Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
EJHaem ; 3(4): 1377-1380, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36467828

ABSTRACT

Due to differences in the protein folding mechanisms, it is exceedingly rare for amyloid light chain (AL) amyloidosis and monoclonal gammopathy of renal significance (MGRS) to coexist. We herein report the first case of concurrent AL amyloidosis and a subclass of MGRS, light chain proximal tubulopathy (LCPT). The 53-year-old female was diagnosed with smoldering myeloma immunoglobulin G kappa and AL amyloidosis with deposits in fat and gastrointestinal tissue. The kidney biopsy did not show amyloid deposits but electron microscopy revealed the presence of LCPT with crystal formation in proximal tubular epithelial cells. This case illustrates the complex pathophysiology of protein deposition in monoclonal gammopathies.

2.
Cancers (Basel) ; 13(18)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34572934

ABSTRACT

Salvage high-dose chemotherapy and autologous stem cell transplantation (HDCT/ASCT) is a treatment option for relapsed and/or refractory multiple myeloma (RRMM). No data are available on salvage HDCT/ASCT following re-induction treatment with state-of-the-art triplet regimens. We retrospectively report on 44 patients receiving salvage HDCT/ASCT following re-induction with carfilzomib/lenalidomide/dexamethasone (KRd). All patients received frontline HDCT/ASCT with median time to progression (TTP1) of 2.9 (1.2-13.5) years, enabling paired comparison of frontline and salvage HDCT/ASCT. After re-induction and before salvage transplant, 25/44 patients (57%) attained ≥ very good partial response (VGPR), which increased to 34/44 (77%) at best response after salvage HDCT/ASCT. Median progression-free survival (PFS) was 23.3 months from salvage HDCT/ASCT. Patients with ≥ VGPR at the time of salvage HDCT/ASCT and those receiving maintenance treatment post salvage HDCT/ASCT had significantly superior PFS (hazard ratio (HR) 0.19, p = 0.001 and HR 0.20, p = 0.009). In patients achieving at least an equal depth of response before salvage HDCT/ASCT as before frontline HDCT/ASCT, PFS after salvage HDCT/ASCT was comparable to the frontline situation (p = 0.3). This is the first report of state-of-the-art triplet re-induction and salvage HDCT/ASCT for RRMM after frontline transplantation. Deep remissions achieved with KRd translate into prolonged PFS following salvage HDCT/ASCT and are enhanced by maintenance treatment.

3.
Ann Hematol ; 97(5): 839-849, 2018 May.
Article in English | MEDLINE | ID: mdl-29359239

ABSTRACT

A promising approach to the treatment of multiple myeloma (MM) involves agents that target not only the myeloma cells directly, but also the tumor microenvironment which promotes tumor cell growth, angiogenesis, and MM bone disease. Here we investigate the orally available multikinase inhibitor, regorafenib (BAY 73-4506), for its therapeutic efficacy in MM. Regorafenib is a potent inhibitor of angiogenic (VEGFR 1-3, PDGFR-b) as well as oncogenic (c-KIT, RET, FGFR, Raf) kinases. We show that regorafenib induces apoptosis in all MM cell lines at below clinically achievable concentrations. Regorafenib overcomes the growth advantage conferred by a stroma cell MM and an endothelial cell MM, co-culture systems, and abrogates growth factor-stimulated MEK, ERK, and AKT phosphorylation at nanomolar to micromolar concentrations. Moreover, it inhibits endothelial cell growth and tubule formation, abrogates both VEGF secretion and VEGF-induced MM cell migration, inhibits osteoclastogenesis, and shows synergistic cytotoxicity with dexamethasone, the immunomodulatory drug pomalidomide, and the p110δ inhibitor idelalisib. Most importantly, regorafenib significantly delays tumor growth in a xenograft mouse model of human MM. These results provide the rationale for further clinical evaluation of regorafenib, alone and in combination, in the treatment of MM.


Subject(s)
Multiple Myeloma/drug therapy , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Administration, Oral , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Nude , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phenylurea Compounds/metabolism , Protein Kinase Inhibitors/metabolism , Pyridines/metabolism , Xenograft Model Antitumor Assays/methods
4.
Hematol Oncol ; 34(4): 200-207, 2016 Dec.
Article in English | MEDLINE | ID: mdl-25784529

ABSTRACT

Therapeutic options in heavily pretreated relapsed/refractory multiple myeloma patients are often very limited because of impaired bone marrow function. Bendamustine is effective in multiple myeloma and has a favourable toxicity profile. We hypothesized that dose-intensified bendamustine (180 mg/m2 , day 1 and 2) followed by autologous blood stem cell support (ASCS) would improve bone marrow function with low post-transplant toxicity in patients with severely impaired haematopoiesis. We analyzed 28 consecutive myeloma patients, with a median of three prior lines of therapy (range 2-7), who had relapsed from the last treatment with very limited bone marrow function and were therefore ineligible for conventional chemotherapy, novel agents or trial enrolment. Dose-intensified bendamustine with ASCS improved haematopoiesis as reflected by increased platelet counts (median 40/nl vs 94/nl, p = 0.0004) and white blood cell counts (3.0/nl vs 4.8/nl, p = 0.02) at day +100. The median time until engraftment of platelets (>50/nl) was 11 days (0-24 days) and of white cell counts (>1.0/nl) 0 days (0-24 days). At least, a minimal response was achieved in 36% of patients. The disease stabilization rate was 50% while the median progression-free survival rate was limited to 2.14 months. Most importantly, patients were once again eligible for alternative treatments including enrolment into clinical trials. We conclude that dose-intensified bendamustine followed by ASCS is safe and feasible for multiple myeloma patients with very limited bone marrow reserve. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Bendamustine Hydrochloride/administration & dosage , Bone Marrow/physiopathology , Hematopoiesis , Multiple Myeloma , Peripheral Blood Stem Cell Transplantation , Transplantation Conditioning , Adult , Aged , Autografts , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/physiopathology , Multiple Myeloma/therapy , Survival Rate
5.
Oncotarget ; 5(21): 10237-50, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25296978

ABSTRACT

The inhibitor-of-apoptosis family member survivin has been reported to inhibit apoptosis and regulate mitosis and cytokinesis. In multiple myeloma, survivin has been described to be involved in downstream sequelae of various therapeutic agents. We assessed 1093 samples from previously untreated patients, including two independent cohorts of 392 and 701 patients, respectively. Survivin expression was associated with cell proliferation, adverse prognostic markers, and inferior event-free and overall survival, supporting the evaluation of survivin as a therapeutic target in myeloma. The small molecule suppressant of survivin--YM155--is in clinical development for the treatment of solid tumors. YM155 potently inhibited proliferation and induced apoptosis in primary myeloma cells and cell lines. Gene expression and protein profiling revealed the critical roles of IL6/STAT3-signaling and the unfolded protein response in the efficacy of YM155. Both pathways converged to down regulate anti-apoptotic Mcl-1 in myeloma cells. Conversely, growth inhibition and apoptotic cell death by YM155 was rescued by ectopic expression of Mcl-1 but not survivin, identifying Mcl-1 as the pivotal downstream target of YM155 in multiple myeloma. Mcl-1 expression was likewise associated with adverse prognostic markers, and inferior survival. Our results strongly support the clinical evaluation of YM155 in patients with multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Naphthoquinones/pharmacology , Unfolded Protein Response , Apoptosis/drug effects , Apoptosis/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cohort Studies , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Interleukin-6/metabolism , Multiple Myeloma/mortality , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Prognosis , STAT3 Transcription Factor/metabolism , Survival Analysis , Survivin , Transcriptome , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
6.
Cancer Res ; 72(20): 5374-85, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22942257

ABSTRACT

In contrast to normal cells, malignant cells are frequently aneuploid and contain multiple centrosomes. To allow for bipolar mitotic division, supernumerary centrosomes are clustered into two functional spindle poles in many cancer cells. Recently, we have shown that griseofulvin forces tumor cells with supernumerary centrosomes to undergo multipolar mitoses resulting in apoptotic cell death. Here, we describe the characterization of the novel small molecule GF-15, a derivative of griseofulvin, as a potent inhibitor of centrosomal clustering in malignant cells. At concentrations where GF-15 had no significant impact on tubulin polymerization, spindle tension was markedly reduced in mitotic cells upon exposure to GF-15. Moreover, isogenic cells with conditional centrosome amplification were more sensitive to GF-15 than parental controls. In a wide array of tumor cell lines, mean inhibitory concentrations (IC(50)) for proliferation and survival were in the range of 1 to 5 µmol/L and were associated with apoptotic cell death. Importantly, treatment of mouse xenograft models of human colon cancer and multiple myeloma resulted in tumor growth inhibition and significantly prolonged survival. These results show the in vitro and in vivo antitumor efficacy of a prototype small molecule inhibitor of centrosomal clustering and strongly support the further evaluation of this new class of molecules.


Subject(s)
Centrosome/drug effects , Griseofulvin/analogs & derivatives , Cell Line, Tumor , Griseofulvin/pharmacology , Humans , In Vitro Techniques , Tissue Distribution
7.
Lancet ; 374(9686): 324-39, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19541364

ABSTRACT

Multiple myeloma is characterised by clonal proliferation of malignant plasma cells, and mounting evidence indicates that the bone marrow microenvironment of tumour cells has a pivotal role in myeloma pathogenesis. This knowledge has already expanded treatment options for patients with multiple myeloma. Prototypic drugs thalidomide, bortezomib, and lenalidomide have each been approved for the treatment of this disease by targeting both multiple myeloma cells and the bone marrow microenvironment. Although benefit was first shown in relapsed and refractory disease, improved overall response, duration of response, and progression-free and overall survival can be achieved when these drugs are part of first-line regimens. This treatment framework promises to improve outcome not only for patients with multiple myeloma, but also with other haematological malignancies and solid tumours.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Multiple Myeloma/drug therapy , Pyrazines/therapeutic use , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Bortezomib , Clinical Trials as Topic , Combined Modality Therapy , Genetic Predisposition to Disease/genetics , Humans , Lenalidomide , Melphalan/therapeutic use , Multiple Myeloma/diagnosis , Multiple Myeloma/epidemiology , Multiple Myeloma/etiology , Neoplasm Staging , Protease Inhibitors/therapeutic use , Remission Induction , Stem Cell Transplantation , Survival Rate , Treatment Outcome
8.
Blood ; 113(7): 1513-21, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19018094

ABSTRACT

Targeting protein kinase C (PKC) isoforms by the small molecule inhibitor enzastaurin has shown promising preclinical activity in a wide range of tumor cells. We further delineated its mechanism of action in multiple myeloma (MM) cells and found a novel role of beta-catenin in regulating growth and survival of tumor cells. Specifically, inhibition of PKC leads to rapid accumulation of beta-catenin by preventing the phosphorylation required for its proteasomal degradation. Microarray analysis and small-interfering RNA (siRNA)-mediated gene silencing in MM cells revealed that accumulated beta-catenin activates early endoplasmic reticulum stress signaling via eIF2alpha, C/EBP-homologous protein (CHOP), and p21, leading to immediate growth inhibition. Furthermore, accumulated beta-catenin contributes to enzastaurin-induced cell death. Sequential knockdown of beta-catenin, c-Jun, and p73, as well as overexpression of beta-catenin or p73 confirmed that accumulated beta-catenin triggers c-Jun-dependent induction of p73, thereby conferring MM cell apoptosis. Our data reveal a novel role of beta-catenin in endoplasmic reticulum (ER) stress-mediated growth inhibition and a new proapoptotic mechanism triggered by beta-catenin on inhibition of PKC isoforms. Moreover, we identify p73 as a potential novel therapeutic target in MM. Based on these and previous data, enzastaurin is currently under clinical investigation in a variety of hematologic malignancies, including MM.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Multiple Myeloma/metabolism , Protein Kinase C/metabolism , beta Catenin/metabolism , Apoptosis/drug effects , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/metabolism , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Nuclear Proteins/genetics , Protein Kinase C/antagonists & inhibitors , RNA, Small Interfering , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Physiological/drug effects , Stress, Physiological/physiology , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
9.
Expert Opin Drug Metab Toxicol ; 4(7): 973-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18624684

ABSTRACT

BACKGROUND: After the tragic events in the early 1960s, thalidomide has re-emerged as therapeutic for multiple myeloma (MM). It was first approved for the treatment of erythema nodosum leprosum, and is now under evaluation for hematologic and non-hematologic disorders. Its complex mechanism of action is not fully understood; however extensive preclinical studies in MM have revealed its antiangiogenic and immunomodulatory properties. OBJECTIVE: In this review, we focus on the importance and toxicity of thalidomide in today's clinical use. METHODS: Key preclinical and clinical trials available as well as data on the pharmacokinetics and pharmacodynamics of thalidomide in humans are summarized. CONCLUSIONS: Thalidomide is widely used as first-line treatment and in relapsed/refractory MM. The most common side effects are fatigue, constipation and peripheral neuropathy, and careful monitoring is required to avoid fetal exposure.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Myeloma/drug therapy , Thalidomide/therapeutic use , Animals , Clinical Trials as Topic , Drug Interactions , Female , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , Male , Pregnancy , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Thalidomide/pharmacology
10.
Br J Haematol ; 141(5): 659-71, 2008 May.
Article in English | MEDLINE | ID: mdl-18410446

ABSTRACT

This study investigated the biological significance of the inhibition of fatty acid synthase (FAS) in multiple myeloma (MM) using the small molecule inhibitor Cerulenin. Cerulenin triggered growth inhibition in both MM cell lines and MM patient cells, and overcame the survival and growth advantages conferred by interleukin-6, insulin-like growth factor-1, and bone marrow stromal cells. It induced apoptosis in MM cell lines with only modest activation of caspase -8, -9, -3 and PARP; moreover, the pan-caspase inhibitor Z-VAD-FMK did not inhibit Cerulenin-induced apoptosis and cell death. In addition, treatment of MM cells with Cerulenin primarily up-regulated apoptosis-inducing factor/endonuclease G, mediators of caspase-independent apoptosis. Importantly, Cerulenin induced endoplasmic reticulum stress response via up-regulation of the Grp78/IRE1alpha/JNK pathway. Although the C-Jun-NH(2)-terminal kinase (JNK) inhibitor SP600215 blocked Cerulenin-induced cytotoxicity, it did not inhibit apoptosis and caspase cleavage. Furthermore, Cerulenin showed synergistic cytotoxic effects with various agents including Bortezomib, Melphalan and Doxorubicin. Our results therefore indicate that inhibition of FAS by Cerulenin primarily triggered caspase-independent apoptosis and JNK-dependent cytotoxicity in MM cells. This report demonstrated that inhibition of FAS has anti-tumour activity against MM cells, suggesting that it represents a novel therapeutic target in MM.


Subject(s)
Cerulenin/therapeutic use , Fatty Acid Synthases/antagonists & inhibitors , Multiple Myeloma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cell Line, Tumor , Cerulenin/pharmacology , Drug Delivery Systems , Drug Evaluation , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4/metabolism , Multiple Myeloma/enzymology , Signal Transduction , Tumor Cells, Cultured , fas Receptor/metabolism
11.
Haematologica ; 93(1): 124-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18166796

ABSTRACT

In the prospective phase 3 HOVON-50/GMMG-HD3 trial, patients randomized to TAD (thalidomide, doxorubicin, dexamethasone) had a significantly higher response rate (at least PR) after induction compared with patients randomized to VAD (vincristine, adriamycin, dexamethasone, 72% vs. 54%, p<0.001). Complete remission (CR) and very good partial remission (VGPR) were also higher after TAD. After High Dose melphalan 200mg/m(2) response was comparable in both arms, 76% and 79% respectively. However, CR plus VGPR were significantly higher in the patients randomized to TAD (49% vs. 32%, p<0.001). CTC grade 3-4 adverse events were similar in both arms.


Subject(s)
Medical Oncology/methods , Multiple Myeloma/drug therapy , Thalidomide/therapeutic use , Adult , Aged , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Dexamethasone/therapeutic use , Female , Humans , Male , Middle Aged , Remission Induction , Thioguanine/therapeutic use , Treatment Outcome , Vincristine/therapeutic use
12.
Br J Haematol ; 139(1): 55-63, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17854307

ABSTRACT

Osteolytic bone disease in multiple myeloma (MM) is associated with upregulation of osteoclast (OCL) activity and constitutive inhibition of osteoblast function. The extracellular signal-regulated kinase 1/2 (ERK1/2) pathway mediates OCL differentiation and maturation. We hypothesized that inhibition of ERK1/2 could prevent OCL differentiation and downregulate OCL function. It was found that AZD6244, a mitogen-activated or extracellular signal-regulated protein kinase (MEK) inhibitor, blocked OCL differentiation and formation in a dose-dependent manner, evidenced by decreased alphaVbeta3-integrin expression and tartrate-resistant acid phosphatase positive (TRAP+) cells. Functional dentine disc cultures showed inhibition of OCL-induced bone resorption by AZD6244. Major MM growth and survival factors produced by OCLs including B-cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL), as well as macrophage inflammatory protein (MIP-1alpha), which mediates OCL differentiation and MM, were also significantly inhibited by AZD6244. In addition to ERK inhibition, NFATc1 (nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1) and c-fos were both downregulated, suggesting that AZD6244 targets a later stage of OCL differentiation. These results indicate that AZD6244 inhibits OCL differentiation, formation and bone resorption, thereby abrogating paracrine MM cell survival in the bone marrow microenvironment. The present study therefore provides a preclinical rationale for the evaluation of AZD6244 as a potential new therapy for patients with MM.


Subject(s)
Benzimidazoles/therapeutic use , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Multiple Myeloma/drug therapy , Osteoclasts/pathology , Acid Phosphatase/metabolism , Biomarkers/analysis , Blotting, Western/methods , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Chemokine CCL3 , Chemokine CCL4 , Cytokines/metabolism , Depression, Chemical , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression/drug effects , Humans , Integrin alphaVbeta3/metabolism , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Inflammatory Proteins/metabolism , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/metabolism , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase , Transcription Factors/metabolism , Tumor Cells, Cultured
13.
Blood ; 110(10): 3744-52, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17715391

ABSTRACT

The interaction between osteoclasts (OCs) and multiple myeloma (MM) cells plays a key role in the pathogenesis of MM-related osteolytic bone disease (OBD). MM cells promote OC formation and, in turn, OCs enhance MM cell proliferation. Chemokines are mediators of MM effects on bone and vice versa; in particular, CCL3 enhances OC formation and promotes MM cell migration and survival. Here, we characterize the effects of MLN3897, a novel specific antagonist of the chemokine receptor CCR1, on both OC formation and OC-MM cell interactions. MLN3897 demonstrates significant impairment of OC formation (by 40%) and function (by 70%), associated with decreased precursor cell multinucleation and down-regulation of c-fos signaling. OCs secrete high levels of CCL3, which triggers MM cell migration; conversely, MLN3897 abrogates its effects by inhibiting Akt signaling. Moreover, MM cell-to-OC adhesion was abrogated by MLN3897, thereby inhibiting MM cell survival and proliferation. Our results therefore show novel biologic sequelae of CCL3 and its inhibition in both osteoclastogenesis and MM cell growth, providing the preclinical rationale for clinical trials of MLN3897 to treat OBD in MM.


Subject(s)
Cell Communication/drug effects , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/physiology , Receptors, CCR1/antagonists & inhibitors , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Fusion , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL3/metabolism , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Genes, fos , Humans , Multiple Myeloma/metabolism , Osteoclasts/metabolism
14.
Blood ; 109(4): 1669-77, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17023575

ABSTRACT

In multiple myeloma (MM) protein kinase C (PKC) signaling pathways have been implicated in cell proliferation, survival, and migration. Here we investigated the novel, orally available PKC-inhibitor enzastaurin for its anti-MM activity. Enzastaurin specifically inhibits phorbol ester-induced activation of PKC isoforms, as well as phosphorylation of downstream signaling molecules MARCKS and PKCmu. Importantly, it also inhibits PKC activation triggered by growth factors and cytokines secreted by bone marrow stromal cells (BMSCs), costimulation with fibronectin, vascular endothelial growth factor (VEGF), or interleukin-6 (IL-6), as well as MM patient serum. Consequently, enzastaurin inhibits proliferation, survival, and migration of MM cell lines and MM cells isolated from multidrug-resistant patients and overcomes MM-cell growth triggered by binding to BMSCs and endothelial cells. Importantly, strong synergistic cytotoxicity is observed when enzastaurin is combined with bortezomib and moderate synergistic or additive effects when combined with melphalan or lenalidomide. Finally, tumor growth, survival, and angiogenesis are abrogated by enzastaurin in an in vivo xenograft model of human MM. Our results therefore demonstrate in vitro and in vivo efficacy of the orally available PKC inhibitor enzastaurin in MM and strongly support its clinical evaluation, alone or in combination therapies, to improve outcome in patients with MM.


Subject(s)
Indoles/pharmacology , Multiple Myeloma/drug therapy , Phorbol Esters/pharmacology , Protein Kinase C/antagonists & inhibitors , Administration, Oral , Animals , Cell Communication , Coculture Techniques , Cytokines/metabolism , Cytokines/pharmacology , Drug Delivery Systems , Enzyme Activation/drug effects , Humans , Mice , Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Phosphorylation , Protein Isoforms , Stromal Cells/cytology , Transplantation, Heterologous , Tumor Burden/drug effects , Tumor Cells, Cultured
15.
Eur J Haematol ; 78(1): 21-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17042762

ABSTRACT

BACKGROUND: We analyzed the hematopoietic reconstitution and outcome of 508 patients with multiple myeloma (MM) with respect to the number of CD34+ cells reinfused at our center. PATIENTS AND METHODS: Each cohort of 390 patients (unselected CD34+ cell transplant) and 118 patients (CD34+ selected transplant) was divided into four subgroups. Among the 390 transplantations, 86 patients received a high dose (HD-) of > or =6.50 x 10(6) unselected CD34+ cells/kg, 116 patients a low dose (LD-) of <3.00 x 10(6) CD34+ cells/kg. Among the patients treated with CD34+ selected PBSC, 34 received > or =6.50 x 10(6) CD34+ cells/kg (HD+) and 16 <3.00 x 10(6) CD34+ cells/kg (LD+). RESULTS: HD- patients experienced a reduced median time to leukocyte (13 d vs. 14 d) (P < 0.001) and platelet reconstitution >20 x 10(9)/L (10 d vs. 12 d) (P < 0.001). Similarly, HD+ showed a reduced median time to leukocyte (12 d vs. 15 d) (P < 0.001) and platelet recovery >20 x 10(9)/L (10 d vs. 11 d) (P = 0.058). CD34+ cell-dose was significant for long-term platelet recovery at day 360 (unselected transplant P = 0.015, selected transplant P = 0.023). Number of transplanted CD34+ cells had no significant impact on transplant related mortality, overall survival or CR/PR rates within 100 d. In terms of supportive care the differences of high-/low-dose grafts were minimal. CONCLUSIONS: These results confirm that high doses of CD34+ PBSC shorten hematopoietic reconstitution and reduce hospitalization. Nevertheless secure engraftment results from transplantation of 2.00-3.00 x 10(6) CD34+ cells/kg. As 60% of our pretreated patients are able to collect > or =5.00 x 10(6) CD34+ cells/kg within a single leukapheresis, division into two or more freezing bags allows safe tandem transplantation in the majority of MM patients.


Subject(s)
Antigens, CD34/biosynthesis , Hematopoiesis , Hematopoietic Stem Cells/immunology , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation , Adult , Aged , Cohort Studies , Dose-Response Relationship, Immunologic , Female , Graft Survival , Hematopoietic Stem Cells/cytology , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Platelet Count , Prognosis , Survival Rate , Transplantation Conditioning , Transplantation, Autologous , Treatment Outcome
16.
Haematologica ; 91(11): 1555-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17082013

ABSTRACT

Patients with multiple myeloma and end-stage renal failure on dialysis are frequently not considered eligible for high-dose therapy (HDT) due to higher transplant-related mortality (TRM). Our aim was to evaluate the toxicity and survival of dialysis-dependent patients after HDT with melphalan (100 mg/m(2)) compared to those of patients without renal insufficiency (melphalan 200 mg/m(2)) in a matched pairs analysis of 34 patients. No significant differences were observed between hematologic toxicity, TRM or disease response. Dialysis patients showed comparable event-free and overall survival. They required significantly extended intravenous antibiotic treatment and longer hospitalization. Thus, melphalan 100 mg/m2 is less toxic, yet equally efficient and improves the prognosis of this group of patients.


Subject(s)
Kidney Failure, Chronic/therapy , Multiple Myeloma/therapy , Renal Dialysis , Stem Cell Transplantation , Adult , Aged , Female , Humans , Kidney Failure, Chronic/mortality , Male , Middle Aged , Multiple Myeloma/mortality , Renal Dialysis/mortality , Stem Cell Transplantation/mortality , Transplantation, Autologous/mortality , Treatment Outcome
17.
Cancer Res ; 66(13): 6675-82, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16818641

ABSTRACT

Recent studies have underscored the role of B-cell-activating factor (BAFF), a member of the tumor necrosis factor superfamily, in promoting the survival of malignant B cells, including human multiple myeloma. We here characterized the functional significance of BAFF in the interaction between multiple myeloma and bone marrow stromal cells (BMSC) and further defined the molecular mechanisms regulating these processes. BAFF is detected on BMSCs derived from multiple myeloma patients as evidenced by flow cytometry. BAFF secretion is 3- to 10-fold higher in BMSCs than in multiple myeloma cells, and tumor cell adhesion to BMSCs augments BAFF secretion by 2- to 5-fold, confirmed by both ELISA and immunoblotting. Adhesion of MM1S and MCCAR multiple myeloma cell lines to KM104 BMSC line transfected with a luciferase reporter vector carrying the BAFF gene promoter (BAFF-LUC) significantly enhanced luciferase activity, suggesting that nuclear factor-kappaB (NF-kappaB) activation induced by multiple myeloma adhesion to BMSCs mediates BAFF up-regulation. Moreover, BAFF (0-100 ng/mL) increases adhesion of multiple myeloma lines to BMSCs in a dose-dependent manner; conversely, transmembrane activator and calcium modulator and cyclophylin ligand interactor-Ig or B-cell maturation antigen/Fc blocked BAFF stimulation. Using adenoviruses expressing dominant-negative and constitutively expressed AKT as well as NF-kappaB inhibitors, we further showed that BAFF-induced multiple myeloma cell adhesion is primarily mediated via activation of AKT and NF-kappaB signaling. Importantly, BAFF similarly increased adhesion of CD138-expressing patient multiple myeloma cells to BMSCs. These studies establish a role for BAFF in localization and survival of multiple myeloma cells in the bone marrow microenvironment and strongly support novel therapeutics, targeting the interaction between BAFF and its receptors in human multiple myeloma.


Subject(s)
Bone Marrow Neoplasms/pathology , Membrane Proteins/physiology , Multiple Myeloma/pathology , Tumor Necrosis Factor-alpha/physiology , B-Cell Activating Factor , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Neoplasms/metabolism , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-4/biosynthesis , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
18.
Cancer Res ; 65(24): 11712-20, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357183

ABSTRACT

SGN-40, a humanized immoglobulin G1 (IgG1) anti-CD40 monoclonal antibody, mediates cytotoxicity against human multiple myeloma (MM) cells via suppression of interleukin (IL)-6-induced proliferative and antiapoptotic effects as well as antibody-dependent cell-mediated cytotoxicity (ADCC). Here, we studied the clinical significance of an immunomodulatory drug lenalidomide on SGN-40-induced cytotoxicity against CD138(+)CD40(+) MM lines and patient MM cells. Pretreatment with lenalidomide sensitized MM cells to SGN-40-induced cell death. Combined lenalidomide and SGN-40 significantly induced MM apoptosis, evidenced by enhanced cleavage of caspase-3/8/poly(ADP-ribose)polymerase and increased sub-G(0) cells, compared with either single agent at the same doses. Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Importantly, pretreatment with lenalidomide or lenalidomide and SGN-40 markedly enhanced NK-cell-mediated lysis of autologous patient MM cells triggered by SGN-40. Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. In addition, lenalidomide induced the CD56(dim) NK subset, which are more potent mediators of ADCC against target MM cells than the CD56(bright) NK subset. Finally, pretreatment of both effector and target MM cells with lenalidomide markedly enhanced SGN-40-mediated ADCC against CD40-expressing MM cells. These studies, therefore, show that the addition of lenalidomide to SGN-40 enhances cytotoxicity against MM cells, providing the framework for combined lenalidomide and SGN-40 in a new treatment paradigm to both target MM cells directly and induce immune effectors against MM.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , CD40 Antigens/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Thalidomide/analogs & derivatives , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Humans , Immunization, Passive , Interleukin-6/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide , Multiple Myeloma/metabolism , Thalidomide/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...