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
. 2010 Jul 15;16(14):3594-606.
doi: 10.1158/1078-0432.CCR-10-0192. Epub 2010 Jul 13.

Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen

Affiliations

Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen

Alena A Chekmasova et al. Clin Cancer Res. .

Abstract

Purpose: Most patients diagnosed with ovarian cancer will ultimately die from their disease. For this reason, novel approaches to the treatment of this malignancy are needed. Adoptive transfer of a patient's own T cells, genetically modified ex vivo through the introduction of a gene encoding a chimeric antigen receptor (CAR) targeted to a tumor-associated antigen, is a novel approach to the treatment of ovarian cancer.

Experimental design: We have generated several CARs targeted to the retained extracellular domain of MUC16, termed MUC-CD, an antigen expressed on most ovarian carcinomas. We investigate the in vitro biology of human T cells retrovirally transduced to express these CARs by coculture assays on artificial antigen-presenting cells as well as by cytotoxicity and cytokine release assays using the human MUC-CD(+) ovarian tumor cell lines and primary patient tumor cells. Further, we assess the in vivo antitumor efficacy of MUC-CD-targeted T cells in SCID-Beige mice bearing peritoneal human MUC-CD(+) tumor cell lines.

Results: CAR-modified, MUC-CD-targeted T cells exhibited efficient MUC-CD-specific cytolytic activity against both human ovarian cell and primary ovarian carcinoma cells in vitro. Furthermore, expanded MUC-CD-targeted T cells infused through either i.p. injection or i.v. infusion into SCID-Beige mice bearing orthotopic human MUC-CD(+) ovarian carcinoma tumors either delayed progression or fully eradicated disease.

Conclusion: These promising preclinical studies justify further investigation of MUC-CD-targeted T cells as a potential therapeutic approach for patients with high-risk MUC16(+) ovarian carcinomas.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Design and in vitro analysis of MUC-CD targeted CARs. (A) Schematic diagram of the first generation 4H11z and second generation 4H11-28z retroviral vectors. 4H11scFv: MUC16 specific scFv derived from the heavy (VH) and light (VL) chain variable regions of the monoclonal antibody 4H11; CD8: CD8 hinge and transmembrane domains; CD28: CD28 transmembrane and cytoplasmic signaling domains; ζ chain: T cell receptor ζ chain cytoplasmic signaling domain; LTR: long terminal repeat; black box: CD8 leader sequence; grey box: (Gly4Ser)3 linker; arrows indicate start of transcription. (B) FACS analysis of human T cells retrovirally transduced to express either the 4H11z or 19z1 CAR. (C) 4H11z+ but not 19z1+ T cells expand on 3T3(MUC-CD/B7.1) AAPC. CAR+ were co-cultured on 3T3(MUC-CD/B7.1) AAPC monolayers at 3 × 106 CAR+ T cells/well of a 6 well plate. Proliferation of CAR+ T cells was assessed by FACS in combination with viable T cell counts as assessed by trypan blue exclusion assays.
Figure 2
Figure 2
In vitro comparison of T cells modified to express the first generation 4H11z CAR to T cells modified to express the second generation co-stimulatory 4H11-28z CAR. (A) CAR+ T cells were co-cultured on MUC-CD monolayers with (right panel) or without B7.1 (left panel). 1 × 106 CAR+ T cells per well were co-cultured on AAPC monolayers in 12 well tissue culture plates in cytokine-free medium. Total viable T cell counts were assessed on days 2, 4 and 7, by trypan blue exclusion assays. (B) Antigen specific cytokine’s secretion by MUC-CD+ targeted T cells. (C) Expansion of CAR+ T cells following 3 cycles of stimulation on 3T3(MUC-CD/B7.1). Arrows indicate 1st and 2nd cycles of restimulation on AAPCs. (D) FACS analysis of the CAR+ T cell fraction of 4H11-28z+ T cells increased following each weekly cycle of stimulation. (I) FACS following initial transduction, (II) FACS at 7 days following first stimulation on AAPCs, (III) FACS at 7 days following second stimulation on AAPCs.
Figure 3
Figure 3
Upon co-culture, 4H11-28z+ T cells specifically expand and lyse MUC-CD+ tumor cells. (A) Cytotoxicity assays of 4H11-28z+ T cells targeting OV-CAR3(MUC-CD) and SK-OV3(MUC-CD) tumor cells demonstrate efficient cytotoxicity mediated by 4H11-28z+ T cells T cells from healthy donors compared to control 19-28z+ T cells. (B) Antigen specific proliferation of MUC-CD targeted T cells after co-culture with OV-CAR3(MUC-CD) or SK-OV3(MUC-CD) tumor cells. CAR+ T cells were co-cultured with wild type or MUC-CD expressing tumor cells at 1:1 ratio for 7 days. Absolute T cell numbers assessed on days 2, 4 and 7 following co-culture with ovarian tumor cells. The T cell seeding density for T cell proliferation assay was 1 × 106/mL. (C) Antigen-specific IL-2 and IFN-γ secretion by 4H11-28z transduced T cells after 2 days of co-culture on ovarian cancer cells. (D) Healthy donor T cells or autologous T cells isolated from ascites, modified to express the 4H11-28z CAR lyse primary patient ascites-derived MUC-CD+ tumor cells when compared to T cells modified to express the control 19-28z CAR.
Figure 4
Figure 4
Eradication of OV-CAR3(MUC-CD) tumors after i.p. treatment with first and second generation of MUC-CD targeted T cells. (A) Intraperitoneal injection of OV-CAR3(MUC-CD) tumors in untreated SCID-Beige mice results in abdominal distension and nodular peritoneal tumors. At 5 weeks post intraperitoneal injection of OV-CAR3(MUC-CD) tumor cells mice developed ascites as evidenced by a distended abdomen (center panel) when compared to a tumor free mouse (left panel). Post mortem visualization of the peritoneum demonstrates nodular tumor masses (arrows) within the abdominal cavity (right panel). (B) Intraperitoneal injection of 4H11z+ and 4H11-28z+ T cells either delay tumor progression or fully eradicate disease. Kaplan-Meier survival curve of SCID-Beige mice treated with first or second generation of MUC-CD targeted T cells.
Figure 5
Figure 5
MUC-CD targeted 4H11-28z+ T cells traffic to peritoneal OV-CAR3(MUC-CD/GFP-FFLuc) tumors following systemic intravenous infusion resulting in efficient anti-tumor efficacy. (A) BLI of tumor progression of representative i.p. and i.v. 4H11-28z+ T cell treated mice with ultimately progressive disease following treatment compared to BLI of tumor progression in a representative control 19-28z+ T cell treated mouse. (B) Kaplan-Meier survival curve of SCID-Beige mice treated i.p. or i.v. with 4H11-28z+ T cells. Tumor eradication is enhanced after either ip or iv infusion of 4H11-28z+ T cells when compared to control treated mice. Both ip and iv 4H11-28z+ T cell treated mice exhibited statistically enhanced survival when compared to 19-28z+ T cell treated control cohorts while survival between the i.p. and i.v. treated 4H11-28z+ T cell cohorts was not statistically significant (p=0.22). (C) Systemically injected CFSE stained 4H11-28z+ T cells traffic to advanced i.p. OV-CAR(MUC-CD) tumors. Presence of i.v. injected CFSE labeled 19-28z+ control T cells (left panel) and 4H11-28z+ T cells (right panel) 1 day following infusion into SCID-Beige mice with OV-CAR(MUC-CD) tumors injected 7 days earlier as assessed by FACS analysis of single cell OV-CAR3(MUC-CD) tumor suspensions.
Figure 6
Figure 6
Eradication of BLI evident OV-CAR3(MUC-CD) tumors in SCID-Beige mice by ip infusion of 4H11-28z+ T cells. (A) BLI of 4H11-28z+ T cell treated mice with either relapsed disease (middle row) or eradicated disease (bottom row) compared to a representative 19-28z+ T cell treated control mouse. (B) Kaplan-Meier survival curve of SCID-Beige mice with advanced OV-CAR3(MUC-CD/GFP-FFLuc) tumors treated i.p. with 4H11-28z+ T cells.

References

    1. Singh AP, Senapati S, Ponnusamy MP, et al. Clinical potential of mucins in diagnosis, prognosis, and therapy of ovarian cancer. Lancet Oncol. 2008;9(11):1076–85. - PMC - PubMed
    1. Sun CC, Ramirez PT, Bodurka DC. Quality of life for patients with epithelial ovarian cancer. Nat Clin Pract Oncol. 2007;4(1):18–29. - PubMed
    1. Brentjens RJ, Latouche JB, Santos E, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9(3):279–86. - PubMed
    1. Hwu P, Yang JC, Cowherd R, et al. In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 1995;55(15):3369–73. - PubMed
    1. Imai C, Mihara K, Andreansky M, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18(4):676–84. - PubMed

Publication types

MeSH terms