2004_breastdis_3-11

Upload: simplerequests

Post on 04-Apr-2018

214 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/30/2019 2004_BreastDis_3-11

    1/11

    Breast Disease 20 (2004) 311 3IOS Press

    Peptide-Based Vaccines in Breast Cancer

    Mary L. Disis, Lupe G. Salazar and Keith L. KnutsonTumor Vaccine Group, Oncology, University of Washington, Seattle, WA, USA

    Abstract. Human tumors are immunogenic and tumor-associated proteins that generate immunity in cancer patients have been

    defined. Many of these proteins are involved in the malignant transformation and play a role in either initiating or maintaining

    the malignant phenotype. Furthermore, due to technical advances in basic immunology over the last decade we have a betterunderstanding of the immune effector cell phenotypes that are potentially involved in tumor eradication and have developed

    methods to quantitate and characterize these immune effectors. Breast cancer is an intriguing model tumor to target with ac-

    tive immunization. Dozens of breast cancer antigens have been defined [1]. Although many patients with breast cancer can

    be rendered free of disease with standard therapy such as surgery, radiation, and chemotherapy, some patients will have their

    disease recur. However, relapse may not occur for many months to years after definitive treatment giving an extended period of

    micrometastatic disease that may be amenable to immune eradication or modulation. Peptide based vaccines are one of the most

    commonly studied vaccine strategies targeting breast cancer.

    Abbreviations: APC: antigen presenting cell; CEA: carcinoembryonic antigen; CTL: cytotoxic T cell; DC: dendritic cell;

    ECD: extracellular domain; GM-CSF: granulocyte macrophage colony stimulating factor; HLA: human leukocyte antigen; IFA:

    incomplete Freunds adjuvant; IFN: interferon; IL: interleukin; KLH: keyhole limpit hemocyanigen; LC: Langerhans cell; MHC:

    major histocompatibility antigen; PBMC: peripheral blood mononuclear cells; TCR: T cell receptor; Th: T helper cell

    THE RATIONALE SUPPORTING PEPTIDE

    BASED VACCINES

    Vaccines have been one of the most successful clin-

    ical interventions for the prevention of human dis-

    ease. Prospective immunization against a variety ofpathogens has resulted in protection from the devel-opment of life-threatening infection. A recent reviewcited data derived from records kept by the Centers of

    Disease Control since 1912 and outlined the decrease

    in infectious disease since vaccines targeting specificpathogens have become widely available [2]. The inci-

    dence of indigenous poliomyelitis has decreased 100%,diphtheria, measles and rubella have decreasedby 99%,

    and whooping cough has had a 97% decrease in inci-dence associated with vaccination. Most cases of vac-

    cine failure can be linked to insufficient immune re-sponses generated after active immunization [2]. The

    Corresponding author: Mary L. Disis, Box 356527, Oncol-ogy, University of Washington, Seattle, WA 98195-6527, USA.Tel.: +1 206 616 1823; Fax: +1 206 685 3128; E-mail: [email protected].

    purpose of a vaccine is to induce an antigen-specific

    immune response that will result in disease prevention.

    The identification of specific tumor antigens has sig-nificantly advanced the field of tumor immunology, in

    particular, the development of breast cancer vaccines.

    Improved understanding of the molecular basis of anti-gen recognition has resulted in the development of ra-

    tionally designed breast cancer antigen-specific vac-cines based on motifs predicted to bind to human class I

    or class II major histocompatibility molecules (MHC).

    The use of synthetic peptides in breast cancer vaccinesoffers practical advantages such as relative ease of con-

    struction and production, chemical stability, and a lack

    of infectious or oncogenic potential. Peptides mayalso allow better manipulation of the immune response

    through the use of epitopes designed for stimulatingparticular subsets of T cells (e.g. helper and cytotoxic

    T cells). In animal models, peptide vaccines are par-

    ticularly effective in generating immune responses toself-proteins [3]. Theoretically, immunization to for-

    eign proteins normally elicits immunity to only a sub-set of potential epitopes, dominant epitopes, whereas

    other potentially immunogenic epitopes, subdominant

    0888-6008/04/$17.00 2004 IOS Press and the authors. All rights reserved

  • 7/30/2019 2004_BreastDis_3-11

    2/11

    4 M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer

    epitopes, are ignored. There is evidence that T cells

    are tolerant to the dominant epitopes of self-proteins,but may respond to the subdominant epitopes [4,5]. As

    many newly defined tumor antigens are self-proteins,

    peptide immunization may play a key role in the ability

    to elicit an immune response to such antigens. Peptide

    based vaccines can be designed to represent subdomi-

    nant epitopes, thus, elicit immunity.

    The development of a rational approach to breast

    cancer vaccination must be based on how the immune

    system is stimulated to respond to breast cancer asso-

    ciated proteins. The goal of many infectious disease

    vaccines is to stimulate a cellular immune response, i.e.

    T cell response that would result in the development

    of long-term memory T cells that would be capable of

    responding to the pathogen when a patient is exposed.

    Generation of T cell immunity would be the goal for a

    cancer vaccine developed to prevent disease in patients

    at high risk for relapse. T cells comprise the major-

    ity of circulating peripheral blood lymphocytes. T cell

    activation depends on recognition of MHC-antigenic

    peptide complexes on the surface of antigen present-

    ing cells (APC). Tumors themselves could present anti-

    genic peptides in MHC molecules expressed on cell

    surface. Vaccines composed of both class I and class II

    peptides derived from breast cancer antigens have been

    studied in human clinical trials [6,7].Peptide-based vaccines have advanced from pre-

    clinical to human clinical studies over the last decade

    and several important issues have been elucidated dur-

    ing this clinical progression. First, investigators have

    developed powerful paradigms to choose peptide(s) for

    immunization and established the pre-clinical evalua-

    tion needed to develop a peptide-based tumor vaccine.

    Secondly, the importance of class II peptides in active

    immunization is being increasingly defined. The gen-

    eration of an endogenous CD4+ T cell response will

    improve the magnitude and maintenance of CD8+ im-

    munity generated with class I binding epitopes. Finally,

    methods have been developed to modify peptides to

    improve the immunogenicity of epitope-specific vac-

    cines. Tumor antigen-specific peptide-based vaccines

    have been an excellent model to test active immuniza-

    tion in breast cancer patients over the last several years.

    CD8+ cytotoxic T cells (CTL) can kill cells express-

    ing an antigen. The action of CD8+ T cells requires

    recognition of antigenic proteins that have been pro-

    cessed and presented as peptide fragments in the con-

    text of MHC class I. Epitopes recognized by CD8+ T

    cells are, in general, peptides of 810 amino acids in

    length anchored at each end within the major groove of

    MHC class I molecules. As a consequence, MHC class

    I molecules exhibit more amino acid sequence speci-ficity based on these anchoring residues, than MHC

    class II molecules which may be more promiscuous in

    the binding of peptides. A potential problem in the de-

    velopment of CTL epitope-based vaccines is the large

    degree of MHC polymorphism and the need for knowl-

    edge of MHC restrictions in the population to be vacci-

    nated. However, it is now known that that MHC class I

    moleculescan be divided into several families or super-

    types based on similar peptide-binding repertoires [8].

    For example, the A2 supertype consists of at least eight

    related molecules and of these, the most frequently ob-

    served are HLA-A*0201, A*0202, A*0203, A*0206,

    and A*6802. Many peptides that bind A*0201 also ex-

    hibit degenerate binding, i.e. binding to multiple alle-

    les, thus, an A2 supertype multi-epitope vaccine could

    be designed to provide broad population coverage [8].

    Initial studies of CTL epitopes were based on the

    elution of naturally processed peptides from MHC

    class I molecules on tumor cells. The identification

    of sequence motifs from eluted peptides, as well as

    crystallographic data of peptide binding to specific

    MHC class I molecules, has allowed predicted bind-

    ing of specific peptides. Recent investigations demon-

    strate that computer algorithms designed for predict-

    ing class I binding can be good predictors of pep-tides that are naturally-processed epitopes of tumor

    antigens. Lu and colleagues used two computer-

    based prediction algorithms that are readily available

    on the Internet; syfpeithi.bmi-heidelberg.com and bi-

    mas.dcrt.nih.gov/molbio/hla bind/hla motif search

    info.html. The algorithms were able to identify three

    candidate peptides of CEA specific for HLA-B7. Af-

    ter constructing the peptides based on motif identifi-

    cation, investigators demonstrated one of the peptides

    generated CTL capable of lysing HLA-matched CEA

    expressing tumors [9]. Similar strategies have been ex-

    tremely useful for identifying class I peptide epitopes

    from a variety of tumor antigens. For example, Rong-

    cun and colleagues identified four HER-2/neu-specific

    HLA-A2.1-restricted CTL epitopes which were able

    to elicit CTL that specifically killed peptide-sensitized

    target cells, and most, importantly, a HER-2/neu-

    transfected cell line and autologous tumor cells. In ad-

    dition, CTL clones specific for particular HER-2/neu

    epitopes were isolated from tumor-specific CTL lines,

    further demonstrating the immunogenicityof these epi-

    topes as candidates for a potential breast cancer vac-

    cine specifically designed to develop a tumor-specific

    CTL response [10]. Thus, peptide based vaccines can

  • 7/30/2019 2004_BreastDis_3-11

    3/11

    M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer 5

    be designed for eliciting CTL immunity and may even

    be able to immunize across multiple class I haplotypes.CD4+ T helper cells (Th) augment the antigen-

    specific immune response via cytokine secretion. Th1

    subsets of CD4+ T cells secrete cytokines such as in-

    terleukin (IL)-2 and interferon (IFN)-that will stimu-

    late the proliferation and activity of CTL. Th2 subsets

    of CD4+ T cells secrete cytokines such as IL-4, -5,

    -6, and -10 that will result in production of high affin-

    ity effective antibody response. The action of CD4+

    T cells requires recognition of antigenic proteins that

    have been processed and presented as peptide frag-

    ments in thecontext of MHC class II. Unlike MHC class

    I molecules, which typically bind peptides of eight to

    ten amino acids in length, MHC class II molecules are

    generally believed to be more permissive in length and

    amino acid sequence of bound peptide [11]. For exam-

    ple, in studies defining the T helper epitopes of tetanus

    toxoid, panels of overlapping peptides were screened

    for their ability to stimulate T cell proliferation in lym-

    phocytes derived from tetanus-immune subjects. Pep-

    tides of greater than 12 amino acids and less than 31

    amino acids and typically in the range of 14 to 16amino

    acids were found to be most efficient in defining rec-

    ognized epitopes [12]. Moreover, T helper peptides

    can be universal or promiscuous and bind to multiple

    alleles. Promiscuous class II helper epitopes have beenidentified for the HER-2/neu [13], NY-ESO [14], and

    human telomerase reverse transcriptase antigens [15],

    three common immunogenic proteins associated with

    breast cancer.

    CD4+ T cells play an important role in the genera-

    tion of an anti-tumor immune response. CD4+ helper

    T cells initiate andmaintainCD8+T cell responses[16,

    17]. Peptide-based cancer vaccines must consider the

    important and potentially critical role of CD4+ T cells.

    Similar to the identification of MHC class I binding

    motifs, computer algorithms have played a major role

    in the identification of MHC class II binding motifs.

    MHC class II antigenic epitopes tend to share a com-

    mon secondary structure, namely amphipathic he-

    lices [18] suggesting antigenic epitopes recognized by

    T cells can be predicted from the primary structure of

    a protein based on predicted secondary structure [19,

    20]. There are now several amphipathicity and hy-

    drophobicity algorithms that have been demonstrated

    to be useful in predicting T cell presented sequences

    within potentially antigenic proteins [18,19,21]. These

    algorithms have been used to identify potential MHC

    class II-bindingepitopes from several tumor-associated

    antigens. In vitro generation of peptide-specific T cells

    from patients with cancer, similar to those used with

    MHC class I peptides, is the endpoint of the identi-fication of epitopes appropriate for use in immuniza-

    tion. This strategy was used to define putative class

    II HER-2/neu binding epitopes for use in breast can-

    cer vaccines [19]. Subsequently, when these peptides

    were tested for binding to HLA-DR molecules in vitro,

    the peptides which were the most immunogenic in vivo

    were also those peptides that had the highest binding

    affinity to HLA-DR [22].

    BREAST CANCER VACCINES COMPOSED OF

    CLASS I PEPTIDES AND DESIGNED TO

    GENERATE A CD8+ CYTOTOXIC T CELL

    (CTL) RESPONSE

    In an initial clinical study, HLA-A2 positive patients

    with metastatic HER-2/neu overexpressing breast,

    ovarian, or colorectal carcinomas were immunized

    with p369-377, a MHC class I peptide defined from

    the HER-2/neu protein sequence, admixed in incom-

    plete Freunds adjuvant (IFA) every three weeks [23].

    Peptide-specific CTL were isolated and expanded from

    the peripheral blood of patients after two or four immu-

    nizations. The CTL could lyse MHC-matched peptide-

    pulsed target cells but could not lyse MHC-matchedtumors expressing the HER-2/neu protein. Even when

    tumors were treated with IFN- to upregulate MHC

    class I, the CTLlines generatedfrom the patients would

    not respond to the peptide presented endogenously on

    tumor cells. A similar study performed with the same

    MHC class I peptide in patients with metastatic breast

    and ovarian cancer demonstrated peptide and tumor-

    specific responses in some patients [24]. Another group

    of investigators, immunizing patients with p369-377,

    used GM-CSF as an adjuvant [6]. GM-CSF is a re-

    cruitment and maturation factor for skin dendritic cells

    (DC), Langerhans cells (LC) and theoretically may al-

    low more efficient presentationof peptide epitopes than

    standard adjuvants such as IFA. Six HLA-A2 patients

    with HER-2/neu-overexpressing cancers received six

    monthly vaccinations with HER-2/neu peptide, p369-

    377, admixed with GM-CSF. The patients had either

    stage III or IV breast or ovarian cancer. Immune re-

    sponses to the p369-377 were examined using an IFN-

    ELIspot assay. Following vaccination, HER-2/neu

    peptide-specific precursors developed to p369-377 in

    just two of four evaluable subjects. The responses were

    short-lived and not detectable at five months after the

    final vaccination. Immunocompetence was evident as

  • 7/30/2019 2004_BreastDis_3-11

    4/11

    6 M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer

    patients had detectable T cell responses to tetanus tox-

    oid and influenza.Another vaccine strategy commonly used in breast

    cancer vaccination is the delivery of MHC class I pep-

    tides loaded on DC and injected subcutaneously or in-

    tradermally. Brossart and colleagues reported a vac-

    cine trial immunizing breast and ovarian cancer patients

    with autologous DC pulsed with MUC-1 or HER-2/neu

    class I binding peptides [25]. In this trial, 10 patients

    were immunized. Half the patients developed CD8+

    T cell precursors to their immunizing peptides. More-

    over, some patients developed new immunity to other

    tumor antigens expressed in their cancers such as CEA

    and MAGE-3, i.e. epitope spreading. Epitope, or de-

    terminant spreading, is a phenomenonfirst described in

    autoimmune disease [26] and represents the generation

    of an immune response to a particular portion of an im-

    munogenic protein and then the natural spread of that

    immunity to other areas of the protein or even to other

    antigens present in the environment. Epitope spreading

    may representthe ability of the initial immuneresponse

    to create a microenvironment at the site of the tumor

    that enhances endogenous local immunity [27].

    These phase I clinical trials demonstrate that patients

    with even advanced stage breast cancer can be immu-

    nized against antigen expressed by their tumors. Al-

    though early trials have focused on only one or two de-fined breast cancer antigens, many class I peptide epi-

    topesare beingdefined for novel immunogenic proteins

    expressed in breast cancer. For example, investigators

    have identified several HLA-A3 binding epitopes of

    mammaglobin-A, a protein highly expressed on about

    80% of breast tumors [28]. Similarly, MHC class I

    binding epitopes have been defined for the near univer-

    sal antigen, survivin [29]. The identification of an ar-

    mamentarium of MHC class I binding peptides derived

    from multiple immunogenic breast cancer proteins will

    lay the foundation of multi-antigen approaches to pre-

    vent breast cancer relapse in antigenically diverse tu-

    mors across a broad patient population.

    BREAST CANCER VACCINES COMPOSED OF

    CLASS II PEPTIDES AND DESIGNED TO

    GENERATE A CD4+ T HELPER CELL (TH)

    RESPONSE

    Potential pitfalls of the use of single MHC class I

    binding peptides are highlighted in the trials described

    above; immunity is short lived and the peptide-specific

    T cells may not recognize endogenously presented

    antigen. A successful vaccine strategy for generating

    peptide-specific CTL capable of lysing tumor express-ing HER-2/neu and resulting in durable immunity in-

    volved immunizing patients with putative T-helper epi-

    topes of HER-2/neu which had, embedded in the nat-

    ural sequence, HLA-A2 binding motifs of HER-2/neu.

    Thus, both CD4+ T cell helper and CD8+ specific epi-

    topes were encompassed in the same vaccine. HLA-

    A2 patients with HER-2/neu-overexpressing cancers

    received a vaccine preparation consisting of putative

    HER-2/neuhelper peptides [6]. Contained within these

    sequences were the HLA-A2 binding motifs. Patients

    developed both HER-2/neu-specific CD4+ and CD8+

    T cell responses. The level of HER-2/neu immunity

    was similar to viral and tetanus immunity. In addi-

    tion, the peptide-specific T cells were able to lyse tu-

    mor, demonstrating generation of T cells that recog-

    nized naturally processed antigen. The responses were

    long-lived and detectable for greater than one year after

    the final vaccination in some patients. These results

    demonstrate that HER-2/neu MHC class II epitopes

    containing encompassed MHC class I epitopes are

    able to induce long-lasting HER-2/neu-specific IFN--

    producing CD8 T cells.

    Stimulating an effective T helper (Th) response, even

    without concomitant CD8+ peptide vaccination, is a

    way to boost antigen-specific immunity as CD4+ Tcells generate the cytokine environment required to

    support an evolving immune response. In one recent

    study, patients with advancedstage HER-2/neu overex-

    pressing breast, ovarian, and non-small cell lung cancer

    were enrolled on a trial of a multi-peptide HER-2/neu

    Th vaccine [7]. Over 90% of patients developed T cell

    immunity to HER-2/neu Th peptides and over 60% to a

    HER-2/neu protein domain. Thus, immunization with

    peptides resulted in the generation of T cells that could

    respond to protein processed by APC. Furthermore, at

    1-year follow-up, immunity to the HER-2/neu protein

    persisted in over a third of patients. Immunity elicited

    by active immunization with CD4+ T helper epitopes

    was durable. An additional finding of this study was

    that epitope spreading was observed in the majority of

    patients and significantly correlatedwith the generation

    of HER-2/neu protein-specific T cell immunity.

    Vaccines targeting HER-2/neu are among the most

    commonly tested immunotherapeutic strategies being

    evaluated in breast cancer patients. However, vacci-

    nation with longer peptides derived from the MUC-1

    breast cancer tumor antigen has undergone extensive

    clinical study. MUC-1 is a membrane-bound glyco-

    protein and is a primary component of the ductal cell

  • 7/30/2019 2004_BreastDis_3-11

    5/11

    M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer 7

    surface of most glandular cells. When a normal cell

    becomes cancerous, MUC-1 is both overexpressed andaberrantly glycosolated. Overexpression of MUC-1 in

    cancer cells interferes with cell adhesion and may fa-

    cilitate the development of metastasis. Several peptide

    based MUC-1 vaccines have been evaluated in clini-

    cal trials using it as a model antigen for eliciting both

    humoral and cellular immune responses. Early trials

    using MUC-1 with BCG as adjuvant induced DTH re-

    sponses, but only in a minority of patients [30]. For-

    mulating MUC-1 as a mannan-MUC-1 fusion protein

    and immunizing patients multiple times resulted in aug-

    mentation of MUC-1 specific antibodies in about 50%

    of patients, with titers higher than 1/20,000. However,

    patient numbers were small [31]. A more recent study

    describes vaccination of breast cancer patients with

    MUC-1/KLH along with QS-21, an adjuvant specif-

    ically developed to stimulate cellular immunity [32].

    While antibody titers were elevated, and remained so

    over 100 days after vaccination, no antigen-specific T

    cell responses were detected. MUC-1 specific anti-

    bodies generated after active immunization have, how-

    ever, been demonstrated to bind to and lyse tumor in

    vitro [33].

    PEPTIDE-SPECIFIC T CELLS ELICITED BYACTIVE IMMUNIZATION AND DIRECTED

    AGAINST A SINGLE BREAST CANCER

    ANTIGENIC EPITOPE CAN BE REMARKABLY

    DIVERSE IN PHENOTYPE AND FUNCTION

    A theoretical drawback of the use of peptide epi-

    topes as immunogens is the risk of generating very

    restricted immune responses, e.g. T cells of a single

    phenotype directed against the epitope. A strongly re-

    stricted response may limit potential therapeutic effi-

    cacy. Dissection of the peptide-specific immune re-

    sponse after immunization has demonstrated a marked

    phenotypic diversity of the vaccinated response [34].

    T cell clones specific for HER-2/neu HLA-A2 pep-

    tide p369-377 were isolated from a patient who had

    been vaccinated with HER-2/neu helper epitopes that

    contained HLA-A2-binding CTL epitopes within their

    sequences. Throughout the course of immunization,

    PBMC from this patient showed strong proliferative

    responses to the HER-2/neu helper epitope p369-384.

    Following vaccination, T cell clones specific for p369-

    377, the HLA-A2 binding peptide were isolated by lim-

    iting dilution and then characterized. The responding T

    cell repertoire generated was both phenotypically and

    functionally diverse. A total of 21 p369-377 T-cell

    clones were isolated from this patient; peptide-specificCD8+, CD4+ and T cell clones. Of note, de-

    spite being generated with an HLA-A2-binding pep-

    tide, CD4+ T cells were generated. Although CD4+

    T cells are predominantly associated with responding

    to peptides associated with MHC class II, at lower fre-

    quencies CD4 T cells play a role in regulating MHC

    class I-restricted T cells, particularly T cells associ-

    ated with cancers such as melanoma, colon, and pan-

    creatic cancer [35,36]. The majority of the clones ex-

    pressed the T cell receptor (TCR), however, two

    clones expressed the TCR. Clones could lyse HLA-

    A2-transfected HER-2/neu overexpressing tumor cells

    as well as peptide loaded MHC-matched cells. In ad-

    dition to their lytic capabilities these clones could be

    induced to produce IFN- specifically in response to

    p369-377 peptide stimulation. The TCR clones

    expressed CD8 and lysed HLA-A2 HER-2/neu posi-

    tive tumor cells, but not HLA-A2 negative HER-2/neu

    overexpressing tumor cells. T cells are involved in

    a wide range of immune responses to infectious and

    non-infectious diseases, including malaria, mycobac-

    terial infections, cancers, as well as autoimmune dis-

    orders such as multiple sclerosis [37]. Often, T

    cells clones are isolated from the tumor-infiltrating lym-

    phocyte population of many tumors including dysger-minoma [38], seminoma [38], renal carcinoma [39],

    lung [40], colorectal [41], and melanoma [42]. The

    recruitment to and role of these unique cells in medi-

    ating anti-tumor immunity is unknown. One hypoth-

    esis is that, given their broad range of regulation by

    multiple mechanisms of antigen presentation and nat-

    ural localization to epithelial tissue, TCR T cells

    are sentinels for the immune system and are capable of

    alerting the immune system to the presence of danger

    (e.g. infection, tumors, etc.). These results suggest

    that a tumor antigen-specific T cell response can be

    markedly polyclonal at multiple levels including T cell

    subset and TCR even if peptide epitopes are used as the

    immunizing antigens.

    BREAST CANCER VACCINES COMPOSED OF

    PEPTIDES AND DESIGNED TO GENERATE

    BREAST CANCER SPECIFIC ANTIBODY

    IMMUNITY

    Another aspect of peptide epitope prediction would

    be to identify peptide portions of a breast cancer

    antigen, such as the HER-2/neu extracellular domain

  • 7/30/2019 2004_BreastDis_3-11

    6/11

    8 M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer

    (ECD), appropriate to target with an antibody re-

    sponse. Several monoclonal antibodies against theHER-2/neu ECD have been isolated and one such an-

    tibody, trastuzumab, has demonstrated clinical efficacy

    in the treatment of metastatic breast cancer [43] . Al-

    though manyHER-2/neu-specific antibodiesinhibit the

    growth of cancer cells, some antibodies have no ef-

    fect on cell growth, while others even actively stimu-

    late cancer growth [44]. This wide range of biologi-

    cal effects is thought to be related to the epitope speci-

    ficity of the antibodies and to consequent changes in

    receptor signaling [44]. An alternative to the use of

    passive antibody therapy would be active immuniza-

    tion against the HER-2/neu ECD. However, inappro-

    priately induced immune responses could have unto-

    ward effects on cancer growth. Therefore, it is crucial

    to identify epitopes on HER-2/neu that are targeted by

    stimulatory and inhibitory antibodies in order to en-

    sure the induction of a beneficial endogenous antibody

    response.

    In a recent study, investigators constructed HER-

    2/neu gene fragment phage display libraries to epitope-

    map a number of HER-2/neu-specific antibodies with

    different biological effects on tumor cell growth [44].

    Regions responsible for opposing effects of antibod-

    ies were identified and then used to immunize mice.

    Mice immunized with the peptides developed antibod-ies that recognized both the peptide and native HER-

    2/neu. More importantly, HER-2/neu-specific antibod-

    ies purified from mouse sera were able to inhibit up to

    85% of tumor cell proliferation in vitro. Furthermore,

    using peptide regions that contain multiple inhibitory B

    cell epitopes is likely to be superior to the use of single

    epitope immunogens [45].

    THE NEXT GENERATION PEPTIDE BASED

    VACCINES FOR USE IN BREAST CANCER

    THERAPY

    Initial clinical trials of peptide-based breast cancer

    vaccines indicated that patients can be immunized, but

    immune responses were generally not to the levels of a

    vaccinated antigen such as tetanus toxoid. Therefore,

    more recent investigations are focused on improving

    the immunogenicity of peptide immunization.

    The choice of adjuvant is an important considera-

    tion in peptide vaccine trials, particularly as peptides

    themselves are typically only weakly immunogenic.

    Due to recent renewed enthusiasm for peptide-based

    tumor vaccines coupled with the search for adjuvants

    that promote cellular as well as antibody responses,

    the list of adjuvants being studied continues to grow.The choice of adjuvant has largely been determined by

    pre-clinical models. Similar to alum, oil-based incom-

    plete Freunds-type adjuvants, such as Montanide ISA-

    51 (Seppic, Paris, France) and TiterMax (CytRx Corp.,

    Norcross, GA), have been used in human peptide vac-

    cine studies [46,47], with their adjuvant effect likely

    mediated by a depot effect,increasing the half lifeof the

    peptide antigen at the site of immunization. Other pro-

    inflammatory biologic adjuvants such as BCG, Detox

    (mycobacterial cell-wall skeleton plus a lipid moiety)

    (Corixa Corp., Seattle, WA), and influenza virosomes

    have been used as adjuvants in peptide- and protein-

    based human vaccine studies [4850]. One study com-

    pared a variety of adjuvants in a rodent model targeting

    human MUC-1 peptide antigens conjugated to keyhole

    limpet hemocyanin (KLH), and found the saponin ad-

    juvant QS-21 (Aquilla Biopharmaceuticals, Worcester,

    MA) to be the most effective in promoting an antigen-

    specific antibody responses [51]. Subsequently, QS-21

    has been used as an adjuvant in human peptide cancer

    vaccine trials [52,53], with the hope that the use of a

    saponin may allow entry of peptides directly into MHC

    complexes to stimulate a T cell response [54]. Another

    type of adjuvant that has been studied in animal models

    is polymer microspheres that encapsulate the peptideantigen, permitting slow release over variable lengths

    of time [55]. Potential advantages to this type of ad-

    juvant includes the ability to use an oral route of de-

    livery [56], continuous release of antigen obviating the

    need for booster immunizations, and the ability to in-

    corporate other biologic adjuvants or cytokines within

    the polymer [57]. Finally, the discovery of various

    cytokines participating in the initiation of immune re-

    sponses has suggested that cytokines themselves may

    be useful immunologic adjuvants. As an example,

    soluble GM-CSF is a vaccine adjuvant, and has the

    ability to induce the differentiation of dendritic cells

    and act as a chemoattractant for various immune cell

    effectors [58,59].

    The relationship between MHC class I affinity and

    tumorantigen epitope immunogenicity is of importance

    because tissue-specific and developmental tumor anti-

    gens, such as those that are breast cancer antigens, are

    expressed on normal tissues at some point in time at

    some location within the body. T cells specific for these

    self antigens could be functionally inactivated by T cell

    tolerance. However, CTL responses to tumor epitopes

    in both volunteer donors and breast cancer patients in-

    dicate that tolerance to these tumor antigens, if it ex-

  • 7/30/2019 2004_BreastDis_3-11

    7/11

    M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer 9

    ists at all, is incomplete [8,60]. It is unknown whether

    T cells recognizing high affinity epitopes have beenselectively eliminated, leaving a repertoire capable of

    recognizing only low affinity epitopes. One investiga-

    tion evaluated several peptides derived from four dif-

    ferent breast cancer tumor antigens, p53, HER-2/neu,

    CEA, and MAGE proteins, for their capacity to in-

    duce CTL in vitro capable of recognizing tumor target

    lines [8]. In order to increase the likelihood of over-

    coming tolerance, fixed anchor analogues that demon-

    strate improved HLA-*0201 affinity and binding, were

    used along with wild type peptides. A total of 20/20

    wild-type and 9/9 single amino acid substitution ana-

    logues were found to be immunogenic in primary invitro CTL induction assays, using normal PBMCs and

    monocyte derived dendritic cells as APC. Cytotoxic T

    cells generated by 13/20 of the wild type epitopes and

    6/9 of single substitution analogues tested recognized

    HLA-matched antigen bearing cancer cell lines. Fur-

    ther analysis suggested that high binding affinity epi-

    topes are frequently generated and can be recognized

    as a result of natural antigen processing. Indeed, the

    immunogenicity of CTL epitopes derived from human

    tumor antigens such as CEA can be improved by alter-

    ing the peptide motif to improve binding to MHC [61].

    Studies such as these demonstrate that recognition of

    self-tumor antigens is within the realm of the T cell

    repertoire and that binding affinity may be an important

    criterion for epitope selection.

    CONCLUSION

    The definition of multiple breast cancer antigen-

    specific peptides, both class I and class II derived epi-

    topes are rapidly being translated into human clinicalstudies. Investigations over the last decade have al-

    lowed the development of technology such as com-

    puter algorithms and high throughput peptide binding

    assays to rapidly screen vaccine candidates. Clinical

    trials of single peptides have demonstrated that cancer

    patients can be immunized against self-tumor antigens

    and some studies have shown early positive clinical re-

    sults. Current effort in the field is focused on improving

    the immunogenicity of individual MHC binding pep-

    tides as well as developing multiple peptide vaccines

    for the prevention and treatment of breast cancer.

    ACKNOWLEDGEMENTS

    This work is supported for MLD by grants from

    the NIH, NCI (R01 CA75163 and CA85374, and K24CA85218) as well as funding from the Cancer Re-search Treatment Foundation, for LGS by NIH train-ing grant T32 (HL07093), and for KLK by a fellow-ship from the Department of Defense Breast CancerProgram (DAMD17-00-1-0492).

    REFERENCES

    [1] M. Disis, Immunologic targets for breast cancer therapy,Breast Disease 15 (2003), 8390.

    [2] G. Ada, Vaccines and vaccination, N Engl J Med 345 (2001),10421053.

    [3] M.L.Disis, J.R.Gralow and H. Bernhard et al., Peptide-based,but not whole protein, vaccines elicit immunity to HER-2/neu,an oncogenic self-protein, J Immunol 156 (1996), 31513158.

    [4] R. Cibotti, J.M. Kanellopoulos and J.P. Cabaniols et al., Tol-erance to a self-protein involves its immunodominant but doesnot involve its subdominant determinants, Proc Natl Acad SciUSA 89 (1992), 416420.

    [5] E.E. Sercarz, P.V. Lehmann and A. Ametani et al., Domi-nance and crypticity of T cell antigenic determinants, Annu

    Rev Immunol 11 (1993), 729766.[6] K.L. Knutson, K. Schiffman and M.A. Cheever et al., Immu-

    nization of cancer patients with a HER-2/neu, HLA-A2 pep-tide, p369-377, results in short-lived peptide-specific immu-nity, Clin Cancer Res 8 (2002), 10141018.

    [7] M.L. Disis, T.A. Gooley and K. Rinn et al., Generation of T-cell immunity to theHER-2/neu protein after active immuniza-tion with HER-2/neu Peptide-based vaccines, J Clin Oncol 20(2002), 26242632.

    [8] E. Keogh, J. Fikes and S. Southwood et al., Identification ofnew epitopes from four different tumor-associated antigens:recognition of naturallyprocessedepitopescorrelateswith hla-a(*)0201-binding affinity, J Immunol 167 (2001), 787796.

    [9] J. Lu and E. Celis, Use of two predictive algorithms of theworld wide web for the identification of tumor-reactive T-cellepitopes, Cancer Res 60 (2000), 52235227.

    [10] Y. Rongcun, F. Salazar-Onfray and J. Charo et al., Identifica-tion of new HER2/neu-derived peptide epitopes that can elicitspecific CTL against autologous and allogeneic carcinomasand melanomas, J Immunol 163 (1999), 10371044.

    [11] A.M. Livingstone and C.G. Fathman, The structure of T-cellepitopes, Annu Rev Immunol 5 (1987), 477501.[12] J.C. Reece, D.L. McGregor and H.M. Geysen et al., Scanning

    for T helper epitopes with human PBMC using pools of shortsynthetic peptides, J Immunol Methods 172 (1994), 241254.

    [13] H. Kobayashi, M. Wood and Y. Song et al., Defining promis-cuous MHC class II helper T-cell epitopes for the HER2/neutumor antigen, 60 (2000), 52285236.

    [14] H.M. Zarour, B. Maillere and V. Brusic et al., NY-ESO-1 119-143 is a promiscuous major histocompatibility complex classII T-helper epitope recognized by Th1- and Th2-type tumor-reactive CD4+ T cells, Cancer Res 62 (2002), 213218.

    [15] R.Schroers, X.F. Huang andJ. Hammeret al., Identification ofHLA DR7-restricted epitopes from human telomerase reversetranscriptase recognized by CD4+ T-helper cells, Cancer Res

    62 (2002), 26002605.

  • 7/30/2019 2004_BreastDis_3-11

    8/11

    10 M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer

    [16] K. Hung, R. Hayashi and A. Lafond-Walker et al., The centralrole of CD4(+) T cells in the antitumor immune response, J

    Exp Med 188 (1998), 23572368.[17] S.A. Kalams and B.D. Walker, The critical need for CD4 help

    in maintaining effective cytotoxic T lymphocyte responses[comment], J Exp Med188 (1998), 21992204.

    [18] C. DeLisi BJ, T-cell antigenic sites tend to be amphipathicstructures, Proc. Natl. Acad. Sci. USA 82 (1985), 70487052.

    [19] D.C. Feller, dlCV. Identifying antigenic T-cell sites, Nature

    349 (1991), 720721.[20] A. OGarra and K. Murphy, T-cell subsets in autoimmunity,

    Curr Opin Immunol 5 (1993), 880886.[21] C.G. Ioannides and T.L. Whiteside, T cell recognition of hu-

    man tumors: implications for molecular immunotherapy ofcancer, Clin Immunol Immunopathol 66 (1993), 91106.

    [22] L. Salazar, J. Fikes and S. Southwood et al., Immunization ofcancer patients with HER-2/neu derived peptides demonstrat-

    ing high affinity binding to multiple class II alleles, Clin CaRes (in press 2003).

    [23] T.Z. Zaks and S.A. Rosenberg, Immunization with a peptideepitope (p369-377) from HER-2/neu leads to peptide-specificcytotoxic T lymphocytes that fail to recognize HER- 2/neu+

    tumors, Cancer Res 58 (1998), 49024908.[24] J.L. Murray, M.E. Gillogly and D. Przepiorka et al., Toxi-

    city, immunogenicity, and induction of E75-specific tumor-lytic CTLs by HER-2 peptide E75 (369-377) combined withgranulocyte macrophage colony-stimulating factor in HLA-A2+ patients with metastatic breast and ovarian cancer, ClinCancer Res 8 (2002), 34073418.

    [25] P. Brossart, S. Wirths and G. Stuhler et al., Induction of cyto-toxic T-lymphocyte responses in vivo after vaccinations withpeptide-pulsed dendritic cells, Blood 96 (2000), 31023108.

    [26] P.V. Lehmann, T. Forsthuber and A. Miller et al., Spreading of

    T-cell autoimmunity to cryptic determinants of an autoantigen,Nature 358 (1992), 155157.

    [27] E.E. Sercarz, Driver clones and determinant spreading, J Au-toimmun 14 (2000), 275277.

    [28] A. Jaramillo, K. Majumder and P.P. Manna et al., Identificationof HLA-A3-restricted CD8+ T cell epitopes derived frommammaglobin-A, a tumor-associated antigen of human breastcancer, Int J Cancer102 (2002), 499506.

    [29] M.H. Andersen, L.O. Pedersen and B. Capeller et al., Spon-taneous cytotoxic T-cell responses against survivin-derivedMHC class I-restricted T-cell epitopes in situ as well as ex vivo

    in cancer patients, Cancer Res 61 (2001), 59645968.[30] M. Reddish, G.D. MacLean and R.R. Koganty et al., Anti-

    MUC1 class I restricted CTLs in metastatic breast cancer pa-tients immunized with a synthetic MUC1peptide, IntJ Cancer

    76 (1998), 817823.[31] V. Karanikas, G. Thynne and P. Mitchell et al., Mannan mucin-

    1 peptide immunization: influence of cyclophosphamide andthe route of injection, J Immunother 24 (2001), 172183.

    [32] T. Gilewski, S. Adluri and G. Ragupathi et al., Vaccination ofhigh-riskbreast cancer patients withmucin-1(MUC1) keyholelimpet hemocyanin conjugate plus QS-21, Clin Cancer Res 6(2000), 16931701.

    [33] F.G. Snijdewint, S. von Mensdorff-Pouilly and A.H. Karuntu-Wanamarta et al., Antibody-dependent cell-mediated cytotox-icity can be induced by MUC1 peptide vaccination of breastcancer patients, Int J Cancer93 (2001), 97106.

    [34] K. Knutson and M. Disis, Clonal diversity of the T cell reper-toirerespondingto a dominant HLA-A2 eptiope of HER-2/neuafter active immunization, Hum Immunol 63 (2002), 547557.

    [35] J.E. de Vries and H. Spits, Cloned human cytotoxic T lym-phocyte (CTL) lines reactive with autologous melanoma cells.I. In vitro generation, isolation, and analysis to phenotype andspecificity, J Immunol 132 (1984), 510519.

    [36] R. Somasundaram, P. Robbins and D. Moonka et al., CD4(+),HLA class I-restricted, cytolytic T-lymphocyte clone againstprimary malignant melanoma cells, Int J Cancer 85 (2000),253259.

    [37] R.Boismenuand W.L.Havran, Gammadelta T cells in host de-fense and epithelial cell biology, Clin Immunol Immunopathol

    86 (1998), 121133.[38] X. Zhao, Y.Q. Wei and Y. Kariya et al., Accumulation of

    gamma/delta T cells in human dysgerminoma and seminoma:roles in autologous tumor killing and granuloma formation,

    Immunol Invest 24 (1995), 607618.[39] A. Choudhary, F. Davodeau and A. Moreau et al., Selective ly-

    sis of autologous tumor cells by recurrent gamma delta tumor-

    infiltrating lymphocytes from renal carcinoma, J Immunol 154(1995), 39323940.

    [40] D. Yu, A. Matin and W. Xia et al., Liposome-mediated in vivo

    E1A gene transfer suppressed dissemination of ovarian cancercells that overexpress HER-2/neu, Oncogene 11 (1995), 1383-1388.

    [41] N. Watanabe, A. Hizuta and N. Tanaka et al., Localizationof T cell receptor (TCR)-gamma delta + T cells into humancolorectal cancer: flow cytometric analysis of TCR-gammadelta expression in tumour-infiltrating lymphocytes, Clin Exp

    Immunol 102 (1995), 167173.[42] H. Bachelez, B. Flageul and L. Degos et al., TCR gamma delta

    bearing T lymphocytes infiltrating human primary cutaneousmelanomas, J Invest Dermatol 98 (1992), 369374.

    [43] C. Vogel, M.A. Cobleigh and D. Tripathy et al., First-line,single-agent Herceptin(R) (trastuzumab) in metastatic breast

    cancer. a preliminary report,Eur J Cancer37 (Suppl 1) (2001),2529.

    [44] Y.L. Yip, G. Smith and J. Koch et al., Identification of epi-tope regions recognized by tumor inhibitory and stimulatoryanti-ErbB-2 monoclonal antibodies: implications for vaccinedesign, J Immunol 166 (2001), 52715278.

    [45] N.K. Dakappagari, D.B. Douglas and P.L. Triozzi et al., Pre-vention of mammary tumors with a chimeric HER-2 B-cellepitope peptide vaccine, Cancer Res 60 (2000), 37823789.

    [46] R.M.W.J. van Driel, G.G. Kenter, R.M. Brandt, E.J. Krul,A.B. van Rossum, E. Schuuring, R. Offringa, T. Bauknecht,A. Tamm-Hermelink, P.A. van Dam, G.J.Fleuren, W.M. Kast,C.J. Melief and J.B. Trimbos, Vaccination with HPV16 pep-tides of patients with advanced cervical carcinoma: clinicalevaluation of a phase I-II trial, Eur J Cancer 35 (1999), 946952.

    [47] F.B.E.Wang, C. Kuniyoshi, L. Spears, G. Jeffery, V. Marty,S. Groshen and J. Weber, Phase I trial of a MART-1 peptidevaccine with incomplete Freunds adjuvant for resected high-risk melanoma, Clin. Cancer Res. 5 (1999), 27562765.

    [48] J.S.E.E. Goydos, T.L. Whiteside, O.J. Finn and M.T. Lotze, Aphase I trial of a synthetic mucin peptide vaccine. Induction ofspecific immune reactivity in patients with adenocarcinoma,

    J. Surg. Res. 63 (1996), 298304.[49] N. Schultz, R. Oratz, D. Chen, A. Zeleniuch-Jacquotte, G.

    Abeles and J.C. Bystryn, Effect of DETOX as an adjuvant formelanoma vaccine, Vaccine 13 (1995), 503508.

    [50] R. Gluck, Immunopotentiating reconstituted influenza viro-somes (IRIVs) and other adjuvants for improved presentationof small antigens, Vaccine 10 (1992), 915919.

  • 7/30/2019 2004_BreastDis_3-11

    9/11

    M.L. Disis et al. / Peptide-Based Vaccines in Breast Cancer 11

    [51] S.K.R.G. Kim, C. Musselli, S.J. Choi, Y.S. Park and P.O. Liv-ingston, Comparison of the effect of different immunologicaladjuvants on the antibody and T-cell response to immunizationwith MUC1-KLH and GD3-KLH conjugate cancer vaccines,Vaccine 18 (1999), 597603.

    [52] T.A.S. Gilewski, G. Ragupathi, S. Zhang, T.J. Yao, K.Panageas, M. Moynahan, A. Houghton, L. Norton and P.O.Livingston, Vaccination of high-risk breast cancer patientswith mucin-1 (MUC1) keyhole limpet hemocyanin conjugateplus QS-21, Clin. Cancer Res. 6 (2000), 16931701.

    [53] J. Pinilla-Ibarz, K. Cathcart and T. Korontsvit et al., Vaccina-tion of patients with chronic myelogenous leukemia with bcr-abl oncogene breakpoint fusion peptides generates specificimmune responses, Blood 95 (2000), 17811787.

    [54] M.J.W.J. Newman, B.H. Gardner, K.J. Munroe, D. Leom-bruno, J. Recchia, C.R. Kensil and R.T. Coughlin, Saponinadjuvant induction of ovalbumin-specific CD8+ cytotoxic T

    lymphocyte responses, J. Immunol. 148 (1992), 23572362.[55] Y.G.B. Men, H.P. Merkle and G. Corradin, Induction of

    sustained and elevated immune responses to weakly im-munogenic synthetic malarial peptides by encapsulation inbiodegradable polymer microspheres, Vaccine 14 (1996),14421450.

    [56] C.O.R.R. Tacket, E.C. Boedeker, G. Losonsky, J.P. Nataro,

    H. Bhagat and R. Edelman, Enteral immunization and chal-lenge of volunteers given enterotoxigenic E. coli CFA/II en-capsulated in biodegradable microspheres, Vaccine 12 (1994),12701274.

    [57] R.C.J. Langer and J. Hanes, New advances in microsphere-based single-dose vaccines, Adv. Drug Deliv. Rev. 28 (1997),97119.

    [58] S.E.E. Markowicz, Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of hu-man peripheral blood dendritic cells in vitro, J. Clin. Invest.

    85 (1990), 955961.[59] R.H.G.D. Weisbart, S.C. Clark, G.G. Wong and J.C. Gasson,

    Human granulocyte-macrophage colony stimulating factor isa neutrophil activator, Nature 314 (1985), 361363.

    [60] I. Kawashima, S.J. Hudson and V. Tsai et al., The multi-epitope approach for immunotherapy for cancer: identifica-tion of several CTL epitopes from various tumor-associated

    antigens expressed on solid epithelial tumors, Hum. Immunol.59 (1998), 114.

    [61] E. Huarte, P. Sarobe and J. Lu et al., Enhancing Immunogenic-ity of a CTL Epitope from Carcinoembryonic Antigen by Se-lective Amino Acid Replacements, Clin Cancer Res 8 (2002),23362344.

  • 7/30/2019 2004_BreastDis_3-11

    10/11

  • 7/30/2019 2004_BreastDis_3-11

    11/11