Skip to main content
  • Correspondence
  • Open access
  • Published:

The role of soluble CD80 in patients with soft tissue tumors

Abstract

Background

Immune checkpoint protein (ICP), which is a central factor group of the immune system, has been reported to have a correlation between the degree of its expression and the prognosis of patients with malignant tumors, and many inhibitors have appeared as therapeutic targets. On the other hand, a soluble form of ICP in circulating blood induced systemic immunosuppression. In this study, we investigated the relationship between the soluble form of CD80 (sCD80) which is a ligand for the inhibitory system CTLA-4, in blood, and clinicopathological parameters in patients with soft tissue tumors.

Methods

A total of 119 patients with primary soft tissue tumors were enrolled in this study. The sCD80 levels were measured by enzyme immunoassay.

Results

There were no significant differences in sCD80 levels between benign (34) and soft tissue sarcoma (STS) patients (85). In STS, the high-sCD80 group had significantly lower metastasis-free survival (MS) and lower overall survival (OS) than the low-sCD80 group at 5 years using the log-rank test (OS: high > 404 pg/mL, low ≤ 404 pg/mL, MS: high > 531 pg/ml, low ≤ 531 pg/ml). On multivariate Cox proportional hazard analysis, the high-sCD80 group had significant differences in 5MS and 5OS compared to the low-sCD80 group.

Conclusions

In conclusion, sCD80 may negatively affect systemic immune circumstances, in STS, and may have potential as a therapeutic target.

Background

Soft tissue sarcoma (STS) is a rare, heterogeneous group of tumors [1]. The incidence of STS is fewer than six per 100,000 cancer cases, which represents 1–2% cases of all cancer in adults [1]. Despite recent advances in the diagnosis and treatment of STS, patients who develop metastasis have mortality rates. Therefore, many studies have attempted to define different factors for predicting the prognosis of patients with STS. Older age and deep, truncal, high-grade, and large size of STS have been reported to be prognostic factors that are linked to poor prognosis [2,3,4]. In addition, the formation of an immune escape mechanism that suppresses the immune response to the tumor is known to have a significant effect on its prognosis. Immune checkpoint protein (ICP), which is a central factor group of the immune system, has been reported to have a correlation between the degree of its expression and the prognosis of patients with malignant tumors, and many inhibitors have appeared as therapeutic targets [5]. C. Perisano et al. evaluated the immunohistochemistry of PD1/PD-L1 expression in 60 adult patients affected by high-grade sarcomas. It showed positivity among the different subgroups of positive PD1 lymphocytes infiltration and PD-L1 expression in high-grade sarcomas [6].

On the other hand, a soluble form of ICP in circulating blood induced systemic immunosuppression [7]. An association between the soluble form of PD-L1 (sPD-L1) and cancer has been reported, such as renal cell carcinoma, hepatocellular carcinoma, esophageal cancer, lung cancer, gastric cancer, rectal cancer, and lymphoma [8,9,10,11,12,13,14]. Elevated sPD-L1 had significant relation with exacerbation of prognosis in cancer patients including STS [15]. In this study, we investigated the relationship between the soluble form of CD80 (sCD80) which is a ligand for the inhibitory system CTLA-4, in blood and clinicopathological parameters in patients with soft tissue tumors.

Materials and methods

Patients

A total of 119 patients who were treated were retrospectively reviewed, including 34 patients with benign soft tissue tumors and 85 patients with STS from 2002 to 2016, and were enrolled in this study. Patients who had local recurrence or who were referred for additional resection after inadequate resection in a previous hospital or who had distant metastasis at the first visit were excluded from this study. This study was approved by the Ethics Committee of the Mie University Graduate School of Medicine. All procedures performed in studies involving human participants were in accordance with the ethical standards of the Ethics Committee of Mie University and with the 1975 Declaration of Helsinki. The histopathological diagnosis and histological grade were verified by independent pathologists.

sCD80 measurement

Blood samples from all patients were obtained prior to initial treatment. All samples were stored at -80 degrees until measurement followed by centrifugation at 1000 × g for 15 min. sCD80 levels were measured using Human CD80 ELISA Kit (B7-1) (Abcam, Cambridge, UK). The minimum detectable level of sCD80 was 31 pg/mL; values under the detectable level were assigned a value of 0 pg/mL.

Statistical analysis

Statistical analysis was performed to compare the serum sCD80 levels to various clinical parameters using the Mann–Whitney U test or the Kruskal–Wallis test for quantitative data. To evaluate the threshold for detecting recurrence, metastasis, or death due to disease, receiver operating characteristic (ROC) curve analysis was performed. The ROC curves were created by plotting sensitivity on the y-axis and the false-positive rate (1-specificity) on the x-axis, and the area under the curve (AUC) was assessed. Local recurrence-free survival (LRFS) was defined as the time from the initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from the initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from the initial treatment to the date of death attributed to the neoplasm. Kaplan–Meier survival plots and log-rank tests were used to assess the differences of LRFS, MFS, and OS. The correlation between immunostaining and sCD80 test results was evaluated by the kappa coefficient test. To adjust for the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered significant. The EZR software program was used for statistical analyses.

Results

Characteristics of the study population

The clinical and pathological characteristics of the study population are summarized in Table 1.

Table 1 Characteristics of patients with soft tissue tumors

Age was significantly different between healthy volunteers, the patients with benign tumors, and the patients with STS. There was no significant difference in sCD80 levels between healthy volunteers, the patients with benign tumors, and the patients with STS. The histopathological diagnosis of the 34 benign tumors was 12 lipomas, 14 schwannomas, 3 myxomas, 2 tenosynovial giant cell tumors, 1 leiomyomas, and 1 others, while those of the 85 STSs were 38 liposarcomas (22 well-differentiated liposarcomas (WLSs), 12 dedifferentiated liposarcomas (DLSs), and 4 myxoid liposarcomas (MLSs)), 13 myxofibrosarcomas (MFSs), 12 undifferentiated pleomorphic sarcomas (UPSs), 8 leiomyosarcomas (LMSs), 5 synovial sarcomas (SSs), 4 malignant peripheral nerve sheath tumors (MPNSTs), and 5 others (Table 2). All patients with benign tumors underwent tumor resection, and 85 patients with STSs received treatment (wide resection in 57 patients, marginal resection in 23 patients, intralesional resection in 3 patients, and ion beam radiotherapy in 2 patients) (Table 3). Although male patients over 60 years old and those with a history of other malignant tumors had higher sCD80 levels, there was only a significant difference in sCD80 levels for age in benign and STS patients (Table 1).

Table 2 Histological classification of tumors
Table 3 Characteristics of patients with STS. sCD80 values were compared for each parameter in STS patients

Characteristics of the STS population

The clinical and pathological characteristics of the STS patients are shown in Table 3. There was a significant difference in sCD80 levels for only age. By histopathological subgroups, the average (standard deviation) of sCD80 levels was as follows: MPNST 1033.6 (495.5) pg/mL; MFS 856.2 (734.0) pg/mL; UPS 476.7 (314.0) pg/mL; SS 586.1 (616.9) pg/mL; WLS 604.5 (571.6) pg/mL; DLS 497.0 (284.7) pg/mL; MLS 394.2 (218.1) pg/mL; LMS 631.5 (320.5) pg/mL; and others 403.9 (230.8) pg/mL (Fig. 1). According to the AJCC classification of STSs, 26 patients were classified as stage I, 15 were classified as stage II, and 44 were classified as stage III. The average of sCD80 concentrations tended to be higher with higher stages than with lower stages, but the difference was not significant.

Fig. 1
figure 1

Scatter plot showing the sCD80 values for each histology

Recurrence, metastasis, and death of disease in the STS group

The mean follow-up in malignant patients was 40 months (range 0.6–208 months). During the period of this study, 23 patients developed recurrence, 36 patients developed metastasis, and 25 patients died of disease.

To confirm the diagnostic accuracy of sCD80 for identifying metastasis, DOD, and recurrence, ROC analysis was performed by evaluating the area under the curve (AUC). The AUCs for identifying DOD, metastasis, and recurrence were 0.641 (95%CI 0.514–0.767), 0.625 (95%CI 0.49–0.761), and 0.50 (95%CI 0.362–0.639), respectively (Fig. 2A–C). Based on the ROC analysis, a cutoff value of 404 pg/mL was used to divide the groups into low (≤ 404 pg/mL) and high (> 404 pg/mL) sCD80 groups for 5OS. High group had a poorer 5OS than low group (Fig. 3A, low sCD80: 89.5%, high sCD80 65.0%, p = 0.015). Like 5OS, to divide sCD80 into two groups for 5MFS, a cutoff value of 531 pg/ml was used to divide the groups into low (≤ 531 pg/ml) and high (> 531 pg/ml) sCD80 groups. The high group had significantly lower 5MFS (Fig. 3B, low sCD80: 75.3%, high sCD80: 44.0%, p = 0.016).

Fig. 2
figure 2

Receiver operating characteristic curve analysis. Diagnostic accuracy is evaluated by the area under the curve for identifying DOD (A AUC: 0.625, 95% CI 0.49–0.761), metastasis (B AUC: 0.641, 95% CI 0.514–0.767), and recurrence (C AUC: 0.50, 95% CI 0.362–0.639). A cutoff of 404 pg/mL results in sensitivity of 84.2% and specificity of 42.4% for identifying DOD, a cutoff of 531 sensitivity of 58.6% and specificity of 71.4% for identifying metastasis, and a cutoff of 284 sensitivity of 85.0% and specificity of 26.2% for identifying recurrence

Fig. 3
figure 3

Kaplan–Meier analysis for STS. 5OS (A) and 5MFS (B) compared in the low- and high-sCD80 groups are shown by Kaplan–Meier analysis. The high-sCD80 group has significantly lower 5MFS (low sCD80 75.3%, high sCD80 44.0%, p = 0.016). For 5OS, the high-sCD80 group has a significantly worse prognosis (low sCD80 89.5%, high sCD80 65.0%, p = 0.015). The x-axis indicates months

In the same way, to divide sCD80 into two groups for 5LRFS, ROC analysis was performed. As AUC was 0.5 for detecting recurrence, further analysis was not evaluated.

sCD80 for high-grade STS

Next, we excluded 23 patients with well-differentiated liposarcoma from our cohort due to their excellent prognosis and examined the relationship between sCD80 value and survival in the remaining 59 patients. The median sCD80 value was 496.4 pg/mL (mean = 611.0, range = 48.8–2947.4) in 59 patients. During the period of this study, 20 patients developed recurrence, 36 patients developed metastasis, and 25 patients died of disease. Like above, to divide sCD80 into two groups for 5OS and 5MFS, a cutoff value of 404 and 531, respectively (Fig. 4A, B.), was used to divide the groups into low- and high-sCD80 groups (Fig. 5A, B). The results of 5LRFS were excluded because the AUC of 5LRFS was low (AUC 0.525) (Fig. 4C).

Fig. 4
figure 4

Receiver operating characteristic curve analysis. Diagnostic accuracy is evaluated by the area under the curve for identifying DOD (A AUC: 0.621, 95% CI 0.467–0.775), metastasis (B AUC: 0.661, 95% CI 0.518–0.805), and recurrence (C AUC: 0.525, 95% CI 0.364–0.686). Excluded the results of 5LRFS because the AUC of 5LRFS was low (AUC 0.525). A cutoff of 404 pg/mL results in sensitivity of 84.2% and specificity of 40.0% for identifying DOD and a cutoff of 531 sensitivity of 58.6% and specificity of 76.7% for identifying metastasis

Fig. 5
figure 5

Kaplan–Meier analysis for STS. 5OS (A) and 5MFS (B) compared in the low- and high-sCD80 groups are shown by Kaplan–Meier analysis in patients with high-grade STS. 5OS shows no significant difference (5OS: low sCD80 83.0%, high sCD80 56.3%, p = 0.617). The high-sCD80 group has significantly lower 5MFS (low sCD80 62.9%, high sCD80 26.5%, p = 0.017). The x-axis indicates months

High group had tend to be a poorer 5OS than low group (low sCD80: 83.0%, high sCD80 56.3%, p = 0.062). The high group had significantly lower 5MFS (low sCD80: 62.9%, high sCD80: 26.5%, p = 0.022). (Fig. 5A, B). In multivariate Cox proportional hazard analysis, sCD80 showed significant difference for 5MFS (HR:2.397, 95% CI 1.143–5.027; p = 0.021) (Table 4).

Table 4 Prognostic factors for 5-year metastasis-free survival (5MFS) in 59 patients with high-grade soft tissue sarcoma

Discussion

Cancer cells, in order to avoid attacks from immune, aggressively use immunosuppressive function of regulatory T cells, bone marrow-derived suppressor cells, and the ICP.

Originally, ICP exists to suppress excessive activation of T cell and not to attack itself, but in the carcinogenic process, cancer cells are used to avoid attack from the immune system and proliferate. Currently, various immune checkpoint molecules and their ligands have been identified. After anti-CTLA4 antibody has been approved, it is becoming a new standard treatment for many cancer types such as melanoma and non-small cell lung cancer [16]. There are also some reports of clinical trials of checkpoint inhibitors in patients with STS and reports of phase II trials of ipilimumab, an anti-CTLA4 inhibitor, in patients with recurrent synovial sarcoma [17]. Despite high expression of CT (cancer–testis) antigens by synovial sarcomas of patients treated in the study, there was neither clinical benefit nor evidence of anti-CT antigen serological responses. In the other phase II trial, the response rate of the combination therapy of nivolumab and ipilimumab was higher (16%) than monotherapy of nivolumab (5%). This mean CTLA-4 is involved in tumor exacerbation of STS [18].

In this study, we focused on CD80, which is a suppressor ligand for CTLA4. CD80 is a transmembrane glycoprotein, a co-stimulator expressed on the surface of activated monocytes, B cells, and T cells [19, 20]. It has been reported that CD80 is also expressed in tumor cells and glomerular epithelial cells [21, 22].

The two B7 family molecules CD80 (B7-1) and CD86 (B7-2) are positioned as co-stimulatory molecules that play the most important role in T cell activation. CD80/86 activates T cells by binding to CD28, which is expressed on T cells; on the other hand, CTLA4 expressed on activated T cells and Treg cells binds to CD80/CD86 with a stronger affinity than CD28 and inhibits T cell activation. In addition, CTLA4 removes CD80/CD86 from APC (antigen-presenting cell), thereby inhibiting co-stimulation signals and suppressing T cell activation [19].

In these days, soluble form of ICPs attracts attention. High level of circulating soluble PD-L1 is significantly correlated with poor prognosis of many malignant neoplasm including STS [1, 9,10,11,12,13,14,15]. Because soluble PD-L1 functionally suppresses systemic immune response, therapeutic targets have been expanding from cellular PD-L1 to cellular and circulating soluble PD-L1 [23]. In this study, we analyzed the relation between soluble CD80 and prognosis in STS patients to present a hypothesis that soluble CD80 may affect for immune system in STS.

There are three reports of the origin of sCD80: shedding of CD80 on the cell membrane [24], CD80 on microvesicles released from cells [25], and splicing variants of CD80 [17]. Of the several splicing variants, the deleted form of the transmembrane domain is released unstable on the cell membrane, resulting in sCD80.

Functionally, the bifacial effect of sCD80 on tumor behavior has been reported. In one aspect, sCD80 inhibits T cell activation by preferentially binding to CTLA4 [19] and elevated levels of sCD80 have been a poor prognosis in patients with blood cancer and prostate cancer [25]. In another aspect, sCD80 inhibits the suppression of T cell activation [26], induces T cell proliferation [27], and inhibits PD-L1/PD1-mediated immunosuppression [28]. In this study, we successfully demonstrated that higher sCD80 was associated with shortened survivals and metastasis-free survival. sCD80 may affect immune system suppressively in STS. To date, seven clinical trials of galiximab, an anti-CD80 antibody, have been conducted (https://clinicaltrials.gov). In one of the lymphoma clinical trials, it has been reported that 49% of patients showed a decrease in tumor volume after administration of galiximab, and there were no grade 4 adverse events or treatment-related deaths [29]. In this study, the high sCD80 value had a poor prognosis, so it is possible that the effect of galiximab can be expected even in the STS region.

Limitations

This study has some limitations. The number of patients was few. STS is rare cancer, a heterogeneous group of tumors. The incidence of STS is fewer than six per 100,000 cancer cases, which represents 1–2% cases of all cancer adults [30]. Therefore, it is difficult to collect samples and may be difficult to decide its cutoff value. In this study, the cutoff value was decided by the ROC curve. More samples may give us a more correct cutoff value of sCD80 and decide the cutoff value in STS. We believe that the level of sCD80 could be a useful prognostic marker in patients with STS.

Availability of data and materials

Not applicable.

Abbreviations

ICP:

Immune checkpoint protein

sCD80:

Soluble form of CD80

STS:

Soft tissue sarcoma

MS:

Metastasis-free survival

OS:

Overall survival

sPD-L1:

Soluble form of PD-L1

ROC:

Receiver operating characteristic

AUC:

Area under the curve

LRFS:

Local recurrence-free survival

WLS:

Well-differentiated liposarcoma

DLS:

Dedifferentiated liposarcoma

MLS:

Myxoid liposarcoma

MFS:

Myxofibrosarcoma

UPS:

Undifferentiated pleomorphic sarcoma

LMSs:

Leiomyosarcoma

SS:

Synovial sarcoma

MPNST:

Malignant peripheral nerve sheath tumor

DOD:

Died of disease

References

  1. Bourcier K, Le Cesne A, Tselikas L, et al. Basic knowledge in soft tissue sarcoma. Cardiovasc Intervent Radiol. 2019;42(9):1255–61.

    Article  Google Scholar 

  2. Sasaki H, Nagano S, Komiya S, et al. Validation of different nutritional assessment tools in predicting prognosis of patients with soft tissue spindle-cell sarcomas. Nutrients. 2018;10(6):765.

    Article  Google Scholar 

  3. Ogura K, Higashi T, Kawai A. Statistics of soft-tissue sarcoma in Japan: report from the bone and soft tissue tumor registry in Japan. Cancer. 2009;115:5243–50.

    Article  Google Scholar 

  4. Stojadinovic A, Leung DH, Hoos A, et al. Analysis of the prognostic significance of microscopic margins in 2,084 localized primary adult soft tissue sarcomas. Ann Surg. 2002;235(3):424–34.

    Article  Google Scholar 

  5. Julian AM, Bhagirathbhai D, Aixa ES, et al. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J HematolOncol. 2018;11(1):39.

    Google Scholar 

  6. Perisano C, Vitiello R, Sgambato A, et al. Evaluation of PD1 and PD-L1 expression in high-grade sarcomas of the limbs in the adults: possible implications of immunotherapy. J Biol Regul Homeost Agents. 2020;34(4 Suppl. 3):289–94.

    CAS  PubMed  Google Scholar 

  7. Gang C, Alexander CH, Wei Z, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1response. Nature. 2018;560(7718):382–286.

    Article  Google Scholar 

  8. Finkelmeier F, Canli O, Tal A, et al. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer. 2016;59:152–9.

    Article  CAS  Google Scholar 

  9. Frigola X, Inman B, Lohse C, et al. Identification of a soluble form of B7–H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res. 2011;17:1915–23.

    Article  CAS  Google Scholar 

  10. Takahashi N, Iwasa S, Sasaki Y, et al. Serum levels of soluble programmed cell death ligand 1 as a prognostic factor on the first-time treatment of metastatic or recurrent gastric cancer. J Cancer Res Clin Oncol. 2016;142:1727–38.

    Article  CAS  Google Scholar 

  11. Yoshida J, Ishikawa T, Doi T, et al. Clinical siginificance of soluble forms of immune checkpoint molecules in advanced esophageal cancer. Med Oncol. 2019;36:60.

    Article  Google Scholar 

  12. Shen H, Ji Y, Zhang Y, et al. Soluble programmed death-ligand 1 are highly expressed in peripheral T-cell lymphoma: a biomarker for prognosis. Hematology. 2019;24:392–298.

    Article  CAS  Google Scholar 

  13. Okuma Y, Hosomi Y, Nakahara Y, et al. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer. 2017;104:1–6.

    Article  Google Scholar 

  14. Tominaga T, Akiyoshi T, Yamamoto N, et al. Clinical significance of soluble programmed cell death-1 and soluble programmed cell death-ligand 1 in patients with locally advanced rectal cancer treated with neoadjuvant chemoradiotherapy. PLoS ONE. 2019;14: e0212978.

    Article  CAS  Google Scholar 

  15. Kunihiro A, Tomoki N, Akinobu H, et al. Soluble programmed death-ligand 1 rather than PD-L1 on tumor cells effectively predicts metastasis and prognosis in soft tissue sarcomas. Sci Rep. 2020;10:9077.

    Article  Google Scholar 

  16. Khan S, Gerber DE, et al. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: a review. Semin Cancer Biol. 2020;64:93–101.

    Article  CAS  Google Scholar 

  17. Robert G. M, Achim A. J, Sacha G, et al. A pilot study of anti-CTLA4 antibody ipilimumab in patients with synovial sarcoma. Sarcoma. 2013;168145.

  18. D’ Angelo SP, Mahoney MR, VanTine BA, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2019; 19(3): 416–426.

  19. Kakoulidou M, Giscombe R, Zhao X, et al. Human soluble CD80 is generated by alternative splicing, and recombinant soluble CD80 binds to CD28 and CD152 influencing T-cell activation. Scand J Immunol. 2007;66:529–37.

    Article  CAS  Google Scholar 

  20. Gu D, Ao X, Yang Y, et al. Soluble immune checkpoints in cancer: production, function and biological significance. J Immunothere Cancer. 2018;6:132.

    Article  Google Scholar 

  21. Dan D, Tuula S, Sirpa S, et al. Molecular crosstalk between cancer cells and tumor microenvironment components suggests potential targets for new therapeutic approaches in mobile tongue cancer. Cancer Med. 2012;1(2):128–40.

    Article  Google Scholar 

  22. Garin EH, Reiser J, Cara-Fuentes G, et al. Case series: CTLA4-IgG1 therapy in minimal change disease and focal segmental glomerulosclerosis. Pediatr Nephrol. 2015;30:469–77.

    Article  Google Scholar 

  23. Sergio AM, Javier DQ, Jose GV, et al. Clinical implications of exosomal PD-L1 in cancer immunotherapy. J Immunol Res. 2021 8839978.

  24. Garin EH, Mu WM, Arthur J, et al. Urinary CD80 is elevated in minimal change disease but not in focal segmental glomerulosclerosis. Kidney Int. 2010;78(3):296–302.

    Article  CAS  Google Scholar 

  25. Esser MT, Graham DR, Coren LV, et al. Differential incorporation of CD45, CD80 (B7–1), CD86 (B7–2), and major histocompatibility complex Class I and II molecules into human immunodeficiency virus type 1 virions and microvesicles: implications for viral pathogenesis and immune regulation. J Virology. 2001;75(13):6173–82.

    Article  CAS  Google Scholar 

  26. Hock BD, Starling GC, Patton WN, et al. Identification of a circulating soluble form of CD80: levels in patients with hematological malignancies. Leuk Lymphoma. 2004;45(10):2111–8.

    Article  CAS  Google Scholar 

  27. He W, Hu ZB, Liu F, et al. In vitro co-stimulation of anti-tumor activity by soluble B7 molecules. Acta Biochim Pol. 2006;53(4):807–13.

    Article  CAS  Google Scholar 

  28. Sturmhoefel K, Lee L, Gray GS, et al. Potent activity of soluble B7-IgG fusion proteins in therapy of established tumors and as vaccine adjuvant. Cancer Res. 1999;59(19):4964–72.

    CAS  PubMed  Google Scholar 

  29. Czuczman MS, Thall A, Witzig TE, et al. Phase I/II Study of Galiximab, an Anti-CD80 antibody, for relapsed or refractory follicular lymphoma. J Clin Oncol. 2005;23(19):4390–8.

    Article  CAS  Google Scholar 

  30. Bourcier K, Le Cesne A, Tselikas L, et al. Basic knowledge in soft tissue sarcoma. Cardiovasc Intervent Radioll. 2019;42(9):1255–61.

    Article  Google Scholar 

Download references

Acknowledgements

None.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

YM performed the experiment, validation, formal analysis, investigation, and visualization, curated the data, and wrote the original draft of the manuscript. KA conceptualized and supervised the study, performed the methodology, validation, formal analysis, investigation, and visualization, collated the resources, curated the data, wrote the original draft of the manuscript, and conducted the project administration. KY, TH, and TI performed the experiment and validation. TN performed the validation and curated the data. AS wrote, reviewed, and edited the manuscript and acquired the funding. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Kunihiro Asanuma.

Ethics declarations

Ethics approval and consent to participate

All experiments were performed in accordance with the interdisciplinary principles and guidelines for the use of animals in research, testing, and education. The study was approved by the Ethics Committee of author’s affiliated institution.

Competing interests

The authors declare that they have 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

Matsuyama, Y., Asanuma, K., Yoshida, K. et al. The role of soluble CD80 in patients with soft tissue tumors. J Orthop Surg Res 17, 404 (2022). https://doi.org/10.1186/s13018-022-03283-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13018-022-03283-2

Keywords