Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Case Rep Hematol ; 2013: 628513, 2013.
Article in English | MEDLINE | ID: mdl-24198984

ABSTRACT

Introduction. Despite its low incidence, acquired factor VIII inhibitor is the most common autoantibody affecting the clotting cascade. The exact mechanism of acquisition remains unclear, but postpartum patients, those with autoimmune conditions or malignancies, and those with exposure to particular drugs appear most susceptible. There have been several case reports describing acquired FVIII inhibitors in patients receiving interferon alpha for HCV treatment and in patients being treated for HIV. To our knowledge, this is the first case of a patient with HCV and HIV who was not actively receiving treatment for either condition. Case Presentation. A 57-year-old Caucasian male with a history of HIV and HCV was admitted to our hospital for a several day history of progressively worsening right thigh bruising and generalized weakness. CTA of the abdominal arteries revealed large bilateral retroperitoneal hematomas. Laboratory studies revealed the presence of a high titer FVIII inhibitor. Conclusion. Our case of a very rare condition highlights the importance of recognizing and understanding the diagnosis of acquired FVIII inhibitor. Laboratory research and clinical data on the role of newer agents are needed in order to better characterize disease pathogenesis, disease associations, genetic markers, and optimal disease management.

2.
Dig Dis Sci ; 54(11): 2538-40, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19104936

ABSTRACT

Anti-CTLA-4 antibodies are human monoclonal antibodies previously studied in the treatment of metastatic melanoma (MM). CTLA-4 is an inhibitory receptor on cytotoxic T cells, blockade of which will activate T cells allowing them to attack malignant cells. Normal host cells may also be affected, and immune-mediated enterocolitis can occur. This is a prospective observational study on the use of corticosteroids and infliximab in the treatment of patients with immune-mediated colitis secondary to anti-CTLA-4 antibody treatment of MM. Five patients presented with colitis after medication administration. Patients were treated with high-dose corticosteroids for 1 week, but diarrhea did not completely abate in any of them. They were then treated successfully with infliximab. One patient had recurrence of symptoms and responded to repeat treatment with infliximab. Patients who develop immune-mediated colitis after administration of anti-CTLA-4 antibodies have previously been reported to respond to corticosteroids, but in our study, all required treatment with infliximab.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Colitis/drug therapy , Gastrointestinal Agents/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Aged , Antibodies, Monoclonal, Humanized , Antigens, CD/immunology , CTLA-4 Antigen , Colitis/chemically induced , Colitis/immunology , Female , Humans , Infliximab , Ipilimumab , Male , Melanoma/drug therapy , Middle Aged , Prospective Studies
3.
J Clin Oncol ; 18(2): 376-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10637253

ABSTRACT

PURPOSE: To determine immune responses and toxicity to the anti-idiotype vaccine, as well as clinical responses and survival, we initiated a clinical trial for patients with advanced melanoma treated with an anti-idiotype antibody (TriGem) that mimics the disialoganglioside GD2. PATIENTS AND METHODS: Forty-seven patients with advanced melanoma received either 1-, 2-, 4-, or 8-mg doses of TriGem (Titan Pharmaceuticals Inc, South San Francisco, CA) mixed with QS-21 adjuvant (Aquila Biopharmaceuticals, Inc, Worcester, MA) 100 microg subcutaneously weekly for 4 weeks and then monthly until disease progression. Median age was 57 years, there were 32 men and 15 women, 43% of patients had undergone prior therapy for metastatic disease, 55% had disease confined to soft tissue, and 45% had visceral metastasis. RESULTS: Hyperimmune sera from 40 of 47 patients showed an anti-anti-idiotype (Ab3) response. Patient Ab3 was truly Ab1' because it specifically bound purified disialoganglioside GD2. The isotypic specificity of the Ab3 antibody consisted of predominantly immunoglobulin (Ig)G, and all IgG subclasses were represented. One patient had a complete response that persisted at 24 months, and 12 patients were stable from 14+ to 37+ months (median, 18+ months). Disease progression occurred in 32 patients on study from 1 to 17 months (median, 5.5 months), and 21 have died at 1 to 16 months (median, 6 months). The Kaplan-Meier-derived overall median survival has not been reached. Median survival has not been reached for the 26 patients with soft tissue disease only and was 13 months for 21 patients with visceral metastasis. Toxicity consisted of local reaction at the site of injection and mild fever and chills. CONCLUSION: TriGem has minimal toxicity and generates robust and specific IgG immune responses against GD2. Objective responses were minimal, but there may be a favorable impact on disease progression and survival that will require prospective randomized trials.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Gangliosides/immunology , Immunoglobulin G/immunology , Melanoma/immunology , Skin Neoplasms/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Anti-Idiotypic/therapeutic use , Antibody Specificity , Disease Progression , Female , Gangliosides/therapeutic use , Humans , Immunization , Male , Melanoma/pathology , Melanoma/therapy , Middle Aged , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Survival Analysis
4.
Cancer ; 88(3): 557-62, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10649247

ABSTRACT

BACKGROUND: The authors conducted a Phase II study to evaluate the activity of the combination of gemcitabine and vinorelbine in patients with advanced nonsmall cell lung carcinoma (NSCLC). METHODS: Patients were eligible if they had Stage IIIB (malignant pleural effusion) or Stage IV NSCLC, no prior chemotherapy, and Cancer and Leukemia Group B performance status (PS) 0-2. Patients with brain metastases were eligible if they were neurologically stable after brain irradiation. Thirty-three patients from participating institutions were enrolled. One patient was ineligible due to untreated brain metastases. Patients were treated with gemcitabine 1250 mg/m(2) over 30 minutes (1000 mg/m(2) for the first 6 patients) and vinorelbine 25 mg/m(2) over 6 minutes, both administered intravenously on Days 1 and 8 every 21 days. Treatment was planned for a total of six cycles or more if the patient had persistent benefit. Growth factors were not allowed. RESULTS: Among all 32 eligible patients, there were 8 partial responses, for an overall response rate of 25% (95% confidence interval [CI], 11.5-43. 4%). The median survival time was 8.3 months and the 1-year survival rate was 38% (95% CI, 24-59%). Patients with PS 0-1 had a median survival of 11.7 months and a 1-year survival rate of 48%. Grade 3 and 4 neutropenia was observed in 13% and 25% of the 148 treatment cycles, respectively. One patient died of neutropenic sepsis. Only 2 episodes of Grade 3 and 4 thrombocytopenia were observed. Nonhematologic toxicity was minimal. CONCLUSIONS: Gemcitabine and vinorelbine is an active and well-tolerated regimen in patients with advanced NSCLC, with response and survival rates at least comparable to those achieved with standard platinum-based regimens. This combination may be particularly suitable for the elderly or for patients who cannot tolerate more toxic platinum-based regimens.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/analogs & derivatives , Lung Neoplasms/drug therapy , Vinblastine/analogs & derivatives , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Confidence Intervals , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasm Staging , Neutropenia/chemically induced , Pleural Effusion, Malignant/drug therapy , Remission Induction , Survival Rate , Thrombocytopenia/chemically induced , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinorelbine , Gemcitabine
5.
Cancer Chemother Pharmacol ; 37(1-2): 168-72, 1995.
Article in English | MEDLINE | ID: mdl-7497588

ABSTRACT

Although collateral sensitivity to gamma radiation has previously been described in multidrug-resistant tumor cell lines, we describe here a multidrug-resistant human T-cell acute lymphatic leukemia cell line, L100, which displayed increased sensitivity to both gamma radiation and cis-platinum. Cis-platinum cytotoxicity of parental L0 cells and L100 cells was enhanced, whereas radiation sensitivity of L0 and L100 cells was unaltered by glutathione depletion. These results indicate that disparate mechanisms are operative in the collateral sensitivity of L100 cells to gamma radiation and cis-platinum.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Radiation Tolerance , Drug Resistance, Multiple , Glutathione/metabolism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured
6.
Exp Hematol ; 21(13): 1640-7, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8243566

ABSTRACT

In autologous bone marrow transplantation in patients with acute myeloid leukemia (AML), 4-hydroperoxycyclophosphamide (4-HC) is a commonly used ex vivo purging agent for leukemic blasts. In the present report, we demonstrate that exposure to high concentrations of 4-HC for 1 hour, as used in ex vivo bone marrow purging, produces internucleosomal DNA fragmentation characteristic of apoptosis, or programmed cell death (PCD), in human myeloid leukemia HL60 cells. Lower concentrations of 4-HC (10, 20, or 50 microM/L) failed to cause this effect, while higher concentrations (> or = 200 microM/L) produced random DNA fragmentation. 4-HC-mediated internucleosomal DNA fragmentation was associated with a marked induction in c-jun and significant reductions in bcl-2 and c-myc oncogene expressions. A combined treatment with interleukin-3 (IL-3) plus IL-6 for 18 hours before an additional, 1-hour concurrent treatment with 4-HC (100 microM/L) significantly increased 4-HC-induced DNA fragmentation as well as colony growth inhibition of HL60 cells. The effects of cotreatment with IL-3 plus IL-6 were also associated with a further, modest decrease in bcl-2 and c-myc and augmentation of c-jun expression. These findings highlight an alternative mechanism of 4-HC-induced leukemic cell death that can be potentially enhanced by cotreatment with IL-3 plus IL-6.


Subject(s)
Apoptosis/drug effects , Cyclophosphamide/analogs & derivatives , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Leukemia, Myeloid/pathology , Blotting, Southern , Bone Marrow Purging , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , DNA, Neoplasm/metabolism , Gene Expression , Genes, jun , Genes, myc , Humans , Interleukin-3/administration & dosage , Interleukin-6/administration & dosage , RNA, Neoplasm/analysis , Tumor Cells, Cultured
7.
Leukemia ; 6(8): 814-9, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1640734

ABSTRACT

4-Hydroperoxycyclophosphamide (4-HC) is widely used as an ex vivo bone marrow purging agent for acute myeloid leukemia (AML) blasts. We have determined the effect of a combined treatment with interleukin 3 (IL-3) plus IL-6 on 4-HC cytotoxicity against normal (CFU-GEMM) versus AML (L-CFU) bone marrow progenitor cells. Following an 18 h exposure to IL-3 plus IL-6, treatment with 4-HC in conjunction with IL-3 and IL-6 for one hour resulted in a significantly greater inhibition of L-CFU versus CFU-GEMM colony growth. In addition, treatment with IL-3 plus IL-6 reduced the inhibitory effects of higher concentrations of 4-HC on CFU-GEMM but not L-CFU growth. IL-3 and IL-6 did not protect the self-renewing, clonogenic, AML blast progenitor cells from the cytotoxic effects of 4-HC. While the total intracellular glutathione (GSH) levels were not significantly different between untreated normal bone marrow mononuclear cells (NBMMC) and AML blasts, greater intracellular GSH-S transferase activity was observed in the NBMMC. 4-HC produced a marked reduction in GSH levels in NBMMC as well as AML blasts. But treatment with IL-3 plus IL-6 in conjunction with 4-HC resulted in significantly higher GSH levels in NBMMC. These differences in intracellular GSH levels and GST activity may offer an explanation for the differential protective effects of IL-3 plus IL-6 treatment against the cytotoxic effects of 4-HC on CFU-GEMM colony growth.


Subject(s)
Cyclophosphamide/analogs & derivatives , Hematopoietic Stem Cells/drug effects , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Leukemia, Myeloid/drug therapy , Acute Disease , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Division/drug effects , Cells, Cultured , Cyclophosphamide/pharmacology , Drug Interactions , Glutathione/metabolism , Glutathione Transferase/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Tumor Cells, Cultured
8.
Gynecol Oncol ; 45(1): 32-9, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1601333

ABSTRACT

The intracellular metabolism and cytotoxic effects of Ara-C and 2'-difluorodeoxycytidine (dFdC or Gemcitabine) administered with or without deoxycytidine (dCyd) were examined in cisplatin-resistant (2008/C13) and -sensitive (2008) human ovarian cystadenocarcinoma cells. Compared to 2008 cells, 2008/C13 cells possess 2.1-fold higher glutathione (GSH) levels, enhanced expressions of GSH S-transferase (GST)-pi mRNA and protein, and significantly greater activity of GST, GSH peroxidase, and GST reductase. Although 2008/C13 cells were slightly cross-resistant to 4-hydroperoxycyclophosphamide, the drug displayed a steep dose-response (colony growth inhibition) effect toward these cells. 2008/C13 cells expressed greater sensitivity toward Ara-C and Gemcitabine. This was associated with intracellular Ara-CTP and dFdCtriphosphate levels in 2008/C13 significantly higher than those in 2008 cells. Against bone marrow progenitor cells, the cytotoxic effects of submicromolar levels of Ara-C or dFdC, produced in plasma following intraperitoneal administration of the drugs, were significantly reversed by cotreatment with high levels of dCyd achieved in plasma following intravenous administration. In contrast, the metabolism and cytotoxic effects of Ara-C and dFdC in 2008 and 2008/C13 cells were not significantly altered by dCyd concentrations that are reached in the peritoneum following intravenous administration. These in vitro data suggest that systematically administered dCyd might protect bone marrow progenitor cells against Ara-C cytotoxicity without impairing antitumor activity of intraperitoneal Ara-C.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Bone Marrow Cells , Cisplatin/therapeutic use , Cystadenocarcinoma/drug therapy , Cystadenocarcinoma/pathology , Cytarabine/adverse effects , Cytarabine/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Stem Cells/drug effects , Antimetabolites, Antineoplastic/administration & dosage , Blotting, Western , Cystadenocarcinoma/chemistry , Cytarabine/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Female , Glutathione Transferase/analysis , Glutathione Transferase/genetics , Humans , Injections, Intraperitoneal , Injections, Intravenous , Ovarian Neoplasms/chemistry , RNA, Messenger/analysis , RNA, Messenger/genetics , Stem Cells/cytology , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Gemcitabine
9.
Am J Clin Oncol ; 14(6): 509-13, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1957839

ABSTRACT

Twenty patients with advanced carcinomas of the colorectum, pancreas, stomach, and breast were enrolled in a Phase I study of a sequential administration of 5-fluorouracil-leucovorin (FU-LV) combination followed by hydroxyurea (HU) with allopurinol protection (HALF regimen). As a weekly regimen for 6 weeks, followed by a rest period of 2 weeks, FU was administered intravenously (i.v.) during infusion of a 2-hour i.v. infusion of LV at a dose of 500 mg/m2. Six hours following the FU-LV combination, HU (1 gm/m2) was administered orally. Allopurinol (300 mg every 8 hours, orally) was given the day before and on the day of the administration of the FU-LV combination. The starting dose of FU was 300 mg/m2, with escalations to 900 mg/m2. Mucositis, diarrhea, and hematologic toxicities were mild and sporadic with FU doses up to 750 mg/m2 and occurred in patients who had received prior treatment with FU and/or radiotherapy. Dose-limiting neurocerebellar toxicity was observed in 2 out of 6 patients who received a FU dose of 900 mg/m2. Three additional patients experienced moderate neuromotor toxicity at this dose level. Among 17 patients evaluable for response, partial responses were seen in 3 of the 9 patients with colorectal cancer, 1 of the 3 patients each with carcinoma of breast and pancreas. Three of the 5 responses occurred in patients who had received prior treatment with FU and/or radiation therapy. An FU dose of 750 mg/m2 is recommended for a Phase II trial of the HALF regimen.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Digestive System Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Digestive System Neoplasms/pathology , Drug Administration Schedule , Drug Evaluation , Female , Fluorouracil/administration & dosage , Humans , Hydroxyurea/administration & dosage , Leucovorin/administration & dosage , Male , Middle Aged
10.
Exp Hematol ; 19(7): 669-73, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1893953

ABSTRACT

We have examined the effect of recombinant interleukin 3 (rIL-3) on the metabolism of high-dose cytosine arabinoside (Ara-C), an S-phase-specific agent, in normal human bone marrow mononuclear cells (BMMC) and leukemic blasts from patients with acute myeloid leukemia (AML). Exposure to rIL-3 for 24 h significantly increased the percentage of cycling S-phase cells in normal BMMC as well as leukemic cells. A concomitant expansion of intracellular deoxycytidine triphosphate (dCTP) levels occurred to a significantly greater extent in normal BMMC. Compared to treatment with Ara-C (10 mumol/liter) alone, prior and coadministration of rIL-3 with Ara-C increased Ara-CTP levels in leukemic blasts. However, an identical treatment produced significantly higher dCTP levels in normal BMMC, resulting in a significantly lower mean Ara-CTP to dCTP pool ratio in normal BMMC compared to that observed in each of the samples of AML blasts. Following treatment with Ara-C plus rIL-3 versus Ara-C alone, the alteration in Ara-C DNA incorporation corresponded with the change in Ara-CTP to dCTP ratio observed in normal BMMC and AML blasts. The differential effect of rIL-3 on the metabolism of high-dose Ara-C in normal versus leukemic cells may indicate a role for rIL-3 in enhancing the selectivity of Ara-C toward leukemic myeloblasts.


Subject(s)
Bone Marrow/metabolism , Cytarabine/metabolism , Interleukin-3/pharmacology , Leukemia, Myeloid, Acute/metabolism , DNA/metabolism , Deoxycytosine Nucleotides/metabolism , Humans , Recombinant Proteins/pharmacology
11.
Cancer Chemother Pharmacol ; 28(2): 93-101, 1991.
Article in English | MEDLINE | ID: mdl-1711935

ABSTRACT

The glycoproteins on the surface of HL-60/S wild-type, drug-sensitive human leukemia cells and HL-60/AR anthracycline-resistant cells which do not overexpress the P-glycoprotein, were characterized by labeling with [35S]-methionine, NaB[3H4], phosphorus 32, or sodium iodide I 125. HL-60/S and HL-60/AR cell lysates and membrane fractions tagged with [35S]-methionine or phosphorus 32 showed no significant differences in their protein patterns as analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and by autoradiography. HL-60/S cells labeled with NaB[3H4] yielded glycoproteins that were smeared predominantly in the molecular-weight range of 210,000 and 160,000 Da, with pI values ranging between pH 4 and pH 4.4. In contrast, NaB[3H4]-labeled HL-60/AR cells showed 7-8 discrete glycoproteins within a molecular-weight range of 170,000 and 140,000 Da, with pI values also ranging between pH 4 and pH 4.4. In addition, [3H]-glucosamine incorporation into HL-60/S and HL-60/AR cells revealed that the latter showed lower uptake of [3H]-glucosamine than did the former. Following treatment with tunicamycin, [3H]-glucosamine uptake in HL-60/S cells decreased, whereas that in HL-60/AR cells remained unchanged. Surface-membrane radioiodination of HL-60/S and HL-60/AR cells showed two distinct protein electrophoretic patterns, with differences being observed in both the high-(220-95 kDa) and low-molecular-weight ranges (21 kDa). Flow cytometric analysis of HL-60/S and HL-60/AR cells using myeloid and lymphoid antigen-specific antibodies demonstrated no antigenic differences between HL-60/S and HL-60/AR cells. HL-60/S cells incubated in the presence of tunicamycin, an inhibitor of N-linked glycosylation, or the protein kinase C agonist phorbol 12-myristate 13-acetate (PMA) developed a glycoprotein pattern similar to that observed in HL-60/AR cells. In addition, tunicamycin treatment of HL-60/S cells decreased daunorubicin (DNR) retention and altered its intracellular distribution as compared with that in HL-60/AR cells. These data indicate that HL-60/AR cells do not possess either de novo or amplified high-molecular-weight surface-membrane proteins; instead, existing proteins are hypoglycosylated. These results also show that HL-60/AR cells exhibit the multidrug-resistant phenotype in association with altered membrane glycoproteins of both high (220-95 kDa) and low molecular weight (21 kDa), but without overexpression of the P-glycoprotein. Furthermore, in HL-60/S cells, the multidrug-resistant phenotype is partially inducible by inhibition of N-linked glycosylation of cell-surface proteins.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Leukemia, Myeloid/metabolism , Membrane Glycoproteins/drug effects , Tunicamycin/pharmacology , Autoradiography , Cell Line , Culture Techniques , Drug Resistance , Electrophoresis, Polyacrylamide Gel , Epitopes/isolation & purification , Glycosylation/drug effects , Humans , Membrane Glycoproteins/metabolism , Phenotype , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology
12.
Cancer Res ; 49(16): 4607-14, 1989 Aug 15.
Article in English | MEDLINE | ID: mdl-2545346

ABSTRACT

Anthracycline-sensitive (HL-60) and -resistant (HL-60/AR) cells, which do not overexpress the P-glycoprotein, each transport and distribute daunorubicin (DNR) into distinct intracellular locations, as visualized by digitized video fluorescence microscopy. At pH 7.4, the fluorescence of DNR in HL-60 cells appears distributed diffusely in both the nucleus and cytoplasm. In contrast, HL-60/AR cells show much less fluorescence in the nucleus and cytoplasm; most of the fluorescence localizes first to the Golgi apparatus and is then gradually shifted to the lysosomes and/or mitochondria. In pharmacokinetic studies, HL-60/AR cells exposed to different extracellular concentrations of [14C]DNR consistently accumulated less radioactive drug than the parent HL-60 cells. Incubation of HL-60/AR cells with sodium azide and deoxyglucose blocked the efflux of [14C]DNR and also prevented the shift of DNR fluorescence from the Golgi apparatus to the lysosomes/mitochondria. The efflux and the intracellular shift of DNR could also be inhibited by lowering the temperature to 18 degrees C, which stops endosomal membrane fusion. When DNR was allowed to accumulate in HL-60 or HL-60/AR cells at pH 5 there was an increase in the proportion of drug fluorescence in the membranes of both HL-60 and HL-60/AR cells; a decrease in the amount of drug retained by HL-60, but not by HL-60/AR cells; and a decrease in the cytostatic effects of DNR on both HL-60 and HL-60/AR cells. These data suggest that DNR resistance is associated with a failure of DNR to pass through membranes and to bind to cytoplasmic and nuclear structures. Instead, most of the drug is taken up by the Golgi apparatus from which it is then shifted to the lysosomes or to mitochondria, or out of the cell.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Daunorubicin/pharmacokinetics , Leukemia, Myeloid/metabolism , Azides/pharmacology , Cell Line , Deoxyglucose/pharmacology , Drug Resistance , Fluorescence , Golgi Apparatus/metabolism , Mitochondria/metabolism , Protons , Sodium Azide , Temperature
13.
Cancer Res ; 49(15): 4120-5, 1989 Aug 01.
Article in English | MEDLINE | ID: mdl-2568167

ABSTRACT

We studied the cellular enzymatic defenses against anthracycline-induced free radical damage in the HL60 human myelogenous leukemia cell line and in its anthracycline-resistant subline, HL60/AR. Intracellular glutathione (GSH) levels and gamma-glutamyl transpeptidase activity were lower in HL60/AR than in HL60 cells. Glutathione-S-transferase (GST) and glutathione peroxidase activities were similar in both cell lines. The intracellular distribution of GSH/GST was visualized by digitized video fluorescence microscopy, utilizing the fluorescent probe monochlorobimane fluorescence microscopy, utilizing the fluorescent probe monochlorobimane (MBCl), which is specifically conjugated to GSH by GST. In HL60 cells stained with the MBCl probe, a bright diffuse cytoplasmic and nuclear fluorescence pattern was observed, whereas in HL60/AR cells, the fluorescence was mostly localized to the Golgi apparatus with a lesser component of diffuse cytoplasmic and nuclear fluorescence. Pretreatment of HL60/AR cells with buthionine sulfoximine (BSO) partially reversed resistance to daunorubicin. This effect of BSO on resistance was associated not only with the abolition of localized MBCl fluorescence to the Golgi apparatus but also with increased intracellular accumulation and retention of daunorubicin. The results of our studies demonstrate that inhibition of GSH synthesis in HL60/AR cells results in significant sensitization to daunorubicin and suggest that changes in the intracellular distribution of GSH/GST and/or increased drug retention may be involved in mediating this effect.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Glutathione Transferase/physiology , Glutathione/physiology , gamma-Glutamyltransferase/physiology , Buthionine Sulfoximine , Cell Survival/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance , Glutathione/analysis , Glutathione Transferase/analysis , Humans , Leukemia, Promyelocytic, Acute/pathology , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , Mitochondria/physiology , Tumor Cells, Cultured , gamma-Glutamyltransferase/analysis
14.
Am J Clin Pathol ; 92(1): 101-4, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2473646

ABSTRACT

Pulmonary interstitial fibrosis, a well-known toxic effect of bleomycin therapy, usually presents radiographically as diffuse reticularity. The authors report an unusual case of biopsy-proven bleomycin toxicity that presented as pulmonary nodules mimicking metastatic tumor. The histologic findings resembled those seen in the diffuse form of toxicity but notably also included foci of bronchiolitis obliterans.


Subject(s)
Bleomycin/adverse effects , Lung Diseases/chemically induced , Adult , Diagnosis, Differential , Humans , Lung Diseases/diagnostic imaging , Lung Diseases/pathology , Male , Radiography
15.
Leukemia ; 2(12): 810-3, 1988 Dec.
Article in English | MEDLINE | ID: mdl-3264363

ABSTRACT

Human recombinant GM-CSF (rGM-CSF) is under investigation as a growth-protective agent for normal hematopoietic elements in phase I trials of myelosuppressive chemotherapy and in bone marrow transplantation. We determined the effect of rGM-CSF on the metabolism of high dose Ara-C in bone marrow mononuclear cells (BMMCs) from healthy volunteers and patients with ANLL. Cells were incubated with rGM-CSF alone, Ara-C alone, or a combination of the two drugs. Treatment with rGM-CSF alone yielded approximately a twofold increment in intracellular dCTP pools in normal BMMCs but not in leukemic blasts. Exposure to rGM-CSF in conjunction with Ara-C corrected Ara-C-mediated declines in dCTP levels and decreased cytosine arabinoside triphosphate (Ara-CTP) accumulation in normal BMMCs but not in their leukemic counterparts. Furthermore, when exposure to Ara-C was preceded by treatment with rGM-CSF for 18 hr, an even greater reduction in the Ara-CTP/dCTP pool ratio was observed in normal versus leukemic elements; however, this did not significantly change Ara-C DNA incorporation in the two cell types. The differential effect of rGM-CSF on the phosphorylation of Ara-C in normal BMMCs versus leukemic blasts has potential implications for the use of a regimen consisting of rGM-CSF and high dose Ara-C in the treatment of ANLL with chemotherapy or autologous bone marrow transplantation.


Subject(s)
Bone Marrow/metabolism , Colony-Stimulating Factors/pharmacology , Cytarabine/metabolism , Hematopoietic Stem Cells/drug effects , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/drug effects , Cell Division/drug effects , Deoxycytosine Nucleotides/metabolism , Drug Synergism , Granulocytes , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Macrophages , Neoplastic Stem Cells/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...