Dysregulated cellular apoptosis and resistance to cell death are hallmarks of neoplastic initiation and disease progression. Therefore, the development of agents that overcome apoptosis dysregulation in tumor cells is an attractive therapeutic approach. Activation of the extrinsic apoptotic pathway is strongly dependent on death receptor (DR) hyperclustering on the cell surface. However, strategies to activate DR5 or DR4 through agonistic antibodies have had only limited clinical success. To pursue an alternative approach for tumor-targeted induction of apoptosis, we engineered a bispecific antibody (BsAb), which simultaneously targets fibroblast-activation protein (FAP) on cancer-associated fibroblasts in tumor stroma and DR5 on tumor cells. We hypothesized that bivalent binding to both FAP and DR5 leads to avidity-driven hyperclustering of DR5 and subsequently strong induction of apoptosis in tumor cells but not in normal cells. Here, we show that RG7386, an optimized FAP-DR5 BsAb, triggers potent tumor cell apoptosis in vitro and in vivo in preclinical tumor models with FAP-positive stroma. RG7386 antitumor efficacy was strictly FAP dependent, was independent of FcR cross-linking, and was superior to conventional DR5 antibodies. In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. FAP-DR5 also demonstrated single-agent activity against FAP-expressing malignant cells, due to cross-binding of FAP and DR5 across tumor cells. Taken together, these data demonstrate that RG7386, a novel and potent antitumor agent in both mono- and combination therapies, overcomes limitations of previous DR5 antibodies and represents a promising approach to conquer tumor-associated resistance to apoptosis. Mol Cancer Ther; 15(5); 946–57. ©2016 AACR.

This article is featured in Highlights of This Issue, p. 765

Programmed cell death (apoptosis) is a crucial process for normal development, tissue homeostasis, and the immune response in multicellular eukaryotic organisms. Impaired apoptosis, in turn, plays a key role in cancer pathogenesis and contributes to poor chemotherapy responses. Thus, development of agents that restore/activate apoptotic pathways specifically in tumor cells has emerged as an important therapeutic strategy. The extrinsic apoptotic pathway is triggered by external signals, including proapoptotic FAS-ligand and TRAIL, which bind to and activate cell surface-anchored death receptors (DR) such as FAS, DR4, or DR5 through receptor oligomerization. DR activation promotes the formation of the death-inducing signaling complex, leading to subsequent activation of caspases 3, 6, and 7 and cell apoptosis (1, 2). Several proapoptotic receptor agonists (PARA), including mAbs, have been developed to target this extrinsic apoptotic pathway (3, 4). However, despite promising preclinical data, none of these approaches have thus far demonstrated significant clinical efficacy (5–7). Although some antibodies appear to be able to autonomously activate DR4 and DR5, many require additional cross-linking to achieve optimal activity in vitro (8–11). In vivo, and as demonstrated for drozitumab, a fully human DR5 agonistic antibody (8), this cross-linking activity is likely provided by activatory or inhibitory Fcγ receptors (FcγR) on leukocytes in the tumor microenvironment (12, 13). Low or variable infiltration of FcγR-expressing leukocytes, reduced targeting of DR agonistic antibodies due to systemic binding to DRs on nontumor cells or to FcRs on circulating cells, and/or competition for FcRs by high levels of endogenous immunoglobulins, may account for the lack of clinical efficacy observed for these agonistic antibodies.

Fibroblast-activation protein (FAP) is a homodimeric, single-pass type II membrane protein expressed in reactive stromal fibroblasts of more than 90% of epithelial malignancies, including breast, colorectal, and lung cancers and on malignant mesenchymal cells of bone and soft tissue sarcomas (14–16). Here, we exploited FAP expression on the tumor stroma to develop novel bispecific antibodies (BsAb) consisting of a DR5 agonist and anti-FAP antibody fusion to enhance targeted delivery of the DR5 agonist, induce avidity-driven DR5 hyperclustering and activate the extrinsic apoptotic pathway specifically in tumor cells. We demonstrate that FAP-DR5 antibodies significantly enhance tumor cell apoptosis in vitro across several tumor indications compared with the parental drozitumab mAb. Importantly, the antitumor activity of the FAP-DR5 antibodies was dependent on the presence of FAP-expressing fibroblasts. Using an optimized FAP-DR5 BsAb RG7386, we further show that the mode of action of the DR5 agonist can be uncoupled from FcγR-dependent cross-linking when fused to the FAP-binding moiety. RG7386 demonstrates in vivo efficacy in several xenograft models, including patient-derived xenograft (PDX) models with FAP expression either on stroma or on malignant cells. Finally, RG7386 in combination with irinotecan or doxorubicin enhances the antitumor effects of the respective monotherapies in vivo and results in complete tumor remission.

Cell lines and host strains

HEK293 EBNA, NIH-3T3, MDA-MB-231, DLD-1, Colo205, and LOX-IMVI were obtained from LGC Standards (European distributor for the ATCC), A549 from ECACC, GM05389 from the Coriell Institute for Medical Research (Camden, NJ), and 1833-PPOP233 from Promega Corp. All cell lines were purchased between 2007 and 2010 and authenticated by morphologic and growth curve analysis. For more details see Supplementary Materials and Methods.

Vector construction

The construction of expression vectors for standard IgGs (immunoglobulin G) and BsAbs was performed according to standard recombinant DNA technologies. Each antibody chain cDNA was amplified by PCR or generated by conventional gene synthesis at Geneart and transiently expressed in HEK293 EBNA cells under the MPSV promoter (myeloproliferative sarcoma virus). Bispecific FAP-DR5 antibodies were generated by fusion of a FAP-binding domain to the DR5 IgG heavy chain at the C-terminus via a (G4S)4 linker. The DR5 portion consisted of the variable-light chain (VL) and variable-heavy chain (VH) of drozitumab (described by Adams in US2007/003141401 A1) or novel DR5 antibodies generated by phage display. To minimize light-chain mispairing side-products, we used CrossMab technology (17). The FAP-binding unit was engineered as a crossed Fab in which the VH was fused to the constant-light chain (CL) and the VL to a CH1 (constant-heavy 1) domain. To prevent FcR binding, three mutations in the Fc region [PGLALA: P329G (patent No. WO 2012130831) and L234A and L235A (18)] were generated by standard site-directed mutagenesis in the indicated BsAbs. All amplified or synthesized genes were confirmed by DNA sequencing.

Production and characterization of anti-DR5 binders

Antibodies with specificity for human DR5 were selected from a generic phage-display antibody library in the Fab format. Specific binders were identified by ELISA assays and clones exhibiting signals on human DR5 but none on human DCR2 and human IgG1 were short-listed for further analyses. All BsAbs and antigens (if not obtained from a commercial source) were transiently produced in HEK293 EBNA cells and purified using protein A affinity purification and size exclusion chromatography. The purity and molecular weight of the BsAb preparations were determined by Microchip capillary electrophoresis (CE)-SDS on Caliper's LabChip GXII using the Protein Express LabChip Kit. For more details see Supplementary Materials and Methods.

Binding of BsAbs and flow cytometry

Simultaneous binding of BsAbs to human DR5 as well as hu or muFAP was assessed by surface plasmon resonance (SPR) using Biacore T100. The binding of BsAbs on cells was determined using a time-resolved fluorescence resonance energy transfer (TR-FRET) assay. In the flow cytometry, anti-FAP and anti-DR5 were used as a primary and goat anti-human Fc specific PE-labeled as a secondary antibody. For more details see Supplementary Materials and Methods.

Apoptosis detection by DNA fragmentation

GM05389 fibroblasts (1 × 104 cells/well), harvested with Cell Dissociation Buffer and resuspended in appropriate medium, were plated overnight at 37°C in 96-well plates. Fibroblasts were then incubated with antibodies (anti-DR5 IgGs or FAP-DR5 BsAbs) alone or together in a 1:1 ratio with a goat anti-human IgG cross-linking antibody (Sigma #I2136) at the indicated concentrations for 10 minutes. Tumor cells (1 × 104 cells/well) were harvested in Cell Dissociation Buffer, resuspended in appropriate medium and were added to the fibroblasts. Cells were incubated with the antibodies for 24 hours at 37°C followed by centrifugation for 10 minutes at 200 × g at room temperature. The amount of fragmented DNA was determined using Cell Death detection ELISAPLUS (Roche Applied Science #11 774 425 001) according to the manufacturer's instructions. Where indicated Vmax values were calculated from absorbance readings taken every minute over 20 minutes at 405 nm using a Versamax Microplate Reader (Molecular Devices). Measurements at 490 nm were used as a reference. In other experiments, end-point absorbance readings were taken at 405 nm using a Tecan Spectra Rainbow Reader.

Mouse xenograft models

All experiments were conducted according to the guidelines of the German Animal Welfare Act (GV-Solas; Felasa; TierschG).

For the cell line–based xenograft models, tumor cells (2 × 106 cells) were co-injected s.c. onto Balb/c nude mice (Charles River Laboratories) together with murine fibroblasts (NIH3T3, 0.4 × 106 cells). Cell inoculation was supplemented with Matrigel (Basement Membrane Matrix, BD Biosciences). The fragment-based colorectal cancer xenograft Co5896 and sarcoma tumor xenograft Sarc4605 originally obtained from patients were passaged approximately three to five times until establishment of stable growth patterns. For the subsequent in vivo PDX studies, tumors were removed from xenografts in serial passage in donor nude mice, cut into fragments (4–5 mm diameter), and placed in PBS until subcutaneous implantation in recipient mice.

Mice (n = 10/group) were subjected to stratified randomization based on primary tumor size (median tumor volume of approximately 100–150 mm3) before treatment. Unless otherwise indicated, FAP-DR5 BsAb RG7386, drozitumab and drozitumab-PGLALA were administered once weekly i.v. Irinotecan (15 mg/kg) was administered intraperitoneally, doxorubicin (5 mg/kg) was given weekly intravenously, and saline was used as the vehicle control. Tumor volume was measured by caliper twice weekly [(length × width2)/2] and the percentage of tumor growth inhibition (TGI) relative to control animals was calculated. All treatments were well tolerated as indicated by stable body weights throughout the duration of the studies.

Bioluminescence detection of apoptosis in vivo

1833-PPOP233 cells (4 × 106 cells/mouse) and NIH3T3 cells (1 × 106 cells/mouse) were subcutaneously coinjected into the right flank of 6- to 8-week-old Scid/beige mice (Charles River Laboratories) with body weights of 18 to 24 g. After treatment with BsAbs, mice were injected with a single dose of d-luciferin (Promega; 150 mg/kg in PBS; i.p.) 10 minutes before bioluminescence imaging using IVIS spectrum (Caliper Life Sciences). The acquired bioluminescence signals in a region of interest (ROI) over the tumor site were measured as average radiance (p/sec/cm3/sr) with Living Image software (Caliper Life Sciences). Every ROI signal was divided by the appropriate baseline ROI signal to calculate fold induction (19, 20).

IHC

IHC protocols for cleaved caspase-3 and FAP were performed on Discovery or BenchMark XT automated strainers. All reagents except the antibodies were obtained from Ventana Medical Systems. For more details see Supplementary Materials and Methods.

Statistical analyses

Nonparametric treatment-to-control-ratios (TCR) based on endpoint analysis and the two-sided nonparametric confidence intervals (CI) compared with the vehicle group were calculated to assess statistical significance in in vivo studies. A TCR value of 0 indicates complete tumor regression and CIs of less than 1 represent statistically significant results. Relative TGI was calculated according to the formula (1−[(T−T0)/(C−C0)]) × 100.

A novel bispecific anti-FAP and anti-DR5 antibody exhibits dual specificity

To overcome the disadvantages of current DR5 antibodies, and to boost antitumor efficacy, we engineered novel BsAbs that simultaneously bind to DR5 on tumor cells and to FAP on the tumor stroma to induce localized DR5 hyper–cross-linking. The BsAbs consisted of a human anti-DR5 agonist (drozitumab-derived huIgG1) with a C-terminal fusion of an anti-FAP binding moiety (mAb007 or affinity-matured mAb082 clone; Fig. 1A). For heterodimerization of this trivalent molecule, the “knobs-into-holes” technology (21) was implemented. All FAP-drozitumab antibodies exhibited similar binding to human DR5 as the parental drozitumab and a nontargeting BsAb (drozitumab fused to non-binding DP47GS Cross Fab as a control), demonstrating that the process of antibody fusion had not impaired antibody binding to the DR5 antigen in cells (Fig. 1B; Supplementary Table S1). Interestingly, we found that the Fab configuration used to fuse FAP to drozitumab was important for preserving BsAb interaction with the FAP antigen. In a VLCH1 format, mAb007_drozitumab demonstrated reduced binding to FAP (>9-fold), compared with parental mAb007 antibody. Binding to FAP was similarly reduced by a BsAb with a monovalent FAP-binding domain (Fig. 1C; Supplementary Table S1), corresponding to a loss in avidity. In contrast, in the VHCL format, mAb007_drozitumab and mAb082_drozitumab demonstrated similar binding to FAP (Ki = 0.32 nmol/L and Ki = 0.53 nmol/L, respectively) that was comparable with the parental antibodies (Fig. 1C; Supplementary Table S1). These data highlight that the VHCL Fab configuration is the most suitable format for generating BsAbs.

Figure 1.

FAP-DR5 BsAb and apoptosis activity. A, principle of tumor-targeted apoptosis by BsAbs consisting of a CrossFab anti-FAP unit (mAb007 or mAb082) fused to the C-terminus of the drozitumab heavy chain using a 20mer GS linker. The mAb007 anti-FAP moiety was fused to drozitumab heavy chain in either a VHCL or VLCH1 configuration. A nontargeting DP47GS_drozitumab antibody was generated in the VHCL format (not shown here). A trivalent mAb007_drozitumab molecule (1+2; VHCL) was also developed. B and C, BsAb binding to antigens DR5 (B) and FAP (C) was assessed using a TagLite competition assay in HEK EBNA cells expressing huDR5 and huFAP extracellular domains, respectively. D–G, analysis of MDA-MB-231 tumor cell apoptosis in coculture with FAP-expressing GM05389 in response to drozitumab alone (D), drozitumab + anti-Fc (cross-linked drozitumab; E) and BsAbs (F–I) at the indicated concentrations. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Vmax values calculated from kinetic measurements taken at 405 nm are shown (mean ± SEM; n = 3 independent experiments). See also Supplementary Fig. S1.

Figure 1.

FAP-DR5 BsAb and apoptosis activity. A, principle of tumor-targeted apoptosis by BsAbs consisting of a CrossFab anti-FAP unit (mAb007 or mAb082) fused to the C-terminus of the drozitumab heavy chain using a 20mer GS linker. The mAb007 anti-FAP moiety was fused to drozitumab heavy chain in either a VHCL or VLCH1 configuration. A nontargeting DP47GS_drozitumab antibody was generated in the VHCL format (not shown here). A trivalent mAb007_drozitumab molecule (1+2; VHCL) was also developed. B and C, BsAb binding to antigens DR5 (B) and FAP (C) was assessed using a TagLite competition assay in HEK EBNA cells expressing huDR5 and huFAP extracellular domains, respectively. D–G, analysis of MDA-MB-231 tumor cell apoptosis in coculture with FAP-expressing GM05389 in response to drozitumab alone (D), drozitumab + anti-Fc (cross-linked drozitumab; E) and BsAbs (F–I) at the indicated concentrations. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Vmax values calculated from kinetic measurements taken at 405 nm are shown (mean ± SEM; n = 3 independent experiments). See also Supplementary Fig. S1.

Close modal

Importantly, SPR experiments confirmed simultaneous binding of the FAP-drozitumab antibodies to both DR5 and FAP recombinant proteins, indicative of a fully functioning dual antibody (Supplementary Fig. S1A).

FAP-DR5 bispecific molecules induce tumor cell apoptosis in vitro in the presence of FAP-expressing fibroblasts

To investigate the antitumor efficacy and specificity of the FAP-drozitumab BsAbs, tumor cell apoptosis (DNA fragmentation) was monitored in the presence or absence of the FAP-positive human GM05389 fibroblast cell line (Supplementary Fig. S1B).

In the absence of FAP-expressing fibroblasts, low concentrations of drozitumab or BsAb resulted in minimal MDA-MB-231 breast cancer cell apoptosis. However, DNA fragmentation was observed at the highest antibody concentrations used (7 and 35 nmol/L; Fig. 1D–I). Induction of drozitumab cross-linking by addition of a secondary anti-human Fc antibody promoted dose-dependent tumor cell apoptosis with a significant 100-fold reduction in EC50 values from 15.2 nmol/L (drozitumab alone) to 0.14 nmol/L (cross-linked drozitumab; Fig. 1D and E; Supplementary Table S2), demonstrating that cross-linking is critical for antibody efficacy.

In the presence of FAP-expressing fibroblasts, all BsAbs substantially increased tumor cell apoptosis (Fig. 1F–I). Under these conditions, BsAb treatment resulted in dose-dependent cell death at concentrations as low as 0.0112 nmol/L with maximum activity at 7 nmol/L. In addition, the EC50 value of the bispecific molecules was approximately 20-fold lower as compared with the anti-Fc cross-linked drozitumab (0.014 vs. 0.33 nmol/L; Supplementary Table S2). Importantly, the parental drozitumab antibody did not trigger apoptosis in the same MDA-MB-231 and fibroblast coculture assay without cross-linking. These data clearly implicate the requirement for FAP expression in the tumor stroma for bispecific FAP-drozitumab antibody function. Interestingly, the antibody configuration did not appear to hinder the in vitro potency of the bispecific molecules (Fig. 1G and H; Supplementary Table S2) despite differential binding to the FAP antigen (Fig. 1B and C). However, it appeared that antibody valence for the FAP antigen was crucial for BsAb potency (EC50 value increased from 0.01 in the bivalent molecule to 0.57 nmol/L in the monovalent format; Supplementary Table S2). Altogether, the BsAb containing the mAb007 anti–FAP-binding moiety appeared to be less specific and induced tumor cell apoptosis in the absence of fibroblasts at lower concentrations. Therefore, the mAb082 anti–FAP-binding moiety was adopted for further BsAb development and in vivo experiments.

The FAP dependency of mAb082_drozitumab to trigger DR5 clustering and apoptosis was further evaluated in the colorectal cancer DLD-1 cell line. In the presence of NIH3T3 fibroblasts that lack FAP expression in vitro (Supplementary Fig. S1B), the antibody failed to trigger tumor cell apoptosis (Supplementary Fig. S1C). In contrast, stable expression of murine FAP in NIH3T3 cells (muFAP NIH3T3; Supplementary Fig. S1B) resulted in significant antibody-mediated tumor cell death (Supplementary Fig. S1D). Moreover, the untargeted control molecule DP47GS_drozitumab, which does not bind FAP, failed to induce apoptosis even in the presence of FAP-expressing cells (Supplementary Fig. S1C–S1D). Together, these data demonstrate the fundamental requirement for FAP-positive cells in cross-linking of the FAP-drozitumab BsAbs and in the activation of the DR5 apoptotic pathway.

BsAb-mediated DR5 cross-linking reveals superior apoptosis induction over untargeted drozitumab in a variety of tumor cell lines

To assess the generalizability of the FAP-drozitumab antibody, five tumor cell lines were evaluated for DR5 cell surface expression and apoptosis sensitivity in response to BsAb treatment. Flow-cytometry analyses revealed two cell lines with high (ACHN; renal carcinoma and DLD-1 colorectal cancer), two with moderate (Colo205; colorectal cancer and MDA-MB-231 breast cancer), and one cell line with low DR5 expression (A549; lung carcinoma; Fig. 2A). All of these tumor cell lines demonstrated susceptibility to FAP-drozitumab antibody-mediated apoptosis (Fig. 2B) in a dose-dependent manner. Moreover, BsAb action required FAP targeting as the untargeted BsAb control (DP47GS_drozitumab) failed to induce apoptosis at low doses (Fig. 2B). As previously discussed, the apoptotic activity of DP47GS_drozitumab at higher concentrations could be attributed to the action of drozitumab alone (Fig. 1D). Intriguingly, and in agreement with a previous publication (22), no obvious correlation was apparent between DR5 cell surface expression and FAP-drozitumab BsAb-induced apoptosis (Fig. 2; Supplementary Table S3).

Figure 2.

FAP-drozitumab antibody induces apoptosis in several tumor cell lines. A, flow-cytometry analysis of DR5 cell surface expression, using drozitumab IgG, in cell lines representing different tumor indications. B, analysis of tumor cell apoptosis in coculture with FAP-expressing GM05389 fibroblasts in response to mAb082_drozitumab or DP47GS_drozitumab nontargeting control. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Vmax values were calculated from kinetic measurements taken at 405 nm and represented as fold induction over untreated cells. Dose–response curves (mean ± SEM; n = 3 independent experiments) are shown.

Figure 2.

FAP-drozitumab antibody induces apoptosis in several tumor cell lines. A, flow-cytometry analysis of DR5 cell surface expression, using drozitumab IgG, in cell lines representing different tumor indications. B, analysis of tumor cell apoptosis in coculture with FAP-expressing GM05389 fibroblasts in response to mAb082_drozitumab or DP47GS_drozitumab nontargeting control. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Vmax values were calculated from kinetic measurements taken at 405 nm and represented as fold induction over untreated cells. Dose–response curves (mean ± SEM; n = 3 independent experiments) are shown.

Close modal

Altogether, these data indicate the potential for the broad application of FAP-drozitumab BsAbs in a range of tumor indications and highlight the importance of FAP targeting for BsAb function.

An optimized bispecific FAP-DR5 antibody, RG7386 induces apoptosis in vitro and inhibits in vivo tumor growth in xenograft mouse tumor models

Conventional anti-DR5 antibodies such as drozitumab rely on FcγR-dependent mechanisms to achieve DR5 clustering (13). Therefore, efficient DR5 activation may be hindered by several factors, including high concentrations of endogenous IgG, present in patient sera, and by the activity of FcγR-expressing immune cells within normal tissue leading to limited antitumor efficacy and unwanted side-effects. In addition, FcγR-dependent cross-linking in the liver has been shown to mediate hepatotoxicity for murine FAS agonistic antibodies (23), and a similar mechanism may play a role in the hepatic side-effects reported for previous DR5 agonists in the clinic (24). Moreover, our data suggested that the parental drozitumab antibody triggers cross-linker–independent cytotoxicity at concentrations above 1 nmol/L (Fig. 1D). To eliminate these confounding variables and to improve future clinical safety profiles, we generated a BsAb composed of another DR5 antibody mAb096, which does not trigger apoptosis in the absence of cross-linking (Supplementary Fig. S2A). Furthermore, to prevent interaction with host FcγR on host immune cells, we introduced previously reported IgG1 point mutations [PGLALA: P329G (patent No. WO 2012130831) and L234A and L235A (18)] in mAb096 that completely abolish FcγR and C1q binding while still retaining cross-linking activity by anti-Fc antibodies.

In the absence of fibroblasts, this optimized FAP-DR5 BsAb (mAb096-mAb082 in VHCL configuration) named RG7386 (characterized in Supplementary Table S4 and Supplementary Fig. S2B), promoted MDA-MB-231 tumor cell apoptosis only with the addition of a secondary anti-Fc antibody similarly to the parent mAb096–anti-Fc combination (Fig. 3A). Importantly, the BsAb displayed more potent tumor cell apoptosis in the presence of FAP-expressing GM05389 fibroblasts (EC50 = 0.3 nmol/L) compared with the cross-linked parent molecule (EC50 = 5.3 nmol/L) and the addition of the anti-Fc secondary antibody had no additive effects on RG7386-mediated MDA-MB-231 tumor cell death in cocultures (EC50 = 0.5 nmol/L; Fig. 3B). In addition, RG7386 had minimal effects on fibroblast cell survival in monocultures (Fig. 3C). These data demonstrate the strong and specific antitumor effects of RG7386 in response to FAP-mediated cross-linking.

Figure 3.

RG7386 induces tumor cell apoptosis in in vitro cocultures. A and B, analysis of MDA-MB-231 cell apoptosis in monocultures (A) and in coculture with FAP-expressing GM05389 fibroblasts (B) in response to the indicated treatments. C, analysis of RG7368-mediated apoptotic effects in GM50389 monocultures. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Endpoint absorbance measurements at 405 nm are shown (representative experiment of n = 3–5). See also Supplementary Fig. S2.

Figure 3.

RG7386 induces tumor cell apoptosis in in vitro cocultures. A and B, analysis of MDA-MB-231 cell apoptosis in monocultures (A) and in coculture with FAP-expressing GM05389 fibroblasts (B) in response to the indicated treatments. C, analysis of RG7368-mediated apoptotic effects in GM50389 monocultures. Apoptosis was determined as a measure of DNA fragmentation in an ELISA assay. Endpoint absorbance measurements at 405 nm are shown (representative experiment of n = 3–5). See also Supplementary Fig. S2.

Close modal

The antitumor efficacy of RG7386 was further investigated in vivo in xenograft mouse models. To obtain comparable FAP expression levels with those observed in the human tumor stroma, FAP-expressing NIH3T3 cells were s.c. coinjected with DLD-1 tumor cells onto nude mice and FAP expression in the tumor microenvironment was confirmed with IHC (Fig. 4A). Dose-optimization studies revealed effective TGI following once weekly administration of RG7386 at 3 and 10 mg/kg relative to vehicle control (Supplementary Fig. S3A). In terms of pharmacokinetics, the exposure to RG7386 (AUC0–72h) appeared to increase in a dose-dependent manner with exposure of the largest dose (10 mg/kg) being somewhat higher than expected (Supplementary Fig. S3B).

Figure 4.

RG7386 induces tumor cell apoptosis in vivo in human xenograft mouse models. A, antitumor efficacy of RG7386 in a mouse xenograft model. Nude mice were subcutaneously coinjected with human DLD-1 and NIH3T3 cells and subjected to RG7386 (twice weekly doses) or Drozitumab_PGLALA antibody treatment when tumor volume was approximately 100 mm3 in size. Shown is a representative FAP IHC of the tumor (median tumor volume and interquartile range (IQR); n = 10 animals/group). B and C, apoptosis was monitored in mice coinjected with bone-metastasizing MDA-MB-231 1833-PPOP233 tumor cells and NIH3T3 cells. B, in vivo analysis of apoptosis using a luminescence caspase 3/7 activation reporter assay in response to the indicated treatments over 72 hours. Luminescence images were taken at 0, 6, and 72 hours posttreatment. Representative luminescence images at 6 hours are shown (mean ± SEM; n = 5 animals/group). C, analysis of TGI in the same model in response to RG7386 (median tumor volume and IQR; n = 10 animals/group). D, IHC validation of caspase-3 activation in response to RG7386. See also Supplementary Fig. S4.

Figure 4.

RG7386 induces tumor cell apoptosis in vivo in human xenograft mouse models. A, antitumor efficacy of RG7386 in a mouse xenograft model. Nude mice were subcutaneously coinjected with human DLD-1 and NIH3T3 cells and subjected to RG7386 (twice weekly doses) or Drozitumab_PGLALA antibody treatment when tumor volume was approximately 100 mm3 in size. Shown is a representative FAP IHC of the tumor (median tumor volume and interquartile range (IQR); n = 10 animals/group). B and C, apoptosis was monitored in mice coinjected with bone-metastasizing MDA-MB-231 1833-PPOP233 tumor cells and NIH3T3 cells. B, in vivo analysis of apoptosis using a luminescence caspase 3/7 activation reporter assay in response to the indicated treatments over 72 hours. Luminescence images were taken at 0, 6, and 72 hours posttreatment. Representative luminescence images at 6 hours are shown (mean ± SEM; n = 5 animals/group). C, analysis of TGI in the same model in response to RG7386 (median tumor volume and IQR; n = 10 animals/group). D, IHC validation of caspase-3 activation in response to RG7386. See also Supplementary Fig. S4.

Close modal

Importantly, treatment with RG7386 (10 mg/kg) significantly inhibited tumor growth (TCR, 0.31; 95% CI, 0.17–0.66; TGI = 89%) whereas drozitumab-PGLALA (mutant drozitumab unable to be cross-linked without addition of a secondary anti-Fc antibody), failed to have any effect on tumor size as compared with vehicle-treated mice (TCR, 1.075; 95% CI, 0.65–2.43; TGI = 22%; Fig. 4A). As RG7386 also lacks these FcγR-binding sites, these data strengthen the notion that FAP-dependent cross-linking drives the antitumor effects of this FAP-DR5 BsAb. Importantly, the in vivo efficacy of the FAP-DR5 antibody complex to inhibit tumor growth was recapitulated using the previous mAb082_drozitumab molecule in the same DLD-1 colorectal cancer model (TCR, 0.12; 95% CI, 0.00–0.33; TGI > 100% compared with vehicle control) and in the MDA-MB-231 breast cancer model (TCR, 0.34; 95% CI, 0.24–0.47; TGI: 77% compared with vehicle control; Supplementary Fig. S4), demonstrating the potential broad application of these bispecific molecules. Moreover, drozitumab coupled to the nontargeting DP47GS molecule did not affect tumor growth, further confirming the FAP-dependent targeting action of the FAP-DR5 antibodies to inhibit tumor growth in vivo (Supplementary Fig. S4).

To determine whether the RG7386-mediated suppression of tumor growth was specifically a result of DR5-activated tumor cell apoptosis and not through an unidentified mode of antibody action, cell apoptosis was monitored noninvasively in a human tumor xenograft mouse model by bioluminescence imaging. The 1833-PPOP233 cells (human bone-metastasizing MDA-MB-231 subclone) expressing a split-luciferase construct with a caspase 3/7 cleavage site (20), together with FAP-expressing NIH3T3 cells were s.c. inoculated in mice. Cleavage of the luciferase construct, resulting in a detectable luminescence signal, was assessed over time and used as a measure of apoptosis. RG7386 treatment (1.0 and 10 mg/kg) of tumor-bearing mice resulted in a rapid and significant increase in the luminescence signal indicative of apoptosis induction downstream of activated caspase-3/7 (Fig. 4B). Importantly, the enhanced luminescence signal denoting increased apoptosis is in agreement with inhibition of tumor growth, in the same mouse model, following RG3786 treatment (Fig. 4C). Increased caspase-3 activation (brown staining) was also confirmed in the vicinity of stromal bands by IHC staining of RG7386-treated DLD-1 / 3T3 coinjected tumor sections (Fig. 4D). These data clearly demonstrate that RG7386 induces tumor cell apoptosis in vivo to restrict tumor growth.

The bispecific FAP-DR5 antibody RG7386 demonstrates in vivo efficacy in PDX models and more than additive effects in combination with irinotecan

To further validate the in vivo efficacy of RG7386 in a more relevant disease background, a patient-derived fragment-based colorectal cancer tumor (Co5896) was s.c. transplanted onto NMRI nude mice and FAP expression was confirmed with IHC analysis (Fig. 5A). RG7386 treatment (30 mg/kg, twice weekly) of these mice led to a significant decrease in tumor burden (TCR, 0.412; 95% CI, 0.24–0.84; TGI = 76%; Fig. 5B). Interestingly, comparison of treatment with control groups revealed that a combination of RG7386 and the standard-of-care irinotecan (chemotherapeutic agent) translated into superior efficacy (TCR, 0.00; 95% CI, 0.00–0.004; TGI > 100%) relative to either RG7386 (TCR, 0.19; 95% CI, 0.05–0.37) or irinotecan monotherapy (TCR, 0.036; 95% CI, 0.02–0.09) alone. Direct comparison between each treatment group further confirmed that the enhanced efficacy of RG7386 and irinotecan combination was more than additive (TCR, 0.00; 95% CI, 0.00–0.019, and TCR, 0.00; 95% CI, 0.00–0.017, compared with RG7386 and irinotecan monotherapies, respectively) and resulted in prolonged antitumor activity with complete eradication of tumors in all mice (Fig. 5C). Together, these data demonstrate the potential cumulative effects of RG7386 when used with the standard-of-care for the treatment of human colorectal cancers.

Figure 5.

RG7386 demonstrates in vivo efficacy and additive effects in combination with irinotecan in a colorectal cancer (CRC) PDX model. A, IHC analysis of FAP expression in a subcutaneous Co5896 PDX model. B, analysis of Co5896 TGI (median tumor volume and IQR; n = 10 animals/group) in response to RG7386 (twice weekly administration). C, combination of RG7386 and irinotecan treatment of colorectal cancer PDX tumors. (Median tumor volume and IQR; n = 10 animals/group). Median tumor volume for vehicle-treated animals extends beyond the y-axis shown here.

Figure 5.

RG7386 demonstrates in vivo efficacy and additive effects in combination with irinotecan in a colorectal cancer (CRC) PDX model. A, IHC analysis of FAP expression in a subcutaneous Co5896 PDX model. B, analysis of Co5896 TGI (median tumor volume and IQR; n = 10 animals/group) in response to RG7386 (twice weekly administration). C, combination of RG7386 and irinotecan treatment of colorectal cancer PDX tumors. (Median tumor volume and IQR; n = 10 animals/group). Median tumor volume for vehicle-treated animals extends beyond the y-axis shown here.

Close modal

RG7386 demonstrates significant efficacy in FAP-positive human mesenchymal tumor models in mice

Thus far, we have focused on the role of RG7386 in initiating tumor cell apoptosis with a complete dependence on the presence of FAP-positive fibroblasts. However, FAP expression is not restricted to the tumor stroma and can be found as a minor or major component of malignant cells in mesenchymal-derived tumors. To investigate whether RG7386 could be used to directly target FAP-positive tumors, LOX-IMVI desmoplastic melanoma cells were s.c. injected onto mice and tumors were allowed to develop. Treatment with 10 mg/kg of RG7386 triggered a significant inhibition of tumor growth compared with vehicle control (TCR, 0.073; 95% CI, 0.03–0.32) and compared with drozitumab-PGLALA (TCR, 0.27; 95% CI, 0.17–0.48), indicating that FAP-dependent cross-linking contributes to enhanced efficacy (Fig. 6A). Moreover, RG7386, administered as a single agent, substantially restricted tumor development (TCR, 0.16; 95% CI, 0.06–0.36; TGI, 99%) in an osteosarcoma Sarc4605 fragment-based PDX model with 100% FAP expression (Fig. 6B). In the same model, complete tumor remission (TGI > 100%) was achieved when RG7386 was combined with the standard-of-care doxorubicin (5 mg/kg; TCR, 0.03; 95% CI, 0.007–0.09; TCR, 0.15; 95% CI, 0.04–0.54 and TCR, 0.06; 95% CI, 0.02–0.14 compared with vehicle, RG7386, and doxorubicin as single agents, respectively; Fig. 6C). Altogether, these data support the use of RG7386 not only in the treatment of multiple carcinomas associated with a FAP-positive stroma but also as a potential viable therapy for mesenchymal tumors directly expressing FAP.

Figure 6.

RG7386 inhibits growth of FAP-positive mesenchymal tumors. A, RG7386-mediated antitumor efficacy was monitored in a FAP-positive LOX-IMVI desmoplastic melanoma cell line (median tumor volume and IQR; n = 6 animals/group) and in a Sarc4605 sarcoma PDX model (median tumor volume and IQR; n = 10 animals/group; B). C, combination of RG7386 and doxorubicin treatment of Sarc4605 tumors (median tumor volume and IQR; n = 10 animals/group). Median tumor volume for vehicle-treated animals extends beyond the y-axis shown here.

Figure 6.

RG7386 inhibits growth of FAP-positive mesenchymal tumors. A, RG7386-mediated antitumor efficacy was monitored in a FAP-positive LOX-IMVI desmoplastic melanoma cell line (median tumor volume and IQR; n = 6 animals/group) and in a Sarc4605 sarcoma PDX model (median tumor volume and IQR; n = 10 animals/group; B). C, combination of RG7386 and doxorubicin treatment of Sarc4605 tumors (median tumor volume and IQR; n = 10 animals/group). Median tumor volume for vehicle-treated animals extends beyond the y-axis shown here.

Close modal

Many cancer therapeutic approaches target the specific molecules/signaling pathways that promote the hyperproliferative/antiapoptotic states in tumor cells. The continuing challenge in these methodologies, as opposed to conventional chemotherapeutics that target all rapidly dividing cells, is to be able to specifically discriminate between tumor cells and their healthy counterparts without loss in efficacy. For conventional DR5 agonistic antibodies, such as drozitumab and conatumumab, efficacy is strongly dependent on cross-linking that is mediated by FcγRs in in vivo mouse models (11, 13, 25). However, despite very convincing and potent in vitro and in vivo preclinical data, this mode of agonistic antibody action has severe drawbacks that may explain the limited clinical efficacy observed for first-generation DR agonists (5, 26–28).

In mouse xenograft models, significant tumoricidal activity of DR5 agonists is dependent on sufficient infiltration of FcγR-expressing immune cells. However, this situation does not appear to be relevant in humans, where FcγR-mediated antibody cross-linking may be reduced by physiologic IgG levels. On the other hand, potential adverse events such as elevated hepatotoxicity are related to nontumor specific, systemic crosslinking of agonistic antibodies via FcγRIIb molecules present in the liver (23). Consequently, second-generation DR agonistic antibodies are currently under development with a focus on both DR cross-linking to enhance efficacy, and on reducing the risk of toxicity due to non–tumor-targeted effects. Conatumumab-coated nanoparticles encasing a cytotoxic drug that allow a sufficient density of antibody paratopes at the tumor site to activate DR5 are currently under investigation (29). APG350 is a hexavalent scTRAIL–RBD (single-chain TRAIL receptor–binding domain), which in contrast with previous agonistic antibodies triggers multimer formation independently of FcγRs (30).

In addition to full-length antibodies, different scaffolds such as atrimers (31) or multivalent monobodies (32, 33) are under evaluation to target DRs. However, the lack of clinical efficacy observed with untargeted DR5 agonistic antibodies illustrates that the activity of these molecules in vitro may not necessarily translate into clinical efficacy. Multivalent DR5 binders might provide a more potent effect as compared with agonistic antibodies, but these also bear the risk of toxic side effects. TAS266, a novel tetravalent and highly potent DR5 agonistic Nanobody, was recently entered into a phase I clinical trial, but was deemed unsafe for human administration due to acute hepatotoxicity (34) that may have been exacerbated by an absence of a tumor-targeting entity. The DR ligand, TRAIL, untargeted, targeted, or in multimeric formats (35–38) also has some disadvantages such as a short half-life and potential toxicity, if oligomerized. In addition, as TRAIL does not only bind to DR4 and DR5, but also to decoy receptors DCR1, DCR2, and OPG (osteoprotegerin; ref.39) in vivo tumor activity may be limited.

In our study, we eliminated FcγRs-dependent formation of antibody multimers by introducing three mutations (PGLALA) in the Fc portion of a novel DR5 agonist (mAb096) that abolish FcγR binding. In addition, we fused the DR5 agonistic antibody to an anti–FAP-binding moiety to take advantage of FAP expression in the tumor stroma as an alternative mechanism to promote hyperclustering of DR5 in a targeted manner. The tumor-associated stroma contributes to a significant proportion of the mass of many malignancies (14) and the stromal fibroblasts play an important role in the development, growth and metastasis of carcinomas (40–42). FAP, normally restricted in healthy adult tissue (except in granulation tissue, e.g., during wound healing), is expressed in tumor stromal fibroblasts of more than 90% of epithelial cancers (15, 16, 43), and therefore is an ideal candidate for targeted delivery of cancer therapeutics. The FAP-DR5 BsAbs we described here, including RG7386, exhibited high specificity and a clear dependency on FAP-based cross-linking to promote tumor cell death. In vitro, apoptosis induction of tumor target cells only occurred in the presence of FAP-expressing fibroblasts and replacement of the FAP CrossFab unit with a non-binding IgG (DP47GS) was sufficient to prevent tumor cell apoptosis. In addition, the FAP-dependent mode of BsAb action successfully translated into in vivo efficacy in several xenograft models, including a colorectal cancer PDX and a sarcoma PDX model. The antitumor effects observed in the sarcoma PDX model also indicate that RG7386 function is not restricted to an environment consisting of a FAP-positive stroma, but can also be applied to mesenchymal-derived tumors that express FAP directly on malignant cells.

The broad application of our FAP-DR5 bispecific molecules was evident in five different tumor cell lines (lung, renal, colorectal, and breast cancer cells). However, the degree of apoptosis was different for each cell line and did not necessarily correlate with DR5 expression levels, indicating that there are additional factors influencing this mechanism. In some cells the DR-initiated signal requires amplification to induce maximum apoptosis. Correspondingly, we demonstrate increased inhibition of tumor growth in patient-derived colorectal cancer and sarcoma models in the presence of both RG7386 with irinotecan or with doxorubicin, respectively, which may be indicative of synergistic extrinsic and intrinsic apoptosis signaling (7, 44). It is, however, important to note that p53, an important tumor suppressor and a key regulator of the intrinsic pathway, is inactivated in many human cancers and the high prevalence of p53 mutants (dominant negative forms) contributes significantly to chemotherapeutic resistance (44–46). As a consequence, activation of the extrinsic pathway offers an attractive target for cancer therapy as the apoptotic cascade is p53 independent, and hence not susceptible to development of apoptosis resistance (47).

Taken together, our preclinical data strongly suggest that RG7386, a novel FAP-DR5 tetravalent BsAb, is superior to existing DR5 agonists as it offers a targeted strategy to specifically activate the extrinsic apoptosis pathway in tumor cells by avidity-driven DR5 hyperclustering. Future studies will be essential for clinical development of RG7386 particularly for the successful application of combination therapies.

C. Klein has ownership interest (including patents) in Roche. No potential conflicts of interest were disclosed by the other authors.

Conception and design: P. Brünker, K. Wartha, T. Friess, S. Grau-Richards, B. Weiser, H. Niu, K. Packman, C. Klein, P. Umaña

Development of methodology: P. Brünker, T. Friess, S. Grau-Richards, M. Majety, V. Runza, H. Niu, K. Packman, N. Feng, S. Daouti, E. Mössner, T.G. Weber, W. Scheuer, H. Sade, C. Shao, B. Liu, P. Wang, C. Klein, P. Umaña

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Friess, S. Grau-Richards, I. Waldhauer, C.F. Koller, M. Majety, H. Niu, N. Feng, S. Daouti, R.J. Hosse, E. Mössner, T.G. Weber, F. Herting, H. Sade, C. Shao, B. Liu, P. Wang, G. Xu, S. Vega-Harring, K. Bosslet, P. Umaña

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): P. Brünker, K. Wartha, T. Friess, S. Grau-Richards, I. Waldhauer, C.F. Koller, B. Weiser, M. Majety, V. Runza, H. Niu, S. Daouti, R.J. Hosse, E. Mössner, T.G. Weber, F. Herting, W. Scheuer, H. Sade, C. Shao, G. Xu, S. Vega-Harring, P. Umaña

Writing, review, and/or revision of the manuscript: P. Brünker, T. Friess, S. Grau-Richards, I. Waldhauer, C.F. Koller, M. Majety, V. Runza, H. Niu, F. Herting, S. Vega-Harring, C. Klein, K. Bosslet, P. Umaña

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): P. Brünker, S. Grau-Richards, B. Weiser, E. Mössner, F. Herting, C. Shao, S. Vega-Harring, K. Bosslet, P. Umaña

Study supervision: T. Friess, M. Majety, H. Niu, F. Herting, K. Bosslet, P. Umaña

Other (conducted preclinical studies and histologic analysis of explanted tumor tissue): W. Scheuer

All authors were employees of Roche at the time of this study. The authors thank Aurexel Ltd. (www.aurexel.com) for editorial support funded by Roche.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ashkenazi
A
,
Dixit
VM
. 
Death receptors: signaling and modulation
.
Science
1998
;
281
:
1305
8
.
2.
Locksley
RM
,
Killeen
N
,
Lenardo
MJ
. 
The TNF and TNF receptor superfamilies: integrating mammalian biology
.
Cell
2001
;
104
:
487
501
.
3.
Wilson
NS
,
Yang
A
,
Yang
B
,
Couto
S
,
Stern
H
,
Gogineni
A
, et al
Proapoptotic activation of death receptor 5 on tumor endothelial cells disrupts the vasculature and reduces tumor growth
.
Cancer Cell
2012
;
22
:
80
90
.
4.
Yang
A
,
Wilson
NS
,
Ashkenazi
A
. 
Proapoptotic DR4 and DR5 signaling in cancer cells: toward clinical translation
.
Curr Opin Cell Biol
2010
;
22
:
837
44
.
5.
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
.
6.
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
.
Ann Oncol
2012
;
23
:
2834
42
.
7.
Wiezorek
J
,
Holland
P
,
Graves
J
. 
Death receptor agonists as a targeted therapy for cancer
.
Clin Cancer Res
2010
;
16
:
1701
8
.
8.
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
.
9.
Chuntharapai
A
,
Dodge
K
,
Grimmer
K
,
Schroeder
K
,
Marsters
SA
,
Koeppen
H
, et al
Isotype-dependent inhibition of tumor growth in vivo by monoclonal antibodies to death receptor 4
.
J Immunol
2001
;
166
:
4891
8
.
10.
Motoki
K
,
Mori
E
,
Matsumoto
A
,
Thomas
M
,
Tomura
T
,
Humphreys
R
, et al
Enhanced apoptosis and tumor regression induced by a direct agonist antibody to tumor necrosis factor-related apoptosis-inducing ligand receptor 2
.
Clin Cancer Res
2005
;
11
:
3126
35
.
11.
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
.
12.
Kim
JM
,
Ashkenazi
A
. 
Fcgamma receptors enable anticancer action of proapoptotic and immune-modulatory antibodies
.
J Exp Med
2013
;
210
:
1647
51
.
13.
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
.
14.
Brennen
WN
,
Isaacs
JT
,
Denmeade
SR
. 
Rationale behind targeting fibroblast activation protein-expressing carcinoma-associated fibroblasts as a novel chemotherapeutic strategy
.
Mol Cancer Ther
2012
;
11
:
257
66
.
15.
Garin-Chesa
P
,
Old
LJ
,
Rettig
WJ
. 
Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers
.
Proc Natl Acad Sci U S A
1990
;
87
:
7235
9
.
16.
Rettig
WJ
,
Garin-Chesa
P
,
Healey
JH
,
Su
SL
,
Ozer
HL
,
Schwab
M
, et al
Regulation and heteromeric structure of the fibroblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin
.
Cancer Res
1993
;
53
:
3327
35
.
17.
Schaefer
W
,
Regula
JT
,
Bahner
M
,
Schanzer
J
,
Croasdale
R
,
Durr
H
, et al
Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies
.
Proc Natl Acad Sci U S A
2011
;
108
:
11187
92
.
18.
Hessell
AJ
,
Hangartner
L
,
Hunter
M
,
Havenith
CE
,
Beurskens
FJ
,
Bakker
JM
, et al
Fc receptor but not complement binding is important in antibody protection against HIV
.
Nature
2007
;
449
:
101
4
.
19.
Weber
TG
,
Osl
F
,
Renner
A
,
Poschinger
T
,
Galban
S
,
Rehemtulla
A
, et al
Apoptosis imaging for monitoring DR5 antibody accumulation and pharmacodynamics in brain tumors noninvasively
.
Cancer Res
2014
;
74
:
1913
23
.
20.
Weber
TG
,
Poschinger
T
,
Galban
S
,
Rehemtulla
A
,
Scheuer
W
. 
Noninvasive monitoring of pharmacodynamics and kinetics of a death receptor 5 antibody and its enhanced apoptosis induction in sequential application with doxorubicin
.
Neoplasia
2013
;
15
:
863
74
.
21.
Ridgway
JB
,
Presta
LG
,
Carter
P
. 
‘Knobs-into-holes’ engineering of antibody CH3 domains for heavy chain heterodimerization
.
Protein Eng
1996
;
9
:
617
21
.
22.
Marconi
M
,
Ascione
B
,
Ciarlo
L
,
Vona
R
,
Garofalo
T
,
Sorice
M
, et al
Constitutive localization of DR4 in lipid rafts is mandatory for TRAIL-induced apoptosis in B-cell hematologic malignancies
.
Cell Death Dis
2013
;
4
:
e863
.
23.
Xu
Y
,
Szalai
AJ
,
Zhou
T
,
Zinn
KR
,
Chaudhuri
TR
,
Li
X
, et al
Fc gamma Rs modulate cytotoxicity of anti-Fas antibodies: implications for agonistic antibody-based therapeutics
.
J Immunol
2003
;
171
:
562
8
.
24.
Li
F
,
Ravetch
JV
. 
Apoptotic and antitumor activity of death receptor antibodies require inhibitory Fcgamma receptor engagement
.
Proc Natl Acad Sci U S A
2012
;
109
:
10966
71
.
25.
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
.
26.
Doi
T
,
Murakami
H
,
Ohtsu
A
,
Fuse
N
,
Yoshino
T
,
Yamamoto
N
, et al
Phase 1 study of conatumumab, a pro-apoptotic death receptor 5 agonist antibody, in Japanese patients with advanced solid tumors
.
Cancer Chemother Pharmacol
2011
;
68
:
733
41
.
27.
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
.
28.
Wakelee
HA
,
Patnaik
A
,
Sikic
BI
,
Mita
M
,
Fox
NL
,
Miceli
R
, et al
Phase I and pharmacokinetic study of lexatumumab (HGS-ETR2) given every 2 weeks in patients with advanced solid tumors
.
Ann Oncol
2010
;
21
:
376
81
.
29.
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
.
Biomaterials
2011
;
32
:
8645
53
.
30.
Gieffers
C
,
Kluge
M
,
Merz
C
,
Sykora
J
,
Thiemann
M
,
Schaal
R
, et al
APG350 induces superior clustering of TRAIL receptors and shows therapeutic antitumor efficacy independent of cross-linking via Fcgamma receptors
.
Mol Cancer Ther
2013
;
12
:
2735
47
.
31.
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
.
32.
Huet
HA
,
Growney
JD
,
Johnson
JA
,
Li
J
,
Bilic
S
,
Ostrom
L
, et al
Multivalent nanobodies targeting death receptor 5 elicit superior tumor cell killing through efficient caspase induction
.
MAbs
2014
;
6
:
1560
70
.
33.
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
.
34.
Papadopoulos
KP
,
Isaacs
R
,
Bilic
S
,
Kentsch
K
,
Huet
HA
,
Hofmann
M
, et al
Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody(R) targeting the DR5 receptor
.
Cancer Chemother Pharmacol
2015
;
75
:
887
95
.
35.
de Bruyn
M
,
Rybczynska
AA
,
Wei
Y
,
Schwenkert
M
,
Fey
GH
,
Dierckx
RA
, et al
Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP)-targeted delivery of soluble TRAIL potently inhibits melanoma outgrowth in vitro and in vivo
.
Mol Cancer
2010
;
9
:
301
14
.
36.
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
.
37.
Siegemund
M
,
Pollak
N
,
Seifert
O
,
Wahl
K
,
Hanak
K
,
Vogel
A
, et al
Superior antitumoral activity of dimerized targeted single-chain TRAIL fusion proteins under retention of tumor selectivity
.
Cell Death Dis
2012
;
3
:
e295
.
38.
Spitzer
D
,
McDunn
JE
,
Plambeck-Suess
S
,
Goedegebuure
PS
,
Hotchkiss
RS
,
Hawkins
WG
. 
A genetically encoded multifunctional TRAIL trimer facilitates cell-specific targeting and tumor cell killing
.
Mol Cancer Ther
2010
;
9
:
2142
51
.
39.
Stuckey
DW
,
Shah
K
. 
TRAIL on trial: preclinical advances in cancer therapy
.
Trends Mol Med
2013
;
19
:
685
94
.
40.
Hewitt
RE
,
Powe
DG
,
Carter
GI
,
Turner
DR
. 
Desmoplasia and its relevance to colorectal tumour invasion
.
Int J Cancer
1993
;
53
:
62
9
.
41.
Pandol
S
,
Edderkaoui
M
,
Gukovsky
I
,
Lugea
A
,
Gukovskaya
A
. 
Desmoplasia of pancreatic ductal adenocarcinoma
.
Clin Gastroenterol Hepatol
2009
;
7
:
S44
7
.
42.
Ronnov-Jessen
L
,
Petersen
OW
,
Bissell
MJ
. 
Cellular changes involved in conversion of normal to malignant breast: importance of the stromal reaction
.
Physiol Rev
1996
;
76
:
69
125
.
43.
Tuxhorn
JA
,
Ayala
GE
,
Smith
MJ
,
Smith
VC
,
Dang
TD
,
Rowley
DR
. 
Reactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodeling
.
Clin Cancer Res
2002
;
8
:
2912
23
.
44.
Lee
JM
,
Bernstein
A
. 
Apoptosis, cancer, and the p53 tumour suppressor gene
.
Cancer Metastasis Rev
1995
;
14
:
149
61
.
45.
Hollstein
M
,
Rice
K
,
Greenblatt
MS
,
Soussi
T
,
Fuchs
R
,
Sorlie
T
, et al
Database of p53 gene somatic mutations in human tumors and cell lines
.
Nucleic Acids Res
1994
;
22
:
3551
5
.
46.
Igney
FH
,
Krammer
PH
. 
Death and anti-death: tumour resistance to apoptosis
.
Nat Rev Cancer
2002
;
2
:
277
88
.
47.
Galligan
L
,
Longley
DB
,
McEwan
M
,
Wilson
TR
,
McLaughlin
K
,
Johnston
PG
. 
Chemotherapy and TRAIL-mediated colon cancer cell death: the roles of p53, TRAIL receptors, and c-FLIP
.
Mol Cancer Ther
2005
;
4
:
2026
36
.