Skip to main content
Antibody Therapeutics logoLink to Antibody Therapeutics
. 2018 Jun 24;1(1):7–12. doi: 10.1093/abt/tby002

Enhancing antibody-dependent cell-mediated cytotoxicity: a strategy for improving antibody-based immunotherapy

David Zahavi 1, Dalal AlDeghaither 1, Allison O’Connell 1, Louis M Weiner 1,
PMCID: PMC7990127  PMID: 33928217

ABSTRACT

The targeting of surface antigens expressed on tumor cells by monoclonal antibodies (mAbs) has revolutionized cancer therapeutics. One mechanism of action of antibody-based immunotherapy is the activation of immune effector cells to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This review will summarize the process of ADCC, its important role in the efficacy of mAb therapy, how to measure it, and finally future strategies for antibody design that can take advantage of it to improve clinical performance.

Keywords: ADCC, monoclonal antibody, immunotherapy, FcγR, Fc glycosylation


Statement of Significance

Targeted mAb therapy represents a promising therapeutic strategy for many cancer types. ADCC is a crucial mechanism underlying targeted antibody-based immunotherapy approaches. Many patients have limited responses to mAb therapy and there is a great need for antibodies with enhanced clinical efficacy. Designing and engineering antibodies with enhanced ADCC eliciting properties will improve patient outcomes.

INTRODUCTION

Targeted therapy utilizing monoclonal antibodies (mAbs) has established immunotherapy as a powerful new tool in the fight against cancer. Antibodies directed towards tumor cell antigens can cause tumor cell death by both direct and indirect mechanisms. Direct mechanisms for therapeutic antibodies to induce cell death include blocking growth factor receptor signaling, direct transmembrane signaling, and acting as targeted vectors for toxic payloads such as radioisotopes [1]. The indirect mechanisms require engagement with components of the host immune system and are comprised of complement-mediated cytotoxicity, antibody-dependent cellular phagocytosis, and antibody-dependent cell-mediated cytotoxicity (ADCC), which is the subject of this review. The phenomenon of ADCC was first described in 1965 by Erna Möller who demonstrated that lymphoid effector cells only induced cytotoxic death of tumor cells when in the presence of anti-serum from rabbits previously inoculated by those tumor cells [2]. It was eventually discovered that the factor in the anti-serum required for cell-mediated cytotoxicity was immunoglobulins [3] and the process was initially termed “antibody-dependent lymphocyte-mediated cytotoxicity” [4]. Subsequent research identified multiple cell types capable of facilitating cytotoxicity. Thus, ADCC was established as an immune mechanism involving three components: effector cells, antibodies, and target cells opsonized by the antibody. As mAb therapy has revolutionized cancer treatment, ADCC has become more relevant in a clinical context. Clinical trials have demonstrated that many mAbs function, in part, by eliciting ADCC. Now that an increasing number of therapeutic antibodies are receiving approval from the FDA and entering the clinic, a significant goal moving forward is to construct more effective mAbs. Future design strategies will have to incorporate an understanding of ADCC and how to best enhance its effects to improve cancer patient outcomes.

ADCC MECHANISMS

ADCC is the process by which antibodies coat a target cell and recruit effector cells to induce target cell death via non-phagocytic mechanisms (Fig. 1). Antibodies can bind to their specific antigens on the target cell surface via their antigen-binding fragment (Fab) portions and interact with effector cells via their fragment crystallizable region (Fc) portions thereby acting as bridges that link the effector to a target. While several classes of human antibodies can mediate ADCC, including IgG, IgA, and IgE, IgG1 is the most used subclass for cancer therapeutic antibodies [5]. In order for an effector cell to carry out ADCC it must express Fc receptors (FcR) that will bind the antibody [6]. The known classes of FcR include FcγR, which bind IgG; FcαR, which bind IgA; and FcεR, which bind IgE. FcγR are the most important for tumor cell clearance by myeloid cells and are comprised of activating FcγRI (CD64), FcγRIIA (CD32A), FcγRIIIA (CD16A), and inhibitory FcγRIIB (CD32B) receptors [7]. Once the FcγR binds antibody it triggers receptor cross-linking and downstream signal propagation. Activating FcγR signal via their immunoreceptor tyrosine-based activation motifs while inhibitory FcγR signal via their immunoreceptor tyrosine-based inhibitory motifs [8]. Many effector cells also express other receptor types such as the inhibitory killer inhibitory receptors (e.g. KIR) and activating NKG2D receptors on natural killer (NK) cells. The delicate balance amongst the activating and inhibitory pathway signaling ultimately determines effector cell response. Myeloid cells capable of acting as ADCC effectors are NK cells, monocytes, macrophages, neutrophils, eosinophils, and dendritic cells [9]. Once these effector cells have been activated they mediate target cell death through three key mechanisms: cytotoxic granule release, Fas signaling, and elaboration of reactive oxygen species. The main and best characterized mechanism utilized in ADCC is the release of perforins and granzymes from effector cell granules. Upon activation, effector cells such as NK cells polarize and exocytose their granules in a calcium-dependent manner [10]. Perforin and Granzyme B work in concert to induce cell death. Perforins create pores in the cell membrane that facilitate granzyme B entry into the target cell, resulting in DNA fragmentation and apoptosis [11]. Additionally, research has suggested that activated effector cells will upregulate Fas ligand expression in order to cause apoptosis in the target via Fas signaling [12]. While multiple myeloid lineage cells are capable of ADCC, in the context of cancer immunotherapy NK cells appear to be the major effector cell type in vivo. The clinical efficacy of many targeted mAb therapies has been demonstrated to be NK cell dependent [13]. NK cells highly express activating FcγRIIIA and do not express the inhibitory FcγRIIB; therefore modifying antibody interactions specifically through FcγRIIIA has become of interest for cancer immunotherapy.

Figure 1.

Figure 1

Diagram of ADCC.

ROLE OF ADCC IN IMMUNOTHERAPY

Currently used mAbs are known to employ a variety of the indirect mechanisms mentioned above to exert cytotoxic effects on tumor cells. Although each of these mechanisms of action is believed to be important, there has been some debate about their role in vivo. Many antitumor mAbs were shown to mediate ADCC in vitro; however for a time the relevance to therapeutic efficacy was unknown. Seminal work from Clynes et al. demonstrated that ADCC was a substantive contributor to the in vivo activity of the mAbs trastuzumab and rituximab using mouse models [14]. Their results indicated that clinical efficacy depended upon the balance of engagement with activating FcγRIII and inhibitory FcγRIIB. This provided the initial rationale for designing mAbs that could preferentially bind the activating receptor. Additional mechanistic studies in mice confirmed that FcγR on innate immune system mononuclear cells were required for tumor clearance [15]. Subsequently, de Haij et al. demonstrated that ADCC was essential for mAb therapy in a novel mouse model where a mutant FcγR incapable of ADCC was unable to clear tumors [16]. One drawback to the majority of these studies was that the mAbs used to assess whether ADCC played a significant role in in vivo efficacy utilized additional mechanisms of action such as signaling perturbation. Therefore, these results proved ADCC was required but not necessarily whether ADCC by itself would be sufficient for tumor clearance. Recent work with mAbs to tumor-associated antigens that rely solely on ADCC as their mechanism of action confirmed that ADCC alone is able to mediate therapeutic benefit [17].

Since FcγR is crucial for mAb efficacy in mouse models, its role in human cancer patients was explored using clinical trial data to correlate FcγR polymorphisms with clinical outcomes. In humans FcγRIIA is polymorphic at position 131 and alleles exist that code for either a histidine (H) or arginine (R). FcγRIIIA is polymorphic at position 158 with alleles for either a valine (V) or phenylalanine (F). Individuals with FcγRIIA-131H/H and FcγRIIIA-158V/V genotypes have FcγR with higher affinity for IgG1 and therefore enhanced ADCC [18,19]. This augmented ADCC was demonstrated in several studies to cause FcγRIIIA-158V/V lymphoma patients to have better clinical response to rituximab, a mAb against CD20 [20–22]. Interestingly, although the inhibitory FcγRIIB is also polymorphic, no association between inhibitory receptor genotype and rituximab response was observed [23]. Both activating FcγRIIA and FcγRIIIA genotypes associated with higher affinities for Fc were strong predictors for improved survival when colorectal patients were treated with cetuximab, a mAb targeting the epidermal growth factor receptor (EGFR), and metastatic breast cancer patients were treated with trastuzumab, a mAb targeting human epidermal growth factor receptor 2 (HER2) [24,25]. However, in nonmetastatic breast cancer patients there was no correlation between FcγR genotypes and trastuzumab efficacy [26]. This suggests that the role of ADCC may be different depending on cancer stage. In more recent studies, the FcγR polymorphisms in patients treated with cetuximab or trastuzumab were directly linked to the ability to carry out ADCC by in vitro studies using patient-derived immune cells [27,28]. In neuroblastoma patients treated with anti-GD2 antibodies, a similar association between the high-affinity FcγRII/IIIA genotypes and survival was found, and appeared to be linked to an increase in ADCC activity [29]. These multiple analyses confirm that patients with higher-affinity FcγR have better clinical outcomes when administered by mAb therapy, irrespective of cancer type and the mAb target antigen.

In addition to examining FcγR polymorphisms, other studies have used patient samples from clinical trials to investigate the relative importance of ADCC to therapeutic success. For example, patients with HER2-positive breast cancer treated with trastuzumab, docetaxel, or another treatment had immunohistochemistry performed on their tumor samples to measure granzyme B expression as a surrogate marker of ADCC activity. Those who received trastuzumab were found to have better overall survival and had much higher levels of ADCC compared to the other cohorts [30]. Furthermore, patient-derived in vitro models demonstrated ADCC as a major therapeutic mechanism of rituximab in non-Hodgkin lymphoma [31] and anti-CD38 antibodies in multiple myeloma [32,33]. There is now strong evidence that ADCC plays a critical role in facilitating mAb antitumor responses in patients. In fact, additional variables that would affect ADCC such as target antigen expression level and density, mAb isotype, and mAb dose all correlate with clinical response [34]. Now that ADCC is recognized as a determining factor for mAb therapy success, research and development of novel mAbs have shifted towards designing mAbs with improved capacity to mediate ADCC.

MEASURING ADCC

Strong evidence for the critical role of ADCC in targeted mAb clinical efficacy has inspired efforts to engineer mAbs with enriched ADCC functionality. Surprisingly, techniques currently employed to quantify ADCC require continued development in order to properly assess and compare the properties of newly developed mAbs. The first assay for calculating cytotoxicity was published in 1968 and involved radiolabeling target cells with 51Cr [35]. 51Cr release could be used as a surrogate for ADCC activity and this quickly became the gold standard assay. However, the use of radioactive substances poses regulatory and safety challenges and generally works best only when high amounts of cytotoxicity are expected. Modern fluorometric techniques have largely replaced radiolabeling with fluorescent markers that allow for more precise measurements of cellular cytotoxicity. Labeling of target cells with fluorescent dyes allowed for sensitive determination of cytotoxicity via flow cytometry [36]. Expanding upon these initial flow cytometric techniques, the VITAL assay was developed to measure cytotoxicity of multiple target populations simultaneously both in vitro and in vivo [37]. There are several newer techniques that rely on similar principles as the 51Cr release assay but replace radioisotopes with natural cell products. As cells are killed they release lactate dehydrogenase and other proteases that can be quantified by supplying fluorogenic substrates in order to more accurately assess cytotoxicity without the need to perform any labeling or manipulation of target cells [38,39]. A potential drawback of relying on reporter molecules is that they may spontaneously release even from live cells. Impedance-based analysis avoids this issue and has shown promise as a quantitative measure of cell-mediated cytotoxicity that can continuously measure ADCC over time [40]. More recently, scientists have recognized the need for a sensitive, reliable, and accurate cytotoxicity assay that is compliant with regulatory requirements. One such method involves combining flow cytometric and labeling-based techniques for high-throughput single-cell computer image analysis [41]. An alternative method genetically modifies target cells to express a reporter protein for a standardized assay [42]. Another variable to consider is the effector cell populations used in the assay. Currently, the most widely used cells for quantifying ADCC are peripheral blood mononuclear cells and purified NK cells from donors. Unfortunately, the FcγR and KIR molecule genotype differences inherent in diverse donor-derived cells will cause high inter-assay variability. In order to address these limitations an immortalized NK cell line that expresses the high-affinity CD16V variant has been developed that allows for more standardized ADCC assays [43]. Additional engineered effector cell lines have been created for single assay thaw and use format to further reduce variability [44]. Robust ADCC assays are not only needed for assessing mAb quality but also for predicting patient responses. Clinically available blood samples can be used without isolating NK cells in a rapid ADCC reporter assay that could be applied to cancer patients [45]. Establishing a new gold standard technique for accurately quantifying cell-mediated cytotoxicity for widespread use will be essential for proper comparison of newly designed mAbs that aim to improve therapeutic benefit by enhancement of ADCC.

IMPROVING THERAPEUTIC BENEFIT BY ENHANCING ADCC

Now that ADCC is recognized as a crucial mechanism of action for targeted mAb cancer therapy there has been increasing interest for clinically testing mAbs engineered with properties that enhance ADCC. While there are many methods to augment ADCC, including supplementation of cytokines or manipulation of the effector cells, we focus here on the modification of therapeutic antibodies’ structures [46,47],. The main strategy to enhance ADCC functionality has been to alter the Fc portion of the mAb to increase binding affinity to the activating FcγRIIIA via site-directed mutagenesis, changing Fc domain glycosylation, and/or removing Fc domain fucosylation. Creation of IgG1 variants with improved binding to activating FcγR by mutagenesis has been an effective strategy for increasing ADCC efficiency in vitro [48,49]. In addition to modification of Fc residues, asymmetrical engineering of the Fc portion to create heterodimers of different heavy chains yielded more stable antibodies with enhanced ADCC functionality [50]. The use of mAbs engineered by mutagenesis for optimal FcγR binding to improve therapeutic efficacy has been further validated by preclinical animal models [51,52]. For several different antibodies, it has also been shown that alterations of the glycosylation patterns can be used to increase affinity for activating FcγR in order to boost ADCC activity [53–55]. In one study, optimization of FcγRIIIA binding only affected the ADCC properties of a subset of effector cells, particularly NK cells, and further optimization for FcγRIIA was required to enhance ADCC mediated by other effector cells [56]. Of the oligosaccharides attached to the Fc domain fucose sugar units appear to play the largest role in determining binding to FcγRIIIA. Removal of fucose improved affinity by up to 50-fold and afucosylated mAbs mediated higher levels of ADCC both in vitro and in vivo [57,58]. These results led to the development of methods for producing mAbs that lacked fucose in their Fc region [59]. Afucosylated versions of commonly used therapeutic mAbs have superior efficacy in vivo [60,61] and have also entered into clinical trials [62,63]. As many of the currently available antibodies are highly fucosylated [64], this represents an important opportunity to improve the clinical efficacy of mAb therapy. Another important mAb therapeutic strategy has been to target inhibitory immune checkpoints on T cells. While mAbs against molecules such as PD-1 are either designed or engineered to eliminate their ADCC activity to prevent immune cell fratricide, novel antibodies targeting the PD-L1 expressed on tumor cells are being designed to both block the immune checkpoint pathway and have enhanced ADCC of the tumor cells [65].

Evaluating antibodies for clinical use based on their ability to facilitate ADCC is not a new concept [66] and mAbs to novel targets are being developed with ADCC in mind [67]. The introduction of targeted mAb therapy has revolutionized the treatment of cancer; however, there is still room for improvement. Detailed analysis of the mechanisms of action of mAbs has revealed that amplifying effector functions, specifically ADCC, is a promising approach to increase clinical benefit. The next generation of therapeutic antibodies must take advantage of engineering strategies to increase benefits for patients. Although many of the new mAbs have shown enhancement of efficacy in preclinical models, studies in patients will ultimately reveal if this approach has general merit. A list of mAbs targeting tumor antigens engineered to augment ADCC that are currently enrolled in or have completed clinical trials is presented in Table 1. These trials will determine if ADCC-augmenting antibodies have acceptable safety profiles and meaningfully impact patient outcomes. The data implicating ADCC as a crucial player in targeted mAb cancer therapy is encouraging and suggests that advances in antibody engineering will ultimately lead to more powerful therapeutic antibodies.

Table 1.

ADCC-Enhancing Antibodies Approved or in Clinical Trials

Antibody Target Modification Status  *
Obinutuzumab CD20 Reduced fucosylation Completed Clinical Trials. FDA Approved for CLL in 2013.
Mogamulizumab CCR4 Afucosylated Completed Clinical Trials. Approved in Japan for T-Cell Lymphoma. FDA Approved for non-Hodgkins lymphoma in 2018.
Margetuximab HER2 Fc Mutagenesis Phase 3 Clinical Trial (NCT02492711)
MOR208 CD19 Fc Mutagenesis Phase 3 Clinical Trial (NCT02763319)
Ublituximab CD20 Reduced fucosylation Phase 3 Clinical Trial (NCT02612311)
MEDI-551 CD19 Afucosylated Phase 2 Clinical Trial (NCT01466153)
Gatipotuzumab MUC1 Altered glycosylation Phase 2 Clinical Trial (NCT01899599)
Tomuzotuximab EGFR Altered glycosylation Phase 2 Clinical Trial (NCT02052960)
Ocaratuzumab CD20 Fc Mutagenesis Phase 2 Clinical Trial
(NCT00003874)
RO5083945 (GA201) EGFR Altered glycosylation Phase 2 Clinical Trial (NCT01326000)
TrasGEX HER2 Altered glycosylation Phase 1 Clinical Trial
(NCT01409343)

*A single representative of the most advanced clinical trial is included.

REFERENCES

  • 1. Adams, G, Weiner, L. Monoclonal antibody therapy of cancer. Nat Biotechnol 2005; 23: 1147–57. [DOI] [PubMed] [Google Scholar]
  • 2. Moller, E. Contact-induced cytotoxicity by lymphoid cells containing foreign isoantigens. Science 1965; 147: 873–9. [DOI] [PubMed] [Google Scholar]
  • 3. MacLennan, ICM, Loewi, G, Harding, B. The role of immunoglobulins in lymphocyte-mediated cell damage, in vitro. I. Comparison of the effects of target cell specific antibody and normal serum factors on cellular damage by immune and non-immune lymphocytes. Immunology 1970; 18: 397–404. [PMC free article] [PubMed] [Google Scholar]
  • 4. Pudifin, DJ, Harding, B, MacLennan, ICM. The differential effect of γ irradiation on the sensitizing and effector stages of antibody dependent lymphocyte mediated cytotoxicity. Immunology 1971; 21: 853–60. [PMC free article] [PubMed] [Google Scholar]
  • 5. Teillaud, J.  Antibody-dependent cellular cytotoxicity (ADCC). Encyclopedia of Life Sciences. 2012, 10.1002/9780470015902.a0000498.pub2. [DOI]
  • 6. Fanger, M, Shen, L, Graziano, Ret al.  Cytotoxicity mediated by human Fc receptors for IgG. Immunol Today 1989; 10: 92–9. [DOI] [PubMed] [Google Scholar]
  • 7. Wallace, P, Howell, A, Fanger, M. Role of Fcγ receptors in cancer and infectious disease. J Leukoc Biol 1994; 55: 816–26. [DOI] [PubMed] [Google Scholar]
  • 8. Shuptrine, C, Surana, R, Weiner, L. Monoclonal antibodies for the treatment of cancer. Semin Cancer Biol 2012; 22: 3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Nimmerjahn, F, Ravetch, J. Fcγ receptors as regulators of immune responses. Nat Rev Immunol 2008; 8: 34–47. [DOI] [PubMed] [Google Scholar]
  • 10. de  Saint Basile, G, Ménasché, G, Fischer, A.  Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules. Nat Rev Immunol 2010; 10: 568–79. [DOI] [PubMed] [Google Scholar]
  • 11. Ackerman, M, Nimmerjahn, F. Antibody Fc. London, Waltham, MA, and San Diego, CA.Elsevier Inc., 2014. [Google Scholar]
  • 12. Eischen, C, Leibson, P. Role for NK-cell-associated Fas ligand in cell-mediated cytotoxicity and apoptosis. Res Immunol 1997; 148: 164–9. [DOI] [PubMed] [Google Scholar]
  • 13. Alderson, K, Sondel, P. Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity. J Biomed Biotechnol 2011; 2011: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Clynes, R, Towers, T, Presta, Let al.  Inhibitory Fc receptors modulate in vivo cytoxicity against tumortargets. Nat Med 2000; 6: 443–6. [DOI] [PubMed] [Google Scholar]
  • 15. Minard-Colin, V, Xiu, Y, Poe, Jet al.  Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcγRI, FcγRIII, and FcγRIV. Blood 2008; 112: 1205–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. de  Haij, S, Jansen, J, Boross, Pet al.  In vivo cytotoxicity of type I CD20 antibodies critically depends on Fc receptor ITAM signaling. Cancer Res 2010; 70: 3209–17. [DOI] [PubMed] [Google Scholar]
  • 17. Hubert, P, Heitzmann, A, Viel, Set al.  Antibody-dependent cell cytotoxicity synapses form in mice during tumor-specific antibody immunotherapy. Cancer Res 2011; 71: 5134–43. [DOI] [PubMed] [Google Scholar]
  • 18. Bibeau, F, Lopez-Crapez, E, Di Fiore, Fet al.  Impact of FcγRIIa-FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol 2009; 27: 1122–9. [DOI] [PubMed] [Google Scholar]
  • 19. Wu, J, Edberg, J, Redecha, Pet al.  A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest 1997; 100: 1059–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Cartron, G. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor Fcgamma RIIIa gene. Blood 2002; 99: 754–8. [DOI] [PubMed] [Google Scholar]
  • 21. Weng, W, Levy, R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol 2003; 21: 3940–7. [DOI] [PubMed] [Google Scholar]
  • 22. Hatjiharissi, E, Xu, L, Santos, Det al.  Increased natural killer cell expression of CD16, augmented binding and ADCC activity to rituximab among individuals expressing the Fc RIIIa-158 V/V and V/F polymorphism. Blood 2007; 110: 2561–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Weng, W, Levy, R. Genetic polymorphism of the inhibitory IgG Fc receptor FcgammaRIIb is not associated with clinical outcome in patients with follicular lymphoma treated with rituximab. Leuk Lymphoma 2009; 50: 723–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Rodríguez, J, Zarate, R, Bandres, Eet al.  Fc gamma receptor polymorphisms as predictive markers of Cetuximab efficacy in epidermal growth factor receptor downstream-mutated metastatic colorectal cancer. Eur J Cancer 2012; 48: 1774–80. [DOI] [PubMed] [Google Scholar]
  • 25. Musolino, A, Naldi, N, Bortesi, Bet al.  Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu–positive metastatic breast cancer. J Clin Oncol 2008; 26: 1789–96. [DOI] [PubMed] [Google Scholar]
  • 26. Hurvitz, S, Betting, D, Stern, Het al.  Analysis of Fcγ receptor IIIa and IIa polymorphisms: lack of correlation with outcome in trastuzumab-treated breast cancer patients. Clin Cancer Res 2012; 18: 3478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Taylor, R, Saloura, V, Jain, Aet al.  Ex vivo antibody-dependent cellular cytotoxicity inducibility predicts efficacy of cetuximab. Cancer Immunol Res 2015; 3: 567–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Boero, S, Morabito, A, Banelli, Bet al.  Analysis of in vitro ADCC and clinical response to trastuzumab: possible relevance of FcγRIIIA/FcγRIIA gene polymorphisms and HER-2 expression levels on breast cancer cell lines. J Transl Med 2015; 13: 324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Siebert, N, Jensen, C, Troschke-Meurer, Set al.  Neuroblastoma patients with high-affinity FCGR2A, -3A and stimulatory KIR 2DS2 treated by long-term infusion of anti-GD2 antibody ch14.18/CHO show higher ADCC levels and improved event-free survival. OncoImmunology 2016; 5: e1235108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Arnould, L, Gelly, M, Penault-Llorca, Fet al.  Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism?  Br J Cancer  2006; 94: 259–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Vermi, W, Micheletti, A, Finotti, Get al.  slan+ monocytes and macrophages mediate CD20-dependent B cell lymphoma elimination via ADCC and ADCP. Cancer Res 2018. 10.1158/0008-5472.can-17-2344. [DOI] [PubMed] [Google Scholar]
  • 32. de  Weers, M, Tai, Y, van derVeer, Met al.  Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol 2010; 186: 1840–8. [DOI] [PubMed] [Google Scholar]
  • 33. van de  Donk, N. Immunomodulatory effects of CD38-targeting antibodies. Immunol Lett 2018; 199: 16–22. [DOI] [PubMed] [Google Scholar]
  • 34. Ferris, R, Jaffee, E, Ferrone, S. Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity, and immunoescape. J Clin Oncol 2010; 28: 4390–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Brunner, KT, Mauel, J, Cerottini, JCet al.  Quantitative assay of the lytic action of immune lymphoid cells on 51-Cr-labelled allogeneic target cells in vitro; inhibition by isoantibody and by drugs. Immunology 1968; 14: 181–96. [PMC free article] [PubMed] [Google Scholar]
  • 36. Radošević, K, Garritsen, H, Van Graft, Met al.  A simple and sensitive flow cytometric assay for the determination of the cytotoxic activity of human natural killer cells. J Immunol Methods 1968; 135: 81–9. [DOI] [PubMed] [Google Scholar]
  • 37. Hermans, I, Silk, J, Yang, Jet al.  The VITAL assay: a versatile fluorometric technique for assessing CTL- and NKT-mediated cytotoxicity against multiple targets in vitro and in vivo. J Immunol Methods 2004; 285: 25–40. [DOI] [PubMed] [Google Scholar]
  • 38. Hassenrück, F, Knödgen, E, Göckeritz, Eet al.  Sensitive detection of the natural killer cell-mediated cytotoxicity of anti-CD20 antibodies and its impairment by B-cell receptor pathway inhibitors. BioMed Res Int 2018; 2018: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Niles, A, Moravec, R, Eric Hesselberth, Pet al.  A homogeneous assay to measure live and dead cells in the same sample by detecting different protease markers. Anal Biochem 2007; 366: 197–206. [DOI] [PubMed] [Google Scholar]
  • 40. Tóth, G, Szöllősi, J, Vereb, G. Quantitating ADCC against adherent cells: Impedance-based detection is superior to release, membrane permeability, or caspase activation assays in resolving antibody dose response. Cytometry A 2017; 91: 1021–9. [DOI] [PubMed] [Google Scholar]
  • 41. Welter, A, Sundararaman, S, Li, Ret al.  High-throughput GLP-capable target cell visualization assay for measuring cell-mediated cytotoxicity. Cells 2018; 7: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Rossignol, A, Bonnaudet, V, Clémenceau, Bet al.  A high-performance, non-radioactive potency assay for measuring cytotoxicity: a full substitute of the chromium-release assay targeting the regulatory-compliance objective. MAbs 2017; 9: 521–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Binyamin, L, Alpaugh, R, Hughes, Tet al.  Blocking NK cell inhibitory self-recognition promotes antibody-dependent cellular cytotoxicity in a model of anti-lymphoma therapy. J Immunol 2008; 180: 6392–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Cheng, Z, Garvin, D, Paguio, Aet al.  Development of a robust reporter-based ADCC assay with frozen, thaw-and-use cells to measure Fc effector function of therapeutic antibodies. J Immunol Methods 2014; 414: 69–81. [DOI] [PubMed] [Google Scholar]
  • 45. Sung, A, Tang, J, Guglielmo, Met al.  An improved method to quantify human NK cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) per IgG FcR-positive NK cell without purification of NK cells. J Immunol Methods 2018; 452: 63–72. [DOI] [PubMed] [Google Scholar]
  • 46. Wang, W, Erbe, A, Hank, Jet al.  NK cell-mediated antibody-dependent cellular cytotoxicity in cancer immunotherapy. Front Immunol 2015; 6: 368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Ochoa, M, Minute, L, Rodriguez, Iet al.  Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol Cell Biol 2017; 95: 347–55. [DOI] [PubMed] [Google Scholar]
  • 48. Shields, R, Namenuk, A, Hong, Ket al.  High resolution mapping of the binding site on human IgG1 for FcγRI, FcγRII, FcγRIII, and FcRn and design of IgG1 variants with improved binding to the FcγR. J Biol Chem 2000; 276: 6591–604. [DOI] [PubMed] [Google Scholar]
  • 49. Tang, Y, Lou, J, Alpaugh, Ret al.  Regulation of antibody-dependent cellular cytotoxicity by IgG intrinsic and apparent affinity for target antigen. J Immunol 2007; 179: 2815–23. [DOI] [PubMed] [Google Scholar]
  • 50. Liu, Z, Gunasekaran, K, Wang, Wet al.  Asymmetrical Fc engineering greatly enhances antibody-dependent cellular cytotoxicity (ADCC) effector function and stability of the modified antibodies. J Biol Chem 2013; 289: 3571–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Lazar, G, Dang, W, Karki, Set al.  Engineered antibody Fc variants with enhanced effector function. Proc Natl Acad Sci 2006; 103: 4005–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zalevsky, J, Leung, I, Karki, Set al.  The impact of Fc engineering on an anti-CD19 antibody: increased Fc receptor affinity enhances B-cell clearing in nonhuman primates. Blood 2009; 113: 3735–43. [DOI] [PubMed] [Google Scholar]
  • 53. Umaña, P, Jean–Mairet, J, Moudry, Ret al.  Engineered glycoforms of an antineuroblastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity. Nat Biotechnol 1999; 17: 176–80. [DOI] [PubMed] [Google Scholar]
  • 54. Davies, J, Jiang, L, Pan, Let al.  Expression of GnTIII in a recombinant anti-CD20 CHO production cell line: expression of antibodies with altered glycoforms leads to an increase in ADCC through higher affinity for FCγRIII. Biotechnol Bioeng 2001; 74: 288–94. [PubMed] [Google Scholar]
  • 55. Sibéril, S, deRomeuf, C, Bihoreau, Net al.  Selection of a human anti-RhD monoclonal antibody for therapeutic use: impact of IgG glycosylation on activating and inhibitory FcγR functions. Clin Immunol 2006; 118: 170–9. [DOI] [PubMed] [Google Scholar]
  • 56. Derer, S, Glorius, P, Schlaeth, Met al.  Increasing FcγRIIa affinity of an FcγRIII-optimized anti-EGFR antibody restores neutrophil-mediated cytotoxicity. MAbs 2013; 6: 409–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Shields, R, Lai, J, Keck, Ret al.  Lack of fucose on human IgG1N-linked oligosaccharide improves binding to human FcγRIII and antibody-dependent cellular toxicity. J Biol Chem 2002; 277: 26733–40. [DOI] [PubMed] [Google Scholar]
  • 58. Liu, S, Chalouni, C, Young, Jet al.  Afucosylated antibodies increase activation of Fc RIIIa-dependent signaling components to intensify processes promoting ADCC. Cancer Immunol Res 2014; 3: 173–83. [DOI] [PubMed] [Google Scholar]
  • 59. Yamane-Ohnuki, N, Kinoshita, S, Inoue-Urakubo, Met al.  Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol Bioeng 2004; 87: 614–22. [DOI] [PubMed] [Google Scholar]
  • 60. Niwa, R, Shoji-Hosaka, E, Sakurada, Met al.  Defucosylated chimeric anti-CC chemokine receptor 4 IgG1 with enhanced antibody-dependent cellular cytotoxicity shows potent therapeutic activity to T-cell leukemia and lymphoma. Cancer Res 2004; 64: 2127–33. [DOI] [PubMed] [Google Scholar]
  • 61. Junttila, T, Parsons, K, Olsson, Cet al.  Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res 2010; 70: 4481–9. [DOI] [PubMed] [Google Scholar]
  • 62. Robak, T. GA-101, a third-generation, humanized and glyco-engineered anti-CD20 mAb for the treatment of B-cell lymphoid malignancies. Curr Opin Investig Drugs 2009; 10: 588–96. [PubMed] [Google Scholar]
  • 63. Ishida, T, Joh, T, Uike, Net al.  Defucosylated anti-CCR4 monoclonal antibody (KW-0761) for relapsed adult T-cell leukemia-lymphoma: a multicenter phase II study. J Clin Oncol 2012; 30: 837–42. [DOI] [PubMed] [Google Scholar]
  • 64. Kubota, T, Niwa, R, Satoh, Met al.  Engineered therapeutic antibodies with improved effector functions. Cancer Sci 2009; 100: 1566–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Boyerinas, B, Jochems, C, Fantini, Met al.  Antibody-dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res 2015; 3: 1148–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Würflein, D, Dechant, M, Stockmeyer, Bet al.  Evaluating antibodies for their capacity to induce cell-mediated lysis of malignant B cells. Cancer Res 1998; 58: 3051–8. [PubMed] [Google Scholar]
  • 66. Romano, S, Moura, V, Simões, Set al.  Anticancer activity and antibody-dependent cell-mediated cytotoxicity of novel anti-nucleolin antibodies. Sci Rep 2018; 8: 7450. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Antibody Therapeutics are provided here courtesy of Oxford University Press

RESOURCES