101
|
Malphettes L, Freyvert Y, Chang J, Liu PQ, Chan E, Miller JC, Zhou Z, Nguyen T, Tsai C, Snowden AW, Collingwood TN, Gregory PD, Cost GJ. Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnol Bioeng 2010; 106:774-83. [PMID: 20564614 DOI: 10.1002/bit.22751] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
IgG1 antibodies produced in Chinese hamster ovary (CHO) cells are heavily alpha1,6-fucosylated, a modification that reduces antibody-dependent cellular cytotoxicity (ADCC) and can inhibit therapeutic antibody function in vivo. Addition of fucose is catalyzed by Fut8, a alpha1,6-fucosyltransferase. FUT8(-/-) CHO cell lines produce completely nonfucosylated antibodies, but the difficulty of recapitulating the knockout in protein-production cell lines has prevented the widespread adoption of FUT8(-/-) cells as hosts for antibody production. We have created zinc-finger nucleases (ZFNs) that cleave the FUT8 gene in a region encoding the catalytic core of the enzyme, allowing the functional disruption of FUT8 in any CHO cell line. These reagents produce FUT8(-/-) CHO cells in 3 weeks at a frequency of 5% in the absence of any selection. Alternately, populations of ZFN-treated cells can be directly selected to give FUT8(-/-) cell pools in as few as 3 days. To demonstrate the utility of this method in bioprocess, FUT8 was disrupted in a CHO cell line used for stable protein production. ZFN-derived FUT8(-/-) cell lines were as transfectable as wild-type, had similar or better growth profiles, and produced equivalent amounts of antibody during transient transfection. Antibodies made in these lines completely lacked core fucosylation but had an otherwise normal glycosylation pattern. Cell lines stably expressing a model antibody were made from wild-type and ZFN-generated FUT8(-/-) cells. Clones from both lines had equivalent titer, specific productivity distributions, and integrated viable cell counts. Antibody titer in the best ZFN-generated FUT8(-/-) cell lines was fourfold higher than in the best-producing clones of FUT8(-/-) cells made by standard homologous recombination in a different CHO subtype. These data demonstrate the straightforward, ZFN-mediated transfer of the Fut8- phenotype to a production CHO cell line without adverse phenotypic effects. This process will speed the production of highly active, completely nonfucosylated therapeutic antibodies.
Collapse
|
102
|
Metastatic melanomas express inhibitory low affinity fc gamma receptor and escape humoral immunity. Dermatol Res Pract 2010; 2010:657406. [PMID: 20672001 PMCID: PMC2905727 DOI: 10.1155/2010/657406] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 04/01/2010] [Indexed: 12/22/2022] Open
Abstract
Our research, inspired by the pioneering works of Isaac Witz in the 1980s, established that 40% of human metastatic melanomas express ectopically inhibitory Fc gamma receptors (FcγRIIB), while they are detected on less than 5% of primary cutaneous melanoma and not on melanocytes. We demonstrated that these tumoral FcγRIIB act as decoy receptors that bind the Fc portion of antimelanoma IgG, which may prevent Fc recognition by the effector cells of the immune system and allow the metastatic melanoma to escape the humoral/natural immune response. The FcγRIIB is able to inhibit the ADCC (antibody dependent cell cytotoxicity) in vitro. Interestingly, the percentage of melanoma expressing the FcγRIIB is high (70%) in organs like the liver, which is rich in patrolling NK (natural killer) cells that exercise their antitumoral activity by ADCC. We found that this tumoral FcγRIIB is fully functional and that its inhibitory potential can be triggered depending on the specificity of the anti-tumor antibody with which it interacts.
Together these observations elucidate how metastatic melanomas interact with and potentially evade humoral immunity and provide direction for the improvement of anti-melanoma monoclonal antibody therapy.
Collapse
|
103
|
Abès R, Dutertre CA, Agnelli L, Teillaud JL. Activating and inhibitory Fcgamma receptors in immunotherapy: being the actor or being the target. Expert Rev Clin Immunol 2010; 5:735-47. [PMID: 20477693 DOI: 10.1586/eci.09.57] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Membrane Fcgamma receptors (FcgammaRs) can act either as potent activators of effector cell functions or as inhibitors of receptor-mediated cell activation following engagement by IgG antibodies bound to their target molecules. The remarkable ability of activating FcgammaRs to trigger antibody-dependent cellular cytotoxicity, cytokine release and phagocytosis/endocytosis followed by antigen presentation has stimulated the development of a number of therapeutic monoclonal antibodies whose Fc regions have been engineered to optimize their effector functions, mostly their killing activities. Conversely, the demonstration that inhibitory FcgammaRs can block or downmodulate effector functions has led to the concept that targeting these receptors is of interest in a number of pathologies. The use of bispecific antibodies leading to the crosslinking of FcgammaRIIB with activating receptors could induce immunomodulation in autoimmune or allergic diseases. Alternatively, the use of cytotoxic/antagonist anti-FcgammaRIIB antibodies could kill FcgammaRIIB-positive tumor cells or prevent the downmodulation of activating receptors. Thus, antibodies engineered to preferentially target activating or inhibitory FcgammaRs are currently being designed for therapeutic use.
Collapse
Affiliation(s)
- Riad Abès
- INSERM UMRS 872, Cordeliers Research Center, Pierre & Marie Curie University and Paris-Descartes University, Paris, France.
| | | | | | | |
Collapse
|
104
|
de Haij S, Jansen JHM, Boross P, Beurskens FJ, Bakema JE, Bos DL, Martens A, Verbeek JS, Parren PWHI, van de Winkel JGJ, Leusen JHW. In vivo cytotoxicity of type I CD20 antibodies critically depends on Fc receptor ITAM signaling. Cancer Res 2010; 70:3209-17. [PMID: 20354182 DOI: 10.1158/0008-5472.can-09-4109] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibody-Fc receptor (FcR) interactions play an important role in the mechanism of action of most therapeutic antibodies against cancer. Effector cell activation through FcR triggering may induce tumor cell killing via antibody-dependent cellular cytotoxicity (ADCC). Reciprocally, FcR cross-linking of antibody may lead to the induction of apoptotic signaling in tumor cells. The relative importance of these bisecting pathways to in vivo antibody activity is unknown. To unravel these roles, we developed a novel mouse model with normal FcR expression but in which FcR signaling was inactivated by mutation of the associated gamma-chain. Transgenic mice showed similar immune complex binding compared with wild-type mice. In contrast, ADCC of cells expressing frequently used cancer targets, such as CD20, epidermal growth factor receptor, Her2, and gp75, was abrogated. Using the therapeutic CD20 antibodies ofatumumab and rituximab, we show that FcR cross-linking of antibody-antigen immune complexes in the absence of gamma-chain signaling is insufficient for their therapeutic activity in vivo. ADCC therefore represents an essential mechanism of action for immunotherapy of lymphoid tumors.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- Antibodies/chemistry
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antibodies, Monoclonal, Murine-Derived
- Antigens, CD20/chemistry
- Apoptosis
- ErbB Receptors/metabolism
- Humans
- Immunotherapy/methods
- Melanoma, Experimental
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Mutation
- Receptors, Fc/chemistry
- Rituximab
- Sequence Homology, Amino Acid
- Signal Transduction
Collapse
Affiliation(s)
- Simone de Haij
- Genmab, Immunotherapy Laboratory, Department of Immunology, University Medical Center, Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Abstract
Rituximab is a mainstay in the therapy for a broad variety of B-cell malignancies. Despite its undeniable therapeutic value, we still do not fully understand the mechanisms of action responsible for rituximab's anti-tumor effects. Direct signaling, complement-mediated cytotoxicity (CMC), and antibody-dependent cellular cytotoxicity (ADCC) all appear to play a role in rituximab efficacy. In vitro, animal model and clinical data addressing each of these mechanisms of action are reviewed, as are data speaking to the complexity of interactions between these mechanisms. Taken together, these data suggest different mechanisms are likely important in different scenarios. Study of the complex mechanisms of action that contribute to the clinical efficacy of rituximab have led to novel clinical trials including novel combinations, schedules, and generation of additional antibodies designed to have even greater effect. Such studies need to be accompanied by rigorous correlative analysis if we are to understand the importance of various mechanisms of action of rituximab and use that information to improve on what is already an indispensable component of therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibody-Dependent Cell Cytotoxicity
- Antigens, CD20/drug effects
- Antigens, CD20/immunology
- Antigens, CD20/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B-Lymphocytes/drug effects
- Cell Death/drug effects
- Cell Line, Tumor/drug effects
- Clinical Trials as Topic
- Complement Activation/drug effects
- Cytotoxicity, Immunologic
- Drug Resistance, Neoplasm/drug effects
- Drug Screening Assays, Antitumor
- Drug Synergism
- GPI-Linked Proteins
- Humans
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/physiology
- Lymphoma/drug therapy
- Lymphoma/immunology
- Membrane Microdomains/drug effects
- Mice
- Receptors, IgG/drug effects
- Receptors, IgG/physiology
- Rituximab
Collapse
Affiliation(s)
- George J Weiner
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242-1002, USA
| |
Collapse
|
106
|
Quaglino E, Mastini C, Amici A, Marchini C, Iezzi M, Lanzardo S, De Giovanni C, Montani M, Lollini PL, Masucci G, Forni G, Cavallo F. A Better Immune Reaction to Erbb-2 Tumors Is Elicited in Mice by DNA Vaccines Encoding Rat/Human Chimeric Proteins. Cancer Res 2010; 70:2604-12. [DOI: 10.1158/0008-5472.can-09-2548] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
107
|
Park HJ, Kim HS, Chung DH. Fcgamma receptors modulate pulmonary inflammation by activating innate immune cells in murine hypersensitivity pneumonitis. Immune Netw 2010; 10:26-34. [PMID: 20228933 PMCID: PMC2837154 DOI: 10.4110/in.2010.10.1.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 01/29/2010] [Accepted: 02/10/2010] [Indexed: 01/21/2023] Open
Abstract
Background Hypersensitivity pneumonitis (HP) is an interstitial lung disease that develops following repeated exposure to inhaled particulate antigens. The family of Fcγ receptors (FcγRs) has emerged as central regulators for modulating both pro-and anti-inflammatory responses. However, the role of FcγRs in the development of HP has not been investigated yet. Methods To explore the functional roles of FcγRs in HP, FcγR-/- and B6 mice were challenged with Saccharopolyspora rectivirgula (SR) antigen intranasally, and compared these mice in terms of the histological change, infiltrated immune cells in BALF and in vitro immune responses. Results FcγR-/- mice exhibited attenuation of HP in terms of histological alterations, and reduced numbers of neutrophils and macrophages in and the increased CD4:CD8 ratio of bronchoalveolar lavage fluid. The lungs of FcγR-/- mice showed high production of Th2 cytokine such as IL-4 and slightly low production of Th1 cytokine, INF-γ compared to those of B6 mice. However, SR-specific adaptive immune responses of FcγR-/- mice were similar to those of B6 mice. Conclusion These results demonstrate that activating Fcγ receptors play an important role in activating neutrophils and macrophages in pulmonary inflammation and inducing Th1 differentiation by regulating cytokine expression in SR-induced HP.
Collapse
Affiliation(s)
- Hyo Jin Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | | | | |
Collapse
|
108
|
Abès R, Teillaud JL. Impact of Glycosylation on Effector Functions of Therapeutic IgG. Pharmaceuticals (Basel) 2010; 3:146-157. [PMID: 27713246 PMCID: PMC3991024 DOI: 10.3390/ph3010146] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 12/30/2009] [Accepted: 01/08/2010] [Indexed: 01/14/2023] Open
Abstract
Human IgG has only one conserved glycosylation site located in the Cγ2 domain of the Fc region that accounts for the presence of two sugar moieties per IgG. These IgG sugar cores play a critical role in a number of IgG effector functions. In the present review, we describe the main characteristics of IgG Fc glycosylation and some abnormalities of serum IgG glycosylation. We also discuss how glycosylation impacts on monoclonal antibodies (mAbs) and IVIg effector functions and how these molecules can be engineered. Several therapeutic antibodies have now been engineered to be no- or low-fucose antibodies and are currently tested in clinical trials. They exhibit an increased binding to activating FcγRIIIA and trigger a strong antibody-dependent cell cytotoxicity (ADCC) as compared to their highly-fucosylated counterparts. They represent a new generation of therapeutic antibodies that are likely to show a better clinical efficacy in patients, notably in cancer patients where cytotoxic antibodies are needed.
Collapse
Affiliation(s)
- Riad Abès
- INSERM UMRS 872, Paris, F-75006 France.
- Cordeliers Research Center, Université Pierre & Marie Curie, UMRS 872, Paris, F-75006, France.
- Université Paris-Descartes, UMRS 872, Paris, F-75006 France.
- Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France.
| | - Jean-Luc Teillaud
- INSERM UMRS 872, Paris, F-75006 France.
- Cordeliers Research Center, Université Pierre & Marie Curie, UMRS 872, Paris, F-75006, France.
- Université Paris-Descartes, UMRS 872, Paris, F-75006 France.
| |
Collapse
|
109
|
Abdel-Motal UM, Wang S, Awad A, Lu S, Wigglesworth K, Galili U. Increased immunogenicity of HIV-1 p24 and gp120 following immunization with gp120/p24 fusion protein vaccine expressing alpha-gal epitopes. Vaccine 2009; 28:1758-65. [PMID: 20034607 DOI: 10.1016/j.vaccine.2009.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 11/23/2009] [Accepted: 12/09/2009] [Indexed: 01/18/2023]
Abstract
Developing an effective HIV-1 vaccine will require strategies to enhance antigen presentation to the immune system. In a previous study we demonstrated a marked increase in immunogenicity of the highly glycosylated HIV-1 gp120 protein following enzymatic addition of alpha-gal epitopes to the carbohydrate chains. In the present study we determined whether gp120(alphagal) can also serve as an effective platform for targeting other HIV-1 proteins to APC and thus increase immunogenicity of both proteins. For this purpose we produced a recombinant fusion protein between gp120 and the HIV-1 matrix p24 protein (gp120/p24). Multiple alpha-gal epitopes were synthesized enzymatically on the gp120 portion of the fusion protein to generate a gp120(alphagal)/p24 vaccine. Immune responses to gp120(alphagal)/p24 compared to gp120/p24 vaccine lacking alpha-gal epitopes were evaluated in alpha1,3galactosyltransferase knockout (KO) mice. These mice lack alpha-gal epitopes and, therefore, are capable of producing the anti-Gal antibody. T cell responses to p24, as assessed by ELISPOT and by CD8+ T cells intracellular staining assays for IFNgamma, was on average 12- and 10-fold higher, respectively, in gp120(alphagal)/p24 immunized mice than in mice immunized with gp120/p24. In addition, cellular and humoral immune responses against gp120 were higher by 10-30-fold in mice immunized with gp120(alphagal)/p24 than in gp120/p24 immunized mice. Our data suggest that the alpha-gal epitopes on the gp120 portion of the fusion protein can significantly augment the immunogenicity of gp120, as well as that of the fused viral protein which lacks alpha-gal epitopes. This strategy of anti-Gal mediated targeting to APC may be used for production of effective HIV-1 vaccines comprised of various viral proteins fused to gp120.
Collapse
Affiliation(s)
- Ussama M Abdel-Motal
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | |
Collapse
|
110
|
Masuda A, Yoshida M, Shiomi H, Morita Y, Kutsumi H, Inokuchi H, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Role of Fc Receptors as a therapeutic target. ACTA ACUST UNITED AC 2009; 8:80-6. [PMID: 19275696 DOI: 10.2174/187152809787582525] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcgammaR), high-affinity IgE receptor (FcepsilonRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcgammaRs have been well known to play an important role in many biologic processes including those associated with the response to infection and cancer as well as in the pathogenesis of immune-mediated diseases. Both positive and negative regulatory function has ascribed to Fc receptors and FcgammaRs in particular which serve to establish a threshold for immune cell activation. In other cases, Fc receptors such as FcRn possess a novel structure and function by playing a major role in the transport of IgG across polarized epithelial barriers at mucosal surfaces and in the regulation of IgG half-life. These diverse functions highlight the potential effectiveness of targeting Fc receptors for therapeutic purposes. This review summarizes new information available in the therapeutic applications of this biology.
Collapse
Affiliation(s)
- Atsuhiro Masuda
- Department of Gastroenterology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Depletion of the C3 component of complement enhances the ability of rituximab-coated target cells to activate human NK cells and improves the efficacy of monoclonal antibody therapy in an in vivo model. Blood 2009; 114:5322-30. [PMID: 19805620 DOI: 10.1182/blood-2009-01-200469] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Growing evidence indicates antibody-dependent cellular cytotoxicity (ADCC) contributes to the clinical response to monoclonal antibody (mAb) therapy of lymphoma. Recent in vitro analysis suggests C3b can inhibit mAb-induced natural killer (NK)-cell activation and ADCC. Further studies were conducted to assess the effect of C3 depletion on mAb-induced NK activation and therapy of lymphoma. Normal human serum inhibited the ability of rituximab-coated lymphoma cells to activate NK cells as previously reported. Serum did not inhibit NK-cell activation when it was preincubated with cobra venom factor (CVF) to deplete C3. Similar results were found when transudative pleural fluid or nonmalignant ascites was used as surrogates for extravascular fluid, suggesting the inhibitory effect of complement may be present in the extravascular compartment, in which many malignant lymphocytes reside. In vivo, C3 was depleted before mAb treatment in a syngeneic murine model of lymphoma. Survival of lymphoma-bearing mice after treatment with CVF plus mAb and with a human C3 derivative with CVF-like functions (HC3-1496) plus mAb was both superior to that of mAb alone. These studies show that complement depletion enhances NK-cell activation induced by rituximab-coated target cells and improves the efficacy of mAb therapy in a murine lymphoma model.
Collapse
|
112
|
Clark AJ, Diamond M, Elfline M, Petty HR. Calicum microdomains form within neutrophils at the neutrophil-tumor cell synapse: role in antibody-dependent target cell apoptosis. Cancer Immunol Immunother 2009; 59:149-59. [PMID: 19593564 DOI: 10.1007/s00262-009-0735-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 06/29/2009] [Indexed: 01/03/2023]
Abstract
Ca(2+) messages are broadly important in cellular signal transduction. In immune cells, Ca(2+) signaling is an essential step in many forms of activation. Neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) is one form of leukocyte activation that plays an important role in tumor cell killing in vitro and in patient care. Using fluorescence methodologies, we found that neutrophils exhibit Ca(2+) signals during ADCC directed against breast fibrosarcoma cells. Importantly, these signals were localized to Ca(2+) microdomains at the neutrophil-to-tumor cell interface where they display dynamic features such as movement, fusion, and fission. These signals were blocked by the intracellular Ca(2+) buffer BAPTA. At the neutrophil-tumor cell synapse, the neutrophil's cytoplasm was enriched in STIM1, a crucial mediator of Ca(2+) signaling, whereas the Ca(2+)-binding proteins calbindin and parvalbumin were not affected. Our findings suggest that Ca(2+) microdomains are due to an active signaling process. As Ca(2+) signals within neutrophils were necessary for specific tumor cell apoptosis, a central role of microdomains in leukocyte-mediated tumor cell destruction is indicated.
Collapse
Affiliation(s)
- Andrea J Clark
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | | | | | | |
Collapse
|
113
|
Cardarelli PM, Moldovan-Loomis MC, Preston B, Black A, Passmore D, Chen TH, Chen S, Liu J, Kuhne MR, Srinivasan M, Assad A, Witte A, Graziano RF, King DJ. In vitro and In vivo Characterization of MDX-1401 for Therapy of Malignant Lymphoma. Clin Cancer Res 2009; 15:3376-83. [DOI: 10.1158/1078-0432.ccr-08-3222] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
114
|
Cassard L, Cohen-Solal JFG, Fournier EM, Camilleri-Broët S, Spatz A, Chouaïb S, Badoual C, Varin A, Fisson S, Duvillard P, Boix C, Loncar SM, Sastre-Garau X, Houghton AN, Avril MF, Gresser I, Fridman WH, Sautès-Fridman C. Selective expression of inhibitory Fcgamma receptor by metastatic melanoma impairs tumor susceptibility to IgG-dependent cellular response. Int J Cancer 2009; 123:2832-9. [PMID: 18798552 DOI: 10.1002/ijc.23870] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
During melanoma progression, patients develop anti-tumor immunity including the production of anti-tumor antibodies. Although the strategies developed by malignant cells to escape anti-tumor cellular immunity have been extensively investigated, little is known about tumor resistance to humoral immunity. The main effect of IgG antibodies is to activate the immune response by binding to host Fc gamma receptors (FcgammaR) expressed by immune cells. We previously reported in a limited study that some human metastatic melanoma cells ectopically express the FcgammaRIIB1, an inhibitory isoform of FcgammaR. By analyzing a large panel of different types of human primary and metastatic solid tumors, we report herein that expression of FcgammaRIIB is restricted to melanoma and is acquired during tumor progression. We show that FcgammaRIIB expression prevents the lysis of human metastatic melanoma cells by NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) in vitro, independently of the intracytoplasmic region of FcgammaRIIB. Using experimental mouse models, we demonstrate that expression of FcgammaRIIB protects B16F0 melanoma tumors from the ADCC induced by monoclonal and polyclonal anti-tumor IgG in vivo. Thus, our results identify FcgammaRIIB as a marker of human metastatic melanoma that impairs the tumor susceptibility to FcgammaR-dependent innate effector responses.
Collapse
Affiliation(s)
- Lydie Cassard
- INSERM, U872, Microenvironnement immunitaire et tumeurs, Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Nakamura A, Kubo T, Takai T. Fc receptor targeting in the treatment of allergy, autoimmune diseases and cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:220-33. [PMID: 19065795 DOI: 10.1007/978-0-387-09789-3_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fc receptors (FcRs) play an important role in the maintenance of an adequate activation threshold of various cells in antibody-mediated immune responses. Analyses of murine models show that the inhibitory FcR, FcyRIIB plays a pivotal role in the suppression of antibody-mediated allergy and autoimmunity. On the other hand, the activating-type FcRs are essential for the development of these diseases, suggesting that regulation of inhibitory or activating FcR is an ideal target for a therapeutic agent. Recent experimental or clinical studies also indicate that FcRs function as key receptors in the treatment with monoclonal antibodies (mAbs) therapy. This review summarizes FcR functions and highlights possible FcR-targeting therapies including mAb therapies for allergy, autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Akira Nakamura
- Department of Experimental Immunology and CREST program of Japan Science and Technology Agency, Institute of Development, Aging and Cancer, Tohoku University, Seiryo 4-1, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
116
|
Saenger YM, Li Y, Chiou KC, Chan B, Rizzuto G, Terzulli SL, Merghoub T, Houghton AN, Wolchok JD. Improved tumor immunity using anti-tyrosinase related protein-1 monoclonal antibody combined with DNA vaccines in murine melanoma. Cancer Res 2008; 68:9884-91. [PMID: 19047169 PMCID: PMC2742375 DOI: 10.1158/0008-5472.can-08-2233] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Passive immunization with monoclonal antibody TA99 targeting melanoma differentiation antigen tyrosinase-related protein-1 (Tyrp1; gp75) and active immunization with plasmid DNA encoding altered Tyrp1 both mediate tumor immunity in the B16 murine melanoma model. We report here that TA99 enhances Tyrp1 DNA vaccination in the treatment of B16 lung metastases, an effect mediated by immunologic mechanisms as Tyrp1 has no known role in regulating tumor growth. TA99 is shown to increase induction of anti-Tyrp1 CD8+T-cell responses to DNA vaccination against Tyrp1 as assessed by IFN-gamma ELISPOT assays. Immunohistochemistry studies reveal that TA99 localizes rapidly and specifically to B16 lung nodules. Augmentation of T-cell responses is dependent on the presence of tumor as well as on activating Fc receptors. Furthermore, TA99 enhances DNA vaccination against a distinct melanoma antigen, gp100(pmel17/silver locus), improving antitumor efficacy, augmenting systemic CD8+ T-cell responses to gp100, and increasing CD8+ T-cell infiltration at the tumor site. Epitope spreading was observed, with CD8+ T-cell responses generated to Tyrp1 peptide in mice receiving gp100 DNA vaccination in the presence of TA99. Finally, we show that TA99 improves therapeutic efficacy of DNA vaccination combined with adoptive T-cell transfer in treatment of established subcutaneous B16 melanoma. In conclusion, TA99 enhances DNA vaccination against both the target antigen Tyrp1 and a distinct melanoma antigen gp100 in an Fc receptor-dependent mechanism, consistent with enhanced cross-presentation of tumor-derived antigen. Monoclonal antibodies should be tested as vaccine adjuvants in the treatment of cancer.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- DNA/genetics
- DNA/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- Humans
- Immunotherapy/methods
- Immunotherapy, Adoptive/methods
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/secondary
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Oxidoreductases/genetics
- Oxidoreductases/immunology
- Receptors, Fc/immunology
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacology
- gp100 Melanoma Antigen
Collapse
Affiliation(s)
- Yvonne M. Saenger
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Yanyun Li
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Karoline C. Chiou
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Brian Chan
- State University of New York Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Gabrielle Rizzuto
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Stephanie L. Terzulli
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Taha Merghoub
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
| | - Alan N. Houghton
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
- Weill Medical College of Cornell University, New York, NY, 10021
| | - Jedd D. Wolchok
- The Swim Across America Laboratory, Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Room Z-1445, New York, NY 10021
- Weill Medical College of Cornell University, New York, NY, 10021
| |
Collapse
|
117
|
Otten MA, van der Bij GJ, Verbeek SJ, Nimmerjahn F, Ravetch JV, Beelen RHJ, van de Winkel JGJ, van Egmond M. Experimental Antibody Therapy of Liver Metastases Reveals Functional Redundancy between FcγRI and FcγRIV. THE JOURNAL OF IMMUNOLOGY 2008; 181:6829-36. [DOI: 10.4049/jimmunol.181.10.6829] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
118
|
Wang SY, Weiner G. Complement and cellular cytotoxicity in antibody therapy of cancer. Expert Opin Biol Ther 2008; 8:759-68. [PMID: 18476787 DOI: 10.1517/14712598.8.6.759] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effective and practical use of mAbs in cancer therapy became a reality with the development of the chimeric anti-CD20 mAb, rituximab. Several additional mAbs have since been approved for clinical use. Despite these successes, the mechanisms by which mAbs mediate antitumor activity are still unclear. Preclinical studies indicate complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) both can contribute to mAb-induced tumor cell lysis. However, evidence related to the relative clinical importance of each mechanism, and whether they are synergistic or antagonistic, is conflicting. New ways to enhance both CDC and ADCC are being developed in attempt to develop a more effective anticancer mAb. Continued research on the mechanisms of mAb therapy will be necessary if we are to take optimal advantage of the current mAbs and develop more effective mAbs in the future.
Collapse
Affiliation(s)
- Siao-Yi Wang
- University of Iowa, 5970 JPP, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
119
|
Abstract
Triggering of tumour cell apoptosis is the foundation of many cancer therapies. Death receptors of the tumour necrosis factor (TNF) superfamily have been largely characterized, as have the signals that are generated when these receptors are activated. TNF-related apoptosis-inducing ligand (TRAIL) receptors (TRAILR1 and TRAILR2) are promising targets for cancer therapy. Herein we review what is known about the molecular control of TRAIL-mediated apoptosis, the role of TRAIL in carcinogenesis and the potential therapeutic utility of recombinant TRAIL and agonistic antibodies against TRAILR1 and TRAILR2.
Collapse
Affiliation(s)
- Ricky W Johnstone
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia.
| | | | | |
Collapse
|
120
|
Arnon TI, Markel G, Bar-Ilan A, Hanna J, Fima E, Benchetrit F, Galili R, Cerwenka A, Benharroch D, Sion-Vardy N, Porgador A, Mandelboim O. Harnessing soluble NK cell killer receptors for the generation of novel cancer immune therapy. PLoS One 2008; 3:e2150. [PMID: 18478075 PMCID: PMC2364651 DOI: 10.1371/journal.pone.0002150] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 04/01/2008] [Indexed: 12/20/2022] Open
Abstract
The natural cytotoxic receptors (NCRs) are a unique set of activating proteins expressed mainly on the surface of natural killer (NK) cells. The NCRs, which include three members; NKp46, NKp44 and NKp30, are critically involved in NK cytotoxicity against different targets, including a wide range of tumor cells derived from various origins. Even though the tumor ligands of the NCRs have not been identified yet, the selective manner by which these receptors target tumor cells may provide an excellent basis for the development of novel anti-tumor therapies. To test the potential use of the NCRs as anti-tumor agents, we generated soluble NCR-Ig fusion proteins in which the constant region of human IgG1 was fused to the extracellular portion of the receptor. We demonstrate, using two different human prostate cancer cell lines, that treatment with NKp30-Ig, dramatically inhibits tumor growth in vivo. Activated macrophages were shown to mediate an ADCC response against the NKp30-Ig coated prostate cell lines. Finally, the Ig fusion proteins were also demonstrated to discriminate between benign prostate hyperplasia and prostate cancer. This may provide a novel diagnostic modality in the difficult task of differentiating between these highly common pathological conditions.
Collapse
Affiliation(s)
- Tal I. Arnon
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Gal Markel
- Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Ahuva Bar-Ilan
- The Shraga Segal Department of Microbiology and Immunology and the National Institute for Biotechnology in the Negev, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Jacob Hanna
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Eyal Fima
- The Shraga Segal Department of Microbiology and Immunology and the National Institute for Biotechnology in the Negev, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Fabrice Benchetrit
- The Shraga Segal Department of Microbiology and Immunology and the National Institute for Biotechnology in the Negev, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Ruth Galili
- Lautenberg Center for General and Tumor Immunology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Adelheid Cerwenka
- Division of Innate Immunity, German Cancer Research Center, Heidelberg, Germany
| | | | | | - Angel Porgador
- The Shraga Segal Department of Microbiology and Immunology and the National Institute for Biotechnology in the Negev, Ben Gurion University of the Negev, Beer Sheva, Israel
- * E-mail: (AP); (OM)
| | - Ofer Mandelboim
- Lautenberg Center for General and Tumor Immunology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
- * E-mail: (AP); (OM)
| |
Collapse
|
121
|
Wang E, Selleri S, Sabatino M, Monaco A, Pos Z, Worschech A, Stroncek DF, Marincola FM. Spontaneous and treatment-induced cancer rejection in humans. Expert Opin Biol Ther 2008; 8:337-49. [PMID: 18294104 DOI: 10.1517/14712598.8.3.337] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Experimental observations suggest that human cancer cells actively interact with normal host cells and this cross-talk results, in most instances, in an increased potential of cancer cells to survive. On the other hand, it is also well documented that on rare occasions tumors can be dramatically destroyed by the host's immune response. OBJECTIVE In this review, we argue that understanding the mechanisms that bring about the immune response and lead to cancer destruction is of paramount importance for the design of future rational therapies. METHODS Here we summarize the present understanding of the phenomenology leading to cancer regression in humans and propose novel strategies for a more efficient study of human cancer under natural conditions and during therapy. CONCLUSION The understanding of tumor/host interactions within the tumor microenvironment is a key component of the study of tumor immunology in humans, much can be learned by a dynamic study of such interactions at time points related to the natural history of the disease or its response to therapy. Such understanding will eventually lead to novel and more effective therapies.
Collapse
Affiliation(s)
- Ena Wang
- National Institutes of Health, Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, Bethesda, Maryland, 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
122
|
Conaghan PJ, Ashraf SQ, Tytherleigh MG, Wilding JL, Tchilian E, Bicknell D, Mortensen NJM, Bodmer WF. Targeted killing of colorectal cancer cell lines by a humanised IgG1 monoclonal antibody that binds to membrane-bound carcinoembryonic antigen. Br J Cancer 2008; 98:1217-25. [PMID: 18349843 PMCID: PMC2359646 DOI: 10.1038/sj.bjc.6604289] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/21/2008] [Accepted: 01/31/2008] [Indexed: 12/12/2022] Open
Abstract
The distribution of carcinoembryonic antigen (CEA) in colorectal cancer (CRC) differs from that in normal colorectal tissue, being found on all borders of the cell membrane and hence enabling access to intravenous antibody, making CEA a good target for antibody-based therapy. The distinctive anti-CEA antibody, PR1A3, binds only membrane-bound CEA. Humanised PR1A3 (hPR1A3) was assessed both in vitro cytotoxicity and binding assays with colorectal cancer cell lines expressing varying levels of CEA. Human peripheral blood mononuclear cells (PBMCs) and purified natural killer (NK) cells were used as effectors. The in vitro assays demonstrated hPR1A3 CEA-specific binding and antibody-dependent and CEA-specific killing of human colorectal cancer cell lines by human PBMCs. The effect increased with increasing concentration of antibody and surface CEA, and was lost by using the parent murine IgG1 PR1A3. Killing was also blocked by antibody to the Fc-gammaIIIA receptor. Purified human NK cells were effective at much lower effector:target ratios than unfractionated PBMCs, indicating that NK cells were the main mediators of hPR1A3-based CEA-specific killing. The results support the development of hPR1A3 for therapy of colorectal cancer.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/pharmacology
- Antibody Specificity
- Antigen-Antibody Complex/metabolism
- Antigens, Neoplasm/metabolism
- Antigens, Surface/metabolism
- Carcinoembryonic Antigen/immunology
- Carcinoembryonic Antigen/metabolism
- Cell Line, Tumor
- Colorectal Neoplasms/therapy
- Cytotoxicity, Immunologic
- Drug Delivery Systems
- Humans
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin G/therapeutic use
- Killer Cells, Natural/immunology
- Receptors, IgG/immunology
Collapse
Affiliation(s)
- P J Conaghan
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford, UK
| | - S Q Ashraf
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford, UK
| | - M G Tytherleigh
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford, UK
| | - J L Wilding
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
| | - E Tchilian
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
| | - D Bicknell
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
| | - N JMcC Mortensen
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford, UK
| | - W F Bodmer
- Cancer Research UK, Cancer & Immunogenetics Laboratory, Weatherall Institute of Molecular Medicine, Headington, Oxford, UK
| |
Collapse
|
123
|
Simon AK, Newsom-Davis T, Frayne MEF, Ch'en PFT, McMichael AJ, Screaton GR. Generation of tumour-rejecting anti-carbohydrate monoclonal antibodies using melanoma modified with Fas ligand. Int Immunol 2008; 20:525-34. [PMID: 18310066 DOI: 10.1093/intimm/dxn011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Carbohydrate antigens such as glycolipids and glycoproteins are over-expressed in a variety of cancers and have therefore been identified as ideal candidates for tumour vaccines. Detection of anti-carbohydrate antibodies is associated with a good prognosis in cancer patients. However, generation of an efficient adaptive immune response has been hampered by the low immunogenicity of carbohydrates due to tolerance. Here, we describe a method by which tumour-rejecting antibodies directed against carbohydrates can be elicited in two different melanoma mouse models. Thus, using the murine melanoma B16F10 over-expressing Fas ligand (FasL), we have generated mAbs against cancer carbohydrate antigens expressed by the melanoma. Importantly, passive transfer of mAbs resulted in rejection of melanoma in vivo. Their protective effect in vivo was dependent on FcR and in vitro antibody-dependent cellular phagocytosis. They were also able to delay tumour growth when injected after the tumour was established. FasL-expressing tumours as an adjuvant are a novel way to generate anti-carbohydrate antibodies able to reject tumours in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/biosynthesis
- Antibody Specificity/immunology
- Antigens, Tumor-Associated, Carbohydrate/immunology
- Cell Line, Tumor
- Fas Ligand Protein/immunology
- Humans
- Injections, Intraperitoneal
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Melanoma, Experimental/immunology
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, IgG/deficiency
- Receptors, IgG/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- A Katharina Simon
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, UK.
| | | | | | | | | | | |
Collapse
|
124
|
|
125
|
Abstract
Monoclonal antibodies (mAb) are presently considered key therapeutic drugs for the treatment of malignancies. They can be designed to specifically target tumour-associated antigens and initiate several effector mechanisms, which potentially leads to elimination of the tumour. Through their Fc tail mAbs interact with Fc receptors (FcR) that are expressed on immune cells. Neutrophils are the most abundant circulating FcR-expressing white blood cells with potent cytotoxic ability that is enhanced in the presence of antitumour mAbs. They furthermore play a role in regulating adaptive immunity, which may lead to the initiation of antitumour immune responses. Yet, neutrophils receive surprisingly little attention as potential effector cell population. This article reviews the scientific data that supports the possibility of exploiting neutrophils for mAb-based immunotherapy of cancer. An increasing awareness and understanding of this topic may allow for future development of new anticancer therapies.
Collapse
Affiliation(s)
- Marjolein van Egmond
- VU University Medical Center, Departments of Surgical Oncology, and Molecular Cell Biology and Immunology, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| |
Collapse
|
126
|
Abstract
Receptors for immunoglobulins [Fc-receptors (FcRs)] are widely expressed throughout the immune system. By binding to the antibody Fc-portion, they provide a link between the specificity of the adaptive immune system and the powerful effector functions triggered by innate immune effector cells. By virtue of coexpression of activating and inhibitory FcRs on the same cell, they set a threshold for immune cell activation by immune complexes (ICs). Besides their involvement in the efferent phase of an immune response, they are also important for modulating adaptive immune responses by regulating B cell and dendritic cell (DC) activation. Deletion of the inhibitory FcR leads to the loss of tolerance in the humoral immune system and the development of autoimmune disease. Uptake of ICs by FcRs on DCs and the concommitant triggering of activating and inhibitory signaling pathways will determine the strength of the initiated T-cell response. Loss of this balanced signaling results in uncontrolled responses that can lead to the damage of healthy tissues and ultimately to the initiation of autoimmune processes. In this chapter, we will discuss how coexpression of different activating and inhibitory receptors on different immune cells of the innate and adaptive immune system modulates cell activity. Moreover, we will focus on exogenous factors that can influence the balanced triggering of activating and inhibitory FcRs, such as the cytokine milieu and the role of differential antibody glycosylation.
Collapse
|
127
|
Strome SE, Sausville EA, Mann D. A mechanistic perspective of monoclonal antibodies in cancer therapy beyond target-related effects. Oncologist 2007; 12:1084-95. [PMID: 17914078 DOI: 10.1634/theoncologist.12-9-1084] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Several monoclonal antibodies are now in clinical use for cancer therapy, and many others are currently undergoing clinical evaluation. These agents offer unique specificity against key molecular targets on tumor cells or in the tumor microenvironment. The clinical efficacy of monoclonal antibodies is generally attributed to target-specific mechanisms resulting from neutralizing or inhibiting a growth factor or receptor that drives cell proliferation and tumor growth. Several targets, including CD20, human epidermal growth factor receptor 2, vascular endothelial growth factor, and epidermal growth factor receptor, have been validated in specific malignancies on the basis of monoclonal antibody efficacy. However, monoclonal antibodies also have the potential to activate immune-mediated effector functions, including antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. These functions result from interactions involving the Fc domain of the antibody, and, consequently, may vary by antibody, isotype, and Fc modification, such as changes in glycosylation. Accordingly, all monoclonal antibodies directed against a given target should not be considered equivalent in their ability to stimulate immune-mediated effector functions.
Collapse
Affiliation(s)
- Scott E Strome
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201-1619, USA.
| | | | | |
Collapse
|
128
|
Stavenhagen JB, Gorlatov S, Tuaillon N, Rankin CT, Li H, Burke S, Huang L, Vijh S, Johnson S, Bonvini E, Koenig S. Fc optimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcgamma receptors. Cancer Res 2007; 67:8882-90. [PMID: 17875730 DOI: 10.1158/0008-5472.can-07-0696] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies (mAb) are widely used in the treatment of non-Hodgkin's lymphoma and autoimmune diseases. Although the mechanism of action in vivo is not always known, the therapeutic activity of several approved mAbs depends on the binding of the Fcgamma regions to low-affinity Fcgamma receptors (FcgammaR) expressed on effector cells. We did functional genetic screens to identify IgG1 Fc domains with improved binding to the low-affinity activating Fc receptor CD16A (FcgammaRIIIA) and reduced binding to the low-affinity inhibitory Fc receptor, CD32B (FcgammaRIIB). Identification of new amino acid residues important for FcgammaR binding guided the construction of an Fc domain that showed a dramatically enhanced CD16A binding and greater than a 100-fold improvement in antibody-dependent cell-mediated cytotoxicity. In a xenograft murine model of B-cell malignancy, the greatest enhancement of an Fc-optimized anti-human B-cell mAb was accounted for by improved binding to FcgammaRIV, a unique mouse activating FcgammaR that is expressed by monocytes and macrophages but not natural killer (NK) cells, consistent with experimental and clinical data suggesting that mononuclear phagocytes, effector cells expressing both activating and inhibitory FcgammaR, are critical mediators of B-cell depletion in vivo. By using mice transgenic for human CD16A, enhanced survival was observed due to expression of CD16A-158(phe) on monocytes and macrophages as well as on NK cells in these mice. The design of new generations of improved antibodies for immunotherapy should aim at Fc optimization to increase the engagement of activating FcgammaR present on the surface of tumor-infiltrating effector cell populations.
Collapse
|
129
|
Optimizing engagement of the immune system by anti-tumor antibodies: an engineer's perspective. Drug Discov Today 2007; 12:898-910. [DOI: 10.1016/j.drudis.2007.08.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 08/03/2007] [Accepted: 08/21/2007] [Indexed: 11/23/2022]
|
130
|
Abstract
It is now commonly recognized that tumor-targeting monoclonal antibodies (mAb) demonstrate significant therapeutic effects in cancer patients. Multiple effector mechanisms are involved in the primary response; however, the induction of tumor-specific immunity has sometimes been a desired outcome and in some cases has contributed to the success of mAb-based therapy. Initiating the antitumor immune response by creating a tumor antigen supply from dying (apoptotic) tumor cells is a goal of tumor-targeting mAb therapy, and ideally a sufficiently immunogenic tumor cell death might provide a platform on which combination immunotherapies can be based. In this review, the authors discuss the possible utility and pitfalls of using such a therapy, which combines tumor cell death with immune activation therapy. The ideas are largely based on the observation that three-mAb (anti-DR5 mAb, anti-CD40 mAb, and anti-CD137 mAb [trimAb]) therapy has been shown to be very effective in a number of mouse experimental tumor models. The authors believe that this rational activation and inhibition of key points in the immune response can be an effective strategy to apply in human cancer patients.
Collapse
Affiliation(s)
- Kazuyoshi Takeda
- Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan.
| | | | | |
Collapse
|
131
|
Bellone S, Frera G, Landolfi G, Romani C, Bandiera E, Tognon G, Roman JJ, Burnett AF, Pecorelli S, Santin AD. Overexpression of epidermal growth factor type-1 receptor (EGF-R1) in cervical cancer: Implications for Cetuximab-mediated therapy in recurrent/metastatic disease. Gynecol Oncol 2007; 106:513-20. [PMID: 17540437 DOI: 10.1016/j.ygyno.2007.04.028] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 04/10/2007] [Accepted: 04/23/2007] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To evaluate and compare epidermal growth factor type-1 receptor (EGF-R1) expression in short term and established cervical cancer cell lines generated from primary and metastatic/recurrent sites of disease. To evaluate the sensitivity of cervical cancer cell lines to treatment with a chimeric MAb against EGFR-1 (Cetuximab). METHODS EGFR-1 expression was evaluated by flow cytometry on 22 cervical cancer cell lines including 14 primary cervical cancer cell lines obtained from cervical biopsies (11 patients) and recurrent sites of disease (three patients) as well as eight established cell lines. Tumor cell lines were tested for sensitivity to Cetuximab-mediated complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) in 51Cr release assays. Finally, Cetuximab-mediated inhibition of cell proliferation was also tested. RESULTS Fourteen out of fourteen (100%) primary tumors and seven out of eight (87.5%) established cervical cancer cell lines expressed EGFR-1 by flow cytometry. Cell lines from recurrent/metastatic sites of disease expressed higher levels of EGFR-1 when compared to those obtained from primary sites (p>0.05). Minimal CDC was detected in the majority of cervical cancer cell lines exposed to complement+/-Cetuximab in the absence of peripheral blood lymphocytes (PBL). In contrast, cervical tumor cell lines were found highly sensitive to Cetuximab-mediated ADCC when challenged with PBL from either healthy donors or cervical cancer patients. Importantly, ADCC was further increased in the presence of complement. Finally, tumor proliferation was significantly inhibited by Cetuximab in all cervical tumors tested. CONCLUSIONS EGFR-1 is highly expressed in primary and recurrent cervical tumors. Cetuximab might be a novel and attractive therapeutic strategy in patients harboring chemotherapy-resistant, recurrent, or metastatic cervical cancer.
Collapse
Affiliation(s)
- Stefania Bellone
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Takeda K, Stagg J, Yagita H, Okumura K, Smyth MJ. Targeting death-inducing receptors in cancer therapy. Oncogene 2007; 26:3745-57. [PMID: 17530027 DOI: 10.1038/sj.onc.1210374] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Deregulated cell death pathways may lead to the development of cancer, and induction of tumor cell apoptosis is the basis of many cancer therapies. Knowledge accumulated concerning the molecular mechanisms of apoptotic cell death has aided the development of new therapeutic strategies to treat cancer. Signals through death receptors of the tumor necrosis factor (TNF) superfamily have been well elucidated, and death receptors are now one of the most attractive therapeutic targets in cancer. In particular, DR5 and DR4, death receptors of TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), are interesting targets of antibody-based therapy, since TRAIL may also bind decoy receptors that may prevent TRAIL-mediated apoptosis, whereas TRAIL ligand itself selectively induces apoptosis in cancer cells. Here, we review the potential therapeutic utility of agonistic antibodies against DR5 and DR4 and discuss the possible extension of this single-antibody-based strategy when combined with additional modalities that either synergizes to cause enhanced apoptosis or further engage the cellular immune response. Rational design of antibody-based therapies combining the induction of tumor cell apoptosis and activation of tumor-specific adaptive immunity enables promotion of distinct steps of the antitumor immune response, thereby enhancing tumor-specific lymphocytes that can eradicate TRAIL/DR5-resistant mutating, large established and heterogeneous tumors in a manner that does not require the definition of individual tumor-specific antigens.
Collapse
Affiliation(s)
- K Takeda
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
133
|
Abdel-Motal UM, Guay HM, Wigglesworth K, Welsh RM, Galili U. Immunogenicity of influenza virus vaccine is increased by anti-gal-mediated targeting to antigen-presenting cells. J Virol 2007; 81:9131-41. [PMID: 17609270 PMCID: PMC1951452 DOI: 10.1128/jvi.00647-07] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
This study describes a method for increasing the immunogenicity of influenza virus vaccines by exploiting the natural anti-Gal antibody to effectively target vaccines to antigen-presenting cells (APC). This method is based on enzymatic engineering of carbohydrate chains on virus envelope hemagglutinin to carry the alpha-Gal epitope (Gal alpha 1-3Gal beta 1-4GlcNAc-R). This epitope interacts with anti-Gal, the most abundant antibody in humans (1% of immunoglobulins). Influenza virus vaccine expressing alpha-Gal epitopes is opsonized in situ by anti-Gal immunoglobulin G. The Fc portion of opsonizing anti-Gal interacts with Fc gamma receptors on APC and induces effective uptake of the vaccine virus by APC. APC internalizes the opsonized virus to transport it to draining lymph nodes for stimulation of influenza virus-specific T cells, thereby eliciting a protective immune response. The efficacy of such an influenza vaccine was demonstrated in alpha 1,3galactosyltransferase (alpha 1,3GT) knockout mice, which produce anti-Gal, using the influenza virus strain A/Puerto Rico/8/34-H1N1 (PR8). Synthesis of alpha-Gal epitopes on carbohydrate chains of PR8 virus (PR8(alpha gal)) was catalyzed by recombinant alpha1,3GT, the glycosylation enzyme that synthesizes alpha-Gal epitopes in cells of nonprimate mammals. Mice immunized with PR8(alpha gal) displayed much higher numbers of PR8-specific CD8(+) and CD4(+) T cells (determined by intracellular cytokine staining and enzyme-linked immunospot assay) and produced anti-PR8 antibodies with much higher titers than mice immunized with PR8 lacking alpha-Gal epitopes. Mice immunized with PR8(alpha gal) also displayed a much higher level of protection than PR8 immunized mice after being challenged with lethal doses of live PR8 virus. We suggest that a similar method for increasing immunogenicity may be applicable to avian influenza vaccines.
Collapse
Affiliation(s)
- Ussama M Abdel-Motal
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, LRB, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
134
|
Durso RJ, Andjelic S, Gardner JP, Margitich DJ, Donovan GP, Arrigale RR, Wang X, Maughan MF, Talarico TL, Olmsted RA, Heston WDW, Maddon PJ, Olson WC. A Novel Alphavirus Vaccine Encoding Prostate-Specific Membrane Antigen Elicits Potent Cellular and Humoral Immune Responses. Clin Cancer Res 2007; 13:3999-4008. [PMID: 17606734 DOI: 10.1158/1078-0432.ccr-06-2202] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is an attractive target for active immunotherapy. Alphavirus vaccines have shown promise in eliciting immunity to tumor antigens. This study investigated the immunogenicity of alphavirus vaccine replicon particles (VRP) that encode PSMA (PSMA-VRP). EXPERIMENTAL DESIGN Cells were infected with PSMA-VRP and evaluated for PSMA expression and folate hydrolase activity. Mice were immunized s.c. with PSMA-VRP or purified PSMA protein. Sera, splenocytes, and purified T cells were evaluated for the magnitude, durability, and epitope specificity of the anti-PSMA response. Antibodies were measured by flow cytometry, and cellular responses were measured by IFN-gamma enzyme-linked immunospot and chromium release assays. Cellular responses in BALB/c and C57BL/6 mice were mapped using overlapping 15-mer PSMA peptides. A Good Laboratory Practice-compliant toxicology study was conducted in rabbits. RESULTS PSMA-VRP directed high-level expression of active PSMA. Robust T-cell and B-cell responses were elicited by a single injection of 2 x 10(5) infectious units, and responses were boosted following repeat immunizations. Anti-PSMA responses were detected following three immunizations with 10(2) infectious units and increased with increasing dose. PSMA-VRP was more immunogenic than adjuvanted PSMA protein. Responses to PSMA-VRP were characterized by Th-1 cytokines, potent CTL activity, and IgG2a/IgG2b antibodies. T-cell responses in BALB/c and C57BL/6 mice were directed toward different PSMA peptides. Immunogenic doses of PSMA-VRP were well tolerated in mice and rabbits. CONCLUSIONS PSMA-VRP elicited potent cellular and humoral immunity in mice, and specific anti-PSMA responses were boosted on repeat dosing. PSMA-VRP represents a promising approach for immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Robert J Durso
- Progenics Pharmaceuticals, Inc., Tarrytown, New York 10591, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Bracher M, Gould HJ, Sutton BJ, Dombrowicz D, Karagiannis SN. Three-colour flow cytometric method to measure antibody-dependent tumour cell killing by cytotoxicity and phagocytosis. J Immunol Methods 2007; 323:160-71. [PMID: 17531261 DOI: 10.1016/j.jim.2007.04.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/02/2007] [Accepted: 04/17/2007] [Indexed: 12/01/2022]
Abstract
We have developed a three-colour flow cytometric method to assay the contributions of cytotoxicity and phagocytosis to antibody-dependent cell-mediated tumour cell killing. In this assay, tumour target cells are pre-labelled with CFSE, and mixed with effector cells and a tumour antigen-specific monoclonal antibody. After incubation of the cells with the antibody, effector cells are labelled with PE and dead cells with PI. Using flow cytometry, dead and phagocytosed tumour cells can be quickly and easily counted and the numbers summed to determine the total number of killed cells. One can thereby measure the phagocytic aspect of antibody-dependent cell-mediated tumour cell killing, otherwise only revealed by microscopic examination. The failure to detect phagocytosed, in addition to live and dead target cells, by standard assays may result in an underestimation of tumour cell killing and hence the potential of an antibody for immunotherapy of cancer. We illustrate the new method by analysing human monocyte-mediated cytotoxic and phagocytic cell killing of IGROV1 ovarian tumour cells by the ovarian tumour antigen-specific anti-folate binding protein monoclonal antibody, MOv18 IgE.
Collapse
Affiliation(s)
- Marguerite Bracher
- Randall Division of Cell and Molecular Biophysics, Kings College London, New Hunts House, London, United Kingdom
| | | | | | | | | |
Collapse
|
136
|
Bueno SM, González PA, Schwebach JR, Kalergis AM. T cell immunity evasion by virulent Salmonella enterica. Immunol Lett 2007; 111:14-20. [PMID: 17583359 DOI: 10.1016/j.imlet.2007.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/13/2007] [Accepted: 05/11/2007] [Indexed: 12/30/2022]
Abstract
Salmonella enterica are Gram-negative bacteria that cause systemic disease in their specific hosts. One of the recently appreciated features of Salmonella pathogenicity is the capacity of the bacteria to impair host adaptive immunity by interfering with DC function and T cell activation. It is likely that this feature of virulent Salmonella is needed to promote systemic dissemination in the host. Recent studies have suggested explanations for some of the molecular mechanisms developed by virulent Salmonella to impair DC and T cell function. Several of these mechanisms require the expression of virulence genes encoded within Salmonella pathogenicity islands. Targeted deletion of these genes diminishes Salmonella pathogenicity and leads to efficient activation of T cells by Salmonella-infected DCs. In this review, recent data that support the subversion of DC function by Salmonella as a means to evade host adaptive immunity and cause systemic infection are discussed. These new findings suggest a new pathogenesis model with DCs as key targets for Salmonella virulence factors.
Collapse
Affiliation(s)
- Susan M Bueno
- Millennium Nucleus on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | | | | | | |
Collapse
|
137
|
Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O'Neill D, Pavlick A, Escalon JB, Cruz CM, Angiulli A, Angiulli F, Mears G, Vogel SM, Pan L, Jungbluth AA, Hoffmann EW, Venhaus R, Ritter G, Old LJ, Ayyoub M. Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci U S A 2007; 104:8947-52. [PMID: 17517626 PMCID: PMC1885608 DOI: 10.1073/pnas.0703395104] [Citation(s) in RCA: 241] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Indexed: 01/05/2023] Open
Abstract
The use of recombinant tumor antigen proteins is a realistic approach for the development of generic cancer vaccines, but the potential of this type of vaccines to induce specific CD8(+) T cell responses, through in vivo cross-priming, has remained unclear. In this article, we report that repeated vaccination of cancer patients with recombinant NY-ESO-1 protein, Montanide ISA-51, and CpG ODN 7909, a potent stimulator of B cells and T helper type 1 (Th1)-type immunity, resulted in the early induction of specific integrated CD4(+) Th cells and antibody responses in most vaccinated patients, followed by the development of later CD8(+) T cell responses in a fraction of them. The correlation between antibody and T cell responses, together with the ability of vaccine-induced antibodies to promote in vitro cross-presentation of NY-ESO-1 by dendritic cells to vaccine-induced CD8(+) T cells, indicated that elicitation of NY-ESO-1-specific CD8(+) T cell responses by cross-priming in vivo was associated with the induction of adequate levels of specific antibodies. Together, our data provide clear evidence of in vivo cross-priming of specific cytotoxic T lymphocytes by a recombinant tumor antigen vaccine, underline the importance of specific antibody induction for the cross-priming to occur, and support the use of this type of formulation for the further development of efficient cancer vaccines.
Collapse
Affiliation(s)
- Danila Valmori
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| | | | - Valeria Tosello
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| | - Nina Bhardwaj
- New York University School of Medicine, New York, NY 10016
| | - Sylvia Adams
- New York University School of Medicine, New York, NY 10016
| | - David O'Neill
- New York University School of Medicine, New York, NY 10016
| | - Anna Pavlick
- New York University School of Medicine, New York, NY 10016
| | | | | | | | | | - Gregory Mears
- Division of Medical Oncology, Columbia University Medical Center, New York, NY 10032; and
| | - Susan M. Vogel
- Division of Medical Oncology, Columbia University Medical Center, New York, NY 10032; and
| | - Linda Pan
- Ludwig Institute for Cancer Research, New York, NY 10158
| | | | | | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Gerd Ritter
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Lloyd J. Old
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Maha Ayyoub
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| |
Collapse
|
138
|
Nimmerjahn F, Ravetch JV. Antibodies, Fc receptors and cancer. Curr Opin Immunol 2007; 19:239-45. [PMID: 17291742 DOI: 10.1016/j.coi.2007.01.005] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 01/31/2007] [Indexed: 11/19/2022]
Abstract
Since the first report of the successful use of a monoclonal antibody for the treatment of human B cell lymphoma in 1982, several antibodies have become incorporated into standard treatment protocols for cancer. One of the most important factors that determine antibody activity in vivo is the efficient interaction with cellular Fc-receptors on innate immune effector cells. It has become clear that the outcome of this interaction is influenced by several factors, such as the antibody isotype-specific affinity to activating and inhibitory receptors, the level of inhibitory FcgammaRIIB expression, and the composition of the sugar side chain attached to the antibody Fc-portion. These novel insights into antibody FcR interactions might be useful to produce the next generation of improved immunotherapeutic molecules.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | |
Collapse
|
139
|
Sibéril S, Dutertre CA, Fridman WH, Teillaud JL. FcgammaR: The key to optimize therapeutic antibodies? Crit Rev Oncol Hematol 2007; 62:26-33. [PMID: 17240158 DOI: 10.1016/j.critrevonc.2006.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 12/05/2006] [Accepted: 12/08/2006] [Indexed: 12/15/2022] Open
Abstract
The binding of IgG to receptors for the Fc region of IgG (FcgammaR) is a critical step for the initiation and the control of effector immune functions. Activating FcgammaR induce antibody-dependent cell cytotoxicity (ADCC), endocytosis of immune complexes followed by antigen presentation, phagocytosis, and release of cytokines or pro-inflammatory mediators. By contrast, inhibitory FcgammaR regulate immune responses by inhibiting the activation of B lymphocytes, monocytes, mast cells and basophils, induced through activating receptors. Studies with FcgammaR-deficient mice support the critical role of the different FcgammaR in the in vivo functional effects of therapeutic monoclonal antibodies. Structural studies have provided detailed insights in the molecular mechanisms that govern IgG/FcgammaR interactions. The importance of the sugar components linked to asparagine 297 in the function of IgG has been also highlighted. These data have led to the engineering of a new generation of monoclonal antibodies for therapeutic use with optimized effector functions.
Collapse
Affiliation(s)
- Sophie Sibéril
- Unité INSERM 255, IFR58, Université René Descartes-Paris 5, Université Pierre et Marie Curie-Paris 6, Centre de Recherches Biomédicales des Cordeliers, Paris, France
| | | | | | | |
Collapse
|
140
|
Seledtsov VI, Niza NA, Felde MA, Shishkov AA, Samarin DM, Seledtsova GV, Seledtsov DV. Xenovaccinotherapy for colorectal cancer. Biomed Pharmacother 2007; 61:125-30. [PMID: 17258887 DOI: 10.1016/j.biopha.2006.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Accepted: 09/27/2006] [Indexed: 01/08/2023] Open
Abstract
The objectives of this phase I-II trial were to assess the toxicity, immunological and clinical responses induced in 37 patients with stage IV colorectal cancer by the subcutaneous administration of a xenogenic polyantigenic vaccine (XPV) prepared from disrupted murine melanoma (B16) and carcinoma (LLC) cells. An inducing course of vaccinotherapy consisted of 10 immunizations (5 at weekly and 5 at fortnight intervals). Twenty-four hours later each of first 5 vaccinations the patient was subcutaneously given a low dose of the recombinant interleukin-2 (IL-2). A consolidating course of vaccinotherapy consisted of monthly vaccinations. No grade III or IV toxicities, but also laboratory and clinical signs of developing systemic autoimmune disorders were noted in any XPV-treated patient. A significant increase in cell-mediated immunoreactivity to both LLC and B16 antigens (Ags) occurred in the patients after inducing vaccinations, as determined by delayed-type hypersensitivity (DTH) skin reactions, as well as by blood lymphocyte proliferation responses. Vaccinations also led to increased cell-mediated reactivity to murine non-tumor, spleen cell (SC)-associated Ags. This reactivity, however, was not as significant as that to tumor-associated antigens (TAAs). XPV was also found to capable of generating IgG antibody-mediated responses. With immunotherapy concentrations of both interferon-gamma (IFN-gamma) and interleukin-4 (IL-4) detectably elevated in patients' sera, suggesting intensification of T helper 1-/T helper 2-mediated responses in the XPV-treated patients. The average survival of the XPV-treated patients was noticeably superior than was that of the clinically comparable control patients (17 vs 7 months). Collectively the results suggest that xenogenic TAAs are safe to use, able to induce measurable cellular and humoral immune responses, and may be clinically effective in certain colorectal cancer patients.
Collapse
Affiliation(s)
- Victor I Seledtsov
- Department of Cell Biotechnology, Institute of Clinical Immunology, Russian Academy of Medical Science, 14 Yadrintsevskaya Street, 630099 Novosibirsk, Russia.
| | | | | | | | | | | | | |
Collapse
|
141
|
Lowe DB, Shearer MH, Jumper CA, Bright RK, Kennedy RC. Fc gamma receptors play a dominant role in protective tumor immunity against a virus-encoded tumor-specific antigen in a murine model of experimental pulmonary metastases. J Virol 2006; 81:1313-8. [PMID: 17108042 PMCID: PMC1797535 DOI: 10.1128/jvi.01943-06] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian virus 40 (SV40) large tumor antigen (Tag) represents a virus-encoded tumor-specific antigen expressed in many types of human cancers and a potential immunologic target for antitumor responses. Fc receptors are important mediators in the regulation and execution of host effector mechanisms against conditions including infectious diseases, autoimmunity, and cancer. By examining tumor protection in SV40 Tag-immunized wild-type BALB/c mice using an experimental pulmonary metastasis model, we attempted to address whether engagement of the immunoglobulin G Fc receptors (FcgammaRs) on effector cells is necessary to mediate antitumor responses. All immunized BALB/c FcgammaR-/- knockout mice developed anti-SV40 Tag antibody responses prior to experimental challenge with a tumorigenic cell line expressing SV40 Tag. However, all mice deficient in the activating FcgammaRI (CD64) and FcgammaRIII (CD16) were unable to mount protective immunologic responses against tumor challenge and developed tumor lung foci. In contrast, mice lacking the inhibitory receptor FcgammaRII (CD32) demonstrated resistance to tumorigenesis. These results underscore the importance of effector cell populations expressing FcgammaRI/III within this murine tumor model system, and along with the production of a specific humoral immune response, antibody-dependent cell-mediated cytotoxicity (ADCC) may be a functioning mechanism of tumor clearance. Additionally, these data demonstrate the potential utility of ADCC as a viable approach for targeting vaccination strategies that promote FcgammaRI/III scavenging pathways against cancer.
Collapse
Affiliation(s)
- Devin B Lowe
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430, USA
| | | | | | | | | |
Collapse
|
142
|
Cassard L, Cohen-Solal J, Camilleri-Broët S, Fournier E, Fridman WH, Sautès-Fridman C. Fc gamma receptors and cancer. ACTA ACUST UNITED AC 2006; 28:321-8. [PMID: 17096153 DOI: 10.1007/s00281-006-0058-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 10/18/2006] [Indexed: 12/14/2022]
Abstract
FcgammaRs are a family of heterogeneous molecules that play opposite roles in immune response and control the effector functions of IgG antibodies. In many cancers, IgG antibodies are produced that recognize cancer cells, form immune complexes and therefore, activate FcgammaR. The therapeutic efficacy of monoclonal IgG antibodies against hematopoietic and epithelial tumors also argue for an important role of IgG antibodies in anti-tumor defenses. Since the 1980s, a series of lines of evidence in experimental models and in humans strongly suggest that FcgammaR are involved in the therapeutic activity of monoclonal IgG antibodies by activating the cytotoxic activity of FcgammaR-positive cells such as NK cells, monocytes, macrophages and neutrophils and by increasing antigen presentation by dendritic cells. Since many cell types co-express activating and inhibitory FcgammaR, the FcgammaR-dependent effector functions of IgG anti-tumor antibodies are counterbalanced by the inhibitory FcgammaRIIB. In addition, some tumor cells express FcgammaR either constitutively, such as B cell lymphomas or ectopically, such as 40% of human metastatic melanoma. The tumor FcgammaR isoform is preferentially FcgammaRIIB, which is functional at least in human metastatic melanoma. This review summarizes these data and discusses how FcgammaRIIB expression may influence the anti-tumor immune reaction and how beneficial or deleterious this expression could be for the efficiency of therapeutics based on monoclonal anti-tumor antibodies.
Collapse
Affiliation(s)
- Lydie Cassard
- INSERM UMRs255, Université Paris 5 René Descartes, Université Paris 6 Pierre et Marie Curie, Centre de Recherches des Cordeliers, 15 rue de L’Ecole de Médecine, 75270, Paris Cedex 06, France,
| | | | | | | | | | | |
Collapse
|
143
|
Rankin CT, Veri MC, Gorlatov S, Tuaillon N, Burke S, Huang L, Inzunza HD, Li H, Thomas S, Johnson S, Stavenhagen J, Koenig S, Bonvini E. CD32B, the human inhibitory Fc-γ receptor IIB, as a target for monoclonal antibody therapy of B-cell lymphoma. Blood 2006; 108:2384-91. [PMID: 16757681 DOI: 10.1182/blood-2006-05-020602] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Abstract
Human CD32B (FcγRIIB), the low-affinity inhibitory receptor for IgG, is the predominant Fc receptor (FcR) present on B cells. Immunohistochemical and expression studies have identified CD32B expression in a variety of B-cell malignancies, suggesting that CD32B is a potential immunotherapeutic target for B-cell malignancies. A high-affinity monoclonal antibody (mAb 2B6), from a novel panel of anti–human CD32B–specific mAbs, was chimerized (ch2B6) and humanized (hu2B6-3.5). Both ch2B6 and hu2B6-3.5 were capable of directing cytotoxicity by peripheral blood mononuclear cells and monocyte-derived macrophages against B-lymphoma lines in vitro. In a human B-cell lymphoma mouse xenograft model, administration of ch2B6 or hu2B6-3.5 reduced tumor growth rate and improved tumor-free survival. Both the in vitro and in vivo activities of 2B6 required an intact Fc, suggesting an FcR-mediated mechanism of action. These data support the hypothesis that CD32B is a viable target for mAb treatment of B-cell lymphoproliferative disorders.
Collapse
|
144
|
Wittman VP, Woodburn D, Nguyen T, Neethling FA, Wright S, Weidanz JA. Antibody Targeting to a Class I MHC-Peptide Epitope Promotes Tumor Cell Death. THE JOURNAL OF IMMUNOLOGY 2006; 177:4187-95. [PMID: 16951384 DOI: 10.4049/jimmunol.177.6.4187] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Therapeutic mAbs that target tumor-associated Ags on the surface of malignant cells have proven to be an effective and specific option for the treatment of certain cancers. However, many of these protein markers of carcinogenesis are not expressed on the cells' surface. Instead these tumor-associated Ags are processed into peptides that are presented at the cell surface, in the context of MHC class I molecules, where they become targets for T cells. To tap this vast source of tumor Ags, we generated a murine IgG2a mAb, 3.2G1, endowed with TCR-like binding specificity for peptide-HLA-A*0201 (HLA-A2) complex and designated this class of Ab as TCR mimics (TCRm). The 3.2G1 TCRm recognizes the GVL peptide (GVLPALPQV) from human chorionic gonadotropin beta presented by the peptide-HLA-A*0201 complex. When used in immunofluorescent staining reactions using GVL peptide-loaded T2 cells, the 3.2G1 TCRm specifically stained the cells in a peptide and Ab concentration-dependent manner. Staining intensity correlated with the extent of cell lysis by complement-dependent cytotoxicity (CDC), and a peptide concentration-dependent threshold level existed for the CDC reaction. Staining of human tumor lines demonstrated that 3.2G1 TCRm was able to recognize endogenously processed peptide and that the breast cancer cell line MDA-MB-231 highly expressed the target epitope. The 3.2G1 TCRm-mediated CDC and Ab-dependent cellular cytotoxicity of a human breast carcinoma line in vitro and inhibited in vivo tumor implantation and growth in nude mice. These results provide validation for the development of novel TCRm therapeutic reagents that specifically target and kill tumors via recognition and binding to MHC-peptide epitopes.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Antibody Specificity
- Apoptosis/immunology
- Binding Sites, Antibody
- Cell Line, Tumor
- Epitopes/immunology
- Epitopes/metabolism
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Mimicry/immunology
- Neoplasm Transplantation
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Vaughan P Wittman
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | | | | | | | | |
Collapse
|
145
|
Abdel-Motal U, Wang S, Lu S, Wigglesworth K, Galili U. Increased immunogenicity of human immunodeficiency virus gp120 engineered to express Galalpha1-3Galbeta1-4GlcNAc-R epitopes. J Virol 2006; 80:6943-51. [PMID: 16809300 PMCID: PMC1489031 DOI: 10.1128/jvi.00310-06] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The glycan shield comprised of multiple carbohydrate chains on the human immunodeficiency virus (HIV) envelope glycoprotein gp120 helps the virus to evade neutralizing antibodies. The present study describes a novel method for increasing immunogenicity of gp120 vaccine by enzymatic replacement of sialic acid on these carbohydrate chains with Galalpha1-3Galbeta1-4GlcNAc-R (alpha-gal) epitopes. These epitopes are ligands for the natural anti-Gal antibody constituting approximately 1% of immunoglobulin G in humans. We hypothesize that vaccination with gp120 expressing alpha-gal epitopes (gp120(alphagal)) results in in vivo formation of immune complexes with anti-Gal, which targets vaccines for effective uptake by antigen-presenting cells (APC), due to interaction between the Fc portion of the antibody and Fcgamma receptors on APC. This in turn results in effective transport of the vaccine to lymph nodes and effective processing and presentation of gp120 immunogenic peptides by APC for eliciting a strong anti-gp120 immune response. This hypothesis was tested in alpha-1,3-galactosyltransferase knockout mice, which produce anti-Gal. Mice immunized with gp120(alphagal) produced anti-gp120 antibodies in titers that were >100-fold higher than those measured in mice immunized with comparable amounts of gp120 and effectively neutralized HIV. T-cell response, measured by ELISPOT, was much higher in mice immunized with gp120(alphagal) than in mice immunized with gp120. It is suggested that gp120(alphagal) can serve as a platform for anti-Gal-mediated targeting of additional vaccinating HIV proteins fused to gp120(alphagal), thereby creating effective prophylactic vaccines.
Collapse
Affiliation(s)
- Ussama Abdel-Motal
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, LRB, Worcester, 01605, USA
| | | | | | | | | |
Collapse
|
146
|
Clynes R. Antitumor Antibodies in the Treatment of Cancer: Fc Receptors Link Opsonic Antibody with Cellular Immunity. Hematol Oncol Clin North Am 2006; 20:585-612. [PMID: 16762726 DOI: 10.1016/j.hoc.2006.02.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Engineered antibody therapeutics have provided new treatment options in cancer. Genetic evidence in man and in the mouse suggests that Fc receptor (FcR) engagement contributes mechanistically to the therapeutic activity of naked antibodies. Preferential activation of activating FcRs and limited engagement of inhibitory FcRs enhance tumor responses in mouse models. Thus, engineered Fc domains with favorable affinities for specific FcR types may prove to be clinically superior.
Collapse
Affiliation(s)
- Raphael Clynes
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
147
|
Liao JCF, Gregor P, Wolchok JD, Orlandi F, Craft D, Leung C, Houghton AN, Bergman PJ. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. CANCER IMMUNITY 2006; 6:8. [PMID: 16626110 PMCID: PMC1976276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 03/09/2006] [Indexed: 05/08/2023]
Abstract
Antitumor immune responses can be elicited in preclinical mouse melanoma models using plasmid DNA vaccines encoding xenogeneic melanosomal differentiation antigens. We previously reported on a phase I clinical trial of human tyrosinase (huTyr) DNA vaccination of 9 dogs with advanced malignant melanoma (World Health Organization stages II-IV), in which we demonstrated the safety of the treatment and the prolongation of the expected survival time (ST) of subjects as compared to historical, stage-matched controls. As a secondary goal of the same study, we report here on the induction of tyrosinase-specific antibody responses in three of the nine dogs vaccinated with huTyr DNA. The antibodies in two of the three responders cross-react with syngeneic canine tyrosinase, demonstrating the ability of this vaccine to overcome host immune tolerance and/or ignorance to or of "self" antigens. Most interestingly, the onset of antibody induction in these three dogs coincides with observed clinical responses and may suggest a means to account for their long-term tumor control and survival.
Collapse
Affiliation(s)
- Jack C. F. Liao
- Flaherty Comparative Oncology Laboratory, Donaldson-Atwood Cancer Clinic, The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA
| | - Polly Gregor
- Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Jedd D. Wolchok
- Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Francesca Orlandi
- Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Diane Craft
- Flaherty Comparative Oncology Laboratory, Donaldson-Atwood Cancer Clinic, The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA
| | - Carrie Leung
- Flaherty Comparative Oncology Laboratory, Donaldson-Atwood Cancer Clinic, The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA
| | - Alan N. Houghton
- Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Philip J. Bergman
- Flaherty Comparative Oncology Laboratory, Donaldson-Atwood Cancer Clinic, The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA
| |
Collapse
|
148
|
Zhang M, Yao Z, Zhang Z, Garmestani K, Goldman CK, Ravetch JV, Janik J, Brechbiel MW, Waldmann TA. Effective therapy for a murine model of human anaplastic large-cell lymphoma with the anti-CD30 monoclonal antibody, HeFi-1, does not require activating Fc receptors. Blood 2006; 108:705-10. [PMID: 16551968 PMCID: PMC1895489 DOI: 10.1182/blood-2005-11-4607] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CD30 is a member of the tumor necrosis factor receptor family. Overexpression of CD30 on some neoplasms versus its limited expression on normal tissues makes this receptor a promising target for antibody-based therapy. Anaplastic large-cell lymphoma (ALCL) represents a heterogeneous group of aggressive non-Hodgkin lymphomas characterized by the strong expression of CD30. We investigated the therapeutic efficacy of HeFi-1, a mouse IgG1 monoclonal antibody, which recognizes the ligand-binding site on CD30, and humanized anti-Tac antibody (daclizumab), which recognizes CD25, in a murine model of human ALCL. The ALCL model was established by intravenous injection of karpas299 cells into nonobese diabetic/severe combined immuno-deficient (SCID/NOD) wild-type or SCID/NOD Fc receptor common gamma chain-deficient (FcRgamma(-/-)) mice. HeFi-1, given at a dose of 100 microg weekly for 4 weeks, significantly prolonged survival of the ALCL-bearing SCID/NOD wild-type and SCID/NOD FcRgamma(-/-) mice (P < .01) as compared with the control groups. In vitro studies showed that HeFi-1 inhibited the proliferation of karpas299 cells, whereas daclizumab did not inhibit cell proliferation. We demonstrated that the expression of FcRgamma on polymorphonuclear leukocytes and monocytes was not required for HeFi-1-mediated tumor growth inhibition in vivo, although it was required for daclizumab.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antigens, Neoplasm/immunology
- Cell Proliferation/drug effects
- Daclizumab
- Disease Models, Animal
- Humans
- Immunoglobulin G/pharmacology
- Immunotherapy/methods
- Ki-1 Antigen/immunology
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Mice
- Mice, Knockout
- Mice, SCID
- Neoplasm Transplantation
- Receptors, Fc/metabolism
- Transplantation, Heterologous
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Meili Zhang
- Metabolism Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Iannello A, Ahmad A. Role of antibody-dependent cell-mediated cytotoxicity in the efficacy of therapeutic anti-cancer monoclonal antibodies. Cancer Metastasis Rev 2006; 24:487-99. [PMID: 16408158 DOI: 10.1007/s10555-005-6192-2] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, interest in anti-cancer therapeutic monoclonal antibodies (mAb) has been renewed. Several of these reagents have been approved for therapy in a variety of cancer patients and many others are in different stages of development. It is believed that multiple mechanisms are involved in the anti-cancer effects of these reagents. However, several in vitro and in vivo studies have demonstrated that antibody-dependent cell-mediated cytotoxicity (ADCC) is their predominant mode of action against cancer cells. The requirement for a direct interaction between mAb and receptors for the Fc region of the antibodies (FcR) has been demonstrated for anti-tumor effects of these antibodies. Consequently, FcR-bearing immune effector cells play an important role in mediating their effects. It is not surprising that cancer cells have developed different strategies to evade these antibodies. Several strategies are proposed to potentiate the mAb-mediated ADCC in cancer patients. They may enhance anti-cancer therapeutic effects of these regents.
Collapse
Affiliation(s)
- Alexandre Iannello
- Department of Microbiology & Immunology, Ste-Justine Hospital Research Center, University of Montreal, Quebec, Canada
| | | |
Collapse
|
150
|
Bevaart L, Jansen MJH, van Vugt MJ, Verbeek JS, van de Winkel JGJ, Leusen JHW. The High-Affinity IgG Receptor, FcγRI, Plays a Central Role in Antibody Therapy of Experimental Melanoma. Cancer Res 2006; 66:1261-4. [PMID: 16452176 DOI: 10.1158/0008-5472.can-05-2856] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the role of FcgammaR in antibody therapy of metastatic melanoma in wild-type and different FcgammaR knock-out mice. Treatment of B16F10-challenged wild-type mice with TA99 antibody specific for the gp75 tumor antigen resulted in a marked decrease in numbers of lung metastases. Treatment of individual FcgammaR knock-out mice revealed the high-affinity IgG receptor, FcgammaRI (CD64), to represent the central FcgammaR for TA99-induced antitumor effects. The potential of immune-modulating agents to further enhance the protective effect induced by monoclonal antibody (mAb) TA99 was examined in combination treatments consisting of mAb TA99 and a TLR-4 agonist, monophosphoryl lipid A (MPL). MPL did potently boost TA99 antibody-induced effects, and combination therapy was, again, found to be dependent on the presence of FcgammaRI.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibody Specificity
- Immunization, Passive/methods
- Lipid A/analogs & derivatives
- Lipid A/immunology
- Lipid A/pharmacology
- Lung Neoplasms/secondary
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, IgG/genetics
- Receptors, IgG/immunology
Collapse
Affiliation(s)
- Lisette Bevaart
- Immunotherapy Laboratory, Department of Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | | | | | | | | | | |
Collapse
|