Corneal Epithelial Findings in Patients with Multiple Myeloma Treated with Antibody-Drug Conjugate Belantamab Mafodotin in the Pivotal, Randomized, DREAMM-2 Study

Asim V Farooq, Simona Degli Esposti, Rakesh Popat, Praneetha Thulasi, Sagar Lonial, Ajay K Nooka, Andrzej Jakubowiak, Douglas Sborov, Brian E Zaugg, Ashraf Z Badros, Bennie H Jeng, Natalie S Callander, Joanna Opalinska, January Baron, Trisha Piontek, Julie Byrne, Ira Gupta, Kathryn Colby, Asim V Farooq, Simona Degli Esposti, Rakesh Popat, Praneetha Thulasi, Sagar Lonial, Ajay K Nooka, Andrzej Jakubowiak, Douglas Sborov, Brian E Zaugg, Ashraf Z Badros, Bennie H Jeng, Natalie S Callander, Joanna Opalinska, January Baron, Trisha Piontek, Julie Byrne, Ira Gupta, Kathryn Colby

Abstract

Introduction: Patients with relapsed or refractory multiple myeloma (RRMM) represent an unmet clinical need. Belantamab mafodotin (belamaf; GSK2857916) is a first-in-class antibody-drug conjugate (ADC; or immunoconjugate) that delivers a cytotoxic payload, monomethyl auristatin F (MMAF), to myeloma cells. In the phase II DREAMM-2 study (NCT03525678), single-agent belamaf (2.5 mg/kg) demonstrated clinically meaningful anti-myeloma activity (overall response rate 32%) in patients with heavily pretreated disease. Microcyst-like epithelial changes (MECs) were common, consistent with reports from other MMAF-containing ADCs.

Methods: Corneal examination findings from patients in DREAMM-2 were reviewed, and the clinical descriptions and accompanying images (slit lamp microscopy and in vivo confocal microscopy [IVCM]) of representative events were selected. A literature review on corneal events reported with other ADCs was performed.

Results: In most patients receiving single-agent belamaf (72%; 68/95), MECs were observed by slit lamp microscopy early in treatment (69% had their first event by dose 4). However, IVCM revealed hyperreflective material. Blurred vision (25%) and dry eye (15%) were commonly reported symptoms. Management of MECs included dose delays (47%)/reductions (25%), with few patients discontinuing due to MECs (1%). The first event resolved in most patients (grade ≥2 MECs and visual acuity [each 77%], blurred vision [67%], and dry eye [86%]), with no reports of permanent vision loss to date. A literature review confirmed that similar MECs were reported with other ADCs; however, event management strategies varied. The pathophysiology of MECs is unclear, though the ADC cytotoxic payload may contribute to on- or off-target effects on corneal epithelial cells.

Conclusion: Single-agent belamaf represents a new treatment option for patients with RRMM. As with other ADCs, MECs were observed and additional research is warranted to determine their pathophysiology. A multidisciplinary approach, involving close collaboration between eye care professionals and hematologist/oncologists, is needed to determine appropriate diagnosis and management of these patients.

Trial registration: ClinicalTrials.gov Identifier, NCT03525678.

Keywords: Antibody–drug conjugate; Belantamab mafodotin; Cornea; In vivo confocal microscopy; Microcyst-like epithelial changes; Monomethyl auristatin F; Multiple myeloma; Oncology.

Figures

Fig. 1
Fig. 1
Belamaf structure and mechanism of action. ADC antibody–drug conjugate, ADCC/ADCP antibody-directed cellular cytotoxicity/phagocytosis, BCMA B-cell maturation antigen, IgG1 immunoglobulin G1, mAb Monoclonal antibody, MM multiple myeloma, MMAF monomethyl auristatin F
Fig. 2
Fig. 2
Time-to-recovery analysis for MECs in patients receiving belamaf (2.5 mg/kg) in DREAMM-2. EoT end of treatment, MEC microcyst-like epithelial changes. EoT defined as the last dose date (for patients with an unresolved event at treatment discontinuation) or the event onset date (for patients with an event that started post treatment discontinuation due to any reason). Analysis included patients who were not recovered at EoT exposure (defined as 20 days after the last infusion) and those who had events that started after treatment discontinuation (n = 39). Twenty-one percent (8/39) of patients died before recovery
Fig. 3
Fig. 3
Representative slit lamp microscopic images of MECs. MECs microcyst-like epithelial changes. Slit lamp microscopic images (ad) demonstrating MECs (see arrowheads). The lesions are small, located within the corneal epithelium, and are seen here in the corneal periphery and mid-periphery. Visualization requires high magnification and is aided by the use of retroillumination off the iris or indirect illumination
Fig. 4
Fig. 4
Case report representative slit lamp and in vivo confocal microscopic images of MECs. MECs microcyst-like epithelial changes. Slit lamp microscopic images of the right (a) and left (b) eyes, demonstrating MECs (arrows). These lesions are best visualized at medium to high magnification. Retroillumination or indirect illumination can be helpful. In vivo confocal microscopic image from the same patient (cf) demonstrating hyperreflective opacities within the corneal epithelium. These opacities were noted to be most prominent in the wing cells (d) and basal cells (e), as compared with the superficial cells (f). They were not visualized within the anterior stroma (f) or endothelium (not shown)
Fig. 5
Fig. 5
Case report representative high-magnification fluorescein staining slit lamp microscopic images. Broad-beam slit lamp microscopic images with cobalt blue light demonstrating fluorescein staining of the right (a) and left (b) eyes. Whorl-like staining was noted in both corneas, most prominent in the inferonasal quadrant of the left eye (arrowhead)
Fig. 6
Fig. 6
Change in BCVA over time with belamaf treatment in the DREAMM-2 case report. BCVA best-corrected visual acuity, KVA keratopathy and visual acuity, MECs microcyst-like epithelial changes. Corneal examination finding (MECs and changes in BCVA) were graded per KVA scale (Table 1), and represented (as grades 1–4) above each line for each examination
Fig. 7
Fig. 7
Proposed mechanism of belamaf-associated MECs. Ep epithelium, BL Bowman’s layer, MECs microcyst-like epithelial changes, St stroma

References

    1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011;364:1046–1060.
    1. Tai YT, Anderson KC. Targeting B-cell maturation antigen in multiple myeloma. Immunotherapy. 2015;7:1187–1199.
    1. National Cancer Institute. Surveillance, epidemiology, and end results program. cancer stat facts: myeloma; 2020. .
    1. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised international staging system for multiple myeloma: a report from International Myeloma Working Group. J Clin Oncol. 2015;33:2863–2869.
    1. Mikhael J. Treatment options for triple-class refractory multiple myeloma. Clin Lymphoma Myeloma Leuk. 2020;20:1–7.
    1. Gandhi UH, Cornell RF, Lakshman A, et al. Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia. 2019;33:2266–2275.
    1. Lonial S, Lee HC, Badros A, et al. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study. Lancet Oncol. 2020;21:207–221.
    1. Trudel S, Lendvai N, Popat R, et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): a dose escalation and expansion phase 1 trial. Lancet Oncol. 2018;19:1641–1653.
    1. Trudel S, Lendvai N, Popat R, et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy from dose expansion phase I study. Blood Cancer J. 2019;9:37.
    1. Tai Y-T, Mayes PA, Acharya C, et al. Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood. 2014;123:3128–3138.
    1. Montes De Oca R, Bhattacharya S, Vitali N, et al. The anti-BCMA antibody-drug conjugate GSK2857916 drives immunogenic cell death and immune-mediated anti-tumor responses, and in combination with an OX40 agonist potentiates in vivo activity. HemaSphere. 2019;3:231.
    1. Lonial S, Lee HC, Badros A, et al. Pivotal DREAMM-2 study: single-agent belantamab mafodotin (GSK2857916) in patients with relapsed/refractory multiple myeloma (RRMM) refractory to proteasome inhibitors (PIs), immunomodulatory agents, and refractory and/or intolerant to anti-CD38 monoclonal antibodies (mAbs). 25th European Haematology Association Annual Congress; June 11–14, 2020; Virtual.
    1. Eaton JS, Miller PE, Mannis MJ, Murphy CJ. Ocular adverse events associated with antibody-drug conjugates in human clinical trials. J Ocul Pharmacol Ther. 2015;31:589–604.
    1. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE), Version 4.03. Rockwell, MD: National Cancer Institute; 2009. .
    1. Kreps EO, Derveaux T, Denys H. Corneal changes in trastuzumab emtansine treatment. Clin Breast Cancer. 2018;18:e427–e429.
    1. Lee BA, Lee MS, Maltry AC, Hou JH. Clinical and histological characterization of toxic keratopathy from depatuxizumab mafodotin (ABT-414), an antibody-drug conjugate: [RETRACTED]. Cornea. 2018.
    1. Younes A, Kim S, Romaguera J, et al. Phase I multidose-escalation study of the anti-CD19 maytansinoid immunoconjugate SAR3419 administered by intravenous infusion every 3 weeks to patients with relapsed/refractory B-cell lymphoma. J Clin Oncol. 2012;30:2776–2782.
    1. Deklerck E, Denys H, Kreps EO. Corneal features in trastuzumab emtansine treatment: not a rare occurrence. Breast Cancer Res Treat. 2019;175:525–530.
    1. Tannir NM, Forero-Torres A, Ramchandren R, et al. Phase I dose-escalation study of SGN-75 in patients with CD70-positive relapsed/refractory non-Hodgkin lymphoma or metastatic renal cell carcinoma. Invest New Drugs. 2014;32:1246–1257.
    1. Tsuda M, Takano Y, Shigeyasu C, Imoto S, Yamada M. Abnormal corneal lesions induced by trastuzumab emtansine: an antibody-drug conjugate for breast cancer. Cornea. 2016;35:1378–1380.
    1. Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17:e328–e346.
    1. Gan HK, Reardon DA, Lassman AB, et al. Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma. Neuro Oncol. 2018;20:838–847.
    1. Thompson JA, Motzer RJ, Molina AM, et al. Phase I trials of anti-ENPP3 antibody-drug conjugates in advanced refractory renal cell carcinomas. Clin Cancer Res. 2018;24:4399–4406.
    1. Corbelli E, Miserocchi E, Marchese A, et al. Ocular toxicity of mirvetuximab. Cornea. 2019;38:229–232.
    1. Clinical and histological characterization of toxic keratopathy from depatuxizumab mafodotin (ABT-414), an antibody-drug conjugate: retraction. Cornea. 2018;37:e55.
    1. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs. 2016;8:659–671.
    1. Goss GD, Vokes EE, Gordon MS, et al. Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor. Cancer. 2018;124:2174–2183.
    1. Lassman AB, van den Bent MJ, Gan HK, et al. Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial. Neuro Oncol. 2019;21:106–114.
    1. Reardon DA, Lassman AB, van den Bent M, et al. Efficacy and safety results of ABT-414 in combination with radiation and temozolomide in newly diagnosed glioblastoma. Neuro Oncol. 2017;19:965–975.
    1. Shapiro GI, Vaishampayan UN, LoRusso P, et al. First-in-human trial of an anti-5T4 antibody-monomethylauristatin conjugate, PF-06263507, in patients with advanced solid tumors. Invest New Drugs. 2017;35:315–323.
    1. van den Bent M, Gan HK, Lassman AB, et al. Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: results from a multi-center, international study. Cancer Chemother Pharmacol. 2017;80:1209–1217.
    1. Banerji U, van Herpen CML, Saura C, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019;20:1124–1135.
    1. de Bono JS, Concin N, Hong DS, et al. Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): a first-in-human, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20:383–393.
    1. Kantarjian HM, Lioure B, Kim SK, et al. A phase II study of coltuximab ravtansine (SAR3419) monotherapy in patients with relapsed or refractory acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk. 2016;16:139–145.
    1. Martin LP, Konner JA, Moore KN, et al. Characterization of folate receptor alpha (FRalpha) expression in archival tumor and biopsy samples from relapsed epithelial ovarian cancer patients: a phase I expansion study of the FRalpha-targeting antibody-drug conjugate mirvetuximab soravtansine. Gynecol Oncol. 2017;147:402–407.
    1. O'Malley DM, Matulonis UA, Birrer MJ, et al. Phase Ib study of mirvetuximab soravtansine, a folate receptor alpha (FRalpha)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol Oncol. 2020;157(2):379–85.
    1. Trneny M, Verhoef G, Dyer MJ, et al. A phase II multicenter study of the anti-CD19 antibody drug conjugate coltuximab ravtansine (SAR3419) in patients with relapsed or refractory diffuse large B-cell lymphoma previously treated with rituximab-based immunotherapy. Haematologica. 2018;103:1351–1358.
    1. Matulonis UA, Birrer MJ, O'Malley DM, et al. Evaluation of prophylactic corticosteroid eye drop use in the management of corneal abnormalities induced by the antibody-drug conjugate mirvetuximab soravtansine. Clin Cancer Res. 2019;25:1727–1736.
    1. Beeram M, Krop IE, Burris HA, et al. A phase 1 study of weekly dosing of trastuzumab emtansine (T-DM1) in patients with advanced human epidermal growth factor 2-positive breast cancer. Cancer. 2012;118:5733–5740.
    1. Ribrag V, Dupuis J, Tilly H, et al. A dose-escalation study of SAR3419, an anti-CD19 antibody maytansinoid conjugate, administered by intravenous infusion once weekly in patients with relapsed/refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2014;20:213–220.
    1. de Goeij BE, Lambert JM. New developments for antibody-drug conjugate-based therapeutic approaches. Curr Opin Immunol. 2016;40:14–23.
    1. Mahalingaiah PK, Ciurlionis R, Durbin KR, et al. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol Ther. 2019;200:110–125.
    1. Liu Z, Carvajal M, Carraway CA, Carraway K, Pflugfelder SC. Expression of the receptor tyrosine kinases, epidermal growth factor receptor, ErbB2, and ErbB3, in human ocular surface epithelia. Cornea. 2001;20:81–85.
    1. Zhao H, Atkinson J, Gulesserian S, et al. Modulation of macropinocytosis-mediated internalization decreases ocular toxicity of antibody-drug conjugates. Cancer Res. 2018;78:2115–2126.
    1. Carpenter RO, Evbuomwan MO, Pittaluga S, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res. 2013;19:2048–2060.
    1. Hollander DA, Aldave AJ. Drug-induced corneal complications. Curr Opin Ophthalmol. 2004;15:541–548.
    1. Bernitt E, Dobereiner HG, Gov NS, Yochelis A. Fronts and waves of actin polymerization in a bistability-based mechanism of circular dorsal ruffles. Nat Commun. 2017;8:15863.
    1. Zhao H, Gulesserian S, Ganesan SK, et al. Inhibition of megakaryocyte differentiation by antibody-drug conjugates (ADCs) is mediated by macropinocytosis: implications for ADC-induced thrombocytopenia. Mol Cancer Ther. 2017;16:1877–1886.
    1. Peng H, Park JK, Lavker RM. Autophagy and macropinocytosis: keeping an eye on the corneal/limbal epithelia. Invest Ophthalmol Vis Sci. 2017;58:416–423.
    1. Nagai N, Ogata F, Otake H, Nakazawa Y, Kawasaki N. Energy-dependent endocytosis is responsible for drug transcorneal penetration following the instillation of ophthalmic formulations containing indomethacin nanoparticles. Int J Nanomedicine. 2019;14:1213–1227.
    1. Pennington MR, Saha A, Painter DF, et al. Disparate entry of adenoviruses dictates differential innate immune responses on the ocular surface. Microorganisms. 2019;7:351.
    1. Guthoff T, Tietze B, Meinhardt B, Becher J, Guthoff R. Cytosine-arabinoside-induced keratopathy: a model of corneal proliferation kinetics. Ophthalmologica. 2010;224:308–311.
    1. Yoon JJ, Ismail S, Sherwin T. Limbal stem cells: central concepts of corneal epithelial homeostasis. World J Stem Cells. 2014;6:391–403.
    1. Lin HP, Singla B, Ghoshal P, et al. Identification of novel macropinocytosis inhibitors using a rational screen of food and drug administration-approved drugs. Br J Pharmacol. 2018;175:3640–3655.
    1. Fathi AT, Borate U, DeAngelo DJ, et al. A phase 1 study of denintuzumab mafodotin (SGN-CD19A) in adults with relapsed or refractory B-lineage acute leukemia (B-ALL) and highly aggressive lymphoma. Blood. 2015;126:1328.
    1. Moskowitz CH, Fanale MA, Shah BD, et al. 182 A phase 1 study of denintuzumab mafodotin (SGN-CD19A) in relapsed/refractory B-lineage non-Hodgkin lymphoma. Blood. 2015;126:182.
    1. Cenedella RJ, Fleschner CR. Kinetics of corneal epithelium turnover in vivo. Studies of lovastatin. Invest Ophthalmol Vis Sci. 1990;31:1957–1962.
    1. Eghrari AO, Riazuddin SA, Gottsch JD. Overview of the cornea: structure, function, and development. Prog Mol Biol Transl Sci. 2015;134:7–23.
    1. Wagoner MD. Chemical injuries of the eye: current concepts in pathophysiology and therapy. Surv Ophthalmol. 1997;41:275–313.
    1. Bashraheel SS, Domling A, Goda SK. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine. Biomed Pharmacother. 2020;125:110009.
    1. Antoun J, Titah C, Cochereau I. Ocular and orbital side-effects of checkpoint inhibitors: a review article. Curr Opin Oncol. 2016;28:288–94.
    1. Heinzerling L, Eigentler TK, Fluck M, Hassel JC, Heller-Schenck D, Leipe J, et al. Tolerability of BRAF/MEK inhibitor combinations: adverse event evaluation and management. ESMO Open. 2019;4:e000491.
    1. Huillard O, Bakalian S, Levy C, Desjardins L, Lumbroso-Le Rouic L, Pop S, et al. Ocular adverse events of molecularly targeted agents approved in solid tumours: a systematic review. Eur J Cancer. 2014;50:638–48.
    1. Omoti AE, Omoti CE. Ocular toxicity of systemic anticancer chemotherapy. Pharm Pract (Granada). 2006;4:55–9.
    1. Srivastava A, Al-Zubidi N, Appelbaum E, Gombos DS, Nader ME, Gidley PW, et al. Immune-Related Oral, Otologic, and Ocular Adverse Events. Adv Exp Med Biol. 2020;1244:295–307.
    1. Ding X, Bishop RJ, Herzlich AA, Patel M, Chan CC. Limbal stem cell deficiency arising from systemic chemotherapy with hydroxycarbamide. Cornea. 2009;28:221–223.
    1. Kim BY, Riaz KM, Bakhtiari P, et al. Medically reversible limbal stem cell disease: clinical features and management strategies. Ophthalmology. 2014;121:2053–2058.
    1. Lichtinger A, Pe'er J, Frucht-Pery J, Solomon A. Limbal stem cell deficiency after topical mitomycin C therapy for primary acquired melanosis with atypia. Ophthalmology. 2010;117:431–437.
    1. Popat R, Warcel D, O'Nions J, et al. Characterisation of response and corneal events with extended follow-up after belantamab mafodotin (GSK2857916) monotherapy for patients with relapsed multiple myeloma: a case series from the first-time-in-human clinical trial. Haematologica. 2020 doi: 10.3324/haematol.2019.235937.

Source: PubMed

3
Iratkozz fel