Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Bone Marrow Transplant ; 52(2): 270-278, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27991895

ABSTRACT

Patients with prior invasive fungal infection (IFI) increasingly proceed to allogeneic hematopoietic cell transplantation (HSCT). However, little is known about the impact of prior IFI on survival. Patients with pre-transplant IFI (cases; n=825) were compared with controls (n=10247). A subset analysis assessed outcomes in leukemia patients pre- and post 2001. Cases were older with lower performance status (KPS), more advanced disease, higher likelihood of AML and having received cord blood, reduced intensity conditioning, mold-active fungal prophylaxis and more recently transplanted. Aspergillus spp. and Candida spp. were the most commonly identified pathogens. 68% of patients had primarily pulmonary involvement. Univariate and multivariable analysis demonstrated inferior PFS and overall survival (OS) for cases. At 2 years, cases had higher mortality and shorter PFS with significant increases in non-relapse mortality (NRM) but no difference in relapse. One year probability of post-HSCT IFI was 24% (cases) and 17% (control, P<0.001). The predominant cause of death was underlying malignancy; infectious death was higher in cases (13% vs 9%). In the subset analysis, patients transplanted before 2001 had increased NRM with inferior OS and PFS compared with later cases. Pre-transplant IFI is associated with lower PFS and OS after allogeneic HSCT but significant survivorship was observed. Consequently, pre-transplant IFI should not be a contraindication to allogeneic HSCT in otherwise suitable candidates. Documented pre-transplant IFI is associated with lower PFS and OS after allogeneic HSCT. However, mortality post transplant is more influenced by advanced disease status than previous IFI. Pre-transplant IFI does not appear to be a contraindication to allogeneic HSCT.


Subject(s)
Aspergillosis , Aspergillus , Candida , Candidiasis , Cord Blood Stem Cell Transplantation , Hematologic Neoplasms , Registries , Adolescent , Adult , Aged , Allografts , Aspergillosis/etiology , Aspergillosis/mortality , Aspergillosis/therapy , Candidiasis/etiology , Candidiasis/mortality , Candidiasis/therapy , Child , Child, Preschool , Disease-Free Survival , Female , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Humans , Infant , Male , Middle Aged , Survival Rate
2.
Leukemia ; 28(10): 1960-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24577530

ABSTRACT

Identification of agents that target human leukemia stem cells is an important consideration for the development of new therapies. The present study demonstrates that rocaglamide and silvestrol, closely related natural products from the flavagline class of compounds, are able to preferentially kill functionally defined leukemia stem cells, while sparing normal stem and progenitor cells. In addition to efficacy as single agents, flavaglines sensitize leukemia cells to several anticancer compounds, including front-line chemotherapeutic drugs used to treat leukemia patients. Mechanistic studies indicate that flavaglines strongly inhibit protein synthesis, leading to the reduction of short-lived antiapoptotic proteins. Notably though, treatment with flavaglines, alone or in combination with other drugs, yields a much stronger cytotoxic activity toward leukemia cells than the translational inhibitor temsirolimus. These results indicate that the underlying cell death mechanism of flavaglines is more complex than simply inhibiting general protein translation. Global gene expression profiling and cell biological assays identified Myc inhibition and the disruption of mitochondrial integrity to be features of flavaglines, which we propose contribute to their efficacy in targeting leukemia cells. Taken together, these findings indicate that rocaglamide and silvestrol are distinct from clinically available translational inhibitors and represent promising candidates for the treatment of leukemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzofurans/therapeutic use , Leukemia/drug therapy , Neoplastic Stem Cells/drug effects , Triterpenes/therapeutic use , Animals , Antigens, CD34/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Leukocytes, Mononuclear/cytology , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/cytology , Phenotype , Reactive Oxygen Species/metabolism , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Stem Cells/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
3.
Leukemia ; 28(3): 658-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23989431

ABSTRACT

The efficacy of reduced intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (HCT) for Philadelphia chromosome positive (Ph+) acute lymphoblastic leukemia (ALL) is uncertain. We analyzed 197 adults with Ph+ ALL in first complete remission; 67 patients receiving RIC were matched with 130 receiving myeloablative conditioning (MAC) for age, donor type and HCT year. Over 75% received pre-HCT tyrosine kinase inhibitors (TKIs), mostly imatinib; 39% (RIC) and 49% (MAC) were minimal residual disease (MRD)(neg) pre-HCT. At a median 4.5 years follow-up, 1-year transplant-related mortality (TRM) was lower in RIC (13%) than MAC (36%; P=0.001) while the 3-year relapse rate was 49% in RIC and 28% in MAC (P=0.058). Overall survival (OS) was similar (RIC 39% (95% confidence interval (CI) 27-52) vs 35% (95% CI 27-44); P=0.62). Patients MRD(pos) pre-HCT had higher risk of relapse with RIC vs MAC (hazard ratio (HR) 1.97; P=0.026). However, patients receiving pre-HCT TKI in combination with MRD negativity pre-RIC HCT had superior OS (55%) compared with a similar MRD population after MAC (33%; P=0.0042). In multivariate analysis, RIC lowered TRM (HR 0.6; P=0.057), but absence of pre-HCT TKI (HR 1.88; P=0.018), RIC (HR 1.891; P=0.054) and pre-HCT MRD(pos) (HR 1.6; P=0.070) increased relapse risk. RIC is a valid alternative strategy for Ph+ ALL patients ineligible for MAC and MRD(neg) status is preferred pre-HCT.


Subject(s)
Bone Marrow Transplantation , Neoplasm, Residual , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Protein-Tyrosine Kinases/antagonists & inhibitors , Remission Induction , Survival Rate , Transplantation Conditioning , Adult , Animals , Female , Guinea Pigs , Humans , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transplantation, Homologous , Young Adult
4.
Bone Marrow Transplant ; 48(11): 1444-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23749109

ABSTRACT

Chemotherapy plus G-CSF (C+G) and G-CSF alone are two of the most common methods used to mobilize CD34(+) cells for autologous hematopoietic SCT (AHSCT). In order to compare and determine the real-world outcomes and costs of these strategies, we performed a retrospective study of 226 consecutive patients at 11 medical centers (64 lymphoma, 162 multiple myeloma), of whom 55% of lymphoma patients and 66% of myeloma patients received C+G. Patients with C+G yielded more CD34(+) cells/day than those with G-CSF alone (lymphoma: average 5.51 × 10(6) cells/kg on day 1 vs 2.92 × 10(6) cells/kg, P=0.0231; myeloma: 4.16 × 10(6) vs 3.69 × 10(6) cells/kg, P<0.00001) and required fewer days of apheresis (lymphoma: average 2.11 vs 2.96 days, P=0.012; myeloma: 2.02 vs 2.83 days, P=0.0015), although nearly all patients ultimately reached the goal of 2 × 10(6) cells/kg. With the exception of higher rates of febrile neutropenia in myeloma patients with C+G (17% vs 2%, P<0.05), toxicities and other outcomes were similar. Mobilization with C+G cost significantly more (lymphoma: median $10,300 vs $7300, P<0.0001; myeloma: $8800 vs $5600, P<0.0001), although re-mobilization adds $6700 for drugs alone. Our results suggest that although both C+G and G-CSF alone are effective mobilization strategies, C+G may be more cost-effective for patients at high risk of insufficient mobilization.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/economics , Granulocyte Colony-Stimulating Factor/economics , Hematopoietic Stem Cell Mobilization/economics , Hematopoietic Stem Cell Transplantation/economics , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Middle Aged , Retrospective Studies , Rituximab , Transplantation, Autologous/economics , Transplantation, Autologous/methods , Treatment Outcome , Young Adult
5.
Bone Marrow Transplant ; 42(5): 297-310, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18660844

ABSTRACT

Stem cell transplants that follow both myeloablative and non-myeloablative conditioning regimens can result in states of mixed chimerism, which can be stable over time. With widespread availability of Y chromosome FISH in sex-mismatched transplantation and DNA-based methodologies for analysis of chimerism in other donor-recipient pairs, further insights have been gained regarding the implications of the mixed chimeric state. In transplants performed for inherited and acquired marrow failure disorders, disease status can be improved with only 10-20% donor cells, and it appears that stable mixed chimerism at that level is an acceptable outcome often leading to a state of tolerance, but an increasing level of recipient cells often precedes graft rejection. In transplants performed for malignant conditions, increasing levels of mixed chimerism may indicate disease relapse, but some cases with stable levels of mixed chimerism have been compatible with prolonged remission states. Understanding when mixed chimerism is an indication of secondary graft failure or impending graft rejection vs a state of tolerance and ongoing propensity for the establishment of a graft-vs-tumor effect is often difficult with currently available technologies and immunologic assays. The ability to understand the implication of mixed chimerism of multiple cell lineages and of varied lymphocyte subsets will remain important areas for future research to best harness the immunologic and other therapeutic benefits of allogeneic transplantation.


Subject(s)
Chromosomes, Human, Y , Graft Rejection/immunology , Graft vs Tumor Effect/immunology , Stem Cell Transplantation , Transplantation Chimera/immunology , Transplantation Tolerance , Bone Marrow Diseases/immunology , Bone Marrow Diseases/therapy , Female , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/therapy , Humans , Male , Neoplasms/immunology , Neoplasms/therapy , Transplantation, Homologous
8.
Bone Marrow Transplant ; 36(9): 747-55, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16044140

ABSTRACT

ABO blood group antigens, of great importance in transplantation and transfusion, are present on virtually all cells, as well as in soluble form in plasma and body fluids. Naturally occurring plasma IgM and IgG antibodies against these antigens are ubiquitous. Nonetheless, the ABO blood group system is widely ignored by many transfusion services, except for purposes of red cell transfusion. We implemented a policy of transfusing only ABO identical platelets and red cells in patients undergoing stem cell transplantation or treatment for hematologic malignancies. Major bleeding episodes have occurred in about 5% of patients undergoing induction therapy for acute leukemia as compared with 15-20% in the literature. Overall survival times appear to be superior to that in historical cohorts. In 2002-2004, treatment-related mortality at 100 days in our Blood and Marrow Transplant Unit was 0.7% for autologous transplants (n=148), 13% for sibling allogeneic transplants (n=110), and 24% (n=62) for matched unrelated allogeneic transplants, suggesting that our approach is safe. We speculate that more rigorous efforts on the part of transfusion services to provide ABO identical blood components, and to remove incompatible supernatant plasma, when necessary, might yield reduced morbidity and mortality in patients undergoing stem cell transplantation.


Subject(s)
ABO Blood-Group System , Bone Marrow Transplantation , Erythrocyte Transfusion , Hemorrhage/therapy , Leukemia/therapy , Platelet Transfusion , Blood Grouping and Crossmatching/methods , Bone Marrow Transplantation/methods , Bone Marrow Transplantation/mortality , Erythrocyte Transfusion/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/mortality , Hemorrhage/etiology , Hemorrhage/mortality , Histocompatibility Testing/methods , Humans , Leukemia/complications , Leukemia/mortality , Platelet Transfusion/methods , Transplantation, Homologous
9.
Bone Marrow Transplant ; 35(2): 171-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15531901

ABSTRACT

Patients with sickle cell disease (N = 3) and thalassemia (N = 1) with high-risk features received hematopoietic stem cell transplantations (HCT) to induce stable (full or partial) donor engraftment. Patients were 9-30 years of age. Fludarabine, rabbit anti-thymocyte globulin (ATG), and 200 cGy total body irradiation were administered pre-transplant. Patients received bone marrow (N = 3) or peripheral blood stem cells (N = 1) from HLA-identical siblings, followed by mycophenolate mofetil and cyclosporine for post-grafting immunosuppression. Significant lymphopenia, but only moderate neutropenia and thrombocytopenia developed post transplant. No grade IV nonhematological toxicities or acute graft-versus-host disease (GVHD) were observed. At 3 months after transplantation, three of four patients had evidence of donor myeloid chimerism (range, 15-100%). However, after post transplant immunosuppression was discontinued, graft rejection occurred in all but one patient. This patient is now doing well 27 months post transplant with full donor engraftment. One patient died after a second transplant, and another patient experienced a stroke as her graft was being rejected. These results suggest that stable donor engraftment after nonmyeloablative HCT is difficult to achieve among immunocompetent patients with hemoglobinopathies and that new approaches will need to be developed before wider application of this transplantation method for hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell/therapy , Hematopoietic Stem Cell Transplantation/methods , Thalassemia/therapy , Vidarabine/analogs & derivatives , Adolescent , Adult , Anemia, Sickle Cell/complications , Antilymphocyte Serum/administration & dosage , Child , Combined Modality Therapy/adverse effects , Female , Graft Rejection , Graft vs Host Disease , Hemoglobinopathies/therapy , Humans , Immunosuppressive Agents/therapeutic use , Infections , Male , Pancytopenia , Transfusion Reaction , Transplantation Chimera , Transplantation, Homologous , Vidarabine/administration & dosage , Whole-Body Irradiation
10.
Semin Oncol ; 31(6 Suppl 18): 59-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15726525

ABSTRACT

We and others have previously shown that the use of amifostine (Ethyol; MedImmune Inc, Gaithersburg, MD) can ameliorate certain regimen-related toxicities of high-dose melphalan (HD-MEL) in the autologous hematopoietic stem cell transplant setting. Our recent experience indicated that the maximum tolerated dose of HD-MEL plus autologous hematopoietic stem cell transplant could be increased from approximately 200 mg/m2 to at least 280 mg/m2 with amifostine. Although a dose-limiting toxicity was not clearly identified, atrial fibrillation was noted in several patients. Phase II trials using this regimen have been reported in lymphoma and myeloma. Nonetheless, it is unlikely that single agent therapy, regardless of dose, will be highly curative in advanced hematologic malignancy. Thus, we used amifostine to permit dose escalation of HD-MEL within the BEAM (BCNU/etoposide/arabinosylcytosine/HD-MEL) combination chemotherapy regimen before autologous hematopoietic stem cell transplant in selected patients with lymphoma. Patient entry at the starting dose (ie, HD-MEL 140 mg/m2) has been completed without the development of severe regimen-related toxicities. This trial is ongoing.


Subject(s)
Amifostine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carmustine/therapeutic use , Cytarabine/therapeutic use , Etoposide/therapeutic use , Hematopoietic Stem Cell Transplantation , Lymphoma/therapy , Melphalan/therapeutic use , Adult , Amifostine/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Carmustine/toxicity , Combined Modality Therapy , Cytarabine/toxicity , Cytoprotection , Etoposide/toxicity , Humans , Melphalan/toxicity , Middle Aged , Radiation-Protective Agents/adverse effects , Radiation-Protective Agents/therapeutic use , Transplantation, Autologous
11.
Leukemia ; 17(9): 1806-12, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970780

ABSTRACT

Patients with acute myelogenous leukemia or myelodysplastic syndrome may respond to farnesyl transferase inhibitors (FTIs) with partial or complete response rates noted in about 30% of such patients. FTIs prevent the attachment of a lipid farnesyl moiety to dependent proteins prior to their insertion into the plasma membrane and thereby prevent activity of these prenylation-dependent proteins, but their mechanism of tumor suppression remains unknown. Many patients receiving FTIs do experience myelosuppression. In this work, the in vitro effects of the FTI, R115777 on normal and leukemic hematopoiesis have been examined as have its effects on apoptosis induction and cell cycle profile in both leukemic blasts and normal CD34+ cells. R115777 was inhibitory to normal CD34+ cell proliferation and to leukemic blast cells, but did not affect long-term culture initiating cell frequency nor NOD-SCID reconstituting capacity. No induction of apoptosis or cell cycle changes were noted in AML blasts. These data suggest that myelosuppression with R115777 occurs largely at the intermediate to late progenitor stage of hematopoiesis and that cyclic use might avoid long-term marrow suppression.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hematopoiesis/drug effects , Leukemia/drug therapy , Quinolones/pharmacology , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Colony-Forming Units Assay , Farnesyltranstransferase , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Poly(ADP-ribose) Polymerases/metabolism , Time Factors , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/transplantation
12.
Bone Marrow Transplant ; 32(3): 293-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12858201

ABSTRACT

The impact of peripheral blood stem cell transplantation (PBSCT) on survival relative to bone marrow transplantation (BMT) remains poorly defined. Several randomized controlled trials (RCTs) comparing HLA-matched related PBSC- and BMT for patients with hematologic malignancies have been published, yielding differing results. We conducted a meta-analysis of published RCTs to more precisely estimate the effect of PBSCT on survival. Seven trials that assessed survival were identified and included in our analysis. Using a fixed effects model, and combining the results of all seven trials, the summary odds ratio for mortality after PBSCT was 0.81 (95% CI, 0.62-1.05) when compared to BMT. Subgroup analysis revealed no association between the median PBSCT 34+ cell dose and relative risk for morality after PBSCT. However, there was an association between the proportion of patients enrolled with advanced-stage disease and the summary odds ratio for mortality. The pooled estimate was 0.64 for studies where patients with intermediate/advanced disease comprised at least 25% of enrollment, and was 1.07 for the studies enrolling a smaller proportion. This finding substantiates results from previously published studies that have demonstrated a survival advantage with PBSCT limited to patients with advanced disease.


Subject(s)
Bone Marrow Transplantation/mortality , Hematologic Neoplasms/therapy , Histocompatibility , Peripheral Blood Stem Cell Transplantation/mortality , Adult , Antigens, CD34 , Cell Count , Disease Progression , Female , HLA Antigens , Hematologic Neoplasms/mortality , Humans , Male , Middle Aged , Odds Ratio , Randomized Controlled Trials as Topic/statistics & numerical data , Risk Factors , Survival Analysis , Transplantation, Homologous
13.
Blood Cells Mol Dis ; 28(3): 315-21, 2002.
Article in English | MEDLINE | ID: mdl-12367578

ABSTRACT

To find a parameter to predict the quality of collected mobilized CD34+ blood as hemopoietic reconstituting cells, the ratio of CFU-GM to CD34+ cells was examined. One hundred six consecutive patients who underwent blood stem cell transplantation at the University of Rochester from 01/01/99 to 12/31/99 were examined retrospectively for the number of days to reach an absolute neutrophil count of 500 or 1000 cells/microl and an absolute platelet count of 20,000 or 50,000 cells/microl without transfusion support as measures of engraftment. Linear regression analyses were conducted to determine factors influencing engraftment. The number of CD34+ cells/kg and CFU-GM/kg correlated highly with the number of nucleated blood cells/kg. In this population, in which 90% of patients received >2 x 10(6) CD34+ cells/kg, neither the number of CD34+ cells/kg nor the number of CFU-GM/kg correlated with the time to engraftment as judged by neutrophil or platelet levels. In contrast, the lower the ratio of CFU-GM to CD34+ cells, the more rapid the reconstitution of platelets to 20,000/microl (P = 0.03) and 50,000/microl (P = 0.02). Thus, a lower ratio of the CFU-GM/CD34+ appended to reflect a greater number of hematopoietic reconstituting cells in the blood cell collection. The CFU-GM/CD34+ ratio is an apparent predictor of earlier platelet engraftment, suggesting that the ratio reflects the engraftment potential of mobilized donor progenitor cells.


Subject(s)
Graft Survival , Hematopoietic Stem Cell Mobilization/standards , Hematopoietic Stem Cells/cytology , Peripheral Blood Stem Cell Transplantation/standards , Adolescent , Adult , Aged , Antigens, CD34/analysis , Blood Cell Count , Child , Child, Preschool , Cohort Studies , Hematopoiesis , Humans , Infant , Kinetics , Middle Aged , Neoplasms/therapy , Prognosis , Regression Analysis , Retrospective Studies
14.
J Hematother Stem Cell Res ; 10(5): 643-55, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11672510

ABSTRACT

Ultrastructural studies of marrow and examination of the in vivo processes of stem cell homing and mobilization show that multipotential hematopoietic progenitors are able to traverse endothelial cells. The regulation of this process by various classes of chemokines was studied in this report, using an in vitro model of transendothelial migration. Human umbilical vein endothelial cells (HUVECs) or bone marrow-derived endothelial cells (BMECs) were grown to confluence on 3-microm microporous membrane inserts and placed in 24-well culture plates. CD34(+) cells isolated from normal volunteer donor marrow by immunoadsorption or magnetic bead selection techniques were added to the inserts and various individual chemokines were added to the lower chamber of the culture plates in serum-free conditions. After 24 h, the percentage of transmigrated cells was determined. A mean of 8.5% of unfractionated marrow CD34(+) populations migrated, and all chemokines tested, with the exception of macrophage inflammatory protein-1alpha (MIP-1alpha), had some positive effect on this migration. The greatest effects were seen with stroma-derived factor-1alpha (SDF-1alpha) and stroma-derived factor-1beta (SDF-1beta), with lesser effects noted for other chemokines and cytokines. When the CD34(+) population was subselected for expression of CD38, a greater fraction of the CD38(-) cells migrated as compared to the CD38(+) fraction. CD34(+) cells isolated from mobilized peripheral blood and cord blood also migrated in response to chemokines. Chemokines of the CC, CXC, and CX(3)C classes as well as other hematopoietic cytokines may modulate the process of stem cell transmigration of endothelial cells. Further understanding of this process may help elucidate the mechanism of stem cell mobilization and homing.


Subject(s)
Antigens, CD34/analysis , Chemokines/pharmacology , Hematopoietic Stem Cells/drug effects , Antibodies/pharmacology , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokines, CC/pharmacology , Chemokines, CX3C/pharmacology , Chemokines, CXC/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Interleukin-8/pharmacology , Receptors, CXCR4/immunology
15.
Am J Hematol ; 66(1): 23-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11426487

ABSTRACT

A Phase II study of GM-CSF with intermediate-dose cytarabine and mitoxantrone was conducted in patients with high-risk myelodysplastic syndrome. It was designed to evaluate if priming with growth factor could increase the efficiency of chemotherapy. In this older population only two of 10 patients achieved a bone marrow CR, including one patient whose leukemic blasts had an "S" phase increase of 2.55x at 48 hr. Unexpected hepatotoxicity was noted. This regimen cannot be recommended for this elderly population of patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Myelodysplastic Syndromes/drug therapy , Aged , Anemia, Refractory, with Excess of Blasts/drug therapy , Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Refractory, with Excess of Blasts/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Marrow/pathology , Chemical and Drug Induced Liver Injury/etiology , Cytarabine/administration & dosage , Cytarabine/adverse effects , DNA Replication/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Hyperbilirubinemia/chemically induced , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Male , Middle Aged , Mitoxantrone/administration & dosage , Mitoxantrone/adverse effects , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Pancytopenia/chemically induced , Pancytopenia/drug therapy , Pilot Projects , Recombinant Proteins , Remission Induction , S Phase/drug effects , Treatment Failure
16.
Blood Cells Mol Dis ; 26(4): 261-75, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11042027

ABSTRACT

ABSTRACT Macrophage inflammatory protein-1alpha (MIP-1alpha) is a C-C chemokine which has antiproliferative effects on early hematopoietic progenitors and stimulatory effects on later progenitors. It also possesses chemotactic and activating properties for monocytes, macrophages, and T-cells. CD34+ progenitors isolated utilizing an avidin-biotin immunoadsorption column produced significant amounts of MIP-1alpha from 24 h onward when cultured in medium with 10% fetal calf serum (>200 pg/ml). Such production persisted through 96 h of culture and was greater when such progenitors were cocultured with a preformed marrow stromal layer (4000 pg/ml at 24 h). The production of MIP-1alpha declined over time of coculture with stromal layers, and stromal layers themselves produced minimal MIP-1alpha as detected by ELISA: <100 pg/ml. In contrast, CD34+ cells isolated by flow cytometry or by magnetic bead adsorption produced minimal MIP-1alpha (0-30 pg/ml). MIP-1alpha production also increased when cells isolated by these two methods were cocultured with stromal layers. The difference in MIP-1alpha production could not be accounted for by differences in purity of the CD34+ population between isolation methods nor on the basis of monocytic or lymphocytic contamination as assessed by the presence of CD14 or CD3 positive cells. CD34+ cells isolated by immune adsorption had increased expression of endothelial and mesenchymal associated antigens, however, suggesting that this subpopulation might account for the MIP-1alpha production observed. Freshly isolated CD34+ cells expressed MIP-1alpha message as assessed by RT-PCR and by in situ hybridization. Coculture of CD34+ cells isolated by any means with stromal cells increased transforming growth factor-beta (TGF-beta) production, in this case by the stromal layer itself. Both MIP-1alpha and TGF-beta have been found to influence cell cycle status and proliferation status of early hematopoietic progenitors, and both have potential effects on accessory cell function. These studies indicate that progenitor-stromal cell interactions may influence local cytokine output, thus potentially influencing progenitor cycling status and accessory cell activation. The method of isolation of CD34+ progenitors may influence secretion of certain cytokines and chemokines.


Subject(s)
Antigens, CD34/metabolism , Macrophage Inflammatory Proteins/biosynthesis , Stem Cells/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta/biosynthesis , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Communication , Cell Separation/methods , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Coculture Techniques , Gene Expression Regulation , Humans , Leukemia/pathology , Macrophage Inflammatory Proteins/pharmacology , RNA/genetics , RNA/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stromal Cells/cytology , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
17.
Bone Marrow Transplant ; 25(5): 489-94, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10713624

ABSTRACT

Marrow-ablative chemo-radiotherapy followed by hematopoietic stem cell rescue from an allogeneic source improves outcomes for children with high-risk acute leukemia. The first effective pre-transplant preparative regimens consisted of high-dose cyclophosphamide (CY) and total body irradiation (TBI). Subsequent attempts have been made to improve leukemia-free survival, by adding other chemotherapy agents to these agents. In previous clinical studies of total body irradiation, etoposide, cyclophosphamide (TBI-VP-16-Cy) in adult allogeneic bone marrow transplantation, there has been a high incidence of severe regimen-related toxicity. In this study, we investigated the safety and efficacy of this combination in 41 children who received TBI (12-14 Gy), VP-16 (30 mg/kg), and CY (60 mg/kg x 2) and then either matched sibling or alternative donor transplants for acute leukemia. There was only one case of fatal regimen-related toxicity. The estimated 3-year event-free survival for patients with early or intermediate stage disease was 68% (53-88%). The estimated event-free survival of patients with advanced disease was 17% (5-59%). TBI-VP16-CY is safe in pediatric transplantation, and it has good efficacy for transplant recipients with less advanced disease. Bone Marrow Transplantation (2000) 25, 489-494.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Transplantation , Leukemia/therapy , Whole-Body Irradiation , Acute Disease , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/toxicity , Blood Grouping and Crossmatching , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Child , Child, Preschool , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/toxicity , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Evaluation , Drug-Related Side Effects and Adverse Reactions , Etoposide/administration & dosage , Etoposide/toxicity , Graft Survival/drug effects , Graft vs Host Disease , Humans , Infant , Myeloablative Agonists/therapeutic use , Myeloablative Agonists/toxicity , Nuclear Family , Recurrence , Stomatitis/chemically induced , Survival Rate , Transplantation, Homologous
18.
Bone Marrow Transplant ; 24(10): 1057-63, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10578155

ABSTRACT

Unmanipulated autologous bone marrow transplant (ABMT) offers patients with chronic myelogenous leukemia (CML) a long-term survival of 10%, at best. Immunotherapy has a role in the myeloid leukemias, and there is increasing evidence that of all hematopoietic neoplasms, CML may be the most susceptible to immune regulation. Roquinimex is known to enhance T cell, NK cell and macrophage activity. A phase II study was initiated in March 1992 to evaluate the role of roquinimex in Ph chromosome-positive CML post ABMT. Patients were conditioned with busulfan/ cyclophosphamide followed by reinfusion of unmanipulated Ph-positive bone marrow stem cells (>1 x 108 NBC/kg). When engraftment of neutrophils (ANC) reached 100/microl, patients received oral roquinimex twice weekly, escalating to a maximal dose of 0.2 mg/kg in 2 weeks. Seventeen patients have entered the study; 11 in first chronic phase (CP1); two in second chronic phase (CP2) and four in accelerated phase (AP). All required significant myelosuppressive therapy prior to ABMT to maintain stable blood counts and most had also received prior interferon therapy. All patients survived the transplant. Subsequent toxicity consisted mainly of musculoskeletal aches and peripheral edema. Additionally, specific skin changes were observed including graft-versus-host-like disease and eccrine sweat gland necrosis. Eight out of 17 patients are alive 28-60 months post ABMT. Of the nine patients who died, two were in CP2 and three in AP. All patients in CP1 went into a complete hematological remission post ABMT and seven of the 11 patients had at least a major cytogenetic response (greater than 65% Ph-negative metaphases) at 1 year or beyond and four of the 11 patients had a complete cytogenetic response at 2 years or beyond. Cytogenetic response post transplant often developed over time and did not simply represent post ABMT engraftment with Ph-negative cells. The clinical and cytogenetic data in these patients are encouraging and suggest that roquinimex may have significant activity when given post ABMT to patients with Ph-positive CML.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation , Hydroxyquinolines/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Combined Modality Therapy , Humans , Hydroxyquinolines/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Middle Aged , Patient Selection , Survival Analysis , Transplantation, Autologous
19.
J Clin Oncol ; 17(8): 2446-53, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10561308

ABSTRACT

PURPOSE: To identify predictors of oral mucositis and gastrointestinal toxicity after high-dose therapy. PATIENTS AND METHODS: Mucositis and gastrointestinal toxicity were prospectively evaluated in 202 recipients of high-dose therapy and autologous or allogeneic stem-cell rescue. Of 10 outcome variables, three were selected as end points: the peak value for the University of Nebraska Oral Assessment Score (MUCPEAK), the duration of parenteral nutritional support, and the peak daily output of diarrhea. Potential covariates included patient age, sex, diagnosis, treatment protocol, transplantation type, stem-cell source, and rate of neutrophil recovery. The three selected end points were also examined for correlation with blood infections and transplant-related mortality. RESULTS: A diagnosis of leukemia, use of total body irradiation, allogeneic transplantation, and delayed neutrophil recovery were associated with increased oral mucositis and longer parenteral nutritional support. No factors were associated with diarrhea. Also, moderate to severe oral mucositis (MUCPEAK > or = 18 on a scale of 8 to 24) was correlated with blood infections and transplant-related mortality: 60% of patients with MUCPEAK > or = 18 had positive blood cultures versus 30% of patients with MUCPEAK less than 18 (P =.001); 24% of patients with MUCPEAK > or = 8 died during the transplantation procedure versus 4% of patients with MUCPEAK less than 18 (P =.001). CONCLUSION: Gastrointestinal toxicity is a major cause of transplant-related morbidity and mortality, emphasizing the need for corrective strategies. The peak oral mucositis score and the duration of parenteral nutritional support are useful indices of gastrointestinal toxicity because these end points are correlated with clinically significant events, including blood infections and treatment-related mortality.


Subject(s)
Antineoplastic Agents/adverse effects , Leukemia/complications , Leukemia/therapy , Mouth Mucosa/drug effects , Parenteral Nutrition , Stem Cell Transplantation , Stomatitis/etiology , Adolescent , Adult , Analysis of Variance , Antineoplastic Agents/therapeutic use , Child , Databases, Factual , Diarrhea/etiology , Female , Humans , Leukemia/mortality , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Severity of Illness Index , Stomatitis/chemically induced , Stomatitis/classification
20.
Cancer Treat Res ; 99: 195-226, 1999.
Article in English | MEDLINE | ID: mdl-9891868

ABSTRACT

Over the past decade, with the advent of hematopoietic growth factors, major strides have been made and multiple studies have attempted to define the use of these cytokines in acute leukemia. It is perhaps disappointing that, after so many studies, so many questions remain. Nevertheless, the role of cytokines in induction therapy seems to be established, although questions remain around the issue of priming therapy. Intriguing data regarding the potential for enhancing antimicrobial function should hopefully be resolved over the next few years. What is perhaps most reassuring is that the issue of safety, which for a considerable period of time precluded the development of clinical trials in acute leukemia, has been firmly laid to rest. The use of growth factors to protect normal stem cells during treatment of leukemia and to induce leukemic cell differentiation has not yet been the subject of many clinical trials. Also, growth factors are likely targets for the interruption of autocrine leukemic blast or progenitor cell growth, but again, few clinical observations are published. With the ongoing cloning of new growth factors active both in normal hematopoiesis and in leukemogenesis, the role of growth factor use in the treatment of AML will likely be the basis for much future preclinical and clinical activity.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Leukemia, Myeloid, Acute/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Cell Differentiation/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Neutropenia/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL
...