51
|
Tang P, Hung MC, Klostergaard J. ADP-ribosylation inhibitors inhibit cellular RNA synthesis but do not affect expression of manganous superoxide dismutase or heat shock protein 70 in tumor necrosis factor alpha-sensitive and -resistant tumor cells. J Interferon Cytokine Res 1995; 15:791-7. [PMID: 8536107 DOI: 10.1089/jir.1995.15.791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We have shown that the cytotoxic response of TNF-sensitive L929 cells and TNF-resistant EMT-6 cells to TNF-alpha can be modulated by ADP-ribosylation inhibitors independently of ADP-ribosylation rates. To explore the possibility that these inhibitors modulate TNF cytotoxicity by interfering with cellular protective mechanisms, we evaluated their effects on general RNA synthesis and on mRNA expression of two proposed protective genes, manganous superoxide dismutase (MnSOD) and heat shock protein 70 (hsp70). We found that ADP-ribosylation inhibitors could inhibit general RNA synthesis in a dose-dependent fashion to a similar extent in both EMT-6 and L929 cells, although these inhibitors increased or decreased the sensitivity of the cells to TNF, respectively. In EMT-6 cells, combination of actinomycin D with these inhibitors further inhibited the RNA synthesis rate, and it actually decreased the TNF sensitivity of the EMT-6 cells. Furthermore, the expression of MnSOD or hsp70 was not regulated by these inhibitors. Thus, TNF resistance must depend on other mechanisms in addition to the expression of these protective genes.
Collapse
Affiliation(s)
- P Tang
- Department of Tumor Biology, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | |
Collapse
|
52
|
van Slooten HJ, Bonsing BA, Hiller AJ, Colbern GT, van Dierendonck JH, Cornelisse CJ, Smith HS. Outgrowth of BT-474 human breast cancer cells in immune-deficient mice: a new in vivo model for hormone-dependent breast cancer. Br J Cancer 1995; 72:22-30. [PMID: 7599056 PMCID: PMC2034136 DOI: 10.1038/bjc.1995.271] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The effect of co-inoculation of basement membrane matrix, Matrigel and two human breast cancer cell lines, BT-474 and SK-BR-3, was tested in immune-deficient mice. Both cell lines strongly overexpress c-ErbB-2 protein, whereas only BT-474 is reported to be oestrogen receptor positive. Co-inoculation of Matrigel and BT-474 cells but not of Matrigel and SK-BR-3 cells resulted in tumour formation in bg-nu-xid mice. Oestrogen supplementation greatly enhanced tumorigenicity, but did not seem to be an absolute requirement. In vivo, BT-474 cells grow as a poorly differentiated adenocarcinoma with a doubling time of 9.4 +/- 1.1 days after inoculation into the neck region. A high proliferative activity appears to be compensated by a relatively high rate of cell loss, as BT-474 tumours contain many cells with the typical morphology of apoptotic cell death. Wild-type p53, known to participate in the induction of apoptosis, is absent from the tumours, whereas Bcl-2, known to inhibit apoptosis, is expressed at intermediate levels. BT-474 tumours tend to metastasise to the regional lymph nodes and are capable of forming micrometastatic lesions in the lung. Flow cytometrical analysis of DNA ploidy demonstrated no change in tumours compared with the cell line. Immunohistochemical and flow cytometrical detection of a number of hormone and growth factor receptors, transcription factors, cell adhesion molecules and proteins involved in proliferation and cell death demonstrated no major changes in ploidy and phenotype of tumours compared with the cell line. High expression of the cell-surface molecules c-ErbB-2 and episialin make it a potentially useful model for research in immune therapy.
Collapse
Affiliation(s)
- H J van Slooten
- Department of Surgery, Leiden University Hospital, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
53
|
Abstract
Over-expression of the major heat-shock protein hsp70 in WEHI-S tumor cells renders them resistant to the cytotoxic effects of tumor necrosis factor (TNF). To study the significance of this resistance in vivo, the tumorigenic potential of WEHI-S cells transfected with human hsp70 in sense and anti-sense orientation was investigated in athymic and in normal syngenic mice. A striking correlation was observed between the level of hsp70 expression and tumorigenicity in athymic mice. Hsp70 expression rendered WEHI cells tumorigenic also in normal mice, but higher numbers of cells were required for tumor formation than in athymic mice. Over-expression of hsp70 in WEHI-S cells did not enhance their anchorage-dependent growth in vitro or their ability to form colonies in soft agar. The hsp70-transfected cells exhibited greatly increased resistance against killing by murine natural cytotoxic cells and macrophages in vitro. A similar tumorigenic phenotype could also be induced independently of hsp70 by prolonged culture of WEHI-S cells with TNF. These results suggest that over-expression of hsp70 increases the tumorigenic potential of WEHI-S cells in mice, by allowing these cells to escape from the early TNF-mediated anti-tumor immune surveillance.
Collapse
Affiliation(s)
- M Jäättelä
- Department of Tumor Cell Biology, Danish Cancer Society Research Center, Copenhagen
| |
Collapse
|
54
|
Delvenne P, al-Saleh W, Gilles C, Thiry A, Boniver J. Inhibition of growth of normal and human papillomavirus-transformed keratinocytes in monolayer and organotypic cultures by interferon-gamma and tumor necrosis factor-alpha. THE AMERICAN JOURNAL OF PATHOLOGY 1995; 146:589-98. [PMID: 7887441 PMCID: PMC1869174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The growth response of normal and human papillomavirus (HPV)-transformed cervical keratinocytes to interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha was investigated in monolayer and organotypic raft cultures. The proliferation rates of monolayer cultures were assessed by [3H]TdR incorporation and fluorimetric DNA titration. The growth of keratinocytes in organotypic cultures was estimated by their ability to stratify on collagen rafts and by immunohistochemistry for Ki67 antigen expression. IFN-gamma reduced the DNA synthesis of normal and HPV-transformed keratinocytes in monolayer cultures and exerted a marked growth inhibitory effect in organotypic raft cultures. In control raft cultures, normal keratinocytes produced an epithelial sheet of approximately 10 cells in thickness that closely resembled normal cervical epithelium and was characterized by sparse Ki67 antigen-positive cells whereas HPV-transformed keratinocytes produced up to 15 poorly differentiated epithelial layers that were reminiscent of high grade cervical lesions seen in vivo and exhibited a full thickness Ki67 antigen expression. When normal and HPV-transformed keratinocytes were maintained in the presence of IFN-gamma, the epithelial sheet was reduced to a few cells in thickness and the density of Ki67 antigen-positive cells was decreased. A more pronounced growth inhibitory effect in monolayer and organotypic cultures was observed when IFN-gamma was associated with tumor necrosis factor-alpha Tumor necrosis factor-alpha alone reduced the DNA synthesis of normal keratinocytes but was significantly less effective than IFN-gamma to inhibit the growth of HPV-transformed keratinocytes. These results suggest that similar responses in vivo to regulatory molecules may play a role in the development of HPV-related lesions.
Collapse
Affiliation(s)
- P Delvenne
- Department of Pathology, University Hospital of Liège, Belgium
| | | | | | | | | |
Collapse
|
55
|
Bacus SS, Zelnick CR, Chin DM, Yarden Y, Kaminsky DB, Bennington J, Wen D, Marcus JN, Page DL. Medullary carcinoma is associated with expression of intercellular adhesion molecule-1. Implication to its morphology and its clinical behavior. THE AMERICAN JOURNAL OF PATHOLOGY 1994; 145:1337-48. [PMID: 7992839 PMCID: PMC1887499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The histological hallmarks for the diagnosis of medullary breast cancer are circumscription, syncytial architecture, diffuse inflammatory infiltrate, and highly atypical nuclei. The biological and prognostic implication is a lower propensity to metastasize. We studied 19 medullary carcinomas for expression of the intercellular adhesion molecule-1 and lymphocyte-function-associated antigen-1, Neu differentiation factor, tumor necrosis factor-alpha, and the expression of HER-2/neu, HER-4, and HER-3 receptors. Our study revealed that all of the 19 medullary carcinomas expressed the intercellular adhesion molecule-1 and lymphocyte function associated antigen. Eighteen of 19 cancers expressed Neu differentiation factor and tumor necrosis factor-alpha. All medullary cancers expressed the HER-2/neu receptor, however, in the majority of the cases, the staining was confined to the cytoplasm. Only 4 of 12 cancers expressed HER-4 and none of the eight medullary cancers tested expressed HER-3. By comparison, in a control group of infiltrating ductal carcinomas, expression of intercellular adhesion molecule-1, lymphocyte function associated antigen-1, and Neu differentiation factor was positive in about 25 to 30% of the cases, HER-4 was expressed in 75% and HER-3 in 95% of the cases. Taken together, our observations suggest that the expression of intercellular adhesion molecule-1, lymphocyte function associated antigen, Neu differentiation factor, and tumor necrosis factor-alpha as factors that may affect the special morphology and the biological behavior that characterizes medullary carcinomas.
Collapse
Affiliation(s)
- S S Bacus
- Advanced Cellular Diagnostics, Elmhurst, Illinois 60126
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Aggarwal BB, Pocsik E, Ali-Osman F, Totpal K. Transfection of cells with transforming growth factor-alpha leads to cellular resistance to the antiproliferative effects of tumor necrosis factor. FEBS Lett 1994; 354:12-6. [PMID: 7957892 DOI: 10.1016/0014-5793(94)01073-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Tumor necrosis factor (TNF) is a growth-modulatory cytokine that inhibits the growth of certain cell lines, stimulates the growth of some, and has no effect on the growth of still others. The molecular basis for this differential regulation of growth by TNF is not understood. We postulate that the growth of normal or tumor cells is determined by the balance between growth-stimulatory and -inhibitory signals. In the present study, we demonstrate that the transfection of cells with the transforming growth factor (TGF)-alpha gene induces resistance to TNF. Colon carcinoma cell lines that express elevated levels of TGF-alpha were also found to be resistant to this cytokine. Exogenous addition of the growth factor was also effective in decreasing the antiproliferative effects of TNF. Transfection of cells with the TGF-alpha gene led to downmodulation of TNF receptors but an increase in intracellular glutathione levels. Thus, these results support our hypothesis that expression of growth factors by certain tumor cells can lead to resistance to antiproliferative agents such as TNF.
Collapse
Affiliation(s)
- B B Aggarwal
- Department of Clinical Immunology and Biological Therapy, University of Texas M.D. Anderson Cancer Center, Houston 77030
| | | | | | | |
Collapse
|
57
|
Dorai H, McCartney JE, Hudziak RM, Tai MS, Laminet AA, Houston LL, Huston JS, Oppermann H. Mammalian cell expression of single-chain Fv (sFv) antibody proteins and their C-terminal fusions with interleukin-2 and other effector domains. BIO/TECHNOLOGY (NATURE PUBLISHING COMPANY) 1994; 12:890-7. [PMID: 7765352 DOI: 10.1038/nbt0994-890] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The production of several single-chain Fv (sFv) antibody proteins was examined by three modes of mammalian cell expression. Our primary model was the 741F8 anti-c-erbB-2 sFv, assembled as either the VH-VL or VL-VH, and expressed alone, with C-terminal cysteine for dimerization, or as fusion proteins with carboxyl-terminal effector domains, including interleukin-2, the B domain of staphylococcal protein A, the S-peptide of ribonuclease S, or hexa-histidine metal chelate peptide. Constructs were expressed and secreted transiently in 293 cells and stably in CHO or Sp2/0 cell lines, the latter yielding up to 10 mg per liter. Single-chain constructs of MOPC 315 myeloma and 26-10 monoclonal antibodies were also expressed, as were hybrids comprising unrelated VH and VL regions. Our results suggest that mammalian expression is a practical and valuable complement to the bacterial expression of single-chain antibodies.
Collapse
Affiliation(s)
- H Dorai
- Creative BioMolecules, Inc., Hopkinton, MA 01748
| | | | | | | | | | | | | | | |
Collapse
|
58
|
Laderoute MP. A new perspective on the nature of the cancer problem: anti-cellular senescence. Mol Carcinog 1994; 10:125-33. [PMID: 7519015 DOI: 10.1002/mc.2940100303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- M P Laderoute
- Molecular Oncology Program, Cross Cancer Institute, Edmonton, Alberta, Canada
| |
Collapse
|
59
|
Aggarwal BB, Totpal K, Ali-Osman F, Budde RJ, Pocsik E. pp60v-src kinase overexpression leads to cellular resistance to the antiproliferative effects of tumor necrosis factor. FEBS Lett 1994; 345:219-24. [PMID: 7911089 DOI: 10.1016/0014-5793(94)00441-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
While some tumor cells are sensitive to the antiproliferative effects of tumor necrosis factor (TNF), others are resistant. The molecular basis for cellular resistance to TNF is not completely understood. Previously we have shown that transfection of cells with an oncogene HER2/neu/erb B2, a receptor tyrosine kinase, leads to resistance to the anticellular effects of TNF [(1988) Proc. Natl. Acad. Sci. USA 85, 5102-5106]. In the present study, we demonstrate that the overexpression of another oncogenic tyrosine kinase, pp60v-src also induces resistance to TNF. In contrast to HER2, however, pp60v-src transfection of cells did not lead to down-modulation of TNF receptors but rather to decreased intracellular glutathione levels. The pp60v-src-induced cellular resistance to TNF could be abrogated by interferon-gamma. Thus, these results indicate that the resistance of certain tumors to TNF may also be due in part to the overexpression of pp60v-src oncogene.
Collapse
Affiliation(s)
- B B Aggarwal
- Department of Clinical Immunology and Biological Therapy, University of Texas, M.D. Anderson Cancer Center, Houston 77030
| | | | | | | | | |
Collapse
|
60
|
Malejczyk J, Malejczyk M, Majewski S, Breitburd F, Luger TA, Jablonska S, Orth G. Increased tumorigenicity of human keratinocytes harboring human papillomavirus type 16 is associated with resistance to endogenous tumor necrosis factor-alpha-mediated growth limitation. Int J Cancer 1994; 56:593-8. [PMID: 8112896 DOI: 10.1002/ijc.2910560421] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The aim of this study was to evaluate the relationship between tumorigenicity of cell sublines derived from weakly tumorigenic SKv-e and SKv-l keratinocytes harboring human papillomavirus type 16 (HPV16) and their susceptibility to autocrine growth limitation mediated by tumor necrosis factor-alpha (TNF-alpha). These sublines displayed different in vitro proliferative potential which correlated with tumorigenicity in nu/nu mice. Recombinant TNF-alpha inhibited in vitro growth of weakly tumorigenic parental SKv cell lines while it did not affect proliferation of their respective highly tumorigenic sublines. Resistance to TNF-alpha correlated with both increased in vitro proliferation and tumorigenicity. Anti-TNF-alpha antibodies (Ab) significantly increased in vitro proliferation of weakly tumorigenic parental SKv cells up to the levels of their highly tumorigenic sublines. Growth of highly tumorigenic SKv cells was not affected. On the other hand, proliferation of SKv cells was affected neither by transforming growth factor-beta (TGF-beta) nor by anti-TGF-beta Ab. All SKv cell sublines tested spontaneously released TNF-alpha, as evaluated by a specific radioimmunoassay; however, the levels of the endogenous cytokine were not related to their proliferative potential and tumorigenicity. An increased resistance to the anti-proliferative effect of TNF-alpha may be associated with decreased expression of TNF-alpha receptors (TNF-alpha R) inasmuch as evaluation of 125I-TNF-alpha binding and Northern-blot analysis of TNF-alpha R-specific mRNA showed that highly tumorigenic SKv cell sublines expressed significantly lower numbers of TNF-alpha R than their respective parental cells. These results show that an increased tumorigenicity of HPV16-harboring SKv keratinocytes may be, at least partially, due to escape from autocrine TNF-alpha-mediated growth limitation.
Collapse
Affiliation(s)
- J Malejczyk
- Department of Histology and Embryology, Warsaw Medical School, Poland
| | | | | | | | | | | | | |
Collapse
|
61
|
Mizutani Y, Yoshida O. Overcoming tumor necrosis factor-alpha resistance of human renal and ovarian carcinoma cells by combination treatment with buthionine sulfoximine and tumor necrosis factor-alpha. Role of tumor necrosis factor-alpha mRNA down-regulation in tumor cell sensitization. Cancer 1994; 73:730-7. [PMID: 8299097 DOI: 10.1002/1097-0142(19940201)73:3<730::aid-cncr2820730338>3.0.co;2-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Previous studies have reported the glutathione plays a central role in a wide range of cellular functions, including protection, detoxification, transport, and metabolism. Buthionine sulfoximine (BSO), a specific inhibitor of gamma-glutamyl-cysteine synthetase, depletes intracellular glutathione. The study investigates the cytotoxic effect of BSO and tumor necrosis factor-alpha (TNF-alpha) used in combination on TNF-alpha-resistant human renal and ovarian cancer cells. METHODS Cytotoxicity was determined by a 1-day microculture tetrazolium dye assay. TNF-alpha mRNA was examined by Northern blot analysis. RESULTS Combination treatment of TNF-alpha-resistant R4 and R11 human renal cell carcinoma cells with BSO and TNF-alpha overcame their resistance to TNF-alpha. In addition, the combination of BSO and TNF-alpha resulted in a synergistic cytotoxic effect on TNF-alpha-resistant OVC-8 and C30 human ovarian cancer cells. Treatment of R4, R11, and OVC-8 cells with TNF-alpha in combination with glutathione or N-acetyl-cysteine (NAC) showed an antagonistic cytotoxic effect. A possible mechanism of resistance to TNF-alpha in tumor cells is the expression of TNF-alpha mRNA or protein. R4 cells and OVC-8 cells constitutively expressed mRNA for TNF-alpha. Treatment of R4 cells or OVC-8 cells with BSO down-regulated the expression of TNF-alpha mRNA; however, treatment with TNF-alpha up-regulated the expression of TNF-alpha mRNA. When BSO was used in combination with TNF-alpha, the level of TNF-alpha mRNA enhanced by TNF-alpha was markedly reduced. Incubation of R4 cells with glutathione or NAC also down-regulated the expression of TNF-alpha mRNA. R11 and C30 cells did not constitutively express mRNA for TNF-alpha, and the BSO treatment had no effect on the TNF-alpha mRNA level. CONCLUSIONS This study demonstrates that the combination of BSO and TNF-alpha can overcome the TNF-alpha resistance of tumor cells and that depletion of intracellular glutathione and down-regulation of TNF-alpha mRNA by BSO may play a role in the enhanced cytotoxicity seen with the combination of BSO and TNF-alpha. There may not be always a correlation between the expression of TNF-alpha mRNA in tumor cells and their resistance to TNF-alpha. The synergistic effect obtained with established renal cell carcinoma cells and ovarian cancer cells suggests that combination treatment with TNF-alpha and BSO could have clinical application in the therapy of TNF-alpha-resistant tumors.
Collapse
Affiliation(s)
- Y Mizutani
- Department of Urology, Faculty of Medicine, Kyoto University, Japan
| | | |
Collapse
|
62
|
|
63
|
Wiltschke C, Tyl E, Speiser P, Steininger A, Zeillinger R, Kury F, Czerwenka K, Kubista E, Preis P, Krainer M. Increased natural killer cell activity correlates with low or negative expression of the HER-2/neu oncogene in patients with breast cancer. Cancer 1994; 73:135-9. [PMID: 7903907 DOI: 10.1002/1097-0142(19940101)73:1<135::aid-cncr2820730123>3.0.co;2-s] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Increased expression of the HER-2/neu oncogene in breast cancer correlates with decreased estrogen receptor concentration and seems to be an important prognostic factor. The authors investigated whether there is a correlation between HER-2/neu expression and immunologic parameters representing tumor defense in patients with breast cancer. METHOD A Western blot analysis was used to investigate HER-2/neu expression, whereas a chromium-release assay using the K562 cell line as target was used to measure natural killer (NK) cell activity. RESULTS In patients with breast cancer, NK cell activity was significantly higher compared with patients with benign tumors (P = 0.006) or healthy control subjects (P = 0.002). Moreover, 23.3% of patients with breast cancer showed an overexpression of HER-2/neu protein. Within this group of patients, NK cell activity was significantly lower (45.6 +/- 16.1%) compared with the group with no HER-2/neu overexpression (57.3 +/- 11.0%). NK cell activity did not increase in patients with HER-2/neu overexpression. Thus, there was a statistically significant correlation of cytolytic effector cell function with HER-2/neu expression of the tumor (P = 0.003), and HER-2/neu overexpression correlated with a negative estrogen receptor status (P = 0.005). CONCLUSION These data add further evidence to previous observations from the authors' laboratory that certain tumor characteristics may be associated with reactions of the host with breast cancer.
Collapse
Affiliation(s)
- C Wiltschke
- First Department of Internal Medicine, University Hospital, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Mochimatsu I, Tsukuda M, Furukawa S, Watanabe S, Kubota A, Yanoma S. The sensitivity of head and neck squamous cell carcinomas to tumor necrosis factor-alpha. BIOTHERAPY (DORDRECHT, NETHERLANDS) 1993; 6:239-44. [PMID: 8018444 DOI: 10.1007/bf01878352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Sensitivities to recombinant human tumor necrosis factor-alpha (TNF-alpha) and chemotherapeutic agents (cisplatin, peplomycin, methotrexate) were evaluated in 20 tumor cells of head and neck squamous cell carcinomas, using a dye uptake method. Also, numbers of TNF receptors of these tumor cells were measured by Scatchard plot analysis. There was no relationship between the number of TNF-alpha receptors and the sensitivity to TNF-alpha. Furthermore, there was no correlation between the sensitivity to TNF-alpha and that to chemotherapeutic drugs, nor between the sensitivity to TNF-alpha and the clinical response to chemotherapy including of cisplatin and peplomycin. The sensitivity to TNF-alpha was higher in poorly differentiated carcinomas than in well differentiated ones.
Collapse
Affiliation(s)
- I Mochimatsu
- Department of Otorhinolaryngology, Yokoham City University, School of Medicine, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
65
|
Kalthoff H, Roeder C, Gieseking J, Humburg I, Schmiegel W. Inverse regulation of human ERBB2 and epidermal growth factor receptors by tumor necrosis factor alpha. Proc Natl Acad Sci U S A 1993; 90:8972-6. [PMID: 8105469 PMCID: PMC47483 DOI: 10.1073/pnas.90.19.8972] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Recombinant human tumor necrosis factor (TNF) alpha decreased the expression of ERBB2 mRNA by stimulating p55 TNF receptors of pancreatic tumor cells. This decrease contrasts with an increase in epidermal growth factor receptor (EGFR) mRNA. Both effects were selectively achieved by TNF-alpha or -beta, whereas interferon alpha or gamma or transforming growth factor beta showed no such effects. The inverse regulatory effects of TNF on ERBB2 and EGFR mRNA levels were evoked by different signaling pathways of p55 TNF receptors. The TNF-mediated ERBB2 mRNA decrease was followed by a reduction in protein. Four of five pancreatic tumor cell lines exhibited this down-regulation. This decrease of ERBB2 is a singular example of a modulation of this growth factor receptor by TNF. Overexpression of ERBB2 has been reported to cause resistance to TNF and other cytotoxic cytokines. In our study we show that the TNF-mediated down-regulation of ERBB2 in pancreatic tumor cells is accompanied by an increase in growth inhibition at low doses of TNF. The simultaneous alteration of the ERBB2/EGFR balance by TNF represents a striking model of cytokine receptor transregulation in the growth control of malignant pancreatic epithelial cells.
Collapse
Affiliation(s)
- H Kalthoff
- Christian-Albrechts-Universität, Klinik für Allgemeine Chirurgie, Kiel, Federal Republic of Germany
| | | | | | | | | |
Collapse
|
66
|
Lewis GD, Figari I, Fendly B, Wong WL, Carter P, Gorman C, Shepard HM. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol Immunother 1993; 37:255-63. [PMID: 8102322 PMCID: PMC11038979 DOI: 10.1007/bf01518520] [Citation(s) in RCA: 371] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/1992] [Accepted: 03/30/1993] [Indexed: 01/28/2023]
Abstract
The HER2 protooncogene encodes a receptor tyrosine kinase, p185HER2. The overexpression of p185HER2 has been associated with a worsened prognosis in certain human cancers. In the present work we have screened a variety of different tumor cell lines for p185HER2 expression using both enzyme-linked immunosorbent and fluorescence-activated cell sorting assays employing murine monoclonal antibodies directed against the extracellular domain of the receptor. Increased levels of p185HER2 were found in breast (5/9), ovarian (1/6), stomach (2/3) and colorectal (5/16) carcinomas, whereas all kidney and submaxillary adenocarcinoma cell lines tested were negative. Some monoclonal antibodies directed against the extracellular domain of p185HER2 inhibited growth in monolayer culture of breast and ovarian tumor cell lines overexpressing p185HER2, but had no effect on the growth of colon or gastric adenocarcinomas expressing increased levels of this receptor. The most potent growth-inhibitory anti-p185HER2 monoclonal antibody in monolayer culture, designated mumAb 4D5 (a murine IgG1 kappa antibody), was also tested in soft-agar growth assays for activity against p185HER2-overexpressing tumor cell lines of each type, with similar results. In order to increase the spectrum of tumor types potentially susceptible to monoclonal antibody-mediated anti-p185HER2 therapies, to decrease potential immunogenicity issues with the use of murine monoclonal antibodies for human therapy, and to provide the potential for antibody-mediated cytotoxic activity, a mouse/human chimeric 4D5 (chmAb 4D5) and a "humanized" 4D5 (rhu)mAb 4D5 HER2 antibody were constructed. Both engineered antibodies, in combination with human peripheral blood mononuclear cells, elicited antibody-dependent cytotoxic responses in accordance with the level of p185HER2 expression. Since this cytotoxic activity is independent of sensitivity to mumAb 4D5, the engineered monoclonal antibodies expand the potential target population for antibody-mediated therapy of human cancers characterized by the overexpression of p185HER2.
Collapse
Affiliation(s)
- G D Lewis
- Genentech Inc., South San Francisco, CA 94080
| | | | | | | | | | | | | |
Collapse
|
67
|
Issing WJ, Dreps A, Heppt WJ, Wustrow TP, Riederer A, Zagury JF. erbB-2/Her-2 gene amplification and overexpression in parotid gland tumors. Eur Arch Otorhinolaryngol 1993; 250:150-3. [PMID: 8102855 DOI: 10.1007/bf00171701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Proto-oncogenes represent a family of normal cellular genes that were identified on the basis of their similarity to genetic sequences with known tumorigenic or transforming potential. Accumulating evidence links alterations in either the structure, copy number, or expression of one or another of these genes to neoplasia. One such gene, called erbB-2/Her-2 was found amplified in an adenocarcinoma of the human salivary gland and has also been found associated with primary human breast cancer. Patients with multiple copies of the gene have had a shorter overall survival. In the present study, 21 tumors of the parotid gland were examined by Southern and Northern blot hybridization for amplification and possible overexpression of the erbB-2/Her-2 oncogene. Normal parotid gland tissue was used as negative control. The parotid gland lesions comprised 7 pleomorphic adenomas, 5 squamous cell carcinomas, 4 cases of chronic fibrotic sialadenosis, 3 mucoepidermoid carcinomas as well as 1 lymphoma and 1 cystadenolymphoma. Gene amplification was found in 1 of the pleomorphic adenomas, with 2 tumors showing a significant overexpression of the erbB-2/Her-2 oncogene. Because 3-5% of all pleomorphic adenomas undergo malignant transformation, close follow-up of patients is currently underway.
Collapse
Affiliation(s)
- W J Issing
- Department of Otorhinolaryngology, University of Munich, Klinikum Grosshadern, Germany
| | | | | | | | | | | |
Collapse
|
68
|
Safrit JT, Belldegrun A, Bonavida B. Sensitivity of human renal cell carcinoma lines to TNF, adriamycin, and combination: role of TNF mRNA induction in overcoming resistance. J Urol 1993; 149:1202-8. [PMID: 8483246 DOI: 10.1016/s0022-5347(17)36348-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have examined 6 human renal cell carcinoma (RCC) cell lines for their sensitivity and resistance to the cytolytic effect of tumor necrosis factor (TNF) and adriamycin (ADR), alone or in combination. The results of cytotoxicity mediated by TNF and ADR showed no direct correlation as TNF resistant lines were sensitive to ADR while the TNF sensitive lines were resistant. The combination of TNF and ADR resulted in enhanced cytotoxicity against the tumor lines. Induction of TNF mRNA and protein has been suggested as a mechanism of resistance to TNF in certain tumors. Resistant and sensitive lines were capable of upregulating TNF mRNA after treatment with TNF or PMA+ionophore for periods as short as 1 hour, but only the resistant lines were able to secrete detectable levels of TNF protein. Therefore, a positive correlation existed between resistance to TNF and production and secretion of TNF by the cell lines. In the presence of the protein synthesis inhibitor cycloheximide (CHX), the TNF mRNA level in the TNF resistant lines was increased while the sensitive lines required an additional signal, such as exogenous TNF, to upregulate the mRNA. Due to the enhanced cytotoxicity seen with the combination of TNF and ADR, we determined the effect of this combination on the levels of TNF mRNA. As examined in a constitutively TNF expressing line, ADR alone reduced the constitutive mRNA level and, in combination with TNF, reduced the level of induction produced by TNF. This downregulation of TNF mRNA by ADR may play a role in the enhanced cytotoxicity seen with combined TNF and ADR treatment. The present study demonstrates that RCC cell lines differ in their sensitivity and/or resistance to TNF. Further, ADR and/or TNF resistant RCC lines can be rendered sensitive by combining TNF and ADR. The constitutive and/or inductive secretion of TNF by certain lines and its relationship to tumor pathogenesis as well as overcoming resistance are discussed.
Collapse
Affiliation(s)
- J T Safrit
- Department of Microbiology and Immunology, UCLA School of Medicine 90024
| | | | | |
Collapse
|
69
|
Fady C, Gardner A, Gera JF, Lichtenstein A. Resistance of HER2/neu-overexpressing tumor targets to lymphokine-activated-killer-cell-mediated lysis: evidence for deficiency of binding and post-binding events. Cancer Immunol Immunother 1993; 36:307-14. [PMID: 8097427 PMCID: PMC11038807 DOI: 10.1007/bf01741169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/1992] [Accepted: 12/04/1992] [Indexed: 01/28/2023]
Abstract
HER2/neu-overexpressing tumor cell lines are relatively resistant to lymphokine-activated killer (LAK) cell cytotoxicity when compared to HER2/neu-nonexpressing lines. HER2/neu+ targets were also resistant to binding by LAK large granular lymphocytes (LGL) as shown by visualization at the single-cell level, a target monolayer binding assay and in "cold" target inhibition experiments. HER2/neu+ LAK-resistant ovarian cell lines demonstrated an absence of ICAM-1 expression while expression of LFA-3, N-CAM, laminin and beta 1 integrins was comparable to that of HER2/neu- targets. In contrast, the HER2/neu+ breast cell line, SKBR-3, which was also resistant to lysis and binding by LAK LGL, demonstrated normal expression of ICAM-1. Anti-ICAM-1 antibodies blocked binding and lysis of HER2/neu- carcinoma targets by LAK cells, further supporting the notion that lack of ICAM-1 expression on HER2/neu+ cells contributes to their resistance. The modest binding and lysis of HER2/neu+ targets by LAK cells was significantly inhibited by anti-LFA-1 antibodies, suggesting the existence of another counter-receptor for LFA-1 on HER2/neu+ targets. The following also supported deficiencies in post-binding events when HER2/neu+ cells resisted the lytic activity of LAK cells: (a) when the relative resistance to effector cell binding was overcome by exogenous lectin. HER2/neu+ cell lines were still resistant to LAK cytolysis, and (b) HER2/neu+ targets were resistant to perforin-containing granule extracts obtained from the CTLL-R8 cytotoxic lymphocyte cell line. These results indicate that deficiency in effector binding as well as post-binding events contributes to the resistance of HER2/neu-overexpressing tumor targets to LAK-cell-mediated lysis.
Collapse
Affiliation(s)
- C Fady
- Department of Medicine, VA Wadsworth-UCLA Medical Center 90073
| | | | | | | |
Collapse
|
70
|
Tripathy D, Benz CC. Activated oncogenes and putative tumor suppressor genes involved in human breast cancers. Cancer Treat Res 1993; 63:15-60. [PMID: 1363356 DOI: 10.1007/978-1-4615-3088-6_2] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytogeneticists first proposed that the karyotypic abnormalities identified on chromosomes 1, 3, 6, 11, 13, 16, 17, and 18 supported a genetic basis for breast cancer. Such abnormal banding patterns, however, may represent either loss-of-function or gain-of-function molecular events. RFLP analyses have since confirmed that 20-60% of primary and spontaneous human breast tumors exhibit allelic losses on these same chromosomes, although the exact genes involved at these chromosomal sites remain largely unknown. Knowledge gained about the Rb-1 and p53 tumor suppressor genes at 13q14 and 17p13 in breast and other human tumors supports the paradigm that for any chromosomal locus, allelic loss associated with a mutation in the remaining tumor allele signifies an involved tumor suppressor gene. Given this paradigm, there are nearly a dozen putative breast tumor suppressor genes under active investigation, with most investigators now focusing on various chromosome 17 loci. Among the known proto-oncogenes found activated in breast cancer, amplification of c-erbB-2 at 17q21 is the most widely studied and clinically significant gain-of-function event uncovered to date, occurring in about 20% of all primary breast tumors. The involvement of this overexpressed membrane receptor has engendered interest in related tyrosine kinase receptors, such as EGFR, IR, and IGF-I-R, as well as their respective ligands, which may be overexpressed in a greater fraction of tumors, contributing to the autocrine and paracrine regulation of breast cancer growth and metastasis. New attention is being given to the potentially oncogenic function of structurally altered nuclear transactivating steroid hormone receptors, such as ER, whose overexpression has long been used to determine endocrine therapy and prognosis for individual breast cancer patients. While c-myc was one of the first known proto-oncogenes to be found amplified and overexpressed in human breast cancers, the actual incidence and clinical significance of its activation remain disputed and in need of further study. Lastly, we can expect greater clarification about the importance of various 11q13 genes found coamplified in nearly 20% of primary breast cancers, and pursuit into the intriguing possibility that a cyclin-encoding gene represents the overexpressed locus of real interest in this amplicon. Virtually all of these important genetic abnormalities identified thus far are associated with but not restricted to human breast cancers. The absence of identifiable molecular defects relating to the tissue specificity of this malignancy must be considered a substantial gap in our basic understanding of breast carcinogenesis.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
|
71
|
Shalaby MR, Shepard HM, Presta L, Rodrigues ML, Beverley PC, Feldmann M, Carter P. Development of humanized bispecific antibodies reactive with cytotoxic lymphocytes and tumor cells overexpressing the HER2 protooncogene. J Exp Med 1992; 175:217-25. [PMID: 1346155 PMCID: PMC2119072 DOI: 10.1084/jem.175.1.217] [Citation(s) in RCA: 142] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The HER2 protooncogene encodes a 185-kD transmembrane phosphoglycoproteins, human epidermal growth factor receptor 2 (p185HER2), whose amplified expression on the cell surface can lead to malignant transformation. Overexpression of HER2/p185HER2 is strongly correlated with progression of human ovarian and breast carcinomas. Recent studies have shown that human T cells can be targeted with bispecific antibody to react against human tumor cells in vitro. We have developed a bispecific F(ab')2 antibody molecule consisting of a humanized arm with a specificity to p185HER2 linked to another arm derived from a murine anti-CD3 monoclonal antibody that we have cloned from UCHT1 hybridoma. The antigen-binding loops for the anti-CD3 were installed in the context of human variable region framework residues, thus forming a fully humanized BsF(ab')2 fragment. Additional variants were produced by replacement of amino acid residues located in light chain complementarity determining region 2 and heavy chain framework region 3 of the humanized anti-CD3 arm. Flow cytometry analysis showed that the bispecific F(ab')2 molecules can bind specifically to cells overexpressing p185HER2 and to normal human peripheral blood mononuclear cells bearing the CD3 surface marker. In additional experiments, the presence of bispecific F(ab')2 caused up to fourfold enhancement in the cytotoxic activities of human T cells against tumor cells overexpressing p185HER2 as determined by a 51Cr release assay. These bispecific molecules have a potential use as therapeutic agents for the treatment of cancer.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies, Monoclonal
- Antigens, CD/genetics
- Antigens, Differentiation, T-Lymphocyte/genetics
- Base Sequence
- Breast Neoplasms/immunology
- CD3 Complex
- Cloning, Molecular
- Cytotoxicity, Immunologic
- Female
- Flow Cytometry
- Genetic Variation
- Humans
- Models, Molecular
- Molecular Sequence Data
- Oligodeoxyribonucleotides
- Oligonucleotides, Antisense
- Polymerase Chain Reaction
- Protein Conformation
- Proto-Oncogene Proteins/genetics
- Proto-Oncogenes
- Receptor, ErbB-2
- Receptors, Antigen, T-Cell/genetics
- Sequence Homology, Nucleic Acid
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- M R Shalaby
- Department of Cell Biology, Genentech, Inc., South San Francisco, California 94080
| | | | | | | | | | | | | |
Collapse
|
72
|
Himeno T, Watanabe N, Yamauchi N, Maeda M, Okamoto T, Tsuji N, Tsuji Y, Akiyama S, Sasaki H, Niitsu Y. Induction of synthesis of manganous superoxide dismutase in L-M(pNTnF) cells carrying an inducible TNF gene. Int J Cancer 1992; 50:458-62. [PMID: 1735614 DOI: 10.1002/ijc.2910500322] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Based on findings that the cytotoxic effects of tumor necrosis factor (TNF) are closely related to levels of intracellular oxygen radicals, and on the results of TNF gene transfection studies, the hypothesis was made that endogenous TNF (enTNF) acts as a protective factor against exogenous TNF by inducing inhibitors or scavengers of oxygen radicals. In order to test this hypothesis, we investigated the intracellular levels of manganous superoxide dismutase (MnSOD) and glutathione (GSH) in L-M(pNTnF) cells carrying a TNF gene induced by dexamethasone (DM). When L-M(pNTnF) cells were treated with DM they expressed enTNF, and acquired resistance to exogenous TNF. There was no change in the GSH concentration after enTNF induction, but a 1.9- to 3.9-fold increase in MnSOD levels was noted. Our findings suggest that enTNF exerts its protective function against the cytocidal effect of exogenous TNF by inducing MnSOD production.
Collapse
Affiliation(s)
- T Himeno
- Department of Internal Medicine (Section 4), Sapporo Medical College, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Ioannides CG, Ioannides MG, O'Brian CA. T-cell recognition of oncogene products: a new strategy for immunotherapy. Mol Carcinog 1992; 6:77-82. [PMID: 1382444 DOI: 10.1002/mc.2940060202] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- C G Ioannides
- Department of Gynecology, University of Texas M. D. Anderson Cancer Center, Houston
| | | | | |
Collapse
|
74
|
Sandberg AA. Cytogenetic and molecular genetic aspects of human prostate cancer: primary and metastatic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1992; 324:45-75. [PMID: 1492627 DOI: 10.1007/978-1-4615-3398-6_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- A A Sandberg
- Cancer Center, Southwest Biomedical Research Institute, Scottsdale, AZ 85251
| |
Collapse
|
75
|
Hudziak R, Ullrich A. Cell transformation potential of a HER2 transmembrane domain deletion mutant retained in the endoplasmic reticulum. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)54400-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
76
|
|
77
|
Scott G, Dodson J, Montgomery P, Johnson R, Sarup J, Wong W, Ullrich A, Shepard H, Benz C. p185HER2 signal transduction in breast cancer cells. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)98683-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
78
|
Shepard HM, Lewis GD, Sarup JC, Fendly BM, Maneval D, Mordenti J, Figari I, Kotts CE, Palladino MA, Ullrich A. Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic. J Clin Immunol 1991; 11:117-27. [PMID: 1679763 DOI: 10.1007/bf00918679] [Citation(s) in RCA: 205] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The HER2 protooncogene encodes a 185-kDa transmembrane protein (p185HER2) with extensive homology to the epidermal growth factor (EGF) receptor. Clinical and experimental evidence supports a role for overexpression of the HER2 protooncogene in the progression of human breast, ovarian, and non-small cell lung carcinoma. These data also support the hypothesis that p185HER2 present on the surface of overexpressing tumor cells may be a good target for receptor-targeted therapeutics. The anti-p185HER2 murine monoclonal antibody (muMAb) 4D5 is one of over 100 monoclonals that was derived following immunization of mice with cells overexpressing p185HER2. The monoclonal antibody is directed at the extracellular (ligand binding) domain of this receptor tyrosine kinase and presumably has its effect as a result of modulating receptor function. In vitro assays have shown that muMAb 4D5 can specifically inhibit the growth of tumor cells only when they overexpress the HER2 protooncogene. MuMAb 4D5 has also been shown to enhance the TNF-alpha sensitivity of breast tumor cells that overexpress this protooncogene. Relevant to its clinical application, muMAb 4D5 may enhance the sensitivity of p185HER2-overexpressing tumor cells to cisplatin, a chemotherapeutic drug often used in the treatment of ovarian cancer. In vivo assays with a nude mouse model have shown that the monoclonal antibody can localize at the tumor site and can inhibit the growth of human tumor xenografts which overexpress p185HER2. Modulation of p185HER2 activity by muMAb 4D5 can therefore reverse many of the properties associated with tumor progression mediated by this putative growth factor receptor. Together with the demonstrated activity of muMAb 4D5 in nude mouse models, these results support the clinical application of muMAb 4D5 for therapy of human cancers characterized by the overexpression of p185HER2.
Collapse
Affiliation(s)
- H M Shepard
- Department of Developmental Biology, Genentech, Inc., South San Francisco, California 94080
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Regulation of phosphorylation of the c-erbB-2/HER2 gene product by a monoclonal antibody and serum growth factor(s) in human mammary carcinoma cells. Mol Cell Biol 1991. [PMID: 1671297 DOI: 10.1128/mcb.11.2.979] [Citation(s) in RCA: 66] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monoclonal antibody (MAb) 4D5 was used to analyze the phosphorylation of p185HER2, the gene product of c-erbB-2/HER2, in SK-BR-3 cells. Culture in the continuous presence of 4D5 reduced the in vivo steady-state levels of p185HER2 phosphorylation by 80% in a dose-dependent manner, suggesting that MAb 4D5 may have interfered with the activation of phosphorylation of p185HER2. The observed MAb-mediated reduction of p185HER2 phosphorylation could not be completely accounted for by down-regulation. When cultures were grown under serum-free conditions, the steady-state levels of p185HER2 phosphorylation were reduced by 56%, and addition of 4D5 further inhibited phosphorylation to 20% of steady-state levels. With continuous exposure to increasing concentrations of newborn calf serum in these cultures, there was a linear increase in tyrosine-specific phosphorylation of p185HER2, reaching a 5.4-fold increase with 10% newborn calf serum. Phosphorylation of p185HER2 in the presence of newborn calf serum was not attributable to stimulation of the epidermal growth factor receptor by epidermal growth factor or by transforming growth factor-alpha. Extension of these observations to two other mammary carcinoma cell lines. MDA-MB-453 and BT-474, also demonstrated a significant capacity of serum to induce p185HER2 phosphorylation. The demonstration of antibody-mediated partial inhibition of phosphorylation under serum-free conditions suggests that mammary carcinoma cells may also produce and secrete a factor or factors which may activate p185HER2. Our observation that growth-inhibitory MAb 4D5 is able to reduce the phosphorylation of p185HER2 by newborn calf serum and by a cellular-derived factor(s) suggests the existence of a growth factor(s) which uses phosphorylation of p185HER2 as a signal transduction pathway to regulate cell proliferation.
Collapse
|
80
|
Soomro S, Shousha S, Taylor P, Shepard HM, Feldmann M. c-erbB-2 expression in different histological types of invasive breast carcinoma. J Clin Pathol 1991; 44:211-4. [PMID: 1672872 PMCID: PMC496939 DOI: 10.1136/jcp.44.3.211] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sections of 149 breast carcinomas were examined for the over-expression of c-erbB-2 oncoprotein using the avidin-biotin immunoperoxidase technique and two different specific antibodies. These included the polyclonal antibody 21N and the monoclonal antibody 4D5. The tumours were divided into two main groups. The first included 75 cases of invasive ductal and classic invasive lobular carcinomas. The second group consisted of 74 cases with histological types known to have a good prognosis, including mucinous, alveolar variant of invasive lobular, medullary, tubular, cribriform and papillary carcinomas. Fifteen (20%) tumours of the first group were positive with the two antibodies. Fourteen of these were of the ductal type and one was a mixed invasive ductal and lobular carcinoma. Ten of the pure ductal cases had areas of comedo carcinoma. The intraductal elements in a further tumour were positively stained with 21N antibody only. None of the second group of tumours, which included histological types known to have good prognosis, stained with 4D5, although one mucinous carcinoma was positively stained with 21N. These findings suggest that in invasive breast carcinoma immunostaining for c-erbB-2 is mainly seen in a subgroup of ductal tumours, and that almost all other histological types, especially those associated with good prognosis, lack this expression.
Collapse
Affiliation(s)
- S Soomro
- Department of Histopathology, Charing Cross and Westminster Medical School, London
| | | | | | | | | |
Collapse
|
81
|
Kumar R, Shepard HM, Mendelsohn J. Regulation of phosphorylation of the c-erbB-2/HER2 gene product by a monoclonal antibody and serum growth factor(s) in human mammary carcinoma cells. Mol Cell Biol 1991; 11:979-86. [PMID: 1671297 PMCID: PMC359762 DOI: 10.1128/mcb.11.2.979-986.1991] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Monoclonal antibody (MAb) 4D5 was used to analyze the phosphorylation of p185HER2, the gene product of c-erbB-2/HER2, in SK-BR-3 cells. Culture in the continuous presence of 4D5 reduced the in vivo steady-state levels of p185HER2 phosphorylation by 80% in a dose-dependent manner, suggesting that MAb 4D5 may have interfered with the activation of phosphorylation of p185HER2. The observed MAb-mediated reduction of p185HER2 phosphorylation could not be completely accounted for by down-regulation. When cultures were grown under serum-free conditions, the steady-state levels of p185HER2 phosphorylation were reduced by 56%, and addition of 4D5 further inhibited phosphorylation to 20% of steady-state levels. With continuous exposure to increasing concentrations of newborn calf serum in these cultures, there was a linear increase in tyrosine-specific phosphorylation of p185HER2, reaching a 5.4-fold increase with 10% newborn calf serum. Phosphorylation of p185HER2 in the presence of newborn calf serum was not attributable to stimulation of the epidermal growth factor receptor by epidermal growth factor or by transforming growth factor-alpha. Extension of these observations to two other mammary carcinoma cell lines. MDA-MB-453 and BT-474, also demonstrated a significant capacity of serum to induce p185HER2 phosphorylation. The demonstration of antibody-mediated partial inhibition of phosphorylation under serum-free conditions suggests that mammary carcinoma cells may also produce and secrete a factor or factors which may activate p185HER2. Our observation that growth-inhibitory MAb 4D5 is able to reduce the phosphorylation of p185HER2 by newborn calf serum and by a cellular-derived factor(s) suggests the existence of a growth factor(s) which uses phosphorylation of p185HER2 as a signal transduction pathway to regulate cell proliferation.
Collapse
Affiliation(s)
- R Kumar
- Laboratory of Receptor Biology, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| | | | | |
Collapse
|
82
|
Park JW, Stagg R, Lewis GD, Carter P, Maneval D, Slamon DJ, Jaffe H, Shepard HM. Anti-p185HER2 monoclonal antibodies: Biological properties and potential for immunotherapy. Cancer Treat Res 1991; 61:193-211. [PMID: 1360232 DOI: 10.1007/978-1-4615-3500-3_10] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
83
|
Bacus SS, Ruby SG, Weinberg DS, Chin D, Ortiz R, Bacus JW. HER-2/neu oncogene expression and proliferation in breast cancers. THE AMERICAN JOURNAL OF PATHOLOGY 1990; 137:103-11. [PMID: 1973597 PMCID: PMC1877709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Amplification of the HER-2/neu proto-oncogene in breast cancer has been reported to correlate with poor patient prognosis. The proliferation, or growth fraction, of cells has also been shown to be of prognostic importance in breast cancer. A study was conducted to evaluate the correlation between HER-2/neu gene expression and proliferation in breast cancer. Quantitative immunohistochemical methods for the detection of the HER-2/neu protein expression and for assessing the proliferation fraction on frozen sections of tumor cells were used. The detection of epidermal growth factor receptor (EGFR) along with quantitative DNA ploidy analysis, also was performed on the same breast cancers. The results indicated two subgroups of invasive ductal carcinoma; 1) HER-2/neu overexpressing cases that were negative for EGFR expression and had low proliferation fraction, and a tetraploid DNA pattern (22 cases), and 2) other combinations of HER-2/neu expression and EGFR expression, with a high proliferation fraction and an aneuploid DNA pattern (38 cases). Eight cases of carcinoma in situ were positive for HER-2/neu overexpression and negative for EGFR expression, and had a high proliferation fraction and a tetraploid DNA pattern. Twenty-six cases of low-grade carcinoma exhibited low proliferation and a diploid DNA pattern.
Collapse
Affiliation(s)
- S S Bacus
- Cell Analysis Systems, Inc., Elmhurst, IL 60126-4944
| | | | | | | | | | | |
Collapse
|
84
|
Shirahata S, Rawson C, Loo D, Chang YJ, Barnes D. ras and neu oncogenes reverse serum inhibition and epidermal growth factor dependence of serum-free mouse embryo cells. J Cell Physiol 1990; 144:69-76. [PMID: 1973170 DOI: 10.1002/jcp.1041440110] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Serum-free mouse embryo cells, cultured in basal nutrient medium supplemented with insulin, transferrin, epidermal growth factor, fibronectin, and high-density lipoprotein, do not exhibit growth crisis, lack detectable chromosomal aberrations, are nontumorigenic in vivo, are dependent on epidermal growth factor for survival, and are growth inhibited by serum or platelet-free plasma. These cells after transfection with the human Ha-ras or rat neu oncogenes no longer required epidermal growth factor for survival, were tumorigenic in vivo, and also proliferated in serum-containing medium. Autocrine activity capable of replacing epidermal growth factor was detected in conditioned medium from ras-transformed cultures, but little such activity was detected in medium from neu-transformed cultures. In addition, the capability of ras or neu-transformed cells to grow in serum-containing medium could not be mimicked in untransformed cells by the addition of growth factors or conditioned medium from transformed cells. These results suggest that the known structural similarity of the neu gene product to the EGF receptor is also reflected in a functional similarity by which the mutationally activated neu protein can replace the ligand-activated EGF receptor. These results also suggest that the ability of ras- and neu-transformed cells to escape the effect of the inhibitory serum activity is a nonautocrine property distinct from the acquisition of EGF autonomy.
Collapse
Affiliation(s)
- S Shirahata
- Department of Biochemistry and Biophysics, Oregon State University 97331-6503
| | | | | | | | | |
Collapse
|
85
|
Donato N, Gallick G, Steck P, Rosenblum M. Tumor Necrosis Factor Modulates Epidermal Growth Factor Receptor Phosphorylation and Kinase Activity in Human Tumor Cells. J Biol Chem 1989. [DOI: 10.1016/s0021-9258(19)47086-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
86
|
Bajzer Z, Myers AC, Vuk-Pavlović S. Binding, Internalization, and Intracellular Processing of Proteins Interacting with Recycling Receptors. J Biol Chem 1989. [DOI: 10.1016/s0021-9258(18)80042-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
87
|
Jäättelä M, Saksela K, Saksela E. Heat shock protects WEHI-164 target cells from the cytolysis by tumor necrosis factors alpha and beta. Eur J Immunol 1989; 19:1413-7. [PMID: 2550247 DOI: 10.1002/eji.1830190810] [Citation(s) in RCA: 66] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Elevated temperatures and a number of other types of stress induce synthesis of a small number of highly conserved proteins, the heat shock proteins, in a wide variety of cells. The structure and regulation of these proteins have been intensively studied but the question of the function of this universal response has remained unanswered. We studied the effect of heat shock on tumor necrosis factor-alpha (TNF-alpha)- and -beta (TNF-beta)-mediated cytolysis of WEHI-164 clone 13 target cells. One hour pretreatment of target cells at 42 degrees C decreased rTNF-alpha-mediated lysis by 65.3%, 50.5% and 44.8% and TNF-beta-mediated lysis by 61.9%, 43.2% and 38.9% at cytokine concentrations of 0.5 ng/ml, 5 ng/ml and 50 ng/ml, respectively, in an 18-h Cr-release assay. The effect was maximal when TNF-alpha was added 1 h after the heat shock and then gradually declined, being almost undetectable after 2 days. This pattern was found to roughly coincide with the kinetics of hsp68, the major heat-induced protein in murine cells. Heat shock treatment had no protective effect when given 1 h after addition of recombinant TNF-alpha. The heat-induced target cell resistance was not associated with decreased binding of recombinant TNF-alpha to its receptor. Inhibition of protein synthesis by cycloheximide diminished this effect by 76% and inhibition of transcription by actinomycin D abolished it completely, suggesting that de novo synthesized heat-induced proteins protect target cells from TNF-mediated lysis in heat shock-treated WEHI cells.
Collapse
Affiliation(s)
- M Jäättelä
- Department of Pathology, University of Helsinki, Finland
| | | | | |
Collapse
|
88
|
p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol 1989. [PMID: 2566907 DOI: 10.1128/mcb.9.3.1165] [Citation(s) in RCA: 414] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The HER2/c-erbB-2 gene encodes the epidermal growth factor receptorlike human homolog of the rat neu oncogene. Amplification of this gene in primary breast carcinomas has been show to correlate with poor clinical prognosis for certain cancer patients. We show here that a monoclonal antibody directed against the extracellular domain of p185HER2 specifically inhibits the growth of breast tumor-derived cell lines overexpressing the HER2/c-erbB-2 gene product and prevents HER2/c-erbB-2-transformed NIH 3T3 cells from forming colonies in soft agar. Furthermore, resistance to the cytotoxic effect of tumor necrosis factor alpha, which has been shown to be a consequence of HER2/c-erbB-2 overexpression, is significantly reduced in the presence of this antibody.
Collapse
|
89
|
Seow HF, Goh CR, Krishnan L, Porter AG. Bacterial Expression, Facile Purification and Properties of Recombinant Human Lymphotoxin (Tumor Necrosis Factor Beta). Nat Biotechnol 1989. [DOI: 10.1038/nbt0489-363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
90
|
Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM, Ullrich A. p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol 1989; 9:1165-72. [PMID: 2566907 PMCID: PMC362707 DOI: 10.1128/mcb.9.3.1165-1172.1989] [Citation(s) in RCA: 182] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The HER2/c-erbB-2 gene encodes the epidermal growth factor receptorlike human homolog of the rat neu oncogene. Amplification of this gene in primary breast carcinomas has been show to correlate with poor clinical prognosis for certain cancer patients. We show here that a monoclonal antibody directed against the extracellular domain of p185HER2 specifically inhibits the growth of breast tumor-derived cell lines overexpressing the HER2/c-erbB-2 gene product and prevents HER2/c-erbB-2-transformed NIH 3T3 cells from forming colonies in soft agar. Furthermore, resistance to the cytotoxic effect of tumor necrosis factor alpha, which has been shown to be a consequence of HER2/c-erbB-2 overexpression, is significantly reduced in the presence of this antibody.
Collapse
Affiliation(s)
- R M Hudziak
- Department of Developmental Biology, Genentech, Inc., South San Francisco, California 94080
| | | | | | | | | | | |
Collapse
|
91
|
Abstract
TNF-alpha is clearly an important mediator of in vitro tumor cell cytotoxicity induced by the activated macrophage. There are a number of other nonspecific mediators of tumor cell cytotoxicity. These include natural killer cells (51, 52), lymphokine-activated killer cells (53), and natural cytotoxic cells (54). The role that TNF-alpha may play in the cytotoxicity induced by these cell types has not been completely elucidated. Neither is it known what role, if any, TNF-alpha may play in major histocompatibility-restricted (T cell)-mediated tumor cell cytotoxicity. Just as in the case of the activated macrophage, activated cytotoxic T cells produce a number of mediators that inhibit the growth of tumor cells or that induce tumor cell cytotoxicity (55). The role that TNF-alpha plays in the whole process of the regulation of tumorigenesis will not become completely defined until an appropriate set of genetic experiments is completed which utilizes transplantable tumor cell lines selected specifically for resistance to this cytokine in in vivo tumor models. The prominance of TNF-alpha as a mediator of macrophage-induced tumor cell cytotoxicity makes it a candidate for analysis in studies of the early stages of tumorigenesis. We have chosen to study mechanisms of resistance to this monokine. Our results have shown that there are multiple pathways leading to resistance to TNF-alpha-induced tumor cell cytotoxicity. These pathways include the production of transforming growth factors by tumor cells and the amplified expression of certain oncogenes. Other pathways will undoubtedly become elucidated as we begin to define the molecular mechanisms giving rise to the resistant phenotype.
Collapse
Affiliation(s)
- H M Shepard
- Department of Pharmacological Sciences, Genentech, Inc., South San Francisco, California 94080
| | | |
Collapse
|