Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Leukemia ; 24(7): 1350-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20508619

ABSTRACT

This phase 1 study (Clinicaltrials.gov: NCT00507442) was conducted to determine the maximum tolerated dose (MTD) of cyclophosphamide in combination with bortezomib, dexamethasone and lenalidomide (VDCR) and to assess the safety and efficacy of this combination in untreated multiple myeloma patients. Cohorts of three to six patients received a cyclophosphamide dosage of 100, 200, 300, 400 or 500 mg/m(2) (on days 1 and 8) plus bortezomib 1.3 mg/m(2) (on days 1, 4, 8 and 11), dexamethasone 40 mg (on days 1, 8 and 15) and lenalidomide 15 mg (on days 1-14), for eight 21-day induction cycles, followed by four 42-day maintenance cycles (bortezomib 1.3 mg/m(2), on days 1, 8, 15 and 22). The MTD was the cyclophosphamide dose below which more than one of six patients experienced a dose-limiting toxicity (DLT). Twenty-five patients were treated. Two DLTs were seen, of grade 4 febrile neutropenia (cyclophosphamide 400 mg/m(2)) and grade 4 herpes zoster despite anti-viral prophylaxis (cyclophosphamide 500 mg/m(2)). No cumulative hematological toxicity or thromboembolic episodes were reported. The overall response rate was 96%, including 20% stringent complete response (CR), 40% CR/near-complete response and 68% >or=very good partial response. VDCR is well tolerated and highly active in this population. No MTD was reached; the recommended phase 2 cyclophosphamide dose in VDCR is 500 mg/m(2), which was the highest dose tested.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/diagnosis , Aged , Boronic Acids/administration & dosage , Bortezomib , Cohort Studies , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Female , Follow-Up Studies , Humans , Lenalidomide , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Pyrazines/administration & dosage , Remission Induction , Survival Rate , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome
2.
Leuk Lymphoma ; 34(1-2): 71-84, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10350334

ABSTRACT

The use of platelet transfusions has increased greatly in the past decade and is likely to continue to escalate because of the risks of thrombocytopenia in patients receiving dose-intensive cancer chemotherapy, the increased use of hematopoietic progenitor cell transplantation, and the prevalence of human immunodeficiency virus infection. Despite marked advances in procedures for ensuring the safety of platelets, including intensive donor screening, infectious disease marker testing, and increased use of leukodepletion techniques, platelet transfusions carry a significant risk for immunologic disorders and transmission of bacterial, viral, and perhaps other diseases and can entail a very high cost. In addition, thrombocytopenia has the potential to interfere with delivery of chemotherapy on schedule and at the planned doses, thus potentially compromising treatment outcome. The limitations of platelet transfusions have prompted the development of agents with the potential to stimulate platelet production and thus reduce or eliminate the need for transfusions. Two such agents, interleukin-11 (IL-11) and thrombopoietin (TPO), have demonstrated promise in clinical trials. In November, 1997, IL-11 received FDA approval for the prevention of severe thrombocytopenia in high risk patients receiving myelosuppressive chemotherapy. Thrombopoietic growth factors have the potential to greatly simplify and increase the safety of transfusion medicine.


Subject(s)
Platelet Transfusion , Animals , Health Care Costs , Humans , Platelet Transfusion/adverse effects , Platelet Transfusion/economics , Platelet Transfusion/methods , Platelet Transfusion/trends , Risk Factors
3.
Blood ; 93(5): 1487-95, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10029575

ABSTRACT

Multiple myeloma (MM) cells express idiotypic proteins and other tumor-associated antigens which make them ideal targets for novel immunotherapeutic approaches. However, recent reports show the presence of Kaposi's sarcoma herpesvirus (KSHV) gene sequences in bone marrow dendritic cells (BMDCs) in MM, raising concerns regarding their antigen-presenting cell (APC) function. In the present study, we sought to identify the ideal source of DCs from MM patients for use in vaccination approaches. We compared the relative frequency, phenotype, and function of BMDCs or peripheral blood dendritic cells (PBDCs) from MM patients versus normal donors. DCs were derived by culture of mononuclear cells in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4. The yield as well as the pattern and intensity of Ag (HLA-DR, CD40, CD54, CD80, and CD86) expression were equivalent on DCs from BM or PB of MM patients versus normal donors. Comparison of PBDCs versus BMDCs showed higher surface expression of HLA-DR (P =.01), CD86 (P =. 0003), and CD14 (P =.04) on PBDCs. APC function, assessed using an allogeneic mixed lymphocyte reaction (MLR), demonstrated equivalent T-cell proliferation triggered by MM versus normal DCs. Moreover, no differences in APC function were noted in BMDCs compared with PBDCs. Polymerase chain reaction (PCR) analysis of genomic DNA from both MM patient and normal donor DCs for the 233-bp KSHV gene sequence (KS330233) was negative, but nested PCR to yield a final product of 186 bp internal to KS330233 was positive in 16 of 18 (88.8%) MM BMDCs, 3 of 8 (37.5%) normal BMDCs, 1 of 5 (20%) MM PBDCs, and 2 of 6 (33.3%) normal donor PBDCs. Sequencing of 4 MM patient PCR products showed 96% to 98% homology to the published KSHV gene sequence, with patient specific mutations ruling out PCR artifacts or contamination. In addition, KHSV-specific viral cyclin D (open reading frame [ORF] 72) was amplified in 2 of 5 MM BMDCs, with sequencing of the ORF 72 amplicon revealing 91% and 92% homology to the KSHV viral cyclin D sequence. These sequences again demonstrated patient specific mutations, ruling out contamination. Therefore, our studies show that PB appears to be the preferred source of DCs for use in vaccination strategies due to the ready accessibility and phenotypic profile of PBDCs, as well as the comparable APC function and lower detection rate of KSHV gene sequences compared with BMDCs. Whether active KSHV infection is present and important in the pathophysiology of MM remains unclear; however, our study shows that MMDCs remain functional despite the detection of KSHV gene sequences.


Subject(s)
Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Dendritic Cells/pathology , Dendritic Cells/virology , Herpesvirus 8, Human/isolation & purification , Multiple Myeloma/pathology , Multiple Myeloma/virology , Adult , Aged , Antigens, CD/immunology , Bone Marrow Cells/immunology , DNA, Viral/analysis , DNA, Viral/genetics , Dendritic Cells/immunology , Female , Flow Cytometry , HLA-DR Antigens/immunology , Herpesvirus 8, Human/genetics , Humans , Immunophenotyping , Male , Middle Aged
4.
Cytotherapy ; 1(3): 175-82, 1999.
Article in English | MEDLINE | ID: mdl-12881173

ABSTRACT

BACKGROUND: Wide ranges i n cell recovery and purity may be observed following CD34(+) cell selection of mobilized HPC componetns. Characteristics of the mobilized HPC, associated with isolation of a high CD34(+) cell yield and purity following cell selection, have yet to be defined. METHODS: Cell number and purities were determined before and after 56 CD34(+) cell-selection procedures, performed using the CellPro Ceprate SC system from April 1997 to February 1998. HPC were collected from 28 patients with multiple myeloma, following cyclophosphamide (60mg/kg) and G-CSF (10microg/kg) mobilization. RESULTS: A medium of 47.9% (range 1.5-109.6%) CD34(+) cells were recovered in the enriched (ENR) fraction. A linear correlation existed between total CD34(+) cells in the ENR fraction and total CD34(+) cells in the START fraction (R2=0.93); there was a logarithmic correlation between CD34 ENR fraction purity and START fraction purity (R2=0.73). A START CD34(+) cell purity > 0.42% improved purity in the ENR fraction. A median of one (range one to nine) procedure was required to isolate 2 x 10 6 CD34(+) cells/kg. Three patients pretreated with alkylating agents failed to mobilized adequate numbers of HPC. DISCUSSION: Isolation of highly purified CD34(+) cell-selected components using the Ceprate SC system in dependent on the CD34(+) purity of the lekapheresis component collected. Mobilization regimens should be used to maximize CD34(+) cell purity in stem cell authografts if CD34(+) cell selection is to be performed. Similar strategies should be used to evaluate other cell-selection devices as they become available.


Subject(s)
Antigens, CD34/biosynthesis , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Multiple Myeloma/therapy , Adult , Aged , Female , Hematopoietic Stem Cells/cytology , Humans , Leukapheresis , Male , Middle Aged , Multiple Myeloma/blood , Stem Cells/cytology , Time Factors
5.
Blood ; 92(12): 4591-601, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9845525

ABSTRACT

Adenoviruses are efficient gene delivery agents for a variety of neoplasms. In the present study, we have investigated the use of adenoviruses for the delivery of the thymidine kinase (tk) gene into multiple myeloma (MM) cells. We first demonstrated that MM cell lines and MM patient cells express both adenovirus receptors as well as the DF3/MUC1 protein, thus providing a rationale for using adenoviruses to selectively deliver genes under the control of the DF3 promoter. By using an adenoviral construct containing beta-galactosidase (beta-gal) gene driven by the DF3 promoter (Ad. DF3-betagal), we demonstrate greater than 80% transduction efficiency in OCI-My5 and RPMI 8226 MM cell lines at a multiplicity of infection of 1 to 100. Importantly, transduction with the tk gene driven by the DF3 promoter (Ad.DF3-tk) followed by treatment with 50 micromol/L ganciclovir (GCV) purged >/=6 log of contaminating OCI-My5 and RPMI 8226 MM cells within bone marrow mononuclear cells. In contrast, normal human hematopoietic progenitor cell number was unaffected under these conditions. Selectivity of DF3/MUC1 promoter was further confirmed, because Ad.DF3-betagal or Ad.DF3-tk did not transduce MUC1-negative HeLa cervical carcinoma cells. In addition, GCV treatment of Ad.DF3-tk-transduced RPMI 8226 MM cells did not induce a significant bystander effect. These findings demonstrate that transduction with Ad vectors using a tumor-selective promoter provides a highly efficient and selective approach for the ex vivo purging of MM cells.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Multiple Myeloma/genetics , Receptors, Vitronectin , Adenoviridae/metabolism , Adenovirus E2 Proteins/genetics , Cell Division/drug effects , Cell Line, Transformed , Coxsackie and Adenovirus Receptor-Like Membrane Protein , DNA, Viral/analysis , Ganciclovir/pharmacology , Gene Expression , Genes, Reporter , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/virology , Humans , Integrins/biosynthesis , Leukocytes, Mononuclear/drug effects , Mucin-1/biosynthesis , Mucin-1/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/virology , Receptors, Virus/genetics , Receptors, Virus/metabolism , Substrate Specificity , Thymidine Kinase/genetics , Transduction, Genetic , Tumor Cells, Cultured , beta-Galactosidase/analysis , beta-Galactosidase/genetics
6.
Blood ; 92(5): 1749-57, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716605

ABSTRACT

Previous studies have suggested that multiple myeloma (MM) cells express estrogen receptors (ER). In the present study, we characterized the effects of estrogen agonists and antagonists (anti-estrogens [AE]) on growth of MM cell lines and MM patient cells. In addition to antagonizing estrogen binding to ER, AE can trigger apoptosis. Hence, we also determined whether estrogens or AE altered MM cell survival. Immunoblotting showed that ER-alpha is expressed in 4 of 5 MM cell lines (ARH-77, RPMI 8226, S6B45, and U266, but not OCI-My-5 cells), as well as in freshly isolated MM cells from 3 of 3 patients. 17beta-estradiol (E2) did not significantly alter proliferation of MM cell lines or MM patient cells. In contrast, two structurally distinct AE, tamoxifen (TAM) and ICI 182,780 (ICI), significantly inhibited the proliferation of all 5 MM cell lines and MM cells from 2 of 2 patients (IC50, 2 to 4 micromol/L). Proliferation of these cell lines was also inhibited by the hydroxylated TAM derivative, 4-hydroxytamoxifen (4HTAM), although this derivative was less potent than TAM (IC50, 3 to 25 micromol/L). In contrast, the dehalogenated TAM derivative toremifene (TOR) did not inhibit MM cell proliferation. We next examined the effects of these agents on MM cell survival. TAM, ICI, and, to a lesser extent, 4HTAM and TOR triggered apoptosis in both ER-alpha-positive as well as ER-alpha-negative MM cell lines and patient MM cells, evidenced both by fluorescence-activated cell sorting (FACS) analysis using propidium iodide staining and the TUNEL assay. TAM-induced growth inhibition and apoptosis of ER-alpha-positive S6B45 MM cells was not blocked by coculture with excess E2. TAM-induced apoptosis of S6B45 MM cells was also unaffected by addition of exogenous interleukin-6. Importantly, both the inhibition of MM cell proliferation and the induction of MM cell apoptosis were achieved at concentrations of TAM (0.5 and 5.0 micromol/L) that did not significantly alter in vitro growth of normal hematopoietic progenitor cells. Similar plasma levels of TAM have been achieved using high-dose oral TAM therapy, with an acceptable toxicity profile. These studies therefore provide the rationale for trials to define the utility of AE therapy in MM.


Subject(s)
Apoptosis/drug effects , Estrogen Antagonists/pharmacology , Multiple Myeloma/pathology , Breast Neoplasms , Cell Division/drug effects , Cell Survival/drug effects , Coloring Agents , Estradiol/analogs & derivatives , Estradiol/pharmacology , Flow Cytometry , Fulvestrant , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunoblotting , Propidium , Receptors, Estrogen/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Toremifene/pharmacology , Tumor Cells, Cultured
7.
J Hematother ; 7(2): 169-74, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9597574

ABSTRACT

We tested the feasibility of sequential selection of CD34+ and CD4+ cell-enriched fractions from aliquots of five autologous leukapheresis components. CD34+ cells were selected from a median 8.0 x 10(8) mononuclear cells using the CellPro CEPRATE LC cell separation system. All cells in the CD34-depleted fraction (median 4.8 x 10(8), range 0.6-10.0 x 10(8) were then incubated with the appropriate antibodies for CD4+ cell selection and passed through a second LC column. Median target cell purities of the CD34+ cell-enriched fraction and CD4+ cell-enriched fraction were 90.5% and 86.0%, respectively. This study demonstrates that high purity CD34+ and CD4+ cell-enriched fractions can be isolated sequentially from leukapheresis components. In addition, CD8+ lymphocytes, implicated in graft-versus-host disease, were depleted in the course of both positive selection procedures. This approach could decrease the number of donor procedures by providing separate CD34(+)-enriched and CD4(+)-enriched populations for allogeneic peripheral blood progenitor cell transplantation and subsequent donor lymphocyte infusion from the same leukapheresis component.


Subject(s)
Antigens, CD34 , CD4-Positive T-Lymphocytes , Lymphocyte Depletion/methods , Cell Separation , Humans , Leukapheresis
9.
Semin Oncol ; 24(1): 141-6, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9045300

ABSTRACT

Patients with acute myeloid leukemia and acute lymphocytic leukemia receive significant numbers of blood products for hematologic support during periods of disease and treatment related cytopenias. These patients may be at increased risk for complications associated with transfusion. This article reviews the indications for platelet and red blood cell transfusion, as well as the incidence and strategies for prevention of the immunohematologic and infectious sequelae arising in the setting of multiple blood component transfusions.


Subject(s)
Erythrocyte Transfusion , Leukemia, Myeloid/blood , Platelet Transfusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Acute Disease , Erythrocyte Transfusion/adverse effects , Humans , Platelet Transfusion/adverse effects
10.
J Clin Oncol ; 15(2): 674-83, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9053493

ABSTRACT

PURPOSE: As an alternative to single-cycle cyclophosphamide, thiotepa, and carboplatin (CTCb) intensification, we evaluated the feasibility of administering one-quarter dose CTCb for four cycles with peripheral-blood progenitor-cell (PBPC) and filgrastim (granulocyte colony-stimulating factor [G-CSF]) in advanced-stage breast cancer patients. PATIENTS AND METHODS: From June 1992 to August 1993, 20 stage IIIB (n = 7) and IV (n = 13) breast cancer patients received 78 cycles of induction with doxorubicin 90 mg/m2 by intravenous (IV) bolus with G-CSF 5 microg/kg/d by subcutaneous injection (SC) repeated every 14 to 21 days for four cycles. PBPC were collected by 2-hour single-blood volume leukapheresis on 2 consecutive days at the time of hematologic recovery from each cycle of doxorubicin. Eighteen patients received 61 cycles of intensification with cyclophosphamide 1,500 mg/m2, thiotepa 125 mg/m2, and carboplatin 200 mg/m2 by IV continuous infusion with G-CSF 10 microg/kg/d SC and PBPC support repeated every 21 to 42 days for four cycles. RESULTS: Twelve of 20 patients (60%) completed all four planned cycles of doxorubicin induction followed by four cycles of one-quarter dose CTCb intensification. Statistically significantly decreases in the yield of mononuclear cells (MNC) (median slope per day, -0.032; P = .03), granulocyte-macrophage colony-forming unit (CFU-GM) (median slope per day, -0.57; P = .0008), and burst-forming unit-erythroid (BFU-E) (median slope per day, -1.18; P = .006) were observed over the course of the eight leukaphereses. Of 18 patients who began CTCb, 12 (67%) completed four cycles. Six patients were removed from study during intensification: two for progressive disease (PD), one refused further treatment, and three for dose-limiting hematologic toxicity. A fourth patient fulfilled the criteria for dose-limiting hematologic toxicity after cycle 4. The toxicity of the multiple cycle CTCb intensification regimen consisted of grade IV leukopenia, grade IV thrombocytopenia, and febrile neutropenia in 100%, 100%, and 26% of cycles, respectively. The median duration of each CTCb cycle was 24 days (range, 18 to 63), and the median duration of an absolute neutrophil count (ANC) < or = 500/microL and platelet count < or = 20,000/microL during each cycle was 6 days (range, 2 to 15) and 4 days (range, 0 to 38), respectively. CONCLUSION: It is feasible to administer repetitive cycles of one-quarter dose CTCb intensification with PBPC and G-CSF. Additional studies are required to determine whether multiple cycles of CTCb intensification might offer a therapeutic advantage over a single high-dose cycle.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Transplantation , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Cyclophosphamide/administration & dosage , Drug Administration Schedule , Feasibility Studies , Female , Filgrastim , Humans , Middle Aged , Recombinant Proteins , Thiotepa/administration & dosage , Treatment Outcome
11.
J Exp Med ; 185(1): 111-20, 1997 Jan 06.
Article in English | MEDLINE | ID: mdl-8996247

ABSTRACT

Hematopoietic progenitor cells migrate in vitro and in vivo towards a gradient of the chemotactic factor stromal cell-derived factor-1 (SDF-1) produced by stromal cells. This is the first chemoattractant reported for human CD34+ progenitor cells. Concentrations of SDF-1 that elicit chemotaxis also induce a transient elevation of cytoplasmic calcium in CD34+ cells. SDF-1-induced chemotaxis is inhibited by pertussis toxin, suggesting that its signaling in CD34+ cells is mediated by seven transmembrane receptors coupled to Gi proteins. CD34+ cells migrating to SDF-1 include cells with a more primitive (CD34+/CD38- or CD34+/DR-) phenotype as well as CD34+ cells phenotypically committed to the erythroid, lymphoid and myeloid lineages, including functional BFU-E, CFU-GM, and CFU-MIX progenitors. Chemotaxis of CD34+ cells in response to SDF-1 is increased by IL-3 in vitro and is lower in CD34+ progenitors from peripheral blood than in CD34+ progenitors from bone marrow, suggesting that an altered response to SDF-1 may be associated with CD34 progenitor mobilization.


Subject(s)
Antigens, CD34 , Antigens, CD , Chemokines, CXC , Chemokines/pharmacology , Chemotaxis/drug effects , Hematopoietic Stem Cells/physiology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antigens, Differentiation , Bone Marrow/pathology , Bone Marrow Cells , Breast Neoplasms/blood , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Calcium/metabolism , Cells, Cultured , Chemokine CCL4 , Chemokine CXCL12 , Chemokines/isolation & purification , Culture Media, Conditioned , Female , Fetal Blood , HLA-DR Antigens , Hematopoietic Stem Cells/drug effects , Humans , Lymphoma, Non-Hodgkin/blood , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Macrophage Inflammatory Proteins/pharmacology , Membrane Glycoproteins , N-Glycosyl Hydrolases , Pregnancy , Stromal Cells
12.
Biol Blood Marrow Transplant ; 3(5): 267-72, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9450922

ABSTRACT

Despite techniques to deplete red cells from major ABO-incompatible allogeneic bone marrow (BM) or to remove recipient isohemagglutinins (IHGs) before transplantation, delayed erythropoiesis and hemolysis, red cell aplasia, and increased red cell transfusion requirements may occur. Twenty-nine recipients of major ABO-incompatible allografts received donor-type frozen fresh plasma (FFP) infusions twice daily to adsorb IHGs in vivo. Engraftment and transfusion requirements were compared between the 29 FFP-treated major ABO-incompatible allograft recipients, 5 recipients of major ABO-incompatible BM who did not receive FFP infusions, 35 recipients of minor ABO-incompatible BM, and 172 recipients of ABO-compatible BM. No significant differences in either transfusion requirements or engraftment were seen in the FFP-treated major ABO-incompatible vs. minor ABO-incompatible or ABO-compatible groups (p values > or = 0.10). The infusion of donor-type FFP represents a simple, effective treatment strategy to neutralize IHGs and to prevent adverse consequences of major ABO incompatibility in the setting of allogeneic BM transplantation. The role of this strategy in the care of patients receiving ABO-incompatible solid organs remains to be defined.


Subject(s)
ABO Blood-Group System/immunology , Blood Component Transfusion , Blood Group Incompatibility , Bone Marrow Cells/immunology , Bone Marrow Transplantation/immunology , Hemagglutinins/chemistry , Plasma , Transplantation, Homologous/immunology , Adsorption , Erythropoiesis , Graft Survival , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Life Tables , Treatment Outcome
13.
Transfusion ; 36(9): 782-8, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8823450

ABSTRACT

BACKGROUND: It is important to compare the incidence of bacterial contamination of components collected from the peripheral blood or bone marrow (BM), as well as of components processed with or without cell selection or depletion, and to evaluate the sequelae of such contamination. STUDY DESIGN AND METHODS: Bacterial contamination rates were compared in 1380 untreated autologous peripheral blood progenitor cells (PBPCs), 291 untreated autologous BM samples, 916 monoclonal antibody (MoAb)-treated autologous and allogeneic BM samples, and in 45 autologous PBPC components from which the CD34+ cells were selected. Bacterial cultures were performed at sequential time points during the processing of MoAb-treated BM. RESULTS: Bacterial contamination was documented in 44 of 2632 components from 1593 patients (1.67% of components, 2.76% of patients) before cryopreservation. Although only 0.65% of untreated PBPCs were contaminated before cryopreservation, each patient was more likely to have given a contaminated PBPC component than a contaminated BM component (2.41% vs. 0%, p < 0.01). Bacterial contamination of MoAb-treated BM was greater during or after manipulation than it was before (2.33% vs. 0.77%, p < 0.05). At thawing, contamination was documented in 42 (1.97%) of 2136 components cultured. Ten (13.7%) of 73 patients who received hematopoietic progenitor cells that were contaminated before cryopreservation or at thawing developed fever or positive blood cultures within 48 hours of transfusion. Fever was associated with bacteremia in two cases, but no irreversible clinical sequelae were noted. CONCLUSION: These studies suggest that, despite careful attention to sterile procedures, low-level contamination of hematopoietic stem cell components can be introduced before or during manipulation as well as at thawing, and that standards for monitoring of the procedures for collection, processing, cryopreservation, thawing, and transfusion of hematopoietic progenitor cells are necessary.


Subject(s)
Bacterial Infections/transmission , Hematopoietic Stem Cell Transplantation/standards , Antibodies, Monoclonal/therapeutic use , Antigens, CD34/analysis , Bone Marrow Cells , Bone Marrow Purging , Freezing , Humans , Lymphocyte Depletion
14.
Transfusion ; 36(2): 160-7, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8614968

ABSTRACT

BACKGROUND: Peripheral blood progenitor cells (PBPCs) are commonly collected and used to reconstitute hematopoiesis after high-dose chemotherapy. However, strategies for optimal collection and assessment of leukapheresis components are not standardized. STUDY DESIGN AND METHODS: Hematopoietic progenitor cell assays were performed on 369 leukapheresis components collected from 95 patients who had received doxorubicin-based chemotherapy and/or granulocyte-colony-stimulating factor (G-CSF). Precollection patient hematologic values, leukapheresis collection values, component hematopoietic progenitor cell assays, and patient outcome measures were summarized. The kinetics of mononuclear cell (MNC) and PBPC mobilization were assessed among four patient groups. RESULTS: Patient group was a significant predictor of the peripheral blood MNC count on the day of collection (p<0.0001), and that value was a significant predictor of granulocyte-macrophage--colony-forming unit (CFU-GM) yield (p<0.0001). This relationship between the peripheral blood MNC count on the day of collection and CFU-GM yield differed according to patient group (p<0.0001). CFU-GM made up a larger fraction of peripheral blood MNCs collected from patients who received chemotherapy plus G-CSF than collected from those who received G-CSF alone. Moreover, the peripheral blood MNC count and the corresponding CFU-GM yield increased significantly on consecutive days of collection in patient groups receiving chemotherapy and G-CSF but were unchanged or decreased in patients receiving G-CSF alone. CONCLUSION: The relationship between peripheral blood MNC count and leukapheresis component CFU-GM yield differed significantly between patients who received chemotherapy and G-CSF and those who received G-CSF alone for the mobilization of PBPCs. Patient peripheral blood MNC count and component CFU-GM yield are useful for both assessing and suggesting revisions to PBPC mobilization and collection strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Separation , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells , Leukocytes, Mononuclear/drug effects , Hematopoietic Stem Cell Transplantation , Humans , Leukapheresis , Leukocyte Count , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...