Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cancer Res ; 59(3): 734-41, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973225

ABSTRACT

Expression and function of the TRAIL apoptotic pathway was investigated in normal and malignant breast epithelial cells. Glutathione-S-transferase (GST)-TRAIL extracellular domain fusion proteins were produced to analyze TRAIL-induced apoptosis. Only GST-TRAIL constructs containing regions homologous to the Fas self-association and ligand binding domains could induce apoptosis. GST-TRAIL induced significant (>90%) apoptosis in just one of eight normal and one of eight malignant breast cell lines. All other lines were relatively resistant to TRAIL-induced apoptosis. Activating TRAIL receptors DR4 and DR5 were expressed in all normal and malignant breast cell lines. The inhibitory receptor TRID was highly expressed in one of four normal and two of seven malignant breast cell lines. DR4, DR5, or TRID expression did not correlate with sensitivity to TRAIL-induced apoptosis. Incubation of cell lines with doxorubicin or 5-fluorouracil significantly augmented TRAIL-induced apoptosis in most breast cell lines. By fractional inhibition analysis, the toxicity of the combination of TRAIL and doxorubicin or 5-fluorouracil was synergistic compared with either agent alone. In contrast, melphalan and paclitaxel augmented TRAIL-induced apoptosis in few cell lines, and methotrexate did not augment it in any cell line. Augmentation of TRAIL-induced apoptosis by doxorubicin or 5-fluorouracil was mediated through caspase activation. This was evidenced by the fact that chemotherapy agents that synergized with TRAIL (e.g., doxorubicin) themselves caused cleavage of caspase-3 and poly(ADP-ribose) polymerase (PARP), and their toxicity was blocked by the caspase inhibitor Z-Val-Ala-Asp(OMe)-CH2 (ZVAD-fmk). The combination of TRAIL and doxorubicin caused significantly greater caspase-3 and PARP cleavage, and the combined toxicity also was inhibited by ZVAD-fmk. In contrast, chemotherapy agents that did not augment TRAIL-induced apoptosis (e.g., methotrexate) caused minimal caspase-3 and PARP cleavage by themselves, and their toxicity was not inhibited by ZVAD-fmk. These drugs also did not increase caspase-3 or PARP cleavage when combined with TRAIL. In summary, few breast cell lines are sensitive to TRAIL-induced apoptosis, and no difference in sensitivity is found between normal and malignant cell lines. Treatment with chemotherapy provides an approach to sensitize breast cancer cells to TRAIL-induced apoptosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Antineoplastic Agents/administration & dosage , Apoptosis Regulatory Proteins , Caspase 3 , Caspases/metabolism , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Enzyme Activation , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Glutathione Transferase/pharmacology , Glutathione Transferase/physiology , Humans , Melphalan/administration & dosage , Melphalan/pharmacology , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/physiology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/physiology
2.
Clin Cancer Res ; 4(9): 2095-102, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9748125

ABSTRACT

The mammalian pulmonary toxin 4-ipomeanol (IPO) is activated by the cytochrome P450 system in bronchial Clara cells in animals. The resulting metabolites bind rapidly to macromolecules, producing localized cytotoxicity. IPO has in vitro and in vivo antitumor activity in non-small cell lung cancer (NSCLC) and thus was proposed as a lung cancer-specific antitumor agent. We have completed a directed Phase I trial in patients with NSCLC. Forty-four patients (34 men and 10 women) with NSCLC were treated with IPO. All but two patients had an Eastern Cooperative Oncology Group performance status of 0 or 1. They received 91 courses of therapy with i.v. IPO; 82 courses were administered daily for five days, and 9 were single bolus doses. The dose-limiting toxicity of elevated serum transaminases was observed in three of seven patients at 922 mg/m2/day. The maximum tolerated dose was 693 mg/m2/day on 5 consecutive days every 3 weeks. One patient developed grade 4 pulmonary toxicity at 167 mg/m2/day. There was no significant hematological or renal toxicity. No objective antitumor responses were observed. Pharmacokinetic analysis of 39 patients from day 1 of IPO administration showed biexponential elimination with mean half-lives of 8.6 (alpha half-life) and 76 min (beta half-life). There was a linear relationship between the area under the plasma drug concentration-time curve and the dose of IPO. There was no significant difference between the pharmacokinetic parameters measured on day 1 and day 5. Using a 4-day in vitro cytotoxicity assay, two tumor cell lines established from patients treated at 693 mg/m2/day had IC50s of approximately 6 mM, a concentration more than 75-fold higher than the plasma levels measured in these patients. Thus, although the total amount of drug administered per cycle on a daily times five dose schedule is more than 2.5-fold higher than the recommended single daily dose, IPO is unlikely to be a useful drug for patients with lung cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Terpenes/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Female , Humans , Lung Diseases/chemically induced , Male , Middle Aged , Partial Thromboplastin Time , Terpenes/adverse effects , Terpenes/pharmacokinetics
3.
Clin Cancer Res ; 3(3): 449-54, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815704

ABSTRACT

Paclitaxel blocks cells in G2-M, and this may result in a schedule-dependent effect on paclitaxel cytotoxicity. To test this hypothesis, we evaluated paclitaxel cytotoxicity in 28 human lung cancer cell lines. Fourteen of the cell lines were derived from patients with non-small cell lung cancer (NSCLC), and 14 were from patients with small cell lung cancer (SCLC). All cell lines were exposed to a range of paclitaxel concentrations for durations of 3, 24, and 120 h, and cytotoxicity was measured with a tetrazolium-based assay. The median IC50 values for all 28 cell lines at exposure durations of 3, 24, and 120 h were >32 microM, 23 microM, and 0.38 microM, respectively. The median IC50 values for the NSCLC cell lines were >32 microM, 9.4 microM, and 0.027 microM at exposure durations of 3, 24, and 120 h, respectively. For the 14 SCLC cell lines, the median IC50 values were >32 microM, 25 microM, and 5.0 microM, respectively. Five of the 14 SCLC cell lines had IC50 values at 120 h of paclitaxel exposure that were 1000-fold less than the remaining SCLC cell lines. The median IC50 values for these five sensitive SCLC cell lines at 3-, 24-, and 120-h exposures were >32 microM, 23 microM, and <0.0032 microM, respectively. These in vitro cytotoxicity results were independent of the paclitaxel diluent, a 1:1 solution of ethanol and Cremophor EL. We conclude that longer durations of paclitaxel exposure result in an increase in the chemosensitivity of some human lung cancer cell lines and that this phenomenon is more consistent within NSCLC cell lines than in SCLC cell lines.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/pathology , Cell Cycle/drug effects , Cell Survival/drug effects , Lung Neoplasms/pathology , Paclitaxel/toxicity , G2 Phase , Humans , Mitosis , Time Factors , Tumor Cells, Cultured
4.
Cancer Res ; 56(20): 4791-8, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8841000

ABSTRACT

Expression and function of the Fas apoptotic pathway was investigated in normal and malignant human breast epithelial cells. Nontransformed mammary epithelial cell lines all expressed high levels of Fas mRNA and protein, but only one of seven breast cancer cell lines (T47D) expressed high levels of Fas. Apoptosis was induced in the nontransformed lines when they were incubated with the anti-Fas antibody. However, all of the breast cancer cell lines tested, except T47D, were resistant to Fas-mediated apoptosis. Four of five Fas-resistant breast cancer cell lines became sensitive to Fas-mediated apoptosis upon treatment with IFN-gamma. Fas mRNA increased slightly in both cell lines that became sensitive and in the cell line that remained resistant to Fas-mediated apoptosis upon IFN-gamma treatment. However, the cell surface expression of Fas showed little or no increase in any of the cell lines tested upon IFN-gamma treatment. In contrast to Fas expression, interleukin-1beta-converting enzyme (ICE) expression increased only in the cell lines that became Fas sensitive after IFN-gamma treatment. The importance of ICE and/or ICE-like proteases in Fas-mediated apoptosis in these cells was confirmed by inhibition of Fas-mediated apoptosis by a specific ICE inhibitor, YVAD-cmk. Fas sensitivity was reconstituted in the IFN-gamma-resistant cell line by transfection of ICE into that cell line. Together, these data suggest that down-regulation of Fas and its pathway may be a step in tumor progression and that modulation of Fas expression may provide an approach to inducing apoptosis in breast cancer cells.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast/metabolism , Cysteine Endopeptidases/biosynthesis , Interferon-gamma/pharmacology , fas Receptor/physiology , Apoptosis/drug effects , Blotting, Northern , Blotting, Western , Breast/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Caspase 1 , Cysteine Endopeptidases/genetics , Enzyme Induction/drug effects , Female , Humans , Transfection , Tumor Cells, Cultured , fas Receptor/metabolism
5.
J Cell Biochem Suppl ; 24: 24-31, 1996.
Article in English | MEDLINE | ID: mdl-8806091

ABSTRACT

More than 200 human small cell lung cancer and non-small cell lung cancer cell lines were established over 15 years mainly by utilizing the serum-free, hormone and growth factor supplemented, defined media HITES and ACL4. Use of modified, established cell culture techniques such as the mechanical spillout method for the releasing of cell aggregates from tumor tissue, ficoll gradient centrifugation for the separation of tumor cells from erythrocytes and tissue debris, and an apparatue consisting of a platinum tubing attached to a suction flask for removal of spent medium have greatly contributed to the success in culturing tumor cells. Characterization of these lung cancer cell lines have extended our knowledge of lung cell biology. Studies elucidating the nutritional requirements of lung cancer cell growth may be helpful for the manipulation of these tumors in patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/pathology , Cell Culture Techniques/methods , Lung Neoplasms/pathology , Ascites/pathology , Bone Marrow/pathology , Cell Culture Techniques/instrumentation , Culture Media, Serum-Free , Humans , National Institutes of Health (U.S.) , Pleural Effusion/pathology , Specimen Handling , Tumor Cells, Cultured , United States
6.
J Cell Biochem Suppl ; 24: 173-85, 1996.
Article in English | MEDLINE | ID: mdl-8806100

ABSTRACT

Clinical protocols for small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) were devised to prospectively select individualized chemotherapy based on in vitro drug sensitivity testing (DST) of cell lines derived from the patient's SCLC tumor cell lines or the patient's fresh NSCLC tumor. DST data derived from SCLC tumor cell lines were available for 33/115 (29%) patients. The DST-selected chemotherapy regimen was administered to 21 (18%) patients, or 64% of patients with DST. In SCLC< the DST-selected chemotherapy was administered either during weeks 13-24 following 12 weeks of etoposide/cisplatin, or at relapse after complete response to etoposide/cisplatin. Several parameters of in vitro drug sensitivity were significantly associated (two-sided P < 0.05) with clinical response to primary therapy and also with response to the DST-selected chemotherapy regimen, but were not associated with survival (P = 0.24). Five patients treated with their DST-selected chemotherapy attained a complete or partial response, compared to 5 of 68 who received an empiric regimen (P = 0.057). A total of 36/165 (22%) NSCLC patients had DST successfully completed. These results directed management for 21/96 (22%) patients who eventually received chemotherapy, or 58% of patients with DST. Response to chemotherapy for the patients treated prospectively with their DST-selected chemotherapy regimen (2/21; 9%) was not significantly different than the response rate for patients treated empirically with etoposide/cisplatin (10/69; 14%) in the absence of in vitro results to direct chemotherapy (P = 0.73). There was no difference in survival by treatment group for the NSCLC patients. The correlation between in vitro and clinical response was not significant for any individual drug or for all drugs considered together, illustrating the poor predictive value of in vitro testing with currently available chemotherapy in NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Lung Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/pathology , Carcinoma, Small Cell/radiotherapy , Carcinoma, Small Cell/surgery , Carmustine/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Etoposide/administration & dosage , Etoposide/pharmacology , Evaluation Studies as Topic , Humans , Lomustine/administration & dosage , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Lung Neoplasms/surgery , Mechlorethamine/pharmacology , Methotrexate/administration & dosage , Palliative Care , Predictive Value of Tests , Prospective Studies , Specimen Handling , Survival Analysis , Treatment Outcome , Tumor Cells, Cultured/drug effects , Vincristine/administration & dosage , Vincristine/pharmacology
7.
Cell Growth Differ ; 4(10): 799-809, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8274449

ABSTRACT

Retinoic acid (RA) and nuclear retinoic acid receptors (RARs) have been implicated in a variety of human malignancies including lung cancer, and RA has been proposed as a chemopreventive agent for bronchogenic carcinoma. Normal human tracheobronchial epithelial cells show dramatic induction of RAR-beta mRNA and significant growth inhibition after RA treatment. In contrast, 17 of 22 small cell lung cancer (SCLC) and 9 of 15 non-SCLC lines treated with 1 microM RA showed no significant growth inhibition. Of interest, 5 SCLC lines with high levels of myc gene family expression related to c-, N-, or L-myc gene amplification exhibited growth inhibition (28-87%), whereas 2 non-SCLC lines actually showed growth stimulation after treatment with 1 microM RA. The lines varied greatly in their constitutive expression of RAR-beta mRNA, and 15 of 20 SCLC and 8 of 15 non-SCLC lines failed to show RAR-beta mRNA induction after RA treatment. Six cell lines showed possible alterations in the coding region of RAR-beta by complementary DNA (cDNA)/polymerase chain reaction (PCR) analysis using primers common to the RAR-beta1,2,3 isoforms, since other regions would undergo cDNA/PCR amplification whereas the DNA binding domain would not. Nonetheless, no abnormal band shift patterns in cDNA amplified by PCR were found by single strand conformation polymorphism analysis covering all 1344 base pairs of the RAR-beta open reading frame. Finally, no abnormalities in RAR-alpha gene structure or expression were identified by Southern and Northern blot analysis, including lines with cytogenetic abnormalities of 17q21. We conclude that abnormalities of the RAR-beta system are common in human lung cancer cell lines.


Subject(s)
Lung Neoplasms/drug therapy , RNA, Messenger/biosynthesis , Receptors, Retinoic Acid/genetics , Tretinoin/therapeutic use , Base Sequence , Blotting, Southern , Cell Division/drug effects , DNA, Complementary/genetics , Drug Resistance , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Cancer Res ; 53(18): 4169-71, 1993 Sep 15.
Article in English | MEDLINE | ID: mdl-8364910

ABSTRACT

IA-1 is a recently isolated novel complementary DNA which encodes a protein of 510 amino acids that contains both a zinc finger DNA-binding domain and a putative prohormone domain. mRNA expression of IA-1 has been found thus far only in tumors of neuroendocrine origin. In this report we describe the expression of IA-1 mRNA in a panel of 64 human lung cancer cell lines. IA-1 mRNA was detected by Northern blot analysis in 97% (30 of 31) of small cell lung cancer cell lines. In contrast, IA-1 mRNA was detected in only 13% (4 of 30) of non-small cell lung cancer cell lines. Nine of the 30 (30%) expressed either chromogranin A mRNA or produced L-dopa decarboxylase. Four of these 9 (44%) had detectable levels of IA-1 mRNA. In most of the lung cancer cell lines examined, IA-1 showed high concordance with the other neuroendocrine markers, L-dopa decarboxylase, and chromogranin A. The one exception was a variant small cell lung cancer cell line which expressed low or nondetectable levels of L-dopa decarboxylase. IA-1 is a candidate marker of neuroendocrine differentiation of human lung tumors.


Subject(s)
Biomarkers, Tumor/analysis , DNA/analysis , Lung Neoplasms/chemistry , Cell Differentiation , Chromogranin A , Chromogranins/analysis , Dopa Decarboxylase/analysis , Humans , Lung Neoplasms/pathology , RNA, Messenger/analysis , Tumor Cells, Cultured
9.
Cancer Res ; 50(18): 6068-74, 1990 Sep 15.
Article in English | MEDLINE | ID: mdl-2168288

ABSTRACT

Synaptophysin is a Mr 38,000 integral membrane glycoprotein expressed by a variety of normal and neoplastic neuroendocrine cells. We studied synaptophysin as an immunocytochemical marker for neuroendocrine differentiation in lung cancer and compared it to the immunocytochemical expression of chromogranin A, a marker for dense core (endocrine) granules, and the biochemical activity of L-dopa decarboxylase (DDC), the key amine-handling enzyme. Of the 250 cell lines available to us, we selected examples representative of the following cell types: bronchial carcinoids (n = 4), small cell lung cancer (SCLC) (n = 7), extrapulmonary small cell carcinomas (n = 4), and non-small cell lung cancers (n = 18) whose neuroendocrine status had been previously determined on the basis of electron microscopy and DDC activity. We demonstrated (a) there was a higher incidence of synaptophysin than chromogranin A immunoreactivity in carcinoid (100 versus 75%), classic SCLC (70 versus 50%), and variant SCLC (57 versus 29%) cell lines; (b) 3 of the 4 (75%) extrapulmonary small cell lung cancer cell lines expressed synaptophysin and chromogranin A; (c) 5 of the 7 (71%) non-small cell lung cancer cell lines previously shown to express multiple neuroendocrine markers were positive for synaptophysin, chromogranin A, and DDC activity; (d) none of the other 11 non-small cell lung cancer cell lines expressed synaptophysin or chromogranin A; and (e) formalin fixation and paraffin embedding reduced synaptophysin immunoreactivity in 11 of 14 (79%) of the cell lines, as compared to freshly prepared specimens fixed in 95% ethanol. Western blot analysis using the synaptophysin antibody (SY38) demonstrated immunoreactive proteins ranging from Mr 43,000 to 45,000 in five representative cell lines. The concordance of expression of all three neuroendocrine markers was statistically significant when values for all cell lines were totalled. Synaptophysin was a more commonly expressed marker for variant SCLC cell lines, which rarely showed DDC activity. We conclude that synaptophysin may be a more sensitive and specific marker for neuroendocrine differentiation, when compared to chromogranin A and DDC in lung cancer cell lines which express only part of the neuroendocrine program.


Subject(s)
Aromatic-L-Amino-Acid Decarboxylases/analysis , Chromogranins/analysis , Dopa Decarboxylase/analysis , Lung Neoplasms/analysis , Membrane Proteins/analysis , Nerve Tissue Proteins/analysis , Neurosecretory Systems/analysis , Carcinoid Tumor/analysis , Carcinoma, Non-Small-Cell Lung/analysis , Carcinoma, Small Cell/analysis , Cell Differentiation , Humans , Lung Neoplasms/pathology , Membrane Proteins/immunology , Molecular Weight , Synaptophysin , Tumor Cells, Cultured
10.
J Natl Cancer Inst ; 82(2): 117-24, 1990 Jan 17.
Article in English | MEDLINE | ID: mdl-2152944

ABSTRACT

We devised a novel clinical protocol for extensive-stage small cell lung cancer (SCLC), selecting chemotherapy whenever possible on the basis of in vitro drug-sensitivity testing (DST) of individual patients' tumor specimens. Most of the specimens were obtained from metastatic sites during routine staging procedures. Increase of tumor cell number by culture in selective media usually was required before DST could be performed. We used the Weisenthal dye exclusion assay to place the seven drugs in rank order and to select the in vitro best regimen (IVBR), a three-drug combination of proved efficacy in SCLC. After initial staging and specimen acquisition, patients received etoposide and cisplatin (primary therapy) and were restaged after 12 weeks. Patients with partial or no responses and those relapsing after a complete response to primary therapy were switched to the IVBR if DST data were available. If DST data were unavailable, an empiric combination, vincristine-doxorubicin-cyclophosphamide, was administered as secondary therapy. Tumor-containing specimens were collected from 60 of the 80 patients (75%). One or more cell lines were established from 28 patients, and DST data were available from 26 patients (33% of total). Several parameters of in vitro drug sensitivity were significantly associated [two-sided P (P2) less than .05] with clinical response to primary therapy and also with response to the IVBR and were marginally associated with length of survival (.07 less than or equal to P2 less than or equal to .08). Sixteen patients (23%) received their IVBR as secondary therapy, and four of these (25%) attained a complete response, compared with three of 43 (7%) who received an empiric regimen (P2 = .16). We concluded that (a) selection of individualized chemotherapy is labor intensive but feasible in extensive-stage SCLC; (b) DST data are associated with clinical response to primary therapy and to secondary therapy with an IVBR; and (c) further observations will be required if we are to determine whether there is a modest therapeutic benefit to administering the IVBR as a secondary therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/drug therapy , Drug Screening Assays, Antitumor , Lung Neoplasms/drug therapy , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/pathology , Clinical Protocols , Clinical Trials as Topic , Coloring Agents , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasm Staging , Prospective Studies , Tumor Cells, Cultured/drug effects
11.
J Clin Oncol ; 7(7): 923-31, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2544686

ABSTRACT

The ability to establish a continuously growing tumor cell line from fresh tumor specimens has been associated with shortened survival in some human malignancies. Therefore, we assessed the relationship between survival and in vitro tumor cell growth from specimens obtained during routine staging procedures in 68 consecutive patients with untreated, extensive-stage small-cell lung cancer (SCLC) who received etoposide/cisplatin chemotherapy. Three groups of SCLC patients could be distinguished: (1) 23 patients in whom a tumor cell line was established in vitro; (2) 28 patients in whom tumor-containing specimens were cultured but in vitro growth did not occur; and (3) 17 patients in whom no tumor-containing specimen could be procured. No significant difference in response rates to chemotherapy of the three groups was noted. Poor performance status (P2 = .001), male gender (P2 = .0008), liver metastases (P2 = .0033), brain metastases (P2 = .0152), and the ability to obtain a tumor-containing specimen from the patient for laboratory culture (P2 = .0005) were all significant independent predictors of decreased survival in this patient population. While the ability to obtain a tumor cell specimen for cell culture using routine staging and diagnostic procedures identified patients with shortened survival, we found no significant survival differences between patients whose tumor cell specimens grew in cell culture v those that did not (median survival of 7 months v 11 months, P2 = .72). Our study indicates that the clinical outcome of extensive-stage SCLC patients from whom tumor cell lines can be established is not significantly different than in those cases from whom tumor-containing specimens could not be grown in vitro.


Subject(s)
Carcinoma, Small Cell/pathology , Lung Neoplasms/pathology , Tumor Cells, Cultured/cytology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/mortality , Cell Division/drug effects , Cisplatin/administration & dosage , Clinical Trials as Topic , Etoposide/administration & dosage , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , Prognosis , Prospective Studies , Random Allocation
12.
Cancer Res ; 48(14): 4078-82, 1988 Jul 15.
Article in English | MEDLINE | ID: mdl-3383200

ABSTRACT

We evaluated the usefulness of L-dopa decarboxylase (DDC) as a tumor marker of neuroendocrine (NE) cell differentiation by measuring its expression in 432 human tumors of diverse types and origins. A subset of these tumors and cell lines derived from them also were studied for expression of two other general NE cell markers, chromogranin A (CgA) and dense core granules (DCG). High concentrations of DDC were present in 96 of 117 (82%) tumors recognized to be of NE or neural origin. As expected, endocrine tumors not recognized to be of NE cell origin, as well as leukemias, lymphomas, sarcomas, melanomas, and germ cell tumors, lacked DDC expression. Of interest, modest concentrations of DDC were present in 46 of 220 (21%) nonendocrine carcinomas, especially non-small cell lung and colorectal carcinomas. We studied concordant expression of the three NE cell markers in lung and colorectal tumors and cell lines. In both tumor types there was nearly 100% concordance between CgA and DCG expression. There was an excellent correlation between DDC and CgA expression in lung cancers, both small cell and non-small cell, but DDC positive colorectal carcinomas usually lacked CgA expression. We conclude: (a) DDC is an excellent cellular marker for tumors of the NE cell system; (b) about 20% of carcinomas not of NE cell origin, especially non-small cell lung and colorectal carcinomas, express DDC, suggesting a common endodermal origin of all of the respiratory and gastrointestinal mucosal cells; and (c) CgA and DCG are expressed concordantly, indicating that CgA expression may be used as a substitute for ultrastructural examination of tumors for DCG expression.


Subject(s)
Aromatic-L-Amino-Acid Decarboxylases/biosynthesis , Biomarkers, Tumor/analysis , Chromogranins/biosynthesis , Cytoplasmic Granules/ultrastructure , Dopa Decarboxylase/biosynthesis , Neoplasms/analysis , Nerve Tissue Proteins/biosynthesis , Neurosecretory Systems/metabolism , Cell Differentiation , Chromogranin A , Humans , Microscopy, Electron , Neoplasms/ultrastructure
13.
Cancer Treat Rep ; 71(7-8): 697-704, 1987.
Article in English | MEDLINE | ID: mdl-3038314

ABSTRACT

Human lung cancer cell lines, established from patients with both small cell cancer (SCLC) and non-small cell cancer (non-SCLC; squamous cell, large cell, anaplastic, and adenocarcinoma), were tested for their in vitro chemosensitivity to a panel of drugs. Drug sensitivity was assayed by either soft agar clonogenicity or a novel dye-exclusion assay. Eleven non-SCLC lines (eight continuous, three recently cultured) and five SCLC lines (all continuous) were tested. Four of eight continuous non-SCLC lines cloned sufficiently to permit limited in vitro drug testing, as did two of the five SCLC lines. All 16 cell lines could be tested for multiple drugs using the dye-exclusion assay. Drug concentrations for the nonclonogenic assay more closely approximated the area under the concentration-time curve for a given concentration of each agent. There was considerable variation in the relative sensitivity of the cell lines and the patterns of individual drug sensitivity. The majority of non-SCLC cell lines were refractory to most drugs. Cell lines derived from two previously treated SCLC patients and from three untreated SCLC patients showed greater sensitivity. Concurrent clonogenic and dye-exclusion assays showed similar drug rankings but different absolute values for percent survival. The nonclonogenic dye-exclusion assay is more rapid than the soft agar clonogenic assay (4 days vs. 2-3 weeks), could be performed on all cell lines tested, and appears to reflect the clinical diversity of human lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/pathology , Colony-Forming Units Assay , Lung Neoplasms/pathology , Tumor Stem Cell Assay , Biotransformation , Cell Division/drug effects , Cell Line/drug effects , Cell Survival/drug effects , Cyclophosphamide/pharmacology , Drug Evaluation, Preclinical/methods , Humans
14.
Life Sci ; 32(5): 487-93, 1983 Jan 31.
Article in English | MEDLINE | ID: mdl-6296591

ABSTRACT

High intracellular levels of BN-like peptides are present in tumors and cell lines of small cell carcinoma of the lung (SCCL) as well as the putative precursor cells of this tumor, the pulmonary endocrine cell. In cell line NCI-H209 the density of bombesin-like peptides was 8.9 +/- 1.1 pmol/mg total protein. Gel filtration chromatography of an extract of these cells revealed one major peak of immunoreactivity which coeluted with synthetic bombesin (1620 daltons). Also, high pressure liquid chromatography revealed one major peak of immunoreactivity was present which eluted before synthetic peptide. Therefore, SCCL bombesin-like peptides may be of similar size but are more hydrophilic than synthetic peptide. Cells maintained in culture continuously release bombesin-like peptides into the growth medium. Also, high concentrations of K+ stimulated the secretion of immunoreactive bombesin from cell lines in a Ca++-dependent manner. These SCCL bombesin-like peptides may function as important regulatory agents in the malignant lung.


Subject(s)
Bombesin/metabolism , Carcinoma, Small Cell/metabolism , Lung Neoplasms/metabolism , Peptides/metabolism , Bombesin/isolation & purification , Calcium Chloride/pharmacology , Cell Line , Humans , Peptides/isolation & purification , Potassium Chloride/pharmacology , Radioimmunoassay , Veratridine/pharmacology
15.
Endocrinology ; 111(2): 684-92, 1982 Aug.
Article in English | MEDLINE | ID: mdl-6284489

ABSTRACT

Activation of T- and B-lymphocytes by a variety of immunological stimuli has been reported to induce specific insulin receptors. The purpose of the present work was to determine whether glucagon receptors are also induced in activated cells. Studies of glucagon and insulin receptors were carried out using normal human mononuclear cells activated by phytohemagglutinin or T-cell growth factor (TCGF), as well as established B- and T-lymphoblastoid cell lines. With phytohemagglutinin, glucagon and insulin binding increased 15- and 36-fold, respectively, and peaked after 5 days in parallel with the rise in thymidine incorporation. Increased binding was associated with an increase in the number of receptors, most marked for insulin, though affinity for the insulin receptor was decreased. Normal human mononuclear cells cultured with TCGF showed an early modest rise in insulin binding due to increased receptor number, without a change in affinity, and a striking and progressive rise up to 50-fold in glucagon binding due to both increased receptor number and affinity. The differences in receptor response to these T-cell mitogens suggest that TCGF selects out a T-lymphoblast subset with very high glucagon receptors. B- and T-lymphoblastoid cells showed patterns of glucagon and insulin receptors that appear to be characteristic for each cell type. Glucagon binding was 7-fold higher (P less than 0.01), while inulin binding was 7-fold lower (P less than 0.01) in T- vs. B-lymphoblastoid cells. T-Cell lines had twice the number of glucagon receptors, whereas B-lines had 4-fold the number of insulin receptors, with much greater affinity for insulin compared with T-line insulin receptors. Induction of both insulin and glucagon receptors on activated lymphoblasts suggests that these receptors may play a significant role in cell function.


Subject(s)
B-Lymphocytes/metabolism , Receptor, Insulin/blood , Receptors, Cell Surface/metabolism , T-Lymphocytes/metabolism , Bacitracin/pharmacology , Glucagon/blood , Humans , Insulin/blood , Interleukin-1 , Lymphocyte Activation , Monocytes/metabolism , Phytohemagglutinins/pharmacology , Proteins/pharmacology , Receptors, Glucagon , Time Factors
18.
Cancer Treat Rep ; 63(4): 587-90, 1979 Apr.
Article in English | MEDLINE | ID: mdl-221120

ABSTRACT

It is relatively easy to propagate EB virus-transformed normal and malignant B lymphocytes in vitro. Normal T lymphocytes divide for short periods of time after exposure to many mitogens. Exposure of normal T lymphocytes to lymphocyte-conditioned medium (LCM) permits them to divide for longer periods of time, but permanent cell lines cannot be established. Intial attempts to grow malignant T cells from patients with cutaneous T-cell lymphomas (CTCL) in unsupplemented growth medium resulted in the establishment of four EB virus-transformed B-lymphoblastoid lines. The responses of CTCL cells to mitogens and LCM varied from unresponsive to near normal. Subsequent attempts to grow CTCL cells in medium supplemented with mitogens or LCM resulted in the establishment of two cell lines lacking B-cell markers or EB virus. The cells of both lines initially had the ability to form E rosettes, although one of the lines has lost this ability with passage. The two lines can be distinguished from normal T cells by having some or all of the following properties: long-term proliferation, tumorigenecity in nude mice, gradual loss of dependence on mitogen or LCM, and convoluted nuclear morphology.


Subject(s)
Cell Line , Mycosis Fungoides , Sezary Syndrome , Skin Neoplasms , Animals , B-Lymphocytes/microbiology , Cell Transformation, Viral , Herpesvirus 4, Human/isolation & purification , Humans , Mice , Mice, Nude , Mitogens , Neoplasm Transplantation , Neoplasms, Experimental , T-Lymphocytes , Transplantation, Heterologous
20.
J Natl Cancer Inst ; 56(2): 423-6, 1976 Feb.
Article in English | MEDLINE | ID: mdl-176389

ABSTRACT

Host-range properties of xenotropic (x-tropic) and ecotropic mouse type-C viruses were determined. NB-tropic viruses replicated only in mouse and rat cells, whereas x-tropic viruses, which do not exogenously infect mouse cells, grew in cells from various other mammalian species. Great variability in susceptibilities to x-tropic viruses was demonstrated in cells from heterologous species and in cells from a single species (human). Although rat cells were susceptible to both types of viruses, hamster cells were uniformly resistant. In addition, the x-tropic viruses crossed class barriers to infect cells of avian species but not insect, fish, or reptile cells.


Subject(s)
Retroviridae/growth & development , Species Specificity , Animals , Birds , Cells, Cultured , Insecta , Mammals , Mice , Rats , Reptiles , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...