Chaoyang Sun's research while affiliated with Tongji Hospital and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (17)


Information of human ovaries and quality control of scRNA-seq data
(a) H&E staining of ovaries from women in young, middle and old groups for scRNA-seq and ST-seq (n = 15). (b) Percentages of mitochondrial genes detected in each sample. Box-and-whisker plots (minimum, 25th percentile, median, 75th percentile, maximum). (c) Left: numbers of unique molecular identifiers (UMIs) (upper) and genes (lower) detected in each cell. Right: UMIs counts and expressed genes are shown by three groups. (d) The number of expressed UMIs (nUMI) and genes (nGene) in different cell types. (e) UMAP plot showing the eight ovarian cell clusters in each sample. (f) UMAP plot showing all ovarian cell clusters. (g) Top three markers of each cell type shown as expression quantity (circle size) and mean expression (color). (h) UMAP cluster map of each slide showing expression of marker genes. (i) Bar plot showing the cell numbers of different cell types and detected genes in human ovaries. (j) Immunofluorescence staining of human GCs markers (AMH and GSTA1) and oocytes markers (ZP3 and TUBB8). The experiment was repeated for three times.
Quality control and summary of ST-seq data
(a) Violin plots showing the percentage of mitochondrial genes detected in each sample. (b) Spatial feature plots of the number of expressed UMI (nUMIs) and genes (nGene) in each sample. (c) Clustered ST spots integrated with scRNA-Seq cell type annotations mapped to the H&E image showing eight cell types in each sample.
Supplement of gene expression in different cell types changed throughout human ovarian aging
(a) Heatmaps showing the upregulated (left) and downregulated (right) DEGs of each cell type between the old and young groups (O/Y), middle and young groups (M/Y), and old and middle groups (O/M). Gene numbers on the left represents from top to bottom, DEGs shared by at least two cell types, DEGs shared by at least two groups, unique DEGs of each cell type in each group. The numbers of unique DEGs are annotated. (b) Venn diagram of DEGs shared by three groups. Y, young; M, middle; O, old. (c) Fluorescence-based-β-Gal staining of human ovaries from young, middle and old group. The scores are listed on the right. Data are presented as the mean ± SEM. n = 10 for each group (one-way ANOVA). (d) Relative mRNA expression of SASPs in human ovaries at different ages by RT–qPCR. Data are presented as the mean ± SEM. n = 9 for each group (one-way ANOVA). (e) Gene set score analysis of NF-κB signaling pathway in eight ovarian cell types of different groups. Two-sided Wilcoxon rank-sum tests. Box-and-whisker plots (minimum, 25th percentile, median, 75th percentile, maximum). n = 3 per age group. (f) Protein expression of cellular senescence-related genes in human ovaries detected by Western blot. Representative images were shown. Data are presented as the mean ± SEM. This test was repeated three times (one-way ANOVA).
Source data
Supplement of gene expression in oocytes changed throughout human ovarian aging
(a) The expression level of stage-specific markers for oocytes. LMOD3 and FOS for primordial oocytes, RPS4X and FIGLA for primary oocytes, SYT5, STK26 and TAF1A for secondary oocytes, UBOX5 and CCDC25 for antral oocytes, HTRA3 and NBPF12 for preovulotary oocytes. (b) Heat map illustrating the DEGs of each oocyte stage. DEG numbers are shown on the left. (c) Representative genes of DNA damage (STAT3, EIF4A1) and DNA repair (APEX1, RAD1) expression in oocytes of three groups. Y, young; M, middle; O, old. (d) Representative images of oocytes by IHC of γH2AX, 8-OHdG and nitrotyrosine between three groups.(e) IHC scores of relative expression for each group. AOD, average optical density. Data are presented as the mean ± SEM. n = 10 for each group (one-way ANOVA).
Supplement of cell communications in oocytes
(a) The receptor-ligands of communication between oocytes and other ovarian cells in the old group. P values were calculated by Fisher’s exact test. (b) ST spot showing the expression of receptor–ligand pair MDK and LRP1 in human ovary.

+13

Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1
  • Article
  • Full-text available

April 2024

·

71 Reads

Nature Aging

·

·

·

[...]

·

Shixuan Wang

Limited understanding exists regarding how aging impacts the cellular and molecular aspects of the human ovary. This study combines single-cell RNA sequencing and spatial transcriptomics to systematically characterize human ovarian aging. Spatiotemporal molecular signatures of the eight types of ovarian cells during aging are observed. An analysis of age-associated changes in gene expression reveals that DNA damage response may be a key biological pathway in oocyte aging. Three granulosa cells subtypes and five theca and stromal cells subtypes, as well as their spatiotemporal transcriptomics changes during aging, are identified. FOXP1 emerges as a regulator of ovarian aging, declining with age and inhibiting CDKN1A transcription. Silencing FOXP1 results in premature ovarian insufficiency in mice. These findings offer a comprehensive understanding of spatiotemporal variability in human ovarian aging, aiding the prioritization of potential diagnostic biomarkers and therapeutic strategies.

Download
Share

309 Cross-ancestry exome sequencing identifies novel endometrial cancer susceptibility genes

March 2024

·

3 Reads

International Journal of Gynecological Cancer

Introduction/Background Endometrial cancer (EC) is the most common gynaecological cancer in high income countries and its incidence is rising globally. Genetic susceptibility of EC have been linked with several loci of common variants, however, these only explained ~10% genetic background and the overall contribution of rare coding variants to EC is unclear. Methodology Here, we performed exome-wide association study to evaluate the penetrance of rare coding variants on cross-ancestry datasets. The UK Biobank conprised of 1,587 EC cases and 159,324 non-cancer controls from Gaussian ancestry. Burden tests from the STAAR pipeline were applied to identify novel genes with excess pathogenic loss-of-function (pLOF) and disruptive missense (d-mis) variants in EC. We replicated these results on in house Chinese EC cohort of 239 patients compared with 10,588 Chinese controls from ChinaMAP. In vitro experients were used to assess effects of these genes. Results Associations between pLOF as well as pLOF plus d-mis (pLOF_ds) variants and EC identified 50 and 26 genes at exome-wide significance (p < 2.5*10–6), respectively, including known EC susceptibility genes MSH6 and MSH2. These results were replicated in Chinese cohort, revealing X genes also contributing to EC in East Asia population. In vitro silence of these genes through small interfering RNA (siRNA) suggested that functional silence of these genes would result in decreased proliferation rates and migration capacity. Conclusion We uncovered novel EC risk genes across European and Asia population and validated their functions. This would provide new insights on genetic background of EC and pave the way for screening of EC. Disclosures The authors declare no competing interests.


216 Risk of ovarian cancer in women with pelvic inflammatory disease under 55: a prospective study in UK biobank database

March 2024

·

1 Read

International Journal of Gynecological Cancer

Introduction/Background Ovarian cancer is a heterogeneous disease with perplexing pathogenesis mechanism and prevention strategies are limited. Pelvic inflammatory disease (PID) has been suggested to increase the risk of developing ovarian cancer (OC), but existing literature have been inconclusive and large prospective cohort studies are inadequate. Methodology We conducted a prospective cohort study of 261624 women participants in the UK Biobank. During a median of 12.8 years of follow-up, we included 901 incident OC cases. Germline homologous recombination (gHR) mutations carriers were determined using paired whole-exome-sequencing data. We used Cox’s regression models to assess the hazard ratios (HRs) of risk of developing OC under PID, with adjustment for confunding factors. We also conducted subgroup analysis by tumor histology subtyping and age. The interactive effect of gHR and PID history was determined using cox regressions. Results We identified 4054 women with PID and 257570 controls in this large prospective cohort study. Among participants 901 had developed ovarian cancer during a median of 12.8 years follow-up period. The adjusted hazard ratio for ovarian cancer in patients with PID was 1.45 (95% CI 0.90–2.32) compared with controls. However, in the age subgroup analysis, we found a positive relation between history of PID and risk of OC in aged younger than 55 years (1.92, 95% CI 1.02–3.63). Furthermore, participants who had a PID history and gHR mutations have the highest risk of developing OC (7.40, 95% CI 1.03–53.10). Conclusion In our study, we found that PID is a potential precursor for ovarian cancer, especially in participants aged younger than 55 years with gHR mutation, as participants who had a PID history and HRD mutations have the highest risk of developing OC. The mechanism between genetic susceptibility, PID and OC remains still obscure and needs to be further investigated. Disclosures The authors declare no competing interests.


215 Spatial tumor evolution panorama of ovarian cancer

International Journal of Gynecological Cancer

Introduction/Background Ovarian cancer is an aggressive disease characterized by extensive intraperitoneal dissemination. However, the mechanisms underlying the facilitation of metastasis within site-specific microenvironments remain unknown. Methodology We analyzed approximately paired 60 whole genome- and bulk RNA-sequencing, ~ 150, 000 of single cells and two million spots of spatial transcriptomics (ST) data to profile site-specific microenvironment and cell-cell contact in situ. We conducted in vitro and in vivo experiments to confirm phenotypes and validate our hypothesis. Results We unveiled a spatiotemporal landscape of ovarian cancer, elucidating the roles of metastatic units (MU), comprised of tripartite ensemble of MMP11+ myCAFs, epithelial cells and SPP1+ macrophages, in promoting tumor spread. In vitro and in vivo experiments indicated that MU could be targeted and disrupted by gene X and gene Y inhibition. We observed dynamic conversions on ST, with immune-activated cell neighborhoods in ovaries transitioning to stromal-like in metastases. Moreover, utilizing our newly designed STARLETS framework, we elucidated a Darwinian evolutionary process driven by hypoxia and immune selections occurring in the ovary while selected clone subsequently migrated to liver. Conclusion Our results provided novel insights into the mechanisms of metastasis and offered a foundation for further investigations into the co-evolution of tumors and host systems, as well as treatments that target the interplay between malignant tumors and immune cells in ascites. Disclosures The authors have no competing interests.



Neoadjuvant camrelizumab plus chemotherapy for locally advanced cervical cancer (NACI study): A prospective, single-arm, phase II trial.

June 2023

·

29 Reads

Journal of Clinical Oncology

5520 Background: First-line treatments for locally advanced cervical cancer (LACC) have limited efficacy and neoadjuvant chemotherapy (NACT) is an emerging approach, however, two-thirds of patients (pts) respond to it and pts without response benefit little. PD-1 inhibitors have shown promising role in recurrent or metastatic cervical cancer. This study aims to evaluate the efficacy and safety of preoperative PD-1 inhibitor camrelizumab combined neoadjuvant therapy for LACC. Methods: The study is designed as a multicenter, open-label, single-arm, prospective phase II study. Pts are enrolled if they had previously untreated LACC (2018 FIGO staged IB3, IIA2 and IIB/IIIC1r (tumor size > 4cm). Eligible pts will receive neoadjuvant chemo-immunotherapy (NACIT), defined as one cycle of cisplatin (75-80 mg/m2, iv) plus nab-paclitaxel (260 mg/m2, iv) NACT and subsequent two cycles of camrelizumab (200mg, iv) combined NACT. Either surgery or concurrent chemoradiotherapy are conducted according to the response as per the Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1. The primary endpoint was objective response rate (ORR), and the secondary endpoints were pathological complete remission (pCR) rate, rate of postoperative adjuvant treatment, event-free survival, overall survival and safety. Results: From Dec 1, 2020 to Feb 1, 2023, 83 pts were enrolled, and 78 pts were evaluated for response. The ORR was 100% (95%CI, 95.38 to 100), with 14 (17.95%) complete response (CR) and 64 (82.05%) partial response. Regarding the pathological findings of 76 pts who underwent radical surgery, 30 (39.47% (95%CI, 28.44 to 51.35)) pts achieved pCR, while 17 (22.37%) needed postoperative adjuvant treatment as indicated in NCCN guideline, of who 14 had positive pelvic nodes, positive surgical margin, and/or positive parametrium and the other three met Sedlis criteria. RECIST CR was significantly associated with pCR ( P = 0.016). Pre-treatment PD-L1 expression (Combined Positive Score) was a predictive biomarker for RECIST CR ( P = 0.036) but not for pCR ( P = 0.078) in these evaluated patients. Grade 3 or 4 treatment-related adverse events occurred in 35 (44.87%) pts during NACIT; the most common were lymphocytopia (25.64%), neutropenia (12.82%) and leucopenia (8.97%). Conclusions: NACIT for LACC demonstrated extremely high ORR and pCR rate with manageable toxicity profile, and greatly reduced the need of postoperative adjuvant therapy. Clinical trial information: NCT04516616 .


Figure 5
Single cell RNA sequencing reveals C5aR1 inhibition to selectively target pro-tumorigenic M2 macrophages reversing PARP inhibitor resistance

November 2022

·

41 Reads

Although Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have been approved in multiple diseases, including BRCA1/2 mutant breast cancer, responses are usually transient thus requiring the deployment of combination therapies that can prevent or reverse PARPi resistance. We thus explored mechanisms underlying sensitivity and resistance to PARPi using two intrinsically sensitive and resistant syngeneic murine breast cancer models. Our data indicate that the PARPi-sensitive tumor model has a high ratio of M1 anti-tumor/M2 pro-tumor macrophages with the M1/M2 ratio being increased by PARPi. In contrast the PARPi-resistant tumor model had very low levels of M1 macrophages and thus a low M1/M2 ratio that was not altered by PARPi. Transplantation of the PARPi-sensitive and the PARPi-resistant tumor in opposite mammary fat pads results in accumulation of M2 macrophages in the sensitive tumor, rendering the sensitive tumor PARPi resistant suggesting that transit of M2 macrophages could contribute to resistance across distant sites both within and between tumors. C5ar1 and Rps19/C5ar1 signaling are selectively elevated in the M2 macrophages that are associated with PARPi resistance. Indeed, C5aR1 positive cells were sufficient to transfer resistance to PARPi. Strikingly targeting C5aR1 decreased M2 macrophage numbers, while sparing M1 macrophages rendering PARPi-resistant tumors sensitive to PARPi in a CD8 T cell dependent manner. Consistent with the murine data, high C5aR1 levels in human breast cancers are associated with a poor response to immune checkpoint blockade. Thus, targeting C5aR1 may represent an approach to selectively deplete M2 macrophages and engender sensitivity to PARPi and potentially other therapies.


Study schema and method for cell‐free DNA (cfDNA) analysis in patients with high‐grade serous ovarian carcinoma. (A) Diagram showing the schema of peripheral blood (PB) sample collection. PB samples are collected at the following time points: pretreatment (B0), then every 3 months after olaparib treatment (B1, B2, B3, and B4). The duration of PB collection is determined according to the follow‐up of patients. (B) Flow diagram showing the processing, detection, and analysis of cfDNA. C, Schema of the method of cfDNA detection. CLAmp‐seq, circular ligation amplification and sequencing; gDNA, genomic DNA; RCA, rolling circle amplification; WBC, white blood cell
Associations between cell‐free DNA (cfDNA) or maximum mutant allele frequency (Max MAF) and the prognosis of patients with high‐grade serous ovarian carcinoma. (A) Distribution maps of progression‐free survival (PFS), cfDNA yield, and Max MAF in pretreatment (B0). (B, C) Pearson correlation analysis between PFS and cfDNA yield (B, p = 0.1484) or Max MAF (C, p = 0.0065) at baseline (B0). (D, E) Differences of cfDNA yield (D, p = 0.9069) or Max MAF (E, p = 0.0081) in patients before treatment (Pre‐T) and after treatment (Post‐T), paired t‐test. (F) Dynamic changes of cfDNA yield or Max MAF with treatment process in different outcomes. (G) Difference of the tendency of the proportion of increased cfDNA yield or Max MAF in different outcomes; χ²‐test, p < 0.0001. (H, I) Survival analyses of the changes of cfDNA yield (H, p = 0.5577) and Max MAF (I, p = 0.0043) in patients. HR, hazard ratio; mPFS, median PFS; PARPi, poly(ADP‐ribose) polymerase inhibitor
Pathogenic germline mutations of homologous recombination repair pathway improve the survival benefits of patients with high‐grade serous ovarian carcinoma (HGSOC). (A) Germline mutation map of HGSOC patients at baseline (B0). (B) Distribution map of pathogenic germline mutations in patients at baseline (B0). (C) Differences in variant counts among different outcomes; ordinary one‐way ANOVA, p = 0.1015. (D) Pearson correlation analysis between progression‐free survival (PFS) and germline mutation load; p = 0.9127. (E) Differences in PFS between carriers and noncarriers of pathogenic mutations; unpaired t‐test, p = 0.0211. (F) Survival analysis in patients with pathogenic germline mutations; p = 0.0229. (G) Differences in PFS between carriers and noncarriers of germline BRCA1/2 (gBRCA) mutations; unpaired t‐test, p = 0.0281. (H) Survival analysis in patients with gBRCA mutations (H, p = 0.1090). (C, E, G) Data are presented as mean ± SEM. HR, hazard ratio; mPFS, median PFS; PARPi, poly(ADP‐ribose) polymerase inhibitor
Associations between somatic mutation profiles and the prognosis of olaparib maintenance therapy in patients with high‐grade serous ovarian carcinoma. (A, B) Distribution maps of somatic mutations in pretreatment (Pre‐T) (A) and post‐treatment (Post‐T) (B) patients. (C) Changes of mutation load in patients before and after olaparib treatment. Paired t‐test: Unrelapsed, p = 0.6109; Relapsed > 12 months, p = 0.8893; Relapsed < 12 months, p = 0.0004. (D) Pearson correlation analysis between progression‐free survival (PFS) and increased mutation load in post‐treatment; p = 0.1817. (E) Differences in the ratio of new mutations among different outcomes. Kruskal–Wallis test with Dunn’s multiple comparisons test: Unrelapsed versus Relapsed > 12 months, p = 0.2162; Unrelapsed versus Relapsed < 12 months, p = 0.0192. (F–H) Survival analyses of somatic mutations (mut) in TP53 (F, p = 0.2762), MRE11A (G, p = 0.0267), and ATM (H, p = 0.0759) in Pre‐T patients. (I–K) Survival analyses of new somatic mutations in MRE11A (I, p = 0.0005), TP53 (J, p = 0.0728 ), and ATM (K, p = 0.1787) in Post‐T patients. (L–N) Survival analyses of combined somatic mutations of pre‐MRE11A + post‐TP53 (L, p = 0.0305), pre‐ATM + post‐MRE11A (M, p = 0.0057), and pre‐ATM + post‐TP53 (N, p = 0.0006) in patients. (C, E) Data are presented as mean ± SEM. HR, hazard ratio; mPFS, median PFS; PARPi, poly(ADP‐ribose) polymerase inhibitor
Associations between somatic mutation sites and the prognosis of olaparib maintenance therapy in patients with high‐grade serous ovarian carcinoma. (A, B) Distribution maps of somatic mutation sites in pretreatment (A) and post‐treatment (B) patients. (C–E) Survival analyses of CHEK2:p.K373E (C, p = 0.0219), CHEK2:p.R406H (D, p = 0.1319), and MRE11A:p.K464R (E, p = 0.0267) in pretreatment. (F, G) Mutation (mut) tracking of CHEK2:p.K373E (F) and CHEK2:p.R406H (G) in cell‐free DNA (cfDNA) detection during olaparib treatment. (H, I) Survival analyses of CHEK2:p.K373E‐higher (H, p = 0.0091) and CHEK2:p.R406H‐higher (I, p = 0.0002) in post‐treatment. (J) Mutation tracking of MRE11A:p.K464R in cfDNA detection during olaparib treatment. (K) Longitudinal representation of MRE11A:p.K464R from all patients with a timepoint available. (L) Survival analysis of newly acquired MRE11A:p.K464R (G, p = 0.0005) in post‐treatment. (M, N) Survival analyses of combined somatic mutation sites of post‐CHEK2:p.K373E + post‐MRE11A:p.K464R (M, p = 0.0120) and post‐MRE11A:p.K464R + post‐CHEK2:p.R406H (N, p < 0.0001) in patients. HR, hazard ratio; mPFS, median PFS; PARPi, poly(ADP‐ribose) polymerase inhibitor
Mutation profiles in circulating cell‐free DNA predict acquired resistance to Olaparib in high‐grade serous ovarian carcinoma

June 2022

·

38 Reads

·

6 Citations

Cancer Science

Cancer Science

Although Poly (ADP-ribose) polymerase (PARP) inhibitors' resistance has gradually become a major challenge in the maintenance therapy for high-grade serous ovarian carcinoma (HGSOC), there are no universal indicators for resistance monitoring in patients. A key resistance mechanism to PARP inhibitors (PARPis) is the restoration of homologous recombination repair (HRR), including BRCA reversion mutations and changes in DNA damage repair proteins. To explore mutation profiles associated with PARPis' resistance, we performed targeted 42-gene deep sequencing of circulating cell-free DNA (cfDNA) extracted from pre-treatment and post-treatment in HGSOC patients with Olaparib maintenance therapy. We found that pathogenic germline mutations in the HRR pathway, including BRCA1/2, were strongly associated with improved clinical outcomes, and newly acquired MRE11A mutations significantly shortened the progression-free survival (PFS) of patients. Furthermore, dynamic fluctuations of somatic mutation sites in CHEK2:p.K373E and CHEK2:p.R406H can be used for evaluating the therapeutic efficacy of patients. MRE11A:p.K464R may be a vital driving factor of Olaparib resistance, which patients who newly acquired MRE11A:p.K464R in post-treatment cfDNA had significantly shorter PFS than those without it. These findings provide potential noninvasive biomarkers for efficacy evaluation and resistance monitoring of Olaparib treatment, and lay the foundation for developing combination treatment after Olaparib resistance.


Spatiotemporal analysis of human ovarian aging at single-cell resolution

May 2022

·

106 Reads

·

5 Citations

Our understanding of how aging affects the cellular and molecular components of the human ovary and contributes to age-related fertility decline is still limited. Here, we link single-cell RNA sequencing and spatial transcriptomics to characterize human ovarian aging. Changes of the molecular signatures of eight types of ovarian cells during aging were defined. We combined single cell types with their spatial location information to divide ovarian granulosa cells into three subtypes and theca & stroma cells into five subtypes. Further analysis revealed increased cellular senescence with age and characterized the transcription factor FOXP1 as a master regulatory gene during ovarian aging. Inhibition of FOXP1 in ovarian cells increased cellular senescence which was alleviated by pharmacological treatment with quercetin or fisetin. These findings provide a comprehensive understanding of the spatiotemporal variability of human ovarian aging, providing resources for developing new diagnostic biomarkers and therapeutic strategies against ovarian aging.



Citations (9)


... In this study, variables with missing data were analyzed using dummy variables, defined as the unknown group. [25][26][27] Radiotherapy was analyzed as a 3-category variable (yes, no, and unknown). We classified the tumor size subgroups following the definitions provided by the AJCC. ...

Reference:

A Competing Risk Nomogram for Prediction of Prognosis in Patients With Primary Squamous Cell Thyroid Carcinoma
Articles The prognosis of patients with small cell carcinoma of the cervix: a retrospective study of the SEER database and a Chinese multicentre registry

... Recent studies have shown the feasibility to detect reversion mutations by cfDNA analysis suggesting its potential clinical use [81][82][83]. Christie et al. [82] conducted a prospective study in 30 patients with HGSOC carrying a germline BRCA1/2 mutation and detected BRCA1/2 reversion mutations in the tumor in 31.3% of patients treated in the recurrent setting, among which 18.8% also had detectable BRCA1/2 reversions in cfDNA [82]. ...

Mutation profiles in circulating cell‐free DNA predict acquired resistance to Olaparib in high‐grade serous ovarian carcinoma
Cancer Science

Cancer Science

... Single-cell analyses of ovarian aging in nonhuman primates identified downregulation of antioxidant programs in aged oocytes and increased apoptosis in aged granulosa cells (GCs) 18 . In addition, single-cell analyses of human ovarian tissue are currently in progress 19 . However, mice represent the model organism most utilized for ovarian aging studies 20 due to their short lifespan and ease of genetic manipulation for mechanistic studies. ...

Spatiotemporal analysis of human ovarian aging at single-cell resolution

... In the ID8 OC xenografts model, Adavosertib and anti-PD-L1 considerably slowed the growth of tumors and improved mouse survival without causing noticeable side effects. 47 These findings provide a rationale for the combination of WEE1i and ICB, though more evidence is needed to verify the feasibility of this strategy before its implementation in clinical settings. ...

WEE1 inhibition induces anti-tumor immunity by activating ERV and the dsRNA pathway

... Although PARPi in combination with chemotherapy have been approved for management of BRCA-associated metastatic triplenegative breast cancer (TNBC) 1,2 , benefits are not durable with almost universal recurrence. The mechanisms underlying PARPi resistance have been explored extensively and include BRCA1/2 reversion mutations, hypomorphic BRCA1/2 alleles, loss of hypermethylation of BRCA1/2, loss of the shieldin complex and activation of pro-survival pathways such as the MAPK and PI3K pathways [3][4][5][6][7] . However, taken together, the identified mechanisms can only explain PARPi resistance in a subset of patients [8][9][10] . ...

Systems approach to rational combination therapy: PARP inhibitors
  • Citing Article
  • May 2020

Biochemical Society Transactions

... The role of protein neddylation on cancer immunogenicity has been investigated using NAE inhibitors. Pevonedistat treatment causes proteome instability and strongly potentiates response to ICB antibodies in mismatch repair-deficient (dMMR) colon cancer cells 52 . In glioblastoma models, pevonedistat up-regulates PD-L1 expression on cancer cells and synergizes with ICB antibodies in mice 53 . ...

Proteome Instability Is a Therapeutic Vulnerability in Mismatch Repair-Deficient Cancer
  • Citing Article
  • February 2020

Cancer Cell

... 25 Recently, Fang et al reported that synergy between PAPRi and WEE1i was most clearly manifest in KRAS or BRAF mutant OC cells that are resistant to PARPi. 31 The same study showed that using sequential rather than concurrent therapy for OC maintained the synergy of PARPi and WEE1i while reducing toxicity. 31 The above research results provide rationale and evidence for further clinical testing. ...

Sequential Therapy with PARP and WEE1 Inhibitors Minimizes Toxicity while Maintaining Efficacy
  • Citing Article
  • June 2019

Cancer Cell

... Next, to prioritize the clinical application potentials, we estimated a priority score for each combination by considering the CI and four additional features of each epigenetic inhibitor (single agent treatment efficacy, cancer dependency defined by genetic screen from the DepMap project, and expression dysregulation and genomic alterations from the TCGA project, Figure 1F). As expected, candidates previously identified by us 15 and other groups, such as BETi [15][16][17] , DNMTi 21,22 , and HDACi 24,25 , were ranked at the top of the list ( Figure 1G and Table S1). ...

BRD4 Inhibition Is Synthetic Lethal with PARP Inhibitors through the Induction of Homologous Recombination Deficiency
  • Citing Article
  • March 2018

Cancer Cell

... 33 A synergy between PARP and MEK inhibitors was observed in pancreatic and ovarian cancer, with mechanistic convergence on the HR repair pathway. 34 Furthermore, a dual PARP-RAD51 inhibitor was developed 35 and showed antineoplastic effects in the absence of radiation. ...

Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers
  • Citing Article
  • May 2017

Science Translational Medicine