Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity - PubMed (original) (raw)

. 2008 Sep 15;68(18):7684-91.

doi: 10.1158/0008-5472.CAN-08-1167. Epub 2008 Sep 3.

Juan R Cubillos-Ruiz, Yolanda C Nesbeth, Uciane K Scarlett, Diana G Martinez, Ronald J Buckanovich, Fabian Benencia, Radu V Stan, Tibor Keler, Pablo Sarobe, Charles L Sentman, Jose R Conejo-Garcia

Affiliations

Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity

Eduardo Huarte et al. Cancer Res. 2008.

Abstract

Dendritic cells (DC) and cytokines that expand myeloid progenitors are widely used to treat cancer. Here, we show that CD11c(+)DEC205(+) DCs coexpressing alpha-smooth muscle actin and VE-cadherin home to perivascular areas in the ovarian cancer microenvironment and are required for the maintenance of tumor vasculature. Consequently, depletion of DCs in mice bearing established ovarian cancer by targeting different specific markers significantly delays tumor growth and enhances the effect of standard chemotherapies. Tumor growth restriction was associated with vascular apoptosis after DC ablation followed by necrosis, which triggered an antitumor immunogenic boost. Our findings provide a mechanistic rationale for selectively eliminating tumor-associated leukocytes to promote antitumor immunity while impeding tumor vascularization and to develop more effective DC vaccines based on a better understanding of the tumor microenvironment.

PubMed Disclaimer

Figures

Fig. 1

Fig. 1. DEC205+CD11b− DCs, but not monocyte/macrophages represent the most abundant leukocyte subset in the microenvironment of solid ovarian carcinoma specimens

(Gated on CD45+ cells) (A) FACS analysis of a mechanically dissociated metastatic stage III ovarian carcinoma specimen. These data are representative of 3 primary and 5 metastatic stage III–IV samples (see also Suppl. Fig.1). (B) FACS analysis of the same tumor and its matching tumor ascites.

Fig. 2

Fig. 2. Elimination of CD11c+ DCs abrogates tumor growth

(A) ITGAX-DTR-GFP mice (n=6/group; 2 independent experiments) were challenged with a subcutaneous injection of 107 ID8-Defb29/Vegf-a cells in 200 µl Matrigel and immediately received 4 ng/g body weight DT in PBS (top) or PBS (bottom). Tumors were removed at 2 months. (B) Administration of DT did not result in reduced tumor growth in wild-type mice. (C) ITGAX-DTR-GFP mice were subcutaneously inoculated with 107 ID8-Defb29/Vegf-a cells (in 200 µl Matrigel). After 10 days, mice received 2 ng/g DT in PBS (ITGAX + DT) or a similar volume of PBS (ITGAX+PBS). (D) DT (Depleted), but not PBS (Control) administration eliminates tumor CD11c+ DCs (GFP+) within 48 h. Magnification is included. Data are representative of 3 independent experiments.

Fig. 3

Fig. 3. Elimination of CD11c+ DCs results in tumor necrosis

(A) Tumor necrosis is obvious 36h after the administration of DT and becomes massive at 48h. N, Necrosis (×40). (B) ITGAX-DTR-GFP mice (n=4) bearing established flank ID8-Defb29/Vegf-a tumors received 4 ng/g body weight DT in PBS (24h, right), or a similar volume of PBS (control, left). Tumors were removed after 24h and stained with PE-labeled anti-CD31 and biotinylated anti-active caspase 3 antibodies (both from BD Pharmingen), plus Alexa-Fluor-350 (×200). (C) In reconstituted mice, depletion of CD11c+ DCs every 4 days for 2 months (DT) decreased tumor burden, compared to animals injected with PBS. Data are representative of two independent experiments. (D) Constant depletion of CD11c+ DCs (DT) increases in the survival of reconstituted ovarian ID8 tumor-bearing mice, compared to reconstituted mice injected with PBS (Control). Pooled data from two independent experiments (n=12/group, total) are displayed.

Fig. 4

Fig. 4. Elimination of DCs through CD11c or DEC205 targeting enhances survival

(A) (left) Depletion of CD11c+ cells with 4 weekly intravenous administrations of anti-CD11c immunotoxin in established flank ovarian tumor-bearing animals (n=5/group; 100 µg/Kg after day 7 or 21) decreases tumor growth, compared to mice receiving a truncated toxin. (right) Formation of necrotic cavities (N) encapsulated by a viable ring (highlighted) in treated mice (IT), compared to mice receiving the harmless truncated IT (Control; ×400). (B) Survival curves of mice bearing intraperitoneal tumors and treated with either: 0.33 mg/Kg/day of truncated toxin (Truncated) or IT (IT) for two weeks, starting at day 3; 7 mg/Kg of topotecan at day 7 (Topotecan); 7 mg/Kg topotecan at day 7 plus 0.33 mg/Kg/day IT for two weeks (IT+Top); and the same regime of topotecan (day 7) plus IT (daily from day 3) administered until mice developed ascites (IT+Topb). (C) Intraperitoneal ovarian carcinoma-bearing mice (n=6/group), depleted of CD11b+ leukocytes by 4 weekly intraperitoneal administrations of anti-CD11b-saporin immunotoxin (90 µg/Kg) survived longer than mice receiving the same dose of unconjugated saporin (both from Advanced Targeting Systems, San Diego, CA). (D) Intraperitoneal ovarian carcinoma-bearing mice (n=6/group), depleted of DEC205+ DCs by intraperitoneal administrations of anti-DEC205-saporin immunotoxin (DEC205SAP; Celldex, 0.25 mg/Kg) survived longer than animals receiving the same amount of unconjugated antibody (DEC205), which in turn exhibited an increase in lifespan compared to animals injected with PBS.

Fig. 5

Fig. 5. Elimination of DCs boosts anti-tumor immune responses

(A) FACS analysis of CD44 and CD69 in tumor ascites from tumor-bearing mice injected with the antiDEC205 IT (Depleted) or PBS (Control). (B) FACS analysis of peritoneal wash samples from tumor-bearing mice injected with the antiCD11c IT (Depleted) or PBS (Control) one month after tumor challenge. (C) OT-I mice (n=4) were intraperitoneally injected with 106 OVA-expressing ID8 cells mixed with 107 ID8-Defb29/Vegf-a tumor ascites cells (containing 30% CD45+CD11+VE-Cadherin+ DCs) and received 0.5 mg/Kg of IT (IT) or truncated toxin (Ctrl) one day later. After seven days, mice received again 107 ID8-Defb29/Vegf-a tumor ascites cells i.p., followed by a new DC depletion or control treatment at day 8. Splenocytes were collected at day 11 and stimulated for 7 days with OVA (Sigma; 1 µg/ml). Interferon-γ ELISPOT was performed against bone marrow-derived DCs pulsed with OVA (1 µg/ml; 10:1, splenocyte:DC ratio). (D) Depletion of CD11c+ DCs in OT-I transgenic mice also enhanced the amount of interferon-γ-producing splenocytes responding to DCs pulsed for 4 h. with gamma and UV irradiated OVA-ID8 tumor cells (10:1, DC:ID8 ratio).

Fig. 6

Fig. 6. DCs support tumor vascularization

Tumor-bearing ITGAX-DTR-GFP mice were perfused with an intracardiac injection of biotinylated Tomato Lectin at different times after tumor inoculation (A) (left) Unperfused (white arrows) and blood transporting (yellow arrows) structures assembled by CD11c+ (GFP+) cells mixed in the growing edge of all specimens analyzed (×200). (right) In central areas of the tumor, CD11c+ (GFP+) leukocytes are irregularly scattered on the wall of most neovessels (×400). (B) Confocal microscopy analysis of the different kinds of structures assembled by CD11c+ (GFP+) DCs (×600). (left) Another example of a different unperfused GFP+ structure. (center) Co-localization of Tomato Lectin and GFP+ cells in selected blood vessels. (right; projection of stack images) CD11c+ cells in big vascular arrangements were predominantly found in an abluminal second layer. (C) FACS analysis of SMA and VE-Cadherin expression on CD11c+ DCs from tumor ascites. (D) CD45+DEC205+ DCs (hDEC205+) or CD3+CD4+ lymphocytes (hCD3+CD4+) were FACS sorted from eight unselected human ovarian carcinoma suspensions. CD45+CD11c+ DCs (mCD11c+) were sorted from the ascites of ID8-Defb29/Vegf-a tumor-bearing mice. Cells (106/µl) were stimulated for 4 h. with PMA/ionomycin (50ng/l µg/ml). Cytokines were determined by Bioplex analysis.

Similar articles

Cited by

References

    1. Ramakrishna V, Ross MM, Petersson M, et al. Naturally occurring peptides associated with HLA-A2 in ovarian cancer cell lines identified by mass spectrometry are targets of HLA-A2-restricted cytotoxic T cells. Int Immunol. 2003;15:751–763. - PubMed
    1. Yen MJ, Hsu CY, Mao TL, et al. Diffuse mesothelin expression correlates with prolonged patient survival in ovarian serous carcinoma. Clin Cancer Res. 2006;12:827–831. - PubMed
    1. Odunsi K, Jungbluth AA, Stockert E, et al. NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res. 2003;63:6076–6083. - PubMed
    1. Hasegawa K, Koizumi F, Noguchi Y, et al. SSX expression in gynecological cancers and antibody response in patients. Cancer Immun. 2004;4:16. - PubMed
    1. Kershaw MH, Westwood JA, Parker LL, 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

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources