Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731936

ABSTRACT

Multiple myeloma is a malignancy characterized by the accumulation of malignant plasma cells in bone marrow and the production of monoclonal immunoglobulin. A hallmark of cancer is the evasion of immune surveillance. Histone deacetylase inhibitors have been shown to promote the expression of silenced molecules and hold potential to increase the anti-MM efficacy of immunotherapy. The aim of the present work was to assess the potential effect of tinostamustine (EDO-S101), a first-in-class alkylating deacetylase inhibitor, in combination with daratumumab, an anti-CD38 monoclonal antibody (mAb), through different preclinical studies. Tinostamustine increases CD38 expression in myeloma cell lines, an effect that occurs in parallel with an increment in CD38 histone H3 acetylation levels. Also, the expression of MICA and MICB, ligands for the NK cell activating receptor NKG2D, augments after tinostamustine treatment in myeloma cell lines and primary myeloma cells. Pretreatment of myeloma cell lines with tinostamustine increased the sensitivity of these cells to daratumumab through its different cytotoxic mechanisms, and the combination of these two drugs showed a higher anti-myeloma effect than individual treatments in ex vivo cultures of myeloma patients' samples. In vivo data confirmed that tinostamustine pretreatment followed by daratumumab administration significantly delayed tumor growth and improved the survival of mice compared to individual treatments. In summary, our results suggest that tinostamustine could be a potential candidate to improve the efficacy of anti-CD38 mAbs.


Subject(s)
ADP-ribosyl Cyclase 1 , Antibodies, Monoclonal , Multiple Myeloma , NK Cell Lectin-Like Receptor Subfamily K , Animals , Humans , Mice , ADP-ribosyl Cyclase 1/drug effects , ADP-ribosyl Cyclase 1/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Drug Synergism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NK Cell Lectin-Like Receptor Subfamily K/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use
2.
Am J Pathol ; 186(8): 2171-2182, 2016 08.
Article in English | MEDLINE | ID: mdl-27301357

ABSTRACT

IL-8 promotes cancer cell growth, survival, angiogenesis, and metastasis in several tumors. Herein, we investigated the sources of IL-8 production in multiple myeloma (MM) and its potential roles in MM pathogenesis. We found that bone marrow cells from patients with MM secreted higher amounts of IL-8 than healthy donors. IL-8 production was detected in cultures of CD138(+) plasma cells and CD138(-) cells isolated from bone marrows of MM patients, and in three of seven human myeloma cell lines (HMCLs) analyzed. Interactions between MM and stromal cells increased IL-8 secretion by stromal cells through cell-cell adhesion and soluble factors. Interestingly, IL8 expression also increased in HMCLs, stromal cells, and osteoclasts after treatment with the antimyeloma drugs melphalan and bortezomib. In fact, the effect of bortezomib on IL-8 production was higher than that exerted by stromal-MM cell interactions. Addition of exogenous IL-8 did not affect growth of HMCLs, although it protected cells from death induced by serum starvation through a caspase-independent mechanism. Furthermore, IL-8 induced by stromal-MM cell interactions strongly contributed to osteoclast formation in vitro, because osteoclastogenesis was markedly reduced by IL-8-specific neutralizing antibodies. In conclusion, our results implicate IL-8 in myeloma bone disease and point to the potential utility of an anti-IL-8 therapy to prevent unwanted effects of IL-8 up-regulation on survival, angiogenesis, and osteolysis in MM.


Subject(s)
Interleukin-8/biosynthesis , Multiple Myeloma/pathology , Osteogenesis/physiology , Cell Separation , Cell Survival , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Humans , Multiple Myeloma/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Polymerase Chain Reaction , Stromal Cells/metabolism , Up-Regulation
3.
Haematologica ; 102(1): 168-175, 2017 01.
Article in English | MEDLINE | ID: mdl-27540138

ABSTRACT

Despite new advances in multiple myeloma treatment and the consequent improvement in overall survival, most patients relapse or become refractory to treatment. This suggests that new molecules and combinations that may further inhibit important survival pathways for these tumor cells are needed. In this context, zalypsis is a novel compound, derived from marine organisms, with a powerful preclinical anti-myeloma effect based on the sensitivity of malignant plasma cells to DNA-damage induction; and it has already been tested in a phase I/II clinical trial in multiple myeloma. We hypothesized that the addition of this compound to the combination of bortezomib plus dexamethasone may improve efficacy with acceptable toxicity. The triple combination demonstrated strong synergy and higher efficacy compared with double combinations; not only in vitro, but also ex vivo and, especially, in in vivo experiments. The triple combination triggers cell death, mainly through a synergistic induction of DNA damage and a decrease in the nuclear localization of nuclear factor kappa B. Our findings support the clinical evaluation of this combination for relapsed and refractory myeloma patients.


Subject(s)
Bortezomib/pharmacology , DNA Damage/drug effects , Dexamethasone/pharmacology , Multiple Myeloma/genetics , Tetrahydroisoquinolines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Drug Synergism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , NF-kappa B/metabolism , Protein Transport/drug effects , Xenograft Model Antitumor Assays
4.
Haematologica ; 102(12): 2113-2124, 2017 12.
Article in English | MEDLINE | ID: mdl-28860344

ABSTRACT

Kinesin spindle protein inhibition is known to be an effective therapeutic approach in several malignancies. Filanesib (ARRY-520), an inhibitor of this protein, has demonstrated activity in heavily pre-treated multiple myeloma patients. The aim of the work herein was to investigate the activity of filanesib in combination with pomalidomide plus dexamethasone backbone, and the mechanisms underlying the potential synergistic effect. The ability of filanesib to enhance the activity of pomalidomide plus dexamethasone was studied in several in vitro and in vivo models. Mechanisms of this synergistic combination were dissected by gene expression profiling, immunostaining, cell cycle and short interfering ribonucleic acid studies. Filanesib showed in vitro, ex vivo, and in vivo synergy with pomalidomide plus dexamethasone treatment. Importantly, the in vivo synergy observed in this combination was more evident in large, highly proliferative tumors, and was shown to be mediated by the impairment of mitosis transcriptional control, an increase in monopolar spindles, cell cycle arrest and the induction of apoptosis in cells in proliferative phases. In addition, the triple combination increased the activation of the proapoptotic protein BAX, which has previously been associated with sensitivity to filanesib, and could potentially be used as a predictive biomarker of response to this combination. Our results provide preclinical evidence for the potential benefit of the combination of filanesib with pomalidomide and dexamethasone, and supported the initiation of a recently activated trial being conducted by the Spanish Myeloma group which is investigating this combination in relapsed myeloma patients.


Subject(s)
Dexamethasone/therapeutic use , Kinesins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Thiadiazoles/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Drug Synergism , Humans , Mice , Thalidomide/therapeutic use , Treatment Outcome
5.
Blood ; 122(22): 3591-8, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24072855

ABSTRACT

Circulating myeloma tumor cells (CTCs) as defined by the presence of peripheral blood (PB) clonal plasma cells (PCs) are a powerful prognostic marker in multiple myeloma (MM). However, the biological features of CTCs and their pathophysiological role in MM remains unexplored. Here, we investigate the phenotypic, cytogenetic, and functional characteristics as well as the circadian distribution of CTCs vs paired bone marrow (BM) clonal PCs from MM patients. Our results show that CTCs typically represent a unique subpopulation of all BM clonal PCs, characterized by downregulation (P < .05) of integrins (CD11a/CD11c/CD29/CD49d/CD49e), adhesion (CD33/CD56/CD117/CD138), and activation molecules (CD28/CD38/CD81). Fluorescence in situ hybridization analysis of fluorescence-activated cell sorter-sorted CTCs also unraveled different cytogenetic profiles vs paired BM clonal PCs. Moreover, CTCs were mostly quiescent and associated with higher clonogenic potential when cocultured with BM stromal cells. Most interestingly, CTCs showed a circadian distribution which fluctuates in a similar pattern to that of CD34(+) cells, and opposite to stromal cell-derived factor 1 plasma levels and corresponding surface expression of CXC chemokine receptor 4 on clonal PCs, suggesting that in MM, CTCs may egress to PB to colonize/metastasize other sites in the BM during the patients' resting period.


Subject(s)
Multiple Myeloma/blood , Neoplastic Cells, Circulating/pathology , Antigens, CD/blood , Cell Cycle , Circadian Rhythm , Cytogenetic Analysis , Humans , Immunophenotyping , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Neoplastic Cells, Circulating/classification , Neoplastic Cells, Circulating/immunology , Plasma Cells/classification , Plasma Cells/immunology , Plasma Cells/pathology , Prognosis , Prospective Studies , Tumor Stem Cell Assay
7.
J Pathol ; 229(1): 36-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22711564

ABSTRACT

Myeloma cell adhesion dependent on α4ß1 integrin is crucial for the progression of multiple myeloma (MM). The α4ß1-dependent myeloma cell adhesion is up-regulated by the chemokine CXCL12, and pharmacological blockade of the CXCL12 receptor CXCR4 leads to defective myeloma cell homing to bone marrow (BM). Sphingosine-1-phosphate (S1P) regulates immune cell trafficking upon binding to G-protein-coupled receptors. Here we show that myeloma cells express S1P1, a receptor for S1P. We found that S1P up-regulated the α4ß1-mediated myeloma cell adhesion and transendothelial migration stimulated by CXCL12. S1P promoted generation of high-affinity α4ß1 that efficiently bound the α4ß1 ligand VCAM-1, a finding that was associated with S1P-triggered increase in talin-ß1 integrin association. Furthermore, S1P cooperated with CXCL12 for enhancement of α4ß1-dependent adhesion strengthening and spreading. CXCL12 and S1P activated the DOCK2-Rac1 pathway, which was required for stimulation of myeloma cell adhesion involving α4ß1. Moreover, in vivo analyses indicated that S1P contributes to optimizing the interactions of MM cells with the BM microvasculture and for their lodging inside the bone marrow. The regulation of α4ß1-dependent adhesion and migration of myeloma cells by CXCL12-S1P combined activities might have important consequences for myeloma disease progression.


Subject(s)
Bone Marrow/metabolism , Cell Adhesion , Chemokine CXCL12/metabolism , Integrin alpha4beta1/metabolism , Lysophospholipids/metabolism , Multiple Myeloma/metabolism , Sphingosine/analogs & derivatives , Stromal Cells/metabolism , Transendothelial and Transepithelial Migration , Animals , Bone Marrow/blood supply , Bone Marrow/immunology , Bone Marrow/pathology , Cell Shape , Coculture Techniques , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/metabolism , Humans , Integrin alpha5beta1/metabolism , K562 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , RNA Interference , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine/metabolism , Stromal Cells/immunology , Stromal Cells/pathology , Talin/metabolism , Time Factors , Transfection , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/metabolism , rac1 GTP-Binding Protein/metabolism
8.
Invest New Drugs ; 31(1): 200-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22773056

ABSTRACT

INTRODUCTION: The RAS/RAF/MEK/ERK signaling pathway plays an important role in osteoclast (OC) differentiation and survival mediated by macrophage-colony stimulating factor (M-CSF). Also, vascular endothelial growth factor (VEGF) may greatly influence OC formation and resorption through VEGFR1 and VEGFR2. RAF265 is a novel, orally bioavailable dual inhibitor of RAF kinase and VEGFR2. METHODS: Effect of RAF265 on osteoclastogenesis from peripheral blood mononuclear cells (PBMCs) and OC resorption on calcium-coated wells was assessed by appropriate in vitro assays. Immunoblotting, real-time RT-PCR and flow cytometry were used to evaluate RAF265 mechanism of action. RESULTS: RAF265 significantly impaired in vitro differentiation of PBMCs to OCs induced by receptor activator of NF-kB ligand (RANKL) and M-CSF (IC(50) ≅ 160 nM). In parallel, RAF265 exerted a potent inhibition of OC resorptive capacity (IC(50) ≅ 20 nM). RAF265 treatment led to ERK inhibition and diminished expression of c-fos and NFATc1 (nuclear factor of activated T cells, calcineurin-dependent 1), which would likely account for inhibition of osteoclastogenesis. The reduced gene expression of aVb3 integrin, CCR1, cathepsin K, carbonic anhydrase II, matrix metalloproteinase 9, urokinase and tissue-type plasminogen activators, vacuolar H(+)-ATPase subunit (ATP6V1A) and Rab7 GTPase would probably mediate RAF265 hindered resorption. RAF265 inhibitory effect on VEGFR2 (noticeable at 10-50 nM) was also found to be implicated in the potent inhibition of this agent on OC function. CONCLUSIONS: We have found a new therapeutic application for RAF265 as an inhibitory agent of osteoclastogenesis and OC function, which might be useful for the treatment of skeletal disorders associated with increased bone resorption.


Subject(s)
Imidazoles/pharmacology , Osteoclasts/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Bone Resorption/drug therapy , Cells, Cultured , Humans , Leukocytes, Mononuclear/cytology , Osteoclasts/cytology
9.
Haematologica ; 97(7): 1110-4, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22315496

ABSTRACT

Although new therapies have doubled the survival of multiple myeloma patients, this remains an incurable disease. It has been postulated that the so-called myeloma cancer stem cells would be responsible for tumor initiation and relapse but their unequivocal identification remains unclear. Here, we investigated in a panel of myeloma cell lines the presence of CD20(+) cells harboring a stem-cell phenotype. Thus, only a small population of CD20(dim+) cells (0.3%) in the RPMI-8226 cell line was found. CD20(dim+) RPMI-8226 cells expressed the plasma cell markers CD38 and CD138 and were CD19(-)CD27(-). Additionally, CD20(dim+) RPMI-8226 cells did not exhibit stem-cell markers as shown by gene expression profiling and the aldehyde dehydrogenase assay. Furthermore, we demonstrated that CD20(dim+) RPMI-8226 cells are not essential for CB17-SCID mice engraftment and show lower self-renewal potential than the CD20(-) RPMI-8226 cells. These results do not support CD20 expression for the identification of myeloma cancer stem cells.


Subject(s)
Antigens, CD20/genetics , Biomarkers, Tumor/genetics , Multiple Myeloma/metabolism , Neoplastic Stem Cells/metabolism , Aldehyde Dehydrogenase/analysis , Animals , Antigens, CD/genetics , Cell Line, Tumor , Flow Cytometry , Gene Expression , Gene Expression Profiling , Humans , Immunophenotyping , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Transplantation, Heterologous
10.
Ann Hematol ; 91(2): 257-69, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21720745

ABSTRACT

Despite the advantage observed with novel drugs such as bortezomib, thalidomide, or lenalidomide, multiple myeloma (MM) remains incurable and there is a clear need for new drugs or combinations based on the pathogenetic mechanism of MM. One of the proposed mechanisms in MM pathogenesis is the involvement of kinase molecules in the growth and survival of myelomatous cells. In this study, we have explored the optimal combination for dasatinib, a tyrosine kinase inhibitor, in MM cells. A clear synergistic effect was observed with the triple combination of dasatinib with bortezomib and dexamethasone which was evident even in the presence of bone marrow microenvironment. Experiments performed on freshly isolated patients' cells also demonstrated potentiation of response in the triple as compared with the agents alone or in double combinations. Gene expression profiling experiments provided some clues on the transcriptional rationale underlying this potentiation, as the triple combination led to significant deregulation of genes involved in cell death, cell growth, proliferation, DNA replication, repair and recombination, and cell-cell signaling. Some of these results were further confirmed by apoptosis and cell cycle experiments and also by Western blot and PCR. These data provide the rationale for the use of this novel combination in MM patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Dexamethasone/therapeutic use , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Transcriptome/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dasatinib , Dexamethasone/pharmacology , Drug Synergism , Gene Expression Profiling , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology
11.
Blood ; 113(16): 3781-91, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19020308

ABSTRACT

Multiple myeloma (MM) remains incurable, and new drugs with novel mechanisms of action are still needed. In this report, we have analyzed the action of Zalypsis, an alkaloid analogous to certain natural marine compounds, in MM. Zalypsis turned out to be the most potent antimyeloma agent we have tested so far, with IC(50) values from picomolar to low nanomolar ranges. It also showed remarkable ex vivo potency in plasma cells from patients and in MM cells in vivo xenografted in mice. Besides the induction of apoptosis and cell cycle arrest, Zalypsis provoked DNA double-strand breaks (DSBs), evidenced by an increase in phospho-histone-H2AX and phospho-CHK2, followed by a striking overexpression of p53 in p53 wild-type cell lines. In addition, in those cell lines in which p53 was mutated, Zalypsis also provoked DSBs and induced cell death, although higher concentrations were required. Immunohistochemical studies in tumors also demonstrated histone-H2AX phosphorylation and p53 overexpression. Gene expression profile studies were concordant with these results, revealing an important deregulation of genes involved in DNA damage response. The potent in vitro and in vivo antimyeloma activity of Zalypsis uncovers the high sensitivity of tumor plasma cells to DSBs and strongly supports the use of this compound in MM patients.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Breaks, Double-Stranded/drug effects , Multiple Myeloma/drug therapy , Tetrahydroisoquinolines/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/therapeutic use , Cell Death , Checkpoint Kinase 2 , Dose-Response Relationship, Drug , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Mutation , Phosphorylation/drug effects , Plasma Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tetrahydroisoquinolines/therapeutic use , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
12.
Haematologica ; 96(5): 687-95, 2011 May.
Article in English | MEDLINE | ID: mdl-21330323

ABSTRACT

BACKGROUND: Although the majority of patients with acute myeloid leukemia initially respond to conventional chemotherapy, relapse is still the leading cause of death, probably because of the presence of leukemic stem cells that are insensitive to current therapies. We investigated the antileukemic activity and mechanism of action of zalypsis, a novel alkaloid of marine origin. DESIGN AND METHODS: The activity of zalypsis was studied in four acute myeloid leukemia cell lines and in freshly isolated blasts taken from patients with acute myeloid leukemia before they started therapy. Zalypsis-induced apoptosis of both malignant and normal cells was measured using flow cytometry techniques. Gene expression profiling and western blot studies were performed to assess the mechanism of action of the alkaloid. RESULTS: Zalypsis showed a very potent antileukemic activity in all the cell lines tested and potentiated the effect of conventional antileukemic drugs such as cytarabine, fludarabine and daunorubicin. Interestingly, zalypsis showed remarkable ex vivo potency, including activity against the most immature blast cells (CD34(+) CD38(-) Lin(-)) which include leukemic stem cells. Zalypsis-induced apoptosis was the result of an important deregulation of genes involved in the recognition of double-strand DNA breaks, such as Fanconi anemia genes and BRCA1, but also genes implicated in the repair of double-strand DNA breaks, such as RAD51 and RAD54. These gene findings were confirmed by an increase in several proteins involved in the pathway (pCHK1, pCHK2 and pH2AX). CONCLUSIONS: The potent and selective antileukemic effect of zalypsis on DNA damage response mechanisms observed in acute myeloid leukemia cell lines and in patients' samples provides the rationale for the investigation of this compound in clinical trials.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Damage , Stem Cells/drug effects , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
13.
Cancers (Basel) ; 13(11)2021 May 22.
Article in English | MEDLINE | ID: mdl-34067236

ABSTRACT

Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.

14.
Cancers (Basel) ; 13(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802806

ABSTRACT

Immunosuppression is a common feature of multiple myeloma (MM) patients and has been associated with disease evolution from its precursor stages. MM cells promote immunosuppressive effects due to both the secretion of soluble factors, which inhibit the function of immune effector cells, and the recruitment of immunosuppressive populations. Alterations in the expression of surface molecules are also responsible for immunosuppression. In this scenario, immunotherapy, as is the case of immunotherapeutic monoclonal antibodies (mAbs), aims to boost the immune system against tumor cells. In fact, mAbs exert part of their cytotoxic effects through different cellular and soluble immune components and, therefore, patients' immunosuppressive status could reduce their efficacy. Here, we will expose the alterations observed in symptomatic MM, as compared to its precursor stages and healthy subjects, in the main immune populations, especially the inhibition of effector cells and the activation of immunosuppressive populations. Additionally, we will revise the mechanisms responsible for all these alterations, including the interplay between MM cells and immune cells and the interactions among immune cells themselves. We will also summarize the main mechanisms of action of the four mAbs approved so far for the treatment of MM. Finally, we will discuss the potential immune-stimulating effects of non-immunotherapeutic drugs, which could enhance the efficacy of immunotherapeutic treatments.

15.
Cells ; 10(3)2021 03 04.
Article in English | MEDLINE | ID: mdl-33806619

ABSTRACT

BH3-mimetics targeting anti-apoptotic proteins such as MCL-1 (S63845) or BCL-2 (venetoclax) are currently being evaluated as effective therapies for the treatment of multiple myeloma (MM). Interleukin 6, produced by mesenchymal stromal cells (MSCs), has been shown to modify the expression of anti-apoptotic proteins and their interaction with the pro-apoptotic BIM protein in MM cells. In this study, we assess the efficacy of S63845 and venetoclax in MM cells in direct co-culture with MSCs derived from MM patients (pMSCs) to identify additional mechanisms involved in the stroma-induced resistance to these agents. MicroRNAs miR-193b-3p and miR-21-5p emerged among the top deregulated miRNAs in myeloma cells when directly co-cultured with pMSCs, and we show their contribution to changes in MCL-1 and BCL-2 protein expression and in the activity of S63845 and venetoclax. Additionally, direct contact with pMSCs under S63845 and/or venetoclax treatment modifies myeloma cell dependence on different BCL-2 family anti-apoptotic proteins in relation to BIM, making myeloma cells more dependent on the non-targeted anti-apoptotic protein or BCL-XL. Finally, we show a potent effect of the combination of S63845 and venetoclax even in the presence of pMSCs, which supports this combinatorial approach for the treatment of MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Thiophenes/therapeutic use , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Multiple Myeloma/pathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology
16.
Nat Commun ; 12(1): 421, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462210

ABSTRACT

Multiple myeloma (MM) progression and myeloma-associated bone disease (MBD) are highly dependent on bone marrow mesenchymal stromal cells (MSCs). MM-MSCs exhibit abnormal transcriptomes, suggesting the involvement of epigenetic mechanisms governing their tumor-promoting functions and prolonged osteoblast suppression. Here, we identify widespread DNA methylation alterations of bone marrow-isolated MSCs from distinct MM stages, particularly in Homeobox genes involved in osteogenic differentiation that associate with their aberrant expression. Moreover, these DNA methylation changes are recapitulated in vitro by exposing MSCs from healthy individuals to MM cells. Pharmacological targeting of DNMTs and G9a with dual inhibitor CM-272 reverts the expression of hypermethylated osteogenic regulators and promotes osteoblast differentiation of myeloma MSCs. Most importantly, CM-272 treatment prevents tumor-associated bone loss and reduces tumor burden in a murine myeloma model. Our results demonstrate that epigenetic aberrancies mediate the impairment of bone formation in MM, and its targeting by CM-272 is able to reverse MBD.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Diseases/drug therapy , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Mesenchymal Stem Cells/drug effects , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Bone Diseases/diagnosis , Bone Diseases/genetics , Bone Diseases/pathology , Bone Marrow/pathology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Female , Femur/diagnostic imaging , Femur/pathology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Xenograft Model Antitumor Assays
17.
Haematologica ; 95(5): 794-803, 2010 May.
Article in English | MEDLINE | ID: mdl-19951978

ABSTRACT

BACKGROUND: Combinations of drug treatments based on bortezomib or lenalidomide plus steroids have resulted in very high response rates in multiple myeloma. However, most patients still relapse, indicating the need for novel combination partners to increase duration of response or to treat relapsed disease. We explored the antimyeloma activity of triple combinations of these well-established schemes with panobinostat, a novel deacetylase inhibitor with a multi-targeted profile. DESIGN AND METHODS: The activity of these combinations was explored in vitro in cell lines by using MTT and annex-in V, ex vivo by flow cytometry, and in vivo using two different murine models of human myeloma: one bearing a subcutaneous plasmacytoma and another with a disseminated myeloma. Moreover, gene expression profiling and immunohistochemical studies were performed. RESULTS: The addition of panobinostat (LBH589) to dexamethasone and either bortezomib or lenalidomide resulted in clear potentiation in multiple myeloma cell lines, freshly isolated plasma cells, and murine models of multiple myeloma. The quantification of the potency of these combinations by using the Chou-Talalay method showed synergistic combination indices for all of them. This effect derived from the deregulation of a cluster of genes that was completely different from the sum of genes affected by the single agents (895 and 1323 genes exclusively deregulated by panobinostat and dexamethasone plus bortezomib or lenalidomide, respectively). Functional experiments, such as annexin V staining, cell cycle analysis, and immunohistochemical studies also supported this potentiation. Anti-myeloma efficacy was confirmed in an extramedullary plasmacytoma model and a disseminated luciferized model, in which panobinostat also provided a marked benefit in bone disease. CONCLUSIONS: The potent activity, together with the exclusive mechanistic profile, provides the rationale for the clinical evaluation of these drug combinations in multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Animals , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Cells, Cultured , Dexamethasone/administration & dosage , Disease Models, Animal , Humans , Hydroxamic Acids/administration & dosage , Indoles , Lenalidomide , Mice , Mice, SCID , Panobinostat , Pyrazines/administration & dosage , Random Allocation , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Xenograft Model Antitumor Assays
18.
Expert Opin Investig Drugs ; 29(1): 5-14, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31815551

ABSTRACT

Introduction: Kinesin spindle protein (KSP) is indispensable for the proper separation of spindle poles during mitosis. Importantly, this protein is expressed only in cells undergoing cell division and hence represents an appealing target for the treatment of cancer. Many KSP inhibitors have demonstrated a strong antitumoral effect in vitro, however, they have exhibited only limited activity in clinical trials. By contrast, the KSP inhibitor filanesib has demonstrated clinical efficacy in patients with multiple myeloma (MM).Areas covered: This article provides a comprehensive overview about the progress to date in the preclinical and clinical development of filanesib for the treatment of cancer, and particularly, MM.Expert opinion: Responses observed with filanesib alone or in combination with dexamethasone were encouraging in MM. However, the subsequent appearance of highly effective novel agents such as monoclonal antibodies, has hindered the development of agents such as filanesib that exhibit a more limited activity. Nevertheless, filanesib has shown interesting results for some patients when combined with carfilzomib and pomalidomide. Most importantly, the availability of a biomarker of response such as alpha 1-acid glycoprotein (AAG), could be key to the identification of patients that could benefit most from these combinations.


Subject(s)
Antimitotic Agents/administration & dosage , Multiple Myeloma/drug therapy , Thiadiazoles/administration & dosage , Animals , Antimitotic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor/metabolism , Drug Development , Humans , Kinesins/antagonists & inhibitors , Multiple Myeloma/pathology , Thiadiazoles/pharmacology
19.
Cancers (Basel) ; 12(10)2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32987735

ABSTRACT

BACKGROUND: Proviral Insertion site for Moloney murine leukemia virus (PIM) kinases are overexpressed in hematologic malignancies, including multiple myeloma. Previous preclinical data from our group demonstrated the anti-myeloma effect of the pan-PIM kinase inhibitor PIM447. METHODS: Based on those data, we evaluate here, by in vitro and in vivo studies, the activity of the triple combination of PIM447 + pomalidomide + dexamethasone (PIM-Pd) in multiple myeloma. RESULTS: Our results show that the PIM-Pd combination exerts a potent anti-myeloma effect in vitro and in vivo, where it markedly delays tumor growth and prolongs survival of treated mice. Mechanism of action studies performed in vitro and on mice tumor samples suggest that the combination PIM-Pd inhibits protein translation processes through the convergent inhibition of c-Myc and mTORC1, which subsequently disrupts the function of eIF4E. Interestingly the MM pro-survival factor IRF4 is also downregulated after PIM-Pd treatment. As a whole, all these molecular changes would promote cell cycle arrest and deregulation of metabolic pathways, including glycolysis and lipid biosynthesis, leading to inhibition of myeloma cell proliferation. CONCLUSIONS: Altogether, our data support the clinical evaluation of the triple combination PIM-Pd for the treatment of patients with multiple myeloma.

20.
Medchemcomm ; 10(9): 1589-1598, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31673316

ABSTRACT

The use of multitarget drugs has evolved as an alternative to "magic bullets" for the treatment of complex diseases such as cancer, in order to affect simultaneously several targets relevant to the disease. We have designed and synthesized a series of dual agents as both Eg5 inhibitors and calcium channel blockers, bearing a 4-aryldihydropyrimidine core. Compound 2 (aryl: 3-nitrophenyl) was selected as potential dual agent due to displaying both activities: it is a vasorelaxant agent (>90% relaxation at 10-5 M in KCl-precontracted aorta rings), it decreases the response to calcium and it is cytotoxic to MCF-7 (breast), HCT-116 (colon) and A-549 (lung) cancer cell lines. The dual mechanism of action was confirmed by blocking (-)-BAY K8644-induced vascular contraction and production of monopolar spindles, typical of Eg5 inhibition. Docking suggests that both (R) and (S)-enantiomers could bind Eg5.

SELECTION OF CITATIONS
SEARCH DETAIL