We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Treatment patterns and outcomes in patients with metastatic synovial sarcoma in France, Germany, Italy, Spain and the UK

    Nashita Patel

    Global Value, Evidence & Outcomes, GSK, Brentford, London, TW8 9GS, UK

    ,
    Shibani Pokras

    *Author for correspondence: Tel.: +1 571 332 9870;

    E-mail Address: shibani.x.pokras@gsk.com

    Value Evidence & Outcomes, GSK, Collegeville, PA 19426, USA

    ,
    Jane Ferma

    Data Science & Advanced Analytics, IQVIA, London, N1 9JY, UK

    ,
    Vicky Casey

    Data Science & Advanced Analytics, IQVIA, London, N1 9JY, UK

    ,
    Fil Manuguid

    Data Science & Advanced Analytics, IQVIA, London, N1 9JY, UK

    ,
    Ken Culver

    Global Medical Affairs, GSK, Collegeville, PA 19426, USA

    &
    Sebastian Bauer

    Department of Medical Oncology & Sarcoma Center, University Hospital, University of Duisburg-Essen, Essen, 45147, Germany

    Published Online:https://doi.org/10.2217/fon-2022-1005

    Abstract

    Aim: Describing the treatment patterns, outcomes by line of treatment (LOT), and healthcare resource utilization (HCRU) in patients with metastatic synovial sarcoma (mSS). Patients & methods: In this descriptive, non-interventional, retrospective cohort study, physicians from five European countries reported on patients with recent pharmacological treatment for mSS. Results: Among 296 patients with mSS, 86.1, 38.9 and 8.4% received 1 LOT (1L), 2 LOTs (2L) and 3+ LOTs (L3+), respectively. Common regimens were doxorubicin/ifosfamide-based (37.4%) for 1L and trabectedin-based for 2L (29.7%). For 1L, median time to next treatment was 13.1 and 6.0 months for living and deceased patients, respectively. Median OS was 22.0, 6.0 and 4.9 months in all patients, 2L and 3L, respectively. HCRU data showed median one inpatient hospital admission, 3 days in hospital and four outpatient visits yearly. Conclusion: This large-scale study documents high unmet needs in patients previously treated for mSS and for more effective therapies.

    Synovial sarcoma (SS) is a rare and aggressive subtype of soft tissue sarcoma (STS), accounting for ≈5–10% of cases [1]. The estimated age-standardized STS incidence is 3.3–4.7 cases/100,000 individuals in Europe [2]; 993 incident SS cases were estimated in 2019 for European Union-28 countries [3]. A recent French study, based on expert pathology review of cases from 2013 to 2016, reported an incidence rate of 1,674/1,000,000/year [4]. Metastatic disease is present in ≈24% of patients at diagnosis and 5070% of patients with localized disease will develop metastatic SS (mSS). Survival prospects for mSS are poor, with a 5-year survival rate of ≈10% [1].

    The Clinical Practice Guidelines of the European Society for Medical Oncology–European Reference Network for rare adult solid cancers cover the treatment of STSs, but due to the rarity of the disease, there are no specific guidelines for SS alone [5]. In Europe, surgical resection with or without radiotherapy is the standard treatment for localized SS [5]. A joint prospective analysis showed that patients with adequately resected ≤5 cm SS, regardless of grade, can be safely treated with a surgery-only approach. Avoiding the use of adjuvant chemotherapy and radiotherapy in this low-risk patient population may decrease both short- and long-term morbidity and mortality [6]. For low-risk patients, surgery can be enough; however, for high-risk patients, neoadjuvant chemotherapy is standard first-line care. In adult patients, neoadjuvant chemotherapy is primarily offered to patients with high risk of mSS [7,8].

    In patients with advanced (unresectable or mSS), standard first line of treatment (LOT) consists of anthracycline chemotherapy (monotherapy or combination) [5]. Given the high activity of ifosfamide (IFOS) in SS, most centers prefer IFOS/anthracycline combinations or IFOS as front-line therapy [8]. Clinical evidence guiding treatment options for patients with advanced/mSS who progress following anthracycline-based chemotherapy is lacking [5]. There is no standard-of-care chemotherapy in later-line settings [9], and while some studies have reported a degree of benefit in approved treatments for STS following progression on chemotherapy (such as pazopanib and trabectedin) [10–15], a recent meta-analysis indicates a lack of consistent evidence supporting a positive impact on overall survival (OS) [16].

    Because patients with metastatic relapse of SS show significant morbidity and mortality with little opportunity for cure [7,17], it is essential to characterize treatment patterns, understand their real-world effectiveness, and examine the impact of disease on healthcare resource utilization (HCRU) [14,18,19]. In a chart review of patients with advanced STS in four European countries (the UK, France, Germany and Spain), the subpopulation of patients with SS comprised only 6% of the overall population, and the most common regimens used (and their duration), OS and overall response rates were only available for first LOT (1L) patients [18]. Typically, details on LOT 2 (2L)+ treatment patterns or HCRU associated with SS are lacking across published studies [9,18,19].

    The aim of this study was to describe the real-world burden of illness in patients with mSS in five European countries (France, Germany, Italy, Spain and the UK), including patient characteristics, treatment patterns and outcomes by LOT, and HCRU.

    Methods

    Study design & objectives

    This was a descriptive, non-interventional, retrospective cohort study of patients with mSS in France, Germany, Italy, Spain and the UK (Figure 1). Primary objectives were to describe the demographic and clinical characteristics of treated patients with mSS and to summarize their treatment patterns and outcomes. A secondary objective was to describe annualized HCRU in the period after diagnosis, stratified by country.

    Figure 1. Study design.

    LOT: Line of treatment; OA: Oncology advantage; mSS: Metastatic synovial sarcoma.

    The study employed a customized data collection effort from Oncology Advantage (OA), an online physician survey reporting on a consecutive set of patients receiving therapy and their previous treatment history, by specific tumor cohort. OA targets large ‘center of excellence' hospitals and smaller local sites, to be representative of real-world treatment patterns and care settings in each country. The survey collected anonymous patient-level data from a specific time point (not designed to capture the same patients prospectively). Physicians reported on their most recent 2–5 consecutive patients treated for mSS in the past 12 months.

    Eligibility criteria

    Physicians eligible to complete the survey were: board certified ≥3 years; currently or previously initiated or recommended/prescribed treatment for patients with SS; seen ≥2 adult patients with SS in the past 12 months; and able to provide details of cytotoxic and/or targeted therapies prescribed for SS. Physicians were considered eligible if they spent >40% of their time seeing patients and >70% in a hospital setting (all other countries), or office-based setting (Germany only). The German healthcare system is organized differently compared with other European countries in that most patients, depending on disease, are treated at office-based sites rather than at main hospitals.

    Eligible patients were: diagnosed with mSS by case definition; ≥18 years of age at the time of data collection; positive for chromosomal translocation between SYT on the X chromosome and SSX1, SXX2, or SSX4 on chromosome 18, or had unknown chromosomal translocation status (i.e., the physician was uncertain whether the patient was tested); alive or died ≤12 months prior to the time of data collection, irrespective of last LOT; if living, currently receiving cytotoxic and/or targeted therapies, or received cytotoxic and/or targeted therapies ≤12 months prior to the time of data collection; or if deceased, received ≥2 cytotoxic and/or targeted therapies ≤12 months prior to the time of data collection.

    Study dates & time periods

    The data collection period for which physicians were surveyed began on 29 August 2019, and ended on 20 November 2019, or sooner if the desired patient quotas per country were fulfilled prior to this date. Physicians were instructed to evaluate all available medical history information when completing the survey and to include information on eligible patients treated most recently ≤12 months prior to the time of data collection.

    The index date was the date of mSS diagnosis. The LOT start date was the date of the first dose of a specified LOT. The study inclusion date was the date of survey completion. Date of death was recorded at month and year level, so date of death was defined as the mid-point of the month at which patient death was recorded. For patients with a primary diagnosis of localized SS prior to the index date, the time from diagnosis to the index date, and prior LOTs received for localized SS, were also documented.

    Follow-up & censoring

    For living patients who had an end date recorded for their most recent LOT, follow-up time started at index date (inclusive) and at the end date (inclusive) of the most recent LOT (recorded closest in time to patient's study inclusion date). For living patients and whose most recent LOT was recorded as ‘ongoing’, or for patients without an end date of their most recent LOT prior to study inclusion date, follow-up time was censored at study inclusion date (inclusive). For deceased patients, follow-up time started at index date (inclusive) and ended at date of death (inclusive).

    Study assessments

    Physician & patient characteristics

    Physician characteristics were described overall and by country, in terms of medical specialty, type of work setting, extent of clinical experience and number of patients treated. Patient baseline demographic and clinical characteristics were summarized overall, by country and by LOT.

    Treatment patterns

    Common treatments were assessed in terms of the proportion of patients in the five countries overall receiving each type of treatment regimen within their current/most recent LOT and current/historic LOT, defined as having a start date closest to the date survey completion or patient's death.

    Treatment outcomes

    Time-to-next-treatment (TTNT) was summarized for the five countries overall and by country. OS was described for the five countries overall. TTNT was defined as the time from the first dose of a given LOT (start date of given LOT; inclusive) until the first dose of the next distinct LOT (start date of next consecutive LOT; exclusive). Patients were considered to have experienced an event if they progressed to the next LOT from the given LOT.

    Patients may not have progressed to their next LOT due to possible ‘censoring' events occurring prior to start date of the next LOT; for living patients (most recent LOT), TTNT was censored at study inclusion date if LOT was ongoing (exclusive: right censoring/end of follow-up). For deceased patients (most recent LOT), TTNT was censored at date of death (exclusive).

    OS was derived within the combined living and deceased patient cohorts. Time to death (in days) was derived for the deceased cohort from the index date (inclusive) to date of death (exclusive). Time to death (OS) from index date (inclusive) for living patients was censored at study inclusion date (exclusive), as future information on date of death for these patients was not collected. Only month and year of death were recorded; therefore, the calculation of OS used date of death defined as the mid-point of the month at which patient death was recorded. Patients who died between the index date and study inclusion date were considered to have experienced an event.

    HCRU

    Annualized HCRU was investigated in the period after mSS diagnosis and stratified by country, including inpatient hospital admissions, total days of inpatient hospitalizations and outpatient visits.

    Data analyses

    Descriptive analyses were conducted for patient demographic and clinical characteristics, treatment patterns, and HCRU.

    Each LOT was defined for chemotherapy/targeted treatments only (surgery and radiotherapy were excluded). Common treatment regimen patterns were reported for current/most recent and current and historic LOTs combined overall and by country. Percentages may not sum to 100% because some patients received no therapy while others received multiple lines of therapy. All 2L and 3L patients are also included in the 1L group.

    The number of HCRU events was reported by the surveyed physician for each patient over their follow-up time. Annualized HCRU for each variable was described continuously and reported for the five countries overall and by country. Annualized rate of HCRU was derived via the equation:

    Number of reported eventsPatient follow  up time×365.25

    Results

    Patient & physician characteristics

    Overall, 296 patients with mSS were included from France (14.9%), Germany (18.6%), Italy (22.3%), Spain (22.3%) and the UK (22.0%; Table 1). At the time of data abstraction, 68.6% of patients were living (median age: 54 years [range: 18–89]). The median number of metastases/patient was 4 (range: 1–30), with lung/respiratory system as the most common metastatic sites (77.7%). Most patients (76.4%) had a primary diagnosis of mSS. Among patients with a primary diagnosis of localized SS (23.6%), median time from diagnosis to index date overall was 502 days (range: 0–6760; Supplementary Table 1).

    Table 1. Patient demographics and clinical characteristics at index (diagnosis of mSS) for the five countries overall and by country.
    VariableMeasureOverall (n = 296)France (n = 44)Germany (n = 55)Italy (n = 66)Spain (n = 66)UK (n = 65)
      n%n%n%n%n%n%
    PatientsLiving cohort20368.63579.54072.74162.14365.24467.7
    Deceased cohort9331.4920.51527.32537.92334.82132.3
    SexMale19666.22965.93665.55075.84162.14061.5
    Female10033.81534.11934.51624.22537.92538.5
    Age at index date, yearsMedian (range)54 (18–89)55 (29–79)59 (18–84)53 (22–80)54 (19–79)46 (18–89)
    Age group at index date≤40 years5819.6511.41018.2812.11116.72436.9
    65 years16856.82965.92341.84669.73857.63249.2
    >65 years7023.61022.72240.01218.21725.8913.8
    Primary diagnosisLocalized disease7023.61227.31527.3913.61725.81726.2
    Metastatic disease22676.43272.74072.75786.44974.24873.8
    Type of mSS diagnosisNOS14448.61329.53054.52639.43857.63756.9
    Spindle cell8227.71943.21730.91624.21624.21421.5
    Epithelioid cell4214.21522.769.1
    Biphasic289.5913.669.1
    Other00
    Tumor grading at index dateG1103.400
    G29532.11840.91425.52233.32030.32132.3
    G318361.82659.13767.34365.23654.54163.1
    Gx82.70000
    Tumor size at index date, cm0.1–5 cm12742.92647.32334.83756.12436.9
    5.1–10 cm14047.32556.81934.53553.03553.8
    >10 cm299.81018.2812.169.2
    Site of SS metastases at index dateLung/respiratory23077.73477.34683.64872.75177.35178.5
    Bone and articular cartilage8027.0818.22240.01928.82131.81015.4
    Liver/biliary tract6321.31125.01221.81827.31218.21015.4
    Mesothelial and soft tissue3612.21323.61015.2812.3
    Brain/meninges/CNS217.169.1
    Head and neck93.0
    Stomach/digestive tract72.400.0
    All others103.400.0712.700.0
    Number of metastases per patient (most recent)n, %12441.92965.91018.22943.92842.42843.1
    Mean (SD)5.0 (4.1)5.0 (3.7)8.6 (3.5)3.2 (1.6)4.1 (3.0)6.2 (6.0)
    Median (range)4 (1–30)4 (1–20)8 (5–17)3 (1–8)3 (1–12)5 (1–30)
    CCI (at index date)010334.81636.42443.61218.22943.92233.8
    110535.51431.8814.53654.52233.32538.5
    24615.5712.71218.269.11218.5
    ≥34214.21629.169.1913.669.2
    Employment status (most recent)Employed9030.61534.11323.62842.41827.31624.6
    Unemployed5418.21534.11421.21116.71218.5
    Unable to work due to mSS6220.91425.5710.61319.72741.5
    Retired5819.6920.52138.269.11421.2812.3
    Smoking status (most recent)Smoker6522.01534.11120.02030.3
    Ex-smoker12341.62147.71934.52842.43248.52335.4
    Never smoked9030.41832.72030.33046.2
    Unknown186.1712.7
    Chromosomal translocation t(X;18)(p11.2;q11.2)Positive11739.51527.34163.1
    Awaiting results134.4
    Not tested16656.12250.03767.34263.64263.6
    Time (days) between index date and date of chromosomal translocation gene testingMean (SD)10.5 (56.8)29.7 (119.3)0 (0)10.0 (29.1)3.9 (14.3)9.4 (47.3)
    Median (range)0 (0–545)0 (0–545)0 (0–0)0 (0–133)0 (0–65)0 (0–302)

    †Index date is the date of diagnosis of metastatic disease.

    CCI: Charlson Comorbidity Index; mSS: Metastatic synovial sarcoma; NOS: Not otherwise specified.

    The majority (86.1%) received 1 LOT; 38.9% received 2 LOTs and 8.4% received 3+ LOTs (Supplementary Table 2). A total of 70.2% 1L, 51.3% 2L and 48.0% of LOT 3 (3L)+ patients had an Eastern Cooperative Oncology Group performance status (ECOG PS) of 0–1. A larger proportion of 2L (47.8%) and 3L+ (52.0%) patients had an ECOG PS ≥2 compared with 1L patients (27.5%).

    Table 2. TTNT by LOT for the five countries, overall and by country.
    LOTMeasureOverall (n = 296)France (n = 44)Germany (n = 55)Italy (n = 66)Spain (n = 66)UK (n = 65)
      n%n%n%n%n%n%
    1L, living patients onlyPatients starting LOT20368.63579.54072.74162.14365.24467.7
    Events (progressed to next LOT)4314.512.31527.3812.11015.2913.8
    Censored16054.13477.32545.53350.03350.03553.8
    Median TTNT, months (95% CI)13.1
    (10.3, 17.7)
    NR
    (11.3, N/A)
    9.9
    (9.3, N/A)
    11.1
    (10.1, N/A)
    14.7
    (8.8, N/A)
    16.5
    (10.3, N/A)
    IQR9.1–18.011.3–N/A9.2–17.76.5–17.38.8–18.010.3–20.9
    Min1.51.92.71.52.42.5
    Max21.312.318.017.321.320.9
    1L, deceased patients onlyPatients starting LOT9331.4920.51527.32537.92334.82132.3
    Events (progressed to next LOT)7926.7613.61323.62233.32233.31624.6
    Censored144.736.823.634.511.557.7
    Median TTNT, months (95% CI)6.0
    (4.9, 8.3)
    8.6
    (6.0, N/A)
    10.8
    (8.3, N/A)
    3.2
    (1.0, 7.0)
    4.8
    (3.2, 10.5)
    6.9
    (4.3, N/A)
    IQR2.8–11.16.0–19.18.3–13.50.8–7.02.8–10.52.8–12.9
    Min00.51.400.50
    Max27.423.720.927.415.223.7
    2LPatients starting LOT12241.2715.92850.93045.53248.52538.5
    Events (progressed to next LOT)217.112.359.134.557.6710.8
    Censored10134.1613.62341.82740.92740.91827.7
    Median TTNT, months (95% CI)12.2
    (10.7, N/A)
    5.2
    (N/A, N/A)
    10.4
    (6.9, N/A)
    11.1
    (11.0, N/A)
    17.8
    (12.2, N/A)
    20.0
    (6.2, N/A)
    IQR10.3–20.05.2–5.26.9–10.711.1–N/A12.2–18.46.3–N/A
    Min0.61.80.60.71.30.9
    Max44.05.211.113.844.022.9
    3LPatients starting LOT217.1
    Events (progressed to next LOT)72.4
    Censored144.7
    Median TTNT, months (95% CI)13.2
    (4.6, N/A)
    IQR4.2–13.2
    Min1.6
    Max13.2
    4LPatients starting LOT72.4
    Events (progressed to next LOT)20.7
    Censored51.7
    Median TTNT, months (95% CI)15.6
    (N/A, N/A)
    IQR15.6–15.6
    Min1.5
    Max15.6
    5LPatients starting LOT20.7
    Events (progressed to next LOT)00.0
    Censored20.7
    Median TTNT, months (95% CI)NR
    (N/A, N/A)
    IQRN/A–N/A
    Min2.6
    Max13.5

    When percentile is beyond survival probability it has been noted as N/A.

    †Overall data only and not by country are presented for 3L and beyond due to small sample size.

    IQR: Interquartile range; LOT: Line of treatment; N/A: Not applicable; NR: Not reached; TTNT: Time-to-next-treatment.

    Seventy-one physicians fulfilled eligibility criteria in France (14.1%), Germany (21.1%), Italy (22.5%), Spain (21.1%) and the UK (21.1%; Supplementary Table 3). Most physicians (77.5%) were medical oncologists, were from teaching or research hospitals (43.7%) and had 15–34 years of experience (57.7%). The median number of patients with mSS who were seen by physicians within the past 12 months was 12 (interquartile range [IQR] 6–30). Physicians in the UK saw the highest median (IQR) number of patients (34 [8–50]), followed by physicians in France (15.5 [10–30]), Spain (12 [6–20]), Italy (10 [3–29]) and Germany (8 [5–10]).

    Table 3. Annualized HCRU for the five countries, overall and by country.
    VariableMeasureOverall (n = 296)France (n = 44)Germany (n = 55)Italy (n = 66)Spain (n = 66)UK (n = 65)
    HCRU
    Inpatient hospital admissionsPatients, n1863733353942
    Mean (SD)2.59 (4.03)3.05 (2.75)2.03 (2.04)1.8 (4.66)1.33 (1.68)4.43 (6.05)
    Median121011.5
    IQR0–31–40–40–20–21–6
    Min000000
    Max2613822626
    Total days of inpatient hospitalizationPatients, n1863733353942
    Mean (SD)9.17 (14.98)10.03 (14.22)12.39 (16.25)3.83 (6.64)6.08 (13.78)13.19 (18.89)
    Median345005.5
    IQR0–122–100–170–80–80–16
    Min000000
    Max905268287690
    Outpatient visitsPatients, n1863733353942
    Mean (SD)7.12 (8.35)3.24 (3.3)8.82 (7.71)3.86 (3.54)10.33 (13.04)8.93 (7.22)
    Median428367
    IQR2–91–44–112–62–143–12
    Min000001
    Max501534155035

    HCRU: Healthcare resource utilization; IQR: Interquartile range.

    Treatment patterns

    The most common 1L regimens overall were doxorubicin/IFOS-based regimens (37.4%), doxorubicin monotherapy (27.0%) and IFOS-based regimens (excluding combination with doxorubicin; 14.9%; Figure 2A). Doxorubicin/IFOS-based regimens were the most common 1L regimens in France (62.2%), Germany (40.7%) and the UK (45.0%). Doxorubicin monotherapy was most common in Spain (47.1%) and IFOS-based regimens most common in Italy (33.3%). In 2L, the most common treatments overall were trabectedin-based regimens (29.7%), followed by IFOS-based (16.8%), pazopanib- or trabectedin-based (15.8%) and doxorubicin-based (13.9%) regimens. Trabectedin-based regimens were most common in the UK (50.0%), Germany (34.8%) and France (33.3%), whereas IFOS-based regimens were most common in Italy (37.0%; Figure 2B). In 3L+, trabectedin-based regimens were reported in 28.6% of patients, whereas 33.3% received ‘other' regimens (Figure 2C). Trabectedin-based regimens were most common in Germany (40.0%) and Italy (66.7%).

    Figure 2. Treatment regimen for current/most recent LOT for the five countries overall, and by country. (A) 1L, (B) 2L and (C) 3L+.

    ‘Other' refers to types of chemotherapy/targeted therapy not listed in categories shown, which included the top five regimens for 1L (EPI, DOX/OLAR_MAB, CYC, DOX/DTIC, GEM), 2L (DOC/GEM, GEM, DOX/OLAR_MAB, ERIB, PAC) and 3L (DOC/GEM, DTIC/GEM, NIVOL/SUNIT, CYC, PEMBR).

    CYC: Cyclophosphamide; DOC: Docetaxel; DOX: Doxorubicin; DTIC: Dacarbazine; EPI: 4-epiadriamycin; ERIB: Eribulin; GEM: Gemcitabine; IFOS: Ifosfamide; LOT: Line of treatment; mono: Monotherapy; NIVOL: Nivolumab; OLAR_MAB: Olaratumab; PAC: Paclitaxel; PEMBR: Pembrolizumab; SUNIT: Sunitinib.

    These observations by country were consistent when current and historic treatment patterns were combined for 1L and 3L+ (Figure 2). The most common reasons for stopping treatment were treatment course completion at 1L (62/122, 50.8%) and distant progression/relapse at 2L (10/21, 47.6%) and 3L+ (5/9, 55.6%). Concurrent surgery was reported in 23.6, 11.9 and 19.0% of 1L, 2L and 3L+ patients, respectively (Supplementary Table 4). Twenty-three percent of 1L, 21.8% of 2L and 14.3% of 3L+ patients had radiotherapy. Most patients starting 1L (81.0%), 2L (77.2%) or 3L+ (90.5%) received supportive care.

    Treatment outcomes

    TTNT

    For 1L, median (IQR) TTNT overall was 13.1 (9.1–18.0) months for living patients and 6.0 (2.8–11.0) months for deceased patients (Table 2). Among all patients (living and deceased), TTNT was 12.2 (10.3–20.0) months in 2L, 13.2 (4.2–13.2) months in 3L, 15.6 (15.6–15.6) months in 4L, and was not reached in 5L. For TTNT by country level, data are limited due to the small number of patients.

    OS

    There were 79 (26.7%) death events overall, and median OS was 22.0 months (95% CI: 18.0–31.2) from the date of first mSS diagnosis for all LOTs. The percentage of living patients at 6, 12 and 24 months of the post-index date was 63.2, 34.1 and 11.1%, respectively. The OS probabilities were 0.94 (95% CI: 0.91–0.97) at 6 months, 0.79 (95% CI: 0.73–0.85) at 12 months and 0.49 (95% CI: 0.40–0.59) at 24 months. Median OS calculated from the date of respective LOT initiation was 6.0 (95% CI: 5.2–9.0) months in 2L (n = 122), 4.9 (4.4–N/A) months in 3L (n = 21), and 7.5 (5.9–N/A) months in 4L (n = 7).

    HCRU

    The median (IQR) number of hospital admissions/year overall was 1 (0–3) (Table 3). Higher median (IQR) numbers of hospital admissions/year were observed in France (2 [1–4]) and the UK (1.5 [1–6]) compared with the other countries. Median (IQR) total days in hospital was 3 (0–12)/year overall; estimates for France (4 [2–10]), Germany (5 [0–17]) and the UK (5.5 [0–16]) were similar and were higher than in Italy and Spain (0 [0–8]).

    The median (IQR) number of outpatient visits/year was 4 (2–9) overall, with the highest number of visits in Germany (8 [4–11]), followed by the UK (7 [3–12]), Spain (6 [2–14]), Italy (3 [2–6]) and France (2 [1–4]; Table 3). The median (IQR) number of visits to the medical/clinical oncologist was 3.5 (1–7), being highest in Spain at 6 (1–10) visits/year and lowest in France (2 [0–4]). The median (IQR) number of visits to the surgeon was 0 (0–1), being highest in the UK at 1 (0–2) visits/year and lowest in France (no visits recorded). The median (IQR) number of visits/year to the radiotherapist was 0 (0–1) overall, and in all countries except for the UK, where the median (IQR) was 1 (0–1) visits/year.

    Discussion

    To our knowledge, this retrospective assessment of the demographic and clinical characteristics, treatment patterns and HCRU among 296 patients diagnosed with mSS in five European countries (France, Germany, Italy, Spain and the UK) is the largest European real-world evidence (RWE) study of patients with mSS, and the first to characterize HCRU in the management of mSS.

    The number of patients seen per physician varied between countries, which may reflect country-specific centralization of care for patients with SS; indeed, a median of 34 patients were seen by UK physicians compared with eight for Germany, which operates a more decentralized healthcare system. Most patients had a primary diagnosis of mSS, and, of these, lung/respiratory metastases were the most common. The median age of patients with mSS was 54 years, which is higher than previously reported in a real-world study of mSS in Italy (37 years) [20] and a review of patients with mSS in 15 European clinical trials (40 years) [21]. It is possible the higher median age observed in this study was a result of the patient selection criteria (including those most recently seen by their treating physician), leading to a degree of survival bias. Owing to the paucity of data for mSS, other like-for-like comparisons to published data are difficult, as most previous studies have provided results for STS overall, with small populations of SS cases included.

    The treatment pattern findings suggest broad adherence to current treatment recommendations in Europe, where doxorubicin (monotherapy or with IFOS) is recommended in 1L; IFOS monotherapy provides an alternative if doxorubicin is contraindicated [5,22,23]. This may particularly be the case if patients have received doxorubicin for localized disease before and have reached the maximum cumulative dose. A small minority of patients (6.3%) were receiving trabectedin or pazopanib as 1L. Trabectedin is approved after progression following anthracyclines and IFOS and for patients unsuited to receive these agents [11], while pazopanib is only approved for mSS following prior chemotherapy, or in patients who have progressed within 12 months of (neo)adjuvant therapy [10]. It is possible that some patients received pazopanib following doxorubicin as perioperative treatment of localized disease and then metachronous metastases. Similar to the results reported in this study, a real-world retrospective review of medical records of 807 patients treated with systemic therapy for advanced histologically diagnosed STS in the UK, Spain, Germany and France by Nagar et al. found that doxorubicin alone, or in combination with ifosfamide, was frequently used at 1L in all four countries [18]. Similarly, Leahy et al. analyzed data from a retrospective medical record review of 213 patients with metastatic STS across nine countries, including the five European countries of our study, and found that the most common 1L regimens were doxorubicin monotherapy (34%) and anthracycline plus ifosfamide (30%) [24]. In the US specifically, Pokras et al. reported that patients with mSS treated in community practices most commonly received doxorubicin and ifosfamide (35.9%) followed by either monotherapy or combination treatment with pazopanib (17.9%) at 1L [25]. Similarly, the Pollack et al. multi-center retrospective study of 249 patients with advanced or mSS from nine sarcoma centers in the US reported that the most common treatment in 1L was doxorubicin and ifosfamide combination (37%) followed by ifosfamide monotherapy (20%) [26].

    Doxorubicin and IFOS-based regimens were also being utilized in later LOTs, which may reflect some degree of ‘rechallenge' with these agents. This is not possible to confirm from the survey data, but research suggests that this approach has clinical activity in STS and may be a viable option for the treatment of advanced/mSS [27,28]. The most common 2L+ regimens were trabectedin-based, which is consistent with product labeling and guideline recommendations [5,11,22,23]. Pazopanib monotherapy and combinations were used in 15.8% and 9.5% of patients in 2L and 3L+, respectively. ECOG PS decreased as patients progressed to later LOTs. Our results were consistent with the findings of a RWE study of 5298 patients with advanced STS across France, Germany, Spain and Italy that trabectedin was the most commonly used choices of treatment in 2L [29]. In contrast to the results of our study, Pollack et al. reported that, in the US, trabectedin was the 5th-most common choice of treatment (12%) for patients with advanced or mSS in 2L [26]. Pokras et al. reported the most common regimens for patients with mSS in the USA involved monotherapy or combination of either ifosfamide (17.9%) or pazopanib (17.9%) in 2L, and monotherapy or combination of pazopanib (20.5%) in 3L [25]. Overall, our findings for treatment patterns across LOTs are similar to published STS RWE in Europe. However, all the RWE so far has been in a broader group of advanced STS, in which SS forms a small population. This is the first RWE study to focus predominately on a larger SS population. Moreover, our study shows differences in treatment pattens at 2L and beyond compared with US studies in patients with SS.

    The median TTNT for the five countries overall was similar for 1L, 2L and 3L (13.1, 12.2 and 13.2 months for living patients, respectively). Differences were observed among individual countries in 1L and 2L, with the highest median TTNT observed in the UK (1L: 16.5 months; 2L: 20.0 months). The TTNT increase observed here may be due to small patient numbers in later lines.

    The estimated median OS (from the date of mSS diagnosis across the five countries for all LOTs) was 22 months, similar to that reported in the recent METAstatic SYNovial sarcoma (METASYN) database study of 417 patients with mSS in France (median OS: 22.3 months [95% CI: 19.7–24.1]) [30]. Another study of patients with mSS treated at a single institution in Italy had an approximate median OS of <18 months (estimated value from a Kaplan–Meier plot showing 5-year OS) [31]. In the USA, Pokras et al. reported that patients with mSS who received systemic treatment at 1L in community practices had a median OS of 24.5 months (95% CI: 17.1–35.0) [25], which is similar to the median OS of 24.7 months (95% CI: 20.9–29.4) reported in Pollack et al. [26]. The median OS for 2L (6.0 months; 95% CI: 5.2–9.0) is slightly lower than other reports, which range from 10.4 months (95% CI: 8.5–12.7) in a meta-analysis of trials and RWE for pazopanib and trabectedin efficacy in 2L+ [16] to 16 months (95% CI: 13.5–18.9) in an analysis of patients with mSS receiving various anticancer therapies at nine major US academic centers [26]. Our estimates may be lower than those reported in the meta-analysis [16] due to a variety of regimens being used, and lower than in the other analysis [26], as that study included patients treated at major academic medical centers. Overall, these findings confirm the need for more effective treatment options in previously treated patients.

    The HCRU data showed that the UK, Germany, and Spain had higher levels of HCRU than France and Italy; the latter had particularly low levels of inpatient and outpatient hospital visits. These findings likely reflect between-country variances in healthcare practices, including utilization of various treatments with different administration routes and/or drug-related adverse event management, rather than being an indication of the severity of disease, which seemed comparable between countries. With the differences noted across countries, it is important for future researchers to ensure HCRU can differ by country for the same condition and disease severity, and this should be considered in cost-effectiveness and economic analyses.

    Strengths of our study include the use of real-world, standardized survey data collected by experienced physicians across specialties and five countries. The study included large center of excellence hospitals and collected patient-level data based on physician-abstracted charts rather than claims data. The paper contains details on patient characteristics, treatment patterns and outcomes. The study had broad patient eligibility criteria and accrued a large sample of patients with mSS, despite the rarity of this disease. These factors help ensure wider applicability and generalizability of the data, and provision of detailed information on mSS treatment patterns by LOT and HCRU addresses some of the gaps in existing literature in these five countries.

    Treatment patterns for 2L reported here differ from those reported for patients with mSS in regions outside of Europe. For example, in a single-center, retrospective analysis in Japan, the most common 2L therapies were doxorubicin with or without IFOS and IFOS plus etoposide [32]. In a retrospective cohort study of patients with mSS treated in US community practices, IFOS- and pazopanib-containing regimens were most common in 2L [25]. However, these studies also highlight the urgent need for new approaches to improve outcomes in patients with mSS in the 2L setting, irrespective of the treatment received [25,32]. In the Japanese study, the 1-year OS rate was 67.1% in patients receiving 2L therapy (median OS was not reported) [32], while in the US community study, median OS was 24.5 months (range: 17.1–35.0) in patients receiving 1L therapy [25].

    The study had the limitation of being a retrospective analysis, with possible selection bias for participating physicians. As the cross-sectional study design did not allow for follow-up after the study inclusion date, the outcomes of ongoing treatment or future LOTs could not be determined. Restricting eligibility to patients who received treatment or died in the past 12 months may have introduced selection bias by excluding patients in remission (likely to have better outcomes), and patients receiving palliative care (likely to have worse outcomes). In the living cohort, inadvertent selection of patients at earlier stages of treatment may have occurred. There may also be an under-representation of living patients in later LOTs compared with the deceased cohort, where eligibility was restricted to patients with ≥2 LOTs. Another limitation of the design is that a certain number of living and deceased patients were preselected, so the OS is not based on natural distribution of death in this population. In addition, LOT was defined by use of chemotherapy/targeted treatments only (information on surgery and radiotherapy was only descriptive), which could potentially affect our understanding of the breadth of treatment used by patients and consequent impact on outcomes. Patients initially diagnosed with localized disease and treated with cytotoxic/targeted regimens who subsequently went on to develop metastatic disease and were unable to take the same regimen were not accounted for in this analysis.

    The study assumed that physicians reported data based on medical record abstraction, which may be subject to errors arising from the abstraction process and comprehension of the data collection tools. While major events such as patient hospitalizations were captured, patients could have received healthcare services in other settings (e.g., pharmacy visits or other specialty clinics outside of the physicians' institutions), which may not have been adequately recorded in patients' medical records. This is likely to be country-specific, influenced by variations in the healthcare systems of the countries sampled and, therefore, represents a potential confounding factor for regional associations observed. In addition, reasons for each healthcare visit and costs were not available. While this is the largest sample of patients with mSS investigated to date, there remain limitations associated with interpreting data in subgroups, in which event numbers are relatively small. For example, further stratification of analyses (e.g., outcomes for current vs historic LOTs) could not be fully explored, and the survey only captured 21 patients being treated in 3L+. However, this might be expected for mSS, where relapsing patients are increasingly likely to be managed in supportive care settings, rather than having regular contact with a specialist. Finally, as the German healthcare system is structured to have more office-based physicians compared with the other four counties, the selection criteria for physician eligibility could not be applied. Consequently, prescribed treatments and outcomes may have been affected.

    Taken together, the variation in treatments across LOTs, the documented worsening of ECOG PS with subsequent LOTs, and poor clinical outcomes highlight the continued unmet need for effective therapies to improve outcomes in patients with mSS. Therapeutic progress for mSS will require mSS-specific clinical trials, and trial designs in Europe should consider regional patient characteristics, treatment patterns and outcomes.

    Conclusion

    The use of real-world, standardized survey data that have broad patient eligibility criteria and accrue a large sample of patients is key for exploring unmet needs and burden of illness, especially in rare populations in which only small sample sizes exist at single centers. In the future, owing to cell and gene therapies in development for metastatic synovial sarcoma, there are expected to be more treatment options available for patients, with possible changes in treatment patterns and patient outcomes.

    Summary points
    • Synovial sarcoma (SS) is a rare and aggressive subtype of soft tissue sarcoma. Owing to the rarity of the disease, there are no studies on the burden of SS specifically. Hence, treatment patterns, real-world effectiveness and impact of disease on healthcare resource utilization  are unknown.

    • To our knowledge, this is the largest European real-world evidence study of patients with metastatic SS (n = 296), and the first to characterize healthcare resource utilization in the management of metastatic synovial sarcoma. Strengths of this study include use of real-world, standardized survey data collected by experienced physicians across centers and countries.

    • The treatment pattern findings in metastatic SS suggest broad adherence to current metastatic soft tissue sarcoma treatment recommendations in Europe, where doxorubicin (monotherapy or with ifosfamide) is recommended in 1L. The most common treatments used in 2L were trabectedin-based regimens; however, with significant variation within and across countries.

    • Median overall survival was 22.0 months in all metastatic patients, 6.0 months in 2L and 4.9 months in 3L; documenting high unmet need in previously treated metastatic patients.

    • Patients typically had one hospital admission per year with a median 3 days in hospital. Higher numbers of hospital admissions per year were noted in France and the UK, and longer hospital stays in France, Germany and the UK.

    • Taken together, the variation in treatments across lines of treatment, high treatment discontinuation due to disease progression and the documented worsening of patient outcomes in 2L and later lines of treatment highlight the continued unmet need for effective therapies to improve outcomes in patients with metastatic synovial sarcoma.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2022-1005

    Acknowledgments

    Data from this study were previously presented at the AACR Annual Meeting 2021 (Poster 2622).

    Financial & competing interests disclosure

    This study was funded by GSK (212253). N Patel is an employee of GSK, a former employee of Merck, and owns stocks/shares in GSK. S Pokras is an employee of GSK and owns stocks/shares in GSK. J Ferma, V Casey and F Manuguid are employees of IQVIA Inc, which has received funding from GSK. K Culver was an employee of GSK at the time of the manuscript submission, and is a former employee of Novartis, and owns stocks/shares in GSK. S Bauer has received honoraria from Bayer, Deciphera, GSK, Novartis, Pfizer and PharmaMar; was in a consulting or advisory role for ADC Therapeutics, Adcendo, Bayer, Blueprint Medicines, Boehringer Ingelheim, Daiichi Sankyo, Deciphera, Exelixis, GSK, Janssen-Cilag, Lilly, Mundipharma and Nanobiotix; has received research funding from Blueprint Medicines, Incyte (institution received funding) and Novartis; and has received travel, accommodations or expenses from PharmaMar. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Editorial support was provided by F Wignall, of Fishawack Indicia, part of Fishawack Health, and T Taylor of Scion (London, UK), and writing support was provided by E Giannakouri of Scion, and was funded by GSK.

    Data availability statement

    GSK makes available anonymized individual participant data and associated documents from interventional clinical studies that evaluate medicines, upon approval of proposals submitted to www.clinicalstudydatarequest.com. To access data for other types of GSK-sponsored research, for study documents without patient-level data and for clinical studies not listed, please submit an enquiry via the website.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Riedel RF, Jones RL, Italiano A et al. Systemic anti-cancer therapy in synovial sarcoma: a systematic review. Cancers (Basel) 10(11), 417 (2018).
    • 2. Stiller CA, Trama A, Serraino D et al. Descriptive epidemiology of sarcomas in Europe: report from the RARECARE project. Eur. J. Cancer 49(3), 684–695 (2013).
    • 3. Joseph N, St Laurent S, Zheng S, Stirnadel-Farrant H, Dharmani C. Epidemiology of synovial sarcoma in EU28 countries. Ann. Oncol. 30, v706–v707 (2019).
    • 4. De Pinieux G, Karanian M, Le Loarer F et al. Nationwide incidence of sarcomas and connective tissue tumors of intermediate malignancy over four years using an expert pathology review network. PLOS ONE 16(2), e0246958 (2021).
    • 5. Casali PG, Abecassis N, Aro HT et al. Soft tissue and visceral sarcomas: ESMO-EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 29(Suppl. 4), iv268–iv269 (2018).
    • 6. Ferrari A, Chi YY, De Salvo GL et al. Surgery alone is sufficient therapy for children and adolescents with low-risk synovial sarcoma: a joint analysis from the European paediatric soft tissue sarcoma Study Group and the Children's Oncology Group. Eur. J. Cancer 78, 1–6 (2017).
    • 7. Desar IME, Fleuren EDG, Van Der Graaf WTA. Systemic treatment for adults with synovial sarcoma. Curr. Treat. Options Oncol. 19(2), 13 (2018).
    • 8. Rothermundt C, Fischer GF, Bauer S et al. Pre- and postoperative chemotherapy in localized extremity soft tissue sarcoma: a European Organization for Research and Treatment of Cancer Expert Survey. Oncologist 23(4), 461–467 (2018).
    • 9. Gelderblom H, Judson IR, Benson C et al. Treatment patterns and clinical outcomes with pazopanib in patients with advanced soft tissue sarcomas in a compassionate use setting: results of the SPIRE study. Acta Oncol. 56(12), 1769–1775 (2017).
    • 10. European Medicines Agency. Votrient (pazopanib). www.ema.europa.eu/en/medicines/human/EPAR/votrient.
    • 11. European Medicines Agency. Summary of Product Characteristics. www.ema.europa.eu/en/documents/product-information/yondelis-epar-product-information_en.pdf.
    • 12. Kasper B, Sleijfer S, Litiere S et al. Long-term responders and survivors on pazopanib for advanced soft tissue sarcomas: subanalysis of two European Organisation for Research and Treatment of Cancer (EORTC) clinical trials 62043 and 62072. Ann. Oncol. 25(3), 719–724 (2014).
    • 13. Van Der Graaf WT, Blay JY, Chawla SP et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 379(9829), 1879–1886 (2012).
    • 14. Sleijfer S, Ray-Coquard I, Papai Z et al. Pazopanib, a multikinase angiogenesis inhibitor, in patients with relapsed or refractory advanced soft tissue sarcoma: a phase II study from the European organisation for research and treatment of cancer-soft tissue and bone sarcoma group (EORTC study 62043). J. Clin. Oncol. 27(19), 3126–3132 (2009).
    • 15. El Beaino M, Araujo DM, Lazar AJ, Lin PP. Synovial sarcoma: advances in diagnosis and treatment identification of new biologic targets to improve multimodal therapy. Ann. Surg. Oncol. 24(8), 2145–2154 (2017).
    • 16. Carroll C, Patel N, Gunsoy NB, Stirnadel-Farrant HA, Pokras S. Meta-analysis of pazopanib and trabectedin effectiveness in previously treated metastatic synovial sarcoma (second-line setting and beyond). Future Oncol 18(32), 3651–3665 (2022). •• This meta-analysis demonstrated the lack of meaningful improvements in survival beyond the 1L setting shown in clinical trials in patients with metastatic soft tissue sarcoma (mSTS). This highlights an unmet need for more effective treatment options for mSTS.
    • 17. Sarker A, Chandrashekar P, Magge A, Cai H, Klein A, Gonzalez G. Discovering cohorts of pregnant women from social media for safety surveillance and analysis. J. Med. Internet Res. 19(10), e361 (2017).
    • 18. Nagar SP, Mytelka DS, Candrilli SD et al. Treatment patterns and survival among adult patients with advanced soft tissue sarcoma: a retrospective medical record review in the United Kingdom, Spain, Germany, and France. Sarcoma 2018, 5467057 (2018). • This large real-world evidence (RWE) study describes treatment patterns and outcomes for patients with advanced soft tissue sarcoma (STS) in the UK, Spain, Germany and France and highlights the need for novel therapies that will improve overall survival (OS) in this patient population.
    • 19. Parikh RC, Lorenzo M, Hess LM, Candrilli SD, Nicol S, Kaye JA. Treatment patterns and survival among older adults in the United States with advanced soft-tissue sarcomas. Clin. Sarcoma Res. 8, 8 (2018).
    • 20. Sanfilippo R, Dileo P, Blay JY et al. Trabectedin in advanced synovial sarcomas: a multicenter retrospective study from four European institutions and the Italian Rare Cancer Network. Anticancer Drugs 26(6), 678–681 (2015).
    • 21. Vlenterie M, Litière S, Rizzo E et al. Outcome of chemotherapy in advanced synovial sarcoma patients: review of 15 clinical trials from the European Organisation for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group; setting a new landmark for studies in this entity. Eur. J. Cancer 58, 62–72 (2016). • This large retrospective study retrieved patient data from 15 European Organisation for Research and Treatment of Cancer advanced 1L STS trials. This study reported that patients with advanced synovial sarcoma (SS), a distinct group of STS, had a better response to systemic chemotherapy and longer progression-free survival (PFS) and OS.
    • 22. Dangoor A, Seddon B, Gerrand C, Grimer R, Whelan J, Judson I. UK guidelines for the management of soft tissue sarcomas. Clin. Sarcoma Res. 6, 20 (2016).
    • 23. Garcia Del Muro X, De Alava E, Artigas V et al. Clinical practice guidelines for the diagnosis and treatment of patients with soft tissue sarcoma by the Spanish group for research in sarcomas (GEIS). Cancer Chemother. Pharmacol. 77(1), 133–146 (2016).
    • 24. Leahy M, Garcia Del Muro X, Reichardt P et al. Chemotherapy treatment patterns and clinical outcomes in patients with metastatic soft tissue sarcoma. The SArcoma treatment and Burden of Illness in North America and Europe (SABINE) study. Ann. Oncol. 23(10), 2763–2770 (2012).
    • 25. Pokras S, Tseng WY, Espirito JL, Beeks A, Culver K, Nadler E. Treatment patterns and outcomes in metastatic synovial sarcoma: a real-world study in the US oncology network. Future Oncol. 10.2217/fon-2022-0477 (2022). •• This retrospective observational study in adults with metastatic synovial sarcoma in the US Oncology Network provides data on treatment patterns and outcomes in this rare sub-population and highlights the ongoing need for effective therapies for mSS.
    • 26. Pollack SM, Somaiah N, Araujo DM et al. Clinical outcomes of patients with advanced synovial sarcoma or myxoid/round cell liposarcoma treated at major cancer centers in the United States. Cancer Med. 9(13), 4593–4602 (2020). •• This large retrospective study assessed real-world clinical outcomes in patients with synovial sarcoma and myxoid/round cell liposarcoma treated at major cancer centers. This paper is of particular interest as survival outcomes for this population were more favorable than those reported in published literature.
    • 27. Noujaim J, Constantinidou A, Messiou C et al. Successful ifosfamide rechallenge in soft-tissue sarcoma. Am. J. Clin. Oncol. 41(2), 147–151 (2018).
    • 28. Tian Z, Yang Y, Yang Y et al. High cumulative doxorubicin dose for advanced soft tissue sarcoma. BMC Cancer 20(1), 1139 (2020).
    • 29. Le Cesne AM-BJ, Reichardt P, Picci P, Blay JY. Current treatment patterns in advanced soft tissue sarcoma: real-world evidence of over 5,000 European patients. Cancer & Chemotherapy Reviews 15(Suppl. 1), 45–54 (2020).
    • 30. Moreau-Bachelard C, Campion L, Toulmonde M et al. Patterns of care and outcomes of 417 patients with METAstatic SYNovial sarcoma (METASYN): real-life data from the French Sarcoma Group (FSG). ESMO Open 7(2), 100402 (2022).
    • 31. Palmerini E, Staals EL, Alberghini M et al. Synovial sarcoma: retrospective analysis of 250 patients treated at a single institution. Cancer 115(13), 2988–2998 (2009).
    • 32. Kojima Y, Shimoi T, Seo T et al. Poor treatment outcomes with second-line chemotherapy in advanced synovial sarcoma. Oncology 100(7), 370–375 (2022).