Purpose:

This phase 1 study (NCT03440437) evaluated the safety, tolerability, pharmacokinetics (PK), and activity of FS118, a bispecific antibody–targeting LAG-3 and PD-L1, in patients with advanced cancer resistant to anti–PD-(L)1 therapy.

Patients and Methods:

Patients with solid tumors, refractory to anti–PD-(L)1–based therapy, received intravenous FS118 weekly with an accelerated dose titration design (800 μg to 0.3 mg/kg) followed by 3+3 ascending dose expansion (1 to 20 mg/kg). Primary objectives were safety, tolerability, and PK. Additional endpoints included antitumor activity, immunogenicity, and pharmacodynamics.

Results:

Forty-three patients with a median of three prior regimens in the locally advanced/metastatic setting, including at least one anti–PD-(L)1 regimen, received FS118 monotherapy. FS118 was well tolerated, with no serious adverse events relating to FS118 reported. No dose-limiting toxicities (DLT) were observed, and an MTD was not reached. The recommended phase 2 dose of FS118 was established as 10 mg/kg weekly. The terminal half-life was 3.9 days. Immunogenicity was transient. Pharmacodynamic activity was prolonged throughout dosing as demonstrated by sustained elevation of soluble LAG-3 and increased peripheral effector cells. The overall disease control rate (DCR) was 46.5%; this disease control was observed as stable disease, except for one late partial response. Disease control of 54.8% was observed in patients receiving 1 mg/kg or greater who had acquired resistance to PD-(L)1–targeted therapy.

Conclusions:

FS118 was well tolerated with no DLTs observed up to and including 20 mg/kg QW. Further studies are warranted to determine clinical benefit in patients who have become refractory to anti–PD-(L)1 therapy.

See related commentary by Karapetyan and Luke, p. 835

Following treatment with immune checkpoint inhibitors, many patients develop resistance, highlighting the need for novel therapeutic approaches. Resistance to anti–PD-(L)1 therapy may be associated with upregulation of LAG-3 (lymphocyte-activation gene-3). FS118 is a first-in-class tetravalent bispecific antibody binding to LAG-3 and PD-L1, resulting in reversal of immune suppression. In this phase 1 first-in-human study, FS118 was well tolerated and exhibited a good safety profile in patients with advanced solid tumors that progressed on prior anti–PD-(L)1 therapy. Sustained, increased levels of soluble LAG-3 and peripheral immune cells indicated a prolonged pharmacodynamic effect beyond the clearance of FS118 from the serum. Acquired resistance and immune checkpoint blockade as last prior therapy correlated with long-lasting disease control in patients with LAG-3+ PD-L1+ tumors. Study data suggest that FS118 is well tolerated, and further studies are warranted to determine how FS118 may provide benefit to patients who are resistant to anti–PD-(L)1 therapies.

Immune checkpoint blockade (ICB) can modulate immune cells within the tumor microenvironment, shifting the cells from an immunosuppressive or exhausted state to a proinflammatory state that favors an antitumor response (1).

Programmed death ligand 1 (PD-L1) is widely accepted as an immune checkpoint receptor, which delivers inhibitory signals and reduces T-cell–mediated effector responses through engagement with its receptors, programmed cell death protein 1 (PD-1) and CD80. PD-1/PD-L1 blockade restores functional T-cell responses in the tumor, leading to the approval of several PD-(L)1–targeting antibodies to treat patients with locally advanced or metastatic disease (2–5). Although these monotherapies have demonstrated meaningful responses and prolonged survival in a subset of patients, a large percentage of patients exhibit resistance to ICB therapy (6). Preclinical studies have indicated that compensatory upregulation of alternative checkpoint proteins such as lymphocyte-activation gene-3 (LAG-3) may be a mechanism of acquired resistance to anti–PD-(L)1 therapy (7, 8).

LAG-3 is an inhibitory checkpoint molecule that is predominantly expressed on the surface of T cells, natural killer (NK) cells, and plasmacytoid dendritic cells. LAG-3 is upregulated on the surface of activated and exhausted T cells and by a proportion of T-regulatory cells, which can confer suppressive activity against T effector cells by engaging peptide–MHC class II complexes (9–11). Although three splice variants of LAG-3 are proposed to exist (12), evidence is limited and soluble LAG-3 is most likely to arise from proteolytic cleavage of surface LAG-3 mediated by a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and 17 (ADAM17; ref. 13). Cleavage of LAG-3 from conventional CD4+ T cells overcame anti–PD-1 resistance, and high LAG-3 and low ADAM-10 expression on CD4+ T cells in patients with squamous cell carcinoma of the head and neck (SCCHN) correlated with poor prognosis (14). Importantly, co-blockade of LAG-3 and the PD-1/L1 pathway modulates T-cell activity and enhanced antitumor T-cell responses in mouse models (8, 15, 16). Co-expression of LAG-3 and PD-1 on tumor-infiltrating lymphocytes (TIL) in clinical samples correlated with T-cell exhaustion and dysfunction (17, 18), and LAG-3 has been described previously as a poor prognostic marker in many cancers (19–21).

Several LAG-3–targeting molecules in clinical development have shown to be well tolerated with some responses as monotherapies reported (22, 23); however, combined targeting of LAG-3 and the PD-(L)1 axis may provide greater clinical benefit (24–26).

FS118 is a first-in-class tetravalent bispecific (mAb2 format) mAb against LAG-3 and PD-L1 (27, 28). A LAG-3–binding site was introduced into the fragment crystallizable (Fc) region of a full-length human IgG1 PD-L1 mAb (27), in addition to two mutations to abrogate Fcγ receptor binding (29). FS118-mediated blockade of LAG-3 and PD-L1 enhanced T-cell activity in vitro, and a mouse surrogate of FS118 significantly suppressed mouse tumor growth (28). Interestingly, a mouse surrogate of FS118 reduced cell-surface LAG-3 on T cells and increased soluble LAG-3 in the serum of mice, whereas the combination of mAbs targeting LAG-3 and PD-L1 increased LAG-3 expression in vivo. Given the compensatory upregulation of LAG-3 in response to anti–PD-(L)1 failures, and the mechanism of FS118-mediated shedding of cell-surface LAG-3 to overcome immune-mediated suppression, these data indicate that FS118 may extend clinical benefit beyond anti–PD-(L)1 monotherapy, providing rationale for this phase 1 clinical trial.

Study design and treatment

This open-label, multiple-dose, first-in-human phase 1 study aimed to characterize the safety, tolerability, pharmacokinetics (PK), and preliminary antitumor activity of FS118 in patients with advanced solid tumors and resistance to PD-(L)1 therapy (clinical trials identifier: NCT03440437). Pharmacodynamics of FS118 activity and immunogenicity measured by anti-drug antibodies (ADA) were also assessed.

Patients enrolled onto the study received FS118 intravenously once a week in three-week treatment cycles. The study followed an accelerated titration design; the first four cohorts (800 μg to 0.3 mg/kg) were enrolled sequentially as single-patient cohorts, and dose levels were progressed if no dose-limiting toxicities (DLT) or ≥ grade 2 adverse events were observed during the first cycle of treatment (intra-patient dose escalation was permitted). This was followed by a 3+3 ascending dose-escalation design (from 1 to 20 mg/kg) to determine the maximum tolerated dose (MTD). Dose-escalation cohorts were expanded at 3 mg/kg (n = 10), 10 mg/kg (n = 13), and 20 mg/kg (n = 13) to further explore PK, pharmacodynamics, and antitumor activity. Patients were considered to have completed treatment following 16 cycles (or 12 months) of FS118 treatment, or following confirmed progressive disease. Patients remaining on treatment at 12 months had the option to continue therapy.

Patient eligibility

Eligible patients had histologically confirmed locally advanced, unresectable or metastatic solid tumors and had exhausted available therapeutic options. Patients had received prior PD-(L)1 immune checkpoint inhibitors for a minimum of 12 weeks and showed subsequent progressive disease before enrolment. Prior treatment with LAG-3–targeting agents was not permitted.

Eligible patients were ≥18 years old, had an Eastern Cooperative Oncology Group performance status of 0–1, measurable disease (as per RECIST v1.1), and a life expectancy of at least three months. Patients were excluded if they had primary central nervous system tumors or metastases, a history of autoimmune disease, known infections, respiratory disease, encephalitis, seizures, or severe immune-related adverse events with prior anti–PD-(L)1–containing therapies. Patients with significant cardiac or laboratory abnormalities or a history of uncontrolled intercurrent illness were excluded (30).

The trial protocol and all amendments were approved by the appropriate institutional review board or independent ethics committee at each trial center. The trial was conducted according to the Declaration of Helsinki and International Conference on Harmonization Guidelines for Good Clinical Practice. All patients gave written informed consent before enrollment.

Study objectives and assessments

The primary objectives were to assess safety, define the MTD and/or recommended phase 2 dose (RP2D), and determine the PK parameters of FS118. The secondary objectives were to assess antitumor activity and characterize the immunogenicity (ADAs) of FS118. Exploratory objectives included characterizing the pharmacodynamic profile and correlating potential primary pharmacology with exposure. Radiographic assessments were conducted every eight weeks as per RECIST v1.1 and iRECIST. Toxicity was evaluated according to the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE, v4.03). The data cutoff value for the assessment of antitumor activity and safety was September 18, 2020.

Immunogenicity analysis

Blood samples taken at each cycle were assessed for the development of ADAs using a validated, quasi-quantitative bridging electrochemiluminescence immunoassay. Samples were analyzed using a three-tier approach (screening, confirmatory, and titer determination). Affinity-purified rabbit anti-FS118 polyclonal antibodies (Envigo) acted as a positive control. The sensitivity of the screening assay was 10 ng/mL of positive control antibody, and the drug tolerance reported at 100 ng/mL of positive control antibody was 4.8 μg/mL.

PK analysis

Serum concentration of free FS118 was measured using a validated ligand-binding assay on the Gyrolab platform, based on capture and detection with the target molecules [biotin-labeled LAG-3 recombinant protein (BPS Bioscience) and Alexa Fluor 647–labeled PD-L1 (Bio-Techne)]. The assay sensitivity was 100 ng/mL. Standard serum PK parameters for FS118 were estimated using two-compartmental analysis.

Soluble receptor analysis

Serum concentration of total soluble LAG-3 (sLAG-3) was quantified using an ELISA. Serum sLAG-3 was complexed with a saturating concentration of FS118, and the complex was captured with a non-competing anti–LAG-3 antibody clone 17B4 (Enzo Life Sciences) and detected with a non-competing anti-FS118 anti-idiotype antibody (Maine Biotechnology Services). Serum concentration data were used to calculate the AUC over the first week of treatment (AUC0–7days) for each patient. The median (with 95% confidence interval) was calculated both for the serum concentration before each dosing cycle, and the AUC0–7days, grouped by dose.

Immunohistochemistry

Tumor biopsies were collected before FS118 treatment for baseline targets expression, and samples were formalin-fixed, processed according to institutional best practices and paraffin-embedded. IHC was implemented on the BenchMark Ultra platform (Ventana/Roche), using anti–PD-L1 antibody clone SP263 (Roche/Ventana) and anti–LAG-3 antibody clone 17B4 (Abcam). The tumor proportion score (TPS) was calculated for PD-L1 expression, and PD-L1–positivity (PD-L1+) was defined as a TPS of ≥1%. For LAG-3 evaluation, five representative high-power fields (HPF, the area visible under the maximum magnification power) within the intra-epithelial tumor component and five HPF within the intra-tumoral stroma were selected and the average of these areas was used to assess the number of positive cells in each field. LAG-3–positivity (LAG-3+) was defined as one or more HPF with LAG-3+ cells.

Immunophenotyping

Immunophenotyping was performed using flow cytometry on live fresh-blood cells. The panel included antibodies to CD45 (HI30, BD Biosciences, 563204), CD3 (SK7, BD Biosciences, 564001), CD19 (HIB19, BioLegend, 302240), CD4 (SK3, BD Biosciences, 563028), CD8 (SK1, BD Biosciences, 564629), CD56 (HCD56, BioLegend, 318334), and Ki67 (20Raj1, eBioscience, 46-5699-42). Immune cell populations were defined as: CD4 T-cells (CD45+/CD3+/CD4+/CD8/CD19), CD8 T-cells (CD45+/CD3+/CD8+/CD4/CD19) and NK cells (CD45+/CD3/CD19/CD56+/CD16+/−). All values were reported as a percentage of total CD45+ leucocytes, and only cell subsets with more than 50 cells were considered.

Statistical analysis

A Mann–Whitney two-tailed test was used to assess correlations between time on FS118 treatment and prior ICB therapy in the context of resistance to PD-(L)1 therapy, as well as to test the statistical significance between dose levels with respect to sLAG-3 in patient serum and the difference in FS118 PK profile between ADA-negative and confirmed positive patients. A paired Wilcoxon signed-rank test was applied to analyze changes in leukocytes in patient blood, between baseline and on treatment values. Multiple P values were corrected with the Benjamani–Hochberg approach. Statistical tests were performed using GraphPad Prism and R (R Core Team (2020). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL https://www.r-project.org/).

Data availability

The data generated in this study are available upon reasonable request from the corresponding author (M. Morrow).

Patient treatment and baseline characteristics

Forty-three patients with advanced solid tumors were treated with FS118 across eight dose-escalation cohorts. Single-patient cohorts were dosed with FS118 at fixed doses of 800 μg (Cohort 1), 2,400 μg (Cohort 2), 0.1 mg/kg (Cohort 3) or 0.3 mg/kg (Cohort 4; Supplementary Fig. S1). Dose-escalation cohorts were then expanded following a 3+3 design to further explore PK, pharmacodynamics, and preliminary antitumor activity, resulting in a further 39 patients treated at 1 mg/kg (n = 3, Cohort 5), 3 mg/kg (n = 10, Cohort 6), 10 mg/kg (n = 13, Cohort 7) or 20 mg/kg (n = 13, Cohort 8).

The representativeness of this study of PD-L1–refractory solid tumors is discussed in Supplementary Table S1. The age of patients across all cohorts ranged from 30 to 85 years with a median age of 59 years (Table 1). The majority of patients were male (60.5%) and white (67.4%). Patients with a wide range of tumor types were enrolled (Supplementary Table S1). Advanced non–small cell lung cancer (NSCLC) and SCCHN were the most common tumor types, comprising 20.9% and 14.0% of all patients, respectively. Patients enrolled on study were heavily pre-treated, having received a median of three prior regimens (range, 1–11) for locally advanced/metastatic disease; 37.2% received four or more prior regimens (Table 1). The median number of prior ICB regimens was one (range, 1–3), with 21.0% of patients receiving two or more prior ICB regimens, and the median total time on prior ICB regimens was 38.9 weeks.

Table 1.

Baseline demographics and prior therapy in patients treated with FS118.

 Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8All cohorts
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n= 13)(n = 43)
Median age (range; y) 54 (47–73) 73 (59–75) 70 (38–78) 59 (43–85) 56 (30–77) 59 (30–85) 
Gender, n (%) 
 Female 1 (25.0) 2 (66.7) 4 (40.0) 6 (46.2) 4 (30.8) 17 (39.5) 
 Male 3 (75.0) 1 (33.3) 6 (60.0) 7 (53.8) 9 (69.2) 26 (60.5) 
Ethnicity, n (%) 
 White 3 (75.0) 3 (100.0) 4 (40.0) 8 (61.5) 11 (84.6) 29 (67.4) 
 Black or African American 1 (25.0) 5 (50.0) 6 (14.0) 
 Asian 3 (23.1) 3 (7.0) 
 Native Hawaiian or Pacific Islander 1 (10.0) 1 (2.3) 
 Other 2 (15.4) 2 (15.4) 4 (9.3) 
ECOG performance score, n (%)  
 0 2 (66.7) 2 (20.0) 1 (7.7) 4 (30.8) 9 (20.9) 
 1 4 (100.0) 1 (33.3) 8 (80.0) 11 (84.6) 9 (69.2) 33 (76.7) 
 2 1 (7.7)a 1 (2.3) 
No. of all regimensb, n (%) 
 1 
 2 1 (25.0) 2 (20.0) 1 (7.7) 5 (38.5) 10 (23.3) 
 3 1 (25.0) 2 (66.6) 2 (20.0) 4 (30.8) 1 (7.7) 9 (20.9) 
 4+ 2 (50.0) 1 (33.3) 6 (60.0) 8 (61.5) 7 (53.8) 24 (55.8) 
 Median number of prior regimens (range) 3.5 (2–5) 3 (3–8) 4 (2–11) 4 (2–6) 4 (2–9) 4 (2–11) 
Prior regimens for locally advanced/metastatic disease 
 No. of all regimens, n (%) 
  1 2 (15.4) 2 (4.7) 
  2 1 (25.0) 1 (33.3) 2 (20.0) 3 (23.1) 5 (38.5) 13 (30.2) 
  3 2 (50.0) 1 (33.3) 3 (30.0) 5 (38.5) 2 (15.4) 12 (27.9) 
  4+ 1 (100.0) 1 (33.3) 5 (50.0) 5 (38.5) 4 (30.8) 16 (37.2) 
  Median number of prior regimens (range) 3 (2–5) 3 (2–6) 3.5 (2–11) 3 (2–6) 2 (1–8) 3 (1–11) 
 No. of prior ICB regimens, n (%) 
  1 3 (75.0) 1 (33.3) 7 (70.0) 11 (84.6) 11 (84.6) 34 (79.1) 
  2 1 (25.0) 2 (66.6) 1 (10.0) 2 (15.4) 1 (7.7) 6 (14.0) 
  3 2 (20.0) 1 (7.7) 3 (7.0) 
  Median number of ICB regimens (range) 1 (1–2) 2 (1–2) 1 (1–3) 1 (1–2) 1 (1–3) 1 (1–3) 
  Mean time (wk) on prior ICB regimens (range) 40.7 (15.3–68.3) 24.3 (15.0–38.3) 26.8 (7.9–67.1) 53.8 (17.6–182.3) 35.5 (12.9–65.0) 38.9 (7.9–182.3) 
 Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8All cohorts
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n= 13)(n = 43)
Median age (range; y) 54 (47–73) 73 (59–75) 70 (38–78) 59 (43–85) 56 (30–77) 59 (30–85) 
Gender, n (%) 
 Female 1 (25.0) 2 (66.7) 4 (40.0) 6 (46.2) 4 (30.8) 17 (39.5) 
 Male 3 (75.0) 1 (33.3) 6 (60.0) 7 (53.8) 9 (69.2) 26 (60.5) 
Ethnicity, n (%) 
 White 3 (75.0) 3 (100.0) 4 (40.0) 8 (61.5) 11 (84.6) 29 (67.4) 
 Black or African American 1 (25.0) 5 (50.0) 6 (14.0) 
 Asian 3 (23.1) 3 (7.0) 
 Native Hawaiian or Pacific Islander 1 (10.0) 1 (2.3) 
 Other 2 (15.4) 2 (15.4) 4 (9.3) 
ECOG performance score, n (%)  
 0 2 (66.7) 2 (20.0) 1 (7.7) 4 (30.8) 9 (20.9) 
 1 4 (100.0) 1 (33.3) 8 (80.0) 11 (84.6) 9 (69.2) 33 (76.7) 
 2 1 (7.7)a 1 (2.3) 
No. of all regimensb, n (%) 
 1 
 2 1 (25.0) 2 (20.0) 1 (7.7) 5 (38.5) 10 (23.3) 
 3 1 (25.0) 2 (66.6) 2 (20.0) 4 (30.8) 1 (7.7) 9 (20.9) 
 4+ 2 (50.0) 1 (33.3) 6 (60.0) 8 (61.5) 7 (53.8) 24 (55.8) 
 Median number of prior regimens (range) 3.5 (2–5) 3 (3–8) 4 (2–11) 4 (2–6) 4 (2–9) 4 (2–11) 
Prior regimens for locally advanced/metastatic disease 
 No. of all regimens, n (%) 
  1 2 (15.4) 2 (4.7) 
  2 1 (25.0) 1 (33.3) 2 (20.0) 3 (23.1) 5 (38.5) 13 (30.2) 
  3 2 (50.0) 1 (33.3) 3 (30.0) 5 (38.5) 2 (15.4) 12 (27.9) 
  4+ 1 (100.0) 1 (33.3) 5 (50.0) 5 (38.5) 4 (30.8) 16 (37.2) 
  Median number of prior regimens (range) 3 (2–5) 3 (2–6) 3.5 (2–11) 3 (2–6) 2 (1–8) 3 (1–11) 
 No. of prior ICB regimens, n (%) 
  1 3 (75.0) 1 (33.3) 7 (70.0) 11 (84.6) 11 (84.6) 34 (79.1) 
  2 1 (25.0) 2 (66.6) 1 (10.0) 2 (15.4) 1 (7.7) 6 (14.0) 
  3 2 (20.0) 1 (7.7) 3 (7.0) 
  Median number of ICB regimens (range) 1 (1–2) 2 (1–2) 1 (1–3) 1 (1–2) 1 (1–3) 1 (1–3) 
  Mean time (wk) on prior ICB regimens (range) 40.7 (15.3–68.3) 24.3 (15.0–38.3) 26.8 (7.9–67.1) 53.8 (17.6–182.3) 35.5 (12.9–65.0) 38.9 (7.9–182.3) 

aProtocol deviation.

bIncluding all regimens in adjuvant and locally advanced/metastatic settings. All percentages are rounded to one decimal place.

Safety and tolerability

A total of 469 treatment-emergent adverse events were observed across all patients enrolled on study, which were present in 42 of the 43 patients enrolled (97.7%; Table 2).

Table 2.

Treatment-emergent adverse events (TEAE) and treatment-related adverse events (TRAE) in patients treated with FS118, by dose level and grade.

 Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8All cohorts
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n = 13)(n = 43)
Number of TEAEs, n 37 119 158 149 469 
Number of patients with TEAEs, n (%) 3 (75.0) 3 (100.0) 10 (100.0) 13 (100.0) 13 (100.0) 42 (97.7) 
Number of patients with TRAEs, n (%) 2 (50.0) 1 (33.3) 7 (70.0) 11 (84.6) 4 (30.8) 25 (58.1) 
Number of patients with ≥grade 3 TRAEs, n (%)a 2 (15.4) 2 (4.7) 
Number of patients with TRAEs leading to drug withdrawal (%)b 1 (7.7) 1 (2.3) 
Number of patients with grade 1 or 2 TRAEs occurring in >1 patient, n (%) 
 Arthralgia 2 (15.4) 2 (4.7) 
 Decreased appetite 1 (25.0) 1 (7.7) 2 (4.7) 
 Diarrhea 1 (25.0) 1 (7.7) 2 (4.7) 
 Dry mouth or throat 1 (25.0) 1 (10.0) 2 (4.7) 
 Headache 1 (10.0) 1 (7.7) 2 (4.7) 
 Hot flush or flushing 2 (20.0) 2 (4.7) 
 Pruritus 2 (20.0) 2 (4.7) 
 Pyrexia 1 (10.0) 1 (7.7) 2 (4.7) 
 Vomiting 2 (20.0) 2 (4.7) 
 Anemia 1 (25.0) 1 (10.0) 1 (7.7) 3 (7.0) 
 Fatigue 1 (10.0) 1 (7.7) 1 (7.7) 3 (7.0) 
 Infusion-related reaction 1 (33.3) 3 (30.0) 1 (7.7) 5 (11.6) 
 Nausea 3 (30.0) 2 (15.4) 1 (7.7) 6 (14.0) 
 Rashc 2 (20.0) 2 (15.4) 2 (15.4) 6 (14.0) 
Number of grade 3 TRAEs, n (%) 
 Elevated ALP 1 (7.7) 1 (2.3) 
 Elevated ALT 1 (7.7) 1 (2.3) 
 Elevated AST 1 (7.7) 1 (2.3) 
 Elevated bilirubin 1 (7.7) 1 (2.3) 
Number of grade 4 or 5 TRAEs, n (%) 
SAEs reported as related to FS118 
 Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8All cohorts
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n = 13)(n = 43)
Number of TEAEs, n 37 119 158 149 469 
Number of patients with TEAEs, n (%) 3 (75.0) 3 (100.0) 10 (100.0) 13 (100.0) 13 (100.0) 42 (97.7) 
Number of patients with TRAEs, n (%) 2 (50.0) 1 (33.3) 7 (70.0) 11 (84.6) 4 (30.8) 25 (58.1) 
Number of patients with ≥grade 3 TRAEs, n (%)a 2 (15.4) 2 (4.7) 
Number of patients with TRAEs leading to drug withdrawal (%)b 1 (7.7) 1 (2.3) 
Number of patients with grade 1 or 2 TRAEs occurring in >1 patient, n (%) 
 Arthralgia 2 (15.4) 2 (4.7) 
 Decreased appetite 1 (25.0) 1 (7.7) 2 (4.7) 
 Diarrhea 1 (25.0) 1 (7.7) 2 (4.7) 
 Dry mouth or throat 1 (25.0) 1 (10.0) 2 (4.7) 
 Headache 1 (10.0) 1 (7.7) 2 (4.7) 
 Hot flush or flushing 2 (20.0) 2 (4.7) 
 Pruritus 2 (20.0) 2 (4.7) 
 Pyrexia 1 (10.0) 1 (7.7) 2 (4.7) 
 Vomiting 2 (20.0) 2 (4.7) 
 Anemia 1 (25.0) 1 (10.0) 1 (7.7) 3 (7.0) 
 Fatigue 1 (10.0) 1 (7.7) 1 (7.7) 3 (7.0) 
 Infusion-related reaction 1 (33.3) 3 (30.0) 1 (7.7) 5 (11.6) 
 Nausea 3 (30.0) 2 (15.4) 1 (7.7) 6 (14.0) 
 Rashc 2 (20.0) 2 (15.4) 2 (15.4) 6 (14.0) 
Number of grade 3 TRAEs, n (%) 
 Elevated ALP 1 (7.7) 1 (2.3) 
 Elevated ALT 1 (7.7) 1 (2.3) 
 Elevated AST 1 (7.7) 1 (2.3) 
 Elevated bilirubin 1 (7.7) 1 (2.3) 
Number of grade 4 or 5 TRAEs, n (%) 
SAEs reported as related to FS118 

Note: Adverse events were graded according to NCI CTCAE v4.03 classification.

Abbreviation: SAEs, serious adverse events.

aTwo patients in total presented with grade 3 TRAEs; one presented with elevated ALT, elevated AST, and increased bilirubin, and another patient presented with elevated ALP.

bOne patient had treatment withdrawn as a result of adverse events related to study drug (grade 3 ALP elevation), which was resolved.

cRash included maculopapular, erythematous, pruritic. All percentages rounded to one decimal place.

Infusion of FS118 was well tolerated, where grade 1–2 infusion-related reactions were observed in five of 43 patients (11.6%; Table 2). In total, five patients discontinued treatment with FS118, which was not drug related in four of these patients (treatment withdrawn due to myocardial infarction, disease progression, abdominal pain, and tracheal obstruction). One patient withdrew due to treatment-related elevation of alkaline phosphatase (ALP) levels (grade 3) following treatment of FS118 at 10 mg/kg. No DLTs were observed, and the MTD was not reached.

Of the 43 patients enrolled on the trial, 25 (58.1%) had treatment-related adverse events (TRAE), of which two patients (4.7%), both of whom were dosed at 10 mg/kg, experienced grade 3 TRAEs (Table 2); one patient presented with elevated alanine aminotransferase, aspartate aminotransferase and bilirubin that resolved without the need to reduce the dose of FS118, and another patient demonstrated elevated ALP that resulted in the discontinuation of treatment. No treatment-related grade 4, 5 or serious adverse events relating to FS118 were reported.

Immunogenicity

Overall, any observed immunogenicity, as defined by the presence of confirmed positive ADAs, was typically transient in nature (i.e., did not persist for longer than three consecutive cycles) and had no significant impact on FS118 levels. Immunogenicity was observed in 21 of 43 patients (48.8%) treated with FS118. Four patients (9.3%) had ADAs present at baseline (Supplementary Table S2). It was noted that a smaller proportion of patients were ADA-positive at higher dose levels compared with patients treated with lower dose levels of FS118 [three out of 13 patients in the 20 mg/kg (23.1%) cohort, compared with seven out of 10 patients in the 3 mg/kg cohort (70%)]. All ADA confirmed positive patients at 10 and 20 mg/kg demonstrated ADAs that were transient, whereas in the 1 and 3 mg/kg cohorts (total of 13 patients), two patients (66.7%) and one patient (10.0%), respectively, demonstrated ADAs for three or more consecutive cycles. In addition, in the higher dose levels of 10 and 20 mg/kg, no patients and two patients (15.4%), respectively, presented with ADAs at the last treatment cycle, compared with two patients (66.7%) and five patients (50.0%) in the 1 and 3 mg/kg cohorts.

Antitumor activity

The median FS118 treatment duration was eight weeks (a range of 0.1–79 weeks, as of the data cutoff value, September 18, 2020). Across all patients, a disease control rate (DCR) of 46.5% was observed (Table 3). In both the 10 and 20 mg/kg cohorts, seven out of 13 patients (53.8%) achieved stable disease (SD) per RECIST v1.1 (hereafter referred to as SD). A DCR of 30% (three out of 10 patients) was observed in the 3 mg/kg cohort. No complete or partial responses (PR) per RECIST v1.1 were observed across all dose levels. Overall, nine of 43 patients (20.9%) surpassed 20 weeks of treatment with FS118 (Fig. 1), which included patients with advanced and heavily pre-treated anaplastic thyroid carcinoma (ATC), leiomyosarcoma, NSCLC, malignant peritoneal mesothelioma, cervical cancer, ovarian cancer, and SCCHN. At the data cutoff value, 41 patients had completed treatment and two patients remained on FS118 treatment at 79 weeks (ATC) and 56.1 weeks (leiomyosarcoma). As of March 2022, the ATC patient remained on FS118 treatment for over three years with disease control, including a late confirmed PR.

Table 3.

Clinical responses in patients treated with FS118.

Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8Cohorts 5–8All patients
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg1–20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n = 13)(n = 39)(n = 43)
DCRa, n (%) 1 (25.0) 2 (66.7) 3 (30.0) 7 (53.8) 7 (53.8) 19 (48.7) 20 (46.5) 
95% CI — 9.4–99.2 6.7–65.2 25.1–80.8 25.1–80.8 32.4–65.2 31.2–62.3 
ORRb, n (%) 
95% CI — 0.0–70.8 0.0–30.8 0.0–24.7 0.0–24.7 0.0–9.0 0.0–8.2 
Best overall response, n (%) 
 CR 
 PR 
 SD 1 (25.0) 2 (66.7) 3 (30.0) 7 (53.8) 7 (53.8) 19 (48.7) 20 (46.5) 
 PD 3 (75.0) 1 (33.3) 5 (50.0) 4 (30.8) 4 (30.8) 14 (35.9) 17 (39.5) 
 Not evaluablec 2 (20.0) 2 (15.4) 2 (15.4) 6 (15.4) 6 (14.0) 
Cohorts 1–4Cohort 5Cohort 6Cohort 7Cohort 8Cohorts 5–8All patients
800 μg to 0.3 mg/kg1 mg/kg3 mg/kg10 mg/kg20 mg/kg1–20 mg/kg
(n = 4)(n = 3)(n = 10)(n = 13)(n = 13)(n = 39)(n = 43)
DCRa, n (%) 1 (25.0) 2 (66.7) 3 (30.0) 7 (53.8) 7 (53.8) 19 (48.7) 20 (46.5) 
95% CI — 9.4–99.2 6.7–65.2 25.1–80.8 25.1–80.8 32.4–65.2 31.2–62.3 
ORRb, n (%) 
95% CI — 0.0–70.8 0.0–30.8 0.0–24.7 0.0–24.7 0.0–9.0 0.0–8.2 
Best overall response, n (%) 
 CR 
 PR 
 SD 1 (25.0) 2 (66.7) 3 (30.0) 7 (53.8) 7 (53.8) 19 (48.7) 20 (46.5) 
 PD 3 (75.0) 1 (33.3) 5 (50.0) 4 (30.8) 4 (30.8) 14 (35.9) 17 (39.5) 
 Not evaluablec 2 (20.0) 2 (15.4) 2 (15.4) 6 (15.4) 6 (14.0) 

Note: Responses according to RECIST v1.1.

aDisease control rate (DCR) defined as the sum of complete response (CR), partial response (PR), and stable disease (SD).

bObjective response rate (ORR) defined as the sum of CR and PR.

cThe response of six patients could not be determined as the patients came off study before a restaging scan. All percentages rounded to one decimal place.

Figure 1.

Clinical responses in patients treated with FS118. A, Time on FS118 treatment in patients with acquired (blue) or primary (red) resistance to ICB therapy by dose level. Yellow diamonds indicate SD and black diamonds indicate PD as per RECIST 1.1. B, Change in target lesions from baseline to last scan during FS118 treatment in patients with acquired (blue) or primary (red) resistance to ICB therapy. C, Time on FS118 treatment in patients treated with ICB or with another treatment as last prior regimen. The Mann–Whitney two-tailed test, *, P < 0.05. ATC, anaplastic thyroid cancer; BLA, bladder; CERV, cervical; CRC, colorectal carcinoma; HCC, hepatocellular carcinoma; ICB, immune checkpoint blockade; MEL, melanoma; MPM, malignant peritoneal mesothelioma; NREN, neuroendocrine; NSCLC, non–small cell lung carcinoma; OTH, other; OVRY, ovarian; PD, progressive disease; PRST, prostate; SARC, sarcoma; SCCHN, head and neck squamous cell carcinoma; SCLC, squamous cell lung carcinoma; SoD, sum of diameter; SD, stable disease.

Figure 1.

Clinical responses in patients treated with FS118. A, Time on FS118 treatment in patients with acquired (blue) or primary (red) resistance to ICB therapy by dose level. Yellow diamonds indicate SD and black diamonds indicate PD as per RECIST 1.1. B, Change in target lesions from baseline to last scan during FS118 treatment in patients with acquired (blue) or primary (red) resistance to ICB therapy. C, Time on FS118 treatment in patients treated with ICB or with another treatment as last prior regimen. The Mann–Whitney two-tailed test, *, P < 0.05. ATC, anaplastic thyroid cancer; BLA, bladder; CERV, cervical; CRC, colorectal carcinoma; HCC, hepatocellular carcinoma; ICB, immune checkpoint blockade; MEL, melanoma; MPM, malignant peritoneal mesothelioma; NREN, neuroendocrine; NSCLC, non–small cell lung carcinoma; OTH, other; OVRY, ovarian; PD, progressive disease; PRST, prostate; SARC, sarcoma; SCCHN, head and neck squamous cell carcinoma; SCLC, squamous cell lung carcinoma; SoD, sum of diameter; SD, stable disease.

Close modal

We hypothesized that a patients’ outcome on their previous anti–PD-(L)1 therapy, and whether they had primary or acquired resistance (6), could influence antitumor activity of FS118. Post hoc analysis explored correlations between FS118 treatment duration and resistance status to prior anti–PD-(L)1 therapy (acquired or primary resistance). It was observed that most patients who received FS118 treatment for greater than 20 weeks had previously achieved a PR, complete response (CR), or SD for more than three months as their best overall response to prior anti–PD-(L)1 therapy (Supplementary Fig. S2A; see Supplementary Data for more details). These patients were considered to have an acquired resistance phenotype (herein referred to as “acquired resistance” patients). Patients with progressive disease per RECIST v1.1 (hereafter referred to as PD) or whose best overall response to prior anti–PD-(L)1 therapy was SD for three months or less were considered to have a primary resistance phenotype (herein referred to as “primary resistance” patients). None of the patients with primary resistance remained on FS118 for more than 20 weeks, whereas it was noted that eight acquired resistance patients remained on study for greater than 20 weeks [seven of these patients for as long as six months (26 weeks); Supplementary Fig. S2A]. We also sought to assess whether the classification of resistance as defined by Kluger and colleagues (31) had any correlation with FS118 treatment duration. Using this classification, when acquired resistance was defined as a best overall response of CR, PR or SD for greater than six months and primary resistance defined as PD or SD for less than six months, a difference in FS118 treatment duration was not observed between acquired resistance versus primary resistance patients (Supplementary Fig. S2B).

Using our three-month resistance definition, no patients with primary resistance were on study beyond 20 weeks of FS118 treatment, compared with a subset of patients with acquired resistance (Supplementary Fig. S2A and Fig. 1A and B). Moreover, the DCR in patients with an acquired resistance phenotype was 54.8% (17 out of 31 patients) in patients receiving dose levels of 1 mg/kg or above, compared with 25.0% (two out of eight patients) in patients with primary resistance. In addition, patients who were treated with ICB therapy as their last prior treatment before treatment with FS118 (this included anti–TIM-3 or anti–CTLA-4 in addition to anti–PD-(L)1 therapy) achieved a significantly longer treatment duration (P = 0.029) on FS118 (median = 11.1 weeks) compared with patients who did not have ICB therapy as their last prior treatment (median = 7.3 weeks; Fig. 1C). Of those nine patients who surpassed 20 weeks treatment, eight patients were characterized by both acquired resistance to anti–PD-(L)1 therapy and receipt of any ICB as their last prior therapy, suggesting that this population of patients may have the greatest benefit from FS118 therapy. No clear correlation between time on prior ICB regimens and FS118 treatment was observed (Supplementary Fig. S3).

Correlations of PD-L1 and LAG-3 target expressions with clinical activity

Suitable pre-treatment tumor core biopsies were obtained from 28 of 43 patients and were evaluated for PD-L1 and LAG-3 tumor expression. Of these, 17 patients were double positive for both PD-L1 and LAG-3 (60.7%), six were LAG-3 single positive (21.4%) and three were single positive for PD-L1 (10.7%; Fig. 2). Two patients were positive for LAG-3 with unknown PD-L1 expression. No patients were negative for both PD-L1 and LAG-3. The patient with ATC, whose PD-L1 and LAG-3 status were unknown when starting on this study, was later confirmed to have tumor-expressing PD-L1 and LAG-3 from an archival biopsy collected 15 months earlier. Although none of the LAG-3 single positive patients remained on study for more than 20 weeks, of the nine evaluable patients that surpassed 20 weeks of FS118 therapy, all with acquired resistance to prior anti–PD-(L)1 therapy, five demonstrated expression of both LAG-3 and PD-L1 in the tumor microenvironment (Fig. 2).

Figure 2.

LAG-3 and PD-L1 baseline expression and treatment duration in FS118-treated patients. LAG-3 and PD-L1 tumor expression in baseline biopsies from patients with acquired (blue) or primary (red) resistance. LAG-3+ defined as one or more high-power fields within the intraepithelial tumor or intratumoural stroma compartments; PD-L1+ defined as a TPS ≥1. ATC, anaplastic thyroid carcinoma; CERV, cervical; MPM, malignant peritoneal mesothelioma; NSCLC, non–small cell lung carcinoma; OVRY, ovarian; SARC, sarcoma; and SCCHN, head and neck squamous cell carcinoma.

Figure 2.

LAG-3 and PD-L1 baseline expression and treatment duration in FS118-treated patients. LAG-3 and PD-L1 tumor expression in baseline biopsies from patients with acquired (blue) or primary (red) resistance. LAG-3+ defined as one or more high-power fields within the intraepithelial tumor or intratumoural stroma compartments; PD-L1+ defined as a TPS ≥1. ATC, anaplastic thyroid carcinoma; CERV, cervical; MPM, malignant peritoneal mesothelioma; NSCLC, non–small cell lung carcinoma; OVRY, ovarian; SARC, sarcoma; and SCCHN, head and neck squamous cell carcinoma.

Close modal

PK and pharmacodynamics

FS118 exhibited dose-linear PK within the evaluated dose range of 1–20 mg/kg (Fig. 3A; Supplementary Table S3) with a rapid phase half-life of 23.3 hours (one day) and a terminal half-life of 92.4 hours (3.9 days), as determined by PK data fit to a biexponential decay, characteristic of a two-compartment model.

Figure 3.

Pharmacokinetics (PK) and pharmacodynamics of FS118. A, Dose-exposure PK analysis of free FS118 in patients’ serum following the first dose of FS118 (cycle 1 and week 1) at 1 mg/kg and above. Data plotted as mean, with error bars showing standard deviation. Assay LLOQ, 100 ng/mL; values calculated BLQ are not included. B, Total soluble LAG-3 (sLAG-3) detected in the serum of patients treated with FS118 at 1 mg/kg or above, over the first 7 days of treatment. Data shown as AUC0–7. Data shown as median values, and the Mann–Whitney two-tailed test with Benjamani–Hochberg correction applied to the P values. *, P < 0.05. C, Total sLAG-3 over treatment cycles. Concentration of sLAG-3 for each cohort of 1 mg/kg or above of FS118, at pre-dose samples for cycles 1, 2, 3, and 4. Data shown as median ± 95% confidence interval. D–F, Percentages of circulating CD4+, CD8+ T cells, and NK cells within CD45+ cells in patients before FS118 treatment (baseline) of 1 mg/kg and above, compared with 6 weeks following the first dose of FS118 (C03D01), by best overall response as per RECIST 1.1. Data from individual patients with acquired (blue) or primary (red) resistance to prior PD-(L)1 ICB therapy. Cell percentage from total leukocyte populations (CD45+). *, P < 0.05 (Wilcoxon signed-rank test). BLQ, below the limit of quantification; LLOQ, lower limit of quantification; PD, progressive disease; SD, stable disease.

Figure 3.

Pharmacokinetics (PK) and pharmacodynamics of FS118. A, Dose-exposure PK analysis of free FS118 in patients’ serum following the first dose of FS118 (cycle 1 and week 1) at 1 mg/kg and above. Data plotted as mean, with error bars showing standard deviation. Assay LLOQ, 100 ng/mL; values calculated BLQ are not included. B, Total soluble LAG-3 (sLAG-3) detected in the serum of patients treated with FS118 at 1 mg/kg or above, over the first 7 days of treatment. Data shown as AUC0–7. Data shown as median values, and the Mann–Whitney two-tailed test with Benjamani–Hochberg correction applied to the P values. *, P < 0.05. C, Total sLAG-3 over treatment cycles. Concentration of sLAG-3 for each cohort of 1 mg/kg or above of FS118, at pre-dose samples for cycles 1, 2, 3, and 4. Data shown as median ± 95% confidence interval. D–F, Percentages of circulating CD4+, CD8+ T cells, and NK cells within CD45+ cells in patients before FS118 treatment (baseline) of 1 mg/kg and above, compared with 6 weeks following the first dose of FS118 (C03D01), by best overall response as per RECIST 1.1. Data from individual patients with acquired (blue) or primary (red) resistance to prior PD-(L)1 ICB therapy. Cell percentage from total leukocyte populations (CD45+). *, P < 0.05 (Wilcoxon signed-rank test). BLQ, below the limit of quantification; LLOQ, lower limit of quantification; PD, progressive disease; SD, stable disease.

Close modal

A dose-dependent increase in total sLAG-3 was observed (Fig. 3B), indicating target engagement and suggesting that FS118 mediates LAG-3 shedding. Levels of sLAG-3 were higher in the 10 and 20 mg/kg cohorts compared with 1 and 3 mg/kg across the first four cycles of treatment (Fig. 3C). Moreover, no statistical differences in the levels of soluble LAG-3 were observed between 10 and 20 mg/kg (Fig. 3B). Peak levels of sLAG-3 were observed two to four days after administration of FS118 and elevation was maintained throughout the dosing interval in the 10 and 20 mg/kg cohorts (Supplementary Fig. S4A and S4B). Trough sLAG-3 concentrations at the end of each cycle were elevated compared with baseline levels (Fig. 3C), demonstrating the prolonged pharmacodynamic effect of FS118. There was no significant difference in baseline levels of sLAG-3 in those patients with acquired resistance compared with those with primary resistance to prior PD-(L)1 therapy (Supplementary Fig. S4C).

Analysis of immune cell subsets in the periphery revealed an increase in the percentage of circulating lymphocytes in patients who demonstrated SD, compared with those with PD, as their best response on FS118 treatment (Fig. 3DF). The proportions of CD4+ and CD8+ T cells in patients with SD were significantly higher (P = 0.015 and P = 0.021, respectively) six weeks after initiating FS118 therapy compared with their baseline level (Fig. 3D and E). In comparison, patients with PD demonstrated significantly reduced circulating CD8+ T cells (P = 0.014) and NK cells (P = 0.014), and a trend toward reduced circulating CD4+ cells following FS118 treatment. The percentage of NK cells also showed a trend toward an increase in patients with SD after six weeks of treatment, but this was not found to be statistically significant (P=0.165; Fig. 3F). In support of the increased peripheral lymphocytes observed in patients with SD, a significant increase in the percentage of proliferating (Ki67+) CD4+ and CD8+ T cells and NK cells (P = 0.039, P = 0.039, and P = 0.0053, respectively) was observed seven days after the first dose of FS118. A similar increase in the percentage of proliferating (Ki67+) CD4+ and CD8+ T cells was also observed in patients with PD (P = 0.010 and P = 0.047, respectively) though as stated above, this did not correlate with an increase in peripheral lymphocytes in these patients (Supplementary Fig. S5A–S5C). These effects on peripheral lymphocytes were shown not to be dose-dependent (Supplementary Fig. S5D–S5F).

On the basis of the evident good safety profile of FS118, lower frequency of immunogenicity at dose levels ≥10 mg/kg, PK and observed pharmacodynamic activity (increased immune cell proliferation and sustained increased sLAG-3 over the dosing period), 10-mg/kg dose level was selected for further evaluation in the clinic.

In this first-in-human phase 1 dose-escalation and expansion study, FS118 was well tolerated with no DLTs observed up to the maximum administered dose of 20 mg/kg, and the MTD was not reached. The RP2D was established at 10 mg/kg QW. PK showed a terminal half-life of 3.9 days. Pharmacodynamic changes consistent with the bispecific mechanism of action of FS118 were observed. SD was observed, particularly in patients with acquired resistance to PD-(L)1 blockade, ICB as their most recent therapy and/or co-expression of LAG-3 and PD-L1 in the tumor.

FS118 demonstrated a good safety profile following weekly intravenous administration of FS118 at all dose levels assessed up to and including 20 mg/kg, characterized by the occurrence of mainly grade 1 and 2 TRAEs. In this study, TRAEs were observed in 58.1% of patients, with grade ≥3 TRAEs observed in only two patients (4.7%), and no adverse events were dose-limiting. The TRAE rate of FS118 compares favorably with PD-L1 inhibitors, where TRAE rates of 63.9%–81.1% (grade ≥3 rates of 4.9-17.0%) have been observed in a phase 1 ICB-naïve clinical setting (32, 33), and to the combination of LAG-3 and PD-1 mAbs, where TRAE rates of 81.1% (grade ≥3 rates of 18.9%) were observed with relatlimab and nivolumab combination (34) and TRAE rates of 69.4% (grade ≥3 rates of 9.1%) were observed with ieramilimab and spartlizumab combination (25). These data indicate that FS118 may have the potential to be better tolerated than PD-L1–targeting therapies alone or a combination of PD-1 and LAG-3–targeting therapies.

The patients enrolled on this study were heavily pre-treated, suggesting that the low rate of grade ≥3 TRAEs observed with FS118 therapy may have resulted from the pre-selection of patients with better tolerance to ICB therapies. We also speculate that the favorable safety profile of FS118 may also be attributed to its unique mechanism of action and localized activity in the tumor microenvironment. Tumor-localized FS118 activity may also explain the relatively rapid clearance of FS118 from the serum when compared with other PD-L1 mAbs (35).

SD was observed in 20 of the heavily pre-treated 43 patients (46.5%), with nine patients (20.9%) demonstrating long-lasting disease control for over 20 weeks. Of these nine patients, two patients had SCCHN and two patients had NSCLC. A patient with advanced ATC and a patient with advanced leiomyosarcoma who both presented with acquired resistance to recent PD-(L)1 therapy, demonstrated the longest time on FS118 treatment of 79 weeks and 56.1 weeks, respectively (at data cutoff value). Moreover, as of March 2022, the ATC patient remained on FS118 treatment for over three years with disease control, including a late confirmed PR.

On the basis of our observations, we speculate that upregulation of LAG-3 and PD-L1 may be markers of acquired resistance. Upregulation of LAG-3 on TILs in anti–PD-1–resistant tumors has been documented preclinically (7, 8, 36, 37) and clinically (36, 38). In this study, 60.7% of evaluable patients’ tumors were positive for both LAG-3 and PD-L1 before the start of FS118 treatment. Studies in melanoma targeting LAG-3 in patients with resistance to anti–PD-(L)1 therapies have demonstrated preliminary clinical benefit (39) and more recently, a study assessing the combination of fianlimab (anti–LAG-3) and cemiplimab (anti–PD-1) reported a much lower overall response rate in patients with melanoma previously exposed to prior PD-(L)1 inhibitors compared with ICB-naïve patients (13.3% and 63.6% respectively; ref. 40). Furthermore, in a phase 1/2 study assessing anti–PD-1 and LAG-3, all responding patients had not received prior checkpoint therapy (25).

In this study, a DCR of 54.8% (17 out of 31 patients) was observed in patients with acquired resistance to prior PD-(L)1 ICB therapy who were treated with at least 1 mg/kg FS118 (Fig. 1A). Given that blockade of the PD-(L)1 axis may result in upregulation of LAG-3 to drive acquired resistance (6, 7, 36), modulation of the LAG-3 pathway in addition to PD-L1 blockade by FS118 may be an approach to address acquired resistance. However, there is a possibility that the disease control observed in this study could be due to re-challenge with PD-L1 blockade, as it has been demonstrated that re-challenge with immune checkpoint inhibitors can result in re-induction of responses in ICB-sensitive tumor types (41). Further clinical studies in acquired resistance patients will be required to assess the potential for FS118 to provide patient benefit in this setting.

Pharmacological activity associated with FS118 was observed as increases in circulating and proliferating lymphocytes. Increases in CD4+ and CD8+ T cells following treatment with FS118 observed in patients with stable disease, but not in patients with progressive disease, may indicate that a T-cell–mediated immune response is a good prognostic indicator. A sustained elevation of soluble LAG-3 was achieved in the serum as a dose-dependent marker of FS118 activity, indicating prolonged pharmacodynamic activity and a saturation of LAG-3 engagement is reached at 10 mg/kg. On the basis of our preclinical data, we hypothesize that LAG-3 shedding drives this increase (42), although it is possible that alternative splicing of LAG-3 contributes to this effect (12). As LAG-3 expression has been associated with an exhaustive T-cell phenotype, and with LAG-3+ NK cells with impaired cytotoxic functions (43), it could be hypothesized that shedding of LAG-3 from the cell surface of effector cells by FS118 may reverse these phenotypes, as supported by data herein demonstrating increased peripheral effector cells following FS118 treatment.

The highest DCR was observed at 10 and 20 mg/kg (53.8% of patients in both cohorts), indicating that optimal pharmacology and antitumor activity were achieved at dose levels of 10 mg/kg and above. This is further supported by the pharmacology data showing that soluble LAG-3 was not markedly increased in serum samples of patients dosed at 20 mg/kg compared with serum samples of patients dosed at 10 mg/kg, suggesting that a higher concentration of FS118 does not necessarily translate to increased activity and a threshold for activity may have already been reached at 10 mg/kg. In further support of this, no pharmacodynamic differences in circulating lymphocytes were observed between dose levels, which suggest saturation of pharmacological effects at lower dose levels. In addition, although the majority of ADAs observed were shown to be transient, two patients from 1 to 3 mg/kg cohorts (from a total of n=3 and n=10 patients, respectively) presented with ADAs for more than three consecutive cycles, consistent with other observations that lower doses of biotherapeutics can be more immunogenic than higher doses (44). Taken together, these data were used to select a RP2D of 10 mg/kg weekly for future FS118 studies.

In this first-in-human study, FS118 was well tolerated without DLT, and the safety profile compared favorably to other ICB therapies. Stabilization of disease was observed in a subset of patients and a RP2D was identified for future studies. Further assessment is warranted to assess the clinical benefit FS118 may bring to patients who have progressed on prior anti–PD-(L)1 therapy. A phase 2 clinical trial exploring the antitumor activity of FS118 in patients with PD-L1- and LAG-3–positive SCCHN with acquired resistance to PD-(L)1 inhibitor therapy is ongoing (ClinicalTrials.gov: NCT03440327).

T.A. Yap reports other support from MD Anderson Cancer Center and Seagen; grants and personal fees from Acrivon, Artios, AstraZeneca, Bayer, BeiGene, Clovis, EMD Serono, F-star, Merck, Pfizer, and Repare; grants from BioNTech, Blueprint, BMS, Constellation, Cyteir, Eli Lilly, Forbius, GlaxoSmithKline, Genentech, Haihe, ImmuneSensor, Ionis, Ipsen, Jounce, Karyophar, KSQ, Kyowa, Mirati, Novartis, Ribon Therapeutics, Regeneron, Rubius, Sanofi, Scholar Rock, Seattle Genetics, Tesaro, Vivace, and Zenith; and personal fees from AbbVie, Acrivon, Adagene, Almac, Aduro, Amphista, Athena, Atrin, Avoro, Axiom, Baptist Health Systems, Boxer, Bristol Myers Squibb, C4 Therapeutics, Calithera, Cancer Research UK, Cybrexa, Diffusion, Genmab, Glenmark, GLG, Globe Life Sciences, GSK, Guidepoint, Idience, Ignyta, I-Mab, ImmuneSensor, Institut Gustave Roussy, Intellisphere, Janssen, Kyn, MEI Pharma, Mereo, Natera, Nexys, Novocure, OHSU, OncoSec, Ono Pharma, Pegascy, PER, Piper-Sandler, Prolynx, ResTORbio, Roche, Schrodinger, Theragnostics, Varian, Versant, Vibliome, Xinthera, Zai Labs, and ZielBio during the conduct of the study. P.M. LoRusso reports other support from AbbVie, Agios, Five Prime, GenMab, Halozyme, Roche-Genentech, Genentech, CytomX, Takeda, SOTIO, Cybrexa, Agenus, Tyme, IQVIA, TRIGR, Pfizer, ImmunoMet, Black Diamond, GlaxoSmithKline, QED Therapeutics, AstraZeneca, EMD Serono, Shattuck, Astellas, Salarius, Silverback, MacroGenics, Kyowa Kirin, Kineta, Zentalis, Molecular Templates, ABL Bio, SK Life Science, STCube, Bayer, I-MAB, Seagen, imCheck, Relay Therapeutics, Stemline, Compass BADX, Mekanist, Mersana, BAKX Therapeutics, Scenic Biotech, Qualigen, Roivant, and NeurotTrials outside the submitted work. D.J. Wong reports grants from F-star during the conduct of the study as well as grants from Bristol-Myers Squibb, Genentech/Roche, Sanofi, Blueprints Medicines, Lilly, Merck Sharp and Dohme, AstraZeneca, Kura Oncology, Enzychem Lifesciences, Elevar Therapeutics, Top Alliance BioSciences, Checkmate Pharmaceuticals, Bicara Therapeutics, and Gilead Sciences and grants and personal fees from Regeneron outside the submitted work. K.P. Papadopoulos reports other support from F-star during the conduct of the study as well as personal fees from Basilia, Bicycle, and Turning Point Therapeutics and other support from 3D Medicines, AbbVie, ADC Therapeutics, Amgen, Anheart, Bayer, Daiichi Sankyo, Incyte, Jounce Therapeutics, Lilly, Linnaeus, Merck, Mersana, Mirati, Pfizer, Regeneron, Revolution Medicines, Syros Pharmaceuticals, Tempest Therapeutics, and Treadwell Therapeutics outside the submitted work. J.-B. Holz reports personal fees from F-star during the conduct of the study as well as personal fees from F-star outside the submitted work. R.C.A. Sainson reports other support from F-star during the conduct of the study. C.J. Shepherd reports holding shares in F-star during the conduct of the study. F. Germaschewski reports service as an independent biomarker consultant supporting Biotech/Pharma and CRUK; however, there is no conflict of interest with F-star with bispecifics or the specific targets; biotechs supported are NovalGen, Evgen Pharma, UCB, Ellipses Pharma, Molecular Templates, and Sqz Biotech; current biotechs supported are Apollo Therapeutics, Tribune Therapeutics, Orchard Therapeutics, Dynamicure, and CRUK. D. Gliddon reports a patent for WO2020229626A1 pending. L. Kayitalire reports other support from F-star during the conduct of the study as well as other support from F-star outside the submitted work. No disclosures were reported by the other authors.

T.A. Yap: Resources, investigation, writing–review and editing. P.M. LoRusso: Resources, investigation, writing–review and editing. D.J. Wong: Resources, investigation, writing–review and editing. S. Hu-Lieskovan: Resources, investigation, writing–review and editing. K.P. Papadopoulos: Resources, investigation, writing–review and editing. J.-B. Holz: Conceptualization, data curation, methodology, project administration, writing–review and editing. U. Grabowska: Validation, project administration, writing–review and editing. C. Gradinaru: Data curation, formal analysis, methodology, writing–review and editing. K.-M. Leung: Supervision, validation, investigation, visualization, writing–review and editing. S. Marshall: Supervision, validation, investigation, visualization, writing–review and editing. C.S. Reader: Data curation, validation, visualization, writing–original draft, writing–review and editing. R. Russell: Data curation, software, formal analysis, validation, investigation, visualization, methodology, writing–review and editing. R.C.A. Sainson: Supervision, validation, writing–review and editing. C.J. Seal: Data curation, validation, visualization, writing–original draft, writing–review and editing. C.J. Shepherd: Data curation, supervision, validation, investigation, writing–original draft, writing–review and editing. F. Germaschewski: Data curation, investigation, methodology, writing–review and editing. D. Gliddon: Conceptualization, supervision, investigation, methodology, writing–review and editing. O. Stern: Investigation, visualization, methodology, writing–original draft, writing–review and editing. L. Young: Conceptualization, supervision, writing–review and editing. N. Brewis: Conceptualization, supervision, writing–review and editing. L. Kayitalire: Supervision, writing–review and editing. M. Morrow: Supervision, writing–original draft, project administration, writing–review and editing.

This study was funded and supported by F-star Therapeutics Ltd., Cambridge, UK. We thank the patients, their families, investigators, and research staff at all study-sites. The authors would also like to thank the F-star team for support with study operations, data analysis and QC, and critical review of this article.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Petitprez
F
,
Meylan
M
,
de Reyniès
A
,
Sautès-Fridman
C
,
Fridman
WH
.
The tumor microenvironment in the response to immune checkpoint blockade therapies
.
Front Immunol
2020
;
11
:
784
.
2.
Okudaira
K
,
Hokari
R
,
Tsuzuki
Y
,
Okada
Y
,
Komoto
S
,
Watanabe
C
, et al
.
Blockade of B7-H1 or B7-DC induces an antitumor effect in a mouse pancreatic cancer model
.
Int J Oncol
2009
;
35
:
741
9
.
3.
Topalian
SL
,
Drake
CG
,
Pardoll
DM
.
Targeting the PD-1/B7-H1(PD-L1) pathway to activate antitumor immunity
.
Curr Opin Immunol
2012
;
24
:
207
212
.
4.
Gettinger
S
,
Rizvi
NA
,
Chow
LQ
,
Borghaei
H
,
Brahmer
J
,
Ready
N
, et al
.
Nivolumab monotherapy for first-line treatment of advanced non–small cell lung cancer
.
J Clin Oncol
2016
;
34
:
2980
2987
.
5.
Stenehjem
DD
,
Tran
D
,
Nkrumah
MA
,
Gupta
S.
PD1/PDL1 inhibitors for the treatment of advanced urothelial bladder cancer
.
Onco Targets Ther
.
2018
;
11
;
5973
-
5989
.
6.
Sharma
P
,
Hu-Lieskovan
S
,
Wargo
JA
,
Ribas A.
Primary, adaptive, and acquired resistance to cancer immunotherapy
.
Cell
.
2017
;
168
:
707
723
.
7.
Koyama
S
,
Akbay
EA
,
Li
YY
,
Herter-Sprie
GS
,
Buczkowski
K A
,
Richards
W G
, et al
.
Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints
.
Nat Commun
2016
;
7
:
10501
.
8.
Huang
R-Y
,
Francois
A
,
McGray
AR
,
Miliotto
A
,
Odunsi
K.
Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer
.
Oncoimmunology
.
2017
;
6
:
e1249561
.
9.
Durham
NM
,
Nirschl
CJ
,
Jackson
CM
,
Elias
J
,
Kochel
CM
,
Anders
RA
, et al
.
Lymphocyte activation Gene 3 (LAG-3) modulates the ability of CD4 T cells to be suppressed in vivo
.
PLoS ONE
2014
;
9
:
e109080
.
10.
Huang
C-T
,
Workman
CJ
,
Flies
D
,
Pan
X
,
Marson
AL
,
Zhou
G
, et al
.
Role of LAG-3 in regulatory T cells
.
Immunity
.
2004
;
21
:
503
513
.
11.
Sega
EI
,
Leveson-Gower
DB
,
Florek
M
,
Schneidawind
D
,
Luong
RH
,
Negrin
RS
.
Role of lymphocyte activation gene-3 (Lag-3) in conventional and regulatory T-cell function in allogeneic transplantation
.
PLoS ONE
2014
;
9
:
e86551
.
12.
Graydon
CG
,
Mohideen
S
,
Fowke
KR
.
LAG3’s enigmatic mechanism of action
.
Front Immunol
2020
;
11
:
615317
.
13.
Li
N
,
Wang
Y
,
Forbes
K
,
Vignali
KM
,
Heale
BS
,
Saftig
P
, et al
.
Metalloproteases regulate T-cell proliferation and effector function via LAG-3
.
EMBO J
2007
;
26
:
494
504
.
14.
Andrews
LP
,
Somasundaram
A
,
Moskovitz
JM
,
Szymczak-Workman
AL
,
Liu
C
,
Cillo
AR
, et al
.
Resistance to PD1 blockade in the absence of metalloprotease-mediated LAG3 shedding
.
Sci Immunol
2020
;
5
:
eabc2728
.
15.
Woo
S-R
,
Turnis
ME
,
Goldberg
MV
,
Bankoti
J
,
Selby
M
,
Nirschl
CJ
, et al
.
Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape
.
Cancer Res
2012
;
72
:
917
927
.
16.
Andrews
LP
,
Marciscano
AE
,
Drake
CG
,
Vignali
DAA
.
LAG3 (CD223) as a cancer immunotherapy target
.
Immunol Rev
2017
;
276
:
80
96
.
17.
Matsuzaki
J
,
Gnjatic
S
,
Mhawech-Fauceglia
P
,
Beck
A
,
Miller
A
,
Tsuji
T
, et al
.
Tumor-infiltrating NY-ESO-1–specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer
.
Proc Natl Acad Sci U S A
2010
;
107
:
7875
7880
.
18.
Baitsch
L
,
Baumgaertner
P
,
Devêvre
E
,
Raghav
SK
,
Legat
A
,
Barba
L
, et al
.
Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients
.
J Clin Invest
2011
;
121
:
2350
2360
.
19.
Seifert
L
,
Plesca
I
,
Müller
L
,
Sommer
U
,
Heiduk
M
,
von Renesse
J
, et al
.
LAG-3-expressing tumor-infiltrating T cells are associated with reduced disease-free survival in pancreatic cancer
.
Cancers
.
2021
;
13
:
1297
.
20.
Lee
I
,
Lee
SJ
,
Kang
BW
,
Chae
YS
,
Kim
HJ
,
Park
SY
, et al
.
LAG-3 expression in tumor infiltrating immune cells is associated with poor prognosis in patients with microsatellite instability high colon cancer
.
Ann Oncol
2017
;
28
;
V196
.
21.
Deng
W-W
,
Mao
L
,
Yu
G-T
,
Bu
L-L
,
Ma
S-R
,
Liu
B
, et al
.
LAG-3 confers poor prognosis and its blockade reshapes antitumor response in head and neck squamous cell carcinoma
.
OncoImmunology
.
2016
;
5
:
e1239005
.
22.
Long
L
,
Zhang
X
,
Chen
F
,
Pan
Q
,
Phiphatwatchara
P
,
Zeng
Y
, et al
.
The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy
.
Genes Cancer
2018
;
9
:
176
189
.
23.
Brignone
C
,
Escudier
B
,
Grygar
C
,
Marcu
M
,
Triebel
F.
A Phase I pharmacokinetic and biological correlative study of IMP321, a Novel MHC class II agonist, in patients with advanced renal cell carcinoma
.
Clin Cancer Res
2009
;
15
:
6225
6231
.
24.
Lythgoe
MP
,
Liu
DSK
,
Annels
NE
,
Krell
J
,
Frampton
A E
.
Gene of the month: lymphocyte-activation gene 3 (LAG-3)
.
J Clin Pathol
2021
;
74
:
543
547
.
25.
Schöffski
P
,
Tan
DSW
,
Martín
M
,
Ochoa-de-Olza
M
,
Sarantopoulos
J
,
Carvajal
RD
, et al
.
Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) ± anti–PD-1 spartalizumab (PDR001) in patients with advanced malignancies
.
J Immunother Cancer
2022
;
10
:
e003776
.
26.
Wang
J
,
Asch
AS
,
Hamad
N
,
Weickhardt
A
,
Tomaszewska-Kiecana
M
,
Dlugosz-Danecka
M
, et al
.
A phase 1, Open-label study of MGD013, a Bispecific DART molecule–binding PD-1 and LAG-3 in patients with relapsed or refractory diffuse large B-cell lymphoma
.
Blood
.
2020
;
136
:
21
22
.
27.
Everett
KL
,
Kraman
M
,
Wollerton
FPG
,
Zimarino
C
,
Kmiecik
K
,
Gaspar
M
, et al
.
Generation of Fcabs targeting human and murine LAG-3 as building blocks for novel bispecific antibody therapeutics
.
Methods
2019
;
154
:
60
69
.
28.
Kraman
M
,
Faroudi
M
,
Allen
NL
,
Kmiecik
K
,
Gliddon
D
,
Seal
C
, et al
.
FS118, a bispecific antibody targeting LAG-3 and PD-L1, enhances T-cell activation resulting in potent antitumor activity
.
Clin Cancer Res
2020
;
26
:
3333
3344
.
29.
Arduin
E
,
Arora
S
,
Bamert
PR
,
Kuiper
T
,
Popp
S
,
Geisse
S
, et al
.
Highly reduced binding to high and low-affinity mouse Fc gamma receptors by L234A/L235A and N297A Fc mutations engineered into mouse IgG2a
.
Mol Immunol
2015
;
63
:
456
463
.
30.
Yap
T
,
Wong
D
,
Hu-Lieskovan
S
,
Papadopoulos
K
,
Morrow
M
,
Grabowska
U
, et al
.
395 A first-in-human study of FS118, a tetravalent bispecific antibody targeting LAG-3 and PD-L1, in patients with advanced cancer and resistance to PD-(L)1 therapy
.
J Immunother Cancer
2020
;
8
:
A240
.
31.
Kluger
HM
,
Tawbi
HA
,
Ascierto
ML
,
Bowden
M
,
Callahan
MK
,
Cha
E
, et al
.
Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce
.
J Immunother Cancer
2020
;
8
:
e000398
.
32.
Heery
CR
,
O'Sullivan-Coyne
G
,
Madan
RA
,
Cordes
L
,
Rajan
A
,
Rauckhorst
M
, et al
.
Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN Solid Tumor): a phase 1a, multicohort, dose-escalation trial
.
Lancet Oncol
2017
;
18
:
587
598
.
33.
Massard
C
,
Gordon
MS
,
Sharma
S
,
Rafii
S
,
Wainberg
Z A
,
Luke
J
, et al
.
Safety and efficacy of durvalumab (MEDI4736), an anti–programmed cell death Ligand-1 immune checkpoint inhibitor, in patients with advanced Urothelial bladder cancer
.
J Clin Oncol
2016
;
34
:
3119
3125
.
34.
Tawbi
HA
,
Schadendorf
D
,
Lipson
EJ
,
Ascierto
PA
,
Matamala
L
,
Gutiérrez
EC
, et al
.
Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma
.
N Engl J Med
2022
;
386
:
24
34
.
35.
Sheng
J
,
Srivastava
S
,
Sanghavi
K
,
Lu
Z
,
Schmidt
BJ
,
Bello
A
, et al
.
Clinical pharmacology considerations for the development of immune checkpoint inhibitors
.
J Clin Pharmacol
.
2017
;
57
;
S26
-S42
36.
Johnson
DB
,
Nixon
MJ
,
Wang
Y
,
Wang
DY
,
Castellanos
E
,
Estrada
MV
, et al
.
Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement
.
JCI Insight
2018
;
3
;
e120360
.
37.
Wierz
M
,
Pierson
S
,
Guyonnet
L
,
Viry
E
,
Lequeux
A
,
Oudin
A
, et al
.
Dual PD1/LAG3 immune checkpoint blockade limits tumor development in a murine model of chronic lymphocytic leukemia
.
Blood
.
2018
;
131
:
1617
1621
.
38.
Gide
TN
,
Quek
C
,
Menzies
AM
,
Tasker
AT
,
Shang
P
,
Holst
J
, et al
.
Distinct immune cell populations define response to anti–PD-1 monotherapy and anti–PD-1/anti–CTLA-4 combined therapy
.
Cancer Cell
.
2019
;
35
;
238
-
255
.e6
39.
Ascierto
PA
,
Bono
P
,
Bhatia
S
,
Melero
I
,
Nyakas
MS
,
Svane
I-M
, et al
.
Efficacy of BMS-986016, a monoclonal antibody that targets lymphocyte activation gene-3 (LAG-3), in combination with nivolumab in pts with melanoma who progressed during prior anti–PD-1/PD-L1 therapy (mel prior IO) in all-comer and biomarker-enriched populations
.
Ann Oncol
2017
;
28
:
v611
–v612.
40.
Hamid
O
,
Wang
D
,
Kim
TM
,
Kim
S-W
,
Lakhani
NJ
,
Johnson
M L
, et al
.
Clinical activity of fianlimab (REGN3767), a human anti–LAG-3 monoclonal antibody, combined with cemiplimab (anti–PD-1) in patients (pts) with advanced melanoma
.
J Clin Oncol
2021
;
39
:
9515
9515
.
41.
Yang
K
,
Li
J
,
Sun
Z
,
Zhao
L
,
Bai
C.
Retreatment with immune checkpoint inhibitors in solid tumors: a systematic review
.
Ther Adv Med Oncol
.
2020
;
27
;12:
1758835920975353
42.
Morrow
M
,
Faroudi
M
,
Chakraborty
K
,
Liao
W
,
Winnewisser
J
,
Seal
C
, et al
.
715 FS118, a tetravalent bispecific antibody targeting LAG-3 and PD-L1, induces LAG-3 shedding resulting in receptor downregulation by T cells via a novel mechanism of action
.
J Immunother Cancer
2020
;
8
:
A428
.
43.
Esen
F
,
Deniz
G
,
Aktas
EC
.
PD-1, CTLA-4, LAG-3, and TIGIT: the roles of immune checkpoint receptors on the regulation of human NK cell phenotype and functions
.
Immunol Lett
2021
;
240
:
15
23
.
44.
Kuriakose
A
,
Chirmule
N
,
Nair
P.
Immunogenicity of Biotherapeutics: causes and association with posttranslational modifications
.
J Immunol Res
2016
;
2016
:
1
18
.

Supplementary data