Abstract
The classical view of therapeutic monoclonal antibodies against tumor associated antigens is that their mechanism of action is dominated by signal blocking or the cytotoxicity of Fc-driven innate immune effector functions. We review here a mounting body of evidence that anti-TAA mAbs are capable of profoundly synergizing with T cell-directed immunotherapies such as checkpoint blockade and adoptive cell therapy. Two key components account for this synergy: 1) a self-vaccinal effect mediated by dendritic cells; and 2) an inflammatory repolarization of the tumor microenvironment. Efficient exploitation of these mechanisms has tremendous therapeutic potential.
Classical anti-TAA mAb pharmacology
The pharmacological objective of killing every single tumor cell is shared by the great majority of cancer drugs, from chemotherapy to targeted kinase inhibitors. However, extended courses of drug administration usually just result in outgrowth of resistant clones [1, 2], shifting the Kaplan-Meier curve midpoint rightward without improving long-term survival. To definitively counter such an evolving pathology, an adaptive therapy is required – and natural selection has provided us a means to accomplish this, in the form of our adaptive immune system.
Although originally extracted from the adaptive humoral immune system, monoclonal antibodies against tumor-associated antigens (anti-TAA mAbs; e.g. cetuximab, trastuzumab, and rituximab) were at first conceptualized as signal blockers rather than triggers of an endogenous therapeutic immune response. It was subsequently found in mouse studies [3] and retrospective analysis of clinical experiences [4] that antibody effector function contributes significantly to anti-TAA mAb efficacy, suggesting that antibody dependent cell-mediated cytotoxicity kills sufficient numbers of tumor cells to directly account for the observed clinical efficacy.
Roles for innate antibody effector function in priming a T cell response
However, in the natural course of an infection, the innate immune response serves primarily as an early restraining action to buy time while initiating the adaptive immune response necessary for long-term protection. Along these lines, it has been proposed that anti-TAA mAbs may foster therapeutic T cell responses [5, 6]. Multiple mouse model studies suggest that anti-TAA mAbs initiate a CD8+ T cell response - that is in turn required for anti-tumor efficacy. In the examples shown in Figure 1, anti-TAA mAb monotherapies cure syngeneic tumors in mice with wild-type immune systems, but lose all efficacy upon depletion of T cells. These results are not more widely known because most mAb preclinical studies in the past have been performed with human tumor cells in mice lacking T cells. A linkage from mAb therapy to T cell responses has also been observed clinically: treatment with trastuzumab [7], cetuximab [8], or a trastuzumab antibody drug conjugate [9] all stimulate T cell responses against tumors in patients.
Figure 1.
Efficacious anti-TAA mAb therapies in syngeneic murine tumor models require the presence of T cells for their mechanism of action. Four different published examples are shown. A) Anti-HER2 mAb fails to cure tumors after depletion of CD8+ T cells [28]. B) Anti-HER2 mAb loses efficacy after anti-CD8 depletion [29]. C) Enhanced survival from anti-TAA mAb therapy plus a TLR4 ligand fails upon depletion of CD4+ and CD8+ T cells [30]. D) Efficacy of anti-TRP1 mAb and extended-lifetime IL-2 fails when CD8+ T cells are depleted [20].
How do anti-tumor antibodies drive a T cell response? Two inter-related mechanisms are supported by significant available evidence: 1) a vaccinal effect following mAb effector-mediated tumor cell killing; and 2) inflammatory reprogramming of the tumor microenvironment. The vaccinal effect results from generating a bolus of antigenic material via tumor cell death, which antigen presenting cells such as dendritic cells or macrophages then cross-present to CD8+ T cells. Analogous vaccinal effects have been shown to occur with chemotherapies that drive immunogenic cell death [10], and with external beam radiation [11]. Particular advantages of antibody-directed immunogenic tumor cell death include: absence of the inadvertent cytotoxicity of chemotherapy and radiation against the very immune effector cells essential for an immune response; efficient concentration and packaging of antibody-bound immune complexes for focused uptake by professional antigen-presenting cells; and stimulation of activating Fc gamma receptors on DCs, which has been demonstrated to potentiate the process of cross-presentation of phagocytosed antigen [12, 13]. The ability of anti-TAA mAbs to drive vaccinal stimulation of T cell responses has been demonstrated in mouse models numerous times [5, 7, 8, 14–19]. Given this extensive convergent evidence, a vaccinal effect should reasonably be assumed to comprise at least a component of the mechanism of action of any anti-TAA mAb possessing an activating isotype (e.g. human IgG1 or murine IgG2a). A particular advantage that immune complexes containing tumor cell debris have over the limited number of synthetic neoantigens in a typical cancer vaccine is the inherent personalization of such self-vaccination, which carries the potential for antigen spreading (which has been observed experimentally [19]). Such antigen spreading could enable the adaptive immune response to track antigenic changes in a genetically labile heterogeneous tumor cell population despite loss of expression of the particular antigens selected for synthetic vaccination.
A second mechanism by which anti-TAA mAbs drive T cell responses is by reprogramming of the tumor microenvironment immune contexture, with significant local increases in inflammatory cytokines and chemokines in syngeneic tumor models (Figure 2). This intratumoral “cytokine storm” is likely a direct consequence of innate effector cell activation during anti-TAA mAb-driven ADCC. Across several of our published studies, consistent and robust increases in MIP-2, G-CSF, and IL-6 in particular have been observed upon addition of an anti-TAA mAb to an immunotherapy cocktail. In one instance [20], these increases were shown to result from a complex interaction amongst NK cells, neutrophils, eosinophils, and macrophages, with macrophages directly increasing MIP-2 production in a fashion reliant on the presence of the other innate immune effector cell types. Not surprisingly, these changes are then correlated with significant intratumoral increases in essentially all types of immune cells examined [19, 20]. The chemokines MIP1α (CCL3), MIP1β (CCL4), and RANTES (CCL5) that are increased intratumorally upon anti-TAA mAb treatment (Figure 2) are among a signature of six chemokines preferentially expressed in human metastatic melanoma biopsies infiltrated with T cells [21]. Given the observed clinical correlation between tumor-infiltrating lymphocytes and response to checkpoint blockade antibodies [22, 23], it is therefore an exciting possibility that existing anti-TAA mAbs may help convert nonresponding patients to obtain benefit from checkpoint blockade antibody treatment. Indeed, we have observed significant anti-TAA mAb potentiation of adoptive cell therapy, vaccines, and immunocytokines in mouse models of cancer (Figure 3;[19, 20, 24]).
Figure 2.
Intratumoral increases in cytokines and chemokines upon addition of an anti-TAA mAb to: Fc/IL-2 (black bars, Zhu et al.); an IgG-based IL-2 immunocytokine (dark grey bars, Tzeng et al.); and combination Fc/IL-2, anti-PD1, and vaccine immunotherapy (light grey bars, Moynihan etal.). In each of these studies, numerous additional analytes were statistically significantly increased; the particular inflammatory molecules most consistently upregulated upon anti-TAA mAb treatment are excerpted and highlighted here.
Figure 3.

Three published examples where addition of an anti-TAA mAb to a T-cell-directed immunotherapy protocol significantly improved its efficacy. A) CD8+ T cell adoptive therapy (ACT) plus IL-2/Fc fusion protein extended survival, but addition of an anti-TAA mAb leads to cures of most mice [20]. B) A vaccine that potently elicits CD8+ Tcell responses, together with an anti-PD-1 antibody and an IL-2/albumin fusion protein (IPV) extends survival of tumor-bearing mice but gives no cures. Addition of an anti-TAA mAb leads to cures for the majority of mice. C) Simultaneous treatment with an anti-TAA mAb, IL-2/Fc fusion, and interferon-alpha extends survival but leads to no cures. However, delaying the interferon-alpha dose by 48 hours leads to cures of all mice, with evidence supporting an effect of improving dendritic cell cross-priming of CD8+ T cell responses [27].
How might this immune response-kindling function be best exploited with anti-TAA mAbs? This mechanism of action is so distinct from simple signaling antagonism as to demand a fundamentally different dosing perspective. In order to kindle adaptive immunity, episodic treatments might be better matched to the natural temporal sequence in an immune response than the sustained high-level exposures typical with chemotherapy or kinase inhibitors. The acute inflammatory response initiates an autonomously feedback-limited time course, terminated by synthesis of anti-inflammatory molecules such as lipid mediators hours to days after onset [25]. There is the distinct possibility that continued saturation with mAbs during the resolution phase of the inflammatory cycle could actually paradoxically lead to an immunosuppressive state of chronic inflammation [26]. Antigen presentation under such tolerizing conditions would clearly be counterproductive to the intended therapeutic effect. Perhaps anti-TAA mAb administration should be reconceptualized as a pulsatile prime or boost component of a vaccination, for which one would never contemplate perpetual saturating dosing.
An anti-TAA mAb’s effector functions generate a significant amount of tumor cell debris in the first days following a bolus dose. One therefore might expect antigen presenting cells to possess an inventory of potential tumor antigens at this time, and not considerably before or after. If one were to administer a pharmacological pulse of dendritic cell stimulation, it would make the most sense to time it so as to coincide with maximal DC antigen loading. We indeed found this to be the case for several immunostimulatory molecules [27]. Coadministration of a type-I interferon with anti-TAA mAb was ineffective against large syngeneic tumors, but delay of the interferon to two days following the mAb administration led to a majority of cures after only two cycles of treatment (Figure 3C). Of particular note, premature administration of a type-I interferon was found to significantly inhibit the immune response to a protein vaccine antigen. This same general time dependence was observed for several other DC immunostimulatory agents [27].
Concluding remarks
Approaching the pharmacology of the extensive industrial portfolios of anti-TAA mAb clinical candidates (and approved drugs) with this vaccinal/immunotherapy mindset could potentially contribute to a considerable amplification of the already impressive inroads that checkpoint blockade antibodies have made against cancer. The pipelines of pharmaceutical companies were once full of anti-RTK antibodies that gave disappointing therapeutic responses despite early promise in preclinical studies in xenografted tumors in nude mice. For example, nine different antibodies against IGF1-R (cixutumumab, ganitumab, dalotuzumab, figitumumab, teprotumumab, robatumumab, AVE1642, BIIB022, and isiratumab) have been tested in clinical trials, but none provided sufficient efficacy for FDA approval. These same anti-TAA mAbs (except figitumumab and BIIB022, which are not human IgG1 isotype) might well be repurposed to serve as innate immune kindlers of a therapeutic CD8+ T cell response, if combined with checkpoint blockade antibodies (see outstanding questions). Dozens of anti-TAA mAbs and antibody drug conjugates are being evaluated in industrial clinical trials, and a case can be made that each of these should be clinically evaluated in a prime/boost dosing schedule together with an anti-PD-1 or anti-PD-L1 antibody.
Acknowledgments
I thank Darrell Irvine, Glenn Dranoff, Stefani Spranger, Michael Birnbaum, Naveen Mehta, Noor Momin, Byong Kang, Emi Lutz, Adrienne Rothschilds, and Kelly Moynihan for helpful critical comments on this piece. Some of the work cited here was supported by NCI174795.
CD8+ T cells are essential for anti-tumor associated antigen monoclonal antibody (α-TAA mAb) efficacy in wild-type mice
α-TAA mAbs amplify the effects of T cell-based immunotherapies
α-TAA mAbs initiate self-vaccination against tumor antigens, and inflame the tumor microenvironment
Can RTK α-TAA mAbs abandoned for lack of monotherapeutic efficacy nevertheless synergize with checkpoint blockade therapy?
Will episodic pulsatile α-TAA mAb dosing schedules provide vaccinal effects superior to chronic saturating dosing?
More and better syngeneic and genetically engineered mouse models are critically needed to perform α-TAA mAb studies in the presence of an intact immune system.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Fedele C, Tothill RW, McArthur GA. Navigating the challenge of tumor heterogeneity in cancer therapy. Cancer Discov. 2014;4(2):146–8. doi: 10.1158/2159-8290.CD-13-1042. [DOI] [PubMed] [Google Scholar]
- 2.Gisselsson D, Egnell R. Cancer - an insurgency of clones. Trends in Cancer. 2017;3(2):73–75. doi: 10.1016/j.trecan.2016.11.010. [DOI] [PubMed] [Google Scholar]
- 3.Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med. 2000;6(4):443–6. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
- 4.Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol. 2003;21(21):3940–7. doi: 10.1200/JCO.2003.05.013. [DOI] [PubMed] [Google Scholar]
- 5.Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol. 2005;23(9):1147–57. doi: 10.1038/nbt1137. [DOI] [PubMed] [Google Scholar]
- 6.Ferris RL, Jaffee EM, Ferrone S. Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity, and immunoescape. J Clin Oncol. 2010;28(28):4390–9. doi: 10.1200/JCO.2009.27.6360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Taylor C, Hershman D, Shah N, Suciu-Foca N, Petrylak DP, Taub R, Vahdat L, Cheng B, Pegram M, Knutson KL, Clynes R. Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy. Clin Cancer Res. 2007;13(17):5133–43. doi: 10.1158/1078-0432.CCR-07-0507. [DOI] [PubMed] [Google Scholar]
- 8.Srivastava RM, Lee SC, Andrade Filho PA, Lord CA, Jie HB, Davidson HC, Lopez-Albaitero A, Gibson SP, Gooding WE, Ferrone S, Ferris RL. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res. 2013;19(7):1858–72. doi: 10.1158/1078-0432.CCR-12-2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Muller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, Savic S, Harbeck N, Nitz U, Gluz O, von Bergwelt-Baildon M, Kreipe H, Reddy S, Christgen M, Zippelius A. Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci Transl Med. 2015;7(315):315ra188. doi: 10.1126/scitranslmed.aac4925. [DOI] [PubMed] [Google Scholar]
- 10.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
- 11.Tang C, Wang X, Soh H, Seyedin S, Cortez MA, Krishnan S, Massarelli E, Hong D, Naing A, Diab A, Gomez D, Ye H, Heymach J, Komaki R, Allison JP, Sharma P, Welsh JW. Combining radiation and immunotherapy: a new systemic therapy for solid tumors? Cancer Immunol Res. 2014;2(9):831–8. doi: 10.1158/2326-6066.CIR-14-0069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dhodapkar KM, Krasovsky J, Williamson B, Dhodapkar MV. Antitumor monoclonal antibodies enhance cross-presentation ofcCellular antigens and the generation of myeloma-specific killer T cells by dendritic cells. J Exp Med. 2002;195(1):125–33. doi: 10.1084/jem.20011097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kim PS, Armstrong TD, Song H, Wolpoe ME, Weiss V, Manning EA, Huang LQ, Murata S, Sgouros G, Emens LA, Reilly RT, Jaffee EM. Antibody association with HER-2/neu-targeted vaccine enhances CD8 T cell responses in mice through Fc-mediated activation of DCs. J Clin Invest. 2008;118(5):1700–11. doi: 10.1172/JCI34333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Abes R, Gelize E, Fridman WH, Teillaud JL. Long-lasting antitumor protection by anti-CD20 antibody through cellular immune response. Blood. 2010;116(6):926–34. doi: 10.1182/blood-2009-10-248609. [DOI] [PubMed] [Google Scholar]
- 15.Wahlin BE, Sundstrom C, Holte H, Hagberg H, Erlanson M, Nilsson-Ehle H, Linden O, Nordstrom M, Ostenstad B, Geisler CH, de Brown PN, Lehtinen T, Maisenholder M, Tierens AM, Sander B, Christensson B, Kimby E. T cells in tumors and blood predict outcome in follicular lymphoma treated with rituximab. Clin Cancer Res. 2011;17(12):4136–44. doi: 10.1158/1078-0432.CCR-11-0264. [DOI] [PubMed] [Google Scholar]
- 16.Carmi Y, Spitzer MH, Linde IL, Burt BM, Prestwood TR, Perlman N, Davidson MG, Kenkel JA, Segal E, Pusapati GV, Bhattacharya N, Engleman EG. Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature. 2015;521(7550):99–104. doi: 10.1038/nature14424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.DiLillo DJ, Ravetch JV. Differential Fc-Receptor Engagement Drives an Anti-tumor Vaccinal Effect. Cell. 2015;161(5):1035–45. doi: 10.1016/j.cell.2015.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Reilly RT, Machiels JP, Emens LA, Ercolini AM, Okoye FI, Lei RY, Weintraub D, Jaffee EM. The collaboration of both humoral and cellular HER-2/neu-targeted immune responses is required for the complete eradication of HER-2/neu-expressing tumors. Cancer Res. 2001;61(3):880–3. [PubMed] [Google Scholar]
- 19.Moynihan KD, Opel CF, Szeto GL, Tzeng A, Zhu EF, Engreitz JM, Williams RT, Rakhra K, Zhang MH, Rothschilds AM, Kumari S, Kelly RL, Kwan BH, Abraham W, Hu K, Mehta NK, Kauke MJ, Suh H, Cochran JR, Lauffenburger DA, Wittrup KD, Irvine DJ. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med. 2016;22(12):1402–1410. doi: 10.1038/nm.4200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zhu EF, Gai SA, Opel CF, Kwan BH, Surana R, Mihm MC, Kauke MJ, Moynihan KD, Angelini A, Williams RT, Stephan MT, Kim JS, Yaffe MB, Irvine DJ, Weiner LM, Dranoff G, Wittrup KD. Synergistic innate and adaptive immune response to combination immunotherapy with anti-tumor antigen antibodies and extended serum half-life IL-2. Cancer Cell. 2015;27(4):489–501. doi: 10.1016/j.ccell.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69(7):3077–85. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12(4):298–306. doi: 10.1038/nrc3245. [DOI] [PubMed] [Google Scholar]
- 23.Spranger S. Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int Immunol. 2016;28(8):383–91. doi: 10.1093/intimm/dxw014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tzeng A, Kwan BH, Opel CF, Navaratna T, Wittrup KD. Antigen specificity can be irrelevant to immunocytokine efficacy and biodistribution. Proc Natl Acad Sci U S A. 2015;112(11):3320–5. doi: 10.1073/pnas.1416159112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6(12):1191–7. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
- 26.Shalapour S, Karin M. Immunity, inflammation, and cancer: an eternal fight between good and evil. J Clin Invest. 2015;125(9):3347–55. doi: 10.1172/JCI80007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tzeng A, Kauke MJ, Zhu EF, Moynihan KD, Opel CF, Yang NJ, Mehta N, Kelly RL, Szeto GL, Overwijk WW, Irvine DJ, Wittrup KD. Temporally Programmed CD8alpha+ DC Activation Enhances Combination Cancer Immunotherapy. Cell Rep. 2016;17(10):2503–2511. doi: 10.1016/j.celrep.2016.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stagg J, Loi S, Divisekera U, Ngiow SF, Duret H, Yagita H, Teng MW, Smyth MJ. Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci U S A. 2011;108(17):7142–7. doi: 10.1073/pnas.1016569108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O, Yang X, Sattar H, Wang Y, Brown NK, Greene M, Liu Y, Tang J, Wang S, Fu YX. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18(2):160–70. doi: 10.1016/j.ccr.2010.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang S, I, Astsaturov A, Bingham CA, McCarthy KM, von Mehren M, Xu W, Alpaugh RK, Tang Y, Littlefield BA, Hawkins LD, Ishizaka ST, Weiner LM. Effective antibody therapy induces host-protective antitumor immunity that is augmented by TLR4 agonist treatment. Cancer Immunol Immunother. 2012;61(1):49–61. doi: 10.1007/s00262-011-1090-7. [DOI] [PMC free article] [PubMed] [Google Scholar]


