Sie sind auf Seite 1von 29

Annu. Rev. Med. 2003. 54:34369 doi: 10.1146/annurev.med.54.101601.152442 Copyright c 2003 by Annual Reviews.

All rights reserved

MONOCLONAL ANTIBODY THERAPY FOR CANCER


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 vonmehren@fccc.edu, gp adams@fccc.edu, lm weiner@fccc.edu
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

Key Words immunoconjugate, immunotoxin, radioimmunotherapy, antibody-dependent cellular cytotoxicity I Abstract Monoclonal antibody therapy has emerged as an important therapeutic modality for cancer. Unconjugated antibodies show signicant efcacy in the treatment of breast cancer, non-Hodgkins lymphoma, and chronic lymphocytic leukemia. Promising new targets for unconjugated antibody therapy include cellular growth factor receptors, receptors or mediators of tumor-driven angiogenesis, and B cell surface antigens other than CD20. Immunoconjugates composed of antibodies conjugated to radionuclides or toxins show efcacy in non-Hodgkins lymphoma. One immunoconjugate containing an antibody and a chemotherapy agent exhibits clinically meaningful antitumor activity in acute myeloid leukemia. Numerous efforts to exploit the ability of antibodies to focus the activities of toxic payloads at tumor sites are under way and show early promise. The ability to create essentially human antibody structures has reduced the likelihood of host-protective immune responses that otherwise limit the duration of therapy. Antibody structures now can be readily manipulated to facilitate selective interaction with host immune effectors. Other structural manipulations that improve the selective targeting properties and rapid systemic clearance of immunoconjugates 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 targeting missiles, have proved much more complex in their targeting and biologic properties than the elds pioneers envisioned them. The ability to manipulate the genes of antibodies with microbiologic techniques has allowed signicant modications of these structures. Murine proteins can be readily transformed into human or humanized formats that are not readily recognized as foreign by the
0066-4219/03/0218-0343$14.00

343

344

VON MEHREN

ADAMS

WEINER

human immune system. In addition, novel antibody-based structures with multiple antigen recognition sites, altered size, or effector domains have been shown to inuence the targeting ability of antibodies. Coupled with the identication of appropriate cancer targets, antibody-based therapeutics are nding increasing applications 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 signicant efcacy and novel approaches with clinical promise.
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 region 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 disulde bonds and noncovalent interactions between several of the domains. The complex is oriented with bilateral symmetry: light chainheavy chainheavy chainlight chain. The IgG molecule is divided into three functional domains, two antigenbinding (Fab) domains with identical antigen specicity connected by a highly exible hinge domain to the Fc (effector) domain that interacts with complement,

Figure 1 Schematic diagram depicting the structures of an IgG molecule and a variety of engineered antibody molecules derived from various IgG domains.

CANCER ANTIBODIES

345

immune effector cells, and receptors involved in maintaining constant concentrations 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 recombinant 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 afnity and less antigen specicity than corresponding intact antibody molecules (1, 3). Single-chain Fv (scFv) molecules, composed of peptide-linked VH and VL domains, are rapidly emerging as key players in the engineered antibody eld. With a size of 25 kDa, these molecules are both effective targeting vehicles in their own right and versatile components for the construction of larger antibody-based regents. ScFv molecules can be produced from genes isolated from hybridomas expressing MAb of a desired specicity (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 specic 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 disulde-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 afnity between 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 bioavailability 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) applications. For RAIT, smaller, more rapidly cleared molecules such as minibodies or diabodies can reduce nontargeted marrow toxicity. Furthermore, because physiologic barriers such as heterogeneous blood supply and elevated interstitial pressure

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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. Receptors 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, immunotoxin, or chemotherapeutic agent, it may be benecial to target a receptor that internalizes upon antibody binding, leading to internalization of the toxic agent.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

MECHANISMS OF ACTION FOR ANTIBODIES AS 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 factor 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 antibodydependent 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 rst 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 efciently with IgM molecules, but it can also occur if IgG molecules are clustered on the cell surface. Cells coated by IgG1 or IgG3 isotype antibodies can also activate effector cells

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 amplied by constructing bispecic 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
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 immunogenicity 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

348

VON MEHREN

ADAMS

WEINER

that they efciently mediate ADCC in vivo. For this to occur, sufcient 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 effector cells 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
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 of which have demonstrated clinically important antitumor activity and have received FDA approval for treatment of cancer. We focus here on antibodies with proven efcacy, classied 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 antibodies reactive 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-specic signature that could be targeted by a customized MAb. The procedures for preparing such antibodies for each patient were laborious, but 50% of treated patients experienced signicant 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 in part 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. This exciting 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, these important observations have informed much of the subsequent work in this eld. CD52

The CAMPATH-1 antibody has specicity for CD52, a glycopeptide that is highly expressed on T and B lymphocytes. It has been tested as a therapeutic

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 transplant grafts. Half of the patients with udarabine-resistant chronic lymphocytic leukemia or B-prolymphocytic leukemia exhibited clinical responses to CAMPATH-1 (29). A larger phase II study reported a 42% response rate in patients 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 rst-line therapy for patients with chronic lymphocytic leukemia. All patients responded with loss of peripheral blood malignant lymphocytes. However, patients with involvement of lymph nodes and/or spleen were less likely to respond completely. There was evidence of reactivation of cytomegalovirus infections. Subcutaneous administration of the antibody was found to be safe and effective. A humanized version of the antibody (CAMPATH-1H) has been extensively 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 refractory 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 frequently 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 carinii pneumonia, cytomegalovirus pneumonitis, pulmonary aspergillosis, disseminated tuberculosis, 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 signicantly decreased graft rejection and graftversus-host disease compared with conventional therapy. However, the frequency of 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). Additionally, 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

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

The testing and evaluation of the chimeric anti-CD20 antibody, IDECC2B8, also known as rituximab, demonstrated impressive clinical responses. Rituximab is the rst MAb to be approved by the FDA for use in human malignancy

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 antibodies 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 relapsed low-grade and intermediate/high-grade non-Hodgkins lymphoma received four weekly infusions of rituximab (37). Thrombocytopenia and B cell lymphocytopenia 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, conrmed the efcacy of this therapy, demonstrating 46% and 48% response rates in two separate studies (38, 39). Although the numbers of circulating B cells were reduced by therapy, there were no documented changes in serum immunoglobulin levels. Bacterial and viral infections were seen in patients with relapsed indolent lymphomas, but in contrast to the CAMPATH-1 experience, treatment with rituximab did not result in signicant 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 signicant activity in intermediate/ high-grade lymphomas, a phase II trial evaluated rituximab in relapsing or refractory diffuse large B cell lymphoma, mantle cell lymphoma, or other intermediateor 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 regimen. 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 rituximab 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-ve 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 responses and 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

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 combination arm compared with 60% complete response rate in the chemotherapy arm without signicant 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 udarabine in low-grade lymphomas. Signicant leukopenias were associated with this combination, requiring a dose reduction in the udarabine. An interim analysis after accruing 30 of the 40 planned patients revealed a response rate of 93% with median response duration of over 14 months. Cytogenetic responses were also observed in some patients (45). The signicance of such responses is unclear. In a study of rituximab with CHOP 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 transplant, 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 reported with a partial response and another two patients with unconrmed complete response. Circulating CD20-positive cells, including lymphoma cells, may affect the efcacy 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 signicant lower response rates. A dosage of eight cycles of weekly rituximab has been used safely in lowgrade and follicular non-Hodgkins lymphoma (48). The efcacy 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 population of tumor cells. At the present time, little is known about mechanisms of resistance (49a). This phenomenon has been documented in a patient with a follicular 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

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

352

VON MEHREN

ADAMS

WEINER

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 include CD22, which is expressed on pro-B and pre-B cell lymphomas, and the major 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 undergoing 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, thrombocytopenia, tumor lysis, bronchospasm, and hypoxemia, requiring discontinuation of 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 (c-erbB-2), a member of the epidermal growth factor receptor (EGFR) family, has been targeted for antibody therapy because it is overexpressed 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 HER2/neu. A phase II trial in women with metastatic breast cancer reported an objective 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 disease, lasting more than 5 months. Overexpression of HER-2/neu that is associated with gene amplication correlates with a clinical response to trastuzumab. Earlier studies utilized variable criteria to dene 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
HER-2/NEU

CANCER ANTIBODIES

353

uorescence in situ hybridization to conrm gene amplication. Intriguingly, preclinical studies have demonstrated decreased expression of vascular endothelial cell growth factor (VEGF) and vascular permeability factor with 4D5 therapy, suggesting that an antiangiogenesis mechanism may account for some of the clinical impact 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 efcacy of combination therapy (59). Patients receiving initial therapy for metastatic breast cancer were treated with cyclophosphamide 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 survival at one year by 16% (60) and improved survival at 29 months by 25% (61). Myocardial dysfunction was observed more frequently in patients receiving doxorubicin 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 participation 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. Although other adenocarcinomas may overexpress HER-2/neu, clinical trials have not documented consistent therapeutic successes in other cancers. The Gynecologic 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/neu has also been found on prostate cancer and nonsmall-cell lung cancer.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 autocrine 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

354

VON MEHREN

ADAMS

WEINER

tyrosine kinases and inhibit growth of normal broblasts (68) as well as tumor cells in culture (69). Also, combining anti-EGFR antibodies with cisplatin signicantly decreases the IC50 of 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 internalization, as occurs with binding of the natural ligand (76). However, receptor processing is slower with antibody engagement than with natural ligand engagement (77). Smaller bivalent F(ab )2 and 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 efcacy of antibody therapy is not dependent on ADCC, as these smaller fragments lack the Fc portion of the antibody required for ADCC. Rather, the efcacy of MAb225 is due to its ability to inhibit binding of the natural ligand, limit receptor phosphorylation and thus downstream signals, and induce receptor internalization. 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 to inhibit 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 ushing, 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). Another phase 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

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

CANCER ANTIBODIES

355

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 benet of adding C225 to radiation therapy 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 development. 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 clinical testing, with some studies suggesting efcacy 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 human 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 such as interferon- (97) and GM-CSF (98). Although these studies have shown evidence for cytokine immunomodulation, no consistent pattern of clinical benet 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 inltration of macrophages and of CD4+ and CD8+ T cells within tumors (103). Induction of T cells against antiidiotypic epitopes has also been evaluated (104). Five of ten patients with antiidiotypic antibodies were also found to have induction of EP-CAM antigen-specic T cells. Four of these patients showed a clinical response and their T cells demonstrated a proliferative response in vitro when stimulated with an anti-idiotypic antibody, in contrast to the six patients without evidence of T cells against antiidiotypic epitopes.

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 gastrointestinal tract with only one intravenous dose of antibody. One patient received an intrahepatic infusion of autologous mononuclear cells mixed with 17-1A, which resulted 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 combinations with cytokines to enhance effector cell number and activity did not result in signicant 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 signicant impact on clinical response (106). Some trials reported evidence of induction of anti-idiotypic antibodies, correlating with clinical response in one trial. The overall lack of efcacy 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 efcacy. 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 patients were followed post-operatively in a similar manner, irrespective of treatment. Of 189 patients, 166 were evaluable for overall survival and disease-free survival. 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 ve 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 benet 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 shift in 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 efcacy of 17-1A on local control is that 11 patients in the observation group received pre- or postoperative 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 conrm 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

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

CANCER ANTIBODIES

357

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

adjuvant therapy with 17-1A would introduce therapies that have different mechanisms 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 no difference 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 benet of questionable signicance in a preliminary analysis. Overall survival was 81.6% for combination therapy versus 78.9% for chemotherapy alone; this was not a statistically signicant difference. Irrespective of the nal 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 rst 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 clinical study 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 conrmed that hypertension is a toxicity of this agent (111). Two patients receiving 3 mg/kg experienced bleeding into their 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 paclitaxel in 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 benet 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

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 with chemotherapy 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-uorouracil 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.
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

RADIOIMMUNOTHERAPY Radioimmunoconjugates
A tumor-specic 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 efcacy with the addition of a cytotoxic radioisotope. For example, although therapy with the anti-CD20 MAb rituximab is associated with a signicant antitumor effect in patients with non-Hodgkins lymphoma, signicantly more patients exhibit complete 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 gure of merit (IFOM) and therapy gure 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 efciency and specicity 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

CANCER ANTIBODIES

359

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

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 a gamma emission. The other commonly used radionuclide, 131I (t1/2 = 8.02 days), requires a trained radiopharmacist and appropriately shielded/vented radiopharmacy 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 radioisotopes limit 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 signicant 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 radioimmunoconjugates to the malignant cells that comprise these neoplasms. Patients with hematologic neoplasms are also less likely to mount HAMA responses when foreign MAbs are employed.

Radioimmunotherapy for Hematologic Malignancies


The CD20 antigen has been a very effective target for RAIT. Press and colleagues 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 highdose 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 efcacious, 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).
B-1

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 of 131I. This antibody is the rst, and to date only, MAb licensed for use in RAIT in the United States.
IBRITUMOMAB TIUXETAN

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 efcacy of a single dose of ibritumomab with a standard treatment course of rituximab in 143 patients with low-grade, follicular 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).
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.
LYM-1 Lym-1 is specic 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, 131I-Lym-1 has shown signicant 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).

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) early hematopoietic stem cells. M195, a murine anti-CD33 MAb, has been used to deliver therapeutic doses of 131I in combination with busulfan or cyclophosphamide to eliminate disease before bone marrow transplantation (125). HuM195, a humanized 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 rst use of alpha particles in RAIT.
M195

Radioimmunotherapy of Solid Malignancies


Signicantly 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 carcinoma, 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, intraperitoneal 177 Lu-labeled CC49 MAb, plus interferon and Taxol led to partial responses in 4 of 17 treated patients and progression-free intervals in patients without measurable

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 characteristics (e.g., domain deletions or signicant antitumor properties as a naked MAb) that may translate into improved results.

Pretargeted Radioimmunotherapy
One new RAIT strategy recently examined in the clinic is pretargeted radioimmunotherapy (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) conjugates were administered to the patient. After the MAb conjugate localized in the tumor, 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 specically 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 lasting longer than one year (132). In the PRIT treatment of patients with colon cancer, 8% (2 of 25) of the patients treated with pretargeted NR-LU-10 MAb and 110 mCi/m2 of 90Y-DOTA-biotin had a partial response, and 16% (4 of 25) exhibited stable disease. However, signicant hematologic and nonhematologic toxicities (diarrhea) were reported (133). A companion trial directed against prostate cancer revealed similar toxicities (I. Horak, personal communication). Signicantly 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 rst-step reagent. Six of seven patients treated with doses of 30 or 50 mCi/m2 of 90Y-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.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

CONCLUSIONS
Therapeutic antibodies have made the transition from concept to clinical reality over the past two decades. Many are now being tested as adjuvant or rst-line therapies to assess their efcacy in improving or prolonging survival. Modied antibody structures, such as bispecic antibodies or smaller antibody fragments, have yet to claim dened therapeutic roles, whereas radioimmunotherapy has clearly demonstrated efcacy in some disease settings.

362

VON MEHREN

ADAMS

WEINER

The Annual Review of Medicine is 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 complementarity-determining region of antitumor antibody. Lancet 346:166266 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. 453:16468 3. Ward ES, Gussow D, Grifths 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 antibody binding sites: recovery of specic activity in an anti-digoxin single-chain Fv analogue produced in E coli. Proc. Natl. Acad. Sci. USA 85:587983 5. Marks JD, Hoogenboom HR, Bonnert TP, et al. 1991. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J. Mol. Biol. 222:58197 6. Begent RH, Verhaar MJ, Chester KA, et al. 1996. Clinical evidence of efcient tumor targeting based on single-chain Fv antibody selected from a combinatorial library. Nat. Med. 2:97984 7. Adams GP, McCartney JE, Tai MS, et al. 1993. Highly specic in vivo tumor targeting 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 bispecic antibody fragments. Proc. Natl. Acad. Sci. USA 90:644448 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 Jain RK. 1990. Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors. Cancer Res. (Suppl.) 50:814s19s Jain RK, Baxter LT. 1988. Mechanisms of heterogeneous distribution of monoclonal antibodies and other macromolecules in tumors: signicance of elevated interstitial pressure. Cancer Res. 48:702232 Trauth B, Klas C, Peters A, et al. 1989. Monoclonal antibody-mediated tumor regression by induction of apoptosis. Science 245:3015 Waldman T. 1991. Monoclonal antibodies in diagnosis and therapy. Science 252:38794 OConnell M, Chen Z, Yang H, et al. 1989. Active specic immunotherapy with antiidiotypic antibodies in patients with solid tumors. Semin. Surg. Oncol. 5:441 47 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 82:124246 Steplewski Z, Lubeck M, Koprowski H. 1983. Human macrophages armed with murine immunoglobulin G2a antibodies to tumors destroy human cancer cells. Science 221:86567 Lindenmann J. 1973. Speculations on Ids and homobodies. Ann. Immunol. 124: 17184 Jerne N. 1974. Towards a network theory of the immune system. Ann. Immunol. 125:37389 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

10.

11.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

12.

13.

14.

15.

16.

17.

18.

19.

CANCER ANTIBODIES 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, Ceriani R. 1995. Selection of tumor-specic epitopes on target antigens for radioimmunotherapy of breast cancer. Cancer Res. 55:5847s51s 22. Badger C, Anasetti C, Davis J, Bernstein I. 1987. Treatment of malignancy with unmodied antibody. Pathol. Immunopathol. Res. 6:41934 23. Holland G, Zlotnik A. 1993. Interleukin10 and cancer. Cancer Invest. 11:75158 24. 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 alters 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 monoclonal antibody directed against a human lymphoma-associated antigen. Cancer Res. 40:314754 27. Vuist W, Levy R, Maloney D. 1994. Lymphoma regression induced by monoclonal anti-idiotypic antibodies correlates with their ability to induce Ig signal transduction and is not prevented by tumor expression of high levels of bcl-2 protein. Blood 83:899906 28. Swisher E, Shawler D, Collins H, et al. 1991. Expression of shared idiotypes in chronic lymphocytic leukemia and small lymphocytic lymphoma. Blood 77:1977 82 29. Bowen A, Zomas A, Emmett E, et al. 1997. Subcutaneous CAMPATH-1H in udarabine-resistant/relapsed chronic lymphocytic and B-prolymphocytic leukaemia. Br. J. Haematol. 96:61719 30. Lundin J, Osterborg A, Brittinger G, et al. 1998. CAMPATH-1H monoclonal antibody in therapy for previously treated

363

31.

32.

33.

34.

35.

36.

37.

38.

low-grade non-Hodgkins lymphomas: a phase II multicenter study. European Study Group of CAMPATH-1H Treatment in Low-Grade Non-Hodgkins Lymphoma. J. Clin. Oncol. 16:325763 Hale G, Zhang M, Bunjes D, et al. 1998. Improving the outcome of bone marrow transplantation by using CD52 monoclonal antibodies to prevent graft-versushost disease and graft rejection. Blood 92:458190 Naparstek E, Delukina M, Or R, et al. 1999. Engraftment of marrow allografts treated with Campath-1 monoclonal antibodies. Exp. Hematol. 27:121018 Novitzky N, Thomas V, Hale G, Waldmann H. 1999. Ex vivo depletion of T cells from bone marrow grafts with CAMPATH-1 in acute leukemia: graftversus-host disease and graft-versusleukemia effect. Transplantation 67:620 26 Hale G, Waldmann H. 1998. Risks of developing Epstein-Barr virusrelated lymphoproliferative disorders after T-cell depleted marrow transplants. CAMPATH Users. Blood 91:307983 Maloney D, Grillo-L opez A, Bodkin D, et al. 1997. IDEC-C2B8: results of a phase I multiple-dose trial in patients with relapsed non-Hodgkins lymphoma. J. Clin. Oncol. 15:326674 Maloney D, Grillo-L opez A, White C, 1997. IDEC-C2B8 (Rituximab) antiCD20 monoclonal antibody therapy in patients with relapsed low-grade nonHodgkins lymphoma. Blood 90:218895 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 McLaughlin P, Grillo-L opez A, Link B, et al. 1998. Rituximab chimeric antiCD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J. Clin. Oncol. 16:282533

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

364

VON MEHREN

ADAMS

WEINER 2002. Immunotherapy with rituximab following high-dose therapy and autologous stem-cell transplantation for mantle cell lymphoma. Semin. Oncol. 29:5669 Piro L, White C, Grillo-L opez A, et al. 1999. Extended rituximab (anti-CD20 monoclonal antibody) therapy for relapsed or refractory low-grade or follicular non-Hodgkins lymphoma. Ann. Oncol. 10:61921 Keating M, OBrien S. 2000. High-dose rituximab therapy in chronic lymphocytic leukemia. Semin. Oncol. 27:8690 Maloney DG, Smith B, Rose A. 2002. Rituximab: mechanisms of action and resistance. Semin. Oncol. 29(1 Suppl. 2):29 Davis T, Czerwinski D, Levy R. 1999. Therapy of B-cell lymphoma with antiCD20 antibodies can result in the loss of CD20 antigen expression. Clin. Cancer Res. 5:61115 Nabhan C, Rosen ST. 2002. Conceptual aspects of combining rituximab and campath-1H in the treatment of chronic lymphocytic leukemia. Semin. Oncol. 29: 7580 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 Byrd J, Waselenko J, Maneatis T, et al. 1999. Rituximab therapy in hematologic malignancy patients with circulating blood tumor cells: association with increased infusion-related side effects and rapid blood tumor clearance. J. Clin. Oncol. 17:79195 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, rituximab). Ann. Hematol. 77:8991 Grillo-L opez A, Hedrick E, Rashford M, Benyunes M. 2002. Rituximab: ongoing and future clinical development. Semin. Oncol. 29:10212

39. Berinstein N, Grillo-L opez A, White C, et al. 1998. Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkins lymphoma. Ann. Oncol. 9:9951001 40. Coifer 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. Blood 92:192732 41. Heinzerling L, Dummer R, Kempf W, et al. 2000. Intralesional therapy with antiCD 20 monoclonal antibody rituximab in primary cutaneous lymphoma. Arch. Dermatol. 136:37478 42. Czuczman M, Grillo-L opez A, White C, et al. 1999. Treatment of patients with low-grade B-cell lymphoma with the combination of chimeric anti-CD20 monoclonal antibody and CHOP chemotherapy. J. Clin. Oncol. 17:26876 43. Demidem A, Lam T, Alas S, et al. 1997. Chimeric anti-CD20 (IDEC-C2B8) monoclonal antibody sensitizes a B cell lymphoma cell line to cell killing by cytotoxic 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 reaction ampliable breakpoint of bcl-2 have residual cells containing the bcl-2 rearrangement at evaluation and after treatment. Blood 78:327580 45. Czuczman M. 2002. Rituximab in combination with CHOP or udarabine in lowgrade lymphoma. Semin. Oncol. 20:36 40 46. Howard O, Gribben J, Neuberg D, et al. 2002. Rituximab and CHOP induction therapy for newly diagnosed mantlecell lymphoma: molecular complete responses are not predictive of progressionfree survival. J. Clin. Oncol. 20:128894 47. Mangel J, Buckstein R, Imrie K, et al.

48.

49.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

49a.

50.

51.

52.

53.

54.

55.

CANCER ANTIBODIES 56. Cobleigh M, Vogel C, Tripathy D, et al. 1999. Multinational study of the efcacy 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 intravenous recombinant humanized antip185HER2 monoclonal antibody in patients 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: angiogenic implications for signal transduction therapy of solid tumors. Am. J. Pathol. 151:152330 59. Slamon D, Leyland-Jones B, Shak S, et al. 2001. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses 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 monoclonal antibody Herceptin (H) in HER2-overexpressing (HER2+) metastatic breast cancer (MBC). Proc. Am. Soc. Clin. Oncol. 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 HER2overexpressing metastatic breast cancer. J. Clin. Oncol. 19:272230 63. Pegram M, Lipton A, Hayes D, et al. 1998. Phase II study of receptor-en-

365

64.

65.

66.

67.

68.

69.

70.

hanced chemosensitivity using recombinant 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 Miller KD, Sisk J, Ansari R, et al. 2001. Gemcitabine, paclitaxel, and trastuzumab in metastatic breast cancer. Oncology 15:S38S40 Bookman M, Darcy K, Clarke-Pearson D, et al. 2002. Evaluation of monoclonal 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 Gynecologic Oncology Group. J. Clin. Oncol. 20: In press 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 the binding of epiregulin to EGFR-expressing tumours. Int. J. Cancer 75:31016 Teramoto T, Onda M, Tokunaga A, Asano G. 1996. Inhibitory effect of an antiepidermal growth factor receptor antibody on human gastric cancer. Cancer 77:163945 Sato J, Kawamoto T, Le A, et al. 1983. Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Mol. Biol. Med. 1:51129 Artega C, Corondo E, Osbore C. 1988. Blockade of the epidermal growth factor receptor inhibits transforming growth factor ainduced but not estrogen-induced growth of hormone-dependent human breast cancer. Mol. Endocrinol. 2:1064 69 Hoffman T, Hafner D, Ballo H, et al. 1997. Antitumor activity of anti-epidermal growth factor receptor monoclonal antibodies and cisplatin in ten human head and neck squamous cell carcinoma lines. Anticancer Res. 17:441925

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

366

VON MEHREN

ADAMS

WEINER tor monoclonal antibodies. Cancer Res. 53:432228 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 Zolfaghari A, Djakiew D. 1996. Inhibition of chemomigration of a human prostatic carcinoma cell (TSU-pr1) line by inhibition of epidermal growth factor receptor function. Prostate 28:23238 Peng D, Fan Z, Lu Y, et al. 1996. Antiepidermal growth factor receptor monoclonal antibody 225 up-regulates p27KIP1 and induces G1 arrest in prostatic cancer cell line DU145. Cancer Res. 56:366669 Wu X, Rubin M, Fan Z, et al. 1996. Involvement of p27KIP1 in G1 arrest mediated by an anti-epidermal growth factor receptor monoclonal antibody. Oncogene 12:1397403 Baselga J, Pster 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 Herbst R, Langer C. 2002. Epidermal growth factor receptors as a target for cancer treatment: the emerging role of IMCC225 in the treatment of lung and head and neck cancers. Semin. Oncol. 29:2736 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 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 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

71. Fan Z, Baselga J, Masui H, Mendelsohn J. 1993. Antitumor effect of anti-epidermal growth factor receptor monoclonal antibodies plus cis-diamminedichloroplatinum 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 sequential treatment with topotecan and antiepidermal growth factor receptor monoclonal antibody C225. Clin. Cancer Res. 5:90916 74. Bonner J, Raisch K, Robert HT, et al. 2000. Enhanced apoptosis with combination C225/radiation treatment serves as the impetus for clinical investigation in head and neck cancers. J. Clin. Oncol. 18 (Suppl.):47s53s 75. Modjtahedi H, Eccles S, Sandle J, et al. 1994. Differentiation or immune destruction: two pathways for therapy of squamous 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 internalized without stimulating receptor phosphorylation. 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 intracellular processing of the epidermal growth factor receptor by anti-receptor monoclonal antibody. J. Cell. Physiol. 142:284 92 78. Fan Z, Masui M, Altas I, Mendelsohn J. 1993. Blockade of the epidermal growth factor receptor function by bivalent and monovalent fragments of 225 anti-epidermal growth factor recep-

79.

80.

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

81.

82.

83.

84.

85.

86.

87.

CANCER ANTIBODIES concomitant chemotherapy. Cancer Res. 59:123643 Haga Y, Sivinski C, Woo D, Tempero M. 1994. Dose related comparison of antibody-dependent cellular cytotoxicity with chimeric and native monoclonal antibody 17-1A. Improved cytolysis of pancreatic cancer cells with chimeric 17-1A. Int. J. Pancreatol. 15:4350 Meredith R, LoBuglio A, Plott W, et al. 1991. Pharmacokinetics, immune response, and biodistribution of iodine-131 labeled chimeric mouse/human IgG1 171A monoclonal antibody. J. Nucl. Med. 32:116268 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: enhancement with interferon gamma. Cancer Immunol. Immunother. 43:22025 Reali E, Guiliani A, Spisani S, et al. 1994. Interferon-gamma enhances monoclonal antibody 17-1Adependent neutrophil cytotoxicity toward colorectal carcinoma cell line SW11-16. Clin. Immunol. Immunopathol. 71:10512 Ragnhammar P, Masucci G, Frodin J, et al. 1992. Cytotoxic functions of blood mononuclear cells in patients with colorectal carcinoma treated with mAb-171A and granulocyte/macrophage colony stimulating factor. Cancer Immunol. Immunother. 35:15864 Ragnhammar P, Magnusson I, Masucci G, Mellstedt H. 1993. The therapeutic use of the unconjugated monoclonal antibodies (MAb) 17-1A in combination with GMCSF in the treatment of colorectal carcinoma (CRC). Med. Oncol. Tumor Pharmacother. 10:6170 Masucci G, Ragnhammar P, Wersall P, Mellstedt H. 1990. Granulocyte-monocyte colony-stimulating factor augments the interleukin-2 induced cytotoxic activity of human lymphocytes in the absence and presence of mouse or chimeric mon-

367

88.

95.

96.

89.
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

97.

90.

98.

91.

99.

92.

100.

93.

101.

94.

102.

oclonal antibodies (mAb 17-1A). Cancer Immunol. Immunother. 31:23135 Wersall P, Masucci G, Mellstedt H. 1991. Interleukin-4 augments cytotoxic capacity of lymphocytes and monocytes in antibody-dependent cellular cytotoxicity. Cancer Immunol. Immunother. 33:4549 Reali E, Spisani S, Gacili R, et al. 1995. IL-8 enhances antibody dependent cellular cytotoxicity in human neutrophils. Immunol. Cell Biol. 73:23438 Weiner L, Steplewski Z, Koprowski H, et al. 1986. Biologic effects of gamma interferon pre-treatment followed by monoclonal antibody 17-1A administration in patients with gastrointestinal carcinoma. Hybridoma 5:S6577 Ragnhammar P, Fagerberg J, Frodin J, et al. 1993. Effect of monoclonal antibody 17-1A and GM-CSF in patients with advanced colorectal carcinoma long-lasting, complete remissions can be induced. Int. J. Cancer 53:75158 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 monoclonal antibody 17-1A (ab1). Relation to the clinical outcomean important antitumoral effector function? Hybridoma 10:42131 Hanzawa Y, Tsujisaki M, Tokuchi S, et al. 1992. Detection of xenogeneic antiidiotypic antibodies specic to murine monoclonal antibody 17-1A in patients with gastrointestinal cancer. Tumour Biol. 13:22636 Fagerberg J, Frodin J, Wigzell H, Mellstedt 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 antianti-idiotypic antibodies (ab3) lead to tumor regression after mAb therapy? Cancer Immunol. Immunother. 37:26470 Ragnhammar P, Fagerberg J, Frodin JE, et al. 1995. Granulocyte/macrophagecolonystimulating factor augments the

368

VON MEHREN

ADAMS

WEINER study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J. Clin. Oncol. 19:843 50 Sledge G, Miller K, Novotny W, et al. 2000. A phase II trial of single agent rhuMAb VEGF (recombinant humanized monoclonal antibody to vascular endothelial cell growth factor) in patients with relapsed metastatic breast cancer. Proc. Am. Soc. Clin. Oncol. 20:A5C (Abstr.) Notovny W, Holmgren E, Grifng S, et al. 2001. Identication of squamous cell histology and central cavitary tumors as possible risk factors for pulmonary hemorrhage (PH) in patients with advanced NSCLC receiving Bevacizumab (B). Proc. Am. Soc. Clin. Oncol. 21:A1318 (Abstr.) DeVore R, Fehrenbacher L, Herbst R, et al. 2000. A randomized phase II trial comparing RhuMAb VEGF (recombinant humanized monoclonal antibody to vascular endothelial growth factor) plus carboplatin/paclitaxel (CP) to CP alone in patients with stage IIIB/IV NSCLC. Proc. Am. Soc. Clin. Oncol. 20:A1896 (Abstr.) Bergsland E, Hurwitz H, Fehrenbacher L, et al. 2000. A randomized phase II trial comparing rhuMAb VEGF (recombinant humanized monoclonal antibody to vascular endothelial cell growth factor) plus 5-uoruracil/leucovorin (FU/LV) in patients with metastatic colorectal cancer. Proc. Am. Soc. Clin. Oncol. 20:A939 (Abstr.) Witzig TE, White CA, Wiseman GA, et al. 1999. Phase I/II trial of IDEC-Y2B8 radioimmunotherapy for treatment of relapsed or refractory CD20 (+) B-cell non-Hodgkins lymphoma. J. Clin. Oncol. 17:3793803 Williams LE, Liu A, Wu AM, et al. 1995. Figures of merit (FOMs) for imaging and therapy using monoclonal antibodies. Med. Phys. 22:202527 Press OW, Eary JF, Appelbaum FR, et al. 1993. Radiolabeled-antibody therapy of

103.

104.
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

105.

106.

107.

108.

109.

110.

induction of antibodies, especially antiidiotypic antibodies to therapeutic monoclonal antibodies. Cancer Immunol. Immunother. 40:36775 Shetye J, Ragnhammar P, Liljefors M, et al. 1998. Immunopathology of metastases in patients of colorectal carcinoma treated with monoclonal antibody 171A and granulocyte macrophage colony stimulating factor. Clin. Cancer Res. 4:192129 Fagerberg J, Frodin J, Ragnhammar P, et al. 1994. Induction of an immune network cascade in cancer patients treated with monoclonal antibodies (ab1). II. Is induction of anti-idiotype reactive T cells (T3) of importance for tumor response to mAb therapy? Cancer Immunol. Immunother. 38:14959 Sears H, Herlyn D, Steplewski Z, Koprowski H. 1985. Phase II clinical trial of a murine monoclonal antibody cytotoxic for gastrointestinal adenocarcinoma. Cancer Res. 45:591013 Weiner L, Harvey E, Padavic-Shaller K, et al. 1993. Phase II multicenter evaluation of prolonged murine monoclonal antibody 17-1a therapy in pancreatic carcinoma. J. Immunother. 13:11016 Reithmuller G, Schneider-Gadicke E, Schlimok G, et al. 1994. Randomised trial of monoclonal antibody for adjuvant therapy of resected Dukes C colorectal carcinoma. German Cancer Aid 17-1A Study Group. Lancet 343:117783 Reithmuller G, Holz E, Schlimok G, et al. 1998. Monoclonal antibody therapy for resected Dukes C colorectal cancer: seven-year outcome of a multicenter randomized trial. J. Clin. Oncol. 16:178894 Dencausse Y, Hartung G, Sturm J, et al. 2001. Prospective randomized study of adjuvant therapy with edrecolomab (Panorex) of stage II colon cancer interim analysis. Proc. Am. Soc. Clin. Oncol. 21:A2198 (Abstr.) Gordon M, Margolin K, Talpaz M, et al. 2001. Phase I safety and pharmacokinetic

111.

112.

113.

114.

115.

116.

117.

CANCER ANTIBODIES B-cell lymphoma with autologous bone marrow support. N. Engl. J. Med. 329: 121924 Kaminski MS, Zasadny KR, Francis IR, et al. 1993. Radioimmunotherapy of B-cell lymphoma with [131I] anti-B1 (anti-CD20) antibody. N. Engl. J. Med. 329:45965 Kaminski MS, Zasadny KR, Francis IR, et al. 1996. Iodine-131anti-B1 radioimmunotherapy for B-cell lymphoma. J. Clin. Oncol. 14:197481 Caron PC, Scheinberg DA. 1993. AntiCD33 monoclonal antibody M195 for the therapy of myeloid leukemia. Leukemia Lymphoma 11(Suppl. 2):16 White CA, Weaver RL, Grillo-L opez AJ. 2001. Antibody-targeted immunotherapy for treatment of malignancy. Annu. Rev. Med. 52:12545 Witzig TE, Gordon LI, Cabanillas F, et al. 2002. Randomized controlled trial of yttrium-90labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkins lymphoma. J. Clin. Oncol. 20:245363 DeNardo GL, DeNardo SJ, Lamborn KR, et al. 1998. Low-dose, fractionated radioimmunotherapy for B-cell malignancies using 131ILym-1 antibody. Cancer Biother. Radiopharm. 13:23954 DeNardo GL, DeNardo SJ, Goldstein DS, et al. 1998. Maximum-tolerated dose, toxicity, and efcacy of 131ILym-1 antibody for fractionated radioimmunotherapy of non-Hodgkins lymphoma. J. Clin. Oncol. 16:324656 Jurcic JG, Caron PC, Nikula TK, et al. 1995. Radiolabeled anti-CD33 monoclonal antibody M195 for myeloid leukemias. Cancer Res. 55(23 Suppl.): 5908s10s Jurcic JG, Divgi CR, McDevitt MR, et al. 1998. Potential for myeloablation

369

118.

127.

119.

128.

120.

129.

121.

122.

130.

131.

123.

132.

124.

133.

125.

134.

126.

with yttrium-90HuM195 (anti-CD33): a phase I trial in advanced myeloid leukemias. Blood 92:613A (Abstr.) Jurcic JG, McDevitt MR, Sgouros G, et al. 1999. Phase I trial of targeted alpha particle therapy for myeloid leukemias with bismuth-213HuM195 (anti-CD33). Proc. Am. Soc. Clin. Oncol. 18:7A (Abstr.) Meredith RF, Khazaeli MB, Macey DJ, et al. 1999. Phase II study of interferonenhanced 131I-labeled high afnity CC49 monoclonal antibody therapy in patients with metastatic prostate cancer. Clin. Cancer Res. 5(Suppl.):3254s58s ODonnell RT, DeNardo SJ, Yuan A, et al. 2001. Radioimmunotherapy with 111 In/90Y-2IT-BAD-m170 for metastatic prostate cancer. Clin. Cancer Res. 7: 156168 Meredith RF, Alvarez RD, Partridge EE, et al. 2001. Intraperitoneal radioimmunochemotherapy of ovarian cancer: a phase I study. Cancer Biother. Radiopharm. 16:30515 DeNardo SJ, Kramer EL, ODonnell RT, et al. 1997. Radioimmunotherapy for breast cancer using indium-111/yttrium90 BrE-3: results of a phase I clinical trial. J. Nucl. Med. 38:118085 Paganelli G, Grana C, Chinol M, et al. 1999. Antibody-guided three-step therapy for high grade glioma with yttrium-90 biotin. Eur. J. Nucl. Med. 26:34857 Knox SJ, Goris ML, Tempero M, et al. 2000. Phase II trial of yttrium-90 DOTA-biotin pretargeted by NR-LU10 antibody/streptavidin in patients with metastatic colon cancer. Clin. Cancer Res. 6:40614 Weiden PL, Breitz HB, Press O, et al. 2000. Pretargeted radioimmunotherapy (PRIT) for treatment of non-Hodgkins lymphoma (NHL): initial phase I/II study results. Cancer. Biother. Radiopharm. 15: 1529

Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

Annual Review of Medicine Volume 54, 2003

CONTENTS
TECHNOLOGY-DRIVEN TRIAGE OF ABDOMINAL TRAUMA: THE EMERGING ERA OF NONOPERATIVE MANAGEMENT, D. Demetriades
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

and G. Velmahos

1 17

ANGIOGENESIS IN ISCHEMIC AND NEOPLASTIC DISORDERS,


Gregg L. Semenza

PATHOGENESIS AND TREATMENT OF GRAFT-VERSUS-HOST DISEASE AFTER BONE MARROW TRANSPLANT, Georgia B. Vogelsang,
Linda Lee, and Debra M. Bensen-Kennedy 29 53 73 89 113

UPDATE IN PALLIATIVE MEDICINE AND END-OF-LIFE CARE,


Janet L. Abrahm

MOLECULAR GENETICS OF LUNG CANCER, Yoshitaka Sekido,


Kwun M. Fong, and John D. Minna

PATHOPHYSIOLOGICAL-BASED APPROACHES TO TREATMENT OF SICKLE CELL DISEASE, Martin H. Steinberg and Carlo Brugnara NEW CONCEPTS IN CHRONIC OBSTRUCTIVE PULMONARY DISEASE,
Peter J. Barnes

CALORIE RESTRICTION, AGING, AND CANCER PREVENTION: MECHANISMS OF ACTION AND APPLICABILITY TO HUMANS,
Stephen D. Hursting, Jackie A. Lavigne, David Berrigan, Susan N. Perkins, and J. Carl Barrett ERECTILE DYSFUNCTION, Ridwan Shabsigh and Aristotelis G. Anastasiadis 131 153 169 185 197 217 235 v

THE HYPEREOSINOPHILIC SYNDROME REVISITED, Florence Roufosse,


Elie Cogan, and Michel Goldman

STANDARDS OF CARE IN GERIATRIC PRACTICE, Robert J. Luchi,


Julie K. Gammack, Victor J. Narcisse, III, and C. Porter Storey, Jr.

NEW CONCEPTS IN THE TREATMENT OF RHEUMATOID ARTHRITIS,


Raphaela Goldbach-Mansky and Peter E. Lipsky

STUDYING CANCER FAMILIES TO IDENTIFY KIDNEY CANCER GENES,


Berton Zbar, Richard Klausner, and W. Marston Linehan

THE AUTOMATED EXTERNAL DEFIBRILLATOR: CRITICAL LINK IN THE CHAIN OF SURVIVAL, Karthik Ramaswamy and Richard L. Page

vi

CONTENTS

THE BIOMEDICAL CHALLENGES OF SPACE FLIGHT, David R. Williams GENETICS AND ARRHYTHMIAS, Robert Roberts and Ramon Brugada OPEN SURGICAL REPAIR VERSUS ENDOVASCULAR THERAPY FOR CHRONIC LOWER-EXTREMITY OCCLUSIVE DISEASE,
Marc L. Schermerhorn, Jack L. Cronenwett, and John C. Baldwin AIDS-RELATED MALIGNANCIES, David T. Scadden

245 257

269 285

DIAGNOSIS AND PREVENTION OF BOVINE SPONGIFORM ENCEPHALOPATHY AND VARIANT CREUTZFELDT-JAKOB DISEASE,
David N. Irani and Richard T. Johnson
Annu. Rev. Med. 2003.54:343-369. Downloaded from www.annualreviews.org by b-on: Universidade de Lisboa (UL) on 02/26/11. For personal use only.

305 321 343

ATHEROPROTECTIVE EFFECTS OF HIGH-DENSITY LIPOPROTEINS,


Gerd Assmann and Jerzy-Roch Nofer

MONOCLONAL ANTIBODY THERAPY FOR CANCER, Margaret von


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

FUNCTIONAL GENOMICS OF THE PARAOXONASE (PON1) POLYMORPHISMS: EFFECTS ON PESTICIDE SENSITIVITY, CARDIOVASCULAR DISEASE, AND DRUG METABOLISM,
Lucio G. Costa, Toby B. Cole, Gail P. Jarvik, and Clement E. Furlong GENETIC PRIVACY, Pamela Sankar THE ANTIPHOSPHOLIPID SYNDROME, Jacob H. Rand 371 393 409 425 437 453 473 491 513 535

ANTI-INTEGRIN THERAPY, Christian W. Hamm PHARMACOGENETICS IN CANCER TREATMENT, R. Nagasubramanian,


F. Innocenti, and M.J. Ratain

GENETICS AND PATHOPHYSIOLOGY OF HUMAN OBESITY,


David E. Cummings and Michael W. Schwartz

MOLECULAR GENETIC RISK SCREENING, Wayne W. Grody THE CURRENT STATUS OF HEMATOPOIETIC CELL TRANSPLANTATION,
Frederick R. Appelbaum

GROWTH HORMONE THERAPY IN ADULTS, David E. Cummings


and George R. Merriam

THE INFLUENCE OF HLA GENOTYPE ON AIDS, Mary Carrington


and Stephen J. OBrien

INDEXES
Subject Index Cumulative Index of Contributing Authors, Volumes 5054 Cumulative Index of Chapter Titles, Volumes 5054 553 585 589

ERRATA
An online log of corrections to Annual Review of Medicine chapters may be found at http://med.annualreviews.org/errata.shtml

Das könnte Ihnen auch gefallen