CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients - PubMed (original) (raw)

. 2011 May;121(5):1822-6.

doi: 10.1172/JCI46110. Epub 2011 Apr 11.

Carlos Almeida Ramos, Enli Liu, Martha P Mims, Michael J Keating, George Carrum, Rammurti T Kamble, Catherine M Bollard, Adrian P Gee, Zhuyong Mei, Hao Liu, Bambi Grilley, Cliona M Rooney, Helen E Heslop, Malcolm K Brenner, Gianpietro Dotti

Affiliations

CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients

Barbara Savoldo et al. J Clin Invest. 2011 May.

Abstract

Targeted T cell immunotherapies using engineered T lymphocytes expressing tumor-directed chimeric antigen receptors (CARs) are designed to benefit patients with cancer. Although incorporation of costimulatory endodomains within these CARs increases the proliferation of CAR-redirected T lymphocytes, it has proven difficult to draw definitive conclusions about the specific effects of costimulatory endodomains on the expansion, persistence, and antitumor effectiveness of CAR-redirected T cells in human subjects, owing to the lack of side-by-side comparisons with T cells bearing only a single signaling domain. We therefore designed a study that allowed us to directly measure the consequences of adding a costimulatory endodomain to CAR-redirected T cells. Patients with B cell lymphomas were simultaneously infused with 2 autologous T cell products expressing CARs with the same specificity for the CD19 antigen, present on most B cell malignancies. One CAR encoded both the costimulatory CD28 and the ζ-endodomains, while the other encoded only the ζ-endodomain. CAR+ T cells containing the CD28 endodomain showed strikingly enhanced expansion and persistence compared with CAR+ T cells lacking this endodomain. These results demonstrate the superiority of CARs with dual signal domains and confirm a method of comparing CAR-modified T cells within individual patients, thereby avoiding patient-to-patient variability and accelerating the development of optimal T cell immunotherapies.

PubMed Disclaimer

Figures

Figure 1

Figure 1. Transduction efficiency and phenotypic/function profile of T cell lines.

(A) FACS and Q-PCR analyses showing transduction efficiency with CAR.CD19ζ and CAR.CD19-28ζ vectors (left). Bars indicate mean values for peripheral blood samples from 6 patients (Supplemental Table 1). Each symbol represents an individual cell line. Representative histograms of T cells transduced with CAR.CD19ζ and CAR.CD19-28ζ vectors from patients number 1 and number 5 are also presented (right). Numbers represent the percentage of CAR+ cells. (B) Results of a 4-hour 51Cr-release assay at an effector/tumor cell (E/T) ratio of 20:1. Target cells were Raji (CD19+, CD80+, CD86–), a Burkitt lymphoma cell line; HLDM-2 (CD19–, CD80+, CD86+), a Hodgkin lymphoma cell line; and K562 (CD19–, CD80–, CD86–), an erythroid leukemia cell line that is susceptible to natural killer cell activity. Both CAR.CD19ζ+ and CAR.CD19-28ζ+ T cells specifically targeted CD19+ tumors. Data are mean ± SD for the 6 T cell lines. (C) Phenotypic composition of CAR.CD19ζ+ or CAR.CD19-28ζ+ T cells. These products contained both CD8+ and CD4+ CAR-expressing T cells that are predominantly CD45RO+CD62L+, with a fraction of them expressing CD28. Each symbol represents an individual cell line, and horizontal bars denote mean group values.

Figure 2

Figure 2. In vivo expansion and persistence of infused CAR.CD19ζ+ versus CAR.CD19-28ζ+ T cell lines as assessed by Q-PCR in peripheral blood.

Data points represent critical postinfusion intervals after the first or second infusion of modified T cells. Patients number 1, number 3, and number 5, who had stable disease or clinical benefit at 6 weeks after the first T cell infusion, received a second infusion of CAR-modified T lymphocytes. Patient number 1 received only CAR.CD19-28ζ+ T cells (2 × 107 cells/m2, the same as for the first infusion), because this was the only product available. Patient number 3 received both CAR.CD19-28ζ+ and CAR.CD19ζ+ T cells, but the cell dose was 60% of their first dose (1 × 108 cells/m2). Patient number 5 received both CAR.CD19-28ζ+ and CAR.CD19ζ+ T cells at the same dose administered during their first infusion (2 × 108 cells/m2). Open arrows indicate the time of T cell infusion, and dashed arrows indicate the time when chemotherapy was initiated for disease progression. Pre, before the first infusion; Pre II, before the second infusion.

Figure 3

Figure 3. Detection of CAR+ T cells in a skin tumor biopsy.

(A) Immunohistochemical examination (diaminobenzidine with hematoxylin counterstaining) of a punch biopsy of a lymphoma skin lesion from patient number 5 at 2 weeks after T cell infusion showed that tumor cells were CD20+ (shown), CD10+, BCL2+, and BCL6+, consistent with involvement by follicular lymphoma with large cell transformation. Scattered CD3+ CD8+ cells infiltrated the tumor. Of note, the infused CAR.CD19-28ζ+ product consisted of 85% CD8+ cells. Scale bar: 100 μm. (B) FACS analysis of a cell suspension obtained from a fragment of the tumor biopsy. Viable cells represented approximately 45% of the preparation. The far left panel shows the gate on CD45+ cells, which represented 12% of the viable cells. The middle panel shows the CD3+ lymphocytes infiltrating the tumor, which accounted for 6.7% of CD45+ cells (0.8% of all viable cells). The far right panel illustrates that 20% of the gated CD3+ lymphocytes cells coexpressed the CAR, as assessed by the Fc-Cy5 monoclonal antibody, which binds to the IgG1-CH2CH3 spacer region of the CD19-specific CARs (~0.16% of all viable cells). SSC, side scatter.

Comment in

Similar articles

Cited by

References

    1. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8(4):299–308. doi: 10.1038/nrc2355. - DOI - PMC - PubMed
    1. Bollard CM, et al. Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer. Blood. 2007;110(8):2838–2845. doi: 10.1182/blood-2007-05-091280. - DOI - PMC - PubMed
    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(11):1264–1270. doi: 10.1038/nm.1882. - DOI - PMC - PubMed
    1. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90(2):720–724. doi: 10.1073/pnas.90.2.720. - DOI - PMC - PubMed
    1. Sadelain M, Brentjens R, Riviere I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009;21(2):215–223. doi: 10.1016/j.coi.2009.02.009. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources