We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Safety and efficacy of mutant neoantigen-specific T-cell treatment combined anti-PD-1 therapy in stage IV solid tumors

    Qi Song‡

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Bo Yang‡

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Wei Sheng‡

    Department of Tissue Repair & Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Zishan Zhou

    Beijing DCTY Biotech Co., Ltd, Beijing, China

    ,
    Tianfu Zhang

    Beijing DCTY Biotech Co., Ltd, Beijing, China

    ,
    Boyu Qin

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    , , ,
    Dan Wang

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Xiaoling Zhang

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Shengjie Sun

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Guoqing Zhang

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Xiao Zhao

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Quan Gan

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    ,
    Qi Xiong

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    , , , ,
    Xiudi Chen

    BeiGene Co., Ltd, Beijing, China

    ,
    Wei Guo

    BeiGene Co., Ltd, Beijing, China

    &
    Shunchang Jiao

    *Author for correspondence: Tel.: +86 138 1156 3597;

    E-mail Address: jiaoshunchang@csco.org.cn

    Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China

    Published Online:https://doi.org/10.2217/imt-2021-0105

    Aims: This trial explored the safety and efficacy of neoantigen-specific T cells (Nas-Ts) combined with anti-PD-1 (Nas-T + anti-PD-1). Patients & methods: This non-randomized trial recruited participants with solid tumors treated with at least two prior systemic treatment lines. For comparison, 1:1-matched controls who received anti-PD-1 alone were recruited. The primary end point was safety. Results: 15 participants were enrolled in the Nas-T + anti-PD-1 group, the objective response rate was 33.3%, and the disease control rate was 93.3%. The median progression-free survival was significantly different between the Nas-T + anti-PD-1 and control groups (13.8 vs 4.2 months; p = 0.024), but no difference in overall survival was found (p = 0.126). The most common adverse events were maculopapular skin reaction (53.3%), rash (53.3%), hepatotoxicity (53.3%) and fever (53.3%) in the Nas-T + anti-PD-1 group. No serious safety issues were experienced. Conclusion: Nas-Ts combined with anti-PD-1 could be more effective than anti-PD-1 alone in prolonging progression-free survival, with good safety.

    Plain language summary

    Cancer immune escape is a major mechanism allowing cancer cells to avoid treatments, and PD-1 is one of those mechanisms. Nevertheless, therapies targeting PD-1 are still somewhat unsatisfactory. In this trial, we explored the safety and efficacy of mutant neoantigen-specific T cells (Nas-Ts) as adoptive cell immunotherapy individualized for each tumor, combined with an anti-PD-1 regimen (Nas-T + anti-PD-1). We recruited participants with solid tumors treated with at least two prior systemic treatment lines: 15 participants were enrolled in the Nas-T + anti-PD-1 group and 15 more in the control group. After the last follow-up, the percentage of patients on whom a therapy had some defined effect as well as the percentage of patients with advanced and metastatic cancer who achieved complete response was significantly higher for those who received Nas-T + anti-PD-1. No serious safety issues were experienced. This study confirmed that Nas-Ts combined with anti-PD-1 could be more effective than anti-PD-1 alone in delaying progression, with good safety.

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1. Bray F, Ferlay J, Soerjomataram I et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68(6), 394–424 (2018).
    • 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J. Clin. 70(1), 7–30 (2020).
    • 3. Vinay DS, Ryan EP, Pawelec G et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 35(Suppl.), 185–198 (2015). •• Provides a description of mechanisms of immune evasion of cancer cells and the potential strategies to counter it.
    • 4. Vanichapol T, Chutipongtanate S, Anurathapan U, Hongeng S. Immune escape mechanisms and future prospects for immunotherapy in neuroblastoma. Biomed. Res. Int. 18, 1812535 (2018).
    • 5. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 236, 219–242 (2010).
    • 6. Fife BT, Pauken KE. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Ann. NY Acad. Sci. 1217(1), 45–59 (2011). •• Provides a description of the role of PD-1 in the immune system.
    • 7. Syn NL, Teng MWL, Mok TSK, Soo RA. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol. 18(12), e731–e741 (2017). •• Describes the reasons for resistance to immune checkpoint inhibitors.
    • 8. Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco. Targets Ther. 9, 5023–5039 (2016).
    • 9. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 100, 88–98 (2016).
    • 10. Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer – preclinical background: CTLA-4 and PD-1 blockade. Semin. Oncol. 37(5), 430–439 (2010).
    • 11. Iorgulescu JB, Braun D, Oliveira G, Keskin DB, Wu CJ. Acquired mechanisms of immune escape in cancer following immunotherapy. Genome Med. 10(1), 87 (2018). •• Also describes the reasons for resistance to immune checkpoint inhibitors.
    • 12. Jiang Y, Zhao X, Fu J, Wang H. Progress and challenges in precise treatment of tumors with PD-1/PD-L1 blockade. Front. Immunol. 11, 339 (2020).
    • 13. Jung CY, Antonia SJ. Tumor immunology and immune checkpoint inhibitors in non-small cell lung cancer. Tuberc. Respir. Dis. (Seoul) 81(1), 29–41 (2018).
    • 14. Robbins PF, Kassim SH, Tran TL 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. 21(5), 1019–1027 (2015).
    • 15. Li Q, Liu M, Wu M et al. PLAC1-specific TCR-engineered T cells mediate antigen-specific antitumor effects in breast cancer. Oncol. Lett. 15(4), 5924–5932 (2018).
    • 16. Oh J, Warshaviak DT, Mkrtichyan M et al. Single variable domains from the T cell receptor beta chain function as mono- and bifunctional CARs and TCRs. Sci. Rep. 9(1), 17291 (2019).
    • 17. Liu S, Matsuzaki J, Wei L et al. Efficient identification of neoantigen-specific T-cell responses in advanced human ovarian cancer. J. Immunother. Cancer 7(1), 156 (2019).
    • 18. Perumal D, Imai N, Lagana A et al. Mutation-derived neoantigen-specific T-cell responses in multiple myeloma. Clin. Cancer Res. 26(2), 450–464 (2020).
    • 19. Tran E, Robbins PF, Lu YC et al. T-cell transfer therapy targeting mutant KRAS in cancer. N. Engl. J. Med. 375(23), 2255–2262 (2016).
    • 20. Tran E, Turcotte S, Gros A et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344(6184), 641–645 (2014).
    • 21. Yossef R, Tran E, Deniger DC et al. Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy. JCI Insight 3(19), e122467 (2018).
    • 22. CD30-targeted CAR T cells show promise in pretreated Hodgkin lymphoma. Cancer Discov. 10(9), 1253 (2020).
    • 23. Keskin DB, Anandappa AJ, Sun J et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 565(7738), 234–239 (2019).
    • 24. Ott PA, Dotti G, Yee C, Goff SL. An update on adoptive T-cell therapy and neoantigen vaccines. Am. Soc. Clin. Oncol. Educ. Book 39, e70–e78 (2019).
    • 25. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45(2), 228–247 (2009).
    • 26. Jia Q, Wu W, Wang Y et al. Local mutational diversity drives intratumoral immune heterogeneity in non-small cell lung cancer. Nat. Commun. 9(1), 5361 (2018).
    • 27. Jia Q, Zhou J, Chen G et al. Diversity index of mucosal resident T lymphocyte repertoire predicts clinical prognosis in gastric cancer. Oncoimmunology 4(4), e1001230 (2015).
    • 28. Postow MA, Manuel M, Wong P et al. Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J. Immunother. Cancer 3, 23 (2015).
    • 29. Parkhurst MR, Yang JC, Langan RC et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther. 19(3), 620–626 (2011).
    • 30. Linette GP, Carreno BM. Dendritic cell-based vaccines: shining the spotlight on signal 3. Oncoimmunology 2(11), e26512 (2013).
    • 31. D’Angelo SP, Melchiori L, Merchant MS et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 (c259)T cells in synovial sarcoma. Cancer Discov. 8(8), 944–957 (2018).
    • 32. Guo Y, Feng K, Liu Y et al. Phase I study of chimeric antigen receptor-modified T cells in patients with EGFR-positive advanced biliary tract cancers. Clin. Cancer Res. 24(6), 1277–1286 (2018).
    • 33. Feng K, Liu Y, Guo Y et al. Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell 9(10), 838–847 (2018).
    • 34. Morgan RA, Chinnasamy N, Abate-Daga D et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 36(2), 133–151 (2013).
    • 35. Morgan RA, Dudley ME, Wunderlich JR et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314(5796), 126–129 (2006).
    • 36. Robbins PF, Morgan RA, Feldman SA et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 29(7), 917–924 (2011).
    • 37. Kageyama S, Ikeda H, Miyahara Y et al. Adoptive transfer of MAGE-A4 T-cell receptor gene-transduced lymphocytes in patients with recurrent esophageal cancer. Clin. Cancer Res. 21(10), 2268–2277 (2015).
    • 38. Beatty GL, O’Hara MH, Lacey SF et al. Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology 155(1), 29–32 (2018).
    • 39. Hege KM, Bergsland EK, Fisher GA et al. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J. Immunother. Cancer 5, 22 (2017).
    • 40. Moon EK, Ranganathan R, Eruslanov E et al. Blockade of programmed death 1 augments the ability of human T cells engineered to target NY-ESO-1 to control tumor growth after adoptive transfer. Clin. Cancer. Res. 22(2), 436–447 (2016).
    • 41. Rizvi NA, Hellmann MD, Snyder A et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348(6230), 124–128 (2015).
    • 42. Kirsch I, Vignali M, Robins H. T-cell receptor profiling in cancer. Mol. Oncol. 9(10), 2063–2070 (2015).
    • 43. Bobisse S, Genolet R, Roberti A et al. Sensitive and frequent identification of high avidity neo-epitope specific CD8 (+) T cells in immunotherapy-naive ovarian cancer. Nat. Commun. 9(1), 1092 (2018).
    • 44. Tumeh PC, Harview CL, Yearley JH et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515(7528), 568–571 (2014).
    • 45. Hogan SA, Courtier A, Cheng PF et al. Peripheral blood TCR repertoire profiling may facilitate patient stratification for immunotherapy against melanoma. Cancer Immunol. Res. 7(1), 77–85 (2019).
    • 46. Wang DY, Eroglu Z, Ozgun A et al. Clinical features of acquired resistance to anti-PD-1 therapy in advanced melanoma. Cancer Immunol. Res. 5(5), 357–362 (2017).
    • 47. Cui P, Li R, Huang Z et al. Comparative effectiveness of pembrolizumab vs. nivolumab in patients with recurrent or advanced NSCLC. Sci. Rep. 10(1), 13160 (2020).
    • 48. Zhou H, Fu X, Li Q, Niu T. Safety and efficacy of anti-PD-1 monoclonal antibodies in patients with relapsed or refractory lymphoma: a meta-analysis of prospective clinic trails. Front. Pharmacol. 10, 387 (2019).