Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
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.
Leukemia ; 32(2): 263-272, 2018 02.
Article in English | MEDLINE | ID: mdl-28674423

ABSTRACT

Prognostic gene expression signatures have been proposed as clinical tools to clarify therapeutic options in acute myeloid leukemia (AML). However, these signatures rely on measuring large numbers of genes and often perform poorly when applied to independent cohorts or those with older patients. Long intergenic non-coding RNAs (lincRNAs) are emerging as important regulators of cell identity and oncogenesis, but knowledge of their utility as prognostic markers in AML is limited. Here we analyze transcriptomic data from multiple cohorts of clinically annotated AML patients and report that (i) microarrays designed for coding gene expression can be repurposed to yield robust lincRNA expression data, (ii) some lincRNA genes are located in close proximity to hematopoietic coding genes and show strong expression correlations in AML, (iii) lincRNA gene expression patterns distinguish cytogenetic and molecular subtypes of AML, (iv) lincRNA signatures composed of three or four genes are independent predictors of clinical outcome and further dichotomize survival in European Leukemia Net (ELN) risk groups and (v) an analytical tool based on logistic regression analysis of quantitative PCR measurement of four lincRNA genes (LINC4) can be used to determine risk in AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , RNA, Long Noncoding/genetics , Transcriptome/genetics , Adolescent , Adult , Female , Gene Expression Profiling/methods , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis , Risk Assessment , Risk Factors , Young Adult
4.
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
5.
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
6.
Bone Marrow Transplant ; 50(12): 1513-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26301967

ABSTRACT

In patients with multiple myeloma (MM) undergoing autologous hematopoietic cell transplantation (auto-HCT), peripheral blood progenitor cells may be collected following mobilization with growth factor alone (GF) or cytotoxic chemotherapy plus GF (CC+GF). It is uncertain whether the method of mobilization affects post-transplant outcomes. We compared these mobilization strategies in a retrospective analysis of 968 patients with MM from the Center for International Blood and Marrow Transplant Research database who received an auto-HCT in the US and Canada between 2007 and 2012. The kinetics of neutrophil engraftment (⩾0.5 × 10(9)/L) was similar between groups (13 vs 13 days, P=0.69) while platelet engraftment (⩾20 × 10(9)/L) was slightly faster with CC+GF (19 vs 18 days, P=0.006). Adjusted 3-year PFS was 43% (95% confidence interval (CI) 38-48) in GF and 40% (95% CI 35-45) in CC+GF, P=0.33. Adjusted 3-year OS was 82% (95% CI 78-86) vs 80% (95% CI 75-84), P=0.43 and adjusted 5-year OS was 62% (95% CI 54-68) vs 60% (95% CI 52-67), P=0.76, for GF and CC+GF, respectively. We conclude that MM patients undergoing auto-HCT have similar outcomes irrespective of the method of mobilization and found no evidence that the addition of chemotherapy to mobilization contributes to disease control.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/blood , Multiple Myeloma/therapy , Adolescent , Adult , Aged , Autografts , Disease-Free Survival , Female , Humans , Leukocyte Count , Male , Middle Aged , Platelet Count , Prospective Studies , Recovery of Function , Survival Rate
8.
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
9.
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
10.
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
11.
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
12.
Intern Med J ; 43(2): 156-61, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22757772

ABSTRACT

BACKGROUND: Multiple myeloma is incurable despite the advance of autologous stem cell transplant (ASCT) and novel agents (thalidomide, bortezomib, lenalidomide). The role of ASCT as salvage therapy in relapsed myeloma remains unclear. AIM: To identify and refine the predictors of survival following salvage ASCT for relapsed multiple myeloma, so that they can be applied clinically for patient selection. METHODS: Retrospective review of patients treated salvage ASCT for relapsed myeloma at our centre from 1992 to 2011. RESULTS: Following an initial ASCT at diagnosis, 30 patients underwent salvage ASCT for subsequent relapse, with the median time to first relapse/progression being 30.2 months. All patients received reinduction, then melphalan-based conditioning with salvage ASCT. Non-relapse mortality at 100 days following salvage ASCT was 3%. The median overall survival and progression-free survival following salvage ASCT were 45 and 22 months respectively. The progression-free interval (PFI) after initial ASCT predicted survival outcomes in a time-dependent manner. With PFI following initial ASCT of <18, 18-36 and ≥36 months, the median progression-free survival following salvage ASCT was 4.2, 13.8 and 49.1 months respectively (P < 0.0001). The median overall survival was 10.7, 30.9 and 86.1 months respectively (P < 0.0001). CONCLUSIONS: Salvage ASCT is an effective and safe treatment option in selected patients and should be considered in patients relapsing ≥36 months after their initial ASCT. The time-dependent relationship between PFI and salvage ASCT outcome is important when stratifying patient groups who may benefit from this procedure.


Subject(s)
Multiple Myeloma/epidemiology , Multiple Myeloma/surgery , Salvage Therapy/methods , Stem Cell Transplantation/methods , Adult , Aged , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Retrospective Studies , Secondary Prevention , Transplantation, Autologous
14.
Intern Med J ; 42(4): 450-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22498118

ABSTRACT

Despite the absence of a robust evidence base, there is growing consensus that effective treatment of iron overload leads to decreased morbidity and premature mortality in patients with good prognosis myelodysplastic syndromes (MDSs). Furthermore, new treatment modalities, including disease-modifying therapies (lenalidamide and azacytidine) and reduced intensity conditioning therapies for allogeneic blood stem cell transplants, are offering the prospect of longer survival for patients with traditionally less favourable prognosis MDS, who might also benefit from iron chelation. This article proposes assessment of patients with MDS and related bone marrow failure syndromes to determine suitability for iron chelation. Iron chelation therapy options and monitoring are discussed.


Subject(s)
Chelation Therapy/methods , Hemoglobinuria, Paroxysmal/drug therapy , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Iron/blood , Myelodysplastic Syndromes/drug therapy , Anemia, Aplastic , Bone Marrow Diseases , Bone Marrow Failure Disorders , Hemoglobinuria, Paroxysmal/blood , Humans , Iron Overload/blood , Myelodysplastic Syndromes/blood , Treatment Outcome
15.
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
16.
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
17.
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
18.
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
19.
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
20.
Intern Med J ; 41(8): 588-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21831119

ABSTRACT

Mobilization and collection of haemopoietic stem and progenitor cells (HSPC) is the cornerstone of autologous and allogeneic stem cell transplantation for a wide variety of haematological and some non-haematological malignancies. Centres providing this service face the challenge of optimizing the likelihood of successful collection of transplantable doses of cells, while maximizing the efficiency of the apheresis unit and minimizing the risk of toxicity as well as mobilization failure. Recent developments in the understanding of the molecular mechanisms of mobilization have led to the emergence of novel strategies for HSPC mobilization, which may assist in meeting these imperatives. The task for clinicians is how to incorporate the use of these strategies into practice, in the light of emerging evidence for efficacy and safety of these agents. Herein, the literature is reviewed, and a proposed algorithm for HSPC mobilization is presented.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Animals , Blood Component Removal/methods , Blood Component Removal/standards , Bone Marrow Cells/physiology , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/standards , Hematopoietic Stem Cell Transplantation/standards , Hematopoietic Stem Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL