Purpose:

CD40 agonists hold great promise for cancer immunotherapy (CIT) as they enhance dendritic cell (DC) activation and concomitant tumor-specific T-cell priming. However, the broad expression of CD40 accounts for sink and side effects, hampering the efficacy of anti-CD40 antibodies. We hypothesized that these limitations can be overcome by selectively targeting CD40 agonism to the tumor. Therefore, we developed a bispecific FAP-CD40 antibody, which induces CD40 stimulation solely in presence of fibroblast activation protein α (FAP), a protease specifically expressed in the tumor stroma.

Experimental Design:

FAP-CD40's in vitro activity and FAP specificity were validated by antigen-presenting cell (APC) activation and T-cell priming assays. In addition, FAP-CD40 was tested in subcutaneous MC38-FAP and KPC-4662-huCEA murine tumor models.

Results:

FAP-CD40 triggered a potent, strictly FAP-dependent CD40 stimulation in vitro. In vivo, FAP-CD40 strongly enhanced T-cell inflammation and growth inhibition of KPC-4662-huCEA tumors. Unlike nontargeted CD40 agonists, FAP-CD40 mediated complete regression of MC38-FAP tumors, entailing long-term protection. A high dose of FAP-CD40 was indispensable for these effects. While nontargeted CD40 agonists induced substantial side effects, highly dosed FAP-CD40 was well tolerated. FAP-CD40 preferentially accumulated in the tumor, inducing predominantly intratumoral immune activation, whereas nontargeted CD40 agonists displayed strong systemic but limited intratumoral effects.

Conclusions:

FAP-CD40 abrogates the systemic toxicity associated with nontargeted CD40 agonists. This enables administration of high doses, essential for overcoming CD40 sink effects and inducing antitumor immunity. Consequently, FAP-targeted CD40 agonism represents a promising strategy to exploit the full potential of CD40 signaling for CIT.

Translational Relevance

Efforts to develop CD40 agonists to harness the CD40 pathway for cancer immunotherapy (CIT) have been ongoing for more than two decades. Although multiple anti-CD40 agonistic antibodies were evaluated in clinical trials, none of these molecules have been approved for cancer treatment. Previously tested CD40 agonists induced systemic CD40 activation, which led to dose-limiting side effects. In addition, CD40 agonists displayed a short serum half-life, indicative of a substantial sink effect due to the broad CD40 receptor expression. We hypothesize that fibroblast activation protein α (FAP)-targeted CD40 agonism can overcome these limitations by reducing systemic toxicity and increasing the therapeutic index. With higher therapeutic doses to mitigate CD40 sink effects, as well as targeting of FAP-CD40 via FAP to the tumor, we aim at increasing CD40 agonism specifically in the tumor. We suggest that this strategy is crucial for unleashing the full therapeutic potential of CD40 agonism and enhancing antitumor immunity in patients with cancer.

The CD40 receptor on antigen-presenting cells (APC) plays a key role in regulating immune responses and triggering the induction of adaptive immunity. Binding of its trimeric ligand CD40L, which is mainly expressed by Th cells, leads to CD40 receptor multimerization, inducing a plethora of downstream signaling pathways (1, 2). As a result, APCs become activated and mature, a process commonly referred to as APC licensing. CD40L-mediated APC activation increases their expression of costimulatory molecules as well as production of proinflammatory cytokines and enhances their capability of antigen processing and presentation (3, 4). Employing agonistic anti-CD40 antibodies to mimic this licensing of APCs for tumor antigen presentation is, therefore, an attractive strategy in the context of cancer immunotherapy (CIT). In particular, the ability of anti-CD40 molecules to activate dendritic cells (DC) and subsequently increase (cross-) priming of tumor-specific T cells (5, 6), has fueled the notion that CD40 agonists could be crucial CIT combination partners for eliciting long-lasting and sustainable antitumor responses (7–9). It is thus not surprising that a number of agonistic anti-CD40 antibodies have been developed over the last 20 years, of which several have undergone clinical evaluation in patients with cancer (10–15). Although some of the tested antibodies triggered clinical responses, major challenges of anti-CD40 agonist treatments were revealed. Across different clinical studies, dose-limiting side effects such as cytokine release syndrome (CRS) and hepatotoxicity were observed (12–15). At the same time, agonistic anti-CD40 molecules displayed a very short serum half-life, indicative of a substantial sink effect due to the broad expression of the CD40 receptor on hematopoietic and nonhematopoietic cells (8, 14). Therefore, target-mediated drug disposition by the peripheral CD40 sink, as well as dose limitations due to on-target systemic toxicity, restrict the amount of systemically administered, first-generation CD40 agonists that can be delivered to the tumor microenvironment to activate APCs locally.

In contrast, various preclinical studies employing intratumoral or peritumoral injection of CD40-activating agents have demonstrated that a stimulation of CD40 limited to the tumor area is well tolerated and induces effective antitumor responses. Despite the confinement of CD40 agonism to a primary tumor, systemic tumor-specific T-cell responses were raised, which enabled the eradication of secondary distant tumors and protected from tumor rechallenge (16, 17).

These data provide a solid rationale for targeting anti-CD40 agonism to the tumor. As intratumoral administration of anti-CD40 agonists is technically challenging and limited to few accessible tumor lesions, we have instead developed a tumor-targeted anti-CD40 agonistic antibody, the bispecific fibroblast activation protein α (FAP)-CD40 molecule, for systemic administration. Unlike nontargeted anti-CD40 antibodies, FAP-CD40 does not induce systemic CD40 stimulation. Instead, FAP-CD40 exclusively triggers APC activation in the presence of FAP, a serine protease that is highly expressed on cancer-associated fibroblasts in the tumor stroma (18). Of note, while a majority of carcinomas display high FAP levels, only low amounts of FAP are expressed in few healthy adult tissues such as the lymph nodes (LN; refs. 19, 20). The highly tumor-specific expression of FAP combined with its presence across many tumor types and recently published evidence describing a preferential localization of APC niches within the stromal area of human tumors (21), account for our choice of tumor target.

Our preclinical data demonstrate that high doses of FAP-CD40 can be administered safely to overcome sink effects, allowing accumulation of our molecule in the tumor. We show that FAP-CD40–mediated DC activation is limited to the tumor and the LNs. This locally restricted CD40 agonism can induce potent immune responses resulting in tumor eradication and durable immunologic memory.

Molecules and reagents

Recombinant biotinylated murine FAP, the human CD40 agonists FAP-huCD40 and its corresponding nontargeted control DP47-huCD40, SGN40 huIgG1, SGN40 moIgG1, selicrelumab (22), DP47, the murine surrogate molecule FAP-moCD40, as well as FGK4.5 moIgG1 were produced at Roche Innovation Center Zurich (RICZ) and Munich (RICM).

The human FAP-targeted CD40 agonist FAP-huCD40 consists of a huIgG1 containing two CD40 binding Fv regions (0817 domain, recognizing human and cynomolgus CD40). Mutations (Pro329Gly, Leu234Ala, and Leu235Ala; PGLALA mutation; ref. 23) to prevent fragment crystallizable γ receptor (FcγR) binding were introduced within the Fc region of the antibody. To ensure correct assembly of light and heavy chains, the knob-into-hole strategy (24) as well as charged mutations in the CH1 domains (EE) and CL domains (RK) were employed. For monovalent FAP binding, one anti-FAP Fab fragment (recognizing human, cynomolgus, and murine FAP) was fused via its VL-CH1 chain to the huIgG1 Fc-knob chain by a 4G4S linker. To prevent mispairings, the light- and heavy-chain variable regions of the anti-FAP Fab were exchanged (25). For the nontargeted DP47-huCD40 molecule, the anti-FAP Fab domain of FAP-huCD40 was replaced by the nonbinding DP47 Fab (described previously in ref. 26). For the conventional human CD40 agonist control molecules, the VH and VL sequences of SGN40 (27) were inserted in an Fc-competent huIgG1 (SGN40 huIgG1) or moIgG1 (SGN40 moIgG1) framework. The control antibody DP47 is based on a PGLALA-mutated huIgG1 with two nonbinding DP47 Fv domains. For the murine surrogate molecule FAP-moCD40, the anti-human CD40 Fv domains of the FAP-huCD40 molecule were replaced by the VH and VL regions of the murine anti-CD40 agonist FGK4.5 (28). FGK4.5 moIgG1 was created by inserting the VH and VL regions of original FGK4.5 molecule, which is a rat IgG2a antibody, into an Fc-functional moIgG1 backbone (see Fig. 1A; Supplementary Fig. S1C for a schematic overview of all anti-CD40 antibody constructs used).

Figure 1.

Molecular properties of the bispecific FAP-targeted anti-CD40 agonist FAP-huCD40. A, Schematic representation of FAP-targeted and nontargeted human CD40 agonists used. FAP-huCD40 consists of a human immunoglobulin G1 (huIgG1) with two N-terminal human CD40-binding moieties (0817 domains), an Fc part with Fc-silencing PGLALA mutations and knob-into-hole (kih) technology promoting heterodimerization of heavy chains. One FAP binding moiety as crossover Fab fragment (exchange of variable regions of Fab heavy and light chain for correct molecular assembly) is fused via the VL-CH1 chain at the C-terminus of the Fc knob chain. The structure of DP47-huCD40 is equivalent to FAP-huCD40 but the anti-FAP domain is replaced by the nonbinding anti-DP47 domain. The Fc-functional SGN40 molecules contain two anti-human CD40-binding sites (SGN40 domains) inserted into a huIgG1 or murine IgG1 (moIgG1) backbone. B–D, Binding of the human CD40 agonists FAP-huCD40 or its nontargeted control DP47-huCD40, SGN40 huIgG1 or nonbinding control antibody DP47 to human CD40 on splenic B cells from huCD40tg mice (B), to murine FAP recombinantly expressed on MC38 cells (C), or to non-FAP expressing parental MC38 cells (D). B–D, Baseline correction was performed by subtracting mean fluorescence intensity (MFI) values of secondary antibody only conditions. Shown is the mean (±SD) of technical duplicates. For the curve fit nonlinear least squares regression using a variable slope model was applied. Results are representative for at least two independent experiments.

Figure 1.

Molecular properties of the bispecific FAP-targeted anti-CD40 agonist FAP-huCD40. A, Schematic representation of FAP-targeted and nontargeted human CD40 agonists used. FAP-huCD40 consists of a human immunoglobulin G1 (huIgG1) with two N-terminal human CD40-binding moieties (0817 domains), an Fc part with Fc-silencing PGLALA mutations and knob-into-hole (kih) technology promoting heterodimerization of heavy chains. One FAP binding moiety as crossover Fab fragment (exchange of variable regions of Fab heavy and light chain for correct molecular assembly) is fused via the VL-CH1 chain at the C-terminus of the Fc knob chain. The structure of DP47-huCD40 is equivalent to FAP-huCD40 but the anti-FAP domain is replaced by the nonbinding anti-DP47 domain. The Fc-functional SGN40 molecules contain two anti-human CD40-binding sites (SGN40 domains) inserted into a huIgG1 or murine IgG1 (moIgG1) backbone. B–D, Binding of the human CD40 agonists FAP-huCD40 or its nontargeted control DP47-huCD40, SGN40 huIgG1 or nonbinding control antibody DP47 to human CD40 on splenic B cells from huCD40tg mice (B), to murine FAP recombinantly expressed on MC38 cells (C), or to non-FAP expressing parental MC38 cells (D). B–D, Baseline correction was performed by subtracting mean fluorescence intensity (MFI) values of secondary antibody only conditions. Shown is the mean (±SD) of technical duplicates. For the curve fit nonlinear least squares regression using a variable slope model was applied. Results are representative for at least two independent experiments.

Close modal

Antibodies were expressed by transient transfection of human embryonic kidney (HEK 293) cells. Antibodies were purified from cell culture supernatants by affinity chromatography using MabSelectSure-Sepharose (GE Healthcare) chromatography. Purified antibodies were characterized by capillary electrophoresis SDS, size-exclusion chromatography, mass spectrometry, and endotoxin determination.

Cell lines

Cells were maintained under sterile conditions at 37°C in a humidified incubator (5% CO2) and passaged regularly upon reaching 80% confluency. MC38-huCEA cells were generated by City of Hope (29) and engineered to express mRFP and ovalbumin (OVA) at RICZ. The plasmid encoding the OVA transgene was kindly provided by ProQinase. MC38-huCEA-mRFP-OVA cells were kept under selection with 1.5 μg/mL puromyin (Invivogen, ant-pr), 100 μg/mL hygromycin (Invivogen, ant-hg), and 300 μg/mL geneticin (Gibco, 10131035).

MC38 cells were obtained from City of Hope and transfected with murine Fap at RICZ. MC38-FAP cells were selected with 6 μg/mL puromycin (Invivogen, ant-pr). To establish a cell line with robust in vivo growth characteristics, MC38-FAP cells were subcutaneously injected in C57BL/6J mice. Tumors that showed good growth were resected, dissociated, and tumor cells were grown and expanded in vitro under puromycin selection for cell line establishment. KPC-4662 cells were received from University of Pennsylvania (Philadelphia, PA; ref. 30) and engineered to express huCEA at RICZ. KPC-4662-huCEA cells were selected for huCEA expression with 500 μg/mL hygromycin. All cell lines used were routinely checked for Mycoplasma using the MycoAlert Mycoplasma Detection Kit (Lonza, LT07–118). Cells were used for experiments one to three weeks after thawing.

Mice

Female mice were used for all studies. Mice were maintained under specific-pathogen-free condition with daily cycles of 12 hours light and 12 hours darkness according to committed guidelines (GV-Solas; Felasa; TierschG). Experimental study protocols were reviewed and approved by the Cantonal Veterinary Office in Zürich (license ZH223/2017, ZH224/2017, and ZH225/2017) and by the Institutional Animal Care and Use Committee of RICZ. After arrival, animals were maintained for one week to get accustomed to the new environment and for observation. They were afterwards implanted with a subcutaneous transponder for identification and maintained one more week for recovery. Continuous health monitoring was carried out on a regular basis. Mice were between 6 to 9 weeks of age at the start of the experiments. In case of tumor-bearing mice, tumors were measured at least two to three times per week. Mice were randomized into different treatment groups according to tumor volume. OT-1 [C57BL/6-Tg(TcraTcrb)1100Mjb/Crl] and C57BL/6J mice were acquired from Charles River France. HuCD40tg mice (background C57BL/6NT) expressing both the human and murine CD40 receptor, were generated by Taconic by oocyte pronuclear injection of a bacterial artificial chromosome construct encoding the human CD40 receptor under control of the human CD40 promoter. The huCD40tg mice were provided by the Baylor research institute. Fap−/− mice were kindly provided by Dr. Junichiro Sonoda (Genentech; ref. 31). HuCEAtg mice [C57BL/6JTgN(CEAGe) 18FJP] were obtained under license agreement from Beckman Research Institute of City of Hope (32). HuCEAtg, huCD40tg and Fap−/− mice were bred by Charles River.

Cynomolgus monkey study

The cynomolgus monkey study was ethically reviewed and approved by the Animal Welfare and Ethics Committee of Charles River Laboratories Edinburgh. The project license application was granted by the British Home Office. Further study details can be found within the Supplementary Material and Methods section.

Flow cytometry

For flow cytometry staining, cells were incubated with 3 μg/mL of Fc receptor blocking mouse IgG isotype control (Thermo Fisher Scientific, 10400C) for 10 minutes at 4°C. Cells were subsequently stained with a mixture of fluorescently labeled antibodies and fixable viability dye (Invitrogen) in Brilliant Stain buffer (BD, 563794; see Supplementary Table S1 for a list of antibodies used) for 30 minutes at 4°C. Afterwards, cells were washed twice with PBS. For intracellular stainings, extracellularly labeled cells were incubated with fixation/permeabilization solution (eBioscience, 00–5523–00) for 30 minutes at 4°C, washed twice with permeabilization buffer, and then stained for 30 minutes at room temperature with intracellular staining antibodies diluted in permeabilization buffer. Cells were washed two times with permeabilization buffer and analyzed the same day using a 5-laser LSR-Fortessa (BD Bioscience with DIVA software). Data analysis was performed using the FlowJo version 10 software (FlowJo, LLC).

Isolation of B cells, DCs, and CD8+ T cells from murine spleens for in vitro assays

To isolate splenic immune cells, murine spleens were cut into small pieces and digested with 0.1 mg/mL collagenase D (Sigma-Aldrich, 11088866001) and 0.05 mg/mL DNase (Sigma-Aldrich, D5025–150KU) for 10 minutes at 37°C. EDTA (Applichem, A4892.1000) was added at a concentration of 0.01 mol/L, followed by a second incubation step at 37°C for 5 minutes. The splenocytes solution was smashed through a 70-μm cell strainer (Corning, 431751). Red blood cells were lysed with cell lysis buffer (BD, 555899). Mouse CD11c UltraPure microbeads (Miltenyi Biotec, 130–108–338), mouse CD8a+ T Cell Isolation Kit (Miltenyi Biotec, 130–104–075) or the Mouse B Cell Isolation Kit (Miltenyi Biotec, 130–090–862) were used according to the manufacturer's instructions to isolate different immune cell populations from the splenocytes suspension. For subsequent in vitro assays with splenic immune cells, cells were resuspended in R10 (RPMI1640; Gibco, 31870–025) supplied with 10% FBS (Gibco, 16140), 1% penicillin–streptomycin (P/S; Gibco, 11548876), 1% l-glutamine (Gibco, 25030–024), 1% sodium-pyruvate (Gibco, 11360–039), 1% nonessential amino acids (Gibco, 11140–035), and 50 μmol/L β-Mercaptoethanol (Gibco, 31350–010).

Binding assays

For binding assays, MC38-FAP, MC38, or B cells from huCD40tg mice were resuspended in flow cytometry staining buffer (eBioscience, 00–4222–57) containing the different antibodies at the indicated range of concentrations and incubated for 120 minutes at 4°C. Afterwards, the cells were washed three times. Cells were further stained with secondary antibody solution containing R-Phycoerythrin (PE) conjugated AffiniPure F(ab')2 Fragment Goat Anti-Human IgG, Fcγ Fragment Specific (1:50 dilution; Jackson ImmunoResearch, 109–116–098) and incubated for 60 minutes at 4°C. Cells were washed twice and resuspended in flow cytometry staining buffer containing 0.2 μg/mL 4′,6-diamidino-2-phenylindole (Roche, 10236276001) and acquired the same day.

In vitro APC activation assays

1.0 × 105 splenic B cells or 0.25 × 105 splenic DCs from huCD40tg mice were seeded per well. Streptavidin Dynabeads (Thermo Fisher Scientific, 11205D) were coated with biotinylated mouse FAP according to the manufacturer's instructions and added to the APCs in a bead:cell ratio of 2:1. As control, noncoated beads were used. For APC activation with cellular FAP, MC38-FAP or parental MC38 cells were irradiated at 50 Gy using a RS 2000 irradiator (Rad Source Technologies) and 0.25 × 105 MC38 or MC38-FAP cells were seeded per well. 0.25 × 105 splenic DCs or 0.5 × 105 splenic B cells from huCD40tg mice were added. SGN40 moIgG1 was crosslinked by preincubation for 30 minutes at room temperature with anti-mouse Fc F(ab')2 (in-house endotoxin-purified, obtained from Jackson, 115–006–071) in a 1:1 ratio. CD40 agonistic antibodies were added at the indicated concentrations. DC activation was measured by flow cytometry after 24 hours, while B-cell activation was assessed after 48 hours.

For APC activation assays with nodal FRCs, FRCs were isolated from inguinal, axial, and brachial LNs of Fap−/− or C57BL/6J mice and in vitro expanded as described by Fletcher and colleagues (33). 0.14 × 105 FRCs per well were seeded in DMEM + 10% FBS + 1% P/S. 0.3 × 105 splenic B cells or 0.25 × 105 splenic DCs from C57BL/6J mice were added one day later to the FRCs. FAP-moCD40 was added at the indicated concentrations. DC activation was measured after one day, while B-cell activation was measured after 3 days by flow cytometry.

Cross-presentation assays

DCs were loaded with OVA protein via the DEC-205 receptor using the OVA Antigen Delivery Reagent (Miltenyi Biotec, 130–094–663) in combination with a biotinylated anti-mouse DEC-205 antibody (Miltenyi Biotec, 130–101–854) according to the manufacturer's protocol. 0.25 × 105 DCs from huCD40tg mice were seeded per well. Murine FAP-coated or noncoated Dynabeads were added to the DCs at a 2:1 bead:cell ratio. Next, different agonistic anti-CD40 antibodies were added at the indicated concentrations. Twenty-four hours later, CD8+ T cells were isolated from the spleens of OT-1 mice added to the OVA-loaded DCs at a DC:T-cell ratio of 1:2. After three days, T cells were either used for flow cytometry analysis of T-cell numbers and activation or T cells were used for coculture with OVA+ target cells. For the killing assay, 5 × 103 MC38-huCEA-mRFP-OVA cells were seeded in R10 and allowed to adhere before DC-primed T cells from the 5 nmol/L FAP-huCD40 plus FAP-beads conditions were transferred. The number of transferred T cells was determined to be 1 × 104 based on flow cytometry analysis. As control, 2.5 × 104 freshly isolated OT-1 T cells were added to the OVA-expressing tumor cells. MC38-huCEA-mRFP-OVA cell killing was measured by determining the red target cell area over time by Incucyte (Sartorius) analysis.

OVA vaccination study

Fap−/− mice were injected subcutaneously in the flank with 12.5 mg/kg OVA alone or in combination with either 13.3 mg/kg FAP-moCD40 or 10 mg/kg FGK4.5 moIgG1. C57BL/6J wild-type mice were injected with the combination of OVA and FAP-moCD40. Animals were sacrificed on day 3 or day 8 after vaccination and spleens as well as inguinal LNs were harvested in PBS.

In vivo immuno-pharmacodynamics and efficacy studies

HuCD40tg mice were injected subcutaneously in the flank with 2 × 106 MC38-FAP cells in RPMI with 50% Matrigel (Corning, 354234). When tumors reached a mean size of 220 to 275 mm3, anti-CD40 agonists FAP-huCD40, SGN40 moIgG1 or selicrelumab were injected intraperitoneally at the indicated concentrations. Mice of the vehicle group received equal volumes of histidine buffer intraperitoneally. Animals were sacrificed at the indicated time points and liver, spleen, inguinal LNs as well as subcutaneous tumor were either harvested in PBS, paraformaldehyde (PFA), or 10% buffered formalin solution or were immediately frozen in liquid nitrogen. Blood was collected in serum tubes (BD, 365967), spun down at 15,000 × g for 10 minutes at 4°C to collect the serum, and serum samples were frozen at −20°C until analyzed.

For the tumor rechallenge experiment, tumor-free mice treated with FAP-huCD40 (9 out of 9 mice) were injected subcutaneously with 2 × 106 MC38-FAP cells into the flank contralateral to the primary injection site 93 days after the first tumor cell injection. Naïve huCD40tg mice were injected simultaneously with tumor cells as control. Animals were sacrificed 54 days after the rechallenge when tumors in the vehicle group reached a mean size of 1,800 mm3.

For the in vivo studies using the KPC-4662-huCEA tumor model, huCEAtg mice received subcutaneous 0.3 × 105 KPC-4662-huCEA pancreatic cells in RPMI with 50% Matrigel into the flank. Randomized mice (average tumor size of around 240 mm3/group) were treated intraperitoneally with vehicle or 13.3 mg/kg moFAP-CD40. Some animals were sacrificed eight days after therapy injection and tumors were resected for immunofluorescence staining analysis.

In vivo drug targeting and tissue biodistribution

HuCD40tg mice were injected subcutaneously in the flank with 2 × 106 MC38-FAP cells in RPMI with 50% Matrigel. When the tumors were between a size of 250 to 350 mm3, mice were intravenously injected with the indicated concentrations of AF647 labeled FAP-huCD40 and selicrelumab or histidine buffer (vehicle mice).

Mice were anesthetized at the indicated time points with 2% isoflurane and the blood collected retro-orbitally in heparin-coated tubes. The animals were sacrificed and organs were extracted, weighed, and placed into 4% PFA. Organs were washed in PBS and placed in the IVIS Spectrum in vivo imaging system (PerkinElmer) to acquire fluorescent images. Fifty microliters of blood was put in an Eppendorf tube for FLI imaging. In vivo imaging software (PerkinElmer) was used for imaging data analysis. Signal intensities for each group of organs were normalized together. A region of interest was drawn to capture the area around the signal. Total flux was captured as the number of photons per second and normalized against the organ weight (total flux at time point t/organ weight = drug concentration at time point t = Ct). No normalization to weight was done for the blood as the same volumes were used for all samples and for the LNs due to their very low weight. The background for each organ (C0) was determined by measuring the total flux normalized to organ weight of the respective organ from vehicle animals at the 2-hour time point. To determine the normalized drug concentrations at each given time point, Ct values were divided by the corresponding C0 values.

To analyze the binding of labeled anti-CD40 agonists on different cell subsets within different organs, mice treated with AF647-labeled molecules were sacrificed 24 hours after injection of the antibodies. Spleen, inguinal LNs, and tumors were harvested in PBS, processed as described below, and analyzed by flow cytometry.

Processing of organs from in vivo studies for immuno-pharmacodynamics analysis

IHC staining of MC38-FAP tumors for murine FAP and CD3 was done as described previously (26).

For flow cytometry analysis spleens, tumors and LNs were cut into small pieces and digested with 0.05 mg/mL DNase and 0.1 mg/mL collagenase D (spleens and LNs) or 1 mg/mL collagenase D (tumors) for 10 minutes (spleens and LNs) or 25 minutes (tumors) at 37°C. EDTA (0.01 mol/L) was added for 5 more minutes of incubation to spleens and LNs. Cell suspensions were smashed through a 70-μm cell strainer. Red blood cell lysis was performed for spleen samples. Cells were subsequently stained for flow cytometry analysis.

Determination of serum and tumor cytokine levels by multiplexed cytokine analysis

Snap frozen tumor pieces were lysed with the Bio-Plex Cell Lysis Kit (Bio-Rad) using Lysing Matrix S tubes (MP Biomedicals) and the Precellys Evolution tissue homogenizer (Bertin Technologies) according to the manufacturers' instructions. Protein content of tumor lysates was determined with the Pierce bicinchoninic acid assay kit. Forty micrograms of tumor protein was used of each sample for multiplexed cytokine analysis. The Bio-Plex Pro Mouse Cytokine 23-plex Assay Kit (Bio-Rad, M60009RDPD) or the Bio-Plex Pro Mouse Chemokine Panel 31-Plex Assay kit (Bio-Rad, 12009159) and the FLEXMAP 3D platform (LUMINEX) were used according to the manufacturers' instructions for analysis of the concentration of cytokines in serum and tumor.

Liver toxicity assessment

For histopathologic assessment, formalin fixed liver samples were trimmed and processed for paraffin embedding. Four-micron–thick slices were stained with hematoxylin and eosin. Stained slides were used for histopathologic evaluation by a board-certified pathologist. Serum samples collected at necropsy were used for analysis of liver enzymes with ADVIA 1800 chemistry system (Siemens) according to the manufacturer's instructions.

Immunofluorescence staining of KPC-4662-huCEA tumors

Tumors were bisected and fixed in Cytofix solution (BD Biosciences, 554655) for 21 hours at 4°C. Half tumors were embedded in 4% low gelling temperature Agarose (Sigma, A0169) and sectioned into 70-μm–thick sections using a Vibratome (Leica 1200s) and common razorblades. Tumor sections were blocked and permeabilized at the same time using home-made “BlockPerm” buffer [Tris 0.1 mol/L (VWR, 89500–584) + 0.3% Triton X (Sigma, T8787) + 1% mouse serum (Jackson, 015–00–120) + 1% BSA (Sigma, A9576)] for 15 minutes. Fluorescently labeled antibodies were diluted in BlockPerm and added to the tumor sections for 15 hours at room temperature. The following antibodies were used: CD8 BV421 (clone 53–6.7, BioLegend, 100738), CD4 BV570 (clone RM4–5, BioLegend, 100542), FoxP3 (clone D6O8R, Cell Signaling Technologies, 12653) conjugated in-house to CF633 via a kit from Biotium (92257), E-cadherin (polyclonal, R&D Systems, AF648) conjugated in-house to CF555 via a kit from Biotium (92254), and FAP (clone 28H1, produced in-house) conjugated in-house to CF647 via a kit from Biotium (92259).

Sections were then washed with Tris 0.1 mol/L two times for 20 minutes and subsequently mounted in Fluoromount G (Thermo Fisher Scientific, 00–4985–02) using #1 coverslips (ROTH). Sections were imaged on a Leica SP8 inverted confocal microscope using a 40× oil immersion objective at 512 by 512 pixels and 1-μm thick virtual z-sections. Individual tiles were stitched together in the microscope software (LAS X, Leica Microsystems) and further processed using Imaris 9.6 (Bitplane). Briefly, in Imaris CD8 and CD4 channels were segmented with the following parameters (Smoothing: 1.52, Background subtraction: Default, Cell splitting: Yes; Quality for splitting: Default, Filter out small objects: below 250 Voxels) and statistics were exported to .csv files. FoxP3 intensity within CD4 segmented objects, as well as positional information (XY coordinates) of CD4 and CD8 objects were assessed in Matlab 2020a (Mathworks) and resulting images and data were exported for either direct use (images of segmented object localization) or subsequent plotting and statistical analysis.

Statistical analysis

Statistical significance was evaluated using the GraphPad Prism Version 8.4.2 (GraphPad Software) with the statistical test indicated. P values of or below 0.05 were considered statistically significant. Significance levels are indicated as: ns, not significant = P > 0.05; *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P ≤ 0.0001.

Illustrations

Illustrations in Fig. 4B and Supplementary Fig. S1B were created with BioRender.com.

Molecular properties and design of the FAP-targeted anti-CD40 agonistic antibody

Our goal was to generate an anti-CD40 agonist that induces a potent CD40 stimulation selectively in the tumor area but not in the periphery. To achieve this, we created a bispecific FAP-huCD40 antibody containing two CD40-binding domains (0817 domain, recognizing human and cynomolgus CD40) and one C-terminally fused FAP binding site (recognizing human, cynomolgus, and murine FAP; Fig. 1A). For both, the anti-FAP and the anti-CD40 moieties, high-affinity binders were selected (Supplementary Fig. S1A). The activation of CD40 by bivalent agonistic anti-CD40 antibodies commonly depends on FcγR crosslinking (17, 34, 35). To prevent FAP-huCD40–mediated CD40 activation by FcγRs in the periphery, mutations in the FAP-huCD40 antibody Fc region were introduced, that abrogate FcγR binding (PGLALA mutations). In addition, the PGLALA mutations ensure that cells bound by FAP-huCD40 are not depleted due to antibody-dependent cellular or complement-dependent cytotoxicity (23).

By introducing the functionally “silent” Fc region in combination with a C-terminal anti-FAP domain into our CD40 agonist, we aimed at designing a molecule that can solely be crosslinked and activated through FAP binding. We envisioned FAP+ cells to function as scaffolds that allow clustering of FAP-huCD40 antibodies, leading to CD40 receptor multimerization and thus to a FAP-restricted induction of CD40 downstream signaling (Supplementary Fig. S1B).

We first tested FAP-huCD40 binding to both its targets CD40 and FAP. To evaluate CD40 binding, we used splenic B cells of mice transgenic for the human CD40 receptor (huCD40tg mice), as the anti-human CD40 moiety of the bispecific molecule is not cross-reactive to murine CD40. The human CD40 binding properties of FAP-huCD40 were comparable with those of the nontargeted human CD40 agonist SGN40 (Fig. 1A and B). Therefore, we considered SGN40 to be a suitable FAP-independent anti-CD40 agonist for comparison in our subsequent assays.

When evaluating FAP binding, we found that FAP-huCD40 but not the corresponding control molecule DP47-huCD40 (DP47 is a non-binding germline control; Fig. 1A) bound to murine FAP recombinantly expressed on MC38 murine colon adenocarcinoma cells (MC38-FAP). In accordance with these observations, no binding of FAP-huCD40 to non-FAP–expressing MC38 cells was detected (Fig. 1C and D).

FAP-CD40 induces FAP-specific APC activation

Next, we assessed FAP-huCD40's potential to activate CD40-expressing APCs in vitro. Splenic DCs or B cells from huCD40tg mice were coincubated with FAP-huCD40 and either FAP-coated or noncoated polystyrene beads. Beads instead of cells were chosen as source of FAP to avoid potential cell-derived APC-activating factors that could affect the potency of our antibody in vitro.

With FAP-huCD40 in combination with FAP-coated beads, DCs and B cells exhibited a strong dose-dependent increase of the activation markers CD70, CD80, CD86, and murine CD40 (huCD40tg mice maintain murine CD40 expression; human CD40 could not be measured by flow cytometry, as FAP-huCD40 competed with labeled anti-human CD40 antibodies). In contrast, when using noncoated beads, FAP-huCD40 did not induce any APC-activating effects. No stimulation of DCs or B cells was observed with DP47-huCD40 in either the presence or absence of FAP (Fig. 2A and B; Supplementary Fig. S2A and S2B).

Figure 2.

FAP-huCD40 induces FAP-specific APC activation and T-cell priming. A and B, Splenic DCs or B cells from huCD40tg mice were coincubated with the human CD40 agonists FAP-huCD40, SGN40 moIgG1, crosslinked SGN40 moIgG1 (SGN40 moIgG1 XL), or nontargeted DP47-huCD40 together with either FAP-coated or noncoated beads. Antibodies were titrated from 0.6 pmol/L to 10 nmol/L (4x dilution). A, HuCD40tg DC activation after 24 hours in the presence (left) or absence (right) of FAP; MFI of the representative activation marker CD70 is shown. B, HuCD40tg B-cell activation after 48 hours in the presence (left) or absence (right) of FAP; MFI of the representative activation marker CD80 is shown. C and D, OVA-pulsed huCD40tg DCs were incubated with 5 pmol/L to 5 nmol/L (10x dilution) of FAP-huCD40, SGN40 XL, or DP47-huCD40 in combination with FAP-coated or noncoated beads. Subsequently OT-1 T cells were added to the DC cultures. C, OT-1 T-cell counts and programmed cell death protein 1 (PD-1; MFI) expression after 72 hours of coculture with OVA-loaded huCD40tg DCs in the presence of different anti-CD40 agonists with (left) or without (right) FAP. D, OT-1 T cells from the 5 nmol/L FAP-huCD40 plus FAP-beads conditions were added to OVA and monomeric red fluorescent protein (mRFP) expressing MC38 cells. Freshly isolated OT-1 T cells served as a control. MC38-mRFP-OVA killing was measured by Incucyte analysis (target cell area, left graph). Significance at 150 hours was calculated using unpaired two-tailed t test. On the right, two representative images show MC38-mRFP-OVA growth after 150 hours in the different conditions. A–D, Mean and SD of technical duplicates are depicted. The results in A–C were confirmed in at least three independent experiments. The killing assay in D was performed twice.

Figure 2.

FAP-huCD40 induces FAP-specific APC activation and T-cell priming. A and B, Splenic DCs or B cells from huCD40tg mice were coincubated with the human CD40 agonists FAP-huCD40, SGN40 moIgG1, crosslinked SGN40 moIgG1 (SGN40 moIgG1 XL), or nontargeted DP47-huCD40 together with either FAP-coated or noncoated beads. Antibodies were titrated from 0.6 pmol/L to 10 nmol/L (4x dilution). A, HuCD40tg DC activation after 24 hours in the presence (left) or absence (right) of FAP; MFI of the representative activation marker CD70 is shown. B, HuCD40tg B-cell activation after 48 hours in the presence (left) or absence (right) of FAP; MFI of the representative activation marker CD80 is shown. C and D, OVA-pulsed huCD40tg DCs were incubated with 5 pmol/L to 5 nmol/L (10x dilution) of FAP-huCD40, SGN40 XL, or DP47-huCD40 in combination with FAP-coated or noncoated beads. Subsequently OT-1 T cells were added to the DC cultures. C, OT-1 T-cell counts and programmed cell death protein 1 (PD-1; MFI) expression after 72 hours of coculture with OVA-loaded huCD40tg DCs in the presence of different anti-CD40 agonists with (left) or without (right) FAP. D, OT-1 T cells from the 5 nmol/L FAP-huCD40 plus FAP-beads conditions were added to OVA and monomeric red fluorescent protein (mRFP) expressing MC38 cells. Freshly isolated OT-1 T cells served as a control. MC38-mRFP-OVA killing was measured by Incucyte analysis (target cell area, left graph). Significance at 150 hours was calculated using unpaired two-tailed t test. On the right, two representative images show MC38-mRFP-OVA growth after 150 hours in the different conditions. A–D, Mean and SD of technical duplicates are depicted. The results in A–C were confirmed in at least three independent experiments. The killing assay in D was performed twice.

Close modal

To compare FAP-huCD40's effects to those of a nontargeted human anti-CD40 agonist, we used the FcγR crosslinking-dependent antibody SGN40. To ensure maximum functionality of the SGN40 molecule in the huCD40tg mouse model, in which human CD40 receptors but murine FcγRs are expressed, the variable regions of the original SGN40 huIgG1 molecule were inserted into an Fc-competent murine IgG1 framework (SGN40 moIgG1; Fig. 1A). The murine IgG1 was selected, as murine CD40 agonists of this subclass are known to be most efficiently activated by FcγR crosslinking (35). SGN40 moIgG1 induced a moderate B-cell activation, while DC activation markers increased only slightly. Although DCs and B cells express FcγRs, this expression might not be sufficient to induce effective hyperclustering of anti-CD40 agonists. We therefore enhanced crosslinking of SGN40 moIgG1 by pre-incubating it with an anti-Fc F(ab')2 (SGN40 moIgG1 XL), resulting in a more pronounced DC as well as B-cell activation. As expected, in contrast to FAP-huCD40, SGN40's activity was independent of the presence of FAP (Fig. 2A and B; Supplementary Fig. S2A and S2B). To confirm FAP-CD40's FAP specificity and to ensure that cellular FAP triggers similar effects as FAP coated to beads, we further tested FAP-huCD40 in presence of MC38-FAP versus MC38 cells. FAP-huCD40 induced APC activation in cocultures of DCs or B cells with MC38-FAP but not MC38 cells (Supplementary Fig. S2C and S2D). Together, our data highlight that FAP-huCD40 mediates a potent and exclusively FAP-dependent activation of DCs and B cells.

FAP-CD40–mediated DC activation leads to enhanced T-cell priming

We next asked whether the observed FAP-huCD40–induced DC activation results in an enhanced capability of DCs to cross-prime CD8+ T cells. To answer this question, OVA was delivered via the antigen uptake receptor DEC-205 to splenic DCs isolated from huCD40tg mice. Targeting of antigen to the endocytic DEC-205 receptor facilitates efficient MHC class I antigen presentation (36). The OVA-loaded DCs were incubated with FAP-huCD40 in the presence of FAP-coated or noncoated beads. Subsequently OT-1 T cells (specific for the OVA-derived SIINFEKL peptide in the context of H-2kb; ref. 37) were added to the DC cultures. OVA-pulsed DCs incubated with FAP-huCD40 in the presence of FAP induced a potent T-cell proliferation and activation. This effect was entirely FAP dependent, as no increase in T-cell numbers or activation markers were observed in conditions with FAP-huCD40 in combination with noncoated beads. Consistent with our previous results and irrespective of the presence of FAP, SGN40 moIgG1 XL induced activation and proliferation of OT-1 T cells, while DP47-huCD40 displayed no signs of activity (Fig. 2C). Of note, activated OT-1 T cells derived from the FAP-huCD40 plus FAP-beads conditions exceeded fresh OT-1 T cells in their capability of killing OVA-expressing MC38 cells (Fig. 2D). These data demonstrate that DC activation by FAP-CD40 in a FAP-specific manner can promote effective T-cell cross-priming.

FAP expressed on murine nodal fibroblastic reticular cells is sufficient to trigger crosslinking of FAP-CD40 and subsequent APC activation

We demonstrated FAP-CD40's effects upon crosslinking by FAP coated on beads or recombinantly expressed by MC38 cells. Because in both cases abundant amounts of FAP are provided, we wanted to investigate whether FAP-CD40 crosslinking and activity can as well be observed in the presence of limited FAP levels. This question seemed in particular relevant as low amounts of FAP are expressed by fibroblastic reticular cells (FRC), a population of myofibroblasts, in the LNs (20).

To validate whether FRCs might trigger FAP-CD40 crosslinking, we isolated them from murine LNs followed by in vitro expansion. Subsequently, these FRCs were coincubated with murine B cells or DCs and the murine surrogate molecule FAP-moCD40, which has equivalent molecular properties as FAP-huCD40, but binds the murine CD40 receptor (Supplementary Fig. S1C and S2E). FAP-moCD40 significantly enhanced the expression of different APC activation markers when added to APC/FRC cocultures. No effect of FAP-moCD40 was observed with in vitro expanded FRCs from FAP knockout (Fap−/−) mice (Fig. 3A; Supplementary Fig. S3A). This indicates that the nodal FRC FAP expression, although being more than 50-fold lower than that of MC38-FAP cells (Supplementary Fig. S3B), is sufficient to induce hyperclustering of FAP-CD40 leading to APC activation.

Figure 3.

FAP expressed by LN FRCs triggers FAP-moCD40 activity. A, Nodal FRCs from wild-type (WT) or Fap−/− mice were coincubated with splenic murine B cells and 6.7 nmol/L of FAP-moCD40. Shown is the activation of B cells based on CD70, CD80, and CD86 upregulation after 48 hours. Mean and SD MFI values of three technical replicates are depicted. To confirm these results, three independent experiments were performed. B and C,Fap−/− mice were vaccinated with 12.5 mg/kg OVA alone or in combination with either 13.3 mg/kg FAP-moCD40 or 10 mg/kg murine CD40 agonist (FGK4.5 moIgG1; matched doses based on molar concentrations). In addition, wild-type mice were injected with the combination of OVA and FAP-moCD40. B, Murine DC activation after three days represented by DC CD86 MFI in spleen and draining as well as nondraining LNs. C, CD8+ T-cell proliferation measured by Ki-67 staining 8 days postinjection in spleen, draining and nondraining LN. Depicted are individual values of 3–5 mice, mean and SD. The data was confirmed in two independent experiments. For graphs A–C, significance was calculated by one-way ANOVA using Tukey multiple comparisons test.

Figure 3.

FAP expressed by LN FRCs triggers FAP-moCD40 activity. A, Nodal FRCs from wild-type (WT) or Fap−/− mice were coincubated with splenic murine B cells and 6.7 nmol/L of FAP-moCD40. Shown is the activation of B cells based on CD70, CD80, and CD86 upregulation after 48 hours. Mean and SD MFI values of three technical replicates are depicted. To confirm these results, three independent experiments were performed. B and C,Fap−/− mice were vaccinated with 12.5 mg/kg OVA alone or in combination with either 13.3 mg/kg FAP-moCD40 or 10 mg/kg murine CD40 agonist (FGK4.5 moIgG1; matched doses based on molar concentrations). In addition, wild-type mice were injected with the combination of OVA and FAP-moCD40. B, Murine DC activation after three days represented by DC CD86 MFI in spleen and draining as well as nondraining LNs. C, CD8+ T-cell proliferation measured by Ki-67 staining 8 days postinjection in spleen, draining and nondraining LN. Depicted are individual values of 3–5 mice, mean and SD. The data was confirmed in two independent experiments. For graphs A–C, significance was calculated by one-way ANOVA using Tukey multiple comparisons test.

Close modal

Our results were substantiated by an in vivo vaccination study in nontumor-bearing wild-type versus Fap−/− mice using OVA as immunogen. Upon subcutaneous injection of OVA and FAP-moCD40, DCs were activated in the LNs (inguinal, adjacent, and contralateral to the injection site) of wild-type but not Fap−/− mice. In the spleen, no DC activation was detected in wild-type mice, in line with the fact that murine splenic FRCs lack FAP expression (Fig. 3B; ref. 20). Surprisingly, FAP-moCD40 failed to activate B cells in the LNs of wild-type mice. A marginal B-cell activation could be detected in the spleen of wild-type but not Fap−/− mice (Supplementary Fig. S3C). OVA combined with a nontargeted murine CD40 agonist (Fc-functional FGK4.5 moIgG1) enhanced DC as well as B-cell activation in the LNs and spleens of Fap−/− mice (Fig. 3B; Supplementary Fig. S3C). In line with the APC activation results, the nontargeted anti-CD40 agonist triggered CD8+ T-cell proliferation in Fap−/− mice. In contrast, T-cell proliferation was induced with FAP-moCD40 in wild-type mice but not in Fap−/− mice (Fig. 3C).

Thus, besides confirming the strict FAP dependency of the FAP-CD40 molecule, our data suggest that even low levels of FAP can induce activity of FAP-CD40. In that context, in particular FAP-expressing FRCs might play a role in crosslinking FAP-CD40 in the LNs, resulting in an in situ nodal DC activation.

FAP-CD40 eradicates tumors in vivo and induces long-lasting antitumor immunity while displaying no signs of toxicity

After verifying FAP-CD40's functionality and FAP specificity, we investigated the antitumor effects of FAP-dependent CD40 stimulation in the syngeneic subcutaneous MC38-FAP tumor model in huCD40tg mice. Compared with human tumors, subcutaneous MC38 tumors show decreased desmoplasia and contain lower amounts of stromal FAP (38). As these reduced murine FAP levels might be a limiting factor for our in vivo studies, we chose to use MC38 cells with recombinant FAP expression. MC38-FAP tumors are characterized by an accumulation of T cells at the tumor border and moderate T-cell infiltration as well as high FAP expression levels at baseline (Fig. 4A; Supplementary Fig. S4A).

Figure 4.

In vivo comparison of FAP-huCD40 and SGN40 moIgG1 reveals superior antitumor efficacy and absence of side effects due to targeted CD40 agonism. A, IHC FAP and corresponding isotype control staining of MC38-FAP tumors in C57BL/6 mice. B, Scheme of the experimental study design: huCD40tg mice were subcutaneously (s.c.) injected with MC38-FAP tumor cells. Once the tumors reached a mean size of 220 mm3, mice were injected intraperitoneally (i.p.) with histidine buffer (vehicle group), 13.3 mg/kg FAP-huCD40, or 10 mg/kg SGN40 moIgG1 (doses were matched based on molar concentrations). Immuno-pharmacodynamics analysis (immune cell activation in inguinal tumor-draining and nondraining LNs, spleen and tumor as well as cytokine analysis) was performed on day three after therapy injection. HuCD40tg mice of the FAP-huCD40 group that rejected the tumor or naive huCD40tg mice were rechallenged with MC38-FAP tumor cells injected subcutaneously into the flank contralateral to the initial tumor injection site 71 days after therapy injection. C, Tumor growth in the different treatment groups and after rechallenge shown as mean tumor volume (±SEM) of nine mice per treatment group. Statistical significance on day 27 was calculated using Kruskal–Wallis test with Dunn multiple comparisons test. D, Body weight change of vehicle-, FAP-huCD40-, and SGN40 moIgG1–treated mice after administration of the treatment (nine mice per group). E, Liver toxicity assessment: liver enzymes alanine aminotransferase (ALT), glutamate dehydrogenase (GLDH), aspartate transaminase (AST), and sorbitol dehydrogenase (SDH) in the serum one day after therapy injection (left). Representative images of hematoxylin and eosin staining of liver sections four days after treatment, showing no noteworthy changes in vehicle and FAP-huCD40–treated mice. With SGN40 moIgG1 treatment, livers displayed foci of hepatocellular degeneration and inflammation, perivascular mixed cell infiltrates, activation of endothelial cells with leukocytes margination, increased cellularity in the sinusoids, Kupffer cells hypertrophy, and reduction of glycogen content in the hepatocytes (right). F, Inflammatory cytokines in tumor (left) and serum (right) three days posttreatment. Colors indicating cytokine levels are based on minimum (Min), average, and maximum (Max) values of each individual column. Each row represents one animal. G, DC activation three days after therapy injection: MFI of the activation markers murine CD40 (top) and CD86 (bottom; n = 4–7 mice per group). H, B-cell activation three days after treatment administration represented by CD69 MFI (n = 4–7 mice per group). I, Expression of CD69 (MFI) on CD8+ T cells (top) and CD8+ T cell to Treg ratio (bottom; n = 4–7 mice per group) three days post therapy injection. The data in C–I result from two independent experiments. Graphs in D, E, and G–I show mean and SD. Significance in E and G–I was calculated using unpaired ordinary one-way ANOVA with Dunnett multiple comparisons test. Shown are significant differences relative to the vehicle group. Immuno-pharmacodynamic effects, side effect and toxicity assessment, as well as effects on tumor growth of FAP-huCD40 were confirmed in three independent experiments. The efficacy comparison of FAP-huCD40 and SGN40 moIgG1 was done once. The tumor rechallenge was performed twice.

Figure 4.

In vivo comparison of FAP-huCD40 and SGN40 moIgG1 reveals superior antitumor efficacy and absence of side effects due to targeted CD40 agonism. A, IHC FAP and corresponding isotype control staining of MC38-FAP tumors in C57BL/6 mice. B, Scheme of the experimental study design: huCD40tg mice were subcutaneously (s.c.) injected with MC38-FAP tumor cells. Once the tumors reached a mean size of 220 mm3, mice were injected intraperitoneally (i.p.) with histidine buffer (vehicle group), 13.3 mg/kg FAP-huCD40, or 10 mg/kg SGN40 moIgG1 (doses were matched based on molar concentrations). Immuno-pharmacodynamics analysis (immune cell activation in inguinal tumor-draining and nondraining LNs, spleen and tumor as well as cytokine analysis) was performed on day three after therapy injection. HuCD40tg mice of the FAP-huCD40 group that rejected the tumor or naive huCD40tg mice were rechallenged with MC38-FAP tumor cells injected subcutaneously into the flank contralateral to the initial tumor injection site 71 days after therapy injection. C, Tumor growth in the different treatment groups and after rechallenge shown as mean tumor volume (±SEM) of nine mice per treatment group. Statistical significance on day 27 was calculated using Kruskal–Wallis test with Dunn multiple comparisons test. D, Body weight change of vehicle-, FAP-huCD40-, and SGN40 moIgG1–treated mice after administration of the treatment (nine mice per group). E, Liver toxicity assessment: liver enzymes alanine aminotransferase (ALT), glutamate dehydrogenase (GLDH), aspartate transaminase (AST), and sorbitol dehydrogenase (SDH) in the serum one day after therapy injection (left). Representative images of hematoxylin and eosin staining of liver sections four days after treatment, showing no noteworthy changes in vehicle and FAP-huCD40–treated mice. With SGN40 moIgG1 treatment, livers displayed foci of hepatocellular degeneration and inflammation, perivascular mixed cell infiltrates, activation of endothelial cells with leukocytes margination, increased cellularity in the sinusoids, Kupffer cells hypertrophy, and reduction of glycogen content in the hepatocytes (right). F, Inflammatory cytokines in tumor (left) and serum (right) three days posttreatment. Colors indicating cytokine levels are based on minimum (Min), average, and maximum (Max) values of each individual column. Each row represents one animal. G, DC activation three days after therapy injection: MFI of the activation markers murine CD40 (top) and CD86 (bottom; n = 4–7 mice per group). H, B-cell activation three days after treatment administration represented by CD69 MFI (n = 4–7 mice per group). I, Expression of CD69 (MFI) on CD8+ T cells (top) and CD8+ T cell to Treg ratio (bottom; n = 4–7 mice per group) three days post therapy injection. The data in C–I result from two independent experiments. Graphs in D, E, and G–I show mean and SD. Significance in E and G–I was calculated using unpaired ordinary one-way ANOVA with Dunnett multiple comparisons test. Shown are significant differences relative to the vehicle group. Immuno-pharmacodynamic effects, side effect and toxicity assessment, as well as effects on tumor growth of FAP-huCD40 were confirmed in three independent experiments. The efficacy comparison of FAP-huCD40 and SGN40 moIgG1 was done once. The tumor rechallenge was performed twice.

Close modal

We first compared the effects of FAP-huCD40 and the FcγR-dependent CD40 agonist SGN40 moIgG1 on tumor growth inhibition and immune activation (Fig. 4B). Strikingly, a single dose of 13.3 mg/kg FAP-huCD40 induced a complete tumor remission in all of the treated mice while an equivalent dose of 10 mg/kg (doses matched based on molar concentrations) SGN40 moIgG1 had no impact on tumor growth. When tumor-free mice in the FAP-huCD40 group were rechallenged with MC38-FAP tumor cells, the tumors were rejected in all the animals (Fig. 4C).

Consistent with observations from other groups using high doses of systemically administered nontargeted anti-CD40 agonists (16, 17), substantial side effects occurred in the SGN40 moIgG1-treated group. Mice receiving SGN40 moIgG1 experienced an average body weight loss of more than 15% four days after therapy injection (Fig. 4D). Of note, 2 out of 9 mice treated with SGN40 moIgG1 had to be sacrificed due to a reduction in body weight of more than 20%. The rest of the mice recovered six days after the weight drop. Moreover, SGN40 moIgG1 administration induced liver damage evidenced by an increase in liver enzymes and hepatocellular degeneration observed by histopathology (Fig. 4E). In contrast, none of the FAP-huCD40–treated animals displayed any signs of toxicity (Fig. 4D and E).

Thus, a single high dose of FAP-huCD40 induced efficient long-term protective antitumor immunity without causing the side effects typically observed upon treatment with systemic CD40 agonists.

We next aimed at obtaining a thorough understanding of the differences in immune cell stimulation triggered by FAP-huCD40 compared with SGN40 moIgG1.

The analysis of the levels of 23 inflammatory cytokines in serum versus tumor of treated mice three days after therapy injection revealed that FAP-huCD40 mediated an immune activation largely confined to the tumor site. Among the cytokines that displayed the strongest tumoral increase in response to FAP-huCD40 were the Th cell type 1 response-inducing IL12, as well as C-C motif chemokine 5 (CCL5), which plays an important role in attracting DCs to the tumor (Fig. 4F; Supplementary Fig. S4B; ref. 39). Unlike FAP-huCD40 and indicative of its systemic immunostimulatory effects, SGN40 moIgG1 failed to evoke a strong cytokine response in the tumor but induced a broad range of cytokines in the serum (Fig. 4F).

Furthermore, FAP-huCD40 significantly enhanced DC activation in the tumor-draining and nondraining LNs as well as in the tumor three days posttreatment. In the spleen, only minor DC activating effects of FAP-huCD40 were observed. In contrast, SGN40 moIgG1 induced a superior nodal and splenic but no tumoral DC activation (Fig. 4G). A strong B-cell activation triggered by SGN40 moIgG1 was detected in the LNs and to a lesser extent in the spleen and tumor. Compared with SGN40 moIgG1, FAP-huCD40 induced no or an inferior activation of B cells in LNs and spleen. Tumoral B-cell activation in FAP-huCD40 and SGN40 moIgG1–treated mice was, depending on the investigated activation marker, similar or more pronounced with SGN40 moIgG1 (Fig. 4H; Supplementary Fig. S4C).

Interestingly, only three days after therapy administration we observed a strongly enhanced expression of the early activation marker CD69 on cytotoxic T cells and an increased CD8+ T cell to regulatory T cell (Treg) ratio in tumors of FAP-huCD40- but not SGN40 moIgG1–treated animals (Fig. 4I).

To substantiate our findings, we compared FAP-huCD40's in vivo effects to those of another nontargeted agonistic anti-CD40 antibody. Selicrelumab is classified as one of the human CD40 agonists with highest potency, mediating CD40 activation in a partially FcγR crosslinking-independent fashion (7, 34, 40, 41).

While a high dose of 13.3 mg/kg FAP-huCD40 induced rapid tumor elimination in 8 out of 10 MC38-FAP tumor-bearing huCD40tg mice, an equivalent dose of selicrelumab merely conferred low growth-inhibitory effects (Fig. 5A; Supplementary Fig. S5A for individual tumor growth curves).

Figure 5.

Highly dosed FAP-huCD40 accumulates to a higher extent in the tumor and induces better efficacy than selicrelumab. A and D, HuCD40tg mice were subcutaneously injected with MC38-FAP tumor cells. Once the tumors reached a mean size of 250 mm3, mice were injected with histidine buffer (vehicle group) or the indicated doses of FAP-huCD40, nontargeted DP47-huCD40, and selicrelumab. Tumor growth depicted as mean (±SEM) of the tumor volume of 10 mice per group. Statistical significance on day 20 was calculated using Kruskal–Wallis test with Dunn multiple comparisons test. The dose-dependent antitumor efficacy of FAP-huCD40 was confirmed in two independent studies. The comparison of efficacy of FAP-huCD40 versus selicrelumab was done once. B, C, and E, HuCD40tg mice were subcutaneously injected with MC38-FAP tumor cells. After tumors had reached an average size of 275 mm3, the mice were injected with histidine buffer (vehicle) or indicated doses of Alexa Fluor 647 (AF647) labeled FAP-huCD40 or selicrelumab.B, Normalized concentration of FAP-huCD40-AF647 and selicrelumab-AF647 measured at different sites after 2, 24, and 72 hours postinjection. Normalized drug concentration was measured as photon flux of the respective organ normalized to organ weight (Ct) relative to background signal of the respective organ in vehicle-treated animals at 2 hours (C0). Shown are mean (±SD) values of three huCD40tg mice per group. Significant differences between treatments for each time point were calculated using two-way ANOVA. C, Labeled FAP-huCD40 and selicrelumab signals in the tumor at different time points detected by fluorescent live imaging (FLI). The color scale indicates the magnitude of flux. E, Signal of labeled antibodies on DCs in tumor, nodes, and spleen 24 hours after treatment. Depicted are individual AF647 MFI values of 2–3 mice per group, mean and SD. Significance was calculated by ordinary one-way ANOVA using Tukey multiple comparisons test. The biodistribution study was performed once.

Figure 5.

Highly dosed FAP-huCD40 accumulates to a higher extent in the tumor and induces better efficacy than selicrelumab. A and D, HuCD40tg mice were subcutaneously injected with MC38-FAP tumor cells. Once the tumors reached a mean size of 250 mm3, mice were injected with histidine buffer (vehicle group) or the indicated doses of FAP-huCD40, nontargeted DP47-huCD40, and selicrelumab. Tumor growth depicted as mean (±SEM) of the tumor volume of 10 mice per group. Statistical significance on day 20 was calculated using Kruskal–Wallis test with Dunn multiple comparisons test. The dose-dependent antitumor efficacy of FAP-huCD40 was confirmed in two independent studies. The comparison of efficacy of FAP-huCD40 versus selicrelumab was done once. B, C, and E, HuCD40tg mice were subcutaneously injected with MC38-FAP tumor cells. After tumors had reached an average size of 275 mm3, the mice were injected with histidine buffer (vehicle) or indicated doses of Alexa Fluor 647 (AF647) labeled FAP-huCD40 or selicrelumab.B, Normalized concentration of FAP-huCD40-AF647 and selicrelumab-AF647 measured at different sites after 2, 24, and 72 hours postinjection. Normalized drug concentration was measured as photon flux of the respective organ normalized to organ weight (Ct) relative to background signal of the respective organ in vehicle-treated animals at 2 hours (C0). Shown are mean (±SD) values of three huCD40tg mice per group. Significant differences between treatments for each time point were calculated using two-way ANOVA. C, Labeled FAP-huCD40 and selicrelumab signals in the tumor at different time points detected by fluorescent live imaging (FLI). The color scale indicates the magnitude of flux. E, Signal of labeled antibodies on DCs in tumor, nodes, and spleen 24 hours after treatment. Depicted are individual AF647 MFI values of 2–3 mice per group, mean and SD. Significance was calculated by ordinary one-way ANOVA using Tukey multiple comparisons test. The biodistribution study was performed once.

Close modal

Coinciding with our previous observations, only the nontargeted anti-CD40 agonist selicrelumab, but not FAP-huCD40 induced a severe body weight loss (Supplementary Fig. S5B). Multiplexed cytokine analysis and investigation of DC activation in different organs confirmed the enhanced intratumoral effects of FAP-huCD40 as opposed to the strong systemic immunostimulatory effects of selicrelumab, similar to those of SGN40 moIgG1 (Supplementary Fig. S5C and S5D). Moreover, analysis of the expression of 518 immune response–related genes in the tumor one day after treatment revealed an increased tumoral effect of FAP-huCD40 compared with selicrelumab. Immunologic pathways such as IFN, toll-like receptor, and cytokine signaling were among the ones particularly impacted by FAP-huCD40 treatment (Supplementary Fig. S5E and Supplementary Table S2).

In accordance with these results, investigation of the biodistribution of the highly dosed fluorescently labeled antibodies over 72 hours showed increased accumulation of FAP-huCD40 versus selicrelumab in the tumor tissue. Both molecules showed accumulation in the tumor-draining and nondraining LNs, with selicrelumab displaying higher nodal levels over time. Clearing of FAP-huCD40 as well as selicrelumab from blood and organs of perfusion (spleen, lung) as well as excretory organs (liver, kidney) started after 2 to 24 hours postinjection. It is noteworthy that selicrelumab showed a delayed clearance in the spleen associated with a higher plateau concentration compared to FAP-huCD40 (Fig. 5B and C).

A high dose of FAP-CD40 is indispensable for efficient antitumor responses

FAP-huCD40 outperformed the nontargeted anti-CD40 agonists SGN40 moIgG1 and selicrelumab in vivo and conferred remarkable efficacy with a single high dose. On the basis of these findings, we next asked whether a lower concentration would diminish our molecule's efficacy. Indeed, potent antitumor responses were only mediated by a FAP-huCD40 dose of 13.3 mg/kg. Lowering the dose to 4 mg/kg resulted in merely a slight tumor growth inhibition and did not induce significantly better anti-tumor efficacy compared with the corresponding low dose of selicrelumab (3 mg/kg; Fig. 5D).

Comparing the 13.3 mg/kg versus the less efficacious 4 mg/kg FAP-huCD40 treatment revealed a major impact of the dosing on intratumoral DCs. A threefold dose increase from 4 mg/kg to 13.3 mg/kg translated to threefold more labled FAP-huCD40 molecule bound to DCs in the tumor (Fig. 5E). These pronounced dose-related differences could not be seen for tumoral B cells (Supplementary Fig. S5F). In the LNs and spleen, dose-dependent effects of FAP-huCD40 on DCs were only moderate or absent. In contrast, high dosing of selicrelumab led to a strong increase of labeled antibody bound to DCs in LNs and spleen but not in the tumor (Fig. 5E). Hence, a high dose of FAP-huCD40 seems in particular crucial for the bispecific antibody to reach intratumoral DCs.

FAP-CD40 induces tumor growth inhibition in a poorly infiltrated pancreatic tumor model with abundant stromal FAP expression

Highly dosed FAP-huCD40 showed a remarkable efficacy in MC38-FAP tumors. To corroborate FAP-CD40's in vivo effects, we tested our bispecific molecule in a second, T-cell–excluded pancreatic cancer model. Human CEA transgenic (huCEAtg) mice were subcutaneously implanted with murine pancreatic KPC-4662 tumor cells recombinantly expressing human CEA (KPC-4662-huCEA). Unlike subcutaneous MC38 tumors, subcutaneous KPC-4662 tumors develop thick stromal banding patterns that can also be frequently observed in human tumors (30, 38). Consequently, we detected a high, stroma-restricted FAP expression in subcutaneous KPC-4662-huCEA tumors (Fig. 6A). FAP-moCD40 mediated distinct tumor growth–inhibitory effects in KPC-4662-huCEA tumor-bearing mice (Fig. 6B). Tumor regressions ranging from 20% to 90% were observed in 6 out of 10 FAP-moCD40-treated animals (Fig. 6C). Strikingly, the FAP-moCD40–induced tumor growth inhibition was preceded by a strong CD4+FoxP3 and CD8+ T-cell tumor infiltration detected eight days after FAP-moCD40 administration (Fig. 6D–F). Thus, our data suggest that in presence of abundant amounts of stromal FAP, a high dose of FAP-CD40 induced inflammation of KPC-4662-huCEA tumors, thereby effectively inhibiting tumor growth.

Figure 6.

FAP-moCD40 induces tumor growth inhibition in a poorly infiltrated pancreatic tumor model with abundant stromal FAP expression. A–F, HuCEAtg mice were subcutaneously injected with KPC-4662-huCEA cells. After an average tumor size of 240 mm3 was reached, histidine buffer (vehicle group) or 13.3 mg/kg FAP-moCD40 was administered. Treatment groups consisted of 15 mice each. Five mice were sacrificed for immuno-pharmacodynamics analysis eight days after therapy injection. A, Immunostaining of KPC-4662-huCEA tumors for FAP (pink) and E-cadherin (mint, marker for tumor cells). B, Tumor growth over time depicted as mean (±SEM) of the tumor volume. Statistical significance on day 22 was calculated using unpaired two-tailed Mann–Whitney U test. C, Variation of tumor volume from baseline for each mouse. Negative values represent tumor regression. Infiltration (D) and quantification (E) of CD4+ (mint) and CD8+ (purple) T cells in KPC-4662-huCEA tumors determined by immunostaining eight days after therapy administration. F, Fraction of Tregs within total intratumoral CD4+ T cells and ratio of CD8+ T cells to Tregs in the tumor of vehicle or FAP-moCD40–treated animals. Graphs in E and F show individual values of four animals per group, mean and SD. Statistical significance was evaluated using unpaired two-tailed t test. The study was performed once.

Figure 6.

FAP-moCD40 induces tumor growth inhibition in a poorly infiltrated pancreatic tumor model with abundant stromal FAP expression. A–F, HuCEAtg mice were subcutaneously injected with KPC-4662-huCEA cells. After an average tumor size of 240 mm3 was reached, histidine buffer (vehicle group) or 13.3 mg/kg FAP-moCD40 was administered. Treatment groups consisted of 15 mice each. Five mice were sacrificed for immuno-pharmacodynamics analysis eight days after therapy injection. A, Immunostaining of KPC-4662-huCEA tumors for FAP (pink) and E-cadherin (mint, marker for tumor cells). B, Tumor growth over time depicted as mean (±SEM) of the tumor volume. Statistical significance on day 22 was calculated using unpaired two-tailed Mann–Whitney U test. C, Variation of tumor volume from baseline for each mouse. Negative values represent tumor regression. Infiltration (D) and quantification (E) of CD4+ (mint) and CD8+ (purple) T cells in KPC-4662-huCEA tumors determined by immunostaining eight days after therapy administration. F, Fraction of Tregs within total intratumoral CD4+ T cells and ratio of CD8+ T cells to Tregs in the tumor of vehicle or FAP-moCD40–treated animals. Graphs in E and F show individual values of four animals per group, mean and SD. Statistical significance was evaluated using unpaired two-tailed t test. The study was performed once.

Close modal

High doses of FAP-CD40 are well tolerated in cynomolgus monkeys and do not result in CD40-related toxicities

On the basis of the finding that high doses of FAP-huCD40 are required to mediate potent antitumor responses, we tested whether these high doses are tolerated in a nonrodent toxicology species. Two groups of cynomolgus monkeys were consecutively dose-escalated weekly with two different dose increments. The first group of animals received three doses from 10 to 100 mg/kg FAP-huCD40 and the second group three doses from 200 to 400 mg/kg (Supplementary Fig. S6A). All treated animals were exposed to high levels of FAP-huCD40 (Supplementary Fig. S6B). High concentrations of systemically administered FAP-huCD40 did not result in mortalities or FAP-huCD40–related effects on clinical observations or body weight (Supplementary Table S3A). Multiple phase I studies with agonistic anti-CD40 antibodies encountered dose-limiting toxicities including CRS, liver function test abnormalities, and a decrease in platelet counts (12–15). Therefore, we examined the FAP-huCD40–treated animals for secretion of eleven different cytokines, and analyzed hematology, clinical chemistry, and histopathology. For most of the cytokines measured, the concentrations were below the limit of quantification. For IL8, CCL2 and CCL5 results were measurable, but showed no obvious dosing effects (Supplementary Table S3B). No changes in liver enzymes (Supplementary Fig. S6C) were observed. Coagulation parameters such as platelet counts, D-dimer, activated partial thromboplastin time (APTT), prothrombin time (PT), and fibrinogen remained similar to untreated controls (Supplementary Fig. S6D). Taken together, administration of FAP-huCD40 at very high doses up to 400 mg/kg was well tolerated and did not result in any CD40-related toxicities in cynomolgus monkeys.

In this study, we introduce for the first time a CD40 agonistic antibody whose activity is strictly dependent on the presence of FAP. By creating a FAP-restricted anti-CD40 molecule, we aimed to enable safe administration of high doses to overcome peripheral CD40 sink effects. We envisioned that the increased therapeutic window but also the targeting of our molecule to the tumor area via FAP could lead to a more efficient and selective activation of tumoral APCs, thereby enhancing tumor antigen (cross-) presentation and consequently antitumor immunity.

The results of our in vitro APC activation and T-cell priming assays clearly indicated that FAP-CD40 provides a strong CD40-stimulating signal in a FAP-specific manner. These findings were recapitulated by in vivo studies in huCD40tg mice, in which FAP-huCD40 predominantly triggered potent immune activation in the tumor in contrast to the nontargeted CD40 agonists SGN40 moIgG1 and selicrelumab, which induced strong peripheral but minor intratumoral effects. Restriction of CD40 stimulation to FAP+ areas abolished the pronounced side effects caused by systemic CD40 activation. A high dose of FAP-huCD40 could therefore be safely administered. The good tolerability of FAP-CD40 was confirmed in cynomolgus monkeys, indicating a favorable safety profile of our therapy for clinical development. Of note, our in vivo efficacy data demonstrated that the administration of a high dose of FAP-targeted anti-CD40 agonist is indispensable for the induction of successful antitumor immunity; only when high amounts of FAP-huCD40 were injected, complete MC38-FAP tumor regression and long-term protection of treated mice were induced. In contrast, minor to no effects on tumor growth inhibition were observed with highly dosed SGN40 moIgG1 and selicrelumab.

With SGN40 moIgG1 and selicrelumab, we selected two nontargeted CD40 agonists that possess different FcγR crosslinking requirements as well as potencies. The huIgG2 selicrelumab is one of the strongest CD40 agonists and acts partially independent of crosslinking (7, 40, 41). It is noteworthy that the affinities of huIgG2s are similar for both the murine and human FcγRIIB (42, 43), the FcγR subclass considered most crucial for Fc-mediated crosslinking of anti-CD40 agonists (35, 44). Hence, in the huCD40tg mouse model, both FcγR-independent and FcγR-dependent activation of selicrelumab is possible.

For SGN40 moIgG1, the Fv domains of the crosslinking-dependent human anti-CD40 agonist SGN40 were combined with a murine IgG1 backbone to enable a good functionality in huCD40tg mice. Of note, murine IgG1s have a higher affinity to the murine FcγRIIB compared to the affinity of human IgG1s for the human FcγRIIB (42, 43). Thus, crosslinking and consequently biological activity of SGN40 moIgG1 in huCD40tg mice is presumably enhanced relative to the activity of SGN40 huIgG1 in humans.

Therefore, it seems unlikely that insufficient crosslinking could account for the lack of efficacy of the nontargeted CD40 agonists in our huCD40tg mouse model.

This was supported by the analysis of immune cell activation induced by FAP-huCD40 versus SGN40 moIgG1 and selicrelumab: the overall immuno-pharmacodynamic effects conferred by the non-targeted anti-CD40 antibodies were not less potent per se. On the contrary, they triggered a stronger activation of B cells, DCs, as well as T cells in the LNs and the spleen, compared with FAP-huCD40. The nontargeted CD40 agonists, however, failed to induce DC or T-cell responses in the tumor, whereas with FAP-huCD40 tumoral DC and, presumably concomitant, tumoral CD8+ T-cell activation was observed. Hence, we hypothesize that stimulation of DCs in the tumor is essential for FAP-huCD40's efficacy. Two factors could account for the superiority of FAP-huCD40 in activating tumor DCs: FAP-mediated crosslinking might be more effective in the tumor than FcγR-crosslinking and, as evidenced by our biodistribution data, targeting to FAP enhanced the intratumoral accumulation of our antibody.

Indeed, our dose–response study supported the hypotheses that (i) the amount of CD40 agonist in the tumor is a limiting factor for efficient tumoral DC activation and that (ii) this tumoral DC activation could be crucial for FAP-CD40's efficacy: decreasing the FAP-huCD40 dose led to a significantly lower amount of antibody reaching tumor DCs, accompanied by a loss of efficacy. In addition, mechanistic studies of the accompanying paper by Labiano and colleagues, entailing the use of cDC1-deficient Batf3−/− mice and T-cell depletion, indicate the requirement of an activation of the cDC1-CD8+ T-cell axis for FAP-CD40–mediated antitumor immunity (45).

While proposing an instrumental role of tumoral DC activation for FAP-CD40's efficacy, we found evidence that low FAP expression by nodal FRCs might as well contribute to FAP-CD40's mode of action. In FAP-huCD40–treated animals, an accumulation of the therapeutic antibody as well as DC activation was not only observed in the tumor, but also, albeit to a lower degree, in tumor-draining and nondraining LNs. In contrast, in FAP-negative spleens no antibody accumulation or substantial effects of FAP-huCD40 on DCs were detected. It is possible that activated nodal DCs result from intratumoral stimulation and subsequent homing to the LNs. However, taken together, our results suggest that in situ DC activation in the LNs is initiated by FAP-CD40. First, DC activation was similar in the tumor-draining versus nondraining LNs of FAP-huCD40–treated animals. This would likely not be the case if only trafficking of tumoral DCs accounted for the observed LN DC activation. Second, our in vitro studies showed that FAP expression on nodal FRCs is sufficient to trigger FAP-moCD40 crosslinking. Third, nodal DC activation was also observed in non–tumor-bearing mice when FAP-moCD40 was administered in a vaccination setting. Thus, it seems likely that FAP-CD40 acts directly in LNs. This might be a beneficial feature, given the importance of, in particular, the tumor-draining LNs for DC-mediated cross-presentation of tumor antigen (46). Further investigations are required to determine to which extent this nodal activity contributes to the efficacy of our molecule.

Surprisingly, in the LNs of both tumor-bearing and non–tumor-bearing mice, FAP-CD40 activated DCs, whereas B-cell activation levels remained unchanged. Moreover, FAP-huCD40 was superior to nontargeted CD40 agonists in activating DCs but not B cells in the tumor. This is in opposition to our in vitro data, which clearly demonstrated the ability of FAP-CD40 to trigger potent B-cell responses. This discrepancy might be partially explained by a lack of colocalization of FAP+ cells and B cells in vivo. FAP-expressing FRCs are known to directly interact with DCs in the nodes via the C-type-lectin-like-2-podoplanin axis (47), which might allow the close contact needed for FAP-CD40–mediated activation. In contrast, the quality or frequency of nodal B cell–FRC interactions might be insufficient. Nonetheless, given the broad FAP expression in our MC38-FAP tumor model, it seems unlikely that a dearth of colocalization alone accounts for the diminished tumoral B-cell activation in FAP-CD40–treated animals.

The overall superior B-cell–activating effects of non-targeted CD40 agonists compared with FAP-huCD40 suggest that in the MC38-FAP tumor model B-cell activation might not play a major role for the potent antitumor activity of FAP-huCD40.

It is worth mentioning that we detected a slight FAP-dependent B-cell activation in the spleens of FAP-CD40–treated mice. As splenic FRCs do not express FAP (20), this effect might be caused by migrating B cells from FAP+ areas rather than by an in situ splenic activation. Low frequencies of FAP-expressing cells are not only reported for the LNs but also other organs, including the bone marrow (48), which were not the subject of our studies but might play a role for the observed splenic B-cell activation.

Our investigations primarily focused on the impact of FAP-CD40 on DCs and B cells; however, numerous studies provided experimental evidence for a beneficial effect of CD40 agonists on macrophages. Activation of this cell subset and reprogramming toward a proinflammatory tumor-associated macrophage phenotype, as well as induction of tumor stroma degradation, are among the described mechanisms by which anti-CD40 antibodies can drive macrophage-mediated antitumor immunity (49, 50). Potential effects of FAP-CD40 on macrophages were not addressed within the scope of this study but are under investigation.

Utilizing the MC38-FAP and KPC-4662-huCEA murine tumor models, we demonstrated that a single high dose of FAP-huCD40 conferred remarkable antitumor efficacy. With MC38-FAP, we used a tumor cell line with recombinant FAP expression, while our studies in KPC-4662-huCEA tumor-bearing mice allowed us to confirm FAP-CD40's antitumor activity in a tumor model in which FAP expression was restricted to the tumor stroma. Regarding the clinical development of our bispecific antibody, it will be pivotal to explore the effects of FAP-CD40 in tumors with varying FAP expression levels and localization patterns. Importantly, the accompanying article by Labiano and colleagues demonstrates a notable antitumor efficacy of FAP-CD40 in combination with radiotherapy in a murine orthotopic head and neck tumor model, which recapitulates the tumor stromal FAP expression patterns of human head and neck squamous cell carcinomas (45).

In addition, as FAP expression can be induced in fibroblasts during wound healing (18), it will be important to evaluate whether patients' surgical wounds could pose a potential risk for side-effects during FAP-CD40 treatment.

Another vital aspect to be considered regarding clinical development of FAP-CD40 will be the selection of combination partners. An attractive combination strategy might be pairing of CD40 agonists with checkpoint inhibitors, which has been shown to enable effective priming and durable T-cell responses (9). Moreover, chemo- or radiotherapy entailing immunogenic cell death and spilling of tumor antigen is believed to function as potent in situ vaccination together with CD40 agonists (8, 51, 52). Indeed, the promising preclinical results that Labiano and colleagues demonstrated by applying hypofractionated radiotherapy plus FAP-CD40 treatment provide a strong rationale for this combinatorial strategy (45).

In summary, our data show that agonizing CD40 and thereby bridging the gap between innate and adaptive immunity, is unequivocally a powerful tool for CIT. Yet, the full therapeutic capacity of the CD40 pathway has not been leveraged by previously tested nontargeted anti-CD40 antibodies. On the basis of our preclinical evidence, we propose that a FAP-targeted CD40 agonist can be safely administered at high doses to unleash its full potential in the tumor area and, with the right combinatorial approach, holds great promise for clinical applications.

E. Sum reports a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG and ownership of Roche stock (options). M. Rapp reports a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG, a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG, and ownership of Roche stock (options). P. Fröbel reports ownership of Roche stock options. M. Le Clech reports a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG and ownership of Roche stock (options). H. Dürr reports a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG and a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG. A.M. Giusti reports ownership of Roche stock options. M. Perro is an employee of Roche at the time of the work described in the manuscript. L. Kunz reports personal fees from Roche Glycart AG during the conduct of the study, as well as personal fees from Roche Glycart AG outside the submitted work. E. Menietti reports personal fees from Roche during the conduct of the study and personal fees from Roche outside the submitted work. P. Brünker reports a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG, a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG, and participation in Roche Connect program (stock options). U. Hopfer reports ownership of Roche non-voting equity securities. M. Lechmann reports participation in Roche Connect and ownership of Roche stocks. V. Nicolini reports other from Roche during the conduct of the study and other from Roche outside the submitted work; in addition, V. Nicolini has a patent 10577429 issued and is a Roche employee. S. Labiano reports grants and personal fees from Roche Glycart during the conduct of the study; in addition, S. Labiano has a patent for EP20207768.1 pending to F. Hoffmann-La Roche AG. F. Weber is an employee of Roche and participates in the "Roche Connect" program (owns stock of Roche). T. Emrich is an employee of Roche Diagnostics GmbH, Germany and reports Roche bonus shares and profit certificates. P. Romero reports grants from Roche pRED during the conduct of the study and personal fees from MaxiVax outside the submitted work; in addition, P. Romero has a patent for AD2838 EP BS pending, has a patent for AC1689 PCT BS pending, and is editor-in-chief of Journal for Immunotherapy of Cancer. C. Trumpfheller reports a patent for EP20207768.1 pending to F. Hoffmann-La Roche AG, a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG, and a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG; C. Trumpfheller also reports ownership of Roche stock (options). P. Umaña reports a patent for WO 2018/185045 A1 pending to F. Hoffmann-La Roche AG, a patent for WO 2020/070041 A1 pending to F. Hoffmann-La Roche AG, and a patent for EP20207768.1 pending to F. Hoffmann-La Roche AG; P. Umaña also reports ownership of Roche shares. No disclosures were reported by the other authors.

E. Sum: Conceptualization, formal analysis, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. M. Rapp: Conceptualization, formal analysis, supervision, validation, investigation, visualization, methodology, project administration, writing–review and editing. P. Fröbel: Formal analysis, investigation, methodology. M. Le Clech: Conceptualization, formal analysis, investigation, visualization, methodology. H. Dürr: Conceptualization, resources. A.M. Giusti: Formal analysis, visualization. M. Perro: Conceptualization, visualization. D. Speziale: Formal analysis, investigation, visualization, writing–original draft. L. Kunz: Formal analysis, investigation, visualization, methodology, writing–original draft. E. Menietti: Formal analysis, investigation, visualization, methodology, writing–original draft. P. Brünker: Conceptualization. U. Hopfer: Conceptualization, formal analysis, visualization, writing–original draft. M. Lechmann: Conceptualization, formal analysis, visualization. A. Sobieniecki: Investigation. B. Appelt: Investigation. R. Adelfio: Investigation. V. Nicolini: Investigation, visualization. A. Freimoser-Grundschober: Resources, investigation. W. Jordaan: Investigation. S. Labiano: Conceptualization. F. Weber: Formal analysis. T. Emrich: Formal analysis. F. Christen: Investigation. B. Essig: Investigation. P. Romero: Conceptualization, supervision. C. Trumpfheller: Conceptualization, supervision, validation, project administration, writing–review and editing. P. Umaña: Conceptualization, supervision, writing–review and editing.

We thank Maximiliane König, Klaus Neff, and Anton Jochner (RICM) for their contributions to design, production, and supply of the bispecific antibody constructs used in this study. We thank Christophe Chabbert and Said Aktas (RICZ) for their help with the statistical and NanoString data analyses. We also thank Junichiro Sonoda (Genentech) for kindly providing the Fap−/− mice. We gratefully acknowledge the RICZ Cell Technologies group for the generation of recombinant cell lines used for in vitro and in vivo studies. Moreover, we would like to thank the members of the RICZ in vivo pharmacology team for their support of the mouse studies. Thanks to Claudia Haftmann (RICZ), Olivera Cirovic (RICB), and Dörte Klostermeyer Rauber (Hoffmann-La Roche Patent Department) for the discussions and critical reading of the manuscript. We are also grateful to the RICZ Biochemistry and Downstream Processing teams for the supply, fluorescent labeling, biotinylation, and endotoxin purification of reagents used in this study. We thank Florian Kast for his support in generating BioRender graphs. This work was funded by F. Hoffmann-La Roche AG. Pedro Romero was supported by grants from Swiss Cancer League (KFS-4404–02–2018) and the Swiss National Science Foundation (310030_182735).

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.
Dadgostar
H
,
Zarnegar
B
,
Hoffmann
A
,
Qin
XF
,
Truong
U
,
Rao
G
, et al
Cooperation of multiple signaling pathways in CD40-regulated gene expression in B lymphocytes
.
Proc Natl Acad Sci U S A
2002
;
99
:
1497
502
.
2.
Gallagher
E
,
Enzler
T
,
Matsuzawa
A
,
Anzelon-Mills
A
,
Otero
D
,
Holzer
R
, et al
Kinase MEKK1 is required for CD40-dependent activation of the kinases Jnk and p38, germinal center formation, B cell proliferation and antibody production
.
Nat Immunol
2007
;
8
:
57
63
.
3.
van Kooten
C
,
Banchereau
J
. 
CD40-CD40 ligand
.
J Leukoc Biol
2000
;
67
:
2
17
.
4.
Quezada
SA
,
Jarvinen
LZ
,
Lind
EF
,
Noelle
RJ
. 
CD40/CD154 interactions at the interface of tolerance and immunity
.
Annu Rev Immunol
2004
;
22
:
307
28
.
5.
French
RR
,
Chan
HT
,
Tutt
AL
,
Glennie
MJ
. 
CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help
.
Nat Med
1999
;
5
:
548
53
.
6.
Sotomayor
EM
,
Borrello
I
,
Tubb
E
,
Rattis
FM
,
Bien
H
,
Lu
Z
, et al
Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40
.
Nat Med
1999
;
5
:
780
7
.
7.
Vonderheide
RH
. 
CD40 agonist agntibodies in cancer immunotherapy
.
Annu Rev Med
2020
;
71
:
47
58
.
8.
Vonderheide
RH
. 
The immune revolution: a case for priming, not checkpoint
.
Cancer Cell
2018
;
33
:
563
9
.
9.
Morrison
AH
,
Diamond
MS
,
Hay
CA
,
Byrne
KT
,
Vonderheide
RH
. 
Sufficiency of CD40 activation and immune checkpoint blockade for T cell priming and tumor immunity
.
Proc Natl Acad Sci U S A
2020
;
117
:
8022
31
.
10.
Grilley-Olson
JE
,
Curti
BD
,
Smith
DC
,
Goel
S
,
Gajewski
T
,
Markovic
S
, et al
SEA-CD40, a non-fucosylated CD40 agonist: interim results from a phase 1 study in advanced solid tumors
.
J Clin Oncol
2018
;
36
:
3093
.
11.
Bendell
JC
,
Fakih
M
,
Infante
JR
,
Bajor
DL
,
Cristea
MC
,
Tremblay
T
, et al
Phase 1 study to evaluate the safety and tolerability of the CD40 agonistic monoclonal antibody APX005M in subjects with solid tumors
.
J Clin Oncol
2016
;
34
:
TPS3110
TPS
.
12.
Vonderheide
RH
,
Flaherty
KT
,
Khalil
M
,
Stumacher
MS
,
Bajor
DL
,
Hutnick
NA
, et al
Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody
.
J Clin Oncol
2007
;
25
:
876
83
.
13.
Advani
R
,
Forero-Torres
A
,
Furman
RR
,
Rosenblatt
JD
,
Younes
A
,
Shankles
B
, et al
SGN-40 (Anti-huCD40 mAb) monotherapy induces durable objective responses in patients with relapsed aggressive non-Hodgkin's lymphoma: evidence of antitumor activity from a phase I study
.
Blood
2006
;
108
:
695
.
14.
Ruter
J
,
Antonia
SJ
,
Burris
HA
,
Huhn
RD
,
Vonderheide
RH
. 
Immune modulation with weekly dosing of an agonist CD40 antibody in a phase I study of patients with advanced solid tumors
.
Cancer Biol Ther
2010
;
10
:
983
93
.
15.
Vonderheide
RH
,
Burg
JM
,
Mick
R
,
Trosko
JA
,
Li
D
,
Shaik
MN
, et al
Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors
.
Oncoimmunology
2013
;
2
:
e23033
.
16.
Fransen
MF
,
Sluijter
M
,
Morreau
H
,
Arens
R
,
Melief
CJ
. 
Local activation of CD8 T cells and systemic tumor eradication without toxicity via slow release and local delivery of agonistic CD40 antibody
.
Clin Cancer Res
2011
;
17
:
2270
80
.
17.
Knorr
DA
,
Dahan
R
,
Ravetch
JV
. 
Toxicity of an Fc-engineered anti-CD40 antibody is abrogated by intratumoral injection and results in durable antitumor immunity
.
Proc Natl Acad Sci U S A
2018
;
115
:
11048
53
.
18.
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
.
19.
Rettig
WJ
,
Garin-Chesa
P
,
Beresford
HR
,
Oettgen
HF
,
Melamed
MR
,
Old
LJ
. 
Cell-surface glycoproteins of human sarcomas: differential expression in normal and malignant tissues and cultured cells
.
Proc Natl Acad Sci U S A
1988
;
85
:
3110
4
.
20.
Denton
AE
,
Roberts
EW
,
Linterman
MA
,
Fearon
DT
. 
Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8+ T cells
.
Proc Natl Acad Sci U S A
2014
;
111
:
12139
44
.
21.
Jansen
CS
,
Prokhnevska
N
,
Master
VA
,
Sanda
MG
,
Carlisle
JW
,
Bilen
MA
, et al
An intra-tumoral niche maintains and differentiates stem-like CD8 T cells
.
Nature
2019
;
576
:
465
70
.
22.
Hunter
TB
,
Alsarraj
M
,
Gladue
RP
,
Bedian
V
,
Antonia
SJ
. 
An agonist antibody specific for CD40 induces dendritic cell maturation and promotes autologous anti-tumour T-cell responses in an in vitro mixed autologous tumour cell/lymph node cell model
.
Scand J Immunol
2007
;
65
:
479
86
.
23.
Schlothauer
T
,
Herter
S
,
Koller
CF
,
Grau-Richards
S
,
Steinhart
V
,
Spick
C
, et al
Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions
.
Protein Eng Des Sel
2016
;
29
:
457
66
.
24.
Carter
P
. 
Bispecific human IgG by design
.
J Immunol Methods
2001
;
248
:
7
15
.
25.
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
.
26.
Claus
C
,
Ferrara
C
,
Xu
W
,
Sam
J
,
Lang
S
,
Uhlenbrock
F
, et al
Tumor-targeted 4–1BB agonists for combination with T cell bispecific antibodies as off-the-shelf therapy
.
Sci Transl Med
2019
;
11
:
eaav5989
.
27.
Kelley
SK
,
Gelzleichter
T
,
Xie
D
,
Lee
WP
,
Darbonne
WC
,
Qureshi
F
, et al
Preclinical pharmacokinetics, pharmacodynamics, and activity of a humanized anti-CD40 antibody (SGN-40) in rodents and non-human primates
.
Br J Pharmacol
2006
;
148
:
1116
23
.
28.
Rolink
A
,
Melchers
F
,
Andersson
J
. 
The SCID but not the RAG-2 gene product is required for S mu-S epsilon heavy chain class switching
.
Immunity
1996
;
5
:
319
30
.
29.
Mizobata
S
,
Tompkins
K
,
Simpson
JF
,
Shyr
Y
,
Primus
FJ
. 
Induction of cytotoxic T cells and their antitumor activity in mice transgenic for carcinoembryonic antigen
.
Cancer Immunol Immunother
2000
;
49
:
285
95
.
30.
Lo
A
,
Wang
LS
,
Scholler
J
,
Monslow
J
,
Avery
D
,
Newick
K
, et al
Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells
.
Cancer Res
2015
;
75
:
2800
10
.
31.
Dunshee
DR
,
Bainbridge
TW
,
Kljavin
NM
,
Zavala-Solorio
J
,
Schroeder
AC
,
Chan
R
, et al
Fibroblast activation protein cleaves and inactivates fibroblast growth factor 21
.
J Biol Chem
2016
;
291
:
5986
96
.
32.
Clarke
P
,
Mann
J
,
Simpson
JF
,
Rickard-Dickson
K
,
Primus
FJ
. 
Mice transgenic for human carcinoembryonic antigen as a model for immunotherapy
.
Cancer Res
1998
;
58
:
1469
77
.
33.
Fletcher
AL
,
Malhotra
D
,
Acton
SE
,
Lukacs-Kornek
V
,
Bellemare-Pelletier
A
,
Curry
M
, et al
Reproducible isolation of lymph node stromal cells reveals site-dependent differences in fibroblastic reticular cells
.
Front Immunol
2011
;
2
:
35
.
34.
Richman
LP
,
Vonderheide
RH
. 
Role of crosslinking for agonistic CD40 monoclonal antibodies as immune therapy of cancer
.
Cancer Immunol Res
2014
;
2
:
19
26
.
35.
Li
F
,
Ravetch
JV
. 
Inhibitory Fcgamma receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies
.
Science
2011
;
333
:
1030
4
.
36.
Bonifaz
L
,
Bonnyay
D
,
Mahnke
K
,
Rivera
M
,
Nussenzweig
MC
,
Steinman
RM
. 
Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance
.
J Exp Med
2002
;
196
:
1627
38
.
37.
Hogquist
KA
,
Jameson
SC
,
Heath
WR
,
Howard
JL
,
Bevan
MJ
,
Carbone
FR
. 
T cell receptor antagonist peptides induce positive selection
.
Cell
1994
;
76
:
17
27
.
38.
Tran
E
,
Chinnasamy
D
,
Yu
Z
,
Morgan
RA
,
Lee
CC
,
Restifo
NP
, et al
Immune targeting of fibroblast activation protein triggers recognition of multipotent bone marrow stromal cells and cachexia
.
J Exp Med
2013
;
210
:
1125
35
.
39.
Bottcher
JP
,
Bonavita
E
,
Chakravarty
P
,
Blees
H
,
Cabeza-Cabrerizo
M
,
Sammicheli
S
, et al
NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control
.
Cell
2018
;
172
:
1022
37
.
40.
White
AL
,
Chan
HT
,
French
RR
,
Willoughby
J
,
Mockridge
CI
,
Roghanian
A
, et al
Conformation of the human immunoglobulin G2 hinge imparts superagonistic properties to immunostimulatory anticancer antibodies
.
Cancer Cell
2015
;
27
:
138
48
.
41.
Yu
X
,
Chan
HTC
,
Fisher
H
,
Penfold
CA
,
Kim
J
,
Inzhelevskaya
T
, et al
Isotype switching converts anti-CD40 antagonism to agonism to elicit potent antitumor activity
.
Cancer Cell
2020
;
37
:
850
66
.
42.
Stewart
R
,
Hammond
SA
,
Oberst
M
,
Wilkinson
RW
. 
The role of Fc gamma receptors in the activity of immunomodulatory antibodies for cancer
.
J Immunother Cancer
2014
;
2
:
29
.
43.
Paul
WE
. 
Fundamental immunology
.
Philadelphia, PA
:
Lippincott Williams & Wilkins
; 
2008
.
44.
White
AL
,
Chan
HT
,
Roghanian
A
,
French
RR
,
Mockridge
CI
,
Tutt
AL
, et al
Interaction with FcgammaRIIB is critical for the agonistic activity of anti-CD40 monoclonal antibody
.
J Immunol
2011
;
187
:
1754
63
.
45.
Labiano
S
,
Roh
V
,
Godfroid
C
,
Hiou-Feige
A
,
Romero
J
,
Sum
E
, et al
CD40 agonist targeted to fibroblast activation protein α synergizes with radiotherapy in murine HPV-positive head and neck tumors
.
Clin Cancer Res
2021
[Epub ahead of print]
.
46.
Roberts
EW
,
Broz
ML
,
Binnewies
M
,
Headley
MB
,
Nelson
AE
,
Wolf
DM
, et al
Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma
.
Cancer Cell
2016
;
30
:
324
36
.
47.
Acton
SE
,
Astarita
JL
,
Malhotra
D
,
Lukacs-Kornek
V
,
Franz
B
,
Hess
PR
, et al
Podoplanin-rich stromal networks induce dendritic cell motility via activation of the C-type lectin receptor CLEC-2
.
Immunity
2012
;
37
:
276
89
.
48.
Kraman
M
,
Bambrough
PJ
,
Arnold
JN
,
Roberts
EW
,
Magiera
L
,
Jones
JO
, et al
Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha
.
Science
2010
;
330
:
827
30
.
49.
Hoves
S
,
Ooi
CH
,
Wolter
C
,
Sade
H
,
Bissinger
S
,
Schmittnaegel
M
, et al
Rapid activation of tumor-associated macrophages boosts preexisting tumor immunity
.
J Exp Med
2018
;
215
:
859
76
.
50.
Long
KB
,
Gladney
WL
,
Tooker
GM
,
Graham
K
,
Fraietta
JA
,
Beatty
GL
. 
IFNgamma and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma
.
Cancer Discov
2016
;
6
:
400
13
.
51.
O Hara
MH
,
O Reilly
E
,
Rosemarie
M
,
Varadhachary
G
,
Wainberg
Z
,
Ko
A
, et al
Abstract CT004: A phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemcitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarcinoma (PDAC) patients
. In:
Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29–Apr 3; Atlanta, GA
.
Philadelphia (PA)
:
AACR
; 
2019
.
Abstract nr CT004
.
52.
Yasmin-Karim
S
,
Bruck
PT
,
Moreau
M
,
Kunjachan
S
,
Chen
GZ
,
Kumar
R
, et al
Radiation and local anti-CD40 generate an effective in situ vaccine in preclinical models of pancreatic cancer
.
Front Immunol
2018
;
9
:
2030
.