Abstract
Multiple myeloma is still incurable due to an intrinsic aggressiveness or, more frequently, to the interactions of malignant plasma cells with the bone marrow (BM) microenvironment. Myeloma cells educate BM cells to support neoplastic cell growth, survival, acquisition of drug resistance resulting in disease relapse. Myeloma microenvironment is characterized by Notch signaling hyperactivation due to the increased expression of Notch1 and 2 and the ligands Jagged1 and 2 in tumor cells. Notch activation influences myeloma cell biology and promotes the reprogramming of BM stromal cells. In this work we demonstrate, in vitro, ex vivo and by using a zebrafish multiple myeloma model, that Jagged inhibition causes a decrease in both myeloma-intrinsic and stromal cell-induced resistance to currently used drugs, i.e. bortezomib, lenalidomide and melphalan. The molecular mechanism of drug resistance involves the chemokine system CXCR4/SDF1α. Myeloma cell-derived Jagged ligands trigger Notch activity in BM stromal cells. These, in turn, secrete higher levels of SDF1α in the BM microenvironment increasing CXCR4 activation in myeloma cells, which is further potentiated by the concomitant increased expression of this receptor induced by Notch activation. Consistently with the augmented pharmacological resistance, SDF1α boosts the expression of BCL2, Survivin and ABCC1. These results indicate that a Jagged-tailored approach may contribute to disrupting the pharmacological resistance due to intrinsic myeloma cell features or to the pathological interplay with BM stromal cells and, conceivably, improve patients’ response to standard-of-care therapies.Introduction
Multiple myeloma (MM) is the second most common hematologic malignancy. It is still incurable, with a median overall survival that has not been substantially extended since the introduction of anti-myeloma agents such as melphalan, lenalidomide, and bortezomib.1 The typical clinical course of MM displays a remission-relapse pattern due to the appearance of drug-resistant malignant cells, reducing the numbers of effective salvage regimens.2 Therefore, a more stable response requires the development of a therapeutic approach that prevents drug resistance.
Multiple myeloma cells accumulate in the bone marrow (BM), where they establish anomalous signaling loops with BM-residing non-tumor cells, resulting in the exchange of anti-apoptotic factors which critically induce drug resistance.3
The Notch pathway includes four transmembrane receptors (Notch1-4) activated by the interaction with five ligands (Jagged1-2 and Dll1-3-4) on adjacent cells.64 Notch receptors and ligands have been found to be aberrantly expressed in MM cells.107 We recently demonstrated that Jagged1 and the Notch transcriptional target HES5 are increasingly expressed in MM and in primary plasma cell leukemia.11 Moreover, Jagged1 and Notch1 are over-expressed during progression from the benign monoclonal gammopathy of uncertain significance (MGUS) to MM,12 while Jagged2 overexpression is already detected at the MGUS stage13 and can be ascribed to aberrant acetylation of its promoter14 or to altered post-translational processing due to aberrant expression of the ubiquitin ligase Skeletrophin.15 Finally, Notch2 hyperexpression is associated with the high-risk translocations t(14;16)(q32;q23) and t(14;20)(q32;q11).16
Recently, we and other groups pointed out the importance of Jagged ligands in providing MM cells with the ability to shape the surrounding microenvironment, interacting with osteoclast progenitors,17 and promoting a release of BM stromal cell (BMSC) key factors, including IL6, IGF1 and VEGF.1311
Aberrant levels of Notch signaling are associated with pharmacological resistance in different tumor settings6 and correlate with the expression of anti-apoptotic genes, such as BCL218 and Survivin/BIRC5,19 or regulates the expression of ABCC1,46 which contributes to multidrug resistance in MM.20
Given this, we hypothesized that the aberrant expression of Notch receptors and ligands in MM cells may foresee the development of drug resistance by inducing autonomous activation of Notch in MM cells, and by triggering Notch signaling in the surrounding BMSC and boosting their ability to support MM cell drug resistance.2221
Previous studies investigated how BMSC support the development of drug resistance in MM cells by activating Notch signaling.2523 Vice versa, here we show that also the overexpression of MM cell-derived Jagged ligands triggers Notch signaling dysregulation in the BM niche and promotes MM cell intrinsic pharmacological resistance as well as BMSC-dependent drug resistance.
Methods
Cell lines and primary cells
The human MM cell lines (HMCL), OPM2 (ACC-50) and U266 (ATCC TIB-196) were purchased from the DSMZ and ATCC, respectively. Primary cells were isolated from patient BM aspirates and MM cells were purified using the Human Whole Blood CD138 Selection Kit EasySep (StemCell Technologies). Detailed information is available in Online Supplementary Table S1. Primary BMSC were isolated as previously reported.11 The Ethical Committee of the Università degli Studi di Milano, Italy, approved this study (approval n. 8/15).
Details of all cell treatments are available in the Online Supplementary Appendix.
Luciferase reporter assay
HS5 cells were transiently transfected with a Notch reporter plasmid pNL2.1 carrying a 6xCSL Notch responsive element26 and with the vector constitutively expressing the firefly luciferase upon the thymidine kinase promoter (pGL4.54[luc2/TK]). After 24 hours (h), HS5 cells were cultured alone or placed in co-culture with scrambled (Scr) or Jagged1 and Jagged2 knockdown (J1/2KD) HMCL and incubated for 24 h. Luciferase activity was measured using Nano-Glo Dual-Luciferase Reporter assay kit (Promega) on the Glowmax instrument (Promega).
In vivo experiments on xenografted zebrafish embryos
Zebrafish AB strains obtained from the Wilson lab, University College London, UK, were maintained according to the national guidelines (Italian Ministerial Decree of 4/03/2014 2014, n. 26). All experiments were conducted within five days post fertilization.
Dechorionated zebrafish embryos were injected with Scr or J1/2KD U266 cells stained with the CM-Dil dye into the yolk (200 cells in 10 nl, 5-20 nl injection volume/embryo) with a manual microinjector (Eppendorf, Germany) using glass microinjection needles.
Xenograft-positive embryos divided randomly into the following groups: Scr-injected embryos treated with DMSO, Scr-injected embryos treated with 10 nM bortezomib, J1/2KD-injected embryos treated with DMSO, and J1/2KD-injected embryos treated with 10 nM bortezomib. Tumor growth was evaluated 48 h post injection (hpi) by fluorescence microscopy. Further details are available in the Online Supplementary Appendix.
Further details and information concerning cell cultures, RNA isolation and quantitative real-time polymerase chain reaction (qRT-PCR), RNAi assay, apoptosis assays, flow cytometry, ELISA, western blot and statistical analysis can be found in the Online Supplementary Appendix: experimental procedures.
Results
Jagged1/2 inhibition improves multiple myeloma cell response to standard-of-care drugs by increasing the anti-apoptotic background
To assess if Jagged1 and Jagged2 contribute to MM intrinsic drug resistance, we took advantage of an established knockdown (KD) approach using specific siRNAs for Jagged ligands1711 and analyzed MM cell response to three standard-of-care drugs: bortezomib (Bor), melphalan (Melph), and lenalidomide (Len). Two HMCL, OPM2 and U266 cells, were transfected with Jagged1 and Jagged2 (J1/2KD) or the scrambled control (Scr) siRNAs and then were treated with 6 nM Bor or 30 μM Melph or with 15 or 30 μM Len (respectively for U266 and OPM2 cells) (Figure 1A). The efficacy of J1/2KD was assessed by evaluating the expression of Jagged ligands and the active forms of the two Notch receptors expressed in MM cells, Notch intracellular domains 1 and 2 (NICD1 and NICD2), by western blot (Figure 1B).
The apoptosis rate of J1/2KD HMCL treated or not with Bor, Melph and Len was analyzed by flow cytometry. Figure 1C shows the effect of Bor, Melph and Len on HMCL normalized on DMSO-treated cells compared to J1/2KD HMCL treated with the drugs and normalized on untreated J1/2KD HMCL. J1/2KD induced an appreciable increase in HMCL sensitivity to standard-of-care drugs, with statistical significance reached in all cases, with the exception of U266 cells treated with Bor and Melph (P=0.06), that in any case confirmed the trend (Figure 1C). The basal apoptotic effect of J1/2KD is shown in Online Supplementary Figure S1. Concerning Len treatment, it is worth mentioning that, although Scr HMCL are resistant to this drug, J1/2KD cells acquire drug sensitivity. The selective inhibition of Jagged1 or Jagged2 is clearly less effective in comparison with the simultaneous J1/2KD, that maximizes the biological outcome (Online Supplementary Figure S2).
These results indicate that the expression of Jagged1 and 2 stimulates autonomous Notch activity in MM cells that, consequently, may be inhibited by Jagged silencing. This evidence prompted us to verify whether the increased pharmacological sensitivity of MM cells induced by J1/2KD was associated to variations in the expression of recognized anti-apoptotic Notch targets, such as BCL218 and Survivin/BIRC5,19 or with the levels of ABCC1 reported to have a significant impact in MM.272019 J1/2KD, validated by the decrease in Jagged1, 2 and HES1 and 6 gene expression, significantly inhibited the expression of the studied anti-apoptotic genes analyzed by qRT-PCR (Figure 2A and B). The effect of J1/2KD on gene expression was seen not to be due to an increased apoptosis rate in HMCL (approx. 15%) (Online Supplementary Figure S1). J1/2KD effect on anti-apoptotic effectors was assessed at protein levels by flow cytometry (Figure 2C and D and Online Supplementary Figure S3) and western blot (Online Supplementary Figure S4). By contrast, the selective inhibition of Jagged1 or Jagged2 was not sufficient to significantly down-regulate the expression of these genes (Online Supplementary Figure S5).
Jagged1 and Jagged2 silencing contributes to multiple myeloma cell ability to promote bone marrow stromal cell-mediated drug resistance
Multiple myeloma cells localize within the BM and interact with several cell types, hijacking their functions to promote tumor progression. BMSC are a crucial target in this process that sustains malignant cell proliferation and survival.22 Since Jagged-mediated activation of Notch pathway is involved in cell-cell communication,6 we hypothesized that MM cell-derived Jagged ligands could activate Notch in BMSC, possibly determining BMSC-mediated drug resistance.
To explore this hypothesis, we first verified that HMCL-derived Jagged1 and Jagged2 were able to trigger the activation of Notch signaling in a BMSC line, HS5, using a Notch reporter assay. Scr HMCL are able to activate Notch signaling in co-cultured HS5 cells (Figure 3A), while this ability is lost by J1/2KD HMCL, indicating that MM-derived Jagged may activate Notch signaling in BMSC.
To verify if Jagged-mediated activation of Notch in BMSC affected the ability of these cells to promote drug resistance in MM cells, we used flow cytometry to analyze the apoptotic rate of Scr or J1/2KD HMCL cultured alone or co-cultured with HS5 cells after treatment with standard-of-care drugs. As expected, HS5 cells show a clear trend of protection of HMCL from apoptosis induced by Bor (15% in OPM2 and 26% in U266), Melph (20% in OPM2 and 11% in U266), and Len (14% in OPM2) (Figure 3B and C), although the statistical significance was reached only in the case of OPM2 treated with Bor. Conversely and more importantly, J1/2KD induced a statistically significant increase in apoptosis, re-establishing HMCL drug sensitivity by hampering BMSC-mediated protection (HS5 cells do not display any significant increase in apoptosis; data not shown). Notably, although U266 cells were resistant to Len treatment in culture alone or in the presence of HS5 cells, apoptotic rate increased up to approximately 20% upon J1/2KD. The basal apoptotic effect of J1/2 KD on MM cells cultured with HS5 cells is reported in Online Supplementary Figure S6. As before, the selective Jagged1 or Jagged2 silencing was less effective than the simultaneous J1/2KD (Online Supplementary Figure S7).
Since HS5 cells could act as a source of paracrine/autocrine Jagged ligands, we wondered why they cannot rescue J1/2KD in MM cells. Western blot analysis indicates that the expression levels of Jagged1 and Jagged2 in HS5 cells are significantly lower than those expressed by OPM2 and U266 cells (Online Supplementary Figure S8). This can reasonably explain why, in our co-culture system, Notch signaling activated in HMCL by BMSC is not sufficient to rescue the loss of Jagged1 and Jagged2 in MM cells.
We further explored whether Jagged-mediated Notch activation in BMSC could promote the pharmacological resistance of MM cells by up-regulating the anti-apoptotic effectors previously analyzed, Survivin, BCL2, and ABCC1. To evaluate gene expression changes, we took advantage of a co-culture system including OPM2 or U266 cells with a non-human mimic model of BMSC, the murine cell line of NIH3T3 fibroblasts. This approach enabled us to precisely assess the expression levels of human (HMCL-derived) anti-apoptotic genes in co-culture by using species-specific primers. Results showed that BMSC were able to promote the expression of the anti-apoptotic effectors Survivin, BCL2, and ABCC1 in Scr HMCL, while BMSC co-cultured with J1/2KD HMCL lost this ability (Figure 4A and B). Importantly, using an entirely human co-culture system, we observed the same effects when we used flow cytometry to measure the protein expression of Survivin, BCL2, and ABCC1 in Scr or J1/2KD HMCL co-cultured with human GFP HS5 (Figure 4C-E and Online Supplementary Figure S9).
The CXCR4/SDF1α axis is a mediator of Notch pathway ability to determine drug resistance in multiple myeloma
To further study the molecular mechanisms underlying BMSC-induced drug resistance generated by Notch activation in the MM microenvironment, we explored the possible involvement of the chemokine system CXCR4/SDF1α, a key player in MM development and progression, and a downstream regulator of Notch signaling.2928 We hypothesized that Notch ability to promote pharmacological resistance in MM cells might be mediated by SDF1α. We reasoned that the main source of SDF1α in the BM was the stromal cell population. Therefore we explored if Jagged ligands, expressed by MM cells, could trigger the BMSC-mediated production of SDF1α and if J1/2KD might inhibit this effect.
The analysis was performed by taking advantage of co-culture systems of Scr or J1/2KD HMCL grown on a layer murine (NIH3T3) or human (HS5) stromal cells to measure the variations in SDF1α gene or protein expression. Results obtained by qRT-PCR with murine-specific primers (Figure 5A) indicate that HMCL promoted the activation of Notch signaling (HES5) and SDF1α gene expression in NIH3T3 cells, which could be reverted by J1/2KD.
Similar results were observed at protein level as assessed by flow cytometry analysis (Figure 5B and Online Supplementary Figure S10) on co-cultures composed of HMCL and the human GFP HS5 cells. Of note, the selective inhibition of Jagged1 or Jagged2 is clearly less effective if compared with the simultaneous J1/2KD, that maximizes the outcome on SDF1α inhibition (Online Supplementary Figure S11). Flow cytometric results were validated by ELISA on conditioned media (Figure 5C) indicating that MM cell-derived Jagged can increase SDF1α production by BMSC. We further confirmed that the variation in SDF1α expression was the consequence of Jagged-activated Notch signaling in BMSC by an assessment that showed that the stimulation with Jagged1 and/or Jagged2 peptides can increase HS5 cell-mediated secretion of SDF1α, measured by ELISA (Figure 5D). Additionally, we knocked down Notch1 expression in HS5 cells (N1KD HS5) by using a specific siRNA, as previously reported,11 and observed that SDF1α expression significantly decreased in comparison to control HS5 cells (Figure 5E). Since Notch1 silencing does not significantly affect HS5 cell viability (Online Supplementary Figure S12), we could exclude the possibility that reduction of SDF1α expression might be due to HS5 cell apoptosis.
On the other hand, we verified that J1/2KD was associated to a reduced CXCR4 expression in HMCL used in co-culture experiments. J1/2KD HMCL significantly decreased CXCR4 expression in comparison to Scr HMCL (Figure 5F and Online Supplementary Figure S13).
We assessed the outcome of SDF1α stimulation on the anti-apoptotic background of HMCL by analyzing the levels of Survivin, BCL2, and ABCC1 in U266 cells treated with 500 ng/mL SDF1α for 48 h. We observed an increase in Survivin, BCL2, and ABCC1 gene expression by qRT-PCR analysis (Figure 5G) confirmed at protein level by western blot (Figure 5H). These results suggest that SDF1α can promote MM cell ability to survive to drug administration, at least in part, by stimulating tumor cell anti-apoptotic defenses (Survivin, BCL2) and detoxification ability (ABCC1). Consistently, the treatment of U266-HS5 co-culture system with 50 μM AMD3100 (an antagonist of SDF1 binding to CXCR4), abrogated BMSC-induced resistance to the analyzed drugs (Figure 5I).
Translational potential of approaches inhibiting Jagged-mediated Notch activation in a multiple myeloma microenvironment
We further verified whether the ability of MM cells to promote BMSC-induced drug resistance was dependent on Jagged1 and Jagged2 expression by using primary co-culture systems of highly purified CD138 MM cells and BMSC isolated from BM aspirates of patients at MM onset (Online Supplementary Table S1).
Primary CD138 cells were transduced with the lentiviral vector pLL3.7 carrying Jagged1/2 shRNAs or Scr shRNAs and the efficiency was assessed by flow cytometry (Online Supplementary Figure S14). In order to maintain CD138 cell viability during ex vivo drug administration, after lentiviral transduction, they were co-cultured with primary BMSC stained with PKH26. Co-cultures were maintained for 72 h and treated for the last 24 h with 6 nM Bor (8 patients) or 30 μM Melph (10 patients), or for the last 48 h with 15 μM Len (9 patients) or the corresponding vehicle. The apoptotic rate of MM cells (expressing the GFP codified by the pLL3.7 vector) was detected by flow cytometry analyzing the GFP/Annexin-V-APC subpopulation (Figure 6A). Results showed that J1/2KD significantly increased apoptosis of primary MM cells treated with all the analyzed drugs, in agreement with the findings obtained in vitro.
To verify if the inhibitory approach based on J1/2KD had a translational potential, we recapitulated the experiments of MM-BMSC interplay by using IGOR1, a novel small molecule recently developed in our laboratory30 to uncouple Notch-Jagged interaction. IGOR1 is able to inhibit Notch activation in OPM2 cells and significantly increases the efficacy of the administered drugs, with a higher efficiency for Mel and Len (Figure 6B and C).
Jagged1 and Jagged2 blockade promotes sensitivity to bortezomib in a zebrafish xenograft myeloma model
Bortezomib is one of the most commonly used drugs for the treatment of newly-diagnosed and refractory MM patients.31 In recent years, several studies have supported the hypothesis that the development of resistance to such treatment is strongly dependent upon the BM microenvironment, with a significant contribution of the CXCR4/SDF1α axis.3432 Due to the results obtained in vitro concerning the role of this chemokine axis in the development of pharmacological resistance to Bor, we validated the effect of J1/2KD on MM cell resistance to Bor by taking advantage of a novel zebrafish xenograft MM model.
Zebrafish embryos were recently validated as a complementary in vivo model for MM that allows the rapid screening of MM cells response to chemotherapeutic drugs.35 Moreover, this model fully recapitulates the cytokine milieu present in the human BM, since zebrafish-secreted growth factors, such as IL6 and SDF1α, support MM cells growth in vivo.3533 To validate our in vitro and ex vivo findings, Scr or J1/2KD U266 cells vitally labeled with the fluorescent dye CM-Dil were injected in the yolk area of 48 hpf zebrafish embryos. Xenotransplanted embryos were visualized by fluorescent microscopy to verify the presence of MM cells at the injection site at 2 hpi (Figure 7A-D), treated or not with 10 nM Bor and, visualized at 48 hpi for tumor cell growth (Figure 7A′-D′). Representative images of whole embryos are shown in Online Supplementary Figure S15.
As shown, the addition of 10 nM Bor to the embryo medium inhibited tumor growth of approximately 57% compared to controls (Figure 7A′ and B′), without affecting embryo viability. A similar effect was induced by J1/2KD (Figure 7A′-C′), while the combination of J1/2KD and Bor significantly reduced tumor growth in comparison to all other experimental groups (-82% in comparison to the control) (Figure 7A′-D′).
Discussion
Multiple myeloma progression is characterized by development of drug resistance causing patient relapse and contributing to the fatal outcome of this disease. The close interaction of MM cells with healthy BM cells represents an important source of factors able to promote malignant cell growth and survival.
The Notch pathway is capable of mediating the cell-cell communication. Current evidence provided by different groups, including ours, highlighted the importance of Jagged ligands in the pathological communication between tumor and healthy cells within the myeloma BM. MM-derived Jagged ligands activate Notch receptors in the nearby BM cells inducing osteoclastogenesis, osteolysis,17 angiogenesis,36 and BMSC-mediated release of key cytokines including IL6, IGF1 and VEGF.1311 Moreover, the activation of Notch signaling in MM cells, induced by tumor cell-derived3837 or BMSC-derived Jagged251 stimulates MM cell proliferation,38 resistance to apoptosis,37 and a decrease in drug sensitivity.25
This work is specifically focused on the pathological communication of MM cells and BMSC mediated by Notch signaling and on its outcome on MM drug resistance. Notably, the Notch pathway is known to be a key player in BM-induced drug resistance in other hematologic malignancies. Indeed, Krampera’s group provided evidence of how the BM-driven activation of Notch3 and Notch4 in B-ALL4139 and Notch1, Notch2 and Notch4 in chronic lymphocytic leukemia,42 results in chemoresistance, while Notch1-Jagged1 crosstalk supports BM-induced drug resistance in AML.43
As far as MM is concerned, in spite of the recent advances in the field, we still do not have a complete picture of the bidirectional crosstalk between BMSC and MM cells, which is indicated by the expression of Notch receptors and ligands on both cell types.442523161211 This work aims to fill some of those gaps by providing novel information about the effects of the aberrant expression of MM-derived Jagged ligands on the intrinsic tumor cell drug resistance and by investigating a key aspect that has never been previously explored, i.e. the outcome of MM-derived Jagged ligands on BMSC-induced drug resistance.
To address these issues, we interfered with the mRNA expression of MM-derived Jagged ligands and investigated J1/2KD outcomes in tumor cells and in surrounding BMSC. We observed in vitro that MM cell-derived Jagged ligands could trigger Notch signaling in the nearby MM cells by homotypic interaction. Notch activation resulted in the increased expression of anti-apoptotic effectors including BCL2, Survivin, and the multidrug resistance transporter ABCC1, along with the increase in MM cell survival to standard-of-care drugs, such as Bor, Len, and Melph. Notably, besides observing homotypic activation of Notch signaling among MM cells, we found that HMCL can trigger Notch signaling in the neighboring BMSC and, in turn, Notch activation boosts the ability of BMSC to increase the pharmacological resistance of MM cells. This effect was clearly dependent on MM-derived Jagged ligands, since J1/2KD completely abrogated BMSC support. At least in part, the pro-tumor effect of Notch-“educated” BMSC was due to their ability to increase SDF1α levels in the BM microenvironment. Indeed, soluble or MM cell-derived Jagged ligands may induce a Notch-dependent increase in SDF1α secretion by BMSC; on the contrary, J1/2KD HMCL lose this ability and N1KD interferes with BMSC to release SDF1α.
The Notch-dependent activation of SDF1α secretion by BMSC is potentially more important than the previously observed secretion induced by Notch activation in MM cells,29 since BMSC are the most effective producers of this cytokine in the BM.
To complete the picture of a Notch signaling effect on the SDF1α/CXCR4 axis in myeloma BM, we also demonstrated that MM cell-derived Jagged ligands may further enhance the anti-apoptotic signaling of SDF1α by stimulating the expression of its receptor CXCR4 on the MM cell surface.
The contribution of the SDF1α/CXCR4 axis to MM pharmacological resistance was confirmed by the ability of the antagonist molecule AMD3100 to abrogate U266 cell resistance to (Bor), (Melph) and (Len) induced by BMSC, consistent with the findings of Azab et al.32
Although the downstream molecular mechanisms of Notch-associated drug resistance in MM still need to be fully elucidated, we showed that the secreted SDF1α can stimulate general mechanisms, including tumor cell anti-apoptotic background, by up-regulating BCL2 and Survivin, or drug extrusion mediated by ABCC1. These antiapoptotic proteins are particularly relevant to MM. Indeed, BCL2 and Survivin are over-expressed in MM cells, where they play an important role in cell survival, and significantly correlate with disease stage;452720 on the other hand, xenobiotic transporters, such as ABCC1, are well known mediators of MM multidrug resistance,20 modulated by Notch in different cancer settings.46
The general validity of these novel findings stems from the observed improvement in drug-response promoted by J1/2KD in ex vivo co-culture systems of CD138 MM cells and BMSC from BM aspirates of newly-diagnosed MM patients.
Additionally, in vivo validation of these findings in a zebrafish xenograft MM model engrafted with U266 cells confirmed that J1/2KD promoted an increased sensitivity to Bor in vivo, showing a wider decrease in tumor burden compared to the control.
The present results provide novel and important information to help improve the current picture on the effect of Notch-mediated communication in myeloma BM. Indeed, since both BMSC and MM cells carry Notch receptors and ligands, their bidirectional crosstalk needs to be taken into consideration. We sought to fill the gap in the available information on the role of MM cells, such as Notch signaling sending cells in the BM. Here we discuss our findings according to the previous literature data in order to summarize the overall picture (Figure 8). Previous work reported the consequences of Notch activity in MM cells (mainly using g-secretase inhibitors), identifying the following molecular mechanisms: i) upregulation of p21 induced MM cell growth inhibition and increased survival;23 ii) Notch/HES1 mediated downregulation of the pro-apoptotic protein Noxa;24 iii) Notch up-regulated expression of integrin αvβ5 resulting in increased adhesion to vitronectin and consequent protection from pro-apoptotic drugs;47iv) upregulation of the enzyme cytochrome P450,44 implicated in drug metabolism and in the onset of several malignancies.48 Concerning the contribution of Notch in BMSC-dependent drug resistance, previous investigations were focused on the autonomous contribution of BMSC-derived Notch ligands in MM cell behavior (Figure 8).442523
In this work, we found that the alteration induced in the BM by the presence of MM cells aberrantly expressing Jagged ligands is a key step in “educating” the tumor microenvironment to a pro-tumor type of behavior.
Indeed, MM cell-derived Jagged1 and 2 may switch on Notch signaling in tumor and non-tumor BMSC by triggering Notch signaling, activating MM cell anti-apoptotic background, increasing SDF1α level in the BM, and, finally, resulting in supporting MM cell resistance to standard-of-care drugs (Figure 8).
Overall, our findings provide the proof-of-principle that selective targeting of Jagged ligands in MM cells can restore tumor cell sensitivity to therapy, laying the foundation for the development of combined low-toxic therapeutic options to restore drug sensitivity and overcome fatal drug resistance of relapsing MM patients. Recently, inhibitory small molecules30 or neutralizing antibodies49 directed to inhibit the activation of Notch signaling mediated by Jagged ligands have been developed. This prompted us to confirm the translational potential of our results by testing the anti-tumor effect of an inhibitory small molecule developed in our laboratory, IGOR1, which was directed to uncouple Notch-Jagged interaction.30 In vitro results showed that IGOR1 had the ability to increase MM cell pharmacological response, with higher efficacy if combined with Melph and Len.
The importance of our results stems from the evidence that a Jagged-tailored therapy might represent a more suitable clinical approach to achieve the inhibition of Notch signaling in the BM of MM patients. Indeed, it lacks the potential adverse effects of pan-Notch blockade obtained with g-secretase inhibitors (GSI), that provided promising results in an in vivo MM model by increasing the chemotherapeutic effect of doxorubicin and melphalan,24 but that were associated with severe gastrointestinal toxicity due to intestine metaplasia.5150
Footnotes
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/105/7/1925
- FundingThis study was supported by grants from Associazione Italiana Ricerca sul Cancro, AIRC Investigator Grant to RC (20614) and AN (16722), My First Grant to AP (18741); Fondazione Italiana per la Ricerca sul Cancro to MC (post-doctoral fellowship 18013) and ET (post-doctoral fellowship 19370); Università degli Studi di Milano to RC (Linea 2B-2017 - Dept. Health Sciences), to NP (post-doctoral fellowship type A), SG, MTP and EL (PhD fellowship in Molecular and Translational Medicine), DG (PhD fellowship in Experimental Medicine).
- Received March 10, 2019.
- Accepted September 26, 2019.
References
- Kumar SK, Rajkumar V, Kyle RA. Multiple myeloma. Nat Rev Dis Primers. 2017; 3:17046. Google Scholar
- Nijhof IS, van de Donk N, Zweegman S, Lokhorst HM. Current and New Therapeutic Strategies for Relapsed and Refractory Multiple Myeloma: An Update. Drugs. 2018; 78(1):19-37. Google Scholar
- Manier S, Sacco A, Leleu X, Ghobrial IM, Roccaro AM. Bone marrow microenvironment in multiple myeloma progression. J Biomed Biotechnol. 2012; 2012:157496. PubMedGoogle Scholar
- Chiaramonte R, Basile A, Tassi E. A wide role for NOTCH1 signaling in acute leukemia. Cancer Lett. 2005; 219(1):113-120. PubMedhttps://doi.org/10.1016/j.canlet.2004.07.022Google Scholar
- Colombo M, Mirandola L, Chiriva-Internati M. Cancer Cells Exploit Notch Signaling to Redefine a Supportive Cytokine Milieu. Front Immunol. 2018; 9:1823. PubMedhttps://doi.org/10.3389/fimmu.2018.01823Google Scholar
- Platonova N, Lesma E, Basile A. Targeting Notch as a Therapeutic Approach for Human Malignancies. Curr Pharm Des. 2017; 23(1):108-134. Google Scholar
- Colombo M, Mirandola L, Platonova N. Notch-directed microenvironment reprogramming in myeloma: a single path to multiple outcomes. Leukemia. 2013; 27(5):1009-1018. PubMedhttps://doi.org/10.1038/leu.2013.6Google Scholar
- Mirandola L, Comi P, Cobos E, Kast WM, Chiriva-Internati M, Chiaramonte R. Notching from T-cell to B-cell lymphoid malignancies. Cancer Lett. 2011; 308(1):1-13. PubMedhttps://doi.org/10.1016/j.canlet.2011.05.009Google Scholar
- Platonova N, Manzo T, Mirandola L. PI3K/AKT signaling inhibits NOTCH1 lysosome-mediated degradation. Genes Chromosomes Cancer. 2015; 54(8):516-526. PubMedhttps://doi.org/10.1002/gcc.22264Google Scholar
- Colombo M, Galletti S, Garavelli S. Notch signaling deregulation in multiple myeloma: A rational molecular target. Oncotarget. 2015; 6(29):26826-26840. Google Scholar
- Colombo M, Galletti S, Bulfamante G. Multiple myeloma-derived Jagged ligands increases autocrine and paracrine interleukin-6 expression in bone marrow niche. Oncotarget. 2016; 7(35):56013-56029. Google Scholar
- Skrtic A, Korac P, Kristo DR, Ajdukovic Stojisavljevic R, Ivankovic D, Dominis M. Immunohistochemical analysis of NOTCH1 and JAGGED1 expression in multiple myeloma and monoclonal gammopathy of undetermined significance. Hum Pathol. 2010; 41(12):1702-1710. PubMedhttps://doi.org/10.1016/j.humpath.2010.05.002Google Scholar
- Houde C, Li Y, Song L. Overexpression of the NOTCH ligand JAG2 in malignant plasma cells from multiple myeloma patients and cell lines. Blood. 2004; 104(12):3697-3704. PubMedhttps://doi.org/10.1182/blood-2003-12-4114Google Scholar
- Ghoshal P, Nganga AJ, Moran-Giuati J. Loss of the SMRT/NCoR2 corepressor correlates with JAG2 overexpression in multiple myeloma. Cancer Res. 2009; 69(10):4380-4387. PubMedhttps://doi.org/10.1158/0008-5472.CAN-08-3467Google Scholar
- Takeuchi T, Adachi Y, Ohtsuki Y. Skeletrophin, a novel ubiquitin ligase to the intracellular region of Jagged-2, is aberrantly expressed in multiple myeloma. Am J Pathol. 2005; 166(6):1817-1826. PubMedhttps://doi.org/10.1016/S0002-9440(10)62491-1Google Scholar
- van Stralen E, van de Wetering M, Agnelli L, Neri A, Clevers HC, Bast BJ. Identification of primary MAFB target genes in multiple myeloma. Exp Hematol. 2009; 37(1):78-86. PubMedhttps://doi.org/10.1016/j.exphem.2008.08.006Google Scholar
- Colombo M, Thummler K, Mirandola L. Notch signaling drives multiple myeloma induced osteoclastogenesis. Oncotarget. 2014; 5(21):10393-10406. Google Scholar
- Ye QF, Zhang YC, Peng XQ, Long Z, Ming YZ, He LY. Silencing Notch-1 induces apoptosis and increases the chemosensitivity of prostate cancer cells to docetaxel through Bcl-2 and Bax. Oncol Lett. 2012; 3(4):879-884. PubMedGoogle Scholar
- Ju JH, Yang W, Oh S. HER2 stabilizes survivin while concomitantly down-regulating survivin gene transcription by suppressing Notch cleavage. Biochem J. 2013; 451(1):123-134. PubMedhttps://doi.org/10.1042/BJ20121716Google Scholar
- Buda G, Ricci D, Huang CC. Polymorphisms in the multiple drug resistance protein 1 and in P-glycoprotein 1 are associated with time to event outcomes in patients with advanced multiple myeloma treated with bortezomib and pegylated liposomal doxorubicin. Ann Hematol. 2010; 89(11):1133-1140. PubMedhttps://doi.org/10.1007/s00277-010-0992-3Google Scholar
- Kawano Y, Moschetta M, Manier S. Targeting the bone marrow microenvironment in multiple myeloma. Immunol Rev. 2015; 263(1):160-172. PubMedhttps://doi.org/10.1111/imr.12233Google Scholar
- Colombo M, Platonova N, Giannandrea D, Palano MT, Basile A, Chiaramonte R. Reestablishing Apoptosis Competence in Bone Associated Cancers via Communicative Reprogramming Induced Through Notch Signaling Inhibition. Front Pharmacol. 2019; 10:145. Google Scholar
- Nefedova Y, Cheng P, Alsina M, Dalton WS, Gabrilovich DI. Involvement of Notch-1 signaling in bone marrow stroma-mediated de novo drug resistance of myeloma and other malignant lymphoid cell lines. Blood. 2004; 103(9):3503-3510. PubMedhttps://doi.org/10.1182/blood-2003-07-2340Google Scholar
- Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008; 111(4):2220-2229. PubMedhttps://doi.org/10.1182/blood-2007-07-102632Google Scholar
- Muguruma Y, Yahata T, Warita T. Jagged1-induced Notch activation contributes to the acquisition of bortezomib resistance in myeloma cells. Blood Cancer J. 2017; 7(12):650. Google Scholar
- Kato H, Taniguchi Y, Kurooka H. Involvement of RBP-J in biological functions of mouse Notch1 and its derivatives. Development. 1997; 124(20):4133-4141. PubMedGoogle Scholar
- Khan N, Kahl B. Targeting BCL-2 in Hematologic Malignancies. Target Oncol. 2018; 13(3):257-267. Google Scholar
- Chiaramonte R, Colombo M, Bulfamante G. Notch pathway promotes ovarian cancer growth and migration via CXCR4/SDF1alpha chemokine system. Int J Biochem Cell Biol. 2015; 66:134-140. Google Scholar
- Mirandola L, Apicella L, Colombo M. Anti-Notch treatment prevents multiple myeloma cells localization to the bone marrow via the chemokine system CXCR4/SDF-1. Leukemia. 2013; 27(7):1558-1566. PubMedhttps://doi.org/10.1038/leu.2013.27Google Scholar
- Platonova N, Parravicini C, Sensi C. Identification of small molecules uncoupling the Notch::Jagged interaction through an integrated high-throughput screening. PloS One. 2017; 12(11):e0182640. Google Scholar
- Richardson PG, Sonneveld P, Schuster M. Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood. 2007; 110(10):3557-3560. PubMedhttps://doi.org/10.1182/blood-2006-08-036947Google Scholar
- Azab AK, Runnels JM, Pitsillides C. CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood. 2009; 113(18):4341-4351. PubMedhttps://doi.org/10.1182/blood-2008-10-186668Google Scholar
- Sacco A, Roccaro AM, Ma D. Cancer Cell Dissemination and Homing to the Bone Marrow in a Zebrafish Model. Cancer Res. 2016; 76(2):463-471. PubMedhttps://doi.org/10.1158/0008-5472.CAN-15-1926Google Scholar
- Waldschmidt JM, Simon A, Wider D. CXCL12 and CXCR7 are relevant targets to reverse cell adhesion-mediated drug resistance in multiple myeloma. Br J Haematol. 2017; 179(1):36-49. Google Scholar
- Lin J, Zhang W, Zhao J-J. A clinically relevant in vivo zebrafish model of human multiple myeloma (MM) to study preclinical therapeutic efficacy. Blood. 2016; 128(2):249-252. PubMedhttps://doi.org/10.1182/blood-2016-03-704460Google Scholar
- Saltarella I, Frassanito MA, Lamanuzzi A. Homotypic and Heterotypic Activation of the Notch Pathway in Multiple Myeloma–Enhanced Angiogenesis: A Novel Therapeutic Target?. Neoplasia. 2019; 21(1):93-105. Google Scholar
- Jia CM, Tian YY, Quan LN, Jiang L, Liu AC. miR-26b-5p suppresses proliferation and promotes apoptosis in multiple myeloma cells by targeting JAG1. Pathol Res Pract. 2018; 214(9):1388-1394. Google Scholar
- Jundt F, Probsting KS, Anagnostopoulos I. Jagged1-induced Notch signaling drives proliferation of multiple myeloma cells. Blood. 2004; 103(9):3511-3515. PubMedhttps://doi.org/10.1182/blood-2003-07-2254Google Scholar
- Nwabo Kamdje AH, Krampera M. Notch signaling in acute lymphoblastic leukemia: any role for stromal microenvironment?. Blood. 2011; 118(25):6506-6514. PubMedhttps://doi.org/10.1182/blood-2011-08-376061Google Scholar
- Nwabo Kamdje AH, Mosna F, Bifari F. Notch-3 and Notch-4 signaling rescue from apoptosis human B-ALL cells in contact with human bone marrow-derived mesenchymal stromal cells. Blood. 2011; 118(2):380-389. PubMedhttps://doi.org/10.1182/blood-2010-12-326694Google Scholar
- Takam Kamga P, Dal Collo G, Midolo M, Adamo A, Delfino P. Inhibition of Notch Signaling Enhances Chemosensitivity in B-cell Precursor Acute Lymphoblastic Leukemia. Cancer Res. 2019; 79(3):639-649. PubMedhttps://doi.org/10.1158/0008-5472.CAN-18-1617Google Scholar
- Nwabo Kamdje AH, Bassi G, Pacelli L. Role of stromal cell-mediated Notch signaling in CLL resistance to chemotherapy. Blood Cancer J. 2012; 2(5):e73-e73. PubMedhttps://doi.org/10.1038/bcj.2012.17Google Scholar
- Takam Kamga P, Bassi G, Cassaro A. Notch signalling drives bone marrow stromal cell-mediated chemoresistance in acute myeloid leukemia. Oncotarget. 2016; 7(16):21713-21727. Google Scholar
- Xu D, Hu J, De Bruyne E. Dll1/Notch activation contributes to bortezomib resistance by upregulating CYP1A1 in multiple myeloma. Biochem Biophys Res Commun. 2012; 428(4):518-524. PubMedhttps://doi.org/10.1016/j.bbrc.2012.10.071Google Scholar
- Romagnoli M, Trichet V, David C. Significant impact of survivin on myeloma cell growth. Leukemia. 2007; 21(5):1070-1078. PubMedGoogle Scholar
- Cho S, Lu M, He X. Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proc Natl Acad Sci U S A. 2011; 108(51):20778-20783. PubMedhttps://doi.org/10.1073/pnas.1019452108Google Scholar
- Ding Y, Shen Y. Notch increased vitronection adhesion protects myeloma cells from drug induced apoptosis. Biochem Biophys Res Commun. 2015; 467(4):717-722. Google Scholar
- Go R-E, Hwang K-A, Choi K-C. Cytochrome P450 1 family and cancers. J Steroid Biochem Mol Biol. 2015; 147:24-30. PubMedGoogle Scholar
- Li D, Masiero M, Banham AH, Harris AL. The Notch Ligand Jagged1 as a Target for Anti-Tumor Therapy. Front Oncol. 2014; 4:254. PubMedhttps://doi.org/10.3389/fonc.2014.00254Google Scholar
- Milano J, McKay J, Dagenais C. Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004; 82(1):341-358. PubMedhttps://doi.org/10.1093/toxsci/kfh254Google Scholar
- Wong GT, Manfra D, Poulet FM. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004; 279(13):12876-12882. PubMedhttps://doi.org/10.1074/jbc.M311652200Google Scholar