Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell ; 39(9): 1214-1226.e10, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34375612

ABSTRACT

PARP7 is a monoPARP that catalyzes the transfer of single units of ADP-ribose onto substrates to change their function. Here, we identify PARP7 as a negative regulator of nucleic acid sensing in tumor cells. Inhibition of PARP7 restores type I interferon (IFN) signaling responses to nucleic acids in tumor models. Restored signaling can directly inhibit cell proliferation and activate the immune system, both of which contribute to tumor regression. Oral dosing of the PARP7 small-molecule inhibitor, RBN-2397, results in complete tumor regression in a lung cancer xenograft and induces tumor-specific adaptive immune memory in an immunocompetent mouse cancer model, dependent on inducing type I IFN signaling in tumor cells. PARP7 is a therapeutic target whose inhibition induces both cancer cell-autonomous and immune stimulatory effects via enhanced IFN signaling. These data support the targeting of a monoPARP in cancer and introduce a potent and selective PARP7 inhibitor to enter clinical development.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Interferon Type I/metabolism , Neoplasms/drug therapy , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism , Small Molecule Libraries/administration & dosage , Adaptive Immunity/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , HeLa Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
2.
Chembiochem ; 22(12): 2107-2110, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33838082

ABSTRACT

PARP14 is an interferon-stimulated gene that is overexpressed in multiple tumor types, influencing pro-tumor macrophage polarization as well as suppressing the antitumor inflammation response by modulating IFN-γ and IL-4 signaling. PARP14 is a 203 kDa protein that possesses a catalytic domain responsible for the transfer of mono-ADP-ribose to its substrates. PARP14 also contains three macrodomains and a WWE domain which are binding modules for mono-ADP-ribose and poly-ADP-ribose, respectively, in addition to two RNA recognition motifs. Catalytic inhibitors of PARP14 have been shown to reverse IL-4 driven pro-tumor gene expression in macrophages, however it is not clear what roles the non-enzymatic biomolecular recognition motifs play in PARP14-driven immunology and inflammation. To further understand this, we have discovered a heterobifunctional small molecule designed based on a catalytic inhibitor of PARP14 that binds in the enzyme's NAD+ -binding site and recruits cereblon to ubiquitinate it and selectively target it for degradation.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Small Molecule Libraries/pharmacology , Humans , Macrophages/drug effects , Macrophages/metabolism , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
3.
Cell Chem Biol ; 28(8): 1158-1168.e13, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33705687

ABSTRACT

PARP14 has been implicated by genetic knockout studies to promote protumor macrophage polarization and suppress the antitumor inflammatory response due to its role in modulating interleukin-4 (IL-4) and interferon-γ signaling pathways. Here, we describe structure-based design efforts leading to the discovery of a potent and highly selective PARP14 chemical probe. RBN012759 inhibits PARP14 with a biochemical half-maximal inhibitory concentration of 0.003 µM, exhibits >300-fold selectivity over all PARP family members, and its profile enables further study of PARP14 biology and disease association both in vitro and in vivo. Inhibition of PARP14 with RBN012759 reverses IL-4-driven protumor gene expression in macrophages and induces an inflammatory mRNA signature similar to that induced by immune checkpoint inhibitor therapy in primary human tumor explants. These data support an immune suppressive role of PARP14 in tumors and suggest potential utility of PARP14 inhibitors in the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Inflammation/drug therapy , Interleukin-4/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Macrophages/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Poly(ADP-ribose) Polymerases/genetics , RAW 264.7 Cells , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Mol Cancer Ther ; 16(5): 850-860, 2017 05.
Article in English | MEDLINE | ID: mdl-28292935

ABSTRACT

The SWI/SNF complex is a major regulator of gene expression and is increasingly thought to play an important role in human cancer, as evidenced by the high frequency of subunit mutations across virtually all cancer types. We previously reported that in preclinical models, malignant rhabdoid tumors, which are deficient in the SWI/SNF core component INI1 (SMARCB1), are selectively killed by inhibitors of the H3K27 histone methyltransferase EZH2. Given the demonstrated antagonistic activities of the SWI/SNF complex and the EZH2-containing PRC2 complex, we investigated whether additional cancers with SWI/SNF mutations are sensitive to selective EZH2 inhibition. It has been recently reported that ovarian cancers with dual loss of the redundant SWI/SNF components SMARCA4 and SMARCA2 are characteristic of a rare rhabdoid-like subtype known as small-cell carcinoma of the ovary hypercalcemic type (SCCOHT). Here, we provide evidence that a subset of commonly used ovarian carcinoma cell lines were misdiagnosed and instead were derived from a SCCOHT tumor. We also demonstrate that tazemetostat, a potent and selective EZH2 inhibitor currently in phase II clinical trials, induces potent antiproliferative and antitumor effects in SCCOHT cell lines and xenografts deficient in both SMARCA2 and SMARCA4. These results exemplify an additional class of rhabdoid-like tumors that are dependent on EZH2 activity for survival. Mol Cancer Ther; 16(5); 850-60. ©2017 AACR.


Subject(s)
Carcinoma, Small Cell/drug therapy , DNA Helicases/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Rhabdoid Tumor/drug therapy , Transcription Factors/genetics , Animals , Carcinoma, Small Cell/diagnosis , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Diagnosis, Differential , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Humans , Hypercalcemia/diagnosis , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Hypercalcemia/pathology , Mice , Mutation , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL