Phase 1 Clinical Trial Evaluating the Safety and Anti-Tumor Activity of ADP-A2M10 SPEAR T-Cells in Patients With MAGE-A10+ Head and Neck, Melanoma, or Urothelial Tumors

David S Hong, Marcus O Butler, Russell K Pachynski, Ryan Sullivan, Partow Kebriaei, Sarah Boross-Harmer, Armin Ghobadi, Matthew J Frigault, Ecaterina E Dumbrava, Amy Sauer, Francine Brophy, Jean-Marc Navenot, Svetlana Fayngerts, Zohar Wolchinsky, Robyn Broad, Dzmitry G Batrakou, Ruoxi Wang, Luisa M Solis, Dzifa Yawa Duose, Joseph P Sanderson, Andrew B Gerry, Diane Marks, Jane Bai, Elliot Norry, Paula M Fracasso, David S Hong, Marcus O Butler, Russell K Pachynski, Ryan Sullivan, Partow Kebriaei, Sarah Boross-Harmer, Armin Ghobadi, Matthew J Frigault, Ecaterina E Dumbrava, Amy Sauer, Francine Brophy, Jean-Marc Navenot, Svetlana Fayngerts, Zohar Wolchinsky, Robyn Broad, Dzmitry G Batrakou, Ruoxi Wang, Luisa M Solis, Dzifa Yawa Duose, Joseph P Sanderson, Andrew B Gerry, Diane Marks, Jane Bai, Elliot Norry, Paula M Fracasso

Abstract

Background: ADP-A2M10 specific peptide enhanced affinity receptor (SPEAR) T-cells are genetically engineered autologous T-cells that express a high-affinity melanoma-associated antigen (MAGE)-A10-specific T-cell receptor (TCR) targeting MAGE-A10-positive tumors in the context of human leukocyte antigen (HLA)-A*02. ADP-0022-004 is a phase 1, dose-escalation trial to evaluate the safety and anti-tumor activity of ADP-A2M10 in three malignancies (https://ichgcp.net/clinical-trials-registry/NCT02989064" title="See in ClinicalTrials.gov">NCT02989064).

Methods: Eligible patients were HLA-A*02 positive with advanced head and neck squamous cell carcinoma (HNSCC), melanoma, or urothelial carcinoma (UC) expressing MAGE-A10. Patients underwent apheresis; T-cells were isolated, transduced with a lentiviral vector containing the MAGE-A10 TCR, and expanded. Patients underwent lymphodepletion with fludarabine and cyclophosphamide prior to receiving ADP-A2M10. ADP-A2M10 was administered in two dose groups receiving 0.1×109 and >1.2 to 6×109 transduced cells, respectively, and an expansion group receiving 1.2 to 15×109 transduced cells.

Results: Ten patients (eight male and two female) with HNSCC (four), melanoma (three), and UC (three) were treated. Three patients were treated in each of the two dose groups, and four patients were treated in the expansion group. The most frequently reported adverse events grade ≥3 were leukopenia (10), lymphopenia (10), neutropenia (10), anemia (nine), and thrombocytopenia (five). Two patients reported cytokine release syndrome (one each with grade 1 and grade 3), with resolution. Best response included stable disease in four patients, progressive disease in five patients, and not evaluable in one patient. ADP-A2M10 cells were detectable in peripheral blood from patients in each dose group and the expansion group and in tumor tissues from patients in the higher dose group and the expansion group. Peak persistence was greater in patients from the higher dose group and the expansion group compared with the lower dose group.

Conclusions: ADP-A2M10 has shown an acceptable safety profile with no evidence of toxicity related to off-target binding or alloreactivity in these malignancies. Persistence of ADP-A2M10 in the peripheral blood and trafficking of ADP-A2M10 into the tumor was demonstrated. Because MAGE-A10 expression frequently overlaps with MAGE-A4 expression in tumors and responses were observed in the MAGE-A4 trial (NCT03132922), this clinical program closed, and trials with SPEAR T-cells targeting the MAGE-A4 antigen are ongoing.

Keywords: ADP-A2M10; HNSCC; MAGE-A10; TCR; adoptive cellular therapy; melanoma; urothelial carcinoma.

Conflict of interest statement

ABG: holds stock options in Adaptimmune. AS, DB, DM, EN, FB, JB, J-MN, JS, RB, RW, SF, and ZW: employees of Adaptimmune. DH: research/ grant funding from AbbVie, Adaptimmune, Aldi-Norte, Amgen, AstraZeneca, Bayer, BMS, Daiichi-Sankyo, Deciphera, Eisai, Erasca, Fate Therapeutics, Genentech, Genmab, Infinity, Kite, Kyowa, Lilly, LOXO, MedImmune, Merck, Mirati, Mologen, Navier, NCI-CTEP, Novartis, Numab, Pfizer, Pyramid Bio, SeaGen, Takeda, Turning Point Therapeutics, Verastem, and VM Oncology; compensation for travel, accommodations, and expenses from AACR, ASCO, Bayer, Genmab, SITC, and Telperian; consulting, speaker, or advisory roles for Acuta, Adaptimmune, Alkermes, Alpha Insights, Amgen, Atheneum, AUM Biosciences, Axiom, Barclays, Baxter, Bayer, Boxer Capital, BridgeBio, CDR-life AG, COG, COR2ed, Ecor1, Genentech, Gilead, GLG, Group H, Guidepoint, HCW Precision, Immunogen, Infinity, Janssen, Liberium, Medscape, Numab, Oncologia Brasil, Pfizer, Pharma Intelligence, POET Congress, Prime Oncology, Seattle Genetics, ST Cube, Takeda, Tavistock, Trieza Therapeutics, Turning Point, WebMD, and Ziopharm; other ownership interests in OncoResponse (Founder) and Telperian Inc (Advisor). ED: grant/research support from Aileron Therapeutics, Amgen, Aprea Therapeutics, Astex Pharmaceuticals, Bayer HealthCare Pharmaceuticals Inc., Bellicum Pharmaceuticals, BOLT Therapeutics, Compugen Ltd, Immunocore LTD, Immunomedics, Mereo BioPharma 5 Inc, NCI, PMV Pharma, Sanofi, SeaGen, TRACON Pharmaceuticals Inc., Triumvira, and Unum Therapeutics; advisory board for BOLT Therapeutics. MB: grant/contract funding for investigator-initiated clinical trials from Merck and Takara Bio; advisory boards for Adaptimmune, BMS, EMD Serono, GlaxoSmithKline, Immunocore, Instil Bio, Iovance, Merck, Novartis, Pfizer, and Sanofi; has attended speaking engagements for BMS, Merck, Novartis, and Pfizer; Safety Review Committees for Adaptimmune and GlaxoSmithKline. MF: consultant for Arcellx, BMS, Iovance, Kite, and Novartis. PF: employee of Adaptimmune; holds stock in Adaptimmune and Bristol-Myers Squibb; has received compensation for travel and congress meetings. RS: research support from Amgen and Merck; has received fees as a consultant or SAB member from Array Biopharma, Asana Biosciences, AstraZeneca, BMS, Eisai, Iovance, Merck, Novartis, OncoSec, Pfizer, and Replimune. RP: grant funding from Janssen; personal fees from AstraZeneca, Bayer, BMS, Dendreon, EMD, Genentech/ Roche, Genomic Health, Jounce Therapeutics, Merck, Sanofi, and Serono/Pfizer; nonfinancial support from BMS and Genentech/Roche. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this study received funding from Adaptimmune Ltd. The funder had the following involvement with the study: study design and analysis and interpretation of the data.

Copyright © 2022 Hong, Butler, Pachynski, Sullivan, Kebriaei, Boross-Harmer, Ghobadi, Frigault, Dumbrava, Sauer, Brophy, Navenot, Fayngerts, Wolchinsky, Broad, Batrakou, Wang, Solis, Duose, Sanderson, Gerry, Marks, Bai, Norry and Fracasso.

Figures

Figure 1
Figure 1
Study design. HLA, human leukocyte antigen; IHC, immunohistochemical; MAGE, melanoma-associated antigen; PD, progressive disease; SPEAR, specific peptide enhanced affinity receptor.
Figure 2
Figure 2
ADP-A2M10 were detected in peripheral blood and tumor tissue after infusion. (A) Persistence of ADP-A2M10 was measured by quantitative PCR of the Psi element sequence in genomic DNA extracted from peripheral blood mononuclear cells. Data points are colored by response. (B) Representative fields for detection of CD3+ and/or ADP-A2M10 TCR+ cells by CD3 immunohistochemical/RNA in situ hybridization assay in tumor tissue of patient 9 collected within 12 weeks after infusion. In the right image, CD3+ cells are shown in teal, ADP-A2M10 TCR+ cells are shown in purple, and nuclei are shown in light blue (hematoxylin stain). (C) Result table for CD3 immunohistochemical/RNA in situ hybridization duplex assays reporting the detection of ADP-A2M10 in two of four post-infusion tumor samples collected from the study patients. H&E, hematoxylin and eosin stain; HNSCC, head and neck squamous cell carcinoma; Mel, melanoma; NE, not evaluable; PD, progressive disease; SD, stable disease; TCR, T-cell receptor; UC, urothelial carcinoma.
Figure 3
Figure 3
Variability of MAGE-A10 and PD-L1 expression in tumor tissue across the study patients. Pre-infusion (screening and baseline) and post-infusion biopsies collected within 12 weeks after infusion were used for (A) MAGE-A10 expression and (B) PD-L1 expression evaluation. (A) MAGE-A10 expression was assessed by MAGE-A10 immunohistochemical staining and plotted as percentage of tumor cells with 1+, 2+ and 3+ intensities. Horizontal lines designate the cut-off of 10% of tumor with ≥1+ intensity of staining. (B) PD-L1 expression was assessed using PD-L1 IHC 22C3 pharmDx assay and plotted in terms of CPS. (A, B) Data points are colored by response. Patient IDs are indicated by shape. CPS, Combined Positive Score; MAGE, melanoma-associated antigen; NE, not evaluable; PD, progressive disease; PD-L1, programmed death ligand 1; Pre, pre-infusion; SD, stable disease.

References

    1. Colli LM, Machiela MJ, Myers TA, Jessop L, Yu K, Chanock SJ. Burden of Nonsynonymous Mutations Among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res (2016) 76(13):3767–72. doi: 10.1158/0008-5472.CAN-16-0170
    1. Martincorena I, Campbell PJ. Somatic Mutation in Cancer and Normal Cells. Science (2015) 349(6255):1483–9. doi: 10.1126/science.aab4082
    1. Grimes JM, Carvajal RD, Muranski P. Cellular Therapy for the Treatment of Solid Tumors. Transfus Apher Sci (2021) 60(1):103056. doi: 10.1016/j.transci.2021.103056
    1. Jones HF, Molvi Z, Klatt MG, Dao T, Scheinberg DA. Empirical and Rational Design of T Cell Receptor-Based Immunotherapies. Front Immunol (2020) 11:585385. doi: 10.3389/fimmu.2020.585385
    1. Kalos M, June CH. Adoptive T Cell Transfer for Cancer Immunotherapy in the Era of Synthetic Biology. Immunity (2013) 39(1):49–60. doi: 10.1016/j.immuni.2013.07.002
    1. Klebanoff CA, Rosenberg SA, Restifo NP. Prospects for Gene-Engineered T Cell Immunotherapy for Solid Cancers. Nat Med (2016) 22(1):26–36. doi: 10.1038/nm.4015
    1. Weber EW, Maus MV, Mackall CL. The Emerging Landscape of Immune Cell Therapies. Cell (2020) 181(1):46–62. doi: 10.1016/j.cell.2020.03.001
    1. Jazaeri AA, Zsiros E, Amaria RN, Artz AS, Edwards RP, Wenham RM, et al. . Safety and Efficacy of Adoptive Cell Transfer Using Autologous Tumor Infiltrating Lymphocytes (LN-145) for Treatment of Recurrent, Metastatic, or Persistent Cervical Carcinoma. J Clin Oncol (2019) 37:2538. doi: 10.1200/JCO.2019.37.15_suppl.2538
    1. Sarnaik A, Khushalani NI, Chesney JA, Kluger HM, Curti BD, Lewis KD, et al. . Safety and Efficacy of Cryopreserved Autologous Tumor Infiltrating Lymphocyte Therapy (LN-144, Lifileucel) in Advanced Metastatic Melanoma Patients Who Progressed on Multiple Prior Therapies Including Anti-PD-1. J Clin Oncol (2019) 37:2518. doi: 10.1200/JCO.2019.37.15suppl.2518
    1. Nagarsheth NB, Norberg SM, Sinkoe AL, Adhikary S, Meyer TJ, Lack JB, et al. . TCR-Engineered T Cells Targeting E7 for Patients With Metastatic HPV-Associated Epithelial Cancers. Nat Med (2021) 27(3):419–25. doi: 10.1038/s41591-020-01225-1
    1. D’Angelo SP, Melchiori L, Merchant MS, Bernstein D, Glod J, Kaplan R, et al. . Antitumor Activity Associated With Prolonged Persistence of Adoptively Transferred NY-ESO-1c259 T Cells in Synovial Sarcoma. Cancer Discov (2018) 8(8):944–57. doi: 10.1158/-17-1417
    1. Hong D, Clarke J, Johanns T, Kebriaei P, Heymach J, Galal A, et al. . Initial Safety, Efficacy, and Product Attributes From the SURPASS Trial With ADP-A2M4CD8, a SPEAR T-Cell Therapy Incorporating an Affinity Optimized TCR Targeting MAGE-A4 and a CD8α Co-Receptor. J Immunother Cancer (2020) 8:A231. doi: 10.1136/jitc-2020-SITC2020.0379
    1. Hong DS, Van Tine BA, Olszanski AJ, Johnson ML, Liebner DA, Trivedi T, et al. . Phase I Dose Escalation and Expansion Trial to Assess the Safety and Efficacy of ADP-A2M4 SPEAR T Cells in Advanced Solid Tumors. J Clin Oncol (2020) 38:102. doi: 10.1200/JCO.2020.38.15_suppl.102
    1. Robbins PF, Kassim SH, Tran TLN, Crystal JS, Morgan RA, Feldman SA, et al. . A Pilot Trial Using Lymphocytes Genetically Engineered With an NY-ESO-1-Reactive T-Cell Receptor: Long-Term Follow-Up and Correlates With Response. Clin Cancer Res (2015) 21(5):1019–27. doi: 10.1158/1078-0432.CCR-14-2708
    1. Van Tine BA, Hong DS, Johnson ML, Liebner DA, Odunsi K, Trivedi T, et al. . (2020). In: Durable Responses in Patients With Synovial Sarcoma in the Phase I Trial of ADP-A2M4 (MAGE-A4). Presented at: Connective Tissue Oncology Society (CTOS) 2020; Virtual.
    1. Daudi S, Eng KH, Mhawech-Fauceglia P, Morrison C, Miliotto A, Beck A, et al. . Expression and Immune Responses to MAGE Antigens Predict Survival in Epithelial Ovarian Cancer. PloS One (2014) 9(8):e104099. doi: 10.1371/journal.pone.0104099
    1. Lin J, Lin L, Thomas DG, Greenson JK, Giordano TJ, Robinson GS, et al. . Melanoma-Associated Antigens in Esophageal Adenocarcinoma: Identification of Novel MAGE-A10 Splice Variants. Clin Cancer Res (2004) 10(17):5708–16. doi: 10.1158/1078-0432.CCR-04-0468
    1. Mengus C, Schultz-Thater E, Coulot J, Kastelan Z, Goluza E, Coric M, et al. . MAGE-A10 Cancer/Testis Antigen Is Highly Expressed in High-Grade Non-Muscle-Invasive Bladder Carcinomas. Int J Cancer (2013) 132(10):2459–63. doi: 10.1002/ijc.27914
    1. Schultz-Thater E, Piscuoglio S, Iezzi G, Le Magnen C, Zajac P, Carafa V, et al. . MAGE-A10 Is a Nuclear Protein Frequently Expressed in High Percentages of Tumor Cells in Lung, Skin and Urothelial Malignancies. Int J Cancer (2011) 129(5):1137–48. doi: 10.1002/ijc.25777
    1. Su C, Xu Y, Li X, Ren S, Zhao C, Hou L, et al. . Predictive and Prognostic Effect of CD133 and Cancer-Testis Antigens in Stage Ib-IIIA Non-Small Cell Lung Cancer. Int J Clin Exp Pathol (2015) 8(5):5509–18.
    1. Weon JL, Potts PR. The MAGE Protein Family and Cancer. Curr Opin Cell Biol (2015) 37:1–8. doi: 10.1016/j.ceb.2015.08.002
    1. Border EC, Sanderson JP, Weissensteiner T, Gerry AB, Pumphrey NJ. Affinity-Enhanced T-Cell Receptors for Adoptive T-Cell Therapy Targeting MAGE-A10: Strategy for Selection of an Optimal Candidate. Oncoimmunology (2019) 8(2):e1532759. doi: 10.1080/2162402X.2018.1532759
    1. Gonzalez-Galarza FF, McCabe A, Santos E, Jones J, Takeshita L, Ortega-Rivera ND, et al. . Allele Frequency Net Database (AFND) 2020 Update: Gold-Standard Data Classification, Open Access Genotype Data and New Query Tools. Nucleic Acids Res (2020) 48(D1):D783–D88. doi: 10.1093/nar/gkz1029
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. . New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (Version 1.1). Eur J Cancer (2009) 45(2):228–47. doi: 10.1016/j.ejca.2008.10.026
    1. Ramachandran I, Lowther DE, Dryer-Minnerly R, Wang R, Fayngerts S, Nunez D, et al. . Systemic and Local Immunity Following Adoptive Transfer of NY-ESO-1 SPEAR T Cells in Synovial Sarcoma. J Immunother Cancer (2019) 7(1):276. doi: 10.1186/s40425-019-0762-2
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. . Current Concepts in the Diagnosis and Management of Cytokine Release Syndrome. Blood (2014) 124(2):188–95. doi: 10.1182/blood-2014-05-552729
    1. US Food and Drug Administration . Testing of Retroviral Vector-Based Human Gene Therapy Products for Replication Competent Retrovirus During Product Manufacture and Patient Follow-Up. Guidance for Industry (2020). Available at: .
    1. US Food and Drug Administration . Long Term Follow-Up After Administration of Human Gene Therapy Products. Guidance for Industry (2020). Available at: .
    1. European Medicines Agency, Committee for Medicinal Products for Human Use (CHMP) . Guideline on Follow-Up of Patients Administered With Gene Therapy Medicinal Products. Available at: .
    1. Şenbabaoğlu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco G, et al. . Tumor Immune Microenvironment Characterization in Clear Cell Renal Cell Carcinoma Identifies Prognostic and Immunotherapeutically Relevant Messenger RNA Signatures. Genome Biol (2016) 17(1):231. doi: 10.1186/s13059-016-1092-z
    1. Rhodes M, Danaher P, Dennis L, Mashadi-Hossein A, Irving L, Beechem J. Using the PanCancer Immune Profiling Advanced Analysis Module (2019). Available at: .
    1. Woroniecka K, Chongsathidkiet P, Rhodin K, Kemeny H, Dechant C, Farber SH, et al. . T-Cell Exhaustion Signatures Vary With Tumor Type and Are Severe in Glioblastoma. Clin Cancer Res (2018) 24(17):4175–86. doi: 10.1158/1078-0432.CCR-17-1846
    1. Grzywa TM, Paskal W, Wlodarski PK. Intratumor and Intertumor Heterogeneity in Melanoma. Transl Oncol (2017) 10(6):956–75. doi: 10.1016/j.tranon.2017.09.007
    1. Meeks JJ, Al-Ahmadie H, Faltas BM, Taylor 3rd JA, Flaig TW, DeGraff DJ, et al. . Genomic Heterogeneity in Bladder Cancer: Challenges and Possible Solutions to Improve Outcomes. Nat Rev Urol (2020) 17(5):259–70. doi: 10.1038/s41585-020-0304-1
    1. Cohen EE, Dunn L, Neupane P, Gibson M, Leidner R, Savvides P, et al. . SPEARHEAD-2 Trial Design: A Phase II Pilot Trial of ADP-A2M4 in Combination With Pembrolizumab in Patients With Recurrent or Metastatic Head and Neck Cancer. Ann Oncol (2020) 31(suppl_4):S685. doi: 10.1016/j.annonc.2020.08.1091
    1. Vasileiou S, Lulla PD, Tzannou I, Watanabe A, Kuvalekar M, Callejas WL, et al. . T-Cell Therapy for Lymphoma Using Nonengineered Multiantigen-Targeted T Cells Is Safe and Produces Durable Clinical Effects. J Clin Oncol (2021) 39(13):1415–25. doi: 10.1200/JCO.20.02224

Source: PubMed

3
Sottoscrivi