PSMA-specific CAR-engineered T cells eradicate disseminated prostate cancer in preclinical models

Gaia Zuccolotto, Giulio Fracasso, Anna Merlo, Isabella Monia Montagner, Maria Rondina, Sara Bobisse, Mariangela Figini, Sara Cingarlini, Marco Colombatti, Paola Zanovello, Antonio Rosato, Gaia Zuccolotto, Giulio Fracasso, Anna Merlo, Isabella Monia Montagner, Maria Rondina, Sara Bobisse, Mariangela Figini, Sara Cingarlini, Marco Colombatti, Paola Zanovello, Antonio Rosato

Abstract

Immunology-based interventions have been proposed as a promising curative chance to effectively attack postoperative minimal residual disease and distant metastatic localizations of prostate tumors. We developed a chimeric antigen receptor (CAR) construct targeting the human prostate-specific membrane antigen (hPSMA), based on a novel and high affinity specific mAb. As a transfer method, we employed last-generation lentiviral vectors (LV) carrying a synthetic bidirectional promoter capable of robust and coordinated expression of the CAR molecule, and a bioluminescent reporter gene to allow the tracking of transgenic T cells after in vivo adoptive transfer. Overall, we demonstrated that CAR-expressing LV efficiently transduced short-term activated PBMC, which in turn were readily stimulated to produce cytokines and to exert a relevant cytotoxic activity by engagement with PSMA+ prostate tumor cells. Upon in vivo transfer in tumor-bearing mice, CAR-transduced T cells were capable to completely eradicate a disseminated neoplasia in the majority of treated animals, thus supporting the translation of such approach in the clinical setting.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. CAR development and validation.
Figure 1. CAR development and validation.
(A) Anti-hPSMA CAR map. EC: extracellular domain; TM: transmembrane domain; IC: intracellular domain. (B) Cytofluorimetric profile of CAR expression in transfected 293 T cells. 293 T cells were transfected with pcDNA3.1 vector bearing CAR sequence and analysed by flow cytometry after 24 hours (dark line). Untransfected cells (grey plot) served as control. (C) CAR expression as assessed by Western blotting at 24 hours (line 1) and 7 days (line 2) post 293 T cell transfection. Negative controls were the medium alone (line 3) and untransfected 293 T cells (line 4). Line 5, molecular weight markers. (D) Linear map of the recombinant viral vector containing the minCMVPGK bidirectional promoter (22). In particular, the reporter gene (eGFP or Luciferase) is under the control of minCMV promoter, while the CAR gene is under control of hPGK promoter. (E) Transduction of Jurkat cells with LV CAR anti-hPSMA/eGFP. Co-expression of c-myc and eGFP in LV-transduced Jurkat cells, as assessed by flow cytometry. Dot plot reports the events gated on total viable cells; more than 90% of cells co-express both c-myc and eGFP. (F) Transduction of Jurkat cells with LV CAR anti-hPSMA/Luciferase. Left panel, c-myc expression (black) in Jurkat cells at 72 h post transduction, as assessed by flow cytometry. Grey plot represents the isotype control. Right panel, Assessment of luciferase activity in LV-transduced Jurkat cells (2×105/well) by BLI.
Figure 2. Generation, expansion and phenotipic properties…
Figure 2. Generation, expansion and phenotipic properties of T-bodies.
(A) Set up of transduction protocol and growth kinetics. Left panel, PBMC were activated with OKT3 at time 0 and transduced concomitantly (Td0), or after 48-h (Td2) or 72-h (Td3) hours. Flow cytometry analysis of c-myc tag expression was performed 72 hours post infection. Central panel reports the expression of CD27, CD28 and CD62L in the three different conditions of activation/infection reported in the left panel. Right panel, Expansion of T-body-hPSMA/fluc and T-body-hPSMA/eGFP transduced after 48 h activation in response to weekly restimulation with PC3-PIP cells. Untransduced PBMC were used as control. (B) Phenotype and CAR expression in transduced T cells upon stimulation. Left panel, Expression of surface markers in T-body-anti-hPSMA/fluc at different time points (3, 11 and 18 days) post transduction, as assessed by flow cytometry. Data are representative of 3 independent experiments. Overlapping results were obtained in T-body-anti-hPSMA/eGFP. Right panel, percentage of c-myc+ cells in LV CAR hPSMA/fluc and LV CAR hPSMA/eGFP-transduced T cell populations at different time points post transduction. Figure shows mean +/− SD of at least three independent experiments. (C) Kinetics of CD4 and CD8 subsets in transduced T cell populations. Expression of CD4 and CD8 markers in c-myc+-gated T-body-hPSMA/eGFP (left panel) and T-body-hPSMA/fluc (right panel) populations at different time points post transduction, as assessed by flow cytometry. Data are from a representative experiment out of three that produced similar results.
Figure 3. Functional characterization of T-bodies.
Figure 3. Functional characterization of T-bodies.
(A) Lytic activity of T-body-hPSMA/eGFP and T-body-hPSMA/fluc. Cytotoxicity was analysed at 3, 11 and 60 days post-transduction; PC3, PC3-PIP and LNCaP cells were used as target cells. Untransduced PBMC served as negative control. In the upper left corner of each panel the percentages of c-myc+ T cells are reported. Figure shows mean +/− SD of 4 independent experiments. (B) IFN-γ secretion upon antigen stimulation. IFN-γ production was analyzed at different time points after PBMC transduction by stimulating T-body-hPSMA/fluc with PC3-PIP hPSMA+ or PC3 hPSMA− cancer cell lines. T-bodies unstimulated or treated with PMA/Ionomycin represented the negative and positive controls, respectively. Similar results were obtained with T-body-hPSMA/eGFP. (C) c-myc expression in T-body-hPSMA/fluc populations tested for IFN-γ production.
Figure 4. Assessment of T-body in vivo…
Figure 4. Assessment of T-body in vivo loco-regional therapeutic efficacy.
(A) Winn Assay. PC3-PIP (left panel) and PC3 (right panel) tumor cells were inoculated s.c. in SCID mice, alone or mixed 1∶1 with T-body-hPSMA/eGFP at opposite flanks of the same animal. Tumor growth was monitored over time by caliper measurement. Number of mice per group, n = 6. (B) Loco-regional therapy. T-body-hPSMA/eGFP at 72 hours post transduction were administered intralesionally and perilesionally in SCID mice 4 days after s.c. injection of PC3-PIP tumor cells (n = 6); untreated animals served as control group (n = 6). Left panel shows tumor volumes, while right panel reports Kaplan-Meyer survival curves of treated and untreated mice. (C) Expression of hPSMA antigen in prostate tumors. PC3-PIP tumor cells from in vitro cultures (first quadrant) or isolated ex-vivo from control or treated mice (second and third quadrant, respectively; dark line) were evaluated for hPSMA expression by flow cytometry. The grey plot corresponds to the isotype control.
Figure 5. Assessment of T-body in vivo…
Figure 5. Assessment of T-body in vivo systemic therapeutic efficacy against disseminated prostate carcinoma.
Rag2−/−/γc−/− mice (n = 6) and NOD/SCID mice (n = 6) bearing established bioluminescent PC3-PIP tumors were injected i.v. with 2×107 T-body-hPSMA/eGFP for 3 times at a two day interval. Untreated animals (n = 6 for both mouse strains) were used as controls. (A) Pictures show two representative Rag2−/−/γc−/− and NOD/SCID mice (left and right panels, respectively) imaged by BLI at different time points, whereas (B) and (C) graphs report cumulative results of the regions of interest (ROI) in lungs and in total body, respectively. Tumor growth was monitored as photon flux and quantified as photon * sec−1 * cm−2 * sr−1. Graphs show mean ± SD of three independent experiments. *: P<0.05. The t-Test was used for statistical analysis. (D) Cumulative Kaplan-Meier survival curves of Rag2−/−/γc−/− (left panel; untreated mice, black line; median survival = 54 days; treated mice, red line; median survival = 74 days; P = 0.046) and NOD/SCID mice (right panel; untreated mice, black line; median survival = 60 days; treated mice, red line; median survival = not evaluable; P<0.001).

References

    1. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME (2008) Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat Rev Cancer 8: 299–308.
    1. Zhang Q, Li H, Yang J, Li L, Zhang B, et al. (2013) Strategies to improve the clinical performance of chimeric antigen receptor-modified T cells for cancer. Curr Gene Ther 13: 65–70.
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365: 725–733.
    1. Kochenderfer JN, Rosenberg SA (2013) Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol 10: 267–276.
    1. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509–1518.
    1. Davila ML, Riviere I, Wang X, Bartido S, Park J, et al. (2014) Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6: 224ra25.
    1. Gilham DE, Debets R, Pule M, Hawkins RE, Abken H (2012) CAR-T cells and solid tumors: Tuning T cells to challenge an inveterate foe. Trends Mol Med 18: 377–384.
    1. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, et al. (2011) Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118: 6050–6056.
    1. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, et al. (2008) Virus-specific T cells engineered to coexpress tumor-specific receptors: Persistence and antitumor activity in individuals with neuroblastoma. Nat Med 14: 1264–1270.
    1. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, et al. (2006) A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 12: 6106–6115.
    1. Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, et al. (2013) Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: Clinical evaluation and management of on-target toxicity. Mol Ther 21: 904–912.
    1. Siegel R, Ma J, Zou Z, Jemal A (2014) Cancer statistics, 2014. CA Cancer J Clin 64: 9–29.
    1. Gade TP, Hassen W, Santos E, Gunset G, Saudemont A, et al. (2005) Targeted elimination of prostate cancer by genetically directed human T lymphocytes. Cancer Res 65: 9080–9088.
    1. Simons JW, Sacks N (2006) Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: The GVAX vaccine for prostate cancer. Urol Oncol 24: 419–424.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, et al. (2010) Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 363: 411–422.
    1. Hillerdal V, Ramachandran M, Leja J, Essand M (2014) Systemic treatment with CAR-engineered T cells against PSCA delays subcutaneous tumor growth and prolongs survival of mice. BMC Cancer 14.
    1. Zhong XS, Matsushita M, Plotkin J, Riviere I, Sadelain M (2010) Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther 18: 413–420.
    1. Ristau BT, O’Keefe DS, Bacich DJ (2014) The prostate-specific membrane antigen: Lessons and current clinical implications from 20 years of research. Urol Oncol 32: 272–279.
    1. Frigerio B, Fracasso G, Luison E, Cingarlini S, Mortarino M, et al. (2013) A single-chain fragment against prostate specific membrane antigen as a tool to build theranostic reagents for prostate cancer. Eur J Cancer 49: 2223–2232.
    1. Bobisse S, Rondina M, Merlo A, Tisato V, Mandruzzato S, et al. (2009) Reprogramming T lymphocytes for melanoma adoptive immunotherapy by T-cell receptor gene transfer with lentiviral vectors. Cancer Res. 69: 9385–9394.
    1. Ghosh A, Wang X, Klein E, Heston WD (2005) Novel role of prostate-specific membrane antigen in suppressing prostate cancer invasiveness. Cancer Res 65: 727–731.
    1. Merlo A, Turrini R, Bobisse S, Zamarchi R, Alaggio R, et al. (2010) Virus-specific cytotoxic CD4+ T cells for the treatment of EBV-related tumors. J Immunol 184: 5895–5902.
    1. Amendola M, Venneri MA, Biffi A, Vigna E, Naldini L (2005) Coordinate dual-gene transgenesis by lentiviral vectors carrying synthetic bidirectional promoters. Nat Biotechnol 23: 108–116.
    1. Keyaerts M, Verschueren J, Bos TJ, Tchouate-Gainkam LO, Peleman C, et al. (2008) Dynamic bioluminescence imaging for quantitative tumour burden assessment using IV or IP administration of D-Luciferin: Effect on intensity, time kinetics and repeatability of photon emission. Eur J Nucl Med Mol Imaging 35: 999–1007.
    1. Rosato A, Milan G, Collavo D, Zanovello P (1999) DNA-based vaccination against tumors expressing the P1A antigen. Methods 19: 187–190.
    1. Ma Q, Gomes EM, Lo AS, Junghans RP (2014) Advanced generation anti-prostate specific membrane antigen designer T cells for prostate cancer immunotherapy. Prostate 74: 286–296.
    1. Abate-Daga D, Lagisetty KH, Tran E, Zheng Z, Gattinoni L, et al. (2014) A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Hum Gene Ther.
    1. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, et al. (2012) CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood 119: 3940–3950.
    1. Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, et al. (2009) Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 17: 1453–1464.
    1. Hombach AA, Rappl G, Abken H (2013) Arming cytokine-induced killer cells with chimeric antigen receptors: CD28 outperforms combined CD28-OX40 “super-stimulation”. Mol Ther 21: 2268–2277.
    1. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, et al. (2011) CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 121: 1822–1826.
    1. Rochford R, Hobbs MV, Garnier JL, Cooper NR, Cannon MJ (1993) Plasmacytoid differentiation of epstein-barr virus-transformed B cells in vivo is associated with reduced expression of viral latent genes. Proc Natl Acad Sci U S A 90: 352–356.
    1. Turrini R, Merlo A, Dolcetti R, Zanovello P, Rosato A (2011) Differential down-modulation of HLA class I and II molecule expression on human tumor cell lines upon in vivo transfer. Cancer Immunol Immunother 60: 1639–1645.
    1. Anurathapan U, Chan RC, Hindi HF, Mucharla R, Bajgain P, et al. (2014) Kinetics of tumor destruction by chimeric antigen receptor-modified T cells. Mol Ther 22: 623–633.
    1. Casucci M, Nicolis di Robilant B, Falcone L, Camisa B, Norelli M, et al. (2013) CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 122: 3461–3472.
    1. Braun CJ, Boztug K, Paruzynski A, Witzel M, Schwarzer A, et al. (2014) Gene therapy for wiskott-aldrich syndrome–long-term efficacy and genotoxicity. Sci Transl Med 6: 227ra33.
    1. Biffi A, Bartolomae CC, Cesana D, Cartier N, Aubourg P, et al. (2011) Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. Blood 117: 5332–5339.
    1. Cesana D, Ranzani M, Volpin M, Bartholomae C, Duros C, et al. (2014) Uncovering and dissecting the genotoxicity of self-inactivating lentiviral vectors in vivo. Mol Ther 22: 774–785.
    1. Sampson JH, Choi BD, Sanchez-Perez L, Suryadevara CM, Snyder DJ, et al. (2014) EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin Cancer Res 20: 972–984.
    1. Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, et al. (2009) A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol 183: 5563–5574.
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, et al. (2010) Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 18: 843–851.
    1. Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, et al. (2011) Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 365: 1673–1683.
    1. Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M (2013) Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol 31: 71–75.

Source: PubMed

3
Prenumerera