You are on page 1of 10

Expert Opinion on Biological Therapy

ISSN: 1471-2598 (Print) 1744-7682 (Online) Journal homepage: https://www.tandfonline.com/loi/iebt20

Avelumab: combining immune checkpoint


inhibition and antibody-dependent cytotoxicity

Gerhard Hamilton & Barbara Rath

To cite this article: Gerhard Hamilton & Barbara Rath (2017) Avelumab: combining immune
checkpoint inhibition and antibody-dependent cytotoxicity, Expert Opinion on Biological Therapy,
17:4, 515-523, DOI: 10.1080/14712598.2017.1294156

To link to this article: https://doi.org/10.1080/14712598.2017.1294156

Accepted author version posted online: 09


Feb 2017.
Published online: 22 Feb 2017.

Submit your article to this journal

Article views: 1066

View Crossmark data

Citing articles: 15 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iebt20
EXPERT OPINION ON BIOLOGICAL THERAPY, 2017
VOL. 17, NO. 4, 515–523
http://dx.doi.org/10.1080/14712598.2017.1294156

DRUG EVALUATION

Avelumab: combining immune checkpoint inhibition and antibody-dependent


cytotoxicity
Gerhard Hamilton and Barbara Rath
Department of Surgery, Medical University of Vienna, Vienna, Austria

ABSTRACT ARTICLE HISTORY


Introduction: Immune checkpoint inhibition holds great promise for selected tumors. The human Received 25 November 2016
monoclonal antibody (mAB) avelumab is directed to programmed death ligand-1 (PD-L1) and is Accepted 8 February 2017
supposed to inhibit the immunosuppressive PD-L1/PD-1 interaction and, furthermore, effect anti- KEYWORDS
body-dependent cytotoxicity (ADCC) lysis of tumor cells. Avelumab; immune
Areas covered: This article presents an overview of the current means to activate the antitumor checkpoints; programmed
immune defense by targeting PD-1 or PD-L1 with mABs and their possible role in ADCC-mediated death ligand-1;
tumor cell elimination. antibody-dependent
Expert opinion: Avelumab contains a Fc region which can bind cognate receptors on immune effector cytotoxicity
cells and induce ADCC-mediated tumor cell lysis, in contrast to other mABs directed to PD-1/PD-L1
which lack the ability to trigger ADCC due to belonging to the IgG4 subclass or possessing a mutated Fc
region. Preclinical and clinical data indicate that avelumab can be safely administered to cancer patients
with a toxicity profile comparable to other mABs and without lysis of PD-L1-positive activated immune
cells. This antibody yielded durable responses in a phase II trial in advanced Merkel cell carcinoma
patients. Tumor cell lysis by avelumab prevents cells from resorting to alternative checkpoints as shown
by targeting PD-1 and the upregulation of TIM-3.

1. Introduction by the immune system and imply that targeting negative


regulatory immune checkpoints would be beneficial for
A hallmark of the malignant phenotype of tumor cells
patients with preexisting T-cell-inflamed tumors [4]. For exam-
includes the ability to override the host immune system
ple, blockade of CTLA-4 or PD-1 has been associated with
and inactivate infiltrating cytotoxic effector cells [1,2]. The
significant clinical responses in metastatic melanoma patients
accumulation of tumor-infiltrating lymphocytes (TILs) within
[5]. CTLA-4 is homologous to the T-cell costimulatory protein,
primary and metastatic tumors could be demonstrated to
CD28, and both molecules bind to CD80/B7-1 and CD86/B7-2
comprise tumor antigen-specific T lymphocytes which can
on antigen-presenting cells (APCs), but CTLA-4 binds CD80
be expanded in vitro. Despite spontaneous antitumor immu-
and CD86 with greater affinity than CD28 and such impairs
nity in distinct tumor types, the disease progresses in most
the stimulatory signal of CD28 [3]. Similarly, binding of pro-
patients, thus indicating the failure of the immune response
grammed death-ligand 1 (PD-L1) to PD-1 or B7-1 transmits an
to control tumor growth and recurrence. Obviously, an
inhibitory signal which reduces the proliferation and survival
effective cellular immune response is blocked within the
of CTLs. Ipilimumab, a fully human monoclonal antibody
tumor microenvironment by inhibitory factors which would
(mAB) against CTLA-4, as well as nivolumab and pembrolizu-
have to be antagonized for a reconstitution of a successful
mab which target PD-1, have been shown to prolong overall
immune defense. Isolated TILs were shown to express a
survival (OS) in patients with advanced melanoma [5,6]. The
range of inhibitory receptors that under normal conditions
combination of a CTLA-4 and a PD-1 inhibitor seems to be
contribute to T-cell regulation. These receptors, including
superior to the monotherapies, especially in patients with PD-
cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), pro-
L1-negative tumors, but increases toxicity.
grammed cell death-1 (PD-1), lymphocyte-activation gene-3,
Metastatic lung cancer, including non–small-cell lung can-
and T-cell Immunoglobulin domain and Mucin domain 3
cer (NSCLC) and small-cell lung cancer, has a poor prognosis
(TIM-3), seem to inactivate the ongoing immune response
with a 5-year survival rate of less than 5% [7]. A subpopula-
and to prevent efficient elimination of the tumor cells under
tion of NSCLC patients whose tumors harbor specific muta-
attack [3].
tions in driver genes can be treated with molecularly
T-cell-inflamed tumors exhibit high expression of immuno-
targeted therapies resulting in improved OS. However, for a
suppressive mechanisms which follow the infiltration of cyto-
large group of NSCLC patients, therapeutic options have
toxic T lymphocytes (CTLs) as a negative feedback control.
been confined to surgery, chemotherapy, and irradiation
Thus, the immunosuppressive pathways are intrinsically driven
alone. Immunotherapies, which aim at eliciting immune-

CONTACT Gerhard Hamilton gerhard.hamilton@meduniwien.ac.at


© 2017 Informa UK Limited, trading as Taylor & Francis Group
516 G. HAMILTON AND B. RATH

mediated destruction of lung tumor cells, may be an alter-


native approach but has had minimal success in lung cancer Box 1. Drug summary.
in the past, leading to the assumption that lung cancer is
Drug name Avelumab
poorly immunogenic [8]. However, the novel targeting of Phase Pre-registration
immune checkpoint proteins by monoclonal antibodies in Indication Cancer
NSCLC was found to prolong survival of suitable patients Pharmacology description PD-L1 antagonist
Route of administration Injectable
significantly [9]. In general, application of immune check- Pivotal trial(s) [64]
point-directed therapy has applied to a growing number of
tumor types so far [10].
immune system attacks [17,18]. Additionally, PD-L1 is
2. Passive immunotherapy for cancer treatment expressed on the surface of resting T cells, B cells, DCs, macro-
phages, vascular endothelial cells, heart, lung, placenta, liver,
Passive immunotherapy is defined as use of agents, such as and pancreatic islet cells [13]. Normally, PD-L1 is involved in
monoclonal antibodies or adaptive cell therapy, that directly self-tolerance, such as protecting peripheral tissues from
target tumor in contrast to administration of cytokines or excess of inflammation and autoimmune pathologies [19,20].
vaccines [10]. CTLs are considered the effectors of immune PD-L1 is induced by various pro-inflammatory cytokines like
responses against tumors, and recognition of tumor antigens IFNγ, tumor necrosis factor-α (TNF-α), vascular endothelial
by specific T cells is a prerequisite for mounting an immune growth factor, granulocyte-macrophage colony-stimulating
cell attack. Naive CD8+ T cells are cross-primed by APCs, factor, and interleukin-10.
invoking a program leading to tumor-specific CTLs which A range of mABs against the checkpoint proteins have been
proliferate and invade the tumor sites. CTLs kill tumor cells developed, including the CTLA-4-directed ipilimumab and treme-
through release of granzymes, perforin, and ligands of the limumab, as well as the PD-1 inhibitors nivolumab and pembro-
tumor necrosis factor (TNF) superfamily, such as Fas ligand. lizumab and PD-L1 inhibitors atezolizumab (MPDL3280A),
The Th1 subpopulation of CD4+ T cells enhances the expan- durvalumab (MEDI4736), and avelumab (MSB0010718C) with
sion, priming, and infiltration of CD8+ T by producing large many more under development (Box 1) [10,21]. Detection of the
amounts of interferon-γ (IFNγ) and chemokines. Furthermore, expression of PD-1/PD-L1 in tumors by immunohistochemistry is
innate immunity effectors which comprise macrophages, gran- used as predictive biomarker of response to immune checkpoint
ulocytes, mast cells, dendritic cells (DCs), and natural killer (NK) therapy, but technical issues and clinical activity of antibodies in
cells can destroy tumor cells directly through several mechan- antigen-negative patients indicate limited reliability of this assay.
isms, including antibody-dependent cellular cytotoxicity Despite attempts for standardization of the immunohistochemical
(ADCC). techniques for confirmation of PD-1/PD-L1 expression, differing
Several checkpoint molecules exist which attenuate the reagents, protocols, and thresholds of positivity impede compar-
T-cell immune response to tumor-associated antigens, and ison of different trials [22].
mABs interacting with two of these molecular pathways, Furthermore, an important precondition for the signifi-
namely CTLA-4 and PD-1, have shown clinical activity in sev- cance of the immune checkpoint inhibitor expression as pre-
eral tumors [3,10]. Some patients with advanced solid tumors, dictor of a therapeutic response is the infiltration of the
like metastatic melanoma, exhibit a prolonged response to respective tumors by immune effector cells. Tumor load of
immunotherapy which lasts for many years after anti-CTLA-4 neoantigens, intratumoral heterogeneity of checkpoint inhibi-
antibody treatment [11]. CTLA-4 is a protein receptor tor expression, and distinct mutations constitute additional
expressed by activated CTLs which binds to CD80 (B7-1) or predictive markers under evaluation [23]. Anti-CTLA-4 antibo-
CD86 (B7-2) on APCs to delimit the activity of T cells under dies can be applied to stimulate immune cell infiltration in
normal conditions. Upregulation of CTLA-4 expression on T combination with PD-1/PD-L1-directed agents. Thus, ipilimu-
cells can occur in response to transforming growth factor-β mab (a CTLA-4 checkpoint inhibitor) and nivolumab (a PD-1
(TGFβ) during the early stage of tumorigenesis. mABs directed checkpoint inhibitor) have been shown to have complemen-
to this ligand inhibit CTLA-4-CD80/CD86 binding and activate tary activity in metastatic melanoma [24]. However, treatment-
CTLs through abolishment of this inhibitory circuit. The main related adverse events (AEs) of grade 3 or 4 occurred in 16.3%
side effects of an anti-CTLA-4 therapy included diarrhea, coli- of the patients in the nivolumab group, 27.3% in the ipilimu-
tis, transaminitis, and pituitary dysfunction [12]. A second mab group, but 55.0% in the antibody combination group,
immune checkpoint molecule, namely programmed death-1 demonstrating increased toxicity of the combined antibodies.
(PD-1), is a T-cell surface receptor expressed on B cells, NK Through blockade of the immune checkpoints, the balance
cells, activated monocytes, and DCs [13,14]. Normally, PD-1 between autoimmunity and immune tolerance is disturbed.
limits autoimmunity as a coinhibitory immune checkpoint Thus, the new immunotherapies are associated with immune-
but in tumors dampens the activity of TILs [15]. PD-1 binds meditated AEs comprising immune-mediated pneumonitis, colitis,
programmed PD-L1/B7-H1 and 2 (PD-L2/B7-DC), inducing hepatitis, endocrinopathies, rash, encephalitis among others [25].
T-cell anergy by interacting with the former receptor on Initiation of appropriate management, usually in the form of
APCs [16]. A range of cancers, including gastric cancer, renal immunosuppression, results in complete reversibility of side
cell cancer, bladder cancer, hepatocellular cancer, cutaneous effects, but inadequate treatment can lead to severe toxicity or
cancer, breast cancer, and melanoma, glioblastoma as well as even death.
NSCLC, express PD-L1 which provides protection against
EXPERT OPINION ON BIOLOGICAL THERAPY 517

2.1. Overview of the market for cancer checkpoint therapies has increasingly been visible, similarly to the situa-
immunotherapies tion observed with the targeted small-molecule inhibitors. In
tumors progressing after initial response to anti-PD-1 therapy,
High response rates and prolonged OS in cancer patients
upregulation of TIM-3, an alternative immune checkpoint
treated with checkpoint inhibitors led to an exponential
receptor, in PD-1 antibody-coated T cells was found [33].
growth of the respective drug development, clinical trials,
Anti-PD-1 antibodies were still active at the time of resistance,
and use of this class of anticancer agents. In 2014, the
but the new blockade could only be reversed by addition of a
NSCLC market was dominated by the chemotherapeutic
TIM-3-blocking antibody. Upregulation of TIM-3 expression
drug pemetrexed, erlotinib, and bevacizumab with costs of
after prolonged exposure to PD-1-blocking antibody was con-
$5.4 billion, but expense is expected to increase exponentially
fined to CTLs in the tumor microenvironment. Cancers seems
due to the novel immunotherapeutic drugs [26]. So far, three
to possess the ability to escape immune checkpoint inhibition
immune checkpoint inhibitors, namely ipilimumab (Yervoy;
by invoking pathways involving a set of alternative immune
Bristol-Myers Squibb, New York, NY, USA), nivolumab
receptors. Genomic and transcriptomic investigations may
(Opdivo; Bristol-Myers Squibb/Ono Pharmaceuticals, Osaka,
help to identify further positive correlates of response or
Japan), and pembrolizumab (Keytruda; Merck Co., Kenilworth,
resistance, such as a high mutational/neoantigen load of the
NJ, USA) are marketed for the treatment of malignant mela-
melanomas and NSCLC, as well as low intratumoral heteroge-
noma [27]. The two PD-1-specific mABs nivolumab and pem-
neity, among others. Resistant tumors have been shown to
brolizumab yield high responses (rates of 24–32%) which are
exhibit distinct alterations, such as loss of Phosphatase and
durable for the majority of patients and are associated with a
Tensin homolog (PTEN), modification of the Januskinase/
low number of AEs, consisting mainly of fatigue, anemia, and
Signal Transducers and Activators of Transcription (JAK/STAT)
dyspnea. The estimated cost of treating all American patients
pathway, and elevated expression of coinhibitory molecules
with nivolumab or pembrolizumab as calculated for data avail-
[23]. Additionally, the cancer–immunity cycle requires the
able in 2016 is $1.57 and $0.97 billion per year, respectively
release of tumor antigens, presentation of these antigens to
[28]. The immuno-oncology market will grow to >$27 billion in
invoke effector cell activation, trafficking, and infiltration of
2021, fueled by the administration of a range of checkpoint
such primed cells into the tumor as well as overcoming an
inhibitors across multiple solid tumor indications [29]. In 2022,
immunosuppressive microenvironment, and thus, tumor cell
the anti-PD-1 agents are expected to hold the highest market
kill is impaired by any failures to successful progress through
share at 72% and anti-CTLA-4 agents and anti-PD-L1 agents
this sequence of events, especially the infiltration step [34].
are expected to account for 20% and 8% of the market,
respectively.
2.3. Antibodies directed to PD-L1

2.2. Antibodies targeting PD-1 Therapeutic PD-L1 antibodies comprise MPDL-3280/Tecentriq,


BMS-936559/MDX-1105 (discontinued), MEDI4736/durvalu-
Immunomodulation with the PD1-targeting mABs has shown mab, and MSB0010718C/avelumab [10,30]. So far, Tecentriq
to mediate tumor shrinkage and to extend OS in several was the first checkpoint inhibitor approved for advanced
pivotal phase I/II studies in melanoma, renal cell carcinoma urothelial carcinoma [35]. The PD-L1 gene promoter region
(RCC), and NSCLC [9,30]. Nivolumab (BMS-936558/MDX1106b, has a response element to the IFN regulatory factor, and upon
Opdivo) is a human IgG4 antibody against PD-1, lacking IFNγ stimulation, PD-L1 is not only expressed on tumor cells,
detectable ADCC, which has been approved by the FDA for but likewise on T cells, NK cells, macrophages, myeloid DCs, B
the application in unresectable or metastatic melanoma, meta- cells, epithelial cells, and vascular endothelial cells [36].
static NSCLC, and advanced RCC. Long-term data on 129 Atezolizumab/MPDL3280A/Tecentriq is a phage-derived
NSCLC patients showed an overall response rate of 17.2% human IgG1 mAB which was engineered with a mutation in
with a median duration of the responses of 18.5 months the Fc domain (298 Asn to Ala) to prevent N-linked glycosyla-
[31]. Drug-related AEs (any grade) occurred in 71% of patients tion and ADCC activity [37,38]. In stage IIIB/IV or recurrent
with NSCLC, with grade 3 to 4 drug-related AEs reported in NSCLC, 19% of objective response rate (ORR) was recorded
14%. However, the drug failed to outperform chemotherapy when atezolizumab used as a first-line therapy compared with
for first-line treatment in a lung cancer trial in patients not 17% when it was a second-line or subsequent therapy. The
preselected for PD-1 expression (Checkmate 026). most commonly reported AEs were fatigue and nausea. In a
Pembrolizumab (MK-3475, lambrolizumab, Keytruda) is a second phase II study, the OS was 12.6 months in patients
highly specific anti-PD-1 humanized IgG4 antibody that con- who received atezolizumab, compared with 9.7 months in
tains a mutation at S228P designed to further suppress Fc- those who received docetaxel but overall responses (OR)
mediated ADCC. First-line treatment of NSCLC has cut progres- with atezolizumab were durable, with a median duration of
sion and the risk of death in half. The median progression-free 14.3 months versus 7.2 months for docetaxel. About 40% of
time was 10.3 months versus 6 months for patients on che- patients in the atezolizumab group experienced grade 3 to 4
motherapy alone [32]. AEs versus 53% in the docetaxel group [10,38,39]. The most
common atezolizumab-related grade 3 AEs were pneumonia
2.2.1. Resistance to anti-PD1 immunotherapy and increased aspartate aminotransferase in serum.
Despite high antitumor activity of antibodies directed to PD-1 BMS-936559 is a PD-L1-specific, fully human, and high-affi-
immune checkpoint in lung cancer, resistance to these nity IgG4 mAB that inhibits the interaction of PD-L1 with PD-1
518 G. HAMILTON AND B. RATH

or CD80 [40]. IgG4 antibodies poorly activate ADCC, and, targeting mechanism [46,49]. The third-generation therapeutic
additionally, its stabilizing S228P Fc mutation prevents Fab mABs are both humanized, and their Fc regions have been
exchange and further impairs ADCC [41]. In a phase I trial, modified for increased binding to FcγRIII [50]. Although each
13% of the patients with NSCLC achieved a partial response of the antigens targeted by these therapeutics is expressed on
and 12% had stable disease. Drug-related AEs were observed non-target cell populations, all of the MAbs have demon-
in 39% of patients and included rash, hypothyroidism, hepati- strated safety and clinical benefit [51].
tis, and one case each of sarcoidosis, endophthalmitis, dia- NK cells lyse target cells by granzyme B- and perforin-
betes mellitus, and myasthenia gravis. BMS-936559 was mediated apoptosis or death receptor ligands [52]. NK cells
discontinued. are therefore able to lyse target cells without priming and
Avelumab (MSB0010718C) represents a fully human anti- secrete cytokines like IFNγ to recruit and activate adaptive
PD-L1 IgG1 mAB [42]. Through the blockade of PD-L1, avelu- immune cells. However, other effector cells, such as macro-
mab reactivates CTLs and, furthermore, can induce ADCC due phages, may be involved in the activity of many anticancer
to the native Fc-region contained [43]. This mAB is discussed antibodies by their expression of all classes of Fcγ receptors
in detail in the following sections. which mediate antibody-dependent phagocytosis. In all tumor
MEDI4736 (durvalumab) is a human IgG1 mAB with high types, the macrophages constitute most of the infiltrating cells
affinity and specificity to PD-L1, which had also been engi- while hardly any NK cells were identified [53,54]. Combining
neered to prevent ADCC [44]. In pretreated bladder cancer tumor-targeting mABs and ADCC-promoting agents that sti-
patients, the ORR was 31.0% in 42 response-evaluable mulate effector cells may hold great clinical potential.
patients, with 46.4% in the PD-L1-positive subgroup and 0% However, most of the fully human or humanized antibodies
in the PD-L1-negative subgroup [45]. The most common treat- directed to PD-1 or PD-L1 are of the IgG4 isotype which
ment-related AEs of any grade were fatigue (13.1%), diarrhea exhibits low activity in ADCC assays. Furthermore, the immune
(9.8%), and decreased appetite (8.2%). checkpoint antibodies of IgG1 isotype are genetically engi-
neered to eliminate activity in ADCC in order to protect PD-
1/PD-L1-positive immune cells [55]. Thus, avelumab is the
3. Role of ADCC in antitumor therapy
single exception of a therapeutic antibody which exploits
The Fc receptor FcγRIII (CD16) provides NK cells with the immune checkpoint inhibition and ADCC-mediated killing of
capacity to mediate ADCC upon recognition of the Fc segment tumor cells simultaneously (Figure1.).
of selected IgGs bound to cell surfaces [46,47]. CD16 (FcγRIII), Another mechanism involving ADCC affects regulatory T
contains an immunotyrosine-activating motif in the cytoplas- cells (Treg), which express CTLA-4 constitutively. Inhibition of
mic domain which upon recognition of an antibody-coated CTLA-4 results in reduced Treg function which may also con-
target cell results in rapid activation and degranulation [48]. tribute to antitumor responses by anti-CTLA-4 treatment. For
Tumor eradication by a range of mAB anticancer therapeutics, example, anti-CTLA-4-IgG2a mediates a rapid reduction of
such as anti-CD20 (rituximab, IgG1), anti-Her2 (trastuzumab, Tregs at the tumor site in experimental animals [56].
IgG1), anti-CD52 (alemtuzumab, IgG1), anti-EGFR (cetuximab, Ipilimumab engages ex vivo FcγRIII (CD16)-expressing, non-
IgG1), and others, relies on this antigen-specific NK cell- classical monocytes resulting in ADCC-mediated lysis of Tregs

CTL NK Cell
Tumor Cell
Other Anti-PD-1
or Anti-PD-L1 AB Avelumab
PD-1 PD-L1 Fc γ RIII

Figure 1. Schematic presentation of tumor cell – CTL – NK cell interactions.


Tumor cells express PD-L1 receptors which are blocked by the anti-PD-L1 avelumab mAB (green). CTL effector cells recognise the MHC class I molecule of tumor cells
via T-cell receptor (left, top) and the CTL PD-1 receptor is blocked by avelumab to inhibit PD-1/PD-L1 binding and generation of immune checkpoint inhibition.
Furthermore, avelumab links NK cells via their Fc receptor to PD-L1 ligands of the tumor cell and induces tumor cell lysis via ADCC. Avelumab may also bind to PD-
L1 of CTLs but, in this case, ADCC seems not to be triggered. Other antibodies directed to PD-1 or PD-L1 (grey) provide blockade of checkpoint inhibition but lack
the ability to invoke NK cell lysis. Full color available online.
EXPERT OPINION ON BIOLOGICAL THERAPY 519

[57]. Accordingly, patients responding to ipilimumab displayed reported in the literature, and data of all other trials is avail-
higher counts of peripheral nonclassical monocytes compared able in abstract form.
with nonresponder patients.
5.1. Phase I study of avelumab
4. Avelumab: preclinical studies
A phase I dose-escalation study of avelumab (1–20 mg/kg) in
Avelumab/MSB0010718C is a fully human IgG1 anti-PD-L1 117 patients has been completed at the National Institutes of
mAB which can lyse a range of PD-L1-positive human Health’s Clinical Center (NCT01772004), with responses and
tumor cells in the presence of peripheral blood mononuc- prolonged disease stabilization observed in several tumor
lear cells (PBMC), or NK effector cells (Figure1) [42,43]. In types [61]. The toxicity profile for avelumab appeared compar-
preclinical experiments, avelumab-mediated ADCC lysis of able to that of other anti-PD1/PD-L1 mABs. Only minor
tumor cells was comparable for NK cells purified from changes were found in analyses of immune cell subsets in
either healthy donors or cancer patients [43]. In contrast, PBMCs of patients in response to avelumab at 15, 43, and
avelumab-mediated lysis of whole PBMCs, including PD-L1- 127 days after initiation of treatment. Thus, avelumab seem
positive subsets as targets, was marginal, corroborating to efficiently mediate ADCC of human tumor cells that express
analyses of PBMC subsets of patients treated with avelu- PD-L1, but only minor levels of avelumab-mediated lysis were
mab [43,51]. However, IFNγ is induced at sites of tumor noted among unstimulated PBMCs.
cell–immune effector cell contact which results in
increased PD-L1 expression of both tumor cells and lym-
5.2. Pharmacokinetics of avelumab
phocytes, thus possibly enhancing ADCC-mediated lysis of
both cell types [43]. For example, PD-L1 expression was The pharmacokinetics of avelumab and the PD-L1 receptor
markedly upregulated by IFNγ in chordoma cell lines, occupancy (RO) were determined in samples collected from
which significantly increased their sensitivity to ADCC [58]. the phase I dose-escalation trial (NCT01772004) [62]. In vitro
In detail, in respect to immune effector cells, addition of data using blood samples from healthy donors had indi-
avelumab to an antigen-specific in vitro stimulation assay cated that 1 µg/ml avelumab was sufficient for >95% satura-
using normal PBMCs markedly reduced the proliferation tion of the cognate antigen. For the avelumab trial, dosage
CD4+ T cells but increased the frequency of activated CD8 was escalated in four steps ranging from 1 and 3 to 10 and
+ T lymphocytes, which are characterized by the production 20 mg/kg antibody. About 50 patients with advanced solid
of IFNγ [43]. An association between low CD4+ T cells and tumors were treated with avelumab, every two weeks (q2w),
an increased magnitude of the CD8+ CTL responses to at the four dose levels and showed a linear increase of Cmax
tumors was reported for an animal model [59]. The mechan- and area under curve (AUC) with dosage. Half-lives for the
isms of the resistance of PD-L1-positive immune cells to antibody were 66, 86, 92, and 115 h for the four dose levels,
avelumab-mediated ADCC are not clear. However, the respectively, with statistically significant differences
IFNγ-induced PD-L1 expression of tumor cells showed no between the lowest and the three higher doses. The phar-
clear correlation to their sensitivity to ADCC-mediated lysis, macokinetics data were consistent with a two-compartment
and IFNγ upregulates major histocompatibility complex model with linear elimination of the antibody. It could be
(MHC) class I expression which is a negative costimulatory demonstrated that trough levels at a dose of 10 mg/kg, but
signal for NK cells and may protect cells from lysis [43]. In not at lower doses, were sufficient for >95% RO. Absolute
conclusion, the IgG1 mAB avelumab did not induce lysis of lymphocyte count was stable, and immune cell subsets
activated PD-L1-positive human immune cells, but increased evaluated exhibited no significant changes due to possible
antigen-specific immune activation and, therefore, seemed avelumab-induced ADCC. Thus, the 10 mg/kg dose of ave-
suitable for PD-L1 blockade in patients. lumab was recommended for the ongoing phase II and III
ADCC of avelumab towards primary mesothelioma cell trials.
lines was evaluated using autologous and allogeneic NK cells
[60]. The lymphocytes present in malignant effusions of malig-
5.3. Avelumab treatment of Merkel cell carcinoma
nant mesothelioma induced IFNγ-mediated PD-L1 expression
on the tumor cell surface. Of three mesothelioma cell lines Merkel cell carcinoma (MCC) is a rare and aggressive neuroen-
tested, two were lysed by avelumab-mediated ADCC in pre- docrine tumor of the skin [63]. Early-stage disease can be
sence of autologous NK cells. Additionally, avelumab treated with surgical resection and radiotherapy, but minimal
enhanced antigen-specific immune activation in PBMCs from residual disease is frequently identified. In addition, responses
metastatic breast cancer (MBC) patients. In conclusion, the to chemotherapy of advanced MCC are short lived, and immu-
preclinical experiments demonstrated checkpoint inhibition notherapy targeting the PD-1/PD-L1 checkpoint holds great
and ADCC without significant negative effects on immune promise as oncogenesis in MCC is linked to polyomavirus
cells for avelumab. integration and frequent ultraviolet-radiation-induced muta-
tions. Avelumab was studied in patients with stage IV MCC
(NCT02155647) that had progressed after cytotoxic che-
5. Avelumab in clinical studies
motherapy [64]. The mAB was administered intravenously at
Avelumab is in an early stage of clinical evaluation, and, there- a dose of 10 mg/kg every 2 weeks. The proportion of patients
fore, the Merkel carcinoma phase II trial is the first study who achieved an objective response was 28/88 patient
520 G. HAMILTON AND B. RATH

(31.8%) including eight complete and 20 partial responses. Stable disease was reported in 34 patients (45.3%), resulting in
Serious treatment-related AEs were reported in five patients a disease control rate of 64.0%, and a median PFS was
(6%): one case of enterocolitis, infusion-related reaction (IRR), 11.6 weeks for all treated patients.
increased aminotransferases, chondrocalcinosis, synovitis, and
interstitial nephritis each. Five grade 3 treatment-related AEs
5.6. Avelumab treatment of breast cancer
occurred in four (5%) patients: lymphopenia in two patients,
blood creatine phosphokinase, aminotransferase, and blood Avelumab was studied in 168 patients with locally advanced
cholesterol increases in one patient each. There were no treat- breast cancer (BC) or MBC refractory to or progressing after
ment-related grade 4 AEs or treatment-related deaths. Thus, standard-of-care therapy (NCT01772004) [68]. Patients had
avelumab showed significant clinical activity in this difficult-to- received a median of three prior therapies with taxane and
treat tumor type. anthracycline. Any grade treatment-related AEs occurred in
120 patients (71.4%) with the most common comprising fati-
gue, nausea, and IRRs. Treatment-related grade ≥3 AEs
5.4. Avelumab for the treatment of chemoresistant
occurred in 24 patients (14.3%) and included (≥1%) fatigue,
ovarian cancer
anemia, increased gamma-glutamyltransferase (GGT), and
A total of 124 relapsed ovarian cancer patients were treated autoimmune hepatitis (each 3 [1.8%]) and arthralgia (2
with avelumab, resulting in a response rate of 9.7% comprised [1.2%]). There were two treatment-related deaths (acute liver
of 12 partial responses with 6 ongoing [65]. Stable disease was failure and respiratory distress). Response rate in the entire
observed in 55 patients (44.4%) yielding a disease control rate cohort was 5.4% (nine patients), with one complete and eight
of 54%. Thus, single-agent avelumab showed an acceptable partial responses. Stable disease was observed in additional 40
safety profile and clinical activity in these heavily pretreated patients (23.8%), yielding an overall disease control rate of
patients. Treatment-related AEs occurred in 66.1% of patients 29.2%. Among all patients with PD-L1-expressing immune
with most common fatigue (13.7%), IRR (12.1%), and diarrhea cells within the tumor, 33.3% (4 of 12) had partial responses.
(11.3%), all of grade 1 to 2. Grade 3 to 4 side effects were In patients with triple-negative BC who had PD-L1-positive
reported in eight patients (6.5%), and there were no treat- immune cells within the tumor, 44.4% (four of nine) had
ment-related deaths. partial responses, compared to 2.6% for triple-negative BC
and PD-L1-negative immune cells, respectively. In conclusion,
avelumab showed an acceptable safety profile and had clinical
5.5. Avelumab treatment of NSCLC
activity in a subset of patients with MBC.
Patients with advanced NSCLC progressing after platinum-
based chemotherapy (NCT01772004) were treated with avelu-
6. Conclusion
mab at 10 mg/kg q2w [66]. Histology was adenocarcinoma
(62%), squamous cell carcinoma (29%), or other (9%). Any Cancer immunotherapy has already yielded promising results
grade AEs occurred in 139 (75.5%) patients including fatigue, in several tumor types, and new mABs for immune checkpoint
nausea, IRRs, chills, decreased appetite, and diarrhea. Drug- inhibition arrive at an accelerated pace. Comparable to the
related AEs grade ≥3 occurred in 22 (12%) patients and three situation experienced with small-molecule inhibitors directed
drug-related deaths were reported (radiation pneumonitis, to mutated kinases in NSCLC, questions addressing the best
acute respiratory failure, and disease progression). mABs, possible combinations of agents, selection of the most
Objective responses were observed in 22 (12%) patients responsive patients, and mechanisms of resistance to the
(1complete response (CR) and 21 partial responses (PR)), and therapeutics are discussed. Targeting of checkpoint inhibitor
stable disease was found in 70 patients (38%). Median pro- has been expanded from CTLA-4 to the PD-1/PD-L1 pathway
gression-free survival was 11.6 weeks and the response in PD- and yielded significant anticancer activity in heavily pretreated
L1-positive patients was 14.4% and 10.0% in PD-L1-negative patients. For the immunotherapies, duration of response is
patients, respectively. Median progression-free survival in PD- suggested as alternative parameter to disease-free survival
L1-positive patients was 11.7 weeks vs. 5.9 weeks in PD-L1- and OS. Tumor PD-L1 expression can be constitutive or induce
negative patients. In conclusion, avelumab was administered by IFNγ, resulting in different levels of expression and intratu-
to pretreated NSCLC patients safely with a toxicity profile moral heterogeneity. In general, intrinsic expression is mostly
similarly to other anti-PD-1/anti-PD-L1 agents. homogeneous throughout the tumors and is better suited for
In a second trial, 145 patients received avelumab and were anti-PD-L1 antibody treatment in contrast to the patchy posi-
monitored for a median duration of 13 weeks (range 0–31) tivity in response to induction by IFNγ [69].
[67]. Median age was 70 years, and performance status was 0 Most of the therapeutic mABs directed to tumor antigens
(31.0%) or 1 (69.0%), with histology of mostly adenocarcinoma exploit ADCC as an important mechanism for highly efficient
(63.4%) and squamous (26.9%) carcinoma. Treatment-related cancer cell eradication. In the past, ADCC was avoided in
AEs occurred in 82 patients (56.6%; all grades); those occurring immune checkpoint inhibition for the concern to damage
in ≥ 10% were IRR in 24 patients (16.6%) and fatigue in 21 and eliminate immune cells which express the relevant target
patients (14.5%). Grade ≥3 treatment-related AEs were antigen in their activated state. Nevertheless, avelumab was
reported in 13 patients (9.0%). There were no treatment- the first mAB to capitalize both on immune checkpoint inhibi-
related deaths. Among 75 patients, unconfirmed overall tion via PD-1/PD-L1 and direct-cell attack via ADCC. Preclinical
response rate was 18.7% (1 CR and 13 PRs; 12 were ongoing). and clinical data support the use of this ADCC-competent
EXPERT OPINION ON BIOLOGICAL THERAPY 521

mAB in checkpoint inhibition, and clinical trials revealed side chemoradiotherapy and innate as well as adaptive immunity
effects comparable to ADCC-disabled IgG therapeutics. in the treatment of cancer is not clear [72]. Chemotherapy,
Advantages of ADCC in immunotherapy directed to check- biological therapies, and radiotherapy can temporarily weaken
points may be increased efficacy and, in particular, preclusion immunity in cancer patients. A panel of in vitro tests, including
of upregulation of alternative immune checkpoint effectors, flow cytometric measurements of PBMC subpopulations, T-cell
resulting eventually in resistance to therapy. The full potency responsiveness to antigenic stimuli, and tests for NK activity
of avelumab is expected to be revealed in subgroups of would provide measures of the immunocompetence of the
patients positive for expression of PD-L1 and for those dis- patients and possibly explain part of failure of immune check-
playing better immunocompetence than heavily pretreated point inhibition. Research on stimulating the immune system
patients. Further trials of avelumab for mesothelioma, gastric in general or individual cell populations, like NK cells, is
cancer, urothelial and adrenocortical carcinoma, renal cancer ongoing, and such methods may be employed as co-medica-
as well as in combination with small-molecule inhibitors are tion. The best modes of application of immune checkpoint
ongoing [70]. inhibitors and their most efficient combinations are studied in
a host of clinical trials. For example, avelumab will be studied
in ovarian cancer in a phase Ib/II trial in combination with
7. Expert opinion
entinostat, a benzamide histone deacetylase inhibitor under-
Immunotherapy for cancer was of limited efficacy until inhibi- going clinical trials for treatment of various cancers [73]. In
tion of the immune defense system by tumor-activated sup- conclusion, clinical data obtained in studies of administration
pressive checkpoints became clear. Accordingly, of avelumab suggests the feasibility to combine immune
administration of an mAB to CTLA-4 prolonged survival in checkpoint inhibition with ADCC-mediated eradication of
melanoma patients. Attempts for immune checkpoint inhibi- tumor cells to increase anticancer activity and prevent a pos-
tion by agents directed to PD-1/PD-1L has generated a range sible switch to alternative checkpoint inhibitors.
of mABs which were found to have clinical activity in mela-
noma, NSCLC, breast cancer, and other tumor types. Avelumab
Funding
is a human IgG1 antibody capable of triggering ADCC as
powerful additional means to eliminate tumor cells. The This manuscript has not been funded.
other mABs directed to PD-L1 have inactivated Fc regions or
are of IgG4 subclass and unable to trigger ADCC. In addition to
the inhibition of the checkpoint blockade, ADCC guided by
Declaration of interest
anti-PD-L1 antibodies may lyse antigen-positive activated The authors have no relevant affiliations or financial involvement with any
immune effector cells known to be induced by IFNγ in the organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
tumor infiltrate. Despite ADCC triggering, preclinical and clin-
employment, consultancies, honoraria, stock ownership or options, expert
ical testing of avelumab revealed no increased toxicity to testimony, grants or patents received or pending, or royalties.
immune cells or elevated toxic side effects compared to simi-
lar therapeutic mABs. Avelumab-guided cancer cell lysis by
ADCC may constitute an important advantage in precluding References
upregulation of alternative suppressive immune checkpoint Papers of special note have been highlighted as either of interest (•) or of
proteins by tumors, as demonstrated for TIM-3 in case of considerable interest (••) to readers.
inhibition of anti-PD-1. Clinical results indicate that enrich- 1. Koller KM, Wang W, Schell TD, et al. Malignant melanoma – the
ment of the patient populations for PD-L1 positivity and dis- cradle of anti-neoplastic immunotherapy. Crit Rev Oncol Hematol.
tinct tumor-specific phenotypes select for the patients with 2016;106:25–54.
2. Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the
highest chance of responses. beginning of the end of cancer? BMC Med. 2016;14:73.
Clinical activity of avelumab in the trials for NSCLC, breast 3. Pardoll DM. The blockade of immune checkpoints in cancer immu-
cancer, and ovarian cancer patients was limited, as found for notherapy. Nat Rev Cancer. 2012;12:252–264.
other immune checkpoint-inhibiting mABs directed to the • Interesting review on cancer immunotherapy.
same receptors. Most of the initial studies of immune check- 4. 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
point inhibitors recruit patients which were heavily pretreated CD8(+) T cells. Sci Transl Med. 2013;5:200ra116.
with chemotherapy. Whereas patients are tested extensively 5. Redman JM, Gibney GT, Atkins MB. Advances in immunotherapy
for expression of PD-1/PD-L1 antigens, there is obviously for melanoma. BMC Med. 2016;14:20.
hardly any attempt to check their immunocompetence 6. Spain L, Larkin J. Combination immune checkpoint blockade with
ipilimumab and nivolumab in the management of advanced mel-
which may constitute an important predictor of successful
anoma. Expert Opin Biol Ther. 2016;16:389–396.
response to checkpoint inhibition and ADCC immune cell 7. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer
attack. Recent data indicate that breast cancer patients have J Clin. 2013;63:11–30.
not recovered from deleterious effects of chemotherapy on 8. Holt GE, Podack ER, Raez LE. Immunotherapy as a strategy for the
the immune system even after 9 months following end of treatment of non-small-cell lung cancer. Therapy. 2011;8:43–54.
9. Herzberg B, Campo MJ, Gainor JF. Immune checkpoint inhibitors in
therapy [71]. Phenotypes of repopulating B and CD4+ T cells
non-small cell lung cancer. Oncologist. 2017;22:81–88.
were significantly different from pre-chemotherapy profiles 10. Aranda F, Vacchelli E, Eggermont A, et al. Trial watch: immunosti-
and showed no signs of recovery. Many treatments adversely mulatory monoclonal antibodies in cancer therapy.
affect NK cell function, and the optimal synergy between Oncoimmunology. 2014;3:e27297.
522 G. HAMILTON AND B. RATH

11. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol.
ipilimumab in patients with metastatic melanoma. N Engl J Med. 2016;17:1497–1508.
2010;363:711–723. 33. Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic
12. di Giacomo AM, Biagioli M, Maio M. The emerging toxicity profiles PD-1 blockade is associated with upregulation of alternative
of anti-CTLA-4 antibodies across clinical indications. Semin Oncol. immune checkpoints. Nat Commun. 2016;7:10501.
2010;37:499–507. • Resistance to PD-1 immunotherapy by upregulation of alter-
13. Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in native checkpoint molecules.
tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. 34. Chen DS, Mellman I. Oncology meets immunology: the cancer-
14. Ceeraz S, Nowak EC, Noelle RJ. B7 family checkpoint regulators in immunity cycle. Immunity. 2013;39:1–10.
immune regulation and disease. Trends Immunol. 2013;34:556–563. 35. Bracarda S, Altavilla A, Hamzaj A, et al. Immunologic checkpoints
15. Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD- blockade in renal cell, prostate, and urothelial malignancies. Semin
L1) pathway to activate anti-tumor immunity. Curr Opin Immunol. Oncol. 2015;42:495–505.
2012;24:207–212. 36. Flies DB, Chen L. The new B7s: playing a pivotal role in tumor
16. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 immunity. J Immunother. 2007;30:251–260.
promotes T-cell apoptosis: a potential mechanism of immune eva- 37. Spigel DR, Gettinger SN, Horn L, et al. Clinical activity, safety, and
sion. Nat Med. 2002;8:793–800. biomarkers of MPDL3280A, an engineered PD-L1 antibody in
17. Yongshu L, Fangfei L, Feng J, et al. A mini-review for cancer patients with locally advanced or metastatic non-small cell lung
immunotherapy: molecular understanding of PD-1/PD-L1 pathway cancer (NSCLC). J Clin Oncol. 2013;31:abstr 8008.
& translational blockade of immune checkpoints. Int J Mol Sci. 38. Soria JC, Cruz C, Balheda R, et al. Clinical activity, safety and
2016;17:1151. biomarkers of PD-L1 blockade in non-small cell lung cancer
18. Zou W, Chen L. Inhibitory B7-family molecules in the tumour (NSCLC): additional analyses from a clinical study of the engineered
microenvironment. Nat Rev Immunol. 2008;8:467–477. antibody MPDL3280A (anti-PDL1). European Cancer Congress
19. Chakravarti N, Prieto VG. Predictive factors of activity of anti-pro- (ECCO-ESMO-ESTRO). Amsterdam, Netherland, 2013.
grammed death-1/programmed death ligand-1 drugs: immunohis- 39. Deng R, Bumbaca D, Pastuskovas CV, et al. Preclinical pharmacoki-
tochemistry analysis. Transl Lung Cancer Res. 2015;4:743–751. netics, pharmacodynamics, tissue distribution, and tumor penetra-
20. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance tion of anti-PD-L1 monoclonal antibody, an immune checkpoint
and autoimmunity. Immunol Rev. 2010;236:219–242. inhibitor. MAbs. 2016;8:593–603.
21. Garon EB. Current perspectives in immunotherapy for non-small 40. Langer CJ, Li Y, Li F, et al. Emerging immunotherapies in the
cell lung cancer. Semin Oncol. 2015;42:S11–S18. treatment of non-small cell lung cancer (NSCLC): the role of
22. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for check- immune checkpoint inhibitors. Am J Clin Oncol. 2015;38:422–430.
point inhibitor-based immunotherapy. Lancet Oncol. 2016;17: 41. Yang X, Ambrogelly A. Enlarging the repertoire of therapeutic
e542–e551. monoclonal antibodies platforms: domesticating half molecule
• Discussion of suitable biomarkers for cancer immunotherapy. exchange to produce stable IgG4 and IgG1 bispecific antibodies.
23. Braun DA, Burke KP, van Allen EM. Genomic approaches to under- Curr Opin Biotechnol. 2014;30:225–229.
standing response and resistance to immunotherapy. Clin Cancer 42. Liu K, Tan S, Chai Y, et al. Structural basis of anti-PD-L1 mono-
Res. 2016;22:5642–5650. clonal antibody avelumab for tumor therapy. Cell Res.
•• Up-to-date review of genomics/transcriptomics in cancer 2017;27:151–153.
immunotherapy. 43. Boyerinas B, Jochems C, Fantini M, et al. Antibody-dependent
24. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelu-
and ipilimumab or monotherapy in untreated melanoma. N Engl J mab (MSB0010718C) on human tumor cells. Cancer Immunol Res.
Med. 2015;373:23–34. 2015;3:1148–1157.
25. Villadolid J, Amin A. Immune checkpoint inhibitors in clinical prac- •• In vitro testing of ADCC activity of avelumab against tumor
tice: update on management of immune-related toxicities. Transl and immune cells.
Lung Cancer Res. 2015;4:560–575. 44. Ibrahim R, Stewart R, Shalabi A. PD-L1 blockade for cancer treat-
26. Nawaz K, Webster RM. The non-small-cell lung cancer drug market. ment: MEDI4736. Semin Oncol. 2015;42:474–483.
Nat Rev Drug Discov. 2016;15:229–230. 45. Massard C, Gordon MS, Sharma S, et al. Safety and efficacy of
• A review of the costs of immunotherapy for NSCLC. durvalumab (MEDI4736), an anti-programmed cell death ligand-1
27. Webster RM. The immune checkpoint inhibitors: where are we immune checkpoint inhibitor, in patients with advanced urothelial
now? Nat Drug Discov. 2014;13:883–884. bladder cancer. J Clin Oncol. 2016;34:3119–3125.
28. Aguiar PN Jr, De Mello R, Tadokoro H, et al. Cost effectiveness and 46. Mentlik James A, Cohen AD, Campbell KS. Combination immune
estimate of economical impact of immune checkpoint inhibitors for therapies to enhance anti-tumor responses by NK cells. Front
NSCLC relative to PD-L1 expression. Annals Oncol. 2016;27S6: Immunol. 2013;4:481.
abstract 1224. 47. Battella S, Cox MC, Santoni A, et al. Natural killer (NK) cells and anti-
29. Webster RM The immuno-oncology market, key players, leading tumor therapeutic mAb: unexplored interactions. J Leukoc Biol.
contenders and the next generation. DCAT Week, 2016 [cited 2016 2016;99:87–96.
Nov 17]. Available from: https://www.dcat.org/Public/Pdfs/ 48. Alderson KL, Sondel PM. Clinical cancer therapy by NK cells via
DCATWeek16/PIO/Rachel%20Webster%20FINAL%203.11.160-OK- antibody-dependent cell-mediated cytotoxicity. J Biomed
Distrib.pdf; Biotechnol. 2011;2011:379123.
30. Lu J, Lee-Gabel L, Nadeau MC, et al. Clinical evaluation of com- 49. Dahlberg CI, Sarhan D, Chrobok M, et al. Natural killer cell-based
pounds targeting PD-1/PD-L1 pathway for cancer immunotherapy. therapies targeting cancer: possible strategies to gain and sustain
J Oncol Pharm Pract. 2015;21:451–467. anti-tumor activity. Front Immunol. 2015;6:605.
31. Brahmer JR, Horn L, Antonia SJ, et al. Survival and long-term 50. Rajasekaran N, Chester C, Yonezawa A, et al. Enhancement of anti-
follow-up of the phase I trial of nivolumab (anti-PD-1; BMS- body-dependent cell mediated cytotoxicity: a new era in cancer
936558; ONO-4538) in patients with previously treated treatment. Immunotargets Ther. 2015;4:91—100.
advanced non-small cell lung cancer (NSCLC). J Clin Oncol. 51. Grenga I, Donahue RN, Lepone LM, et al. A fully human IgG1 anti-
2013;31:suppl; abstr 8030. PD-L1 MAb in an in vitro assay enhances antigen-specific T-cell
32. Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and responses. Clin Transl Immunol. 2016;5:e83.
pemetrexed with or without pembrolizumab for advanced, •• In vitro testing of ADCC activity of avelumab against immune
non-squamous non-small-cell lung cancer: a randomised, phase cells.
EXPERT OPINION ON BIOLOGICAL THERAPY 523

52. Rezvani K, Rouce RH. The application of natural killer cell immu- 63. Cassler NM, Merrill D, Bichakjian CK, et al. Merkel cell carcinoma
notherapy for the treatment of cancer. Front Immunol. 2015;6:578. therapeutic update. Curr Treat Options Oncol. 2016;17:36.
53. Wang W, Erbe AK, Hank JA, et al. NK cell-mediated antibody- 64. Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with
dependent cellular cytotoxicity in cancer immunotherapy. Front chemotherapy-refractory metastatic Merkel cell carcinoma: a multi-
Immunol. 2015;6:368. centre, single-group, open-label, phase 2 trial. Lancet Oncol.
54. Weiskopf K, Weissman IL. Macrophages are critical effectors of 2016;17:1374–1385.
antibody therapies for cancer. MAbs. 2015;7:303–310. •• Pivotal clinical trial of avelumab in Merkel cell carcinoma
55. Stevenson GT. Three major uncertainties in the antibody therapy of patients.
cancer. Haematologica. 2014;99:1538–1546. 65. Disis ML, Patel MR, Pant S, et al. Avelumab (MSB0010718C; anti-PD-
56. Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA-4 antibodies of L1) in patients with recurrent/refractory ovarian cancer from the
IgG2a isotype enhance antitumor activity through reduction of intra- JAVELIN solid tumor phase Ib trial: safety and clinical activity. J Clin
tumoral regulatory T cells. Cancer Immunol Res. 2013;1:32–42. Oncol. 2016;34:suppl; abstr 5533.
57. Romano E, Kusio-Kobialka M, Foukas PG, et al. Ipilimumab-depen- 66. Gulley JL, Spigel D, Kelly K, et al. Avelumab (MSB0010718C), an
dent cell-mediated cytotoxicity of regulatory T cells ex vivo by anti-PD-L1 antibody, in advanced NSCLC patients: a phase 1b,
nonclassical monocytes in melanoma patients. Proc Natl Acad Sci open-label expansion trial in patients progressing after platinum-
U S A. 2015;112:6140–6145. based chemotherapy. J Clin Oncol. 2015;33:Abstract 8034.
58. Fujii R, Friedman ER, Richards J, et al. Enhanced killing of chordoma 67. Verschraegen CF, Chen F, Spigel DR, et al. Avelumab
cells by antibody-dependent cell-mediated cytotoxicity employing (MSB0010718C; anti-PD-L1) as a first-line treatment for patients
the novel anti-PD-L1 antibody avelumab. Oncotarget. 2016;7:33498– with advanced NSCLC from the JAVELIN solid tumor phase 1b
33511. trial: safety, clinical activity, and PD-L1 expression. J Clin Oncol.
59. Yu P, Lee Y, Liu W, et al. Intratumor depletion of CD4+ cells 2016;34S:abstract 9036.
unmasks tumor immunogenicity leading to the rejection of late- 68. Dirix LY, Takacs I, Nikolinakos P, et al. Avelumab (MSB0010718C), an
stage tumors. J Exp Med. 2005;201:779–791. anti-PD-L1 antibody, in patients with locally advanced or meta-
60. Khanna S, Thomas A, Abate-Daga D, et al. Malignant mesothelioma static breast cancer: a phase IB JAVELIN solid tumor trial. Cancer
effusions are infiltrated by CD3+ T cells highly expressing PD-L1 Res. 2016;76:S1–04.
and the PD-L1+ tumor cells within these effusions are susceptible 69. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative
to ADCC by the anti-PD-L1 antibody avelumab. J Thorac Oncol. mean?. J Exp Med. 2016;213:2835–2840.
2016;11:1993–2005. • Review of the significance of PD-L1 detection in
61. Heery CR, O’Sullivan Coyne GH, Madan R.A, et al. Phase I open- immunohistochemistry.
label, multiple ascending dose trial of MSB0010718C, an anti-PD-L1 70. ASCO Meeting 2016 cited 2016 Nov 25; Available from: http://
monoclonal antibody, in advanced solid malignancies. J Clin Oncol. meetinglibrary.asco.org/abstracts
2014;32:5S, abstr. 3064 71. Verma R, Foster RE, Horgan K, et al. Lymphocyte depletion and
62. Heery CR, O’Sullivan Coyne GH, Marte JL, et al. Pharmacokinetic repopulation after chemotherapy for primary breast cancer. Breast
profile and receptor occupancy of avelumab (MSB0010718C), an Cancer Res. 2016;18:10.
anti-PD-L1 monoclonal antibody, in a phase I, open-label, dose 72. Rosental B, Appel MY, Yossef R, et al. The effect of chemotherapy/
escalation trial in patients with advanced solid tumors. J Clin radiotherapy on cancerous pattern recognition by NK cells. Curr
Oncol. 2015;33: suppl; abstr 3055. Med Chem. 2012;19:1780–1791.
• Pharmacokinetic characterization of avelumab in patients with 73. Weintraub K. Take two: combining immunotherapy with epigenetic
solid tumors. drugs to tackle cancer. Nat Med. 2016;22:8–10.

You might also like