Elsevier

Cryobiology

Volume 42, Issue 1, February 2001, Pages 49-58
Cryobiology

Regular Article
In Vivo Destruction of Tumor Tissue by Cryoablation Can Induce Inhibition of Secondary Tumor Growth: An Experimental Study

https://doi.org/10.1006/cryo.2001.2302Get rights and content

Abstract

Background. Cryoablation has been used successfully for the local treatment of several cancers. Besides local destruction, a systemic antitumor response has been postulated after cryoablation of tumor tissue. In this study we evaluate the possible systemic antitumor response induced by cryodestruction of tumor tissue in two mouse tumor models. Methods. Mice received two subcutaneously placed tumor implants (thigh and flank) of the nonimmunogenic mouse colon tumor cell line, colon 26-B. After 7 days, the thigh implant was treated by cryoablation or excision and the effect on secondary tumor growth was determined by volume measurement of the nontreated flank tumor. Cytokine (IL-1α and TNF-α) levels in plasma were measured after treatment. Similar experiments were performed in nude mice using a human melanoma cell line (MV3). Moreover, in this model the effect of cryoablation on development of spontaneous lung metastases was evaluated. Results. In the colon 26-B tumor model treatment of primary tumor implants by cryoablation resulted in a significant inhibition of secondary tumor growth compared to animals treated by surgical excision (P < 0.01). Six hours after treatment, plasma levels of IL-1α and TNF-α were higher after cryoablation than after excision (P < 0.01). Also in the nude mice model cryoablation resulted in inhibition of secondary tumor growth, though not significant. Mice treated by cryoablation showed significantly less lung metastases compared to those treated by excision (P = 0.03). Conclusions. Cryoablation of tumor tissue can result in inhibition of secondary and metastatic tumor growth. A cytokine response induced by cryoablation of tumor tissue may attribute to this feature.

References (34)

  • E.A. Carswell et al.

    An endotoxin-induced serum factor that causes necrosis of tumors

    Proc. Natl. Acad. Sci. USA

    (1975)
  • T.H. Corbett et al.

    A mouse colon-tumor model for experimental therapy

    Cancer Chemother. Rep.

    (1975)
  • S.A. El-Shakhs et al.

    Effective hepatic cryoablation: Does it enhance tumor dissimenation?

    World J. Surg.

    (1999)
  • R.P. Faraci et al.

    In vitro demonstration of cryosurgical augmentation of tumor immunity

    Surgery

    (1975)
  • A.A. Gage

    History of cryosurgery

    Semin. Surg. Oncol.

    (1998)
  • D.M. Grace

    Comparison of cryotherapy and surgery for the treatment of spontaneous mouse tumor

    Can. J. Surg.

    (1977)
  • Cited by (81)

    • Immunomodulatory effects of ablation therapy on tumors: Potentials for combination with immunotherapy

      2020, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      However, these DCs have a poor tumor-killing effect in vivo, and only retard the growth of re-challenged tumors. The frequency of cryoablation has been reported to affect tumor growth and local T cell recruitment, as well as immune activation or immunosuppression after cryoablation [59–62]. A previous study [63] analyzed the immune activation effect of cryoablation by regulating the freezing rate in a mouse breast cancer model.

    • Interventional therapy combined with radiotherapy for pancreatic carcinoma

      2020, Integrative Pancreatic Intervention Therapy: A Holistic Approach
    • Percutaneous ablative cryoimmunotherapy for micrometastaic abscopal effect: No complications

      2018, Cryobiology
      Citation Excerpt :

      Cryoablation has a beneficial immunogenic profile compared with other ablative techniques due to the propensity to induce a high necrosis/apoptosis ratio, disrupting the cellular membrane while leaving TAAs intact; which would be degraded to a higher extent by thermal techniques [1]. Furthermore, cryoablation has been demonstrated to induce statistically significant reductions in secondary tumor growth and propensity for pulmonary metastasis in a mouse model, due to its ability to expose TAAs that have previously evaded immunosurveillance [10]. Combination cryoablation with adjuvant pharmacologic induction of the tumor-specific immune response showed enhanced tumor rejection as well as increased intratumoral cytotoxic (CD8) TIL activity [16].

    • Systemic antitumor immune response following reconstruction using frozen autografts for total en bloc spondylectomy

      2014, Spine Journal
      Citation Excerpt :

      The tumor antigens released from the necrotic tumor cells in tumor-bearing grafted bone inside the cage activate the tumor-specific immune response. Previous reports have suggested immune system activation brought about by cryosurgery [4,5,8,9]. Nishida et al. [10] reported that reimplantation of tumor tissue frozen using liquid nitrogen induces antitumor activity against murine osteosarcoma and the serum levels of INF-γ and IL-12 increased after treatment [11].

    View all citing articles on Scopus

    This work was funded by institutional sources.

    View full text