<<

Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Review Clinical Research Immunotherapy of Malignant Disease with Tumor –Specific Monoclonal

Michael Campoli1, Robert Ferris2,3,6, Soldano Ferrone2,4,5,6, and Xinhui Wang2,6

Abstract A few tumor antigen (TA)–specific monoclonal antibodies (mAb) have been approved by the Food and Drug Administration for the treatment of several major malignant diseases and are commercially avail- able. Once in the clinic, mAbs have an average success rate of ∼30% and are well tolerated. These results have changed the face of cancer therapy, bringing us closer to more specific and more effective biological therapy of cancer. The challenge facing tumor immunologists at present is represented by the identifica- tion of the mechanism(s) underlying the patients' differential clinical response to mAb-based immu- notherapy. This information is expected to lead to the development of criteria to select patients to be treated with mAb-based immunotherapy. In the past, in vitro and in vivo evidence has shown that TA- specific mAbs can mediate their therapeutic effect by inducing tumor apoptosis, inhibiting the tar- geted antigen function, blocking tumor cell signaling, and/or mediating complement- or cell-dependent lysis of tumor cells. More recent evidence suggests that TA-specific mAb can induce TA-specific cytotoxic T- cell responses by enhancing TA uptake by dendritic cells and cross- of T cells. In this review, we briefly summarize the TA-specific mAbs that have received Food and Drug Administration approval. Next, we review the potential mechanisms underlying the therapeutic efficacy of TA-specific mAbs with empha- sis on the induction of TA-specific cellular immune responses and their potential to contribute to the clinical efficacy of TA-specific mAb-based immunotherapy. Lastly, we discuss the potential negative effect of immune escape mechanisms on the clinical efficacy of TA-specific mAb-based immunotherapy. Clin Cancer Res; 16(1); 11–20. ©2010 AACR.

The concept that antibodies could be used for the treat- quate response rates (RR) observed with first generation ment of malignant disease originated more than a century TA-specific mAb most likely reflected their murine origins, ago (1). The first generation of -based therapies resulting in high immunogenicity and poor ability to re- were based on the use of tumor antigen (TA)–specific al- cruit immune effector mechanisms (4–6). These hurdles logeneic, autologous, or xenogeneic , have been recently overcome by the generation of chime- which were ill suited as cancer-specific therapies because ric, humanized, and human mAb, resulting in reduced or of their limited or lack of specificity and reproducibility. lack of immunogenicity and improved ability to recruit ef- It was not until the development of the hybridoma tech- fector cells (7). nology (2) that antibody-based immunotherapy of malig- Today, TA-specific mAb have been established as highly nant diseases became a practical reality. The hybridoma sensitive and reproducible probes in the diagnostic arena technology enabled the production of a large number of (8–13) as well as in clinically and commercially successful human TA–specific murine monoclonal antibodies therapies for a variety of malignant diseases (3). The average (mAb). The clinical application of some of them yielded clinical success rate of TA-specific mAb-based immunother- a handful of promising results that were however oversha- apy, which manifests itself as statistically significant disease- dowed by disappointing outcomes in early clinical trials free interval and survival prolongation as well as reduction implemented with TA-specific mouse mAb in patients of tumor mass in some of the treated patients, is ∼30% with with various types of cancer (3). In hindsight, the inade- ranges from 0% to 60% (3, 14). Only little is known about why merely a limited percentage of the treated patients re- spond clinically to TA-specific mAb-based immunotherapy. Authors' Affiliations: 1Department of Dermatology, University of Colorado The mechanisms underlying the patients' differential clini- Health Science Center, Denver, Colorado; Departments of 2Immunology, cal response to TA-specific mAb-based immunotherapy 3Otolaryngology, 4Surgery, and 5Pathology, University of Pittsburgh Cancer Institute, and 6Cancer Program, School of Medicine, represent a topic of intense research at present, because this Pittsburgh, University of Pittsburgh, Pennsylvania information has both basic research and clinical relevance. Corresponding Author: Soldano Ferrone, University of Pittsburgh Cancer It will contribute to our understanding of the molecular ba- Institute, 5117 Centre Avenue, Pittsburgh, PA 15213. Phone: 412-623- sis of the clinical efficacy of TA-specific mAb-based immu- 5040; Fax: 412-623-4840; E-mail: [email protected]. notherapy and it will lead to the development of criteria to doi: 10.1158/1078-0432.CCR-09-2345 select patients to be treated with TA-specific mAb-based ©2010 American Association for Cancer Research. immunotherapy. In this review, we first summarize the

www.aacrjournals.org 11

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Campoli et al.

14). Clinical responses have been found to include com- Translational Relevance plete and partial responses as well as statistically sig- nificant increases in progression-free survival and in – Tumor antigen specific monoclonal antibodies disease-free interval and survival in patients with several (mAb) have been successfully implemented into stan- malignant diseases (Table 1). In general, compared with dard treatment regimens for patients with a variety of solid tumors, hematologic have been proven malignant diseases. Despite the clinical successes, scant easier to target with TA-specific mAb-based therapies be- information is available about the variables underlying cause therapeutic efficacy can be achieved at lower doses patients' differential clinical response to mAb-based and tumor penetration is more readily achieved (3, 14). immunotherapy. The scant information in this area Moreover, radioimmunotherapy has been more successful has a negative effect on the optimization of the use for hematologic malignancies such as non–Hodgkin's – of tumor antigen specific mAb in therapeutic strate- lymphoma. However, fewer of these agents are entering gies and represents a major obstacle to the selection clinical trials due to complexities in manufacture and safe- of patients to be treated with mAb-based immunother- ty concerns compared with unconjugated mAb (3, 14). An apy. This review discusses the potential mechanisms additional limitation of radioimmunotherapy relates to underlying the therapeutic efficacy of mAb-based im- the delivery of the dose of radioactivity to the tumor com- munotherapy in patients with malignant disease. This pared with normal tissues (3, 14). The poor specificity in- information is expected to contribute to our under- dex is due to slow pharmacokinetics and slow tumor standing of the molecular basis of the clinical efficacy perfusion. of mAb-based immunotherapy and to lead to the de- Compared with traditional chemothearpy- and radio- velopment of criteria to select patients with malignant therapy-based regimens, in general, the side effects of disease to be treated with mAb-based immunotherapy. immunotherapy with nonconjugated TA-specific mAb are fairly mild (3, 14). Nonetheless, toxicities do occur and may be classified as mechanism-independent and TA-specific mAbs that have received Food and Drug Admin- mechanism-dependent (Table 2). Most of the toxicities re- istration approval. Second, we describe the potential me- lated to TA-specific mAb are mechanism-independent and chanisms underlying the therapeutic efficacy of TA-specific are related to allergic or reactions caused by mAb with emphasis on the induction of TA-specific cellular a containing xenogeneic sequences (3, 14). These immune responses. Finally, we discuss the potential nega- reactions occur during or just after the first injection and tive effect of tumor immune escape mechanisms on the are summarized in Table 2. Moreover, human anti-mouse clinical efficacy of TA-specific mAb-based immunotherapy. immunoglobulin antibodies, including anti-idiotypic anti- bodies, can complex with circulating therapeutic mAb, TA-Specific mAb and inhibit their targeting of tumor cells. It should be not- ed that conjugated antibodies often have a lower therapeu- In recent years, the regulatory approval and sales of new tic index than nonconjugated TA-specific mAb, and for this human medicines indicates an increasing number of bio- reason, they often result in more side effects (3, 14). Rare, logical therapies, i.e., small molecular inhibitors and mAb but more serious side effects of TA-specific mAb-based im- specifically designed to target malignant cells (15).7 In munotherapy are often related to mechanism-dependent fact, the global sale of mAb-derived biological therapies toxicities and result from the binding of a therapeutic in 2006 was $20.6 billion dollars, indicating a major par- antibody to its target antigen (Table 2; refs. 3, 14). adigm shift in industrial research and development from pharmaceutical to biological therapies (15).7 At the end of 2007, >30 years of clinical studies have resulted in the Molecular Mechanisms Underlying the approval of six unconjugated, humanized, or chimeric TA- Therapeutic Efficacy of TA-Specific mAb-Based specific mAbs for cancer therapy along with one drug im- Immunotherapy munoconjugate and two radioisotope immunoconjugates (Table 1; refs. 3, 14). The results of clinical trials in pa- The majority of nonconjugated TA-specific mAb ap- tients treated with TA-specific mAb have shown that the proved for clinical use display intrinsic antitumor effects goal of cancer-targeted therapy is realistic and may be mediated by one or more of the mechanisms outlined superior to older nonspecific conventional - in Table 3. They can be broadly divided into those that and radiotherapy-based regimens, at a minimum enhanc- require immune effector cells and those that do not. It ing their activity. Once in the clinic, TA-specific mAb are should be noted that these mechanisms do not function well tolerated with an average success rate of 30% (3, independently, but extensively interact with each other. The relative importance of each mechanism varies with the type of tumor and the treatment administered. More- 7 Datamonitor. Big Pharma' Turns To Biologics For Growth To 2010: Fi- over, it should be stressed that TA-specific mAb have been nancial And Strategic Segmentation of The 'Big Pharma' Sector By Drug Technology. Datamonitor, May 4, 2006). http://www.marketresearch. clearly shown to be able to inhibit their specific receptor com/map/prod/1285675.html. and induce apoptosis in the targeted tumor cell without

12 Clin Cancer Res; 16(1) January 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

mAb-Based Immunotherapy, T Cells, Escape Mechanisms

the influence of immune cells in vitro. Nevertheless, it is factor-α, chemokines, and that recruit immune still debated whether immune effector cells or receptor effector cells. Consequently, tumor cell proliferation and blockade is the dominant mode of action in vivo or if both angiogenesis are inhibited; antigen presentation is pathways need to cooperate to achieve therapeutic effect. increased; and tumor cells are lysed (31, 32, 34). TA-specific mAb can block activation signals that are Several of the clinically approved TA-specific mAbs, needed for continued malignant cell growth and/or viabil- such as rituximab, cetuximab, trastuzumab, alemtuzumab, ity by blocking the interactions between the ligand and its panitumumab, and 131I-tositumomab, have been shown receptor, inducing modulation of the receptor or interfer- to activate ADCC and CDC in vitro and when administered ing with ligand binding and/or dimerization of the recep- to mice transplanted with TA-expressing tumor cells. In tor (reviewed in refs. 3, 14). The latter mechanisms seem many cases, it is difficult to unravel whether the therapeu- to be particularly important for the epidermal growth tic efficacy of TA-specific mAb depends more on ADCC or factor receptor (EGFR)– (16–21), CD20- (22–26), and CDC; however, there has been some work in this area. It is vascular endothelial growth factor–specific (27–30) noteworthy that the most convincing evidence for ADCC mAb. Alternatively, some TA-specific mAb may exert their and CDC in the antitumor activity of TA-specific mAb effects through Fc-based mechanisms such as antibody- comes from hematologic malignancies, i.e., the CD52- dependent cell-mediated (ADCC) and com- and CD20-specific mAbs alemtuzumab and rituximab, plement-dependent cytotoxicity (CDC; refs. 31, 32). respectively (3, 14). Whether this finding reflects the acces- ADCC and CDC are dependent on interactions of anti- sibility of tumor cells to both TA-specific mAb and plasma body Fc domains with cellular Fcγ receptors (FcγR) ex- of the complement cascade as well as immune ef- pressed on immune accessory cells and with the classic fector cells remains to be determined. Nevertheless the complement-activating protein C1q, respectively. The role of CDC in the antitumor activity of mAb, which rec- potential to mediate ADCC and CDC is a function of an ognize expressed by malignant lymphoid cells, is antibody's subclass and is also influenced by the nature of suggested by experimental and clinical findings. First, its glycosylation (33). Triggering of both ADCC and CDC alemtuzumab mediates significant CDC of chronic lym- not only activates natural killer (NK) cells, , phocytic leukemia (CLL) cells in vitro (35). Second, the mononuclear , and/or dendritic cells (DC), ability of rituximab to cure immunocompetent mice chal- but also induces secretion of IFN-γ, tumor necrosis lenged with murine lymphoma EL4 cells stably transfected

Table 1. FDA-approved TA-specific mAb for human

mAb Target FDA-approved disease

Rituximab CD20 Chimeric IgG1 CD20 (+) low-grade lymphoma,* diffuse large B-cell lymphoma,* follicular lymphoma* 90Y Ibritumomab + tiuxetan CD20 Radiolabeled murine IgG1 CD20(+) low-grade lyphoma† 131I Tositumomab CD20 Radiolabeled murine IgG1 CD20(+) low-grade lyphoma† Alemtuzumab CD53 Humanized IgG1 Chronic lymphocytic leukemia‡ Gemtuzumab + ozogamicin CD33 Recombinant humanized Acute myelogenous leukemia§ IgG4-conjugated to calicheamicin Trastuzumab HER2/neu Humanized IgG1 Her2/neu (+) ∥ Cetuximab EGFR Chimeric IgG1 EGFR(+) Colon cancer¶ Panitumumab EGFR Fully human IgG2 EGFR(+) Colon cancer† Bevacizumab VEGF Humanized IgG1 Colon cancer,** recurrent or advance non–small cell lung cancer, metastatic breast cancer

Abbreviations: FDA, Food and Drug Administration; VEGF, vascular endothelial growth factor. *Low-grade lymphoma: second-line monotherapy; diffuse large B-cell lymphoma and follicular lymphoma: first-line chemoimmune therapy as well as maintenance for follicular lymphoma. †Second-line monotherapy. ‡First- and second-line monotherapy. §>60 y of age, second-line monotherapy. ∥Second-line monotherapy, adjuvant, and first-line chemoimmunetherapy. ¶Second-line monotherapy or chemoimmune therapy. **First-line chemoimmune therapy.

www.aacrjournals.org Clin Cancer Res; 16(1) January 1, 2010 13

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Campoli et al.

Table 2. Toxicities related to TA-specific mAb for human cancers

Mechanism dependent Mechanism independent

Rituximab Transitory B depletion Hypersensitivity reactions* Reactivation of hepatitis B/C, cytomegalovirus, Urticarial or morbilliform and parvovirus B19 skin eruptions Reversible ventricular tachycardia Fever, chills, headache 90Y Ibritumomab + tiuxetan Transitory lymphocyte B depletion Gastrointestinal upset Myelosuppression† Anaphylactoid reactions 131I Tositumomab Hypothyroidism Hypotension Hematologic toxicities Angioedema Thrombocytopenia, neutropenia, and anemia Development of human Transitory lymphocyte B depletion anti-mouse Ig antibodies Alemtuzumab Pancytopenia Autoimmune idiopathic thrombocytopenia and hemolytic anemia ‡ Gemtuzumab + ozogamicin Myelosuppression Trastuzumab Cardiac dysfunction§ Cetuximab Severe infusion reactions∥ Flushing, seborrheic dermatitis, and acneiform eruptions Panitumumab Severe infusion reactions∥ Pulmonary fibrosis Hypomagnesemia Bevacizumab Hypertension, proteinuria, minor bleeding, or thrombosis Bowel perforation¶ Wound healing complications and bleeding**

*Caused by mAb-containing xenogeneic sequences. These reactions occur during or just after the first injection. †Patients must have >25% bone marrow involvement to be considered eligible for treatment. ‡Pneumocystis jiroveci pneumonia and herpes prophylaxis is recommended for patients being treated with alemtuzumab. §When administered in combination with paclitaxel or an anthracycline and cyclophosphamide. ∥Rare and usually occur within minutes of the initial infusion. ¶Alone or in combination with chemotherapy. **In patients treated within 60 d from surgery.

with human CD20 is completely abolished in syngeneic tumor challenge using several antigen/antibody systems. knockout mice lacking C1q (36). Lastly, complement con- Furthermore, the removal of the Fc portion from TA- sumption has been observed after administration of the specific mAb reduces TA-specific mAb-related side effects CD20-specific mAb rituximab to patients with lymphoma as well as their antitumor activity. In contrast, deletion (32, 37). It is noteworthy that target antigen density seems of the inhibitory type II FcγR(FcγRIIb) results in an in- to be a critical factor for CDC, since Golay et al. (39) have creased protective effect, suggesting that FcγRIIb modu- shown that the success of rituximab in mediating CDC lates TA-specific ADCC activity in vivo (31). The role of against malignant B cells is highly dependent on CD20 interactions with cellular FcγR in the clinical efficacy of density. Whether the lack of convincing evidence for the TA-specific mAb-based immunotherapy is further sup- role of CDC in the antitumor activity of TA-specific mAb ported by the statistically significant correlation between used for solid tumors reflects inadequate TA density re- improved clinical RR to mAb-based immunotherapy and mains to be determined. particular “high responder” FcγR polymorphisms in pa- The role of ADCC in the antitumor activity of TA- tients with (a) CD20(+) follicular cell lymphoma treated specific mAb is also suggested by experimental and clinical with rituximab, (b) metastatic colon cancer treated with findings. First, transgenic mice that lack type I and type III cetuximab, or (c) metastatic breast cancer treated with FcγR have provided the conclusive evidence that mAbs are trastuzumab (3). Nevertheless, it should be stressed that capable of targeting immune effector cells to cancer cells the type of FcγR polymorphism does not seem to be asso- in vivo (31, 39). In this regard, FcγR(−)mice,unlike ciated with improved clinical RR in every patient with a wild-type mice, fail to show protective against certain disease and in every disease. For example, Fcγ

14 Clin Cancer Res; 16(1) January 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

mAb-Based Immunotherapy, T Cells, Escape Mechanisms

RIIIa-158F polymorphism is an indicator of improved immunity through the production of , chemo- clinical RR in patients with colon cancer and breast can- kines, and opsonins, which ultimately lead to (a) amplifi- cer treated with cetuximab and trastuzumab, respectively, cation of ADCC and CDC, (b) recruitment and activation of but is linked to poor RR in patients with hematologic immune effector cells, (c) maturation of APC, (d) enhance- malignancies treated with rituximab (3). Moreover, ment of antigen presentation, and (d)generationofTA- FcγRII and RIII polymorphisms are not associated with specific CD4(+) and CD8(+) T-cell immunity (Fig. 1). improved clinical RR in chronic lymphocytic leukemia Moreover, generation of cleavage products from compo- patients treated with alemtuzumab or rituximab, in dif- nents of the complement pathway, e.g., C3b, may acti- fuse large B-cell lymphoma patients treated with rituxi- vate complement receptor 3 on the surface of effector cells mab, and in follicular cell lymphoma non–Hodgkin's and induce complement receptor 3–dependent cellular lymphoma patients treated with sequential cyclophos- cytotoxicity (42). phamide-Adriamycin-vincristine-prednisone (Oncovin) The possibility that TA-specific mAb can enhance the and rituximab (3). Whether these conflicting findings re- immunogenicity of TA and induce TA-specific cellular flect the effect of other variables, such as characteristics immunity is supported by the following lines of preclin- of the TA-specific mAb used, induction of TA-specific ical and clinical evidence. First, incubation of ovarian CTLs, tumor sensitivity to apoptosis, and host immune cancer cells with the CA125-specific mouse IgG1 mAb cell dysfunction, on the role of FcγR polymorphisms in oregovomab in vitro can induce CA125-specific and the patients' clinical response to antibody-based immu- autologous ovarian TA-specific CTL (43). Second, cross- notherapy is not known. presentation mediated by FcγRs on human DC can en- At present, the variables underlying the differential clini- hance the presentation of multiple myeloma antigens to pa- cal RR of the patients treated with antibody-based immu- tient-derived T cells, thus suggesting that uptake of notherapy have not been identified. The data we have antibody-opsonized tumor cells and cellular fragments by summarized suggest that ADCC and CDC play a part in APCs could lead to antigen/ spreading and induction the clinical efficacy of at least some TA-specific mAb. How- of immunity to several TA (40). Third, in vitro incubation of ever, other mechanism(s) are likely to underlie the durable cells with trastuzumab results in augmentation of HER2- clinical responses observed in some patients treated with specific CTL killing of HER2(+) tumors, through the ability TA-specific mAbs, because every tumor cell is not complete- of trastuzumab to mediate HER2 internalization and to en- ly eradicated during therapy with TA-specific mAbs. Given hance HLA class I antigen–restricted presentation of endoge- the number of interactions that are predicted to occur nous HER2 through proteasomal processing (44). Fourth, among different components of the , sever- immunization of mice with DC pulsed with antibody- al investigators have begun to examine the ability of TA-spe- opsonized TA acquired through either FcγR-mediated cific mAb to induce TA-specific cytotoxic T-cell responses in (40, 42, 45) or (43) induces TA-specific mAb-treated patients. In this regard, TA-specific CD4(+) and CD8(+) T-cell–mediated tumor immunity. mAbs are likely to enhance antigen-presenting cell (APC) Lastly, induction of a TA-specific CTL response has been internalization and antigen presentation of TA, as well as recently documented in patients treated with TA-specific cross-priming of T cells through endocytosis and phagocy- mAb-based immunotherapy (46). Six of 10 evaluable tosis of TA-containing immune complexes and antibody- breast cancer patients showed augmented HER2-specific opsonized tumor target cells, respectively (40, 41). Further- CD4 T-cell responses during therapy with trastuzumab. Fur- more, the activation of both ADCC as well as CDC can fur- thermore, the number of patients with detectable HER2- ther augment and focus the generation of TA-specific T-cell specific antibodies among the 27 treated with trastuzumab

Table 3. Molecular mechanisms underlying therapeutic efficacy of TA-specific mAb-based therapy

Immune effector cell independent* Immune effector cell dependent

Induce apoptosis Activation of complement mediated: Induce alteration in intracellular signaling Phagocytosis Inhibit growth factor binding to its cognate receptor CDC Inhibit growth factor receptor activation Trigger antibody-dependent cellular cytotoxicity Induction of tumor cell necrosis or apopotsis leading to Presentation of TA by APC Activation of CD(+) T-cell–mediated kill Activation of B cells and Activation of TA-specific CTL

*Ultimately results in inhibition of cancer cell proliferation, tumor-induced angiogenesis, cancer cell invasion and metastasis, as well as potentating antitumor activity of cytotoxic drugs and radiotherapy.

www.aacrjournals.org Clin Cancer Res; 16(1) January 1, 2010 15

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Campoli et al.

Fig. 1. Triggering of immune effector mechanisms by TA-specific mAb-based immunotherapy. TA-specific mAb may participate in host-dependent immune effector mechanisms including (A) activation of (i) complement-mediated phagocytosis and/or (ii) CDC, (B) induction of ADCC, and/or (C) induction of tumor cell necrosis or apoptosis. The latter mechanisms result in the release of TA as well as the production of cytokines and opsonins that lead to (I) TA uptake, APC maturation and presentation of TA by APC, (II) generation of CD8(+) TA-specific CTL through CD4(+) T-cell help, and (III) activation of CD4(+) T helper cells which leads to activation of NK cells, , and through release of Th1 cytokines, CD4(+) T-cell–mediated killing, and activation of B cells and eosinophils through release of Th2 cytokines.

increased from 8 (29%) before treatment to 15 (56%) dur- FcγR, have been reported to present HLA class I anti- ing treatment (P < 0.001). Similar results have been gen–restricted exogenous antigens (47, 48). An alterna- obtained by one of us (RLF) in studies in progress. Specifi- tive, although not exclusive, possibility is represented cally, HLA class I antigen–restricted, TA-specific CTL were by the induction of anti-idiotypic antibodies, i.e., anti- detected in four of seven HLA-A*0201+ patients with head bodies specific for the variable region of the administered and neck squamous cell cancer treated with the EGFR- TA-specific mAb (49, 50). In some cases, these anti-idio- specific mAb cetuximab. typic antibodies may mimic the TA recognized by the TA-specific mAb are likely to prime antigen-specific original TA-specific mAb and function as a surrogate an- CD4(+) and/or CD8(+) T cells by cross-presentation of tigen (49, 50). The anti-idiotypic antibody as such or TA released by dying cells. In this regard, once TA-specific complexed with the TA-specific mAb may be taken up mAb mediate tumor cell lysis by any of the mechanisms by professional APC and induce a TA-specific T-cell im- outlined above, they can also participate in the cross- mune response (49, 50). presentation of antigen to professional APC (Fig. 2). The term cross-presentation has been used to describe Tumor Cell Resistance to TA-Specific CTL the uptake and representation of cell-associated antigens primarily in the HLA class I antigen pathway of profes- Despite appropriate TA expression, patients may not sional APC (47, 48). Among them, B cells and most have a clinical response to TA-specific mAb-based immu- prominently DC and , all of which express notherapy and/or may develop resistance to therapy during

16 Clin Cancer Res; 16(1) January 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

mAb-Based Immunotherapy, T Cells, Escape Mechanisms

the course of treatment. The multiple mechanisms by cells may change in the expression of molecules such as which TA-specific mAb may exert their antitumor effects TA, HLA class I antigens, and/or antigen-processing ma- make it difficult to determine which “escape” mechanism chinery components; all of them play a crucial role in (s) is/are most important in patients. Furthermore, the the generation of the HLA class I antigen–TA peptide com- intrinsic genetic instability of tumor cells (51) limits our plex, which mediates the recognition of tumor cells by the ability to predict how successful TA-specific mAb-based im- host's CTL. Approximately 10% to 30% of tumor cells may munotherapy will be. Nevertheless, several mechanisms of fail to express TA and a variable degree of interlesional tumor cell resistance to TA-specific mAb-based immu- and intralesional heterogeneity may also be present in notherapy have been suggested (reviewed in refs. 3, 14). a patient (52). Furthermore, tumor cells may present For the purpose of this review, we have focused on the abil- TA-derived peptide analogues with antagonist activity re- ity of tumor cells to evade immune recognition and de- sulting in suboptimal T-cell activation (53). These defects struction by TA-specific CTL. have been found to render malignant cells ineffective tar- If T cells do play a role in the clinical efficacy of TA- gets for TA-specific T cells. Moreover, HLA class I antigen specific mAb-based immunotherapy, then the escape me- downregulation or loss by tumor cells occurs with a fre- chanisms, which have been identified in patients treated quency of about 10% to 80% depending on the type of ma- with T-cell–based immunotherapies, will have relevance. lignancy (53). These abnormalities are caused by distinct The latter include changes in TA, HLA antigen, and anti- molecular mechanisms (reviewed in ref. 54). In some of gen-processing machinery component expression as well these cases, HLA class I antigen expression can be restored as CD4(+)CD25(+) T regulatory cells, each of which mod- by cytokines, providing the potential for benefit by combin- ulates the interaction between tumor cells and T cells. Spe- ing TA-specific mAb-based immunotherapy with adminis- cifically, because of their genetic instability (51), tumor tration of cytokines.

Fig. 2. Induction of TA-specific CTL responses by TA-specific mAb-based immunotherapy. TA-specific mAb may enhance the generation and promote the survival of TA-specific CTL through several mechanisms. TA-specific mAb may (i) induce tumor cell death or activate (ii) ADCC and (iii) CDC. The latter results in (a) the formation of the lytic membrane-attack complex (MAC); (b) the generation of opsonins (C3b), and (d) the release of the C3a and C5a. The culmination of the above events (i-iii) leads to the release of Th1 cytokines, (iv) the formation of TA-specific mAb-TA complexes, and (v) the uptake of TA and TA-specific mAb-TA complexes by APC. Ultimately, mature DC present processed TA to CD4(+) and CD8(+) T cells, and promote the generation of (vi) TA-specific CTL.

www.aacrjournals.org Clin Cancer Res; 16(1) January 1, 2010 17

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Campoli et al.

It is noteworthy that the role of HLA class I antigen ab- mAb to induce more clinically effective TA-specific T-cell normalities in the clinical course of the disease is high- immune responses. First, the type of TA targeted by the lighted by their increased frequency in recurrent cancers TA-specific T-cell responses generated by TA-specific in patients treated with T-cell–based immunotherapy mAb-based immunotherapy may be very important. In (53). These findings have important implications if the this regard, most of the currently used TA in T-cell–based generation of TA-specific CTL underlies, at least in part, immunotherapy trials represent differentiation and/or the clinical efficacy of TA-specific mAb-based immu- shared TA that have been identified from tumor metasta- notherapy. Specifically, the loss of HLA class I antigens ses and CTL from patients who, in the majority of cases, will facilitate the outgrowth of tumor cells that have ac- have failed to reject their cancer. Moreover, many of these quired the ability to escape T-cell recognition because of TA are selected on the basis of their immunogenicity and these defects. If this interpretation is correct, the outgrowth tissue distribution without paying much attention to their of tumor cells with HLA class I antigen abnormalities is function in tumor cell biology. Whether any of the identi- likely to reflect escape of tumor cells from immune recog- fied TA are tumor rejection antigens and whether stage IV nition more than dormancy or ignorance of the patient's cancer patients represent the most appropriate source to immune system. This possibility is supported by the dis- identify clinically relevant TA is not known at present. Sec- ease progression frequently observed in patients with ma- ond, TA-specific mAbs have the potential to generate adap- lignancy in spite of the development and/or persistence of tive immune responses against the entire TA repertoire of a a TA-specific CTL response (54). Moreover, given the high particular patient's tumor because they are able to enhance rate of in tumor cells, the generation of TA- cross-presentation of multiple TA through their direct anti- specific CTL through TA-specific mAb therapy, even when tumor effects. Third, the adaptive immunity potentially successful, is not likely to be able to completely eradicate a generated by TA-specific mAb provides a means to match patient's malignancy, because the eventual outcome is therapeutic regimens with changing tumor antigenic pro- likely to be outgrowth of tumor cells with HLA class I an- files that are likely to occur during the course of the dis- tigen abnormalities. ease. Fourth, the potential ability of TA-specific mAb to HLA class I antigen–TA peptide complex loss by tumor generate adaptive immunity against multiple TA increases cells may not solely be responsible for their immune resis- the chances to also target unique TAs that have been sug- tance because in several cases, TA-specific CTL coexist with gested to be more effective targets of T-cell–based immu- cancer cells expressing the target components required for notherapy than shared TA (60). Lastly, the polyvalent recognition (55). Factors such as the type of T-cell re- vaccine–like potential of TA-specific mAb not only elimi- sponse induced (i.e., tolerance), lack of CD4+ helper-T-cell nates the requirement for patient selection based on HLA activation, host immune cell dysfunction, activating type, but may also be able to counteract escape mechan- antigen-specific CD4(+)CD25(+) T regulatory cells, pro- isms caused by selective loss of HLA class I allospecificities duction of inflammatory and lytic molecules by tumor and TA. However, it should be noted that one disadvan- cells, and/or high tumor burden may also be responsible tage of the use of TA-specific mAb is the lack of knowledge for the lack of association between self–TA-specific T-cell about the identity of the TA-mediating tumor recognition responses and clinical outcome (55–59). that are targeted in adaptive immune responses; the lack of this information hinders the standardization of the assays Toward Improving Immunotherapy designed to monitor immune responses in patients treated with TA-specific mAb-based immunotherapy. Clearly we are at an early stage in our understanding of The ability of TA-specific mAb to induce more clinical- the molecular mechanisms by which TA-specific mAb- ly effective TA-specific T-cell immune responses may also based immunotherapy mediates effective clinical re- stem from their ability to activate multiple arms of the sponses. Although not conclusive, the information we host's immune system including ADCC, CDC, innate im- have reviewed has focused attention on the potential role mune effector cells, DC, NK, B cells, and T cells. This is in of TA-specific adaptive immunity in patients treated with contrast to the narrow view of strictly activating TA-spe- TA-specific mAb-based immunotherapy. Such responses cific CTL that has been attempted in the majority of the would be desirable, because they provide a mechanism T-cell–based vaccine strategies to date. In this regard, re- for long-term protection and immunologic memory. cent progress into our understanding of the mechanisms Moreover, the generation of TA-specific adaptive immuni- underlying activation and proliferation of the adaptive ty provides a mechanism to explain the improved clinical arm of the immune response provides support for the responses in at least some patients treated with repeated concept that tumor cells may directly and/or indirectly administrations of some TA-specific mAb. lead to dysfunction and/or death of immune cells in If T cells do play a role in the clinical efficacy of TA- the tumor microenvironment (55). Therefore, it is unlike- specific mAb-based immunotherapy, one might ask why ly that T-cell–based vaccination strategies will be success- T cells activated in this setting are effective at controlling ful in the setting of tumor cell–induced immune tumor cell growth but not when patients are vaccinated suppression. Through their ability to activate ADCC, with T-cell–based activation strategies. Several possibilities CDC, and APC, TA-specific mAbs may promote a Th1 cy- can be envisioned to underlie the ability of TA-specific tokine–rich environment, thereby enhancing both the

18 Clin Cancer Res; 16(1) January 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

mAb-Based Immunotherapy, T Cells, Escape Mechanisms

number and function of DC, B cells, as well as NK cells Consequently, even when successful, it is likely that im- and ultimately preventing premature death of T-cell effec- munotherapy will facilitate the emergence and expansion tors. It is noteworthy that the potential ability of TA-spe- of tumor cell populations with TA and/or HLA antigen cific mAb to activate NK cells may prove significant, defects and eventually the recurrence of malignant le- because there is growing evidence that NK cells are re- sions. The latter suggests that immunotherapy for the quired for the activation of DC as well as the generation treatment of cancer may only be successful in a limited of antigen-specific T- and B-cell response both in vitro number of patients. Even when successful, it is likely that and in vivo (61–63). the selective pressure imposed by immunotherapy will If it is determined that the induction of such TA-specific facilitate the emergence and expansion of tumor cell po- T-cell immune responses does contribute to the clinical ef- pulations with TA and/or HLA antigen defects and, even- ficacy of TA-specific mAb-based immunotherapy, in vivo tually, the recurrence of malignant lesions. Therefore, it induction of ADCC or CDC leading to TA-specific CTL will be important to combine immunotherapy with other could be viewed as potential biomarkers of clinical re- types of immunologic and nonimmunologic strategies, sponse. In this regard, it should be noted that the ability which use distinct mechanisms to control tumor growth. of TA-specific mAb to induce ADCC, CDC, and TA-specific In this regard, the concomitant targeting of other cells in T-cell immune responses might have important implica- the tumor microenvironment, which are crucial for ma- tions for the development of future TA-specific mAb. As lignant cell survival and proliferation, may counteract noted above, the potential of a TA-specific mAb to medi- the negative effect of tumor cell genetic instability on im- ate ADCC and CDC is a function of not only its subclass munologic and nonimmunologic therapies. but also its degree and type of glycosylation (33). Notably, γ deglycosylated antibodies are unable to bind Fc R and the Disclosure of Potential Conflicts of Interest presence of sialic acid residues confers anti-inflammatory properties to mAb. Therefore, the type and amount of R. Ferris, commercial research grant, Amgen; consultant, glycosylation of TA-specific mAb is likely to play an im- Bristol Myers Squibb. portant role in balancing the proinflammatory versus an- ti-inflammatory properties of administered antibodies. It is possible that this vaccine-like property of TA-specific Grant Support mAb-based immunotherapy could be exploited to selec- tively amplify or bias the resulting adaptive immune re- ASDS Cutting Edge Research Grant (M. Campoli) and by sponse by promoting antigen presentation, host antibody PHS grants IPSO CA097190 (R. Ferris, S. Ferrone), production, and expansion of TA-specific CTL. Nonethe- RO1CA110249 (S. Ferrone), RO1CA113861 (S. Ferrone), less, it must be kept in mind that if indeed T cells are RO1CA10494 (S. Ferrone), R01CA105500 (S. Ferrone), major players in the clinical efficacy of TA-specific RO1 DE19727 (R. Ferris) and RO3 RFA PA-08-209 (X. mAb-based immunotherapy, we will continue to face Wang) awarded by the National Cancer Institute. the negative effect on the outcome of immunotherapy Received 8/27/09; revised 11/2/09; accepted 11/3/09; of escape mechanisms selected for by immune pressure. published OnlineFirst 12/22/09.

References 1. Ehrlich P. Collected studies on immunity. New York, NY: J. Wiley & perative probes and 90 imaging probes. Eur J Nucl Med 1999;26: Sons; 1906. 913–35. 2. Kohler G, Milstein C. Continuous cultures of fused cells secreting 10. Dvorak HF. Vascular permeability factor/vascular endothelial growth antibody of predefined specificity. Nature 1975;256:495–7. factor: a critical in tumor angiogenesis and a potential target 3. Campoli M, Ferrone S. Immunotherapy of malignant disease: The for diagnosis and therapy. J Clin Oncol 2002;20:4368–80. coming age of therapeutic monoclonal antibodies.. In: DeVita V, 11. Kobayashi H, Brechbiel MW. Dendrimer-based nanosized MRI con- Hellman S, Rosenberg S, editors. Cancer: Principles & Practice of trast agents. Curr Pharm Biotechnol 2004;5:539–49. Oncology. 23. New York, NY: Lippincott Williams and Wilkins; 2009, 12. Mohammed AA, Shergill IS, Vandal M, Gujral SS. ProstaScint and its p. 1–18. role in the diagnosis of prostate cancer. Expert Rev Mol Diagn 2007; 4. Badger C, Anasetti C, Davis J, Berstein I. Treatment of malignancy 7:345–9. with unmodified antibody. Pathol Immunopathol Res 1987;6:419–34. 13. Nie S, Xing Y, Kim GJ, Simons JW. Nanotechnology applications in 5. Khazaeli MB, Conry RM, LoBuglio AF. Human immune response to cancer. Annu Rev Biomed Eng 2007;9:257–88. monoclonal antibodies. J Immunother 1994;15:42–52. 14. Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal 6. Groner B, Hartmann C, Wels W. Therapeutic antibodies. Curr Mol antibody successes in the clinic. Nat Biotechnol 2005;23:1073–8. Med 2004;4:539–47. 15. Buelow R, Schooten WV. The future of antibody therapy. Immu- 7. Adams GP, Weiner LM. therapy of cancer. Nat notherapy in 2020. Berlin, Germany: Springer Berlin Heidelberg; Biotechnol 2005;23:1147–57. 2007, p. 83–106. 8. Martin EW, Jr., Thurston MO. The use of monoclonal antibodies 16. Sunada H, Magun BE, Mendelsohn J, MacLeod CL. Monoclonal (MAbs) and the development of an intraoperative hand-held probe antibody against epidermal growth factor receptor is internalized for cancer detection. Cancer Invest 1996;14:560–71. without stimulating receptor phosphorylation. Proc Natl Acad Sci 9. Hoffman EJ, Tornai MP, Janecek M, Patt BE, Iwanczyk JS. Intrao- U S A 1986;83:3825–9.

www.aacrjournals.org Clin Cancer Res; 16(1) January 1, 2010 19

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Campoli et al.

17. Yang X, Jia XC, Corvalan JR, Wang P, Davis CG, Jakobovits A. Erad- in vitro susceptibility to rituximab and complement of B-cell chronic ication of established tumors by a fully human monoclonal antibody lymphocytic leukemia: further regulation by CD55 and CD59. Blood to the epidermal growth factor receptor without concomitant chemo- 2001;98:3383–9. therapy. Cancer Res 1999;59:1236–43. 40. Rafiq K, Bergtold A, Clynes R. -mediated anti- 18. Mendelsohn J, Baselga J. Status of epidermal growth factor receptor gen presentation induces tumor immunity. J Clin Invest 2002; antagonists in the biology and treatment of cancer. J Clin Oncol 110:71–9. 2003;21:2787–99. 41. Dhodapkar KM, Krasovsky J, Williamson B, Dhodapkar MV. Antitu- 19. Li S, Schmitz KR, Jeffrey PD, Wiltzius JJ, Kussie P, Ferguson KM. mor monoclonal antibodies enhance cross-presentation of cellular Structural basis for inhibition of the epidermal growth factor receptor antigens and the generation of myeloma-specific killer T cells by by cetuximab. Cancer Cell 2005;7:301–11. dendritic cells. J Exp Med 2002;195:125–33. 20. Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva FJ. Mechanisms of 42. Kalergis AM, Ravetch JV. Inducing tumor immunity through the se- disease: understanding resistance to HER2-targeted therapy in hu- lective engagement of activating Fc receptors on dendritic cells. man breast cancer. Nat Clin Pract Oncol 2006;3:269–80. J Exp Med 2002;195:1653–9. 21. Hudis CA. Trastuzumab-mechanism of action and use in clinical 43. Gelderman KA, Tomlinson S, Ross GD, Gorter A. Complement func- practice. N Engl J Med 2007;357:39–51. tion in mAb-mediated . Trends Immunol 2004; 22. Alas S, Bonavida B. Rituximab inactivates signal transducer and ac- 25:158–64. tivation of 3 (STAT3) activity in B-non-Hodgkin's lym- 44. Ma J, Samuel J, Kwon GS, Noujaim AA, Madiyalakan R. Induction of phoma through inhibition of the interleukin 10 autocrine/paracrine anti-idiotypic humoral and cellular immune responses by a murine loop and results in down-regulation of Bcl-2 and sensitization to cy- monoclonal antibody recognizing the ovarian carcinoma antigen totoxic drugs. Cancer Res 2001;61:5137–44. CA125 encapsulated in biodegradable microspheres. Cancer Immu- 23. Bezombes C, Grazide S, Garret C, et al. Rituximab antiproliferative nol Immunother 1998;47:13–20. effect in B-lymphoma cells is associated with acid-sphingomyelinase 45. Zum Büschenfelde CM, Hermann C, Schmidt B, Peschel C, Bernhard activation in raft microdomains. Blood 2004;104:1166–73. H. Antihuman epidermal growth factor receptor 2 (HER2) monoclonal 24. Janas E, Priest R, Wilde JI, White JH, Malhotra R. Rituxan (anti-CD20 antibody trastuzumab enhances cytolytic activity of class I-restricted antibody)-induced translocation of CD20 into lipid rafts is crucial for HER2-specific T against HER2-overexpressing tumor calcium influx and apoptosis. Clin Exp Immunol 2005;139:439–46. cells. Cancer Res 2002;62:2244–7. 25. Bonavida B. Rituximab-induced inhibition of antiapoptotic cell sur- 46. Akiyama K, Ebihara S, Yada A, et al. Targeting apoptotic tumor cells vival pathways: implications in chemo/immunoresistance, rituximab to FcR provides efficient and versatile vaccination against tumors by unresponsiveness, prognostic and novel therapeutic interventions. dendritic cells. J Immunol 2003;170:1641–8. 2007;26:3629–36. 47. Taylor C, Hershman D, Shah N, et al. Augmented HER-2 specific im- 26. Ghetie MA, Crank M, Kufert S, Pop I, Vitetta E. Rituximab but not other munity during treatment with trastuzumab and chemotherapy. Clin anti-CD20 antibodies reverses multidrug resistance in 2 B lymphoma Cancer Res 2007;13:5133–43. cell lines, blocks the activity of P- (P-gp), and induces 48. Thery C, Amigorena S. The cell biology of antigen presentation in P-gp to translocate out of lipid rafts. J Immunother 2006;29:536–44. dendritic cells. Curr Opin Immunol 2001;13:45–51. 27. Zhu Z, Hattori K, Zhang H, et al. Inhibition of human leukemia in an 49. Burgdorf S, Kurts C. Endocytosis mechanisms and the cell biology of animal model with human antibodies directed against vascular endo- antigen presentation. Curr Opin Immunol 2008;20:89–95. thelial growth factor receptor 2. Correlation between antibody affinity 50. de Cerio AL, Zabalegui N, Rodríguez-Calvillo M, Inogés S, Bendandi and biological activity. Leukemia 2003;17:604–11. M. Anti- antibodies in cancer treatment. Oncogene 2007;26: 28. Lin MI, Sessa WC. Antiangiogenic therapy: creating a unique “win- 3594–602. dow” of opportunity. Cancer Cell 2004;6:529–31. 51. Pejawar-Gaddy S, Finn OJ. Cancer vaccines: accomplishments and 29. Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normaliza- challenges. Crit Rev Oncol Hematol 2008;67:93–102. tion by VEGFR2 blockade governs brain tumor response to radiation: 52. Onyango P. Genomics and cancer. Curr Opin Oncol 2002;14:79–85. role of oxygenation, angiopoietin-1, and matrix metalloproteinases. 53. Neller MA, López JA, Schmidt CW. Antigens for cancer immunother- Cancer Cell 2004;6:553–63. apy. Semin Immunol 2008;20:286–95. 30. Cao Y. Molecular mechanisms and therapeutic development of an- 54. Chang CC, Campoli M, Ferrone S. Classical and nonclassical HLA giogenesis inhibitors. Adv Cancer Res 2008;100:113–31. class I antigen and NK Cell-activating ligand changes in malignant 31. Cassard L, Cohen-Solal J, Camilleri-Broët S, et al. Fc γ receptors and cells: current challenges and future directions. Adv Cancer Res cancer. Springer Semin Immunopathol 2006;28:321–8. 2005;93:189–234. 32. Wang SY, Weiner G. Complement and cellular cytotoxicity in anti- 55. Xue SA, Stauss HJ. Enhancing immune responses for cancer thera- body therapy of cancer. Expert Opin Biol Ther 2008;8:759–68. py. Cell Mol Immunol 2007;4:173–84. 33. Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of 56. Whiteside T, Campoli M, Ferrone S. Tumor induced immune resulting from Fc sialylation. Science 2006;313: suppression mechanisms and possible solutions. In: Nagorsen F, 670–3. Marincola M, editors. Analyzing Responses. How to analyze 34. Strome SE, Sausville EA, Mann D. A mechanistic perspective of cellular immune responses against tumor associated antigens. monoclonal antibodies in cancer therapy beyond target-related ef- Netherlands: Springer-Verlag; 2005, p. 43–81. fects. Oncologist 2007;12:1084–95. 57. Nizar S, Copier J, Meyer B, et al. T-regulatory cell modulation: the 35. Lopez-Albaitero A, Lee SC, Morgan S, et al. Role of polymorphic Fc γ future of cancer immunotherapy? Br J Cancer 2009;100:1697–703. receptor IIIa and EGFR expression level in cetuximab mediated, NK 58. Ferrone S. Tumour immune escape. Semin Cancer Biol 2002;12. cell dependent in vitro cytotoxicity of head and neck squamous cell 59. Rivoltini L, Carrabba M, Huber V, et al. Immunity to cancer: attack carcinoma cells. Cancer Immunol Immunother 2009;58:1855–64. and escape in T lymphocyte-tumor cell interaction. Immunol Rev 36. Zent CS, Secreto CR, LaPlant BR, et al. Direct and complement 2002;188:97–113. dependent cytotoxicity in CLL cells from patients with high-risk 60. Yu Z, Restifo NP. Cancer vaccines: progress reveals new complex- early-intermediate stage chronic lymphocytic leukemia (CLL) trea- ities. J Clin Invest 2002;110:289–94. ted with alemtuzumab and rituximab. Leuk Res 2008;32:1849–56. 61. Andoniou CE, Coudert JD, Degli-Esposti MA. Killers and beyond: 37. Di Gaetano N, Cittera E, Nota R, et al. Complement activation deter- NK-cell-mediated control of immune responses. Eur J Immunol mines the therapeutic activity of rituximab in vivo. J Immunol 2003; 2008;38:2938–42. 171:411581–7. 62. Werner H. Tolerance and reactivity of NK cells: two sides of the same 38. Coiffi B. Rituximab therapy in malignant lymphoma. Oncogene 2007; coin? Eur J Immunol 2008;38:2930–3. 26:3603–13. 63. Zimmer J, Andrès E, Hentges F. NK cells and Treg cells: a fascinating 39. Golay J, Lazzari M, Facchinetti V, et al. CD20 levels determine the dance cheek to cheek. Eur J Immunol 2008;38:2942–5.

20 Clin Cancer Res; 16(1) January 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst December 22, 2009; DOI: 10.1158/1078-0432.CCR-09-2345

Immunotherapy of Malignant Disease with Tumor Antigen− Specific Monoclonal Antibodies

Michael Campoli, Robert Ferris, Soldano Ferrone, et al.

Clin Cancer Res 2010;16:11-20. Published OnlineFirst December 22, 2009.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-09-2345

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/16/1/11. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research.