Phase I study of a multitargeted recombinant Ad5 PSA/MUC-1/brachyury-based immunotherapy vaccine in patients with metastatic castration-resistant prostate cancer (mCRPC)

Marijo Bilusic, Sheri McMahon, Ravi A Madan, Fatima Karzai, Yo-Ting Tsai, Renee N Donahue, Claudia Palena, Caroline Jochems, Jennifer L Marté, Charalampos Floudas, Julius Strauss, Jason Redman, Houssein Abdul Sater, Shahrooz Rabizadeh, Patrick Soon-Shiong, Jeffrey Schlom, James L Gulley, Marijo Bilusic, Sheri McMahon, Ravi A Madan, Fatima Karzai, Yo-Ting Tsai, Renee N Donahue, Claudia Palena, Caroline Jochems, Jennifer L Marté, Charalampos Floudas, Julius Strauss, Jason Redman, Houssein Abdul Sater, Shahrooz Rabizadeh, Patrick Soon-Shiong, Jeffrey Schlom, James L Gulley

Abstract

Background: Antitumor vaccines targeting tumor-associated antigens (TAAs) can generate antitumor immune response. A novel vaccine platform using adenovirus 5 (Ad5) vectors [E1-, E2b-] targeting three TAAs-prostate-specific antigen (PSA), brachyury, and MUC-1-has been developed. Both brachyury and the C-terminus of MUC-1 are overexpressed in metastatic castration-resistant prostate cancer (mCRPC) and have been shown to play an important role in resistance to chemotherapy, epithelial-mesenchymal transition, and metastasis. The transgenes for PSA, brachyury, and MUC-1 all contain epitope modifications for the expression of CD8+ T-cell enhancer agonist epitopes. We report here the first-in-human trial of this vaccine platform.

Methods: Patients with mCRPC were given concurrently three vaccines targeting PSA, brachyury, and MUC-1 at 5×1011 viral particles (VP) each, subcutaneously every 3 weeks for a maximum of three doses (dose de-escalation cohort), followed by a booster vaccine every 8 weeks for 1 year (dose-expansion cohort only). The primary objective was to determine the safety and the recommended phase II dose. Immune assays and clinical responses were evaluated.

Results: Eighteen patients with mCRPC were enrolled between July 2018 and September 2019 and received at least one vaccination. Median PSA was 25.58 ng/mL (range, 0.65-1006 ng/mL). The vaccine was tolerable and safe, and no grade >3 treatment-related adverse events or dose-limiting toxicities (DLTs) were observed. One patient had a partial response, while five patients had confirmed PSA decline and five had stable disease for >6 months. Median progression-free survival was 22 weeks (95% CI: 19.1 to 34). Seventeen (100%) of 17 patients mounted T-cell responses to at least one TAA, whereras 8 (47%) of 17 patients mounted immune responses to all three TAAs. Multifunctional T-cell responses to PSA, MUC-1, and brachyury were also detected after vaccination in the majority of the patients.

Conclusions: Ad5 PSA/MUC-1/brachyury vaccine is well tolerated. The primary end points were met and there were no DLTs. The recommended phase II dose is 5×1011 VP. The vaccine demonstrated clinical activity, including one partial response and confirmed PSA responses in five patients. Three patients with prolonged PSA responses received palliative radiation therapy. Further research is needed to evaluate the clinical benefit and immunogenicity of this vaccine in combination with other immuno-oncology agents and/or palliative radiation therapy.

Trial registration number: NCT03481816.

Keywords: immunogenicity; prostatic neoplasms; vaccine.

Conflict of interest statement

Competing interests: ImmunityBio authors are employees of ImmunityBio, Inc.

© 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
PSA changes. This spider plot of PSA measurement data shows percentage change over time: (A) all patients and (B) PSA responders only. PSA, prostate-specific antigen.
Figure 2
Figure 2
Waterfall plot of best PSA response. PSA response was confirmed in five patients. PSA, prostate-specific antigen.

References

    1. Cunha AC, Weigle B, Kiessling A, et al. . Tissue-specificity of prostate specific antigens: comparative analysis of transcript levels in prostate and non-prostatic tissues. Cancer Lett 2006;236:229–38. 10.1016/j.canlet.2005.05.021
    1. Cole G, McCaffrey J, Ali AA, et al. . Dna vaccination for prostate cancer: key concepts and considerations. Cancer Nanotechnol 2015;6:2. 10.1186/s12645-015-0010-5
    1. Coffey DS, Isaacs JT. Prostate tumor biology and cell kinetics--theory. Urology 1981;17:40–53.
    1. Gulley JL, Borre M, Vogelzang NJ, et al. . Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J Clin Oncol 2019;37:1051–61. 10.1200/JCO.18.02031
    1. Imler JL. Adenovirus vectors as recombinant viral vaccines. Vaccine 1995;13:1143–51. 10.1016/0264-410X(95)00032-V
    1. Tatsis N, Ertl HCJ. Adenoviruses as vaccine vectors. Mol Ther 2004;10:616–29. 10.1016/j.ymthe.2004.07.013
    1. Gabitzsch ES, Jones FR. New recombinant Ad5 vector overcomes Ad5 immunity allowing for multiple safe, homologous immunizations. J Clin Cellular Immunol 2012;2012:1–4.
    1. Osada T, Yang XY, Hartman ZC, et al. . Optimization of vaccine responses with an E1, E2b and E3-deleted Ad5 vector circumvents pre-existing anti-vector immunity. Cancer Gene Ther 2009;16:673–82. 10.1038/cgt.2009.17
    1. Gabitzsch ES, Xu Y, Yoshida LH, et al. . A preliminary and comparative evaluation of a novel Ad5 [E1-, E2b-] recombinant-based vaccine used to induce cell mediated immune responses. Immunol Lett 2009;122:44–51. 10.1016/j.imlet.2008.11.003
    1. Gabitzsch ES, Xu Y, Balint JP, et al. . Induction and comparison of SIV immunity in Ad5 naïve and Ad5 immune non-human primates using an Ad5 [E1-, E2b-] based vaccine. Vaccine 2011;29:8101–7. 10.1016/j.vaccine.2011.08.038
    1. Gabitzsch ES, Tsang KY, Palena C, et al. . The generation and analyses of a novel combination of recombinant adenovirus vaccines targeting three tumor antigens as an immunotherapeutic. Oncotarget 2015;6:31344–59. 10.18632/oncotarget.5181
    1. Gatti-Mays ME, Redman JM, Donahue RN, et al. . A Phase I Trial Using a Multitargeted Recombinant Adenovirus 5 (CEA/MUC1/Brachyury)-Based Immunotherapy Vaccine Regimen in Patients with Advanced Cancer. Oncologist 2020;25:479–899. 10.1634/theoncologist.2019-0608
    1. Balint JP, Gabitzsch ES, Rice A, et al. . Extended evaluation of a phase 1/2 trial on dosing, safety, immunogenicity, and overall survival after immunizations with an advanced-generation Ad5 [E1-, E2b-]-CEA(6D) vaccine in late-stage colorectal cancer. Cancer Immunol Immunother 2015;64:977–87. 10.1007/s00262-015-1706-4
    1. Madan RA, Mohebtash M, Arlen PM, et al. . Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012;13:501–8. 10.1016/S1470-2045(12)70006-2
    1. Heery CR, Palena C, McMahon S, et al. . Phase I study of a Poxviral TRICOM-Based vaccine directed against the transcription factor Brachyury. Clin Cancer Res 2017;23:6833–45. 10.1158/1078-0432.CCR-17-1087
    1. Morse MA, Chaudhry A, Gabitzsch ES, et al. . Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother 2013;62:1293–301. 10.1007/s00262-013-1400-3
    1. Gabitzsch ES, Xu Y, Balint JP, et al. . Anti-tumor immunotherapy despite immunity to adenovirus using a novel adenoviral vector Ad5 [E1-, E2b-]-CEA. Cancer Immunol Immunother 2010;59:1131–5. 10.1007/s00262-010-0847-8
    1. Gabitzsch ES, Xu Y, Balcaitis S, et al. . An Ad5[E1-, E2b-]-HER2/neu vector induces immune responses and inhibits HER2/neu expressing tumor progression in Ad5 immune mice. Cancer Gene Ther 2011;18:326–35. 10.1038/cgt.2010.82
    1. Gatti-Mays ME, Strauss J, Donahue RN, et al. . A phase I dose-escalation trial of BN-CV301, a recombinant Poxviral vaccine targeting MUC1 and CEA with costimulatory molecules. Clin Cancer Res 2019;25:4933–44. 10.1158/1078-0432.CCR-19-0183
    1. Huang J, Jochems C, Anderson AM, et al. . Soluble CD27-pool in humans may contribute to T cell activation and tumor immunity. J Immunol 2013;190:6250–8. 10.4049/jimmunol.1300022
    1. Schlom J, Jochems C, Gulley JL, et al. . The role of soluble CD40L in immunosuppression. Oncoimmunology 2013;2:e22546. 10.4161/onci.22546
    1. Huang J, Jochems C, Talaie T, et al. . Elevated serum soluble CD40 ligand in cancer patients may play an immunosuppressive role. Blood 2012;120:3030–8. 10.1182/blood-2012-05-427799
    1. Wimmers F, Aarntzen EHJG, Duiveman-deBoer T, et al. . Long-lasting multifunctional CD8+ T cell responses in end-stage melanoma patients can be induced by dendritic cell vaccination. Oncoimmunology 2016;5:e1067745. 10.1080/2162402X.2015.1067745
    1. Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst 2013;105:256–65. 10.1093/jnci/djs629
    1. Garnett-Benson C, Hodge JW, Gameiro SR. Combination regimens of radiation therapy and therapeutic cancer vaccines: mechanisms and opportunities. Semin Radiat Oncol 2015;25:46–53. 10.1016/j.semradonc.2014.07.002
    1. Formenti SC. Silvia Formenti on the promise of combining radiotherapy and immunotherapy to treat cancer. Oncology 2016;30:289–92.
    1. Gameiro SR, Jammeh ML, Wattenberg MM, et al. . Radiation-Induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 2014;5:403–16. 10.18632/oncotarget.1719
    1. Chakraborty M, Abrams SI, Camphausen K, et al. . Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 2003;170:6338–47. 10.4049/jimmunol.170.12.6338
    1. Griffith TS, Brunner T, Fletcher SM, et al. . Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 1995;270:1189–92. 10.1126/science.270.5239.1189
    1. Abrahams VM, Kamsteeg M, Mor G. The Fas/Fas ligand system and cancer: immune privilege and apoptosis. Mol Biotechnol 2003;25:19–30. 10.1385/MB:25:1:19
    1. Gameiro SR, Ardiani A, Kwilas A, et al. . Radiation-Induced survival responses promote immunogenic modulation to enhance immunotherapy in combinatorial regimens. Oncoimmunology 2014;3:e28643. 10.4161/onci.28643
    1. Dewan MZ, Galloway AE, Kawashima N, et al. . Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 2009;15:5379–88. 10.1158/1078-0432.CCR-09-0265
    1. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer 2004;4:45–60. 10.1038/nrc1251
    1. Kufe DW. Functional targeting of the MUC1 oncogene in human cancers. Cancer Biol Ther 2009;8:1197–203. 10.4161/cbt.8.13.8844
    1. Yin L, Kufe D. Muc1-C oncoprotein blocks terminal differentiation of chronic myelogenous leukemia cells by a ROS-mediated mechanism. Genes Cancer 2011;2:56–64. 10.1177/1947601911405044
    1. Roselli M, Fernando RI, Guadagni F, et al. . Brachyury, a driver of the epithelial-mesenchymal transition, is overexpressed in human lung tumors: an opportunity for novel interventions against lung cancer. Clin Cancer Res 2012;18:3868–79. 10.1158/1078-0432.CCR-11-3211
    1. Kilic N, Feldhaus S, Kilic E, et al. . Brachyury expression predicts poor prognosis at early stages of colorectal cancer. Eur J Cancer 2011;47:1080–5. 10.1016/j.ejca.2010.11.015
    1. Pinto F, Pértega-Gomes N, Pereira MS, et al. . T-Box transcription factor Brachyury is associated with prostate cancer progression and aggressiveness. Clin Cancer Res 2014;20:4949–61. 10.1158/1078-0432.CCR-14-0421
    1. Palena C, Roselli M, Litzinger MT, et al. . Overexpression of the EMT driver Brachyury in breast carcinomas: association with poor prognosis. J Natl Cancer Inst 2014;106. 10.1093/jnci/dju054. [Epub ahead of print: 09 May 2014].
    1. Haro A, Yano T, Kohno M, et al. . Expression of Brachyury gene is a significant prognostic factor for primary lung carcinoma. Ann Surg Oncol 2013;20 Suppl 3:509–16. 10.1245/s10434-013-2914-9
    1. Wong N, Major P, Kapoor A, et al. . Amplification of MUC1 in prostate cancer metastasis and CRPC development. Oncotarget 2016;7:83115–33. 10.18632/oncotarget.13073
    1. Collins JM, Donahue RN, Tsai Y-T, et al. . Phase I trial of a modified vaccinia Ankara priming vaccine followed by a fowlpox virus boosting vaccine modified to express Brachyury and costimulatory molecules in advanced solid tumors. Oncologist 2020;25:560–1006. 10.1634/theoncologist.2019-0932
    1. Heery CR, Singh BH, Rauckhorst M, et al. . Phase I trial of a yeast-based therapeutic cancer vaccine (GI-6301) targeting the transcription factor Brachyury. Cancer Immunol Res 2015;3:1248–56. 10.1158/2326-6066.CIR-15-0119
    1. Duggan MC, Jochems C, Donahue RN, et al. . A phase I study of recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)-TRICOM vaccines with GM-CSF and IFN-α-2b in patients with CEA-expressing carcinomas. Cancer Immunol Immunother 2016;65:1353–64. 10.1007/s00262-016-1893-7
    1. Abdul Sater H, Marté JL, Donahue RN, et al. . Neoadjuvant PROSTVAC prior to radical prostatectomy enhances T-cell infiltration into the tumor immune microenvironment in men with prostate cancer. J Immunother Cancer 2020;8:e000655. 10.1136/jitc-2020-000655

Source: PubMed

3
Subscribe