Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 May 1.
Published in final edited form as: Expert Opin Biol Ther. 2016 Feb 25;16(5):655–674. doi: 10.1517/14712598.2016.1152256

Enhancing the safety of antibody-based immunomodulatory cancer therapy without compromising therapeutic benefit: Can we have our cake and eat it too?

Expert Opinion on Biological Therapy

Joseph M Ryan *, Jeffrey S Wasser #, Adam J Adler *, Anthony T Vella *
PMCID: PMC4955764  NIHMSID: NIHMS794649  PMID: 26855028

Abstract

Introduction

Monoclonal antibodies (mAbs) targeting checkpoint inhibitors have demonstrated clinical benefit in treating patients with cancer and have paved the way for additional immune-modulating mAbs such as those targeting costimulatory receptors. The full clinical utility of these agents, however, is hampered by immune-related adverse events (irAEs) that can occur during therapy.

Areas covered

We first provide a general overview of tumor immunity, followed by a review of the two major classes of immunomodulatory mAbs being developed as cancer therapeutics: checkpoint inhibitors and costimulatory receptor agonists. We then discuss therapy-associated adverse events. Finally, we describe in detail the mechanisms driving their therapeutic activity, with an emphasis on interactions between antibody fragment crystallizable (Fc) domains and Fc receptors (FcR).

Expert Opinion

Given that Fc-FcR interactions appear critical in facilitating the ability of immunomodulatory mAbs to elicit both therapeutically useful as well as adverse effects, the engineering of mAbs that can effectively engage their targets while limiting interaction with FcRs might represent a promising future avenue for developing the next generation of immune-enhancing tumoricidal agents with increased safety and retention of efficacy.

Keywords: Accessory cell, adverse events, antibody, antitumor, biologic, cancer, checkpoint inhibitor, costimulatory agonist, cytotoxic, effector, efficacy, Fc, Fc receptor, FcR, immunomodulatory, immunosuppression, immunotherapy, irAE, mAb, monoclonal, side effects, tolerance, toxicity, tumor microenvironment, tumoricidal

1. Introduction

There is an extensive list of drugs and treatment options at the disposal of clinical oncologists. Surgery, chemotherapy, and radiation have long been the standard of care therapies for most cancers. Nevertheless, surgery cannot eliminate highly metastatic disease, and chemotherapy and radiation cause significant side effects due to their inability to specifically target malignant cells. Furthermore, tumors that may initially respond to therapy often develop resistance, leading to disease recurrence.

The identification of genetic mutations common to particular types of cancer has facilitated the development of targeted therapies, such as the tyrosine kinase inhibitor imatinib, which is highly effective in chronic myelogenous leukemia (CML) patients harboring the corresponding Bcr-Abl fusion gene mutant 1. Monoclonal antibodies (mAbs) are also used in cancer treatment. These mAbs can be categorized into two groups based on their function. Tumor-targeting mAbs function within the tumor microenvironment (TME), where they bind directly to tumor cells or stromal trophic factors 2. The vascular endothelial growth factor (VEGF) mAb inhibitor bevacizumab, for example, interferes with survival-promoting signaling pathways important to tumor progression 3, 4. The anti-CD20 mAb rituximab works by a different mechanism, first binding to tumor cells (as well as to some healthy cells) expressing surface CD20, then engaging innate effector mechanisms for elimination 58. Tumor-targeting mAbs can also be chemically linked to radioactive or toxic entities so that the cytotoxic payload is selectively delivered to tumor cells without harming healthy tissue 9, 10.

The second group of mAbs modulate the immune system and work by promoting tumoricidal activity in a variety of immune effector cells by lowering their thresholds for initiating antitumor responses 11. Tumors are generally considered to be non-immunogenic since they arise from the body’s own cells 12; thus these mAbs are in essence “breaking” peripheral tolerance to self. These immunomodulatory mAbs can work by either antagonizing cell surface receptors that transmit inhibitory signals (i.e., checkpoint inhibitors) or, alternatively, activating stimulatory receptors (i.e., costimulatory agonists). The net effect in both cases is an enhancement of tumoricidal immune effector cell activity. This review describes the antitumor efficacies and the associated adverse events seen with antibody-based immunomodulatory cancer therapies and then considers if it might be possible to optimize their design in order to limit adverse events without compromising therapeutic benefit.

2. Tumor immunity

The immune system constantly surveys the body for potential threats, such as pathogens, with the goal of neutralizing them to maintain homeostasis. The T lymphocyte (T cell) compartment of the adaptive immune system is particularly effective at employing this “search and destroy” strategy. Individual T cells possess exquisite specificities endowed by the expression of a unique T cell receptor (TCR) capable of recognizing distinct peptides presented by major histocompatibility complex (MHC) molecules on the surface of cells. Due to recombinations between V, D, and J gene segments that comprise the TCR, the diversity of specificities is immense, allowing for the recognition of virtually any antigen 13, 14. Given the potential for many of these TCRs to recognize self-antigens, a variety of tolerance mechanisms must be employed, operating both centrally during thymic development 15, 16, as well as in the periphery 1720, to prevent autoimmunity. Consequently, tumor cells, are more difficult to detect since the majority of their antigens overlap with those expressed in their healthy progenitors to which T cells have already been tolerized. In some cases, vaccination against these non-mutated tumor-associated antigens (TAAs) can elicit effector T cells that simultaneously mediate tumor immunity and autoimmunity 2123. Alternatively, the genetic instability characteristic of malignant cells 24, 25 gives rise to mutations in protein-coding genes, resulting in the formation of tumor-specific (neo)-antigens (TSAs) that can be targeted by T cells without causing autoimmune disease 26, 27.

Naïve T cells are activated when they encounter cognate peptides complexed to MHC molecules on the surface of antigen-presenting cells (APCs). This event, referred to as “signal 1”, is the first of three sequential signals required to program proliferation, differentiation, and survival. Dendritic cells (DCs) represent the most potent APC population, in part owing to their efficiency in capturing and processing antigens into peptide fragments suitable for MHC presentation. Upon sensing pathogen-associated molecular patterns (PAMPs), DCs are also induced to express costimulatory ligands and pro-inflammatory cytokines, delivering signals 2 and 3, respectively 2830. Costimulation occurs in two waves. First, the immunoglobulin (Ig) superfamily molecules CD80 (B7.1) and CD86 (B7.2) on DCs ligate CD28 on the antigen-responding T cells to facilitate the initial phase of activation 3133. Tumor necrosis factor receptor (TNFR) superfamily members then deliver a second costimulatory wave. This process fine-tunes T cell responses by regulating the magnitude of clonal expansion, determining which particular cytokines the T cells become capable of secreting, and dictating whether long-lived memory is established 34, 35. The cytokines that constitute signal 3 further contribute to fine-tuning the T cell response 36.

In the absence of inflammation, however, steady-state DCs express only minimal amounts of costimulatory ligands and cytokines. Under these conditions, T cells encountering cognate antigens (e.g., self-antigens acquired from healthy tissues by DCs) undergo an abortive/tolerogenic response 3739. This process not only serves to prevent autoimmunity mediated by self-reactive T cells that escape negative selection in the thymus 40, but also can dampen tumor immunity by tolerizing T cells specific for TAAs 23, 41 and TSAs 42, 43. A critical factor in programming effective antitumor T cell responses is the amount and variety of costimulation and cytokines accompanying DC presentation of tumor epitopes. The PAMPs that help facilitate the potent activation of DCs during infection are generally absent from tumor sites, often resulting in a tolerogenic outcome (as described above). Nevertheless, productive T cell responses to tumor antigens do occur in some cases 4446. DC activation can be achieved when tumor cells undergoing certain forms of programmed death 47 release damage-associated molecular patterns (DAMPs), such as heat shock proteins 48, uric acid 49 and HMGB1 50, which can act in a manner analogous to that of PAMPs.

Even when tumor-reactive T cells are successfully primed, with the capacity to migrate into tumors and kill target cells, the efficiency and overall impact of the response can be inadequate. In a process termed “immunoediting”, the most immunogenic tumor cell variants are eliminated during the initial stages of tumorigenesis, while less immunogenic tumor cells are spared and gradually expand 51. Whether T cells become tolerant of tumor antigens at the earliest stages of tumorigenesis due to a lack of DC activation, or alternatively following a period of immunoediting, clinically detectable tumors are inherently non-immunogenic. Furthermore, the resulting non-immunogenic tumor engages a variety of powerful immunosuppressive mechanisms that can thwart the activity of infiltrating effector T cells and foster a tumor-favorable microenvironment. For example, the recruitment of regulatory T cells (Tregs) 5255 and myeloid-derived suppressor cells (MDSCs) 56 establishes an immune barrier around tumor sites, effectively protecting malignant cells from immune intrusion. Additional mechanisms include the secretion of immune-regulating cytokines such as transforming growth factor beta (TGF-β) 57, 58 and enzymes such as indoleamine 2,3-dioxygenase (IDO) 59, all of which are capable of altering the immune response within the TME.

An understanding of the mechanisms underlying tumor-induced immunosuppression is paramount to the development of effective cancer therapies, and as such has become a major focus of investigation; a comprehensive review on the subject is provided by Rabinovich and colleagues 60. To be effective, T cell-based immunotherapies will likely need to compensate for insufficiencies during the initial phase of T cell priming, as well as confer the capacity to overcome potent immunosuppression within the tumor microenvironment. Therefore, the most effective cancer therapies will likely consist of multiple agents targeting a variety of cancer-promoting pathways and mechanisms. Combination therapies have already begun to showcase their clinical potential, in some cases demonstrating enhanced efficacy and superiority over standard of care monotherapies. Although combining multiple agents can cause more frequent and severe adverse events, increases in efficacy may exceed increases in toxicity, suggesting combination approaches as one possibility for improving safety without sacrificing therapeutic benefit.

3. Checkpoint inhibitors

The immune system prevents excessive inflammation or autoimmunity via the engagement of inhibitory feedback networks. A commonly used analogy equates the immune system to an automotive vehicle with the antigen-stimulated TCR representing the transmission in drive, CD28 costimulatory receptor acting as the accelerator, the TNFR costimulators functioning as turbo boosters, and checkpoint inhibitory receptors serving as the brake.

Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) was the first checkpoint receptor identified and is the most extensively studied. Following the initial activation of naïve T cells, in which DCs provide signal 1 (MHC-peptide) along with signal 2 in the form of B7.1 and B7.2 ligating CD28, intracellularly stored CTLA-4 is relocated to the T cell surface where it competes with CD28 for binding to B7.1 and B7.2 61, thereby helping to avoid excessive CD28-mediated stimulation. Upon ligation, CTLA-4 engages the protein phosphatases SHP2 and PP2A, which deactivate signaling proteins downstream from TCR engagement 62. The net effect is the prevention of T cell hyper-responsiveness 6367.

Mice harboring a null mutation in the gene encoding CTLA-4 develop fatal lymphoproliferative disease characterized by autoreactive T cell infiltration and extensive tissue destruction 68. This dramatic phenotype results not only from the predicted effect of releasing the brakes on self-reactive T cells, but also from impairment in the function of Tregs, which constitutively express surface CTLA-4 to maintain tolerance and suppress auto-reactivity 19, 6973. That CTLA-4 functions on multiple cell types to maintain immune homeostasis raised the critical question of whether it could be targeted therapeutically without unleashing fatal adverse events. Nevertheless, preclinical studies using a variety of mouse tumor models demonstrated that a blocking mAb to CTLA-4 altering the activities of both tumor-reactive effector T cells and Tregs can produce therapeutic benefit, albeit with toxicity 7478.

This work paved the way for the development of a humanized CTLA-4 antagonist, ipilimumab 79. A decade of clinical trials culminated in an international phase III trial that led to its 2011 approval by the FDA for treating melanoma, making it one of the first immune-based biologic therapies, and the first checkpoint inhibitor, to be used clinically 80. In this study, 676 patients with late-stage melanoma were given either ipilimumab, a vaccine targeting the melanoma TAA gp100, or ipilimumab plus vaccine. Objective response rates (ORR) for ipilimumab alone and gp100 alone were 10.9% and 1.5%, respectively. Importantly, ipilimumab (with or without vaccine) boosted median overall survival (OS) by 10 months, compared to 6.4 months for gp100 vaccination alone (Table 1). These encouraging results earned for ipilimumab its approval for the treatment of unresectable or metastatic melanoma.

Table 1.

Summary of clinical results for representative immunomodulatory mAbs used to treat cancer

Checkpoint Inhibitors Costimulatory Agonists
Target CTLA-4 PD-1 PD-L1 CD40 CD134 (OX40) CD137 (4-1BB)
Agent* Ipilimumab Nivolumab Atezolizumab CP-870,893** MEDI6469**** Urelumab
mAb Isotype IgG1 IgG4 IgG1 IgG2 murine IgG1 IgG4
Company Bristol-Myers Squibb Bristol-Myers Squibb Genentech/Roche Pfizer/Roche MedImmune/AstraZeneca Bristol-Myers Squibb
Highest Phase of Testing Approved/Marketed Approved/Marketed Phase III Phase I Phase I/II Phase II
Marketed/registered Indications Malignant melanoma Malignant melanoma, NSCLC, RCC - - - -
Phase III Indications Glioblastoma, NSCLC, RCC, SCLC, prostate Glioblastoma, gastric, HNC, SCLC Bladder, breast, NSCLC, RCC - - -
Phase II Indications Gastric, MDS, esophageal, ovarian, UGC, lymphoma, brain metastases AML, CLL, breast, DLBCL, MDS, UGC, esophageal, ovarian, follicular and Hodgkin’s lymphomas CRC, solid tumors, soft tissue sarcoma - - CLL, malignant melanoma
Phase I/II Indications Solid tumors CRC, HCC, NHL, solid tumors, hematological malignancies DLBCL, follicular lymphoma - Breast, DLBCL, prostate cancer, solid tumors NHL, solid tumors
Phase I Indications - CML, hepatitis C Malignant melanoma, MM, MDS, hematological malignancies PDA, solid tumors CRC, HNC MM, CRC, HNC
Efficacy ORR 10.9%, phase III, stage III/IV melanoma vs 1.5% gp100 alone OS=10.1 months vs 6.4 (gp100 alone) 80
ORR 19.0%, phase III, stage III/IV melanoma 106
ORR 40.0%, phase III, stage III/IV melanoma, vs 13.9% for dacarbazine 103
ORR 43.7%, phase III, stage III/IV melanoma 106
ORR 38%, phase II, advanced NSCLC, PD-L1+; compared to 13% for docetaxel 262 ORR 20% (6 PR), phase I, with chemothera py, solid tumors 138 No PR, but tumor shrinkage in 40% (12 of 30) following single dose; induction of HAMA prevented additional doses being given 140*** ORR 5.6% (3 of 54), phase I, melanoma; phase II study terminated due to hepatotoxicity ; newer trials using lower dosages 141
Adverse Events Toxicities generally immune-related; rash, colitis, diarrhea, hepatotoxicity, endocrinopathies, neuropathies 80, 106, 263 Generally more tolerable than ipilimumab; fatigue, rash, diarrhea, pruritis, pneumonitis, fever can occur 100, 101, 106 Toxicities similar to those seen with anti-PD-1; grades 3/4 adverse events have been uncommon 262 Fatigue (81%) most common; does-limiting grade 3 CRS, grade 4 TIA 138 Well-tolerated, brief grade 3 lymphopeni a, fatigue, fevers, chills, mild rash observed 140*** Cytopenias and liver inflammation (elevated ALT/AST) most concerning, dose dependent; also fatigue, rash, fever 141

Information on clinical trials obtained from ClinicalTrials.gov.

*

Particular agents listed here are only representative; there are a number of other agents to the same and/or other targets in various stages of development.

**

Pfizer’s CP-870,893 is now licensed to Roche and has been renamed RO7009789; ongoing clinical trials are listed under RO7009789.

***

Efficacy and adverse events based on phase I testing of a different OX40 agonist, 9B12.

****

MedImmune/AstraZeneca has recently begun recruiting for a phase I trial with a humanized anti-CD134 agonist (MEDI0562) in adults with selected advanced solid tumors (NCT02318394).

NSCLC, non-small cell lung carcinoma; RCC, renal cell carcinoma; SCLC, small cell lung carcinoma; MDS, myelodysplastic syndromes; UGC, urogenital carcinoma; HNC, head and neck cancers; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; DLBCL, diffuse large B cell lymphoma; NHL, non-Hodgkin’s lymphoma; CML, chronic myeloid leukemia; CRC, colorectal cancer; MM, multiple myeloma; PDA, pancreatic ductal adenocarcinoma; CRS, cytokine release syndrome; TIA, transient ischemic attack; ORR, objective response rate; PR, partial response; HAMA, human anti-mouse antibodies.

Another checkpoint molecule, programmed cell death protein 1 (PD-1), is expressed primarily on already-activated effector T cells and regulates their activity in the periphery 81, 82. Upon binding of its ligands, PD-L1 83 or PD-L2 84, PD-1 engages phosphatases that inactivate signaling molecules downstream of the TCR to limit proliferation, survival, and cytokine production 85. PD-1 is highly expressed on CD8+ T cells residing within the TME 86. PD-L1 can be constitutively expressed by tumor cells 87, 88 due to oncogenic signaling pathway aberrations 89, 90 or induced in response to localized T cell secretion of cytokines (e.g., interferons) 9193 in a process referred to as adaptive immune resistance 94. Tumor-infiltrating myeloid cells and lymphocytes can express PD-L1, and their co-localization and interaction with PD-1-expressing cells further define PD-1-mediated activity within the TME 95, 96. PD-1 also plays a role in Treg function 97, and thus, similar to CTLA-4 blockade, targeting the PD-1/PD-L axis may enhance tumoricidal capacity through effects on both effector and regulatory T cells. Indeed, preclinical testing in mouse models demonstrated that mAb-mediated disruption of PD-1/PD-L1 interaction can boost tumor immunity 98, 99.

Two humanized antagonist mAbs targeting PD-1, pembrolizumab and nivolumab, have been approved after yielding significant therapeutic responses in phase III clinical trials 100102. A double-blind phase III trial of 418 patients receiving either nivolumab or dacarbazine found ORR of 40.0% and 13.9%, respectively, and 1 year OS rates of 72.9% and 42.1%, respectively 103 (Table 1). Response to treatment has also been shown to correlate with tumor expression of PD-L1 100. Additional clinical data show similarly encouraging therapeutic responses in patients treated with anti-PD-L1 mAb 104, and provide preliminary evidence in support of using intratumoral PD-L1 expression as a biomarker to determine likelihood of therapeutic success on a patient-to-patient basis 95, 96, 105, 106.

Checkpoint-inhibiting biologics are now being used routinely to treat patients with malignant melanoma, and numerous ongoing clinical studies raise the likelihood that they will soon be approved for treating patients with a variety of cancers including kidney, bladder, prostate, and lung 79. Recently, nivolumab was approved for additional indications including non-small cell lung carcinoma (NSCLC) and renal cell carcinoma (RCC). Thus, in addition to the durability of treatment response, the potential for use in a number of different clinical conditions sets these mAbs apart from other oncologic agents. As shown in Table 1, checkpoint inhibitors and costimulatory agonists have demonstrated efficacy in both solid tumors and hematologic malignancies 107. Particularly noteworthy is their efficacy in solid tumors, which can be especially difficult to treat due to the existence of localized immunosuppression in the TME 5259. Intriguingly, immunomodulatory mAbs enable antitumor activities that are relatively nonspecific with respect to cancer subsets, but relatively specific in terms of preferential targeting of cancerous cells while for the most part sparing healthy cells, in essence, approaching the “holy grail” of cancer therapy.

mAbs blocking both CTLA-4 and PD-1/PD-L1, enhance tumor immunity by disrupting T cell inhibitory signaling. However, they act during distinct early (CTLA-4) and late (PD-1) phases of the anti-tumor response. Thus, the possibility is raised that a combination therapy might be superior to either agent alone. Improved response has been confirmed in mice 108, and human data obtained thus far have been encouraging. In one study the combination of nivolumab and ipilimumab given concurrently yielded objective clinical responses 109 exceeding those reported previously for either monotherapy 80, 100, although direct comparison was not made. While objective-response rates may not predict long-term benefit to patients, and survival data are needed, these early studies suggest a potential advantage of combination treatment strategies 109.

More recently, a phase III study 106 comparing nivolumab alone, ipilimumab alone, and combination of the two in previously-untreated metastatic melanoma (major findings highlighted in Table 2), demonstrated greater progression-free survival (PFS) for nivolumab, alone or in combination (both 14.0 months), compared with ipilimumab alone (3.9 months). Notably, in the context of PD-L1-positive tumors, the addition of ipilimumab to nivolumab did not improve PFS but did increase the overall rates of grades 3/4 adverse events, from 43.5% to 68.7%. In this case it would appear that combination therapy is not as safe as nivolumab monotherapy (Table 2). However, combination therapy proved superior efficacy to either monotherapy in the context of PD-L1-negative tumors (Median PFS of 11.2 months, compared to 5.3 months and 2.8 months for nivolumab and ipilimumab, respectively). Although the advantage over nivolumab monotherapy dissipated by 16 months of treatment (not shown in Table 2), these data suggest the possibility of value in combined therapy regimens 106. Although this study showed no evidence of synergy, the additive effects of combination therapy seen with PD-L1-negative tumors provides some evidence that this multi-pronged approach may offer unique advantages over single-agent therapies in some tumor types. Nonetheless, definitive conclusions from this study regarding the value of supplementing nivolumab with ipilimumab cannot be drawn until overall survival data is obtained.

Table 2.

Combination immunotherapy with ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) in previously-untreated metastatic melanoma.

This table represents an abbreviated version of findings from the randomized, double-blind, phase 3 study 106 deemed most important

Treatment* Ipilimumab Nivolumab Both
Efficacy ORR** 19.0% 43.7% 57.6%
Median PFS (Intention-to-treat) 2.9 months 6.9 months 11.5 months
Median PFS (PD-L1-positive) 3.9 months 14.0 months 14.0 months
Median PFS PD-L1-negative 2.8 months 5.3 months 11.2 months
Adverse events Rate of any adverse event 99% 99.4% 99.7%
Severe*** 55.6% 43.5% 68.7%
*

Number of patients per group (n) ranged from 311–316.

**

Response rates determined by the investigator using Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1.

***

Defined as grades 3/4 based on severity determined using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), version 4.0.

ORR, objective response rate; PFS, progression-free survival.

4. Costimulatory agonists

Agonist mAbs targeting costimulatory receptors on antigen-primed T cells represent the second major category of immunomodulatory antibodies. Primarily targeting receptors of the TNFR superfamily that provide the second wave of signal 2, these antibodies are designed to mimic the activity of endogenous costimulatory ligands whose availability tends to be low in the context of tumor antigen presentation. In contrast to Ig superfamily costimulatory receptors such as CD28 that modulate activity of signaling proteins located proximally to the TCR, TNFR superfamily costimulatory receptors engage various combinations of TNFR-associated factors (TRAFs) 1–6, which control several downstream signaling pathways that include NF-κB and MAP kinase 34, 35.

CD40 is a TNFR family member expressed on DCs that, when bound by its ligand CD40L (CD154) expressed on cognate CD4+ helper T cells, induces expression of MHC molecules, costimulatory ligands (B7.1 and B7.2) and cytokines such as IL-12, which collectively function to boost the response of CD8+ cytotoxic T lymphocytes (CTL) 110113. A CD40 agonist mAb can thus activate DCs when CD4+ T cell help is unavailable and lead to robust effector T cell responsiveness and antitumor immunity 114116.

Agonists can also directly target TNFR costimulatory receptors on T cells. For instance, CD134 (OX40) expressed on T cells following TCR stimulation 117, 118 can be ligated by OX40L (CD252) expressed on activated, but not steady state, DCs 119 to program T cell effector differentiation, survival, and memory 118, 120122. CD134 agonist thus enables antigen-primed T cells to develop potent effector and tumoricidal capacity under otherwise tolerogenic (i.e., steady state) conditions 123130. Similarly, T cell responsiveness and tumor immunity in mice can also be elicited with agonists to several other T cell TNFR family costimulatory receptors such as CD137 (4-1BB) 131, glucocorticoid-induced TNFR-related protein (GITR) 132 and CD27 133.

Extensive mouse studies characterizing the therapeutic potential of TNFR agonists have fueled ongoing efforts to develop humanized therapeutics. Although clinical testing of these agents has lagged behind that of the checkpoint inhibitors by several years, initial results for costimulatory agonists have been encouraging. For instance, a phase II trial evaluating the CD40 agonist dacetuzumab for use in the treatment of relapsed diffuse large B-cell lymphoma demonstrated clinical benefit in a subset of patients 134, complementing previous data from phase I testing of this agent in other cancers, which found only moderate toxicities associated with therapy 135137. For example, one study demonstrated that out of 44 patients with multiple myeloma treated with dacetuzumab, adverse events of mild cytokine release syndrome, non-infectious ocular inflammation, and elevated liver enzymes were infrequent and well tolerated, with less than 10% experiencing serious toxicities 137. Additional phase I trials evaluating dacetuzumab in other cancers such as non-Hodgkin’s lymphoma (NHL) 135 and chronic lymphocytic leukemia (CLL) 136 have generated similar results.

CP-870,893, a fully-human IgG2 mAb considered to be a more potent anti-CD40 agonist, has yielded relatively encouraging efficacy data, including a phase I ORR of 20% in combination with carboplatin and paclitaxel to treat various advanced solid tumors 138 (Table 1) and another phase I ORR of 19% with 4 partial responses (PRs) in combination with gemcitabine to treat advanced pancreatic ductal adenocarcinoma (PDA) 139, with generally tolerable side effects. CP-870,893 has since been licensed to Roche and is now, under the name RO7009789, undergoing phase I testing in patients with locally advanced and/or metastatic solid tumors and newly diagnosed resectable PDA.

Alternatively, a mouse mAb agonist specific for human CD134 given in a phase I trial to patients with various advanced cancers elicited objective clinical responses (regression of at least one tumor nodule) in 12 out of 30 patients 140 (Table 1). Although none of the patients demonstrated responses according to Response Evaluation Criteria in Solid Tumors (RECIST, tumor shrinkage of at least 30%), the fact that these results were achieved using a murine mAb that could only be given in a single course (due to the production of human anti-mouse antibodies (HAMA) in patients) is highly encouraging as it suggests that fully humanized CD134 agonists might elicit significant tumor regression, especially if given in multiple courses. A humanized CD137 agonist urelumab has also demonstrated therapeutic activity in phase I testing 141, although liver toxicities led to the termination of phase II testing (Table 1). It was later determined that the toxicities that had occurred were dose-dependent, and careful dosing of the agent could avoid liver damage. Humanized agonists to other TNFR members are currently in various stages of clinical development 142144.

Given the possibility that a variety of humanized TNFR agonists may gain FDA approval for the treatment of human cancer patients, it would be useful to consider if they might be combined to improve therapeutic efficacy, like the checkpoint inhibitors 108, 109. The rationale for combining agonists targeting different TNFR family members stems from their non-identical tissue expression patterns as well as the partial (but not complete) overlap in the signaling pathways they engage. These subtle differences likely reflect the potential of each member to uniquely contribute to the fine-tuning of T cell responses needed to optimally handle particular immunogenic challenges 34, 35. In a clinical setting, it might be possible to exploit this variation by combining particular TNFR agonists to tailor optimal antitumor responses 145. For instance, co-administration of agonists to CD134 and CD137 146150 or death receptor 5 (DR5, also known as TRAIL receptor 5) plus CD40 and CD137 151 can be synergistic in programming T cell clonal expansion and effector function enabling the control of tumor growth in mice. Effective combination strategies might also involve the administration of a checkpoint inhibitor with a TNFR agonist, as has been demonstrated with anti-CTLA-4 plus anti-CD137 152 and also with anti-PD-1 plus anti-CD134 153.

5. Adverse events

The elicitation of adverse events is a risk inherent to virtually any type of therapy. With immune-based therapies, adverse events can potentially involve several mechanisms, which vary depending on the particular nature of therapy. For example, therapeutically-administered cytokines can give rise to nonspecific, systemic immune reactivity due to their widespread sites of action, whereas other immunotherapies such as adoptive cell therapies, tumor vaccines, and immunomodulatory antibodies tend to trigger more localized tissue destruction due to their more targeted and specific immune cell activation 154. With immunomodulatory mAbs, tumoricidal effector T cells that recognize non-mutated TAAs can destroy healthy cells expressing that same TAA. The classic example of this type of immune-related adverse event (irAE) is vitiligo, in which T cells primed to non-mutated melanocytic antigens destroy both melanoma cells and healthy melanocytes 22, 155. While vitiligo may be an acceptable trade-off for clearing aggressive melanomas (and other cancer types), autoimmune disease obviously would be more problematic when treating tumors derived from vital organs.

Another mechanism leading to irAEs is the disengagement of inhibitory pathways critical for maintaining tolerance to self and immune homeostasis. For instance, since part of the overall mechanism by which ipilimumab boosts tumor immunity involves neutralizing the function of Tregs 76, 78, it might be expected that ipilimumab-treated patients would experience irAEs that would otherwise be kept in check by Treg oversight. Consistent with this prediction, in the landmark melanoma phase III clinical trial mentioned previously, 60% of ipilimumab-treated patients experienced irAEs, 10–15% of which were severe (grades 3/4) that included diarrhea, vitiligo, colitis, and endocrinopathies, as well as infusion reactions 80 (Table 1). Careful examination of the specific mechanism(s) involved for individual antibodies (shown for a representative sample of mAbs in Table 4) can also shed light on how specific toxicities arise due to therapy. For example, because anti-CTLA-4 works, in part, by depleting Tregs and/or altering their function, the occurrence of diarrhea, colitis, and other gastrointestinal (GI) manifestations can be attributed to the abundant Treg populations located in the GI tract and their critical role in maintaining immune tolerance 156. Tissue-specific and antibody-specific adverse events are discussed in greater depth elsewhere 11.

Table 4.

Characterizing potential Fc-FcR interactions required for efficacy and/or adverse events of representative immunomodulatory targets

FcR Requirements for Biological Functions FcR Independent FcR Dependent
mAb Class mAb Target Block ligand binding Counter-regulatory Signaling* Activate Costim. Deplete/Alter Tregs in TME Deplete/Alter Myeloid Cells in TME Tumor Depletion/Apoptosis Additional comments
Checkpoint Inhibitors CTLA-4 Yes Yes - Yes - -
PD-1 Yes Yes - - - - FcR engagement diminishes efficacy
PD-L1 Yes - - - Yes Yes**
Costimulatory Agonists CD40 - - Yes - - Yes** FcγRIIB-dependent***
CD134 (OX40) - - Yes Yes - - Treg depletion due to ADCC and/or phenotypic conversion
D137 (4-1BB) - - Yes Yes - - FcγRIII diminishes efficacy; FcγRIIB-dependent***
GITR - - Yes Yes - - Treg depletion due to ADCC and/or phenotypic conversion
PARA DR5 - - - - - Yes FcγRIIB-dependent****
CD95 (Fas) - - - - - Yes FcγRIIB-dependent****

Mechanisms that occur only during particular circumstances are italicized.

*

Possibly FcR-independent; not yet characterized.

**

Some tumors cells can express surface this target.

***

Can be activating FcR-dependent and/or FcR-independent in certain contexts.

****

Can be activating FcR-dependent in specific circumstances.

Fc, fragment crystallizable; FcR, Fc receptor; mAb, monoclonal antibody; Treg, regulatory T cell; TME, tumor microenvironment; ADCC, antibody-dependent cell-mediated cytotoxicity; PARA, pro-apoptotic receptor agonists.

In contrast, irAEs caused by targeting the PD-1 axis tend to be fewer in frequency and less severe 100, 101, 104. Early human testing revealed tolerability of adverse events in addition to impressive efficacy, with response rates ranging from 18–27% when used to treat NSCLC, RCC, and melanoma, with grades 3/4 adverse events occurring in 14% of patients 100. Adverse events observed during anti-PD-1 therapy have included hepatitis, colitis, hypophysitis, thyroiditis, pneumonitis, and vitiligo (Table 1). Antibodies targeting PD-L1 have demonstrated response rates and safety profiles comparable to those targeting PD-1 104. This difference in adverse event profiles, along with the fact that PD-1 blockade can be effective in ipilimumab-refractory melanoma 109, suggest that these particular checkpoint inhibitors act via distinct cellular and molecular mechanisms. PD-1/PD-L1 interactions primarily take place peripherally at sites where activated T cells interact with PD-L1/2-expressing cells (e.g., within the TME), whereas B7/CD28/CTLA-4 interactions occur primarily in lymphoid organs 157 harboring bountiful populations of T cells, and Tregs in particular (e.g., Peyer’s patches in the lining of the small intestines). Thus, the action of mAbs targeting PD-1 are likely concentrated within the TME as opposed to anti-CTLA-4 mAbs, manifesting in a decreased likelihood of off-target effects and greater tolerability.

Combination therapies targeting CTLA-4 and PD-1 have generated positive data showing that combining immunomodulatory mAbs might have advantages over monotherapy with either agent, albeit with greater side effect frequency and severity. The phase 3 study discussed previously and outlined in Table 2 found that combination of nivolumab plus ipilimumab, compared with nivolumab alone, resulted in a greater incidence of grades 3/4 adverse events (68.7% versus 43.5%, respectively 106). There were no drug-related deaths seen with combination therapy, and most adverse events (85–100%) could be managed successfully using standardized treatment guidelines, including steroids and infliximab 106, but endocrine-related events were the most resistant to management. Thus, combined therapy targeting CTLA-4 and PD-1 might be advantageous in patient subsets determined by screening for the presence of predictive biomarkers, such as tumor PD-L1 expression.

Differences among checkpoint inhibitors are also reflected in their dosing regimens (outlined in their respective package inserts). For the treatment of melanoma, ipilimumab, for example, is given every 3 weeks for a total of four doses, at which point therapy must be stopped. On the other hand, nivolumab, with its milder side effect profile, is given every 2 weeks indefinitely, until the patient experiences either lack of clinical benefit or unacceptable toxicity. Nivolumab can also be combined with ipilimumab, with the two agents being co-administered every 3 weeks for 4 doses, after which point the normal dosing scheme of nivolumab is followed. Importantly, nivolumab is also indicated for NSCLC and RCC. Pembrolizumab, the other approved PD-1 antagonist, is currently indicated for melanoma and NSCLC, and is given every 3 weeks. For either anti-PD-1 therapy, treatment is maintained regardless of whether the tumor completely resolves or its size has stabilized. Thus a major advantage of anti-PD-1 mAbs is greater tolerability, making them more suitable for maintenance therapy in appropriate patient subsets.

Costimulatory agonists might be expected to elicit irAEs associated with the production of inflammatory mediators that drive T cell expansion and effector differentiation, but on the other hand also have potential toxic properties when present in large quantities systemically. For instance, CD40 ligation on DCs induces secretion of IL-12 110, which facilitates beneficial tumoricidal effector T cell function 158, but when present at high systemic levels can be toxic 159. Perhaps the most dramatic example of this type of irAE occurred during a phase I clinical trial evaluating the safety of TGN1412, a superagonist to CD28 that had been designed to activate Tregs, but unfortunately ended in disaster when all six trial participants in the treatment group rapidly developed symptoms characteristic of cytokine release syndrome (cytokine storm involving rapid spikes in systemic TNF-α, IFN-γ, IL-6, IL-2, IL-1β and others, as illustrated in Fig. 1), such as headache, nausea, diarrhea, vasodilation, hypotension, and eventually (non-fatal) multi-organ failure 160.

Figure 1. Role of Fc-FcR interactions in mediating therapeutic efficacy and adverse events of antibody-based immunomodulatory therapy (using anti-CD137 agonist as an example).

Figure 1

Monoclonal antibodies (mAbs) bind costimulatory tumor necrosis factor receptor (TNFR) superfamily members bivalently, which is insufficient to initiate costimulatory signaling in the T cell (A) unless fragment crystallizable receptor (FcR) is present on nearby innate accessory cells to facilitate costimulatory receptor clustering (B). Clustering of activating FcR can also signal to the FcR-bearing accessory cell (C), leading to the release of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, etc.), which can potentially contribute to cytokine release syndrome. Conversely, cytotoxic or phagocytic innate cells expressing activating FcR can kill therapeutically-irrelevant cells expressing the mAb target via antibody-dependent cell-mediated cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP), respectively (D). It might also be directed against the T cells undergoing costimulation (not shown), which could account for recent findings that activating FcR engagement can negatively impact efficacy in certain scenarios. The precise nature of FcR-triggered responses by accessory cells depends ultimately on a variety of factors, such as the relative ratio of activating to inhibitory FcRs engaged (not depicted here), as well as the structural/geometric context of FcR ligation; for example, large immune complexes comprised of IgG-coated soluble antigens have been shown to increase inflammatory cytokine production (E), whereas interaction with soluble, unbound IgG (i.e., under homeostatic conditions) or small immune complexes appears to suppress hyper-activation of immune cells by inducing the secretion of anti-inflammatory cytokines, such as IL-10, or by suppressing the secretion or signaling of inflammatory cytokine secretion (F). Although pro-inflammatory cytokines play a causative role in therapy-associated adverse events, their effects could also contribute to overall antitumor efficacy. This last point is an important consideration, since disruption of Fc-FcR interactions could impact both adverse events and efficacy, depending on the particular context. Also worth noting is that avoiding FcR-induced inflammatory responses would not completely prevent therapy-associated adverse events, which is thought to be mediated in part through the activation of specific effector T cells themselves.

Several mechanisms have been suggested for cytokine release induced by TGN1412. One viewpoint implicates a toxic inflammatory response due to T cell hyper-activation mediated by excessive costimulatory receptor stimulation (depicted in Fig. 1B, in this case using CD137 as a representative example) 161, 162. Another school of thought points to excessive activation of innate effector cell populations (e.g., monocytes and NK cells) via antibody fragment crystallizable (Fc) engagement of Fc receptor (FcR) (discussed in the next section), thus triggering the massive release of pro-inflammatory cytokines 163, 164 (Fig. 1C). Although the severe symptoms elicited by TGN1412 were impossible to predict, it was already known that infusion of immune-reactivity antibodies can trigger responses resembling cytokine release syndrome 165167. It is likely that both mechanisms were, at least to some extent, involved in mediating toxicity in these patients. Evidence supporting the latter assertion comes from recent work describing contributions of stimulation-induced FcγR signaling to the induction of pro-inflammatory cytokines 168, 169, in addition to the suggested role of pro-inflammatory M1 macrophages in mediating immunotherapy-associated toxicity 170.

In the case of the anti-CD137 agonist urelumab, phase II testing had to be terminated due to unanticipated severe toxicity in a substantial portion of patients. Grade 4 hepatitis was the most serious adverse event, although additional toxicities were commonly noted, including fatigue, rash, diarrhea, and neutropenia (Table 1). These toxicities may have been related in part to the production of pro-inflammatory cytokines, such as types I and II interferons (IFN) and tumor necrosis factor alpha (TNF-α) 107, 141. These results were perhaps not entirely surprising since mouse studies have demonstrated that CD137 agonist administered chronically or at high dosage can induce immunologic dysfunction leading to organ-specific toxicities that correlate with increased CD8+ T cell infiltration into those tissues and organs 171, 172. Subsequent clinical testing of CD137 agonists has utilized modified protocols with lower dosages, avoiding the dose-dependent hepatotoxicities observed previously 142.

During initial phase I clinical testing of a CD134 agonist for the treatment of various advanced cancers, including melanoma, RCC, squamous cell carcinoma (SCC) of the urethra, prostate cancer, and cholangiocarcinoma, fewer irAEs occurred and an acceptable toxicity profile was observed 140. 12 out of 30 patients had regression of at least one tumor nodule, and in an additional 6 patients there were no changes in target lesion measurements (Table 1); however none of the patients demonstrated responses according to RECIST criteria, which mandate tumor shrinkage of at least 30% to be considered positive clinical responses 140. Differences in adverse events seen with anti-CD134 versus anti-CD137 may in part be attributed to the more widespread expression pattern of CD137 that encompasses not only activated T cells and Tregs but also several innate cell types such as natural killer (NK) cells and DCs 173175, in contrast to CD134, whose expression appears to be more restricted to T cell subsets.

Management of irAEs should be guided by the severity of symptoms and typically involves early detection, temporary cessation of therapy, careful monitoring, supportive care such as oral hydration, and tailored symptomatic relief. For more severe irAEs, immunosuppressive drugs such as corticosteroids are often successful in mitigating symptoms, and infliximab (which neutralizes TNF-α) can be helpful in managing cytokine release syndrome. Additionally, there are clinical algorithms to aid in determining the appropriate strategy to best manage irAEs 107, 154, 176, 177. As mentioned previously, symptomatic relief using these agents has been shown not to negatively impact treatment efficacy, which substantially enhances the clinical utility of immunomodulatory mAbs.

6. Mechanistic contributions of Fc-FcR interactions

An understanding of the structural features of immunomodulatory mAbs illuminates their efficacy and adverse event profiles. Such mAbs generally are IgG isotypes and thus share a common structure composed of two identical, variable fragment antigen-binding (Fab) domains that confer target specificity, as well as a single, non-variable Fc domain corresponding to the particular IgG subtype (1–4 in human; 1, 2a, 2b and 3 in mouse). The Fc domain can bind FcRs on the surface of innate immune effector cells (macrophages, DCs, NK cells, etc.) and B cells. Depending on cell type and environmental context, Fc binding to surface FcR will engage signaling pathways in the FcR-bearing cell and help shape functional responses. For example, interaction of macrophage FcR with the Fc domains present on an IgG-coated cell can induce that macrophage to initiate effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cell-mediated phagocytosis (ADCP) 58, 178. Fc-FcR interaction thus serves as a homing signal to direct immune effector mechanisms to a specific target, effectively bridging adaptive (e.g., B cell-generated IgG) and innate (e.g., phagocytosis) immunity and enabling a number of important biological activities 179.

The canonical FcRs belong to the Ig superfamily and can either transmit an activating signal (FcγRI, IIA, IIC, IIIA and IIIB in human; FcγRI, III and IV in mouse) or alternatively an inhibitory signal (FcγRIIB in both human and mouse). The different IgG subtypes possess variable affinities for specific FcRs and thus differ in the balance of activating versus inhibitory signals they transmit into FcR-expressing cells 8. Table 3 provides a summary of IgG-FcR interactions relevant to immunomodulatory mAb-based therapy. Due to the fact that human FcγRIIC (expressed in only 20% of patients) and FcγRIIIB (a GPI-linked receptor mainly restricted to neutrophils) have not been well characterized 180, they have been left out of Table 3 and will not be considered further. Similarly, human IgG3, which is not used therapeutically due to relative instability and immunogenicity 180, and mouse IgG3, which does not interact appreciably with FcγRs 180, 181, are also excluded from Table 3 and from further discussion.

Table 3.

FcγR properties, expression patterns and IgG isotype binding affinities in mice and humans

Species Mouse Species Human
Receptor FcγRI FcγRIII FcγRIV FcγRII B Receptor FcγRI FcγRIIa FcγRIIIa FcγRII B
Function Activating Activating Activating Inhibitory Function Activating Activating Activating Inhibitory
Signaling ITAM ITAM ITAM ITIM Signaling ITAM ITAM ITAM ITIM
Allotype n/a n/a n/a n/a Allotype* n/a H131 R131 F158 V158 n/a
IgG1 ++ +++ IgG1 ++++ +++ +++ ++ +++ ++
IgG2a ++++ ++ +++ ++ IgG2 ++ + +
IgG2b ++++ ++ ++ +++ IgG4 ++++ ++ ++ ++ ++ ++
Expression patterns ** DC DC
Mac
PMN
NK
DC
Mac
PMN
NK
DC
Mac
PMN
NK
T***
Expression patterns** DC
Mac
PMN
DC
Mac
PMN
Mac
PMN
NK
B
DC
Mac
PMN
NK****
*

Only human allotypes that have been sufficiently characterized are included. For each receptor, the more common variant appears first.

**

Low expression levels indicated by italics.

***

Expressed by mouse memory CD8+ T cells.

****

Expressed by a subset of human NK cells.

ITAM, immunoreceptor tyrosine-based activation motifs; ITIM, immunoreceptor tyrosine-based inhibitory motifs; B, B cell; DC, dendritic cell; Mac, macrophage/monocyte; PMN, polymorphonuclear cell (neutrophil); NK, NK cell; T, T cell.

FcγRs can be either activating or inhibitory, as determined mainly by their intracellular signaling components. Activating receptors contain intracellular immunoreceptor tyrosine-based activation motifs (ITAMs) which transduce stimulatory signals to the receptor-bearing cell when the extracellular portion of the receptor is ligated. Conversely, inhibitory receptors contain immunoreceptor tyrosine-based inhibitory motifs (ITIMs), which transduce suppressive signals when bound by ligand 178 (Table 3). The opposing nature of the activating and inhibitory FcγRs allows for fine-tuning of FcR-mediated immune responses based on the summation of stimulatory and suppressive inputs 182 (see Fig. 1). The immune-regulating activities mediated by FcRs have been reviewed elsewhere 178.

Recently, it was found that inhibitory ITAM (ITAMi) signaling, in the context of low-valency receptor binding 183, such as that occurring with unbound, soluble IgG (Fig. 1F), represents another source of FcR-mediated inputs 184. ITAMi signaling after FcγRI (an activating receptor) binding by immune complexes has been shown, for example, to attenuate IFN-γ signaling and induce the secretion of immunosuppressive IL-10 169. The size of immune complexes seems to be an important factor in determining whether FcR signaling generates a pro- or anti-inflammatory response 168, with larger complexes generally triggering pro-inflammatory cytokine activity (Fig. 1E) and smaller immune complexes (and soluble, monomeric IgG) appearing to promote anti-inflammatory conditions (Fig. 1F) . This phenomenon likely accounts for the clinical benefits of IVIG therapy in treating patients with rheumatoid arthritis 185, idiopathic thrombocytopenic purpura (ITP) 186, and other autoimmune disorders 187, 188. Our understanding of Fc-FcR interactions and their involvements in numerous biological processes, however, is far from complete, and their complexities are only now beginning to be unraveled. Overviews of Fc-FcR interactions and their role in mAb-based cancer therapy (specifically, their contributions to treatment efficacy) are available 7, 8, 180, 189, 190.

The role of FcR-mediated interactions in the function of cytotoxic mAbs such as rituximab arises predictably from the involvement of innate effector cells in their mechanisms of cellular elimination 191194, but defies straightforward prediction in the case of immunomodulatory mAbs. Since checkpoint inhibitors disrupt inhibitory ligand-receptor interactions, the involvement of FcR would not seem necessary, for instance, to prevent CTLA-4 from disrupting B7.1/B7.2 interaction with CD28 during T cell priming in draining lymph nodes. However, the potency of ipilimumab was found to depend on intratumoral depletion of Tregs 195 due to their constitutive expression of the CTLA-4. Depletion was shown to depend on activating FcγRIV, suggesting macrophage-mediated ADCC as the driving mechanism 78, 196, 197 (Table 4).

The story is further complicated by the fact that ADCC may not be the only mechanism of Treg depletion. There is evidence that Tregs possess a degree of plasticity and under certain circumstances can undergo a phenotypic conversion, in which they lose their suppressive properties and gain effector functionality 198. CD134 agonists were shown to inhibit de novo Treg induction, expansion, and suppressive function 199, in addition to their ability to promote effector CD4+ and CD8+ T cell proliferation 125, 128. Similarly, GITR agonism was shown to downregulate Treg-specific transcription factors and cytokines (with complete loss of Foxp3 expression) and upregulate T-bet and Eomesodermin (Eomes) 200. More recent evidence suggests that costimulation with CD134 plus CD137 agonists can induce Tregs to adopt a cytotoxic CD4+ effector phenotype, expressing Eomes, Granzyme B (GzmB), and IFN-γ, while retaining Foxp3 expression 201. Additionally, CD137 costimulated Foxp3+ Tregs that express Eomes and GzmB can kill tumor target cells 202. Thus it appears that immunomodulatory mAb therapy can exploit Treg plasticity to reverse tumor-mediated immunosuppression and add to the overall effector T cell response (Table 4).

Costimulatory signaling by TNFR family members is initiated under physiological conditions when they undergo trimerization upon binding their natural ligands; however, mAbs can only bind a target bivalently (Fig. 1A). Thus, generally speaking, costimulatory agonists cannot on their own induce costimulation. Nevertheless, FcR-mediated, mAb-induced clustering of receptors facilitates the formation of higher order signaling complexes capable of initiating costimulation (Fig. 1B) and appears to be required for the efficacy of at least some agonists. Since engagement of activating FcR on DCs enhances their antigen presentation and costimulatory capacity 203, 204, one might predict that activating FcR would be the most potent in facilitating therapeutic effects of costimulatory agonists. To the contrary, activity of the proapoptotic receptor agonists (PARA) targeting death receptor (DR) 4, DR5, and CD95 (Fas) was found to depend on either activating or inhibitory FcR, and in some contexts, inhibitory FcγRIIB was shown to be sufficient in mediating this activity 205. Similar FcR requirements were later demonstrated for anti-CD40 205207, thus extending the finding to non-apoptosis-inducing costimulatory TNFR agonists (Table 4). These observations suggested a mechanisms whereby FcγRIIB engagement on nearby FcR-expressing cells by TNFR-bound agonist mAbs induces clustering of TNFRs, thus enabling sufficient induction of costimulatory signaling (Fig. 1B). This proposed model was further supported by the discovery that efficacy did not depend on FcR-dependent signaling in the accessory cell that expresses it 208. Thus, FcR appears to facilitate agonist-induced costimulation through receptor clustering.

CD40, CD28, and CD137 agonists containing a subdomain from the Fc hinge region of the B isoform of human IgG2 can function independently of Fc-FcR interactions, retaining biological effects even after enzymatic removal of antibody Fc domains. The authors propose that the constrained disulfide hinge configuration facilitates uniquely compact divalent receptor clustering and thereby the induction of costimulatory signaling 209. On the other hand, activating FcR are required to enable other TNFR agonists, such as GITR 197, CD27 210 and CD134 211, to deplete intratumoral Tregs (as with anti-CTLA-4 mAbs 78, 196, 197), implicating innate effector mechanisms (reviewed in 189 and shown in Table 4). Whether activating or inhibitory FcR are involved in the mechanism(s) underlying immunomodulatory agonist-induced boosting of initial antigen-priming in lymphoid organs, however, remains to be determined in vivo. Interestingly, the therapeutic activity of CD137 agonist in a mouse lymphoma model was actually enhanced in the absence of activating FcγRIII 212. This is a surprising finding, particularly in light of earlier work demonstrating FcR-dependent CD137 induction on NK cells 213, whose tumoricidal function is well known to be engaged by CD137 agonist 173. As such, the outcome of FcR-mediated events occurring following administration of CD137 agonist can vary in a context-dependent manner.

Most recently, Dahan et. al., described the Fc-FcR interactions involved in the targeting of the PD-1/PD-L1 axis using a variety of antagonist mAbs 214. This study revealed that antagonistic anti-PD-1 mAbs mediate biological activity in a manner that is not dependent on FcR interaction. As with anti-CD137 agonist 212, engagement of activating FcR leads to a decrease in efficacy (Table 4). This inhibition seems to be mediated by FcR-induced ADCC directed toward T cells expressing PD-1, depleting the cells intended to be activated. An additional proposed mechanism of FcR-related diminution of efficacy implicates FcγRIIB-induced clustering and consequent stimulation (as with TNFR agonists), in essence converting the intended blockade of PD-1 into agonistic PD-1 stimulation 214.

The same study uncovered entirely different FcR-dependent mechanisms at work in mAbs targeting PD-L1, which require the presence of activating FcR-expressing cells for optimal efficacy. These findings suggest that ADCC-mediated depletion of PD-L1-expressing myeloid cells within the TME is important for the efficacy of PD-L1-targeted immunomodulatory therapy (Table 4). Furthermore, this mechanism likely synergizes with the FcR-independent disruption of PD-1/PD-L signaling facilitated by the antagonistic mAbs 106. These data are in agreement with previous work showing that immunosuppressive myeloid cell subsets, such as MDSCs 56, can foster a pro-cancer environment barricading the tumor from immune intervention.

The complexities of Fc-FcR interactions and the roles they play in mediating the activity of immunomodulatory antibodies is an area of active investigation and has been reviewed extensively elsewhere 168, 178, 182, 215. Additionally, Offringa and Glennie provide in a Cancer Cell preview 216 to Dahan et. al.’s report 214 a concise overview of Fc-FcR roles in immunomodulatory therapy that includes those just uncovered for anti-PD-1 and anti-PD-L1 antagonists.

7. Expert opinion: Is it possible to selectively increase safety while maintaining efficacy?

Modulation of the immune system with checkpoint inhibitors and costimulatory agonists has emerged as a highly promising approach to cancer therapy. Improved durability and rates of response, more tolerable side effect profiles and extensive clinical applicability (in both solid and liquid tumors) set immunomodulatory mAbs apart from previous generations of cancer therapeutics. A growing body of evidence supports the notion that immunomodulatory agents will soon become standard first-line therapies. Table 1 summarizes the current development status for representative mAbs and lists for each phase of clinical testing specific conditions/indications in which they are being evaluated. Data on PFS, OS, and best overall response (BOR) rates are essential to evaluate different treatments. Additionally, although rates of grades 3/4 side effects are included in clinical trials and can be used as an indirect measure of quality of life (QOL), direct assessment will be of great value moving forward.

Evidence has shown that the efficacy and adverse event profile of a given agent can vary significantly from patient to patient. The identification of biomarkers to predict patient likelihood of response and the potential of treatment toxicity is essential to the design of patient-specific treatment strategies, as well as to the proper clinical testing of these agents. Importantly, cancer patients must be screened to assess their eligibility, since inclusion in clinical trials of patients unable to respond can dilute overall efficacy determined by the study and potentially mask the true clinical potency of new agents being developed 156. One potential biomarker identified for ipilimumab response is the cancer/testis antigen NY-ESO-1, which is expressed normally in the testis and placenta, as well as aberrantly in a subset of advanced melanomas. It was found that NY-ESO-1 seropositivity, and to greater extent the existence of NY-ESO-1 specific CD8+ T cells in the periphery, correlates positively with clinical responsiveness to ipilimumab 217. It was also found that patients with lactate dehydrogenase levels exceeding twice the upper limit of normal were far less likely to benefit from ipilimumab therapy in the long-term 218. Moreover, recent evidence has demonstrated potential value in using PD-L1 expression as a biomarker to determine the likelihood of patient response to PD-1 antagonism and the likelihood of enhanced clinical benefit from supplementing anti-PD-1 with additional therapeutics in combination 106 (Table 2).

Interestingly, it was recently shown that gut microbiota composition can influence the efficacy of anti-CTLA-4 therapy. Specifically, the gut microbiota is required for antitumor responses with the anti-CTLA-4 mAb 9D9 in multiple mouse models of melanoma, sarcoma, and colon cancer. Data also showed that the microbiota was necessary for mAb-induced activation of effector CD4+ T cells in the spleen 219. These findings align with previous knowledge that Tregs are abundant in the GI tract. The identification of individual bacterial species associated with anti-CTLA-4 efficacy suggested that their presence and relative abundance in a patient’s gut microbiota could serve as a biomarker to predict likelihood of treatment response. Furthermore, feces harvested from ipilimumab-treated patients were found to differ in microbiota composition, which gradually shifted in favor of the efficacy-enhancing species. Intriguingly, fecal microbial transplants into tumor-inoculated mice with patient feces samples containing significant numbers of these microbes helped to generate effective antitumor responses, whereas transplantation of feces containing fewer of these specific microbes were unable to respond 219.

There also appears to be a genetic basis for the likelihood of beneficial response to immunomodulatory therapy. Genetic analysis of 64 ipilimumab-treated melanoma patients uncovered a correlation between mutational load and clinical benefit and identified a neo-antigen landscape specific to tumors that are highly responsive to treatment 220. Some specific genetic mutations 221 and the presence of particular single-nucleotide polymorphisms 222 have also been shown to predict clinical response. Interestingly, the single-nucleotide polymorphisms (SNPs) that correlated with beneficial clinical response did not show any correlation with the occurrence or severity of adverse events 222, suggesting that the mechanisms underlying efficacy and toxicity do not overlap entirely and can thus be differentially targeted when designing future immunomodulatory mAbs for greater therapeutic index. This study also supports an earlier hypothesis that some mutations considered silent passengers should instead be classified as immune determinants 223 that collectively define immunogenicity and susceptibility to antitumor effector mechanisms employed by the immune system. Accordingly, genetic screening could be employed to identify likely responders to therapy and help ensure that patients are offered only those treatments from which they are likely to benefit.

The process of selecting new immunomodulatory antibodies for clinical development also needs to be improved at the preclinical level in order to better predict their effects in humans. The use of in vitro screening assays is convenient, but results obtained must be interpreted with caution, because numerous factors such as cell culture density, microenvironment architecture, and stimulation timeframes often differ substantially from those in real patients 224. The unanticipated consequences of the TGN1412 trial, for example, was later determined to result from failure to take into consideration differences in CD28 expression patterns between humans and cynomolgus macaques 225, 226. This unfortunate event has helped lead to the development of new screening techniques and safety measures, which are already being utilized in designing and testing new therapies 226. Prevention of such incidents will be essential to continued progress in this field.

The difference in response kinetics when comparing immunomodulatory mAbs to traditional cancer therapeutics is also important to consider in future clinical testing protocols. Immunomodulatory antibodies, which function indirectly via immune system activation, can take longer to mount an attack and thus warrant adjustment to currently defined endpoints and other measures used to evaluate agents 227. The importance of this last point is illustrated in the premature halting of a phase III trial with the anti-CTLA-4 agent tremelimumab due to apparent lack of improvement in survival. However, follow-up investigation of study participants eventually revealed improvements occurring after 2 years 228, reflective of the delayed evidence of therapeutic response seen with these agents. Similarly, as discussed earlier, the absence of efficacy reported for an anti-OX40 agonist antibody despite initially promising data demonstrates the incompatibility of traditional RECIST criteria with immunomodulatory therapies 140. In fact, even with successful therapeutic outcomes tumor shrinkage would not be expected to occur for some time after treatment is initiated. This is because tumoricidal responses induced by immunomodulatory mAbs rely on effector cell infiltration into the TME; thus upon measurement, any tumor shrinkage would likely be difficult to detect due to lymphocytic influx localized to tumor sites. Therefore, appropriate additional criteria, such as immune cell phenotypes, frequencies, and overall abundances, both in circulation and within TMEs, need to be incorporated for improved evaluation of clinical trials 229. Only then can we be confident that we are accurately characterizing future generations of immunomodulatory therapeutic candidates.

As mentioned earlier, the ability to selectively enhance efficacy and/or limit toxicity likely depends on the specific mechanism(s) responsible for each. There is considerable overlap in the immune effector mechanisms involved in beneficial therapeutic antitumor effects and harmful adverse events. For instance, TNF-α has potent tumoricidal activity, but can be a causative factor in cachexia, insulin resistance and cytokine release syndrome 230233. Further, initial clinical trials testing ipilimumab found a positive correlation between therapeutic responses and the prevalence and severity of adverse events 177, 234, 235, raising the critical question as to what extent adverse events can be limited without compromising therapeutic efficacy. Fortunately, extensive clinical testing of ipilimumab has provided encouraging results in this regard, specifically that ipilimumab-responsive patients who develop irAEs can continue to benefit from an anti-tumor response even after pharmacologic reversal of associated symptoms with, for example, administration of corticosteroids 106, 176. Reversal of irAEs and cytokine release syndrome without loss of efficacy was also achieved in other T cell-mediated cancer therapies, in which inhibition of TNF-α with etanercept 236 and inhibition of IL-6 signaling with tocilizumab 236, 237 were effective. Alternative agents that might be useful in managing cytokine release syndrome during immunotherapy include inhibitors to MCP-1, MIP-1β, IL-2R, and IL-1R 238 (depicted in Fig. 1).

Combinations of immunotherapies 109, as well as immunotherapies used in conjunction with traditional treatment options such as chemotherapy 239 and radiotherapy 240 have been attempted in search of treatment approaches with enhanced efficacy and improved safety profiles. Combination therapy has opened the possibility of limiting the occurrence of irAEs due to additive (and for certain combinations perhaps even synergistic) effects of multiple agents, which could allow for administration of lower doses. Although the frequency and severity of adverse events have been greater with combinations than with individual monotherapies thus far 106 (Table 2), the ability to titrate dosages could potentially avoid many adverse events directly. A detailed mechanistic understanding of how these therapeutics elicit their associated beneficial and toxic effects is crucial to designing strategies that selectively block the latter. Ideally, these approaches will facilitate an overall advantage in favor of enhanced efficacy over adverse events. Ultimately, the benefits and risks of specific combinations will need to be addressed in future trials. Melero and colleagues recently published a comprehensive review summarizing general concepts, specific examples under development, and important considerations in the design of combination immunotherapies 241.

It seems likely that Fc-FcR interactions also play a role in mediating at least some of the adverse events elicited by immunomodulatory mAbs. For instance, target expression on therapeutically-irrelevant cell types could potentially lead to collateral damage of these cells via ADCC or ADCP (Fig. 1D). Thus, while ADCC/ADCP might be therapeutically beneficial by depleting intratumoral Tregs with mAbs to CTLA-4, GITR and OX40 78, 196, 197, 211 (Table 4), it might be problematic, for instance, with anti-CD137, since CD137 is expressed on a variety of cell types, some of which appear not to be involved in the antitumor response 242. Neurons, astrocytes and microglia 243, for example, are among these therapeutically-irrelevant, CD137-expressing cell types. Additionally, endothelia can express CD137 in response to inflammatory mediators 244 and in diseases such as atherosclerosis 245.

Furthermore, expression of soluble forms of some targets (e.g., CD134 and CD137) has been found in a variety of pathological conditions 246248, raising the potential for the formation of immune complexes which can trigger inflammation (Fig. 1E) 168 and may potentially be involved in mechanisms driving pathological immune hyper-reactivity, such as that occurring in IgG-mediated anaphylaxis 249. Finally, surface coating of IgG on either therapeutically-relevant or -irrelevant cells could cause innate cells expressing activating FcγR to release pro-inflammatory cytokines contributing to adverse events 168, 250. In sum, although Fc-FcR interactions may be necessary for the full therapeutic efficacy of immunomodulatory mAbs, they might also be responsible for therapeutically-complicating adverse events through a variety of potential mechanisms. Further, these adverse events might be exacerbated by other pathologies (e.g., underlying infection or autoimmune predisposition) from which cancer patients might be suffering. Nevertheless, seemingly contradictory evidence showing both pro- and anti-inflammatory cytokine secretion in response to FcR engagement illustrates our incomplete understanding of FcR-mediated immune functions.

Despite these gaps in understanding, Fc-FcR interactions are likely responsible for at least some of the toxicities seen with immunomodulatory mAb therapy. In such cases, preventing these interactions may help limit adverse events to increase therapeutic index and enhance precision medicine. This notion is additionally supported by the heterogeneity of responses to FcR-dependent mAb activity. For instance, the absence of a human homolog to the high-avidity FcγRIIB-binding of mouse IgG1 may limit efficacy of some costimulatory agonists (Table 3). Furthermore, patient response heterogeneity can arise from relative levels of FcR expression and functional status, which can vary widely between tissues and cell types. For example, although most innate cells express both activating and inhibitory FcγRs, NK cells are known to express only activating receptors in mice and humans (except for a recently discovered subset of NK cells expressing FcγRIIB), whereas B cells only express inhibitory receptor 178. The different distributions of such cells likely explains the tissue- and organ-specific differences in overall response to therapy 11. Interestingly, it was recently suggested that in a murine model peritoneal and visceral adiposity can influence the magnitude of cytokine-mediated toxicity associated with the presence of resident M1-polarized macrophages 170. Other factors include current immune status (e.g., active responses to infection), and patient-specific differences in both FcR-mediated signaling summation patterns (i.e., activating vs. inhibitory FcR inputs) and FcR-specific avidities to various IgG isotypes (Table 3). Additionally, the existence of allotypic variants with different binding affinities contributes to patient response heterogeneity; so far, human FcγRIIa and FcγRIIIa each have had two variants characterized 251.

Establishing FcR-independent mechanisms of immunomodulatory mAb activity is therefore a highly desirable goal. Antibody multimerization has been shown to be an effective strategy to bypass FcR requirements, enabling the preservation of anti-CD40 agonist activity in vitro and in vivo, as well as in a model of lymphoma with mice completely lacking all FcγRs 252 (Table 4). Immunomodulatory therapy can also be optimized using mAb engineering to alter FcR binding properties. Early studies focused on increasing binding avidity of IgG to activating FcR, which could be accomplished using protein engineering 253, 254, as well as modification to antibody glycosylation 255, 256. Additionally, selective enhancement of FcγRIIB binding was used to generate an anti-CD137 agonist with increased agonistic activity in vitro 257. Individual FcRs have also been shown to exhibit differential contributions to cellular functions; for example, it has been demonstrated that FcγRI was largely responsible for secretion of both anti-inflammatory IL-10 258 and pro-inflammatory cytokines 259. A suggested implication of this findings was that macrophage-mediated ADCC and ADCP could potentially be uncoupled from antibody-dependent cytokine release (ADCR), a feat which was accomplished recently by Kinder et. al., using an Fc-engineering approach to modulate cytokine release while leaving cell-killing activity intact 259. The ability to selectively fine-tune or prevent cytokine release thus opens the possibility of limiting cytokine-related adverse events without losing effector capacity and tumoricidal activity. Contrarily, in situations where the secretion of anti-inflammatory cytokines might be hindering efficacy, selective attenuation of ADCR would be advantageous.

Additionally, the human IgG2 isotype (specifically the B isoform) was shown to possess the unique ability to exhibit activity in an entirely FcR-independent manner, likely due to an especially compact geometry and constrained disulfide hinge configuration 209. The authors propose that these properties enable sufficient clustering of TNFRs to reach a signaling threshold beyond which stimulation ensues. Notably, this phenomenon was shown for agonists targeting multiple costimulatory receptors, including CD40, CD28, and CD137, and it was further demonstrated that selective mutagenesis could generate a wide range of agonistic activities, without altering receptor binding avidity (Table 4). That human IgG2 is capable of exerting superior agonistic activity in an FcR-independent manner potentially accounts for data from clinical trials demonstrating superior potency of the anti-CD40 agonist CP-870,893, an IgG2 isotype (Table 1), compared to those of other isotypes 260, 261.

A greater understanding of the mechanisms behind immunomodulatory mAb therapy will be critical for the future optimization of these therapeutic agents. Importantly, the mechanisms underlying efficacy and adverse events need to be further delineated. Recent characterization of PD-1/PD-L1-targeted therapies 214 further adds to the ever-expanding complexity of the mechanisms at work in immunomodulatory therapy, while also illustrating the importance of proper antibody isotype selection and the value of Fc domain modification in order to optimize efficacy. Much work is also needed to explain the context-dependent pro- and anti-inflammatory responses (Figs. 1E and 1F, respectively) induced by FcR binding to mAb Fc domain. While many data support the idea that avoiding FcR activity will help limit adverse events, the anti-inflammatory responses to FcR signaling in certain contexts now pose the possibility that FcR engagement actually contributes negatively to therapeutic efficacy, and that circumvention of FcR interaction could lead to an enhancement of activity (perhaps even concurrently with attenuation of adverse events). Conversely, it is possible that FcR-mediated pro-inflammatory cytokine secretion contributes to overall antitumor efficacy, representing a benefit that would be lost if FcR engagement were avoided. Only with continued investigation will we be able to accurately predict how adjustments to Fc-FcR interactions will impact efficacy and toxicity. Therapeutic approaches that do not engage FcR, yet retain the ability to effectively stimulate their target pathways, will potentially be capable of inducing beneficial therapeutic responses in cancer patients while sparing them from having to endure the dangerous adverse events of today’s therapies. Hence, perhaps we can have our cake and eat it too.

Article highlights box.

  • Immunomodulatory antibodies (mAbs), including checkpoint inhibitors and costimulatory agonists, represent a promising new approach to cancer therapy.

  • These mAbs work by enhancing immunity, thereby increasing antitumor activities. In doing so, however, a variety of immune-related adverse events (irAEs) can occur.

  • Successful translation of these agents depends on selectively limiting the frequency and severity of irAEs without losing therapeutic efficacy.

  • Although the mechanisms involving efficacy and irAEs have much overlap, evidence supports the idea that these effects can be isolated and fine-tuned.

  • Interactions between Fc and FcR are essential in mediating biological activity, but are also implicated in the eliciting of adverse events.

  • Modifying the structures of these mAbs so that they avoid Fc-FcR interactions, while still retaining therapeutic efficacy, might be a viable strategy for the future development of immunomodulatory antibodies with more favorable therapeutic indices.

Acknowledgments

Authors thank Dr. James Buturla (UConn Health) for his critical comments and editorial contributions.

Footnotes

Declaration of interests

A Vella and A Adler received grants from the NIH (AI094640 and CA109339). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Bibliography

  • 1.Druker BJ, Guilhot F, O’Brien SG, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–17. doi: 10.1056/NEJMoa062867. [DOI] [PubMed] [Google Scholar]
  • 2.Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol. 2010;10:317–27. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391–400. doi: 10.1038/nrd1381. [DOI] [PubMed] [Google Scholar]
  • 4.Michielsen AJ, Ryan EJ, O’Sullivan JN. Dendritic cell inhibition correlates with survival of colorectal cancer patients on bevacizumab treatment. Oncoimmunology. 2012;1:1445–47. doi: 10.4161/onci.21318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lim SH, Beers SA, French RR, et al. Anti-CD20 monoclonal antibodies: historical and future perspectives. Haematologica. 2010;95:135–43. doi: 10.3324/haematol.2008.001628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Beers SA, Chan CH, French RR, et al. CD20 as a target for therapeutic type I and II monoclonal antibodies. Semin Hematol. 2010;47:107–14. doi: 10.1053/j.seminhematol.2010.01.001. [DOI] [PubMed] [Google Scholar]
  • 7**.Nimmerjahn F, Gordan S, Lux A. FcgammaR dependent mechanisms of cytotoxic, agonistic, and neutralizing antibody activities. Trends Immunol. 2015;36:325–36. doi: 10.1016/j.it.2015.04.005. Reviews FcR-dependent mechanisms for different classes of mAbs. [DOI] [PubMed] [Google Scholar]
  • 8**.DiLillo DJ, Ravetch JV. Fc-Receptor Interactions Regulate Both Cytotoxic and Immunomodulatory Therapeutic Antibody Effector Functions. Cancer Immunol Res. 2015;3:704–13. doi: 10.1158/2326-6066.CIR-15-0120. Reviews FcR-dependent mechanisms and discusses Fc-engineering. [DOI] [PubMed] [Google Scholar]
  • 9.Witzig TE, Gordon LI, Cabanillas F, et al. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma. J Clin Oncol. 2002;20:2453–63. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
  • 10.Kaminski MS, Estes J, Zasadny KR, et al. Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience. Blood. 2000;96:1259–66. [PubMed] [Google Scholar]
  • 11**.Melero I, Grimaldi AM, Perez-Gracia JL, Ascierto PA. Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination. Clin Cancer Res. 2013;19:997–1008. doi: 10.1158/1078-0432.CCR-12-2214. Good clinical overview of immunostimulatory (immunomodulatory) mAbs. [DOI] [PubMed] [Google Scholar]
  • 12.Pardoll D. Does the immune system see tumors as foreign or self? Annu Rev Immunol. 2003;21:807–39. doi: 10.1146/annurev.immunol.21.120601.141135. [DOI] [PubMed] [Google Scholar]
  • 13.Schatz DG, Oettinger MA, Schlissel MS. V(D)J recombination: molecular biology and regulation. Annu Rev Immunol. 1992;10:359–83. doi: 10.1146/annurev.iy.10.040192.002043. [DOI] [PubMed] [Google Scholar]
  • 14.Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of T-cell repertoire diversity. Nat Rev Immunol. 2004;4:123–32. doi: 10.1038/nri1292. [DOI] [PubMed] [Google Scholar]
  • 15.Kappler J, Roehm M, Marrack P. T cell tolerance by clonal elimination in the thymus. Cell. 1987;49:273–80. doi: 10.1016/0092-8674(87)90568-x. [DOI] [PubMed] [Google Scholar]
  • 16.Hogquist KA, Baldwin TA, Jameson SC. Central tolerance: learning self-control in the thymus. Nat Rev Immunol. 2005;5:772–82. doi: 10.1038/nri1707. [DOI] [PubMed] [Google Scholar]
  • 17.Krueger A, Fas SC, Baumann S, Krammer PH. The role of CD95 in the regulation of peripheral T-cell apoptosis. Immunol Rev. 2003;193:58–69. doi: 10.1034/j.1600-065x.2003.00047.x. [DOI] [PubMed] [Google Scholar]
  • 18.Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305–34. doi: 10.1146/annurev.immunol.21.120601.141110. [DOI] [PubMed] [Google Scholar]
  • 19.Sakaguchi S. Regulatory T cells: key controllers of immunologic self-tolerance. Cell. 2000;101:455–8. doi: 10.1016/s0092-8674(00)80856-9. [DOI] [PubMed] [Google Scholar]
  • 20.Morgan DJ, Kreuwel HT, Fleck S, et al. Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity. J Immunol. 1998;160:643–51. [PubMed] [Google Scholar]
  • 21.Bowne WB, Srinivasan R, Wolchok JD, et al. Coupling and uncoupling of tumor immunity and autoimmunity. J Exp Med. 1999;190:1717–22. doi: 10.1084/jem.190.11.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Overwijk WW, Lee DS, Surman DR, et al. Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4(+) T lymphocytes. Proc Natl Acad Sci U S A. 1999;96:2982–7. doi: 10.1073/pnas.96.6.2982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Colella TA, Bullock TN, Russell LB, et al. Self-tolerance to the murine homologue of a tyrosinase-derived melanoma antigen: implications for tumor immunotherapy. J Exp Med. 2000;191:1221–32. doi: 10.1084/jem.191.7.1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature. 1998;396:643–9. doi: 10.1038/25292. [DOI] [PubMed] [Google Scholar]
  • 25.Stephens PJ, Greenman CD, Fu B, et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell. 2011;144:27–40. doi: 10.1016/j.cell.2010.11.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–91. doi: 10.1158/0008-5472.CAN-11-3722. [DOI] [PubMed] [Google Scholar]
  • 27.Duan F, Duitama J, Al Seesi S, et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J Exp Med. 2014;211:2231–48. doi: 10.1084/jem.20141308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 29.Jenkins MK, Khoruts A, Ingulli E, et al. In vivo activation of antigen-specific CD4 T cells. Annu Rev Immunol. 2001;19:23–45. doi: 10.1146/annurev.immunol.19.1.23. [DOI] [PubMed] [Google Scholar]
  • 30.Janeway CA, Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. doi: 10.1146/annurev.immunol.20.083001.084359. [DOI] [PubMed] [Google Scholar]
  • 31.Jenkins MK, Taylor PS, Norton SD, Urdahl KB. CD28 delivers a costimulatory signal involved in antigen-specific IL-2 production by human T cells. J Immunol. 1991;147:2461–6. [PubMed] [Google Scholar]
  • 32.Harding FA, McArthur JG, Gross JA, et al. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature. 1992;356:607–9. doi: 10.1038/356607a0. [DOI] [PubMed] [Google Scholar]
  • 33.Norton SD, Zuckerman L, Urdahl KB, et al. The CD28 ligand, B7, enhances IL-2 production by providing a costimulatory signal to T cells. J Immunol. 1992;149:1556–61. [PubMed] [Google Scholar]
  • 34.Croft M. Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat Rev Immunol. 2003;3:609–20. doi: 10.1038/nri1148. [DOI] [PubMed] [Google Scholar]
  • 35.Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005;23:23–68. doi: 10.1146/annurev.immunol.23.021704.115839. [DOI] [PubMed] [Google Scholar]
  • 36.Mescher MF, Curtsinger JM, Agarwal P, et al. Signals required for programming effector and memory development by CD8+ T cells. Immunol Rev. 2006;211:81–92. doi: 10.1111/j.0105-2896.2006.00382.x. [DOI] [PubMed] [Google Scholar]
  • 37.Hawiger D, Inaba K, Dorsett Y, et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med. 2001;194:769–79. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kurts C, Cannarile M, Klebba I, Brocker T. Dendritic cells are sufficient to cross-present self-antigens to CD8 T cells in vivo. J Immunol. 2001;166:1439–42. doi: 10.4049/jimmunol.166.3.1439. [DOI] [PubMed] [Google Scholar]
  • 39.Hagymasi AT, Slaiby AM, Mihalyo MA, et al. Steady state dendritic cells present parenchymal self-antigen and contribute to, but are not essential for, tolerization of naive and Th1 effector CD4 cells. J Immunol. 2007;179:1524–31. doi: 10.4049/jimmunol.179.3.1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Luckashenak N, Schroeder S, Endt K, et al. Constitutive crosspresentation of tissue antigens by dendritic cells controls CD8+ T cell tolerance in vivo. Immunity. 2008;28:521–32. doi: 10.1016/j.immuni.2008.02.018. [DOI] [PubMed] [Google Scholar]
  • 41.Hu J, Kindsvogel W, Busby S, et al. An evaluation of the potential to use tumor-associated antigens as targets for antitumor T cell therapy using transgenic mice expressing a retroviral tumor antigen in normal lymphoid tissues. J Exp Med. 1993;177:1681–90. doi: 10.1084/jem.177.6.1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sotomayor EM, Borrello I, Rattis FM, et al. Cross-presentation of tumor antigens by bone marrow-derived antigen-presenting cells is the dominant mechanism in the induction of T-cell tolerance during B-cell lymphoma progression. Blood. 2001;98:1070–7. doi: 10.1182/blood.v98.4.1070. [DOI] [PubMed] [Google Scholar]
  • 43.Willimsky G, Blankenstein T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature. 2005;437:141–6. doi: 10.1038/nature03954. [DOI] [PubMed] [Google Scholar]
  • 44.Nguyen LT, Elford AR, Murakami K, et al. Tumor growth enhances cross-presentation leading to limited T cell activation without tolerance. J Exp Med. 2002;195:423–35. doi: 10.1084/jem.20010032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ochsenbein AF, Sierro S, Odermatt B, et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature. 2001;411:1058–64. doi: 10.1038/35082583. [DOI] [PubMed] [Google Scholar]
  • 46.Spiotto MT, Yu P, Rowley DA, et al. Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells. Immunity. 2002;17:737–47. doi: 10.1016/s1074-7613(02)00480-6. [DOI] [PubMed] [Google Scholar]
  • 47.Yatim N, Jusforgues-Saklani H, Orozco S, et al. RIPK1 and NF-kappaB signaling in dying cells determines cross-priming of CD8+ T cells. Science. 2015 doi: 10.1126/science.aad0395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Basu S, Binder RJ, Suto R, et al. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol. 2000;12:1539–46. doi: 10.1093/intimm/12.11.1539. [DOI] [PubMed] [Google Scholar]
  • 49.Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–21. doi: 10.1038/nature01991. [DOI] [PubMed] [Google Scholar]
  • 50.Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  • 51.Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–8. doi: 10.1038/ni1102-991. [DOI] [PubMed] [Google Scholar]
  • 52.Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 53.Yu P, Lee Y, Liu W, et al. Intratumor depletion of CD4+ cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors. J Exp Med. 2005;201:779–91. doi: 10.1084/jem.20041684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cao X, Cai SF, Fehniger TA, et al. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity. 2007;27:635–46. doi: 10.1016/j.immuni.2007.08.014. [DOI] [PubMed] [Google Scholar]
  • 55.Malchow S, Leventhal DS, Savage PA. Organ-specific regulatory T cells of thymic origin are expanded in murine prostate tumors. Oncoimmunology. 2013;2:e24898. doi: 10.4161/onci.24898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253–68. doi: 10.1038/nri3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Torre-Amione G, Beauchamp RD, Koeppen H, et al. A highly immunogenic tumor transfected with a murine transforming growth factor type beta 1 cDNA escapes immune surveillance. Proc Natl Acad Sci U S A. 1990;87:1486–90. doi: 10.1073/pnas.87.4.1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med. 2001;7:1118–22. doi: 10.1038/nm1001-1118. [DOI] [PubMed] [Google Scholar]
  • 59.Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9:1269–74. doi: 10.1038/nm934. [DOI] [PubMed] [Google Scholar]
  • 60*.Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–96. doi: 10.1146/annurev.immunol.25.022106.141609. Extensive review of tumor-induced immunosuppression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Egen JG, Allison JP. Cytotoxic T lymphocyte antigen-4 accumulation in the immunological synapse is regulated by TCR signal strength. Immunity. 2002;16:23–35. doi: 10.1016/s1074-7613(01)00259-x. [DOI] [PubMed] [Google Scholar]
  • 62.Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev. 2009;229:12–26. doi: 10.1111/j.1600-065X.2009.00770.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Freeman GJ, Borriello F, Hodes RJ, et al. Uncovering of functional alternative CTLA-4 counter-receptor in B7-deficient mice. Science. 1993;262:907–9. doi: 10.1126/science.7694362. [DOI] [PubMed] [Google Scholar]
  • 64.Freeman GJ, Gribben JG, Boussiotis VA, et al. Cloning of B7–2: a CTLA-4 counter-receptor that costimulates human T cell proliferation. Science. 1993;262:909–11. doi: 10.1126/science.7694363. [DOI] [PubMed] [Google Scholar]
  • 65.Azuma M, Ito D, Yagita H, et al. B70 antigen is a second ligand for CTLA-4 and CD28. Nature. 1993;366:76–9. doi: 10.1038/366076a0. [DOI] [PubMed] [Google Scholar]
  • 66.Linsley PS, Greene JL, Brady W, et al. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity. 1994;1:793–801. doi: 10.1016/s1074-7613(94)80021-9. [DOI] [PubMed] [Google Scholar]
  • 67.Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  • 68.Tivol EA, Borriello F, Schweitzer AN, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3:541–7. doi: 10.1016/1074-7613(95)90125-6. [DOI] [PubMed] [Google Scholar]
  • 69.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shevach EM. Certified professionals: CD4(+)CD25(+) suppressor T cells. J Exp Med. 2001;193:F41–6. doi: 10.1084/jem.193.11.f41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 72.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–61. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 73.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–42. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
  • 74.Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:1734–6. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
  • 75.Hurwitz AA, Yu TF, Leach DR, Allison JP. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. Proc Natl Acad Sci U S A. 1998;95:10067–71. doi: 10.1073/pnas.95.17.10067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Peggs KS, Quezada SA, Chambers CA, et al. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–25. doi: 10.1084/jem.20082492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hurwitz AA, Foster BA, Kwon ED, et al. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res. 2000;60:2444–8. [PubMed] [Google Scholar]
  • 78*.Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–710. doi: 10.1084/jem.20130579. Shows that FcR-mediated ADCC against intratumoral Tregs contributes to efficacy of anti-CTLA-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi: 10.1126/science.aaa8172. [DOI] [PubMed] [Google Scholar]
  • 80**.Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. Landmark phase III trial showing improved overall survival with ipilimumab in advanced melanoma patients, leading to FDA approval in 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nishimura H, Nose M, Hiai H, et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–51. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
  • 82.Nishimura H, Okazaki T, Tanaka Y, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291:319–22. doi: 10.1126/science.291.5502.319. [DOI] [PubMed] [Google Scholar]
  • 83.Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–8. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
  • 85.Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209:1201–17. doi: 10.1084/jem.20112741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ahmadzadeh M, Johnson LA, Heemskerk B, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87*.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. First study to describe the cancer therapeutic potential of PD-1/PD-L1 blockade. [DOI] [PubMed] [Google Scholar]
  • 88.Rosenwald A, Wright G, Leroy K, et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med. 2003;198:851–62. doi: 10.1084/jem.20031074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13:84–8. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
  • 90.Atefi M, Avramis E, Lassen A, et al. Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin Cancer Res. 2014;20:3446–57. doi: 10.1158/1078-0432.CCR-13-2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Spranger S, Spaapen RM, Zha Y, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med. 2013;5:200ra116. doi: 10.1126/scitranslmed.3006504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bald T, Landsberg J, Lopez-Ramos D, et al. Immune cell-poor melanomas benefit from PD-1 blockade after targeted type I IFN activation. Cancer Discov. 2014;4:674–87. doi: 10.1158/2159-8290.CD-13-0458. [DOI] [PubMed] [Google Scholar]
  • 93.Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013;73:3591–603. doi: 10.1158/0008-5472.CAN-12-4100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–71. doi: 10.1038/nature13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7. doi: 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206:3015–29. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Blank C, Brown I, Peterson AC, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64:1140–5. doi: 10.1158/0008-5472.can-03-3259. [DOI] [PubMed] [Google Scholar]
  • 99.Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99:12293–7. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100**.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690. Phase I study of nivolumab showing durable responses against melanoma, RCC, and NSCLC; efficacy in multiple tumor types. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32:1020–30. doi: 10.1200/JCO.2013.53.0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Robert C, Schachter J, Long GV, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372:2521–32. doi: 10.1056/NEJMoa1503093. [DOI] [PubMed] [Google Scholar]
  • 103**.Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–30. doi: 10.1056/NEJMoa1412082. Phase III study showing improved OS and PFS for nivolumab compared to dacarbazine in previously untreated melanoma; superior to standard of care therapies such as chemotherapeutic regimens. [DOI] [PubMed] [Google Scholar]
  • 104**.Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. Phase I trial of anti-PD-L1 showing durable regression and prolonged stabilization of melanoma, RCC, and NSCLC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064–74. doi: 10.1158/1078-0432.CCR-13-3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106**.Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030. Phase III study comparing nivolumab, ipilimumab, and combination therapy with both in untreated melanoma, emphasizes the need to biomarkers and eligibility screening of patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107**.Gelao L, Criscitiello C, Esposito A, et al. Immune checkpoint blockade in cancer treatment: a double-edged sword cross-targeting the host as an “innocent bystander”. Toxins. 2014;6:914–33. doi: 10.3390/toxins6030914. Reviews clinical data, adverse events, and potential correlations with outcome for immunomodulatory mAbs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107:4275–80. doi: 10.1073/pnas.0915174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109*.Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–33. doi: 10.1056/NEJMoa1302369. Phase I study showing possible advantage of nivolumab plus ipilimumab concurrent therapy in melanoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Cella M, Scheidegger D, Palmer-Lehmann K, et al. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med. 1996;184:747–52. doi: 10.1084/jem.184.2.747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Bennett SR, Carbone FR, Karamalis F, et al. Help for cytotoxic-T-cell response is mediated by CD40 signalling. Nature. 1998;393:478–80. doi: 10.1038/30996. [DOI] [PubMed] [Google Scholar]
  • 112.Schoenberger SP, Toes RE, van der Voort EI, et al. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393:480–3. doi: 10.1038/31002. [DOI] [PubMed] [Google Scholar]
  • 113.Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998;393:474–8. doi: 10.1038/30989. [DOI] [PubMed] [Google Scholar]
  • 114.Maxwell JR, Campbell JD, Kim CH, Vella AT. CD40 activation boosts T cell immunity in vivo by enhancing T cell clonal expansion and delaying peripheral T cell deletion. J Immunol. 1999;162:2024–34. [PubMed] [Google Scholar]
  • 115.Sotomayor EM, Borrello I, Tubb E, et al. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40. Nat Med. 1999;5:780–7. doi: 10.1038/10503. [DOI] [PubMed] [Google Scholar]
  • 116.Diehl L, den Boer AT, Schoenberger SP, et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy. Nat Med. 1999;5:774–9. doi: 10.1038/10495. [DOI] [PubMed] [Google Scholar]
  • 117.Mallett S, Fossum S, Barclay AN. Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes--a molecule related to nerve growth factor receptor. EMBO J. 1990;9:1063–8. doi: 10.1002/j.1460-2075.1990.tb08211.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Gramaglia I, Weinberg AD, Lemon M, Croft M. Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses. J Immunol. 1998;161:6510–7. [PubMed] [Google Scholar]
  • 119.Murata K, Ishii N, Takano H, et al. Impairment of antigen-presenting cell function in mice lacking expression of OX40 ligand. J Exp Med. 2000;191:365–74. doi: 10.1084/jem.191.2.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Flynn S, Toellner KM, Raykundalia C, et al. CD4 T cell cytokine differentiation: the B cell activation molecule, OX40 ligand, instructs CD4 T cells to express interleukin 4 and upregulates expression of the chemokine receptor, Blr-1. J Exp Med. 1998;188:297–304. doi: 10.1084/jem.188.2.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Rogers PR, Song J, Gramaglia I, et al. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity. 2001;15:445–55. doi: 10.1016/s1074-7613(01)00191-1. [DOI] [PubMed] [Google Scholar]
  • 122.So T, Song J, Sugie K, et al. Signals from OX40 regulate nuclear factor of activated T cells c1 and T cell helper 2 lineage commitment. Proc Natl Acad Sci U S A. 2006;103:3740–5. doi: 10.1073/pnas.0600205103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Weinberg AD, Rivera MM, Prell R, et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol. 2000;164:2160–9. doi: 10.4049/jimmunol.164.4.2160. [DOI] [PubMed] [Google Scholar]
  • 124.Maxwell JR, Weinberg A, Prell RA, Vella AT. Danger and OX40 receptor signaling synergize to enhance memory T cell survival by inhibiting peripheral deletion. J Immunol. 2000;164:107–12. doi: 10.4049/jimmunol.164.1.107. [DOI] [PubMed] [Google Scholar]
  • 125.Weatherill AR, Maxwell JR, Takahashi C, et al. OX40 ligation enhances cell cycle turnover of Ag-activated CD4 T cells in vivo. Cell Immunol. 2001;209:63–75. doi: 10.1006/cimm.2001.1783. [DOI] [PubMed] [Google Scholar]
  • 126.Lathrop SK, Huddleston CA, Dullforce PA, et al. A signal through OX40 (CD134) allows anergic, autoreactive T cells to acquire effector cell functions. J Immunol. 2004;172:6735–43. doi: 10.4049/jimmunol.172.11.6735. [DOI] [PubMed] [Google Scholar]
  • 127.Huddleston CA, Weinberg AD, Parker DC. OX40 (CD134) engagement drives differentiation of CD4+ T cells to effector cells. Eur J Immunol. 2006;36:1093–103. doi: 10.1002/eji.200535637. [DOI] [PubMed] [Google Scholar]
  • 128.Redmond WL, Gough MJ, Charbonneau B, et al. Defects in the acquisition of CD8 T cell effector function after priming with tumor or soluble antigen can be overcome by the addition of an OX40 agonist. J Immunol. 2007;179:7244–53. doi: 10.4049/jimmunol.179.11.7244. [DOI] [PubMed] [Google Scholar]
  • 129.Bandyopadhyay S, Long M, Qui HZ, et al. Self-antigen prevents CD8 T cell effector differentiation by CD134 and CD137 dual costimulation. J Immunol. 2008;181:7728–37. doi: 10.4049/jimmunol.181.11.7728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Qui HZ, Hagymasi AT, Bandyopadhyay S, et al. CD134 plus CD137 dual costimulation induces Eomesodermin in CD4 T cells to program cytotoxic Th1 differentiation. J Immunol. 2011;187:3555–64. doi: 10.4049/jimmunol.1101244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Melero I, Shuford WW, Ashe N, et al. Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nature Med. 1997;3:682–5. doi: 10.1038/nm0697-682. [DOI] [PubMed] [Google Scholar]
  • 132.Cohen AD, Diab A, Perales MA, et al. Agonist anti-GITR antibody enhances vaccine-induced CD8(+) T-cell responses and tumor immunity. Cancer Res. 2006;66:4904–12. doi: 10.1158/0008-5472.CAN-05-2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Roberts DJ, Franklin NA, Kingeter LM, et al. Control of established melanoma by CD27 stimulation is associated with enhanced effector function and persistence, and reduced PD-1 expression of tumor infiltrating CD8(+) T cells. J Immunother. 2010;33:769–79. doi: 10.1097/CJI.0b013e3181ee238f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.de Vos S, Forero-Torres A, Ansell SM, et al. A phase II study of dacetuzumab (SGN-40) in patients with relapsed diffuse large B-cell lymphoma (DLBCL) and correlative analyses of patient-specific factors. J Hematol Oncol. 2014;7:44. doi: 10.1186/1756-8722-7-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Advani R, Forero-Torres A, Furman RR, et al. Phase I study of the humanized anti-CD40 monoclonal antibody dacetuzumab in refractory or recurrent non-Hodgkin’s lymphoma. J Clin Oncol. 2009;27:4371–7. doi: 10.1200/JCO.2008.21.3017. [DOI] [PubMed] [Google Scholar]
  • 136.Furman RR, Forero-Torres A, Shustov A, Drachman JG. A phase I study of dacetuzumab (SGN-40, a humanized anti-CD40 monoclonal antibody) in patients with chronic lymphocytic leukemia. Leuk Lymphoma. 2010;51:228–35. doi: 10.3109/10428190903440946. [DOI] [PubMed] [Google Scholar]
  • 137.Hussein M, Berenson JR, Niesvizky R, et al. A phase I multidose study of dacetuzumab (SGN-40; humanized anti-CD40 monoclonal antibody) in patients with multiple myeloma. Haematologica. 2010;95:845–8. doi: 10.3324/haematol.2009.008003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138*.Vonderheide RH, Burg JM, Mick R, et al. Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors. Oncoimmunology. 2013;2:e23033. doi: 10.4161/onci.23033. Phase I study demonstrating FcR-independent activity with CP-870,893 due to IgG2 isotype. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res. 2013;19:6286–95. doi: 10.1158/1078-0432.CCR-13-1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140*.Curti BD, Kovacsovics-Bankowski M, Morris N, et al. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013;73:7189–98. doi: 10.1158/0008-5472.CAN-12-4174. Phase I study of CD134, which showed evidence of tumor regression in 12 of 30 patients with a single dose. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ascierto PA, Simeone E, Sznol M, et al. Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol. 2010;37:508–16. doi: 10.1053/j.seminoncol.2010.09.008. [DOI] [PubMed] [Google Scholar]
  • 142.Schaer DA, Hirschhorn-Cymerman D, Wolchok JD. Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy. J Immunother Cancer. 2014;2:7. doi: 10.1186/2051-1426-2-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wajant H. Principles of antibody-mediated TNF receptor activation. Cell Death Differ. 2015;22:1727–41. doi: 10.1038/cdd.2015.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Marquez-Rodas I, Cerezuela P, Soria A, et al. Immune checkpoint inhibitors: therapeutic advances in melanoma. Ann Transl Med. 2015;3:267. doi: 10.3978/j.issn.2305-5839.2015.10.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145*.Adler AJ, Vella AT. Betting on improved cancer immunotherapy by doubling down on CD134 and CD137 co-stimulation. Oncoimmunology. 2013;2:e22837. doi: 10.4161/onci.22837. Discusses preclinical antitumor synergy observed with CD134 plus CD137 dual costimulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Lee SJ, Myers L, Muralimohan G, et al. 4-1BB and OX40 dual costimulation synergistically stimulate primary specific CD8 T cells for robust effector function. J Immunol. 2004;173:3002–12. doi: 10.4049/jimmunol.173.5.3002. [DOI] [PubMed] [Google Scholar]
  • 147.Lee SJ, Rossi RJ, Lee SK, et al. CD134 Costimulation Couples the CD137 Pathway to Induce Production of Supereffector CD8 T Cells That Become IL-7 Dependent. J Immunol. 2007;179:2203–14. doi: 10.4049/jimmunol.179.4.2203. [DOI] [PubMed] [Google Scholar]
  • 148.Munks MW, Mourich DV, Mittler RS, et al. 4-1BB and OX40 stimulation enhance CD8 and CD4 T-cell responses to a DNA prime, poxvirus boost vaccine. Immunology. 2004;112:559–66. doi: 10.1111/j.1365-2567.2004.01917.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Cuadros C, Dominguez AL, Lollini PL, et al. Vaccination with dendritic cells pulsed with apoptotic tumors in combination with anti-OX40 and anti-4-1BB monoclonal antibodies induces T cell-mediated protective immunity in Her-2/neu transgenic mice. Int J Cancer. 2005;116:934–43. doi: 10.1002/ijc.21098. [DOI] [PubMed] [Google Scholar]
  • 150.Gray JC, French RR, James S, et al. Optimising anti-tumour CD8 T-cell responses using combinations of immunomodulatory antibodies. Eur J Immunol. 2008;38:2499–511. doi: 10.1002/eji.200838208. [DOI] [PubMed] [Google Scholar]
  • 151.Uno T, Takeda K, Kojima Y, et al. Eradication of established tumors in mice by a combination antibody-based therapy. Nat Med. 2006;12:693–8. doi: 10.1038/nm1405. [DOI] [PubMed] [Google Scholar]
  • 152.Curran MA, Kim M, Montalvo W, et al. Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. PLoS One. 2011;6:e19499. doi: 10.1371/journal.pone.0019499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Guo Z, Wang X, Cheng D, et al. PD-1 blockade and OX40 triggering synergistically protects against tumor growth in a murine model of ovarian cancer. PLoS One. 2014;9:e89350. doi: 10.1371/journal.pone.0089350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154**.Weber JS, Yang JC, Atkins MB, Disis ML. Toxicities of Immunotherapy for the Practitioner. J Clin Oncol. 2015;33:2092–9. doi: 10.1200/JCO.2014.60.0379. Reviews adverse events associated with immunotherapy, with a clinically-oriented focus; will be of interest to practicing physicians. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Rosenberg SA, White DE. Vitiligo in patients with melanoma: normal tissue antigens can be targets for cancer immunotherapy. J Immunother Emphasis Tumor Immunol. 1996;19:81–4. [PubMed] [Google Scholar]
  • 156.Quirk SK, Shure AK, Agrawal DK. Immune-mediated adverse events of anticytotoxic T lymphocyte-associated antigen 4 antibody therapy in metastatic melanoma. Transl Res. 2015;166:412–24. doi: 10.1016/j.trsl.2015.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Robert C, Soria JC, Eggermont AM. Drug of the year: programmed death-1 receptor/programmed death-1 ligand-1 receptor monoclonal antibodies. Eur J Cancer. 2013;49:2968–71. doi: 10.1016/j.ejca.2013.07.001. [DOI] [PubMed] [Google Scholar]
  • 158.Curtsinger JM, Gerner MY, Lins DC, Mescher MF. Signal 3 availability limits the CD8 T cell response to a solid tumor. J Immunol. 2007;178:6752–60. doi: 10.4049/jimmunol.178.11.6752. [DOI] [PubMed] [Google Scholar]
  • 159.Lacy MQ, Jacobus S, Blood EA, et al. Phase II study of interleukin-12 for treatment of plateau phase multiple myeloma (E1A96): a trial of the Eastern Cooperative Oncology Group. Leuk Res. 2009;33:1485–9. doi: 10.1016/j.leukres.2009.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160**.Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018–28. doi: 10.1056/NEJMoa063842. Reports results for the TGN1412 trial and discusses the adverse events that occurred. [DOI] [PubMed] [Google Scholar]
  • 161.Bartholomaeus P, Semmler LY, Bukur T, et al. Cell contact-dependent priming and Fc interaction with CD32+ immune cells contribute to the TGN1412-triggered cytokine response. J Immunol. 2014;192:2091–8. doi: 10.4049/jimmunol.1302461. [DOI] [PubMed] [Google Scholar]
  • 162.Hussain K, Hargreaves CE, Roghanian A, et al. Upregulation of FcgammaRIIb on monocytes is necessary to promote the superagonist activity of TGN1412. Blood. 2015;125:102–10. doi: 10.1182/blood-2014-08-593061. [DOI] [PubMed] [Google Scholar]
  • 163.Sandilands GP, Wilson M, Huser C, et al. Were monocytes responsible for initiating the cytokine storm in the TGN1412 clinical trial tragedy? Clin Exp Immunol. 2010;162:516–27. doi: 10.1111/j.1365-2249.2010.04264.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Puellmann K, Beham AW, Kaminski WE. Cytokine storm and an anti-CD28 monoclonal antibody. N Engl J Med. 2006;355:2592–3. doi: 10.1056/NEJMc062750. author reply 93–4. [DOI] [PubMed] [Google Scholar]
  • 165.Winkler U, Jensen M, Manzke O, et al. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8) Blood. 1999;94:2217–24. [PubMed] [Google Scholar]
  • 166.Gaston RS, Deierhoi MH, Patterson T, et al. OKT3 first-dose reaction: association with T cell subsets and cytokine release. Kidney Int. 1991;39:141–8. doi: 10.1038/ki.1991.18. [DOI] [PubMed] [Google Scholar]
  • 167.Wing MG, Moreau T, Greenwood J, et al. Mechanism of first-dose cytokine-release syndrome by CAMPATH 1-H: involvement of CD16 (FcgammaRIII) and CD11a/CD18 (LFA-1) on NK cells. J Clin Invest. 1996;98:2819–26. doi: 10.1172/JCI119110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168**.Vogelpoel LT, Baeten DL, de Jong EC, den Dunnen J. Control of cytokine production by human fc gamma receptors: implications for pathogen defense and autoimmunity. Front Immunol. 2015;6:79. doi: 10.3389/fimmu.2015.00079. Reviews FcγRs and their role in cytokine secretion. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Swisher JF, Feldman GM. The many faces of FcgammaRI: implications for therapeutic antibody function. Immunol Rev. 2015;268:160–74. doi: 10.1111/imr.12334. [DOI] [PubMed] [Google Scholar]
  • 170.Mirsoian A, Bouchlaka MN, Sckisel GD, et al. Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J Exp Med. 2014;211:2373–83. doi: 10.1084/jem.20140116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Niu L, Strahotin S, Hewes B, et al. Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and thrombocytopenia in anti-CD137-treated mice. J Immunol. 2007;178:4194–213. doi: 10.4049/jimmunol.178.7.4194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Lee SW, Salek-Ardakani S, Mittler RS, Croft M. Hypercostimulation through 4-1BB distorts homeostasis of immune cells. J Immunol. 2009;182:6753–62. doi: 10.4049/jimmunol.0803241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Melero I, Johnston JV, Shufford WW, et al. NK1, 1 cells express 4-1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by anti-4-1BB monoclonal antibodies. Cell Immunol. 1998;190:167–72. doi: 10.1006/cimm.1998.1396. [DOI] [PubMed] [Google Scholar]
  • 174.Futagawa T, Akiba H, Kodama T, et al. Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells. Int Immunol. 2002;14:275–86. doi: 10.1093/intimm/14.3.275. [DOI] [PubMed] [Google Scholar]
  • 175.Pauly S, Broll K, Wittmann M, et al. CD137 is expressed by follicular dendritic cells and costimulates B lymphocyte activation in germinal centers. J Leukoc Biol. 2002;72:35–42. [PubMed] [Google Scholar]
  • 176.Downey SG, Klapper JA, Smith FO, et al. Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin Cancer Res. 2007;13:6681–8. doi: 10.1158/1078-0432.CCR-07-0187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Pages C, Gornet JM, Monsel G, et al. Ipilimumab-induced acute severe colitis treated by infliximab. Melanoma Res. 2013;23:227–30. doi: 10.1097/CMR.0b013e32835fb524. [DOI] [PubMed] [Google Scholar]
  • 178**.Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol. 2008;8:34–47. doi: 10.1038/nri2206. Reviews FcγRs and their functional role in immunity. [DOI] [PubMed] [Google Scholar]
  • 179.Ravetch JV, Bolland S. IgG Fc receptors. Annu Rev Immunol. 2001;19:275–90. doi: 10.1146/annurev.immunol.19.1.275. [DOI] [PubMed] [Google Scholar]
  • 180**.Stewart R, Hammond S, Oberst M, Wilkinson R. The role of Fc gamma receptors in the activity of immunomodulatory antibodies for cancer. J Immunother Cancer. 2014;2:1–10. Reviews FcγRs, their role in therapy, and translational considerations. [Google Scholar]
  • 181.Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity. 2005;23:41–51. doi: 10.1016/j.immuni.2005.05.010. [DOI] [PubMed] [Google Scholar]
  • 182.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 183.Ivashkiv LB. How ITAMs inhibit signaling. Sci Signal. 2011;4:pe20. doi: 10.1126/scisignal.2001917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184*.Blank U, Launay P, Benhamou M, Monteiro RC. Inhibitory ITAMs as novel regulators of immunity. Immunol Rev. 2009;232:59–71. doi: 10.1111/j.1600-065X.2009.00832.x. Discusses what is known about ITAMi signaling. [DOI] [PubMed] [Google Scholar]
  • 185.Ben Mkaddem S, Hayem G, Jonsson F, et al. Shifting FcgammaRIIA-ITAM from activation to inhibitory configuration ameliorates arthritis. J Clin Invest. 2014;124:3945–59. doi: 10.1172/JCI74572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Fehr J, Hofmann V, Kappeler U. Transient reversal of thrombocytopenia in idiopathic thrombocytopenic purpura by high-dose intravenous gamma globulin. N Engl J Med. 1982;306:1254–8. doi: 10.1056/NEJM198205273062102. [DOI] [PubMed] [Google Scholar]
  • 187.Gelfand EW. Intravenous immune globulin in autoimmune and inflammatory diseases. N Engl J Med. 2012;367:2015–25. doi: 10.1056/NEJMra1009433. [DOI] [PubMed] [Google Scholar]
  • 188.Aloulou M, Ben Mkaddem S, Biarnes-Pelicot M, et al. IgG1 and IVIg induce inhibitory ITAM signaling through FcgammaRIII controlling inflammatory responses. Blood. 2012;119:3084–96. doi: 10.1182/blood-2011-08-376046. [DOI] [PubMed] [Google Scholar]
  • 189**.Furness AJ, Vargas FA, Peggs KS, Quezada SA. Impact of tumour microenvironment and Fc receptors on the activity of immunomodulatory antibodies. Trends Immunol. 2014;35:290–8. doi: 10.1016/j.it.2014.05.002. Reviews FcγRs, their role within the TME and contributions to efficacy, and discusses clinical development and synergy. [DOI] [PubMed] [Google Scholar]
  • 190.Kim JM, Ashkenazi A. Fcgamma receptors enable anticancer action of proapoptotic and immune-modulatory antibodies. J Exp Med. 2013;210:1647–51. doi: 10.1084/jem.20131625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:235–42. doi: 10.1056/NEJMoa011795. [DOI] [PubMed] [Google Scholar]
  • 192.McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16:2825–33. doi: 10.1200/JCO.1998.16.8.2825. [DOI] [PubMed] [Google Scholar]
  • 193.Sorbye SW, Kilvaer T, Valkov A, et al. High expression of CD20+ lymphocytes in soft tissue sarcomas is a positive prognostic indicator. Oncoimmunology. 2012;1:75–77. doi: 10.4161/onci.1.1.17825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Weiner GJ. Building better monoclonal antibody-based therapeutics. Nat Rev Cancer. 2015;15:361–70. doi: 10.1038/nrc3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195*.Marabelle A, Kohrt H, Sagiv-Barfi I, et al. Depleting tumor-specific Tregs at a single site eradicates disseminated tumors. J Clin Invest. 2013;123:2447–63. doi: 10.1172/JCI64859. Showed that intratumoral co-injection of anti-OX40 and anti-CTLA-4 induced Treg depletion and generated a systemic antitumor response. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196*.Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1:32–42. doi: 10.1158/2326-6066.CIR-13-0013. Showed that IgG2a isotype anti-CTLA-4 reduces intratumoral Tregs, whereas an FcγR-nonbinding IgG1 isotype did not. [DOI] [PubMed] [Google Scholar]
  • 197*.Bulliard Y, Jolicoeur R, Windman M, et al. Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med. 2013;210:1685–93. doi: 10.1084/jem.20130573. Showed that activating FcγRs were necessary for antitumor efficacy of anti-GITR and anti-CTLA-4 mAbs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Zhou X, Bailey-Bucktrout SL, Jeker LT, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–7. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199*.Voo KS, Bover L, Harline ML, et al. Antibodies targeting human OX40 expand effector T cells and block inducible and natural regulatory T cell function. J Immunol. 2013;191:3641–50. doi: 10.4049/jimmunol.1202752. Shows that human anti-CD134 inhibit de novo Treg induction, expansion, and suppressive function. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Schaer DA, Budhu S, Liu C, et al. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013;1:320–31. doi: 10.1158/2326-6066.CIR-13-0086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Mittal P, St Rose MC, Wang X, et al. Tumor-Unrelated CD4 T Cell Help Augments CD134 Plus CD137 Dual Costimulation Tumor Therapy. J Immunol. 2015;195:5816–26. doi: 10.4049/jimmunol.1502032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Akhmetzyanova I, Zelinskyy G, Littwitz-Salomon E, et al. CD137 Agonist Therapy Can Reprogram Regulatory T Cells into Cytotoxic CD4+ T Cells with Antitumor Activity. J Immunol. 2016;196:484–92. doi: 10.4049/jimmunol.1403039. [DOI] [PubMed] [Google Scholar]
  • 203.Regnault A, Lankar D, Lacabanne V, et al. Fcgamma receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization. J Exp Med. 1999;189:371–80. doi: 10.1084/jem.189.2.371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Kalergis AM, Ravetch JV. Inducing tumor immunity through the selective engagement of activating Fcgamma receptors on dendritic cells. J Exp Med. 2002;195:1653–9. doi: 10.1084/jem.20020338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205*.Wilson NS, Yang B, Yang A, et al. An Fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 2011;19:101–13. doi: 10.1016/j.ccr.2010.11.012. Provided first evidence that FcγRs can play a role in cancer immunotherapy not mediated by ADCC (i.e., immunomodulation) [DOI] [PubMed] [Google Scholar]
  • 206.Li F, Ravetch JV. Inhibitory Fcgamma receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies. Science. 2011;333:1030–4. doi: 10.1126/science.1206954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.White AL, Chan HT, Roghanian A, et al. Interaction with FcgammaRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody. J Immunol. 2011;187:1754–63. doi: 10.4049/jimmunol.1101135. [DOI] [PubMed] [Google Scholar]
  • 208**.Li F, Ravetch JV. Antitumor activities of agonistic anti-TNFR antibodies require differential FcgammaRIIB coengagement in vivo. Proc Natl Acad Sci U S A. 2013;110:19501–6. doi: 10.1073/pnas.1319502110. Describes the trans-acting role of FcγRIIB and differential requirements for efficacy for various TNFR agonists. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209**.White AL, Chan HT, French RR, et al. Conformation of the human immunoglobulin G2 hinge imparts superagonistic properties to immunostimulatory anticancer antibodies. Cancer Cell. 2015;27:138–48. doi: 10.1016/j.ccell.2014.11.001. Characterizes the FcγR-independent activity of human IgG2 mAbs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Vitale LA, He LZ, Thomas LJ, et al. Development of a human monoclonal antibody for potential therapy of CD27-expressing lymphoma and leukemia. Clin Cancer Res. 2012;18:3812–21. doi: 10.1158/1078-0432.CCR-11-3308. [DOI] [PubMed] [Google Scholar]
  • 211.Bulliard Y, Jolicoeur R, Zhang J, et al. OX40 engagement depletes intratumoral Tregs via activating FcgammaRs, leading to antitumor efficacy. Immunol Cell Biol. 2014;92:475–80. doi: 10.1038/icb.2014.26. [DOI] [PubMed] [Google Scholar]
  • 212*.Sallin MA, Zhang X, So EC, et al. The anti-lymphoma activities of anti-CD137 monoclonal antibodies are enhanced in FcgammaRIII(-/-) mice. Cancer Immunol Immunother. 2014;63:947–58. doi: 10.1007/s00262-014-1567-2. Shows that activating FcγRs diminish efficacy of 4-1BB agonists, contrary to agonists of other TNFRs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Lin W, Voskens CJ, Zhang X, et al. Fc-dependent expression of CD137 on human NK cells: insights into “agonistic” effects of anti-CD137 monoclonal antibodies. Blood. 2008;112:699–707. doi: 10.1182/blood-2007-11-122465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214**.Dahan R, Sega E, Engelhardt J, et al. FcgammaRs Modulate the Anti-tumor Activity of Antibodies Targeting the PD-1/PD-L1 Axis. Cancer Cell. 2015;28:285–95. doi: 10.1016/j.ccell.2015.08.004. Identifies the different FcR requirements for efficacy of mAbs targeting PD-1 and PD-L1. [DOI] [PubMed] [Google Scholar]
  • 215**.Bruhns P. Properties of mouse and human IgG receptors and their contribution to disease models. Blood. 2012;119:5640–9. doi: 10.1182/blood-2012-01-380121. Discusses FcγRs in both mouse and human, and how they may be involved in different diseases. [DOI] [PubMed] [Google Scholar]
  • 216**.Offringa R, Glennie MJ. Development of Next-Generation Immunomodulatory Antibodies for Cancer Therapy through Optimization of the IgG Framework. Cancer Cell. 2015;28:273–5. doi: 10.1016/j.ccell.2015.08.008. Provides a concise review of FcR requirements of various immunomodulatory mAbs, including those targeting PD-1 and PD-L1. [DOI] [PubMed] [Google Scholar]
  • 217.Yuan J, Adamow M, Ginsberg BA, et al. Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc Natl Acad Sci U S A. 2011;108:16723–8. doi: 10.1073/pnas.1110814108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Kelderman S, Heemskerk B, van Tinteren H, et al. Lactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanoma. Cancer Immunol Immunother. 2014;63:449–58. doi: 10.1007/s00262-014-1528-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Vetizou M, Pitt JM, Daillere R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–84. doi: 10.1126/science.aad1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99. doi: 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res. 2015;3:288–95. doi: 10.1158/2326-6066.CIR-14-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Queirolo P, Morabito A, Laurent S, et al. Association of CTLA-4 polymorphisms with improved overall survival in melanoma patients treated with CTLA-4 blockade: a pilot study. Cancer Invest. 2013;31:336–45. doi: 10.3109/07357907.2013.793699. [DOI] [PubMed] [Google Scholar]
  • 223.Srivastava PK, Duan F. Harnessing the antigenic fingerprint of each individual cancer for immunotherapy of human cancer: genomics shows a new way and its challenges. Cancer Immunol Immunother. 2013;62:967–74. doi: 10.1007/s00262-013-1422-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Weiner GJ. Rituximab: mechanism of action. Semin Hematol. 2010;47:115–23. doi: 10.1053/j.seminhematol.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225*.Eastwood D, Findlay L, Poole S, et al. Monoclonal antibody TGN1412 trial failure explained by species differences in CD28 expression on CD4+ effector memory T-cells. Br J Pharmacol. 2010;161:512–26. doi: 10.1111/j.1476-5381.2010.00922.x. Exemplifies shortcomings of preclinical models, and also illustrates how receptor expression levels can give rise to heterogeneity of efficacy and adverse events. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Stebbings R, Eastwood D, Poole S, Thorpe R. After TGN1412: recent developments in cytokine release assays. J Immunotoxicol. 2013;10:75–82. doi: 10.3109/1547691X.2012.711783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Hoos A, Eggermont AM, Janetzki S, et al. Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst. 2010;102:1388–97. doi: 10.1093/jnci/djq310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Hoos A, Britten C. The immuno-oncology framework: Enabling a new era of cancer therapy. Oncoimmunology. 2012;1:334–39. doi: 10.4161/onci.19268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Alatrash G, Jakher H, Stafford PD, Mittendorf EA. Cancer immunotherapies, their safety and toxicity. Expert Opin Drug Saf. 2013;12:631–45. doi: 10.1517/14740338.2013.795944. [DOI] [PubMed] [Google Scholar]
  • 230.Old LJ. Tumor necrosis factor (TNF) Science. 1985;230:630–2. doi: 10.1126/science.2413547. [DOI] [PubMed] [Google Scholar]
  • 231.Calzascia T, Pellegrini M, Hall H, et al. TNF-alpha is critical for antitumor but not antiviral T cell immunity in mice. J Clin Invest. 2007;117:3833–45. doi: 10.1172/JCI32567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Poehlein CH, Hu HM, Yamada J, et al. TNF plays an essential role in tumor regression after adoptive transfer of perforin/IFN-gamma double knockout effector T cells. J Immunol. 2003;170:2004–13. doi: 10.4049/jimmunol.170.4.2004. [DOI] [PubMed] [Google Scholar]
  • 233.Hotamisligil GS, Spiegelman BM. Tumor necrosis factor alpha: a key component of the obesity-diabetes link. Diabetes. 1994;43:1271–8. doi: 10.2337/diab.43.11.1271. [DOI] [PubMed] [Google Scholar]
  • 234.Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–20. doi: 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
  • 235.Weber J, Thompson JA, Hamid O, et al. A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res. 2009;15:5591–8. doi: 10.1158/1078-0432.CCR-09-1024. [DOI] [PubMed] [Google Scholar]
  • 236.Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–18. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Teachey DT, Rheingold SR, Maude SL, et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood. 2013;121:5154–7. doi: 10.1182/blood-2013-02-485623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 2014;20:119–22. doi: 10.1097/PPO.0000000000000035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Reck M, Bondarenko I, Luft A, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol. 2013;24:75–83. doi: 10.1093/annonc/mds213. [DOI] [PubMed] [Google Scholar]
  • 240.Slovin SF, Higano CS, Hamid O, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013;24:1813–21. doi: 10.1093/annonc/mdt107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241**.Melero I, Berman DM, Aznar MA, et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015;15:457–72. doi: 10.1038/nrc3973. Comprehensive review of combination therapies and their clinical development. [DOI] [PubMed] [Google Scholar]
  • 242.Kwon B. Regulation of Inflammation by Bidirectional Signaling through CD137 and Its Ligand. Immune Netw. 2012;12:176–80. doi: 10.4110/in.2012.12.5.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Reali C, Curto M, Sogos V, et al. Expression of CD137 and its ligand in human neurons, astrocytes, and microglia: modulation by FGF-2. J Neurosci Res. 2003;74:67–73. doi: 10.1002/jnr.10727. [DOI] [PubMed] [Google Scholar]
  • 244.Teijeira A, Palazon A, Garasa S, et al. CD137 on inflamed lymphatic endothelial cells enhances CCL21-guided migration of dendritic cells. FASEB J. 2012;26:3380–92. doi: 10.1096/fj.11-201061. [DOI] [PubMed] [Google Scholar]
  • 245.Olofsson PS, Soderstrom LA, Wagsater D, et al. CD137 is expressed in human atherosclerosis and promotes development of plaque inflammation in hypercholesterolemic mice. Circulation. 2008;117:1292–301. doi: 10.1161/CIRCULATIONAHA.107.699173. [DOI] [PubMed] [Google Scholar]
  • 246.Ilzecka J. Serum soluble OX40 in patients with amyotrophic lateral sclerosis. Acta Clin Croat. 2012;51:3–7. [PubMed] [Google Scholar]
  • 247.Tu TH, Kim CS, Kang JH, et al. Levels of 4-1BB transcripts and soluble 4-1BB protein are elevated in the adipose tissue of human obese subjects and are associated with inflammatory and metabolic parameters. Int J Obes (Lond) 2014;38:1075–82. doi: 10.1038/ijo.2013.222. [DOI] [PubMed] [Google Scholar]
  • 248.Yan J, Wang C, Chen R, Yang H. Clinical implications of elevated serum soluble CD137 levels in patients with acute coronary syndrome. Clinics (Sao Paulo) 2013;68:193–8. doi: 10.6061/clinics/2013(02)OA12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Miyajima I, Dombrowicz D, Martin TR, et al. Systemic anaphylaxis in the mouse can be mediated largely through IgG1 and Fc gammaRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE- or IgG1-dependent passive anaphylaxis. J Clin Invest. 1997;99:901–14. doi: 10.1172/JCI119255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Heller T, Gessner JE, Schmidt RE, et al. Cutting edge: Fc receptor type I for IgG on macrophages and complement mediate the inflammatory response in immune complex peritonitis. J Immunol. 1999;162:5657–61. [PubMed] [Google Scholar]
  • 251**.Mellor JD, Brown MP, Irving HR, et al. A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer. J Hematol Oncol. 2013;6:1. doi: 10.1186/1756-8722-6-1. Discusses how different FcγR allotypes can give rise to patient response heterogeneity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252**.White AL, Dou L, Chan HT, et al. Fcgamma receptor dependency of agonistic CD40 antibody in lymphoma therapy can be overcome through antibody multimerization. J Immunol. 2014;193:1828–35. doi: 10.4049/jimmunol.1303204. Shows that the FcγR requirements for anti-CD40 agonist are not absolute and can be overcome using strategies such as multimerization. [DOI] [PubMed] [Google Scholar]
  • 253.Stavenhagen JB, Gorlatov S, Tuaillon N, et al. Fc optimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcgamma receptors. Cancer Res. 2007;67:8882–90. doi: 10.1158/0008-5472.CAN-07-0696. [DOI] [PubMed] [Google Scholar]
  • 254.Zalevsky J, Leung IW, Karki S, et al. The impact of Fc engineering on an anti-CD19 antibody: increased Fcgamma receptor affinity enhances B-cell clearing in nonhuman primates. Blood. 2009;113:3735–43. doi: 10.1182/blood-2008-10-182048. [DOI] [PubMed] [Google Scholar]
  • 255.Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 2005;310:1510–2. doi: 10.1126/science.1118948. [DOI] [PubMed] [Google Scholar]
  • 256.Mossner E, Brunker P, Moser S, et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity. Blood. 2010;115:4393–402. doi: 10.1182/blood-2009-06-225979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257**.Mimoto F, Katada H, Kadono S, et al. Engineered antibody Fc variant with selectively enhanced FcgammaRIIb binding over both FcgammaRIIa(R131) and FcgammaRIIa(H131) Protein Eng Des Sel. 2013;26:589–98. doi: 10.1093/protein/gzt022. Demonstrates that modification to mAb Fc domains can impact FcγR binding and enhance or diminish activity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Sutterwala FS, Noel GJ, Salgame P, Mosser DM. Reversal of proinflammatory responses by ligating the macrophage Fcgamma receptor type I. J Exp Med. 1998;188:217–22. doi: 10.1084/jem.188.1.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259**.Kinder M, Greenplate AR, Strohl WR, et al. An Fc engineering approach that modulates antibody-dependent cytokine release without altering cell-killing functions. mAbs. 2015;7:494–504. doi: 10.1080/19420862.2015.1022692. Shows that modification of Fc domains can selectively alter FcγR-mediated cytokine release without impacting ADCC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Richman LP, Vonderheide RH. Anti-human CD40 monoclonal antibody therapy is potent without FcR crosslinking. Oncoimmunology. 2014;3:e28610. doi: 10.4161/onci.28610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261*.Richman LP, Vonderheide RH. Role of crosslinking for agonistic CD40 monoclonal antibodies as immune therapy of cancer. Cancer Immunol Res. 2014;2:19–26. doi: 10.1158/2326-6066.CIR-13-0152. Shows that the efficacy of anti-CD40 human IgG2 agonist CP-870,893 is FcγR-independent. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Spira AI, Park K, Mazieres J. Efficacy, safety and predictive biomarker results from a randomized phase II study comparing atezolizumab vs docetaxel in 2L/3L NSCLC (POPLAR) J Clin Oncol. 2015;33(suppl) Abstract 8010. [Google Scholar]
  • 263.Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]

RESOURCES