2019
DOI: 10.1080/2162402x.2019.1638207
|View full text |Cite
|
Sign up to set email alerts
|

Enhancing direct cytotoxicity and response to immune checkpoint blockade following ionizing radiation with Wee1 kinase inhibition

Abstract: Tumor cells activate the G2/M cell cycle checkpoint in response to ionizing radiation (IR) and effector immune cell-derived granzyme B to facilitate repair and survival. Wee1 kinase inhibition reverses the ability of tumor cells to pause at G2/M. Here, we hypothesized that AZD1775, a small molecule inhibitor of Wee1 kinase, could sensitize tumor cells to IR and T-lymphocyte killing and improve responses to combination IR and programmed death (PD)-axis immune checkpoint blockade (ICB). Multiple models of head a… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
23
0

Year Published

2020
2020
2024
2024

Publication Types

Select...
7
1

Relationship

0
8

Authors

Journals

citations
Cited by 39 publications
(24 citation statements)
references
References 43 publications
0
23
0
Order By: Relevance
“…In preclinical studies, WEE1 kinase inhibitor, AZD1775, prevented tumour cells halting at G2/M phase of the cell cycle after ionising radiation treatment thus inducing cell death. Combination of AZD1775 and ionising radiation also increased cytotoxic T lymphocyte‐mediated killing of tumour cells 171 . Ionising radiation increased tumour PD‐L1 expression and the combination of ionising radiation or AZD1775 with anti‐PD‐1 antibody induced tumour rejection in 27–33% of the treated mice, whereas the triple combination of ionising radiation, AZD1775 and anti‐PD‐1 antibody significantly delayed tumour growth and induced tumour rejection in 73% of mice, thus prolonging survival.…”
Section: Combination Of Immune Checkpoint Inhibitor Therapiesmentioning
confidence: 97%
See 2 more Smart Citations
“…In preclinical studies, WEE1 kinase inhibitor, AZD1775, prevented tumour cells halting at G2/M phase of the cell cycle after ionising radiation treatment thus inducing cell death. Combination of AZD1775 and ionising radiation also increased cytotoxic T lymphocyte‐mediated killing of tumour cells 171 . Ionising radiation increased tumour PD‐L1 expression and the combination of ionising radiation or AZD1775 with anti‐PD‐1 antibody induced tumour rejection in 27–33% of the treated mice, whereas the triple combination of ionising radiation, AZD1775 and anti‐PD‐1 antibody significantly delayed tumour growth and induced tumour rejection in 73% of mice, thus prolonging survival.…”
Section: Combination Of Immune Checkpoint Inhibitor Therapiesmentioning
confidence: 97%
“…Ionising radiation increased tumour PD‐L1 expression and the combination of ionising radiation or AZD1775 with anti‐PD‐1 antibody induced tumour rejection in 27–33% of the treated mice, whereas the triple combination of ionising radiation, AZD1775 and anti‐PD‐1 antibody significantly delayed tumour growth and induced tumour rejection in 73% of mice, thus prolonging survival. This therapeutic effect is dependent on CD8 + T cells 171 . Cured mice were subsequently rechallenged with tumour cells and rejection of tumour cells was observed, suggesting that the triple combination also conferred immunological memory 171 .…”
Section: Combination Of Immune Checkpoint Inhibitor Therapiesmentioning
confidence: 99%
See 1 more Smart Citation
“…AZD6738 increased cell proliferation, infiltration, and interferon-gamma (IFN-γ) production of tumor-infiltrating lymphocyte CD8 + T cells, resulting in decreased T cells and tumor-infiltrating lymphocyte Tregs in mice xenografts. Moreover, Patel et al [ 151 ] indicated that adavosertib and ionizing radiation combination therapy enhanced the sensitivity to T-lymphocyte, tumor-specific cytotoxicity, and programmed death-axis immune checkpoint blockade response in various cancers such as melanoma, lung carcinoma, and head and neck carcinoma in vitro and in vivo. The addition of adavosertib after ionizing radiation reversed the G2/M cell cycle checkpoint activation and led to cell death.…”
Section: Preclinic and Clinical Development Of Synthetic Lethalitymentioning
confidence: 99%
“…However, other mechanisms, such as DDR aberrations, nucleotide starvation, replicative stress, and, as more recently found, loss of the ATRX chromatin remodeler gene [ 36 ] and low phosphatase and tensin homolog (PTEN) expression [ 37 ], contribute to sensitize cancer cells to WEE1 inhibition, which, thus, proved monotherapy activity even in TP53 -wild-type cancer cells [ 29 , 30 , 38 ]. Moreover, WEE1 inhibition showed efficacy also in combination with inhibitors of other DDR factors, such as PARP [ 39 , 40 , 41 , 42 ], CHK1 [ 29 , 43 , 44 , 45 , 46 , 47 ], and ataxia telangiectasia and Rad3 related (ATR) kinase [ 48 , 49 , 50 ], and also when combined with different anticancer targeted agents [ 29 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 ] and immunotherapeutic approaches [ 29 , 62 , 63 , 64 ].…”
Section: Introductionmentioning
confidence: 99%