Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
10.
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
11.
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
12.
Blood ; 90(2): 562-70, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9226155

ABSTRACT

Inorganic arsenic trioxide (As2O3) was recently shown to induce apoptosis in NB4 promyelocytic leukemic cells. We have investigated the effects of the organic arsenical, melarsoprol (a drug used for treatment of trypanosomiasis), upon induction of apoptosis in cell lines representative of chronic B-cell lymphoproliferative disorders. An Epstein-Barr virus (EBV)-transformed B-prolymphocytic cell line (JVM-2), an EBV-transformed B-cell chronic lymphocytic leukemia (B-CLL) cell line (I83CLL), and one non-EBV-transformed B-CLL cell line (WSU-CLL) were used as targets. Dose-response experiments with melarsoprol (10(-7) to 10(-9) mol/L) were performed over 96 hours. Unexpectedly, we found that melarsoprol caused a dose- and time-dependent inhibition of survival and growth in all three cell lines. In contrast, As2O3 at similar concentrations had no effect on either viability or growth. After 24 hours, all three cell lines treated with melarsoprol (10(-7) mol/L) exhibited morphologic characteristics of apoptosis. We also observed prominent concentration-dependent downregulation of bcl-2 mRNA after 24 hours of exposure to melarsoprol in WSU-CLL, I83CLL, and JVM-2 cells. Decrease of bcl-2 protein expression was also observed in all three cell lines, whereas As2O3 had no effect on this parameter. We conclude that melarsoprol may inhibit the growth of lymphoid leukemic cell by promoting programmed cell death. Results of these studies suggest that melarsoprol shows promising therapeutic activity in these diseases, and a study to evaluate clinical effects of this drug has been initiated.


Subject(s)
Antineoplastic Agents/toxicity , Arsenic Poisoning , Arsenicals , Melarsoprol/toxicity , Oxides/toxicity , Apoptosis/drug effects , Arsenic Trioxide , Cell Division/drug effects , Cell Line, Transformed , Cell Survival/drug effects , DNA Damage , DNA Fragmentation , Drug Screening Assays, Antitumor , Herpesvirus 4, Human , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA, Messenger/biosynthesis , Time Factors , Transcription, Genetic/drug effects , Tumor Cells, Cultured
13.
Leukemia ; 11(2): 258-65, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9009090

ABSTRACT

Basic fibroblast growth factor (bFGF) is a pleiotropic cytokine which has recently been shown to delay fludarabine-induced apoptosis in B cell chronic lymphocytic leukemia (B-CLL) cells. To investigate the potential mechanism of bFGF-mediated delay of apoptosis, two EBV-transformed B prolymphocytic cell lines (JVM-2, JVM-13), one EBV-transformed B-CLL cell line (I83CLL), and one non-EBV-transformed B-CLL cell line (WSU-CLL) were used as a model for chronic lymphoid malignancies. Viability data of cells treated with fludarabine alone or in combination with bFGF demonstrated that the addition of bFGF to the cells resulted in prolonged survival. Quantitative assessment of apoptosis-associated DNA strand breaks by in situ TdT labeling showed a protective effect of bFGF on fludarabine-treated cells. The potential effect of bFGF on bcl-2 mRNA expression was analyzed by Northern blotting. Stimulation with bFGF led to a time-dependent accumulation of bcl-2 specific mRNA in all three cell lines. Maximal levels of bcl-2 mRNA expression were detected after 8 h in JVM-2, and after 18 h in JVM-13 and I83CLL. Intracellular bcl-2 protein was also found to be increased upon bFGF stimulation in both EBV- and non-EBV-transformed cells. In addition, exposure of cells from three patients with B-CLL to bFGF showed an upregulation of bcl-2 protein after 4-8 h. Our data demonstrate that bFGF upregulates the expression of bcl-2 in these cells, suggesting that this increase in bcl-2 expression may play a role in the delay of fludarabine-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line, Transformed , DNA Fragmentation , Dactinomycin/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Tumor Cells, Cultured/drug effects , Vidarabine/analogs & derivatives , Vidarabine/antagonists & inhibitors , Vidarabine/pharmacology
14.
Mt Sinai J Med ; 64(1): 20-5, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8997072

ABSTRACT

BACKGROUND: It has long been suspected that sex steroids play a key role in the pathogenesis of benign prostatic hyperplasia (BPH). Prostatic diseases do not occur in males castrated before puberty or in males with heritable disorders of androgen production or action. Both estrogens and androgens have been shown to induce BPH in experimental animals. METHODS: Clinical studies utilizing hormonal therapies to treat BPH were reviewed. Studies that used total medical castration therapy via the use of a long-acting gonadotropin-releasing hormone (GnRH agonist), partial androgen blockade via the use of the 5 alpha-reductase inhibitor finasteride, and estrogen blockade (via the use of aromatase inhibitors) were analyzed. RESULTS AND CONCLUSIONS: Both the GnRH agonists and finasteride result in prostatic size reduction and alleviate symptoms in some patients. Both therapies are more effective in men with larger prostates (> 40 cc). Finasteride is less efficacious in terms of size reduction than the GnRH agonists but also has fewer side effects. To date, clinical trials with aromatase inhibitors have not yielded dramatic positive results in the treatment of BPH.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Enzyme Inhibitors/therapeutic use , Estrogen Antagonists/therapeutic use , Finasteride/therapeutic use , Leuprolide/therapeutic use , Prostatic Hyperplasia/drug therapy , Estrogens/metabolism , Humans , Male , Prostatic Hyperplasia/etiology , Testosterone/metabolism
15.
Growth Factors ; 15(1): 41-60, 1997.
Article in English | MEDLINE | ID: mdl-9401817

ABSTRACT

We investigated the roles of basic fibroblast growth factor (bFGF) in the transformation and survival of NIH 3T3 cells. We constructed NIH 3T3-derived cell lines expressing human bFGF using retroviral gene transfer with an N2-based vector. Clonally derived cell lines containing a single copy of the vector overexpress bFGF mRNA and produce more immunoreactive protein (0.407 +/- 0.010-3.028 +/- 0.087 ng bFGF/10(6) cells) which is biologically active than nontransduced (0.151 +/- 0.013 ng bFGF/10(6) cells) or N2-transduced (0.211 +/- 0.029 ng bFGF/10(6) cells) NIH 3T3 cells. All cells producing excess amounts of bFGF achieve greater density at confluence, show delayed apoptosis and increased survival and have elevated intracellular levels of Bcl-2. However, only cells expressing from 8-15 times background levels of bFGF are phenotypically transformed. The transformed cells form dense foci at confluence, have decreased adherence to tissue culture plates and grow colonies in soft agar. Exogenous bFGF induces higher Bcl-2 levels in a dose dependent manner and recapitulates the antiapoptotic effects of the overexpressed species but fails to induce changes associated with the transformed phenotype. In this study, we demonstrate a dissociation between phenotypic transformation secondary to bFGF overexpression and upregulation of cellular Bcl-2 that correlates with a delay in programmed cell death. Although low level expression of bFGF or exogenous bFGF is sufficient to upregulate Bcl-2 and delay apoptosis, high intracellular levels are required for cellular transformation. These data suggest that overexpression of bFGF modulates cellular transformation and Bcl-2-mediated inhibition of apoptosis through alternate molecular mechanisms.


Subject(s)
Cell Transformation, Neoplastic , Fibroblast Growth Factor 2/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , 3T3 Cells , Animals , Apoptosis , Cell Division , Cell Survival/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , DNA/analysis , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , Gene Expression Regulation , Humans , Immunohistochemistry , Mice , Proto-Oncogene Proteins/metabolism , RNA, Messenger/analysis , Up-Regulation , bcl-2-Associated X Protein
16.
Blood ; 83(4): 907-10, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-7509211

ABSTRACT

Stem cell factor (SCF) and basic fibroblast growth factor (bFGF) are hematopoietic cytokines produced by bone marrow stromal cells. It is known that, although SCF and bFGF have limited clonogenic activity on their own, they can augment colony-stimulating factor (CSF)-mediated progenitor cell growth. Because these factors are both sequestered by stromal cells, we examined their interaction on progenitor cell growth in conjunction with granulocyte-macrophage-CSF (GM-CSF). In this study, we show that clonogenic growth derived from low-density bone marrow cells stimulated by GM-CSF is significantly augmented (P < .001) in the presence of maximal (100 ng/mL) concentrations of SCF in combination with 100 ng/mL of bFGF. When CD34+ cells are used, the synergistic effect of bFGF and SCF for GM-CSF-mediated progenitor cell growth is further increased, resulting in as much as a sevenfold increase in detectable colony-forming units granulocyte-macrophage (P < .001). These data suggest that the synergistic activity of bFGF and SCF is mediated directly on hematopoietic precursors. These observations suggest that bFGF and SCF, concentrated locally on stromal cell surfaces, might interact in concert with other hematopoietic cytokines to regulate stem cell proliferation and differentiation in hematopoietic niches in the bone marrow.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/cytology , Antigens, CD/analysis , Antigens, CD34 , Bone Marrow Cells , Cell Adhesion Molecules/pharmacology , Cell Division/drug effects , Drug Synergism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/pharmacology , Recombinant Proteins/pharmacology , Stem Cell Factor
17.
J Immunother Emphasis Tumor Immunol ; 15(1): 59-66, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8110732

ABSTRACT

To assess the toxicity, pharmacokinetics, and local and systemic effects of the intraperitoneal (i.p.) administration of granulocyte-macrophage colony-stimulating factor (GM-CSF) at various dosages, 13 patients with predominantly i.p. malignancies refractory to standard chemotherapy were studied. GM-CSF was administered intravenously (i.v.) for 5 consecutive days; 21 days later the same dosage of GM-CSF was administered i.p. for 5 consecutive days. Four dosage levels were studied: 1, 2, 4, and 8 micrograms/kg/day. GM-CSF was well tolerated after i.v. and i.p. administration at doses up to 8 micrograms/kg/day. A transient fall followed by an elevation of circulating white cells was observed over a 24-h period after both i.v. and i.p. GM-CSF administration (mean minimum +/- SE as % baseline): 38 +/- 8% at 30 min after i.v. administration, 21 +/- 5% at 60 min after i.p. administration; mean maximum: 220 +/- 41% at 6 h after i.v. administration, 202 +/- 39% at 12 h after i.p. administration). The magnitude and time course of these changes were very similar for the two routes despite an up to 400-fold difference in serum GM-CSF levels at the same time points. Changes in leukocyte count and differential and neutrophil function were also similar over the 3-week period after both i.v. and i.p. administration. In the only patient who had i.p. GM-CSF levels assayed, i.p. administration achieved high levels of GM-CSF in peritoneal fluid (Cmax 343 ng/ml) with maintenance of high concentrations over 24 h (C24h 128 ng/ml).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunotherapy , Neoplasms/therapy , Adult , Aged , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/blood , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics
18.
Blood ; 82(2): 430-5, 1993 Jul 15.
Article in English | MEDLINE | ID: mdl-8329701

ABSTRACT

Basic fibroblast growth factor (bFGF), a multifunctional growth factor produced by bone marrow stromal cells, is known to be a potent modulator of hematopoiesis. Because bFGF is present in both human megakaryocytes (MKs) and platelets, we have hypothesized that this growth factor might affect human megakaryocytopoiesis. To test this hypothesis, either low density bone marrow (BM) cells (LDBM), a human BM subpopulation (CD34+ DR+) enriched for the colony-forming unit megakaryocyte (CFU-MK) or a BM subpopulation (CD34+ DR-) enriched for the more primitive burst-forming unit megakaryocyte (BFU-MK) were assayed in the presence of this growth factor. The effect of bFGF on MK colony formation differed according to the cell population assayed. bFGF alone had on MK colony-stimulating activity (MK-CSA) when either CD34+ DR+ or CD34+ DR- BM cells were cloned, but exhibited MK-CSA equivalent to that of interleukin-3 (IL-3) when LDBM cells were used as the target cell population. The MK-CSA of bFGF was inhibited by the addition of neutralizing antisera to either IL-3 and/or granulocyte-macrophage colony-stimulating factor (GM-CSF) but not IL-6. The addition of excess amounts of either IL-3 or GM-CSF to cultures containing bFGF plus anti-IL-3 or anti-GM-CSF reversed the inhibition by the corresponding antisera. The addition of bFGF and IL-3 to assays containing CD34+ DR+ or CD34+ DR- cells increased the size of both CFU-MK- and BFU-MK-derived colonies, respectively, when compared with assays containing IL-3 alone. This increase in MK colony size mediated by bFGF was not affected by addition of either an anti-GM-CSF or anti-IL-6 neutralizing antisera. When LDBM cells were assayed, bFGF alone increased CFU-MK-derived colony size when compared with control values. However, this potentiation of MK colony size by bFGF could be reversed by the addition of either anti-IL-3 or anti-GM-CSF but not anti-IL-6 antisera. In addition, the effects of bFGF and IL-3 on the size of MK colonies cloned from LDBM were not additive. These results suggest that bFGF affects human megakaryocytopoiesis by directly promoting MK progenitor cell proliferation and stimulating BM accessory cells to release growth factor(s) with MK-CSA, such as IL-3 and GM-CSF. We conclude that bFGF, likely produced by cellular components of the BM microenvironment, plays an important role in the control of human megakaryocytopoiesis.


Subject(s)
Bone Marrow Cells , Fibroblast Growth Factor 2/pharmacology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Antibodies , Bone Marrow/drug effects , Cell Division , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/immunology , Interleukin-3/pharmacology , Interleukin-6/immunology , Megakaryocytes/drug effects , Platelet Count , Recombinant Proteins/pharmacology
20.
Blood ; 81(7): 1745-54, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-7681702

ABSTRACT

Interleukin-1 (IL-1) modulation of cytokine receptors (human IL-1 receptor [hIL-1R], human granulocyte colony-stimulating factor [hG-CSFR], human granulocyte-macrophage CSF receptor [hGM-CSFR], and human tumor necrosis factor receptor [hTNFR]) on human neutrophils was studied both in vitro and in vivo. In vitro, incubation of neutrophils with IL-1 at 37 degrees C for 0.5 or 8 hours caused a reduction of IL-1 binding in a dose-dependent manner, but did not demonstrably affect binding of the other cytokines tested. In vivo, neutrophils from patients with gastrointestinal malignancies who were participating in a clinical trial of recombinant human IL-1 beta (rhIL-1 beta) demonstrated modulation of cytokine receptors in an IL-1 beta dose- and time-dependent manner. At the two highest dose levels of IL-1 beta (0.068 and 0.1 microgram/kg), reduction (> 40%) of G-CSF binding and elevation (twofold to sixfold) of IL-1 binding to neutrophils was observed after 1 hour and 4 to 8 hours, respectively. In addition, IL-1 beta rapidly elevated G-CSF and glucocorticoid levels in plasma. Patients at the lowest dose level (0.002 microgram/kg) had a less dramatic change in these parameters. Further in vitro studies showed that synthetic glucocorticoids and G-CSF synergistically up-modulated IL-1 binding to neutrophils in a dose- and time-dependent manner. Scatchard analysis of binding data showed that this in vitro synergistic modulation was due to an increase in receptor numbers, rather than an increase in binding affinity. In addition, both human umbilical cord blood and bone marrow neutrophils responded to G-CSF and dexamethasone (Dex) with a superadditive increase in IL-1 binding. Therefore, one of mechanisms for IL-1 up-modulation of IL-1R on human neutrophils in vivo was due to the fact that IL-1 rapidly elevates serum levels of G-CSF and glucocorticoids.


Subject(s)
Interleukin-1/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, Interleukin-1/drug effects , Animals , Cells, Cultured , Dexamethasone/pharmacology , Drug Synergism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Interleukin-1/metabolism , Mice , Protein Binding , Receptors, Cell Surface/drug effects , Receptors, Granulocyte Colony-Stimulating Factor/drug effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Receptors, Tumor Necrosis Factor , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...