Cancer/testis (C/T) antigens are genes whose expression is silenced in healthy adult tissues except for male germ cells. Because C/T antigens are expressed in various cancers, they are potential targets for targeted therapies.1 MAGE-A1, the first identified C/T antigen, was characterized as the target of an autologous cytotoxic T-cell clone recognizing the melanoma cell line MZ2E.2 MAGE-A1 expression was described in biopsies from patients with multiple myeloma (MM),3 an aggressive plasma cell malignancy, and was associated with poor prognosis in one study.4 Despite recent advances in the treatment of MM, such as chimeric antigen receptor (CAR )T-cell therapy and bispecific T-cell engagers (BiTE),5 MM remains incurable, and antigen loss has been described as resistance mechanism for immunotherapies.6 Therefore, there is a high need for new treatment options and therapeutic targets. T-cell receptor (TCR) therapy represents another T-cell-based immunotherapeutic approach in cancer.7 TCR T-cell therapy allows to target intracellular proteins presented by major histocompatibility (MHC) class I molecules with a higher antigen sensitivity compared to CAR T cells.8,9 We have previously described TCR-T1367 with optimal affinity against MAGE-A1 for development of TCR-based cellular immunotherapy.10 In this study, we present the analysis of a large cohort of MM patients for MAGE-A1 expression and its association with specific clinical and disease characteristics. Furthermore, we describe TCR-1367 T-cell production in an academic setting, and present clinical data from the phase I study (EudraCT: 2017-001208-30) investigating TCR-1367 T cells in two patients. MAGE-A1 expression was investigated by immunohisto-chemistry (IHC) in 252 formalin-fixed, paraffin-embedded histological samples from 213 patients, collected from 2012 to 2022, using the commonly used anti-MAGE-A1 antibody MA454. The study was approved by the ethical committee of Charité-Universitätsmedizin Berlin (EA4/133/23). Statistical tests included Mann-Whitney test for comparing two categories and Kruskal-Wallis test for more than two categories. Clinical and genetic characteristics of our cohort are presented in the Online Supplementary Table S1. Out of the 252 samples, 27% presented with ≥30% MAGE-A1 positive (MAGE-A1+) MM cells, 23% with a lower fraction and 50% without MAGE-A1 expression (Figure 1A). An exemplary slide of MAGE-A1 expressing MM cells is shown in Figure 1B. The fraction of MAGE-A1+ samples (≥30% of MAGE-A1+ MM cells), increased from 18% at diagnosis to 33% during relapse (Figure 1A) with a significant increase of the mean proportion of MAGE-A1+ MM cells in all samples (15% vs. 26%; P=0.0002). For 131 patients cytogenetics and fluorescence in situ hybridization (FISH) results were available, with 59 classified as standard-risk and 72 as high-risk based on cytogenetic aberrations defined as del(17p), t(4;14), t(14;16), gain or amplification (1q21).11,12 High-risk patients presented with a slightly higher proportion of MAGE-A1+ samples (31%) compared to the standard-risk group (23%), but without significant difference in mean MAGE-A1 expression (24% vs. 18%; P=0.0788; Figure 1C).
Furthermore, we investigated the association of MAGE-A1 expression with extramedullar disease (EMD). In bone marrow (BM) samples, 22% had ≥30% MAGE-A1+ cells, compared to 55% in bone-related extramedullar myeloma (EM-B) and 46% in extraosseous extramedullar myeloma (EM-E) samples. The mean proportion of MAGE-A1+ MM cells in positive samples was higher in EM-B (83%; P=0.3918) and EM-E (85%; P=0.0417) samples compared to BM (68%) as shown in Figure 1D. A significant higher proportion of MAGE-A1+ MM cells in positive BM samples were found in patients with documented EMD compared to patients without EMD (82% vs. 58%; P=0.0017; Figure 1E). Analyzing 11 matched EMD and BM samples collected from the same time point and same patient we found only a weak correlation between the proportion of MAGE-A1+ cells between these samples (R2=0.3883; P=0.0405). Survival data were available for 99 newly diagnosed MM patients of which 83 were MAGE-A1-negative (<30% MAGE-A1+ MM cells) and 16 positive (≥30% MAGE-A1+ MM cells). Kaplan-Meier survival analysis revealed that MAGE-A1 expression at diagnosis was associated with impaired overall survival (OS) resulting in 2-year survival rates of 95% for negative and 54% for positive patients (median OS not-reached for both; hazard ratio [HR]=0.21; 95% confidence interval [CI]: 0.04-1.16; P=0.0015, log-rank test; Figure 1F). To investigate MAGE-A1 as a therapeutic target, we applied the MAGE-A1278-286- epitope-directed TCR-T1367 sequence10 and transduced autologous T cells from patients with MM with retrovirus encoding TCR-T1367. TCR-1367 T cells were manufactured at Zellkulturlabor für Klinische Prüfung (ZKP), the GMP Facility of the Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin. An overview of the manufacturing process is shown in Figure 2A. The manufacturing process was validated in three healthy donor validation runs. TCR-1367 T cells were manufactured for three patients (patients 001, 004,
To evaluate safety and efficacy of TCR-1367 T cells, we conducted a one-armed, single-center, open-label, phase I clinical trial (EudraCT: 2017-001208-30). The summarized study design is shown in Figure 3A. The main inclusion criteria were age ≥18 years, relapsed and/or refractory disease requiring therapy, at least three prior lines of therapy, HLA-A*02:01 genotype, and at least 30% of MAGE-A1+ MM cells assessed by IHC. The primary objective was to evaluate the safety and tolerability of TCR-1367 T cells. It was planned to enroll 12 patients in four cohorts with ascending doses of TCR-1367 T cells (105; 106; 107 and 5x107 cells/kg body weight [BW] ± 20%). However, based on limited recruitment potential upon availability of BCMA-CAR T cells, and competing clinical studies investigating BiTE, the trial was closed by the sponsor after treating two patients (patients 004 and 006). All patients provided written informed consent and the trial was approved by the local ethical committee in Berlin, Germany (17/0259-EK13). The study was conducted in accordance with principles of good clinical practice and the Declaration of Helsinki. The patient characteristics are shown in Online Supplementary Table S2. Both patients were treated in the first dosing cohort and were eligible for safety and efficacy analysis. The time between apheresis to application of TCR-1367 T cells was 64 days (004) and 55 days (006). Altogether, we observed 18 treatment-emergent adverse events (TEAE) in the two patients with 11 classified as possibly study treatment-related. Four of the TEAE were CTCAE (version 4.03)13 grade 3 or 4 and two were serious AE (febrile neutropenia and cancer pain), both likely related to chemotherapy and disease progression, respectively. All AE are listed in Online Supplementary Table S3. The best response according to the IMWG criteria14 was minimal response for patient 004, while patient 006 experienced progressive disease. The time to next treatment was 110 days for patient 004 and 64 days for patient 006. In patient 004, the proportion of MAGE-A1+ MM cells subsequently decreased from 80% to 60% to 30% 3 months after administration of TCR-1367 T cells (Figure 3B). MM cell infiltration decreased from 60% to 40%. However, the fraction of MAGE-A1+ MM cells started to rise again 7 months after the administration of TCR-1367 T cells, reaching 40% (Figure 3B). As shown in Figure 3C for patient 006, no measurable effect of the TCR-1367 therapy on the MAGE-A1 expression was found. Patient 004 achieved a complete response under following BiTE treatment and is still alive 35 months after administration of TCR-1367 T cells. Patient 006 further progressed and died 3 months after receiving the study treatment from myeloma progression and disease-related pancytopenia with an infection of unknown focus, considered not related to the study treatment, resulting in an OS of 91 days. No TCR-1367 T cells could be detected by flow cytometry or quantitative polymerase chain reaction (qPCR) in pharmacokinetics samples, possibly due to the low cell number administered.
MAGE-A1, identified as a frequently expressed antigen in MM in our diagnostic study, could emerge as a valuable new target, especially considering loss of commonly targeted antigens like BCMA and GPRC5D under current therapies.6 MAGE-A1 expression was associated with EMD and lower OS, aligning with findings in other MM patient cohorts.3,15 In the phase I clinical trial investigating MAGE-A1-direct-ed TCR-1367 T cells, we treated only two patients due to premature closure of the trial by the sponsor, making it impossible to provide conclusive safety and efficacy data. While we observed no severe TEAE, durable responses were not achieved, possibly due to the low dose of TCR-1367 T cells administered in the first dose cohort (1x105 cells/kg BW).
In conclusion, MAGE-A1 is an antigen expressed by a subset of MM patients associated with advanced disease and EMD. MAGE-A1-directed TCR-1367 therapy appears feasible for the tested dose in this patient group. Further clinical studies are required within the multi-refractory patient population, especially those relapsing after currently approved T-cell redirecting therapies.
Footnotes
- Received June 28, 2024
- Accepted September 2, 2024
Correspondence
Disclosures
AN discloses consultancy for Celgene, Janssen, Roche, Takeda, Alexion, Sanofi, GSK and BMS and receives research funding from BMS, Janssen and Celgene. VS, EK and DH are full-time employees of T-knife Therapeutics Inc. TB and EK are shareholders in T-knife Therapeutics Inc. TB is a founder and scientific advisory board member of T-knife Therapeutics. MO and TB are inventors of a patent applied by the Max-Delbrück Center describing the TCR used in this study. The remaining authors have non conflicts of interest to disclose.
Funding
Acknowledgments
The protocol for determining the vector copy number for pharmacokinetics analysis by qPCR was kindly provided by Dr. Michael Rothe, Hannover Medical School, Hannover, Germany. The sponsor of the trial was Charité-Universitätsmedizin Berlin.
References
- Yang P, Meng M, Zhou Q. Oncogenic cancer/testis antigens are a hallmarker of cancer and a sensible target for cancer immunotherapy. Biochim Biophys Acta Rev Cancer. 2021; 1876(1):188558. Google Scholar
- van der Bruggen P, Traversari C, Chomez P. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991; 254(5038):1643-1647. Google Scholar
- Dhodapkar MV, Osman K, Teruya-Feldstein J. Expression of cancer/testis (CT) antigens MAGE-A1, MAGE-A3, MAGE-A4, CT-7, and NY-ESO-1 in malignant gammopathies is heterogeneous and correlates with site, stage and risk status of disease. Cancer Immun. 2003; 3:9. Google Scholar
- Chari A, Cho HJ, Dhadwal A. A phase 2 study of panobinostat with lenalidomide and weekly dexamethasone in myeloma. Blood Adv. 2017; 1(19):1575-1583. Google Scholar
- Holstein SA, Grant SJ, Wildes TM. Chimeric antigen receptor T-cell and bispecific antibody therapy in multiple myeloma: moving into the future. J Clin Oncol. 2023; 41(27):4416-4429. Google Scholar
- Lee H, Ahn S, Maity R. Mechanisms of antigen escape from BCMA- or GPRC5D-targeted immunotherapies in multiple myeloma. Nat Med. 2023; 29(9):2295-2306. Google Scholar
- Rapoport AP, Stadtmauer EA, Binder-Scholl GK. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015; 21(8):914-921. Google Scholar
- Salter AI, Rajan A, Kennedy JJ. Comparative analysis of TCR and CAR signaling informs CAR designs with superior antigen sensitivity and in vivo function. Sci Signal. 2021; 14(697):eabe2606. Google Scholar
- Teppert K, Wang X, Anders K, Evaristo C, Lock D, Künkele A. Joining forces for cancer treatment: rrom “TCR versus CAR” to “TCR and CAR”. Int J Mol Sci. 2022; 23(23):14563. Google Scholar
- Obenaus M, Leitão C, Leisegang M. Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat Biotechnol. 2015; 33(4):402-407. Google Scholar
- D’Agostino M, Cairns DA, Lahuerta JJ. Second revision of the International Staging System (R2-ISS) for overall survival in multiple myeloma: a European Myeloma Network (EMN) report within the HARMONY project. J Clin Oncol. 2022; 40(29):3406-3418. Google Scholar
- Palumbo A, Avet-Loiseau H, Oliva S. Revised International Staging System for multiple myeloma: a report from International Myeloma Working Group. J Clin Oncol. 2015; 33(26):2863-2869. Google Scholar
- National Cancer Institute. Common terminology criteria for adverse events (CTCAE) Version 4.0. 2024. Publisher Full TextGoogle Scholar
- Kumar S, Paiva B, Anderson KC. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016; 17(8):e328-e346. Google Scholar
- Adebayo OO, Dammer EB, Dill CD. Multivariant transcriptome analysis identifies modules and hub genes associated with poor outcomes in newly diagnosed multiple myeloma patients. Cancers (Basel). 2022; 14(9):2228. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.