Skip to main content

ATM, BLM, and CDH1 gene co-mutations in a high-grade endometrial stromal sarcoma patient with multiple abdominal cavity metastases: a case report and literature review

Abstract

Background

High-grade endometrial stromal sarcoma (HG-ESS) is a rare malignant tumor with poor prognosis. To overcome the limitations of current treatment for advanced patients, the intervention of targeted drug therapy is urgently needed.

Case presentation

A 74-year-old married woman who presented with abdominal distension and lower abdominal pain was admitted to Hebei General Hospital. After surgery, immunohistochemical staining revealed a malignant tumor which was consistent with HG-ESS. Tumor recurrence occurred 2 months after surgery. Then the patient underwent chemotherapy with two courses but responded poorly. Subsequently we observed ATM, BLM, and CDH1 co-mutations by Next Generation Sequencing (NGS). Then the patient received pamiparib, which resulted in a 10-month progression-free survival (PFS) and is now stable with the administration of sintilimab in combination with pamiparib and anlotinib.

Conclusions

Due to the successful use of poly ADP-ribose polymerase inhibitor (PARPi) on HG-ESS, we suggest that the selection of effective targeted drugs combined with anti- programmed death-1 (PD-1) drug therapy based on genetic testing may become a new option for the treatment of homologous repair deficient (HR-deficient) HG-ESS.

Peer Review reports

Background

Endometrial stromal sarcoma (ESS), derived from endometrial stromal cells, accounts for approximately 1% of all uterine malignancies. According to the latest 2020 WHO classification, ESSs can be divided into four categories: endometrial stromal nodule (ESN), low-grade endometrial stromal sarcoma (LG-ESS), high-grade endometrial stromal sarcoma (HG-ESS) and undifferentiated uterine sarcoma (UUS) [1]. HG-ESS, a highly rare event with a high rate of focal and metastatic recurrence and poor prognosis, is of unique biological behaviors. The treatment for early (stage I to stage II) HG-ESS patients is mainly surgery, including total hysterectomy plus bilateral adnexectomy. However, due to its uncertainty in improving patient prognosis, whether tumor cell reduction should be performed for advanced (stage III to IV) HG-ESS patients is unclear. Moreover, radiotherapy or chemotherapy, the most common postoperative adjuvant therapy strategy, is not effective for treating HG-ESS. In this study, we report a case of HG-ESS mutated with the ATM, BLM, and CDH1 genes, which achieved satisfactory clinical results with the oral targeted drug pamiparib.

Case presentation

A 74-year-old married woman who presented with abdominal distension and lower abdominal pain in April 2022 was admitted to Hebei General Hospital. Pelvic abdominal computed tomography (CT) suggested that there was a mixed-density space-occupying lesion at the base of the uterus as well as abdominal pelvic effusion (Fig. 1A). Gynecological ultrasound indicated that there was a mixed echo mass in the uterine (129 × 124 × 105 mm mixed echo mass, the inner part of which was heterogeneous high echo with strong echo spots) and a pelvic-abdominal cavity mixed echo mass (233 × 208 × 131 mm mixed echo mass above the uterus) with an abdominal fluid dark area. Serological examination revealed a high level of CA125 (162.300U/ml). The patient underwent exploratory laparotomy under general anesthesia on 2022-04-20.

The laparotomy findings were an irregular cystic solid mass with a diameter of approximately 30 cm in the pelvic-abdominal cavity adhering to the posterior wall of the uterus, intestinal tubes, and peritoneum, and the surface of the greater omentum was covered with focal nodules. Intraoperative frozen sections revealed spindle malignancy cells. Subsequently, total abdominal hysterectomy, bilateral adnexectomy, partial greater omentectomy, pelvic abdominal mass resection, and pelvic adhesion release were performed. In histopathological analysis, the resected specimen exhibited diffusely distributed tumor cells in high-magnification images. Tumor cells rendered short spindle shapes and scattered multinucleated tumor giant cells could be seen. Nuclear chromatin was fine and uniform, and the mitotic index was > 10/10 in high-power fields (Fig. 2A). Immunohistochemistry showed positive staining for cell CyclinD1, CD10, Caldesmon, and Vimentin in the tumor cells, with some tumor cells also demonstrating positive labeling for MyoD1, CKpan, and Myogenin (Fig. 2B, C, D). Tumor cells were negative for estrogen receptor, smooth muscle actin, CD117, and S100. The Ki-67 labeling index was approximately 40%. Based on immunohistochemical staining, it was consistent with HG-ESS of the endocervix with cartilage, bone, and striated muscle differentiation.

The patient refused further radiotherapy and chemotherapy. Unfortunately, tumor recurrence occurred 2 months after surgery (Fig. 1B). Thus, the patient was given injections of liposomal doxorubicin and ifosfamide for 2 cycles. The growth of solid component enhancement in an outer rim of the tumor in the right flank and septal enhancement were revealed using enhanced CT (Fig. 1C). The patient exhibited ongoing abdominal distention with a progressive increase in abdominal circumference, and the reaction of the cancer to chemotherapy was unsatisfactory. Moreover, the patient had poor tolerance to chemotherapy. Subsequently, hybridization capture-based targeted Next Generation Sequencing (NGS) was performed on the illumina MiSeq platform (Life Healthcare Group Limited, Beijing, China). The patient underwent testing with a 176-gene panel associated with molecularly targeted drugs, immunotherapy, and chemotherapeutic agents for solid tumors. The proportion of quality score of the sequencing data above Q30 in the samples was more than 92% and passed the quality control. It was found that ATM, BLM, and CDH1 genes were mutated. Specifically, a substitution of c. 5908 C > T (p.Gln1970*) was identified in exon 39 of the ATM gene with a mutation abundance of 1.17%. Additionally, a mutation of c.1937G > T (p.Ser646Ile) was detected in exon 8 of the BLM gene with a mutation abundance of 32.05%. The third significant genetic change was a c.2024 A > G mutation in exon13 of the CDH1 gene (p.Lys675Arg) with 45.72% mutation abundance. The results showed that the tumor belonged to microsatellite stable (MSS) phenotype. Subsequently, the patient received pamiparib 80 mg a day as well as an intraperitoneal infusion of recombinant human endostatin (45 mg d1,60 mg d4,60 mg d8) combined with cisplatin (40 mg d2, d5, d8), respectively. Efficacy was evaluated for partial response (PR) (Fig. 1D). Subsequent single-agent administration of pamiparib 40 mg 2/day maintenance therapy resulted in a 10-month progression-free survival (PFS) (Fig. 1E). Unfortunately, tumor progression reappeared after 10 months and is now stable with the administration of sintilimab in combination with pamiparib and anlotinib (Fig. 1F) (Last follow-up 2023-12-29).

Fig. 1
figure 1

Radiologic features at different time points. A Preoperative CT scan (unenhanced CT) (2022-4). B Tumor recurrence after surgery (enhanced CT) (2022-6). C After 2 cycles of chemotherapy, no significant relief was revealed using enhanced CT (2022-8). D After 6 months of pamiparib treatment, an enhanced CT scan indicated PR (2023-2). E A 10-month PFS was achieved using pamiparib (enhanced CT) (2023-6). F The latest enhanced CT review (2023-9)

Fig. 2
figure 2

Histopathologic features of HG-ESS. A Hematoxylin and eosin (HE) staining (40×) revealed striated muscle differentiation in HG-ESS. B immunohistochemical (IHC) examinations (100×) of HG-ESS tissue was positive for CD10. C IHC examinations for CyclinD1. D IHC examinations for MyoD1

Discussion

The clinical presentation of HG-ESS is often nonspecific, possessing a high degree of malignancy and aggressiveness associated with a poor prognosis. Histopathological examination remains the definitive standard for diagnosis. Risk factors that have been found to influence overall survival in HG-ESS include disease stage, tumor size, minimum and average CA125 levels, menopausal status, history of uterine smooth muscle tumors, and endometriosis [2]. A study showed that for patients with stage III, the 1-year disease-specific survival (DSS) rate was 26.7%, and 0% for those with stage IV [3]. Analysis of the data revealed that the median survival time (95% CI) for HG-ESS is only 19.9 (17.1–22.1) months, and for each additional 1 cm in tumor size, the survival rate decreases by 2%[4].

The current treatment for HG-ESS predominantly involves surgical intervention complemented by adjuvant systemic therapy and local radiotherapy. However, the efficacy of lymphadenectomy for HG-ESS is controversial [5]. Current studies suggest that a multimodal approach combining surgery, radiotherapy, and chemotherapy may enhance PFS exclusively in patients with early-stage disease [6]. For patients with advanced recurrent metastases, there is currently no optimal standard treatment, and the National Comprehensive Cancer Network guidelines recommend that patients with NTRK-like family member 4 (SLITRK4) gene fusions choose targeted therapies such as larotrectinib or entrectinib.

The combination of CDK4/6 inhibitors and aromatase inhibitors has recently been studied as an option for treating ER-positive patients with BCOR-related metastatic HG-ESS [7].

In this case, a 74-year-old woman was diagnosed with ATM, BLM, and CDH1 co-mutations in HG-ESS with a lesion characterized by a large mass (30 cm) and late-stage disease, and she did not receive adjuvant therapy after surgery. The patient, whose tumor recurred two months post-surgery, and the response to chemotherapy was unsatisfactory, achieved remission following treatment with the poly ADP-ribose polymerase inhibitor (PARPi) pamiparib.

Advancements in molecular analysis techniques have led to the detection of mutations in various genes associated with HG-ESS, including YWHAE, NUTM2, EPC1, SUZ12, BCOR, PHF1, ZC3H7B, EML4, COL1A1, PDGFB, STRN, MDM2, CDK4, SLITRK4, etc. [8,9,10,11,12,13,14,15,16,17]. With the development of NGS technologies, other mutated genes may also be identified, and could serve as potential therapeutic targets.

In this report, NGS confirmed mutations within the ATM, BLM, and CDH1 genes, among which the ATM and BLM genes were classified as homologous recombination repair (HRR) genes.

The ataxia-telangiectasia mutated (ATM) protein is the most critical initiator of the DNA damage response (DDR) [18], and its signaling pathway involves hundreds of downstream targets that regulate DDR, proliferation, metabolism, and other physiological activities of cells [19, 20]. According to previous reports, the ATM gene is associated with an increased risk of various cancers, such as breast cancer, lung cancer, pancreatic cancer, and melanoma [21,22,23,24]. Studies have shown that mutations in the ATM gene can induce sensitivity to PARPi [25,26,27,28].

Bloom syndrome protein (BLM), a member of the RecQ family of helicases using the energy from ATP hydrolysis to unwind duplex DNA, plays a crucial role in correcting mismatched bases to reduce DNA damage induced by itself or the external environment [29]. Recently, the relationship between BLM and tumor development has been discovered [30, 31]. For instance, as a potential biomarker for prostate cancer, BLM has attracted the attention of many investigators. Several reports have suggested that BLM mutations in prostate cancer increase the sensitivity of patients to PARPi_olaparib [32].

E-cadherin gene (CDH1) mutations are considered to be noteworthy contributors to tumor migration and invasion [33]. Studies have elucidated that the reduction of CDH1 expression increases the cytotoxic effect of PARPi on triple-negative breast cancer cells with or without BRCA defects by inducing DNA damage, checkpoint activation, cell cycle arrest, and cell apoptosis [34].

Few studies have investigated ATM, BLM, and CDH1 gene mutations in high-grade endometrial stromal sarcoma. In the present study, ATM, BLM, and CDH1 mutations were detected by NGS. The patient who received PARPi obtained a 10-month-PFS after 2 cycles of peritoneal perfusion of cisplatin and anti-vascular drugs. PARPi combined with anlotinib was applied after subsequent progression.

Alterations in DDR genes have been linked to genomic instability and increased tumor mutational burden, potentially enhancing tumor immunogenicity [35]. The STING signaling pathway is activated with incompletely repaired DNA damage accumulation, thereby enhancing the immune response [36].

Treatment with PARPi may further increase the level of DNA damage and promote the release of neoantigens and the expression of tumor programmed death-ligand 1 (PD-L1) [37]. The potential synergistic effect between PARPi and PD-1/PD-L1 inhibitors has been confirmed in preclinical studies across various tumor types [38]. Accordingly, we managed to control the disease by administering immunotherapy in combination with PARPi and anlotinib following the patient’s disease re-progression.

Conclusion

Due to the low prevalence of HG-ESS, there is a lack of consensus on diagnostic and treatment strategies, and there is currently no standard treatment for advanced patients with recurrent metastases. Thus, we anticipate that treatment based on the identification of genetic mutations that can be targeted could lead to the exploration of novel therapeutic approaches for HG-ESS. We identified a case of postoperative HG-ESS recurrence in a patient with mutations in the ATM, BLM, and CDH1 genes. This patient responded poorly to chemotherapy. PFS resulting from the PARPi (pamiparib) was nearly 10 months in the first stage. After progression, the PARPi was used in combination with multi-targeted tyrosine kinase inhibitor (anlotinib) and anti-PD-1 (sintilimab)to achieve a PR. To our knowledge, this is the first case report of HG-ESS with ATM, BLM, and CDH1 mutations successfully treated with PARPi based on genetic alteration information generated by NGS, suggesting that the selection of effective targeted drugs combined with anti-PD-1 drug therapy on the basis of genetic testing may become a new option for the treatment of homologous repair (HR-deficient) HG-ESS.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

HG-ESS:

High-grade endometrial stromal sarcoma

NGS:

Next Generation Sequencing

PFS:

progression-free survival

PARPi:

Poly ADP-ribose polymerase inhibitor

HR:

Homologous recombination

ESS:

Endometrial stromal sarcoma

ESN:

endometrial stromal nodule

LG-ESS:

low-grade endometrial stromal sarcoma

UUS:

Undifferentiated uterine sarcoma

CT:

Computed tomography

PR:

Partial response

DSS:

Disease specific survival

HE:

Hematoxylin and eosin

IHC:

immunohistochemical

HRR:

Homologous recombination repair

DDR:

DNA damage response

ATM:

Ataxia-telangiectasia mutated

BLM:

Bloom syndrome protein

CDH1:

E-cadherin gene

PD-1:

Programmed death-1

PD-L1:

Programmed death-ligand 1

References

  1. Gadducci A. Endometrial stromal tumors of the uterus: Epidemiology, pathological and biological features, treatment options and clinical outcomes. Gynecol Oncol. 2023.

  2. Zhang YY, Li Y, Qin M, Cai Y, Jin Y, Pan LY. High-grade endometrial stromal sarcoma: a retrospective study of factors influencing prognosis. Cancer Manag Res. 2019;11:831–7. https://doi.org/10.2147/CMAR.S187849.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Bai H, Yuan F, Liang B, et al. Clinicopathological characteristics and treatment of patients with high-grade endometrial stromal sarcoma. Med (Baltim). 2022;101(2):e28490. https://doi.org/10.1097/MD.0000000000028490.

    Article  CAS  Google Scholar 

  4. Seagle BLL, Shilpi A, Buchanan S, Goodman C, Shahabi S. Low-grade and high-grade endometrial stromal sarcoma: a National Cancer Database study. Gynecol Oncol. 2017;146(2):254–62. https://doi.org/10.1016/j.ygyno.2017.05.036.

    Article  PubMed  Google Scholar 

  5. Dos Santos LA, Garg K, Diaz JP, et al. Incidence of lymph node and adnexal metastasis in endometrial stromal sarcoma. Gynecol Oncol. 2011;121(2):319–22. https://doi.org/10.1016/j.ygyno.2010.12.363.

    Article  PubMed  Google Scholar 

  6. Benson C, Miah AB. Uterine sarcoma – current perspectives. Int J Womens Health. 2017;9:597–606. https://doi.org/10.2147/IJWH.S117754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li C, Wang C. LG-ESSs and HG-ESSs: underlying molecular alterations and potential therapeutic strategies. J Zhejiang Univ-Sci B. 2021;22(8):633–46. https://doi.org/10.1631/jzus.B2000797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Brahmi M, Franceschi T, Treilleux I, et al. Molecular classification of endometrial stromal sarcomas using RNA sequencing defines nosological and prognostic subgroups with different natural history. Cancers. 2020;12(9):2604. https://doi.org/10.3390/cancers12092604.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lee C, Nucci MR. Endometrial stromal sarcoma – the new genetic paradigm. Histopathology. 2015;67(1):1–19. https://doi.org/10.1111/his.12594.

    Article  PubMed  Google Scholar 

  10. Juckett LT, Lin DI, Madison R, Ross JS, Schrock AB, Ali S. A Pan-cancer Landscape Analysis reveals a subset of Endometrial Stromal and Pediatric tumors defined by Internal Tandem duplications of BCOR. Oncology. 2019;96(2):101–9. https://doi.org/10.1159/000493322.

    Article  CAS  PubMed  Google Scholar 

  11. Dickson BC, Lum A, Swanson D, et al. Novel EPC1 gene fusions in endometrial stromal sarcoma. Genes Chromosomes Cancer. 2018;57(11):598–603. https://doi.org/10.1002/gcc.22649.

    Article  CAS  PubMed  Google Scholar 

  12. Cotzia P, Benayed R, Mullaney K, et al. Undifferentiated uterine sarcomas represent underrecognized high-Grade Endometrial stromal sarcomas. Am J Surg Pathol. 2019;43(5):662–9. https://doi.org/10.1097/PAS.0000000000001215.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Chiang S, Lee CH, Stewart CJR, et al. BCOR is a robust diagnostic immunohistochemical marker of genetically diverse high-Grade Endometrial stromal sarcoma, including tumors exhibiting variant morphology. Mod Pathol off J U S Can Acad Pathol Inc. 2017;30(9):1251–61. https://doi.org/10.1038/modpathol.2017.42.

    Article  CAS  Google Scholar 

  14. Chiang S, Cotzia P, Hyman DM, et al. NTRK fusions define a novel uterine sarcoma subtype with features of Fibrosarcoma. Am J Surg Pathol. 2018;42(6):791–8. https://doi.org/10.1097/PAS.0000000000001055.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Croce S, Hostein I, Longacre TA, et al. Uterine and vaginal sarcomas resembling fibrosarcoma: a clinicopathological and molecular analysis of 13 cases showing common NTRK-rearrangements and the description of a COL1A1-PDGFB fusion novel to uterine neoplasms. Mod Pathol. 2019;32(7):1008–22. https://doi.org/10.1038/s41379-018-0184-6.

    Article  CAS  PubMed  Google Scholar 

  16. Grindstaff SL, DiSilvestro J, Hansen K, DiSilvestro P, Sung CJ, Quddus MR. COL1A1-PDGFB fusion uterine fibrosarcoma: a case report with treatment implication. Gynecol Oncol Rep. 2019;31:100523. https://doi.org/10.1016/j.gore.2019.100523.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Michal M, Hájková V, Skálová A, Michal M. STRN-NTRK3-rearranged mesenchymal tumor of the Uterus: expanding the morphologic spectrum of tumors with NTRK fusions. Am J Surg Pathol. 2019;43(8):1152–4. https://doi.org/10.1097/PAS.0000000000001292.

    Article  PubMed  Google Scholar 

  18. Xie X, Zhang Y, Wang Z, et al. ATM at the crossroads of reactive oxygen species and autophagy. Int J Biol Sci. 2021;17(12):3080–90. https://doi.org/10.7150/ijbs.63963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ueno S, Sudo T, Hirasawa A. ATM: functions of ATM kinase and its relevance to Hereditary tumors. Int J Mol Sci. 2022;23(1):523. https://doi.org/10.3390/ijms23010523.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhou H, Chen H, Cheng C, et al. A quality evaluation of the clinical practice guidelines on breast cancer using the RIGHT checklist. Ann Transl Med. 2021;9(14):1174. https://doi.org/10.21037/atm-21-2884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen H, Ye Z, Xu X, et al. ALDOA inhibits cell cycle arrest induced by DNA damage via the ATM-PLK1 pathway in pancreatic cancer cells. Cancer Cell Int. 2021;21:514. https://doi.org/10.1186/s12935-021-02210-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Saunders RA, Michniacki TF, Hames C, et al. Elevated inflammatory responses and targeted therapeutic intervention in a preclinical mouse model of ataxia-telangiectasia lung disease. Sci Rep. 2021;11:4268. https://doi.org/10.1038/s41598-021-83531-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dalmasso B, Pastorino L, Nathan V, et al. Germline ATM variants predispose to melanoma: a joint analysis across the GenoMEL and MelaNostrum consortia. Genet Med. 2021;23(11):2087–95. https://doi.org/10.1038/s41436-021-01240-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang YC, Lee KW, Tsai YS, et al. Downregulation of ATM and BRCA1 predicts poor outcome in Head and Neck Cancer: implications for ATM-Targeted therapy. J Pers Med. 2021;11(5):389. https://doi.org/10.3390/jpm11050389.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mateo J, Carreira S, Sandhu S, et al. DNA-Repair defects and Olaparib in metastatic prostate Cancer. N Engl J Med. 2015;373(18):1697–708. https://doi.org/10.1056/NEJMoa1506859.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Young K, Starling N, Cunningham D. Targeting deficient DNA damage repair in gastric cancer. Expert Opin Pharmacother. 2016;17(13):1757–66. https://doi.org/10.1080/14656566.2016.1217992.

    Article  CAS  PubMed  Google Scholar 

  27. Rimar KJ, Tran PT, Matulewicz RS, Hussain M, Meeks JJ. The emerging role of homologous recombination repair and PARP inhibitors in Genitourinary malignancies. Cancer. 2017;123(11):1912. https://doi.org/10.1002/cncr.30631.

    Article  CAS  PubMed  Google Scholar 

  28. Wang W, Zhang X, Fang Y, et al. Case Report: Olaparib shows satisfactory clinical outcomes against small cell esophageal carcinoma with ATM Mutation. Front Oncol. 2022;12:808801. https://doi.org/10.3389/fonc.2022.808801.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Cox RL, Hofley CM, Tatapudy P, et al. Functional conservation of RecQ helicase BLM between humans and Drosophila melanogaster. Sci Rep. 2019;9:17527. https://doi.org/10.1038/s41598-019-54101-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Datta A, Dhar S, Awate S, Brosh RM. Synthetic Lethal interactions of RECQ helicases. Trends Cancer. 2021;7(2):146–61. https://doi.org/10.1016/j.trecan.2020.09.001.

    Article  CAS  PubMed  Google Scholar 

  31. Mojumdar A. Mutations in conserved functional domains of human RecQ helicases are associated with diseases and cancer: a review. Biophys Chem. 2020;265:106433. https://doi.org/10.1016/j.bpc.2020.106433.

    Article  CAS  PubMed  Google Scholar 

  32. Zhang D, Xu X, Wei Y, et al. Prognostic role of DNA damage response genes mutations and their Association with the sensitivity of olaparib in prostate Cancer patients. Cancer Control J Moffitt Cancer Cent. 2022;29:10732748221129451. https://doi.org/10.1177/10732748221129451.

    Article  Google Scholar 

  33. Atkinson MJ, Reich U, Becker I. E-Cadherin Gene Mutations Provide Clues to Diffuse Type Gastric Carcinomas.

  34. Li J, Lan M, Peng J, et al. Cdh1 Deficiency sensitizes TNBC cells to PARP inhibitors. Genes. 2022;13(5):803. https://doi.org/10.3390/genes13050803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mouw KW, Goldberg MS, Konstantinopoulos PA, D’Andrea AD. DNA damage and repair biomarkers of Immunotherapy Response. Cancer Discov. 2017;7(7):675–93. https://doi.org/10.1158/2159-8290.CD-17-0226.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15(12):760–70. https://doi.org/10.1038/nri3921.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Peyraud F, Italiano A, Combined PARP. Inhibition and Immune Checkpoint Therapy in Solid tumors. Cancers. 2020;12(6):1502. https://doi.org/10.3390/cancers12061502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang Z, Sun K, Xiao Y, et al. Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Sci Rep. 2019;9:1853. https://doi.org/10.1038/s41598-019-38534-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by Natural Science Foundation of Hebei Province(H2023307006).

Author information

Authors and Affiliations

Authors

Contributions

Na.L convinced this study. Nan.L and Y.X.Y wrote the main manuscript. Y.X.L led the pathological assessment. C.W.D and Y.Y.Y interpreted the clinical images. All authors edited the manuscript and approved the final manuscript.

Corresponding author

Correspondence to Na Li.

Ethics declarations

Ethics approval and consent to participate

Collection and use of patient samples were approved by The Ethics Committee of Hebei General Hospital.

Consent for publication

Written informed consent was obtained by the patient for publication of this report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, N., Yan, Y., Li, Y. et al. ATM, BLM, and CDH1 gene co-mutations in a high-grade endometrial stromal sarcoma patient with multiple abdominal cavity metastases: a case report and literature review. BMC Geriatr 24, 603 (2024). https://doi.org/10.1186/s12877-024-05201-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12877-024-05201-z

Keywords