Yonsei Med J.  2005 Dec;46(6):835-842.

Combined Treatment of an Intratumoral Injection of Dendritic Cells and Systemic Chemotherapy (Paclitaxel) for Murine Fibrosarcoma

Affiliations
  • 1Department of Dermatology, Inha University College of Medicine, Inchon, Korea.
  • 2Department of Hematology-Oncology, Inha University College of Medicine, Inchon, Korea.
  • 3Clinical Research Center, Inha University College of Medicine, Inchon, Korea.
  • 4Department of Dermatology, Yonsei University College of Medicine, Seoul, Korea.

Abstract

A novel combined treatment of conventional chemotherapy with an intratumoral injection of syngeneic dendritic cells (DCs) has emerged as a potent cancer treatment strategy. In this study, we evaluated the synergistic effect of an intraperitoneal (i.p.) injection of a chemotherapeutic drug, paclitaxel, and an intratumoral (i.t.) injection of syngeneic bone marrow- derived DCs for the treatment of pre-existing fibrosarcoma. Subcutaneous tumors were established using MCA102 fibrosarcoma cells in syngeneic C57BL/6 mice. The results demonstrated that the combined treatment of paclitaxel chemotherapy and the injection of DCs led to complete tumor regression, in contrast to only partial eradication of the tumors with chemotherapy or DCs alone. Furthermore, the tumor-free mice were able to resist a repeat challenge with the same type of tumor. These findings suggest that a combination therapy of systemic chemotherapy along with the intratumoral administration of DCs is a potent treatment strategy for fibrosarcoma.

Keyword

Dendritic cells; paclitaxel; immunotherapy

MeSH Terms

Treatment Outcome
Transplantation, Isogeneic
Phenotype
Paclitaxel/administration & dosage/*therapeutic use
Mice
Injections, Intraperitoneal
Immunologic Memory
Fibrosarcoma/drug therapy/pathology/*therapy
Dendritic Cells/cytology/*transplantation
Combined Modality Therapy
Cells, Cultured
Cell Line, Tumor
Bone Marrow Cells/cytology
Antineoplastic Agents, Phytogenic/administration & dosage/*therapeutic use
Animals

Figure

  • Fig. 1 Phenotyping of cultured dendritic cells. Primary DCs were obtained from syngeneic mouse bone marrow precursors by culturing in the presence of GM-CSF and IL-4; their phenotypes were verified by flow cytometry. The results showed that the DCs isolated were CD11c (B), CD44 (C), and I-Ad (D) positive. Both CD40 (E) and CD80 (F) expression was very low.

  • Fig. 2 in vitro cytotoxic effect of paclitaxel on MCA102 murine fibrosarcoma cells and DCs. The cytotoxicity effects of etoposide, ifosfamide, doxorubicine, and paclitaxel on MCA102 tumor cells (A) and the cytotoxicity effects of paclitaxel on DCs (B) were measured by an MTT assay in vitro. Paclitaxel was found to have the most profound cytotoxic effect on MCA102 tumor cells.

  • Fig. 3 Significant regression of tumor growth in the combined treatment group in contrast to the control groups. Using a well-characterized tumor model (MCA102 sarcoma in C57BL/6 mouse), a synergistic effect of a systemic paclitaxel administration by i.p. together with an i.t. injection of DCs was evaluated in comparison to chemotherapy or DC treatment alone. The data represent the mean ± SD of three independent experiments.

  • Fig. 4 Representative photographs of animals showing the tumor masses 15 days and 42 days after the tumor cell injection. The mouse treated with the combined therapy showed the most significant suppression of tumor growth. (A, B, C, and D) mice treated with medium, paclitaxel, DC, or paclitaxel plus DC, respectively, on day 15. (E, F, G, and H) mice treated with medium, paclitaxel, DC, or paclitaxel plus DC, respectively, on day 42.

  • Fig. 5 Determination of apoptosis of MCA102 tumor cells in the tumor tissues by the TUNEL assay. Apoptotic cells stained brown were easily observed in the tumor tissues treated with the combined treatment (D) or paclitaxel treatment alone (B) but not in tissues treated with DC treatment alone (C) or medium injection (A). Arrows indicate the apoptotic cells.

  • Fig. 6 Immunohistochemical staining of the tumor tissue after the combined treatment. When the border areas between the tumor tissue and normal tissue were observed, more immune cells, such as macrophages and neutrophils, were detected in the mouse specimen from the combined treatment (B) than that from the medium control specimen (A). Secondary antibody alone control (C).

  • Fig. 7 Persistent antitumor memory after the combined treatment. A second challenge of the same MCA102 tumor cells to the mice, in which tumors had completely regressed in the previous experiments, was carried out. A very small-sized tumor appeared around day nine after the second injection, but it disappeared quickly thereafter. Age-matched normal mice were also injected with the same type of tumor for controls. The data represent the means ± SD of triplicate experiments.


Reference

1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998. 392:245–252.
2. Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991. 9:271–296.
3. Celluzzi CM, Mayordomo JI, Storkus WJ, Lotze MT, Falo LD Jr. Peptide-pulsed dendritic cells induce antigen-specific CTL-mediated protective tumor immunity. J Exp Med. 1996. 183:283–287.
4. Carbone FR, Kurts C, Bennett SR, Miller JF, Heath WR. Cross-presentation: a general mechanism for CTL immunity and tolerance. Immunol Today. 1998. 19:368–373.
5. Grabbe S, Beissert S, Schwarz T, Granstein RD. Dendritic cells as initiators of tumor immune responses: a possible strategy for tumor immunotherapy? Immunol Today. 1995. 16:117–121.
6. Gabrilovich DI, Nadaf S, Corak J, Berzofsky JA, Carbone DP. Dendritic cells in antitumor immune responses. II. Dendritic cells grown from bone marrow precursors, but not mature DC from tumor-bearing mice, are effective antigen carriers in the therapy of established tumors. Cell Immunol. 1996. 170:111–119.
7. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998. 4:328–332.
8. Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996. 184:465–472.
9. Condon C, Watkins SC, Celluzzi CM, Thompson K, Falo LD Jr. DNA-based immunization by in vivo transfection of dendritic cells. Nat Med. 1996. 2:1122–1128.
10. Gong J, Avigan D, Chen D, Wu Z, Koido S, Kashiwaba M, Kufe D. Activation of antitumor cytotoxic T lymphocytes by fusions of human dendritic cells and breast carcinoma cells. Proc Natl Acad Sci USA. 2000. 97:2715–2718.
11. Song SY, Kim HS. Strategies to improve dendritic cell-based immunotherapy against cancer. Yonsei Med J. 2004. 45:Suppl. 48–52.
12. Tong Y, Song W, Crystal RG. Combined intratumoral injection of bone marrow-derived dendritic cells and systemic chemotherapy to treat pre-existing murine tumors. Cancer Res. 2001. 61:7530–7535.
13. Inoue N, Yamasaki S, Kondo K, Kan T, Furumoto K, Imamura M. Dendritic cells coinjected with tumor cells treated with an anticancer drug to induce tumor rejection. Surg Today. 2003. 33:269–276.
14. Shin JY, Lee SK, Kang CD, Chung JS, Lee EY, Seo SY, et al. Antitumor effect of intratumoral administration of dendritic cell combination with vincristine chemotherapy in a murine fibrosarcoma model. Histol Histopathol. 2003. 18:435–447.
15. Yu B, Kusmartsev S, Cheng F, Paolini M, Nefedova Y, Sotomayor E, et al. Effective combination of chemotherapy and dendritic cell administration for the treatment of advanced-stage experimental breast cancer. Clin Cancer Res. 2003. 9:285–294.
16. Schiff PB, Fant J, Horwitz SB. Promotion of microtubule assembly in vitro by taxol. Nature. 1979. 277:665–667.
17. Joo HG. Altered maturation of dendritic cells by taxol, an anticancer drug. J Vet Sci. 2003. 4:229–234.
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr