CX-072 (pacmilimab), a Probody PD-L1 inhibitor, in combination with ipilimumab in patients with advanced solid tumors (PROCLAIM-CX-072): a first-in-human, dose-finding study

Rachel E Sanborn, Omid Hamid, Elisabeth Ge de Vries, Patrick A Ott, Javier Garcia-Corbacho, Valentina Boni, Johanna Bendell, Karen A Autio, Daniel C Cho, Ruth Plummer, Mark Stroh, Lawrence Lu, Fiona Thistlethwaite, Rachel E Sanborn, Omid Hamid, Elisabeth Ge de Vries, Patrick A Ott, Javier Garcia-Corbacho, Valentina Boni, Johanna Bendell, Karen A Autio, Daniel C Cho, Ruth Plummer, Mark Stroh, Lawrence Lu, Fiona Thistlethwaite

Abstract

Background: Probody® therapeutics are antibody prodrugs designed to be activated by tumor-associated proteases. This conditional activation restricts antibody binding to the tumor microenvironment, thereby minimizing 'off-tumor' toxicity. Here, we report the phase 1 data from the first-in-human study of CX-072 (pacmilimab), a Probody immune checkpoint inhibitor directed against programmed death-ligand 1 (PD-L1), in combination with the anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) antibody ipilimumab.

Methods: Adults (n=27) with advanced solid tumors (naive to PD-L1/programmed cell death protein 1 or CTLA-4 inhibitors) were enrolled in the phase 1 combination therapy dose-escalation portion of this multicenter, open-label, phase 1/2 study (NCT03013491). Dose-escalation pacmilimab/ipilimumab followed a standard 3+3 design and continued until the maximum tolerated dose (MTD) was determined. Pacmilimab+ipilimumab was administered intravenously every 3 weeks for four cycles, followed by pacmilimab administered every 2 weeks as monotherapy. The primary objective was identification of dose-limiting toxicities and determination of the MTD. Other endpoints included the rate of objective response (Response Evaluation Criteria In Solid Tumors v.1.1).

Results: Twenty-seven patients were enrolled in pacmilimab (mg/kg)+ipilimumab (mg/kg) dose-escalation cohorts: 0.3+3 (n=6); 1+3 (n=3); 3+3 (n=3); 10+3 (n=8); 10+6 (n=6); and 10+10 (n=1). Dose-limiting toxicities occurred in three patients, one at the 0.3+3 dose level (grade 3 dyspnea/pneumonitis) and two at the 10+6 dose level (grade 3 colitis, grade 3 increased aspartate aminotransferase). The MTD and recommended phase 2 dose was pacmilimab 10 mg/kg+ipilimumab 3 mg/kg administered every 3 weeks. Pacmilimab-related grade 3-4 adverse events (AEs) and grade 3-4 immune-related AEs were reported in nine (33%) and six (22%) patients, respectively. Three patients (11%) discontinued treatment because of AEs. The overall response rate was 19% (95% CI 6.3 to 38.1), with one complete (anal squamous cell carcinoma) and four partial responses (cancer of unknown primary, leiomyosarcoma, mesothelioma, testicular cancer). Responses lasted for >12 months in four patients.

Conclusions: The MTD and recommended phase 2 dose of pacmilimab (10 mg/kg)+ipilimumab (3 mg/kg) every 3 weeks is active and has a favorable tolerability profile.

Keywords: B7-H1 antigen; CTLA-4 antigen; immunotherapy; investigational; therapies.

Conflict of interest statement

Competing interests: RES: Honoraria from Amgen and AstraZeneca. Advisory boards for AstraZeneca, Blueprint Medicines, Celldex, Daiichi Sankyo, EMD Serono, Genentech/Roche, Janssen, and Seagen. Research support from AstraZeneca and Merck. OH: Consulting/advisory boards for Aduro, Akeso, Amgen, Beigene, Bioatla, BMS, Roche Genentech, GSK, lmmunocore, ldera, lncyte, Janssen, Merck, Nextcure, Novartis, Pfizer, Sanofi/Regeneron, Seattle Genetics, Tempus, and Zelluna. Speakers’ bureau for BMS, Novartis, Pfizer, and Sanofi/Regeneron. Institutional financial support for contracted research from Arcus, Aduro, Akeso, Amgen, Bioatla, BMS, CytomX, Exelixis, Roche Genentech, GSK, lmmunocore, ldera, lncyte, lovance, Merck, Moderna, Merck-Serano, NextCure, Novartis, Pfizer, Sanofi/Regeneron, Seattle Genetics, Torque, and Zelluna. EGEdV: Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, Bayer, Chugai Pharma, G1 Therapeutics Genentech, Nordic Nanovector, Radius Health, Regeneron, Roche, Servier, and Synthon. Institutional financial support for advisory boards from Daiichi Sankyo, Merck, NSABP, Pfizer, and Sanofi; all outside the submitted work. PAO: Research funding from and has advisory role for Neon Therapeutics, Bristol-Meyers Squibb, Merck, CytomX, Pfizer, Novartis, Celldex, Amgen, Array, AstraZeneca/MedImmune, Armo BioSciences and Roche/Genentech. JG-C: Speaker’s bureau for Bayer; Fees to support registration and attending scientific meetings from BMS, Novartis. VB: Consulting/advisory role for CytomX Therapeutics, Guidepoint, Ideaya Biosciences; Loxo Therapeutics, Oncoart, and Puma Biotechnology. Institutional financial support for clinical trials from Abbvie, ACEO, Adaptaimmune, Amcure, Amgen, Astellas, AstraZeneca, BeiGene, BMS, Boehringer Ingelheim, Boston Therapeutics, CytomX Therapeutics, Daiichi, DebioPharm, Dynavax, GSK, Genentech/Roche, H3, Incyte, Innovio, Janssen, Kura, Lilly, Loxo, Mersana, Nektar, Macrogenics, Menarini, Merck, Merus, Millenium, MSD, Nanobiotix, Novartis, ORCA, Pfizer, PharmaMar, Principia, PsiOxus, PUMA, Regeneron, Rigontec, Sanofi, Seattle Genetics, Spectrum, Synthon, Taiho, Tesaro, Transgene, Takeda, and Zenith. JB: Institutional financial support for clinical trials or contracted research from AbbVie, Acerta Pharma, ADC, Agios, Amgen, Apexigen, Arch Oncology, Arcus Bio, ARMO, Array, Arrys, AstraZeneca, AtlasMedx, Bayer, Beigene, Bellicum, BI, Bicycle Therapeutics, Blueprint, BMS, Boston Biomedical, CALGB, Calithera, Celgene, Celldex, Cyteir Therapeutics, Cytomx, Daiichi Sankyo, Effector, Eisai, EMD Serono, Evelo, Five Prime, FORMA, Forty Seven, Foundation Bio, Genentech / Roche, Gilead, Gossamer Bio, GSK, Harpoon, Hutchinson MediPharma, IGM Biosciences, Imclone, Incyte, Innate, Innate Pharma, Ipsen, Jacobio, Koltan, LEAP, Lilly, Mabspace, Macrogenics, Marshall Edwards, MedImmune, Merck, Merrimack, Mersana, Merus, Millennium, Morphotex, Nektar, NeoImmune Tech, NGM Biopharma, Novartis, Novocare, NuMab, Oncogenex, OncoMed, Ongologie, Onyx, Pfizer, Pieris, Prelude Oncology, PureTech Health, Regeneron, Relay Therapeutics, REPARE Therapeutics, Revolution Medicines, Rgenix, Sanofi, Scholar Rock, Seattle Genetics, Shattuck Labs, Sierra, Stemcentrx, SynDevRex, Synthorx, Taiho, Takeda, Tarveda, TempestTx, TG Therapeutics, Tracon, Treadwell Therapeutics, Tyrogenex, Unum Therapeutics, Vyriad, and Zymeworks. Institutional financial support for advisory boards/consulting from Array, Agios, Amgen, Apexigen, Arch Oncology, ARMO, AstraZeneca, Bayer, Beigene, BI, Bicycle Therapeutics, BMS, Celgene, Continuum Clinical, Cyteir, Daiichi Sankyo, Evelo, Five Prime, FORMA, Fusion Therapeutics, Genentech / Roche, Gilead, GSK, Incyte, Innate, Ipsen, Janssen, LEAP, Lilly, Macrogenics, MedImmune, Merck, Merrimack, Moderna Therapeutics, Molecular Partners, Novartis, Oncogenex, OncoMed, Pfizer, Phoenix Bio, Piper Biotech, Prelude Therapeutics, Relay Therapeutics, Samsung Bioepios, Sanofi, Seattle Genetics, Taiho, Tanabe Research Laboratories, TD2 (Translational Drug Development), TG Therapeutics, Tizona, Tolero, and Torque. Food, beverages and/or travel expenses from ARMO, BI, BMS, Celgene, FORMA, Genentech / Roche, Gilead, Ipsen, Lilly, MedImmune, Merck, Novartis, Oncogenex, OncoMed, and Taiho. KAA: Advisory board for CytomX (unpaid). Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, CytomX, GSK, Merck, Pfizer, and Tizona. DCC: Consultant for Nektar Therapeutics, GSK, Torque, PureTech, and HUYA. RP: Honoria for attending advisory boards from Pierre Faber, Bayer, Octimet, Clovis Oncology, Novartis, Karus Therapeutics, Biosceptre, BMS, Cybrexa, Ellipses, CV6 Therapeutics, Astex Therapeutics and Sanofi Aventis. Fees for delivery of educational talks or chairing educational meetings by AstraZeneca, Novartis, Bayer, Tesaro and BMS. Funds to support attendance at conferences from BMS and MSD. MS is a former employee and Lawrence Lu is a current employee of CytomX Therapeutics and both own stock in CytomX Therapeutics. FT: Research funding and conference registration from Novartis. Consultancy/advisory role for GSK, Achilles Therapeutics, BMS, Zelluna, Enara Bio, Bayer, T-Knife.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Patient disposition.
Figure 2
Figure 2
(A) Best percentage change in target lesion size from baseline and (B) percentage change in tumor burden over time. *Indicates that the patient is still on treatment. H, high PD-L1 expression; L, low PD-L1 expression; NE, not evaluable; Neg, no PD-L1 expression; Unk, unknown PD-L1 expression.
Figure 3
Figure 3
Ipilimumab PK after (A) administration of pacmilimab followed by ipilimumab 3 mg/kg q3w×4; (B) pacmilimab followed by ipilimumab 6 mg/kg q3w×4; and (C) pacmilimab followed by ipilimumab 10 mg/kg q3w×4. Data presented are observed ipilimumab plasma concentrations (points) versus population predicted ipilimumab concentrations (line and shaded area). Line and shaded regions represent median and 90% prediction intervals of ipilimumab concentrations from a published population PK model for ipilimumab when given as monotherapy. PK, pharmacokinetic; q3w, once every 3 weeks.

References

    1. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol 2019;12:92. 10.1186/s13045-019-0779-5
    1. Vaddepally RK, Kharel P, Pandey R, et al. . Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers 2020;12:738. 10.3390/cancers12030738
    1. Schmidt EV, Chisamore MJ, Chaney MF, et al. . Assessment of clinical activity of PD-1 checkpoint inhibitor combination therapies reported in clinical trials. JAMA Netw Open 2020;3:e1920833. 10.1001/jamanetworkopen.2019.20833
    1. Yang Y, Jin G, Pang Y, et al. . Comparative efficacy and safety of nivolumab and nivolumab plus ipilimumab in advanced cancer: a systematic review and meta-analysis. Front Pharmacol 2020;11:40. 10.3389/fphar.2020.00040
    1. Wang DY, Salem J-E, Cohen JV, et al. . Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol 2018;4:1721–8. 10.1001/jamaoncol.2018.3923
    1. Zhang B, Wu Q, Zhou YL, et al. . Immune-Related adverse events from combination immunotherapy in cancer patients: a comprehensive meta-analysis of randomized controlled trials. Int Immunopharmacol 2018;63:292–8. 10.1016/j.intimp.2018.08.014
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015;373:23–34. 10.1056/NEJMoa1504030
    1. Bristol Myers Squibb . OPDIVO (nivolumab) injection, for intravenous use [Package Insert], 2020.
    1. Wolchok JD, Kluger H, Callahan MK, et al. . Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122–33. 10.1056/NEJMoa1302369
    1. Puzanov I, Diab A, Abdallah K, et al. . Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for immunotherapy of cancer (SITC) toxicity management Working group. J Immunother Cancer 2017;5:95. 10.1186/s40425-017-0300-z
    1. Baxi S, Yang A, Gennarelli RL, et al. . Immune-Related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ 2018;360:k793. 10.1136/bmj.k793
    1. Feng Y, Roy A, Masson E, et al. . Exposure-Response relationships of the efficacy and safety of ipilimumab in patients with advanced melanoma. Clin Cancer Res 2013;19:3977–86. 10.1158/1078-0432.CCR-12-3243
    1. Polu KR, Lowman HB. Probody therapeutics for targeting antibodies to diseased tissue. Expert Opin Biol Ther 2014;14:1049–53. 10.1517/14712598.2014.920814
    1. Desnoyers LR, Vasiljeva O, Richardson JH, et al. . Tumor-Specific activation of an EGFR-targeting probody enhances therapeutic index. Sci Transl Med 2013;5:207ra144. 10.1126/scitranslmed.3006682
    1. Vasiljeva O, Hostetter DR, Moore SJ, et al. . The multifaceted roles of tumor-associated proteases and harnessing their activity for prodrug activation. Biol Chem 2019;400:965–77. 10.1515/hsz-2018-0451
    1. Vasiljeva O, Menendez E, Nguyen M, et al. . Monitoring protease activity in biological tissues using antibody prodrugs as sensing probes. Sci Rep 2020;10:5894. 10.1038/s41598-020-62339-7
    1. Autio KA, Boni V, Humphrey RW, et al. . Probody therapeutics: an emerging class of therapies designed to enhance on-target effects with reduced off-tumor toxicity for use in immuno-oncology. Clin Cancer Res 2020;26:984–9. 10.1158/1078-0432.CCR-19-1457
    1. Tipton KA, Chan C, Wong KR. PD-1 ProbodyTM therapeutic anti-tumor efficacy and protection against autoimmunity in preclinical models [poster]. Annual Meeting of the American Association for Cancer Research, 2016.
    1. Giesen D, Broer LN, Lub-de Hooge MN, et al. . Probody therapeutic design of 89Zr-CX-072 promotes accumulation in PD-L1-expressing tumors compared to normal murine lymphoid tissue. Clin Cancer Res 2020;26:3999–4009. 10.1158/1078-0432.CCR-19-3137
    1. Lyman SK, Naing A, Zein IA. Evidence of intratumoral localization, activation, and immunomodulatory effect of CX-072, a PROBODY therapeutic targeting PD-L1, in a phase I/II trial [poster]. American Society of Clinical Oncology Virtual Meeting, 2020.
    1. Bohnsack O, Hoos A, Ludajic K. Adaptation of the immune related response criteria: Irrecist. Annals of Oncology 2014;25:iv369–72. 10.1093/annonc/mdu342.23
    1. Stroh M, Green M, Millard BL, et al. . Model-informed drug development of the masked anti-PD-L1 antibody CX-072. Clin Pharmacol Ther 2021;109:383–93. 10.1002/cpt.1985
    1. Brookmeyer R, Crowley J. A confidence interval for the median survival time. Biometrics 1982;38:29–41. 10.2307/2530286
    1. Feng Y, Masson E, Dai D, et al. . Model-Based clinical pharmacology profiling of ipilimumab in patients with advanced melanoma. Br J Clin Pharmacol 2014;78:106–17. 10.1111/bcp.12323
    1. Naing A, Thistlethwaite F, De Vries E, et al. . CX-072 (pacmilimab), a probody PD-L1 inhibitor, in advanced or recurrent solid tumors (PROCLAIM-CX-072): an open label dose-finding and first-inhuman study. J Immunother Cancer 2021. 10.1136/jitc-2021-002447
    1. Long GV, Atkinson V, Cebon JS, et al. . Standard-Dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1B trial. Lancet Oncol 2017;18:1202–10. 10.1016/S1470-2045(17)30428-X
    1. Bristol Myers Squibb . YERVOY® (ipilimumab) injection, for intravenous use [Package Insert], 2020.
    1. Janjigian YY, Bendell J, Calvo E, et al. . CheckMate-032 study: efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol 2018;36:2836–44. 10.1200/JCO.2017.76.6212
    1. Sharma P, Siefker-Radtke A, de Braud F, et al. . Nivolumab alone and with ipilimumab in previously treated metastatic urothelial carcinoma: CheckMate 032 nivolumab 1 mg/kg plus ipilimumab 3 mg/kg expansion cohort results. J Clin Oncol 2019;37:1608–16. 10.1200/JCO.19.00538
    1. Overman MJ, Lonardi S, Wong KYM, et al. . Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol 2018;36:773–9. 10.1200/JCO.2017.76.9901
    1. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. . Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med 2019;381:2020–31. 10.1056/NEJMoa1910231
    1. Motzer RJ, Tannir NM, McDermott DF, et al. . Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018;378:1277–90. 10.1056/NEJMoa1712126

Source: PubMed

3
Abonnere