Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Leukemia ; 27(2): 315-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22751450

ABSTRACT

Arsenic trioxide (ATO) induces disease remission in acute promyelocytic leukemia (APL) patients, but not in non-APL acute myeloid leukemia (AML) patients. ATO at therapeutic concentrations (1-2 µM) induces APL NB4, but not non-APL HL-60, cells to undergo apoptosis through the mitochondrial pathway. The role of antiapoptotic protein Mcl-1 in ATO-induced apoptosis was determined. The levels of Mcl-1 were decreased in NB4, but not in HL-60, cells after ATO treatment through proteasomal degradation. Both glycogen synthase kinase-3ß (GSK-3ß) inhibitor SB216763 and siRNA blocked ATO-induced Mcl-1 reduction as well as attenuated ATO-induced apoptosis in NB4 cells. Silencing Mcl-1 sensitized HL-60 cells to ATO-induced apoptosis. Both ERK and AKT inhibitors decreased Mcl-1 levels and enhanced ATO-induced apoptosis in HL-60 cells. Sorafenib, an Raf inhibitor, activated GSK-3ß by inhibiting its phosphorylation, decreased Mcl-1 levels and decreased intracellular glutathione levels in HL-60 cells. Sorafenib plus ATO augmented reactive oxygen species production and apoptosis induction in HL-60 cells and in primary AML cells. These results indicate that ATO induces Mcl-1 degradation through activation of GSK-3ß in APL cells and provide a rationale for utilizing ATO in combination with sorafenib for the treatment of non-APL AML patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Glycogen Synthase Kinase 3/metabolism , Leukemia, Myeloid, Acute/pathology , Oxides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Arsenic Trioxide , Blotting, Western , Down-Regulation , Enzyme Activation/drug effects , Flow Cytometry , Glutathione/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
2.
Pituitary ; 15 Suppl 1: S6-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-20703943

ABSTRACT

Describe a case of apoplexy of an ACTH-producing pituitary adenoma which resulted not only in an empty sella with concurrent hypothyroidism, hypoprolactinemia, and hypogonadism but persistent hypercortisolemia from two distinct extrasellar remnants of the original adenoma. Review the literature to identify other similar cases. The patient's medical history, physical exam, lab data, imaging exams and histopathological results were analyzed and compiled into a case report, and an extensive review of the literature was performed. Endocrinological data revealed hypercortisolism and an elevated ACTH with an otherwise suppressed pituitary axis. A pituitary MRI showed a macroadenoma in the left cavernous sinus in addition to an empty sella. An octreotide scan revealed lesions in the left sella turcica and the right sphenoid sinus. Tissue samples of both lesions stained positive for ACTH and negative for GH, prolactin, FSH, LH, and TSH. The lesions were surgically removed, and the patient treated with radiation and ketoconazole. This resulted in a significant decrease in ACTH and cortisol as well as a marked improvement in blood glucose control. The review of literature revealed the absence of any similar cases in the past. The patient presented with apoplexy of an ACTH-secreting pituitary macroadenoma with two hormonally active extrasellar remnants. Several cases in the literature describe recurrence of Cushing's disease following infarction of ACTH-secreting adenomas. This is the first documented case of infarction of an ACTH-producing adenoma resulting in two distinct ACTH-producing remnants without recurrence of the original adenoma.


Subject(s)
ACTH-Secreting Pituitary Adenoma/pathology , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Adult , Female , Humans , Magnetic Resonance Imaging
4.
Ann Oncol ; 18(11): 1842-50, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17872903

ABSTRACT

INTRODUCTION: HDT/ASCT is standard for relapsed and refractory DLCL patients responding to second-line chemotherapy. We incorporated a thrombopoietic agent into the ICE chemotherapy program to potentially: decrease platelet associated toxicities, augment stem cell collection and maintain dose intensity. METHODS: This randomized, double-blind, placebo-controlled phase I/II trial examines PEG-rHuMGDF versus placebo with ICE chemotherapy. Phase I compared three cohorts and defined a clinically effective dose (CED). Phase II evaluated the CED versus placebo. Outcome measures included safety, hematological end-points, stem cell collection and the impact of dose-intensity on outcome. RESULTS: Forty-one patients with primary refractory (16) or relapsed DLCL (25) were treated; Response rates for evaluable patients are: 75% (12/16) for placebo and 82% (18/22) for PEG-rHuMGDF. PEG-rHuMGDF treated patients had significantly less grade IV thrombocytopenia, higher median platelet nadirs, and less platelet transfusion per cycle. ICE dose intensity was improved with PEG-rHuMGDF versus placebo: 75 versus 42% (P = 0.008). At 8.5 years median follow-up, overall and event-free survival are 47 and 31%, respectively. Patients treated on PEG-rHuMGDF versus placebo had improved survival (59 versus 31%, P = 0.06). CONCLUSION: PEG-rHuMGDF ameliorated thrombocytopenia, improved platelet recovery, and maintained ICE dose intensity. Potential survival advantages conferred by maintaining dose intensity require validation with newer thrombopoietic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/mortality , Neoplasm Recurrence, Local/mortality , Polyethylene Glycols/administration & dosage , Thrombopoietin/administration & dosage , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Ifosfamide/administration & dosage , Karnofsky Performance Status , Lymphoma, Non-Hodgkin/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Probability , Prognosis , Recombinant Proteins/administration & dosage , Reference Values , Survival Analysis , Treatment Outcome
5.
Endocr Pract ; 13(4): 408-12, 2007.
Article in English | MEDLINE | ID: mdl-17669719

ABSTRACT

OBJECTIVE: To present a case of concomitant secretion of cortisol, androgens, and 11-deoxycorticosterone (DOC) by an adrenocortical carcinoma and review the literature in an attempt to identify similar cases. METHODS: The patient's medical history, physical examination, laboratory data, computed tomographic scan, and histopathologic results were analyzed and summarized in a case report, and an extensive review of the literature was performed. RESULTS: Endocrinologic data showed excess cortisol production, substantially elevated testosterone and androstenedione levels, and profoundly increased DOC in the setting of suppressed aldosterone. An abdominal computed tomographic scan showed a left adrenal tumor. A left adrenalectomy was performed, and the histopathologic diagnosis was stage II adrenocortical carcinoma. The review of the pertinent literature revealed the absence of any identical cases in the past. CONCLUSION: Our patient presented with a rare case of cosecretion of cortisol, testosterone, androstenedione, and DOC by an adrenocortical carcinoma, resulting in a clinical picture consistent with Cushing's syndrome, hyperandrogenism, and primary hypermineralocorticoidism. We recommend the routine performance of a DOC assay in the setting of mineralocorticoid excess in association with low plasma aldosterone levels.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/metabolism , Hydrocortisone/blood , Mineralocorticoids/blood , Testosterone/blood , Adrenal Cortex Neoplasms/diagnostic imaging , Adrenocortical Carcinoma/diagnostic imaging , Adult , Androstenedione/blood , Desoxycorticosterone/blood , Female , Humans , Tomography, X-Ray Computed
8.
Oncologist ; 6 Suppl 5: 4-7, 2001.
Article in English | MEDLINE | ID: mdl-11700386

ABSTRACT

Recent research has focused on the role of angiogenic growth factors and their ability to mediate tumor growth and metastases, both in solid tumors and in hematologic malignancies. The bone marrow microenvironment is the setting for a wealth of complex interactions that include cell-to-cell contacts as well as secretion of and response to soluble factors. Abundant evidence supports the role of basic fibroblast growth factor (bFGF) in contributing to the dysregulation of apoptosis that is the hallmark of chronic lymphocytic leukemia (CLL). In fact, CLL cells themselves express bFGF; intracellular levels of this cytokine correlate with clinical CLL stage. Other stromal factors mediate the inhibition of apoptosis in CLL as well, suggesting that strategies to block the responses of CLL cells to these factors may represent effective therapies. More broadly, the class of agents known as angiogenesis inhibitors may offer important advantages with respect to the treatment of numerous types of malignancies. Currently, a number of clinical trials are under way to evaluate the clinical potential of several different angiogenesis inhibitors in several hematologic neoplasms.


Subject(s)
Apoptosis/physiology , Fibroblast Growth Factor 2/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Vidarabine/analogs & derivatives , Angiogenesis Inhibitors/therapeutic use , Apoptosis/drug effects , Cytokines/physiology , Fibroblast Growth Factor 2/pharmacology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/physiopathology , Humans , Immunosuppressive Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Vidarabine/therapeutic use
9.
Semin Hematol ; 38(3 Suppl 7): 1-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11523022

ABSTRACT

In the postgenome era, development of novel targeted therapeutics is anticipated to accelerate, with the promise of specifically tailored strategies to treat specific molecularly characterized disease entities. Greater understanding of disease pathophysiology and pharmacologic actions at the molecular, cellular, and tissue levels have provided the basis for expanding the clinical application of available therapies such as recombinant human erythropoietin (r-HuEPO, epoetin alfa). The role of epoetin alfa in anemic cancer patients receiving chemotherapy has grown as our understanding of the relationship between anemia and quality of life in this patient population has evolved. With anemia and tumor hypoxia associated with poorer outcomes in various solid tumors, the potential impact of epoetin alfa on outcomes, as well as quality of life, in patients undergoing radiation or chemoradiation is of considerable interest. Anemia occurs almost universally in critically ill patients, resulting in substantial transfusion requirements. In this setting, the anemia appears to be consistent with anemia of chronic inflammatory disease and is potentially treatable by epoetin alfa. Recent preclinical studies indicate that erythropoietin exhibits neuroprotective effects in models of central nervous system injury, suggesting additional potential novel clinical applications for epoetin alfa worthy of careful investigation. Advances in the understanding of the ras genes and their functional proteins in signaling pathways involved in the development of cancer, particularly hematologic malignancies, over the last decade have prompted development of a new class of agents, farnesyl protein transferase inhibitors (FTIs), designed specifically to inhibit the initial step in Ras protein activation. Initial clinical evaluation of FTIs is ongoing, and preliminary results demonstrate antitumor activity in hematologic malignancies. Further identification and understanding of the function and complex interactions of proteins involved in diseases holds the promise of targeted therapies and improved patient outcomes.


Subject(s)
Anemia/drug therapy , Neoplasms/drug therapy , Alkyl and Aryl Transferases/antagonists & inhibitors , Anemia/etiology , Anemia/prevention & control , Enzyme Inhibitors/therapeutic use , Erythropoietin/therapeutic use , Farnesyltranstransferase , Humans , Neoplasms/blood , Neoplasms/complications , Quinolones/therapeutic use , Recombinant Proteins
10.
J Clin Oncol ; 19(11): 2875-82, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11387360

ABSTRACT

PURPOSE: To prospectively evaluate the effectiveness, safety, and clinical benefits of once-weekly epoetin alfa therapy as an adjunct to chemotherapy in anemic cancer patients. PATIENTS AND METHODS: A total of 3,012 patients with nonmyeloid malignancies who received chemotherapy were enrolled onto this multicenter, open-label, nonrandomized study conducted in 600 United States community-based practices. Patients received epoetin alfa 40,000 U once weekly, which could be increased to 60,000 U once weekly after 4 weeks dependent on hemoglobin response. Treatment was continued for a maximum of 16 weeks. RESULTS: Among the 2,964 patients assessable for efficacy, epoetin alfa therapy resulted in significant increases in hemoglobin levels, decreases in transfusion requirements, and improvements in functional status and fatigue as assessed by the linear analog scale assessment (energy level, ability to perform daily activities, and overall quality of life) and the anemia subscale of the Functional Assessment of Cancer Therapy-Anemia questionnaire. Improvements in quality-of-life parameters correlated significantly (P <.001) with increased hemoglobin levels. The direct relationship between hemoglobin and quality-of-life improvement was sustained during the 16-week study period, which is similar to findings of large community-based trials of three-times-weekly dosing. Once-weekly epoetin alfa was well tolerated, with most adverse events attributed to the underlying disease or concomitant chemotherapy. CONCLUSION: The results from this large, prospective, community-based trial suggest that once-weekly epoetin alfa therapy increases hemoglobin levels, decreases transfusion requirements, and improves quality of life in patients with cancer and anemia who undergo concomitant chemotherapy. Based on the results of this study, the clinical benefits and the adverse event profile of once-weekly epoetin alfa therapy in community-based practice are similar to those observed in the historical experience with the three-times-weekly dosage schedule.


Subject(s)
Anemia/drug therapy , Antineoplastic Agents/adverse effects , Erythropoietin/pharmacology , Hematinics/pharmacology , Activities of Daily Living , Adolescent , Adult , Aged , Aged, 80 and over , Anemia/pathology , Antineoplastic Agents/therapeutic use , Blood Transfusion , Drug Administration Schedule , Epoetin Alfa , Erythropoietin/administration & dosage , Fatigue , Female , Hematinics/administration & dosage , Hemoglobins/analysis , Humans , Injections, Subcutaneous , Male , Middle Aged , Neoplasms/complications , Neoplasms/drug therapy , Prospective Studies , Quality of Life , Recombinant Proteins , Treatment Outcome
11.
Semin Hematol ; 37(4 Suppl 6): 1-3, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11068948

ABSTRACT

The final stages of maturation of erythroid cells Into mature red blood cells are regulated by the growth factor erythropoietin. Circulating levels of erythropoietin are remarkably consistent across the range of normal hemoglobin levels; levels Increase markedly as hemoglobin declines below 12 g/dL In a manner suggesting that mechanisms in addition to the level of tissue oxygen are important in regulating increases in erythropoietin production and erythropoiesis. The erythropoietin receptor is found on a number of cell types in addition to erythroid progenitor cells, suggesting that erythropoietin may have specific biologic effects on other tissues, still to be carefully discerned. Clinical trials have demonstrated the effectiveness of recombinant human erythropoietin (epoetin alfa) in increasing hemoglobin level in iatrogenic and disease-related anemias. This increase has been associated with improving fatigue symptoms and enhancing overall quality of life. Questions remain, however, regarding the optimal increases in hemoglobin to be achieved in anemic patients with such therapy and whether optimal levels might vary in different patient groups.


Subject(s)
Anemia , Erythropoiesis , Erythropoietin , Humans
12.
J Pediatr ; 137(1): 96-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10891829

ABSTRACT

OBJECTIVE: To assess the role of granulocyte colony-stimulating factor (G-CSF) in autoimmune neutropenia (AIN). DESIGN: Serum G-CSF levels were measured in 57 children with AIN. Two different G-CSF-dependent assays were used: a solid-phase "sandwich" enzyme-linked immunosorbent assay and a proliferation assay. Sera from healthy persons and from patients with severe congenital neutropenia were used for negative and positive controls. RESULTS: The median G-CSF level in healthy persons (n = 13) was low, 45.6 pg/mL (range <39 to 141 pg/mL). The median G-CSF level in patients with AIN (n = 57) was very similar, 45.5 pg/mL (range <39 to 2500 pg/mL). Forty-five (79%) of 57 patients with AIN had levels within the range of the control group. Seven (12%) had marginally increased G-CSF levels (141 to 400 pg/mL), and only 5 (9%) had levels higher than 400 pg/mL. The G-CSF levels measured by enzyme-linked immunosorbent assay correlated well with levels measured by the proliferation assay, thus demonstrating that antibodies present in patient sera did not affect the biologic activity of G-CSF. CONCLUSION: G-CSF production in AIN is not increased despite the low neutrophil count, similar to thrombopoietin in immune thrombocytopenic purpura.


Subject(s)
Autoimmune Diseases/blood , Granulocyte Colony-Stimulating Factor/blood , Neutropenia/blood , Enzyme-Linked Immunosorbent Assay , Humans , Immunoassay , Infant , Sensitivity and Specificity , Tumor Cells, Cultured
13.
Endocr Pract ; 6(2): 143-7, 2000.
Article in English | MEDLINE | ID: mdl-11421530

ABSTRACT

OBJECTIVE: To review the patient profiles, laboratory data, and diagnostic approaches in factitious administration of glucocorticoids. METHODS: Four cases of surreptitious use of glucocorticoids are presented. Clinical and laboratory data as well as imaging studies are summarized. Pertinent case reports in the literature are reviewed. RESULTS: We report four cases of surreptitious use of glucocorticoids encountered within a 2-year period. All four patients were women without significant psychiatric histories. In three patients, the question of factitious Cushing's syndrome was suspected because of physical evidence or symptoms of Cushing's syndrome (or both) in the setting of suppressed cortisol levels. The fourth patient had undetectable cortisol levels in both serum and 24-hour urine samples but did not have signs or symptoms of adrenal insufficiency. In three cases, the diagnosis was confirmed by direct measurement of synthetic glucocorticoids in the patient's urine or serum. The fourth case was diagnosed by correlating increased cortisol levels with decreased precursor adrenal steroids. CONCLUSIONS: Exogenous corticosteroid use in the absence of a medical indication poses a serious risk to a patient. This possibility should be considered in patients with signs and symptoms consistent with Cushing's syndrome but with low serum and urinary cortisol levels. Similarly, this diagnosis should be suggested in patients without symptoms of adrenal insufficiency and with low cortisol levels. Laboratory measurement of synthetic steroids can be helpful in confirming the diagnosis.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Cushing Syndrome/diagnosis , Munchausen Syndrome/diagnosis , 17-Hydroxycorticosteroids/blood , Adrenal Cortex Hormones/blood , Adrenocorticotropic Hormone/blood , Cushing Syndrome/psychology , Dehydroepiandrosterone Sulfate/blood , Female , Hormones/blood , Humans , Hydrocortisone/blood , Middle Aged , Munchausen Syndrome/psychology
14.
N Engl J Med ; 339(19): 1341-8, 1998 Nov 05.
Article in English | MEDLINE | ID: mdl-9801394

ABSTRACT

BACKGROUND: Two reports from China have suggested that arsenic trioxide can induce complete remissions in patients with acute promyelocytic leukemia (APL). We evaluated this drug in patients with APL in an attempt to elucidate its mechanism of action. METHODS: Twelve patients with APL who had relapsed after extensive prior therapy were treated with arsenic trioxide at doses ranging from 0.06 to 0.2 mg per kilogram of body weight per day until visible leukemic cells were eliminated from the bone marrow. Bone marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization, reverse-transcription-polymerase-chain-reaction (RT-PCR) assay for PML-RAR-alpha fusion transcripts, and Western blot analysis for expression of the apoptosis-associated proteins caspases 1, 2, and 3. RESULTS: Of the 12 patients studied, 11 achieved complete remission after treatment that lasted from 12 to 39 days (range of cumulative doses, 160 to 495 mg). Adverse effects were relatively mild and included rash, lightheadedness, fatigue, and musculoskeletal pain. Cells that expressed both CD11b and CD33 (antigens characteristic of mature and immature cells, respectively), and which were found by fluorescence in situ hybridization to carry the t(15;17) translocation, increased progressively in number during treatment and persisted in the early phase of complete remission. Eight of 11 patients who initially tested positive for the PML-RAR-alpha fusion transcript by the RT-PCR assay later tested negative; 3 other patients, who persistently tested positive, relapsed early. Arsenic trioxide induced the expression of the proenzymes of caspase 2 and caspase 3 and activation of both caspase 1 and caspase 3. CONCLUSIONS: Low doses of arsenic trioxide can induce complete remissions in patients with APL who have relapsed. The clinical response is associated with incomplete cytodifferentiation and the induction of apoptosis with caspase activation in leukemic cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/therapeutic use , Adolescent , Adult , Aged , Antigens, CD/analysis , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Apoptosis , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/adverse effects , Bone Marrow Cells/immunology , Caspases , Cell Differentiation , Child , Humans , Immunophenotyping , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/drug effects , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/drug effects , Oxides/administration & dosage , Oxides/adverse effects , Recurrence , Remission Induction/methods
15.
J Clin Endocrinol Metab ; 83(11): 3786-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9814447

ABSTRACT

A 33-yr-old woman was found to have Cushing's syndrome due to a left adrenal cortical tumor. The tumor and the surrounding adrenal gland were removed intact and in toto. Four years later, she noticed recurrent symptoms of Cushing's syndrome, and 6 yr postoperatively, an adrenal tumor was demonstrable on computed tomography. Fourteen years after the initial procedure, a left adrenal tumor, presumably arising in ectopic adrenal tissue, was removed with relief of her symptoms of Cushing's syndrome. The site and functional capacity of ectopic adrenal tissues are reviewed.


Subject(s)
Adrenal Cortex Neoplasms/surgery , Adrenocortical Adenoma/surgery , Choristoma , Cushing Syndrome/etiology , Neoplasm Recurrence, Local/complications , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/complications , Adrenocortical Adenoma/metabolism , Adult , Female , Humans , Hydrocortisone/metabolism , Magnetic Resonance Imaging , Recurrence , Tomography, X-Ray Computed
16.
J Cell Physiol ; 177(3): 411-25, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9808150

ABSTRACT

Basic fibroblast growth factor (bFGF, FGF-2) is progressively lost from mammary epithelial cells as they become malignant. To investigate the effects of restoring the expression of bFGF in breast cancer cells, we constructed MCF-7 cells that permanently overexpress 18-kD cytoplasm-localizing bFGF (MCF-7/deltaA(FGF)(18) cells) and cells that express both the 18-kD along with the 22- and 24-kD nucleus-localizing bFGF peptides (MCF-7/NCF(FGF)(18,22,24) cells), using retroviral transduction. These stable cell constructs grew more slowly and had a larger fraction of their populations in the G0/G1 phase of the cell cycle than control cells. All forms of bFGF were eluted from MCF-7/NCF(FGF)(18,22,24) cell monolayers with 2 M NaCl, in contrast to fibroblasts that were demonstrated to secrete only the 18-kD bFGF isoform. High-affinity binding of 18-kD 125I-bFGF to these cells was significantly decreased, probably because of competitive binding by the autocrine-secreted bFGF. Recombinant 18-kD bFGF that was previously demonstrated in our laboratory to inhibit proliferation, activate MAP kinase, and induce the cyclin-dependent kinase inhibitor p21WAF1/CIP1 in MCF-7 cells, further inhibited MCF-7/deltaA(FGF)(18) cells but had no effect on MCF-7/NCF(FGF)(18,22,24) cells. The total cellular content of the high-affinity FGF receptors 1-3 was unchanged, but FGF receptor 4 was decreased in MCF-7/NCF(FGF)(18,22,24) cells. Both cell types overexpressing bFGF isoforms had elevated levels of the cyclin-dependent kinase inhibitor p27Kip1 but not that of p21WAF1/CIP1. In MCF-7/deltaA(FGF)(18) cells, FGFR1 and MAP kinase were constitutively phosphorylated. Exogenous recombinant 18-kD bFGF did not accentuate these effects but did induce an increase in the levels of p21WAF1/CIP1 corresponding to the further inhibition induced by exogenous bFGF in these cells. In MCF-7/NCF(FGF)(18,22,24) cells, FGFR1 and MAP kinase were not phosphorylated at baseline nor upon stimulation with recombinant bFGF, and exogenous bFGF only had a minimal effect on low steady-state p21WAF1/CIP1 levels. However, stimulation of these cells with phorbol ester or insulin did result in MAP kinase phosphorylation. While growth-inhibited in the G1 phase of the cell cycle, MCF-7/NCF(FGF)(18,22,24) cells retained active isoforms of cdk2 and the hyperphosphorylated form of Rb. These data suggest that high molecular weight forms of bFGF overexpressed in MCF-7 cells do not activate the receptor-mediated MAP kinase pathway, and do not induce p21WAF1/CIP1 in an autocrine manner, but inhibit proliferation through other, possibly direct nuclear signalling mechanisms.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Fibroblast Growth Factor 2/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/physiology , Cyclins/physiology , Enzyme Activation/physiology , G1 Phase/physiology , Humans , Phosphorylation , Phosphotransferases/antagonists & inhibitors , Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Transduction, Genetic , Tumor Cells, Cultured
17.
Blood ; 92(5): 1497-504, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716575

ABSTRACT

Inorganic arsenic trioxide (As2O3) and the organic arsenical, melarsoprol, were recently shown to inhibit growth and induce apoptosis in NB4 acute promyelocytic leukemia (APL) and chronic B-cell leukemia cell lines, respectively. As2O3 has been proposed to principally target PML and PML-RARalpha proteins in APL cells. We investigated the activity of As2O3 and melarsoprol in a broader context encompassing various myeloid leukemia cell lines, including the APL cell line NB4-306 (a retinoic acid-resistant cell line derived from NB4 that no longer expresses the intact PML-RARalpha fusion protein), HL60, KG-1, and the myelomonocytic cell line U937. To examine the role of PML in mediating arsenical activity, we also tested these agents using murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors in which the PML gene had been inactivated by homologous recombination. Unexpectedly, we found that both compounds inhibited cell growth, induced apoptosis, and downregulated bcl-2 protein in all cell lines tested. Melarsoprol was more potent than As2O3 at equimolar concentrations ranging from 10(-7) to 10(-5) mol/L. As2O3 relocalized PML and PML-RARalpha onto nuclear bodies, which was followed by PML degradation in NB4 as well as in HL60 and U937 cell lines. Although melarsoprol was more potent in inhibiting growth and inducing apoptosis, it did not affect PML and/or PML-RARalpha nuclear localization. Moreover, both As2O3 and melarsoprol comparably inhibited growth and induced apoptosis of PML+/+ and PML-/- MEFs, and inhibited colony-forming unit erythroid (CFU-E) and CFU granulocyte-monocyte formation in BM cultures of PML+/+ and PML-/- progenitors. Together, these results show that As2O3 and melarsoprol inhibit growth and induce apoptosis independent of both PML and PML-RARalpha expression in a variety of myeloid leukemia cell lines, and suggest that these agents may be more broadly used for treatment of leukemias other than APL.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Leukemia, Myeloid/pathology , Melarsoprol/pharmacology , Neoplasm Proteins/physiology , Nuclear Proteins , Oxides/pharmacology , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Animals , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Division/drug effects , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Leukemia, Promyelocytic, Acute/pathology , Mice , Neoplasm Proteins/analysis , Promyelocytic Leukemia Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Retinoic Acid/analysis , Transcription Factors/analysis , Tumor Cells, Cultured , Tumor Suppressor Proteins
19.
Clin Cancer Res ; 4(1): 171-6, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9516967

ABSTRACT

Actinonin, an antibiotic and CD13/aminopeptidase N (APN) inhibitor, has been shown to be cytotoxic to tumor cell lines in vitro. We investigated the antiproliferative effects of actinonin on human and murine leukemia and lymphoma cells. Actinonin inhibited growth of NB4 and HL60 human cell lines and AKR mouse leukemia cells in vitro with an IC50 of about 2-5 micrograms/ml. The inhibitory effect on CD13-positive cells was not blocked by pretreatment with the anti-CD13/APN monoclonal antibody F23, which binds with high affinity to the active site of CD13/APN and blocks its enzymatic activity. Moreover, F23 alone was not inhibitory to CD13-positive cells. Furthermore, a similar inhibitory IC50 of actinonin was seen in the CD13-negative cell lines RAJI and DAUDI human lymphoma. These data suggest that the inhibitory effect of actinonin is not mediated by inhibition of CD13/APN. Cell cycle analysis showed that actinonin induces a G1 arrest in HL60 and NB4 cells; apoptosis was observed in 20-35% of the cells as measured by intracellular flow cytometry. To assess whether these effects could be seen in vivo, the effect of actinonin on the syngeneic AKR mouse leukemia model was evaluated. Actinonin showed dose-dependent antitumor effects on AKR leukemia in vivo, resulting in a survival advantage. In conclusion, apoptosis, growth inhibition, and therapeutic effects in vivo are induced by actinonin and are not likely to be mediated by CD13/APN.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Female , G1 Phase/drug effects , Humans , Hydroxamic Acids/pharmacology , Leukemia, Experimental/drug therapy , Mice , Mice, Inbred AKR , Tumor Cells, Cultured
20.
Blood ; 90(11): 4307-12, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9373241

ABSTRACT

Flavopiridol is a novel, potent inhibitor of cyclin-dependent kinases (CDK). This synthetic flavone has been reported to exhibit antitumor activity in murine and human tumor cell lines in vitro and in vivo and is currently undergoing clinical phase I evaluation. In the present study, 1 Epstein-Barr virus (EBV)-transformed B-prolymphocytic cell line (JVM-2), 1 EBV-transformed B-CLL cell line (I83CLL), and 1 non-EBV transformed B-CLL cell line (WSU-CLL) were used as targets. Treatment of the cells with flavopiridol (100 nmol/L to 400 nmol/L) led to a marked dose- and time-dependent inhibition of cell growth and survival as determined using trypan blue exclusion. Morphologic analysis showed characteristic apoptotic changes such as chromatin condensation and fragmentation, membrane blebbing, and formation of apoptotic bodies. Furthermore, quantitative assessment of apoptosis-associated DNA strand breaks by in situ TdT labeling showed that a significant number of flavopiridol-treated cells underwent apoptosis. These cellular effects were associated with a significant decrease in bcl-2 expression as observed by Northern and Western blotting. The results showed that flavopiridol downregulates bcl-2 mRNA and bcl-2 protein expression within 24 hours. Genistein and quercetin, two flavonoids that do not inhibit CDKs, did not affect bcl-2 expression. These data suggest an additional mechanism of action of this new flavone which might be useful as an agent in the treatment of chronic lymphoid malignancies.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Leukemia, B-Cell/pathology , Piperidines/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Cell Survival , Humans , Leukemia, B-Cell/metabolism , RNA, Messenger/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...