32
Accepted Manuscript Title: Death receptor agonist therapies for cancer, which is the right TRAIL? Author: <ce:author id="aut0005" biographyid="vt0005"> Pamela M. Holland PII: S1359-6101(13)00107-X DOI: http://dx.doi.org/doi:10.1016/j.cytogfr.2013.12.009 Reference: CGFR 762 To appear in: Cytokine & Growth Factor Reviews Received date: 22-11-2013 Accepted date: 15-12-2013 Please cite this article as: Holland PM, Death receptor agonist therapies for cancer, which is the right TRAIL?, Cytokine & Growth Factor Reviews (2013), http://dx.doi.org/10.1016/j.cytogfr.2013.12.009 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Death receptor agonist therapies for cancer, which is the right TRAIL?

Embed Size (px)

Citation preview

Page 1: Death receptor agonist therapies for cancer, which is the right TRAIL?

Accepted Manuscript

Title: Death receptor agonist therapies for cancer, which is theright TRAIL?

Author: <ce:author id="aut0005" biographyid="vt0005">Pamela M. Holland

PII: S1359-6101(13)00107-XDOI: http://dx.doi.org/doi:10.1016/j.cytogfr.2013.12.009Reference: CGFR 762

To appear in: Cytokine & Growth Factor Reviews

Received date: 22-11-2013Accepted date: 15-12-2013

Please cite this article as: Holland PM, Death receptor agonist therapies forcancer, which is the right TRAIL?, Cytokine & Growth Factor Reviews (2013),http://dx.doi.org/10.1016/j.cytogfr.2013.12.009

This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

Page 2: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 1 of 31

Accep

ted

Man

uscr

ipt

1  

 

 

Death receptor agonist therapies for cancer, which is the right TRAIL?

Pamela M. Holland

1Therapeutic Innovation Unit, Amgen Inc., 360 Binney St., Cambridge, MA 02142

Correspondence: Pamela M. Holland, Amgen Inc., 360 Binney St., Cambridge, MA 02142,

Office: (617) 444-5530, E-mail: [email protected]

Keywords: Apoptosis, Apo2L/TRAIL, death receptor, cancer

Conflict of interest statement: The author is an employee of Amgen Inc. and has received stock/stock options from Amgen Inc.

Page 3: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 2 of 31

Accep

ted

Man

uscr

ipt

2  

Abstract

The activation of cell-surface death receptors represents an attractive therapeutic strategy to

promote apoptosis of tumor cells. Several investigational therapeutics that target this extrinsic

pathway, including recombinant human Apo2L/TRAIL and monoclonal agonist antibodies

directed against death receptors -4 (DR4) or -5 (DR5), have been evaluated in the clinic.

Although Phase 1/1b studies provided encouraging preliminary results, findings from

randomized Phase 2 studies failed to demonstrate significant clinical benefit. This has raised

multiple questions as to why pre-clinical data was not predictive of clinical response. Results

from clinical studies and insight into why current agents have failed to yield robust responses are

discussed. In addition, new strategies for the development of next generation death receptor

agonists, are reviewed.

Introduction

Apoptosis is integral to normal, physiological processes that regulate cell number, and results in

the removal of unnecessary or damaged cells. Evasion of apoptosis by tumor cells is key to the

pathogenesis and progression of cancer, and advancements in our understanding of the regulation

of programmed cell death pathways has led to the development of novel agents to reactivate

apoptosis in malignant cells. Activation of cell-surface death receptors by tumor necrosis factor-

related apoptosis inducing ligand (Apo2L/TRAIL, TNFSF10) and death-receptor agonists is one

approach aimed at promoting apoptosis of tumor cells via activation of the extrinsic pathway.

The early observation that Apo2L/TRAIL preferentially triggers apoptosis in tumor cells over

normal cells highlighted its potential as a candidate therapeutic in cancer. Several investigational

Page 4: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 3 of 31

Accep

ted

Man

uscr

ipt

3  

therapeutics that target this pathway, including soluble recombinant human Apo2L/TRAIL

(dulanermin) and agonist monoclonal antibodies directed against death receptors 4 (DR4) or 5

(DR5), have been developed and evaluated in phase 1 and 2 trials, either as single agents or in

combination with cytotoxic chemotherapy or other targeted agents. These studies demonstrated

that this class of agents is well tolerated and provided preliminary evidence of activity.

However, findings from randomized Phase 2 studies have not demonstrated strong clinical

activity, and no death receptor agonist therapies have advanced into Phase 3 [1-6].

Why have death receptor agonist therapies underperformed in the clinic? One possibility

is that clinically, tumors are inherently resistant to death receptor agonism, despite showing

potent activity in pre-clinical models. In this case, resistance might be attributed to extrinsic

pathway-specific issues, and agonists with distinct features with respect to their receptor

selectivity, cross-linking requirements or pharmacokinetics, might all be expected to yield poor

activity. On the other hand, it is possible that modality-specific issues have contributed to the

weak clinical findings. In this case, liabilities associated with the unique characteristics of

agonist antibodies or soluble Apo2L/TRAIL may have independently resulted in similarly weak

outcomes. It is likely that multiple factors have influenced the clinical findings generated to

date, and the feasibility and supporting data for these alternatives is discussed. A greater

understanding of these factors should provide insight into why death receptor agonist therapies

have not lived up to their potential in the clinic to date, and may provide approaches to

reconsider extrinsic pathway activation as a cancer therapeutic.

1.1 Apo2L/TRAIL and its receptors

Page 5: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 4 of 31

Accep

ted

Man

uscr

ipt

4  

Apo2L/TRAIL is a member of the TNF ligand superfamily, and its physiological role is to

modulate immune responses. Apo2L/TRAIL is expressed on many cells of the innate and

adaptive immune system in a stimulus dependent manner [7, 8]. Apo2L/TRAIL has potent anti-

viral activity in vitro and in mice can function as a key effector molecule in NK cell mediated

cytotoxicity [8]. Apo2L/TRAIL induction on NK cells also plays a critical role in the anti-

metastatic effects mediated by IFNγ in vivo [7].

Apo2L/TRAIL was identified based on its sequence homology to the extracellular

domains of TNF and CD95/FasL [9, 10]. Although the sequence identity across the family is

only 25-30%, all ligands are type II transmembrane proteins that exist in the membrane or can be

shed from the cell surface and are active as self-assembling non-covalent trimers [11]. Like

TNF, lymphotoxin, and CD40L, the structure of Apo2L/TRAIL is a homotrimeric jelly roll

protein formed by antiparallel β-pleated sheets, that binds the extracellular portion of three

cysteine-rich receptors, thereby inducing oligomerization of intracellular death domains [12, 13].

Crystallography studies have also highlighted the presence of a unique zinc binding site buried at

the trimer interface which is critical for maintaining the native structure, stability and biological

activity of Apo2L/TRAIL [14]. Preparations of recombinant Apo2L/TRAIL lacking zinc have

been shown to have reduced solubility, and tend to aggregate, potentially explaining the

associated toxicity reported by some in early studies [15, 16].

Apo2L/TRAIL binds multiple receptors with high affinity, presumably to enhance

regulatory flexibility and signaling complexity in the physiological setting. Two of these

receptors, DR4 (TR-1, TNFRSF10A) and DR5 (TR-2, TNFRSF10B), recruit adaptor proteins via

death domain interactions and initiate the formation of the death inducing signaling complex

(DISC), leading to the induction of apoptosis [17] (Fig. 1). Increasing evidence has suggested

Page 6: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 5 of 31

Accep

ted

Man

uscr

ipt

5  

that optimal signal transduction mediated by DR4 and DR5 occurs when the receptors are

clustered and aggregated into lipid rafts, which can be enhanced by multiple agents [18-20].

Therefore, receptor architecture may be an important component of facilitating DISC formation

and optimal caspase activation.

Apo2L/TRAIL binds additional receptors that share close homology within the

extracellular domain but do not signal cell death. DcR1 (TR-3, TNFRSF10C) lacks a

cytoplasmic tail and is membrane anchored via a glycophosphatidylinositol (GPI) moiety, and

DcR2 (TR-4, TNFRSF10D) has a truncated, nonfunctional death domain [17]. In some settings

DcR1 and DcR2 may use distinct mechanisms to attenuate Apo2L/TRAIL-induced apoptosis.

For example, DcR2 can be co-recruited to DR5 in a ligand dependent manner to prevent initiator

caspase activation, and may also sequester DR5 through a pre-ligand assembly domain (PLAD)

shared by both receptors, independent of ligand engagement [21, 22]. A distinct functional role

for DcR1 other than competing for Apo2L/TRAIL binding has not been described.

Apo2L/TRAIL also binds to osteoprotegerin (OPG), a soluble decoy receptor for

RANKL that blocks the RANK-RANKL interaction and limits osteoclastogenesis [23, 24]. The

issue of whether Apo2L/TRAIL can influence osteoclastogenesis remains controversial, although

Apo2L/TRAIL deficient mice show no evidence of altered bone density and no alterations in

frequency or in vitro differentiation of bone marrow precursor osteoclasts [25-27].

1.2 Apo2L/TRAIL signaling

Binding of homotrimeric Apo2L/TRAIL to DR4 and DR5 induces oligomerization of the

receptors and initiation of a pathway mediated by proteases called caspases [17]. This pathway,

Page 7: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 6 of 31

Accep

ted

Man

uscr

ipt

6  

sometimes referred to as the extrinsic pathway to denote its responsiveness to extracellular

signals, is initiated by the recruitment of the initiator caspases-8 and -10 via the adaptor protein

FADD to generate a death-inducing signaling complex (DISC) [28] (Fig. 1). The initiator

caspases in turn activate the downstream effector caspases-3, -6 and -7, which cleave a variety of

cellular substrates in order to execute the apoptotic program (Fig. 1).

In some cells, referred to as type 1, the death receptor-initiated extrinsic pathway

generates a signal strong enough to initiate apoptosis by itself. However, in the majority of cells,

referred to as type 2, the death receptor-initiated signal needs to be amplified in order to induce

apoptosis. This amplification can be achieved by cross talk between the extrinsic pathway and

the Bcl-2-regulated mitochondrial, or intrinsic, pathway [17]. One mechanism by which the

extrinsic pathway recruits the intrinsic pathway involves caspase-8-mediated cleavage of the pro-

apoptotic BH3-only Bcl-2 family member, Bid, to generate active truncated Bid (tBid). Active

tBid antagonizes the function of pro-survival Bcl-2 family members, such as Bcl-2, Bcl-XL and

Mcl-1, triggering a sequence of events that culminates in release of apoptotic factors from the

mitochondrion [28]. These apoptotic factors mediate activation of caspase-9 and antagonize the

activity of inhibitors of apoptosis (IAPs) that otherwise function to suppress caspase activity.

Thus, the intrinsic pathway can function together with the extrinsic pathway to promote caspase

activation and apoptosis.

Whereas the predominant feature of Apo2L/TRAIL signaling is the induction of apoptosis,

it is also capable of weakly activating NFkB and the MAP kinase pathways [29, 30].

Apo2L/TRAIL induced IKK and JNK activation is dependent on the protein kinase RIP, which

is dispensable for Apo2L/TRAIL induced apoptosis, but forms a secondary intracellular complex

with FADD, Traf2 and caspase 8, leading to kinase activation [29, 30]. The physiological

Page 8: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 7 of 31

Accep

ted

Man

uscr

ipt

7  

significance of Apo2L/TRAIL-induced activation of these pathways is unclear. Some reports

have suggested that Apo2L/TRAIL-induced NFkB activation promotes survival and proliferation

of apoptosis-resistant tumor cells and vascular smooth muscle cells [31-33]. Others have shown

that the NFkB subunits RelA (p65) and c-Rel mediate opposing effects on Apo2L/TRAIL

survival or death [34]. The current data is complex and controversial, yet suggests that the

biological outcome of Apo2L/TRAIL-induced NFkB activation is likely to be context dependent

and may positively and negatively influence cellular outcomes. It is worth noting that compared

to other cytokines such as TNF, the magnitude of NFkB or JNK induction by Apo2L/TRAIL is

considerably smaller. An increased understanding of the molecular mechanisms that determine

Apo2L/TRAIL-mediated apoptotic and non-apoptotic signaling is warranted. To this end, the

identification of novel factors that influence Apo2L/TRAIL signaling may provide insight.

Ubiquitination, which is a known critical regulator of many TNF induced immune and apoptotic

responses, including NFkB regulation, has recently been shown to also play a role in

Apo2L/TRAIL signaling [35, 36]. Apo2L/TRAIL can promote ubiquitination of caspase 8 via

the E3 ligase CUL3 in the DISC, leading to caspase 8 aggregation and increased apoptosis [36].

This raises the possibility that the ubiquitination states of proteins such as caspase 8 and RIP1

within Apo2L/TRAIL signaling complexes could influence cellular outcomes in response to

Apo2L/TRAIL.

1.3 Pre-clinical validation of death receptor agonists

Extensive pre-clinical validation evaluating the anti-tumor effects of death receptor agonists has

been performed. Apo2L/TRAIL as well as agonist antibodies targeting either DR4 or DR5 have

Page 9: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 8 of 31

Accep

ted

Man

uscr

ipt

8  

shown activity in vitro and in vivo against a wide range of tumor cell lines, including lung,

colon, pancreatic, non-Hodgkin’s lymphoma (NHL), multiple myeloma, glioma and breast [28,

37]. In addition, Apo2L/TRAIL has shown activity against primary tumor explants derived from

patient pancreatic and colorectal tumors [38, 39]. Not all cancer cells are sensitive to

Apo2L/TRAIL, and anti-tumor activity is enhanced when Apo2L/TRAIL is combined with

chemotherapy, radiotherapy, IFNγ or other targeted therapies [37]. Apo2L/TRAIL resistance

appears to be multi-factorial and can be influenced by the level of pro- and anti-apoptotic factors

within a cell. For example over-expression of anti-apoptotic Bcl-2 family proteins or XIAP

diminishes Apo2L/TRAIL induced apoptosis and cells may be re-sensitized by Bcl-2 or IAP

antagonists [28]. In many combination settings, including those with apoptosis pathway

regulators, growth factor inhibitors, or proteasome and histone deacetylase inhibitors,

cooperativity is directly or indirectly achieved by simultaneous activation of the extrinsic and

intrinsic pathways. However, positive interactions may also be achieved in combination with

agents that kill tumors cells via independent mechanisms, such as ADCC, or via targeting cells

within the tumor microenvironment, such as osteoclast activity in a bone metastasis setting [40,

41]. Collectively, all of these agents may lead to increased stress of tumor cells, which may

already be preferentially poised to die over normal cells due to multiple genetic alterations.

Death receptor agonist antibodies, either alone or with conventional chemotherapeutics and

targeted agents, also yielded promising preclinical results, warranting the evaluation of these

agents in a clinical setting [42-47].

1.4 Clinical experience of death receptor agonist therapies

Page 10: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 9 of 31

Accep

ted

Man

uscr

ipt

9  

Several agents for agonizing Apo2L/TRAIL-death receptors have been evaluated in the clinic

(Table 1). Dulanermin is an optimized, zinc-coordinated, homotrimeric recombinant

Apo2L/TRAIL protein consisting of amino acids 114-281 of the endogenous polypeptide, and is

the only agonist that engages both Apo2L/TRAIL death receptors, DR4 and DR5.

Conatumumab, drozitumab, tigatuzumab, and lexatumumab are all monoclonal agonist

antibodies selectively targeting DR5, whereas mapatumumab is a fully human agonist antibody

against DR4 [42-47].

Several phase 1, monotherapy, dose-escalation, safety studies in patients with advanced

solid tumors have been completed. These trials demonstrated that death receptor agonists were

tolerable as monotherapies, and concerns regarding fulminant hepatotoxicity, as observed with

FasL [48], have not been substantiated. No consistent dose-limiting toxicities have been

reported. The pharmacokinetics of the antibody agonists maintain drug concentrations that

predict activity in preclinical models, with less frequent dosing compared to dulanermin. Single

agent anti-tumor activity, however, has been modest, with only a few durable partial responses

reported across different tumor histologies, including chondrosarcoma [1, 49], follicular

lymphoma [50] and non-small cell lung cancer [2, 51-54](Table 1) . Increases in circulating cell

death markers after treatment have suggested that there was pharmacodynamic activity but have

not been useful as predictive markers of response, in part due to the small number of clinical

responses observed [55].

Several phase 1b safety studies of death receptor agonists in combination with

chemotherapy and/or targeted agents, which would be predicted to enhance the anti-tumor

activity of the class through cross-talk between the intrinsic and extrinsic pathways, have also

been reported in studies of advanced solid tumors and specific tumor histologies [3, 56-65]

Page 11: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 10 of 31

Accep

ted

Man

uscr

ipt

10  

(Table 1). Death receptor agonists were safely combined with standard doses of cancer

therapeutics in small cohorts of patients. These combinations include single-agent cytotoxics

(gemcitabine, doxorubicin, and pemetrexed), cytotoxic combinations (FOLFIRI,

carboplatin/paclitaxel, and cisplatin/gemcitabine), targeted agents (rituximab, panitumumab,

bortezomib, and vorinostat), and cytotoxic targeted agent combinations (FOLFOX/bevacizumab,

carboplatin/paclitaxel/bevacizumab, and irinotecan/cetuximab) (Table 1). Importantly, in these

studies, the combinations did not seem to significantly further sensitize normal cells to apoptosis,

and no significant drug-drug pharmacokinetic interactions were reported. However, efficacy in

small single-arm trials is difficult to formally assess in combination with another agent.

Single-agent phase 2 trials of the fully human DR4 agonist antibody mapatumumab have

been completed in non-small cell lung cancer (NSCLC), colorectal cancer (CRC) and NHL.

Two partial responses and one complete response among 40 patients with pretreated follicular

NHL were observed [50]; however, there were no objective responses in treatment-refractory

NSCLC and CRC patients [66, 67]. Randomized Phase 2 studies in NSCLC with either

dulanermin or drozitumab showed no improved response rates or progression-free survival

benefit [6, 68]. In randomized Phase 2 studies with conatumumab and chemotherapy in

pancreatic and non-small cell lung cancer, the addition of conatumumab failed to improve

outcomes [3, 69]. Results from Phase 2 studies with Tigatuzumab have not been reported.

1.5 Potential limitations of current death receptor agonist therapies

The limited activity observed in Phase 1/2 studies with death receptor agonist therapies has not

warranted initiation of Phase 3 trials. This has raised questions as to why compelling pre-clinical

Page 12: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 11 of 31

Accep

ted

Man

uscr

ipt

11  

findings from multiple agents did not translate into more robust clinical efficacy. For agonist

antibodies, one possible reason may be attributed to the cross-linking requirement for effective

receptor engagement and clustering. In vitro, this may be achieved by the addition of exogenous

cross-linking agents. In vivo, cross-linking is mediated by engagement with activating and

inhibitory Fcγ receptors (FcγR) expressed on the surface of immune cells. The DR5 agonist

antibody conatumumab is dependent on cross-linking for activity in vitro and in vivo [42].

Similarly, drozitumab also requires FcγR binding to trigger tumor-cell apoptosis [70]. In a

patient setting, the presence of infiltrating immune cells expressing the relevant FcγR in the

tumor microenvironment would be required to facilitate cross-linking and subsequent agonist

antibody activity. Therefore, reduced numbers of FcγR-expressing immune cells within a tumor

could contribute to limited antibody cross-linking and diminished anti-tumor response mediated

by death receptor agonist antibodies.

The effects of antibody cross-linking may be multi-factorial. Importantly, cross-linking

enhances death receptor clustering on the tumor cell surface to generate robust caspase

activation. In addition, antibody effector function might also contribute to the mechanism of

action of death receptor agonist antibodies. For example, the DR4 antibody mapatumumab was

reported to mediate antibody-dependent cellular cytotoxicity (ADCC) against DR4 expressing

target cells in vitro [71]. The DR5 antibody conatumumab can also induce ADCC of target cells

in vitro, and this is influenced by the allelic polymorphism of FcγR [72], which leads to high or

low affinity forms of the receptor. Collectively, this raises the possibility that death receptor

agonist antibodies may bring additional immune-mediated mechanisms to bear on the tumor. In

this respect, an agonist antibody directed against the mouse TRAIL death receptor can also

induce tumor specific effector and memory T cells [73]. Interestingly, in randomized phase 2

Page 13: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 12 of 31

Accep

ted

Man

uscr

ipt

12  

trials conducted with conatumumab in patients with both advanced non-small cell lung cancer

and metastatic colorectal cancer, trends toward a stronger treatment effect on overall survival

were observed in patients carrying a high affinity allele of the FcγRIIIa receptor, suggesting that

the high affinity form of the receptor may be associated with enhanced responsiveness to

conatumumab [72]. However, no similar trends were observed in a phase 2 pancreatic cancer

study, therefore the predictive value of FcγRIIIa polymorphisms for conatumumab is unclear and

requires additional studies [72].

The limited efficacy observed with dulanermin could be attributed to its limited exposure.

Dulanermin has a half-life of less than 1 hour in humans [1]. In addition, some soluble forms of

TNF family ligands have been shown to have different activity profiles compared to their

corresponding membrane form. For example, the soluble form of FasL has limited apoptosis

inducing activity [74-76]. The soluble form of Apo2L/TRAIL has been reported to retain

receptor activating potential for DR4, but is a weak inducer of apoptosis mediated through DR5

[77-80].

1.6 New strategies to target the extrinsic pathway

The results from clinical studies have prompted investigators to explore alternative

methodologies to improve the efficacy of death receptor agonists. Several approaches have been

employed to promote oligomerization or improve the stability of Apo2L/TRAIL, whose poor

pharmacokinetic properties might influence its anti-tumor activity [1]. For example, fusion of

Apo2L/TRAIL to a tenascin C (TNC) oligomerization domain, or fusion of two single-chain

Apo2L/TRAIL molecules to the Fc portion of a human IgG to generate a “hexameric”

Apo2L/TRAIL (www.apogenix.com), have been reported to increase apoptosis inducing-

Page 14: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 13 of 31

Accep

ted

Man

uscr

ipt

13  

activity[80] (Table 2). Fusion to human serum albumin (HSA), or PEGylation was also shown

to improve the stability and activity of Apo2L/TRAIL [81-83]. The use of poly (lactic-co-

glycolic acid) (PLGA) microspheres or nanoparticles for Apo2L/TRAIL encapsulation and

delivery to tumor sites has also been described, and shown to have superior antitumor activity in

vivo compared to free recombinant Apo2L/TRAIL [84, 85]. Other mechanisms to promote

oligomerization or mimic a membrane-anchored form of Apo2L/TRAIL have been achieved by

the fusion of soluble ligand to a single chain fragment of a variable region (scFv) antibody

fragment. Fusion to a tumor-specific target antigen can also make these agents tumor selective.

The scFv-targeted ligand is inactive but becomes activated upon antibody mediated binding and

cross-linking to the tumor cell surface (reviewed in [86, 87]). In order to also enhance T-cell

responsiveness to tumors, a gene immunotherapy approach using Apo2L/TRAIL-overexpressing

lymphocytes combined with an EpCAMxCD3 bi-specific antibody to enhance anti-tumor

responsiveness has been described [88]. Other delivery methods reported to selectively express

Apo2L/TRAIL in the tumor environment include oncolytic viruses and stem cell delivery. A

recombinant oncolytic herpes simplex virus (oHSV) type-1 bearing a secretable Apo2L/TRAIL

was shown to inhibit tumor growth and invasiveness of mice bearing Apo2L/TRAIL- and oHSV-

resistant intracerebral glioblastoma tumors [89]. Stem cell delivery would provide a cell-based

delivery system to continually produce Apo2L/TRAIL within the tumor, thereby increasing its

efficacy. By taking advantage of the inherent tropism of stem cells to malignant lesions and

modifying them to express antitumor agents, a variety of adult stem cell types have been

engineered to express soluble Apo2L/TRAIL, and successfully applied in a variety of pre-

clinical cancer models (reviewed in [90]). Two recent reports also described the use of

Page 15: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 14 of 31

Accep

ted

Man

uscr

ipt

14  

Apo2L/TRAIL-expressing stem cells encapsulated in biodegradable, synthetic extracellular

matrix which would promote more sustained therapeutic release [91, 92].

Methodologies to overcome the limitations of bivalent death receptor agonist antibodies

have also been an area of active investigation (Table 2). Although some studies have shown that

monovalent anti-DR5 scFv antibodies can directly activate DR5 and induce cell death [93],

multimerization of scFv antibody fragments can improve pharmacokinetic properties and

increase affinity. Wange et al engineered a molecule designed to mimic the affinity of naturally

occurring pentavalent molecules, termed a “combody”. This pentameric structure is composed

of a scFv directed against DR5 and fused to the coiled-coil domain of COMP (cartilage

oligomeric protein 48), which significantly increased its affinity [94]. The DR5 combody was

shown to have antitumor activity in vitro and in vivo. In another example, tetranectin, an

abundant C-type lectin domain (CTLD)-containing protein found in serum and tissue, was used

as a platform to specifically engineer DR4-specific “Atrimer” complexes [95]. Loop regions

within the CTLD were randomized to create a phage display library used to select DR4 binding

complexes with subnanomolar affinity to DR4, and showed DR4-specific tumor cell killing in

vitro. Similarly, a scaffold based on the third fibronecting type III domain of tenascin C (Tn3)

that resembles an antibody variable region was used to generate a phage display library, and

panned against DR5-Fc [96]. Linear fusion of DR5-specific Tn3 modules into multivalent

constructs and additional optimization resulted in increased agonist activity in vitro and in vivo.

In a separate study, a DR5 agonist antibody decorated on PLGA nanoparticles which

encapsulated camptothecin was shown to have activity on tumor cells in vitro [97]. Although the

majority of engineered death receptor agonist therapies have only been evaluated pre-clinically

to date, one agent, TAS266, was reported to have entered clinical trials in late 2012

Page 16: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 15 of 31

Accep

ted

Man

uscr

ipt

15  

(www.clinicaltrials.gov), but the study was quickly terminated for unknown reasons. TAS266 is

a tetrameric nanobody, consisting of a single, heavy chain domain (VHH) antibody, which occurs

naturally in the camelid species. TAS266 is humanized and linked to form a tetrameric structure,

which should not be dependent on cross-linking by Fcγ receptors in the tumor microenvironment

for activity [98]. Collectively, these examples demonstrate that generation of more potent and

tumor selective death receptor agonist therapies is feasible and begin to pave the path toward

how these approaches might lead to clinical applications. It is worth noting that although these

agents have shown activity in pre-clinical models, the clinical therapeutic potential for more

potent next generation death receptor agonists will be influenced by their safety index, potential

for immunogenicity or aggregation, and ease of manufacturability.

 

1.7 Conclusion and Perspective

The last decade has brought forth significant advances in our understanding of the

mechanisms of cell death and evasion of apoptosis as a tumor survival mechanism. Many novel

therapeutics aimed at restoring apoptosis induction in cancer cells have emerged and undergone

clinical evaluation. Although a foundation to support apoptosis induction as a therapeutic

strategy is in place, much remains to be learned about the underlying molecular intricacies and

regulatory components. Importantly, it is difficult to distinguish the observed weak clinical

activity from either insufficient pathway activation by current death receptor agonist therapies, or

a lack of significance of the Apo2L/TRAIL extrinsic pathway in human tumors, and the

development of more potent death receptor agonist therapies may help to address this. Our

understanding of mechanisms of resistance in cell lines suggests that multiple factors contribute

to a resistant phenotype. These factors include receptor expression, the ability of the receptor to

Page 17: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 16 of 31

Accep

ted

Man

uscr

ipt

16  

initiate a signal, and the threshold sensitivity of the cell to the initiating signal. The same factors

are likely to contribute to the resistance of primary human tumors but it is unclear whether the

contribution of these individual factors to the resistant phenotype is different in cell lines versus

primary human tumors. In addition, identifying patients with the greatest likelihood of response

by the application of informative, validated markers will be key to the success of these strategies.

Ongoing research continues to provide additional insight into the functions of proteins that

regulate cell death. Our increased knowledge into the regulation of TNF-family death receptor

signaling, in combination with advances in methodologies to promote receptor clustering in a

tumor selective and receptor selective manner, provides the potential for superior clinical activity

over first-generation approaches to target death receptors that have been tested to date. The

clinical development of next generation death receptor agonist therapies is eagerly awaited.

Page 18: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 17 of 31

Accep

ted

Man

uscr

ipt

17  

References

1. Herbst RS, Eckhardt SG, Kurzrock R, Ebbinghaus S, O'Dwyer PJ, Gordon MS, et al. Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. J Clin Oncol. 2010;28:2839-46. 2. Herbst RS, Kurzrock R, Hong DS, Valdivieso M, Hsu CP, Goyal L, et al. A first-in-human study of conatumumab in adult patients with advanced solid tumors. Clin Cancer Res. 2010;16:5883-91. 3. Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ, Jr., Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Annals of Oncol. 2012;23:2834-42. 4. Paz Ares L, Balint B, de Boer R, van Meerbeeck J, Wierzbicki R, De Souza P, et al. A Randomized, Phase 2 Study of Paclitaxel (P) and Carboplatin (C) +/- Conatumumab (CON) for First-Line Treatment of Non-Small Cell Lung Cancer (NSCLC). ESMO Congress. 2010;Abstract 412P. 5. Soria JC, Smit E, Khayat D, Besse B, Yang X, Hsu CP, et al. Phase 1b Study of Dulanermin (recombinant human Apo2L/TRAIL) in Combination With Paclitaxel, Carboplatin, and Bevacizumab in Patients With Advanced Non-Squamous Non-Small-Cell Lung Cancer. J Clin Oncol. 2010;28:1527-33. 6. Soria JC, Mark Z, Zatloukal P, Szima B, Albert I, Juhasz E, et al. Randomized phase II study of dulanermin in combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung cancer. J Clin Oncol. 2011;29:4442-51. 7. Cretney E, Shanker A, Yagita H, Smyth MJ, Sayers TJ. TNF-related apoptosis-inducing ligand as a therapeutic agent in autoimmunity and cancer. Immunology and cell biology. 2006;84:87-98. 8. Falschlehner C, Schaefer U, Walczak H. Following TRAIL's path in the immune system. Immunol. 2009;127:145-54. 9. Wiley SR, Schooley K, Smolak PJ, Din WS, Huang CP, Nicholl JK, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995;3:673-82. 10. Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A, Ashkenazi A. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 1996;271:12687-90. 11. Idriss HT, Naismith JH. TNF alpha and the TNF receptor superfamily: structure-function relationship(s). Microsc Res Tech. 2000;50:184-95. 12. Eck MJ, Sprang SR. The structure of tumor necrosis factor-alpha at 2.6 A resolution. Implications for receptor binding. J Biol Chem. 1989;264:17595-605. 13. Karpusas M, Hsu YM, Wang JH, Thompson J, Lederman S, Chess L, et al. 2 A crystal structure of an extracellular fragment of human CD40 ligand. Structure. 1995;3:1031-9. 14. Hymowitz SG, O'Connell MP, Ultsch MH, Hurst A, Totpal K, Ashkenazi A, et al. A unique zinc-binding site revealed by a high-resolution X-ray structure of homotrimeric Apo2L/TRAIL. Biochemistry. 2000;39:633-40. 15. Jo M, Kim TH, Seol DW, Esplen JE, Dorko K, Billiar TR, et al. Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand. Nat Med. 2000;6:564-7. 16. Lawrence D, Shahrokh Z, Marsters S, Achilles K, Shih D, Mounho B, et al. Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions. Nat Med. 2001;7:383-5. 17. Ashkenazi A. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2002;2:420-30. 18. Song JH, Tse MC, Bellail A, Phuphanich S, Khuri F, Kneteman NM, et al. Lipid rafts and nonrafts mediate tumor necrosis factor related apoptosis-inducing ligand induced apoptotic and nonapoptotic signals in non small cell lung carcinoma cells. Cancer Res. 2007;67:6946-55.

Page 19: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 18 of 31

Accep

ted

Man

uscr

ipt

18  

19. Psahoulia FH, Drosopoulos KG, Doubravska L, Andera L, Pintzas A. Quercetin enhances TRAIL-mediated apoptosis in colon cancer cells by inducing the accumulation of death receptors in lipid rafts. Mol Cancer Ther. 2007;6:2591-9. 20. Xu L, Qu X, Zhang Y, Hu X, Yang X, Hou K, et al. Oxaliplatin enhances TRAIL-induced apoptosis in gastric cancer cells by CBL-regulated death receptor redistribution in lipid rafts. FEBS Lett. 2009;583:943-8. 21. Merino D, Lalaoui N, Morizot A, Schneider P, Solary E, Micheau O. Differential inhibition of TRAIL-mediated DR5-DISC formation by decoy receptors 1 and 2. Mol Cell Biol. 2006;26:7046-55. 22. Clancy L, Mruk K, Archer K, Woelfel M, Mongkolsapaya J, Screaton G, et al. Preligand assembly domain-mediated ligand-independent association between TRAIL receptor 4 (TR4) and TR2 regulates TRAIL-induced apoptosis. Proc Natl Acad Sci U S A. 2005;102:18099-104. 23. Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S, Silverman C, et al. Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. J Biol Chem. 1998;273:14363-7. 24. Vitovski S, Phillips JS, Sayers J, Croucher PI. Investigating the interaction between osteoprotegerin and receptor activator of NF-kappaB or tumor necrosis factor-related apoptosis-inducing ligand: evidence for a pivotal role for osteoprotegerin in regulating two distinct pathways. J Biol Chem. 2007;282:31601-9. 25. Labrinidis A, Liapis V, Thai le M, Atkins GJ, Vincent C, Hay S, et al. Does Apo2L/TRAIL play any physiologic role in osteoclastogenesis? Blood. 2008;111:5411-2. 26. Zauli G, Secchiero P. Role of TRAIL in osteoclastogenesis. Blood. 2008;111:5413. 27. Sedger LM, Glaccum MB, Schuh JC, Kanaly ST, Williamson E, Kayagaki N, et al. Characterization of the in vivo function of TNF-alpha-related apoptosis-inducing ligand, TRAIL/Apo2L, using TRAIL/Apo2L gene-deficient mice. Eu J Immunol. 2002;32:2246-54. 28. Ashkenazi A. Directing cancer cells to self-destruct with pro-apoptotic receptor agonists. Nat Rev Drug Discov. 2008;7:1001-12. 29. Wajant H. TRAIL and NFkappaB signaling--a complex relationship. Vitam Horm. 2004;67:101-32. 30. Varfolomeev E, Maecker H, Sharp D, Lawrence D, Renz M, Vucic D, et al. Molecular determinants of kinase pathway activation by Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand. J Biol Chem. 2005;280:40599-608. 31. Baader E, Toloczko A, Fuchs U, Schmid I, Beltinger C, Ehrhardt H, et al. Tumor necrosis factor-related apoptosis-inducing ligand-mediated proliferation of tumor cells with receptor-proximal apoptosis defects. Cancer Res. 2005;65:7888-95. 32. Ishimura N, Isomoto H, Bronk SF, Gores GJ. Trail induces cell migration and invasion in apoptosis-resistant cholangiocarcinoma cells. Am J Physiol Gastrointest Liver Physiol. 2006;290:G129-36. 33. Kavurma MM, Schoppet M, Bobryshev YV, Khachigian LM, Bennett MR. TRAIL stimulates proliferation of vascular smooth muscle cells via activation of NF-kappaB and induction of insulin-like growth factor-1 receptor. J Biol Chem. 2008;283:7754-62. 34. Chen X, Kandasamy K, Srivastava RK. Differential roles of RelA (p65) and c-Rel subunits of nuclear factor kappa B in tumor necrosis factor-related apoptosis-inducing ligand signaling. Cancer Res. 2003;63:1059-66. 35. Bhoj VG, Chen ZJ. Ubiquitylation in innate and adaptive immunity. Nature. 2009;458:430-7. 36. Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR, et al. Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell. 2009;137:721-35. 37. Johnstone RW, Frew AJ, Smyth MJ. The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nat Rev Cancer. 2008;8:782-98. 38. Hylander BL, Pitoniak R, Penetrante RB, Gibbs JF, Oktay D, Cheng J, et al. The anti-tumor effect of Apo2L/TRAIL on patient pancreatic adenocarcinomas grown as xenografts in SCID mice. J Transl Med. 2005;3:22.

Page 20: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 19 of 31

Accep

ted

Man

uscr

ipt

19  

39. Sugamura K, Gibbs JF, Belicha-Villanueva A, Andrews C, Repasky EA, Hylander BL. Synergism of CPT-11 and Apo2L/TRAIL against two differentially sensitive human colon tumor xenografts. Oncology. 2008;74:188-97. 40. Daniel D, Yang B, Lawrence DA, Totpal K, Balter I, Lee WP, et al. Cooperation of the proapoptotic receptor agonist rhApo2L/TRAIL with the CD20 antibody rituximab against non-Hodgkin lymphoma xenografts. Blood. 2007;110:4037-46. 41. Holland PM, Miller R, Jones J, Douangpanya H, Piasecki J, Roudier M, et al. Combined therapy with the RANKL inhibitor RANK-Fc and rhApo2L/TRAIL/dulanermin reduces bone lesions and skeletal tumor burden in a model of breast cancer skeletal metastasis. Cancer Biol Ther. 2010;9. 42. Kaplan-Lefko PJ, Graves JD, Zoog SJ, Pan Y, Wall J, Branstetter DG, et al. Conatumumab, a fully human agonist antibody to death receptor 5, induces apoptosis via caspase activation in multiple tumor types. Cancer Biol Ther. 2010;9:618-31. 43. Adams C, Totpal K, Lawrence D, Marsters S, Pitti R, Yee S, et al. Structural and functional analysis of the interaction between the agonistic monoclonal antibody Apomab and the proapoptotic receptor DR5. Cell Death Differ. 2008;15:751-61. 44. Yada A, Yazawa M, Ishida S, Yoshida H, Ichikawa K, Kurakata S, et al. A novel humanized anti-human death receptor 5 antibody CS-1008 induces apoptosis in tumor cells without toxicity in hepatocytes. Annals of Oncol. 2008;19:1060-7. 45. Zeng Y, Wu XX, Fiscella M, Shimada O, Humphreys R, Albert V, et al. Monoclonal antibody to tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) induces apoptosis in primary renal cell carcinoma cells in vitro and inhibits tumor growth in vivo. Int J Oncol. 2006;28:421-30. 46. Georgakis GV, Li Y, Humphreys R, Andreeff M, O'Brien S, Younes M, et al. Activity of selective fully human agonistic antibodies to the TRAIL death receptors TRAIL-R1 and TRAIL-R2 in primary and cultured lymphoma cells: induction of apoptosis and enhancement of doxorubicin- and bortezomib-induced cell death. Br J Heametol. 2005;130:501-10. 47. Pukac L, Kanakaraj P, Humphreys R, Alderson R, Bloom M, Sung C, et al. HGS-ETR1, a fully human TRAIL-receptor 1 monoclonal antibody, induces cell death in multiple tumour types in vitro and in vivo. Br J Cancer. 2005;92:1430-41. 48. Nagata S. Apoptosis by death factor. Cell. 1997;88:355-65. 49. Subbiah V, Brown RE, Buryanek J, Trent J, Ashkenazi A, Herbst R, et al. Targeting the apoptotic pathway in chondrosarcoma using recombinant human Apo2L/TRAIL (dulanermin), a dual proapoptotic receptor (DR4/DR5) agonist. Mol Cancer Ther. 2012;11:2541-6. 50. Younes A, Vose JM, Zelenetz AD, Smith MR, Burris HA, Ansell SM, et al. A Phase 1b/2 trial of mapatumumab in patients with relapsed/refractory non-Hodgkin's lymphoma. Br J Cancer. 2010;103:1783-7. 51. Tolcher AW, Mita M, Meropol NJ, von Mehren M, Patnaik A, Padavic K, et al. Phase I pharmacokinetic and biologic correlative study of mapatumumab, a fully human monoclonal antibody with agonist activity to tumor necrosis factor-related apoptosis-inducing ligand receptor-1. J Clin Oncol. 2007;25:1390-5. 52. Hotte SJ, Hirte HW, Chen EX, Siu LL, Le LH, Corey A, et al. A phase 1 study of mapatumumab (fully human monoclonal antibody to TRAIL-R1) in patients with advanced solid malignancies. Clin Cancer Res. 2008;14:3450-5. 53. Camidge DR, Herbst RS, Gordon MS, Eckhardt SG, Kurzrock R, Durbin B, et al. A phase I safety and pharmacokinetic study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malignancies. Clin Cancer Res. 2010;16:1256-63. 54. Forero-Torres A, Shah J, Wood T, Posey J, Carlisle R, Copigneaux C, et al. Phase I trial of weekly tigatuzumab, an agonistic humanized monoclonal antibody targeting death receptor 5 (DR5). Cancer Biother Radiopharm. 2010;25:13-9.

Page 21: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 20 of 31

Accep

ted

Man

uscr

ipt

20  

55. Pan Y, Xu R, Peach M, Huang CP, Branstetter D, Novotny W, et al. Evaluation of pharmacodynamic biomarkers in a Phase 1a trial of dulanermin (rhApo2L/TRAIL) in patients with advanced tumours. Br J Cancer. 2011;105:1830-8. 56. Demetri GD, Le Cesne A, Chawla SP, Brodowicz T, Maki RG, Bach BA, et al. First-line treatment of metastatic or locally advanced unresectable soft tissue sarcomas with conatumumab in combination with doxorubicin or doxorubicin alone: a phase I/II open-label and double-blind study. Eur J Cancer. 2012;48:547-63. 57. Leong S, Cohen RB, Gustafson DL, Langer CJ, Camidge DR, Padavic K, et al. Mapatumumab, an antibody targeting TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid malignancies: results of a phase I and pharmacokinetic study. J Clin Oncol. 2009;27:4413-21. 58. Mom CH, Verweij J, Oldenhuis CN, Gietema JA, Fox NL, Miceli R, et al. Mapatumumab, a fully human agonistic monoclonal antibody that targets TRAIL-R1, in combination with gemcitabine and cisplatin: a phase I study. Clin Cancer Res. 2009;15:5584-90. 59. Paz-Ares L, Sánchez Torres JM, Diaz-Padilla I, Links M, Reguart N, Boyer M, et al. Safety and efficacy of AMG 655 in combination with paclitaxel and carboplatin (PC) in patients with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2009;27:Abstract 19048 60. Peeters M, Infante JR, Rougier P, Van Laethem J, Uronis HE, Stephenson J, et al. Phase Ib/II trial of conatumumab and panitumumab (pmab) for the treatment (tx) of metastatic colorectal cancer (mCRC): Safety and efficacy. J Clinical Oncology. 2010;2010 Gastrointestinal Cancers Symposium:Abstract 443. 61. Fuchs CS, Fakih M, Schwartzberg L, Cohn AL, Yee L, Dreisbach L, et al. TRAIL receptor agonist conatumumab with modified FOLFOX6 plus bevacizumab for first-line treatment of metastatic colorectal cancer: A randomized phase 1b/2 trial. Cancer. 2013;Oct, ePub. 62. Sikic BI, Wakelee HA, von Mehren M, Lewis N, Calvert AH, Plummer ER, et al. A phase Ib study to assess the safety of lexatumumab, a human monoclonal antibody that activates TRAIL-R2, in combination with gemcitabine, pemetrexed, doxorubicin or FOLFIRI. J Clin Oncol 2007;25:Abstract 14006. 63. Yee L, Burris HA, Kozloff M, Wainberg Z, Pao M, Skettino S, et al. Phase Ib study of recombinant human Apo2L/TRAIL plus irinotecan and cetuximab or FOLFIRI in metastatic colorectal cancer (mCRC) patients (pts): Preliminary results. J Clin Oncol. 2009;27:Abstract 4129. 64. Yee L, Fanale M, Dimick K, Calvert S, Robins C, Ing J, et al. A phase IB safety and pharmacokinetic (PK) study of recombinant human Apo2L/TRAIL in combination with rituximab in patients with low-grade non-Hodgkin lymphoma. J Clin Oncol. 2007;25:Abstract 8078. 65. Younes A, Kirschbaum M, Sokol L, Yee L, Romaguera J, Goyal L, et al. Safety and Tolerability of Conatumumab in Combination with Bortezomib or Vorinostat in Patients with Relapsed or Refractory Lymphoma. ASH 51st Annual Meeting. 2009:Abstract 1708. 66. Greco FA, Bonomi P, Crawford J, Kelly K, Oh Y, Halpern W, et al. Phase 2 study of mapatumumab, a fully human agonistic monoclonal antibody which targets and activates the TRAIL receptor-1, in patients with advanced non-small cell lung cancer. Lung Cancer. 2008;61:82-90. 67. Kanzler S, Trarbach T, Heinemann V, Köhne CH, Sneller V, Bieber F, et al. Results of a Phase 2 trial of HGS-ETR1 (agonistic human monoclonal antibody to TRAIL receptor 1) in subjects with relapsed or refractory colorectal cancer (CRC). ECCO 13 2005:Abstract 630. 68. Karapetis CS, Clingan PR, Leighl NB, Durbin-Johnson B, O'Neill V, Spigel DR. Phase II study of PRO95780 plus paclitaxel, carboplatin, and bevacizumab (PCB) in non-small cell lung cancer (NSCLC). J Clin Oncol 2010;28: Abstract 7535. 69. Paz-Ares L, Balint B, de Boer RH, van Meerbeeck JP, Wierzbicki R, De Souza P, et al. A randomized phase 2 study of paclitaxel and carboplatin with or without conatumumab for first-line treatment of advanced non-small-cell lung cancer. J Thorac Oncol. 2013;8:329-37. 70. Wilson NS, Yang B, Yang A, Loeser S, Marsters S, Lawrence D, et al. An Fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 2011;19:101-13.

Page 22: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 21 of 31

Accep

ted

Man

uscr

ipt

21  

71. Maddipatla S, Hernandez-Ilizaliturri FJ, Knight J, Czuczman MS. Augmented antitumor activity against B-cell lymphoma by a combination of monoclonal antibodies targeting TRAIL-R1 and CD20. Clin Cancer Res. 2007;13:4556-64. 72. Pan Y, Haddad V, Sabin T, Baker N, Yong-Jiang H, Galimi F, et al. Predictive value of Fc gamma receptor IIIa genotype in response to Conatumumab in three Phase II studies. J Clin Oncol. 2011;29:Abstract 3103. 73. Takeda K, Yamaguchi N, Akiba H, Kojima Y, Hayakawa Y, Tanner JE, et al. Induction of tumor-specific T cell immunity by anti-DR5 antibody therapy. J Exp Med. 2004;199:437-48. 74. Suda T, Hashimoto H, Tanaka M, Ochi T, Nagata S. Membrane Fas ligand kills human peripheral blood T lymphocytes, and soluble Fas ligand blocks the killing. J Exp Med. 1997;186:2045-50. 75. Schneider P, Holler N, Bodmer JL, Hahne M, Frei K, Fontana A, et al. Conversion of membrane-bound Fas(CD95) ligand to its soluble form is associated with downregulation of its proapoptotic activity and loss of liver toxicity. J Exp Med. 1998;187:1205-13. 76. O'Reilly L, Tai L, Lee L, Kruse EA, Grabow S, Fairlie WD, et al. Membrane-bound Fas ligand only is essential for Fas-induced apoptosis. Nature. 2009;461:659-63. 77. Muhlenbeck F, Schneider P, Bodmer JL, Schwenzer R, Hauser A, Schubert G, et al. The tumor necrosis factor-related apoptosis-inducing ligand receptors TRAIL-R1 and TRAIL-R2 have distinct cross-linking requirements for initiation of apoptosis and are non-redundant in JNK activation. J Biol Chem. 2000;275:32208-13. 78. Wajant H, Moosmayer D, Wuest T, Bartke T, Gerlach E, Schonherr U, et al. Differential activation of TRAIL-R1 and -2 by soluble and membrane TRAIL allows selective surface antigen-directed activation of TRAIL-R2 by a soluble TRAIL derivative. Oncogene. 2001;20:4101-6. 79. Kelley RF, Totpal K, Lindstrom SH, Mathieu M, Billeci K, Deforge L, et al. Receptor-selective mutants of apoptosis-inducing ligand 2/tumor necrosis factor-related apoptosis-inducing ligand reveal a greater contribution of death receptor (DR) 5 than DR4 to apoptosis signaling. J Biol Chem. 2005;280:2205-12. 80. Berg D, Lehne M, Muller N, Siegmund D, Munkel S, Sebald W, et al. Enforced covalent trimerization increases the activity of the TNF ligand family members TRAIL and CD95L. Cell Death Differ. 2007;14:2021-34. 81. Muller N, Schneider B, Pfizenmaier K, Wajant H. Superior serum half life of albumin tagged TNF ligands. BBRC. 2010;396:793-9. 82. Kim TH, Youn YS, Jiang HH, Lee S, Chen X, Lee KC. PEGylated TNF-related apoptosis-inducing ligand (TRAIL) analogues: pharmacokinetics and antitumor effects. Bioconj Chem. 2011;22:1631-7. 83. Kim TH, Jo YG, Jiang HH, Lim SM, Youn YS, Lee S, et al. PEG-transferrin conjugated TRAIL (TNF-related apoptosis-inducing ligand) for therapeutic tumor targeting. J Controlled Rel. 2012;162:422-8. 84. Kim H, Jeong D, Kang HE, Lee KC, Na K. A sulfate polysaccharide/TNF-related apoptosis-inducing ligand (TRAIL) complex for the long-term delivery of TRAIL in poly(lactic-co-glycolic acid) (PLGA) microspheres. J Pharm & Pharmacol. 2013;65:11-21. 85. Perlstein B, Finniss SA, Miller C, Okhrimenko H, Kazimirsky G, Cazacu S, et al. TRAIL conjugated to nanoparticles exhibits increased anti-tumor activities in glioma cells and glioma stem cells in vitro and in vivo. Neuro-Oncol. 2013;15:29-40. 86. Wajant H, Gerspach J, Pfizenmaier K. Engineering death receptor ligands for cancer therapy. Cancer Lett. 2013;332:163-74. 87. de Bruyn M, Bremer E, Helfrich W. Antibody-based fusion proteins to target death receptors in cancer. Cancer Lett. 2013;332:175-83. 88. Groth A, Salnikov AV, Ottinger S, Gladkich J, Liu L, Kallifatidis G, et al. New gene-immunotherapy combining TRAIL-lymphocytes and EpCAMxCD3 Bispecific antibody for tumor targeting. Clin Cancer Res. 2012;18:1028-38. 89. Tamura K, Wakimoto H, Agarwal AS, Rabkin SD, Bhere D, Martuza RL, et al. Multimechanistic tumor targeted oncolytic virus overcomes resistance in brain tumors. Molec Ther. 2013;21:68-77.

Page 23: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 22 of 31

Accep

ted

Man

uscr

ipt

22  

90. Stuckey DW, Shah K. TRAIL on trial: preclinical advances in cancer therapy. Trends Molec Med. 2013;19:685-94. 91. Kauer TM, Figueiredo JL, Hingtgen S, Shah K. Encapsulated therapeutic stem cells implanted in the tumor resection cavity induce cell death in gliomas. Nat Neurosci. 2012;15:197-204. 92. Reagan MR, Seib FP, McMillin DW, Sage EK, Mitsiades CS, Janes SM, et al. Stem cell implants for cancer therapy: TRAIL-expressing mesenchymal stem cells target cancer cells in situ. J Breast Cancer. 2012;15:273-82. 93. Park KJ, Lee SH, Kim TI, Lee HW, Lee CH, Kim EH, et al. A human scFv antibody against TRAIL receptor 2 induces autophagic cell death in both TRAIL-sensitive and TRAIL-resistant cancer cells. Cancer Res. 2007;67:7327-34. 94. Wang W, He W, Wang L, Zhang G, Gao B. Pentamerisation of a scFv directed against TRAIL receptor 2 increases its antitumour efficacy. Immunol Cell Biol. 2013;91:360-7. 95. Allen JE, Ferrini R, Dicker DT, Batzer G, Chen E, Oltean DI, et al. Targeting TRAIL death receptor 4 with trivalent DR4 Atrimer complexes. Mol Cancer Ther. 2012;11:2087-95. 96. Swers JS, Grinberg L, Wang L, Feng H, Lekstrom K, Carrasco R, et al. Multivalent scaffold proteins as superagonists of TRAIL receptor 2-induced apoptosis. Mol Cancer Ther. 2013;12:1235-44. 97. Fay F, McLaughlin KM, Small DM, Fennell DA, Johnston PG, Longley DB, et al. Conatumumab (AMG 655) coated nanoparticles for targeted pro-apoptotic drug delivery. Biomat. 2011;32:8645-53. 98. Huet H, Schuller A, Li J, Johnson J, Dombrecht B, Meerschaert K, et al. TAS266, a novel tetrameric nanobody agonist targeting death receptor 5 (DR5), elicits superior antitumor efficacy than conventional DR5-targeted approaches Cancer Res. 2012;72:Abstract 3853. 99. Cohn AL, Tabernero J, Maurel J, Nowara E, Sastre J, Chuah BY, et al. A randomized, placebo-controlled phase 2 study of ganitumab or conatumumab in combination with FOLFIRI for second-line treatment of mutant KRAS metastatic colorectal cancer. Annals Oncol. 2013;24:1777-85. 100. Chawla SP, Tabernero J, Kindler HL, Chiorean EG, LoRusso P, Hsu M, et al. Phase I evaluation of the safety of conatumumab (AMG 655) in combination with AMG 479 in patients (pts) with advanced, refractory solid tumors. J Clin Oncol. 2010;28:Abstract 3102. 101. Rocha Lima CM, Bayraktar S, Flores AM, MacIntyre J, Montero A, Baranda JC, et al. Phase Ib study of drozitumab combined with first-line mFOLFOX6 plus bevacizumab in patients with metastatic colorectal cancer. Cancer Invest. 2012;30:727-31. 102. Baron AD, O'Bryant CL, Choi Y, Ashkenazi A, Royer-Joo S, Portera CC. Phase Ib studyof drozitumab combined with cetuximab (CET) plus irinotecan (IRI) or with FOLFIRI +/- bevacizumab (BV) in previously treated patients (Pts) with metastatic colorectal cancer (mCRC). J Clin Oncol. 2011;29:Abstract 3581. 103. Wittebol S, Ferrant A, Wickham NW, Fehrenbacher L, Durbin-Johnson B, Bray GL. Phase II study of PRO95780 plus rituximab in patients with relapsed follicular non-Hodgkin's lymphoma (NHL). J Clin Oncol. 2010;28:Abstract e18511. 104. Chawla S, G D, J D, al e. Initial results of a Phase II study of the safety and efficacy of the Apomab DR5 agonist antibody in advanced chondrosarcoma and synovial sarcoma patients. CTOS Annual Meeting. 2008:Abstract 35010. 105. Wainberg ZA, Messersmith WA, Peddi PF, Kapp AV, Ashkenazi A, Royer-Joo S, et al. A Phase 1B study of Dulanermin in combination with modified FOLFOX6 plus Bevacizumab in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2013:Sept ePub. 106. Kasubhai SM, Bendell JC, Kozloff MF, Kapp AV, Ashkenazi A, Royer-Joo S, et al. Phase Ib study of dulanermin combined with FOLFIRI (with or without bevacizumab [BV]) in previously treated patients with metastatic colorectal cancer (mCRC). J Clin Oncol. 2012;30:Abstract 3543. 107. Lim B, Scicchitano A, Beachler C, Gusani N, Sarwani N, Yang Z, et al. FOLFIRI plus dulanermin (rhApo2L/TRAIL) in a patient with BRAF-mutant metastatic colon cancer. Cancer Biol Ther. 2013;14:711-9.

Page 24: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 23 of 31

Accep

ted

Man

uscr

ipt

23  

108. Belada D, Mayer J, Czuczman MS, Flinn IW, Durbin-Johnson B, Bray GL. Phase II study of dulanermin plus rituximab in patients with relapsed follicular non-Hodgkin's lymphoma (NHL). J Clin Oncol. 2010;28:Abstract 8104. 109. Sharma S, de Vries EG, Infante JR, Oldenhuis CN, Gietema JA, Yang L, et al. Safety, pharmacokinetics, and pharmacodynamics of the DR5 antibody LBY135 alone and in combination with capecitabine in patients with advanced solid tumors. Invest New Drugs. 2013:April ePub. 110. Plummer R, Attard G, Pacey S, Li L, Razak A, Perrett R, et al. Phase 1 and pharmacokinetic study of lexatumumab in patients with advanced cancers. Clin Cancer Res. 2007;13:6187-94. 111. Merchant MS, Geller JI, Baird K, Chou AJ, Galli S, Charles A, et al. Phase I trial and pharmacokinetic study of Lexatumumab in pediatric patients with solid tumors. J Clin Oncol. 2012;30:4141-7. 112. Trarbach T, Moehler M, Heinemann V, Kohne CH, Przyborek M, Schulz C, et al. Phase II trial of mapatumumab, a fully human agonistic monoclonal antibody that targets and activates the tumour necrosis factor apoptosis-inducing ligand receptor-1 (TRAIL-R1), in patients with refractory colorectal cancer. Br J Cancer. 2010;102:506-12. 113. Sun W, Nelson D, Alberts SR, Poordad F, Leong S, Teitelbaum UR, et al. Phase Ib study of mapatumumab in combination with sorafenib in patients with advanced hepatocellular carcinoma (HCC) and chronic viral hepatitis. J Clin Oncol. 2011;29:Abstract 261. 114. Belch A, Sharma A, Spencer A, S T, Bahlis N, Doval D, et al. A multicenter randomized Phase II Trial of Mapatumumab, a TRAIL-R1 agonist monoclonal antibody, in combination with Bortezomib in patients with relapsed/refractory Multiple Myeloma (MM). ASH Annual Meeting 2010;116:Abstract 5031. 115. Von Pawel J, Harvey JH, Spigel DR, Dediu M, Reck M, Cebotaru CL, et al. A randomized phase II trial of mapatumumab, a TRAIL-R1 agonist monoclonal antibody, in combination with carboplatin and paclitaxel in patients with advanced NSCLC. J Clin Oncol. 2010;28:Abstract LBA7501. 116. Kim G, Borad M, Pitot H, Rubin J, Greenberg J, McCroskery P, et al. Pilot study of Tigatuzumab (CS-1008) in combination with FOLFIRI in patients with metastatic colorectal cancer (CRC). ESMO. 2012:Abstract 2284. 117. Reck M, Krzakowski M, Chmielowska E, Sebastian M, Hadler D, Fox T, et al. A randomized, double-blind, placebo-controlled phase 2 study of tigatuzumab (CS-1008) in combination with carboplatin/paclitaxel in patients with chemotherapy-naive metastatic/unresectable non-small cell lung cancer. Lung Cancer. 2013:Oct ePub. 118. Holland PM. Targeting Apo2L/TRAIL receptors by soluble Apo2L/TRAIL. Cancer Lett. 2013;332:156-62.

Page 25: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 24 of 31

Accep

ted

Man

uscr

ipt

24  

Figure Legend

Figure 1. Apo2L/TRAIL apoptosis signaling.

Apo2L/TRAIL-mediated apoptosis is triggered upon binding to pro-apoptotic receptors

DR4 and DR5 on the surface of a target cell. Apoptosis may be attenuated by ligand binding to

DcR1, DcR2 or OPG, as these receptors do not induce cell death. Binding of death receptor

agonists to the receptors DR4 and/or DR5 results in DISC (death-inducing signaling complex)

formation. DISC formation involves recruitment of the adaptor protein FADD to the receptor

via the death domain (DD) and the inactive pro-caspases 8 and 10. This facilitates activation and

self-processing of caspase 8 and 10, leading to their release into the cytoplasm, where they

activate effector caspases 3 and 7. c-FLIP is a negative regulator and the ratio of caspase 8 to c-

FLIP in the DISC is an important determinant of response to death receptor engagement. Once

activated, caspases 3 and 7 cleave intracellular substrates, resulting in cell death. XIAP is

another negative regulator of the extrinsic pathway and functions to bind and sequester active

caspase 3. The intrinsic, or Bcl-2 regulated mitochondrial pathway, is responsive to a variety of

cellular stresses and DNA damaging agents, leading to activation of the tumor suppressor p53

and upregulation of the pro-apoptotic Bcl-2 members Puma and Noxa. These facilitate Bax and

Bak activation, and the release of cytochrome C and Smac/DIABLO from the mitochondrion.

Cytochrome C complexes with Apaf-1 and caspase 9 to form the apoptosome and further

activate caspase-3, -6 and -7, thereby providing signal amplification. Extrinsic and intrinsic

pathway cross-talk is mediated by Bid, a caspase-8 substrate that translocates to the mitochondria

and activates Bax and Bak upon cleavage. Figure adapted from Holland, Cancer Letters [118].

Page 26: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 25 of 31

Accep

ted

Man

uscr

ipt

25  

Table 1. Clinical Trials of Death Receptor Agonist Therapies

Page 27: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 26 of 31

Accep

ted

Man

uscr

ipt

26  

Agent/Target Phase/Indication Combination agents Results

Bevacizumab/FOLFOX No improvement over chemo, 63 pts [61]

FOLFIRI, mutant KRAS Trend to improved 6.5 month PFS [99] Ph 2, Colorectal cancer

Panitumumab No evidence of activity, 52 pts [60]

Ph2, Non-small cell lung cancer Carboplatin/paclitaxel

Trend to improved OS for pts on maintenance monotherapy among FcγRIIIa V-allele carriers[69]

Ph 2, Pancreatic cancer Gemcitabine Trend to improved 6 month PFS, 41 pts [3]

Ph 1/2, Sarcoma Doxorubicin No improvement over dox, 86 pts [56]

Monotherapy PR in 1/37 pts (NSCLC), metabolic PR in 1/37 pts (CRC) [2]

Bortezomib or vorinostat CR in 3/33 pts (1 CR bortezomib arm, 2 CR vorinostat arm) [65]

Conatumumab Fully human DR5 agonist antibody (Amgen)

Ph 1b, Solid tumors or lymphoma

Ganitumab SD in 3/9 pts [100]

Bevacizumab/FOLFOX PR in 5/9 pts, SD in 3/9 pts, 1 no assessment [101] Ph 1b, Colorectal cancer

Cetuximab/irinotecan or FOLFIRI in mCRC

PR in 3/20 pts, SD in 13/20 pts, PD in 4/20 pts [102]

Ph2, Non-Hodgkin’s lymphoma Rituximab No improvement over rituximab, 40 pts [103]

Ph 2, Non-small cell lung cancer

Bevacizumab/carboplatin/ paclitaxel No improvement over backbone, 120 pts [68]

Ph 2, Sarcoma Monotherapy No objective responses [104]

Drozitumab Fully human DR5 agonist antibody (Genentech/Roche)

Ph 1, Solid tumors Monotherapy No objective responses, 50 pts [53]

Bevacizumab/FOLFOX PR in 13/23 pts, SD in 7/23 pts, PD in 3/23 pts [105]

Ph 1b, Colorectal cancer Cetuximab/irinotecan or FOLFIRI in mCRC

PR in 6/27 pts, SD in 17/27 pts, PD in 3/27 pts, 1 no assessment [63, 106]. PR in 1 pt with BRAF mutant mCRC [107]

Ph 1b/2, Non-Hodgkins lymphoma Rituximab No improved overall response [64, 108]

Ph 2, Non-small cell lung cancer

Bevacizumab/carboplatin/ paclitaxel

No improved RR or progression free survival [6]

Dulanermin Recombinant human ligand DR4, DR5 dual agonist (Amgen/Genentech)

Ph 1, Solid tumors, NHL Monotherapy 2 PR in chondrosarcoma [1] LBY135 Recombinant chimeric DR5 agonist antibody (Novartis)

Ph 1, Solid tumors Monotherapy, capecitabine SD in 21/38 pts monotherapy, PR in 2/27 pts combination, SD in 12/27 pts combination. Immunogenicity in 16/73 pts [109]

Monotherapy, adult or pediatric pts

SD in 12/37 pts (3 sarcoma), DLT in 3/7 pts at 20 mg/kg [110] SD in 5/24 pts, no PR, CR [111]

Multiple chemotherapy regimens Unspecified PR with FOLFIRI, doxorubicin combinations [62]

Lexatumumab Fully human DR5 agonist antibody (Human Genome Sciences/GSK)

Ph 1/1b, Solid tumors or lymphoma

Interferon gamma No results reported

Ph 1/2, Cervical cancer Cisplatin/radiation Ongoing Ph 2, Colorectal cancer Monotherapy No activity, 38 pts [112]

Ph 1b, Hepatocellular carcinoma Sorafenib PR in 2/19 pts, SD in 4/19 pts [113]

Ph 2, Multiple myeloma Bortezomib No improvement over bortezomib, 104 pts [114]

Ph 1b/2, Non-Hodgkin lymphoma Monotherapy CR in 2/40 pts, PR in 1/40 pts, both follicular

lymphoma [50]

Ph2, Non-small cell lung cancer

Monotherapy Carboplatin/paclitaxel

SD in 9/32 pts [66] No improvement over chemo, 111 pts [115]

Mapatumumab Fully human DR4 (Human Genome Sciences/GSK) Dagonist antibody

Monotherapy SD in 19/49 pts [51]

Page 28: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 27 of 31

Accep

ted

Man

uscr

ipt

27  

Abbreviations: CR, complete response, DLT, dose-limiting toxicity, OS, overall survival, PD, progressive disease, PFS, progression-free survival, PR, partial response, Pts, patients, SD, stable disease.

Page 29: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 28 of 31

Accep

ted

Man

uscr

ipt

28  

Table 2. Apo2L/TRAIL and death receptor antibody modifications to improve agonist stability and activity.

 

Target Multimerization Agent Activity Reference

TNC oligomerization domain In vitro [80]

APG 350, hexameric TRAIL-Fc In vitro, in vivo www.apogenix.com

HSA-TRAIL In vitro, in vivo [81]

Peg-TRAIL In vivo [82, 83]

TRAIL-loaded PLGA microspheres In vivo [84]

Nanoparticle TRAIL In vitro, in vivo [85]

scFv-TRAIL bi-functional fusion proteins In vitro, in vivo Rev. in [86, 87] TRAIL-over expressing lymphocytes with EpCAMxCD3 bi-specific antibody In vitro, in vivo [88]

oHSV type-1 with secretable TRAIL In vitro, in vivo [89]

Apo2L/TRAIL

TRAIL stem cells In vivo [90-92]

DR4 Atrimer In vitro [95]

Combody-pentameric DR5-directed scFV fused to COMP In vitro, in vivo [94]

Tn3 tandem repeats In vitro, in vivo [96]

PLGA nanoparticles In vitro [97]

DR5

TAS 266, tetrameric camelid nanobody In vitro, in vivo, clinical trials [98], www.clinical.trials.gov

Page 30: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 29 of 31

Accep

ted

Man

uscr

ipt

29  

Pamela M. Holland biosketch 

Pamela Holland received her BS in Animal Physiology from the University of California, San Diego, in June 1985.  Before attending graduate school, she worked at Cetus Corporation in Emeryville, CA on early PCR applications, and made significant contributions to the development of TaqMan.  She began her graduate studies in the Biochemistry Department at the University of WA in 1992, and was the recipient of a National Science Foundation Pre‐Doctoral Fellowship.  She received her PhD in 1999 from the Fred Hutchinson Cancer Research Center, where she cloned and characterized the JNK‐activating kinase MKK7.  She then joined Immunex Corporation in Seattle, WA as a post‐doctoral fellow and characterized the function of the RIP‐family kinase RIP4.  Upon appointment to a scientist position at Immunex (later Amgen) in 2002, she supported pre‐clinical and clinical development of the death receptor agonist therapies AMG 655 (conatumumab) and Apo2L/TRAIL (dulanermin).  Since that time she has worked on the development of large and small molecule therapeutics aimed at promoting tumor cell apoptosis.  Her current research interests also include tumor immunology and the development of novel agents to enhance immune‐mediated clearance of tumor cells. 

 

Page 31: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 30 of 31

Accep

ted

Man

uscr

ipt

*Author Photo

Page 32: Death receptor agonist therapies for cancer, which is the right TRAIL?

Page 31 of 31

Accep

ted

Man

uscr

ipt

Fig. 1 Apo2L/TRAIL signaling

c-FLIP

Caspase-8, 10

Caspase-3, 7

Cellular

substrates

DR4/DR5

Apoptosis

FADD

DISC

Apo2L/TRAIL

DcR1

DcR2

OPG

XIAP

Caspase-9 Apaf

Apoptosome Cyto C

Bcl-2 Bid

Bax/Bak

Smac

Stress

DNA damage

p53

Puma/Noxa

Intrinsic pathway

Extrinsic pathway

Figure