Your browser doesn't support javascript.
loading
SPOP Mutations Target STING1 Signaling in Prostate Cancer and Create Therapeutic Vulnerabilities to PARP Inhibitor-Induced Growth Suppression.
Geng, Chuandong; Zhang, Man-Chao; Manyam, Ganiraju C; Vykoukal, Jody V; Fahrmann, Johannes F; Peng, Shan; Wu, Cheng; Park, Sanghee; Kondraganti, Shakuntala; Wang, Daoqi; Robinson, Brian D; Loda, Massimo; Barbieri, Christopher E; Yap, Timothy A; Corn, Paul G; Hanash, Samir; Broom, Bradley M; Pilié, Patrick G; Thompson, Timothy C.
Afiliación
  • Geng C; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Zhang MC; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Manyam GC; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Vykoukal JV; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Fahrmann JF; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Peng S; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Wu C; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Park S; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Kondraganti S; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Wang D; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Robinson BD; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York.
  • Loda M; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York.
  • Barbieri CE; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York.
  • Yap TA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York.
  • Corn PG; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York.
  • Hanash S; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York.
  • Broom BM; Department of Urology, Weill Cornell Medicine, New York, New York.
  • Pilié PG; Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Thompson TC; Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
Clin Cancer Res ; 29(21): 4464-4478, 2023 11 01.
Article en En | MEDLINE | ID: mdl-37581614
ABSTRACT

PURPOSE:

Speckle-type POZ protein (SPOP) is important in DNA damage response (DDR) and maintenance of genomic stability. Somatic heterozygous missense mutations in the SPOP substrate-binding cleft are found in up to 15% of prostate cancers. While mutations in SPOP predict for benefit from androgen receptor signaling inhibition (ARSi) therapy, outcomes for patients with SPOP-mutant (SPOPmut) prostate cancer are heterogeneous and targeted treatments for SPOPmut castrate-resistant prostate cancer (CRPC) are lacking. EXPERIMENTAL

DESIGN:

Using in silico genomic and transcriptomic tumor data, proteomics analysis, and genetically modified cell line models, we demonstrate mechanistic links between SPOP mutations, STING signaling alterations, and PARP inhibitor vulnerabilities.

RESULTS:

We demonstrate that SPOP mutations are associated with upregulation of a 29-gene noncanonical (NC) STING (NC-STING) signature in a subset of SPOPmut, treatment-refractory CRPC patients. We show in preclinical CRPC models that SPOP targets and destabilizes STING1 protein, and prostate cancer-associated SPOP mutations result in upregulated NC-STING-NF-κB signaling and macrophage- and tumor microenvironment (TME)-facilitated reprogramming, leading to tumor cell growth. Importantly, we provide in vitro and in vivo mechanism-based evidence that PARP inhibitor (PARPi) treatment results in a shift from immunosuppressive NC-STING-NF-κB signaling to antitumor, canonical cGAS-STING-IFNß signaling in SPOPmut CRPC and results in enhanced tumor growth inhibition.

CONCLUSIONS:

We provide evidence that SPOP is critical in regulating immunosuppressive versus antitumor activity downstream of DNA damage-induced STING1 activation in prostate cancer. PARPi treatment of SPOPmut CRPC alters this NC-STING signaling toward canonical, antitumor cGAS-STING-IFNß signaling, highlighting a novel biomarker-informed treatment strategy for prostate cancer.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Neoplasias de la Próstata / Neoplasias de la Próstata Resistentes a la Castración Tipo de estudio: Prognostic_studies Límite: Humans / Male Idioma: En Año: 2023 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Neoplasias de la Próstata / Neoplasias de la Próstata Resistentes a la Castración Tipo de estudio: Prognostic_studies Límite: Humans / Male Idioma: En Año: 2023 Tipo del documento: Article