Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Tumor Biology

Down-Regulation of the erbB-2 Receptor by Trastuzumab (Herceptin) Enhances Tumor Necrosis Factor-related Apoptosis-inducing Ligand-mediated Apoptosis in Breast and Ovarian Cancer Cell Lines that Overexpress erbB-2

Mauricio Cuello, Seth A. Ettenberg, Amy S. Clark, Maccon M. Keane, Reuben H. Posner, Marion M. Nau, Phillip A. Dennis and Stan Lipkowitz
Mauricio Cuello
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Seth A. Ettenberg
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Amy S. Clark
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maccon M. Keane
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Reuben H. Posner
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marion M. Nau
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Phillip A. Dennis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stan Lipkowitz
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published June 2001
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

We investigated whether combined treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and trastuzumab could enhance the specific killing of cells that overexpress the erbB-2 receptor. The combination resulted in an enhancement of TRAIL-mediated apoptosis in all cell lines overexpressing erbB-2 receptor compared with either reagent alone. In contrast, there was no effect in cell lines with low levels of the erb-B2 receptor. Trastuzumab treatment resulted in down-regulation of the erbB-2 receptor in all erbB-2-overexpressing cell lines. Similar enhancement of TRAIL toxicity was observed when the erbB-2 receptor was down-regulated using antisense oligodeoxynucleotides. Down-regulation of the erbB-2 receptor protein by trastuzumab or antisense oligodeoxynucleotides decreased Akt kinase activation but not mitogen-activated protein kinase activation. Down-regulation of Akt kinase activity by a phosphatidylinositol 3′-kinase inhibitor (LY294002) also resulted in enhancement of TRAIL-mediated apoptosis. Expression of a constitutively active form of Akt kinase in an erbB-2-overexpressing cell line completely abrogated the increase in TRAIL-mediated apoptosis by trastuzumab and significantly reduced the biological effect of either reagent alone. Therefore, down-regulation of the erbB-2 receptor by trastuzumab enhances TRAIL-mediated apoptosis by inhibiting Akt kinase activity. These data suggest that the combination of trastuzumab and TRAIL may allow enhanced therapeutic efficacy and specificity in the treatment of erbB-2-overexpressing tumors.

INTRODUCTION

Death receptors of the TNF 3 receptor family (e.g., TNF receptor and Fas receptor) induce apoptosis on binding to their specific ligands (e.g., TNF and Fas ligand, respectively) via activation of caspases (1, 2, 3) . These death receptor pathways offer an attractive method to induce apoptosis in cancer cells and may thereby serve as a treatment of cancer. This approach has been hampered due to lack of efficacy and prohibitive toxicity [e.g., TNF fails to induce apoptosis in most cancer cells (4) , and Fas induces lethal liver damage (5)] . TRAIL binds to the death receptors TRAIL-R1 (also called DR4) and TRAIL-R2 (also called DR5/TRICK-2/KILLER) and induces caspase-mediated apoptosis (6, 7, 8) . TRAIL has been reported to induce apoptosis selectively in a variety of cancer cell lines but not in normal cells (9, 10, 11, 12, 13, 14, 15, 16) . Also, animal studies have shown that TRAIL can induce regression of cancer xenografts without toxicity to normal tissues (11 , 17) . The selective induction of apoptosis in cancer cells but not in normal cells has prompted investigation into the use of TRAIL in cancer therapy.

However, not all cancer cells undergo apoptosis when treated with TRAIL. We have previously shown that a majority of breast cancer cells lines are resistant to TRAIL-mediated apoptosis (18) . Similar results have been reported in other cancer cell lines (12 , 19, 20, 21, 22) . Several mechanisms have been described that may control sensitivity to TRAIL-mediated apoptosis. These include the expression of decoy receptors (TRAIL-R3/DcR1 and TRAIL-R4/DcR2) that bind to TRAIL but do not activate the caspase cascade (6 , 7 , 10 , 15) ; the expression of inhibitory downstream molecules such as FLIPs (23 , 24) and IAPs (25) ; and the activation of antiapoptotic transcription factors such as nuclear factor κB (26, 27, 28, 29, 30) . Studies in resistant cancer cell lines have failed to identify the major determinants of TRAIL sensitivity (18 , 20) . Recently, we reported that the combination of chemotherapy and TRAIL enhances TRAIL-mediated apoptosis in breast cancer cell lines. However, this combination also resulted in increased death of normal breast epithelial cells (18) . Thus, it is important not only to overcome resistance to TRAIL but also to target therapy specifically to the cancer cells.

erbB-2 is a member of the epidermal growth factor receptor family and is overexpressed in breast (30%) and ovarian (15–30%) carcinomas (31, 32, 33, 34) . The overall survival rate and time to relapse for patients whose tumors overexpress erbB-2 are significantly shorter than those in patients whose tumors lack erbB-2 overexpression (35 , 36) . erbB-2 overexpression has been demonstrated to enhance proliferative, prosurvival, and metastatic signals in breast cancer cell lines (37, 38, 39) . For example, Akt kinase activity and MAPK activity are up-regulated in cells overexpressing erbB-2 (39, 40, 41) . The anti-erbB-2 antibody, trastuzumab (Herceptin), has clinical activity alone and in combination with chemotherapy in metastatic breast cancer (42, 43, 44) . The mechanisms that account for the biological effect of trastuzumab alone or in combination with chemotherapy are not known completely (45) . In vitro studies suggest that trastuzumab down-regulates the erbB-2 receptor and consequently inhibits downstream pathways involved in cell survival, cell proliferation, and metastasis (46) . As a consequence of erbB-2 down-regulation in breast cancer cell lines, inhibition of proliferation and enhancement of apoptosis have been described (40 , 42) .

In the present work, we investigate whether the combination of trastuzumab and TRAIL could enhance selective TRAIL-mediated apoptosis in cancer cells overexpressing the erbB-2 receptor.

MATERIALS AND METHODS

Cell Lines.

Three erbB-2-overexpressing cell lines (SKBr-3, MDA-MB-453, and SKOv-3) and three cell lines with low erbB-2 expression (MDA-MB-468, SW-626, and MDA-MB-231) were obtained from American Type Culture Collection (Manassas, VA) and grown in culture according to the instructions provided with them. The MCF-7 cell line (with normal erbB-2 expression) was a gift provided by Dr. Ed Chu (Yale Comprehensive Cancer Center, New Haven, CT) and was cultured in RPMI 1640 supplemented with 10% FCS and 1% penicillin/streptomycin. The low erbB-2 receptor-expressing cell line MDA-MB-231 has been demonstrated to be highly sensitive to TRAIL and was used as a positive control for TRAIL treatment (18) .

To generate stable clones, HA-tagged myristylated Akt kinase cDNA in PSRα (gift provided by Dr. Philip Tsichlis; Fox Chase Cancer Research Center, Philadelphia, PA) and the PSRα vector were introduced into the MDA-MB-453 cell line using LipofectAMINE transfection (Life Technologies, Inc., Gaithersburg, MD). Clones were isolated after selection in 800 μg/ml G418 antibiotic (Life Technologies, Inc.).

GST and GST-TRAIL Fusion Protein Production.

The GST and GST-TRAIL fusion protein have been described previously (18) . To produce GST and active GST-TRAIL, GST and GST-TRAIL cDNA plasmids were transformed into DH5α Escherichia coli, and protein expression was induced with 100 mm isopropylthio-β-d-galactosidase (Amersham Pharmacia Biotech Inc., Piscataway, NJ). Bacteria were harvested and lysed by sonication in 0.1% TONE buffer [20 mm Tris-HCl (pH 7.5), 100 mm NaCl, 1 mm EDTA, and 0.1% n-octyl-β-d-glucopyranoside). GST and GST-TRAIL proteins were purified by precipitation with glutathione-Sepharose beads (Amersham Pharmacia Biotech Inc.) and then eluted from the beads with 50 mm glutathione in 0.5% TONE buffer (pH 8.5). The buffer was exchanged for PBS using a PD10 Sephadex column (Amersham Pharmacia Biotech Inc.). The proteins were analyzed by fractionation on 10% SDS-PAGE and visualized with Chromaphor reagent (Promega, Madison, WI). The protein concentrations were measured using a Bio-Rad colorimetric assay (Bio-Rad, Hercules, CA). GST-TRAIL proteins were stable when stored bound to glutathione-Sepharose beads at 4°C for up to 3 months. All experiments were performed with freshly eluted GST-TRAIL proteins because TRAIL activity decreased rapidly on storage at 4°C after elution.

Trastuzumab and TRAIL-mediated Toxicity.

To assess TRAIL-mediated cytotoxicity on trastuzumab (Genentech, South San Francisco, CA) treatment, cells were plated at 2 × 104 cells/well in 96-well microtiter plates and allowed to adhere to the plates overnight. As an isotype-matched antibody, the humanized mouse IgG anti-CD20 rituximab (Genentech) was used. The adherent cells were incubated in the presence or absence of trastuzumab (or rituximab) as indicated in the figure legends for 96 h. Freshly eluted GST-TRAIL fusion protein was added for the last 16 h at the indicated concentrations. Cell viability was assessed by the MTS dye reduction assay (Cell Titer 96 AQueous One Solution Cell Proliferation Assay; Promega).

All MTS data points were repeated six times, and each experiment was carried out at least three times. Results of representative experiments are given as the mean ± SD, and results of multiple experiments are given as the mean ± SE.

Flow Cytometric Detection of Apoptosis.

To assess apoptosis, cells were plated at 5 × 105 cells/60-mm dish, allowed to adhere overnight, and then treated using the same conditions described in the MTS assay. The cells were trypsinized, washed with PBS, fixed in ice-cold methanol, and stored at −20°C overnight. Fixed cells were washed twice with PBS and incubated with DNase-free RNase (1 unit/ml; Roche Molecular Biochemicals, Indianapolis, IN) for 30 min at 37°C. After incubation, nuclei were stained with propidium iodide at 50 μg/ml (Roche Molecular Biochemicals). Stained cells were stored at 4°C and protected from light until flow cytometric analysis. Cells undergoing apoptosis were determined as a percentage of cells with sub-G0-G1 DNA content in the DNA histogram compared with the total number of cells present using the FACSort system (Becton Dickinson, Mansfield, MA) and Cell Quest Software (Becton Dickinson, San Jose, CA).

Inhibitors of Caspase Activation and PI3k Activity.

The tetrapeptide caspase inhibitor ZVAD-fmk (Biomol Research Laboratories Inc., Plymouth, PA) was resuspended in DMSO (Sigma Chemical Co., St. Louis, MO) at a concentration of 1 mm. ZVAD-fmk was added to cells treated with or without trastuzumab (as described) at a final concentration of 50 μm 1 h before TRAIL treatment. Control cells were incubated with DMSO at the same concentration as test cells. Cell viability was analyzed by the MTS assay after 16 h of incubation with TRAIL.

The inhibitor of PI3k activity, LY294002 (Alexis, San Diego, CA), was stored at a concentration of 25 mg/ml in DMSO. LY294002 was added to a final concentration of 10 μm for 6 h before TRAIL treatment. Control cells were incubated with DMSO at the same concentration as test cells. Cell viability was analyzed by the MTS assay after 16 h of incubation with TRAIL.

ODNs and TRAIL-mediated Toxicity.

Antisense (5′-CTCCATGGTGCTCAC-3′) and sense (5′-GTGAGCACCATGGAG-3′) phosphorothioate ODNs targeting the 5′ region of the erbB-2 mRNA molecule were obtained from Sigma Chemical Co./Genosys (The Woodlands, TX). The lyophilized ODNs were reconstituted in sterile distilled water to 1 mm, filter-sterilized, and stored in aliquots at −20°C as stock solutions. For subsequent experiments, the stock solutions of ODNs were diluted to give a final concentration of 1 μm. To assess TRAIL-mediated toxicity on ODN treatment, cells were plated at 2 × 104 in 96-well plates, allowed to adhere overnight, and transfected with the ODNs. Diluted ODNs were mixed with 2 μg/ml LipofectAMINE (Life Technologies, Inc.), cells were exposed to the mixture for 4 h, the transfection medium was replaced with fresh culture medium, and the cells were incubated for an additional 48 h. Freshly eluted TRAIL at the concentrations indicated in the figure legends was added to the cells, which were incubated for an additional 16 h. Cell viability was analyzed by the MTS assay.

Isolation and Analysis of Protein Lysates.

Protein was extracted from cells by detergent lysis [1% Triton X-100, 10 mm Tris-HCl (pH 7.5), 150 mm NaCl, 5 mm EDTA, 10% glycerol, 2 mm sodium vanadate, and protease inhibitors (Complete tabs; Roche Molecular Biochemicals)]. Protein lysates were cleared of debris by centrifugation at 15,000 × g for 10 min at 4°C, and the concentration was assessed by the Bio-Rad colorimetric assay (Bio-Rad). Protein samples were boiled in an equal volume of sample buffer [20% glycerol, 4% SDS, 0.2% bromphenol blue, 125 mm Tris-HCl (pH 7.5), and 640 mm β-mercaptoethanol], fractionated by 10–12% SDS-PAGE, and transferred to polyvinylidene fluoride membranes (Millipore, Bedford, MA). Polyclonal rabbit anti-erbB-2 antibody (RB-103-P; Neomarkers, Fremont, CA; 2 μg/ml), mouse monoclonal horseradish peroxidase-conjugated anti-phosphotyrosine antibody (clone 4G10; Upstate Biotechnology, Lake Placid, NY; 0.5 μg/ml), rabbit polyclonal anti-TRAIL-R1 antibody (Calbiochem, San Diego, CA; 2 μg/ml), rabbit polyclonal anti-TRAIL-R2 antibody (Imgenex, San Diego, CA; 1 μg/ml), rabbit polyclonal anti-TRAIL-R3 antibody (Affinity Bioreagents, Golden, CO; 1 μg/ml), anti-TRAIL-R4 antibody (Oncogene Research Products, Cambridge, MA; 1 μg/ml), rabbit polyclonal anti-IAP-1 antibody (R&D Systems Inc., Minneapolis, MN; 1 μg/ml), rabbit polyclonal anti-IAP-2 antibody (R&D Systems Inc.; 1.5 μg/ml), rabbit polyclonal anti-FLIP antibody (Calbiochem; 2 μg/ml), rabbit polyclonal anti-Akt antibody (9272; New England Biolabs, Beverly, MA; 2 μg/ml), rabbit polyclonal anti-phospho-Akt antibody (9271; New England Biolabs; 2 μg/ml), rabbit polyclonal anti-erk-2 antibody (C-14; Santa Cruz Biotechnology, Santa Cruz Biotechnology, CA; 1 μg/ml), mouse monoclonal anti-phospho-erk (Thr 202/Tyr 204) antibody (9106; New Englands Biolabs; 0.5 μg/ml), mouse monoclonal anti-phospho-gsk-3 α/β (9331; New England Biolabs; 1 μg/ml), mouse monoclonal anti-HA (Roche Molecular Biochemicals; 2 μg/ml), rabbit polyclonal anti-HA (Y-11, sc-805; Santa Cruz Biotechnology), and mouse monoclonal anti-α-tubulin antibody (T9026; Sigma Chemical Co.) were used for immunoblotting. Goat antirabbit and antimouse antibodies conjugated with horseradish peroxidase (Amersham Pharmacia Biotech Inc.) was used to visualize immunoreactive proteins at a 1:5000 dilution using SuperSignal (Pierce, Rockford, IL) detection reagent.

Assessment of Cell Surface Death Receptor Expression.

The total TRAIL surface binding was determined by flow cytometry by measuring the binding of GST-TRAIL or GST alone. After 72 h of incubation in the presence or absence of trastuzumab, the SKBr-3 cells were washed once with cold PBS and harvested using EDTA (2.5 μm) in cold PBS. The cells were pelleted and resuspended in cold PBS containing 1% FCS, 0.02 mm sodium azide, and 0.5 mm EDTA. The cells were then incubated with freshly eluted GST or GST-TRAIL at 20 μg/ml for 1 h at 4°C. After this, the cells were washed once with cold PBS, resuspended in the solution described above, and then incubated with a mouse FITC-conjugated anti-GST antibody (Santa Cruz Biotechnology) at 10 μg/ml for 45 min at 4°C. Finally, the cells were washed once in cold PBS, and surface staining was determined using the FACSort system (Becton Dickinson) and Cell Quest Software (Becton Dickinson). The specific surface expression of TRAIL-R1 and TRAIL-R2 was determined by measuring the binding of a mouse anti-TRAIL-R1 antibody (Santa Cruz Biotechnology) or a mouse anti-TRAIL-R2 antibody (Imgenex), respectively. A purified mouse IgG1 immunoglobulin (PharMingen, San Diego, CA) was used as an isotype-matched antibody. Cells were treated and incubated as described above. After the incubation with mouse anti-TRAIL-R1 (20 μg/ml), mouse anti-TRAIL-R2 (20 μg/ml), or mouse isotype immunoglobulin (20 μg/ml), the cells were incubated with a goat FITC-conjugated antimouse IgG (Sigma Chemical Co.; 10 μg/ml). The surface staining was determined as described above.

Akt Kinase Assays.

Stable clones of MDA-MB-453 cells expressing the HA-tagged myristylated Akt kinase were plated at 2 × 106 in 100-mm dishes, allowed to adhere overnight, and treated with trastuzumab as described above. HA-tagged Akt kinase was immunoprecipitated by using mouse monoclonal anti-HA antibody, and Akt kinase activity was assayed by detecting phosphorylation of gsk-3 α/β protein under conditions recommended by the manufacturer (Akt kinase assay kit; New England Biolabs). The assay was performed in triplicate.

Statistical Analysis.

Statistical comparison of mean values was performed using Student’s t test (paired and unpaired). All Ps are two-tailed. Interactions between TRAIL and trastuzumab were classified by the fractional inhibition method as follows: when expressed as the fractional inhibition of cell viability, additive inhibition produced by both inhibitors (i) occurs when i1,2 = i1 + i2; synergism occurs when i1,2 > i1 + i2; and antagonism occurs when i1,2 < i1 + i2 (47) . The synergism was confirmed by dose-effect analysis using Syncalc software (Biososft, Cambridge, United Kingdom; Ref. 48 ).

RESULTS

Trastuzumab Enhances TRAIL-mediated Apoptosis in erbB-2-overexpressing Cancer Cell Lines.

Breast and ovarian cancer cell lines expressing different levels of the erbB-2 receptor were assessed for sensitivity to TRAIL-mediated apoptosis in the presence or absence of trastuzumab pretreatment (Fig. 1A) ⇓ . Treatment of erbB-2-overexpressing cell lines (SKBr-3, MDA-MB-453, and SKOv-3) with trastuzumab significantly enhances TRAIL-mediated growth inhibition. These cells show low or modest sensitivity to either reagent alone, but the combination results in 30–40% growth inhibition. The combined effect of trastuzumab and TRAIL in the erbB-2-overexpressing cell lines was similar to the effect of TRAIL alone in the TRAIL-sensitive cell line MDA-MB-231 (18) . In contrast, in cells with low erbB-2 receptor expression, there was no enhancement of TRAIL-mediated toxicity by pretreatment with trastuzumab in either the TRAIL-resistant (MCF-7, MDA-MB-468, and SW-626) or TRAIL-sensitive (MDA-MB-231) cell lines. The level of the erbB-2 receptor for each cell line is shown in the bottom panel of Fig. 1A ⇓ . Longer exposure demonstrated the presence of low levels of erbB-2 receptor in MDA-MB-468, SW-626, and MDA-MB-231 cells (data not shown). The enhancement of TRAIL-mediated toxicity by trastuzumab antibody required pretreatment for at least 48 h. Cotreatment or shorter pretreatments did not result in enhanced TRAIL-mediated toxicity. The enhancement of TRAIL-mediated toxicity by trastuzumab does not reflect a shift in the kinetics of TRAIL-mediated apoptosis. Longer incubation with TRAIL alone (i.e., 48 or 72 h) resulted in only slight increases in TRAIL-mediated toxicity. Pretreatment with trastuzumab resulted in a significant increase of TRAIL-mediated toxicity when TRAIL was added for 24, 48, or 72 h. Similarly, no shift in kinetics was seen when the length of trastuzumab preincubation was varied (data not shown). The lack of inhibitory effect by trastuzumab on cells expressing low levels of the erbB-2 receptor is consistent with previous reports that demonstrated that only cells overexpressing erbB-2 are inhibited by trastuzumab (49) . Interestingly, in cells expressing low levels of the erbB-2 receptor (i.e., MCF-7, SW-626, or MDA-MB-231), trastuzumab induced slight growth enhancement. Trastuzumab, like other antibodies against erbB-2, is a weak agonist for the erbB-2 receptor (42 , 50 , 51) , and this may account for the growth in cell lines expressing low amounts of erbB-2.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Trastuzumab enhances TRAIL-mediated apoptosis in erbB-2-overexpressing cell lines. A, top panel, seven cell lines [three erbB-2-overexpressing cell lines (SKBr-3, MDA-MB-453, and SKOv-3) and four cell lines with normal to low erbB-2 levels (MCF-7, MDA-MB-468, SW-626, and MDA-MB-231)] were incubated with trastuzumab alone (□), TRAIL alone ( Embedded Image), or the combination (▪). Cells were incubated with or without trastuzumab (1 μm) for 96 h, and TRAIL was added to a final concentration of 1 μg/ml for the final 16 h in culture. The low erbB-2 receptor-expressing cell line MDA-MB-231 was used as a positive control for TRAIL treatment (18) . Cell viability was measured by the MTS assay, and the data represent growth inhibition as a percentage of control cells. Data points show the mean ± SE for a minimum of three experiments for each cell line. The negative effect observed on trastuzumab incubation in MCF-7, SW-626, and MDA-MB-231 cell lines was not statistically different compared with the controls. A, bottom panel, Western blot probed with an erbB-2 antibody shows levels of expression in all cancer cell lines. MAPK expression is shown as a loading control. Molecular weights (in thousands) are shown to the left. B, an erbB-2-overexpressing cancer cell line (SKOv-3) was incubated with TRAIL alone (□), TRAIL + rituximab ( Embedded Image), or TRAIL + trastuzumab (▪). Cells were incubated with trastuzumab (1 μm) or rituximab (1 μm) for a total of 96 h, and TRAIL was added to the concentrations indicated for the final 16 h in culture. Cell viability was measured by the MTS assay, and the data represent growth inhibition as a percentage of control cells. Data points show the mean ± SE for a minimum of three experiments.

To assess the effect of trastuzumab further and to demonstrate the specificity of trastuzumab, the SKOv-3 cells were incubated without antibody, with trastuzumab, or with the isotype-matched antibody rituximab and then treated with varying concentrations of TRAIL (Fig. 1B) ⇓ . A significant enhancement of TRAIL-mediated toxicity by trastuzumab was seen, even at concentrations of TRAIL that alone had little or no toxic effect (i.e., 0.025 or 0.1 μg/ml). In contrast, rituximab had no toxic effect alone or in combination with TRAIL. Rituximab also had no toxic effect alone or in combination with TRAIL in other erbB-2-expressing or non-erbB-2-expressing cells (data not shown).

When cells were incubated with varying concentrations of trastuzumab and then treated with TRAIL (1 μg/ml), enhancement of TRAIL-mediated toxicity could be seen at 10-fold lower doses of trastuzumab (data not shown). When the sum of each treatment was compared with the combination, the combined treatment was statistically greater than the sum of the individual treatments (P < 0.04 and P < 0.01 by unpaired and paired two-tailed t tests, respectively). Thus the enhancement of TRAIL-mediated toxicity by trastuzumab in the erbB-2-overexpressing cell lines was more than additive. By both fractional inhibition analysis (47) and dose-effect analysis (48) , the effects of the combination were synergistic compared with either reagent alone.

TRAIL has been demonstrated to induce apoptosis by a caspase-dependent mechanism (3 , 7) . To determine whether the increased inhibition by the combination of trastuzumab plus TRAIL seen in the MTS assays was due to the induction of apoptosis, the effect of caspase inhibition was measured (Fig. 2A) ⇓ . SKBr-3 cells were incubated with or without trastuzumab, and 1 h before the addition of TRAIL, the caspase inhibitor ZVAD-fmk was added to the cultures. ZVAD-fmk completely inhibited the toxicity of TRAIL either alone or in combination with trastuzumab. Caspase inhibition only slightly blocked the toxicity of trastuzumab alone. The partial inhibition of trastuzumab-induced toxicity results because there was no inhibitor present for most of the time that trastuzumab was present. When ZVAD-fmk is added at the same time as the trastuzumab, an almost complete inhibition of trastuzumab-induced toxicity is observed (data not shown). This is consistent with trastuzumab inducing apoptosis via a caspase-dependent mechanism. In previous work (18) , we have shown that chemotherapy could induce TRAIL-mediated apoptosis by augmenting activation of caspase-3. By itself, TRAIL weakly activates caspase-3, but trastuzumab did not increase this activation (data not shown). This is consistent with the demonstrated existence of both caspase-3-dependent and -independent mechanisms of apoptosis (52) .

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Inhibition of caspase activation blocks TRAIL-mediated apoptosis. A, the erbB-2-overexpressing cancer cell line SKBr-3 was incubated with trastuzumab alone, TRAIL alone, or the combination. Cells were incubated with trastuzumab (1 μm) for 96 h, and TRAIL was added to a final concentration of 1 μg/ml for the final 16 h in culture. One h before the addition of TRAIL, caspase inhibitor ZVAD-fmk (50 μm ; ▪) or PBS alone (□) was added to the culture. Cell viability was measured by the MTS assay. The data represent the growth inhibition, as a percentage of control cells. Data points show the mean ± SE for a minimum of three experiments. The Ps comparing each treatment with control cells are shown above the data. B, trastuzumab enhances TRAIL-mediated apoptosis as measured by using flow cytometry. The erbB-2-overexpressing cancer cell line SKBr-3 was incubated with trastuzumab alone, TRAIL alone, or the combination. Cells were incubated without (control) or with trastuzumab (1 μm) for 96 h, and TRAIL (1 μg/ml) was added for the final 16 h in culture. Cells were collected and stained with propidium iodide. Apoptosis was measured as a percentage of cells with sub-G0-G1 DNA content in the DNA histogram compared with the total number of cells present. Data points show the mean ± SE for a minimum of three experiments. The Ps comparing each treatment with control cells are shown above the data.

To quantitate the induction of apoptosis in the cultures, the fraction of cells with sub-G0-G1 DNA content was measured by flow cytometry (Fig. 2B) ⇓ . Whereas trastuzumab or TRAIL alone each significantly increased the fraction of apoptotic cells compared with control cells, the combination of trastuzumab plus TRAIL results in a synergistic increase in the fraction of apoptotic cells. In these experiments, TRAIL induced greater apoptosis than trastuzumab, whereas Fig. 1A ⇓ shows that the trastuzumab treatment resulted in greater growth inhibition than TRAIL. The measurement of apoptosis by flow cytometry (Fig. 2B) ⇓ shows only those cells undergoing apoptosis at the time of the measurement, whereas the MTS assay shows the cumulative effect of ongoing apoptosis over the entire period. Trastuzumab was present in the medium for 72 h before TRAIL treatment. Thus, the greater growth inhibition induced by trastuzumab as compared with TRAIL observed in Fig. 1A ⇓ likely reflects ongoing apoptosis over the entire 96-h incubation. Together, the results of caspase inhibition and flow cytometry demonstrate that the enhancement of TRAIL-mediated toxicity by trastuzumab is due to the induction of apoptosis.

We evaluated whether the enhancement of TRAIL-mediated apoptosis by trastuzumab preincubation resulted from changes in the expression of proteins known to modulate TRAIL sensitivity (Fig. 3A) ⇓ . There was no change in protein expression of TRAIL-Rs, IAP-1, IAP-2, or FLIP. In addition, on trastuzumab treatment, there was no change in the surface expression of the death-inducing receptors (TRAIL-R1 and TRAIL-R2; Fig. 3B ⇓ ). There was also no change in total TRAIL binding, indicating that there is also no change in the surface levels of the decoy receptors (TRAIL-R3 and TRAIL-R4). Thus, alterations in expression of different components of the TRAIL pathway do not account for the increase in TRAIL-mediated apoptosis.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Treatment with trastuzumab does not change the expression of TRAIL-Rs or proteins involved in the TRAIL-mediated apoptosis pathway. A, two different cell lines overexpressing erbB-2 (SKBr-3 and SKOv-3) and a cell line expressing low levels of erbB-2 (MDA-MB-231) were incubated in medium supplemented with 1 μm trastuzumab (+) or PBS (−) for 4 days. Cell lysates were prepared and immunoblotted with TRAIL-R1, TRAIL-R2, TRAIL-R3, TRAIL-R4, IAP-1, IAP-2, cFlip, or MAPK antibody, as indicated to the right. MAPK is shown as a loading control. Molecular weights (in thousands) are shown to the left. B, the erbB-2-overexpressing cell line SKBr-3 was incubated with PBS or trastuzumab (1 μm) for 72 h to determine the total surface expression of the death receptors and the surface expression of TRAIL-R1 and TRAIL-R2. The total surface TRAIL binding was determined by measuring GST-TRAIL or GST binding with a FITC-conjugated anti-GST antibody by flow cytometry. The surface expression of TRAIL-R1 and TRAIL-R2 was determined by measuring the binding of a mouse anti-TRAIL-R1, anti-TRAIL-R2, or isotype IgG antibody (as a control) with a FITC-conjugated antimouse antibody by flow cytometry. The top, middle, and bottom panels represent the total surface expression, the TRAIL-R1 surface expression, and the TRAIL-R2 surface expression, respectively. The curves represent the control (thin black line) and the surface expression on PBS (thick black line) and trastuzumab (dashed line) incubation. The curves showing surface expression on incubation with PBS or trastuzumab overlap.

Trastuzumab Down-Regulates the erbB-2 Receptor.

Incubation of cells overexpressing the erbB-2 receptor with trastuzumab for 96 h resulted in down-regulation of the erbB-2 protein and a decrease in the amount of phosphorylated erbB-2 receptor (Fig. 4) ⇓ . Short incubations (5 min to 8 h) of cells with trastuzumab resulted in increased activation of the erbB-2 protein (as measured by phosphorylation of the receptor) and no decrease in protein levels (data not shown). Longer incubations (>24 h) resulted in a progressive decrease and inactivation of the erbB-2 protein with maximum down-regulation at 72–96 h. This time course of erbB-2 down-regulation is consistent with the long preincubation necessary for trastuzumab to enhance TRAIL toxicity.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Trastuzumab down-regulates the erbB-2 receptor. Three different cell lines overexpressing erbB-2 (SKBr-3, MDA-MB-453, and SKOv-3) were incubated in medium supplemented with trastuzumab at 1 μm (+) or PBS (−) for 4 days. Cell lysates were prepared and immunoblotted with erbB-2, phosphotyrosine (p-tyr), or tubulin antibody as indicated to the right. Tubulin is shown as a loading control. Molecular weights (in thousands) are shown to the left.

To assess whether down-regulation of the erbB-2 receptor is sufficient to enhance TRAIL-mediated apoptosis, cells were incubated with antisense ODNs. Antisense ODNs have previously been shown to down-regulate the erbB-2 receptor in breast cell lines (53 , 54) . The SKBr-3 cell line was transfected with antisense or sense ODNs or mock-transfected, and then cells were treated with TRAIL (Fig. 5A) ⇓ . The antisense ODNs induced significantly more toxicity (15–20%) than mock-transfection (P < 0.02) or sense ODNs (P < 0.02). A slight increase in toxicity by sense ODNs alone (5–10%) was observed compared with the mock transfection, but this increase was not statistically significant (P = 0.15). There was a significant increase in TRAIL-mediated apoptosis in cells transfected with antisense ODNs (30–40%; P < 0.001) but not in cells transfected with sense ODNs (P = 0.25) compared with mock-transfected cells (Fig. 5A) ⇓ . By fractional inhibition analysis, the interaction between antisense ODNs and TRAIL was synergistic. Lysates obtained from cells transfected under the same conditions showed a significant down-regulation of the erbB-2 receptor in cells transfected with antisense ODNs, but not in mock-transfected cells or in cells transfected with sense ODNs (Fig. 5B) ⇓ . Thus, down-regulation of the erbB-2 receptor by two distinct mechanisms enhanced TRAIL-mediated apoptosis.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Down-regulation of erbB-2 by antisense ODNs enhances TRAIL-mediated toxicity. A, the erbB-2-overexpressing cancer cell line SKBr-3 was transfected with antisense ODNs (AS) at 1 μm or sense ODNs (S) at 1 μm or mock-transfected (M). Cells were incubated for an additional 72 h after transfection, and TRAIL was added at a final concentration of 1 μg/ml for the final 16 h in culture. Cell viability was measured by the MTS assay, and the data represent growth inhibition as a percentage of control cells in absence (□) or presence of TRAIL (▪). Data points show the mean ± SE for a minimum of three experiments. The statistical analysis is summarized as follows: antisense ODNs versus mock-transfection, P < 0.02; antisense ODNs versus sense ODNs, P < 0.02; sense ODNs versus mock-transfection, P = 0.15; antisense ODNs + TRAIL versus TRAIL alone, P < 0.001; and sense ODNs + TRAIL versus TRAIL alone, P = 0.25. B, cell lysates prepared from cells transfected as described above were immunoblotted with erbB-2 or tubulin antibody as indicated to the right. Tubulin is shown as a loading control. Molecular weights (in thousands) are shown to the left.

Down-Regulation of the erbB-2 Receptor Inhibits Akt Kinase Activity but not MAPK Activity.

erbB-2 overexpression results in activation of different downstream pathways such as the Akt kinase and MAPK pathways, which leads to cell proliferation and cell survival (39 , 41) . To assess whether down-regulation of the erbB-2 receptor affects these downstream pathways, cells overexpressing the erbB-2 receptor (SKBr-3 and MDA-MB-453) or cells with low erbB-2 expression (MDA-MB-468) were treated with trastuzumab, and then the activities of the Akt kinase and MAPK pathways were measured. Both the Akt kinase and MAPK are activated in response to growth factor receptor activation by phosphorylation of specific serine and threonine residues (55 , 56) . The activation of Akt kinase and MAPK can be measured using phospho-specific antibodies (55 , 56) . Trastuzumab treatment resulted in a significant reduction of Akt kinase activity as measured by a phospho-Akt kinase-specific antibody, whereas there was no decrease in total Akt protein levels (Fig. 6A) ⇓ . Therefore, the change in phosphorylation represents a decrease in activity and not down-regulation of the Akt protein. Trastuzumab induced a similar decrease in Akt kinase activity in the SKOv-3 cell line (data not shown). In contrast, there was no change in the activity of MAPK as measured by a phospho-MAPK-specific antibody (Fig. 6A) ⇓ . In cells expressing low levels of the erbB-2 receptor (MDA-MB-468 and MDA-MB-231), trastuzumab did not induce changes in either Akt kinase or MAPK activity. Interestingly, the MDA-MB-231 cell line, which is very sensitive to TRAIL-mediated apoptosis, has very low levels of phospho-Akt. These pathways were also assessed in cells treated with antisense or sense ODNs (Fig. 6B) ⇓ . As observed with trastuzumab, there was a significant reduction in Akt kinase activation and no change in MAPK activation in cells transfected with antisense ODNs. There was no change in the activity of either of these kinases in mock-transfected or sense ODN-transfected cells.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Down-regulation of the erbB-2 receptor by trastuzumab decreases Akt kinase activation but not MAPK activation. A, two erbB-2-overexpressing cancer cell lines (SKBr-3 and MDA-MB-453) and two cell lines expressing low levels of erbB-2 receptor (MDA-MB-468 and MDA-MB-231) were incubated with trastuzumab (1 μm) for 4 days. Cell lysates were prepared and immunoblotted with antibodies to phospho-Akt (p-Akt), Akt, phospho-MAPK (p-MAPK), MAPK, and tubulin as indicated to the right. Tubulin is shown as a loading control. The bracket at the right indicates p-MAPK. The relative intensity of phosphorylation of erk1 and erk2 varied among different cell lines. Molecular weights (in thousands) are shown to the left. B, the erbB-2-overexpressing cell line SKBr-3 was transfected with antisense (AS) or sense (S) ODNs or mock-transfected (M). After transfection, cells were incubated for an additional 72 h. Lysates were prepared and immunoblotted with antibodies as described in A. Molecular weights (in thousands) are shown to the left.

Decrease in Akt Kinase Activity Enhances TRAIL Toxicity.

The data above suggest that the enhancement of TRAIL-mediated apoptosis by down-regulation of the erbB-2 receptor (by either trastuzumab or antisense ODNs) results from decreased Akt kinase activity. The activation of Akt kinase by growth factor receptors is mediated by PI3k activity (56) . To assess whether the decrease of Akt kinase activity is responsible for the observed increase in TRAIL-mediated apoptosis described above, cells overexpressing erbB-2 receptor (SKBr-3) or expressing a low amount of erbB-2 receptor (MDA-MB-468) were treated with a PI3k inhibitor (LY294002) before the addition of TRAIL. In both cell lines, LY294002 was toxic by itself. There was a significant enhancement in TRAIL-mediated toxicity in both cell lines by pretreatment with LY294002 (Fig. 7A) ⇓ . Immunoblotting with phospho-Akt demonstrated that LY294002 pretreatment resulted in decreased Akt activity (Fig. 7B) ⇓ .

Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

Inhibition of Akt kinase activity enhances TRAIL-mediated apoptosis. A, two breast cancer cell lines, one overexpressing erbB-2 (SKBr-3) and the other with low erbB-2 levels (MDA-MB-468), were incubated in the presence or absence of a PI3k inhibitor, LY294002 at 10 μm in complete medium (10% fetal bovine serum) for 6 h. After inhibitor incubation, TRAIL was added at a final concentration of 1 μg/ml for the final 16 h. □, inhibition of growth induced by LY294002 alone; Embedded Image, inhibition of growth induced by TRAIL alone; and ▪, inhibition of growth by the combination of TRAIL + LY294002 as a percentage of control cells. Cell viability was measured by the MTS assay. Data points show the mean ± SE for a minimum of three experiments. The Ps (*) compare TRAIL + LY294002 with TRAIL alone in each cell line. B, cell lysates from both cell lines incubated in the absence (−) or presence (+) of LY294002 (Ly) were prepared and immunoblotted with phospho-Akt (p-Akt) and Akt antibodies as indicated to the right. Molecular weights (in thousands) are shown to the left.

To confirm that the Akt kinase is the critical target affected by erbB-2 down-regulation, a constitutively active (membrane-targeted) Akt kinase was introduced into the MDA-MB-453 cell line, which overexpresses the erbB-2 receptor. Vector control or the activated Akt kinase clones were treated with trastuzumab alone, TRAIL alone, or the combination, and toxicity was assayed as described in the Fig. 1 ⇓ legend. Treatment of the vector clone with the combination of trastuzumab + TRAIL resulted in 35–40% inhibition compared with the control cells. This was similar to the effect in the parental MDA-MB-453 cells, as seen in Fig. 1A ⇓ . Expression of the constitutively active Akt kinase in two independent clones resulted in a significant reduction of toxicity with each reagent alone or the combination (Fig. 8A) ⇓ . There was almost complete abrogation of the synergism between trastuzumab and TRAIL in the clones expressing the active Akt kinase. Lysates from these clones were immunoblotted using erbB-2 and phospho-Akt antibodies (Fig. 8B) ⇓ . The vector and the active Akt kinase clones all showed down-regulation of the erbB-2 receptor. Reduced levels of phospho-Akt after trastuzumab incubation were observed. However, because the transfected constitutively active Akt kinase is a small fraction of the total Akt kinase and is myristylated, the phosphorylation status does not reflect its activity accurately. The exogenous constitutively active Akt kinase was immunoprecipitated via its HA epitope, and its activity was measured by an in vitro kinase assay using a specific phospho-gsk-3 antibody (Fig. 8C) ⇓ . No suppression of the exogenous Akt kinase activity by trastuzumab treatment was seen.

Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Constitutively active Akt kinase inhibits the enhancement of TRAIL-mediated apoptosis by trastuzumab. A, stable clones expressing a constitutively active form of Akt kinase were generated in an erbB-2-overexpressing cell line (MDA-MB-453). The vector control and two different clones (2/7 and 2/8) were incubated with trastuzumab alone (□), TRAIL alone ( Embedded Image), or the combination (▪). Cells were incubated with or without trastuzumab (1 μm) for 96 h, and TRAIL was added at a final concentration of 1 μg/ml for the final 16 h in culture. Cell viability was measured by the MTS assay, and the data represent growth inhibition as a percentage of control cells. Data points show the mean ± SE for a minimum of three experiments for each cell line. B, cell lysates from all of the clones incubated in the absence (−) or presence (+) of trastuzumab (1 μm) for 4 days were immunoblotted with erbB-2, phospho-Akt (p-Akt), Akt, or MAPK antibodies as indicated to the right. MAPK is shown as a loading control. Molecular weights (in thousands) are shown to the left. C, exogenous Akt kinase was immunoprecipitated with anti-HA antibody and immunoblotted with phospho-gsk-3 (p-gsk-3) and HA antibodies as indicated to the right. Molecular weights (in thousands) are shown to the left.

DISCUSSION

In the work presented here, we demonstrate that down-regulation of the erbB-2 receptor by either trastuzumab or antisense ODNs enhances TRAIL-mediated apoptosis in breast and ovarian cancer cells that overexpress erbB-2 (Figs. 1 ⇓ 2 ⇓ 3 ⇓ 4 ⇓ 5 ⇓ ). Cross-linking of the erbB-2 receptor by trastuzumab activates the receptor (57) . Recent reports have demonstrated that down-regulation of erbB-2 by anti-erbB-2 antibodies results in recruitment of the cbl protein, ubiquitination, and proteasome-dependent degradation of the activated receptors (46) . Antisense ODNs, on the other hand, block synthesis of the erbB-2 protein (53 , 58) . Thus, the enhancement of TRAIL-mediated apoptosis by erbB-2 down-regulation is independent of the mechanism through which down-regulation is achieved.

Trastuzumab specifically enhances TRAIL-mediated apoptosis in erbB-2-overexpressing cells. There was no enhancement of TRAIL-mediated apoptosis in all of the cell lines tested that express low levels of the erbB-2 receptor (Fig. 1A) ⇓ . The selectivity of this effect results in targeting TRAIL only to those cells that overexpress erbB-2 receptor, thus potentially avoiding toxicity to normal cells that express low levels of erbB-2. Furthermore, other ways to down-regulate the receptor, such as antisense ODNs, might also be expected to selectively sensitize only the cancer cells that overexpress erbB-2.

Down-regulation of the erbB-2 receptor did not alter total or surface expression of TRAIL-Rs or other proteins that have been shown to modulate TRAIL-mediated apoptosis (Fig. 3A) ⇓ . The mechanism underlying the enhancement of TRAIL-mediated apoptosis is inhibition of the prosurvival Akt kinase pathway. Akt/protein kinase B, the cellular homologue of the viral oncoprotein v-Akt, is a serine/threonine kinase that has been identified as an important component of prosurvival signaling pathways (59) . Once activated, Akt exerts antiapoptotic effects through phosphorylation of substrates that directly regulate the apoptotic machinery such as Bad and caspase-9 (60) . In addition, Akt also phosphorylates substrates that indirectly inhibit apoptosis such as the human telomerase reverse transcriptase subunit, the forkhead transcription family members, or the IκB kinases (61, 62, 63) . All cell lines overexpressing the erbB-2 receptor, which are resistant to TRAIL-mediated apoptosis, show high levels of Akt kinase activity. Similarly, two cell lines expressing low levels of erbB-2 receptor (MDA-MB-468 and SW-626) also show high levels of Akt activity and resistance to TRAIL-mediated apoptosis. In contrast, the MDA-MB-231 cell line, which expresses low levels of erbB-2 receptor, has low Akt activity and is extremely sensitive to TRAIL-mediated apoptosis.

In all of the erbB-2-overexpressing cell lines tested, there was a decrease in activation of the Akt kinase when the erbB-2 receptor was down-regulated by trastuzumab or antisense ODNs (Fig. 6) ⇓ . Although erbB-2 down-regulation was seen in cells with low erbB-2 expression, there was no change in Akt kinase activation or biological effect by either agent alone or the combination. erbB-2 down-regulation did not decrease MAPK activation. Both Akt kinase and MAPK are activated in response to growth factor receptor activation and up-regulated when erbB-2 receptor is overexpressed (39 , 40 , 42) . The reason for the selective decrease in Akt kinase activity is unknown.

The Akt kinase is activated via the PI3k signaling pathway (59) . Similar enhancement in TRAIL-mediated apoptosis was observed when the phosphorylation of Akt kinase was blocked by using an inhibitor of PI3k activity. However, LY294002 treatment resulted in a significant enhancement of TRAIL-mediated apoptosis in erbB-2-overexpressing cells as well as in cells with low erbB-2 expression (Fig. 7) ⇓ . These data suggest that other pathways keep the Akt kinase active in the cell lines with low erbB-2 expression. The expression of a constitutively active form of Akt kinase in erbB-2-overexpressing cell lines almost abrogated the effect of trastuzumab alone, TRAIL alone, or the combination, indicating that Akt kinase is a major determinant in cell survival in erbB-2-overexpressing cell lines (Fig. 8) ⇓ . Therefore, down-regulation of Akt kinase activity will result in an enhancement of TRAIL-mediated apoptosis independent of the underlying mechanisms. Thus, targeting the Akt kinase pathway may enhance TRAIL-mediated apoptosis in many cell types, even those that do not overexpress the erbB-2 receptor.

It is not surprising that inhibition of the antiapoptotic Akt kinase pathway would sensitize cells to death receptor-induced apoptosis. Consistent with this, previously published work has shown that erbB-2 overexpression inhibits TNF-induced apoptosis by activation of Akt kinase and that an anti-erbB-2 antibody enhances TNF-induced apoptosis (64 , 65) . It is also likely that the enhanced efficacy of chemotherapy by trastuzumab is mediated through this Akt kinase-dependent mechanism.

In summary, down-regulation of erbB-2 by trastuzumab enhances TRAIL-mediated apoptosis specifically in erbB-2-overexpressing breast and ovarian cancer cell lines. The mechanism behind the interaction can be explained at least in part by down-regulation of the prosurvival Akt kinase pathway. Trastuzumab has been described to have collateral toxicity in normal tissues (66) . More recent data suggest that TRAIL may be toxic to normal human liver cells (67) . Therefore, combinations of therapy that maintain the efficacy of TRAIL and trastuzumab at lower doses may be important to their clinical usefulness. Our results indicate that the interaction between trastuzumab and TRAIL is still present after reducing the dose of each reagent. The trastuzumab concentrations that sensitized cells to TRAIL-mediated apoptosis were within a range clinically achieved in patients (68) . Therefore, the identification of this synergistic interaction between these two drugs with effects targeted specifically to cancer cells is attractive as a new treatment modality. These data are in vitro experiments with cancer cell lines, and hence the efficacy and specificity of this combination remain to be tested in animal models.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 Present address: Department of Oncology, University College Hospital Galway, Galway, Republic of Ireland.

  • ↵2 To whom requests for reprints should be addressed, at Genetics Department, Medicine Branch, National Cancer Institute, Building 8, Room 5101, National Naval Medical Center, Bethesda, MD 20889. Phone: (301) 402-4276; Fax: (301) 496-0047; E-mail: Stan_Lipkowitz{at}nih.gov

  • ↵3 The abbreviations used are: TNF, tumor necrosis factor; GST, glutathione S-transferase; MAPK, mitogen-activated protein kinase; MTS, [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium]; ODN, oligodeoxynucleotide; PI3k, phosphatidylinositol 3′-kinase; TRAIL, TNF-related apoptosis-inducing ligand; TRAIL-R, TRAIL receptor; ZVAD-fmk, Z-Val-Ala-Asp (OMe)-CH2F; HA, hemagglutinin; erk, extracellular signal-regulated kinase; FLIP, Flice-inhibitory proteins; IAP, inhibitor of apoptosis protein.

  • Received September 6, 2000.
  • Accepted April 9, 2001.
  • ©2001 American Association for Cancer Research.

References

  1. ↵
    Schulze-Osthoff K., Ferrari D., Los M., Wesselborg S., Peter M. E. Apoptosis signaling by death receptors. Eur. J. Biochem., 254: 439-459, 1998.
    OpenUrlPubMed
  2. ↵
    Muzio M. Signalling by proteolysis: death receptors induce apoptosis. Int. J. Clin. Lab. Res., 28: 141-147, 1998.
    OpenUrlCrossRefPubMed
  3. ↵
    Ashkenazi A., Dixit V. M. Death receptors: signaling and modulation. Science (Wash. DC), 281: 1305-1308, 1998.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Smith C. A., Farrah T., Goodwin R. G. The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell, 76: 959-962, 1994.
    OpenUrlCrossRefPubMed
  5. ↵
    Ogasawara J., Watanabe-Fukunaga R., Adachi M., Matsuzawa A., Kasugai T., Kitamura Y., Itoh N., Suda T., Nagata S. Lethal effect of the anti-Fas antibody in mice. Nature (Lond.), 364: 806-809, 1993.
    OpenUrlCrossRefPubMed
  6. ↵
    Ashkenazi A., Dixit V. M. Apoptosis control by death and decoy receptors. Curr. Opin. Cell Biol., 11: 255-260, 1999.
    OpenUrlCrossRefPubMed
  7. ↵
    Griffith T. S., Lynch D. H. TRAIL: a molecule with multiple receptors and control mechanisms. Curr. Opin. Immunol., 10: 559-563, 1998.
    OpenUrlCrossRefPubMed
  8. ↵
    Marsters S. A., Pitti R. A., Sheridan J. P., Ashkenazi A. Control of apoptosis signaling by Apo2 ligand. Recent Prog. Horm. Res., 54: 225-234, 1999.
  9. ↵
    Wiley S. R., Schooley K., Smolak P. J., Din W. S., Huang C. P., Nicholl J. K., Sutherland G. R., Smith T. D., Rauch C., Smith C. A., et al Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity, 3: 673-682, 1995.
    OpenUrlCrossRefPubMed
  10. ↵
    Sheridan J. P., Marsters S. A., Pitti R. M., Gurney A., Skubatch M., Baldwin D., Ramakrishnan L., Gray C. L., Baker K., Wood W. I., Goddard A. D., Godowski P., Ashkenazi A. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science (Wash. DC), 277: 818-821, 1997.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Walczak H., Miller R. E., Ariail K., Gliniak B., Griffith T. S., Kubin M., Chin W., Jones J., Woodward A., Le T., Smith C., Smolak P., Goodwin R. G., Rauch C. T., Schuh J. C., Lynch D. H. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat. Med., 5: 157-163, 1999.
    OpenUrlCrossRefPubMed
  12. ↵
    Bonavida B., Ng C. P., Jazirehi A., Schiller G., Mizutani Y. Selectivity of TRAIL-mediated apoptosis of cancer cells and synergy with drugs: the trail to non-toxic cancer therapeutics. Int. J. Oncol., 15: 793-802, 1999.
    OpenUrlPubMed
  13. ↵
    Gura T. TRAIL kills cancer cells, but not normal cells. Science (Wash. DC), 277: 768 1997.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    French L. E., Tschopp J. The TRAIL to selective tumor death. Nat. Med., 5: 146-147, 1999.
    OpenUrlCrossRefPubMed
  15. ↵
    Pan G., Ni J., Wei Y. F., Yu G., Gentz R., Dixit V. M. An antagonist decoy receptor and a death domain-containing receptor for TRAIL. Science (Wash. DC), 277: 815-818, 1997.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Jeremias I., Herr I., Boehler T., Debatin K. M. TRAIL/Apo-2-ligand-induced apoptosis in human T cells. Eur. J. Immunol., 28: 143-152, 1998.
    OpenUrlCrossRefPubMed
  17. ↵
    Roth W., Isenmann S., Naumann U., Kugler S., Bahr M., Dichgans J., Ashkenazi A., Weller M. Locoregional Apo2L/TRAIL eradicates intracranial human malignant glioma xenografts in athymic mice in the absence of neurotoxicity. Biochem. Biophys. Res. Commun., 265: 479-483, 1999.
    OpenUrlCrossRefPubMed
  18. ↵
    Keane M. M., Ettenberg S. A., Nau M. M., Russell E. K., Lipkowitz S. Chemotherapy augments TRAIL-induced apoptosis in breast cell lines. Cancer Res., 59: 734-741, 1999.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Snell V., Clodi K., Zhao S., Goodwin R., Thomas E. K., Morris S. W., Kadin M. E., Cabanillas F., Andreeff M., Younes A. Activity of TNF-related apoptosis-inducing ligand (TRAIL) in haematological malignancies. Br. J. Haematol., 99: 618-624, 1997.
    OpenUrlCrossRefPubMed
  20. ↵
    Zhang X. D., Franco A., Myers K., Gray C., Nguyen T., Hersey P. Relation of TNF-related apoptosis-inducing ligand (TRAIL) receptor and FLICE-inhibitory protein expression to TRAIL-induced apoptosis of melanoma. Cancer Res., 59: 2747-2753, 1999.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Bretz J. D., Rymaszewski M., Arscott P. L., Myc A., Ain K. B., Thompson N. W., Baker J. R., Jr. TRAIL death pathway expression and induction in thyroid follicular cells. J. Biol. Chem., 274: 23627-23632, 1999.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Mizutani Y., Yoshida O., Miki T., Bonavida B. Synergistic cytotoxicity and apoptosis by Apo-2 ligand and Adriamycin against bladder cancer cells. Clin. Cancer Res., 5: 2605-2612, 1999.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Kim K., Fisher M. J., Xu S. Q., el-Deiry W. S. Molecular determinants of response to TRAIL in killing of normal and cancer cells. Clin. Cancer Res., 6: 335-346, 2000.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Irmler M., Thome M., Hahne M., Schneider P., Hofmann K., Steiner V., Bodmer J. L., Schroter M., Burns K., Mattmann C., Rimoldi D., French L. E., Tschopp J. Inhibition of death receptor signals by cellular FLIP. Nature (Lond.), 388: 190-195, 1997.
    OpenUrlCrossRefPubMed
  25. ↵
    Deveraux Q. L., Reed J. C. IAP family proteins: suppressors of apoptosis. Genes Dev., 13: 239-252, 1999.
    OpenUrlFREE Full Text
  26. ↵
    Degli-Esposti M. A., Dougall W. C., Smolak P. J., Waugh J. Y., Smith C. A., Goodwin R. G. The novel receptor TRAIL-R4 induces NF-κB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain. Immunity, 7: 813-820, 1997.
    OpenUrlCrossRefPubMed
  27. ↵
    Schneider P., Thome M., Burns K., Bodmer J. L., Hofmann K., Kataoka T., Holler N., Tschopp J. TRAIL receptors 1 (DR4) and 2 (DR5) signal FADD-dependent apoptosis and activate NF-κB. Immunity, 7: 831-836, 1997.
    OpenUrlCrossRefPubMed
  28. ↵
    Wang C. Y., Mayo M. W., Korneluk R. G., Goeddel D. V., Baldwin A. S., Jr. NF-κB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science (Wash. DC), 281: 1680-1683, 1998.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Goke R., Goke A., Goke B., Chen Y. Regulation of TRAIL-induced apoptosis by transcription factors. Cell. Immunol., 201: 77-82, 2000.
    OpenUrlCrossRefPubMed
  30. ↵
    Keane M. M., Rubinstein Y., Cuello M., Ettenberg S. A., Banerjee P., Nau M. M., Lipkowitz S. NF-κB and TRAIL-mediated apoptosis in breast cancer cell lines. Breast Cancer Res. Treat., 64: 211-219, 2000.
    OpenUrlCrossRefPubMed
  31. ↵
    Salomon D. S., Brandt R., Ciardiello F., Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit. Rev. Oncol. Hematol., 19: 183-232, 1995.
    OpenUrlCrossRefPubMed
  32. ↵
    Tyson F. L., Boyer C. M., Kaufman R., O’Briant K., Cram G., Crews J. R., Soper J. T., Daly L., Fowler W. C., Jr., Haskill J. S., et al Expression and amplification of the HER-2/neu (c-erbB-2) protooncogene in epithelial ovarian tumors and cell lines. Am. J. Obstet. Gynecol., 165: 640-646, 1991.
    OpenUrlPubMed
  33. ↵
    Slamon D. J., Godolphin W., Jones L. A., Holt J. A., Wong S. G., Keith D. E., Levin W. J., Stuart S. G., Udove J., Ullrich A., et al Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science (Wash. DC), 244: 707-712, 1989.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Zhang X., Silva E., Gershenson D., Hung M. C. Amplification and rearrangement of c-erb B proto-oncogenes in cancer of human female genital tract. Oncogene, 4: 985-989, 1989.
    OpenUrlPubMed
  35. ↵
    Berchuck A., Kamel A., Whitaker R., Kerns B., Olt G., Kinney R., Soper J. T., Dodge R., Clarke-Pearson D. L., Marks P., et al Overexpression of HER-2/neu is associated with poor survival in advanced epithelial ovarian cancer. Cancer Res., 50: 4087-4091, 1990.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Ross J. S., Fletcher J. A. The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells, 16: 413-428, 1998.
    OpenUrlCrossRefPubMed
  37. ↵
    Hung M. C., Lau Y. K. Basic science of HER-2/neu: a review. Semin. Oncol., 26: 51-59, 1999.
  38. ↵
    Ignatoski K. M., Maehama T., Markwart S. M., Dixon J. E., Livant D. L., Ethier S. P. ERBB-2 overexpression confers PI 3′ kinase-dependent invasion capacity on human mammary epithelial cells. Br. J. Cancer, 82: 666-674, 2000.
    OpenUrlCrossRefPubMed
  39. ↵
    Tzahar E., Yarden Y. The ErbB-2/HER2 oncogenic receptor of adenocarcinomas: from orphanhood to multiple stromal ligands. Biochim. Biophys. Acta, 1377: M25-M37, 1998.
    OpenUrlPubMed
  40. ↵
    Liu W., Li J., Roth R. A. Heregulin regulation of Akt/protein kinase B in breast cancer cells. Biochem. Biophys. Res. Commun., 261: 897-903, 1999.
    OpenUrlCrossRefPubMed
  41. ↵
    Tari A. M., Lopez-Berestein G. Serum predominantly activates MAPK and akt kinases in EGFR- and ErbB2-over-expressing cells, respectively. Int. J. Cancer, 86: 295-297, 2000.
    OpenUrlCrossRefPubMed
  42. ↵
    Sliwkowski M. X., Lofgren J. A., Lewis G. D., Hotaling T. E., Fendly B. M., Fox J. A. Nonclinical studies addressing the mechanism of action of trastuzumab (Herceptin). Semin. Oncol., 26: 60-70, 1999.
  43. ↵
    Baselga J., Tripathy D., Mendelsohn J., Baughman S., Benz C. C., Dantis L., Sklarin N. T., Seidman A. D., Hudis C. A., Moore J., Rosen P. P., Twaddell T., Henderson I. C., Norton L. Phase II study of weekly intravenous trastuzumab (Herceptin) in patients with HER2/neu-overexpressing metastatic breast cancer. Semin. Oncol., 26: 78-83, 1999.
    OpenUrlPubMed
  44. ↵
    Pegram M. D., Slamon D. J. Combination therapy with trastuzumab (Herceptin) and cisplatin for chemoresistant metastatic breast cancer: evidence for receptor-enhanced chemosensitivity. Semin. Oncol., 26: 89-95, 1999.
    OpenUrlPubMed
  45. ↵
    Baselga J., Norton L., Albanell J., Kim Y. M., Mendelsohn J. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu-overexpressing human breast cancer xenografts. Cancer Res., 58: 2825-2831, 1998.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Klapper L. N., Waterman H., Sela M., Yarden Y. Tumor-inhibitory antibodies to HER-2/ErbB-2 may act by recruiting c-Cbl and enhancing ubiquitination of HER-2. Cancer Res., 60: 3384-3388, 2000.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Webb J. L. Effects of more than one inhibitor 1st ed. Webb J. L. eds. . Enzymes and Metabolic Inhibitors, : 487-512, Academic Press New York 1963.
  48. ↵
    Chou T. C., Motzer R. J., Tong Y., Bosl G. J. Computerized quantitation of synergism and antagonism of Taxol, topotecan, and cisplatin against human teratocarcinoma cell growth: a rational approach to clinical protocol design. J. Natl. Cancer Inst. (Bethesda), 86: 1517-1524, 1994.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Lewis G. D., Figari I., Fendly B., Wong W. L., Carter P., Gorman C., Shepard H. M. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol. Immunother., 37: 255-263, 1993.
    OpenUrlCrossRefPubMed
  50. ↵
    Harwerth I. M., Wels W., Marte B. M., Hynes N. E. Monoclonal antibodies against the extracellular domain of the erbB-2 receptor function as partial ligand agonists. J. Biol. Chem., 267: 15160-15167, 1992.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Shawver L. K., Mann E., Elliger S. S., Dugger T. C., Arteaga C. L. Ligand-like effects induced by anti-c-erbB-2 antibodies do not correlate with and are not required for growth inhibition of human carcinoma cells. Cancer Res., 54: 1367-1373, 1994.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Hakem R., Hakem A., Duncan G. S., Henderson J. T., Woo M., Soengas M. S., Elia A., de la Pompa J. L., Kagi D., Khoo W., Potter J., Yoshida R., Kaufman S. A., Lowe S. W., Penninger J. M., Mak T. W. Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell, 94: 339-352, 1998.
    OpenUrlCrossRefPubMed
  53. ↵
    Roh H., Pippin J., Drebin J. A. Down-regulation of HER2/neu expression induces apoptosis in human cancer cells that overexpress HER2/neu. Cancer Res., 60: 560-565, 2000.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Roh H., Hirose C. B., Boswell C. B., Pippin J. A., Drebin J. A. Synergistic antitumor effects of HER2/neu antisense oligodeoxynucleotides and conventional chemotherapeutic agents. Surgery, 126: 413-421, 1999.
    OpenUrlPubMed
  55. ↵
    Marte B. M., Graus-Porta D., Jeschke M., Fabbro D., Hynes N. E., Taverna D. NDF/heregulin activates MAP kinase and p70/p85 S6 kinase during proliferation or differentiation of mammary epithelial cells. Oncogene, 10: 167-175, 1995.
    OpenUrlPubMed
  56. ↵
    Kandel E. S., Hay N. The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell Res., 253: 210-229, 1999.
    OpenUrlCrossRefPubMed
  57. ↵
    Kumar R., Shepard H. M., Mendelsohn J. Regulation of phosphorylation of the c-erbB-2/HER2 gene product by a monoclonal antibody and serum growth factor(s) in human mammary carcinoma cells. Mol. Cell. Biol., 11: 979-986, 1991.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Roh H., Pippin J., Boswell C., Drebin J. A. Antisense oligonucleotides specific for the HER2/neu oncogene inhibit the growth of human breast carcinoma cells that overexpress HER2/neu. J. Surg. Res., 77: 85-90, 1998.
    OpenUrlCrossRefPubMed
  59. ↵
    Downward J. Mechanisms and consequences of activation of protein kinase B/Akt. Curr. Opin. Cell Biol., 10: 262-267, 1998.
    OpenUrlCrossRefPubMed
  60. ↵
    Fujita E., Jinbo A., Matuzaki H., Konishi H., Kikkawa U., Momoi T. Akt phosphorylation site found in human caspase-9 is absent in mouse caspase-9. Biochem. Biophys. Res. Commun., 264: 550-555, 1999.
    OpenUrlCrossRefPubMed
  61. ↵
    Kops G. J., Burgering B. M. Forkhead transcription factors: new insights into protein kinase B (c- akt) signaling. J. Mol. Med., 77: 656-665, 1999.
    OpenUrlCrossRefPubMed
  62. ↵
    Kang S. S., Kwon T., Kwon D. Y., Do S. I. Akt protein kinase enhances human telomerase activity through phosphorylation of telomerase reverse transcriptase subunit. J. Biol. Chem., 274: 13085-13090, 1999.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    Brunet A., Bonni A., Zigmond M. J., Lin M. Z., Juo P., Hu L. S., Anderson M. J., Arden K. C., Blenis J., Greenberg M. E. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell, 96: 857-868, 1999.
    OpenUrlCrossRefPubMed
  64. ↵
    Zhou B. P., Hu M. C., Miller S. A., Yu Z., Xia W., Lin S. Y., Hung M. C. HER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-κB pathway. J. Biol. Chem., 275: 8027-8031, 2000.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    Hudziak R. M., Lewis G. D., Winget M., Fendly B. M., Shepard H. M., Ullrich A. p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol. Cell. Biol., 9: 1165-1172, 1989.
    OpenUrlAbstract/FREE Full Text
  66. ↵
    Feldman A. M., Lorell B. H., Reis S. E. Trastuzumab in the treatment of metastatic breast cancer: anticancer therapy versus cardiotoxicity. Circulation, 102: 272-274, 2000.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Jo M., Kim T. H., Seol D. W., Esplen J. E., Dorko K., Billiar T. R., Strom S. C. Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand. Nat. Med., 6: 564-567, 2000.
    OpenUrlCrossRefPubMed
  68. ↵
    Tokuda Y., Watanabe T., Omuro Y., Ando M., Katsumata N., Okumura A., Ohta M., Fujii H., Sasaki Y., Niwa T., Tajima T. Dose escalation and pharmacokinetic study of a humanized anti-HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. Br. J. Cancer, 81: 1419-1425, 1999.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 61 (12)
June 2001
Volume 61, Issue 12
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Down-Regulation of the erbB-2 Receptor by Trastuzumab (Herceptin) Enhances Tumor Necrosis Factor-related Apoptosis-inducing Ligand-mediated Apoptosis in Breast and Ovarian Cancer Cell Lines that Overexpress erbB-2
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Down-Regulation of the erbB-2 Receptor by Trastuzumab (Herceptin) Enhances Tumor Necrosis Factor-related Apoptosis-inducing Ligand-mediated Apoptosis in Breast and Ovarian Cancer Cell Lines that Overexpress erbB-2
Mauricio Cuello, Seth A. Ettenberg, Amy S. Clark, Maccon M. Keane, Reuben H. Posner, Marion M. Nau, Phillip A. Dennis and Stan Lipkowitz
Cancer Res June 15 2001 (61) (12) 4892-4900;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Down-Regulation of the erbB-2 Receptor by Trastuzumab (Herceptin) Enhances Tumor Necrosis Factor-related Apoptosis-inducing Ligand-mediated Apoptosis in Breast and Ovarian Cancer Cell Lines that Overexpress erbB-2
Mauricio Cuello, Seth A. Ettenberg, Amy S. Clark, Maccon M. Keane, Reuben H. Posner, Marion M. Nau, Phillip A. Dennis and Stan Lipkowitz
Cancer Res June 15 2001 (61) (12) 4892-4900;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Abstract 6119: RNAi rat models for drug discovery
  • Abstract 3834: Histone methyltransferase SET8 is regulated by miR-192/-215 and induces oncogene-induced senescence via p53-dependent DNA damage in human gastric carcinoma cells
  • Abstract 3788: CircHMGCS1 interacts with RNA binding protein HuR and maintains stem-like cells in gliomas
Show more Tumor Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement