151
|
Abstract
Activation of intracellular mitogenic signal transduction pathways driven by the ErbB family of receptor tyrosine kinases has been implicated in a variety of cancers. Amongst these, the tumorigenic roles of the Epidermal Growth Factor receptor (EGFR) and HER-2 have been most extensively studied. Therapeutic antibodies and small molecule kinase inhibitors targeting EGFR have recently received regulatory agency approval for the treatment of colon and lung cancer, respectively. In this review, I briefly describe these agents and their potential use in inhibiting the growth of tumors that overexpress HER-2. I also discuss other therapeutics currently available or being developed specifically to target HER-2 dependent tumors.
Collapse
|
152
|
Faltus T, Yuan J, Zimmer B, Krämer A, Loibl S, Kaufmann M, Strebhardt K. Silencing of the HER2/neu gene by siRNA inhibits proliferation and induces apoptosis in HER2/neu-overexpressing breast cancer cells. Neoplasia 2004; 6:786-95. [PMID: 15720805 PMCID: PMC1531682 DOI: 10.1593/neo.04313] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Revised: 07/29/2004] [Accepted: 08/06/2004] [Indexed: 11/18/2022]
Abstract
In eukaryotes, double-stranded (ds) RNA induces sequence-specific inhibition of gene expression referred to as RNA interference (RNAi). We exploited RNAi to define the role of HER2/neu in the neoplastic proliferation of human breast cancer cells. We transfected SK-BR-3, BT-474, MCF-7, and MDA-MB-468 breast cancer cells with short interfering RNA (siRNA) targeted against human HER2/neu and analyzed the specific inhibition of HER2/neu expression by Northern and Western blots. Transfection with HER2/neu-specific siRNA resulted in a sequence-specific decrease in HER2/neu mRNA and protein levels. Moreover, transfection with HER2/neu siRNA caused cell cycle arrest at G0/G1 in the breast cancer cell lines SK-BR-3 and BT-474, consistent with a powerful RNA silencing effect. siRNA treatment resulted in an antiproliferative and apoptotic response in cells overexpressing HER2/neu, but had no influence in cells with almost no expression of HER2/neu proteins like MDA-MB-468 cells. These data indicate that HER2/neu function is essential for the proliferation of HER2/neu-overexpressing breast cancer cells. Our observations suggest that siRNA targeted against human HER2/neu may be valuable tools as antiproliferative agents that display activity against neoplastic cells at very low doses.
Collapse
Affiliation(s)
- Timo Faltus
- Department of Obstetrics and Gynecology, Medical School, J. W. Goethe University, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | | | | | | | | | | | | |
Collapse
|
153
|
Le XF, Lammayot A, Gold D, Lu Y, Mao W, Chang T, Patel A, Mills GB, Bast RC. Genes affecting the cell cycle, growth, maintenance, and drug sensitivity are preferentially regulated by anti-HER2 antibody through phosphatidylinositol 3-kinase-AKT signaling. J Biol Chem 2004; 280:2092-104. [PMID: 15504738 DOI: 10.1074/jbc.m403080200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The molecular mechanisms by which the anti-HER2 antibodies trastuzumab and its murine equivalent 4D5 inhibit tumor growth and potentiate chemotherapy are not fully understood. Inhibition of signaling through the phosphatidylinositol 3-kinase (PI3K)-AKT pathway may be particularly important. Treatment of breast cancer cells that overexpress HER2 with trastuzumab inhibited HER2-HER3 association, decreased PDK1 activity, reduced Thr-308 and Ser-473 phosphorylation of AKT, and reduced AKT enzymatic activity. To place the role of PI3K-AKT in perspective, gene expression was studied by using Affymetrix microarrays and real time reverse transcription-PCR. Sixteen genes were consistently down-regulated 2.0-4.9-fold in two antibody-treated breast cancer cell lines. Fourteen of the 16 genes were involved in three major functional areas as follows: 7 in cell cycle regulation, particularly of the G(2)-M; 5 in DNA repair/replication; and 2 in modifying chromatin structure. Of the 16 antibody-regulated genes, 64% had roles in cell growth/maintenance and 52% contributed to the cell cycle. Direct inhibition of PI3K with an inhibitor markedly reduced expression of 14 genes that were also affected by the antibody. Constitutive activation of AKT1 blocked the effect of the anti-HER2 antibody on cell cycle arrest and on eight differentially expressed genes. The antibody enhanced docetaxel-induced growth inhibition but did not increase the fraction of apoptotic cells induced with docetaxel alone. In contrast, the antibody plus docetaxel markedly down-regulated two genes, HEC and DEEPEST, required for passage through G(2)-M. Thus, anti-HER2 antibody preferentially affects genes contributing to cell cycle progression and cell growth/maintenance, in part through the PI3K-AKT signaling. Transcriptional regulation by anti-HER2 antibody through PI3K-AKT pathway may potentiate the growth inhibitory activity of docetaxel by affecting cell cycle progression.
Collapse
Affiliation(s)
- Xiao-Feng Le
- Department of Experimental Therapeutics, the University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Kute T, Lack CM, Willingham M, Bishwokama B, Williams H, Barrett K, Mitchell T, Vaughn JP. Development of Herceptin resistance in breast cancer cells. Cytometry A 2004; 57:86-93. [PMID: 14750129 DOI: 10.1002/cyto.a.10095] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Herceptin, a humanized antibody to HER-2, is now utilized in the clinic for metastatic breast cancer treatment. The response rate for HER-2+ patients is only 30% and little is known as to mechanisms of resistance. The mechanism of Herceptin action is also unknown but has been related to cell cycle inhibition. METHODS The effects of Herceptin and other antibody treatments were determined by cell counting and cell cycle analysis. HER-2 and p27 expression levels were analyzed by flow cytometry and levels of activated AKT were compared by Western blot analysis. Cellular HER-2 and p27 expression was measured by immunofluorescence. RESULTS Herceptin treatment of BT-474 cells results in inhibition of cell growth and arrest in the G1 phase. The efficacy of growth arrest was not directly correlated to the binding affinity of antibodies to Her-2. Our laboratory has developed cell lines that are resistant to Herceptin treatment. In resistant cell lines, binding of antibodies is not hindered. However, Herceptin has completely lost the ability to inhibit cell proliferation. Yet, the mouse isotype 4D5 maintains significant inhibitory activity upon Herceptin-resistant clones. CONCLUSIONS Herceptin binds effectively to Her-2 on the cell surface of Herceptin-resistant cell lines and the level of Her-2 expression on the cell surface is not downregulated. Herceptin resistance is not due to downregulation of levels of AKT protein expression, although, phosphorylation of AKT is enhanced in resistant lines and could have a role in resistance. Resistance appears to correlate with the loss of nuclear expression of the cyclin-dependent kinase inhibitor, p27, as defined by immunofluorescence and flow cytometry studies and cdk-2 binding studies.
Collapse
Affiliation(s)
- Timothy Kute
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1072, USA.
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Koziczak M, Hynes NE. Cooperation between fibroblast growth factor receptor-4 and ErbB2 in regulation of cyclin D1 translation. J Biol Chem 2004; 279:50004-11. [PMID: 15377668 DOI: 10.1074/jbc.m404252200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alterations in ErbB2 or fibroblast growth factor receptor-4 (FGFR-4) expression and activity occur in a significant fraction of breast cancers. Because signaling molecules and pathways cooperate to drive cancer progression, simultaneous targeting of multiple pathways is an appealing therapeutic strategy. With this in mind, we examined breast tumor cells for their sensitivity to the ErbB2 and FGFR inhibitors, PKI166 and PD173074, respectively. Simultaneous blocking of ErbB2 and FGFR-4 in MDA-MB-453 tumor cells had a stronger anti-proliferative effect than treatment with individual inhibitors. Examination of cell cycle regulators revealed a novel translation-mediated mechanism whereby ErbB2 and FGFR-4 cooperate to regulate cyclin D1 levels. Our results showed that FGFR-4 and ErbB2 via the MAPK and the phosphatidylinositol 3-kinase/protein kinase B pathways, respectively, both contribute to the maintenance of constitutive activity of the mammalian target of rapamycin translational pathway. Dual inhibition of these receptors strongly blocked S6 kinase 1 (S6K1) activity and cyclin D1 translation, as attested by a decrease in cyclin D1 mRNA association with polysomes. Ectopic expression of active protein kinase B or active S6K1 abrogated the dual inhibitor-mediated down-regulation of cyclin D1 expression, demonstrating the importance of these FGFR-4/ErbB2 signaling targets in regulating cyclin D1 translation. S6K1 has the central role in this process, since small interfering RNA-targeted S6K1 depletion led to a decrease in cellular S6K1 activity and, as a consequence, repression of cyclin D1 expression. Thus, we propose a novel mechanism for controlling cyclin D1 expression downstream of combined activity of ErbB2 and FGFR-4 that involves S6K1-mediated translation.
Collapse
Affiliation(s)
- Magdalena Koziczak
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | | |
Collapse
|
156
|
Casalini P, Iorio MV, Galmozzi E, Ménard S. Role of HER receptors family in development and differentiation. J Cell Physiol 2004; 200:343-50. [PMID: 15254961 DOI: 10.1002/jcp.20007] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Members of the epidermal growth factor receptor family of receptor tyrosine kinases play a critical role in both development and oncogenesis. The latter is suggested by the frequent overexpression of HER-2, EGFR, and HER-3 in some human carcinomas, primarily breast and squamous cancer. The biological activities of the EGFR family are exerted through various ligand-receptor and receptor-receptor interactions. One receptor that plays a central role in this signaling network is HER-2/Neu, which is considered the preferred heterodimerization partner for other members of the EGFR family. The role of these receptors and their ligands in development is discussed, with particular emphasis on their ability to mediate a variety of pathways and cellular responses, including proliferation, differentiation, and apoptosis.
Collapse
Affiliation(s)
- Patrizia Casalini
- Molecular Targeting Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | |
Collapse
|
157
|
Nahta R, Takahashi T, Ueno NT, Hung MC, Esteva FJ. P27(kip1) down-regulation is associated with trastuzumab resistance in breast cancer cells. Cancer Res 2004; 64:3981-6. [PMID: 15173011 DOI: 10.1158/0008-5472.can-03-3900] [Citation(s) in RCA: 225] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Trastuzumab (Herceptin) is a recombinant humanized monoclonal antibody directed against HER-2. The objective response rate to trastuzumab monotherapy is 12-34% for a median duration of 9 months, by which point most patients become resistant to treatment. We created two trastuzumab-resistant (TR) pools from the SKBR3 HER-2-overexpressing breast cancer cell line to study the mechanisms by which breast cancer cells escape trastuzumab-mediated growth inhibition. Both pools maintained her-2 gene amplification and protein overexpression. Resistant cells demonstrated a higher S-phase fraction by flow cytometry and a faster doubling time of 24-36 h compared with 72 h for parental cells. The cyclin-dependent kinase inhibitor p27(kip1) was decreased in TR cells, and cyclin-dependent kinase 2 activity was increased. Importantly, exogenous addition of p27(kip1) increased trastuzumab sensitivity. Additionally, resistant cells displayed heightened sensitivity to the proteasome inhibitor MG132, which induced p27(kip1) expression. Thus, we propose that trastuzumab resistance may be associated with decreased p27(kip1) levels and may be susceptible to treatments that induce p27(kip1) expression.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030-4009, USA
| | | | | | | | | |
Collapse
|
158
|
Menendez JA, Vellon L, Mehmi I, Oza BP, Ropero S, Colomer R, Lupu R. Inhibition of fatty acid synthase (FAS) suppresses HER2/neu (erbB-2) oncogene overexpression in cancer cells. Proc Natl Acad Sci U S A 2004; 101:10715-20. [PMID: 15235125 PMCID: PMC490000 DOI: 10.1073/pnas.0403390101] [Citation(s) in RCA: 268] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Indexed: 12/11/2022] Open
Abstract
Fatty acid synthase (FAS) activity is a potential therapeutic target to treat cancer and obesity. Here, we have identified a molecular link between FAS and HER2 (erbB-2) oncogene, a marker for poor prognosis that is overexpressed in 30% of breast and ovarian cancers. Pharmacological FAS inhibitors cerulenin and C75 were found to suppress p185(HER2) oncoprotein expression and tyrosine-kinase activity in breast and ovarian HER2 overexpressors. Similarly, p185(HER2) expression was dramatically down-regulated when FAS gene expression was silenced by using the highly sequence-specific mechanism of RNA interference (RNAi). Pharmacological and RNAi-mediated silencing of FAS specifically down-regulated HER2 mRNA and, concomitantly, caused a prominent up-regulation of PEA3, a transcriptional repressor of HER2. A cytoplasmic redistribution of p185(HER2) was associated with marked morphological changes of FAS RNAi-transfected cells, whereas chemical inhibitors of FAS promoted a striking nuclear accumulation of p185(HER2). The simultaneous targeting of FAS and HER2 by chemical FAS inhibitors and the humanized antibody directed against p185(HER2) trastuzumab, respectively, was synergistically cytotoxic toward HER2 overexpressors. Similarly, concurrent RNAi-mediated silencing of FAS and HER2 genes synergistically stimulated apoptotic cell death in HER2 overexpressors. p185(HER2) was synergistically down-regulated after simultaneous inhibition of FAS and HER2 by either pharmacological inhibitors or small interfering RNA. These findings provide evidence of an active role of FAS in cancer evolution by specifically regulating oncogenic proteins closely related to malignant transformation, strongly suggesting that HER2 oncogene may act as the key molecular sensor of energy imbalance after the perturbation of tumor-associated FAS hyperactivity in cancer cells.
Collapse
MESH Headings
- 4-Butyrolactone/analogs & derivatives
- 4-Butyrolactone/metabolism
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antifungal Agents/metabolism
- Antineoplastic Agents/metabolism
- Apoptosis
- Biomarkers, Tumor
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line
- Cell Size
- Cell Survival
- Cerulenin/metabolism
- Fatty Acid Synthases/antagonists & inhibitors
- Fatty Acid Synthases/genetics
- Fatty Acid Synthases/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Genes, erbB-2
- Humans
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Signal Transduction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Trastuzumab
Collapse
Affiliation(s)
- Javier A Menendez
- Department of Medicine, Evanston Northwestern Healthcare, 1001 University Place, Evanston, IL 60201, USA
| | | | | | | | | | | | | |
Collapse
|
159
|
Abstract
The ErbB family of receptors and ligands is a complex, delicately balanced system involved in the growth and differentiation of normal human cells as well as neoplasms. Targeting this system with therapies that inhibit ErbB receptor activity in cancer patients has been somewhat successful, but resistance to ErbB inhibitor monotherapy is substantial. An understanding of the biology of ErbB receptor inhibitors is necessary to determine how best to utilize them in treatment regimens. Experimental evidence has provided valuable insights regarding mechanisms involved in resistance, and indicates that resistance can be reversed in some models. Ongoing studies in patients are evaluating whether agents that target both the epidermal growth factor (EGFR, ErbB-1) and ErbB-2 (HER-2) receptors can prevent or delay resistance in patients with metastatic breast cancer that overexpresses HER-2. With a better understanding of the biology of breast cancer, and with several novel ErbB receptor inhibitors in development, continued progress for improved patient outcomes is expected.
Collapse
Affiliation(s)
- Kathy D Miller
- Indiana University School of Medicine, Indianapolis, 46202, USA.
| |
Collapse
|
160
|
Vera-Román JM, Rubio-Martínez LA. Comparative Assays for the HER-2/neu Oncogene Status in Breast Cancer. Arch Pathol Lab Med 2004; 128:627-33. [PMID: 15163239 DOI: 10.5858/2004-128-627-caftno] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Abstract
Context.—Tumor marker assays, especially those used to indicate the right therapy, should be standardized.
Objective.—To analyze the current methods for the HER-2/neu (h2n) oncogene status by immunohistochemical (IHC) analysis, fluorescence in situ hybridization (FISH), and chromogenic in situ hybridization (CISH) and compare those results with the chromosome 17 copy number and the status of the topoisomerase II alpha (TPIIα) gene.
Design.—We tested 50 infiltrating ductal breast carcinomas (pTNM status varied from pT1 N0 to pT4 N1) using the Food and Drug Administration (FDA)–approved methods HercepTest and Pathway for overexpression of h2n. We also used FISH and CISH to test for h2n amplification and CISH to test for chromosome 17 (c17) and TPIIα. The p53 and Ki-67 factors were also evaluated by IHC analysis.
Results.—h2n overexpression (3+) and amplification were observed in only 6 (12%) of 50 cases by IHC analysis, FISH, and CISH. Three cases that initially scored 3+ and 2+ had 4 to 5.95 signals (equivocal) by FISH but when corrected by the h2n/c17 ratio were nonamplified. TPIIα isomerase was amplified in only 2 (4%) of the 50 cases. Nineteen (38%) of the 50 cases were aneuploidic. All h2n amplified cases had high proliferative activity, but only 2 of 6 had p53 protein alterations.
Conclusions.—The HercepTest and Pathway IHC assay h2n were fully concordant for the 3+ cases. The 3+ cases had to be confirmed in 75% of the tumor area examined. These 2 IHC assays were fully concordant with FISH and CISH. The 2 in situ hybridization (ISH) assays were 94% concordant for the 50 cases. The cutoff signal points for both ISH assays should be 6 or more. Thus, there is no need for the c17 ratio correction. Tumor heterogeneity appears not be a major problem, but our percentage of amplified cases is lower than previously reported. The FDA-approved IHC and ISH assays should give relatively uniform results when used following our recommendations.
Collapse
Affiliation(s)
- José María Vera-Román
- Department of Pathology, Hospital General de Castellón, Castellón de la Plana, Spain.
| | | |
Collapse
|
161
|
Abstract
The epidermal growth factor (EGF)-related peptides bind the ErbB receptors, inducing the formation of different homo- and heterodimers. Receptor dimerization promotes activation of the intrinsic kinase, leading to phosphorylation of specific tyrosines located in the ErbB's cytoplasmic region. These phosphorylated residues serve as docking sites for a variety of signaling molecules whose recruitment stimulates intracellular signaling cascades, which ultimately control diverse genetic programs. Particular ligand-receptor complexes have essential roles in embryonic development as well as in the adult. Finally, ErbB receptors are being pursued as therapeutic targets because aberrant ErbB activity has been observed in many human cancers. In this review, we discuss these data in more detail, illustrating the importance of tightly regulated ErbB signaling throughout life.
Collapse
Affiliation(s)
- Thomas Holbro
- Friedrich Miescher Institute for Biomedical Research, 4002 Basel, Switzerland.
| | | |
Collapse
|
162
|
Koziczak M, Holbro T, Hynes NE. Blocking of FGFR signaling inhibits breast cancer cell proliferation through downregulation of D-type cyclins. Oncogene 2004; 23:3501-8. [PMID: 15116089 DOI: 10.1038/sj.onc.1207331] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Overexpression of fibroblast growth factor receptor (FGFR) tyrosine kinases has been found in many human breast cancers and has been associated with poor patient prognosis. In order to understand the mechanism by which FGFR mediates breast cancer cell proliferation, we used a low molecular weight compound, PD173074, that selectively inhibits FGFR tyrosine kinase activity and autophosphorylation. This potential anticancer agent caused a G1 growth arrest of MDA-MB-415, MDA-MB-453 and SUM 52 breast cancer cells. Our analyses revealed that FGFR signaling links to the cell cycle machinery via D-type cyclins. PD173074-mediated inhibition of FGFR activity caused downregulation of cyclin D1 and cyclin D2 expression, inhibition of cyclin D/cdk4 activity and, as a consequence, reduction of pRB phosphorylation. Retroviral-mediated ectopic expression of cyclin D1 prevented pRB hypophosphorylation and the cell cycle G1 block in PD173074-treated cells, suggesting a central role for D cyclins in proliferation of FGFR-driven breast cancer cells. The repression of FGFR activity caused downregulation of MAPK in MDA-MB-415 and MDA-MB-453 cells. In SUM 52 cells, both MAPK and PI3K signaling pathways were suppressed. In conclusion, results shown here describe a mechanism by which FGFR promotes proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Magdalena Koziczak
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | | | | |
Collapse
|
163
|
Abstract
Emerging results from clinical trials with epidermal growth factor receptor (EGFR) inhibitors indicate good tolerability and, at best, modest to no clinical activity in a variety of epithelial tumors, including breast carcinomas. Although the EGFR is widely expressed in epithelial cancers, there is no evidence of frequent EGFR alterations at the DNA level in human tumors. This lack of molecular evidence to suggest a pathogenic role for the EGFR in breast cancer questions the notion that use of single-agent EGFR inhibitors in this disease is a viable therapeutic approach. The lack of selection of EGFR-dependent breast tumors into trials with EGFR inhibitors suggests the possibility that these were not true-negative studies. A point of view regarding challenges in the development of anti-EGFR therapies for patients with breast cancer and possible approaches to overcome them is presented in this article.
Collapse
Affiliation(s)
- Carlos L Arteaga
- Department of Medicine, Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
164
|
Boulay A, Zumstein-Mecker S, Stephan C, Beuvink I, Zilbermann F, Haller R, Tobler S, Heusser C, O'Reilly T, Stolz B, Marti A, Thomas G, Lane HA. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res 2004; 64:252-61. [PMID: 14729632 DOI: 10.1158/0008-5472.can-3554-2] [Citation(s) in RCA: 265] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The orally bioavailable rapamycin derivative RAD001 (everolimus) targets the mammalian target of rapamycin pathway and possesses potent immunosuppressive and anticancer activities. Here, the antitumor activity of RAD001 was evaluated in the CA20948 syngeneic rat pancreatic tumor model. RAD001 demonstrated dose-dependent antitumor activity with daily and weekly administration schedules; statistically significant antitumor effects were observed with 2.5 and 0.5 mg/kg RAD001 administered daily [treated tumor versus control tumor size (T/C), 23% and 23-30%, respectively], with 3-5 mg/kg RAD001 administered once weekly (T/C, 14-36%), or with 5 mg/kg RAD001 administered twice weekly (T/C, 36%). These schedules were well tolerated and exhibited antitumor potency similar to that of the cytotoxic agent 5-fluorouracil (T/C, 23%). Moreover, the efficacy of intermittent treatment schedules suggests a therapeutic window allowing differentiation of antitumor activity from the immunosuppressive properties of this agent. Detailed biochemical profiling of mammalian target of rapamycin signaling in tumors, skin, and peripheral blood mononuclear cells (PBMCs), after a single administration of 5 mg/kg RAD001, indicated that RAD001 treatment blocked phosphorylation of the translational repressor eukaryotic initiation factor 4E-binding protein 1 and inactivated the translational activator ribosomal protein S6 kinase 1 (S6K1). The efficacy of intermittent treatment schedules was associated with prolonged inactivation of S6K1 in tumors and surrogate tissues (> or =72 h). Furthermore, detailed analysis of the dose dependency of weekly treatment schedules demonstrated a correlation between antitumor efficacy and prolonged effects (> or =7 days) on PBMC-derived S6K1 activity. Analysis of human PBMCs revealed that S6K1 also underwent a concentration-dependent inactivation after RAD001 treatment ex vivo (>95% inactivation with 20 nM RAD001). In contrast, human PBMC-derived eukaryotic initiation factor 4E-binding protein 1 was present predominantly in the hypophosphorylated form and was unaffected by RAD001 treatment. Taken together, these results demonstrate a correlation between the antitumor efficacy of intermittent RAD001 treatment schedules and prolonged S6K1 inactivation in PBMCs and suggest that long-term monitoring of PBMC-derived S6K1 activity levels could be used for assessing RAD001 treatment schedules in cancer patients.
Collapse
Affiliation(s)
- Anne Boulay
- Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Bennasroune A, Gardin A, Aunis D, Crémel G, Hubert P. Tyrosine kinase receptors as attractive targets of cancer therapy. Crit Rev Oncol Hematol 2004; 50:23-38. [PMID: 15094157 DOI: 10.1016/j.critrevonc.2003.08.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2003] [Indexed: 12/24/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are the main mediators of the signaling network that transmit extracellular signals into the cell, and control cellular differentiation and proliferation. Recent and rapid advances in our understanding of cellular signaling by receptor tyrosine kinases, in normal and malignant cells, have brought to light the potential of RTKs as selective anti-cancer targets. Their activity is normally tightly controlled and regulated. Overexpression of RTK proteins or functional alterations caused by mutations in the corresponding genes or abnormal stimulation by autocrine growth factor loops contribute to constitutive RTK signaling, resulting in dysregulated cell growth and cancer. The mechanisms of uncontrolled RTK signaling that leads to cancer has provided the rationale for anti-RTK drug development. Herceptin, Gleevec, and Iressa are the first examples of drugs which have successfully translated basic research on oncogenes into cancer therapeutics. RTKs can be viewed as multifunctional targets, and strategies towards the prevention and inhibition of RTK signaling include antibodies, antagonist ligands, small molecule inhibitors of protein kinase activity, and inhibitors of protein-protein interactions. Progresses in the field of rational drug design and computational chemistry will vastly benefit from the availability of increasing structural knowledge of both the kinase domains and the ligand-binding sites of these receptors.
Collapse
Affiliation(s)
- Amar Bennasroune
- INSERM Unit 575, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | | | | | |
Collapse
|
166
|
Yang C, Ionescu-Tiba V, Burns K, Gadd M, Zukerberg L, Louis DN, Sgroi D, Schmidt EV. The role of the cyclin D1-dependent kinases in ErbB2-mediated breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1031-8. [PMID: 14982856 PMCID: PMC1614725 DOI: 10.1016/s0002-9440(10)63190-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Intact cyclin D1 functions are essential for transformation by erbB2 in tissue culture and murine models. Because cyclin D1 may alter cell proliferation through a variety of mechanisms, we used transgenic models and human tumor samples to particularly address the role of cyclin D1-cyclin-dependent kinases in transformation by erbB2. The p16 tumor suppressor specifically blocks cyclin-dependent kinase 4 and 6 activity. Here we show that an MMTV-p16 transgene blocked tumorigenesis by erbB2, demonstrating that deregulation of the cyclin-dependent kinase partner of cyclin D1 is an essential target of erbB2. ErbB2 overexpression was a determining factor in deregulation of cyclin D1-cdk4/6 interactions because neither transgenic cyclin D1 nor loss of p16 accelerated tumorigenesis in MMTV-erbB2-transgenic mice. ErbB2 was also a deciding factor in deregulation of cyclin D1-cdk4/6 in human tumors because no loss of pRb or p16 was found in tumors overexpressing erbB2, although erbB2-negative invasive breast adenocarcinomas frequently lacked expression of p16 or pRb. We conclude that deregulation of cyclin D1-Cdk4/6 interactions is a critical target of erbB2 function in human and mouse breast tumors, and erbB2's overexpression may be sufficient to deregulate cyclin D1-cdk4/6 activity in breast cancer.
Collapse
Affiliation(s)
- Chuanwei Yang
- Massachusetts General Hospital (MGH) Cancer Research Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
167
|
Marmor MD, Skaria KB, Yarden Y. Signal transduction and oncogenesis by ErbB/HER receptors. Int J Radiat Oncol Biol Phys 2004; 58:903-13. [PMID: 14967450 DOI: 10.1016/j.ijrobp.2003.06.002] [Citation(s) in RCA: 267] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2003] [Accepted: 06/25/2003] [Indexed: 12/31/2022]
Abstract
Growth factors enable cells to escape irradiation-induced death (apoptosis). One important family of growth factors share an epidermal growth factor motif, and all bind to ErbB transmembrane receptors. In response to growth factor ligands, ErbB receptor tyrosine kinases induce a variety of cellular responses, including proliferation, differentiation and motility. Signal transduction pathways are initiated upon ligand-induced receptor homo- or heterodimerization and activation of tyrosine kinase activity. The complement of induced signaling pathways, as well as their magnitude and duration, determines the biological outcome of signaling, and in turn, is regulated by the identity of the ligand and the receptor composition. Recent insights into the structural basis for receptor dimerization, as provided by crystallographic analysis, are described, as is the differential activation of signaling pathways and downregulatory mechanisms. Further, dysregulation of the ErbB network is implicated in a variety of human cancers, and the nature of aberrant signaling through ErbB proteins, as well as current therapeutic approaches, are discussed, highlighting the role of the highly oncogenic ErbB-2 molecule.
Collapse
Affiliation(s)
- Mina D Marmor
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
168
|
Spadaro M, Lanzardo S, Curcio C, Forni G, Cavallo F. Immunological inhibition of carcinogenesis. Cancer Immunol Immunother 2004; 53:204-16. [PMID: 14722672 PMCID: PMC11032997 DOI: 10.1007/s00262-003-0483-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2003] [Accepted: 10/24/2003] [Indexed: 10/26/2022]
Abstract
The combination of new information provided by fundamental immunology, along with the refinement of genetic engineering techniques has given scientists the capacity to produce vaccines able to inhibit the growth of most if not every transplantable tumor. However, when faced with already established tumors, vaccines fail to afford any significant protection. Many studies are underway which seek to overcome this gloomy situation. However, another possibility is to follow the indications provided by a large quantity of experimental data and to evaluate the possibility of using immunotherapy to prevent the initial stages of tumor growth. Is it possible to prevent an autologous tumor by means of a vaccination performed before tumor onset? Could antitumor vaccines be a new form of preventive medicine in the wake of Jenner, Pasteur, and other pioneers? In this paper it is our intention to review the results obtained by our laboratory in the attempt to use natural and adaptive immunity in the control of carcinogenesis. Natural immunity boosted by IL-12 and IL-2 significantly hampers the progression of mammary lesions occurring in HER-2/neu transgenic mice genetically predestined to develop lethal mammary carcinomas. Specific immunity elicited by DNA vaccination provides a much stronger inhibition of the development of mammary lesions, and a significant number of transgenic mice are tumor free at 1 year of age. These experimental data suggest the possibility of using immunity as a means of controlling preneoplastic lesions and protecting healthy persons at risk of developing cancer.
Collapse
Affiliation(s)
- Michela Spadaro
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Stefania Lanzardo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Claudia Curcio
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Guido Forni
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Federica Cavallo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| |
Collapse
|
169
|
Abstract
p27Kip1 is an important regulator of the G1 to S transition. While a potent inhibitor of cyclin-dependent-kinase (Cdk)2, p27 is also involved in assembly of cyclin D/Cdk4 complexes. Although rarely mutated, p27 is functionally downregulated in many human cancers by mechanisms involving enhanced degradation, cytoplasmic mislocalization, and/or sequestration by cyclin D/Cdk complexes in response to oncogenic signals. Therefore, low levels and/or cytoplasmic localized p27 have been associated with enhanced malignancy and poor patient prognosis in many neoplasias including breast cancer. Recent data discussed below suggest that a threshold of p27 is required for response to antiestrogens and, conversely, that low levels predict for antiestrogen resistance. These results imply that hormone receptor-positive tumors with low and/or cytosolic p27 respond poorly to antiestrogens and should be considered for alternative therapeutic strategies.
Collapse
Affiliation(s)
- Carlos L Arteaga
- Departments of Medicine and Cancer Biology and Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
170
|
Argiris A, Wang CX, Whalen SG, DiGiovanna MP. Synergistic Interactions between Tamoxifen and Trastuzumab (Herceptin). Clin Cancer Res 2004; 10:1409-20. [PMID: 14977844 DOI: 10.1158/1078-0432.ccr-1060-02] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE HER-2/neu and estrogen receptor (ER) are critical in the biology of breast carcinoma, and both are validated therapeutic targets. Extensive interactions between the signaling pathways of these receptors have been demonstrated. This suggests that targeting both receptors simultaneously may have a dramatic effect on the biology of breast cancer. This hypothesis was tested in cell culture experiments. EXPERIMENTAL DESIGN ER-positive, HER-2/neu-overexpressing BT-474 human breast carcinoma cells were cultured in the presence of the anti-HER-2/neu therapeutic antibody trastuzumab (Herceptin), the antiestrogen tamoxifen, or both. The effects on cell growth, cell cycle distribution, clonogenicity, survival, and the level and activity of HER-2/neu were examined. RESULTS The combination of tamoxifen and Herceptin resulted in synergistic growth inhibition and enhancement of cell accumulation in the G(0)-G(1) phase of the cell cycle, with a decrease in cells in S phase. Clonogenicity was inhibited in the presence of each drug and more so by the combination, although prior exposure to drugs did not affect subsequent clonogenicity in drug-free media, and neither drug nor the combination induced apoptosis. Herceptin, but not tamoxifen, inhibited signaling by HER-2/neu. CONCLUSIONS The combination of tamoxifen and Herceptin is formally demonstrated to result in synergistic growth inhibition and enhancement of G(0)-G(1) cell cycle accumulation. In vitro, the individual drugs or combination produces a cytostatic effect. These results suggest that combined inhibition of ER and HER-2/neu signaling may represent a powerful approach to the treatment of breast cancer.
Collapse
Affiliation(s)
- Athanassios Argiris
- Yale University School of Medicine, Department of Internal Medicine, New Haven, Connecticut 06510, USA
| | | | | | | |
Collapse
|
171
|
Albanell J, Codony J, Rovira A, Mellado B, Gascón P. Mechanism of action of anti-HER2 monoclonal antibodies: scientific update on trastuzumab and 2C4. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 532:253-68. [PMID: 12908564 DOI: 10.1007/978-1-4615-0081-0_21] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The HER family of transmembrane tyrosine kinase receptors is composed of four members, BER1 to HER4. HER2 is a ligand-orphan receptor expressed in many human tumors and overexpressed in 25-30% of breast cancers. HER2 amplifies the signal provided by other receptors of the HER family by forming heterodimers. The essential role of HER2 in the HER signaling network led to the development of anti-HER2 monoclonal antibodies (MAbs) for cancer therapy. In particular, the humanized MAb trastuzumab (Herceptin) has antitumor activity against HER2-overexpressing human breast tumor cells and is widely used for the treatment of women with HER2 overexpressing breast cancers. Trastuzumab induces HER2 receptor downmodulation and, as a result, inhibits critical signalling pathways (i.e. ras-Raf-MAPK and PI3K/Akt) and blocks cell cycle progression by inducing the formation of p27/Cdk2 complexes. Trastuzumab also inhibits HER2 cleavage, preceding antibody-induced receptor downmodulation, and this effect might contribute to its antitumor activity in some cancers. In vivo, trastuzumab inhibits angiogenesis and induces antibody-dependent cellular cytotoxicity. A limitation of trastuzumab is that its activity is largely restricted to breast cancers with the highest level of HER2 overexpression or HER2 gene amplification. However, there is a large population of breast cancers and of many other tumors that have low or moderate HER2 expression. In such tumors, HER2 functions as a preferred coreceptor to form heterodimers with HER1 (EGFR), HER3 or HER4. For this reason, a humanized monoclonal antibody, called 2C4, that targets the role of HER2 as a coreceptor is under active development. 2C4 binds to a different epitope of HER2 ectodomain than trastuzumab and sterically hinders HER2 recruitment in heterodimers with other HER receptors. This results in the inhibition of signalling by HER2-based heterodimers both in cells with low and high HER2 expression. In vitro and in vivo antitumor activity has been reported in a range of breast and prostate tumor models. Therefore, 2C4 may have potential against a wide variety of solid tumors. Phase I trials are underway.
Collapse
Affiliation(s)
- Joan Albanell
- ICMHO, Laboratory of Oncology Research, Medical Oncology Service, Hospital Clinic i Provincial de Barcelona, Spain.
| | | | | | | | | |
Collapse
|
172
|
Nicholson KM, Streuli CH, Anderson NG. Autocrine signalling through erbB receptors promotes constitutive activation of protein kinase B/Akt in breast cancer cell lines. Breast Cancer Res Treat 2004; 81:117-28. [PMID: 14572154 DOI: 10.1023/a:1025765215765] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The protein kinase PKB/Akt plays a pivotal role in promoting cell survival and proliferation. This study investigated the regulation of PKB/Akt activity in breast cancer cells. In primary invasive breast cancers PKB/Akt exhibited elevated phosphorylation at regulatory site Ser473 in 80% of cases, using immunohistochemistry. The degree of phospho-PKB/Akt immunoreactivity was positively correlated with the extent of its nuclear accumulation. Moderate/strong staining was seen in 31% of the samples but was absent in tumour-associated normal breast epithelia. To examine the mechanisms of PKB/Akt activation, we studied its phosphorylation in a panel of breast cancer cell lines. PKB/Akt was constitutively phosphorylated on both regulatory sites (Thr308 and Ser473) in the absence of serum growth factors in 7 of 8 lines but not in two cell lines derived from normal breast epithelia. Further analysis revealed that constitutive PKB/Akt phosphorylation was associated with loss of PTEN phosphatase expression (CAL51, MDA-MB-468, BT549 cells) and constitutive activation of erbB2 (SKBR3, BT474 cells). In two further breast cancer lines (T47D and HS578T) PKB/Akt phosphorylation was dependent upon autocrine factors acting primary through the epidermal growth factor receptor (EGFR) and erbB2. Conditioned medium from HS578T cells stimulated EGFR-dependent PKB/Akt phosphorylation in normal breast cells. These results demonstrate that PKB/Akt is frequently activated in breast cancer through diverse mechanisms, including autocrine signalling via erbB receptors.
Collapse
Affiliation(s)
- K M Nicholson
- Division of Cancer Studies, School of Medicine, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
173
|
Lu Y, Zi X, Pollak M. Molecular mechanisms underlying IGF-I-induced attenuation of the growth-inhibitory activity of trastuzumab (Herceptin) on SKBR3 breast cancer cells. Int J Cancer 2004; 108:334-41. [PMID: 14648698 DOI: 10.1002/ijc.11445] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The clinical usefulness of trastuzumab (Herceptin; Genentech, San Francisco, CA) in breast cancer treatment is limited by the rapid development of resistance. We previously reported that IGF-I signaling confers resistance to the growth-inhibitory actions of trastuzumab in a model system, but the underlying molecular mechanism remains unknown. We used SKBR3/neo cells (expressing few IGF-I receptors) and SKBR3/IGF-IR cells (overexpressing IGF-I receptor) as our experimental model. IGF-I antagonized the trastuzumab-induced increase in the level of the Cdk inhibitor p27(Kip1). This resulted in decreased association of p27(Kip1) with Cdk2, restoration of Cdk2 activity and attenuation of cell-cycle arrest in G(1) phase, all of which had been induced by trastuzumab treatment in SKBR3/IGF-IR cells. We also found that the decrease in p27(Kip1) induced by IGF-I was accompanied by an increase in expression of Skp2, which is a ubiquitin ligase for p27(Kip1), and by increased Skp2 association with p27(Kip1). A specific proteasome inhibitor (LLnL) completely blocked the ability of IGF-I to reduce the p27(Kip1) protein level, while IGF-I increased p27(Kip1) ubiquitination. This suggests that the action of IGF-I in conferring resistance to trastuzumab involves targeting of p27(Kip1) to the ubiquitin/proteasome degradation machinery. Finally, specific inhibitors of MAPK and PI3K suggest that the IGF-I-mediated reduction in p27(Kip1) protein level by increased degradation predominantly involves the PI3K pathway. Our results provide an example of resistance to an antineoplastic therapy that targets one tyrosine kinase receptor by increased signal transduction through an alternative pathway in a complex regulatory network.
Collapse
Affiliation(s)
- Yuhong Lu
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
174
|
Musgrove EA, Davison EA, Ormandy CJ. Role of the CDK inhibitor p27 (Kip1) in mammary development and carcinogenesis: insights from knockout mice. J Mammary Gland Biol Neoplasia 2004; 9:55-66. [PMID: 15082918 DOI: 10.1023/b:jomg.0000023588.55733.84] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The cyclin-dependent kinase inhibitor p27 (Kip1) is an important cell cycle regulatory gene in breast cancer, and decreased p27 expression is associated with poor prognosis. Some investigations of its role in mammary development have demonstrated reduced cyclin D1 expression and consequent lack of lobuloalveolar development, but others have found increased cyclin E-Cdk2 activity and increased proliferation balanced by increased apoptosis. It is unclear at present why these apparently divergent results have been obtained. Mice with reduced p27 gene dosage alone do not develop mammary carcinomas but do display substantially shorter tumor latency upon overexpression of erbB2, consistent with a role for p27 as a mammary tumor suppressor gene. In this review we summarize these and other data addressing the role of p27 in normal mammary epithelium and experimental models of mammary carcinogenesis.
Collapse
Affiliation(s)
- Elizabeth A Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, Australia.
| | | | | |
Collapse
|
175
|
Alkarain A, Jordan R, Slingerland J. p27 deregulation in breast cancer: prognostic significance and implications for therapy. J Mammary Gland Biol Neoplasia 2004; 9:67-80. [PMID: 15082919 DOI: 10.1023/b:jomg.0000023589.00994.5e] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
p27 is a key regulator of G1-to-S phase progression. It prevents premature activation of cyclin E-cdk2 in G1 and promotes the assembly and activation of D-type cyclin-cdks. While the p27 gene is rarely mutated in human cancers, the action of p27 is impaired in breast and other human cancers through accelerated p27 proteolysis, sequestration by cyclin D-cdks, and by p27 mislocalization in tumor cell cytoplasm. Reduced p27 protein is strongly associated with high histopathologic tumor grade, reflecting a lack of tumor differentiation. Loss of p27 is also an indicator of poor patient outcome in a majority of breast cancer studies, including node negative disease. The broad application of p27 in the clinical evaluation of breast cancer prognosis will require a consensus on methods of tumor fixation, staining, and scoring. This review will focus on mechanisms of p27 regulation in normal cells and how deregulation of p27 may arise in breast and other human cancers. The prognostic significance of p27 in human breast cancer and the possible therapeutic implications of these findings will also be reviewed.
Collapse
Affiliation(s)
- A Alkarain
- Molecular and Cell Biology, Sunnybrook and Women's Health Sciences Centre, University of Toronto, Bayview Avenue, Toronto, Ontario, Canada
| | | | | |
Collapse
|
176
|
Marches R, Uhr JW. Enhancement of the p27Kip1-mediated antiproliferative effect of trastuzumab (Herceptin) on HER2-overexpressing tumor cells. Int J Cancer 2004; 112:492-501. [PMID: 15382077 DOI: 10.1002/ijc.20378] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The oncogenic activity of the overexpressed HER2 tyrosine kinase receptor requires its localization in the plasma membrane. The antitumor effect of anti-HER2 antibodies (Abs) is mainly dependent on receptor downregulation and comprises p27Kip1-mediated G1 cell cycle arrest. However, one major limitation of anti-HER2 therapy is the reversibility of tumor growth inhibition after discontinuation of treatment caused by the mitogenic signaling associated with cell surface receptor re-expression. We found that the level of p27Kip1 upregulation, inhibition of Cdk2 activity and magnitude of G1 arrest induced by the humanized Ab trastuzumab (Herceptin, HCT) on BT474 and SKBr3 HER2-overexpressing breast cancer cells correlates with the level of cell surface receptor. Thus, continuous exposure of cells to HCT for 72 hr results in downregulation of the cell surface receptor and a concurrent increase in the level of p27Kip1 protein. Discontinuation of Ab exposure after the first 8 hr results in failure to upregulate p27Kip1 and arrest of cell cycle progression. We show that the lysosomotropic amine chloroquine (CQ) augments receptor internalization in HER2-overexpressing cells either pretreated or continuously treated with HCT and leads to an increased and sustained inhibitory effect. The enhanced CQ-dependent loss of functional HER2 from the cell surface resulted in sustained inactivation of the serine/threonine kinase Akt, upregulation of p27Kip1 protein and inhibition of cyclin E/Cdk2 activity. Potentiation of the inhibitory effect of HCT by CQ was directly related to loss of HER2 from the plasma membrane since prevention of Ab-mediated receptor endocytosis by engagement of the receptor with immobilized HCT abrogated the effect of CQ.
Collapse
Affiliation(s)
- Radu Marches
- Cancer Immunobiology Center and Department of Microbiology, University of Texas Southwestern Medical School, Dallas, TX, USA.
| | | |
Collapse
|
177
|
Alkarain A, Slingerland J. Deregulation of p27 by oncogenic signaling and its prognostic significance in breast cancer. Breast Cancer Res 2003; 6:13-21. [PMID: 14680481 PMCID: PMC314445 DOI: 10.1186/bcr722] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
p27 is a key regulator of progression from G1 to S phase. Although the gene encoding p27 is rarely mutated in human cancers, p27 is functionally inactivated in a majority of human cancers through accelerated p27 proteolysis, through sequestration by cyclin D-cyclin-dependent kinase complexes and by cytoplasmic mislocalization. Here we review mechanisms whereby oncogenic activation of receptor tyrosine kinase and Ras pathways lead to accelerated p27 proteolysis and p27 mislocalization in cancer cells. The prognostic significance of p27 in human breast cancer is also reviewed.
Collapse
Affiliation(s)
- Angel Alkarain
- Sunnybrook and Women's Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Joyce Slingerland
- Braman Breast Cancer Institute, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
178
|
Hudelist G, Köstler WJ, Attems J, Czerwenka K, Müller R, Manavi M, Steger GG, Kubista E, Zielinski CC, Singer CF. Her-2/neu-triggered intracellular tyrosine kinase activation: in vivo relevance of ligand-independent activation mechanisms and impact upon the efficacy of trastuzumab-based treatment. Br J Cancer 2003; 89:983-91. [PMID: 12966413 PMCID: PMC2376939 DOI: 10.1038/sj.bjc.6601160] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Proteolytic cleavage of the Her-2/neu extracellular domain (ECD) has been shown to initiate receptor phosphorylation representing Her-2/neu activation in vitro. The present investigation was performed to evaluate the clinical relevance of ECD cleavage for Her-2/neu activation and the consequences of active intracellular Her-2/neu signalling reflected by tyrosine kinase phosphorylation in patients treated with the anti-Her-2/neu antibody trastuzumab. Sera from 62 patients receiving trastuzumab-based treatment for Her-2/neu overexpressing metastatic breast cancer were assessed for pretreatment ECD levels using an enzyme-linked immunosorbent assay. In parallel, Her-2/neu activation status of tumour specimens was assessed by immunohistochemistry using a Her-2/neu phosphorylation state specific antibody (PN2A) and correlated with the patients' ECD levels and clinical course of disease. Serum ECD levels were significantly higher in 15 (24%) patients with tumours exhibiting activated Her-2/neu as compared to those without detectable Her-2/neu phosphorylation (median 148.2 vs 28.5 ng ml(-1), P=0.010). Whereas response rate only showed a trend to be higher in patients with Her-2/neu-phosphorylated breast cancer (47 vs 34%, P=0.197), both uni- and multivariate analyses revealed that the median progression-free survival under trastuzumab-based treatment was significantly longer in patients with Her-2/neu-phosphorylated breast cancer-11.7 (95% CI 5.2-18.3) months-when compared to the progression-free survival of 4.5 (95% CI 3.4-5.6) months observed in patients with tumours lacking phosphorylated Her-2/neu (P=0.001). Proteolytic cleavage of the ECD represents a biologically relevant ligand-independent mechanism of Her-2/neu activation in vivo. The influence of Her-2/neu activation status upon the outcome of trastuzumab-based therapies merits further investigation in larger prospective trials.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Enzyme Activation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoglobulin G/metabolism
- Ligands
- Middle Aged
- Phosphorylation
- Phosphotyrosine/metabolism
- Prognosis
- Protein-Tyrosine Kinases/metabolism
- Receptor, ErbB-2/metabolism
- Receptors, Progesterone/metabolism
- Survival Rate
- Trastuzumab
- Treatment Outcome
Collapse
Affiliation(s)
- G Hudelist
- Clinical Division of Special Gynaecology, Department of Obstetrics and Gynaecology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
| | - W J Köstler
- Clinical Division of Oncology, Department of Medicine I, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
- Clinical Division of Oncology, Department of Medicine I, University Hospital of Vienna, 18-20 Waehringer Guertel, A-1090 Vienna, Austria. E-mail:
| | - J Attems
- Department of Pathology, Otto Wagner Hospital, Vienna, Austria
| | - K Czerwenka
- Division of Gynaecopathology, Department of Pathology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
| | - R Müller
- Division of Gynaecopathology, Department of Pathology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
| | - M Manavi
- Clinical Division of Special Gynaecology, Department of Obstetrics and Gynaecology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
| | - G G Steger
- Clinical Division of Oncology, Department of Medicine I, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
| | - E Kubista
- Clinical Division of Special Gynaecology, Department of Obstetrics and Gynaecology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Clinical-Experimental Oncology, Vienna, Austria
| | - C C Zielinski
- Clinical Division of Oncology, Department of Medicine I, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Clinical-Experimental Oncology, Vienna, Austria
| | - C F Singer
- Clinical Division of Special Gynaecology, Department of Obstetrics and Gynaecology, and Center for Excellence in Clinical and Experimental Oncology, University Hospital, Vienna, Austria
- Ludwig Boltzmann Institute for Clinical-Experimental Oncology, Vienna, Austria
| |
Collapse
|
179
|
Abstract
Identification of the key role of protein kinases as potential oncoproteins has led to the emergence of a new era of target-directed therapies. Among a variety of novel therapeutic strategies two have shown the most promise and led to a variety of therapeutic agents in clinical development. One approach utilises humanised monoclonal antibodies generated against the extracellular domain of transmembrane protein kinases. The second approach is the generation of small molecule ATP analogues targeting the kinase domain itself. The approval of agents such as Herceptin for the treatment of advanced breast cancer and Gleevec for chronic myelogenous leukemia and gastrointestinal stromal tumours are the first examples of gene-based cancer drugs and represent the first example of a novel strategy in anti-cancer therapy.
Collapse
Affiliation(s)
- Oliver M Fischer
- Max-Planck-Institute of Biochemistry, Department of Molecular Biology, Am Klopferspitz 18A, D-82152 Martinsried, Germany
| | | | | | | |
Collapse
|
180
|
Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF, Hynes NE. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci U S A 2003; 100:8933-8. [PMID: 12853564 PMCID: PMC166416 DOI: 10.1073/pnas.1537685100] [Citation(s) in RCA: 761] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
ErbB2 is a receptor tyrosine kinase whose activity in normal cells depends on dimerization with another ligand-binding ErbB receptor. In contrast, amplification of c-erbB2 in tumors results in dramatic overexpression and constitutive activation of the receptor. Breast cancer cells overexpressing ErbB2 depend on its activity for proliferation, because treatment of these cells with ErbB2-specific antagonistic antibodies or kinase inhibitors blocks tumor cells in the G1 phase of the cell cycle. Intriguingly, loss of ErbB2 signaling is accompanied by a decrease in the phosphotyrosine content of ErbB3. On the basis of these results, it has been proposed that ErbB3 might be a partner for ErbB2 in promoting cellular transformation. To test this hypothesis and directly examine the role of the "kinase dead" ErbB3, we specifically ablated its expression with a designer transcription factor (E3). By infection of ErbB2-overexpressing breast cancer cells with a retrovirus expressing E3, we show that ErbB3 is an essential partner in the transformation process. Loss of functional ErbB2 or ErbB3 has similar effects on cell proliferation and cell cycle regulators. Furthermore, expression of constitutively active protein kinase B rescues the proliferative block induced as a consequence of loss of ErbB2 or ErbB3 signaling. These results demonstrate that ErbB2 overexpression and activity alone are insufficient to promote breast tumor cell division. Furthermore, we identify ErbB3's role, which is to couple active ErbB2 to the phosphatidylinositol 3-kinase/protein kinase B pathway. Thus, the ErbB2/ErbB3 dimer functions as an oncogenic unit to drive breast tumor cell proliferation.
Collapse
Affiliation(s)
- Thomas Holbro
- Friedrich Miescher Institute, P.O. Box 2543, 4002 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
181
|
Rudolph P, Kühling H, Alm P, Fernö M, Baldetorp B, Olsson H, Parwaresch R. Differential prognostic impact of the cyclins E and B in premenopausal and postmenopausal women with lymph node-negative breast cancer. Int J Cancer 2003; 105:674-80. [PMID: 12740917 DOI: 10.1002/ijc.11132] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Searching for new prognostic factors, we investigated the influence of cyclin expression on breast cancer prognosis. A total of 273 archival tumor specimens from patients with pT1/pT2 N0 breast cancers treated by surgery and local irradiation were immunostained for cyclins E, A and B. Outcome was evaluated as metastasis-free (MFS) and disease-specific survival (DSS) over a median observation period of 99 months. In postmenopausal women, DSS was significantly predicted by cyclin E, and in premenopausal patients by cyclin B. No statistical significance was found for cyclin A. When the prognostic impact of cyclins was compared to that of standard prognostic indicators in a multivariate analysis, both cyclin E and cyclin B were selected as independent predictors of survival in postmenopausal and premenopausal patients, respectively. After inclusion of Ki-67 in the model, cyclin E lost its significance, whereas cyclin B remained the only independent prognostic factor with a hazard ratio of 4.5 (p = 0.026) for tumor-related death. Assessment of cyclin expression may, therefore, refine current prognostic models if considered in relation to menopausal status. The prognostic relevance of cyclins is likely attributable to an influence on proliferation, cell survival and genetic instability. Awareness of the molecular mechanisms leading to deregulated cyclin expression may guide decisions for risk-adapted therapy regimens.
Collapse
Affiliation(s)
- Pierre Rudolph
- Department of Pathology, University of Kiel, Michaelisstrasse 11, 24105 Kiel, Germany.
| | | | | | | | | | | | | |
Collapse
|
182
|
Le XF, Claret FX, Lammayot A, Tian L, Deshpande D, LaPushin R, Tari AM, Bast RC. The role of cyclin-dependent kinase inhibitor p27Kip1 in anti-HER2 antibody-induced G1 cell cycle arrest and tumor growth inhibition. J Biol Chem 2003; 278:23441-50. [PMID: 12700233 DOI: 10.1074/jbc.m300848200] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclin-dependent kinase (CDK) inhibitor p27Kip1 binds to the cyclin E.CDK2 complex and plays a major role in controlling cell cycle and cell growth. Our group and others have reported that anti-HER2 monoclonal antibodies exert inhibitory effects on HER2-overexpressing breast cancers through G1 cell cycle arrest associated with induction of p27Kip1 and reduction of CDK2. The role of p27Kip1 in anti-HER2 antibody-induced cell cycle arrest and growth inhibition is, however, still uncertain. Here we have provided several lines of evidence supporting a critical role for p27Kip1 in the anti-HER2 antibody-induced G1 cell cycle arrest and tumor growth inhibition. Induction of p27Kip1 and G1 growth arrest by anti-HER2 antibody, murine 4D5, or humanized trastuzumab (Herceptin) are concentration-dependent, time-dependent, irreversible, and long-lasting. The magnitude of G1 cell cycle arrest induced by trastuzumab or 4D5 is well correlated with the level of p27Kip1 protein induced. Up-regulation of p27Kip1 and G1 growth arrest could no longer be removed with as little as 14 h of treatment with trastuzumab. Anti-HER2 antibody-induced p27Kip1 protein, G1 arrest, and growth inhibition persist at least 5 days after a single treatment. The magnitude of growth inhibition of breast cancer cells induced by anti-HER2 antibody closely parallels the level of p27Kip1 induced. Induced expression of exogenous p27Kip1 results in a p27Kip1 level-dependent G1 cell cycle arrest and growth inhibition similar to that obtained with anti-HER2 antibodies. Reducing p27Kip1 expression using p27Kip1 small interfering RNA blocks anti-HER2 antibody-induced p27Kip1 up-regulation and G1 arrest. Treatment with anti-HER2 antibody significantly increases the half-life of p27Kip1 protein. Inhibition of ubiquitin-proteasome pathway, but not inhibition of calpain and caspase activities, up-regulates p27Kip1 protein to a degree comparable with that obtained with anti-HER2 antibodies. We have further demonstrated that anti-HER2 antibody significantly decreases threonine phosphorylation of p27Kip1 protein at position 187 (Thr-187) and increases serine phosphorylation of p27Kip1 protein at position 10 (Ser-10). Expression of S10A and T187A mutant p27Kip1 protein increases the fraction of cells in G1 and reduces a further antibody-induced G1 arrest. Consequently, p27Kip1 plays an important role in the anti-HER2 antibody-induced G1 cell cycle arrest and tumor growth inhibition through post-translational regulation. Regulation of the phosphorylation of p27Kip1 protein is one of the post-translational mechanisms by which anti-HER2 antibody upregulates the protein.
Collapse
Affiliation(s)
- Xiao-Feng Le
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, Rosen N, Tsurutani J, Dennis PA, Mills GB, Arteaga CL. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 2003; 22:2812-22. [PMID: 12743604 DOI: 10.1038/sj.onc.1206388] [Citation(s) in RCA: 336] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We have examined the possible mechanisms of resistance to the epidermal growth factor receptor (EGFR) inhibitors in tumor cells with variable levels of EGFR. ZD1839 (Iressa) is a small-molecular-weight, ATP-mimetic that specifically inhibits the EGFR tyrosine kinase. A431 cell growth was markedly inhibited by ZD1839 (IC(50)< or =0.1 microM) whereas the MDA-468 cells were relatively resistant (IC(50)2 microM). Low doses of ZD1839 delayed cell cycle progression and induced apoptosis in A431 cells but not in MDA-468 cells. In both cell lines, 0.1 microM ZD1839 eliminated EGFR phosphorylation. However, the basal activity of the phosphatidylinositol-3 kinase (PI3 K) target Akt was eliminated in A431 but not in MDA-468 cells, implying that their Akt activity is independent of EGFR signals. A431 cells express PTEN/MMAC1/TEP, a phosphatase that can dephosphorylate position D3 of phosphatidylinositol-3,4,5 trisphosphate, the site that recruits the plecstrin-homology domain of Akt to the cell membrane. On the contrary, MDA-468 cells lack the phosphatase and tensin homolog (PTEN), potentially setting Akt activity at a high threshold that is unresponsive to EGFR inhibition alone. Therefore, we reintroduced (PTEN) by retroviral infection in MDA-468 cells. In MDA-468/PTEN but not in vector controls, treatment with ZD1839 inhibited P-Akt levels, induced relocalization of the Forkhead factor FKHRL1 to the cell nucleus, and increased FKHRL1-dependent transcriptional activity. ZD1839 induced a greater degree of apoptosis and cell cycle delay in PTEN-reconstituted than in control cells. These data suggest that loss of PTEN, by permitting a high level of Akt activity independent of receptor tyrosine kinase inputs, can temporally dissociate the inhibition of the EGFR with that of Akt induced by EGFR inhibitors. Thus, in EGFR-expressing tumor cells with concomitant amplification(s) of PI3K-Akt signaling, combined blockade of the EGFR tyrosine kinase and Akt should be considered as a therapeutic approach.
Collapse
Affiliation(s)
- Roberto Bianco
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Dakappagari NK, Pyles J, Parihar R, Carson WE, Young DC, Kaumaya PTP. A chimeric multi-human epidermal growth factor receptor-2 B cell epitope peptide vaccine mediates superior antitumor responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4242-53. [PMID: 12682258 DOI: 10.4049/jimmunol.170.8.4242] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunotherapeutic approaches to cancer should focus on novel undertakings that modulate immune responses by synergistic enhancement of antitumor immunological parameters. Cancer vaccines should preferably be composed of multiple defined tumor Ag-specific B and T cell epitopes. To develop a multiepitope vaccine, 12 high ranking B cell epitopes were identified from the extracellular domain of the human epidermal growth factor receptor-2 (HER-2) oncoprotein by computer-aided analysis. Four novel HER-2 B cell epitopes were synthesized as chimeras with a promiscuous T cell epitope (aa 288-302) from the measles virus fusion protein (MVF). Two chimeric peptide vaccines, MVF HER-2(316-339) and MVF HER-2(485-503) induced high levels of Abs in outbred rabbits, which inhibited tumor cell growth. In addition, Abs induced by a combination of two vaccines, MVF HER-2(316-339) and MVF HER-2(628-647) down-modulated receptor expression and activated IFN-gamma release better than the individual vaccines. Furthermore, this multiepitope vaccine in combination with IL-12 caused a significant reduction (p = 0.004) in the number of pulmonary metastases induced by challenge with syngeneic tumor cells overexpressing HER-2. Peptide Abs targeting specific sites in the extracellular domain may be used for exploring the oncoprotein's functions. The multiepitope vaccine may have potential application in the treatment of HER-2-associated cancers.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/metabolism
- Antibodies, Neoplasm/pharmacology
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Cancer Vaccines/chemical synthesis
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Cross Reactions
- Epitopes, B-Lymphocyte/biosynthesis
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/physiology
- Growth Inhibitors/chemical synthesis
- Growth Inhibitors/immunology
- Growth Inhibitors/pharmacology
- Humans
- Interleukin-12/immunology
- Interleukin-12/pharmacology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Measles virus/genetics
- Measles virus/immunology
- Mice
- Mice, Inbred ICR
- Molecular Sequence Data
- Peptide Fragments/chemical synthesis
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/physiology
- Protein Structure, Secondary/genetics
- Rabbits
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/physiology
- Recombinant Fusion Proteins/chemical synthesis
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/physiology
- Tumor Cells, Cultured
- Vaccines, Combined/chemical synthesis
- Vaccines, Combined/genetics
- Vaccines, Combined/immunology
- Vaccines, Combined/pharmacology
Collapse
Affiliation(s)
- Naveen K Dakappagari
- Department of Obstetrics and Gynecology, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
185
|
Di Gennaro E, Barbarino M, Bruzzese F, De Lorenzo S, Caraglia M, Abbruzzese A, Avallone A, Comella P, Caponigro F, Pepe S, Budillon A. Critical role of both p27KIP1 and p21CIP1/WAF1 in the antiproliferative effect of ZD1839 ('Iressa'), an epidermal growth factor receptor tyrosine kinase inhibitor, in head and neck squamous carcinoma cells. J Cell Physiol 2003; 195:139-50. [PMID: 12599217 DOI: 10.1002/jcp.10239] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
High expression of the epidermal growth factor receptor (EGFR) has been implicated in the development of squamous-cell carcinomas of head and neck (SCCHN). ZD1839 ('Iressa') is an orally active, selective EGFR-TKI (EGFR-tyrosine kinase inhibitor) that blocks signal transduction pathways implicated in proliferation and survival of cancer cells, and other host-dependent processes promoting cancer growth. We have demonstrated that ZD1839 induces growth arrest in SCCHN cell lines by inhibiting EGFR-mediated signaling. Cell cycle kinetic analysis demonstrated that ZD1839 induces a delay in cell cycle progression and a G1 arrest together with a partial G2/M block; this was associated with increased expression of both p27(KIP1) and p21(CIP1/WAF1) cyclin-dependent kinase (CDK) inhibitors. The activity of CDK2, the main target of CIP/KIP CDK inhibitors, was reduced in a dose-dependent fashion after 24 h of ZD1839 treatment and this effect correlated to the increased amount of p27(KIP1) and p21(CIP1/WAF1) proteins associated with CDK2-cyclin-E and CDK2-cyclin-A complexes. In addition, ZD1839-induced growth inhibition was significantly reduced in cell transfectants expressing p27(KIP1) or p21(CIP1/WAF1) antisense constructs. Overall, these results as well as the timing of the effect of ZD1839 on G1 arrest and p27(KIP1) and p21(CIP1/WAF1) upregulation, suggest a mechanistic connection between these events.
Collapse
Affiliation(s)
- Elena Di Gennaro
- Dipartimento di Oncologia Sperimentale, Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Spataro VJ, Litman H, Viale G, Maffini F, Masullo M, Golouh R, Martinez-Tello FJ, Grigolato P, Shilkin KB, Gusterson BA, Castiglione-Gertsch M, Price K, Lindtner J, Cortés-Funes H, Simoncini E, Byrne MJ, Collins J, Gelber RD, Coates AS, Goldhirsch A. Decreased immunoreactivity for p27 protein in patients with early-stage breast carcinoma is correlated with HER-2/neu overexpression and with benefit from one course of perioperative chemotherapy in patients with negative lymph node status: results from International Breast Cancer Study Group Trial V. Cancer 2003; 97:1591-600. [PMID: 12655514 DOI: 10.1002/cncr.11224] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The objective of this study was to clarify the prognostic and predictive value of immunoreactivity for the cyclin-dependent kinase inhibitor p27(Kip1) in patients with early-stage breast carcinoma and to investigate its relation with clinicopathologic features and other markers. METHODS Immunoreactivity for p27 protein was analyzed on tumor slides from 461 patients who were enrolled in the International Breast Cancer Study Group (IBCSG) Trial V (median follow-up, 13 years), including 198 patients with lymph node negative disease and 263 patients with lymph node positive disease. Tumors with < 50% immunoreactive neoplastic cells were considered low expressors. Immunoreactivity for p27 was correlated with several clinicopathologic characteristics. Disease free survival (DFS) and overall survival were analyzed according to p27 immunoreactivity and treatment group. RESULTS In the lymph node negative population, decreased p27 immunoreactivity was associated with higher tumor grade (P = 0.001) and HER-2/neu overexpression (P = 0.04). In the lymph node positive population, low p27 expression was associated with higher tumor grade (P = 0.01), low expression of thymidylate synthase (P = 0.001), and higher Ki-67 expression (P = 0.007). DFS was not significantly different according to p27 status in either lymph node negative patients (10-year DFS: low p27 expression, 53% +/- 5%; high p27 expression, 55% +/- 5%) or in lymph node positive patients (10 year DFS: low p27 expression, 33% +/- 4%; high p27 expression, 32% +/- 4%). However, in the lymph node negative population, the benefit of one course of perioperative chemotherapy with cyclophosphamide, methotrexate, and 5-fluorouracil was confined exclusively to patients with tumors that showed reduced p27 immunoreactivity (P = 0.03; test for interaction). CONCLUSIONS This analysis indicates that p27 immunoreactivity has little if any prognostic value in patients with early-stage breast carcinoma. However, these findings suggest that, in patients with breast carcinoma who have negative lymph node status, reduced p27 immunoreactivity is associated with HER-2/neu overexpression and may be predictive of a benefit from the early administration of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Vito J Spataro
- Department of Medical Oncology, Ospedale San Giovanni, Bellinzona, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Shimizu H, Seiki T, Asada M, Yoshimatsu K, Koyama N. Alpha6beta1 integrin induces proteasome-mediated cleavage of erbB2 in breast cancer cells. Oncogene 2003; 22:831-9. [PMID: 12584562 DOI: 10.1038/sj.onc.1206203] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ErbB2 and alpha6 integrin have been implicated in malignancy of breast cancer cells. Here we have determined the influence of alpha6beta1 integrin on erbB2 signaling in anchorage-independent growth, using MDA-MB435 breast cancer cells. Firstly, we transfected the cells with erbB2 cDNA, and isolated cells with high or low levels of alpha6beta1 integrin by cell sorting (alpha6H-ErbB and alpha6L-ErbB). We found that an erbB ligand, heregulin beta1, enhanced growth activity of alpha6L-ErbB cells, but not alpha6H-ErbB cells. Secondly, we established cells expressing a beta4 integrin deletion mutant (beta4-deltacyt), which selectively inhibited alpha6beta1 integrin expression and adhesion to laminin-1. Again, heregulin beta1 enhanced the growth of erbB2 cDNA-transfected beta4-deltacyt cells, but not mock cells. Western blot analysis revealed that heregulin beta1 stimulated phosphorylation of Akt and its downstream molecules, GSK3beta and p70S6kinase, and that the extent of phosphorylation was greater in ErbB2/beta4-deltacyt cells than ErbB2/mock cells. Furthermore, we found that the erbB2 cytoplasmic domain was truncated in ErbB2/mock cells, which was independent of ligand stimulation and adhesion, and was suppressed by proteasome inhibitors. These results suggest that alpha6beta1 integrin inhibits erbB2 signals by inducing proteasome-dependent proteolytic cleavage of the erbB2 cytoplasmic domain, and may thereby contribute to the regulation of tumor growth.
Collapse
Affiliation(s)
- Hajime Shimizu
- Tsukuba Research Laboratories, Eisai Co, Ltd, Ibaraki, Japan.
| | | | | | | | | |
Collapse
|
188
|
Civenni G, Holbro T, Hynes NE. Wnt1 and Wnt5a induce cyclin D1 expression through ErbB1 transactivation in HC11 mammary epithelial cells. EMBO Rep 2003; 4:166-71. [PMID: 12612606 PMCID: PMC1315833 DOI: 10.1038/sj.embor.embor735] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2002] [Revised: 10/17/2002] [Accepted: 12/02/2002] [Indexed: 12/16/2022] Open
Abstract
Constitutive expression of Wnt1 and Wnt5a in HC11 mammary cells led to elevated TCF transcriptional activity. Intriguingly, Wnt-expressing cells also displayed activation of ErbB1 and mitogen-activated protein kinase (MAPK), in contrast to control HC11 cells, which did not. Furthermore, conditioned media harvested from Wnt-expressing cells stimulated ErbB1 and the MAPK cascade when added to control cells. This process was rapid and could be blocked by an ErbB1 antibody that interferes with ligand binding and by matrix metalloproteinase (MMP) inhibitors. These results suggest that in mammary cells Wnt binding to its receptor, Frizzled (Fz), transactivates ErbB1, probably by MMP-mediated release of soluble ErbB1 ligands. Importantly, Wnt-transactivated ErbB1 was responsible for MAPK activation and the increased levels of cyclin D1 present in the Wnt-expressing HC11 cells. Our finding that Wnts transactivate ErbB1 in addition to stimulating the prototypic beta-catenin/TCF pathway may help to explain why wnt1 is a potent oncogene in the mammary gland.
Collapse
Affiliation(s)
- Gianluca Civenni
- Friedrich Miescher Institute, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Thomas Holbro
- Friedrich Miescher Institute, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | - Nancy E. Hynes
- Friedrich Miescher Institute, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
- Tel: +41 61 697 8107; Fax: +41 61 697 8102;
| |
Collapse
|
189
|
Banerjee S, Saxena N, Sengupta K, Tawfik O, Mayo MS, Banerjee SK. WISP-2 gene in human breast cancer: estrogen and progesterone inducible expression and regulation of tumor cell proliferation. Neoplasia 2003; 5:63-73. [PMID: 12659671 PMCID: PMC1502127 DOI: 10.1016/s1476-5586(03)80018-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
WISP-2 mRNA and protein was overexpressed in preneoplastic and cancerous cells of human breast. Statistical analyses show a significant association between WISP-2 expression and estrogen receptor (ER) positivity. In normal breast, the expression was virtually undetected. The studies showed that WISP-2 is an estrogen-induced early response gene in MCF-7 cells and the expression was continuously increased to reach a maximum level at 24 h. The estrogen effect was inhibited by a pure antiestrogen (ICI 182,780). Human mammary epithelial cells, in which WISP-2 expression was undetected or minimally detected, responded to 17beta-estradiol by upregulating the WISP-2 gene after transfection with ER-alpha, providing further evidences that WISP-2 expression is mediated through ER-alpha. Overexpression of WISP-2 mRNA by estrogen may be accomplished by both transcriptional activation and stabilization. MCF-7 cells exposed to progesterone had a rapid but transient increase in WISP-2 expression, and PR antagonist RU38486 blocked this mRNA induction. In combination with estradiol, progesterone acted as an antagonist inhibiting the expression of WISP-2 mRNA. Moreover, disruption of WISP-2 signaling in MCF-7 cells by use of antisense oligomers caused a significant reduction in tumor cell proliferation. The results are consistent with the conclusion that WISP-2 expression is a requirement for breast tumor cells proliferation.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Blotting, Northern
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- CCN Intercellular Signaling Proteins
- Case-Control Studies
- Cell Division/drug effects
- Dactinomycin/pharmacology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Estrogens/pharmacology
- Female
- Fulvestrant
- Gene Expression Regulation, Neoplastic/drug effects
- Hormone Antagonists/pharmacology
- Humans
- Immunoenzyme Techniques
- In Situ Hybridization
- Intercellular Signaling Peptides and Proteins
- Male
- Middle Aged
- Mifepristone/pharmacology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Oligodeoxyribonucleotides, Antisense/pharmacology
- Polymerase Chain Reaction
- Progesterone/pharmacology
- Protein Synthesis Inhibitors/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Repressor Proteins
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Snigdha Banerjee
- Cancer Research Unit, Research Division, V.A. Medical Center, Kansas City, MO 64128, USA.
| | | | | | | | | | | |
Collapse
|
190
|
Arteaga CL. Trastuzumab, an appropriate first-line single-agent therapy for HER2-overexpressing metastatic breast cancer. Breast Cancer Res 2003; 5:96-100. [PMID: 12631388 PMCID: PMC154152 DOI: 10.1186/bcr574] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2002] [Revised: 01/02/2003] [Accepted: 01/16/2002] [Indexed: 12/18/2022] Open
Abstract
Overexpression of the HER2/Neu (ErbB2) proto-oncogene is associated with breast cancer progression and poor patient prognosis. Herceptin (trastuzumab) is a humanized IgG1 against the ectodomain of the HER2 receptor. In combination with chemotherapy, it induces regression of HER2-overexpressing metastatic breast tumors and prolongs patient survival. Single-agent Herceptin in patients with HER2-amplified breast tumors also induces a definite objective response and clinical benefit rates, and is well tolerated. These data suggest that Herceptin is an effective first-line single-agent therapy for a predictable cohort of metastatic breast cancers and can therefore be used as a platform for therapeutic discovery against tumors that overexpress HER2.
Collapse
Affiliation(s)
- Carlos L Arteaga
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
191
|
Glöckner S, Buurman H, Kleeberger W, Lehmann U, Kreipe H. Marked intratumoral heterogeneity of c-myc and cyclinD1 but not of c-erbB2 amplification in breast cancer. J Transl Med 2002; 82:1419-26. [PMID: 12379776 DOI: 10.1097/01.lab.0000032371.16521.40] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Intratumoral heterogeneity mirrors subclonal diversity and might affect treatment response. To investigate molecular heterogeneity of primary breast cancer specimens, we determined the amplification status of growth regulatory genes (c-erbB2, topoisomerase IIalpha, c-myc, and cyclinD1) in macroscopically and microscopically separate areas of individual tumors (n = 21). Using laser-assisted microdissection and quantitative PCR, we found marked intratumoral heterogeneity with different patterns for each gene. Molecular heterogeneity in amplification pattern could be demonstrated between both macroscopically (0.5 to several centimeters) and microscopically (10 to several hundred micrometers) distant tumor areas. C-erbB2 amplification proved to be the most stable amplification in individual tumors, with heterogeneity occurring in only 36% of amplified cases. By contrast, amplification of c-myc and cyclinD1 revealed varying patterns in the vast majority of amplified cases (100% and 83%). The constancy of c-erbB2 amplification underlines its presumed importance in breast cancer biology. We conclude that the molecular heterogeneity of breast cancer as evidenced in this study requires thorough and representative sampling of different tumor areas when the biologic significance of somatic mutations is considered. Patterns of heterogeneity can be used to trace the clonal evolution within different compartments of an individual tumor.
Collapse
Affiliation(s)
- Sabine Glöckner
- Institute of PathologyMedizinische Hochschule Hannover, Hannover, Germany
| | | | | | | | | |
Collapse
|
192
|
Abstract
Therapy directed against specific biologic targets has long been used in the treatment of breast cancer; the estrogen receptor is a validated prognostic and therapeutic target, and antiestrogen therapy has been used effectively for decades. Recently, scientific progress and increased comprehension of mechanisms of breast cancer pathogenesis have led to the proliferation of both potential molecular targets and new therapeutic agents. The success of traztuzumab (Herceptin, Genentech, South San Francisco, CA), an anti-HER2 antibody, has spurred the development of other biologically directed therapeutics. In this overview, I discuss three targets relevant to breast cancer (the epidermal growth factor receptor family, angiogenesis, and NF-kappa B), and therapeutic approaches directed against these targets are discussed.
Collapse
Affiliation(s)
- Carolyn I Sartor
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA.
| |
Collapse
|
193
|
Timms JF, White SL, O'Hare MJ, Waterfield MD. Effects of ErbB-2 overexpression on mitogenic signalling and cell cycle progression in human breast luminal epithelial cells. Oncogene 2002; 21:6573-86. [PMID: 12242655 DOI: 10.1038/sj.onc.1205847] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2002] [Revised: 06/12/2002] [Accepted: 07/05/2002] [Indexed: 01/23/2023]
Abstract
Most breast cancers arise from luminal epithelial cells and 25-30% of these tumours overexpress the ErbB-2 receptor. Herein, a non-transformed, immortalized cell system was used to investigate the effects of ErbB-2 overexpression in luminal epithelial cells. The phenotypic consequence of ErbB-2 overexpression is a shortening of the G1 phase of the cell cycle and early S phase entry, which leads to hyperproliferation. We show that this effect was mediated through the up-regulation of cdk6 and cyclins D1 and E, and enhanced degradation and relocalization of p27(Kip1). These changes were effected predominantly through enhanced MAPK signalling, resulting in cdk2 hyperactivation. PI3K signalling also participated in cell cycle progression, since PI3K and MAPK coordinately regulated changes in cyclin D1 and cdk6 expression. Cdk4 activity was not required for cell cycle progression in these cells, and was constitutively inhibited through its association with p16(INK4A). MAPK-dependent induction of p21(Cip1) was also necessary for G1 phase progression, although its degradation by the proteasome was required for S phase entry. These data provide new insights into the complex molecular mechanisms underlying mitogenic cell cycle control in luminal epithelial cells, the cell type relevant to primary breast cancer, and show how ErbB-2 overexpression subverts this normal control.
Collapse
Affiliation(s)
- John F Timms
- Ludwig Institute for Cancer Research, 91 Riding House Street, London W1W 7BS, UK.
| | | | | | | |
Collapse
|
194
|
Shin I, Yakes FM, Rojo F, Shin NY, Bakin AV, Baselga J, Arteaga CL. PKB/Akt mediates cell-cycle progression by phosphorylation of p27(Kip1) at threonine 157 and modulation of its cellular localization. Nat Med 2002; 8:1145-52. [PMID: 12244301 DOI: 10.1038/nm759] [Citation(s) in RCA: 599] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2002] [Accepted: 08/14/2002] [Indexed: 12/17/2022]
Abstract
We have shown a novel mechanism of Akt-mediated regulation of the CDK inhibitor p27(kip1). Blockade of HER2/neu in tumor cells inhibits Akt kinase activity and upregulates nuclear levels of the CDK inhibitor (Kip1). Recombinant Akt and Akt precipitated from tumor cells phosphorylated wild-type p27 in vitro. p27 contains an Akt consensus RXRXXT(157)D within its nuclear localization motif. Active (myristoylated) Akt phosphorylated wild-type p27 in vivo but was unable to phosphorylate a T157A-p27 mutant. Wild-type p27 localized in the cytosol and nucleus, whereas T157A-p27 localized exclusively in the nucleus and was resistant to nuclear exclusion by Akt. T157A-p27 was more effective than wild-type p27 in inhibiting cyclin E/CDK2 activity and cell proliferation; these effects were not rescued by active Akt. Expression of Ser(473) phospho Akt in primary human breast cancers statistically correlated with expression of p27 in tumor cytosol. These data indicate that Akt may contribute to tumor-cell proliferation by phosphorylation and cytosolic retention of p27, thus relieving CDK2 from p27-induced inhibition.
Collapse
Affiliation(s)
- Incheol Shin
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | |
Collapse
|
195
|
Xia W, Mullin RJ, Keith BR, Liu LH, Ma H, Rusnak DW, Owens G, Alligood KJ, Spector NL. Anti-tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene 2002; 21:6255-63. [PMID: 12214266 DOI: 10.1038/sj.onc.1205794] [Citation(s) in RCA: 494] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2002] [Revised: 06/06/2002] [Accepted: 06/18/2002] [Indexed: 02/06/2023]
Abstract
Dual EGFR/erbB2 inhibition is an attractive therapeutic strategy for epithelial tumors, as ligand-induced erbB2/EGFR heterodimerization triggers potent proliferative and survival signals. Here we show that a small molecule, GW572016, potently inhibits both EGFR and erbB2 tyrosine kinases leading to growth arrest and/or apoptosis in EGFR and erbB2-dependent tumor cell lines. GW572016 markedly reduced tyrosine phosphorylation of EGFR and erbB2, and inhibited activation of Erk1/2 and AKT, downstream effectors of proliferation and cell survival, respectively. Complete inhibition of activated AKT in erbB2 overexpressing cells correlated with a 23-fold increase in apoptosis compared with vehicle controls. EGF, often elevated in cancer patients, did not reverse the inhibitory effects of GW572016. These observations were reproduced in vivo, where GW572016 treatment inhibited activation of EGFR, erbB2, Erk1/2 and AKT in human tumor xenografts. Erk1/2 and AKT represent potential biomarkers to assess the clinical activity of GW572016. Inhibition of activated AKT in EGFR or erbB2-dependent tumors by GW572016 may lead to tumor regressions when used as a monotherapy, or may enhance the anti-tumor activity of chemotherapeutics, since constitutive activation of AKT has been linked to chemo-resistance.
Collapse
Affiliation(s)
- Wenle Xia
- Department of Discovery Medicine, GlaxoSmithKline, Five Moore Drive, Research Triangle Park, North Carolina, NC 27709-3398, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Fujita N, Sato S, Katayama K, Tsuruo T. Akt-dependent phosphorylation of p27Kip1 promotes binding to 14-3-3 and cytoplasmic localization. J Biol Chem 2002; 277:28706-13. [PMID: 12042314 DOI: 10.1074/jbc.m203668200] [Citation(s) in RCA: 254] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In many human cancers, the cyclin-dependent kinase inhibitor p27(Kip1) is expressed at low or undetectable levels. The decreased p27(Kip1) expression allows cyclin-dependent kinase activity to cause cells to enter into S phase and correlates with poor patient survival. Inhibition of serine/threonine kinase Akt signaling by some pharmacological agents or by PTEN induces G(1) arrest, in part by up-regulating p27(Kip1). However, the role of Akt-dependent phosphorylation in p27(Kip1) regulation is not clear. Here, we show that Akt bound directly to and phosphorylated p27(Kip1). Screening p27(Kip1) phosphorylation sites identified the COOH-terminal Thr(198) residue as a novel site. Further analysis revealed that 14-3-3 proteins bound to p27(Kip1) through Thr(198) only when it was phosphorylated by Akt. Although Akt also phosphorylated p27(Kip1) at Ser(10) and Thr(187), these two sites were not involved in the binding to 14-3-3 proteins. p27(Kip1) phosphorylated at Thr(198) exists only in the cytoplasm. Therefore, Akt promotes cell-cycle progression through the mechanisms of phosphorylation-dependent 14-3-3 binding to p27(Kip1) and cytoplasmic localization.
Collapse
Affiliation(s)
- Naoya Fujita
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo 113-0032, Japan
| | | | | | | |
Collapse
|
197
|
Clarke R, Howell A, Anderson E. Control of Proliferation in the Normal and Neoplastic Breast. Breast Cancer 2002. [DOI: 10.1201/b14039-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
198
|
Neve RM, Holbro T, Hynes NE. Distinct roles for phosphoinositide 3-kinase, mitogen-activated protein kinase and p38 MAPK in mediating cell cycle progression of breast cancer cells. Oncogene 2002; 21:4567-76. [PMID: 12085235 DOI: 10.1038/sj.onc.1205555] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2001] [Revised: 03/20/2002] [Accepted: 03/27/2002] [Indexed: 12/18/2022]
Abstract
Addition of the ErbB-ligand, Heregulinbeta1 (HRG), to breast tumour-derived T47D cells promotes D-cyclin expression, p21(cip1) synthesis, cyclin-dependent kinase (CDK) activation through re-distribution of p27(kip1) and DNA synthesis. In contrast EGF has no effect on T47D cell cycle progression. By comparing these two ligands and the use of specific inhibitors for phosphatidylinositol-3 kinase (PI3K), mitogen-activated protein kinase (MAPK) and p38MAPK, we have identified several molecular mechanisms required for ErbB receptor-mediated proliferation. The PI3K, MAPK and p38MAPK pathways each displayed distinct activation profiles in response to either HRG or EGF, with obvious differences in both the intensity and duration of signal output. Through inhibition of each of these pathways it is apparent that each pathway is necessary, yet insufficient alone, to stimulate proliferation. Each pathway regulates distinct subsets of essential cell cycle regulators and integration of these signal networks is required for the timely expression of these components, which culminates in cell cycle progression. Significantly, the mechanisms controlling ligand-stimulated proliferation through ErbB2 are strikingly similar to the mechanisms through which overexpressed, constitutively activated, ErbB2 orchestrates uncontrolled proliferation in cancer cells. This suggests that downstream effectors of ErbB receptors represent good therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Richard M Neve
- Department of Hematology/Oncology, UCSF San Francisco and Buck Institute, Novato, California 94945, USA.
| | | | | |
Collapse
|
199
|
Fantin VR, Berardi MJ, Scorrano L, Korsmeyer SJ, Leder P. A novel mitochondriotoxic small molecule that selectively inhibits tumor cell growth. Cancer Cell 2002; 2:29-42. [PMID: 12150823 DOI: 10.1016/s1535-6108(02)00082-x] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tumorigenesis results from events that impinge on a variety of collaborating metabolic pathways. To assess their role in this process, we utilized a cell-based assay to perform a high-throughput, chemical library screen. In so doing, we identified F16, a small molecule that selectively inhibits proliferation of mammary epithelial, neu-overexpressing cells, as well as a variety of mouse mammary tumor and human breast cancer cell lines. F16 belongs to a group of structurally similar molecules with a delocalized positive charge. The compound is accumulated in mitochondria of responsive cells, driven by the membrane potential, and it compromises their functional integrity. Mitochondrial hyperpolarization is a shared feature of many tumor cell lines, explaining the broad action spectrum of this novel delocalized lipophilic cation.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cations/pharmacology
- Cations/toxicity
- Cell Cycle/drug effects
- Cell Division/drug effects
- Cell Line, Transformed
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Genes, erbB-2/genetics
- Growth Inhibitors/chemistry
- Growth Inhibitors/pharmacology
- Growth Inhibitors/toxicity
- Humans
- Indoles/metabolism
- Indoles/pharmacology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Experimental/metabolism
- Membrane Potentials
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- Pyridinium Compounds/metabolism
- Pyridinium Compounds/pharmacology
- Receptor, ErbB-2/metabolism
- Signal Transduction/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Valeria R Fantin
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
200
|
Price-Schiavi SA, Jepson S, Li P, Arango M, Rudland PS, Yee L, Carraway KL. Rat Muc4 (sialomucin complex) reduces binding of anti-ErbB2 antibodies to tumor cell surfaces, a potential mechanism for herceptin resistance. Int J Cancer 2002; 99:783-91. [PMID: 12115478 DOI: 10.1002/ijc.10410] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Muc4 (also called sialomucin complex), the rat homolog of human MUC4, is a heterodimeric glycoprotein complex that consists of a peripheral O-glycosylated mucin subunit, ASGP-1, tightly but noncovalently linked to a N-glycosylated transmembrane subunit, ASGP-2. The complex is expressed in a number of normal, vulnerable epithelial tissues, including mammary gland, uterus, colon, cornea and trachea. Muc4/SMC is also overexpressed or aberrantly expressed on a number of human tumors including breast tumors. Overexpression of Muc4/SMC has been shown to block cell-cell and cell-matrix interactions, protect tumor cells from immune surveillance and promote metastasis. In addition, as a ligand for ErbB2, Muc4/SMC can potentiate phosphorylation of ErbB2 and potentially alter signals generated from this receptor. Using A375 human melanoma cells and MCF7 human breast adenocarcinoma cells stably transfected with tetracycline regulatable Muc4, we have investigated whether overexpression of Muc4/SMC can repress antibody binding to cell surface-expressed ErbB2. Overexpression of Muc4/SMC does not affect the level of ErbB2 expression in either cell line, but it does reduce binding of a number of anti-ErbB2 antibodies, including Herceptin. Interestingly, overexpression of ErbB2 does not block binding of other unrelated antibodies of the same isotype, suggesting that the reduction in ErbB2 antibody binding is due to complex formation of Muc4/SMC and ErbB2. Furthermore, capping of Muc4/SMC with anti-Muc4/SMC antibodies reduces antibody binding to ErbB2 instead of increasing binding, again suggesting that reduced antibody binding to ErbB2 is due to steric hindrance from complex formation of Muc4/SMC and ErbB2. Thus, overexpression of Muc4/SMC on tumor cells may have both prognostic and therapeutic relevance.
Collapse
Affiliation(s)
- Shari A Price-Schiavi
- Department of Cell Biology and Anatomy, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | | | |
Collapse
|