Phase 1 study of fianlimab, a human lymphocyte activation gene-3 monoclonal antibody, in combination with cemiplimab in advanced melanoma: Expansion cohort analysis Omid Hamid,1 Karl Lewis,2 Amy Weise,3 Meredith McKean,4 Kyriakos P Papadopoulos,5 John Crown,6 Tae Min Kim,7 Nehal J Lakhani,8 John Kaczmar,9 Ragini Kudchadkar,10 Alexander Spira,11 Guilherme Rabinowits,12 Kevin Kim,13 Richard Carvajal,14 Stephen Williamson,15 Ella Ioffe,16 Shuquan Chen,16 Jayakumar Mani,16 Vladimir Jankovic,16 Laura Brennan,16 Glenn Kroog,16 Tasha Sims,16* Israel Lowy,16 Giuseppe Gullo16 1The Angeles Clinical and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, CA, USA; 2University of Colorado Hospital, CO, USA; 3Henry Ford Hospital, Detroit, MI, USA; 4Sarah Cannon Research Institute/Tennessee Oncology PLLC, Nashville, TN, USA; 5START San Antonio, San Antonio, TX, USA; 6St Vincent’s University Hospital, Dublin, Ireland; 7Seoul National University Hospital, Seoul, South Korea; 8START Midwest, Grand Rapids, MI, USA; 9MUSC Hollings Cancer Center, North Charleston, SC, USA; 10Emory University School of Medicine, Atlanta GA, USA; 11Virginia Cancer Specialists, Fairfax, VA, USA; 12Miami Cancer Institute, Miami, FL, USA; 13Sutter Health Research Enterprise, San Francisco, CA, USA; 14Columbia University Medical Center, New York, NY, USA; 15University of Kansas Medical Center, Fairway, KS, USA; 16Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA *Formerly with Regeneron Pharmaceuticals, Inc. Funding Funding was provided by Regeneron Pharmaceuticals, Inc. Table 1. Baseline characteristics and disposition: Anti–PD-L1 naive (Cohorts 6 + 15)† Characteristic Anti–PD-L1 naive† Cohorts 6 + 15 (N=80)Cohort 6 (N=40) Cohort 15 (N=40) Age Median (range), years 69.5 (27–85) 69.0 (24–88) 69.0 (24–88) ≥65 years, % (n) 60.0 (24) 62.5 (25) 61.3 (49) Male, % (n) 62.5 (25) 57.5 (23) 60.0 (48) White, % (n) 90.0 (36) 90.0 (36) 90.0 (72) SOD of TL at baseline, median (range), mm 51 (15–214) 52 (11–173) 51.5 (11–214) BRAF mutant, % (n) 27.5 (11) 30.0 (12) 28.8 (23) Melanoma subtype, % (n) Acral 7.5 (3) 5.0 (2) 6.2 (5) Mucosal 2.5 (1) 0 1.2 (1) Cutaneous nonacral 90.0 (36) 95.0 (38) 92.5 (74) Metastasis stage at baseline, % (n) M0 15.0 (6) 5.0 (2) 10.0 (8) M1 82.5 (33) 87.5 (35) 85.0 (68) M1c‡ 45.0 (18) 22.5 (9) 33.8 (27) LDH > ULN, % (n) 42.5 (17) 27.5 (11) 35.0 (28) Liver metastases, % (n) 35.0 (14) 12.5 (5) 23.8 (19) Previous systemic therapy, % (n) 20.0 (8)§ 0 10.0 (8) †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. ‡M1 category includes M1, M1a, M1b, and M1c. §Two patients received prior treatment for advanced disease and six patients received prior adjuvant treatment. LDH, lactate dehydrogenase; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; SOD, sum of diameters; TL, target lesion; ULN, upper limit of normal. Table 2. Tumor response among anti–PD-L1–naive patients (Cohorts 6 + 15)† % (n), unless otherwise stated Anti–PD-L1 naive† Cohorts 6 + 15 (N=80)Cohort 6 (N=40) Cohort 15 (N=40) ORR, % (95% CI) 62.5 (45.8, 77.3) 65 (48.3, 79.4) 63.8 (52.2, 74.2) Complete response 15.0 (6) 2.5 (1) 8.8 (7) Partial response 47.5 (19) 62.5 (25) 55.0 (44) Stable disease 17.5 (7) 15.0 (6) 16.3 (13) Progressive disease 15.0 (6) 15.0 (6) 15.0 (12) NE 5.0 (2) 5.0 (2) 5.0 (4) DCR 80.0 (32) 80.0 (32) 80.0 (64) KM-estimated PFS, median (95% CI), months 24 (4.2, NE) NR (7.5, NE) 24 (9.9, NE) DOR, median (95% CI), months NR (11.9, NE) NR (6.3, NE) NR (22.6, NE) ORR: baseline LDH, n/N1 (%) LDH > ULN 10/17 (58.8) 6/11 (54.5) 16/28 (57.1) LDH normal 15/23 (65.2) 18/24 (75.0) 33/47 (70.2) ORR: liver metastasis, n/N2 (%) Yes 6/14 (42.9) 3/5 (60.0) 9/19 (47.4) No 19/26 (73.1) 23/35 (65.7) 42/61 (68.9) †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. CI, confidence interval; DCR, disease control rate; DOR, duration of response; KM, Kaplan-Meier; LDH, lactase dehydrogenase; N1, proportion of patients with the listed LDH status; N2, proportion of patients with the listed liver metastasis status; NE, not evaluable; NR, not reached; ORR, objective response rate; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; PFS, progression-free survival; ULN, upper limit of normal. Table 6. Safety for anti–PD-L1–naive and –experienced patients % (n), unless otherwise stated Anti–PD-L1 naive† (N=80) Anti–PD-L1 experienced (N=15) Duration of exposure, median (range), weeks 30.9 (2.0–110.0) 9.0 (6.0–57.0) Patients with treatment-emergent AEs regardless of attribution Any grade Grade 3–5 Any grade Grade 3–5 Overall 96.3 (77) 40.0 (32) 80.0 (12) 46.7 (7) Serious 28.8 (23) 25.0 (20) 33.3 (5) 26.7 (4) Patients with treatment-related AEs Overall 80.0 (64) 20.0 (16) 53.3 (8) 20.0 (3) Serious 13.8 (11) 13.8 (11) 13.3 (2) 13.3 (2) Treatment-emergent immune-mediated AEs, % (n) Any grade Grade 3–5 Any grade Grade 3–5 Overall 65.0 (52) 11.3 (9) 33.3 (5) 13.3 (2) Occurred in >5% of patients (any grade) Rash 23.8 (19) 0 26.7 (4) 0 Pruritis 15.0 (12) 0 0 0 Hypothyroidism 13.8 (11) 0 0 0 Arthralgia 12.5 (10) 0 6.7 (1) 0 Diarrhea 12.5 (10) 0 13.3 (2) 0 Myalgia 10.0 (8) 0 6.7 (1) 0 Adrenal insufficiency 8.8 (7) 2.5 (2) 6.7 (1) 0 Colitis 7.5 (6) 3.8 (3) 0 0 Pneumonitis 6.3 (5) 0 6.7 (1) 6.7 (1) †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. AE, adverse event; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1. Table 3. Patient disposition among anti–PD-L1–naive patients (Cohorts 6 + 15)† % (n), unless otherwise stated Anti–PD-L1 naive† Cohorts 6 + 15 (N=80)Cohort 6 (N=40) Cohort 15 (N=40) Patients completed planned treatment‡ 15.0 (6) 5.0 (2) 10.0 (8) Ongoing treatment 15.0 (6) 52.5 (21) 33.8 (27) Discontinued treatment 70.0 (28) 42.5 (17) 56.3 (45) Disease progression 45.0 (18) 17.5 (7) 31.3 (25) AE 15.0 (6) 15.0 (6) 15.0 (12) Patient decision 5.0 (2) 0 2.5 (2) Death 2.5 (1) 5.0 (2) 3.8 (3) Physician decision 2.5 (1) 5.0 (2) 3.8 (3) Duration of exposure, median (range), weeks 37.1 (2–110) 24.2 (3–56) 30.9 (2–110) †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. ‡Planned treatment; 51 weeks + additional 51 weeks given based on investigator discretion. AE, adverse event; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1. Table 5. Clinical activity among anti–PD-L1–experienced patients (Cohort 7) % (n), unless otherwise stated Total (N=15) ORR, % (95% CI) 13.3 (1.7, 40.5) Complete response 0 Partial response 13.3 (2) Stable disease 26.7 (4) Progressive disease 53.3 (8) NE 6.7 (1) DCR 40.0 (6) KM-estimated PFS, median (95% CI), months 1.5 (1.3, 7.7) DOR, median (95% CI), months NR (3.4, NE) ORR by LAG-3 expression, % <1% NA ≥1% 18.2 ORR by PD-L1 expression, % <1% 18.2 ≥1% 0 CI, confidence interval; DCR, disease control rate; DOR, duration of response; KM, Kaplan-Meier; LAG-3, lymphocyte activation gene-3; NA, not available; NE, not evaluable; NR, not reached; ORR, objective response rate; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; PFS, progression-free survival. Table 4. Clinical activity based on PD-L1 and LAG-3 levels (Cohort 6) Patients, % (n) ORR, % (n) PFS, median (95% CI), months Overall 100.0 (40) 62.5 (25) 24 (4.2, NE) LAG-3 expression ≥1% 67.5 (27) 74.1 (20) 24 (5.6, NE) LAG-3 expression <1% 12.5 (5) 40.0 (2) NR (1.4, NE) PD-L1 expression ≥1% 45.0 (18) 77.8 (14) 24 (9.9, NE) PD-L1 expression <1% 40.0 (16) 56.3 (9) 8.5 (2.8, NE) PD-L1 ≥1% and LAG-3 ≥1% 45.0 (18) 77.8 (14) 24 (9.9, NE) PD-L1 <1% and LAG-3 ≥1% 22.5 (9) 66.7 (6) 5.6 (1.2, NE) PD-L1 <1% and LAG-3 <1% 12.5 (5) 40.0 (2) NR (1.4, NE) There were no patients with PD-L1 ≥1% and LAG-3 <1%. CI, confidence interval; IHC, immunohistochemistry; LAG-3, lymphocyte activation gene-3; NA, not available; NE, not evaluable; ORR, objective response rate; PD-L1, programmed cell death-ligand 1; PFS, progression-free survival. Results Baseline demographics and disease characteristics • As of July 1, 2022, 40 patients were enrolled and received treatment in each Cohort 6 and Cohort 15, and 15 patients received treatment in Cohort 7. • Median age among the anti–PD-L1–naive patients (Cohorts 6 + 15) was 69.0 years (range: 24–88), 60.0% of patients were male, and 90.0% were White (Table 1). • For anti–PD-1/PD-L1–experienced patients (Cohort 7), median age was 59.0 years, 46.7% were male, and 60.0% were White. • The median sum of diameters of the target lesion was 51.5 mm (range: 11–214) among patients in Cohorts 6 + 15 (Table 1). • Among anti–PD-L1–naive patients 33.8% had stage M1c at baseline, 35% had lactate dehydrogenase (LDH) levels above the upper limit of normal (ULN), and 23.8% had liver metastases (Table 1). Methods • Adult patients with advanced melanoma who had no prior anti–PD-1/PD-L1 treatment (naive; Cohort 6 and 15) or had prior anti–PD-1/PD–L1 treatment within 3 months of screening (experienced; Cohort 7) received fianlimab 1600 mg + cemiplimab 350 mg intravenously (IV) every 3 weeks (Q3W), for up to 51 weeks (Figure 1). – Prior systemic therapies, including prior adjuvant therapies, were excluded for Cohort 15. – Patients in Cohort 7 must have tolerated prior anti–PD-1/PD-L1 therapy for at least 6 weeks and must not have discontinued treatment due to toxicity. – Patients had an option to continue fianlimab + cemiplimab treatment for an additional 51 weeks. • Tumor measurements were performed every 6 weeks for the first 24 weeks, then 9 weeks for the subsequent 27 weeks. • The data cut-off date was July 1, 2022. Figure 2. Efficacy overview among anti–PD-L1–naive patients (Cohorts 6 + 15)† †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. §Patients with ongoing status (missing study complete status). CI, confidence interval; CR, complete response; DOR, duration of response; NE, not estimable; NR, not reached; PD, progressive disease; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; PFS, progression-free survival; PR, partial response; SD, stable disease; SOD, sum of diameters. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 100 80 60 40 20 0 -20 -40 -60 -80 -100 PD SD PR CR B e s t p e rc e n t c h a n g e fr o m b a s e lin e Patients (N=76) 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 100 80 60 40 20 0 -20 -40 -60 -80 -100 P e rc e n t o f S O D c h a n g e fr o m b a s e lin e Months 0 Total (Cohorts 6 + 15) No. at risk: 2 4 6 8 10 12 14 16 18 20 22 24 26 28 3230 80 64 56 42 32 28 20 16 16 12 8 7 6 6 4 01 1.0 0.8 0.6 0.4 0.2 0 P ro b a b ili ty o f p ro g re s s io n -f re e s u rv iv a l Months Baseline On treatment (after 33 weeks) PD SD PR CR NR 76% of patients had any level of tumor reduction Median DOR not yet reached Kaplan-Meier estimation of PFS Anti–PD-L1 naive † by investigator assessment (N=80) PFS, median (95% CI), months 24.0 (9.9, NE) Estimated event-free probability at 12 months, % (95% CI) 55.0 (41.6, 66.5) Figure 3. Clinical activity among anti–PD-L1–experienced patients (Cohort 7) §Patients with ongoing status (missing study complete status). CR, complete response; PD, progressive disease; PD-1, programmed cell death-1; PR, partial response; SD, stable disease; SOD, sum of diameters. 100 80 60 40 20 0 -20 -40 -60 -80 -100 PD SD PR B e s t p e rc e n t c h a n g e fr o m b a s e lin e 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 100 80 60 40 20 0 -20 -40 -60 -80 -100P e rc e n t o f S O D c h a n g e fr o m b a s e lin e Months 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Patients (N=14) PD SD PR CR Figure 1. Study schema †Prior systemic therapies, including prior adjuvant therapies, excluded for Cohort 15. ‡Defined as patients who had progressed on prior anti–PD-1/PD-L1 treatment within 3 months of screening. Patients must have tolerated therapy for a ≥6 weeks and must not have discontinued treatment due to toxicity. §With an option for an additional 51 weeks. IIResponse assessments were every 6 weeks for the first 24 weeks, then 9 weeks for the subsequent 27 weeks. ADA, antidrug antibody; ECOG PS, Eastern Cooperative Oncology Group performance score; IV, intravenous; LAG-3, lymphocyte activation gene-3; mAb, monoclonal antibody; MHC, major histocompatibility complex; ORR, objective response rate; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; PD-L2, programmed cell death-ligand 2; PK, pharmacokinetics; RECIST 1.1, Response Evaluation Criteria in Solid Tumors version 1.1. Expansion Cohorts 6 and 15† Anti–PD-1/PD-L1 naive Fianlimab 1600 mg + cemiplimab 350 mg IV every 3 weeks, for up to 51 weeks§ • Tumor response assessed by investigators • Response assessments every 6 or 9II weeks (RECIST 1.1) to determine ORR Primary endpoint • ORR per RECIST 1.1 criteria Secondary endpoints • Safety, PK, and ADA Key inclusion criteria • ≥18 years of age • ECOG PS of 0 or 1 • At least one lesion measurable by RECIST 1.1 • Metastatic or inoperable locally advanced nonuveal melanoma Key exclusion criteria • Prior treatment with LAG-3–targeting biologic or small molecule • Radiation therapy within 2 weeks prior to enrollment Expansion Cohort 7 Anti–PD-1/PD-L1 experienced‡ Objectives • To assess preliminary anti-tumor activity of fianlimab + cemiplimab as measured by ORR per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 criteria in patients with advanced melanoma. • To assess the safety profile of fianlimab + cemiplimab in patients with advanced melanoma. Key takeaway • This analysis provides additional data that supports the use of fianlimab + cemiplimab combination treatment for patients with advanced melanoma. Conclusions • In two sequential expansion cohorts (total N=40/cohort), fianlimab + cemiplimab independently and reproducibly demonstrated clinically meaningful activity among patients with anti–PD-L1–naive advanced melanoma. – ORR was 63.8% (7 complete responses and 44 partial responses) and median DOR was not reached (95% CI: 22.6, NE). – Kaplan-Meier estimation of PFS was 24 months (95% CI: 9.9, NE). – Clinical activity was observed in poor prognosis subgroups (i.e., LDH > ULN, liver metastases), as well as in patients with high and low PD-L1 expression levels. • Observed clinical activity in the anti–PD-L1–exposed population (ORR 13.3%) was consistent with previous reports of anti–LAG-3 + anti–PD-L1 combination treatment in this setting. • The fianlimab + cemiplimab combination demonstrated an acceptable risk/benefit profile similar to that observed with cemiplimab monotherapy and other anti–PD-1 agents. In the anti–PD-L1– naive population: – 96.3% of patients experienced treatment-emergent AEs (TEAEs) of any grade. – 28.8% of patients experienced serious TEAEs. – 16.3% of patients discontinued treatment due to a TEAE. • A Phase 3 trial (NCT05352672) of fianlimab + cemiplimab in patients with advanced melanoma is ongoing. Background • Combination anti–lymphocyte activation gene-3 (LAG-3) and anti–programmed cell death-1 (PD-1) treatment demonstrated higher median progression free survival (PFS) and objective response rate (ORR) compared with anti–PD-1 monotherapy in a Phase 2/3 clinical trial of patients with untreated advanced melanoma.1 • The RELATIVITY-047 study showed an ORR of 43.1%.2 • Fianlimab (REGN3767) and cemiplimab are both high-affinity, human, hinge-stabilized immunoglobulin 4 (IgG4) monoclonal antibodies derived using VelocImmune technology. • Fianlimab blocks LAG-3 and major histocompatibility complex (MHC) class II–driven T-cell inhibition.3 • Cemiplimab blocks interactions of PD-1 with PD-ligand 1 (PD-L1) and PD-L2.4 • In an initial expansion cohort, fianlimab + cemiplimab in patients with advanced melanoma gave an impressive efficacy of >60% ORR.5 • Here we present clinical activity and safety follow-up data of fianlimab + cemiplimab in Phase 1 expansion cohorts of patients with advanced melanoma, and a confirmatory expansion cohort (NCT03005782). Presented at Winter Clinical Derm 2023, January 13–18, 2023 (encore of previous presentation at the European Society of Medical Oncology [ESMO] 2022, September 8–13, Hamid O et al.). Reused with permission. Tumor response • The ORR for anti–PD-L1–naive patients was 63.8% (95% confidence interval [CI]: 52.2, 74.2%; Table 2). – Seven (8.8%) patients had a complete response, and 44 (55.0%) patients had a partial response. • Eight (10.0%) anti–PD-L1–naive patients completed planned treatment; 33.8% of patients are ongoing treatment, and 56.3% of patients discontinued treatment (Table 3). • Among anti–PD-L1–experienced patients, ORR was 13.3% (95% CI: 1.7, 40.5); 2 (13.3%) patients had a partial response (Table 5, Figure 3). • The median Kaplan-Meier estimated PFS among anti–PD-L1 experienced patients was 1.5 months (95% CI: 1.3, 7.7) (Table 5). • Clinical activity based on PD-L1 and LAG-3 levels was assessed for patients in Cohort 6 (Table 4). – Formalin-fixed, paraffin-embedded baseline tumor samples were used to determine LAG-3 and PD-L1 expression levels by IHC. – LAG-3 levels were reported as the percentage of positively staining immune cells in the viable tumor area using the 17B4 clone. Safety data • The safety profile of fianlimab + cemiplimab combination treatment was similar to anti–PD-L1 therapies. • Median duration of treatment exposure was 30.9 weeks (range: 2–110) among anti–PD-L1–naive patients and 9.0 weeks (range: 6–57) among anti–PD-L1–experienced patients (Table 6). • In the anti–PD-L1–naive population: – Rate of grade ≥3 treatment-related adverse events (AE) was 20.0%. – Rate of discontinuation due to treatment-related AEs was 15.0%. – Treatment-related AEs leading to death occurred in two patients (2.5%): one experienced colitis and one experienced cardiac shock. – The patient who experienced cardiac shock also had COVID-19 with pulmonary edema concurrently. – Rate of treatment-emergent adrenal insufficiency was 10%. References 1. Tawbi HA et al. N Engl J Med. 2022;386:24–34. 2. Long GV et al. J Clin Oncol. 2022;40(suppl 36):360385. 3. Burova E et al. Mol Cancer. 2019;18:2051–2062. 4. Burova E et al. Mol Cancer. 2017;16:861–870. 5. Hamid O et al. J Clin Oncol. 2021;39(suppl 15):9515. Acknowledgments The authors thank the patients, their families, all other investigators and all investigational site members involved in this study. The study was funded by Regeneron Pharmaceuticals, Inc., and Sanofi. Medical writing support and typesetting was provided by Jenna Lee, MSc, of Prime, Knutsford, UK, funded by Regeneron Pharmaceuticals, Inc., and Sanofi. Disclosure Omid Hamid reports honoraria from Bristol-Myers Squibb, Novartis, Pfizer, Sanofi and Regeneron Pharmaceuticals, Inc.; and consulting or advisory roles with Aduro Biotech, Akeso Biopharma, Amgen, Arcus Biosciences, BioAtla, Bristol-Myers Squibb, CytomX Therapeutics, Exelixis, Genentech, GlaxoSmithKline, Idera, Immunocore, Incyte, Iovance Biotherapeutics, Merck, Merck Serono, Moderna Therapeutics, NextCure, Novartis, Pfizer, Regeneron Pharmaceuticals, Inc., Roche, Sanofi, Seattle Genetics, Torque and Zelluna. Scan the QR code for co-author disclosures