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
  • 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
  • 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

Advances in Brief

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

R. Todd Reilly, Jean-Pascal H. Machiels, Leisha A. Emens, Anne M. Ercolini, Francesca I. Okoye, Rachel Y. Lei, Diane Weintraub and Elizabeth M. Jaffee
R. Todd Reilly
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jean-Pascal H. Machiels
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Leisha A. Emens
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anne M. Ercolini
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesca I. Okoye
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rachel Y. Lei
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Diane Weintraub
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elizabeth M. Jaffee
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published February 2001
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

HER-2/neu (neu) transgenic mice (neu-N mice), which express the nontransforming rat proto-oncogene, demonstrate immunological tolerance to neu that is similar to what is encountered in patients with neu-expressing breast cancer. We have shown previously that a significant increase in neu-specific T cells, but no induction of neu-specific antibody, is seen after neu-specific vaccination in neu-N mice. In contrast, a significant induction of both neu-specific T-cell and antibody responses is found in nontoleragenic FVB/N mice after vaccination. These mice are fully protected from a s.c. challenge with NT cells, a mammary tumor cell line derived from a spontaneous tumor that arose in a neu-N mouse, whereas neu-N mice are not. In this study, we demonstrate that CD4+ T cell-depleted FVB/N mice show no induction of neu-specific IgG after vaccination and are unable to reject an NT challenge (0 of 10 mice were tumor free). Conversely, the depletion of natural killer cells has no effect on vaccine-mediated tumor rejection (100% of mice were tumor free). In CD8+ T cell-depleted animals, where vaccine-induced neu-specific IgG titers were normal, NT growth was delayed, but only 10% of mice remained tumor free, demonstrating that neu-specific IgG alone is insufficient for protection from NT challenge. To directly assess the necessity for the combination of neu-specific cellular and humoral immune responses, severe combined immunodeficient mice were given an adoptive transfer of CTLs plus IgG derived from FVB/N mice. Animals that were given CTLs that recognized an irrelevant antigen plus neu-specific IgG developed tumors at a rate similar to CD8+ T cell-depleted FVB/N mice. Animals receiving an adoptive transfer of neu-specific CTLs plus control IgG derived from naive FVB/N mice were only partially protected from NT challenge (50% of animals were tumor free). However, only animals receiving the combination of neu-specific CTLs and neu-specific IgG were fully protected from NT challenge (100% of animals were tumor free). These studies specifically define the immunological requirements for the eradication of neu-expressing tumors in this model system, demonstrating that both cellular and humoral neu-specific responses are necessary for protection from an NT challenge. These data suggest that vaccines optimized to induce maximal T- and B-cell immunity to neu, and possibly to similar putative tumor-rejection antigens, may lead to more potent in vivo antitumor immunity.

Introduction

Current vaccine strategies for the treatment of solid tumors tend to focus on the cellular arm of the immune response. However, the success of passive immunotherapy through the administration of monoclonal antibodies that target neu 3 (1) , CD19 (2) , or the epidermal growth factor receptor (3) has generated renewed interest in the application of humoral immunity in tumor eradication. Trastuzumab, a recombinant humanized monoclonal antibody to neu administered as a single agent or in combination with chemotherapy, produces durable objective responses in women with neu-overexpressing breast cancer (4) . Similarly, passive immunotherapy with monoclonal antibodies against neu was shown to have a dramatic effect on spontaneous tumor development in transgenic mice expressing rat neu (5) . We have documented the existence of immunological tolerance to neu in these mice similar to what is observed in patients with neu-expressing breast cancers (6) . Although neu-N mice are capable of generating CTLs against neu after neu-specific vaccination, there is little or no vaccine-mediated induction of neu-specific IgG in mice vaccinated after 8 weeks of age (6) . Furthermore, although the growth of neu-expressing transplantable tumors in vaccinated neu-N mice is significantly delayed relative to control animals, tumor growth is not prevented completely (6) . This is in stark contrast to what is seen in the absence of tolerance. In nontransgenic FVB/N mice, a significant induction of both neu-specific CTLs and neu-specific IgG is seen, and mice are completely protected from a transplantable tumor challenge (6) . In this study, data are presented demonstrating the induction of neu-specific humoral and cellular immunity in FVB/N mice by active immunization. Furthermore, the combination of neu-specific humoral and cellular immune responses fully protects from a neu-expressing tumor challenge, where the absence of either the cellular or humoral arm leads to incomplete protection. These data have important implications for the development of vaccines that induce immunity against antigens that are targets of both B- and T-cell responses.

Materials and Methods

Mice.

neu-N mice, 8–10 weeks of age, which express the normal rat proto-oncogene (line N#202; Ref. 7 ), FVB/N mice (National Cancer Institute, Bethesda, MD), and BALB/c SCID mice (National Cancer Institute) were used. All experiments were performed in accordance with protocols approved by the Animal Care and Use Committee of the Johns Hopkins University School of Medicine.

Cell Lines and Media.

The neu-expressing mouse mammary tumor line, NT, was described previously (6) . The 3T3wt cell line (American Type Culture Collection, Rockville, MD), composed of NIH-3T3 cells, and the 3T3-neu cell line (CRL-1915; American Type Culture Collection), composed of NIH-3T3 cells expressing rat neu (8) , were maintained as described (6) . NIH-3T3 cells transfected with a plasmid encoding HA were maintained under selection with 1 mg/ml geneticin (Life Technologies, Inc., Grand Island, NY). HA expression was verified by fluorescence-activated cell sorter analysis (data not shown). The 3T3wt and 3T3-neu lines were genetically modified to express mGM-CSF by retroviral transduction as described (6) .

T-Cell Lines.

The neu-specific CD8+ T-cell line Fneu-CTL was established from an FVB/N mouse after neu-specific plasmid DNA vaccination, followed by NT challenge. This animal was tumor free for> 100 days, after which time the animal was sacrificed and splenectomized. Splenocytes were maintained in culture in RPMI 1640 (Life Technologies, Inc.) supplemented with 10% fetal bovine serum (Hyclone, Logan, UT), 0.5% l-glutamate, and 1% penicillin/streptomycin (JRH Biosciences, Lenexa, KS), and 0.1% 2-mercaptoethanol (Sigma Chemical Co., St. Louis, MO) at 37°C and 5% CO2. Fneu-CTL cells have been in culture for> 24 months and are stimulated every 9 days by coculture with mitomycin-treated 3T3-neu cells and irradiated syngeneic splenocytes. The Fneu-CTL line is 100% CD8+, expresses a single T-cell receptor β-chain variable region (Vβ4) as determined by flow cytometry, and shows a high degree of specificity for neu-expressing cell lines as determined by chromium release assay (data not shown). Fneu-CTL cells were suspended in sterile HBSS (Life Technologies, Inc.) at a concentration of 4 × 107 cells/ml for adoptive transfer. Control CD8+ T cells specific for HA (FHA-CTL) were similarly obtained from FVB/N mice after vaccination with 3 × 107 plaque-forming units of HA recombinant vaccinia virus. The FHA-CTL line has been maintained in vitro for ∼1 year and is stimulated as described above for Fneu-CTL except with NIH-3T3 cells transfected with a plasmid encoding HA. These cells are specific for the HA protein and do not lyse neu-expressing cell lines (data not shown).

Isolation of Neu-specific IgG.

Female FVB/N mice, 8 weeks of age, were primed with a s.c. injection of 5 × 106 neu-expressing NT cells. After 14 days, animals were sacrificed, and blood was obtained by cardiac puncture, pooled, and allowed to coagulate. The serum was then pooled, and the total IgG was obtained by ammonium sulfate precipitation and dialyzed against PBS (Life Technologies, Inc.). neu-specific immunoreactivity of the total IgG was verified by flow cytometry as described previously (6) . Control IgG was obtained from naive FVB/N mice. Protein concentrations were determined using the Lowry Assay (Sigma) according to the manufacturer’s directions. Total IgG samples were adjusted to a final concentration of 10 mg/ml using HBSS prior to injection.

For quantitation of neu-specific serum IgG, serum samples were obtained by tail bleed 1 day prior to tumor challenge (neu-N and FVB/N mice) or 1 week after the transfer of neu-specific serum (SCID mice) and neu-specific IgG titers determined as described (6) .

Whole-Cell Vaccinations.

Vaccinations with 3T3/GM cells, composed of 3T3wt cells producing mGM-CSF, or 3T3-neu/GM cells, composed of 3T3-neu cells producing mGM-CSF, were performed as described (6) .

Chromium-Release Assays.

Animals were vaccinated with 3T3-neu/GM or 3T3/GM and CTL prepared as described (6) . Lytic function was determined against 3T3wt, 3T3-neu, and NT cells in a 4-h 51Cr-release assay. The percentage of neu-specific lysis was determined by the following formula: % neu-specific lysis = (% lysis against 3T3-neu targets) − (% lysis against 3T3wt targets).

Depletion Studies.

The depletion of CD4+ and CD8+ T-cell subsets and NK cells was accomplished by i.p. injection of 500 μg of GK1.5 (anti-CD4) or 2.43 (anti-CD8) antibody or pk136 (anti-NK) antibody, respectively, as described (6) .

Tumor Challenge Experiments.

Tumor challenge consisted of 5 × 10 4 (for neu-N mice), 1 × 106 (for SCID mice), or 5 × 106 (for FVB/N mice) NT cells s.c. in the right hind limb 14 days after receiving a whole-cell vaccine (neu-N and FVB/N mice) or 1 day prior to adoptive transfer (SCID mice). Animals were monitored twice weekly for the development of palpable (>5 mm in diameter) tumors. Animals were sacrificed before tumors reached a diameter >12 mm.

Adoptive Transfer Experiments.

One day after s.c. tumor challenge, female SCID mice, 8 weeks of age, received 2 × 107 T cells via tail vein injection and/or passive infusion of 1 mg of IgG given i.p. T cells were maintained in vivo by daily i.p. injections of 20,000 international units of recombinant human IL-2 (Chiron, Emeryville, CA). IgG injections were given weekly throughout the experiment. Animals were monitored as described above.

Statistical Analyses.

Statistical analyses were performed using the Statview software program (SAS Institute Inc., Cary, NC). Kaplan-Meier nonparametric regression analyses for tumor challenge experiments were performed, and significance was determined using the Mantel-Cox log-rank test.

Results and Discussion

We previously described the existence of tolerance to neu in neu-N mice (6) . One measure of neu-specific tolerance in neu-N mice is the absence of an inducible neu-specific humoral response above low baseline levels of neu-specific IgG (6) . In addition, splenocytes from neu-N mice taken 14 days after neu-specific vaccination, although capable of neu-specific lysis of 3T3-neu cell targets (6) , cannot lyse NT cells in a 4-h 51Cr-release assay (data not shown). The net result of this is incomplete protection from tumor challenge and partial prevention of spontaneous tumor formation in vaccinated animals. In contrast, FVB/N mice, in which tolerance to the rat neu protein does not exist, generate robust neu-specific antibody and CTL responses to vaccination and are fully protected from NT challenge (6) . In this model of neu-expressing tumor eradication, successful antitumor immunity appears to be associated with the induction of both neu-specific humoral and cellular immunity in nontolerized mice. Therefore, we used the nontoleragenic FVB/N mice to define the immunological requirements for the successful rejection of neu-expressing tumors.

To determine the relative importance of the various T-cell subsets in the antitumor response, FVB/N mice were depleted of CD4+ T cells, CD8+ T cells, or NK cells by antibody injection (GK1.5, 2.43, and pk136, respectively). The animals were then vaccinated, challenged 2 weeks later with NT cells, and monitored for tumor development. In addition, serum samples were obtained prior to tumor challenge, and neu-specific IgG titers were determined. These data, summarized in Table 1 ⇓ , demonstrate an absolute requirement for both CD4+ T cells as well as CD8+ T cells in mediating tumor-free survival. The results confirm recently reported data demonstrating a similar CD4+ and CD8+ T-cell dependence for protection from an NT challenge in neu-N mice (6) . The tumor-free survival of CD4-depleted mice is essentially indistinguishable from that of immunocompetent mice receiving a mock vaccination (P = 0.53 versus no deplete, 3T3/GM). In the absence of CD4+ T-cell help, these animals would be incapable of generating either a T-cell or B-cell effector response; the lack of neu-specific B-cell effectors is confirmed by the absence of a neu-specific IgG response in these animals. Mice depleted of CD8+ T cells, where CD4+ T-cell help is still in place, develop neu-specific IgG at titers that are identical to that obtained in undepleted animals receiving a neu-specific vaccination (no deplete, 3T3-neu/GM). Additionally, these mice demonstrate a significant delay in the appearance of palpable tumors relative to the mock vaccine controls (P < 0.001 versus no deplete, 3T3/GM). However, only 10% of the CD8+-depleted animals remained tumor free beyond day 60 after challenge, whereas all undepleted animals receiving a neu-specific vaccination were tumor free.

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

Effects of lymphocyte subset depletion on tumor-free survival in FVB/N mice

FVB/N mice were depleted of CD4+ T cells, or NK cells as described in “Materials and Methods.” Depletion was verified by flow cytometric analysis of splenocytes (data not shown), and depletion was maintained by twice-weekly injections of the depleting antibody. Animals were then vaccinated with 3T3/GM (control vaccine) or 3T3-neu/GM (neu-specific vaccine), followed 14 days later by s.c. NT challenge. The mice were monitored for the development of palpable tumors (>5 mm diameter). The average tumor-free survival in days ± SD as well as the number of animals tumor free at the completion of the experiment (day 60 after challenge) are reported for each group. In addition, serum samples were taken from each animal 1 day prior to tumor challenge and analyzed for neu-specific IgG as described in “Materials and Methods.” The titer reported is the greatest dilution of serum for which a shift in mean fluorescence intensity of binding to 3T3-neu cells is seen relative to an irrelevant control antibody. The values shown are the average titer ± SD from several (n ≥ 4) from each group. These data are combined from two separate experiments.

The fact that FVB/N mice depleted of CD8+ T cells were not fully protected from NT challenge, despite the presence of normal neu-specific IgG titers, suggested that a neu-specific antibody response, although growth inhibitory, was not sufficient for tumor eradication. To more directly assess the necessity for neu-specific antibody in tumor rejection, we sought to reconstitute the neu-specific immune response of FVB/N mice in a SCID model system. SCID mice were given a s.c. tumor challenge on day 0. Animals were then divided into groups receiving either: (a) Fneu-CTL cells plus daily IL-2; (b) total IgG derived from FVB/N mice primed against rat neu; (c) Fneu-CTL cells plus neu-specific IgG plus IL-2; or (d) total IgG derived from naive FVB/N mice plus FHA-CTL plus IL-2. An in vitro analysis of the lytic ability of the Fneu-CTL line in a 4-h 51Cr-release assay demonstrated excellent specificity for neu (Fig. 1A) ⇓ . A similar study of the lytic ability of the FHA-CTL line showed excellent specificity for HA-expressing cells with no recognition of neu-expressing targets (Fig. 1B) ⇓ . The dose of Fneu-CTL used in these experiments was based on studies in which FVB/N mice received a s.c. NT challenge followed by the adoptive transfer of either 1 × 107, 2 × 107, or 3 × 107 Fneu-CTL. In these experiments, there was no statistical difference in the tumor-free survival of the animals (data not shown). Six days after the first injection of neu-specific IgG, SCID mice had serum antibody titers of ∼150 (data not shown), similar to what we have reported in FVB/N mice after 3T3-neu/GM vaccination (Table 1) ⇓ . The tumor-free survival of these mice is shown in Fig. 2 ⇓ . Consistent with our observations in the FVB/N depletion studies, SCID mice given neu-specific IgG alone showed a significant delay in the appearance of palpable tumors (P < 0.001 relative to IL-2 alone); however, no animals remained tumor free. This study also confirmed the importance of a potent neu-specific CTL response because 50% of animals given neu-specific CD8+ T cells were tumor free beyond the 50-day end point of the experiment. Additional control groups in which animals received either FHA-CTL cells or total IgG derived from naive FVB/N mice were indistinguishable from animals receiving IL-2 alone (data not shown). Furthermore, animals receiving neu-specific T cells plus IgG from naive mice were indistinguishable from animals receiving only neu-specific T cells (data not shown). Most striking, however, was the finding that the combination of neu-specific IgG and neu-specific CTLs gave significantly better protection from tumor challenge than either treatment alone; all animals in the CTL/IgG combination group were tumor free beyond the end point of the experiment. These data demonstrate that, although neu-specific CTLs play a dominant role in tumor rejection, the greatest level of protection is seen when both neu-specific CD8+ T cells and antibodies are present.

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

The Fneu-CTL and FHA-CTL T-cell lines are specific for neu and HA, respectively. Fneu-CTL (A) and FHA-CTL (B) CD8+ T cells were used in a 4-h 51Cr-release assay at the E:T ratios indicated. The percentage of lysis is indicated for incubation with 3T3-wt (□), 3T3-neu (⋄), and 3T3-HA (○; NIH-3T3 cells expressing HA). Data were obtained in triplicate. The mean value is shown.

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

SCID mice require both neu-specific CTLs and neu-specific IgG for tumor eradication. SCID mice were given an s.c. NT challenge and then 2 × 107 neu-specific CD8+ T cells (○), 1 mg/week IgG from neu-primed mice (□), or both (•). Control animals received HA-specific T cells, 1 mg/week IgG from naive mice, and IL-2 (♦). This experiment was repeated twice (n ≥ 5 animals/group/experiment) with similar results. The cumulative data are shown.

There is a large body of evidence in the literature to support the idea that antibodies directed against neu can inhibit the growth of neu-expressing tumors (9, 10, 11) through several mechanisms, including antibody-dependent cell cytotoxicity and the inhibition of signal transduction through neu. Thus, it is likely that the neu-specific antibody response in CD8-depleted mice is mediating the growth-inhibitory effects in these studies. The fact that NK-depleted mice are fully protected from tumor challenge suggests that NK-mediated mechanisms of immunity do not play a significant role in the delayed tumor growth seen in the CD8-depleted group. This does not, however, exclude a role for macrophage- or monocyte-mediated antibody-dependent cell cytotoxicity in addition to complement fixation and/or the direct growth inhibition of NT cells by antibody-mediated blockade of signaling through neu. Clynes et al. (10) reported recently that the in vivo growth-inhibitory activity of trastuzumab, a Food and Drug Administration-approved monoclonal antibody against the human neu gene product, is only partially mediated by the direct interaction of the antibody with surface neu on a neu-overexpressing target tumor cell line. In addition to the antibody-mediated tumor growth inhibition, the data demonstrated a major role for Fc receptor-dependent mechanisms in tumor eradication (10) . Although the data did not exclude a specific role for NK cells in tumor rejection, a dependence on cells expressing both FcγRIIb and FcγRIII (i.e., monocytes and macrophages) was demonstrated. This is consistent with our observation that NK cells are not required for protection from s.c. tumor challenge in vaccinated FVB/N mice (Table 1) ⇓ .

The Fneu-CTL line used in the adoptive transfer experiments described above was derived from an FVB/N mouse and demonstrates significant lysis of neu-expressing targets. However, the neu-specific T-cell repertoire of neu-N mice is functionally distinct from that of FVB/N mice in its lytic ability. 4 Similarly, patients with neu-positive tumors would be expected to express a neu-specific T-cell repertoire that reflects tolerance to this “self” protein. It is possible that under these conditions of more limited neu-specific CTL lytic ability, the presence of significant neu-specific IgG titers is even more important. Whether the effect of the antibody and CTL interaction is synergistic or simply additive remains to be determined.

neu-specific CTL and antibody responses have been found in patients with neu-expressing tumors (12, 13, 14, 15) . In addition, several groups have demonstrated vaccine-induced neu-specific CTL and antibody responses in animal model systems with vaccine-mediated protection from neu-expressing tumor challenge as well as spontaneous tumor formation (16, 17, 18, 19, 20) . However, there has been no evidence to date correlating protective antitumor immunity with both antigen-specific CTL and antibody responses. Here we show that the induction of neu-specific CTL and IgG responses in nontolerized mice after rat neu-specific vaccination is potent enough to fully protect these animals from challenge with a rat neu-expressing tumor line. Significantly, we have also demonstrated that optimal antitumor immunity is achieved only when both neu-specific CTLs and neu-specific IgG are present. Together, these studies suggest that vaccines optimized to induce maximal T-cell and B-cell immunity to neu, and possibly to similar putative tumor rejection antigens, may lead to more potent in vivo antitumor immunity.

Footnotes

  • ↵1 Supported by Department of Defense Grant DAMD17-96-6138 (to E. M. J.), Department of Defense National Cooperative Drug Discovery Group Grant U19CA72108 (to E. M. J.), NIH Grant T32A107247 (to R. T. R.), the AACR-Cancer Research Foundation of America Fellowship in Prevention Research (to R. T. R.), a grant from Belgian Fonds National de la Recherche Scientifique-Televie (to J-P. H. M.), and a grant from Oeuvre Belge du Cancer (to J-P. H. M.).

  • ↵2 To whom requests for reprints should be addressed, at The Johns Hopkins School of Medicine, Bunting-Blaustein Cancer Research Building, Room 4 M07, 1650 Orleans Street, Baltimore, MD 21231. E-mail: ejaffee{at}jhmi.edu

  • ↵3 The abbreviations used are: neu, HER-2/neu; IL, interleukin; SCID, severe combined immunodeficient; NK, natural killer; mGM-CSF, murine granulocyte/macrophage-colony stimulating factor; HA, hemagglutinin.

  • ↵4 E. M. Jaffee, R. T. Reilly, and A. M. Ercolini, unpublished data.

    • ©2001 American Association for Cancer Research.

    References

    1. ↵
      Shak S. Overview of the trastuzumab (Herceptin) anti-HER2 monoclonal antibody clinical program in HER2-overexpressing metastatic breast cancer.. Herceptin Multinational Investigator Study Group. Semin. Oncol., 26: 71-77, 1999.
      OpenUrlPubMed
    2. ↵
      Maloney D. G. Preclinical and Phase I and II trials of rituximab.. Semin. Oncol., 26: 74-78, 1999.
      OpenUrlPubMed
    3. ↵
      Baselga J., Pfister D., Cooper M. R., Cohen R., Burtness B., Bos M., D’Andrea G., Seidman A., Norton L., Gunnett K., Falcey J., Anderson V., Waksal H., Mendelsohn J. Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin.. J. Clin. Oncol., 18: 904-914, 2000.
      OpenUrlAbstract/FREE Full Text
    4. ↵
      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
    5. ↵
      Katsumata M., Okudaira T., Samanta A., Clark D. P., Drebin J. A., Jolicoeur P., Greene M. I. Prevention of breast tumor development in vivo by downregulation of the p185neu receptor.. Nat. Med., 1: 644-648, 1995.
      OpenUrlCrossRefPubMed
    6. ↵
      Reilly R. T., Gottlieb M. B., Ercolini A. M., Machiels J. P., Kane C. E., Okoye F. I., Muller W. J., Dixon K. H., Jaffee E. M. HER-2/neu is a tumor rejection target in tolerized HER-2/neu transgenic mice.. Cancer Res., 60: 3569-3576, 2000.
      OpenUrlAbstract/FREE Full Text
    7. ↵
      Guy C. T., Webster M. A., Schaller M., Parsons T. J., Cardiff R. D., Muller W. J. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease.. Proc. Natl. Acad. Sci. USA, 89: 10578-10582, 1992.
      OpenUrlAbstract/FREE Full Text
    8. ↵
      Drebin J. A., Link V. C., Weinberg R. A., Greene M. I. Inhibition of tumor growth by a monoclonal antibody reactive with an oncogene-encoded tumor antigen.. Proc. Natl. Acad. Sci. USA, 83: 9129-9133, 1986.
      OpenUrlAbstract/FREE Full Text
    9. ↵
      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.
    10. ↵
      Clynes R. A., Towers T. L., Presta L. G., Ravetch J. V. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets.. Nat. Med., 6: 443-446, 2000.
      OpenUrlCrossRefPubMed
    11. ↵
      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
    12. ↵
      Ioannides C. G., Fisk B., Fan D., Biddison W. E., Wharton J. T., O’Brian C. A. Cytotoxic T cells isolated from ovarian malignant ascites recognize a peptide derived from the HER-2/neu proto-oncogene.. Cell. Immunol., 151: 225-234, 1993.
      OpenUrlCrossRefPubMed
    13. ↵
      Disis M. L., Calenoff E., McLaughlin G., Murphy A. E., Chen W., Groner B., Jeschke M., Lydon N., McGlynn E., Livingston R. B., et al Existent T-cell and antibody immunity to HER-2/neu protein in patients with breast cancer.. Cancer Res., 54: 16-20, 1994.
      OpenUrlAbstract/FREE Full Text
    14. ↵
      Fisk B., Blevins T. L., Wharton J. T., Ioannides C. G. Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte lines.. J. Exp. Med., 181: 2109-2117, 1995.
      OpenUrlAbstract/FREE Full Text
    15. ↵
      Disis M. L., Pupa S. M., Gralow J. R., Dittadi R., Menard S., Cheever M. A. High-titer HER-2/neu protein-specific antibody can be detected in patients with early-stage breast cancer.. J. Clin. Oncol., 15: 3363-3367, 1997.
      OpenUrlAbstract/FREE Full Text
    16. ↵
      Amici A., Venanzi F. M., Concetti A. Genetic immunization against neu/erbB2 transgenic breast cancer.. Cancer Immunol. Immunother., 47: 183-190, 1998.
      OpenUrlCrossRefPubMed
    17. ↵
      Cefai D., Morrison B. W., Sckell A., Favre L., Balli M., Leunig M., Gimmi C. D. Targeting HER-2/neu for active-specific immunotherapy in a mouse model of spontaneous breast cancer.. Int. J. Cancer, 83: 393-400, 1999.
      OpenUrlCrossRefPubMed
    18. ↵
      Esserman L. J., Lopez T., Montes R., Bald L. N., Fendly B. M., Campbell M. J. Vaccination with the extracellular domain of p185neu prevents mammary tumor development in neu transgenic mice.. Cancer Immunol. Immunother., 47: 337-342, 1999.
      OpenUrlCrossRefPubMed
    19. ↵
      Amici A., Smorlesi A., Noce G., Santoni G., Cappelletti P., Capparuccia L., Coppari R., Lucciarini R., Petrelli C., Provinciali M. DNA vaccination with full-length or truncated neu induces protective immunity against the development of spontaneous mammary tumors in HER-2/neu transgenic mice.. Gene Ther., 7: 703-706, 2000.
      OpenUrlCrossRefPubMed
    20. ↵
      Dakappagari N. K., Douglas D. B., Triozzi P. L., Stevens V. C., Kaumaya P. T. Prevention of mammary tumors with a chimeric HER-2 B-cell epitope peptide vaccine.. Cancer Res., 60: 3782-3789, 2000.
      OpenUrlAbstract/FREE Full Text
    PreviousNext
    Back to top
    Cancer Research: 61 (3)
    February 2001
    Volume 61, Issue 3
    • Table of Contents

    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.
    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
    (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
    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
    R. Todd Reilly, Jean-Pascal H. Machiels, Leisha A. Emens, Anne M. Ercolini, Francesca I. Okoye, Rachel Y. Lei, Diane Weintraub and Elizabeth M. Jaffee
    Cancer Res February 2 2001 (61) (3) 880-883;

    Citation Manager Formats

    • BibTeX
    • Bookends
    • EasyBib
    • EndNote (tagged)
    • EndNote 8 (xml)
    • Medlars
    • Mendeley
    • Papers
    • RefWorks Tagged
    • Ref Manager
    • RIS
    • Zotero
    Share
    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
    R. Todd Reilly, Jean-Pascal H. Machiels, Leisha A. Emens, Anne M. Ercolini, Francesca I. Okoye, Rachel Y. Lei, Diane Weintraub and Elizabeth M. Jaffee
    Cancer Res February 2 2001 (61) (3) 880-883;
    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 and Discussion
      • Footnotes
      • References
    • Figures & Data
    • Info & Metrics
    • PDF
    Advertisement

    Related Articles

    Cited By...

    More in this TOC Section

    • Down-Regulation of Regulatory Subunit Type 1A of Protein Kinase A Leads to Endocrine and Other Tumors
    • Activating Mutations of the Noonan Syndrome-Associated SHP2/PTPN11 Gene in Human Solid Tumors and Adult Acute Myelogenous Leukemia
    • Recombinant Listeria Vaccines Containing PEST Sequences Are Potent Immune Adjuvants for the Tumor-Associated Antigen Human Papillomavirus-16 E7
    Show more Advances in Brief
    • 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