Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2015 May;21(5):524-9.
doi: 10.1038/nm.3833. Epub 2015 Apr 13.

Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes

Affiliations

Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes

Ignazio Caruana et al. Nat Med. 2015 May.

Abstract

Adoptive transfer of chimeric antigen receptor (CAR)-redirected T lymphocytes (CAR-T cells) has had less striking therapeutic effects in solid tumors than in lymphoid malignancies. Although active tumor-mediated immunosuppression may have a role in limiting the efficacy of CAR-T cells, functional changes in T lymphocytes after their ex vivo manipulation may also account for the reduced ability of cultured CAR-T cells to penetrate stroma-rich solid tumors compared with lymphoid tissues. We therefore studied the capacity of human in vitro-cultured CAR-T cells to degrade components of the extracellular matrix (ECM). In contrast to freshly isolated T lymphocytes, we found that in vitro-cultured T lymphocytes lack expression of the enzyme heparanase (HPSE), which degrades heparan sulfate proteoglycans, the main components of ECM. We found that HPSE mRNA is downregulated in in vitro-expanded T cells, which may be a consequence of p53 (officially known as TP53, encoding tumor protein 53) binding to the HPSE gene promoter. We therefore engineered CAR-T cells to express HPSE and showed their improved capacity to degrade the ECM, which promoted tumor T cell infiltration and antitumor activity. The use of this strategy may enhance the activity of CAR-T cells in individuals with stroma-rich solid tumors.

PubMed Disclaimer

Conflict of interest statement

Disclosure conflict-of-interest disclosure: G.D. and B.S. have ownership interest (including patents) in the field of T-cell and gene-modified T-cell therapy for cancer and have a collaborative research agreement with Celgene and Bluebird bio.

Figures

Figure 1
Figure 1. LTE-T show reduced invasion of ECM and loss of the enzyme HPSE
(a) ECM invasion assay of CD14+ monocytes (M), freshly isolated T lymphocytes (FI-T), briefly activated T cells (BA-T) and long-term ex vivo expanded T cells (LTE-T). Monocytes freshly isolated from peripheral blood showed the highest capacity to degrade ECM (63% ± 23%). BA-T showed superior invasion of ECM compared to FI-T (*p=0.05). Conversely, LTE-T had significantly reduced ability to degrade ECM compared to both BA-T (**p=0.01) and FI-T (***p=0.022). Data summarize means ± SD of 5 donors. We compared all four cell subsets for each donor. (b) Western blot showing the expression of HPSE in M, CD4+ and CD8+ T cells at different time points of culture. Data are representative of 4 donors. Positive controls are HPSE transfected 293T cells. (c) Immunofluorescence staining for HPSE in MCF-7 cells, M, FI-T and LTE-T. Nuclei are stained with DAPI and shown in blue, while HPSE is stained with red-fluorescent dye (Alexa Fluor 555). Magnification is 20× (50 micron scale bar). (d) qRT-PCR of HPSE in CD4+ and CD8+ T cells at different time points of culture. Data summarize means ± SD of 4 donors. (e) HPSE enzymatic activity assessed in supernatants collected from CD4+ and CD8+ T cells at different time points of culture. Monocytes and tumor cell lines CHLA-255, A549 and DU-145 are positive controls. Data summarize means ± SD of 4 donors. In (b), (d), (e) the condition “day 15” indicates HPSE expression in LTE-T cultured for 14 days, and re-stimulated with immobilized OKT3 and anti-CD28 Abs for 24 hrs to assess whether TCR re-stimulation can re-induce HPSE expression. (f) Western blot showing the expression of HPSE and full-length p53 protein in T cells before (T=0) and after activation with immobilized OKT3 and anti-CD28 Abs for 18 and 72 hrs. Shown are results from 1 representative of 3 donors. (g) p53 ChIP in LTE-T cultured for 14 days, and (h) in CD45RA+ cells before (T=0) and after stimulation with OKT3 and anti-CD28 mAbs (T=72h). Input is DNA sonicated but not immunoprecipitated; IgG and p53 are DNA immunoprecipitated by the isotype and p53-specific Ab that recognizes the full-length protein. Relative quantification was performed comparing the intensities of PCR bands of IgG and p53 to input PCR band. For this representative sample relative quantifications are: IgG 20% and p53 90% for LTE-T (g); IgG 2% and p53 4% at T=0 and IgG 53% and p53 100% at T=72h for CD45RA+ cells (h). Shown is 1 representative of 3 donors.
Figure 2
Figure 2. LTE-T modified to express HPSE show enhanced degradation of ECM
We transduced LTE-T with a retroviral vector encoding HPSE and GFP [HPSE(I)GFP]. (a) GFP expression by both CD4+ and CD8+ LTE-T at day 12 – 14 of culture. (b) Western blot showing the expression of HPSE in control and transduced LTE-T at day 12 – 14 of culture. (c) qRT-PCR for HPSE in control and HPSE(I)GFP+ LTE-T starved in culture from IL-2 for 3, 7 and 10 days. Data are representative of 2 donors. (d) ECM invasion assay of control and HPSE(I)GFP+ LTE-T, with or without selection based on GFP expression. Data summarize mean ± SD of 9 donors, *p=0.025; **p<0.001. (e) ECM invasion assay of HPSE-transduced LTE-T in the presence or in the absence of the inhibitor, heparin H1. Data summarize mean ± SD of 4 donors, ***p<0.01.
Figure 3
Figure 3. LTE-T co-expressing HPSE and GD2-specific CAR retain GD2 specificity and have enhanced capacity to degrade ECM
We transduced LTE-T with retroviral vectors encoding either the GD2-specific CAR alone (CAR) or both the GD2-specific CAR and HPSE [CAR(I)HPSE]. (a) Flow cytometry analysis to detect CAR expression by control and transduced LTE-T. (b) Western blot to detect HPSE in control and transduced LTE-T. Data are representative of 5 donors. (c) Cytotoxic activity of control, CAR+ and CAR+HPSE+ LTE-T assessed by 51Cr-release assay at a 20:1 effector:target ratio. We used LAN-1 and CHLA-255 (GD2+), and Raji (GD2) as target cells. (d) Transduced LTE-T release both IL-2 and IFN-γ in response to GD2+ tumor cells. (e) Invasion of ECM by control, CAR+ and CAR+HPSE+ LTE-T. Data in (c-e) summarize mean ± SD from 5 donors, *p=0.004. (f,g) We plated control and transduced LTE-T in the upper part of either ECM assay or insert assay, and LAN-1-GFP+ cells in the lower chamber. After day 3 of culture, we collected cells in the lower chamber to quantify CD3+ T cells and GFP+ tumor cells by flow cytometry. (f) illustrates representative dot plots, while (g) summarizes mean ± SD of 5 donors, **p=0.009. In all cases we analyzed LTE-T by day 12 –14 of culture.
Figure 4
Figure 4. CAR-GD2+ LTE-T co-expressing HPSE show enhanced tumor infiltration and improve overall survival in xenograft tumor models
(a) Kaplan-Meier analysis of mice engrafted i.p. with the tumor cell line CHLA-255 and treated i.p. with control, CAR+ and CAR(I)HPSE+ LTE-T. Shown are data from 3 independent experiments using LTE-T generated from 3 donors; control n=16, CAR n=22, CAR(I)HPSE n=26 mice; *p<0.007, **p<0.0001. (b) Kaplan-Meier analysis of mice engrafted i.p. with the tumor cell line LAN-1 and treated i.p. with control, CAR+ and CAR(I)HPSE+ LTE-T. For these experiments, we generated LTE-T from 2 donors; control n=12, CAR n=18, CAR(I)HPSE n=14 mice; *p=0.039, **p<0.0001. (c) Flow cytometry analysis of CD3+ T cells detected within the tumor samples. Dot plots are representative of 3 mice per group from mice infused with LTE-T generated from the same donor. (d) Weight of the tumors collected from euthanized mice engrafted with LAN-1 tumor. (e,f) Immunohistochemistry showing CD3+ T-cell infiltration in tumors implanted in the kidney of mice infused with either CAR+ or CAR-GD2+HPSE+ LTE-T. 100× magnification (e) and 200× magnification (f) (100 micron scale bar). (g) The graph shows the numbers of infiltrating CD3+ T cells per 10 high power fields in tumors collected from mice treated with either CAR+ or CAR(I)HPSE+ LTE-T, *p=0.028. (h) Kaplan-Meier analysis of tumor-bearing mice in the kidney infused i.v. with either CAR+ or CAR(I)HPSE+ LTE-T. For these experiments, we generated LTE-T from 2 donors; CAR n=21, CAR(I)HPSE n=21 mice; *p=0.0006.

References

    1. Pule MA, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270. - PMC - PubMed
    1. Kershaw MH, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12:6106–6115. - PMC - PubMed
    1. Bawley VS, et al. T Cells Redirected Against HER2 for Adoptive Immunotherapy for HER2-positive Osteosarcoma. Molecular Therapy. 2012;2010(Supplement 1)(20):S206. Ref Type: Abstract.
    1. Kalos M, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73. - PMC - PubMed
    1. Brentjens RJ, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra38. - PMC - PubMed

Publication types

MeSH terms