Article Text
Abstract
Introduction Triple-negative breast cancer (TNBC) prognosis is poor. Immunotherapies to enhance the antibody-induced natural killer (NK) cell antitumor activity are emerging for TNBC that is frequently immunogenic. The aspartic protease cathepsin D (cath-D), a tumor cell-associated extracellular protein with protumor activity and a poor prognosis marker in TNBC, is a prime target for antibody-based therapy to induce NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC). This study investigated whether Fc-engineered anti-cath-D antibodies trigger ADCC, their impact on antitumor efficacy and tumor-infiltrating NK cells, and their relevance for combinatory therapy in TNBC.
Methods Cath-D expression and localization in TNBC samples were evaluated by western blotting, immunofluorescence, and immunohistochemistry. The binding of human anti-cath-D F1M1 and Fc-engineered antibody variants, which enhance (F1M1-Fc+) or prevent (F1M1-Fc−) affinity for CD16a, to secreted human and murine cath-D was analyzed by ELISA, and to CD16a by surface plasmon resonance and flow cytometry. NK cell activation was investigated by flow cytometry, and ADCC by lactate dehydrogenase release. The antitumor efficacy of F1M1 Fc-variants was investigated using TNBC cell xenografts in nude mice. NK cell recruitment, activation, and cytotoxic activity were analyzed in MDA-MB-231 cell xenografts by immunophenotyping and RT-qPCR. NK cells were depleted using an anti-asialo GM1 antibody. F1M1-Fc+ antitumor effect was assessed in TNBC patient-derived xenografts (PDXs) and TNBC SUM159 cell xenografts, and in combination with paclitaxel or enzalutamide.
Results Cath-D expression on the TNBC cell surface could be exploited to induce ADCC. F1M1 Fc-variants recognized human and mouse cath-D. F1M1-Fc+ activated NK cells in vitro and induced ADCC against TNBC cells and cancer-associated fibroblasts more efficiently than F1M1. F1M1-Fc− was ineffective. In the MDA-MB-231 cell xenograft model, F1M1-Fc+ displayed higher antitumor activity than F1M1, whereas F1M1-Fc− was less effective, reflecting the importance of Fc-dependent mechanisms in vivo. F1M1-Fc+ triggered tumor-infiltrating NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts. NK cell depletion impaired F1M1-Fc+ antitumor activity, demonstrating their key role. F1M1-Fc+ inhibited growth of SUM159 cell xenografts and two TNBC PDXs. In combination therapy, F1M1-Fc+ improved paclitaxel and enzalutamide therapeutic efficacy without toxicity.
Conclusions F1M1-Fc+ is a promising immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens.
- TNBC
- ADCC
- NK
- human antibody-based therapy
- protease
- paclitaxel
- enzalutamide
Data availability statement
Data are available on reasonable request.
This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/.
Statistics from Altmetric.com
WHAT IS ALREADY KNOWN ON THIS TOPIC
Cathepsin D (cath-D) is a tumor cell-associated extracellular protein with protumor activity, a poor prognosis marker, and a target for antibody-based therapy in triple-negative breast cancer (TNBC).
WHAT THIS STUDY ADDS
This study first shows that cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger antibody-dependent cellular cytotoxicity (ADCC).
The Fc-optimized F1M1-Fc+ antibody (derived from F1M1 by introducing the S239D, H268F, S324T, and I332E mutations to enhance the affinity for CD16a) promotes ADCC induction, improves antitumor potency, and triggers natural killer (NK) cell recruitment, activation and cytotoxic activity in tumors. NK cell depletion impaired F1M1-Fc+ therapeutic efficacy, proving their key role.
F1M1-Fc+ improves paclitaxel and enzalutamide therapeutic efficacy in combination, demonstrating its clinical relevance.
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
F1M1-Fc+ is a promising immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens.
Introduction
Triple-negative breast cancers (TNBCs; 15% of all breast cancers (BCs)) lack estrogen, progesterone and HER2 receptors and are a clinically aggressive BC subtype (highest metastatic potential and recurrence in the first 5 years after diagnosis).1 The prognosis of patients with TNBC is poor, mainly due to disease heterogeneity and lack of targeted therapies. Although conventional chemotherapy remains the standard treatment, immunotherapy is changing the paradigm of anticancer treatment and is emerging as an alternative treatment for TNBC, classified as an immunogenic BC subtype.2 3
Recent studies and clinical trials highlighted that natural killer (NK) cell-based immunotherapy can awake the innate anticancer response, particularly against tumor metastases,4–6 and sustain BC dormancy.7 Hence, different immunotherapeutic strategies are tested to improve the efficacy of antibody-induced NK cell-mediated antitumor activity. Glycoengineering and protein-engineering of the Fc region of tumor-targeting antibodies have been exploited to modulate their interaction with activating or inhibitory members of the FcγR family.8 Afucosylation and selected amino acid substitutions increase the affinity for FcγRs, thus enhancing NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC),8 a major mechanism contributing to the therapeutic efficacy of antibodies.9 Many clinically relevant anticancer antibodies, such as rituximab, cetuximab and trastuzumab, induce NK cell-mediated ADCC in vitro. 6 ,8 NK cells bind to the Fc of antibody-labeled tumor cells, predominantly through a single activating FcγR (FcγRIIIA, also named CD16a), a low-affinity receptor for IgG.10 Binding of IgG to CD16a can activate NK cells and stimulate the release of lytic granules that contain molecules, such as perforin and granzymes11 12 ultimately leading to the target cell lysis. FcγR ligation also affects NK cell survival and proliferation13 and induces the release of cytokines and chemokines that promote the recruitment and activation of tumor-infiltrating immune cells.10
The aspartyl protease cathepsin D (cath-D), a marker of poor prognosis in BC14 and TNBC,15 16 is overexpressed and hypersecreted in the tumor microenvironment17 and exhibits protumor activity.18–26 In TNBC, cath-D is a tumor cell-associated extracellular biomarker and a target for antibody-based therapy.27 We previously showed that F1, a fully human anti-cath-D IgG1 originally isolated by phage display, inhibits growth of TNBC cell xenografts and TNBC patient-derived xenografts (PDX), activates NK cells, and prevents the tumor recruitment of M2-polarized tumor-associated macrophages and myeloid-derived immunosuppressive populations in nude mice.27 We found that an Fc-aglycosylated F1 anti-cath-D antibody, in which the N297A mutation prevents Fcγ receptor (FcγR) binding, no longer induces NK cell activation. This was associated with reduced tumor growth inhibition.27 This demonstrated the essential role of the Fc part of the F1 antibody for anti-cath-D antibody-based therapy through NK cell activation. Therefore, cath-D is a prime target for monoclonal antibody-based therapies to induce NK cell-mediated ADCC.
Here, we showed that cath-D expression on the TNBC cell surface can be exploited to induce ADCC. We applied Fc-engineering to F1M1, a novel fully human anti-cath-D IgG1 antibody in which the variable regions were derived from F1,27 to generate three antibody variants that harbor the wild-type (F1M1) or mutated Fc region to enhance (F1M1-Fc+) or prevent (F1M1-Fc−) binding to CD16a on NK cells. This study explored whether targeted therapy with Fc-engineered anti-cath-D antibodies triggers ADCC, its impact on antitumor efficacy and tumor-infiltrating NK cells, and the relevance of combination therapies with anti-cath-D antibodies in TNBC.
Materials and methods
The materials used, cell lines, conditioned medium, western blotting, ELISA, immunoprecipitation, fluorescence microscopy, immunohistochemistry (IHC), surface plasmon resonance, quantitative RT-qPCR as well as in vivo experiments and study approvals are described in online supplemental file 1.
Supplemental material
Binding of human Fc-engineered anti-cath-D antibodies to human CD16a (hCD16a)
To analyze the anti-cath-D antibody binding to human FcγRIIIA (hCD16a), hCD16a 158V or 158F-transduced NK92 cells (2×104) were incubated in the absence or in the presence of increasing concentrations of the F1M1-Fc−, F1M1, or F1M1-Fc+ antibodies (4°C, 30 min). Then, cells were incubated with APC-conjugated 3G8 (anti-hCD16a antibody; 1:100) (4°C, 30 min), washed twice in phosphate-buffered saline (PBS) at 4°C, and analyzed by flow cytometry. Fluorescence was acquired with a MACSQuant cytometer (Miltenyi) and analyzed with the Kaluza software V.2.1 (Beckman Coulter). Results were expressed as percentage of fluorescent 3G8 binding inhibition: (MFI without anti-cath-D antibody - MFI with anti-cath-D antibody)×100/(MFI without anti-cath-D antibody). This allowed the standardization and comparison between hCD16a 158V- and 158F-transduced NK92 cells.
In vitro stimulation and functional responses of hCD16a-transduced NK92 cells
For in vitro stimulation and functional responses of hCD16a-transduced NK92 cells, NUNC Maxisorp 96-well plates were directly sensitized with F1M1-Fc−, F1M1, F1M1-Fc+, or 3G8 (5 µg/mL) at 4°C for 12 hours or precoated with the M2E8 anti-cath-D antibody in PBS (2500 ng/well), then incubated or not with increasing concentrations of cath-D (3–50 nM), and finally incubated with F1M1-Fc+ (5 µg/mL). After three washes with PBS/Tween-20 solution (PBS-T; 45 µL Tween-20 in 100 mL PBS), plates were saturated with 1% bovine serum albumin for 1 hour, then washed three times with PBS-T. NK92 hCD16a 158V or 158 F cells (1×105) in 100 µL RPMI were then incubated on non-sensitized or sensitized plates in the presence of the anti-CD107a-PC5 antibody (1:20) and 0.1 µg/mL BD GolgiPlug containing brefeldin A (BD Biosciences) at 37°C in 5% CO2 humidified air for 4 hours. Cells were then fixed and permeabilized using the BD Cytofix/Cytoperm Plus Kit (BD Biosciences) and stained with an anti-IFNγ-PE antibody (1:20) at 4°C for 30 min to detect intracellular IFNγ. The proportions of responding (degranulating (CD107a+), IFNγ-producing (IFNγ+) NK cells and cells with both responses (CD107a+ IFNγ+) were analyzed by flow cytometry with a Cytoflex S cytometer and the Kaluza V.2.1 software (Beckman Coulter).
NK cell-mediated ADCC toward TNBC cells and cancer-associated fibroblasts
MDA-MB-231 cells (1×104 target; T) or human breast cancer-associated fibroblasts (hCAF1) cells (2×104 target; T) were plated in 96-well plates. After 24 hours, they were first incubated with F1M1-Fc+, F1M1, F1M1-Fc− or cetuximab (positive control) at 37°C for 30 min. Then, cells were incubated with NK92 hCD16a 158V or 158F at an effector:target (E:T) ratio of 10:1, or human primary expanded NK cells (effector; E) at an E:T ratio of 3:1 at 37°C for 24 hours. MDA-MB-231 cell lysis (ADCC) or hCAF1 cell lysis was evaluated by measuring lactate dehydrogenase (LDH) release with the Cytotox 96 Non-radioactive Cytotoxicity Assay (Promega). The percentage of specific lysis in each sample was determined using the following formula: percentage of specific lysis=(sample LDH release–[E cell+T cell] spontaneous LDH release)/(T cell maximum LDH release -T cell spontaneous LDH release)×100. For ADCC in cell spheroids, target MDA-MB-231 cells (5×103) were seeded on Ultra-Low Attachment 96-well plates (#7007, Corning). After 72 hours, MDA-MB-231 cell spheroids were first incubated with F1M1-Fc+, F1M1-Fc−, or cetuximab at 37°C for 30 min, and then with effector NK92 hCD16a+ 158V cells (E:T ratio of 20:1) for 24 hours. ADCC was assessed using the Cytotox 96 Non-radioactive Cytotoxicity Assay (Promega). The percentage of specific lysis in each sample was determined as described above.
In vivo isolation and immunophenotyping of tumor-infiltrating NK cells
Tumors were shredded and digested with a mixture of collagenase IV (1 mg/mL) and DNase I (200 U/mL) in RPMI at 37°C. The mixture was transferred into a gentleMACS C tube for enzymatic and mechanical digestion using a gentleMACS Octo Dissociator (Miltenyi Biotec) at 37°C. After digestion, tumor suspensions were passed through a 70 µm nylon cell strainer (#22-363-548, Thermo Fisher Scientific), centrifuged and resuspended in FACS Buffer (PBS pH 7.2, 1% decomplemented fetal calf serum, 2 mM EDTA, and 0.02% sodium azide). Cells were blocked with FACS Buffer containing 1% (v/v) of mouse Fc block for 30 min and stained with fluorescent-conjugated antibodies against the cell surface markers CD45, CD3, CD19, CD11c, F4/80, NKp46, CD27, CD11b, and CD107a for 45 min. Then, cells were fixed, permeabilized and stained with fluorescent-conjugated antibodies against the intracellular marker granzyme B overnight. After fixation with 1% paraformaldehyde in PBS, cell samples were analyzed by flow cytometry using a Beckman and Coulter Cytoflex flow cytometer and FlowJo V.10.8.1. Living immune cells were defined as Viakrome IR808− CD45+. NK cells were defined as NKp46+ cells within the gate excluding CD3+ T and NKT cells, CD19+ B cells, F4/80+ macrophages, and CD11c+ dendritic cells. NK cell maturation stage was analyzed with the CD27 and CD11b cell surface markers. Functional markers (CD107a, granzyme B) were studied in the NK cell subpopulations.
Statistical analysis
A linear mixed regression model was used to determine the relationship between tumor growth and number of days postxenograft. The variables included in the fixed part of the model were: number of days postxenograft and treatment groups. Their interaction also was evaluated. Random intercepts and random slopes were included to take into account the time effect. The model coefficients were estimated by maximum likelihood. Survival rates were estimated using the Kaplan-Meier method and compared with the log-rank test. Comparisons of more than two groups were done using the non-parametric Kruskal-Wallis test; the Mann-Whitney test was used for pairwise comparisons. Statistical significance was set at the 0.05 level. All statistical analyses were done with STATA V.16.0.
Results
In TNBC, cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger ADCC
ADCC initiation requires the presence of antibody-antigen complexes at the cell surface. Cath-D localizes at the cell surface through its binding to the M6P/IGF2 receptor via its mannose-6-phosphate (M6P) motif (BC cells and stromal fibroblasts)28 29 and to LRP1 (fibroblasts).23 We first examined the expression of cath-D, M6P/IGF2 receptor and LRP1 in a panel of TNBC cell lines (MDA-MB-231, SUM-159, MDA-MB-436, MDA-MB-453, BT-549) and hCAF1 cells (figure 1A). We detected the fully mature cellular lysosomal protease cath-D (34 kDa heavy chain) and the IGF2/M6P receptor in all TNBC cell lines and hCAF1s. We could not detect cath-D only in BT549 cells. Similarly, we did not detect LRP1 expression in TNBC cells, except for SUM159 and BT549 cells. Conversely, it was strongly expressed in hCAF1 cells. The 52-kDa cath-D precursor (pro-cath-D) was secreted in the culture medium of all TNBC cell lines and of hCAFs and was detectable in cell lysates, in agreement with its cell surface association after secretion (figure 1A). Moreover, we detected the 52-kDa cath-D precursor and IGF2/M6P receptor in the cytosol (ie, whole tumor lysate) of eight primary TNBC samples and in MDA-MB-231 and SUM-159 cell xenografts (figure 1B). Immunofluorescence analysis of cath-D in MDA-MB-231 cell xenografts showed intense, punctate staining at the tumor periphery and in cell membrane protrusions, potentially in areas of exocytosis (figure 1C), reflecting cath-D expression profile in TNBC biopsy samples (online supplemental figure 1). This is in agreement with recent IHC results showing membrane-associated cath-D expression in 85.7% of TNBC samples in a tissue microarray.27 These observations strongly suggest that cath-D is a tumor microenvironment antigen eligible for Fc-engineered antibody targeted therapy to trigger ADCC.
Supplemental material
Generation of anti-cath-D F1M1 Fc-variant antibodies with different CD16a binding properties
We next evaluated whether manipulation of the Fc portion of F1, our lead anti-cath-D antibody, by protein-engineering to increase ADCC could potentiate its antitumor effectiveness in TNBC. We first improved F1 format by introducing two-point mutations (one in VL CDR3 and one in VH FR3) to abrogate N-glycosylation at these two sites (F1M1 antibody) that may lead to the production of glycoforms associated with the risk of immunogenic responses in patients, as described for cetuximab.30 The migration profiles of F1 and F1M1 in the IgG1 format showed that the light and heavy chains molecular weights were lower in F1M1 than F1, as expected due to reduced glycosylation (online supplemental figure 2A). Sandwich ELISA using human cath-D secreted from MDA-MB-231 cells showed that F1M1 (online supplemental figure 2B) had good binding capacities (0.04 nM), as previously shown for F1 (0.2 nM).27 Sandwich ELISA using cath-D secreted from mouse E0771 TNBC cells showed that F1M1 and F1 (online supplemental figure 2C) retained good binding capacities also toward mouse cath-D (EC50=0.9 nM and 26.3 nM, respectively). The lower EC50 of F1M1 indicated that the two-point mutations leading to Fab aglycosylation enhanced F1M1 capacity to recognize human and mouse cath-D, presumably because the VL-CDR3 glycosylation decreased F1 affinity due to steric hindrance. To compare the antitumor efficacy of F1M1 and F1, we xenografted athymic Foxn1nu nude mice with MDA-MB-231 cells. When tumor volume reached 50 mm3, we treated mice with F1M1, F1, or negative isotype control (rituximab) (15 mg/kg) by ip three times per week for 35 days (days 21–56 postgraft), and then sacrificed all mice at treatment end. Both F1M1 and F1 significantly delayed tumor growth compared with negative control (online supplemental figure 2D; p=0.003 for F1M1, p<0.001 for F1). Thus, the VH and VL aglycosylated F1M1 antibody appears to be a suitable candidate for future clinical development.
We then designed an Fc-optimized F1M1 (F1M1-Fc+), in which the S239D, H268F, S324T, I332E mutations should increase the binding affinity to CD16a (FcγRIIIA) to enhance NK cell activation, and a F1M1 Fc-silent (F1M1-Fc−) in which the L234A, L235A and P329G mutations prevented binding to FcγRs8 and ADCC induction. F1M1-Fc− activities should rely exclusively on Fab-mediated effector functions. Antibody preparations were highly pure, and after SDS-PAGE in reducing conditions, the heavy and light chains were of the expected molecular mass (50 and 25 kDa respectively) (online supplemental figure 3A). The three F1M1 antibody variants showed almost identical, concentration-dependent binding to secreted cath-D with EC50 values in the low nanomolar range (EC50=0.07 nM, 0.051 nM, and 0.048 nM for F1M1, F1M1-Fc− and F1M1-Fc+, respectively) (figure 1D). Moreover, F1M1-Fc+ binding to pro-cath-D was comparable at pH values from 7.2 to 5.6 (figure 1E). All F1M1 variant antibodies immunoprecipitated secreted cath-D at pH values from 7.5 to 5.5 (online supplemental filgure 3B), suggesting that the highly acidic tumor microenvironment does not inhibit cath-D recognition and binding by antibodies. Then, we investigated by surface plasmon resonance F1M1-Fc+, F1M1 and F1M1-Fc− binding to hCD16a (158V or 158F allotype). F1M1 binding to hCD16a 158V was 3.9-fold higher than to hCD16a 158F (KD=42 nM and KD 164 nM, respectively) (figure 1F). Compared with F1M1, F1M1-Fc+ binding to hCD16a 158V was increased by 21.2-fold (KD=2 nM for F1M1-Fc+ and 42.4 nM for F1M1), and even more to hCD16a 158F (by 54.6-fold) (KD=3 nM for F1M1-Fc+, and 164 nM F1M1). As F1M1-Fc− showed no detectable binding to both hCD16a allotypes, we used it as control devoid of effector function. Finally, we investigated by flow cytometry F1M1-Fc+, F1M1 and F1M1-Fc− binding to hCD16a (158V or 158F allotype) expressed on the surface of human NK92 cells. It has been shown that the hCD16a 158 V/F polymorphism in NK cells influences its binding to human IgG1.31 Specifically, hCD16a V158 displays increased affinity for human IgG1, resulting in increased NK cell activation. F1M1 binding to hCD16a 158V was approximately fivefold higher than to hCD16a 158F at all tested concentrations (figure 1G). Compared with F1M1, F1M1-Fc+ binding to hCD16a 158V was increased (2–3 fold), and even more to hCD16a 158F (7–20 fold).
F1M1-Fc+ increases NK cell activation in vitro, and ADCC of TNBC cells and CAFs
We next compared F1M1-Fc+, F1M1 and F1M1-Fc− in vitro capacity to activate NK cells by analyzing cell surface CD107a expression, as a functional marker of NK cell degranulation, and intracellular IFNγ, a cytokine secreted following NK cell activation,32 in hCD16a-transduced human NK92 cell line. The percentages of CD107a+ NK, IFNγ+ NK and CD107a+ IFNγ+ NK subsets were increased similarly by 2–3 fold following incubation with F1M1-Fc+, compared with F1M1, in hCD16a 158V-expressing or 158F-expressing NK cells (figure 2A). Conversely, F1M1-Fc− did not activate NK effector functions. Moreover, the percentages of CD107a+ NK, IFNγ+ NK and CD107a+ IFNγ+ NK cells were increased in a dose-dependent manner in the presence of increasing concentrations of cath-D bound to F1M1-Fc+ (figure 2B). Overall, F1M1-Fc+ was the most potent antibody to activate NK cell response. Next, we determined whether the F1M1-Fc+-enhanced binding to hCD16a-expressing NK cells translated into increased cytotoxic activity. First, incubation of MDA-MB-231 cells with F1M1-Fc+ significantly increased ADCC by 5-fold (p<0.0001) and 18-fold (p=0.0002), compared with F1M1, in the presence of NK92-hCD16a 158 V cells and NK92-hCD16a 158 F cells, respectively (figure 2C). F1M1-Fc− did not induce ADCC. Moreover, F1M1-Fc+ triggered ADCC in a dose-dependent manner in the presence of NK92-hCD16a 158V cells (figure 2D). F1M1-Fc+-induced ADCC was exclusively dependent on Fc binding to the hCD16 receptor on NK92-hCD16a 158V cells, because addition of a Fc block completely inhibited ADCC, to a level similar to that observed with F1M1-Fc− (figure 2E). We then evaluated ADCC against MDA-MB-231 cells using primary human NK cells (158V/F allotype) (online supplemental figure 4A). ADCC was significantly increased by 3.5-fold on incubation with F1M1-Fc+, compared with F1M1 (p<0.0001) (online supplemental figure 4B).
As cath-D localizes at the surface of BC cells and stromal fibroblasts by binding to the M6P/IGF2 receptor,28 we assessed whether M6P/IGF2 receptor-bound cath-D was involved in ADCC induction by F1M1-Fc+. Thus, we evaluated ADCC against MDA-MB-231 cells in the presence of NK92-hCD16a 158 V cells and of excess M6P to compete with cath-D binding to the M6P/IGF2 receptor (figure 2F). Addition of M6P, but not glucose-6-phosphate (negative control for M6P), significantly inhibited ADCC induction by F1M1-Fc+ by 51% (p<0.0001), indicating that cath-D bound to its M6P/IGF2 receptor at the cell surface was involved in ADCC. Similarly, using primary human NK cells (158 V/V allotype) (online supplemental figure 4C), ADCC of MDA-MB-231 cells was inhibited by 70% (p<0.0001) in the presence of M6P (online supplemental figure 4D). Moreover, F1M1-Fc+ efficiently triggered ADCC against hCAF1 cells in the presence of NK92-hCD16a 158 V cells, and excess M6P significantly inhibited ADCC by 62% (p<0.0001) (figure 2G). Lastly, to better recapitulate the in vivo cancer cell environment, we prepared spheroids of MDA-MB-231 cells co-cultured with NK92 hCD16a 158V cells. F1M1-Fc+ also induced ADCC of MDA-MB-231 cell spheroids (figure 2H). Live imaging of ADCC in calcein-labeled MDA-MB-231 cell spheroids showed their time-dependent lysis in response to F1M1-Fc+ exposure (online supplemental figure 5A,B). At 24 hours, the fluorescence signal of calcein-labeled MDA-MB-231 spheroids was significantly decreased by 29.5% in response to F1M1-Fc+ compared with F1M1-Fc− (p=0.005) (online supplemental figure 5C). Overall, our results demonstrated that F1M1-Fc+ is the most potent anti-cath-D antibody to trigger ADCC in TNBC cells and CAFs.
F1M1-Fc+ is the best candidate to reduce MDA-MB-231 cell xenograft growth and improve mouse survival
To compare the in vivo antitumor efficacy of F1M1-Fc+, F1M1, and F1M1-Fc−, we used athymic Foxn1nu nude mice subcutaneously xenografted with MDA-MB-231 cells. When MDA-MB-231 tumors reached 50 mm3, we treated mice with F1M1-Fc+, F1M1, F1M1-Fc−, or the anti-human CD20 IgG1 rituximab, as negative isotype control (Ctrl) (15 mg/kg) by ip three times per week for 35 days (days 13–48 postgraft), and sacrificed mice when tumor volume reached 2000 mm3. Both F1M1-Fc+ and F1M1 significantly delayed tumor growth compared with negative control (p=0.001 for F1M1-Fc+, p=0.011 for F1M1). F1M1-Fc− slightly delayed tumor growth (p=0.057) (figure 3A). F1M1-Fc+ tended to be more effective than F1M1-Fc− (p=0.055). At day 48 (treatment end), tumor volume was significantly reduced by 77% (p=0.0002) in the F1M1-Fc+ group, by 60% (p=0.0007) in the F1M1 group, and by 40.5% (p=0.0289) in the F1M1-Fc− group compared with control (figure 3B). Tumor volume was significantly smaller in the F1M1-Fc+ group than in the F1M1-Fc− group (p=0.0041). The overall survival rate, reflected by a tumor volume <2000 mm3, was significantly longer in the F1M1-Fc+, F1M1, and F1M1-Fc− groups than in controls. The median survival was 59, 57, 54.5, and 50 days in the F1M1-Fc+, F1M1, F1M1-Fc−, and control groups, respectively (figure 3C; Kaplan-Meier survival analysis, p=0.0003 for F1M1-Fc+, p=0.0011 for F1M1, p=0.0275 for F1M1-Fc− compared with control). The overall survival rate was significantly longer in mice treated with F1M1-Fc+ than with F1M1-Fc− (p=0.0253). Moreover, all treated mice gained weight (figure 3D) and displayed normal activities, suggesting no apparent toxicity. Overall, the results suggest that F1M1-Fc+ is the anti-cath-D antibody with the best antitumor activity.
F1M1-Fc+ triggers in vivo recruitment, activation, and cytotoxic activity of NK cells to induce ADCC in MDA-MB-231 TNBC cell xenografts
To investigate the in vivo mechanisms underlying the antitumor effect of F1M1-Fc+, F1M1, and F1M1-Fc−, we treated nude mice xenografted with MDA-MB-231 cells with F1M1-Fc+, F1M1, F1M1-Fc−, or rituximab as negative isotype control (same schedule as before), and then sacrificed all mice at treatment end (day 48). F1M1-Fc+, F1M1, and F1M1-Fc− inhibited tumor growth compared with rituximab, but only F1M1-Fc+ resulted in significant tumor growth inhibition compared with rituximab (p=0.003) (figure 4A). At day 48, tumor volume was significantly reduced by 60.3% (p=0.0005) in the F1M1-Fc+ group, by 42.9% (p=0.02) in the F1M1 group, and by 36.6% (p=0.0368) in the F1M1-Fc− group compared with control group (figure 4B,C). Tumor volume was significantly more reduced in the F1M1-Fc+ group than in the F1M1-Fc− group (p=0.0368). Human cath-D expression level in tumors was not affected by F1M1-Fc+, F1M1, or F1M1-Fc− treatment, as shown by IHC and western blot analyses (online supplemental figure 6A–C). To study the impact of the Fc part of F1M1 on NK cell recruitment and activation, we identified and quantified by flow cytometry tumor-infiltrating NK cells in MDA-MB-231 xenografts after treatment end (day 48). The overall percentage of living cells was not different between untreated (control) and treated groups (online supplemental figure 7). The percentage of NK (CD45+ F4/80- CD3- CD19- CD11c- NKp46+) cells within the living immune CD45+ cell population was significantly increased by 207.2% in the F1M1-Fc+ group compared with control (p=0.0053), but not in the F1M1 and F1M1-Fc− groups (figure 4D). Linear regression analysis showed that the NK cell percentage was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.2139, p=0.0062) (figure 4E). This suggested that in the F1M1-Fc+ group, NK cell recruitment in the tumor contributed to the antitumor response.
We also determined the NK cell phenotype based on the expression of the NK cell surface maturation markers CD11b and CD27.33 The immature CD27−CD11b− NK cell population was significantly increased by 268% in F1M1-Fc+-treated animals (p=0.0016 compared with the control group), but not in the F1M1-treated and F1M1-Fc−-treated groups (figure 4F). The CD27+CD11b− NK cell population (mature stage 1) also was increased in the F1M1-Fc+ group by 206.9% (p=0.0360 compared with the control group) (figure 4F). This suggests that F1M1-Fc+ treatment promoted the recruitment of immature NK cells and at an early stage of the maturation process. NK cell development and maturation is orchestrated by a network of transcription factors, including Eomes.34 Eomes is mainly expressed in immature NK cells, promotes survival of maturing NK cells, and plays a major role in the induction of genes associated with NK cell cytotoxicity, such as Prf1.35 RT-qPCR analysis of tumor samples showed that Eomes expression was significantly upregulated by 479.3% in the F1M1-Fc+ group (p=0.0006 compared with control), but not in the F1M1 and F1M1-Fc− groups (figure 4G). This strongly suggested that F1M1-Fc+-based therapy triggers the recruitment of immature NK cells that might become cytotoxic in the MDA-MB-231 TNBC model.
Next, we analyzed cell surface expression of CD107a, as a functional marker of NK cell degranulation, by flow cytometry in tumor-infiltrating NKp46+ cells in MDA-MB-231 cell xenografts at day 48. The percentage of CD107a+ NK cells within the NK cell population was significantly increased by 142% in F1M1-Fc+-treated animals (p=0.0464 compared with control), but not in the F1M1 and F1M1-Fc− groups (figure 4H). The percentage of CD107a+ NK cells was higher (by 142.6%) in the F1M1-Fc+ group compared with the F1M1-Fc− group (p=0.0079). Linear regression analysis showed that the percentage of CD107a+ NK cells was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.1703, p=0.0211) (online supplemental figure 8A). This suggested that in the F1M1-Fc+ group, in vivo tumor infiltration by activated CD107a+ NK cells may contribute to the antitumor response.
Lastly, we analyzed intracellular granzyme B (GZMB) expression as a marker of NK cell cytotoxic activity by flow cytometry in tumor-infiltrating NKp46+ cells in MDA-MB-231 cell xenografts at day 48. The percentage of GZMB+ NK cells was significantly increased by 173.8% (p=0.0079) and by 185.4% (p=0.0289) in the F1M1-Fc+ and F1M1 groups, respectively, compared with control, but not in the F1M1-Fc− group (figure 4I). It also was higher (by 193%) in the F1M1-Fc+ group than in the F1M1-Fc− group (p=0.0418). Linear regression analysis showed that the percentage of GZMB+ NK cells was inversely correlated with tumor volume in all animals (four treatment groups together) (R2=0.3085, p=0.0012) (online supplemental figure 8B). This suggested that in the F1M1-Fc+ and F1M1 groups, tumor infiltration by NK cells with cytotoxic activity contributed to the antitumor response.
In agreement, granzyme B (Gzmb) and perforin (Prf1) mRNA levels, as a read-out of NK cell cytotoxic activity, were upregulated (up to 166.3% and 173.8%, respectively) in the F1M1-Fc+ group compared with control group (p=0.3969 and p=0.2319), and also compared with the F1M1-Fc− group (up to 256.3% and 243.4%; p=0.0207 and p=0.0379, respectively) (online supplemental figure 9A,B). The antitumor cytokine Tnf, secreted by activated NK cells, was also upregulated by 171.5% (p=0.0059) in the F1M1-Fc+ group and by 160.3% (p=0.0541) in the F1M1 group compared with control (online supplemental figure 9C). Tnf was upregulated (by 266.8%) (p=0.003) in the F1M1-Fc+ group compared with the F1M1-Fc− group. Altogether, these results demonstrated that F1M1-Fc+ triggers NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts, and strongly suggest the key role of the Fc part of this anti-cath-D antibody in its antitumor activity via NK cells.
NK cell depletion impairs F1M1-Fc+ therapeutic efficacy in MDA-MB-231 cell xenografts
The significant antitumor effects of F1M1-Fc+ and the F1M1-Fc+-mediated induction of NK cell recruitment, activation and cytotoxic activity strongly suggested that NK cells are crucial for F1M1-Fc+-antitumor activity. To confirm this hypothesis, we depleted NK cells in MDA-MB-231-bearing nude mice by intraperitoneal injection of an anti-asialo-GM1 antibody (αGM1). First, to determine the efficiency of NK cell depletion, we treated non-grafted nude mice with control (saline solution) or αGM1 at day 0 and day 3 (online supplemental figure 10A). Analysis of blood samples at day 4 and day 7 confirmed the efficacy of NK cell depletion, even 4 days (ie, day 7) after the last αGM1 injection (online supplemental figure 10B,C). Other immune cells (B cells, neutrophils, dendritic cells and macrophages) were not affected by αGM1 treatment, as indicated by their quantification in blood and spleen (online supplemental figure 10D,E).
Then, to investigate NK cell role in the antitumor effect of F1M1-Fc+, we treated nude mice harboring MDA-MB-231 cell xenografts with F1M1-Fc+ or rituximab (negative isotype control) in the presence or absence of αGM1, and then sacrificed all mice at treatment end (day 48), according to the schedule described in figure 5A. As previously observed, F1M1-Fc+ significantly inhibited tumor growth compared with control (p<0.001) (figure 5B). In the presence of αGM1, F1M1-Fc+ antitumor activity (tumor growth reduction) was decreased (p=0.021 vs control, and p=0.015 vs F1M1-Fc+ alone), revealing the crucial role of NK cells (figure 5B). At day 48, compared with the control group, tumor volume was significantly reduced by 63.3% (p=0.0003) in the F1M1-Fc+ group, but only by 30.7% (p=0.0881) in the F1M1-Fc++αGM1 group (figure 5C). Moreover, tumor volume was significantly smaller in the F1M1-Fc+ group than in the F1M1-Fc++αGM1 group (p=0.0281) (figure 5C). The number of tumor-infiltrating NK cells (CD45+ CD3− CD19− Ly6G− NKp46+) within the living immune CD45+ cell population in tumors was significantly increased by 248.7% in the F1M1-Fc+ group compared with control (p=0.0312). Conversely, very few NK cells were detected in the F1M1-Fc++αGM1 group (figure 5D). Analysis of blood samples at day 45 and of the spleens at day 48 validated NK cell depletion in the F1M1-Fc++αGM1 group (online supplemental figure 11). This demonstrated the key role of NK cells in F1M1-Fc+ therapeutic efficiency.
Drug-induced neutropenia is a potentially serious and life-threatening adverse event that may occur after therapy with various agents, including antibodies (eg, immune checkpoint inhibitors). Analysis by flow cytometry of blood from mice from the control and F1M1-Fc+ treated groups (from figure 5) showed that neutrophil count at day 45 was not affected, indicating the absence of neutropenia (online supplemental figure 12A). Similarly, no sign of leukopenia, thrombopenia, and anemia was observed after F1M1-Fc+ treatment, suggesting the absence of toxicity (online supplemental figure 12B).
F1M1-Fc+ inhibits tumor growth of patient-derived TNBC xenografts and improves mouse survival
Next, we tested F1M1-Fc+ therapeutic effect in mice harboring TNBC-PDXs (B1995 and B3977).36 First, we analyzed the expression of cath-D and the M6P/IGF2 receptor in PDX B1995 and B3977 xenografts (the two fastest growing PDXs in nude mice among a PDX collection),36 and in MDA-MB-231 and SUM-159 cell xenografts by western blot analysis. Cath-D and the M6P/IGF2 receptor were similarly expressed in all samples (figure 6A). Immunostaining of the PDX B1995 and B3977 confirmed that cath-D was expressed in tumor cells and in the microenvironment (figure 6B), as previously observed in TNBC biopsy samples (online supplemental figure 1). These results indicated these PDXs are representative of the disease, at least concerning cath-D expression. We then xenografted Swiss nude mice with the PDX B1995 or B3977. Tumor growth was significantly reduced by F1M1-Fc+ (15 mg/kg by ip three times per week) compared with control (rituximab) in both models (p<0.001) (figure 6C,D, left panel). At treatment end (day 48 for the PDX B1995 and day 42 for the PDX B3977), tumor volume was significantly reduced by 61.8% (p=0.0002) and by 44.6% (p=0.0018), respectively, in the F1M1-Fc+ group compared with the control group (figure 6C,D, middle panels). The overall survival rate, reflected by a tumor volume <1500 mm3, was significantly longer in mice treated with F1M1-Fc+ than in control group (median survival of 64 and 49 days for the F1M1-Fc+ group and for control animals in mice xenografted with PDX B1995, and of 57 and 46 days for the F1M1-Fc+ group and for control animals in mice xenografted with PDX B3977) (figure 6C,D, right panel; Kaplan-Meier survival analysis, p=0.0002 and p=0.0024, respectively). These results showed that F1M1-Fc+ monotherapy very efficiently delayed tumor growth in nude mice xenografted with TNBC PDXs.
F1M1-Fc+ improves paclitaxel therapeutic efficacy
Next, we investigated the therapeutic benefit of combining F1M1-Fc+ with paclitaxel. First, we confirmed paclitaxel cytotoxicity in MDA-MB-231 cells in vitro (IC50=53.4 nM) (online supplemental figure 13A). Then, we showed that incubation of MDA-MB-231 cells with 5 nM paclitaxel for 48 hours (dose equivalent to the IC20; online supplemental figure 13A) did not affect cath-D expression (cell lysates) and secretion (conditioned media) (online supplemental figure 13B). As paclitaxel is frequently administered to patients with a weekly schedule, we treated nude mice bearing MDA-MB-231 cell xenografts with 1 mg/kg, 4 mg/kg, or 7 mg/kg paclitaxel (PTX) weekly. Higher PTX doses resulted in excessive toxicity (data not shown). PTX induced tumor growth inhibition in a dose-dependent manner (online supplemental figure 13C) that was significant at 4 mg/kg (p=0.007) and 7 mg/kg (p<0.001) compared with control (NaCl).
We then tested the combination therapy (paclitaxel and F1M1-Fc+) in nude mice bearing MDA-MB-231 cell xenografts using a low dose (PTX LD, 1 mg/kg) and a medium dose of paclitaxel (PTX MD, 4 mg/kg) that inhibited tumor growth on its own; online supplemental figure 13C). When tumor volume reached 50 mm3 at day 15 postgraft, we treated mice with F1M1-Fc+ (15 mg/kg; three times per week), PTX LD (1 mg/kg; once per week), F1M1-Fc+ (15 mg/kg; three times per week)+PTX LD (1 mg/kg; once per week), or control (rituximab, three times per week+saline, once per week; all ip) for 37 days. Both F1M1-Fc+ and PTX LD+F1M1-Fc+ significantly delayed tumor growth compared with control (p=0.039 for F1M1-Fc+, p=0.032 for PTX LD+F1M1-Fc+) (figure 7A). At day 52 (treatment end), tumor volume was reduced by 37.4% (p=0.07) in the F1M1-Fc+ group, by 18.7% (p=0.0005) in the PTX LD group, and by 50.5% (p=0.002) in the PTX LD+F1M1-Fc+ group compared with control group (figure 7B). Tumor volume was significantly smaller in the PTX LD+F1M1-Fc+ group than in the PTX LD group (p=0.0293). The overall survival rate, reflected by a tumor volume <2000 mm3, was significantly longer in mice treated with F1M1-Fc+ and with PTX LD+F1M1-Fc+ than in controls (median survival of 59 and 66 days for the F1M1-Fc+ and PTX LD+F1M1-Fc+ groups, respectively, compared with 55 days for control animals) (figure 7C; Kaplan-Meier survival analysis, p=0.0313 for F1M1-Fc+, p=0.0009 for PTX LD+F1M1-Fc+). The overall survival rate was significantly longer in mice treated with PTX LD+F1M1-Fc+ (66 days) than in mice treated with PTX LD (57 days) (p=0.0219). Overall, these results demonstrated that in combination therapy, F1M1-Fc+ improved the therapeutic efficacy of a suboptimal dose of paclitaxel that was poorly effective in vivo (online supplemental figure 13C).
Similarly, F1M1-Fc+, PTX MD, and PTX MD+F1M1-Fc+ significantly delayed tumor growth compared with control (p=0.039 for F1M1-Fc+, p=0.042 for PTX MD, p=0.013 for PTX MD+F1M1-Fc+) (figure 7D). At day 52 (treatment end), tumor volume was reduced by 37.4% (p=0.07) in the F1M1-Fc+ group, by 32.9% (p=0.0289) in the PTX MD group, and by 55.5% (p=0.0012) in the PTX MD+F1M1-Fc+ group compared with control (figure 7E). The overall survival rate was significantly longer in mice treated with F1M1-Fc+, PTX MD and PTX MD+F1M1-Fc+ than in control (median survival of 59, 59 and 69 days for the F1M1-Fc+, PTX MD, and PTX MD+F1M1-Fc+ groups, respectively, compared with 55 days for control animals) (figure 7F; Kaplan-Meier survival analysis, p=0.0313 for F1M1-Fc+, p=0.0312 for PTX MD, p=0.001 for PTX MD+F1M1-Fc+). The overall survival rate was significantly longer in mice treated with PTX MD+F1M1-Fc+ (69 days) than with PTX MD (59 days) (p=0.041). All mice treated with the combination therapy gained weight (figure 7G) and displayed normal activities, suggesting no apparent toxicity. Overall, these results demonstrated that F1M1-Fc+ improved PTX therapeutic efficacy without increasing toxicity.
F1M1-Fc+ improves the therapeutic efficacy of the antiandrogen enzalutamide
We previously found that coexpression of cath-D and androgen receptor (AR) in non-metastatic TNBC is an independent prognostic factor of worse overall survival, suggesting the possibility of adjuvant combination therapy with anti-androgen drugs and anti-cath-D antibodies.16 We first showed that in the AR-expressing SUM159 TNBC cell line (online supplemental figure 14A), incubation with enzalutamide, an antiandrogen drug, did not decrease cath-D expression and secretion (online supplemental figure 14B), and incubation with F1M1-Fc+ did not reduce AR expression (online supplemental figure 14C).
Then, we treated mice harboring SUM159 cell xenografts with F1M1-Fc+ alone. Tumor volume increase was significantly slowed down in mice treated with F1M1-Fc+ (15 mg/kg by ip three times per week) compared with control (rituximab) (p<0.001) (figure 8A). Tumor volume at day 48 (treatment end) was significantly reduced by 71.9% in the F1M1-Fc+ compared with control group (p<0.0001) (figure 8B). The overall survival rate, reflected by a tumor volume <1000 mm3, was significantly longer in mice treated with F1M1-Fc+ than in the control group (median survival of 62 days and 55 days, respectively) (figure 8C; Kaplan-Meier survival analysis, p=0.0024), without any apparent toxicity (figure 8D).
We then treated nude mice bearing SUM159 cell xenografts with F1M1-Fc+ (15 mg/kg, twice per week, ip) or/and enzalutamide (Enza, 30 mg/kg, five times per week, per os). F1M1-Fc+ and Enza+F1M1-Fc+ significantly delayed tumor growth compared with control (p=0.001 for F1M1-Fc+, p=0.003 for Enza+F1M1-Fc+) (figure 8E). Tumor growth was significantly more reduced in the Enza+F1M1-Fc+ group than in the Enza group (p=0.008). At day 50 (treatment end), tumor volume was reduced by 55.7% (p=0.0019) in the F1M1-Fc+ group, by 43.4% (p=0.02) in the Enza group, and by 71% (p=0.0002) in the Enza+F1M1-Fc+ group compared with control (figure 8F). Tumor volume was significantly smaller in the Enza+F1M1-Fc+ group than in the Enza group (p=0.0082). Overall survival was significantly longer in mice treated with F1M1-Fc+, Enza, and Enza+F1M1-Fc+ than in controls (median survival of 63, 61, 64, and 57 days, respectively) (figure 8G; Kaplan-Meier survival analysis, p=0.0126 for F1M1-Fc+, p=0.0196 for Enza, p<0.0001 for Enza+F1M1-Fc+), without apparent toxicity (figure 8H). Overall survival was significantly longer in the Enza+F1M1-Fc+ group than in the Enza group (64 vs 61 days, p=0.017) and in the F1M1-Fc+ group (64 vs 63 days) (p=0.0091) (figure 8G). Overall, these results demonstrated that F1M1-Fc+ improves enzalutamide therapeutic efficacy.
Discussion
Here, we showed that F1M1-Fc+, a novel human Fc-engineered anti-cath-D antibody, enhanced ADCC and tumor growth inhibition, and triggered NK cell recruitment, activation, and cytotoxic activity in the MDA-MB-231 cell xenograft TNBC model. In addition, NK cell depletion impaired F1M1-Fc+ therapeutic efficacy in this model. Furthermore, in combination therapy, F1M1-Fc+ increased the efficacy of chemotherapy (paclitaxel) and antiandrogens (enzalutamide). These data suggest that F1M1-Fc+ may represent a first-in-class treatment for patients with TNBC.
Numerous afucosylated antibodies and Fc-protein engineered antibodies are in clinical trials. Two afucosylated antibodies, obinutuzumab (against CD20) and mogamulizumab (against CCR4), and two Fc-optimized antibodies, tafasitamab (against CD19) and margetuximab (against HER2) that harbor mutations (S329D, I332E and F243L, R292P, Y300L, V305I, P396L, respectively) to increase ADCC, are approved for clinical use.6 8 Here, to improve the NK cell-mediated effector functions of F1M1, an anti-cath-D antibody that binds to both human and mouse cath-D in vitro and exhibits antitumor activity in vivo without apparent toxicity, we generated the F1M1-Fc+ antibody with four mutations (S239D, H268F, S324T, I332E) in the Fc domain to increase its affinity for FcγRIIIA (CD16a) and enhance NK cell-mediated ADCC.37 These four mutations substantially improved F1M1-Fc+ binding to CD16a 158V and 158F expressed in human NK92 cells compared with F1M1. Conversely, F1M1-Fc− (L234A, L235A, and P329G substitutions) showed no detectable binding to CD16a 158V or CD16a 158F.38 Many clinically relevant anti-cancer antibodies, such as rituximab, trastuzumab and cetuximab, induce NK cell-mediated ADCC in vitro.6 8 However, the clinical effects of these ADCC-inducing therapeutic antibodies could be limited by the presence of the V158F polymorphism on the FcγRIIIA/CD16a receptor. Indeed, the CD16a 158F/F and CD16a 158F/V allotypes have been associated with a poorer clinical response to antibody treatment in some clinical trials,39–41 but not in others.41 42
We then showed that compared with F1M1, F1M1-Fc+ activated more potently the functional response of NK cells in vitro (ie, degranulation and intracellular IFNγ production) in NK92 cells expressing CD16a 158V or CD16a 158F, while F1M1-Fc− was completely ineffective. Moreover, F1M1-Fc+ was the most potent antibody to trigger ADCC in vitro against MDA-MB-231 TNBC cells in the presence of NK92 cells that express CD16a 158V or CD16a 158F and also of human primary NK cells that express endogenous CD16a 158V/F. These results strongly highlighted the relevance of potentiating the NK cell-mediated Fc effector functions of F1M1 by Fc-engineering.
This is the first study showing that cell surface cath-D can be targeted by a human antibody to mediate ADCC in TNBC cells. To our knowledge, only incubation with a humanized antibody against cathepsin S, another protease, triggered ADCC mechanism in colorectal and pancreatic cancer cells.43 Importantly, cath-D can also be associated with cell surface receptors on CAFs,23 29 and we found that F1M1-Fc+ also induced ADCC against breast CAFs. This suggests the possibility of a cytotoxic effect in the tumor stroma following its targeting, which has never been described before. We also found that the M6P/IGF2 receptor was involved in F1M1-Fc+-mediated ADCC on both TNBC cells and CAFs.
Next, we showed that compared with F1M1 and F1M1-Fc−, F1M1-Fc+ inhibited more potently MDA-MB-231 tumor growth and prolonged survival of mice harboring TNBC cell xenografts. F1M1-Fc− was significantly less effective, reflecting the importance of Fc-dependent mechanisms also in vivo, and suggesting an important role of immune cells in mice. In addition, nude mice treated with F1M1-Fc+ showed no sign of leukopenia, thrombopenia, anemia, and neutropenia, suggesting the absence of toxicity. Importantly, F1M1-Fc+, our most potent antibody, inhibited tumor growth also of two TNBC PDXs (B1995 and B3977) and prolonged survival of these mice. PDX B1995 was isolated from a patient with TNBC after neo-adjuvant therapy, whereas PDX B3977 was isolated from a patient with TNBC resistant to neoadjuvant chemotherapy.36 Therefore, our results suggest that F1M1-Fc+ may represent a new therapeutic strategy for patient with chemotherapy-resistant TNBC.
Recent studies and clinical trials highlighted that NK cell-based immunotherapy can awake the innate anticancer response, particularly against tumor metastases.4–6 Using mouse TNBC models, it was shown that NK cells are selectively increased in the dormant milieu, and that adjuvant IL15-based immunotherapy ensures an abundant pool of NK cells to sustain dormancy, thus preventing liver metastasis formation and prolonging survival.7 By phenotyping the NK cell infiltrates in MDA-MB-231 cell xenografts, we observed that only treatment with F1M1-Fc+ led to the tumor recruitment of immature CD27-CD11b- NK and mature stage 1 CD27+CD11b− NK cells that were undergoing activation, as indicated by the concomitant upregulation of the Eomes transcription factor.35 Moreover, following F1M1-Fc+ treatment, the cell surface expression of CD107a, a functional marker of NK cell degranulation, and the intracellular expression of granzyme B, a marker of NK cell cytotoxic activity, were significantly increased in NK cells that infiltrated MDA-MB-231 cell xenografts. This was corroborated by the upregulation of granzyme B and perforin in MDA-MB-231 tumor xenografts and also of the antitumor cytokine TNFα. These effects were limited when using F1M1-Fc−, suggesting that F1M1-Fc+ can target NK cells to tumors and possibly also to metastases, sustaining dormancy.7 We also found that in nude mice, NK cell depletion, by treatment with the anti-asialo GM1 antibody, impaired F1M1-Fc+ therapeutic efficacy in MDA-MB-231 TNBC cell xenografts, demonstrating the key role of tumor-infiltrating NK cells in F1M1-Fc+ effect. Interestingly, the limited antitumor activity of F1M1-Fc+ observed in mice treated with the anti-asialo GM1 antibody was very similar to that observed with F1M1-Fc−, which depends solely on Fab-mediated effector functions. This strongly suggests that NK cells are the main effectors of the Fc-dependent response with F1M1-Fc+. In addition, our unpublished experiments in MDA-MB-231 cells strongly suggest that F1M1-Fc+ only slightly triggered ADCP in the presence of THP1 M1-polarized macrophages and did not induce CDC (data not shown). However, macrophage depletion experiments are needed to conclude on the potential role of macrophages in F1M1-Fc+ antitumor effect. Taken together, these results demonstrated that the Fc-mediated effector functions of F1M1-Fc+ are dependent on NK cells. The mechanism by which F1M1-Fc+ induces NK cell recruitment to the tumor remains unexplained. F1M1-Fc+ via its improved Fc-part might bind to immature NK cells in the bloodstream and direct them to the tumor where they become activated.
The immunomodulating effects of standard and novel chemotherapeutic approaches can also be exploited in combination with tumor-targeting antibodies.44 In metastatic TNBC, pembrolizumab (anti-PD-L1 antibody) in association with chemotherapy, sacituzumab govitecan (anti-TROP2 antibody-drug conjugate, ADC), and trastuzumab-deruxtecan (ADC against HER2low tumors) showed improved overall and progression-free survival compared with chemotherapy alone.3 45 Paclitaxel induces cell-cycle arrest and also promotes tumor immunity,46 guiding tumor-associated macrophages toward the M1-like antitumor phenotype.47 As neoadjuvant chemotherapy (nanoparticle-bound paclitaxel, epirubicin, and cyclophosphamide) decreases by ~50% the frequency of NK cells,48 combining chemotherapy with immunotherapy that recruits and/or reactivates NK cell cytotoxic activity is an attractive treatment option. Therefore, we analyzed the combination of paclitaxel and F1M1-Fc+. We found that this combination significantly enhanced paclitaxel therapeutic effect in MDA-MB-231 tumor xenografts. As incubation of TNBC cells with paclitaxel in vitro did not have any effect on cath-D expression and secretion, adding a protease-targeting agent, such as F1M1-Fc+, may benefit chemotherapy-based treatment regimens for patients with TNBC. Moreover, combined cytotoxic effects of chemotherapy and NK cells, should favor priming of CD4+ and CD8+ T cells and increase efficacy of adaptative antitumoral response.49
Recently, we performed a preclinical pilot study to characterize the biological TNBC subtypes that express cath-D by IHC using a TMA of 147 TNBC to identify patients who could benefit most of anti-cath-D antibody based-therapy.16 We found that AR/cath-D coexpression in non-metastatic TNBC is an independent prognostic factor of worse overall survival, suggesting the possibility of adjuvant anti-androgen therapy combined with anti-cath-D antibodies.16 The AR antagonist enzalutamide has clinical activity and is well tolerated in patients with early and advanced AR+ TNBC.50 51 A recent clinical trial showed the safety and efficacy of enzalutamide in combination with the PI3K inhibitor taselisib in patients with metastatic AR+ TNBC.52 Here, we demonstrated that the combination of F1M1-Fc+ with enzalutamide improved the therapeutic efficacy of enzalutamide in AR+ SUM159 TNBC tumor xenografts. We also showed that in vitro cath-D secretion and AR expression in SUM159 cells were not affected by incubation with enzalutamide and with F1M1-Fc+, respectively. Thus, the F1M1-Fc++ enzalutamide combination may have a therapeutic advantage for patients with AR+ TNBC. In this study (mouse model), we injected 15 mg/kg F1M1-Fc+ 3 times/week ip for 5 weeks. As the ip route is less efficient than the iv route in terms of PK/PD, we increased the dose to 15 mg/kg (3–4 fold more than the intravenous dose in humans). Therefore, a dose similar (4–8 mg/kg) to the one classically used for naked anti-HER2 trastuzumab, according to the FDA and EMA recommendations, could be used when the naked F1M1-Fc+ is transferred to the clinic.
Conclusion
Overall, our data indicate that the Fc-enhanced anti-cath-D F1M1-Fc+ antibody increased ADCC, triggered the recruitment, activation and cytotoxic activity of tumor-infiltrating NK cells, had higher antitumor efficacy, and prolonged survival of mice harboring TNBC cell xenografts with no associated toxicity. Our preclinical proof-of-concept study validated the feasibility and efficacy of combining an Fc-optimized anti-cath-D antibody with chemotherapy or antiandrogens for TNBC treatment.
Data availability statement
Data are available on reasonable request.
Ethics statements
Patient consent for publication
Ethics approval
This study involves human participants for immunohistochemistry (IHC) and TNBC cytosols. TNBC biopsy samples were provided by the biological resource center (Biobank number BB-0033-00059) after approval by the Montpellier Cancer Institute Institutional Review Board, following the French national Ethics and Legal dispositions for patients’ information and consent. For TNBC cytosols, patient samples were processed according to the French Public Health Code (law no 2004-800, articles L. 1243-4 and R. 1243-61), and the biological resources center has been authorized (authorization number: AC-2008-700; Val d’Aurelle, ICM, Montpellier) to deliver human samples for scientific research. All patients were informed before surgery that their surgical specimens might be used for research purposes. For human primary NK cell isolation and expansion, this work benefited from umbilical cord blood units and John De Vos’ expertise (head of the Biological Resource Center Collection of the University Hospital of Montpellier, http://www.chu-montpellier.fr/en/platforms; BIOBANQUES Identifier: BB-0033-00031).The study approval for PDXs was previously published. Participants gave informed consent to participate in the study before taking part.
Acknowledgments
We thank Cecile Dejou (MRI, Montpellier, France) for flow cytometry support. We thank Charles Theillet for providing PDXs. We thank Béatrice Clémenceau and Henri Vié (INSERM U892, Nantes, France) for kindly providing NK92 cells. We thank Hervé Watier (University of Tours, France), Bruno Robert (IRCM, Montpellier, France) and William Jacot (ICM, Montpellier, France) for helpful discussions and support. We thank the IRCM’s animal facility unit members (BioCampus RAM-PEFO, IRCM, Montpellier, France) for animal care and for in vivo studies. SPR experiments were carried out using the facilities of the Montpellier Proteomics Platform (PPM-PP2I, BioCampus Montpellier). This publication is based upon work from COST Action ProteoCure, CA20113, supported by COST (European Cooperation in Science and Technology).
References
Supplementary materials
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Footnotes
Twitter @LAJOIE Laurie
TC and LG contributed equally.
Contributors PDdR, LL, TC, LG and EL-C designed the experiments and prepared the manuscript. PDdR, LL, LG, AM, GN, LR, LBA, LF, VG and EL-C performed the experiments. PDdR, LL, LG, AM, LBA, HM, LC, TD, FBM, M-CC, MJ, MV, TC, PM, VL-M, PR, SG, TC and EL-C provided material and analyzed data. PDdR, TC and EL-C analyzed data and proof-read and finalized the manuscript. EL-C acted as guarantor.
Funding This work was supported by a public grant overseen by the French National Research Agency (ANR) as part of the “Investissements d’Avenir” program (reference: LabEx MabImprove ANR-10-LABX-53- 01), University of Montpellier, Inserm Transfert, Région Occitanie, the association "Ligue Régionale du Gard CD30", "Ligue Régionale de l’Herault CD34", and "Ligue Régionale de la Charente Maritime CD17".
Competing interests None declared.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.