Long-Term Clinical Responses of Neoadjuvant Dendritic Cell Infusions and Radiation in Soft Tissue Sarcoma

Shailaja Raj, Marilyn M Bui, Gregory Springett, Anthony Conley, Sergio Lavilla-Alonso, Xiuhua Zhao, Dungsa Chen, Randy Haysek, Ricardo Gonzalez, G Douglas Letson, Steven Eric Finkelstein, Alberto A Chiappori, Dmitry I Gabrilovitch, Scott J Antonia, Shailaja Raj, Marilyn M Bui, Gregory Springett, Anthony Conley, Sergio Lavilla-Alonso, Xiuhua Zhao, Dungsa Chen, Randy Haysek, Ricardo Gonzalez, G Douglas Letson, Steven Eric Finkelstein, Alberto A Chiappori, Dmitry I Gabrilovitch, Scott J Antonia

Abstract

Purpose. Patients with large >5 cm, high-grade resectable soft tissue sarcomas (STS) have the highest risk of distant metastases. Previously we have shown that dendritic cell (DC) based vaccines show consistent immune responses. Methods. This was a Phase I single institution study of neoadjuvant radiation with DC injections on 18 newly diagnosed high-risk STS patients. Neoadjuvant treatment consisted of 50 Gy of external beam radiation (EBRT), given in 25 fractions delivered five days/week, combined with four intratumoral injections of DCs followed by complete resection. The primary endpoint was to establish the immunological response to neoadjuvant therapy and obtain data on its clinical safety and outcomes. Results. There were no unexpected toxicities or serious adverse events. Twelve out of 18 (67%) patients were alive, of which an encouraging 11/18 (61%) were alive with no systemic recurrence over a period of 2-8 years. Favorable immunological responses correlated with clinical responses in some cases. Conclusions. This study provides clinical support to using dendritic cell injections along with radiation in sarcomas, which when used optimally in combination can help clinical outcomes in soft tissue sarcoma. Study registration number is NCT00365872.

Figures

Figure 1
Figure 1
(a) Kaplan Meir curves showing the disease-free survival at 1, 2, 3, and 4 years (94%, 83%, 72%, and 66%, resp.) of patients who received neoadjuvant intratumoral injection of dendritic cells and radiation in high-grade soft tissue sarcomas over 5 cm. (b) Kaplan Meir curve showing overall survival of patients in months on patients who received neoadjuvant intratumoral injection of dendritic cells and radiation in high-grade soft tissue sarcomas over 5 cm. (c) Kaplan Meir curves showing tumor necrosis less than 70% or ≥70% in patients who received intratumoral dendritic cell injections followed by radiation and then surgery with resection of tumors to determine the percentage of necrosis after surgery p = 0.76.
Figure 2
Figure 2
The eighteen patients who received neoadjuvant radiation and DC vaccine were divided into two groups based on their immune response (positive and negative) and then correlated with the overall survival in days and progression free survival.
Figure 3
Figure 3
Pre- and postsurgery CAT scan of a 46-year-old lady with history of dedifferentiated liposarcoma patient who completed the DC vaccine + RT treatment, underwent surgical resection, and approximately 10 months later presented with (a and b) pre- and post-CT scans showing the presence of the lung nodules (two) and then postsurgical imaging study after its resection. (c) Pathological analysis showed tissue necrosis, hyalinization, and infiltration of lymphocytes and is currently free of systemic or local recurrence over 8 years.

References

    1. Spira A. I., Ettinger D. S. The use of chemotherapy in soft-tissue sarcomas. The Oncologist. 2002;7(4):348–359. doi: 10.1634/theoncologist.7-4-348.
    1. Brennan M. F., Casper E. S., Harrison L. B., Shiu M. H., Gaynor J., Hajdu S. I. The role of multimodality therapy in soft-tissue sarcoma. Annals of Surgery. 1991;214(3):328–336. doi: 10.1097/00000658-199109000-00015.
    1. Rosenberg S. A., Tepper J., Glatstein E., et al. The treatment of soft-tissue sarcomas of the extremities: prospective randomized evaluations of (1) limb-sparing surgery plus radiation therapy compared with amputation and (2) the role of adjuvant chemotherapy. Annals of Surgery. 1982;196(3):305–315. doi: 10.1097/00000658-198209000-00009.
    1. Pisters P. W., Harrison L. B., Leung D. H., Woodruff J. M., Casper E. S., Brennan M. F. Long-term results of a prospective randomized trial of adjuvant brachytherapy in soft tissue sarcoma. Journal of Clinical Oncology. 1996;14(3):859–868.
    1. Rooser B., Gustafson P., Rydholm A. Is there no influence of local control on the rate of metastases in high-grade soft tissue sarcoma? Cancer. 1990;65(8):1727–1729. doi: 10.1002/1097-0142(19900415)65:8<1727::AID-CNCR2820650811>;2-X.
    1. Coindre J.-M., Terrier P., Guillou L., et al. Predictive value of grade for metastasis development in the main histologic types of adult soft tissue sarcomas. Cancer. 2001;91(10):1914–1926.
    1. Tierney J. F. Adjuvant chemotherapy for localised resectable soft-tissue sarcoma of adults: meta-analysis of individual data. The Lancet. 1997;350(9092):1647–1654. doi: 10.1016/s0140-6736(97)08165-8.
    1. Frustaci S., Gherlinzoni F., De Paoli A., et al. Adjuvant chemotherapy for adult soft tissue sarcomas of the extremities and girdles: results of the Italian randomized cooperative trial. Journal of Clinical Oncology. 2001;19(5):1238–1247.
    1. Nelson D., Fisher S., Robinson B. The ‘trojan Horse’ approach to tumor immunotherapy: targeting the tumor microenvironment. Journal of Immunology Research. 2014;2014:14. doi: 10.1155/2014/789069.789069
    1. Banchereau J., Steinman R. M. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245–252. doi: 10.1038/32588.
    1. Schreibelt G., Tel J., Sliepen K. H. E. W. J., et al. Toll-like receptor expression and function in human dendritic cell subsets: implications for dendritic cell-based anti-cancer immunotherapy. Cancer Immunology, Immunotherapy. 2010;59(10):1573–1582. doi: 10.1007/s00262-010-0833-1.
    1. Finkelstein S. E., Iclozan C., Bui M. M., et al. Combination of external beam radiotherapy (EBRT) with intratumoral injection of dendritic cells as neo-adjuvant treatment of high-risk soft tissue sarcoma patients. International Journal of Radiation Oncology, Biology, Physics. 2012;82(2):924–932. doi: 10.1016/j.ijrobp.2010.12.068.
    1. Chakraborty M., Abrams S. I., Coleman C. N., Camphausen K., Schlom J., Hodge J. W. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Research. 2004;64(12):4328–4337. doi: 10.1158/0008-5472.CAN-04-0073.
    1. Park B., Yee C., Lee K.-M. The effect of radiation on the immune response to cancers. International Journal of Molecular Sciences. 2014;15(1):927–943. doi: 10.3390/ijms15010927.
    1. Nikitina E., Gabrilovich D. I. Combination of γ-irradiation and dendritic cell administration induces a potent antitumor response in tumor-bearing mice: approach to treatment of advanced stage cancer. International Journal of Cancer. 2001;94(6):825–833. doi: 10.1002/1097-0215(20011215)94:6>825::aid-ijc1545<;2-5.
    1. Teitz-Tennenbaum S., Li Q., Rynkiewicz S., et al. Radiotherapy potentiates the therapeutic efficacy of intratumoral dendritic cell administration. Cancer Research. 2003;63(23):8466–8475.
    1. Rusakiewicz S., Semeraro M., Sarabi M., et al. Immune infiltrates are prognostic factors in localized gastrointestinal stromal tumors. Cancer Research. 2013;73(12):3499–3510. doi: 10.1158/0008-5472.CAN-13-0371.
    1. Brinkrolf P., Landmeier S., Altvater B., et al. A high proportion of bone marrow T cells with regulatory phenotype (CD4+CD25hiFoxP3+) in Ewing sarcoma patients is associated with metastatic disease. International Journal of Cancer. 2009;125(4):879–886. doi: 10.1002/ijc.24461.
    1. Berghuis D., Santos S. J., Baelde H. J., et al. Pro-inflammatory chemokine-chemokine receptor interactions within the Ewing sarcoma microenvironment determine CD8+ T-lymphocyte infiltration and affect tumour progression. The Journal of Pathology. 2011;223(3):347–357. doi: 10.1002/path.2819.
    1. Pisarev V., Yu B., Salup R., Sherman S., Altieri D. C., Gabrilovich D. I. Full-length dominant-negative survivin for cancer immunotherapy. Clinical Cancer Research. 2003;9(17):6523–6533.
    1. Altieri D. C. Validating survivin as a cancer therapeutic target. Nature Reviews Cancer. 2003;3(1):46–54. doi: 10.1038/nrc968.
    1. Geiger J. D., Hutchinson R. J., Hohenkirk L. F., et al. Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Research. 2001;61(23):8513–8519.
    1. Finkelstein S. E., Carey T., Fricke I., et al. Changes in dendritic cell phenotype after a new high-dose weekly schedule of interleukin-2 therapy for kidney cancer and melanoma. Journal of Immunotherapy. 2010;33(8):817–827. doi: 10.1097/cji.0b013e3181ecccad.
    1. Lesterhuis W. J., De Vries I. J. M., Schreibelt G., et al. Route of administration modulates the induction of dendritic cell vaccine-induced antigen-specific T cells in advanced melanoma patients. Clinical Cancer Research. 2011;17(17):5725–5735. doi: 10.1158/1078-0432.ccr-11-1261.

Source: PubMed

3
Abonnieren