOXO1 is known to regulate the transcriptional activity of this protein, which has been shown to activate apoptotic pathways and cell cycle arrest15, 30. Together, these studies provide a molecular basis for the observed induction of apoptosis ON123300-treated cells.
Safety studies in mice suggest that ON123300 is orally bio-available and is minimally toxic when administered orally or intraperitoneally. The in vivo efficacy of ON123300 was examined using Z138 cells grown as xenografts in nude mice. These studies, presented in Figure 6, showed that treatment of these tumor-bearing mice with ON123300 markedly inhibited tumor growth over a 14-day period, further suggesting that this compound could be a possible candidate for the initiation of clinical trials. Previous studies have demonstrated that ON123300 readily crosses the blood-brain barrier34, 35, and considering the fact that a small percentage of MCL patients experience lymphomatous spread to the meninges and central nervous system36, this agent could also be useful for patients with CNS involvement.
Recent studies have shown that constitutive activation of BCR signaling via PI3 kinases is essential for the proliferation of a large portion of MCLs, particularly in relapsed or refractory disease37. This observation was further supported by the finding that ibrutinib, a covalent inhibitor of BTK, has a profound effect on disease progression of MCL patients16. MCLs that acquire resistance to or relapse from treatment with ibrutinib due to the BTKC481S mutation, as well as those that exhibit primary resistance to the drug show increased AKT and ERK activition19, 38. Interestingly, these drug-resistant MCLs exhibit a bias in CDK4-driven proliferation, which is sensitive to combined inhibition of the CDK4 and PI3K pathways19. We examined the effects of ON123300 on primary tumor samples that were derived from MCL patients who either responded to ibrutinib or who acquired resistance following ibrutinib treatment. Both cell types were sensitive to the pro-apoptotic effects of ON123300 as measured by increased Annexin V staining and reductions in the levels of phospho-Rb, phospho-AKT and S6RB phosphorylation, consistent with inhibition of the mTOR pathway. These results suggest that ON123300 is likely to be an effective treatment option for both ibrutinib-responsive and resistant patients.
Supplementary Material
Acknowledgements
We thank Chen Ren for assistance with LC/MS studies. This work was supported by grants from the NIH (P01CA-130821), (NIH-RO1-CA-158209) and Onconova Therapeutics Inc to EPR.
EPR is an equity holder, board member and a paid consultant of Onconova Therapeutics, Inc. In addition, EPR and MVRR are named inventors on pending and issued patents filed by Temple University. MVRR is a stockholder and paid consultant of Onconova Therapeutics Inc. SCC and SJB are paid consultants of Onconova Therapeutics Inc.
Footnotes
Conflict-of-interest disclosure: All other authors declare no conflicts of interest.
Supplementary Information accompanies this paper on the Leukemia website (http://www.nature.com/leu)
References
- 1.Weisenburger DD, Sanger WG, Armitage JO, Purtilo DT. Intermediate lymphocytic lymphoma: immunophenotypic and cytogenetic findings. Blood. 1987 Jun;69(6):1617–1621. [PubMed] [Google Scholar]
- 2.Vandenberghe E, De Wolf-Peeters C, van den Oord J, Wlodarska I, Delabie J, Stul M, et al. Translocation (11;14): a cytogenetic anomaly associated with B-cell lymphomas of non-follicle centre cell lineage. J Pathol. 1991 Jan;163(1):13–18. doi: 10.1002/path.1711630104. [DOI] [PubMed] [Google Scholar]
- 3.Bosch F, Jares P, Campo E, Lopez-Guillermo A, Piris MA, Villamor N, et al. PRAD-1/cyclin D1 gene overexpression in chronic lymphoproliferative disorders: a highly specific marker of mantle cell lymphoma. Blood. 1994 Oct 15;84(8):2726–2732. [PubMed] [Google Scholar]
- 4.Dreyling M, Ferrero S, Vogt N, Klapper W European Mantle Cell Lymphoma N. New paradigms in mantle cell lymphoma: is it time to risk-stratify treatment based on the proliferative signature? Clin Cancer Res. 2014 Oct 15;20(20):5194–5206. doi: 10.1158/1078-0432.CCR-14-0836. [DOI] [PubMed] [Google Scholar]
- 5.Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004 Nov;3(11):1427–1438. [PubMed] [Google Scholar]
- 6.Leonard JP, LaCasce AS, Smith MR, Noy A, Chirieac LR, Rodig SJ, et al. Selective CDK4/6 inhibition with tumor responses by PD0332991 in patients with mantle cell lymphoma. Blood. 2012 May 17;119(20):4597–4607. doi: 10.1182/blood-2011-10-388298. [DOI] [PubMed] [Google Scholar]
- 7.Baker SJ, Reddy EP. CDK4: A Key Player in the Cell Cycle, Development, and Cancer. Genes Cancer. 2012 Nov;3(11–12):658–669. doi: 10.1177/1947601913478972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Parekh S. Taking it up a notch in MCL. Blood. 2012 Mar 1;119(9):1957–1958. doi: 10.1182/blood-2012-01-403626. [DOI] [PubMed] [Google Scholar]
- 9.Parekh S, Weniger MA, Wiestner A. New molecular targets in mantle cell lymphoma. Semin Cancer Biol. 2011 Nov;21(5):335–346. doi: 10.1016/j.semcancer.2011.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Castellano E, Downward J. RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes Cancer. 2011 Mar;2(3):261–274. doi: 10.1177/1947601911408079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010 Jan 7;463(7277):88–92. doi: 10.1038/nature08638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ramadani F, Bolland DJ, Garcon F, Emery JL, Vanhaesebroeck B, Corcoran AE, et al. The PI3K isoforms p110alpha and p110delta are essential for pre-B cell receptor signaling and B cell development. Sci Signal. 2010;3(134):ra60. doi: 10.1126/scisignal.2001104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Tabe Y, Jin L, Konopleva M, Shikami M, Kimura S, Andreeff M, et al. Class IA PI3K inhibition inhibits cell growth and proliferation in mantle cell lymphoma. Acta Haematol. 2014;131(1):59–69. doi: 10.1159/000353164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dal Col J, Zancai P, Terrin L, Guidoboni M, Ponzoni M, Pavan A, et al. Distinct functional significance of Akt and mTOR constitutive activation in mantle cell lymphoma. Blood. 2008 May 15;111(10):5142–5151. doi: 10.1182/blood-2007-07-103481. [DOI] [PubMed] [Google Scholar]
- 15.Rudelius M, Pittaluga S, Nishizuka S, Pham TH, Fend F, Jaffe ES, et al. Constitutive activation of Akt contributes to the pathogenesis and survival of mantle cell lymphoma. Blood. 2006 Sep 1;108(5):1668–1676. doi: 10.1182/blood-2006-04-015586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013 Aug 8;369(6):507–516. doi: 10.1056/NEJMoa1306220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jessen KA, Lannutti BJ. Putative roles for PI3K alpha and delta isoforms in refractory mantle cell lymphoma. Acta Haematol. 2014;131(4):218–219. doi: 10.1159/000355636. [DOI] [PubMed] [Google Scholar]
- 18.Blachly JS, Baiocchi RA. Targeting PI3-kinase (PI3K), AKT and mTOR axis in lymphoma. Br J Haematol. 2014 Oct;167(1):19–32. doi: 10.1111/bjh.13065. [DOI] [PubMed] [Google Scholar]
- 19.Chiron D, Di Liberto M, Martin P, Huang X, Sharman J, Blecua P, et al. Cell-cycle reprogramming for PI3K inhibition overrides a relapse-specific C481S BTK mutation revealed by longitudinal functional genomics in mantle cell lymphoma. Cancer Discov. 2014 Sep;4(9):1022–1035. doi: 10.1158/2159-8290.CD-14-0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chiron D, Martin P, Di Liberto M, Huang X, Ely S, Lannutti BJ, et al. Induction of prolonged early G1 arrest by CDK4/CDK6 inhibition reprograms lymphoma cells for durable PI3Kdelta inhibition through PIK3IP1. Cell Cycle. 2013 Jun 15;12(12):1892–1900. doi: 10.4161/cc.24928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Reddy MV, Akula B, Cosenza SC, Athuluridivakar S, Mallireddigari MR, Pallela VR, et al. Discovery of 8-cyclopentyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-7-oxo-7,8-dihydro-pyrid o[2,3-d]pyrimidine-6-carbonitrile (7×) as a potent inhibitor of cyclin-dependent kinase 4 (CDK4) and AMPK-related kinase 5 (ARK5) J Med Chem. 2014 Feb 13;57(3):578–599. doi: 10.1021/jm401073p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kusakai G, Suzuki A, Ogura T, Kaminishi M, Esumi H. Strong association of ARK5 with tumor invasion and metastasis. J Exp Clin Cancer Res. 2004 Jun;23(2):263–268. [PubMed] [Google Scholar]
- 23.Suzuki A, Lu J, Kusakai G, Kishimoto A, Ogura T, Esumi H. ARK5 is a tumor invasion-associated factor downstream of Akt signaling. Mol Cell Biol. 2004 Apr;24(8):3526–3535. doi: 10.1128/MCB.24.8.3526-3535.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Marzec M, Kasprzycka M, Lai R, Gladden AB, Wlodarski P, Tomczak E, et al. Mantle cell lymphoma cells express predominantly cyclin D1a isoform and are highly sensitive to selective inhibition of CDK4 kinase activity. Blood. 2006 Sep 1;108(5):1744–1750. doi: 10.1182/blood-2006-04-016634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Andjelkovic M, Maira SM, Cron P, Parker PJ, Hemmings BA. Domain swapping used to investigate the mechanism of protein kinase B regulation by 3-phosphoinositide-dependent protein kinase 1 and Ser473 kinase. Mol Cell Biol. 1999 Jul;19(7):5061–5072. doi: 10.1128/mcb.19.7.5061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005 Feb 18;307(5712):1098–1101. doi: 10.1126/science.1106148. [DOI] [PubMed] [Google Scholar]
- 27.Peterson RT, Beal PA, Comb MJ, Schreiber SL. FKBP12-rapamycin-associated protein (FRAP) autophosphorylates at serine 2481 under translationally repressive conditions. J Biol Chem. 2000 Mar 10;275(10):7416–7423. doi: 10.1074/jbc.275.10.7416. [DOI] [PubMed] [Google Scholar]
- 28.Dufner A, Andjelkovic M, Burgering BM, Hemmings BA, Thomas G. Protein kinase B localization and activation differentially affect S6 kinase 1 activity and eukaryotic translation initiation factor 4E-binding protein 1 phosphorylation. Mol Cell Biol. 1999 Jun;19(6):4525–4534. doi: 10.1128/mcb.19.6.4525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Choe G, Horvath S, Cloughesy TF, Crosby K, Seligson D, Palotie A, et al. Analysis of the phosphatidylinositol 3'-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res. 2003 Jun 1;63(11):2742–2746. [PubMed] [Google Scholar]
- 30.Zhang X, Tang N, Hadden TJ, Rishi AK. Akt, FoxO and regulation of apoptosis. Biochim Biophys Acta. 2011 Nov;1813(11):1978–1986. doi: 10.1016/j.bbamcr.2011.03.010. [DOI] [PubMed] [Google Scholar]
- 31.Rahal R, Frick M, Romero R, Korn JM, Kridel R, Chan FC, et al. Pharmacological and genomic profiling identifies NF-kappaB-targeted treatment strategies for mantle cell lymphoma. Nat Med. 2014 Jan;20(1):87–92. doi: 10.1038/nm.3435. [DOI] [PubMed] [Google Scholar]
- 32.Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006 Apr 10;24(11):1770–1783. doi: 10.1200/JCO.2005.03.7689. [DOI] [PubMed] [Google Scholar]
- 33.Kahl BS, Spurgeon SE, Furman RR, Flinn IW, Coutre SE, Brown JR, et al. A phase 1 study of the PI3Kdelta inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL) Blood. 2014 May 29;123(22):3398–3405. doi: 10.1182/blood-2013-11-537555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lv H, Zhang X, Sharma J, Reddy MV, Reddy EP, Gallo JM. Integrated pharmacokinetic-driven approach to screen candidate anticancer drugs for brain tumor chemotherapy. AAPS J. 2013 Jan;15(1):250–257. doi: 10.1208/s12248-012-9428-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhang X, Lv H, Zhou Q, Elkholi R, Chipuk JE, Reddy MV, et al. Preclinical pharmacological evaluation of a novel multiple kinase inhibitor, ON123300, in brain tumor models. Mol Cancer Ther. 2014 May;13(5):1105–1116. doi: 10.1158/1535-7163.MCT-13-0847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Montserrat E, Bosch F, Lopez-Guillermo A, Graus F, Terol MJ, Campo E, et al. CNS involvement in mantle-cell lymphoma. J Clin Oncol. 1996 Mar;14(3):941–944. doi: 10.1200/JCO.1996.14.3.941. [DOI] [PubMed] [Google Scholar]
- 37.Iyengar S, Clear A, Bodor C, Maharaj L, Lee A, Calaminici M, et al. P110alpha-mediated constitutive PI3K signaling limits the efficacy of p110delta-selective inhibition in mantle cell lymphoma, particularly with multiple relapse. Blood. 2013 Mar 21;121(12):2274–2284. doi: 10.1182/blood-2012-10-460832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ma J, Lu P, Guo A, Cheng S, Zong H, Martin P, et al. Characterization of ibrutinib-sensitive and -resistant mantle lymphoma cells. Br J Haematol. 2014 Sep;166(6):849–861. doi: 10.1111/bjh.12974. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






