Your browser doesn't support javascript.
loading
Pharmacological prevention of surgery-accelerated metastasis in an animal model of osteosarcoma.
Kallis, Michelle P; Maloney, Caroline; Blank, Brandon; Soffer, Samuel Z; Symons, Marc; Steinberg, Bettie M.
Afiliação
  • Kallis MP; The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA.
  • Maloney C; The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
  • Blank B; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA.
  • Soffer SZ; The Elmezzi Graduate School of Molecular Medicine, Northwell Health, Manhasset, NY, USA.
  • Symons M; The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
  • Steinberg BM; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA.
J Transl Med ; 18(1): 183, 2020 04 30.
Article em En | MEDLINE | ID: mdl-32354335
ABSTRACT

BACKGROUND:

Osteosarcoma is a highly metastatic primary bone tumor that predominantly affects adolescents and young adults. A mainstay of treatment in osteosarcoma is removal of the primary tumor. However, surgical excision itself has been implicated in promoting tumor growth and metastasis, an effect known as surgery-accelerated metastasis. The underlying mechanisms contributing to surgery-accelerated metastasis remain poorly understood, but pro-tumorigenic alterations in macrophage function have been implicated.

METHODS:

The K7M2-BALB/c syngeneic murine model of osteosarcoma was used to study the effect of surgery on metastasis, macrophage phenotype, and overall survival. Pharmacological prevention of surgery-accelerated metastasis was examined utilizing gefitinib, a receptor interacting protein kinase 2 inhibitor previously shown to promote anti-tumor macrophage phenotype.

RESULTS:

Surgical excision of the primary tumor resulted in increases in lung metastatic surface nodules, overall metastatic burden and number of micrometastatic foci. This post-surgical metastatic enhancement was associated with a shift in macrophage phenotype within the lung to a more pro-tumor state. Treatment with gefitinib prevented tumor-supportive alterations in macrophage phenotype and resulted in reduced metastasis. Removal of the primary tumor coupled with gefitinib treatment resulted in enhanced median and overall survival.

CONCLUSIONS:

Surgery-accelerated metastasis is mediated in part through tumor supportive alterations in macrophage phenotype. Targeted pharmacologic therapies that prevent pro-tumor changes in macrophage phenotype could be utilized perioperatively to mitigate surgery-accelerated metastasis and improve the therapeutic benefits of surgery.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Ósseas / Osteossarcoma / Neoplasias Pulmonares Limite: Animals Idioma: En Revista: J Transl Med Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Ósseas / Osteossarcoma / Neoplasias Pulmonares Limite: Animals Idioma: En Revista: J Transl Med Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Estados Unidos