814
Views
5
CrossRef citations to date
0
Altmetric
Review

Lonely killers

Effector cell- and complement-independent non-proapoptotic cytotoxic antibodies inducing membrane lesions

&
Pages 528-534 | Received 15 Jul 2011, Accepted 15 Aug 2011, Published online: 04 Nov 2011

Abstract

The majority of the most effective monoclonal antibodies (mAbs) currently in the clinics bind to cancer or immune cells. Classic mechanisms of cell killing by therapeutic mAbs include antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity and induction of apoptosis by engagement of specific cell ligands. A few reports have described mAbs whose cytotoxic activity is Fc-independent and that do not induce the morphological and biochemical changes associated with the apoptosis-type of cell death. Even fewer works describe mAbs able to directly induce membrane lesions. Here, we discuss the available data on those molecules and their cell killing activity, with particular attention to the case of a mAb specific for the tumor-associated N-glycolyl (Neu5Gc)-GM3 ganglioside (GM3(Neu5Gc)). Some similarities are found in the cell death pathways triggered by these mAbs, but data are not abundant. We conclude that the usefulness of mAbs with a direct cytotoxic activity for immunotherapeutic strategies deserves deeper research.

Targeting Cells with Therapeutic Antibodies

Substantial research efforts are currently devoted to elucidating the mechanisms of action of therapeutic antibodies. These molecules are leading products of the biopharmaceutical industry worldwide, both from the clinicalCitation1Citation3 and the marketCitation4 points of view. Although one approved monoclonal antibody (mAb) is directed against an infectious agent (anti-respiratory syncytial virus palivizumab), the majority of both approved and pipeline mAbs are evaluated as treatments for chronic non-transmissible diseases, particularly cancer.Citation5

In the field of tumor immunotherapy, mAbs are well ahead of cancer vaccinesCitation6Citation8 in terms of clinical efficacy and approval by regulatory agencies.Citation5 Cancer is a very complex and diverse pathology. To date, it has been postulated that a normal cell may acquire at least ten capabilities in becoming a “successful” tumor: sustained proliferative signaling; the evasion of growth suppressors; avoidance of an immune response; the possibility of replicative immortality; induction of tumor-promoting inflammation; invasiveness and metastatic potential; induction of neoangiogenesis; genome instability with accumulation of mutations; insensitivity to normal cell death pathways; and deregulation of the energetic metabolism.Citation9 Each of these steps is susceptible to different therapeutic strategies, which increasingly includes the use of mAbs either as single agents or in combination with other cancer drugs.

Approved anti-cancer mAbs target not only tumor-associated antigens, but also molecules important for the tumor microenvironment or displayed by immune cells,Citation3,Citation10 e.g., anti-CD20 rituximab (Rituxan®), anti-CD52 alemtuzumab (Campath®), anti-HER2 trastuzumab (Herceptin®), and anti-epidermal growth factor receptor (EGFR) cetuximab (Erbitux®) and nimotuzumab (CIMAher), which belong to the first group; anti-vascular endothelial growth factor (VEGF) bevacizumab (Avastin®), and anti-CTLA4 ipilimumab (Yervoy®), from the second and third groups, respectively. An alternative approach to cancer immunotherapy is the use of anti-idiotypic vaccines. In this case, the antibodies generated against the immunoglobulin acting as immunogen are supposed to recognize the tumor-associated antigen.Citation11 Although understanding the carcinogenesis process and its interaction with the immune system is leading to more effective and combined treatments, currently the majority of the mAbs with clinical efficacy directly target tumor cells. Bound mAbs can then trigger a number of cell death mechanisms that may or may not involve immune effectors.

Most therapeutic mAbs against autoimmune diseases neutralize soluble and membrane-bound proinflammatory cytokines,Citation2 e.g., anti-tumor necrosis factor (TNF) infliximab (Remicade®) and adalimumab (Humira®), which can also induce cell death. Other mAbs target surface molecules on immune cells, e.g., rituximab and alemtuzumab, which exert cytotoxicity by different mechanisms over B lymphocytes; and anti-CD3 otelixizumab (TRX4), teplizumab (MGA031), ior t3,Citation2,Citation12 and anti-CD6 itolizumab (T1hT),Citation13 which all modulate T-cell function.

In this review, we analyze the published data on non-proapoptotic mAbs still able to kill target cells without the intervention of cytotoxic cells or complement. In particular, we focus on mAbs that cause cell death by affecting membrane integrity, particularly an antibody specific for the tumor-associated N-glycolyl (Neu5Gc)-GM3 ganglioside [GM3(Neu5Gc)].Citation14

Considerations on “Classic” Antibody-Mediated Cell Death Mechanisms

Antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and apoptosis are currently regarded as the main effector functions of therapeutic mAbs for the killing of target cells.Citation2,Citation3,Citation15 For instance, from the above mentioned mAbs that bind to cells, rituximab is known to exert all of them.Citation16,Citation17

Although still the subject of some controversy, methods to measure and characterize the different types of cell death are being unified.Citation18 It is of paramount importance to assign to each cell killing activity the correct classification, thus contributing to understand the mechanism of action of therapeutic mAbs. Probably the most popular current technique is cytofluorimetry, which is widely used to quantify the above described mechanisms. Interestingly, a cytofluorimetric assay was recently proposed to differentiate ADCC from antibody-dependent cell-mediated phagocytosis (ADCP).Citation19,Citation20 Briefly, target cells stained with a vital dye and incorporating another non-vital one are considered to undergo cytotoxic killing, while those being phagocytized are identified by staining the phagocytic cell with a specific antibody-fluorophore conjugate. Using this assay, these authors postulated that the ADCC activity assigned to known therapeutic mAbs may be overestimated, while the ADCP is normally not determined.Citation21 In fact, they demonstrated that when trastuzumab isotype (gamma1) is switched to epsilon (thus obtaining a therapeutic IgE), the relative contribution of ADCC and ADCP to the cell killing properties of the IgG1 and the IgE mAbs is evidenced. Surprisingly, the IgE antibody mediated more ADCC than the IgG1, while the latter was more efficient in ADCP.Citation21 The potential use of therapeutic IgE to treat cancer, termed allergooncology, is an emerging field with interesting preliminary results.Citation22

Another polemic issue is the unequivocal definition of the apoptosis-type of cell death. Apoptosis is often considered equivalent to death with caspase activation,Citation23 but today it is well-documented that this is not always true.Citation18,Citation24,Citation25 There are several methods for identifying apoptosis, the most frequently used of which includes nevertheless caspase activation, as well as the activation of proapoptotic proteins, DNA fragmentation and phosphatidylserine exposure, among others.Citation18,Citation25 On the other hand, the necrosis-type of cell death, which is sometimes considered as an unregulated processCitation23 despite evidence that it can occur through well-controlled pathways, is defined by exclusion criteria, i.e., neither apoptosis nor cell dying with autophagy.Citation18 The fourth type of cell death is cornification, which is a protective mechanism exclusive of the skin.Citation18 Therefore, it can easily be inferred that the mAbs to whom the title of this review refers to, induce necrosis on cancer cells.

As mentioned earlier, tumors “evolve” to avoid destruction by the immune system, and the killing activities of therapeutic mAbs are also influenced by different patient factors.Citation26,Citation27 ADCC and CDC effectiveness is modulated by the host polymorphism in the Fcγ receptors (FcγRs) and key components of complement, such as C1q, respectively. CDC can also be impaired through the expression by the tumor of complement inhibitor surface proteins. Apoptosis is susceptible to alteration as a consequence of the modification of several cellular pathways.Citation26,Citation27 However, the most obvious way to circumvent the antibody attack is the change in antigen density or distribution in the malignant cell. It has thus been suggested that a successful strategy for cancer immunotherapy should be based in the simultaneous targeting of at least two independent molecules.Citation28

Active effects of passive antibodies.

The study of the mechanisms of action of therapeutic mAbs targeting cells directly have recently gone beyond their passive effects, i.e., tumor burden reduction by direct killing of cells through ADCC, CDC or apoptosis. This has come from the observation of long-lasting immune responses after the termination of mAb administration, which suggests the activation of endogenous responses.Citation3,Citation16,Citation29

The vaccinal effect of mAbs has been well documented for rituximab.Citation30 A recent example is that of 7A7, a mAb used as preclinical model of nimotuzumab.Citation31 Besides inducing ADCC, administration of 7A7 activated an anti-tumoral T-cell response.Citation32 It was later demonstrated that this antibody induces an immunogenic apoptosis-type of cell death.Citation96 Although apoptosis is generally considered to be silent in terms of immune system activation,Citation33,Citation34 under certain conditions, such as delayed phagocytosis of apoptotic bodies,Citation35 apoptotic cells can provide signals able to prime adaptive immune responses.Citation36 Therefore, the relative contribution of each type of cell death for the vaccinal effect of therapeutic mAbs remains controversial because necrotic cells can also be poorly immunogenic or even have tumor-promoting effects.Citation9,Citation37

Cytotoxic Antibodies Affecting Membrane Integrity

Antibodies with the ability to kill cells by themselves, in a non-apoptotic way, are rarely described in literature. BR96 is a mAb specific for Lewis y (Ley)Citation38 that has been used as immunotoxin in clinical trials of patients with carcinomas expressing this antigen.Citation39Citation41 The antibody was demonstrated to induce in vitro a direct cytotoxic activityCitation38 that involved loss of membrane integrity.Citation42

In 1995, a novel mechanism of cell killing by an antibody, which did not meet apoptosis criteria, was described. The antibody, RE2, was obtained by immunizing rats with a lysate of mouse T cells. The most striking finding was the induction on activated mouse T cells of extensive lesions (described by the authors as “giant holes”) that were perfectly visible by scanning electron microscopy.Citation43

A group of mAbs recognizing lymphocytes, but in this case human B cells, were reported to have a similar cytotoxic effect. Cells incubated with 216 mAb and similar human IgMs, which bind to a surface carbohydrate similar to “i” antigen on cord erythrocytes, exhibited large membrane lesions (or “pores,” according to these authors).Citation44,Citation45 A mouse IgM specific for a surface receptor on human Jurkat T cells, named anti-Porimin (derived from the phrase “pro-oncosis receptor inducing membrane injury”), also induced direct cell death with the formation of membrane pores.Citation46 Cells transfected with this receptor were also killed by this antibody with cell membrane injury.Citation47

RAV12 is a chimeric antibody that recognizes the N-linked carbohydrate antigen RAAG12.Citation48,Citation49 It was constructed from the mouse mAb KID3. Both antibody versions induced membrane rupture upon binding to a colon tumor cell line.Citation48 RAV12 was evaluated in a Phase 1 clinical study of patients with recurrent adenocarcinoma.Citation50

An IgM mAb that recognizes human embryonic stem cells, specifically through the podocalyxin-like protein-1, was found to be directly cytotoxic to these cells.Citation51 The 84 mAb induced membrane pores in treated cells.Citation52 Another well characterized antibody exhibiting this effect on plasma membrane is 14F7 mAb,Citation53 which targets the tumor-associated GM3(Neu5Gc).Citation14

14F7 mAb.

Neu5Gc-sialoconjugates are absent from human normal tissues, but tumors can differentially incorporate them from exogenous sources;Citation54 thus, they become like tumor-specific antigens. GM3(Neu5Gc) is a ganglioside whose expression has been detected in some human tumors, including breast and melanoma.Citation53,Citation55,Citation56 Several therapeutic strategies have been developed against this target,Citation14 e.g., vaccines (ganglioside-basedCitation57 and anti-idiotypicCitation58Citation63) and mAbs.Citation55 14F7 mAb is specific for this ganglioside and unable to bind its N-acetylated (Neu5Ac) counterpart.Citation53 Its exquisite specificity for GM3(Neu5Gc), which differs from GM3(Neu5Ac) only in the presence of a hydroxyl group instead of a hydrogen atom, has been explored with structural studies.Citation64 14F7 mAb was able to recognize breast and melanoma tumors by immunohistochemistry,Citation53 and also breast cancer by radioimmunoscintigraphy, in a Phase 1/2 diagnostic clinical study.Citation55

14F7 mAb displayed anti-tumor properties in vivo against a GM3(Neu5Gc)-expressing murine myeloma. In vitro, the antibody was shown to induce both ADCC and CDC, but the most interesting finding was a direct cytotoxic activity over the cells.Citation65 Cells treated with 14F7 mAb showed big “holes” on their plasma membrane,Citation66 similar to those reported previously for RE2,Citation43 216 Citation44,Citation45 and 84 Citation52 mAbs. A humanized version of 14F7 mAb, obtained by the modification of potential human T-cell epitopes on the variable region of both antibody chains,Citation67 retained this ability.Citation68 Interestingly, the recombinant antibody-producing cell line was also sensitive to 14F7 mAb-induced cytotoxicity. Therefore, for scaling up the production process, it was necessary to genetically modify these cells in order to impair the synthesis of GM3(Neu5Gc). This was achieved by transducing the cells with a lentiviral vector encoding a short hairpin RNA specific for the hydroxylase responsible for the synthesis of Neu5Gc.Citation68

Mechanism of membrane lesion formation.

The available data on the molecular mechanism(s) underlying the appearance of these plasma membrane holes or pores triggered by the above described mAbs are scattered and incomplete (). This cell death pathway, involving cell and organelle swelling, vacuolization, blebbing, membrane rupture and karyolisis (as opposed to apoptotic karyorhexis), was previously referred to as “oncosis,”Citation69 but is now recognized as an initial step of necrosis.Citation18 A feature shared by these mAbs is the induction of rapid cell death, often within 5–20 min of incubation.Citation42Citation44,Citation46,Citation51

Cytoskeleton reorganization in treated cells has been demonstrated for RE2Citation43,Citation70 and 14F7Citation66 mAbs. Addition of the actin polymerization inhibitor cytochalasin impaired the cytotoxicity induced by these mAbs. Moreover, it was demonstrated that in the RE2 mAb killing mechanism, the LFA-1 integrin plays an important role,Citation70 and 14F7 mAb binding induces phosphorylation of ezrin.Citation66 This protein is part of a triad known as ERM (ezrin-radixin-moesin), which regulates the association between membrane proteins and the cytoskeleton and participates in signal transduction pathways.Citation71 Upon phosphorylation, ezrin links the plasma membrane to actin cytoskeleton, and also interacts with transmembrane proteins. Notably, podocalyxin binds to actin through ezrin,Citation72 and the antigen recognized by 84 mAb is a podocalyxin-like protein.Citation51 In the case of cells treated with 84 mAb52 and KID3/RAV12,Citation48 a disruption of the actin cytoskeleton was observed.

BR96 is an internalizing antibody.Citation38,Citation42 Its cytotoxic activity thus begins with membrane infolding, with its subsequent internalization, cell surface and intracellular vesicle formation and loss of membrane integrity. This process required antigen cross-linking and occurred also at 4°C.Citation42 These two latter features are shared by RE2 Citation70 and 14F7 Citation66 mAbs. 84 mAb was also cytotoxic at 4°C.Citation51 Interestingly, in the case of 216 mAb cytotoxicity levels increased at this temperature when compared with incubation at 37°C.Citation44

Additionally, neither the presence of sodium azide nor of EDTA dampened the killing activity of RE2,Citation70 216 Citation44 and 14F7 Citation66 mAbs, which indicates independence from metabolic energy and calcium influx, respectively. Furthermore, with the respective cycloheximide and actinomycin inhibitors, it was demonstrated that the cytotoxic activity of 14F7 mAb does not require de novo protein or mRNA synthesis.Citation66 For KID3/RAV12, it was demonstrated that sodium influx was necessary for cell killing.Citation48 No data are available for the anti-Porimin mAb.Citation46,Citation47

In summary, there are similarities in the killing mechanism exerted by the above described mAbs (); nevertheless, the relative contribution for this activity of both the antibody isotype and the nature of the antigen recognized on the target cells is not fully elucidated.

Affinity vs. cytotoxicity.

Although generally a desired feature for therapeutic mAbs, high affinities can also have shortcomings. Cetuximab and nimotuzumab are representative examples of this phenomenon. Both mAbs bind to the EGFR, which is a validated target for cancer immunotherapy.Citation73 Nimotuzumab has a lower affinity,Citation74 and coincidently the adverse effects (mainly a skin rash) it provokes are much less serious compared with those of cetuximab.Citation75Citation77 Non-mutually exclusive hypotheses have been offered to explain its toxicity profile, including its preferential accumulation in tumor tissues, which have a higher antigen density;Citation75 the need of bivalent binding for exertion of its activity;Citation78 and its unique binding site on the EGFR extracellular domain III.Citation74

Nevertheless, in the case of 14F7 mAb-induced cell death, affinity plays a central role. Evidences of an affinity maturation process were found in 14F7 mAb variable region.Citation79 P3 is a mAb that also recognizes GM3(Neu5Gc), but also other Neu5Gc-gangliosides and sulfatides.Citation80,Citation81 In contrast to 14F7, the chimeric version of P3 Citation82 was unable to kill GM3(Neu5Gc)-expressing cells.Citation66 Originally a germ-line IgM,Citation83 its lower affinity was the explanation given to this observation.Citation66 This was later proved with a more reactive mutated variant of chimeric P3. By replacing a glutamate by an arginine residue at the heavy chain variable region (VH) of P3, thus increasing the number of this residue in the heavy chain complementary determining region 3 (H-CDR3), an antibody able to bind more strongly to P3 mAb glycolipidic ligands, without affecting its interaction with its anti-idiotypic mAbs was obtained.Citation84

Similar observations were made for 216 mAb and its family of human IgMs. These mAbs, which belong to the same VH family, bound a common anti-idiotypic antibody with similar affinities. However, the reactivity to B cells varied, and this positively correlated with the cytotoxicity. Although these are predominantly unmutated IgMs, the strongest binders display basic residues-enriched H-CDR3, with a predominance of arginines.Citation45 The importance of these latter residues for the binding of antibodies to several self-antigens, such as chromatin,Citation85 cardiolipinCitation86 and other phospholipids,Citation87 and gangliosides has been demonstrated.Citation88,Citation89

Unlike the parental antibody, the arginine-enriched mutated P3 variant displayed cytotoxic activity over cells with high expression of the ganglioside. Surprisingly, in contrast to 14F7, this antibody was able to kill cells, although to a lesser extent, devoid of gangliosides but not of Neu5Gc-sialoconjugates in general, suggesting that binding to non-glycolipidic ligands containing this variant of sialic acid can also mediate this effect. Additionally, preliminary results suggested that the mechanism of cell death induced by this mutated P3 antibody might have differences with respect to that of 14F7 mAb, as the characteristic cell swelling observed with the latter was absent in cells treated with the former.Citation84 The possible induction of membrane lesions and the determination of markers of apoptosis are pending.

P3 mAb was used to obtain racotumomab (1E10),Citation90 the anti-idiotypic antibody that is being used as a vaccine for inducing anti-GM3(Neu5Gc) antibodies.Citation58Citation61,Citation63 A positive correlation between the development of such antibodies and patient survival was found.Citation63 Despite the lack of direct cytotoxicity by P3 mAb,Citation66 the antibodies generated by the anti-idiotypic vaccine were able to directly kill GM3(Neu5Gc)-expressing cells.Citation91 Furthermore, these antibodies induced a 14F7-like type of cell death, including the formation of the membrane lesions on target cells, even at 4°C.Citation91

Other Examples Of Non-Proapoptotic Cytotoxic Antibodies

AD5-10 mAbCitation92 recognizes DR5, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 2.Citation93 This antibody induced apoptosis in several tumor cell lines, but was not toxic to human normal hepatocytes. Interestingly, using a pan-caspase inhibitor, it was observed that AD5-10 also induced non-apoptotic cell death, as shown by the absence of the typical DNA fragmentation and the simultaneous demonstration of phosphatidylserine exposure and staining with the cell membrane-impermeable dye propidium iodide.Citation92

A prostate stem cell antigen (PSCA)-specific mAb, named 1G8, was shown to directly kill antigen-expressing cells without caspase activation or apoptotic DNA fragmentation.Citation94 A cytotoxic activity with the same features was observed for alemtuzumab over chronic lymphocytic leukemia cells. Binding of this mAb caused the aggregation of glycolipid-enriched domains and the triggering of a cell death pathway.Citation95

Although cells treated with the AD5-10 mAb and the pan-caspase inhibitor exhibited a particular morphology,Citation94 the appearance of membrane lesions in these examples was not reported.

Future Directions

A critical unanswered question regarding non-proapoptotic direct cytotoxicity-inducing mAbs is why some of them only kill in vitro a small proportion of target cells, in spite of recognizing almost the whole population. Deciphering their mechanism of action at the molecular level, could help to design combined strategies to increase their cytotoxic activity, e.g., by simultaneously targeting accessory molecules participating in the cell death pathway. Currently, data on the cellular processes triggered upon antigen binding are scarce. It would be particularly interesting to describe the formation of the spectacular membrane lesions induced by these mAbs. This cytotoxic activity would contribute to other better described mechanisms such as ADCC and CDC, increasing their potential for immunotherapy of malignancies and autoimmune diseases. Also, it would be useful to determine whether cells dying this way are able to activate the adaptive immune system, i.e., whether this is an immunogenic type of cell death.

Figures and Tables

Table 1 Features of non-proapoptotic membrane lesion-inducing cytotoxic mAbs

Acknowledgments

This work was supported by the Center of Molecular Immunology.

References

  • Campoli M, Ferris R, Ferrone S, Wang X. Immunotherapy of malignant disease with tumor antigen-specific monoclonal antibodies. Clin Cancer Res 2010; 16:11 - 20; PMID: 20028761; http://dx.doi.org/10.1158/1078-0432.CCR-09-2345
  • Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and inflammation. Nat Rev Immunol 2010; 10:301 - 316; PMID: 20414204; http://dx.doi.org/10.1038/nri2761
  • Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol 2010; 10:317 - 327; PMID: 20414205; http://dx.doi.org/10.1038/nri2744
  • Scolnik PA. mAbs: a business perspective. MAbs 2009; 1:179 - 184; PMID: 20061824; http://dx.doi.org/10.4161/mabs.1.2.7736
  • Reichert JM. Antibody-based therapeutics to watch in 2011. MAbs 2011; 3:76 - 99; PMID: 21051951; http://dx.doi.org/10.4161/mabs.3.1.13895
  • Goldman B, DeFrancesco L. The cancer vaccine roller coaster. Nat Biotechnol 2009; 27:129 - 139; PMID: 19204689; http://dx.doi.org/10.1038/nbt0209-129
  • Carballido E, Fishman M. Sipuleucel-T: Prototype for development of anti-tumor vaccines. Curr Oncol Rep 2011; 13:112 - 119; PMID: 21243538; http://dx.doi.org/10.1007/s11912-011-0152-5
  • Klebanoff CA, Acquavella N, Yu Z, Restifo NP. Therapeutic cancer vaccines: are we there yet?. Immunol Rev 2011; 239:27 - 44; PMID: 21198663; http://dx.doi.org/10.1111/j.1600-065X.2010.00979.x
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:646 - 674; PMID: 21376230; http://dx.doi.org/10.1016/j.cell.2011.02.013
  • Beck A, Wurch T, Bailly C, Corvaia N. Strategies and challenges for the next generation of therapeutic antibodies. Nat Rev Immunol 2010; 10:345 - 352; PMID: 20414207; http://dx.doi.org/10.1038/nri2747
  • Lopez-Requena A, Burrone OR. Anti-idiotypic antibodies and “tumor-only” antigens: an update. Open Immunol J 2009; 2:1 - 8; http://dx.doi.org/10.2174/1874226200902010001
  • Hinojosa LE, Hernandez T, de Acosta CM, Montero E, Perez R, Lopez-Requena A. Construction of a recombinant non-mitogenic anti-human CD3 antibody. Hybridoma (Larchmt) 2010; 29:115 - 124; PMID: 20443703; http://dx.doi.org/10.1089/hyb.2009.0042
  • Alonso R, Huerta V, de Leon J, Piedra P, Puchades Y, Guirola O, et al. Towards the definition of a chimpanzee and human conserved CD6 domain 1 epitope recognized by T1 monoclonal antibody. Hybridoma (Larchmt) 2008; 27:291 - 301; PMID: 18707547; http://dx.doi.org/10.1089/hyb.2008.0007
  • Fernandez LE, Gabri MR, Guthmann MD, Gomez RE, Gold S, Fainboim L, et al. NGcGM3 ganglioside: a privileged target for cancer vaccines. Clin Dev Immunol 2010; 2010:814397; PMID: 21048926; http://dx.doi.org/10.1155/2010/814397
  • Liu XY, Pop LM, Vitetta ES. Engineering therapeutic monoclonal antibodies. Immunol Rev 2008; 222:9 - 27; PMID: 18363992; http://dx.doi.org/10.1111/j.1600-065X.2008.00601.x
  • Cartron G, Watier H, Golay J, Solal-Celigny P. From the bench to the bedside: ways to improve rituximab efficacy. Blood 2004; 104:2635 - 2642; PMID: 15226177; http://dx.doi.org/10.1182/blood-2004-03-1110
  • Weiner GJ. Rituximab: mechanism of action. Semin Hematol 2010; 47:115 - 123; PMID: 20350658; http://dx.doi.org/10.1053/j.seminhematol.2010.01.011
  • Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ 2009; 16:3 - 11; PMID: 18846107; http://dx.doi.org/10.1038/cdd.2008.150
  • Karagiannis SN, Bracher MG, Hunt J, McCloskey N, Beavil RL, Beavil AJ, et al. IgE-antibody-dependent immunotherapy of solid tumors: cytotoxic and phagocytic mechanisms of eradication of ovarian cancer cells. J Immunol 2007; 179:2832 - 2843; PMID: 17709497
  • Karagiannis SN, Bracher MG, Beavil RL, Beavil AJ, Hunt J, McCloskey N, et al. Role of IgE receptors in IgE antibody-dependent cytotoxicity and phagocytosis of ovarian tumor cells by human monocytic cells. Cancer Immunol Immunother 2008; 57:247 - 263; PMID: 17657488; http://dx.doi.org/10.1007/s00262-007-0371-7
  • Karagiannis P, Singer J, Hunt J, Gan SK, Rudman SM, Mechtcheriakova D, et al. Characterisation of an engineered trastuzumab IgE antibody and effector cell mechanisms targeting HER2/neu-positive tumour cells. Cancer Immunol Immunother 2009; 58:915 - 930; PMID: 18941743; http://dx.doi.org/10.1007/s00262-008-0607-1
  • Jensen-Jarolim E, Achatz G, Turner MC, Karagiannis S, Legrand F, Capron M, et al. Allergo Oncology: the role of IgE-mediated allergy in cancer. Allergy 2008; 63:1255 - 1266; PMID: 18671772; http://dx.doi.org/10.1111/j.1398-9995.2008.01768.x
  • Okada H, Mak TW. Pathways of apoptotic and non-apoptotic death in tumour cells. Nat Rev Cancer 2004; 4:592 - 603; PMID: 15286739; http://dx.doi.org/10.1038/nrc1412
  • Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol 2001; 2:589 - 598; PMID: 11483992; http://dx.doi.org/10.1038/35085008
  • Kepp O, Galluzzi L, Lipinski M, Yuan J, Kroemer G. Cell death assays for drug discovery. Nat Rev Drug Discov 2011; 10:221 - 237; PMID: 21358741; http://dx.doi.org/10.1038/nrd3373
  • Ferris RL, Jaffee EM, Ferrone S. Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity and immunoescape. J Clin Oncol 2010; 28:4390 - 4399; PMID: 20697078; http://dx.doi.org/10.1200/JCO.2009.27.6360
  • Reslan L, Dalle S, Dumontet C. Understanding and circumventing resistance to anticancer monoclonal antibodies. MAbs 2009; 1:222 - 229; PMID: 20065642; http://dx.doi.org/10.4161/mabs.1.3.8292
  • Alpizar YA, Chain B, Collins MK, Greenwood J, Katz D, Stauss HJ, et al. Ten years of progress in vaccination against cancer: the need to counteract cancer evasion by dual targeting in future therapies. Cancer Immunol Immunother 2011; 60:1127 - 1135; PMID: 21479639; http://dx.doi.org/10.1007/s00262-011-0985-7
  • Perez R, Moreno E, Garrido G, Crombet T. EGFR-targeting as a biological therapy: understanding Nimotuzumab's clinical effects. Cancers 2011; 3:2014 - 2031; http://dx.doi.org/10.3390/cancers3022014
  • Hilchey SP, Hyrien O, Mosmann TR, Livingstone AM, Friedberg JW, Young F, et al. Rituximab immunotherapy results in the induction of a lymphoma idiotype-specific T-cell response in patients with follicular lymphoma: support for a “vaccinal effect” of rituximab. Blood 2009; 113:3809 - 3812; PMID: 19196657; http://dx.doi.org/10.1182/blood-2008-10-185280
  • Garrido G, Sanchez B, Rodriguez HM, Lorenzano P, Alonso D, Fernandez LE. 7A7 MAb: a new tool for the pre-clinical evaluation of EGFR-based therapies. Hybrid Hybridomics 2004; 23:168 - 175; PMID: 15312307; http://dx.doi.org/10.1089/1536859041224280
  • Garrido G, Lorenzano P, Sanchez B, Beausoleil I, Alonso DF, Perez R, et al. T cells are crucial for the anti-metastatic effect of anti-epidermal growth factor receptor antibodies. Cancer Immunol Immunother 2007; 56:1701 - 1710; PMID: 17415565; http://dx.doi.org/10.1007/s00262-007-0313-4
  • Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000; 407:784 - 788; PMID: 11048729; http://dx.doi.org/10.1038/35037722
  • Matzinger P. An innate sense of danger. Ann NY Acad Sci 2002; 961:341 - 342; PMID: 12081934; http://dx.doi.org/10.1111/j.1749-6632.2002.tb03118.x
  • Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol 2008; 8:279 - 289; PMID: 18340345; http://dx.doi.org/10.1038/nri2215
  • Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol 2009; 9:353 - 363; PMID: 19365408; http://dx.doi.org/10.1038/nri2545
  • Zitvogel L, Casares N, Pequignot MO, Chaput N, Albert ML, Kroemer G. Immune response against dying tumor cells. Adv Immunol 2004; 84:131 - 179; PMID: 15246252; http://dx.doi.org/10.1016/S0065-2776(04)84004-5
  • Hellström I, Garrigues HJ, Garrigues U, Hellstrom KE. Highly tumor-reactive, internalizing, mouse monoclonal antibodies to Le(y)-related cell surface antigens. Cancer Res 1990; 50:2183 - 2190; PMID: 1690595
  • Ajani JA, Kelsen DP, Haller D, Hargraves K, Healey D. A multi-institutional phase II study of BMS-182248-01 (BR96-doxorubicin conjugate) administered every 21 days in patients with advanced gastric adenocarcinoma. Cancer J 2000; 6:78 - 81; PMID: 11069223
  • Saleh MN, Sugarman S, Murray J, Ostroff JB, Healey D, Jones D, et al. Phase I trial of the anti-Lewis Y drug immunoconjugate BR96-doxorubicin in patients with lewis Y-expressing epithelial tumors. J Clin Oncol 2000; 18:2282 - 2292; PMID: 10829049
  • Posey JA, Khazaeli MB, Bookman MA, Nowrouzi A, Grizzle WE, Thornton J, et al. A phase I trial of the single-chain immunotoxin SGN-10 (BR96 sFv-PE40) in patients with advanced solid tumors. Clin Cancer Res 2002; 8:3092 - 3099; PMID: 12374676
  • Garrigues J, Garrigues U, Hellstrom I, Hellstrom KE. Ley specific antibody with potent anti-tumor activity is internalized and degraded in lysosomes. Am J Pathol 1993; 142:607 - 622; PMID: 8434651
  • Matsuoka S, Asano Y, Sano K, Kishimoto H, Yamashita I, Yorifuji H, et al. A novel type of cell death of lymphocytes induced by a monoclonal antibody without participation of complement. J Exp Med 1995; 181:2007 - 2015; PMID: 7759995; http://dx.doi.org/10.1084/jem.181.6.2007
  • Bhat NM, Bieber MM, Stevenson FK, Teng NN. Rapid cytotoxicity of human B lymphocytes induced by VH4-34 (VH4.21) gene-encoded monoclonal antibodies. Clin Exp Immunol 1996; 105:183 - 190; PMID: 8697629; http://dx.doi.org/10.1046/j.1365-2249.1996.d01-733.x
  • Bhat NM, Bieber MM, Hsu FJ, Chapman CJ, Spellerberg M, Stevenson FK, et al. Rapid cytotoxicity of human B lymphocytes induced by VH4-34 (VH4.21) gene-encoded monoclonal antibodies, II. Clin Exp Immunol 1997; 108:151 - 159; PMID: 9097924; http://dx.doi.org/10.1046/j.1365-2249.1997.d01-976.x
  • Zhang C, Xu Y, Gu J, Schlossman SF. A cell surface receptor defined by a mAb mediates a unique type of cell death similar to oncosis. Proc Natl Acad Sci USA 1998; 95:6290 - 6295; PMID: 9600958; http://dx.doi.org/10.1073/pnas.95.11.6290
  • Ma F, Zhang C, Prasad KV, Freeman GJ, Schlossman SF. Molecular cloning of Porimin, a novel cell surface receptor mediating oncotic cell death. Proc Natl Acad Sci USA 2001; 98:9778 - 9783; PMID: 11481458; http://dx.doi.org/10.1073/pnas.171322898
  • Loo D, Pryer N, Young P, Liang T, Coberly S, King KL, et al. The glycotope-specific RAV12 monoclonal antibody induces oncosis in vitro and has antitumor activity against gastrointestinal adenocarcinoma tumor xenografts in vivo. Mol Cancer Ther 2007; 6:856 - 865; PMID: 17363480; http://dx.doi.org/10.1158/1535-7163.MCT-06-0581
  • Coberly SK, Chen FZ, Armanini MP, Chen Y, Young PF, Mather JP, et al. The RAV12 monoclonal antibody recognizes the N-linked glycotope RAAG12: expression in human normal and tumor tissues. Arch Pathol Lab Med 2009; 133:1403 - 1412; PMID: 19722746
  • Burris HA 3rd, Rosen LS, Rocha-Lima CM, Marshall J, Jones S, Cohen RB, et al. Phase 1 experience with an anti-glycotope monoclonal antibody, RAV12, in recurrent adenocarcinoma. Clin Cancer Res 2010; 16:1673 - 1681; PMID: 20179219; http://dx.doi.org/10.1158/1078-0432.CCR-09-2263
  • Choo AB, Tan HL, Ang SN, Fong WJ, Chin A, Lo J, et al. Selection against undifferentiated human embryonic stem cells by a cytotoxic antibody recognizing podocalyxin-like protein-1. Stem Cells 2008; 26:1454 - 1463; PMID: 18356574; http://dx.doi.org/10.1634/stemcells.2007-0576
  • Tan HL, Fong WJ, Lee EH, Yap M, Choo A. mAb 84, a cytotoxic antibody that kills undifferentiated human embryonic stem cells via oncosis. Stem Cells 2009; 27:1792 - 1801; PMID: 19544435; http://dx.doi.org/10.1002/stem.109
  • Carr A, Mullet A, Mazorra Z, Vazquez AM, Alfonso M, Mesa C, et al. A mouse IgG1 monoclonal antibody specific for N-glycolyl GM3 ganglioside recognized breast and melanoma tumors. Hybridoma 2000; 19:241 - 247; PMID: 10952412; http://dx.doi.org/10.1089/02724570050109639
  • Varki A. Colloquium paper: uniquely human evolution of sialic acid genetics and biology. Proc Natl Acad Sci USA 2010; 107:8939 - 8946; PMID: 20445087; http://dx.doi.org/10.1073/pnas.0914634107
  • Oliva JP, Valdes Z, Casaco A, Pimentel G, Gonzalez J, Alvarez I, et al. Clinical evidences of GM3 (NeuGc) ganglioside expression in human breast cancer using the 14F7 monoclonal antibody labelled with (99m)Tc. Breast Cancer Res Treat 2006; 96:115 - 121; PMID: 16322892; http://dx.doi.org/10.1007/s10549-005-9064-0
  • Marquina G, Waki H, Fernandez LE, Kon K, Carr A, Valiente O, et al. Gangliosides expressed in human breast cancer. Cancer Res 1996; 56:5165 - 5171; PMID: 8912852
  • Mulens V, de la Torre A, Marinello P, Rodriguez R, Cardoso J, Diaz R, et al. Immunogenicity and safety of a NeuGcGM3 based cancer vaccine: Results from a controlled study in metastatic breast cancer patients. Hum Vaccin 2010; In press PMID: 20855939
  • Alfonso M, Diaz A, Hernandez AM, Perez A, Rodriguez E, Bitton R, et al. An anti-idiotype vaccine elicits a specific response to N-glycolyl sialic acid residues of glycoconjugates in melanoma patients. J Immunol 2002; 168:2523 - 2529; PMID: 11859147
  • Díaz A, Alfonso M, Alonso R, Saurez G, Troche M, Catala M, et al. Immune responses in breast cancer patients immunized with an anti-idiotype antibody mimicking NeuGc-containing gangliosides. Clin Immunol 2003; 107:80 - 89; PMID: 12763476; http://dx.doi.org/10.1016/S1521-6616(03)00036-6
  • Guthmann MD, Castro MA, Cinat G, Venier C, Koliren L, Bitton RJ, et al. Cellular and humoral immune response to N-Glycolyl-GM3 elicited by prolonged immunotherapy with an anti-idiotypic vaccine in high-risk and metastatic breast cancer patients. J Immunother 2006; 29:215 - 223; PMID: 16531822; http://dx.doi.org/10.1097/01.cji.0000188502.11348.34
  • Neninger E, Diaz RM, de la Torre A, Rives R, Diaz A, Saurez G, et al. Active immunotherapy with 1E10 anti-idiotype vaccine in patients with small cell lung cancer: report of a phase I trial. Cancer Biol Ther 2007; 6:145 - 150; PMID: 17218777; http://dx.doi.org/10.4161/cbt.6.2.3574
  • Alfonso S, Diaz RM, de la Torre A, Santiesteban E, Aguirre F, Perez K, et al. 1E10 anti-idiotype vaccine in non-small cell lung cancer: experience in stage IIIb/IV patients. Cancer Biol Ther 2007; 6:1847 - 1852; PMID: 18075301; http://dx.doi.org/10.4161/cbt.6.12.5000
  • Hernández AM, Toledo D, Martinez D, Grinan T, Brito V, Macias A, et al. Characterization of the antibody response against NeuGcGM3 ganglioside elicited in non-small cell lung cancer patients immunized with an anti-idiotype antibody. J Immunol 2008; 181:6625 - 6634; PMID: 18941253
  • Krengel U, Olsson LL, Martinez C, Talavera A, Rojas G, Mier E, et al. Structure and molecular interactions of a unique antitumor antibody specific for N-glycolyl GM3. J Biol Chem 2004; 279:5597 - 5603; PMID: 14627696; http://dx.doi.org/10.1074/jbc.M311693200
  • Carr A, Mesa C, del Carmen Arango M, Vazquez AM, Fernandez LE. In vivo and in vitro anti-tumor effect of 14F7 monoclonal antibody. Hybrid Hybridomics 2002; 21:463 - 468; PMID: 12573110; http://dx.doi.org/10.1089/153685902321043990
  • Roque-Navarro L, Chakrabandhu K, de Leon J, Rodriguez S, Toledo C, Carr A, et al. Anti-ganglioside antibody-induced tumor cell death by loss of membrane integrity. Mol Cancer Ther 2008; 7:2033 - 2041; PMID: 18645013; http://dx.doi.org/10.1158/1535-7163.MCT-08-0222
  • Mateo C, Lombardero J, Moreno E, Morales A, Bombino G, Coloma J, et al. Removal of amphipathic epitopes from genetically engineered antibodies: production of modified immunoglobulins with reduced immunogenicity. Hybridoma 2000; 19:463 - 471; PMID: 11152398; http://dx.doi.org/10.1089/027245700750053959
  • Fernandez-Marrero Y, Roque-Navarro L, Hernandez T, Dorvignit D, Molina-Perez M, Gonzalez A, et al. A cytotoxic humanized anti-ganglioside antibody produced in a murine cell line defective of N-glycolylated-glycoconjugates. Immunobiology 2011; In press
  • Majno G, Joris I. Apoptosis, oncosis and necrosis. An overview of cell death. Am J Pathol 1995; 146:3 - 15; PMID: 7856735
  • Matsuoka S, Tsurui H, Abe M, Terashima K, Nakamura K, Hamano Y, et al. A monoclonal antibody to the alpha2 domain of murine major histocompatibility complex class I that specifically kills activated lymphocytes and blocks liver damage in the concanavalin A hepatitis model. J Exp Med 2003; 198:497 - 503; PMID: 12885869; http://dx.doi.org/10.1084/jem.20021301
  • Bretscher A, Edwards K, Fehon RG. ERM proteins and merlin: integrators at the cell cortex. Nat Rev Mol Cell Biol 2002; 3:586 - 599; PMID: 12154370; http://dx.doi.org/10.1038/nrm882
  • Takeda T, McQuistan T, Orlando RA, Farquhar MG. Loss of glomerular foot processes is associated with uncoupling of podocalyxin from the actin cytoskeleton. J Clin Invest 2001; 108:289 - 301; PMID: 11457882
  • Pal SK, Pegram M. Epidermal growth factor receptor and signal transduction: potential targets for anti-cancer therapy. Anticancer Drugs 2005; 16:483 - 494; PMID: 15846113; http://dx.doi.org/10.1097/00001813-200506000-00003
  • Talavera A, Friemann R, Gomez-Puerta S, Martinez-Fleites C, Garrido G, Rabasa A, et al. Nimotuzumab, an antitumor antibody that targets the epidermal growth factor receptor, blocks ligand binding while permitting the active receptor conformation. Cancer Res 2009; 69:5851 - 5859; PMID: 19584289; http://dx.doi.org/10.1158/0008-5472.CAN-08-4518
  • Crombet T, Osorio M, Cruz T, Roca C, del Castillo R, Mon R, et al. Use of the humanized anti-epidermal growth factor receptor monoclonal antibody h-R3 in combination with radiotherapy in the treatment of locally advanced head and neck cancer patients. J Clin Oncol 2004; 22:1646 - 1654; PMID: 15117987; http://dx.doi.org/10.1200/JCO.2004.03.089
  • Ramos TC, Figueredo J, Catala M, Gonzalez S, Selva JC, Cruz TM, et al. Treatment of high-grade glioma patients with the humanized anti-epidermal growth factor receptor (EGFR) antibody h-R3: report from a phase I/II trial. Cancer Biol Ther 2006; 5:375 - 379; PMID: 16575203; http://dx.doi.org/10.4161/cbt.5.4.2522
  • Rodríguez MO, Rivero TC, del Castillo Bahi R, Muchuli CR, Bilbao MA, Vinageras EN, et al. Nimotuzumab plus radiotherapy for unresectable squamous-cell carcinoma of the head and neck. Cancer Biol Ther 2010; 9:343 - 349; PMID: 20448462; http://dx.doi.org/10.4161/cbt.9.5.10981
  • Garrido G, Tikhomirov IA, Rabasa A, Yang E, Gracia E, Iznaga N, et al. Bivalent binding by intermediate affinity of nimotuzumab: a contribution to explain antibody clinical profile. Cancer Biol Ther 2011; 11:373 - 382; PMID: 21150278; http://dx.doi.org/10.4161/cbt.11.4.14097
  • Rodríguez M, Roque-Navarro L, Lopez-Requena A, Moreno E, Mateo de Acosta C, Perez R, et al. Insights into the immunogenetic basis of two ganglioside-associated idiotypic networks. Immunobiology 2007; 212:57 - 70; PMID: 17270710; http://dx.doi.org/10.1016/j.imbio.2006.08.005
  • Vázquez AM, Alfonso M, Lanne B, Karlsson KA, Carr A, Barroso O, et al. Generation of a murine monoclonal antibody specific for N-glycolylneuraminic acid-containing gangliosides that also recognizes sulfated glycolipids. Hybridoma 1995; 14:551 - 556; PMID: 8770642; http://dx.doi.org/10.1089/hyb.1995.14.551
  • Moreno E, Lanne B, Vazquez AM, Kawashima I, Tai T, Fernandez LE, et al. Delineation of the epitope recognized by an antibody specific for N-glycolylneuraminic acid-containing gangliosides. Glycobiology 1998; 8:695 - 705; PMID: 9621110; http://dx.doi.org/10.1093/glycob/8.7.695
  • López-Requena A, Mateo de Acosta C, Perez A, Valle A, Lombardero J, Sosa K, et al. Chimeric anti-N-glycolyl-ganglioside and its anti-idiotypic mAbs: immunodominance of their variable regions. Hybrid Hybridomics 2003; 22:235 - 243; PMID: 14511569; http://dx.doi.org/10.1089/153685903322328965
  • Perez A, Lombardero J, Mateo C, Mustelier G, Alfonso M, Vazquez AM, et al. Immunogenetic analysis of variable regions encoding AB1 and gamma-type AB2 antibodies from the NeuGc-containing ganglioside family. Hybridoma 2001; 20:211 - 221; PMID: 11604106; http://dx.doi.org/10.1089/027245701753179785
  • Fernández-Marrero Y, Hernandez T, Roque-Navarro L, Talavera A, Moreno E, Grinan T, et al. Switching on cytotoxicity by a single mutation at the heavy chain variable region of an anti-ganglioside antibody. Mol Immunol 2011; 48:1059 - 1067; PMID: 21306777; http://dx.doi.org/10.1016/j.molimm.2011.01.008
  • Guth AM, Zhang X, Smith D, Detanico T, Wysocki LJ. Chromatin specificity of anti-double-stranded DNA antibodies and a role for Arg residues in the third complementarity-determining region of the heavy chain. J Immunol 2003; 171:6260 - 6266; PMID: 14634143
  • Giles I, Lambrianides N, Latchman D, Chen P, Chukwuocha R, Isenberg D, et al. The critical role of arginine residues in the binding of human monoclonal antibodies to cardiolipin. Arthritis Res Ther 2005; 7:47 - 56; PMID: 15642142; http://dx.doi.org/10.1186/ar1449
  • Giles I, Lambrianides N, Pattni N, Faulkes D, Latchman D, Chen P, et al. Arginine residues are important in determining the binding of human monoclonal antiphospholipid antibodies to clinically relevant antigens. J Immunol 2006; 177:1729 - 1736; PMID: 16849482
  • López-Requena A, De Acosta CM, Moreno E, Gonzalez M, Puchades Y, Talavera A, et al. Gangliosides, Ab1 and Ab2 antibodies I. Towards a molecular dissection of an idiotype-anti-idiotype system. Mol Immunol 2007; 44:423 - 433; PMID: 16581129
  • Talavera A, Eriksson A, Okvist M, Lopez-Requena A, Fernandez-Marrero Y, Perez R, et al. Crystal structure of an anti-ganglioside antibody and modelling of the functional mimicry of its NeuGc-GM3 antigen by an anti-idiotypic antibody. Mol Immunol 2009; 46:3466 - 3475; PMID: 19748674; http://dx.doi.org/10.1016/j.molimm.2009.07.032
  • Vázquez AM, Perez A, Hernandez AM, Macias A, Alfonso M, Bombino G, et al. Syngeneic anti-idiotypic monoclonal antibodies to an anti-NeuGc-containing ganglioside monoclonal antibody. Hybridoma 1998; 17:527 - 534; PMID: 9890708; http://dx.doi.org/10.1089/hyb.1998.17.527
  • Hernández AM, Rodriguez N, Gonzalez JE, Reyes E, Rondon T, Grinan T, et al. Anti-NeuGcGM3 antibodies, actively elicited by idiotypic vaccination in nonsmall cell lung cancer patients, induce tumor cell death by an oncosis-like mechanism. J Immunol 2011; 186:3735 - 3744; PMID: 21300821; http://dx.doi.org/10.4049/jimmunol.1000609
  • Guo Y, Chen C, Zheng Y, Zhang J, Tao X, Liu S, et al. A novel anti-human DR5 monoclonal antibody with tumoricidal activity induces caspase-dependent and caspase-independent cell death. J Biol Chem 2005; 280:41940 - 41952; PMID: 16234248; http://dx.doi.org/10.1074/jbc.M503621200
  • Wu GS, Kim K, el-Deiry WS. KILLER/DR5, a novel DNA-damage inducible death receptor gene, links the p53-tumor suppressor to caspase activation and apoptotic death. Adv Exp Med Biol 2000; 465:143 - 151; PMID: 10810622; http://dx.doi.org/10.1007/0-306-46817-4_13
  • Gu Z, Yamashiro J, Kono E, Reiter RE. Anti-prostate stem cell antigen monoclonal antibody 1G8 induces cell death in vitro and inhibits tumor growth in vivo via a Fc-independent mechanism. Cancer Res 2005; 65:9495 - 9500; PMID: 16230414; http://dx.doi.org/10.1158/0008-5472.CAN-05-2086
  • Mone AP, Cheney C, Banks AL, Tridandapani S, Mehter N, Guster S, et al. Alemtuzumab induces caspase-independent cell death in human chronic lymphocytic leukemia cells through a lipid raft-dependent mechanism. Leukemia 2006; 20:272 - 279; PMID: 16341049; http://dx.doi.org/10.1038/sj.leu.2404014
  • Garrido G, Rabasa A, Sánchez B, López MV, Blanco R, López A, et al. Induction of immunogenic apoptosis by blockade of epidermal growth factor receptor activation with a specific antibody. J Immunol 2011; In press PMID: 21984704; http://dx.doi.org/10.4049/jimmunol.1003477

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.