Mehren,2003

download Mehren,2003

of 29

Transcript of Mehren,2003

  • 8/13/2019 Mehren,2003

    1/29

    Annu. Rev. Med. 2003. 54:34369doi: 10.1146/annurev.med.54.101601.152442

    Copyright c 2003 by Annual Reviews. All rights reserved

    MONOCLONALANTIBODYTHERAPY FORCANCER

    Margaret von Mehren, Gregory P. Adams,and Louis M. Weiner

    Department of Medical Oncology, Fox Chase Cancer Center, 7701 Burholme Avenue,

    Philadelphia, Pennsylvania 19111; e-mail: m [email protected],

    gp [email protected], lm [email protected]

    Key Words immunoconjugate, immunotoxin, radioimmunotherapy,antibody-dependent cellular cytotoxicity

    Abstract Monoclonal antibody therapy has emerged as an important therapeuticmodality for cancer. Unconjugated antibodies show significant efficacy in the treat-ment of breast cancer, non-Hodgkins lymphoma, and chronic lymphocytic leukemia.Promising new targets for unconjugated antibody therapy include cellular growth fac-tor receptors, receptors or mediators of tumor-driven angiogenesis, and B cell surfaceantigens other than CD20. Immunoconjugates composed of antibodies conjugated toradionuclides or toxins show efficacy in non-Hodgkins lymphoma. One immunocon-

    jugate containing an antibody and a chemotherapy agent exhibits clinically meaningfulantitumor activity in acute myeloid leukemia. Numerous efforts to exploit the abilityof antibodies to focus the activities of toxic payloads at tumor sites are under way andshow early promise. The ability to create essentially human antibody structures hasreduced the likelihood of host-protective immune responses that otherwise limit theduration of therapy. Antibody structures now can be readily manipulated to facilitateselective interaction with host immune effectors. Other structural manipulations thatimprove the selective targeting properties and rapid systemic clearance of immuno-conjugates should lead to the design of effective new treatments, particularly for solid

    tumors.

    INTRODUCTION

    The description of techniques for the production of monoclonal antibodies (MAb)

    by Kohler & Milstein 25 years ago led to the development of multiple reagents

    employing these structures. Antibodies, which initially were viewed as target-

    ing missiles, have proved much more complex in their targeting and biologic

    properties than the fields pioneers envisioned them. The ability to manipulate thegenes of antibodies with microbiologic techniques has allowed significant mod-

    ifications of these structures. Murine proteins can be readily transformed into

    human or humanized formats that are not readily recognized as foreign by the

    byb

    on:UniversidadedeLisboa(UL)

    on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    2/29

    344 VON MEHREN ADAMS WEINER

    human immune system. In addition, novel antibody-based structures with multi-

    ple antigen recognition sites, altered size, or effector domains have been shown

    to influence the targeting ability of antibodies. Coupled with the identification

    of appropriate cancer targets, antibody-based therapeutics are finding increasingapplications in cancer treatment, and they can be effective alone, in conjunction

    with chemotherapy or radiation therapy, or when conjugated to toxic moieties

    such as toxins, chemotherapy agents, or radionuclides. This review focuses on

    antibody-based agents with significant efficacy and novel approaches with clinical

    promise.

    IMMUNOGLOBULIN STRUCTURE

    IgG

    IgG molecules are the most common antibodies employed in cancer therapy. They

    are comprised of two identical light chains and two identical heavy chains, each

    composed of variable domains and constant domains (Figure 1). The variable re-

    gion contains the hypervariable or complementarity-determining regions (CDRs).

    These short, highly variable amino acid sequences are the major site of interaction

    with antigen. There are three CDRs (CDR1, CDR2, and CDR3) on each variable

    chain. Both the light chains and the heavy chains are held together by disulfide

    bonds and noncovalent interactions between several of the domains. The complexis oriented with bilateral symmetry: light chainheavy chainheavy chainlight

    chain. The IgG molecule is divided into three functional domains, two antigen-

    binding (Fab) domains with identical antigen specificity connected by a highly

    flexible hinge domain to the Fc (effector) domain that interacts with complement,

    Figure 1 Schematic diagram depicting the structures of an IgGmolecule and a variety

    of engineered antibody molecules derived from various IgG domains.

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    3/29

    CANCER ANTIBODIES 345

    immune effector cells, and receptors involved in maintaining constant concentra-

    tions of IgG in the circulation.

    Antibody Fragments

    For many years, IgG molecules have been enzymatically digested into smaller

    functional fragments [Fab and F(ab)2]. More recently, the genes encoding the

    variable and constant domains have been used to construct a variety of recombi-

    nant antibody-based fragments. These range in size from peptides comprised of an

    individual CDR (1) to multidomained molecules with four antigen binding sites

    (2). The smallest antibody-based fragments, individual CDRs and single-variable

    domains, often exhibit lower affinity and less antigen specificity than correspond-

    ing intact antibody molecules (1, 3). Single-chain Fv (scFv) molecules, composedof peptide-linked VH and VL domains, are rapidly emerging as key players in

    the engineered antibody field. With a size of25 kDa, these molecules are both

    effective targeting vehicles in their own right and versatile components for the con-

    struction of larger antibody-based regents. ScFv molecules can be produced from

    genes isolated from hybridomas expressing MAb of a desired specificity (4) or can

    be isolated by panning (selection) from a combinatorial phage display library (5).

    The small size of scFv molecules mediates their rapid renal elimination, leading

    to highly specific tumor localization. Because of this property, radiolabeled scFv

    molecules are highly attractive agents for use in tumor detection (6).ScFv can be dimerized to yield larger molecules that exhibit a slower systemic

    clearance and a greater avidity for cells that overexpress their target antigen, making

    them potentially useful in tumor therapy. Various strategies can be employed to

    dimerize scFv molecules. These include the creation of disulfide-linked (scFv)2 (7)

    or gene-fused (scFv)2, with a peptide spacer joining two scFv, and the shortening of

    the linker between the light and heavy chain, forcing the production of diabodies by

    noncovalent associations between two molecules (8). Larger divalent molecules,

    termed minibodies, can be produced by fusing the gene for an scFv to that for a

    CH3 domain (9). An additional advantage of the minibody format is the greater

    stability of the complex conferred by the affinitybetween the CH3 domains. Tandem

    diabodies, with four antigen-binding sites, can be produced by joining two diabody

    components with peptide spacer (2).

    The ideal antibody-based molecule depends on the application. If a naked

    antibody can directly mediate an antitumor effect, the greatest possible bioavail-

    ability is desired. This is best achieved by leaving the antibody in the native IgG

    format, which will exhibit a prolonged residence in the circulation. However, the

    native IgG format could lead to unacceptable bone marrow toxicity if the antibody

    is conjugated to a therapeutic radioisotope for radioimmunotherapy (RAIT) appli-

    cations. For RAIT, smaller, more rapidly cleared molecules such as minibodies or

    diabodies can reduce nontargeted marrow toxicity. Furthermore, because physio-

    logic barriers such as heterogeneous blood supply and elevated interstitial pressure

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    4/29

    346 VON MEHREN ADAMS WEINER

    (10) impede the penetration of antibodies into tumors, these smaller molecules may

    have other advantages over intact IgG. Jain & Baxter (11) determined that an intact

    IgG molecule would take 54 h to move 1 mm into a solid tumor, whereas a Fab

    fragment would travel the same distance in only 16 h.

    DESIRABLE CHARACTERISTICS OF ANTIBODY TARGETS

    A variety of protein and carbohydrate molecules on the surface of cancer cells are

    potential targets for antibody-directed therapies. Clinically useful targets may be

    uniquely expressed by cancer cells or expressed at higher levels by cancer cells than

    by normal cells. As a result, antibodies will selectively bind to cancer cells. Recep-

    tors on the surface of cancer cells are particularly attractive targets for therapeutic

    antibodies if binding of the receptor perturbs a downstream signaling event, or

    inhibits the binding of a natural ligand to its receptor and therefore interferes with

    a normal signaling pathway. If an antibody is linked to a radioactive particle, im-

    munotoxin, or chemotherapeutic agent, it may be beneficial to target a receptor that

    internalizes upon antibody binding, leading to internalization of the toxic agent.

    MECHANISMS OF ACTION FOR ANTIBODIESAS THERAPEUTIC AGENTS

    Antibodies may exert antitumor effects by inducing apoptosis (12), interfering with

    ligand-receptor interactions (13), or preventing the expression of proteins that are

    critical to the neoplastic phenotype. In addition, antibodies have been developed

    that target components of the tumor microenvironment, perturbing vital structures

    such as the formation of tumor-associated vasculature. Some antibodies target

    receptors whose ligands are growth factors, such as the epidermal growth fac-

    tor receptor. The antibody thus inhibits natural ligands that stimulate cell growth

    from binding to targeted tumor cells. Alternatively, antibodies may induce an

    anti-idiotype network (14), complement-mediated cytotoxicity (15), or antibody-

    dependent cellular cytotoxicity (ADCC) (16). An anti-idiotypic network results

    from the recognition of the antibody, called Ab1, by the host immune system. The

    antigenic binding site within the variable regions of the antibody is seen as foreign.

    When an antibody, termed Ab2, is formed against this binding site, it recapitulates

    the three-dimensional structure of the antigen targeted by Ab1. This chain of events

    can keep occurring, thus perpetuating the immune response against the primary

    antigen (17, 18). Some classes of immunoglobulins can activate complement or

    natural killer (NK) cells. The first component of the complement cascade, C1, is

    capable of binding the Fc portion of IgM and IgG molecules. C1 activation triggers

    the classical complement cascade, leading to the recruitment of phagocytic cells

    and death of the antibody-bound cell. This process occurs more efficiently with

    IgM molecules, but it can also occur if IgG molecules are clustered on the cell sur-

    face. Cells coated by IgG1 or IgG3 isotype antibodies can also activate effector cells

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    5/29

    CANCER ANTIBODIES 347

    via the binding of the terminal Fc portion of the antibody to Fc receptors, found

    on NK cells, neutrophils, mononuclear phagocytes, some T cells, and eosinophils.

    ADCC occurs with the release of cytoplasmic granules containing perforins and

    granzymes from these effector cells. This phenomenon can be amplified by con-structing bispecific antibodies that contain two binding domains, one that targets

    a tumor antigen and one that targets the immunoglobulin Fc receptor on immune

    effector cells, thereby activating the effector cells for tumor lysis.

    ANTIBODY LIMITATIONS

    Initial clinical trials with MAb produced some striking antitumor effects (19),

    but most of the early experiences illustrated the obstacles to successful therapy

    (Table 1). Most of the MAb employed in early clinical trials were derived from

    mice, and patients exposed to them developed human antimouse antibody (HAMA)

    responses, which limited the number of treatments patients could safely receive

    (20). Molecular engineering techniques have overcome this obstacle by grafting

    critical sequences in the human heavy-chain backbone onto the xenogeneic murine

    antibody structure, paring the interspecies differences and reducing the immuno-

    genicity of the resulting antibodies. Numerous techniques have further reduced

    immunogenicity, and it is now possible to create fully human immunoglobulins

    with limited capacity to induce HAMA. However, these approaches do not inhibit

    the production of anti-idiotype antibodies (see previous section).

    Some tumor antigens are shed or secreted. Antibodies that target these antigens

    will bind their targets in the circulation, limiting the amount of unbound antibody

    available to bind to tumor (21). Barriers that impede antibody distribution within

    tumors include (a) disordered tumor vasculature, (b) increased hydrostatic pressure

    within tumors and (c) heterogeneity of antigen distribution within tumors (11). It

    has been estimated that, due to these barriers, an IgG molecule will take 2 days to

    travel 1 mm and 78 months to travel 1 cm within a tumor. Estimates for a smaller

    antibody-based molecule, such as a Fab fragment, are 1 day to travel 1 mm and

    2 months to travel 1 cm. If antibodies do reach their targets, there is little evidence

    TABLE 1 Obstacles to effective antibody

    therapy

    1. Immunogenicity of xenogeneic antibodies

    2. Shedding of antigen into circulation

    3. Disordered vasculature in tumors

    4. Increased hydrostatic pressure in tumors

    5. Heterogeneity of antigen on tumor surface

    6. Limited numbers of effector cells at tumor

    7. Immunosuppressive tumor microenvironment

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    6/29

    348 VON MEHREN ADAMS WEINER

    that they efficiently mediate ADCC in vivo. For this to occur, sufficient numbers

    of effector cells, such as macrophages, NK cells, or cytotoxic T cells, must be

    present in the tumor (22). Finally, many tumors are known to secrete compounds

    that downregulate the immune response (23, 24), or to have decreased effectorcells as a consequence of hypoxia (25). Despite these impediments, preclinical and

    clinical data with improved antibody-based molecules continue to demonstrate a

    role for antibody-based therapy as a component of the oncologic armamentarium.

    ANTIBODY THERAPEUTICS

    The ability to identify therapeutic targets and produce antibodies with limited

    immunogenicity has led to the production and testing of a host of agents, several ofwhich have demonstrated clinically important antitumor activity and have received

    FDA approval for treatment of cancer. We focus here on antibodies with proven

    efficacy, classified according to the tumor or antigen system that is targeted.

    Hematologic Malignancy Antigens

    B CELL ANTIGENS Initial studies employing antibodies directed against B cell

    determinants showed that the passive administration of these antibodies led to

    clearance of circulating tumor cells and rare objective clinical responses (26). In

    a series of landmark studies, Levy and colleagues prepared customized antibodiesreactive with a given lymphoma patients idiotype that was uniquely expressed

    on the surface of the malignant B cell clone. Each patients idiotype served as a

    tumor-specific signature that could be targeted by a customized MAb. The pro-

    cedures for preparing such antibodies for each patient were laborious, but 50%

    of treated patients experienced significant clinical responses, with some patients

    achieving durable complete remissions (19). The addition of chemotherapy agents,

    interferon, or other cytokines did not appreciably improve treatment outcomes.

    Effective therapy had to overcome circulating lymphoma idiotype proteins that

    diverted antibodies from their cellular targets, and resistance to therapy resulted inpart from the emergence of idiotype-negative variants. The mechanisms underlying

    responses to these antibodies have not been completely elucidated but may include

    mechanisms such as ADCC (see below) and perturbation of signal transduction

    through idiotype engagement (27). Due to the immunosuppression of lymphoma

    patients, relatively few patients developed HAMA that interfered with repeated

    therapeutic antibody administration. Thisexciting approach was very cumbersome,

    as it required the generation of patient-customized reagents. These results could

    not be replicated using antibodies that recognize shared idiotypes expressed by a

    large proportion of lymphoma patients (28). Despite this set of impediments, theseimportant observations have informed much of the subsequent work in this field.

    CD52 The CAMPATH-1 antibody has specificity for CD52, a glycopeptide that

    is highly expressed on T and B lymphocytes. It has been tested as a therapeutic

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    7/29

    CANCER ANTIBODIES 349

    agent for chronic lymphocytic and promyelocytic leukemias, as well as other

    non-Hodgkins lymphomas, and as a means to deplete T cells from allogeneic trans-

    plant grafts. Half of the patients with fludarabine-resistant chronic lymphocytic

    leukemia or B-prolymphocytic leukemia exhibited clinical responses toCAMPATH-1 (29). A larger phase II study reported a 42% response rate in pa-

    tients with relapsed or refractory chronic lymphocytic leukemia, but at the cost of

    an increase in opportunistic infections and septicemia. CAMPATH-1 has also been

    evaluated as first-line therapy for patients with chronic lymphocytic leukemia. All

    patients responded with loss of peripheral blood malignant lymphocytes. How-

    ever, patients with involvement of lymph nodes and/or spleen were less likely to

    respond completely. There was evidence of reactivation of cytomegalovirus in-

    fections. Subcutaneous administration of the antibody was found to be safe and

    effective. A humanized version of the antibody (CAMPATH-1H) has been exten-sively tested.

    In a phase II multicenter study of CAMPATH-1H in previously treated patients

    with low-grade non-Hodgkins lymphomas, 50 patients with relapsed or refrac-

    tory disease were treated with 30 mg of CAMPATH-1H 3 times weekly for up

    to 12 weeks (30). Infection, anemia, and thrombocytopenia were common, and

    myocardial infarction occurred in one patient with a prior history of angina and

    congestive heart failure. The overall response rate was 20% (16% partial response,

    4% complete reponse). Responses were short in duration, with a median time to

    progression of 4 months. Patients with mycosis fungoides responded more fre-quently and had a longer time to progression (10 months) than did patients with

    low-grade non-Hodgkins lymphoma (4 months). Treatment was associated with

    reactivation of herpes simplex, oral candidiasis,pneumocystis cariniipneumonia,

    cytomegalovirus pneumonitis, pulmonary aspergillosis, disseminated tuberculo-

    sis, and seven cases of pneumonia and septicemia.

    CAMPATH-1 has been used to deplete T cells from allogeneic transplant grafts

    in patients with hematologic malignancies (31, 32). The initial study suggested

    that the addition of CAMPATH-1 significantly decreased graft rejection and graft-

    versus-host disease compared with conventional therapy. However, the frequencyof graft rejection and graft failure was higher. A retrospective review of patients

    with acute lymphocytic or acute myelogenous leukemia who underwent allogeneic

    transplantation with CAMPATH-1purged marrow suggested no impact on the

    graft-versus-leukemia effect, as there was no increase in leukemia relapse (33). Ad-

    ditionally, the incidence of Epstein-Barr virus (EBV)related lymphoproliferative

    disorders was decreased in allogeneic transplant patients who had T cell depletion

    with CAMPATH-1 therapy compared with other methods of T cell depletion (e.g.,

    E-rosettes or other MAb) (34). CAMPATH-1 eliminated B cells within the graft,

    thus eliminating a potential reservoir of EBV or targets for subsequent infection.

    CD20 The testing and evaluation of the chimeric anti-CD20 antibody, IDEC-

    C2B8, also known as rituximab, demonstrated impressive clinical responses.

    Rituximab is the first MAb to be approved by the FDA for use in human malignancy

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    8/29

    350 VON MEHREN ADAMS WEINER

    (35, 36). Rituximab is humanized and multiple doses can be safely administered.

    In vitro studies have demonstrated multiple mechanisms by which anti-CD20 an-

    tibodies lead to cell death: ADCC, complement-mediated lysis, and apoptosis that

    is diminished by inhibitors of Lck and Fyn tyrosine kinases, calcium chelators,and caspase inhibitors (37).

    In the phase I study to determine the maximum tolerated dose, patients with re-

    lapsed low-grade and intermediate/high-grade non-Hodgkins lymphoma received

    four weekly infusions of rituximab (37). Thrombocytopenia and B cell lympho-

    cytopenia were observed. The lymphocytopenia persisted for 36 months. Six of

    18 patients, all of whom had low-grade lymphomas, demonstrated partial responses

    (33%). Phase II studies using the maximum tolerated dose, 375 mg/m2, confirmed

    the efficacy of this therapy, demonstrating 46% and 48% response rates in two

    separate studies (38, 39). Although the numbers of circulating B cells were re-duced by therapy, there were no documented changes in serum immunoglobulin

    levels. Bacterial and viral infections were seen in patients with relapsed indo-

    lent lymphomas, but in contrast to the CAMPATH-1 experience, treatment with

    rituximab did not result in significant morbidity due to infections. Patients with

    small-lymphocytic-B-cell lymphoma had lower response rates, probably related

    to the lesser expression of CD20 on these tumor cells.

    Although phase I testing had not suggested significant activity in intermediate/

    high-grade lymphomas, a phase II trial evaluated rituximab in relapsing or refrac-

    tory diffuse large B cell lymphoma, mantle cell lymphoma, or other intermediate-or high-grade B cell non-Hodgkins lymphomas (40). The study randomized

    54 patients to either 8 weekly treatments of 375 mg/m2 intravenous rituximab or

    375 mg/m2 in week one followed by 7 weekly intravenous infusions of 500 mg/m2.

    Five complete responses and 12 partial responses were observed, for an overall

    response rate of 31%; there was no evidence of superiority of either treatment reg-

    imen. Patients with refractory disease and those with histologies other than diffuse

    large B cell lymphoma appeared to have lower response rates. A novel use of rit-

    uximab has been reported for cutaneous B cell lymphoma; patients who received

    intralesional injections of the antibody showed partial clearing of nodules (41).Rituximab has been tested in conjunction with chemotherapy (42). Preclinical

    data show that this antibody can sensitize chemotherapy-resistant cell lines to the

    cytotoxic effects of chemotherapy (43). Forty patients with low-grade or follicular

    B cell non-Hodgkins lymphoma were enrolled in the study. Thirty-five patients

    received all six planned cycles of CHOP every 21 days, with six infusions of

    rituximab at a dose of 375 mg/m2 given before, during, and after chemotherapy.

    Three patients did not complete treatment due to intercurrent infections (n = 2)

    and patient choice (n = 1), and two patients were withdrawn from the study prior to

    therapy. The overall response rate was 95% (38/40), with 55% complete responsesand 40% partial responses. Fewer complete responses were noted in patients with

    bulky disease. Median response duration and time to progression had not been

    reached after 29+ months of follow-up. Seven of 8 patients who had initially

    been positive for the bcl-2 translocation became negative for the translocation

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    9/29

    CANCER ANTIBODIES 351

    by PCR assay after therapy; this has not been seen with CHOP chemotherapy

    alone (44). A preliminary analysis of a randomized study in elderly patients with

    diffuse large-cell lymphoma comparing standard CHOP chemotherapy to CHOP

    with rituximab demonstrated a 76% complete response rate in the combinationarm compared with 60% complete response rate in the chemotherapy arm without

    significant differences in toxicity between the two groups (40). Furthermore, the

    addition of rituximab prolonged event-free survival and overall survival.

    Rituximab also has been evaluated in conjunction with fludarabine in low-grade

    lymphomas. Significant leukopenias were associated with thiscombination, requir-

    ing a dose reduction in the fludarabine. An interim analysis after accruing 30 of the

    40 planned patients revealed a response rate of 93% with median response dura-

    tion of over 14 months. Cytogenetic responses were also observed in some patients

    (45). The significance of such responses is unclear. In a study of rituximab withCHOP chemotherapy in patients with mantle cell lymphoma, loss of a cytogenetic

    marker was not associated with improved progression-free survival (46).

    Another setting in which rituximab has been tested is following high-dose

    chemotherapy with autologous transplantation (47). Patients with at least a 75%

    reduction in tumor volume following CHOP chemotherapy underwent high-dose

    chemotherapy with stem cell transplantation. Two and six months following trans-

    plant, patients received four weekly treatments with rituximab. A preliminary

    report of this study describes four patients who have received one post-transplant

    cycle and eight who have received two post-transplant cycles. Two patients are re-ported with a partial response and another two patients with unconfirmed complete

    response.

    Circulating CD20-positive cells, including lymphoma cells, may affect the ef-

    ficacy of rituximab. Peak levels of circulating antibody inversely correlate with

    pretreatment B cell counts as well as the bulk of tumor (38, 39). Greater numbers

    of peripheral lymphocytes and/or tumor bulk serve as an antigen sink, removing

    antibody from the circulation. For patients with bulky disease, a higher antibody

    dose or a greater number of cycles may be warranted, since patients with lower

    serum rituximab concentrations have had statistically significant lower responserates. A dosage of eight cycles of weekly rituximab has been used safely in low-

    grade and follicular non-Hodgkins lymphoma (48). The efficacy of rituximab in

    chronic lymphocytic leukemia is clearly affected by lower circulative levels of

    the antibody. Initial studies revealed a 20% response rate. Chronic lymphocytic

    leukemia has lower antigen density than many lymphomas that express CD20, and

    the cells circulate, acting as an antigen sink. A dose-escalation study demonstrated

    a clear dose-response relationship (49).

    Rituximab therapy rarely selects for the emergence of an antigen-negative pop-

    ulation of tumor cells. At the present time, little is known about mechanisms ofresistance (49a). This phenomenon has been documented in a patient with a follic-

    ular mixed small- and large-cell lymphoma, who was treated with rituximab after

    multiple chemotherapy regimens (50). Approaches to overcome antigen-negative

    populations include combining rituximab with chemotherapy or with antibodies

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    10/29

    352 VON MEHREN ADAMS WEINER

    targeting other antigens expressed by the lymphoma or leukemia being treated.

    For example, chronic lymphocytic leukemia expresses both CD20 and CD52.

    Studies are evaluating combination therapy with both rituxan and CAMPATH-1H

    (51). Other antigens that could target B cell lymphomas and leukemias includeCD22, which is expressed on pro-B and pre-B cell lymphomas, and the ma-

    jor histocompatability class II antigen, HLA-DR, found on B lymphocytes as

    well as macrophages and dendritic cells. Epratuzumab, which targets CD22, and

    apolizumab, which targets a subset of HLA-DR molecules, are currently undergo-

    ing testing (52). Clearly these agents are candidates for combination therapy with

    rituximab.

    In some patients with circulating blood tumor cells, rituximab therapy has

    induced an infusion-related syndrome characterized by fever, rigors, thrombocy-

    topenia, tumor lysis, bronchospasm, and hypoxemia, requiring discontinuationof the antibody infusion. Symptoms typically resolve with supportive care and

    patients may continue further therapy without sequelae (53). Another case report

    documents rapid tumor lysis in a patient with B cell chronic lymphocytic leukemia

    with a pretreatment lymphocytosis of>100 109/liter (54). Occasional severe

    mucocutaneous reactions have been reported, some of which have been fatal.

    These present as paraneoplastic pemphigus, Stevens-Johnson syndrome, and toxic

    epidermal necrolysis (55).

    Solid Tumor Antigens

    HER-2/NEU HER-2/neu(c-erbB-2), a member of the epidermal growth factor re-

    ceptor (EGFR) family, has been targeted for antibody therapy because it is over-

    expressed on 25% of breast cancers, as well as other adenocarcinomas of the

    ovary, prostate, lung, and gastrointestinal tract. Trastuzumab (56, 57), also known

    as Herceptin and rhuMAb HER2, is a humanized antibody derived from 4D5, a

    murineMAb, which recognizes an epitope on the extracellular domain of HER-

    2/neu. A phase II trial in women with metastatic breast cancer reported an objec-

    tive response rate of 11.6%, with responses seen in the liver, mediastinum, lymph

    nodes, and chest wall. Patients received ten or more treatments with the antibody,

    and none developed an antibody response against trastuzumab. In a second phase

    II study, 222 women with metastatic breast cancer were treated with 2 mg/kg of

    trastuzumab weekly, with an objective response rate of 16% (56). The median

    response duration was 9.1 months, with a median overall survival of 13 months,

    both of which are superior to outcomes reported for second-line chemotherapy in

    metastatic disease. In each of these trials, about 30% of the patients had stable dis-

    ease, lasting more than 5 months. Overexpression of HER-2/neuthat is associated

    with gene amplification correlates with a clinical response to trastuzumab. Earlier

    studies utilized variable criteria to define HER2 positivity that may account for

    the differences in the response rates. The standard assay for determining HER2

    expression is the Hercept test. Breast tumors with 3+ expression by this assay

    correlate with gene overexpression; those with 2+ expression require testing by

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    11/29

    CANCER ANTIBODIES 353

    fluorescence in situ hybridization to confirm gene amplification. Intriguingly, pre-

    clinical studies have demonstrated decreased expression of vascular endothelial

    cell growth factor (VEGF) and vascular permeability factor with 4D5 therapy, sug-

    gesting that an antiangiogenesis mechanism may account for some of the clinicalimpact of this antibody (58). Trastuzumab continues to be evaluated clinically in

    diverse adenocarcinoma types.

    A large, randomized phase III trial evaluating cytotoxic chemotherapy alone

    and with trastuzumab has shown the efficacy of combination therapy (59). Patients

    receiving initial therapy for metastatic breast cancer were treated with cyclophos-

    phamide plus doxorubicin or epirubicin, or with paclitaxel if they had received

    an anthracycline in the adjuvant setting. Patients were randomized to receive this

    chemotherapy alone or in combination with weekly antibody therapy. Response

    rates for combination therapy with an anthracycline regimen increased from 43%to 52% with the addition of trastuzumab. Using a taxane regimen, response rates

    increased from 16% to 42% with the addition of trastuzumab. In addition, there

    was evidence that the addition of trastuzumab to chemotherapy improved sur-

    vival at one year by 16% (60) and improved survival at 29 months by 25% (61).

    Myocardial dysfunction was observed more frequently in patients receiving dox-

    orubicin or epirubicin when trastuzumab was added. Therefore, trastuzumab is not

    recommended in combination with anthracyclines.

    Based on these clinical trial results, trastuzumab was approved by the FDA

    to treat women with metastatic breast cancer with HER-2/neu overexpression,given either alone or in combination with paclitaxel. Herceptin also has activity in

    combination with vinorelbine (62), docetaxel, cisplatin (63), and the combination

    of gemcitabine and paclitaxel (64). In addition, breast cancer patients with lymph

    node involvement whose cancers overexpress HER-2/neu are candidates for partic-

    ipation in a randomized phase III trial evaluating standard adjuvant chemotherapy

    with or without trastuzumab.

    The use of Herceptin for breast cancer has been a therapeutic success. Al-

    though other adenocarcinomas may overexpress HER-2/neu, clinical trials have

    not documented consistent therapeutic successes in other cancers. The Gyneco-logic Oncology Group evaluated Herceptin in patients with recurrent or refractory

    ovarian or primary peritoneal carcinoma (65). An overall response rate of 7.3% was

    found with a median treatment of 8 weeks and median progression-free interval of

    2 months. HER-2/neuhas also been found on prostate cancer and nonsmall-cell

    lung cancer.

    Epidermal Growth Factor Receptor

    EGFR is overexpressed on many cancers. The receptor and its ligands, epidermal

    growth factor (EGF) and transforming growth factoralpha (TGF), act in an au-

    tocrine loop to stimulate the growth of breast cancer cells. In vitro, some anti-EGFR

    antibodies have been shown to inhibit the binding of the receptor ligands (66, 67).

    Antibodies that block the binding of EGFR ligands limit receptor activation by

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    12/29

    354 VON MEHREN ADAMS WEINER

    tyrosine kinases and inhibit growth of normal fibroblasts (68) as well as tumor cells

    in culture (69). Also, combining anti-EGFR antibodies with cisplatin significantly

    decreases the IC50of cisplatin (70), and cures of established tumors have occurred

    when anti-EGFR antibodies are combined with cisplatin (71) or doxorubicin (72).Sequential treatment with topotecan followed by C225 (a chimerized version of the

    anti-EGFR antibody MAb225) in vitro leads to supra-additive growth inhibition in

    breast, ovary, and colon cancer cell lines (73). C225 delivered simultaneously with

    radiation reduces phosphorylation of the EGFR and STAT-3, enhancing apoptosis

    (74). In vitro and in vivo studies suggest that anti-EGFR antibodies may lead to

    terminal differentiation of squamous cell carcinoma cells, with accumulation of

    cells in G0G1 phases of the cell cycle and expression of cell surface markers such

    as involucrin and cytokeratin-10 (75).

    MAb225 blocks in vitro phosphorylation of the EGFR and induces receptor in-ternalization, as occurs with binding of the natural ligand (76). However, receptor

    processing is slower with antibody engagement than with natural ligand engage-

    ment (77). Smaller bivalent F(ab)2and univalent Fab forms of this antibody also

    inhibit growth and decrease receptor phosphorylation, although the bivalent form

    is superior to the monovalent form (78). Since the smaller fragments lead to tumor

    regressions, the efficacy of antibody therapy is not dependent on ADCC, as these

    smaller fragments lack the Fc portion of the antibody required for ADCC. Rather,

    the efficacy of MAb225 is due to its ability to inhibit binding of the natural ligand,

    limit receptor phosphorylation and thus downstream signals, and induce receptorinternalization.

    The chimeric form of MAb225, C225, has been evaluated in vitro and in vivo in

    hormone-sensitive and hormone-refractory prostate cancer (79). EGF is a strong

    chemoattractant for prostate cancer cells. Blocking the EGFR receptor with C225

    in vitro decreases migration of prostate cancer cells in a dose-dependent manner by

    decreasing phosphorylation of the EGFR (80). C225 has also been shown to cause

    cell cycle arrest and decrease proliferation of prostate cancer cells (81, 82). The

    binding of C225 to the EGFR results in multiple events that decrease proliferation

    and possibly metastatic potential in prostate cancer. It has also been shown toinhibit the expression of VEGF and vascular permeability factor, both of which

    are involved in the induction of angiogenesis (59).

    Phase I clinical trials of C225 alone or in combination with cisplatin have been

    conducted in patients with cancers overexpressing EGFR (83). Skin toxicity in

    the form of flushing, seborrheic dermatitis, and acneiform rash were observed at

    doses higher than 100 mg/m2. The rash commonly presents on the head, neck,

    and trunk and resolves without scarring once the treatment has been discontinued

    (84). Epiglottitis resulting in severe shortness of breath was noted when C225

    was combined with cisplatin at 100 mg/m2, but not at 60 mg/m2 (83). Anotherphase I study, in which patients with recurrent head and neck cancer received

    cisplatin every three weeks and C225 weekly, found no such toxicity (85). Of the

    9 evaluable patients in that study, 2 patients previously treated with cisplatin had

    complete responses and an additional 4 had partial responses. In addition, tumor

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    13/29

    CANCER ANTIBODIES 355

    biopsies demonstrated a dose-dependent saturation of the EGFR and loss of EGFR

    tyrosine kinase activity following repeated doses of the antibody. Phase III trials

    are now evaluating the impact of adding C225 to cisplatin in patients with recurrent

    or metastatic head and neck cancer and the benefit of adding C225 to radiationtherapy in patients with locally advanced head and neck cancer. Studies are also

    ongoing in patients with nonsmall-cell lung cancer, who receive C225 along with

    various chemotherapy regimens including carboplatin and paclitaxel, gemcitabine

    and carboplatin, and docetaxel (84).

    ABX-EGF, another antibody that targets EGFR, is also under clinical develop-

    ment. This antibody was produced in a transgenic mouse in which murine antibody

    genes were inactivated and replaced by genes that encode the human sequences

    (86). The resulting fully human antibody has been shown to cause inhibition of

    the tyrosine phosphorylation of the receptor and internalization of the receptor.The inhibitory concentration of the antibody is lower than that of MAb225, the

    parent antibody of C225. Preclinical studies in animals have shown eradication

    of EGFR tumors with ABX-EGF antibody therapy alone, whereas similar studies

    with C225 have required the addition of chemotherapy to produce similar results

    (87). ABX-EGF is currently in clinical trials.

    Ep-CAM (EGP-2/GA 733-2)

    The 17-1A antibody, which recognizes Ep-CAM, has undergone extensive clinicaltesting, with some studies suggesting efficacy in colorectal carcinomas. The murine

    antibody has been replaced by a human chimeric construct that offers increased

    mononuclear cell-mediated ADCC (88), a longer half-life, no development of hu-

    man antimouse antibody (HAMA), and radiolocalization to known sites of disease

    (89). Clinical trials have also incorporated cytokines because of in vitro data that

    demonstrate improved target cell killing by apoptosis with interferon- (90) and

    increased ADCC with interferon- (91), GM-CSF (92, 93), the combination of

    GM-CSF and interleukin (IL)-2 (94), IL-4 (95), and IL-8 (96). These studies have

    formed the basis for clinical trials incorporating the antibody and cytokines suchas interferon- (97) and GM-CSF (98). Although these studies have shown ev-

    idence for cytokine immunomodulation, no consistent pattern of clinical benefit

    has emerged from therapy with these combinations.

    The therapeutic use of 17-1A has also been shown to induce potentially effective

    anti-idiotypic antibodies (99101), an effect that was increased by concomitant

    therapy with GM-CSF (102), and to increase infiltration of macrophages and of

    CD4+ and CD8+ T cells within tumors (103). Induction of T cells against anti-

    idiotypic epitopes has also been evaluated (104). Five of ten patients with anti-

    idiotypic antibodies were also found to have induction of EP-CAM antigen-specificT cells. Four of these patients showed a clinical response and their T cells demon-

    strated a proliferative response in vitro when stimulated with an anti-idiotypic

    antibody, in contrast to the six patients without evidence of T cells against anti-

    idiotypic epitopes.

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    14/29

    356 VON MEHREN ADAMS WEINER

    Initial phase I studies with 17-1A yielded promising results. Several studies

    demonstrated responses in patients with metastatic cancers of the gastrointesti-

    nal tract with only one intravenous dose of antibody. One patient received an

    intrahepatic infusion of autologous mononuclear cells mixed with 17-1A, whichresulted in regression of hepatic metastases. Antibody therapy was well tolerated

    with mild nausea, vomiting, or diarrhea. Phase II studies in colon and pancreatic

    cancers were less encouraging (105, 106). Repeat-dose injections and combina-

    tions with cytokines to enhance effector cell number and activity did not result in

    significant response rates, although in vitro assays of patient effector cells revealed

    increased activity with cytokine therapy. Repeat-dose schedules with higher doses

    were theorized to induce tolerance to the murine antibody, but this maneuver had

    no significant impact on clinical response (106). Some trials reported evidence

    of induction of anti-idiotypic antibodies, correlating with clinical response in onetrial. The overall lack of efficacy seen in these studies may have resulted from the

    large tumor burden or the associated immunosuppression in these patients with

    metastatic disease.

    The initial study evaluating 17-1A in the adjuvant setting suggested possible

    efficacy. A phase III clinical trial of patients with lymph-nodepositive colorectal

    cancer randomized patients to observation or therapy with 17-1A. The surgical

    approach was standardized and agreed to by all participating surgeons. All pa-

    tients were followed post-operatively in a similar manner, irrespective of treat-

    ment. Of 189 patients, 166 were evaluable for overall survival and disease-freesurvival. Therapy with 17-1A was well tolerated except for malaise, low-grade

    fevers and chills, and mild gastrointestinal discomfort. Four anaphylactic reactions

    were treated without sequelae. At five years of follow-up, the death rate was 36%

    in the 17-1A group in contrast to 51% in the observation group, and the calculated

    recurrence rate was 48.7% versus 66.5% (107). At seven years the death rates were

    43% (17-1A) and 63% (observation), and the calculated recurrence rates were 52%

    and 68%, respectively, demonstrating a continued benefit in patients who received

    17-1A (108). Treatment with 17-1A was associated with a decreased incidence

    of metastatic disease but did not alter the incidence of local failure. This shiftin failure pattern was felt to represent the ability of 17-1A to eradicate isolated

    metastatic cancer cells but not bulkier disease. Alternatively, altered vasculature

    due to surgery and scar tissue may have limited antibody diffusion to tumor cells.

    Another factor potentially accounting for the apparent lack of efficacy of 17-1A

    on local control is that 11 patients in the observation group received pre- or post-

    operative radiation therapy alone or in combination with chemotherapy. This trial

    has been criticized because of higher rates of recurrence and death in the control

    arm than would be anticipated. However, it is intriguing because it demonstrates

    an effect of antibody-based therapy in the adjuvant setting of colorectal cancer.Recent clinical trials were designed to confirm these results in stage II colon

    cancers and to test the value of adding 17-1A to standard chemotherapy in patients

    with stage III disease. Therapy with antibody alone is unlikely to be effective in all

    patients owing to the antigenic heterogeneity of cancer cells. Combining standard

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    15/29

    CANCER ANTIBODIES 357

    adjuvant therapy with 17-1A would introduce therapies that have different mech-

    anisms of action, are cell-cycledependent and -independent, and allow death of

    cancer cells irrespective of antigen expression patterns. A preliminary report of

    the phase III trial of 17-1A versus no therapy in the adjuvant setting revealed nodifference in overall survival in patients with stage II colorectal at a median follow

    up of 13.4 months (109); a randomized study of chemotherapy with or without the

    antibody in patients with stage III disease has shown a minimal benefit of ques-

    tionable significance in a preliminary analysis. Overall survival was 81.6% for

    combination therapy versus 78.9% for chemotherapy alone; this was not a statisti-

    cally significant difference. Irrespective of the final analyses of these large studies,

    the value of treating colorectal cancer patients with this antibody is unproven and

    likely to be low.

    Vascular Endothelial Growth Factor

    Molecules that are selectively expressed in the tissue matrix surrounding tumor

    are potentially important targets for antibody therapy. The first of these targets

    to be exploited resides in the vasculature that feeds tumor cells. Bevacizumab is

    a humanized MAb that blocks binding of the vascular endothelial growth factor

    (VEGF) to its receptor on vascular endothelium. VEGF is produced by many

    cancers to stimulate the growth of new blood vessels, and in some studies its

    expression at tumor sites is correlated with risk for metastases. A phase I clinicalstudy of bevacizumab tested doses of 0.110 mg/kg infused on days 1, 28, 35, and

    42 (110). No severe toxicities were found. At doses of 310 mg/kg, increases in

    systolic and diastolic blood pressure of>10 mm Hg were noted during therapy

    (110), and subsequent studies have confirmed that hypertension is a toxicity of this

    agent (111). Two patients receiving3 mg/kg experiencedbleeding intotheir tumors.

    One patient with hepatocellular carcinoma bled into a previously undiagnosed

    brain metastasis. Another patient with an extremity sarcoma and lung metastases

    bled into the extremity mass and experienced hemoptysis.

    In a phase II study of bevacizumab combined with carboplatin and paclitaxelin patients with nonsmall-cell lung cancer, 6 of 66 patients developed pulmonary

    hemorrhage, 4 of whom died (112). Patients with squamous cell lung cancers and

    tumors with evidence of cavitation or squamous cell histology appeared to be at

    higher risk of hemoptysis. Interestingly, a study of bevacizumab and chemotherapy

    in metastatic colorectal cancer found an increase in thrombotic episodes as well

    as hemorrhage. The incidence of thrombotic events was higher at the 5 mg/kg

    dose level than at the 10 mg/kg dose level. The majority of cases were deep venous

    thromboses, but one cerebral vascular accident and one pulmonary embolism were

    reported.Clinical trials have suggested a therapeutic benefit from bevacizumab. Although

    the phase I study demonstrated no objective responses, 12 of 23 patients had

    stable disease during the trial (70 days) and an additional patient with renal cell

    carcinoma experienced a minor response (110). A phase II study was conducted

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    16/29

    358 VON MEHREN ADAMS WEINER

    in 25 women whose metastatic breast cancer had progressed following at least one

    anthracycline or taxane-based regimen. Clinical responses were noted at doses of

    3 mg/kg (n = 1/18) and 10 mg/kg (n = 2/17) (111). All responses were in lymph

    nodes or subcutaneous skin nodules. Bevacizumab has also been combined withchemotherapy in patients with nonsmall-cell lung cancer (113) and metastatic

    colorectal cancer (114). The response rate and time to progression in patients

    with lung cancer improved when carboplatin and paclitaxel were combined with

    bevacizumab at 10 mg/kg, but not at 5 mg/kg. A similar result was found in patients

    with colorectal cancer receiving 5-fluorouracil and folinic acid plus bevacizumab,

    but the improved response rate was seen at the 5 mg/kg dose. Time to progression

    improved at the both 5 mg/kg and 10 mg/kg, but to a greater extent at 5 mg/kg.

    RADIOIMMUNOTHERAPY

    Radioimmunoconjugates

    A tumor-specific antibody that does not alter tumor growth properties can be

    armed with a radioisotope to create a cytotoxic agent. Even antibodies that are

    effective antitumor agents in an unconjugated state can have enhanced efficacy

    with the addition of a cytotoxic radioisotope. For example, although therapy with

    the anti-CD20 MAb rituximab is associated with a significant antitumor effect

    in patients with non-Hodgkins lymphoma, significantly more patients exhibitcomplete responses when the 90Y parent MAb Y2B8 is added to the treatment

    protocol (115). The recent availability of a panel of antibody-based constructs with

    a range of in vivo half-lives now makes it possible to select antibody/radioisotope

    combinations with matched biologic and physical half-lives. This type of pairing

    enables the majority of the decay, and therefore cytotoxic emissions, to occur

    while the greatest quantity of labeled antibody is localized in the tumor. Williams

    et al. developed formulas, termed the imaging figure of merit (IFOM) and therapy

    figure of merit (TFOM), to determine which isotopes best pair with a series of

    antibodies and fragments based on the rate of MAb clearance, isotope half-life, and

    emission track length (116). This approach can be used to optimize the efficiency

    and specificity of an imaging or treatment system.

    Until recently, most RAIT studies employed 131I, but improvements in labeling

    strategies and the availability of large quantities of radiopharmaceutical-grade 90Y

    have enabled the study of 90Y conjugates. Now, a wide variety of therapeutic

    radioisotopes are available for RAIT. They fall into two general categories, those

    that emit shorttrack-length (e.g., one cell length) alpha particles with a high

    linear energy transfer (LET) and those that emit longertrack-length (e.g., 50 cell

    lengths) beta particles with a low LET. Because the LET is a measure of the energy

    deposited along the track of the emitted particle, a high LET is associated with a

    greater probability of tumor cell death. This is in part dictated by the difference

    in the size of the particles. An alpha particle is equivalent to a helium nucleus

    whereas a beta particle is an electron. Alpha particles tend to kill cells by causing

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    17/29

    CANCER ANTIBODIES 359

    irreparable double-strand DNA breaks; beta particles lead to single-strand DNA

    breaks.

    Of the available beta-emitting radioisotopes, 90Y (t1/2 = 64 h) is rapidly gaining

    dominance in most RAIT protocols owing to its ease of labeling and absence of agamma emission. The other commonly used radionuclide, 131I (t1/2 = 8.02 days),

    requires a trained radiopharmacist and appropriately shielded/vented radiophar-

    macy because of its volatile nature. Other beta-emitting radioisotopes that have

    shown promise for radioimmunotherapy include 67Cu (t1/2 = 61.8 h) and 177Lu

    (t1/2 = 6.73 days). There are also a number of alpha-emitting radioisotopes that are

    potential partners with antibody-based molecules for RAIT. These include 213Bi

    (t1/2 = 45 min), 212Bi (t1/2 = 61 min), and

    211At (t1/2 = 7.2 h). Whereas the

    half-life of 211At is compatible with the pharmacokinetics of several engineered

    antibody-based molecules, the very short half-lives of the bismuth radioisotopeslimit their utility to the setting of disseminated disease or to pretargeted RAIT

    (described below).

    Some clinical trials have demonstrated partial, short-lived clinical responses in

    some patients with advanced, solid tumors, but most of the significant advances in

    RAIT have occurred in the treatment of hematologic neoplasms. Lymphomas and

    leukemias remain the most sensitive tumor targets for RAIT, presumably because

    of their intrinsic sensitivity to radiation and the relatively good access of radioim-

    munoconjugates to the malignant cells that comprise these neoplasms. Patients

    with hematologic neoplasms are also less likely to mount HAMA responses whenforeign MAbs are employed.

    Radioimmunotherapy for Hematologic Malignancies

    B-1 The CD20 antigen has been a very effective target for RAIT. Press and col-

    leagues used high, marrow-ablative doses of 131I-B1 (tositumomab or Bexxar)

    anti-CD20 RAIT in the setting of autologous bone marrow transplantation. In a

    total of 42 patients with chemotherapy-refractory lymphomas, 24 exhibited MAb

    distributions that were predictive of a favorable response and 19 received high-

    dose RAIT. Of the RAIT group, 84% exhibited a complete remission, and 11%

    had a partial remission (117). Many of the complete remissions observed with this

    approach have proven to be durable, with 62% exhibiting progression-free survival

    at 2 years. Low-dose, non-myeloablative RAIT with 131I-B1 has also proven effi-

    cacious, showing 50% response rates, with a median duration of 16.5 months, in

    patients with chemotherapy-refractory B cell lymphoma (118, 119). Preliminary

    studies have also shown that RAIT may be useful as a conditioning regimen prior to

    bone marrow transplantation in patients with acute myelogenous leukemia (120).

    IBRITUMOMAB TIUXETAN Ibritumomab tiuxetan (Zevalin) is another MAb that

    targets an epitope on the CD20 antigen that is distinct from the B1 epitope. Another

    distinction between the ibritumomab trials and the tositumomab trials described

    above is that Ibritumomab employs 90Y in place of131I. This antibody is the first,

    and to date only, MAb licensed for use in RAIT in the United States.

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    18/29

    360 VON MEHREN ADAMS WEINER

    A number of clinical trials have evaluated ibritumomab in over 230 patients

    with relapsed or refractory low- or intermediate-grade non-Hodgkins lymphoma

    (reviewed in Reference 121). The most comprehensive study was a randomized

    phase III trial that compared the safety and efficacy of a single dose of ibritumomabwith a standard treatment course of rituximab in 143 patients with low-grade, fol-

    licular or transformed non-Hodgkins lymphoma. In this trial, an overall response

    rate of 80% and a complete response rate of 30% was observed in the group treated

    with ibritumomab versus an overall response rate of 56% and a complete response

    rate of 16% in the group treated with rituximab (122).

    LYM-1 Lym-1 is specific for a human leukocyte antigen (HLA-DR) expressed

    in >95% of B cell tumors. This murine MAb has not been humanized. In phase

    I/II clinical trials,131

    I-Lym-1 has shown significant antitumor activity. Low-dose,fractionated treatment of 30 patients (25 with non-Hodgkins lymphoma and 5

    with chronic lymphocytic leukemia) resulted in 3 complete responses and 14 partial

    responses (123). When treated at the non-myeloablative maxiumum tolerated dose

    of 100 mCi/m2, 11 of 21 patients responded favorably (7 complete responses, 4

    partial responses) (124).

    M195 The cell surface antigen CD33 is expressed on most myeloid leukemic blasts

    and leukemic progenitor cells. Its normal tissue expression is limited to committed

    normal myelomonocytic and erythroid progenitor cells and (at low levels) earlyhematopoietic stem cells. M195, a murine anti-CD33 MAb, has been used to de-

    liver therapeutic doses of131I in combination with busulfan or cyclophosphamide

    to eliminate disease before bone marrow transplantation (125). HuM195, a hu-

    manized version of M195, has been employed as a vehicle for the RAIT of acute

    and chronic myelogenous leukemia. HuM195 RAIT resulted in minor responses

    in 8 of 12 patients treated with 90Y-conjugated Mab and 13 of 18 patients treated

    with 213Bi-conjugated Mab (126, 127). The 213Bi studies were the first use of alpha

    particles in RAIT.

    Radioimmunotherapy of Solid Malignancies

    Significantly less progress has been made in the treatment of solid malignancies

    with RAIT. In most reported trials in patients with solid tumors (e.g., breast car-

    cinoma, colon carcinoma, ovarian carcinoma), RAIT is associated with, at best,

    stable disease. Although reports of a greater response do appear, their rarity gives

    them the aura of an Elvis sighting. A phase II trial of 75 mCi/m2 of 131I-labeled

    CC49 MAb with interferon (to enhance target antigen expression) in patients with

    metastatic prostate cancer reported minor radiographic responses and relief of pain

    (128). Similar results were reported for a phase I study of 90Y-2IT-BAD-M170 in

    patients with androgen-independent metastatic prostate cancer (129). In a recent

    phase I trial in patients with recurrent or persistent ovarian cancer, intraperitoneal177Lu-labeled CC49 MAb, plus interferon and Taxolled to partial responses in 4 of

    17 treated patients and progression-free intervals in patients without measurable

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    19/29

    CANCER ANTIBODIES 361

    disease (130). Transient clinical responses were also observed in a phase I trial in

    metastatic breast cancer patients treated with 90Y-BrE-3 (131). Still, several new

    agents are currently undergoing clinical evaluation, many of which have novel char-

    acteristics (e.g., domain deletions or significant antitumor properties as a nakedMAb) that may translate into improved results.

    Pretargeted Radioimmunotherapy

    One new RAIT strategy recently examined in the clinic is pretargeted radioim-

    munotherapy (PRIT). PRIT differs from traditional RAIT in that the antibody and

    the radioisotope are delivered to the tumor in separate steps. In the PRIT protocols

    employed to date, MAb-streptavidin (MAb-SA) or MAb-biotin (MAb-B) conju-

    gates were administered to the patient. After the MAb conjugate localized in the tu-

    mor, a clearing agent [e.g., a biotingalactosehuman serum albumin conjugate

    or streptavidin (SA)] is administered that binds to the circulating MAb conjugate

    and directs its clearance by the liver. After an additional 24 h, a biotin-chelate-90Y

    complex is administered that binds specifically to the MAb-SA or MAB-B-SA

    prelocalized in the tumor.

    PRIT clinical trials have been performed in patients with glioma (132), colon

    cancer (133), prostate cancer, and non-Hodgkins lymphoma (134). Of 48 glioma

    patients treated by PRIT at doses of 6080 mCi/m2, 25% (12 patients) exhibited

    a reduction in tumor mass ranging from>25% to 100%, with 8 responses lastinglonger than one year (132). In the PRIT treatment of patients with colon can-

    cer, 8% (2 of 25) of the patients treated with pretargeted NR-LU-10 MAb and

    110 mCi/m2 of90Y-DOTA-biotin had a partial response, and 16% (4 of 25) exhib-

    ited stable disease. However, significant hematologic and nonhematologic toxic-

    ities (diarrhea) were reported (133). A companion trial directed against prostate

    cancer revealed similar toxicities (I. Horak, personal communication). Signifi-

    cantly more success was observed in the PRIT of non-Hodgkins lymphoma. In

    this trial, rituximab, which has antitumor properties in an unconjugated form, was

    conjugated to SA as the first-step reagent. Six of seven patients treated with dosesof 30 or 50 mCi/m2 of90Y-DOTA-biotin exhibited objective regressions, with one

    partial response and three complete responses (134). The toxicities reported in this

    trial were also less severe and were limited to grade I/II nonhematologic toxicity

    and transient grade III hematologic toxicity.

    CONCLUSIONS

    Therapeutic antibodies have made the transition from concept to clinical real-

    ity over the past two decades. Many are now being tested as adjuvant or first-line

    therapies to assess their efficacy in improving or prolonging survival. Modified an-

    tibody structures, such as bispecific antibodies or smaller antibody fragments, have

    yet to claim defined therapeutic roles, whereas radioimmunotherapy has clearly

    demonstrated efficacy in some disease settings.

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    20/29

    362 VON MEHREN ADAMS WEINER

    TheAnnual Review of Medicineis online at http://med.annualreviews.org

    LITERATURE CITED

    1. Sivolapenko GB, Douli V, Pectasides D,

    et al. 1995. Breast cancer imaging with

    radiolabelled peptide from complemen-

    tarity-determining region of antitumor an-

    tibody.Lancet346:166266

    2. Le Gall F, Kipriyanov SM, Moldenhauer

    G, Little M. 1999. Di-, tri- and tetrameric

    single chain Fv antibody fragments

    against human CD19: effect of valency on

    cell binding.FEBS Lett.453:164683. Ward ES, Gussow D, Griffiths AD, et al.

    1989. Binding activities of a repertoire of

    single immunoglobulin variable domains

    secreted from Escherichia coli. Nature

    341:54446

    4. Huston JS, Levinson D, Mudgett-Hunter

    M, et al. 1988. Protein engineering of an-

    tibody binding sites: recovery of specific

    activity in an anti-digoxin single-chain Fv

    analogue produced in E coli. Proc. Natl.

    Acad. Sci. USA85:587983

    5. Marks JD, Hoogenboom HR, Bonnert

    TP, et al. 1991. By-passing immuniza-

    tion. Human antibodies from V-gene li-

    braries displayed on phage. J. Mol. Biol.

    222:58197

    6. Begent RH, Verhaar MJ, Chester KA,

    et al. 1996. Clinical evidence of efficient

    tumor targeting based on single-chain Fvantibody selected from a combinatorial li-

    brary.Nat. Med.2:97984

    7. Adams GP, McCartney JE, Tai MS, et al.

    1993. Highly specific in vivo tumor tar-

    geting by monovalent and divalent forms

    of 741F8 anti-c-erbB-2 single-chain Fv.

    Cancer Res.53:402634

    8. Holliger P, Prospero T, Winter G. 1993.

    Diabodies: small bivalent and bispecific

    antibody fragments. Proc. Natl. Acad. Sci.USA90:644448

    9. Hu SSL, Raubitschek A, Sherman M,

    et al. 1996. Minibody: a novel engineered

    anti-carcinoembryonic antigen antibody

    fragment (single-chain Fv-CH3) which

    exhibits rapid, high-level targeting of

    xenografts. Cancer Res.56:305561

    10. Jain RK. 1990. Physiological barriers to

    delivery of monoclonal antibodies and

    other macromolecules in tumors. Cancer

    Res.(Suppl.) 50:814s19s

    11. Jain RK, Baxter LT. 1988. Mechanisms of

    heterogeneous distribution of monoclonal

    antibodies and other macromolecules in

    tumors: significance of elevated intersti-tial pressure.Cancer Res.48:702232

    12. Trauth B, Klas C, Peters A, et al. 1989.

    Monoclonal antibody-mediated tumor re-

    gression by induction of apoptosis. Sci-

    ence245:3015

    13. Waldman T. 1991. Monoclonal antibod-

    ies in diagnosis and therapy. Science

    252:38794

    14. OConnell M, Chen Z, Yang H, et al. 1989.

    Active specific immunotherapy with an-

    tiidiotypic antibodies in patients with

    solid tumors.Semin. Surg. Oncol.5:441

    47

    15. Houghton A, Mintzer D, Cordon-Cardo

    C, et al. 1985. Mouse monoclonal IgG3

    antibody detecting GD3 ganglioside: a

    phase I trial in patients with malignant

    melanoma. Proc. Natl. Acad. Sci. USA

    82:12424616. Steplewski Z, Lubeck M, Koprowski H.

    1983. Human macrophages armed with

    murine immunoglobulin G2a antibodies

    to tumors destroy human cancer cells. Sci-

    ence221:86567

    17. Lindenmann J. 1973. Speculations on Ids

    and homobodies. Ann. Immunol. 124:

    17184

    18. Jerne N. 1974. Towards a network the-

    ory of the immune system.Ann. Immunol.125:37389

    19. Miller R, Maloney D, Warnke R, Levy

    R. 1982. Treatment of B-cell lymphoma

    with monocolonal anti-idiotype antibody.

    N. Engl. J. Med. 306:51722

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    21/29

    CANCER ANTIBODIES 363

    20. Khazaeli M, Conry R, LoBuglio A. 1994.

    Human immune responses to monoclonal

    antibodies.J. Immunother.15:4252

    21. Peterson J, Couto J, Taylor M, Ceri-

    ani R. 1995. Selection of tumor-specific

    epitopes on target antigens for radioim-

    munotherapy of breast cancer. Cancer

    Res.55:5847s51s

    22. Badger C, Anasetti C, Davis J, Bern-

    stein I. 1987. Treatment of malignancy

    with unmodified antibody. Pathol. Im-

    munopathol. Res.6:41934

    23. Holland G, Zlotnik A. 1993. Interleukin-

    10 and cancer.Cancer Invest.11:7515824. Norgaard P, Hougaard S, Poulsen H,

    Thomsen M. 1995. Transforming growth

    factor and cancer. Cancer Treat. Rev.

    21:367403

    25. Lee J, Fenton BM, Koch CJ, et al. 1998.

    Interleukin 2 expression by tumor cells al-

    ters both the immune response and the

    tumor microenvironment. Cancer Res.

    58:147885

    26. Nadler L, Stashenko P, Hardy R, et al.1980. Serotherapy of a patient with a mon-

    oclonal antibody directed against a human

    lymphoma-associated antigen. Cancer

    Res.40:314754

    27. Vuist W, Levy R, Maloney D. 1994. Lym-

    phoma regression induced by monoclonal

    anti-idiotypic antibodies correlates with

    their ability to induce Ig signal transduc-

    tion and is not prevented by tumor expres-

    sion of high levels of bcl-2 protein.Blood83:899906

    28. Swisher E, Shawler D, Collins H, et al.

    1991. Expression of shared idiotypes in

    chronic lymphocytic leukemia and small

    lymphocytic lymphoma.Blood77:1977

    82

    29. Bowen A, Zomas A, Emmett E, et al.

    1997. Subcutaneous CAMPATH-1H in

    fludarabine-resistant/relapsed chronic

    lymphocytic and B-prolymphocytic leuk-

    aemia.Br. J. Haematol.96:61719

    30. Lundin J, Osterborg A, Brittinger G, et al.

    1998. CAMPATH-1H monoclonal anti-

    body in therapy for previously treated

    low-grade non-Hodgkins lymphomas: a

    phase II multicenter study. European

    Study Group of CAMPATH-1H Treat-

    ment in Low-Grade Non-Hodgkins Lym-

    phoma. J. Clin. Oncol.16:325763

    31. Hale G, Zhang M, Bunjes D, et al. 1998.

    Improving the outcome of bone marrow

    transplantation by using CD52 mono-

    clonal antibodies to prevent graft-versus-

    host disease and graft rejection. Blood

    92:458190

    32. Naparstek E, Delukina M, Or R, et al.

    1999. Engraftment of marrow allografts

    treated with Campath-1 monoclonal anti-bodies.Exp. Hematol.27:121018

    33. Novitzky N, Thomas V, Hale G, Wald-

    mann H. 1999. Ex vivo depletion of

    T cells from bone marrow grafts with

    CAMPATH-1 in acute leukemia: graft-

    versus-host disease and graft-versus-

    leukemia effect.Transplantation67:620

    26

    34. Hale G, Waldmann H. 1998. Risks of de-

    veloping Epstein-Barr virusrelated lym-phoproliferative disorders after T-cell

    depleted marrow transplants. CAMPATH

    Users.Blood91:307983

    35. Maloney D, Grillo-Lopez A, Bodkin D,

    et al. 1997. IDEC-C2B8: results of a phase

    I multiple-dose trial in patients with re-

    lapsed non-Hodgkins lymphoma.J. Clin.

    Oncol.15:326674

    36. Maloney D, Grillo-Lopez A, White C,

    1997. IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in

    patients with relapsed low-grade non-

    Hodgkins lymphoma.Blood90:218895

    37. Shan D, Ledbetter J, Press O. 2000.

    Signaling events involved in anti-CD20

    induced apoptosis of malignant human

    B cells. Cancer Immunol. Immunother.

    48:67383

    38. McLaughlin P, Grillo-Lopez A, Link B,

    et al. 1998. Rituximab chimeric anti-

    CD20 monoclonal antibody therapy for

    relapsed indolent lymphoma: half of pa-

    tients respond to a four-dose treatment

    program. J. Clin. Oncol.16:282533

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    22/29

    364 VON MEHREN ADAMS WEINER

    39. Berinstein N, Grillo-Lopez A, White

    C, et al. 1998. Association of serum

    Rituximab (IDEC-C2B8) concentration

    and anti-tumor response in the treat-

    ment of recurrent low-grade or follicu-

    lar non-Hodgkins lymphoma. Ann. On-

    col.9:9951001

    40. Coiffier B, Haioun C, Ketterer N, et al.

    1998. Rituximab (anti-CD20 monoclonal

    antibody) for the treatment of patients

    with relapsing or refractory aggressive

    lymphoma: a multicenter phase II study.

    Blood92:192732

    41. Heinzerling L, Dummer R, Kempf W,et al. 2000. Intralesional therapy with

    antiCD 20 monoclonal antibody ritux-

    imab in primary cutaneous lymphoma.

    Arch. Dermatol.136:37478

    42. Czuczman M, Grillo-Lopez A, White

    C, et al. 1999. Treatment of patients

    with low-grade B-cell lymphoma with the

    combination of chimeric anti-CD20 mon-

    oclonal antibody and CHOP chemother-

    apy.J. Clin. Oncol.17:2687643. Demidem A, Lam T, Alas S, et al. 1997.

    Chimeric anti-CD20 (IDEC-C2B8) mo-

    noclonal antibody sensitizes a B cell lym-

    phoma cell line to cell killing by cyto-

    toxic drugs. Cancer Biother. Radiopharm.

    12:17786

    44. Gribben J, Freedman A, Woo S, et al.

    1991. All advanced stage non-Hodgkins

    lymphomas with a polymerase chain reac-

    tion amplifiable breakpoint of bcl-2 haveresidual cells containing the bcl-2 rear-

    rangement at evaluation and after treat-

    ment.Blood78:327580

    45. Czuczman M. 2002. Rituximab in combi-

    nation with CHOP or fludarabine in low-

    grade lymphoma. Semin. Oncol. 20:36

    40

    46. Howard O, Gribben J, Neuberg D, et al.

    2002. Rituximab and CHOP induction

    therapy for newly diagnosed mantle-

    cell lymphoma: molecular complete re-

    sponses are not predictive of progression-

    free survival.J. Clin. Oncol.20:128894

    47. Mangel J, Buckstein R, Imrie K, et al.

    2002. Immunotherapy with rituximab fol-

    lowing high-dose therapy and autologous

    stem-cell transplantation for mantle cell

    lymphoma. Semin. Oncol.29:5669

    48. Piro L, White C, Grillo-Lopez A, et al.

    1999. Extended rituximab (anti-CD20

    monoclonal antibody) therapy for re-

    lapsed or refractory low-grade or follic-

    ular non-Hodgkins lymphoma.Ann. On-

    col.10:61921

    49. Keating M, OBrien S. 2000. High-dose

    rituximab therapy in chronic lymphocytic

    leukemia.Semin. Oncol.27:8690

    49a. Maloney DG, Smith B, Rose A. 2002. Rit-uximab: mechanisms of action and resis-

    tance.Semin. Oncol.29(1 Suppl. 2):29

    50. Davis T, Czerwinski D, Levy R. 1999.

    Therapy of B-cell lymphoma with anti-

    CD20 antibodies can result in the loss of

    CD20 antigen expression. Clin. Cancer

    Res.5:61115

    51. Nabhan C, Rosen ST. 2002. Concep-

    tual aspects of combining rituximab and

    campath-1H in the treatment of chroniclymphocytic leukemia.Semin. Oncol.29:

    7580

    52. Leonard J, Link B. 2002. Immunotherapy

    of non-Hodgkins lymphoma with hLL2

    (Epratuzumab, an anti-CD22 monoclonal

    antibody) and HuIDI0 (Apolizumab).

    Semin. Oncol.29:8186

    53. Byrd J, Waselenko J, Maneatis T, et al.

    1999. Rituximab therapy in hemato-

    logic malignancy patients with circulat-ing blood tumor cells: association with in-

    creased infusion-related side effects and

    rapid blood tumor clearance. J. Clin. On-

    col.17:79195

    54. Jensen M, Winkler U, Manze O, et al.

    1998. Rapid tumor lysis in a patient with

    B-cell chronic lymphocytic leukemia and

    lymphocytosis treated with an anti-CD20

    monoclonal antibody (IDEC-C2B8, ritux-

    imab).Ann. Hematol.77:8991

    55. Grillo-Lopez A, Hedrick E, Rashford M,

    Benyunes M. 2002. Rituximab: ongoing

    and future clinical development. Semin.

    Oncol.29:10212

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    23/29

    CANCER ANTIBODIES 365

    56. Cobleigh M, Vogel C, Tripathy D, et al.

    1999. Multinational study of the effi-

    cacy 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. Oncol.17:263948

    57. Baselga J, Tripathy D, Mendelsohn J,

    et al. 1996. Phase II study of weekly in-

    travenous recombinant humanized anti-

    p185HER2 monoclonal antibody in pa-

    tients with HER2/neu-overexpressing

    metastatic breast cancer. J. Clin. Oncol.14:73744

    58. Petit A, Rak J, Hung M, et al. 1997.

    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: angio-

    genic implications for signal transduction

    therapy of solid tumors. Am. J. Pathol.

    151:15233059. Slamon D, Leyland-Jones B, Shak S,

    et al. 2001. Use of chemotherapy plus

    a monoclonal antibody against HER2

    for metastatic breast cancer that overex-

    presses HER2.N. Engl. J. Med.344:783

    92

    60. Norton L, Slamon D, Leyland-Jones B.

    1999. Overall survival (OS) advantage to

    simultaneous chemotherapy (CRx) plus

    the humanized anti-HER2 monoclonalantibody Herceptin (H) in HER2-over-

    expressing (HER2+) metastatic breast

    cancer (MBC). Proc. Am. Soc. Clin. On-

    col.18:A127 (Abstr.)

    61. Baselga J. 2001. Clinical trials of

    Herceptin(trastuzumab).Eur. J. Cancer

    37:S18S24

    62. Burstein HJ, Kuter I, Campos SM, et al.

    2001. Clinical activity of trastuzumab

    and vinorelbine in women with HER2-

    overexpressing metastatic breast cancer.

    J. Clin. Oncol.19:272230

    63. Pegram M, Lipton A, Hayes D, et al.

    1998. Phase II study of receptor-en-

    hanced chemosensitivity using re-

    combinant humanized anti-p185her2/neu

    monoclonal antibody plus cisplatin in

    patients with HER2/neu-overexpressing

    metastatic breast cancer refractory to

    chemotherapy treatment. J. Clin. Oncol.

    16:265971

    64. Miller KD, Sisk J, Ansari R, et al. 2001.

    Gemcitabine, paclitaxel, and trastuzumab

    in metastatic breast cancer. Oncology

    15:S38S40

    65. Bookman M, Darcy K, Clarke-Pearson

    D, et al. 2002. Evaluation of mono-

    clonal humanized anti-HER2 antibody(Trastuzumab, Herceptin) in patients with

    recurrent or refractory ovarian or primary

    peritoneal carcinoma with overexpression

    of HER2: a phase II trial of the Gyneco-

    logic Oncology Group.J. Clin. Oncol. 20:

    In press

    66. Modjtahedi H, Komurasaki T, Toyoda H,

    Dean C. 1998. Anti-EGFR monoclonal

    antibodies which act as EGF, TGF alpha,

    HB-EGF, and BTC antagonists block thebinding of epiregulin to EGFR-expressing

    tumours. Int. J. Cancer75:31016

    67. Teramoto T, Onda M, Tokunaga A, Asano

    G. 1996. Inhibitory effect of an anti-

    epidermal growth factor receptor anti-

    body on human gastric cancer. Cancer

    77:163945

    68. Sato J, Kawamoto T, Le A, et al. 1983. Bi-

    ological effects in vitro of monoclonal an-

    tibodies to human epidermal growth fac-tor receptors.Mol. Biol. Med.1:51129

    69. Artega C, Corondo E, Osbore C. 1988.

    Blockade of the epidermal growth factor

    receptor inhibits transforming growth fac-

    tor ainduced but not estrogen-induced

    growth of hormone-dependent human

    breast cancer. Mol. Endocrinol. 2:1064

    69

    70. Hoffman T, Hafner D, Ballo H, et al. 1997.

    Antitumor activity of anti-epidermal

    growth factor receptor monoclonal anti-

    bodies and cisplatin in ten human head

    and neck squamous cell carcinoma lines.

    Anticancer Res.17:441925

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    24/29

    366 VON MEHREN ADAMS WEINER

    71. Fan Z, Baselga J, Masui H, Mendelsohn

    J. 1993. Antitumor effect of anti-epi-

    dermal growth factor receptor monoclo-

    nal antibodies plus cis-diamminedi-

    chloroplatinum on well established A431

    cell xenografts. Cancer Res. 53:4637

    42

    72. Baselga J, Norton L, Masui H, et al.

    1993. Antitumor effects of doxorubicin in

    combination with anti-epidermal growth

    factor receptor monoclonal antibodies. J.

    Natl. Cancer Inst.85:132733

    73. Ciardiello F, Bianco R, Damiano V,

    et al. 1999. Antitumor activity of sequen-tial treatment with topotecan and anti-

    epidermal growth factor receptor mono-

    clonal antibody C225. Clin. Cancer Res.

    5:90916

    74. Bonner J, Raisch K, Robert HT, et al.

    2000. Enhanced apoptosis with combina-

    tion C225/radiation treatment serves as

    the impetus for clinical investigation in

    head and neck cancers.J. Clin. Oncol.18

    (Suppl.):47s53s75. Modjtahedi H, Eccles S, Sandle J, et al.

    1994. Differentiation or immune destruc-

    tion: two pathways for therapy of squa-

    mous cell carcinomas with antibodies

    to the epidermal growth factor receptor.

    Cancer Res.54:1695701

    76. Sunada H, Magun B, Mendelsohn J,

    MacLeod C. 1986. Monoclonal antibody

    against the EGF receptor is internal-

    ized without stimulating receptor phos-phorylation. Proc. Natl. Acad. Sci. USA

    83:382528

    77. Sunada H, Yu P, Peacock J, Mendelsohn

    J. 1990. Modulation of tyrosine serine and

    threonine phosphorylation and intracellu-

    lar processing of the epidermal growth

    factor receptor by anti-receptor mono-

    clonal antibody.J. Cell. Physiol. 142:284

    92

    78. Fan Z, Masui M, Altas I, Mendel-

    sohn J. 1993. Blockade of the epider-

    mal growth factor receptor function by

    bivalent and monovalent fragments of

    225 anti-epidermal growth factor recep-

    tor monoclonal antibodies. Cancer Res.

    53:432228

    79. Prewett M, Rockwell R, Giorgio N, et al.

    1996. The biologic effects of C225, a

    chimeric monoclonal antibody to the

    EGFR, on human prostate carcinoma. J.

    Immunother. Emphasis Tumor Immunol.

    19:41927

    80. Zolfaghari A, Djakiew D. 1996. Inhibition

    of chemomigration of a human prostatic

    carcinoma cell (TSU-pr1) line by inhibi-

    tion of epidermal growth factor receptor

    function.Prostate28:23238

    81. Peng D, Fan Z, Lu Y, et al. 1996. Anti-epidermal growth factor receptor mono-

    clonal antibody 225 up-regulates

    p27KIP1 and induces G1 arrest in pro-

    static cancer cell line DU145. Cancer

    Res.56:366669

    82. Wu X, Rubin M, Fan Z, et al. 1996. In-

    volvement of p27KIP1 in G1 arrest me-

    diated by an anti-epidermal growth factor

    receptor monoclonal antibody.Oncogene

    12:139740383. Baselga J, Pfister D, Cooper M,et al. 2000.

    Phase I studies of anti-epidermal growth

    factor receptor chimeric antibody C225

    alone and in combination with cisplatin.

    J. Clin. Oncol.18:90914

    84. Herbst R, Langer C. 2002. Epidermal

    growth factor receptors as a target for can-

    cer treatment: the emerging role of IMC-

    C225inthetreatmentoflungandheadand

    neck cancers.Semin. Oncol.29:273685. Shin D, Donato N, Perez-Soler R, et al.

    2001. Epidermal growth factor receptor

    targeted therapy with C225 and cisplatin

    in patients with head and neck cancer.

    Clin. Cancer Res.7:120413

    86. Mendez M, Green L, Corvalan J, et al.

    1997. Functional transplant of megabase

    human immunoglobulin loci recapitulates

    human antibody response in mice. Nat.

    Genet.15:14656

    87. Yang X, Jia X, Corvalan J, et al. 1999.

    Eradication of established tumors by a

    fully human monoclonal antibody to the

    epidermal growth factor receptor without

    byb

    on:UniversidadedeLisboa(UL)on02/26/11.Forpersonaluseon

    ly.

  • 8/13/2019 Mehren,2003

    25/29

    CANCER ANTIBODIES 367

    concomitant chemotherapy. Cancer Res.

    59:123643

    88. Haga Y, Sivinski C, Woo D, Tempero

    M. 1994. Dose related comparison of

    antibody-dependent cellular cytotoxicity

    with chimeric and native monoclonal an-

    tibody 17-1A. Improved cytolysis of pan-

    creatic cancer cells with chimeric 17-1A.

    Int. J. Pancreatol.15:4350

    89. Meredith R, LoBuglio A, Plott W, et al.

    1991. Pharmacokinetics, immune respon-

    se, and biodistribution of iodine-131

    labeled chimeric mouse/human IgG1 17-

    1A monoclonal antibody. J. Nucl. Med.32:116268

    90. Takamuku K, Baba K, Aringa S, et al.

    1996. Apoptosis in antibody-dependent

    monocyte-mediated cytotoxicity with

    monoclonal antibody 17-1A against

    human colorectal carcinoma cells: en-

    hancement with interferon gamma. Can-

    cer Immunol. Immunother.43:22025

    91. Reali E, Guiliani A, Spisani S, et al.

    1994. Interferon-gamma enhances mon-oclonal antibody 17-1Adependent neu-

    trophil cytotoxicity toward colorectal car-

    cinoma cell line SW11-16.Clin. Immunol.

    Immunopathol. 71:10512

    92. Ragnhammar P, Masucci G, Frodin J,

    et al. 1992. Cytotoxic functions of blood

    mononuclear cells in patients with col-

    orectal carcinoma treated with mAb-17-

    1A and granulocyte/macrophage colony

    stimulating factor. Cancer Immunol. Im-munother.35:15864

    93. Ragnhammar P, Magnusson I, Masucci G,

    Mellstedt H. 1993. The therapeutic use of

    the unconjugated monoclonal antibodies

    (MAb) 17-1A in combination with GM-

    CSF in the treatment of colorectal carci-

    noma (CRC). Med. Oncol. Tumor Phar-

    macother.10:6170

    94. Masucci G, Ragnhammar P, Wersall P,

    Mellstedt H. 1990. Granulocyte-mono-

    cyte colony-stimulating factor augments

    the interleukin-2 induced cytotoxic activ-

    ity of human lymphocytes in the absence

    and presence of mouse or chimeric mon-

    oclonal antibodies (mAb 17-1A).Cancer

    Immunol. Immunother.31:23135

    95. Wersall P, Masucci G, Mellstedt H. 1991.

    Interleukin-4 augments cytotoxic capac-

    ity of lymphocytes and monocytes in

    antibody-dependent cellular cytotoxicity.

    Cancer Immunol. Immunother.33:4549

    96. Reali E, Spisani S, Gacili R, et al. 1995.

    IL-8 enhances antibody dependent cellu-

    lar cytotoxicity in human neutrophils.Im-

    munol. Cell Biol.73:23438

    97. Weiner L, Steplewski Z, Koprowski H,

    et al. 1986. Biologic effects of gamma in-

    terferon pre-treatment followed by mon-oclonal antibody 17-1A administration in

    patients with gastrointestinal carcinoma.

    Hybridoma5:S6577

    98. Ragnhammar P, Fagerberg J, Frodin J,

    et al. 1993. Effect of monoclonal an-

    tibody 17-1A and GM-CSF in patients

    with advanced colorectal carcinoma

    long-lasting, complete remissions can be

    induced. Int. J. Cancer53:75158

    99. Frodin J, Faxas M, Hagstrom B, et al.1991. Induction of anti-idiotypic (ab2)

    and anti-anti-idiotypic (ab3) antibodies in

    patients treated with the mouse mono-

    clonal antibody 17-1A (ab1). Relation to

    the clinical outcomean important an-

    titumoral effector function? Hybridoma

    10:42131

    100. Hanzawa Y, Tsujisaki M, Tokuchi S,

    et al. 1992. Detection of xenogeneic anti-

    idiotypic antibodies specific to murinemonoclonal antibody 17-1A in patients

    with gastrointestinal cancer. Tumour Biol.

    13:22636

    101. Fagerberg J, Frodin J, Wigzell H, Mell-

    stedt H. 1993. Induction of an immune

    network cascade in cancer patients treated

    with monoclonal antibodies (ab1). I. May

    induction of ab-1 reactive T cells and anti-

    anti-idiotypic antibodies (ab3) lead to tu-

    mor regression after mAb therapy? Can-

    cer Immunol. Immunother.37:26470

    102. Ragnhammar P, Fagerberg J, Frodin J-

    E, et al. 1995. Granulocyte/macrophage-

    colonystimulating factor augments the

    byb

    on:Univ