Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Haematologica ; 106(12): 3176-3187, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-33147936

ABSTRACT

Multiple myeloma (MM) disease progression is dependent on the ability of MM plasma cells (PCs) to egress from the bone marrow (BM), enter the circulation and disseminate to distal BM sites. Expression of the chemokine CXCL12 by BM stromal cells is crucial for MM PC retention within the BM. However, the mechanisms which overcome CXCL12-mediated retention to enable dissemination are poorly understood. We have previously identified that treatment with the CCR1 ligand CCL3 inhibits the response to CXCL12 in MM cell lines, suggesting that CCL3/CCR1 signalling may enable egress of MM PC from the BM. Here, we demonstrated that CCR1 expression was an independent prognostic indicator in newly diagnosed MM patients. Furthermore, we showed that CCR1 is a crucial driver of dissemination in vivo, with CCR1 expression in the murine MM cell line 5TGM1 being associated with an increased incidence of bone and splenic disseminated tumours in C57BL/KaLwRij mice. Furthermore, we demonstrated that CCR1 knockout in the human myeloma cell line OPM2 resulted in a >95% reduction in circulating MM PC numbers and BM and splenic tumour dissemination following intratibial injection in NSG mice. Therapeutic targeting of CCR1 with the inhibitor CCX9588 significantly reduced OPM2 or RPMI-8226 dissemination in intratibial xenograft models. Collectively, our findings suggest a novel role for CCR1 as a critical driver of BM egress of MM PCs during tumour dissemination. Furthermore, these data suggest that CCR1 may represent a potential therapeutic target for the prevention of MM tumour dissemination.


Subject(s)
Multiple Myeloma , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Plasma Cells , Receptors, CCR1/genetics
2.
Nat Genet ; 51(4): 694-704, 2019 04.
Article in English | MEDLINE | ID: mdl-30926971

ABSTRACT

Acute erythroid leukemia (AEL) is a high-risk leukemia of poorly understood genetic basis, with controversy regarding diagnosis in the spectrum of myelodysplasia and myeloid leukemia. We compared genomic features of 159 childhood and adult AEL cases with non-AEL myeloid disorders and defined five age-related subgroups with distinct transcriptional profiles: adult, TP53 mutated; NPM1 mutated; KMT2A mutated/rearranged; adult, DDX41 mutated; and pediatric, NUP98 rearranged. Genomic features influenced outcome, with NPM1 mutations and HOXB9 overexpression being associated with a favorable prognosis and TP53, FLT3 or RB1 alterations associated with poor survival. Targetable signaling mutations were present in 45% of cases and included recurrent mutations of ALK and NTRK1, the latter of which drives erythroid leukemogenesis sensitive to TRK inhibition. This genomic landscape of AEL provides the framework for accurate diagnosis and risk stratification of this disease, and the rationale for testing targeted therapies in this high-risk leukemia.


Subject(s)
Leukemia, Erythroblastic, Acute/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Genomics/methods , Homeodomain Proteins/genetics , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Tumor Suppressor Protein p53/genetics , Young Adult , fms-Like Tyrosine Kinase 3/genetics
3.
Cancer Res ; 77(20): 5452-5463, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28855206

ABSTRACT

Disease progression and relapse in multiple myeloma is dependent on the ability of the multiple myeloma plasma cells (PC) to reenter the circulation and disseminate throughout the bone marrow. Increased bone marrow hypoxia is associated with increased recirculation of multiple myeloma PCs. Accordingly, we hypothesized that during chronic hypoxia, activation of HIF-2α may overcome the bone marrow retention signal provided by stromal-derived CXCL12, thereby enabling dissemination of multiple myeloma PCs. Here we demonstrate that HIF-2α upregulates multiple myeloma PC CXCL12 expression, decreasing migration toward CXCL12 and reducing adhesion to mesenchymal stromal cells in vitro We also found that HIF-2α strongly induced expression of the chemokine receptor CCR1 in multiple myeloma PCs. CCR1 activation potently induces multiple myeloma PC migration toward CCL3 while abrogating the multiple myeloma PC migratory response to CXCL12. In addition, increased CCR1 expression by multiple myeloma PCs conferred poor prognosis in newly diagnosed multiple myeloma patients and was associated with an increase in circulating multiple myeloma PCs in these patients. Taken together, our results suggest a role for hypoxia-mediated CCR1 upregulation in driving the egress of multiple myeloma PCs from the bone marrow. Targeting CCR1 may represent a novel strategy to prevent dissemination and overt relapse in multiple myeloma. Cancer Res; 77(20); 5452-63. ©2017 AACR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chemokine CXCL12/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Plasma Cells/pathology , Receptors, CCR1/metabolism , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Plasma Cells/metabolism , Tumor Cells, Cultured
4.
Am J Hematol ; 92(6): 508-514, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28247421

ABSTRACT

RBC-transfusion dependency (RBC-TD) is an independent prognostic factor for poor overall survival (OS) in the WHO classification-based prognostic scoring system (WPSS) for MDS patients. However, WPSS did not include cytopenia, whereas revised International Prognostic Scoring System (IPSS-R) did not include RBC-TD. Thus, neither of these prognostic scoring systems incorporates both cytopenia and RBC-TD. We aimed to test whether RBC-TD adds prognostic value to the IPSS-R. We analyzed MDS patients not treated with disease-modifying therapy, and enrolled in SA-MDS Registry (derivation cohort; n = 295) and Dusseldorf registry (Germany; validation cohort; n = 113) using time-dependent Cox proportional regression and serial landmark analyses. In the derivation cohort, RBC-TD patients had inferior OS compared to RBC transfusion-independent (RBC-TI) patients (P < 0.0001) at 6- (18 vs. 64 months), 12- (24 vs. 71 months), and 24-months (40 vs. 87 months). In a Cox proportional regression analysis, RBC-TD was an independent adverse prognostic marker in addition to age, sex, and IPSS-R variables (P < 0.0001). A prognostic index (PI) was derived using these Cox-proportional regression model variables. In the validation cohort, this PI classified patients into four prognostic groups with significantly different OS (P < 0.001) as in the derivation cohort. In conclusion, multivariate analysis by Cox proportional hazards regression and serial landmark analyses clearly demonstrates that development of RBC-TD at any time during the course of MDS is associated with poor OS, independent of IPSS-R. This study demonstrates that dynamic assessment of RBC-TD provides additional prognostic value to IPSS-R and should be included in treatment decision algorithms for MDS patients.


Subject(s)
Erythrocyte Transfusion , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Adult , Aged , Aged, 80 and over , Australia , Cause of Death , Combined Modality Therapy , Disease Management , Erythrocyte Transfusion/methods , Female , Germany , Humans , Incidence , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Prognosis , Proportional Hazards Models , Registries , Reproducibility of Results , Treatment Outcome , Young Adult
5.
Blood ; 125(17): 2649-55, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25762180

ABSTRACT

Pacritinib (SB1518) is a Janus kinase 2 (JAK2), JAK2(V617F), and Fms-like tyrosine kinase 3 inhibitor that does not inhibit JAK1. It demonstrated a favorable safety profile with promising efficacy in phase 1 studies in patients with primary and secondary myelofibrosis (MF). This multicenter phase 2 study further characterized the safety and efficacy of pacritinib in the treatment of patients with MF. Eligible patients had clinical splenomegaly poorly controlled with standard therapies or were newly diagnosed with intermediate- or high-risk Lille score. Patients with any degree of cytopenia were eligible. Thirty-five patients were enrolled. At entry, 40% had hemoglobin <10 g/dL and 43% had platelets <100 000× 10(9)/L. Up to week 24, 8 of 26 evaluable patients (31%) achieved a ≥35% decrease in spleen volume determined by magnetic resonance imaging and 14 of 33 (42%) attained a ≥50% reduction in spleen size by physical examination. Median MF symptom improvement was ≥50% for all symptoms except fatigue. Grade 1 or 2 diarrhea (69%) and nausea (49%) were the most common treatment-emergent adverse events. The study drug was discontinued in 9 patients (26%) due to adverse events (4 severe). Pacritinib is an active agent in patients with MF, offering a potential treatment option for patients with preexisting anemia and thrombocytopenia. This trial was registered at www.clinicaltrials.gov as #NCT00745550.


Subject(s)
Bridged-Ring Compounds/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Pyrimidines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Bridged-Ring Compounds/adverse effects , Female , Humans , Male , Middle Aged , Primary Myelofibrosis/complications , Primary Myelofibrosis/pathology , Pyrimidines/adverse effects , Spleen/drug effects , Spleen/pathology , Splenomegaly/complications , Splenomegaly/drug therapy , Splenomegaly/pathology
6.
Exp Cell Res ; 332(1): 24-38, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25637218

ABSTRACT

BACKGROUND: Increased expression of the tetraspanin TSPAN7 has been observed in a number of cancers; however, it is unclear how TSPAN7 plays a role in cancer progression. METHODS: We investigated the expression of TSPAN7 in the haematological malignancy multiple myleoma (MM) and assessed the consequences of TSPAN7 expression in the adhesion, migration and growth of MM plasma cells (PC) in vitro and in bone marrow (BM) homing and tumour growth in vivo. Finally, we characterised the association of TSPAN7 with cell surface partner molecules in vitro. RESULTS: TSPAN7 was found to be highly expressed at the RNA and protein level in CD138(+) MM PC from approximately 50% of MM patients. TSPAN7 overexpression in the murine myeloma cell line 5TGM1 significantly reduced tumour burden in 5TGM1/KaLwRij mice 4 weeks after intravenous adminstration of 5TGM1 cells. While TSPAN7 overexpression did not affect cell proliferation in vitro, TSPAN7 increased 5TGM1 cell adhesion to BM stromal cells and transendothelial migration. In addition, TSPAN7 was found to associate with the molecular chaperone calnexin on the cell surface. CONCLUSION: These results suggest that elevated TSPAN7 may be associated with better outcomes for up to 50% of MM patients.


Subject(s)
Multiple Myeloma/metabolism , Nerve Tissue Proteins/genetics , Tetraspanins/genetics , Animals , Calnexin/genetics , Calnexin/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Gene Expression , Humans , Mice, Inbred C57BL , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , Proportional Hazards Models , Tetraspanins/metabolism , Up-Regulation
7.
Biol Blood Marrow Transplant ; 19(5): 760-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23298856

ABSTRACT

Autologous hematopoietic cell transplantation (AHCT) as initial therapy of patients with multiple myeloma (MM) improves survival. However, data to support this approach for relapsed/progressive disease after initial AHCT (AHCT1) are limited. Using Center for International Blood and Marrow Transplant Research data, we report the outcomes of 187 patients who underwent a second AHCT (AHCT2) for the treatment of relapsed/progressive MM. Planned tandem AHCT was excluded. Median age at AHCT2 was 59 years (range, 28 to 72), and median patient follow-up was 47 months (range, 3 to 97). Nonrelapse mortality after AHCT2 was 2% at 1 year and 4% at 3 years. Median interval from AHCT1 to relapse/progression was 18 months, and median interval between transplantations was 32 months. After AHCT2, the incidence of relapse/progression at 1 and 3 years was 51% and 82%, respectively. At 3 years after AHCT2, progression-free survival was 13%, and overall survival was 46%. In multivariate analyses, those relapsing ≥36 months after AHCT1 had superior progression-free (P = .045) and overall survival (P = .019). Patients who underwent AHCT2 after 2004 had superior survival (P = .026). AHCT2 is safe and feasible for disease progression after AHCT1. In this retrospective study, individuals relapsing ≥36 months from AHCT1 derived greater benefit from AHCT2 compared with those with a shorter disease-free interval. Storage of an adequate graft before AHCT1 will ensure that the option of a second autologous transplantation is retained for patients with relapsed/progressive MM.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/surgery , Salvage Therapy/methods , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Recurrence , Treatment Outcome
8.
Nat Genet ; 45(3): 242-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334668

ABSTRACT

The genetic basis of hypodiploid acute lymphoblastic leukemia (ALL), a subtype of ALL characterized by aneuploidy and poor outcome, is unknown. Genomic profiling of 124 hypodiploid ALL cases, including whole-genome and exome sequencing of 40 cases, identified two subtypes that differ in the severity of aneuploidy, transcriptional profiles and submicroscopic genetic alterations. Near-haploid ALL with 24-31 chromosomes harbor alterations targeting receptor tyrosine kinase signaling and Ras signaling (71%) and the lymphoid transcription factor gene IKZF3 (encoding AIOLOS; 13%). In contrast, low-hypodiploid ALL with 32-39 chromosomes are characterized by alterations in TP53 (91.2%) that are commonly present in nontumor cells, IKZF2 (encoding HELIOS; 53%) and RB1 (41%). Both near-haploid and low-hypodiploid leukemic cells show activation of Ras-signaling and phosphoinositide 3-kinase (PI3K)-signaling pathways and are sensitive to PI3K inhibitors, indicating that these drugs should be explored as a new therapeutic strategy for this aggressive form of leukemia.


Subject(s)
Aneuploidy , Chromosome Aberrations , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Haploidy , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Transplantation, Heterologous , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Leuk Res ; 36(1): 110-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21993314

ABSTRACT

Krüppel-like factor 5 (KLF5) has been implicated as a tumor suppressor in various solid tumors such as breast and prostate, and recent studies have demonstrated a role for this protein in neutrophil differentiation of acute promyelocytic leukemia cells in response to ATRA. Here, we show that KLF5 expression increases during primary granulocyte differentiation and that expression of KLF5 is a requirement for granulocyte differentiation of 32D cells. In AML, we show that KLF5 mRNA expression levels are reduced in multiple French-American-British subtypes compared to normal controls, and also in leukemic stem cells relative to normal hematopoietic stem cells. We demonstrate that in selected AML cases, reduced expression is associated with hypermethylation of the KLF5 locus in the proximal promoter and/or intron 1, suggesting that this may represent a Class II genetic lesion in the development of AML.


Subject(s)
DNA Methylation , Gene Silencing , Granulocytes/metabolism , Kruppel-Like Transcription Factors/genetics , Leukemia, Myeloid, Acute/genetics , Cell Line, Tumor , DNA Methylation/physiology , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Gene Silencing/physiology , Genetic Loci , Granulocytes/pathology , Humans , Kruppel-Like Transcription Factors/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Microarray Analysis , Promoter Regions, Genetic/genetics , Up-Regulation
10.
Leukemia ; 26(5): 1091-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22042147

ABSTRACT

There are limited data on hematopoietic cell transplantation (HCT) in primary plasma cell leukemia (pPCL), an aggressive plasma cell disorder. We report outcomes of 147 patients with pPCL receiving autologous (n=97) or allogeneic (n=50) HCT within 18 months after diagnosis between 1995 and 2006. Median age was 56 years and 48 years for autologous HCT and allogeneic HCT, respectively. Progression-free survival (PFS) at 3 years was 34% (95% confidence interval (CI), 23-46%) in the autologous group and 20% (95% CI, 10-34%) in the allogeneic group. Cumulative incidence of relapse at 3 years was 61% (95% CI, 48-72%) in the autologous group and 38% (95% CI, 25-53%) in the allogeneic group. Overall survival (OS) at 3 years was 64% (95% CI, 52-75%) in the autologous group and 39% (95% CI, 26-54%) in the allogeneic group. Non-relapse mortality (NRM) at 3 years was 5% (95% CI, 1-11%) in the autologous group and 41% (95% CI, 28-56%) in the allogeneic group. The encouraging OS after autologous HCT, establishes the safety and feasibility of this consolidative treatment option after initial induction therapy for pPCL. Allogeneic HCT, although associated with a significantly lower relapse rate, carries a much higher risk of NRM and no OS benefit.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Plasma Cell/surgery , Adult , Cohort Studies , Female , Humans , Male , Middle Aged
11.
Pathology ; 43(6): 547-65, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921732

ABSTRACT

This review evaluates the latest information on the mobilisation of haemopoietic stem cells for transplantation, with the focus on what is the current best practice and how new understanding of the bone marrow stem cell niche provides new insights into optimising mobilisation regimens. The review then looks at the mobilisation of mesenchymal stromal cells, immune cells as well as malignant cells and what clinical implications there are.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Transplantation Conditioning/methods , Animals , Humans , Models, Animal
12.
Nat Genet ; 43(10): 1012-7, 2011 Sep 04.
Article in English | MEDLINE | ID: mdl-21892162

ABSTRACT

We report the discovery of GATA2 as a new myelodysplastic syndrome (MDS)-acute myeloid leukemia (AML) predisposition gene. We found the same, previously unidentified heterozygous c.1061C>T (p.Thr354Met) missense mutation in the GATA2 transcription factor gene segregating with the multigenerational transmission of MDS-AML in three families and a GATA2 c.1063_1065delACA (p.Thr355del) mutation at an adjacent codon in a fourth MDS family. The resulting alterations reside within the second zinc finger of GATA2, which mediates DNA-binding and protein-protein interactions. We show differential effects of the mutations on the transactivation of target genes, cellular differentiation, apoptosis and global gene expression. Identification of such predisposing genes to familial forms of MDS and AML is critical for more effective diagnosis and prognosis, counseling, selection of related bone marrow transplant donors and development of therapies.


Subject(s)
GATA2 Transcription Factor/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Quantitative Trait, Heritable , Amino Acid Sequence , Animals , COS Cells , Cell Differentiation , Cell Proliferation , Chlorocebus aethiops , Chromosome Mapping , DNA, Complementary , Female , GATA2 Transcription Factor/metabolism , Genetic Predisposition to Disease , HEK293 Cells , Haplotypes , Humans , Male , Molecular Sequence Data , Pedigree , Plasmids , Polymorphism, Single Nucleotide
13.
Blood ; 118(17): 4530-40, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21832280

ABSTRACT

Transplantation with 2-5 × 10(6) mobilized CD34(+)cells/kg body weight lowers transplantation costs and mortality. Mobilization is most commonly performed with recombinant human G-CSF with or without chemotherapy, but a proportion of patients/donors fail to mobilize sufficient cells. BM disease, prior treatment, and age are factors influencing mobilization, but genetics also contributes. Mobilization may fail because of the changes affecting the HSC/progenitor cell/BM niche integrity and chemotaxis. Poor mobilization affects patient outcome and increases resource use. Until recently increasing G-CSF dose and adding SCF have been used in poor mobilizers with limited success. However, plerixafor through its rapid direct blockage of the CXCR4/CXCL12 chemotaxis pathway and synergy with G-CSF and chemotherapy has become a new and important agent for mobilization. Its efficacy in upfront and failed mobilizers is well established. To maximize HSC harvest in poor mobilizers the clinician needs to optimize current mobilization protocols and to integrate novel agents such as plerixafor. These include when to mobilize in relation to chemotherapy, how to schedule and perform apheresis, how to identify poor mobilizers, and what are the criteria for preemptive and immediate salvage use of plerixafor.


Subject(s)
Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation/methods , Hemoglobinuria, Paroxysmal/therapy , Transplantation Conditioning/methods , Anemia, Aplastic , Bone Marrow Diseases , Bone Marrow Failure Disorders , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Humans , Models, Biological , Treatment Failure
14.
Blood ; 114(23): 4859-70, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19805619

ABSTRACT

Deregulated cell survival programs are a classic hallmark of cancer. We have previously identified a serine residue (Ser585) in the betac subunit of the granulocyte-macrophage colony-stimulating factor receptor that selectively and independently promotes cell survival. We now show that Ser585 phosphorylation is constitutive in 20 (87%) of 23 acute myeloid leukemia (AML) patient samples, indicating that this survival-only pathway is frequently deregulated in leukemia. We performed a global expression screen to identify gene targets of this survival pathway and report a 138-gene betac Ser585-regulated transcriptome. Pathway analysis defines a gene network enriched for PI3-kinase target genes and a cluster of genes involved in cancer and cell survival. We show that one such gene, osteopontin (OPN), is a functionally relevant target of the Ser585-survival pathway as shown by siRNA-mediated knockdown of OPN expression that induces cell death in both AML blasts and CD34(+)CD38(-)CD123(+) leukemic progenitors. Increased expression of OPN at diagnosis is associated with poor prognosis with multivariate analysis indicating that it is an independent predictor of overall patient survival in normal karyotype AML (n = 60; HR = 2.2; P = .01). These results delineate a novel cytokine-regulated Ser585/PI3-kinase signaling network that is deregulated in AML and identify OPN as a potential prognostic and therapeutic target.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid/genetics , Neoplasm Proteins/physiology , Osteopontin/physiology , Adult , Aged , Cell Survival , Cytokine Receptor Common beta Subunit/metabolism , Female , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Gene Regulatory Networks , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteopontin/biosynthesis , Osteopontin/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphoserine/metabolism , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
15.
Stem Cells ; 27(9): 2293-300, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19544531

ABSTRACT

Limited cell numbers in a unit restricts cord blood transplantation (CBT) in adults. We evaluated whether cotransplantation of placental mesenchymal stromal cells (MSCs) would enhance engraftment. Plastic adherent cells from placenta demonstrated typical characteristics of MSCs. In six individual experiments, 4 cohorts of 24 nonobese diabetic/severe combined immune deficient (NOD/SCID) mice were evaluated. Cohort 1 received 5 x 10(4) CD34+ cells from unit (U) one (SCBT); cohort 2 received 5 x 10(4) CD34+ cells from U1 + 4 x 10(4) MSCs (SCBT+MSCs); cohort 3 received 2.5 x 10(4) CD34+ cells from U1 + 2.5 x 10(4) CD34+ cells from U2 (double cord blood transplant [DCBT]); cohort 4 received 2.5 x 10(4) CD34+ cells from U1 + 2.5 x 10(4) CD34+ cells from U2 + 4 x 10(4) MSCs (DCBT+MSCs). Hematopoietic engraftment evaluated after 6 to 8 weeks, was similar in recipients of SCBT and DCBT. MSC cotransplantation demonstrated enhanced engraftment in DCBT (51.8 +/- 6.8% versus 14.9 +/- 6.5%; p = .04) with an increased trend in SCBT (48.7 +/- 7.7% versus 17.5 +/- 6.1%; p = .07). In DCBT, cotransplantation of placental MSCs reduced single cord dominance. Self-renewal capacity was assessed by serial transplantation in secondary recipients infused with engrafted human cells from primary mice transplanted with or without MSCs. In secondary transplant experiments, 13 of 17 evaluable mice engrafted at levels of 1% to 6.5%. Despite enhanced engraftment in primary mice, long-term engraftment capacity was unaltered with MSC cotransplantation. Imaging studies showed MSCs migrated to pelvic region and improved cord blood (CB) CD34+ homing. Cotransplantation of placental MSCs enhanced cord blood engraftment and may act by improving homing of CD34+ cells.


Subject(s)
Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Placenta/cytology , Stromal Cells/cytology , Animals , Antigens, CD34/metabolism , Cells, Cultured , Cord Blood Stem Cell Transplantation/methods , Female , Flow Cytometry , Humans , Karyotyping , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Pregnancy , Stromal Cells/metabolism , Stromal Cells/physiology
16.
J Bone Miner Res ; 24(7): 1150-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19335218

ABSTRACT

The plasma cell (PC) malignancy, multiple myeloma (MM), is unique among hematological malignancies in its capacity to cause osteoclast (OC)-mediated skeletal destruction. We have previously shown that elevated plasma levels of PC-derived CXCL12 are associated with presence of X-ray detectable osteolytic lesions in MM patients. To further investigate this relationship, plasma levels of CXCL12 and betaCrossLaps, a marker of bone loss, were measured. A strong correlation between levels of CXCL12 and OC-mediated bone resorption was identified. To confirm the OC-activating potential of MM PC-derived CXCL12 in vivo, we established a model of MM-mediated focal osteolysis, wherein MM PC lines, such as RPMI-8226, were injected into the tibias of nude mice. Implanting RPMI-8226 gave rise to osteolytic lesions proximal to the tumor, resulting in a 5% decrease in bone volume (BV) compared with vehicle control. Importantly, bone loss was significantly inhibited with systemic administration of the CXCL12/CXCR4 antagonist T140. Furthermore, implanting CXCL12-overexpressing RPMI-8226 cells resulted in a 13% decrease in BV and was associated with increased OC recruitment proximal to the tumor, increased serum matrix metalloproteinase activity, and increased levels of collagen I degradation products. These findings confirm our hypothesis that MM PC-derived CXCL12 stimulates the recruitment and activity of OC, thereby contributing to the formation of MM osteolytic lesions.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Multiple Myeloma/drug therapy , Oligopeptides/pharmacology , Osteolysis/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Chemokine CXCL12/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Transplantation , Osteoclasts/metabolism , Osteoclasts/pathology , Osteolysis/metabolism , Osteolysis/pathology , Receptors, CXCR4/metabolism
17.
Biol Blood Marrow Transplant ; 14(10): 1118-1124, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18804041

ABSTRACT

Relapse is the overwhelming cause of treatment failure after autologous transplantation for multiple myeloma (MM). For patients with a syngeneic donor, twin transplants provide a healthy graft that is free of myeloma. The relative impact of the graft on posttransplant relapse can be estimated by comparing risk of relapse after hematopoietic cell transplantation from genetically identical twins versus autotransplants because confounding differences in minor or major histocompatibility antigens are absent in the syngeneic transplant setting. Outcomes of 43 subjects who received twin transplants for MM were compared to 170 matched autotransplant recipients reported to the Center for International Blood and Marrow Transplant Research (CIBMTR). Multivariate analysis was performed by fitting a Cox model stratified on matched pairs. The matched transplant patients studied were similar with respect to subject-, disease-, and transplant-related characteristics. Cumulative incidence of relapse/progression was significantly lower, and progression-free survival (PFS) was significantly higher following twin transplants. In multivariate analysis, the probability of relapse/progression was lower in twins (relative risk [RR] = 0.49, 95% confidence interval [CI] 0.28-0.86, P = .011). Twin transplants have a significantly lower relapse risk than autotransplants in MM, suggesting that graft composition may impact outcomes following high-dose chemotherapy.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , Transplantation Immunology , Adult , Aged , Analysis of Variance , Disease-Free Survival , Female , Humans , Male , Middle Aged , Recurrence , Transplantation, Autologous/immunology , Transplantation, Isogeneic/immunology , Twins, Monozygotic
18.
Biol Blood Marrow Transplant ; 14(10): 1134-1140, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18804043

ABSTRACT

Nonsecretory myeloma (NSM) accounts for <5% of cases of multiple myeloma (MM). The outcome of these patients following autologous stem cell transplantation (ASCT) has not been evaluated in clinical trials. We compared the outcomes after ASCT for patients with NSM reported to the Center for International Blood and Marrow Transplant Research (CIBMTR) between 1989 and 2003, to a matched group of 438 patients (4 controls for each patient) with secretory myeloma (SM). The patients were matched using propensity scores calculated using age, Durie-Salmon stage, sensitivity to pretransplant therapy, time from diagnosis to transplant, and year of transplant. Disease characteristics were similar in both groups at diagnosis and at transplant except higher risk of anemia, hypoalbuminemia, and marrow plasmacytosis (in SM) and plasmacytoma (more in NSM). Cumulative incidence of treatment-related mortality (TRM), relapse, progression-free survival (PFS), and overall survival (OS) were similar between the groups. In multivariate analysis, based on a Cox model stratified on matched pairs and adjusted for covariates not considered in the propensity score, we found no difference in outcome between the NSM and SM groups. In this large cohort of patients undergoing ASCT, we found no difference in outcomes of patients with NSM compared to those with SM.


Subject(s)
Hematopoietic Stem Cell Transplantation/statistics & numerical data , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Adult , Analysis of Variance , Humans , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/mortality , Recurrence , Retrospective Studies , Survival Analysis , Transplantation, Autologous , Treatment Outcome
19.
Clin Cancer Res ; 14(12): 3881-8, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559609

ABSTRACT

PURPOSE: The organic cation transporter OCT-1 mediates active transport of imatinib. We recently showed that low OCT-1 activity is a major contributor to suboptimal response in chronic myeloid leukemia (CML) patients treated with imatinib. The relevance of OCT-1 activity and efflux pumps in determining intracellular uptake and retention (IUR) of dasatinib was assessed. EXPERIMENTAL DESIGN: The effect of OCT inhibitors on [14C]dasatinib and [14C]imatinib IUR was compared using peripheral blood mononuclear cells from newly diagnosed CML patients. The role of efflux transporters was studied using ABCB1- and ABCG2-overexpressing cell lines and relevant inhibitors. RESULTS: Unlike imatinib, there was no significant difference in the dasatinib IUR at 37 degrees C and 4 degrees C (P = 0.8), and OCT-1 inhibitors including prazosin did not reduce dasatinib IUR significantly. In CML mononuclear cells, prazosin inhibitable IUR was significantly higher for imatinib than dasatinib (6.38 versus 1.48 ng/200,000 cells; P = 0.002; n = 11). Patients with high OCT-1 activity based on their imatinib uptake had IC50(dasatinib) values equivalent to patients with low OCT-1 activity. Dasatinib IUR was significantly lower in ABCB1-overexpressing cell lines compared with parental cell lines (P < 0.05). PSC833 (ABCB1 inhibitor) significantly increased the dasatinib IUR (P < 0.05) and reduced IC50(dasatinib) (from 100 to 8 nmol/L) in K562-DOX cell line. The ABCG2 inhibitor Ko143 significantly increased dasatinib IUR in ABCG2-overexpressing cell lines and reduced IC(50)(dasatinib). CONCLUSION: Unlike imatinib, dasatinib cellular uptake is not significantly affected by OCT-1 activity, so that expression and function of OCT-1 is unlikely to affect response to dasatinib. Dasatinib is a substrate of both efflux proteins, ABCB1 and ABCG2.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/physiology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzamides , Biological Transport/drug effects , Dasatinib , Drug Evaluation, Preclinical , Fusion Proteins, bcr-abl/metabolism , HL-60 Cells , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Mice , Neoplasm Proteins/physiology , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Piperazines/pharmacokinetics , Temperature , Tumor Cells, Cultured
20.
Blood ; 111(5): 2538-47, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18042796

ABSTRACT

Imatinib inhibits tyrosine kinases important in osteoclast (c-Fms) and osteoblast (platelet-derived growth factor receptor [PDGF-R], c-Abl) function, suggesting that long-term therapy may alter bone homeostasis. To investigate this question, we measured the trabecular bone volume (TBV) in iliac crest bone biopsies taken from chronic myeloid leukemia (CML) patients at diagnosis and again after 2 to 4 years of imatinib therapy. Half the patients (8 of 17) showed a substantive increase in TBV (> 2-fold), after imatinib therapy, with the TBV in the posttreatment biopsy typically surpassing the normal upper limit for the patient's age group. Imatinib-treated patients exhibited reduced serum calcium and phosphate levels with hypophosphatemia evident in 53% (9 of 17) of patients. In vitro, imatinib suppressed osteoblast proliferation and stimulated osteogenic gene expression and mineralized-matrix production by inhibiting PDGF receptor function. In PDGF-stimulated cultures, imatinib dose-dependently inhibited activation of Akt and Crk-L. Using pharmacologic inhibitors, inhibition of PI3-kinase/Akt activation promoted mineral formation, suggesting a possible molecular mechanism for the imatinib-mediated increase in TBV in vivo. Further investigation is required to determine whether the increase in TBV associated with imatinib therapy may represent a novel therapeutic avenue for the treatment of diseases that are characterized by generalized bone loss.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Osteogenesis/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Adipogenesis/drug effects , Adipogenesis/genetics , Adult , Aged , Benzamides , Calcification, Physiologic/drug effects , Calcium/blood , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Middle Aged , Organ Size/drug effects , Osteogenesis/genetics , Phosphates/blood , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...