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
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • 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
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Advances in Brief

Targeting Multiple Her-2 Epitopes with Monoclonal Antibodies Results in Improved Antigrowth Activity of a Human Breast Cancer Cell Line in Vitro and in Vivo

Camelia I. Spiridon, Maria-Ana Ghetie, Jonathan Uhr, Radu Marches, Jia-Ling Li, Guo-Liang Shen and Ellen S. Vitetta
Camelia I. Spiridon
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maria-Ana Ghetie
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jonathan Uhr
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Radu Marches
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jia-Ling Li
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Guo-Liang Shen
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ellen S. Vitetta
Cancer Immunobiology Center, U. T. Southwestern Medical School, Dallas, Texas 75390-8576
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published June 2002
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Her-2 (p185erbB-2) is a transmembrane tyrosine kinase receptor, which is encoded by the Her-2/neu proto-oncogene. Her-2 is overexpressed on 30% of highly malignant breast cancers. Monoclonal antibodies (MAbs) against Her-2 inhibit the growth of Her-2-overexpressing tumor cells and this occurs by a variety of mechanisms. One such MAb, Herceptin (Trastuzumab), has been approved for human use. We have generated a panel of murine anti-Her-2 MAbs against nine different epitopes on the extracellular domain of Her-2 and have evaluated the antitumor activity of three of these MAbs alone and in combination, both in vitro and in vivo. We found that MAbs (against different epitopes) make a highly effective mixture, which was more effective than the individual MAbs in treating s.c. tumor nodules of BT474 cells in SCID mice. In vitro, the MAb mixture was also more effective than the single MAbs in inducing antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity, inhibiting cell growth and inducing apoptosis, and inhibiting the secretion of vascular endothelial growth factor. Taken together, these activities might explain the superior performance of the MAb mixture in vivo.

Introduction

The erbB family of tyrosine kinase receptors includes epidermal growth factor receptor, erbB-2 (Her-2), erbB-3 (Her-3), and erbB-4 (Her-4; Ref. 1 ). These transmembrane receptors are involved in the regulation of various physiological processes, including cell growth, differentiation, cell-cell interactions, and cytokine signaling (2 , 3) . The overexpression of the erbB-2 (Her-2) receptor occurs in various human cancers, including breast, ovary, prostate, gastric, lung, bladder, and kidney carcinomas (4) and is generally associated with an enhanced metastatic potential and a poor prognosis (5) . The levels of Her-2 expression correlate with the degree of breast tumor differentiation, estrogen receptor status, and clinical outcome (6) . Several mechanisms are responsible for the aggressive behavior of Her-2+ tumor cells (7, 8, 9) . These include the up-regulation of growth factor-mediated tumor angiogenesis that has been recently associated with the overexpression of Her-2 (10, 11, 12, 13) . One of these proangiogenic factors, VEGF3 , is down-regulated by antibodies against Her-2, resulting in the inhibition of tumor growth (14 , 15) . Because Her-2 is expressed at lower levels on cells in many normal tissues (16) , it makes an attractive target for immunotherapy.

There has been considerable interest in developing agents that block the signaling function of Her-2, as well as VEGF secretion or activity. Particularly effective are the MAbs raised against Her-2, which inhibit the in vitro and in vivo growth of Her-2-overexpressing tumor cells (17 , 18) . Several anti-Her-2 MAbs recognizing different epitopes on the EC domain of Her-2 have been described, and the epitopes recognized have been mapped to different parts of the molecule (19, 20, 21, 22, 23) . The humanized anti-Her-2 MAb, 4D5 (Herceptin), has been approved by the Food and Drug Administration for human use and has increased the survival of relapsed patients with Her-2-overexpressing breast tumors (24 , 25) .

In this study, we demonstrate that MAbs against several epitopes on Her-2 make highly effective antitumor agents in vivo when used as a mixture. The MAb mixture is superior to individual MAbs in vitro with regard to inhibiting tumor cell growth and VEGF secretion, as well as mediating ADCC and CDC.

Materials and Methods

Cells.

The human breast cancer cell line BT474 (American Type Culture Collection, Manassas, VA) was maintained in monolayer culture in MEM (Life Technologies, Inc., Grand Island, NY) supplemented with 1% vitamins, 2 mm l-glutamine, 0.1 mm nonessential amino acids, 1 mm sodium pyruvate, 1% HEPES buffer (all from Life Technologies, Inc.), and 10% heat-inactivated FCS (Hyclone, Logan, UT). Cultures were grown at 37°C and 5% CO2, and cells were passaged when they were 70% confluent. Before passage, cells were removed from the flasks after a 5-min incubation with trypsin/EDTA (Life Technologies, Inc.) at room temperature and were washed twice in CM.

MAbs.

Mouse anti-Her-2 MAbs were generated in our laboratory by immunizing BALB/c mice s.c. with the EC domain of Her-2.4 Spleen cells from the immunized mice were harvested and fused with the myeloma cell line SP2/0. The hybridomas were subcloned and assayed by ELISA for their ability to secrete immunoglobulin. Immunoglobulin-containing SNs from positive clones were tested by ELISA for reactivity against the EC domain of Her-2. Positive SNs were then tested by indirect fluorescence on a panel of breast cancer cell lines and analyzed on a FACS (Becton Dickinson, Franklin Lakes, NJ). The MAbs were affinity purified on protein G-Sepharose (Pharmacia, Piscataway, NJ) and further characterized.

Characterization of MAbs.

Twelve MAbs recognizing nine different epitopes on Her-2 were characterized for binding to BT474 cells using indirect immunofluorescence assays and for epitope specificity by cross blocking in direct immunofluorescence assays. Indirect immunofluorescence assays were carried out on BT474 cells suspended at 1 × 105/ml in CM and treated with 0.01–1 μg/ml MAbs for 30 min on ice. The excess MAbs were washed out with CM containing 0.05% sodium azide, and the cells were then treated with 2 μl of the secondary FITC-labeled goat antimouse immunoglobulin and analyzed on a FACScan to determine the percentage of cells that are stained and their MFI.

Direct immunofluorescence assays were carried out with FITC-anti-Her-2 MAbs to determine the amount of MAbs needed to self-block and to cross-block. BT474 cells were treated as described above with saturating concentrations of unlabeled anti-Her-2 MAbs, washed twice, and then treated with various amounts of FITC-labeled anti-Her-2 MAbs. The percentage of cross-blocking was determined by comparing the percentage of positive cells treated with unlabeled anti-Her-2 MAbs followed by FITC-anti-Her-2 MAbs versus the percentage of positive cells treated with FITC-anti-Her-2 MAbs alone. Cross-blocking was considered positive if the binding of the FITC-anti-Her-2 MAbs was reduced by at least 50%.

The isotype of each of the anti-Her-2 MAbs was determined using the ISOStrip Mouse Monoclonal Antibody Isotyping kit (Boehringer Manheim, Indianapolis, IN) according to the manufacturer’s directions. All of the MAbs were of the IgG1k isotype.

Of these 12 MAbs, three IgG1 MAbs (HER-50, HER-66, and HER-70), which recognize three different epitopes on the EC domain of Her-2, were selected for in vivo evaluation because of their superior ability to inhibit the growth of the Her-2-overexpressing BT474 cell line in vitro.

Growth of BT474 s.c. Tumors in SCID Mice.

Male CB.17-SCID mice were used. The sera from all of the mice were screened by ELISA for the presence of mouse immunoglobulin. If serum levels exceeded 10 μg/ml, the mice were considered “leaky” and were not used because tumor-take in these mice is less reliable (26) . The mice were then irradiated with 150 cGy and given a single s.c. injection of 4 × 106 BT474 cells/mouse. Tumor nodules were allowed to grow for 10 weeks or until they reached 400 mm3. The diameter of s.c. tumors was measured twice a week using Venier calipers, and the volumes were calculated according to the formula: volume (mm3 ) = length (mm) × width (mm2).

IHC.

s.c. tumors were removed from mice at the end of the experiment after anesthetizing the animals and perfusing them with heparinized saline followed by 4% paraformaldehyde. Mouse tumors were fixed in 10% formalin for 24 h and then embedded in paraffin. Three-μm paraffin sections were prepared at the U. T. Southwestern Pathology Laboratory. Paraffin-embedded tumors were stained with 5 μg/ml affinity-purified polyclonal rabbit antihuman c-erbB-2 antibody (Dako Co., Carpinteria, CA), followed by 2.5 μg/ml biotinylated goat antirabbit IgG (27, 28, 29) . The same sections were also immunostained with 20 μg/ml HER-50, HER-66, or HER-70 MAbs, or the isotype-matched (negative control) anti-CD25 MAb, RFT5, followed by 2.5 μg/ml biotinylated horse antimouse IgG. The endothelial cells were immunostained with 5 μg/ml rat antimouse CD31 (PECAM-1), followed by 2.5 μg/ml biotinylated rabbit antirat IgG. Positive reactions were visualized by horseradish peroxidase-conjugated streptavidin-biotin complex and diaminobenzidine. Sections were counterstained with hematoxylin.

ADCC Assays.

ADCC assays were performed as previously described (30) with some modifications. The peripheral blood mononuclear cells used as human effector cells were separated from the blood of healthy donors on Ficoll-Hypaque density gradients. Cells were washed once in PBS and then incubated with the green fluorescence cytoplasmic dye, CFSE (Molecular Probes, Inc., Eugene, OR), using 25 μl of a 10-μm solution/106 cells for 7 min at 37°C in a water bath. Murine effector cells were obtained from the spleens of BALB/c mice and were stimulated with concanavalin A (2.5 μg/1 × 106 cells) for 24 h. BT474 (target) cells were harvested, washed, and incubated with different dilutions of either a single anti-Her-2 MAb (HER-50, HER-66, and HER-70), the MAb mixture, Herceptin, or isotype-matched murine or human IgGs (controls). After 30 min at 4°C, excess MAbs were washed out with medium containing 1% FCS plus 0.05% sodium azide. Target cells (BT474) were mixed with either murine or human effector cells at various E:T cell ratios (10:1, 50:1, and 100:1) and incubated for 7 h in a 37°C, 5% CO2 incubator. PI was used at 50 μg/ml to stain dead cells before analyzing the cells on a Becton Dickinson FACS.

CDC Assays.

The CDC assay was similar to the ADCC assay with some modifications (30) . For the murine MAbs, mouse serum (Sigma) was used as a source of complement. For Herceptin, human serum isolated from freshly drawn blood was used as one source of complement and mouse serum (Sigma) was used as another. BT474 target cells, treated as described previously, were mixed with either mouse or human sera diluted 1/10, 1/50, or 1/100, and the cells were incubated for 4 h in a 37°C, 5% CO2 incubator. PI was used at 50 μg/ml to stain dead cells before FACS analysis.

Measurement of VEGF in the SNs of BT474 Cells by ELISA.

5 × 105 BT474 tumor cells/well were seeded in 24-well plates (Falcon). Cells were allowed to adhere overnight, and they were then treated with different concentrations of either HER-50, HER-66, HER-70, the MAb mixture, or Herceptin. Cells were removed by centrifugation at different time points, and the SNs were filter sterilized using a 0.22-μm pore size filter. VEGF levels were measured using the commercially available competitive ELISA kit from Cytimmune Sciences, Inc. The amount of VEGF present in the SNs was extrapolated from the VEGF standard curve and expressed in ng/ml. The levels of VEGF that could be detected in this assay ranged from 0.195–200 ng/ml.

Growth Inhibition Assay.

BT474 cells, at 2.5 × 106 cells/ml in 10% MEM, were plated in triplicate (100 μl/well) in 96-well microtiter plates and were allowed to adhere overnight. The cells were then treated with 100 μl of different concentrations of HER-50, HER-66, or HER-70, a combination of all three (mixture), or Herceptin diluted in the same medium. Nine wells of untreated cells were included in each plate as controls. The plates were incubated for 72 h at 37°C in a 5% CO2 incubator and then pulsed for 6 h with 1 μCi/well [3H]thymidine. Plates were harvested using a Skatron cell harvester, and the radioactivity was measured using a liquid scintillation counter (LKB Wallac). Inhibition of growth was determined by plotting the percentage of [3H]thymidine incorporation compared with untreated cells that were taken as 100%. The IC50s (m) concentration, which inhibited [3H]thymidine incorporation by 50%, were compared.

Annexin-V Assay.

This assay measures early events in apoptosis (31) . BT474 cells were plated at 2.5 × 105/ml in 24-well Falcon plates, allowed to adhere overnight, and treated with 100 μg/ml of the individual anti-Her-2 MAbs using amounts determined in preliminary experiments to inhibit the growth of BT474 cells by 50% as determined by [3H]thymidine incorporation. The MAb mixture was used at the same final concentration. Untreated cells were used as negative controls, and sodium azide was used as a positive control. After a 4-h incubation, the cells were harvested by trypsinization and washed and stained with FITC-Annexin-V and PI according to the manufacturer’s instructions. A Becton Dickinson FACS was used to quantify Annexin+ (apoptotic) cells and PI+ (necrotic) cells.

Results

Characterization of the Three Anti-Her-2 MAbs.

We have developed a panel of murine anti-Her-2 MAbs that recognize nine different epitopes on the EC domain of Her-2. All of the MAbs were of the IgG1k isotype as determined using the ISOStrip Mouse Monoclonal Antibody Isotyping kit. The MAbs were tested for their ability to bind to specific epitopes on the EC domain of Her-2, and the epitopes recognized were determined by cross-blocking (32 , 33) .

Each MAb and Herceptin could self-block but none could cross-block, suggesting that they recognized four different epitopes on the Her-2 molecule (Table 1)⇓ . BT474 cells were stained with each of the anti-Her-2 MAbs followed by FITC-labeled goat antimouse immunoglobulin to determine the percentage of positive cells and the density of Her-2 epitopes recognized by the MAbs (MFI). Over 90% of the cells stained positively with HER-50 or HER-66, and >80% of the cells stained positively with HER-70 or Herceptin. However, the epitopes recognized by HER-70 were expressed at the highest density (highest MFI), followed by the epitopes recognized by HER-66 and HER-50. When the relative-binding affinities of all MAbs were determined (by plotting the percentage of positive cells versus the concentration and calculating the concentration required to reach 50% saturation), Herceptin and HER-50 showed the highest relative-binding affinities on BT474 cells. HER-70 had the lowest relative-binding affinity, and HER-66 was intermediary.

View this table:
  • View inline
  • View popup
Table 1

Characterization of the anti-Her-2 MAbs

Growth Inhibition and Apoptotic Effect of the Anti-Her-2 MAbs on BT474 Cells.

We next determined whether the 3 MAbs would inhibit [3H]thymidine incorporation in BT474 cells. The MAbs were incubated for 72 h with BT474 cells, and the levels of [3H]thymidine incorporation in the MAb-treated cells were measured and compared with those of untreated cells to determine the IC50. As shown in Fig. 1⇓ and Table 2⇓ , the IC50 of the MAb mixture was decreased 3–200-fold as compared with any one of the three MAbs alone or to Herceptin. Table 2⇓ summarizes the means ± SD of three experiments.

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

Killing of BT474 cells by the anti-HER-2 MAbs. BT474 cells were treated with varying concentrations of HER-50 (•), HER-66 (○), HER-70 (▾), combination of the three MAbs (▪), or Herceptin (▿) for 72 h, then pulsed with [3H]thymidine for 6 h. Growth inhibition was calculated by comparing [3H]thymidine incorporation in treated versus untreated cells. The difference between the IC50s of the MAb mixture and the individual MAbs is statistically significant with Ps < 0.05. This is one representative experiment of three performed.

View this table:
  • View inline
  • View popup
Table 2

A combination of three anti-Her-2 antibodies induces more significant growth inhibition and apoptosis in vitro

We also determined whether the MAb mixture would induce apoptosis of BT474 cells more effectively than the individual MAbs. Table 2⇓ shows the means ± SD of three experiments. As measured by Annexin-V staining, the MAb mixture induced the highest percentage of apoptotic BT474 cells, i.e., 20–27% higher than that induced by individual MAbs and 34% higher than that induced by Herceptin. Cell death, as determined by trypan blue exclusion and PI staining, was similar for the 3 MAbs and Herceptin and marginally superior for the MAb mixture. Levels of apoptosis and necrosis in the untreated controls were negligible. The positive control, sodium azide, induced significant apoptosis. Because the percentage of apoptotic cells induced by the MAbs surpassed the percentage of necrotic cells, it is possible that apoptosis is the major type of cell death induced by the MAbs.

The Therapeutic Effect of Anti-Her-2 MAbs in SCID Mice with s.c. BT474 Tumors.

We next determined whether HER-50, HER-66, and HER-70 would inhibit the growth of the BT474 tumor in mice. The in vivo model used to evaluate therapy has been described previously (34) ; in this model, there is 100% tumor-take and consistent tumor growth after s.c. injection of BT474 cells into preirradiated male SCID mice. Because the growth of BT474 cells is estrogen independent, (35, 36, 37) male mice could be used. This is important because estrogen can down-regulate Her-2 expression on tumor cells (38 , 39) . Individual variations of tumor nodules were included in the mean values ± SD for the five mice in each treatment group.

To determine the dose of the MAbs required to inhibit the growth of established s.c. BT474 tumors in SCID mice, we first studied the blood levels that could be achieved by injecting different amounts of 125I-HER-66. Doses that gave levels that reached an IC50 in vitro were chosen (data not shown). Biodistribution of 125I-HER-66 was also determined after i.p. injection of 125I-HER-66 into both normal and s.c tumor-bearing SCID mice. We observed that 1.5% of the injected dose was taken up by the s.c. breast tumor nodule as compared with 0.7% by the kidneys and liver, 0.4% by the lungs, and 0.09% by the spleen (data not shown). The MAb persisted at the tumor site for at least 4 days.

Therefore, SCID mice with advanced s.c. BT474 tumors measuring 400 mm3 were then treated with 15 μg/g mouse of the individual anti-Her-2 MAbs, the MAb mixture, or Herceptin. The MAbs were administered as three i.p. injections at 3-week intervals.

As shown in Fig. 2A⇓ , mice treated with the MAb mixture showed continuous regression of tumor nodules starting after the first course of treatment. After the third course, tumor nodules became nonpalpable for a total of 3 weeks. Individual MAbs induced 75–90% reductions in tumor volume after 2 weeks, and these reductions were maintained over the next 4.5 weeks, followed by relapse. Herceptin was inefficient initially, then induced a 40% reduction in tumor volume after the third week and progressively reduced tumor volumes to 60% of their original size in the following 3.5 weeks, followed by relapse. The reasons for this delay are unclear but may be related to poor ADCC/CDC initially, followed by delayed antigrowth activity. Fig. 2B⇓ shows that the MAb mixture also prolonged the survival of SCID/BT474 mice more effectively than either the individual MAbs or Herceptin. Thus, 100% of mice treated with the MAb mixture survived for 7 weeks after the first injection, whereas 40–60% of mice treated with individual MAbs and 5% of the mice treated with Herceptin survived for the same length of time. Untreated mice were sacrificed when tumor burdens exceeded 1000 mm2 volume or 10% of their total body weight.

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

BT474 cells are killed most effectively in vivo by the MAb mixture. A, preirradiated male SCID mice with 400 mm3 s.c. BT474 breast tumors were injected i.p. with three equal doses of 300 μg (15 μg/g mouse) 10 weeks after tumor inoculation on days 0, 24, and 48. The following treatments were administered to groups of five mice: HER-50 (•); HER-66 (○); HER-70 (▾); combination of the three MAbs (▪); Herceptin (▿); and control (injected with saline; □). Tumors were measured twice a week using calipers, and the mean tumor volume (mm3) ± SD for the five mice in each group was plotted. This represents one experiment of three carried out. The difference between the effects of each treatment versus the control, and between the MAb mixture and each individual MAb are significant with Ps < 0.00715 and < 0.05, respectively. B, one hundred percent of mice treated with the combination of three MAbs survived 7 weeks after the first dose injected. A total of 40–60% of mice treated with individual MAbs and 5% of Herceptin-treated mice survived after 7 weeks when the experiment was terminated. Animals were euthanized when tumor burden exceeded 1000 mm3 or 10% of total body weight.

When mice with smaller, 200 mm3 tumors were treated with the MAb mixture, tumors became nonpalpable 2 weeks after a single dose (data not shown). Remissions were sustained for 4 weeks and all of the mice treated with the mixture survived for 7 weeks after treatment. None of the individual MAbs or Herceptin induced 100% inhibition of s.c. tumor growth, nor did they prolong survival of mice as long as the MAb mixture. Histopathological examination indicated that none of the treated or untreated mice had metastases in their lungs, livers, spleens, or kidneys (data not shown). These results confirm earlier findings that metastases can only be detected when s.c. tumors are >800 mm3 (34) . In all treated animals, tumors resumed growing 2–3 weeks after the last treatment. However, the regressions induced by the MAb mixture generally lasted 1 week longer.

These experiments demonstrate that the MAb mixture is more effective in treating larger tumors than either the individual MAbs alone or Herceptin. When smaller tumors were treated, significant growth inhibition effect was achieved by only one dose of the mixture and was more rapid and long lived.

The Effects of MAb Treatment on the Expression of Her-2 Epitopes on Surviving Tumor Cells.

One mechanism by which anti-Her-2 MAbs act is to induce receptor down-modulation (40, 41, 42) . Therefore, we determined whether the tumor cells which grew back after therapy was discontinued, lacked the Her-2 epitopes recognized by the MAbs used for treatment. Tumors were removed from mice at the end of the experiment, fixed in 10% formalin, embedded in paraffin, and immunostained with HER-50, HER-70, or HER-66 MAbs, the isotype-matched anti-CD25 antibody, RFT5 (negative control), or the anti-c-erbB-2 polyclonal antibody (Dako Co.). As shown in Fig. 3⇓ , tumor nodules removed from relapsed mice expressed the same levels of Her-2 as the original tumor. This indicates that tumor cells that grew back either had not down-regulated Her-2 or that down-regulation was reversible. These findings suggest that the relapsed tumors do not lack the targeted epitope, but rather escape killing by the MAbs for other reasons, e.g., inaccessibility, mutations in signaling pathways, and so on.

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

Relapsed tumors show similar expression of the Her-2 epitopes recognized by each MAb used for treatment. IHC was performed on formalin-fixed, paraffin-embedded s.c. tumors. Tumors were removed from untreated and treated mice and immunostained with HER-50, HER-66, and HER-70 versus the rabbit antihuman c-erbB-2 polyclonal antibody and the isotype-matched control anti-CD25 MAb RFT5. Dark brown areas are indicative of Her-2 expression.

The Ability of the Anti-Her-2 MAbs to Mediate ADCC and CDC.

Although the MAb mixture was highly effective at inhibiting cell growth in vitro, it has been documented that in vivo the Fc portion of MAbs can be of major importance in efficacy (43, 44, 45) . Furthermore, the binding of MAbs to three different epitopes on Her-2 might be more efficient in mediating effector functions because of higher concentrations of total antibody bound. This should contribute to a superior performance of the MAb mixture in vivo. To explore this possibility, we compared the effector functions of the individual MAbs to those of the MAb mixture in vitro. As shown in Fig. 4A⇓ , the individual MAbs induced 10–20% lysis of target cells, whereas the MAb mixture induced 30% lysis at an E:T cell ratio of 100:1. Herceptin mediated potent ADCC in the presence of human peripheral blood mononuclear cells at an E:T cell ratio of 100:1, inducing up to 50% lysis of target cells. However, Herceptin mediated only 10% lysis in the presence of murine effector cells (data not shown), at least partially explaining its inferior activity in mice as compared with the murine MAbs. As shown in Fig. 4B⇓ , the MAbs also fixed complement, resulting in dose-dependent lysis of the target cells. At the same dilution of complement, the MAb mixture induced 2-fold more lysis than HER-50 or HER-66 alone and 10% more lysis than Herceptin or HER-70. Herceptin induced only 10% lysis in the presence of mouse serum, again partially explaining why it was less effective in mice. The isotype-matched negative controls did not mediate either ADCC or CDC.

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

The MAb mixture induces ADCC (A) and CDC (B) against BT474 breast cancer cells in vitro. Flow cytometric analysis was carried out on the CFSE-stained effector cells (peripheral blood mononuclear cells or mouse spleen cells) using PI to stain target BT474 cells treated with 10 ng/1 × 104 cells of HER-50 (•), HER-66 (○), HER-70 (▾), a combination (▪), RFT5 (□; IgG1-isotype-matched control), Herceptin (▿), or Human IgG (♦; IgG1-isotype-matched control). A, the E:T cell ratios were 10:1, 50:1, and 100:1. B, target BT474 cells treated with 100 ng/1 × 105 cells of the same MAbs were mixed with mouse serum diluted 1/10-, 1/50-, and 1/100-fold (as source of complement for the CDC assays). Relative to the isotype-matched control, the effect of each treatment is statistically significant with Ps < 0.05. Both figures illustrate one representative experiment of three performed.

The Effect of Anti-Her-2 MAbs Treatment on VEGF Secretion by Tumor Cells in Vitro.

As described by others (15) , treatment with anti-Her-2 MAbs may also exert an antiangiogenic effect in vivo by down-regulating the production of one or more proangiogenic factors produced by the tumor cells. We have documented the presence of newly formed vessels in the s.c. breast tumor nodules by immunostaining tumors with MAbs that react with the endothelial cell marker PECAM-1 (CD31; data not shown). All tumors contain a high density of vessels, which are immunoreactive with the anti-CD31 antibody. These results suggest that angiogenesis is involved in the growth of s.c. BT474 tumors.

Therefore, we used a competitive ELISA assay to determine whether the three anti-Her-2 MAbs could suppress VEGF production by BT474 cells in vitro. We measured VEGF levels in cells treated with either the individual MAbs, the MAb mixture, or Herceptin. As shown in Fig. 5⇓ , untreated BT474 cells secreted ∼50 ng VEGF/ml/5 × 105 cells over 48 h. Increasing concentrations of the anti-Her-2 MAbs induced a dose-dependent decrease in VEGF production. The MAb mixture completely inhibited VEGF production at a concentration of 1 μg/ml. HER-50 and Herceptin induced appreciable but not complete inhibition of VEGF secretion, whereas HER-66 and HER-70 had marginal activity at the same concentration. These results indicate that all of the MAbs block the secretion of VEGF but that the MAb mixture is superior.

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

HER-50, HER-66, HER-70, and the MAb mixture inhibit the secretion of VEGF by BT474 cells in vitro. Cell supernatants were harvested from 5 × 105 BT474 cells treated with various concentrations of HER-50 (•), HER-66 (○), HER-70 (▾), combination of the three MAbs (▪), and Herceptin (▿). Untreated BT474 cells secrete ∼50 ng VEGF/ml/ 5 × 105 cells over 48 h (□). Relative to the untreated control, the effect of each treatment is statistically significant with Ps < 0.001. VEGF level was measured using a competitive ELISA kit. This assay can detect 0.195–200 ng/ml VEGF. This is a representative experiment of three performed.

Discussion

In this study, we have compared the antitumor activity of three new anti-Her-2 MAbs (HER-50, HER-66, and HER-70) alone, as a MAb mixture, and versus Herceptin. Initially, 12 MAbs recognizing nine epitopes on Her-2 were generated and tested for their ability to bind to specific epitopes on the EC domain of Her-2. The three MAbs described in this study were selected based on their potent inhibition of the growth of BT474 cells in vitro. Herceptin is the fully humanized variant of the anti-Her-2 MAb, 4D5, produced by Genentech and was used as a reference MAb. Herceptin has been approved by the Food and Drug Administration for treating women with Her-2-overexpressing breast carcinomas (24 , 25 , 46, 47, 48) . Currently, Herceptin is being used in patients in several combinatorial regimens.

We (49 , 50) and others (51, 52, 53) have previously demonstrated that hypercrosslinking of cell surface molecules on lymphoma cells is important for negative signaling. Hypercrosslinking can be achieved by homodimerizing individual MAbs or by mixing high affinity MAbs directed against different epitopes on the same molecule. Therefore, we determined whether a combination of three anti-Her-2 MAbs that would be predicted to hypercrosslink Her-2 more effectively on breast cancer cells would have more potent cytotoxic activity than the individual MAbs. The major findings to emerge from this study are as follows: (a) as compared with individual MAbs (including Herceptin), in vivo therapy with the MAb mixture induced more rapid tumor regressions, increased the time to relapse, and prolonged the survival of the treated mice; and (b) as compared with individual MAbs, the MAb mixture showed improved Fc-mediated effector functions, improved blocking of VEGF secretion, and improved direct antiproliferative and proapoptotic effects.

Our in vivo results indicate that in SCID mice with 400 mm3 BT474 xenografts, the MAb mixture, as compared with the individual MAbs, induced complete but transient regressions of tumor nodules and also prolonged both survival and the time to relapse. As determined by IHC, BT474 tumor nodules removed from relapsed mice expressed the same levels of Her-2 epitopes recognized by the MAbs used for treatment as the original tumor. Hence, relapses were not due to the lack of targetable epitopes but rather to escape from killing by the MAbs for other reasons, e.g., mutations in signaling pathways, reversible down-regulation of Her-2 epitopes, inaccessibility, and so on. It is possible that cures might have been achieved with additional or larger doses.

To determine why the MAb mixture was superior to the individual MAbs in vivo, we compared their mechanisms of action (54) . In three [3H]thymidine incorporation experiments, the IC50 of the MAb mixture was decreased as compared with the individual MAbs, demonstrating that it had superior antigrowth activity. The mixture induced apoptosis in BT474 cells more effectively than the individual MAbs.

It has been reported previously that anti-Her-2 MAbs can signal tumor cells to undergo growth arrest or cell death by competitive binding to growth factor receptors, inhibition of ligand binding, and suppression of transcription of growth factors (54, 55, 56) . MAbs that target growth factors or cellular growth factor receptors that are required for tumor cell survival mediate cytostatic or cytotoxic effects (57) . Antiepidermal growth factor receptor MAbs have antitumor activity and effectively induce cell cycle arrest or apoptosis in Her-2+ tumor cells (48 , 58 , 59) . Anti-Her-2 MAbs induce down-modulation of Her-2 epitopes and prevent the growth of tumors in vivo (40, 41, 42 ,, 60 , 61) . Anti-Her-2 MAbs can also kill tumor cells indirectly through immunological mechanisms, including ADCC and CDC (62, 63, 64, 65) , and these effects are dependent upon the immunoglobulin isotype of the MAb. With regard to murine MAbs, IgG2a and IgG3 MAbs are the most effective at mediating ADCC and CDC (66 , 67) . Anti-Her-2 MAbs might also inhibit angiogenesis by blocking Her-2-mediated signal transduction pathways that up-regulate the expression of proangiogenic factors such as VEGF (10, 11, 12, 13) .

Regarding the in vitro efficacy of murine anti-Her-2 MAbs, the 4D5 MAb is one of the most potent growth inhibitory anti-Her-2 MAbs (68) . Treatment of breast cancer cells with 4D5 partially blocks Heregulin activation of Her-2-Her-3 complexes, as measured by receptor phosphorylation assays (69) . 4D5 exhibited significant antiproliferative activity in vitro against cell lines that overexpress Her-2 (70) and induced dose-dependent cell cycle arrest with maximal inhibitory activity occurring at MAb concentrations > 1 μg/ml (71) . 4D5 sensitized Her-2-overexpressing cell lines to tumor necrosis factor α(64) and restored adhesion molecules E-cadherin and α2 integrin to normal levels, turning off the metastatic process and malignant progression (72) . The fully humanized variant of 4D5, Herceptin, binds to the EC domain of Her-2 with an affinity (Kd) of 0.1 nmol/liter, which is 3-fold greater than that of 4D5 (73) .

In several models of MAb therapy, MAbs have demonstrated antigrowth activity and the ability to induce apoptosis (57 , 69 , 74, 75, 76, 77) . There is also evidence that the superior antitumor activity of some MAbs in vivo can be attributable to enhanced negative signaling and that signaling can be enhanced by hypercrosslinking using homodimers (49 , 51) or mixture of antibodies (78) . Wolff et al. (51) previously reported that homodimers of a breast tumor-reactive MAb, Chi BR96, were 10-fold more effective both in vitro and in vivo. Homodimers also showed improved antitumor activity as compared with the monomers in nude mice bearing human lung adenocarcinoma xenografts (51) . In this regard, genetically engineered multimeric IgG constructs offer the advantage of increased therapeutic potential. As described previously, homodimeric IgG was 100-fold more potent in mediating CDC and ADCC than monomers (52 , 53) . Homodimers of Rituxan (chimeric anti-CD20 MAb) were superior in signaling G0/G1 arrest and in inducing apoptosis in several B-lymphoma cell lines in vitro. Treatment with homodimers, compared with monomers, also rendered tumor cell lines more sensitive to chemotherapeutic agents and synergized with an anti-CD22-immunotoxin in vitro (49 , 50) . Similar results have been described for a mixture of immunotoxins that target more than one surface antigen, which have demonstrated better in vivo activity in experimental animals than single immunotoxins (79 , 80) .

Although the inhibition of cell growth is often important in vitro, the effector functions of MAbs can also be equally or more important in vivo (62, 63, 64, 65) . In addition to superior apoptotic activity, the MAb mixture induced a 3-fold increase in cell lysis in our in vitro ADCC assays as compared with each of the individual MAbs. Hence, improved ADCC may also be involved in the superior performance of the MAb mixture in vivo.

The inferior activity of Herceptin versus the murine MAbs in vivo is not surprising and in no way predicts that it would have less activity in humans. Thus, the effectiveness of a MAb depends upon whether the Fc portion of the antibody is of the same species as the one being treated (81 , 82) . Hence, the Fc portion of murine MAbs can fix mouse murine complement and interact efficiently with FcRs on mouse immune effector cells. In contrast, the binding of a human IgG1 to FcγRIII on murine natural killer cells, macrophages, or neutrophils, as well as to mouse complement, is relatively weak (81) . Our data confirm these results, indicating that although Herceptin mediated ADCC/CDC works well in vitro with human immune effector cells/serum, it works poorly with murine immune effector cells and serum. Hence, its activity in mice with human tumor xenografts gives a poor estimate of its potency in humans.

With regard to in vivo activity in mice, the antitumor activity of Herceptin relative to a control human IgG1 was compared with that of its murine parent MAb, 4D5 (83) . There was a 10-fold decrease in tumor size using 4D5 at 25 mg/kg and Herceptin at 100 mg/kg. Thus, the murine MAb was a more effective antitumor agent in mice than Herceptin. Our in vivo results also correlate with previously reported data on Herceptin where dose-dependent antitumor activity using doses ranging from 0.1 to 1 mg/kg and a plateau at doses > 1 mg/kg occurred in nude mice bearing BT474 breast carcinoma xenografts (83) . Pegram et al. (71) described dose-dependent antitumor activity with 3–100 mg/kg in athymic mice bearing MCF7 breast carcinoma xenografts. BT474 xenografts are therefore more sensitive to Herceptin treatment than the MCF7 xenografts.

In addition to inducing apoptosis and mediating effector function, anti-Her-2 MAbs could also exert antitumor activity by inhibiting angiogenesis. Angiogenesis is critical for the growth and survival of solid tumors (84 , 85) . VEGF is one of the most important mediators of tumor angiogenesis (86) and is also an important factor required for the growth of BT474 tumors (15) . It has been demonstrated that the treatment of BT474 tumor cells with anti-Her-2 MAbs decreases VEGF production because of the suppression of VEGF gene expression (15) . Suppression of angiogenesis enhanced the activity of anti-Her-2 MAb therapy in vivo (15) . Our results are in agreement with these findings, suggesting that angiogenesis is involved in the growth of s.c. BT474 tumors, and that each of the three murine anti-Her-2 MAbs, the MAb mixture, and/or Herceptin all induced a dose-dependent decrease in VEGF production by BT474 cells in vitro. The MAb mixture was the most effective and completely inhibited VEGF secretion at a concentration of 1 μg/ml. HER-50 and Herceptin induced appreciable but not complete inhibition of VEGF secretion, whereas HER-66 and HER-70 inhibition was marginal at the same concentration. These results indicate that all of the MAbs block the secretion of VEGF but that the MAb mixture is superior.

In summary, the results presented in this study suggest that the murine anti-Her-2 MAb mixture, as compared with individual murine MAbs, has improved antitumor activity against Her-2-overexpressing breast carcinoma cell line BT474 in vivo and in vitro. This superior activity is probably attributable to a combination of factors, including enhanced antiproliferative and antiapoptotic effects, improved Fc-mediated effector functions, and an increased ability to inhibit VEGF secretion. Although it is not possible to make any valid comparisons between humanized Herceptin and our murine MAbs in mice, our results suggest that a mixture of humanized anti-Her-2 MAbs might be superior to Herceptin alone in humans.

Acknowledgments

We thank Lien Li, Y. Chinn, Ming Mei Lui, and Ana Firan for technical assistance and Linda Owens for administrative assistance.

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 Supported by Department of Defense Grant DAMD17-98-1-8640.

  • ↵2 To whom requests for reprints should be addressed, at Cancer Immunobiology Center, U. T. Southwestern Medical School, 6000 Harry Hines Boulevard, Dallas, TX 75390-8576. Phone: (214) 648-1200; Fax: (214) 648-1204.

  • ↵3 The abbreviations used are: VEGF, vascular endothelial growth factor; MAb, monoclonal antibody; ADCC, antibody-dependent cellular cytotoxicity; CDC, complement-dependent cytotoxicity; CM, complete medium; EC, extracellular; SN, supernatant; FACS, fluorescence-activated cell sorter; MFI, mean fluorescence intensity; IHC, immunohistochemistry; PI, propidium iodide.

  • ↵4 J. L. Li, G. L. Shen, and E. S. Vitetta, unpublished observations.

  • Received November 27, 2001.
  • Revision received February 6, 2002.
  • Accepted February 25, 2002.

References

  1. ↵
    Stern D. A. Tyrosine kinase signaling in breast cancer-erbB family receptor tyrosine kinase. Breast Cancer Res., 2: 176-183, 2000.
    OpenUrlCrossRefPubMed
  2. ↵
    Riese D. J., Stern D. F. Specificity within the EGF family/ErbB receptor family signaling network. Bioassays, 20: 41-48, 1997.
    OpenUrl
  3. ↵
    Alroy I., Yarden Y. The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial receptor-ligand interactions. FEBS Lett., 410: 83-86, 1997.
    OpenUrlCrossRefPubMed
  4. ↵
    Slamon D. J., Clark G. M., Wong S. G., Levin W. J., Ullrich A., McGuire W. L. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science (Wash. DC), 235: 177-182, 1987.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Dougall W. C., Qian X., Peterson N. C., Miller M. J., Samanta A., Greene M. I. The neu-oncogene: signal transduction pathways, transformation mechanisms and evolving therapies. Oncogene, 9: 2109-2123, 1994.
    OpenUrlPubMed
  6. ↵
    Révillion F., Bonneterre J., Peyrat J. P. ERBB2 oncogene in human breast cancer and its clinical significance. Eur. J. Cancer, 34: 791-808, 1998.
    OpenUrlCrossRefPubMed
  7. ↵
    Di Fiore P. P., Pierce J. H., Kraus M. H., Segatto O., King C. R., Aaronson S. A. erbB-2 is a potent oncogene when overexpressed in NIH/3T3 cells. Science (Wash. DC), 237: 178-182, 1987.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Hudziak R. M., Schlessinger J., Ullrich A. Increased expression of the putative growth factor receptor p185HER2 causes transformation and tumorigenesis of NIH 3T3 cells. Proc. Natl. Acad. Sci. USA, 84: 7159-7163, 1987.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Hayes D. F., Mesa-Tejada R., Papsidero L., Croghan G., Korzun A., Norton L., Wood W., Strauchen J., Grimes M., Weiss R., Lee H., Thor A., Koerner F., Rice M., Barcos M., Kufe D. W. Prediction of prognosis in primary breast cancer by detection of a high molecular weight mucin-like antigen using MAbs DF3, F36/22, and CU18: a Cancer and Leukemia Group B study. J. Clin. Oncol., 9: 1113-1123, 1991.
    OpenUrlAbstract
  10. ↵
    Kerbel R. S., Viloria-Petit A., Okada F., Rak J. Establishing a link between oncogenes and tumor angiogenesis. Mol. Med., 4: 286-295, 1998.
    OpenUrlPubMed
  11. ↵
    Kerbel R. S., Viloria-Petit A., Rak K. J. Accidental antiangiogenic drugs: antioncogene directed signal transduction inhibitors and conventional chemotherapeutic agents as examples. Eur. J. Cancer, 36: 1248-1257, 2000.
    OpenUrlCrossRefPubMed
  12. ↵
    Rak J., Filmus J., Finkenzeller G., Grugel S., Marme D., Kerbel R. S. Oncogenes as inducers of tumor angiogenesis. Cancer Metastasis Rev., 14: 263-277, 1995.
    OpenUrlCrossRefPubMed
  13. ↵
    Koukourakis M. I., Giatromanolaki A., O’Byrne K. J., Cox J., Karmmer B., Gatter K. C., Harris A. L. bcl-2 and c-erbB-2 proteins are involved in the regulation of VEGF and of tyrosine phosphorylase angiogenic activity in non-small cell lung cancer. Clin. Exp. Metastasis, 17: 545-554, 1999.
    OpenUrlCrossRefPubMed
  14. ↵
    Perrotte P., Matsumoto T., Inoue K., Kuniyasu H., Eve B. Y., Hicklin D. J., Radinsky R., Dinney C. P. N. Antiepidermal growth factor receptor antibody C225 inhibits angiogenesis in human transitional cell carcinoma growing orthotopically in nude mice. Clin. Cancer Res., 5: 257-264, 1999.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Viloria-Petit A. M., Rak J., Hung M. C., Rockwell P., Goldstein N., Fendly B., Kerbel K. S. Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo. Am. J. Pathol., 151: 1523-1530, 1997.
    OpenUrlPubMed
  16. ↵
    Press M. F., Cordon-Cardo C., Slamon D. J. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene, 5: 953-962, 1990.
    OpenUrlPubMed
  17. ↵
    Disis M. L., Cheever M. A. HER-2/neu protein: a target for antigen-specific immunotherapy of human cancer. Adv. Cancer Res., 71: 343-371, 1997.
    OpenUrlCrossRefPubMed
  18. ↵
    Frankel A. E., Ring D. B., Tringale F., Hsieh-Ma S. T. Tissue distribution of breast cancer-associated antigens defined by monoclonal antibodies. J. Biol. Response Modif., 4: 273-286, 1985.
    OpenUrlPubMed
  19. ↵
    Ring D. B., Kassel J. A., Hsieh-Ma S. T., Bjorn M. J., Tringale F., Eaton A. M., Reid S. A., Frankel A. E., Nadji M. Distribution and physical properties of BCA200, a Mr 200,000 glycoprotein selectively associated with human breast cancer. Cancer Res., 49: 3070-3080, 1989.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Ring D. B., Clark R., Saxena A. Identity of BCA200 and c-erbB-2 indicated by reactivity of monoclonal antibodies with recombinant c-erbB-2. Mol. Immunol., 28: 915-917, 1991.
    OpenUrlCrossRefPubMed
  21. ↵
    Xu F. J., Boyer C. M., Bae D. S., Wu S., Greenwald M., O’Briant K., Yu Y. H., Mills G. B., Bast R. C., Jr. The tyrosine kinase activity of the C-erbB-2 gene product (p185) is required for inhibition by anti-p185 antibodies but not for the cytotoxicity of an anti-p185-ricin-A chain immunotoxin. Int. J. Cancer, 59: 242-247, 1994.
    OpenUrlCrossRefPubMed
  22. ↵
    Xu F., Lupu R., Rodriguez G. C., Whitaker R. S., Boente M. P., Berchuck Y., Yu Y., DeSombre K. A., Boyer C. M., Bast R. C. Antibody-induced growth inhibition is mediated through immunochemically and functionally distinct epitopes on the extracellular domain of the c-erbB-2 (HER-2/neu) gene product p185. Int. J. Cancer, 53: 401-408, 1993.
    OpenUrlCrossRefPubMed
  23. ↵
    Rodriguez G. C., Boente M. P., Berchuck A., Whitaker R. S., O’Briant K. C., Xu F., Bast R. C., Jr. The effect of antibodies and immunotoxins reactive with HER-2/neu on growth of ovarian and breast cancer cell lines. Am. J. Obstet. Gynecol., 168: 228-232, 1993.
    OpenUrlPubMed
  24. ↵
    Baselga J., Tripathy D., Mendelsohn J. Phase II study of weekly intravenous recombinant humanized anti-p185 (HER2) monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J. Clin. Oncol., 14: 737-744, 1996.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Baselga J. Clinical trials of Herceptin (trastuzumab). Eur. J. Cancer, 37 (Suppl.1): 18-24, 2001.
    OpenUrl
  26. ↵
    Bosma G. C., Fried M., Custer R. P., Carroll A., Gibson D. M., Bosma M. J. Evidence of functional lymphocytes in some (leaky) SCID mice. J. Exp. Med., 167: 1016-1033, 1988.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Prange W., Schrimacher P. Absence of therapeutical relevant c-erbB2 expression in human hepatocellular carcinoma. Oncol. Rep., 8: 727-730, 2001.
    OpenUrlPubMed
  28. ↵
    Malcolm S., Michael N. L. The role of immunohistochemistry and fluorescence in situ hybridization for HER-2/neu in assessing the prognosis of breast cancer. Semin. Oncol., 26: 108-116, 1999.
    OpenUrl
  29. ↵
    Pegram M. D., Pauletti G., Slamon D. J. HER-2/neu as a predictive marker of response to breast cancer therapy. Breast Cancer Res. Treat., 52: 65-77, 1998.
    OpenUrlCrossRefPubMed
  30. ↵
    Flieger D., Gruber R., Schlimok G., Reiter C., Pantel K., Riethmuller G. A novel nonradioactive cellular cytotoxicity test based on the differential assessment of living and killed target and effector cells. J. Immunol. Methods, 180: 1-13, 1995.
    OpenUrlCrossRefPubMed
  31. ↵
    Vermes I., Haanen C., Steffens-Nakken H., Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein-labelled Annexin V. J. Immunol. Methods, 184: 39-51, 1995.
    OpenUrlCrossRefPubMed
  32. ↵
    Hamann D., Eichler W., Fiebig, VanLier R. A. W. Conformational changes in CD45 upon monoclonal antibody cross-linking. Hybridoma, 15: 11-16, 1996.
    OpenUrlPubMed
  33. ↵
    Cragg M., Zhang L., French R., Glennie M. Analysis of the interaction of monoclonal antibodies with surface IgM on neoplastic B-cells. Br. J. Cancer, 79: 850-857, 1999.
    OpenUrlCrossRefPubMed
  34. ↵
    Clinchy B., Rabinovsky R., Gazdar A., Yefenof E., Gordon B., Vitetta E. S. The growth and metastasis of human, HER-2/neu-overexpressing tumor cell lines in male SCID mice. Breast Cancer Res., : 217-228, 1999.
  35. ↵
    LeRoy X., Escot C., Boriullet J. P., Theillet C., Maudelonde T., Simony-Lafontaine J., Pujol H., Rochefort H. Decrease of c-erbB-2 and c-myc RNA levels in tamoxifen-treated breast cancer. Oncogene, 6: 431-437, 1991.
    OpenUrlPubMed
  36. ↵
    Yarden R. I., Lauber A. H., El-Ashry D., Chrysogelos S. A. Bimodal regulation of epidermal growth factor receptor by estrogen in breast cancer cells. Endocrinology, 137: 2739-2747, 1996.
    OpenUrlCrossRefPubMed
  37. ↵
    van Slooten H-J., Bonsing B. A., Hiller A. J., Colbern G. T., van Dierendonck J. H., Cornelisse C. J., Smith H. S. Outgrowth of BT-474 human breast cancer cells in immune-deficient mice: a new in vivo model for hormone-dependent breast cancer. Br. J. Cancer, 72: 22-30, 1995.
    OpenUrlPubMed
  38. ↵
    Read L. D., Keith D., Slamon D. J., Katzenellenbogen B. S. Hormonal modulation of HER-2/neu proto-oncogene messenger ribonucleic acid and p185 protein expression in human breast cancer cell lines. Cancer Res., 50: 3947-3951, 1990.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Taverna D., Antoniotti S., Maggiora P., Dati C., De Bortoli M., Hynes N. E. erbB-2 expression in estrogen receptor-positive breast tumor cells is regulated by growth-modulatory reagents. Int. J. Cancer, 56: 522-528, 1994.
    OpenUrlCrossRefPubMed
  40. ↵
    Katsumata M., Okudaira T., Samanta A., Clark D. P., Drebin J. A., Jolicoeur P., Greene M. I. Prevention of breast tumour development in vivo by down-regulation of the p185neu receptor. Nat. Med., 1: 644-648, 1995.
    OpenUrlCrossRefPubMed
  41. ↵
    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
  42. ↵
    Molina M. A., Servat J. C., Albanell J., Rojo F., Arribas J., Baselga J. Trastuzumab (Herceptin), a humanized anti-HER2 receptor monoclonal antibody, inhibits basal and activated HER2 ectodomain cleavage in breast cancer cells. Cancer Res., 61: 4744-4749, 2001.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Li M., Wirthmuller U., Ravetch J. V. Reconstitution of human Fc γ RIII cell type specificity in transgenic mice. Exp. Med., 183: 1259-1263, 1996.
    OpenUrl
  44. ↵
    Clynes R. A., Towers T. L., Presta L. G., Ravetch J. V. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat. Med., 6: 443-446, 2000.
    OpenUrlCrossRefPubMed
  45. ↵
    Samuelson A., Towers T. L., Ravetch J. V. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science (Wash. DC), 291: 484-486, 2001.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Cobleigh M., Vogel C., Tripathy D., Robert N. J., Scholl S., Fehrenbacher L., Wolter J. M., Paton V., Shak S., Lieberman G., Slamon D. J. Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J. Clin. Pathol. (Lond.), 37: 2639-2648, 1999.
    OpenUrl
  47. ↵
    Scott A. M., Welt S. Antibody-based immunological therapies. Curr. Opin. Immunol., 9: 717-722, 1997.
    OpenUrlCrossRefPubMed
  48. ↵
    Shepard H. M., Lewis G. D., Sarup J. C., Fendly B. M., Maneval D., Mordenti J., Figari I., Kotts C. E., Palladino M. A., Ullrich A., Slamon D. Monoclonal antibody therapy of human cancer: taking the HER2 proto-oncogene to the clinic. J. Clin. Immunol., 11: 117-127, 1991.
    OpenUrlCrossRefPubMed
  49. ↵
    Ghetie M-A., Podar E. M., Ilgen A., Gordon B. E., Uhr J. W., Vitetta E. S. Homodimerization of tumor-reactive monoclonal antibodies markedly increases their ability to induce growth arrest or apoptosis of tumor cells. Proc. Natl. Acad. Sci. USA, 94: 7509-7514, 1997.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Ghetie M. A., Bright H., Vitetta E. S. Homodimers but not monomers of Rituxan (chimeric anti-CD20) induce apoptosis in human B-lymphoma cells and synergize with a chemotherapeutic agent and an immunotoxin. Blood, 97: 1392-1398, 2001.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Wolff E. A., Schreiber G. J., Cosand W. L., Raff H. V. Monoclonal antibody homodimers: enhanced antitumor activity in nude mice. Cancer Res., 53: 2560-2565, 1993.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Shopes B. A genetically engineered human IgG mutant with enhanced cytolytic activity. J. Immunol., 148: 2918-2922, 1992.
    OpenUrlAbstract
  53. ↵
    Caron P. C., Laird W., Co M. S., Avdalovic N. M., Queen C., Scheinberg D. A. Engineered humanized dimeric forms of IgG are more effective antibodies. J. Exp. Med., 176: 1191-1195, 1992.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Reese D. M., Slamon D. J. HER-2/neu signal transduction in human breast and ovarian cancer. Stem Cells, 158: 1-8, 1997.
    OpenUrl
  55. ↵
    Le X. F., Ratna V., McWatters A., Bae D. S., Mills G. B., Kumar R., Bast R. C., Jr. Differential signaling by an anti-p185HER2 antibody and heregulin. Cancer Res., 60: 3522-3531, 2000.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Yu D., Hung M. C. Overexpression of erbB2 in cancer and ErbB2-targeting strategies. Oncogene, 19: 6115-6121, 2000.
    OpenUrlCrossRefPubMed
  57. ↵
    Baselga J., Mendelson J. Receptor blockade with monoclonal antibodies as anticancer therapy. Pharmacol. Ther., 64: 127-154, 1994.
    OpenUrlCrossRefPubMed
  58. ↵
    Debatin K-M., Krammer P. H. Resistance to APO-1 (CD95) induced apoptosis in T-ALL is determined by a BCL-2 independent antiapoptotic program. Leukemia (Baltimore), 9: 815-821, 1995.
    OpenUrlPubMed
  59. ↵
    Wu X., Fan Z., Masui H., Rosen S., Mendelsohn J. Apoptosis induced by an antiepidermal growth factor receptor monoclonal antibody in a human colorectal carcinoma cell line and its delay by insulin. J. Clin. Investig., 95: 1897-1905, 1995.
    OpenUrlCrossRefPubMed
  60. ↵
    Cuello M., Ettenberg S. A., Clark S. A., Keane M. M., Posner R. H., Nau M. M., Dennis P. A. L. S. 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 erbB2. Cancer Res., 61: 4892-4900, 2001.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Konecny G., Untch M., Arboleda J., Wilson C., Kahlert S., Boettcher B., Felter M., Beryt M., Lude S., Hepp H., Slamon D., Pegram M. HER2/neu and urokinase-type plasminogen activator and its inhibitor in breast cancer. Clin. Cancer Res., 7: 2448-2457, 2001.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    Pegram M., Slamon D. Biological rationale for HER2/neu (c-erbB2) as a target for monoclonal antibody therapy. Semin. Oncol., 27: 13-19, 2000.
    OpenUrl
  63. ↵
    Reilly R. T., Machiels J. P., Emens L. A., Ercolini A. M., Okoye F. L., Lei R. Y., Weintraub D., Jaffee E. M. The collaboration of both humoral and cellular HER-2/neu-targeted immune responses is required for the complete eradication of HER-2/neu-expressing tumors. Cancer Res., 61: 880-883, 2001.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Cooley S., Burns L. J., Repka T., Miller J. S. Natural killer cell cytotoxicity of breast cancer targets is enhanced by two distinct mechanisms of antibody dependent cellular cytotoxicity against LFA-3 and HER2/neu. Exp. Hematol. (Charlottesville Va.), 27: 1533-1541, 1999.
    OpenUrlCrossRefPubMed
  65. ↵
    Watanabe M., Wallace P. K., Keler T., Deo Y. M., Akewanlop C., Hayes D. F. Antibody dependent cellular phagocytosis (ADCP) and antibody dependent cellular cytotoxicity (ADCC) of breast cancer cells mediated by bispecific antibody MDX-210. Breast Cancer Res. Treat., 53: 199-207, 1999.
    OpenUrlCrossRefPubMed
  66. ↵
    Dyer M. J. S., Hale G., Hayhoe F. G. J., Waldmann H. Effects of CAMPATH-1 antibodies in vivo in patients with lymphoid malignancies: influence of antibody isotype. Blood, 73: 1431-1439, 1989.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Lobuglio A. F., Saleh M. N. Advances in monoclonal antibody therapy of cancer. Am. J. Med. Sci., 304: 214-224, 1992.
    OpenUrlCrossRefPubMed
  68. ↵
    Fendly B. M., Winget M., Hudziak R. M., Lipari M. T., Napier M. A., Ullrich A. Characterization of murine monoclonal antibodies reactive to either the human epidermal growth factor receptor or HER2/neu gene product. Cancer Res., 50: 1550-1558, 1990.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    Lewis G. D., Lofgren J. A., Mcmurtrey A. E. Growth regulation of human breast and ovarian tumor cells by heregulin: evidence for the requirement of ErbB2 as a critical component in mediating heregulin responsiveness. Cancer Res, 56: 1457-1465, 1996.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Lewis G. D., Figari I., Fendly B. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol. Immunother., 37: 255-263, 1993.
    OpenUrlCrossRefPubMed
  71. ↵
    Pegram M., Hsu S., Lewis G., Pietras R., Beryt M., Sliwkowski M. X., Coombs D., Bally D., Kabbinavar F., Slamon D. Inhibitory effects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancers. Oncogene, 18: 2241-2251, 1999.
    OpenUrlCrossRefPubMed
  72. ↵
    D’Souza B., Taylor-Papadimitriou J. Overexpression of ERBB2 in human mammary epithelial cells signals inhibition of transcription of the E-cadherin gene. Proc. Natl. Sci. USA, 91: 7202-7206, 1994.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    Carter P., Presta L., Gorman C. M., Ridgway J. B. B., Henner D., Wong W. L. T., Rowland A. M., Kotts C., Carver M. E., Shepard H. M. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc. Natl. Acad. Sci. USA, 89: 4285-4289, 1992.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    Harari D., Yarden Y. Molecular mechanisms underlying ErbB2/HER2 action in breast cancer. Oncogene, 19: 6102-6114, 2000.
    OpenUrlCrossRefPubMed
  75. ↵
    Brennan P. J., Kumagai T., Berezov A., Murali R., Greene M. I. HER2/neu: mechanism of dimerization/oligomerization. Oncogene, 19: 6093-6101, 2000.
    OpenUrlCrossRefPubMed
  76. ↵
    Qian X., O’Rourke D. M., Drebin J., Zhao H., Wang Q., Greene M. I. Identification of p185neu sequences required for monoclonal antibody- or ligand-mediated receptor signal attenuation. DNA Cell Biol., 16: 1395-1405, 1997.
    OpenUrlPubMed
  77. ↵
    Robinson D. R., Wu Y. M., Lin S. F. The protein tyrosine kinase family of the human genome. Oncogene, 19: 5548-5557, 2000.
    OpenUrlCrossRefPubMed
  78. ↵
    Marches R., Racila E., Tucker T. F., Picker L., Mongini P., Hsueh R., Vitetta E. S., Scheuermann R. H., Uhr J. W. Tumor dormancy and cell signalling III: role of hypercrosslinking of IgM and CD40 on the induction of cell cycle arrest and apoptosis in B lymphoma cells. Ther. Immunol., 2: 125-136, 1996.
    OpenUrl
  79. ↵
    Flavell D. J., Boehm D. A., Emery L., Noss A., Ramsay A., Flavell S. U. Therapy of human B-cell lymphoma-bearing SCID mice is more effective with anti-CD19- and anti-CD38-saporin immunotoxins used in combination than with either immunotoxin used alone. Int. J. Cancer, 62: 1-8, 1995.
    OpenUrlPubMed
  80. ↵
    Ghetie M. A., Tucker K., Richardson J., Uhr J. W., Vitetta E. S. The antitumor activity of an anti-CD22 immunotoxin in SCID mice with disseminated Daudi lymphoma is enhanced by either an anti-CD19 antibody or an anti-CD19 immunotoxin. Blood, 80: 2315-2320, 1992.
    OpenUrlAbstract/FREE Full Text
  81. ↵
    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.
    OpenUrlPubMed
  82. ↵
    Pietersz G. A., Wenjun L., Sutton V. R., Burgess J., McKenzie I. F. C., Zola H., Trapani J. A. In vitro and in vivo antitumor activity of a chimeric anti-CD19 antibody. Cancer Immunol. Immunother., 41: 53-60, 1995.
    OpenUrlCrossRefPubMed
  83. ↵
    Pietras R. J., Pegram M. D., Finn R. S., Slamon D. J. Remission of human breast cancer xenografts of therapy with humanized monoclonal antibody to HER-2 receptor and DNA-reactive drugs. Oncogene, 17: 2235-2249, 1998.
    OpenUrlCrossRefPubMed
  84. ↵
    Folkman J. What is the evidence that tumors are angiogenesis dependent?. J. Natl Cancer Inst. (Bethesda), 82: 4-6, 1990.
    OpenUrlCrossRefPubMed
  85. ↵
    Weidner N. Tumor angiogenesis and metastasis correlation in invasive breast carcinoma. N. Engl. J. Med., 324: 1-8, 1991.
    OpenUrlCrossRefPubMed
  86. ↵
    Ferrara N., Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr. Rev., 18: 4-25, 1997.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
June 2002
Volume 8, Issue 6
  • 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 Clinical 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.
Targeting Multiple Her-2 Epitopes with Monoclonal Antibodies Results in Improved Antigrowth Activity of a Human Breast Cancer Cell Line in Vitro and in Vivo
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Targeting Multiple Her-2 Epitopes with Monoclonal Antibodies Results in Improved Antigrowth Activity of a Human Breast Cancer Cell Line in Vitro and in Vivo
Camelia I. Spiridon, Maria-Ana Ghetie, Jonathan Uhr, Radu Marches, Jia-Ling Li, Guo-Liang Shen and Ellen S. Vitetta
Clin Cancer Res June 1 2002 (8) (6) 1720-1730;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Targeting Multiple Her-2 Epitopes with Monoclonal Antibodies Results in Improved Antigrowth Activity of a Human Breast Cancer Cell Line in Vitro and in Vivo
Camelia I. Spiridon, Maria-Ana Ghetie, Jonathan Uhr, Radu Marches, Jia-Ling Li, Guo-Liang Shen and Ellen S. Vitetta
Clin Cancer Res June 1 2002 (8) (6) 1720-1730;
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
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

Advances in Brief

  • Zoledronic Acid Induces Significant and Long-Lasting Modifications of Circulating Angiogenic Factors in Cancer Patients
  • CpG Island Methylator Phenotype Is an Independent Predictor of Survival Benefit from 5-Fluorouracil in Stage III Colorectal Cancer
  • Haptoglobin-α Subunit As Potential Serum Biomarker in Ovarian Cancer
Show more Advances in Brief

Experimental Therapeutics, Preclinical Pharmacology

  • Intermittent Hypoxia Induces Proteasome-Dependent Down-Regulation of Estrogen Receptor α in Human Breast Carcinoma
  • Efficacy and Safety Evaluation of Human Reovirus Type 3 in Immunocompetent Animals
  • Preparation and Biological Evaluation of Copper-64–Labeled Tyr3-Octreotate Using a Cross-Bridged Macrocyclic Chelator
Show more Experimental Therapeutics, Preclinical Pharmacology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

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

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement