Your browser doesn't support javascript.
loading
The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity.
Palakurthi, Sangeetha; Kuraguchi, Mari; Zacharek, Sima J; Zudaire, Enrique; Huang, Wei; Bonal, Dennis M; Liu, Jeffrey; Dhaneshwar, Abha; DePeaux, Kristin; Gowaski, Martha R; Bailey, Dyane; Regan, Samuel N; Ivanova, Elena; Ferrante, Catherine; English, Jessie M; Khosla, Aditya; Beck, Andrew H; Rytlewski, Julie A; Sanders, Catherine; Laquerre, Sylvie; Bittinger, Mark A; Kirschmeier, Paul T; Packman, Kathryn; Janne, Pasi A; Moy, Christopher; Wong, Kwok-Kin; Verona, Raluca I; Lorenzi, Matthew V.
Afiliação
  • Palakurthi S; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Kuraguchi M; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Zacharek SJ; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Zudaire E; Janssen, Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania.
  • Huang W; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Bonal DM; Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Liu J; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Dhaneshwar A; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • DePeaux K; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Gowaski MR; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Bailey D; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Regan SN; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Ivanova E; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Ferrante C; Janssen, Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania.
  • English JM; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Khosla A; PathAI, Boston, Massachusetts.
  • Beck AH; PathAI, Boston, Massachusetts.
  • Rytlewski JA; Adaptive Biotechnologies, Seattle, Washington.
  • Sanders C; Adaptive Biotechnologies, Seattle, Washington.
  • Laquerre S; Janssen, Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania.
  • Bittinger MA; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Kirschmeier PT; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Packman K; Janssen, Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania.
  • Janne PA; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Moy C; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Wong KK; Janssen, Pharmaceutical Companies of Johnson & Johnson, Spring House, Pennsylvania.
  • Verona RI; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Lorenzi MV; Laura & Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York University, New York, New York.
Cancer Immunol Res ; 7(9): 1457-1471, 2019 Sep.
Article em En | MEDLINE | ID: mdl-31331945
ABSTRACT
The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti-PD-1 alone was ineffective, the erdafitinib and anti-PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C-mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti-PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti-PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores de Fatores de Crescimento de Fibroblastos / Receptor de Morte Celular Programada 1 / Antineoplásicos Imunológicos / Imunidade / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores de Fatores de Crescimento de Fibroblastos / Receptor de Morte Celular Programada 1 / Antineoplásicos Imunológicos / Imunidade / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article