Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Cancer Epidemiol Biomarkers Prev ; 32(6): 744-747, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37012203

ABSTRACT

INTRODUCTION: Molecularly targeted therapies such as tyrosine kinase inhibitors (TKI) are effective treatments for B-cell receptor (BCR)-ABL-bearing leukemias. We evaluated the impact of TKIs on historical chronic myeloid leukemia (CML) mortality trends compared with acute lymphoblastic leukemia (ALL) and chronic lymphoblastic leukemia (CLL). METHODS: Because mortality trends reflect combined effects of leukemia incidence and survival, we also evaluated the contribution of incidence and survival trends to mortality trends by subtypes. We used data from 13 U.S. (SEER) registries (1992-2017) among U.S. adults. We utilized histology codes to identify cases of CML, ALL, and CLL and death certificate data to calculate mortality. We used Joinpoint to characterize incidence (1992-2017) and mortality (1992-2018) trends by subtype and diagnosis year. RESULTS: For CML, mortality rates started declining in 1998 at an average rate of 12% annually. Imatinib was approved by the FDA for treating CML and ALL in 2001, leading to clear benefits for patients with CML. Five-year CML survival increased dramatically over time, especially between 1996 to 2011, 2.3% per year on average. ALL incidence increased 1.5% annually from 1992 to 2017. ALL mortality decreased 0.6% annually during 1992 to 2012 and then stopped declining. CLL incidence fluctuated during 1992 to 2017 while mortality decreased 1.1% annually during 1992 to 2011 and at a faster rate of 3.6% per year from 2011. Five-year survival increased 0.7% per year on average during 1992 to 2016. CONCLUSIONS: Survival benefit from TKIs and other novel therapies for treating leukemia subtypes has been demonstrated in clinical trials. IMPACT: Our study highlights the impact of molecularly targeted therapies at the population level.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Adult , Humans , Imatinib Mesylate/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Registries
3.
Clin Cancer Res ; 29(14): 2573-2579, 2023 07 14.
Article in English | MEDLINE | ID: mdl-36688922

ABSTRACT

Myelodysplastic syndromes (MDS) have historically been challenging diseases for drug development due to their biology, preclinical modeling, and the affected patient population. In April 2022, the FDA convened a panel of regulators and academic experts in MDS to discuss approaches to improve MDS drug development. The panel reviewed challenges in MDS clinical trial design and endpoints and outlined considerations for future trial design in MDS to facilitate drug development to meaningfully meet patient needs. Challenges for defining clinical benefit in patients with MDS include cumbersome response criteria, standardized transfusion thresholds, and application and validation of patient reported outcome instruments. Clinical trials should reflect the biology of disease evolution, the advanced age of patients with MDS, and how patients are treated in real-world settings to maximize the likelihood of identifying active drugs. In patients with lower-risk disease, response criteria for anemic patients should be based on baseline transfusion dependency, improvement in symptoms, and quality of life. For higher-risk patients with MDS, trials should include guidance to prevent dose reductions or delays that could limit efficacy, specify minimal durations of treatment (in the absence of toxicity or progression), and have endpoints focused on overall survival and durable responses. MDS trials should be designed from the outset to allow the practicable application of new therapies in this high-needs population, with drugs that can be administered and tolerated in community settings, and with endpoints that meaningfully improve patients' lives over existing therapies.


Subject(s)
Myelodysplastic Syndromes , Quality of Life , Humans , Myelodysplastic Syndromes/therapy
4.
Clin Cancer Res ; 29(12): 2179-2183, 2023 06 13.
Article in English | MEDLINE | ID: mdl-36547666

ABSTRACT

The FDA has an accelerated approval program for drugs that have been identified as promising treatments for serious conditions when the available data suggest that the benefits outweigh the foreseeable risks. All of the currently available treatment options for chronic myeloid leukemia (CML) initially went through the accelerated approval program. Here, a group of academic CML experts, patient panelists, and members from the FDA convened to discuss the utility of the accelerated approval program as it pertains to CML, and the utility of this program in future drug development in this disease. The results of that discussion are summarized here.


Subject(s)
Drug Approval , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , United States , Humans , United States Food and Drug Administration , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
5.
Clin Cancer Res ; 28(16): 3411-3416, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35435961

ABSTRACT

On July 7, 2020, the Food and Drug Administration approved Inqovi (Otsuka Pharmaceutical Co.), an oral fixed-dose combination tablet comprising 35 mg decitabine, a hypomethylating agent, and 100 mg cedazuridine, a cytidine deaminase inhibitor (abbreviated DEC-C) for treatment of adult patients with myelodysplastic syndromes (MDS). Evidence of effectiveness of DEC-C was established in phase III ASTX727-02 (N = 133) in adults with MDS. The study involved a two-sequence crossover comparing DEC-C and intravenous (IV) decitabine 20 mg/m2 once daily for the first 5 days of each 28-day cycle in the first 2 cycles. From cycle 3 onward, patients received DEC-C. Five-day cumulative area under the curve (5-d AUC) of decitabine for DEC-C was similar to that of IV decitabine, with geometric mean ratio 0.99 (90% confidence interval: 0.93-1.06). Clinical benefit was supported by study ASTX727-02 and the similarly designed phase II study ASTX727-01-B (n = 80), with complete remission (CR) of 21% and 18% and median duration of CR 7.5 and 8.7 months, respectively. Adverse reactions were consistent with IV decitabine. Postmarketing assessments were issued to address the effect of cedazuridine on QT prolongation, food effect, moderate and severe hepatic impairment, and severe renal impairment on the pharmacokinetics and safety of DEC-C.


Subject(s)
Azacitidine , Myelodysplastic Syndromes , Adult , Azacitidine/adverse effects , Decitabine/adverse effects , Humans , Myelodysplastic Syndromes/drug therapy , Tablets/therapeutic use , Treatment Outcome , Uridine/analogs & derivatives
6.
Pediatr Blood Cancer ; 64(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-28627134

ABSTRACT

The U.S. Food and Drug Administration (FDA) approved eltrombopag for pediatric patients with chronic immune (idiopathic) thrombocytopenia (ITP) ages ≥6 on June 11, 2015, and ages ≥1 on August 24, 2015. Approval was based on the FDA review of two randomized trials that included 159 pediatric patients with chronic ITP who had an insufficient response to corticosteroids, immunoglobulins, or splenectomy. This manuscript describes the basis for approval of these applications. The FDA concluded that eltrombopag has shown efficacy and a favorable benefit to risk profile for pediatric patients with chronic ITP.


Subject(s)
Benzoates/therapeutic use , Hydrazines/therapeutic use , Pyrazoles/therapeutic use , Thrombocythemia, Essential/drug therapy , United States Food and Drug Administration , Adolescent , Benzoates/administration & dosage , Child , Child, Preschool , Chronic Disease , Drug Approval , Female , Humans , Hydrazines/administration & dosage , Infant , Male , Pyrazoles/administration & dosage , United States
7.
Clin Cancer Res ; 22(9): 2111-3, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26783289

ABSTRACT

Sponsors of human drug and biologic products subject to an investigational new drug (IND) application are required to distribute expedited safety reports of serious and unexpected suspected adverse reactions to participating investigators and the FDA to assure the protection of human subjects participating in clinical trials. On September 29, 2010, the FDA issued a final rule amending its regulations governing expedited IND safety reporting requirements that revised the definitions used for reporting and clarified when to submit relevant and useful information to reduce the number of uninformative reports distributed by sponsors. From January 1, 2006, to December 31, 2014, the FDA's Office of Hematology and Oncology Products received an average of 17,686 expedited safety reports per year. An analysis of FDA submissions by commercial sponsors covering this time period suggested a slight increase in the number of expedited safety reports per IND per year after publication of the final rule. An audit of 160 randomly selected expedited safety reports submitted to the FDA's Office of Hematology and Oncology Products in 2015 revealed that only 22 (14%) were informative. The submission of uninformative expedited safety reports by commercial sponsors of INDs continues to be a significant problem that can compromise detection of valid safety signals. Clin Cancer Res; 22(9); 2111-3. ©2016 AACR.


Subject(s)
Drugs, Investigational/adverse effects , Drugs, Investigational/therapeutic use , Drug Approval/methods , Humans , Investigational New Drug Application/methods , Medical Oncology/methods , United States , United States Food and Drug Administration
8.
Cell Cycle ; 14(3): 388-98, 2015.
Article in English | MEDLINE | ID: mdl-25659036

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a cancer of immature T cells that exhibits heterogeneity of oncogenic lesions, providing an obstacle for development of more effective and less toxic therapies. Inherited deficiency of ATM, a regulator of the cellular DNA damage response, predisposes young humans and mice to T-ALLs with clonal chromosome translocations. While acquired ATM mutation or deletion occurs in pediatric T-ALLs, the role of somatic ATM alterations in T-ALL pathogenesis remains unknown. We demonstrate here that somatic Atm inactivation in haematopoietic cells starting as these cells differentiate in utero predisposes mice to T-ALL at similar young ages and harboring analogous translocations as germline Atm-deficient mice. However, some T-ALLs from haematopoietic cell specific deletion of Atm were of more mature thymocytes, revealing that the developmental timing and celluar origin of Atm inactivation influences the phenotype of ATM-deficient T-ALLs. Although it has been hypothesized that ATM suppresses cancer by preventing deletion and inactivation of TP53, we find that Atm inhibits T-ALL independent of Tp53 deletion. Finally, we demonstrate that the Cyclin D3 protein that drives immature T cell proliferation is essential for transformation of Atm-deficient thymocytes. Our study establishes a pre-clinical model for pediatric T-ALLs with acquired ATM inactivation and identifies the cell cycle machinery as a therapeutic target for this aggressive childhood T-ALL subtype.


Subject(s)
Cyclin D3/metabolism , Gene Silencing , Hematopoietic Stem Cells/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Susceptibility , Gene Deletion , Genomic Instability , Heterozygote , Mice, Inbred C57BL , Mice, Transgenic , Thymocytes/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Cell Cycle ; 13(19): 3076-82, 2014.
Article in English | MEDLINE | ID: mdl-25486566

ABSTRACT

ATM is the master regulator of the cellular response to DNA double strand breaks (DSBs). Deficiency of ATM predisposes humans and mice to αß T lymphoid cancers with clonal translocations between the T cell receptor (TCR) α/δ locus and a 450 kb region of synteny on human chromosome 14 and mouse chromosome 12. While these translocations target and activate the TCL1 oncogene at 14q32 to cause T cell pro-lymphocytic leukemia (T-PLL), the TCRα/δ;14q32 translocations in ATM-deficient T cell acute lymphoblastic leukemia (T-ALL) have not been characterized and their role in cancer pathogenesis remains unknown. The corresponding lesion in Atm-deficient mouse T-ALLs is a chromosome t(12;14) translocation with Tcrδ genes fused to sequences on chromosome 12; although these translocations do not activate Tcl1, they delete the Bcl11b haploinsufficient tumor suppressor gene. To assess whether Tcrδ translocations that inactivate one copy of Bcl11b promote transformation of Atm-deficient cells, we analyzed Atm(-/-) mice with mono-allelic Bcl11b deletion initiating in thymocytes concomitant with Tcrδ recombination. Inactivation of one Bcl11b copy had no effect on the predisposition of Atm(-/-) mice to clonal T-ALLs. Yet, none of these T-ALLs had a clonal chromosome t(12;14) translocation that deleted Bcl11b indicating that Tcrδ translocations that inactivate a copy of Bcl11b promote transformation of Atm-deficient thymocytes. Our data demonstrate that antigen receptor locus translocations can cause cancer by deleting a tumor suppressor gene. We discuss the implications of these findings for the etiology and therapy of T-ALLs associated with ATM deficiency and TCRα/δ translocations targeting the 14q32 cytogenetic region.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Repressor Proteins/genetics , Translocation, Genetic , Tumor Suppressor Proteins/genetics , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosomes , Flow Cytometry , Humans , Karyotyping , Metaphase , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism
10.
Hematol Oncol Clin North Am ; 21(6): 1035-49, viii, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17996587

ABSTRACT

Multiple myeloma is a plasma cell malignancy characterized by the frequent development of osteolytic bone lesions. The multiple myeloma-induced bone destruction is a result of the increased activity of osteoclasts that occurs adjacent to multiple myeloma cells. This activity is accompanied by suppressed osteoblast differentiation and activity, resulting in severely impaired bone formation and development of devastating osteolytic lesions. Recently the biologic mechanism involved in the imbalance between osteoclast activation and osteoblast inhibition induced by multiple myeloma cells has begun to be clarified. In this article, the pathophysiology underlying the imbalanced bone remodeling and potential new strategies for the treatment of bone disease in multiple myeloma are reviewed.


Subject(s)
Multiple Myeloma/physiopathology , Osteoclasts/physiology , Osteolysis/physiopathology , Cell Communication/physiology , Humans , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Osteoblasts/drug effects , Osteoblasts/physiology , Osteolysis/etiology , Signal Transduction/physiology
11.
Clin Cancer Res ; 12(22): 6826-35, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121904

ABSTRACT

PURPOSE: The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (mTOR) pathway and the heat shock protein family are up-regulated in multiple myeloma and are both regulators of the cyclin D/retinoblastoma pathway, a critical pathway in multiple myeloma. Inhibitors of mTOR and HSP90 protein have showed in vitro and in vivo single-agent activity in multiple myeloma. Our objective was to determine the effects of the mTOR inhibitor rapamycin and the HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) on multiple myeloma cells. EXPERIMENTAL DESIGN: Multiple myeloma cell lines were incubated with rapamycin (0.1-100 nmol/L) and 17-AAG (100-600 nmol/L) alone and in combination. RESULTS: In this study, we showed that the combination of rapamycin and 17-AAG synergistically inhibited proliferation, induced apoptosis and cell cycle arrest, induced cleavage of poly(ADP-ribose) polymerase and caspase-8/caspase-9, and dysregulated signaling in the phosphatidylinositol 3-kinase/AKT/mTOR and cyclin D1/retinoblastoma pathways. In addition, we showed that both 17-AAG and rapamycin inhibited angiogenesis and osteoclast formation, indicating that these agents target not only multiple myeloma cells but also the bone marrow microenvironment. CONCLUSIONS: These studies provide the basis for potential clinical evaluation of this combination for multiple myeloma patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzoquinones/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/therapeutic use , Multiple Myeloma/drug therapy , Protein Kinases/metabolism , Sirolimus/therapeutic use , Apoptosis/drug effects , Benzoquinones/administration & dosage , Benzoquinones/pharmacology , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/pharmacology , Models, Biological , Neovascularization, Physiologic/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
12.
Ann N Y Acad Sci ; 1068: 240-3, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16831924

ABSTRACT

Eosinophil chemotactic factor-L (ECF-L) is a novel stimulator of osteoclast (OCL) formation that acts at the differentiation/fusion stage of OCL formation, and is a cofactor for RANK ligand (RANKL). We examined the effects of ECF-L on the intracellular signaling pathways utilized by RANKL, and on the expression of ICAM-1/LFA-1 to determine its mechanism of action. RAW 264.7 and bone marrow cells were treated with RANKL and/or ECF-L Fc protein to determine their effect on NF-kappaB and AP-1 activity. ECF-L by itself only modestly increased NF-kappaB binding and JNK activity in RAW 264.7 cells, which were further enhanced by RANKL. In contrast, ECF-L Fc increased LFA-1alpha and ICAM-1 mRNA levels 1.8-fold in mouse marrow cultures, and anti-ICAM-1 almost completely inhibited OCL formation induced by 10(-10) M 1,25-(OH)2D3, and ECF-L Fc. Furthermore, ECF-L Fc did not enhance OCL formation by ICAM-1 knockout (KO) cells. Increased expression of ICAM-1 by ECF-L appears to be critical for its effects on OCL formation.


Subject(s)
Chemokines/pharmacology , Chemotactic Factors, Eosinophil/pharmacology , Intercellular Adhesion Molecule-1/genetics , Osteoclasts/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Communication/physiology , Cell Line , Gene Expression Regulation/drug effects , Macrophages/drug effects , Macrophages/physiology , Mice , Osteoclasts/drug effects
13.
Immunol Rev ; 208: 252-66, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16313353

ABSTRACT

The osteoclast (OCL) is the primary cell involved in the pathogenesis of Paget's disease (PD) and the destructive bone process in multiple myeloma (MM). Both of these diseases are characterized by increased numbers of OCLs actively resorbing bone, but they differ in that bone formation is greatly increased in PD and is suppressed in MM. The marrow microenvironment plays a critical role in both disease processes, through the increased expression of inflammatory cytokines that enhance osteoclastogenesis and, in the case of MM, also suppress osteoblast (OBL) activity. In addition, the OCLs in PD are intrinsically abnormal, are markedly increased in number and size, and are hyper-responsive to inflammatory cytokines and 1,25-(OH)2D3. This article discusses the role of immune cells and inflammatory cytokines and chemokines in the increased OCL activity in PD and MM bone disease, as well as the potential role of interleukin-3 in the suppression of OBL activity in MM.


Subject(s)
Cytokines/physiology , Multiple Myeloma/etiology , Osteitis Deformans/etiology , T-Lymphocytes/physiology , Animals , Carrier Proteins/physiology , Chemokine CCL4 , Dendritic Cells/physiology , Humans , Interleukin-3/physiology , Interleukin-6/physiology , Macrophage Inflammatory Proteins/physiology , Measles virus/pathogenicity , Membrane Glycoproteins/physiology , Osteitis Deformans/genetics , Osteoclasts/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
14.
Blood ; 106(4): 1407-14, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15878977

ABSTRACT

Bone destruction in multiple myeloma is characterized both by markedly increased osteoclastic bone destruction and severely impaired osteoblast activity. We reported that interleukin-3 (IL-3) levels are increased in bone marrow plasma of myeloma patients compared with healthy controls and that IL-3 stimulates osteoclast formation. However, the effects of IL-3 on osteoblasts are unknown. Therefore, to determine if IL-3 inhibits osteoblast growth and differentiation, we treated primary mouse and human marrow stromal cells with IL-3 and assessed osteoblast differentiation. IL-3 inhibited basal and bone morphogenic protein-2 (BMP-2)-stimulated osteoblast formation in a dose-dependent manner without affecting cell growth. Importantly, marrow plasma from patients with high IL-3 levels inhibited osteoblast differentiation, which could be blocked by anti-IL-3. However, IL-3 did not inhibit osteoblast differentiation of osteoblastlike cell lines. In contrast, IL-3 increased the number of CD45+ hematopoietic cells in stromal-cell cultures. Depletion of the CD45+ cells abolished the inhibitory effects of IL-3 on osteoblasts, and reconstitution of the cultures with CD45+ cells restored the capacity of IL-3 to inhibit osteoblast differentiation. These data suggest that IL-3 plays a dual role in the bone destructive process in myeloma by both stimulating osteoclasts and indirectly inhibiting osteoblast formation.


Subject(s)
Interleukin-3/pharmacology , Multiple Myeloma/pathology , Osteoblasts/drug effects , Animals , Bone Marrow Cells/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Hematopoietic Stem Cells , Humans , Leukocyte Common Antigens , Mice , Multiple Myeloma/drug therapy , Osteoblasts/pathology , Osteoclasts/drug effects , Stromal Cells/pathology
15.
Exp Hematol ; 33(3): 272-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15730850

ABSTRACT

OBJECTIVES: Macrophage inflammatory protein-1alpha (MIP-1alpha), an osteoclast (OCL) stimulatory factor produced by primary multiple myeloma (MM) cells, increases bone destruction and tumor burden in murine models of MM. Several chemokine receptors (CCR1, CCR5, and CCR9) mediate the effects of MIP-1alpha. In this study, we determined which of these mediates the effects of MIP-1alpha on human OCL formation and myeloma cells. METHODS: We employed RT-PCR analysis, neutralizing antibodies to CCR1 and CCR5 as well as a CCR1-specific antagonist and OCL formation assays to identify the MIP-1alpha receptors involved in MIP-1alpha's effects on myeloma cells and OCL formation. RESULTS: RT-PCR analysis demonstrated that both CCR1 and CCR5 were expressed by highly purified human OCL precursors, myeloma cell lines, and purified marrow plasma cells from MM patients. Neutralizing antibodies to CCR1 or CCR5 inhibited MIP-1alpha-induced OCL formation. Furthermore, monocyte chemotactic protein-3 (MCP-3), which binds CCR1 but not CCR5 and the CCR1-specific antagonist, BX471, markedly inhibited OCL formation stimulated with MIP-1alpha. Anti-CCR1, anti-CCR5, or BX471 also inhibited the upregulation of beta1 integrin mRNA in myeloma cells induced by MIP-1alpha, as well as the adherence of myeloma cells to stromal cells and IL-6 production by stromal cells in response to myeloma cells. CONCLUSION: These data demonstrate that MIP-1alpha utilizes either CCR1 or CCR5 for its effects on OCL formation and myeloma cells, and that blocking either CCR1 or CCR5 inhibits OCL formation and myeloma cell adhesion to stromal cells.


Subject(s)
Bone Marrow Cells/cytology , Macrophage Inflammatory Proteins/metabolism , Multiple Myeloma/metabolism , Osteoclasts/physiology , Receptors, CCR5/metabolism , Receptors, Chemokine/metabolism , Bone Marrow Cells/physiology , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Chemokine CCL3 , Chemokine CCL4 , Gene Expression Regulation, Neoplastic , Humans , Multiple Myeloma/pathology , Osteoclasts/cytology , Receptors, CCR1 , Receptors, CCR5/genetics , Receptors, Chemokine/genetics , Stromal Cells/cytology , Stromal Cells/physiology
16.
Blood ; 103(6): 2308-15, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14615378

ABSTRACT

Macrophage inflammatory protein-1 alpha (MIP-1 alpha) gene expression is abnormally regulated in multiple myeloma (MM) owing to imbalanced expression of the acute myeloid leukemia-1A (AML-1A) and AML-1B transcription factors. We hypothesized that the increased expression ratios of AML-1A to AML-1B also induced abnormal expression of other hematopoietic and bone-specific genes that contribute to the poor prognosis of MM patients with high levels of MIP-1 alpha. We found that interleukin-3 (IL-3) was also induced by the imbalanced AML-1A and AML-1B expression in myeloma. IL-3 mRNA levels were increased in CD138+ purified myeloma cells with increased AML-1A-to-AML-1B expression from MM patients, and IL-3 protein levels were significantly increased in freshly isolated bone marrow plasma from MM patients (66.4 +/- 12 versus 22.1 +/- 8.2 pg/mL; P = .038). IL-3 in combination with MIP-1 alpha or receptor activator of nuclear factor-kappa B ligand (RANKL) significantly enhanced human osteoclast (OCL) formation and bone resorption compared with MIP-1 alpha or RANKL alone. IL-3 stimulated the growth of interleukin-6 (IL-6)-dependent and IL-6-independent myeloma cells in the absence of IL-6, even though IL-3 did not induce IL-6 expression by myeloma cells. These data suggest that increased IL-3 levels in the bone marrow microenvironment of MM patients with imbalanced AML-1A and AML-1B expression can increase bone destruction and tumor cell growth.


Subject(s)
Interleukin-3/genetics , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Osteoclasts/cytology , Osteoclasts/physiology , Antibodies/pharmacology , Bone Marrow Cells/physiology , Bone Resorption/physiopathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Line, Tumor , Chemokine CCL4 , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Interleukin-3/immunology , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RANK Ligand , RNA, Messenger/analysis , Receptor Activator of Nuclear Factor-kappa B , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...