Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
. 2017 Jun 15;9(8):533–550. doi: 10.1177/1758834017712963

Treatment of advanced, metastatic soft tissue sarcoma: latest evidence and clinical considerations

Gino K In 1,2, James S Hu 3,4, William W Tseng 5,
PMCID: PMC5524246  PMID: 28794805

Abstract

Soft tissue sarcoma (STS) is a biologically heterogeneous malignancy with over 50 subtypes. Historically, there have been few systemic treatment options for this relatively rare disease. Traditional cytotoxic agents, such as anthracyclines, alkylating agents, and taxanes have limited clinical benefit beyond the first-line setting; across all high-grade STS subtypes, median overall survival remains approximately 12–18 months for advanced metastatic disease. The development of targeted therapies has led to recent US Food and Drug Administration approval of four new treatments for high-grade STS in the advanced metastatic setting. Among these, olaratumab is most notable for its improvement in overall survival for patients with anthracycline-naïve disease. Further progress in STS management will rely on novel trial design, subtype-specific therapies and validation of biomarkers to tailor therapy. Immunotherapy has shown promise as a new, but yet undiscovered frontier in the management of STS.

Keywords: eribulin, high-grade, immunotherapy, metastatic, olaratumab, pazopanib, sarcoma, soft tissue, trabectedin

Introduction

Soft tissue sarcomas (STSs) remain one of the most challenging diseases to treat for the medical oncologist. STSs are mesenchymal neoplasms that can arise from any site within the body, including the extremity, trunk, retroperitoneum, and head and neck. These are biologically heterogeneous diseases, with over 50 subtypes that vary by molecular, histological, and clinical characteristics. The most common subtypes of high grade STS include undifferentiated pleomorphic sarcoma (UPS), liposarcoma (LPS), leiomyosarcoma (LMS), synovial sarcoma (SS), and malignant peripheral nerve sheath tumors (MPNSTs). Collectively, STSs are rare, accounting for <1% of adult cancers, with an estimated 12,310 new cases in 2016 in the United States.1 Unfortunately, up to 50% of high-risk patients with high-grade STS develop metastases and die from their disease.2 Among the young adult and pediatric population under 20-years old, STS is one of the top five causes of cancer-related death.1 The median overall survival (OS) for advanced, metastatic STS has historically been in the range of 12 months, while more recent randomized studies have noted survival approximating 18–19 months.310 Nonetheless, improvements in the management of high-grade STS are needed.

Because of the relatively low annual incidence and heterogeneous nature of these neoplasms, positive studies using newer active systemic agents for sarcomas are few. In recent years, the understanding and management of STS have been improved by the molecular and histological subclassification of STS, as well as development of novel drugs for these subtypes. In the age of personalized medicine, the medical oncologist is challenged with the task of properly tailoring and sequencing therapies for STS in an individualized manner. In this review, we provide an update of systemic options for high-grade STS, including a discussion of how best to incorporate their use amongst the growing arsenal of active agents. Specifically, we focus on the subtypes mentioned above: UPS, LPS, LMS, SS and MPNST. While there have been advances in treatment of other sarcoma subtypes, including dermatofibrosarcoma protuberans and gastrointestinal stromal tumors, a discussion of these and other sarcoma subtypes is beyond the scope of this paper and should be sought elsewhere.

Anthracycline-based therapy: first-line therapy across all subtypes

As of 2016, anthracyclines (e.g. doxorubicin) remain the standard of care for first-line therapy in high-grade STS, regardless of subtype, presentation and patient characteristics. Anthracyclines intercalate into deoxyribonucleic acid (DNA), thereby blocking DNA and ribonucleic acid (RNA) synthesis, while also interfering with topoisomerase II, leading to DNA breakage.11 Doxorubicin has single-agent activity in high-grade STS, and demonstrates a dose–response relationship, with doses lower than 60 mg/m2 associated with inferior efficacy.12,13 Because of its toxicity profile however, most dose-intensive treatment schedules administer doxorubicin at 75 mg/m2 for otherwise fit patients.14,15 Alkylating agents (e.g. ifosfamide) exert their antineoplastic effects by cross-linking strands of DNA.16 Ifosfamide also has single-agent activity in STS,17 and while they are not synergistic with anthracyclines, the two are often combined on the basis of improved response rates (RRs), progression-free survival (PFS) and palliation.13,14,18 In the first-line setting, the combination of doxorubicin and ifosfamide (AI) achieves an RR up to 12–34%, dependent on subtypes, and disease control rates (DCRs) as high as 45–77%.14,15,1821 There has been no definitive improvement in survival, however, with median OS still between 12 and 18 months.1315,18,22,23 This remains an area of active investigation, with one particular criticism being the lower doses of anthracyclines that may have confounded results in earlier studies.24

The phase III, multi-institutional EORTC 62012 study was designed to address this continuing debate.14 The investigators compared doxorubicin 75 mg/m2 in combination with ifosfamide 10 g/m2 every 3 weeks for six cycles (AI) versus doxorubicin 75 mg/m2 every 3 weeks for six cycles in 455 patients with advanced or metastatic high-grade STS. Patients younger than 60-years old with good performance status were eligible. The AI combination showed a higher RR, 26.5% versus 13.6% (p < 0.0006), respectively, compared with doxorubicin alone. AI significantly improved PFS from 4.6 months to 7.4 months [hazard ratio (HR) 0.74, p = 0.003], as well. However, there was no significant difference between AI and doxorubicin alone in terms of OS (14.3 months versus 12.8 months, HR 0.83, p = 0.076). Despite its efficacy, there was higher toxicity when using the combination, with 18% of patients receiving AI and 3% of patients receiving doxorubicin alone being unable to complete the planned six cycles of therapy due to adverse events. It should be noted the most common grade 3–4 toxicities when administering AI are related to bone marrow suppression,15,18,19 which highlights the need for regular blood-count monitoring, supportive transfusions and growth factor support. Cardiotoxicity is a rare, but significant toxicity related to anthracyclines that occurs in a dose-dependent manner, with increased risk of cardiomyopathy occurring with cumulative doses above 550 mg/m2.2527

Retrospective studies indicate that young age and good performance status are good prognostic factors for survival,28,29 while a study by Van Glabbeke noted that young age may also be predictive of response to anthracycline-based therapy.30 Meanwhile, higher histological grade is associated with better RR to chemotherapy but worse OS,28,30 likely reflecting that higher-grade tumors are more chemosensitive, but will also relapse and progress quickly. Other characteristics, such as gender, location of primary tumor, location of metastases, and histology subtype have also been investigated as additional prognostic and predictive factors, and warrant further investigation. Negative phase III trials of other anthracycline-based combinations (e.g. evofosfamide, palifosfamide),10,31 indicate the need for better patient selection, and putative biomarkers will be crucial towards improving future clinical trials.

In consideration of these findings, our practice is to offer AI combination therapy in the front-line setting to select patients who are fit and highly appropriate based on burden of disease, particularly with the goals of tumor shrinkage and resectability, or potentially life-threatening disease. We recommend the dose of doxorubicin 75 mg/m2 (25 mg/m2 per day on days 1, 2, and 3) in combination with ifosfamide 7.5 g/m2 (2.5 g/m2 per day on days 1, 2 and 3) with granulocyte colony-stimulating factor, every 3 weeks for a maximum of six cycles. For total doses of doxorubicin above 450 mg/m2, use of cardioprotectants (e.g. desroxazone) and cardiac monitoring should be considered. For less fit patients, in whom the risks of toxicity outweigh the benefits of AI combination chemotherapy, alternatives may include single-agent doxorubicin, single-agent ifosfamide, or liposomal doxorubicin (Doxil).3234 As of November 2016, the US Food and Drug Administration (FDA) granted accelerated approval for olaratumab, an anti-PDGF R (as in anti-PDGFR) monoclonal antibody, for use in combination with anthracyclines; this will be discussed in further detail below.

It should be noted that other than the five primary STS subtypes mentioned above (UPS, LPS, LMS, SS, MPNST), there are a number of other less common subtypes which are anthracycline resistant, and for which AI therapy is not recommended first line. For these subtypes, specific-histology-based treatment is discussed elsewhere,3540 and the reader is encouraged to also consider clinical trials and genomic profiling.

Second-line therapy and beyond: the basis for treating by histology subtypes

Beyond first-line therapy, we recommend that treatment be tailored to histology and molecular subtype, as well as patient characteristics. While a number of second-line regimens exist, any one agent has not proven superior across all subtypes. We will discuss these options, and provide our recommendations for when they should be appropriately sequenced (Table 1).

Table 1.

Proposed treatment sequence for advanced or metastatic, high-grade soft tissue sarcoma.

Sarcoma subtype First line Second line Third line Fourth linee
UPS aAnthracycline-based regimen Gemcitabine + docetaxel Pazopanib
LPS aAnthracycline-based regimen bTrabectedin Eribulin
LMS aAnthracycline-based regimen cGemcitabine + docetaxel Trabectedin Pazopanib
SS aAnthracycline-based regimen dHigh-dose ifosfamide Pazopanib
MPNST aAnthracycline-based regimen Pazopanib
a

Anthracycline-based regimens include: single-agent doxorubicin, doxorubicin and ifosfamide, doxorubicin and olaratumab, or liposomal doxorubicin.

b

Trabectedin particularly effective for myxoid/round cell LPS.

c

Gemcitabine + docetaxel particularly effective for uterine LMS.

d

High-dose ifosfamide only recommended for select patients with good performance status and preserved renal function.

e

Clinical trials are recommended for eligible patients.

UPS, undifferentiated pleomorphic sarcoma; LPS, liposarcoma; LMS, leiomyosarcoma; SS, synovial sarcoma; MPNST, malignant peripheral nerve sheath tumor.

High-dose ifosfamide

For SS (i.e. harboring the t(x;18) or SYT (synaptotagmin) / SSX (synovial sarcoma, X breakpoint) translocation) patients, who are fit and have good performance status, our practice is to rechallenge with high-dose ifosfamide (HDI) as second-line therapy. As previously mentioned, ifosfamide has single-agent activity in front-line and relapsed STS, with RR ranging from 25% to 50%. Ifosfamide demonstrates a dose–response relationship, particularly at doses from 5 to 12 g/m2,4144 while at doses above 16 g/m2, there are higher rates of toxicity without any further efficacy, and worse pharmacokinetics.4,43,4547 The primary toxicities associated with ifosfamide include nausea, vomiting, myelosuppression, hemorrhagic cystitis, nephrotoxicity (renal tubular acidosis, salt-wasting nephropathy), asthenia and encephalopathy.4,43,4548 Ifosfamide can be administered as a prolonged infusion with mesna via portable pumps with good physicochemical stability.49 As such, a number of studies have explored the potential for HDI in salvaging refractory STS patients, especially those who progressed on prior anthracycline-based regimens, including AI combination therapy.

In a phase I dose-escalation study by Elias et al., 20 patients with refractory STS were treated with a 4-day continuous infusion of ifosfamide, with doses escalated from 8 up to 18 g/m2 total, and supportive mesna at an equivalent dose.50 Dose-limiting toxicities at the 18 g/m2 dose, including renal insufficiency, myelosuppression, and mucositis, were all dose dependent, while severe CNS toxicity appeared even at lower doses. Among 20 patients, there were six partial responses (PRs) and one complete response (CR) for an overall RR of 35%. A number of additional phase II and retrospective studies further support the role of salvage HDI in STS, with RRs ranging from 25% to 53.8% for pretreated patients.5,42,43,48,51,52 However, many dose-dense regimens, such as 6-day ifosfamide with 2 g/m2 per day every 3 weeks, or 4-day ifosfamide with 3 g/m2 per day every 3 weeks, resulted in significant myelosuppression, sometimes as high as 40–75% grade 3–4, despite use of growth factor support.42,43,48,53

Investigators at the Royal Marsden Hospital, UK, explored a schedule of 14-day ifosfamide dosing, with 1 g/m2 per day, continuous infusion every 4 weeks as first- or second-line therapy.54 There were seven patients with PR (20%), as well as 10 patients with stable disease (SD) (29%) for a DCR of 49%. Although there was significant encephalopathy, including 34% among all grades and 17% grade 3–4, myelosuppression was significantly improved with 46% all grades and only 5.7% grade 3–4. Another retrospective analysis at the Royal Marsden Hospital rechallenged 67 patients with STS using HDI, either following prior adjuvant AI or front-line palliative ifosfamide-based regimens.55 This regimen showed significant activity, including responses in the second-line, third-line and fourth-line settings. Although the clinical benefit was greater for the SS subtype, responses were also seen in LPS and LMS, as well.

Despite its toxicities, as well as its costly and time-intensive scheduling, we endorse HDI as an appropriate second-line therapy for SS after failing anthracycline-based treatment. We recommend a 14-day continuous dosing schedule with 1 g/m2 per day, every 4 weeks with granulocyte colony-stimulating factor, as this regimen is well tolerated and has good efficacy as a salvage regimen. However, we advise this regimen be reserved to tertiary, academic centers with a high level of experience and appropriate support including adequate mesna administration. Additional precaution should be taken with patients at risk of nephrotoxicity, such as those with pre-existing renal insufficiency or history of nephrectomy.4547 For the highly select and fit patient then, especially those who demonstrated prior sensitivity to ifosfamide, this regimen is an option for salvage therapy.

Further study is needed to elicit which subtypes may be eligible for HDI treatment. Although it is generally agreed that RRs are highest for the SS subtype,41 there is evidence that specific subtypes of LPS, including myxoid/round cell LPS and well-differentiated/dedifferentiated LPS, also respond to HDI.54,56 Meanwhile, LMS is poorly responsive to HDI and should not be considered in this setting.4,41,42

Gemcitabine and docetaxel

For the second-line treatment of STS with either UPS or LMS histology, we recommend salvage with the combination of gemcitabine and docetaxel (GD). Gemcitabine is a nucleoside analog which inhibits DNA synthesis, and demonstrated single-agent activity in phase II studies of STS that progressed after anthracyclines or alkylators, including responses among UPS and angiosarcoma histology.57,58 Phase II studies of gemcitabine in combination with either vinorelbine or dacarbazine, showed only limited efficacy for STS.5961 Meanwhile, docetaxel is a taxane with microtubule-inhibiting activity, and also has single-agent activity as second-line therapy.62,63 Given that GD have different mechanisms of action from either anthracyclines or alkylating agents, as well as preclinical evidence of their synergy, there was rationale for combining these agents for treating STS.

The first phase II study of GD, conducted by Hensley et al., treated 34 patients with unresectable LMS, including 29 uterine LMS, as well as 18 treatment-naïve patients, with gemcitabine 900 mg/m2 on days 1 and 8, and docetaxel 100 mg/m2 on day 8, with granulocyte colony-stimulating factor every 3 weeks.64 Among the 34 patients, there were 3 CR and 15 PR, for an overall response rate (ORR) of 53%. There were also seven patients with SD, resulting in a DCR of 73.5%. The regimen was well tolerated, with grade 3–4 neutropenia occurring in 21% of patients and grade 3–4 thrombocytopenia in 29%.

In the follow-up phase II, open-label SARC 002 study, 122 STS patients with zero-to-three prior lines of chemotherapy, were randomized to either gemcitabine 1200 mg/m2 on days 1 and 8 every 3 weeks alone or to GD. Histology subtypes included LMS, LPS, UPS, as well as others not otherwise specified. Patients on both arms received a median of four cycles of therapy, and up to a maximum of eight cycles. The GD combination resulted in higher ORR (16% versus 8%), higher median PFS (6.2 months versus 3.2 months), and higher median OS (17.9 months versus 11.5 months), compared with gemcitabine alone, across all subtypes, but with the best outcomes primarily among LMS and UPS.6

The randomized, multicenter phase II TAXOGEM study compared GD with single-agent gemcitabine for the second-line treatment of LMS.65 A total of 90 patients were stratified as uterine LMS or nonuterine LMS, and randomized to gemcitabine or GD. For patients with uterine LMS, the ORR was 19% for gemcitabine alone versus 24% for GD; for nonuterine LMS, the ORRs were 14% and 5%, respectively. For uterine LMS, median PFS was 5.5 months versus 4.7 months, and for nonuterine LMS, median PFS was 6.3 months and 3.8 months, respectively. Given its efficacy and better tolerability, the authors also recommended that single-agent gemcitabine could be considered in second-line treatment for LMS.

In the multi-institutional European GeDDiS trial, GD failed to improve outcomes in the first-line setting compared with anthracycline-based therapy.66 A total of 257 patients with previously untreated advanced or metastatic STS were randomized 1:1 to receive doxorubicin 75 mg/m2 on day 1 every 3 weeks for six cycles versus GD for six cycles. Histology subtypes included uterine LMS (27%), SS (4%), UPS (12%) and other sarcomas (56%). Median PFS, OS and RR trended in favor of doxorubicin compared with GD. In addition, the anthracycline arm was less toxic, thereby corroborating that GD should not displace anthracycline as first-line therapy for STS.66 In a retrospective analysis of 246 patients with metastatic UPS treated at Memorial Sloan Kettering, there was no statistically significant difference in median time to progression between anthracycline-based therapy or GD in the first-, second- or third-line setting. ORR was marginally higher for anthracycline-based regimens in the first line, 26% compared with 22% among GD-treated patients.67

As with HDI, responses to GD appear to be heavily dependent on STS histology subtype. In particular, multiple studies demonstrate the efficacy of GD in LMS, with RR of 36–53% in the first-line setting and 24–27% in the second line.7,64,68,69 Furthermore, it appears that uterine LMS is more sensitive to GD, compared with nonuterine LMS.6,64,70,71 Smaller studies suggest GD has activity in other less common STS subtypes including angiosarcoma and epithelioid sarcoma.72,73

Given its modest efficacy as second-line therapy, our practice is to use the combination of gemcitabine, 900 mg/m2 on days 1 and 8, and docetaxel 100 mg/m2 on day 8, with granulocyte colony-stimulating factor every 3 weeks, as second-line therapy for refractory LMS and UPS. This regimen may be particularly effective for uterine LMS, as opposed to nonuterine LMS. Although practices vary by institution, many oncologists may prefer a lower dose of docetaxel 75mg/m2, based on the toxicity data from SARC002. For less robust patients, single-agent gemcitabine may also be considered an option.

Trabectedin

Trabectedin is FDA approved for second-line treatment and beyond for LPS and LMS. Trabectedin (ecteinascidin-743) is a synthetic compound derived from the Caribbean sea squirt, Ecteinascidia turbinata.74 Unlike other alkylating agents which target the DNA major groove, trabectedin binds and alkylates the minor groove, bending towards the major groove, disrupting the late S-phase and G2 phase, and induces p-53-independent apoptosis.7577 Other effects include modulating inflammation in the tumor microenvironment and inducing caspase-8-mediated apoptosis of tumor-associated macrophages, with reduced angiogenesis.78,79 Preclinical studies indicate trabectedin is also effective in modulating the transcription of oncogenic fusion proteins, and thus may be particularly useful in treating translocation-associated sarcomas.80,81

In a phase II study of 36 STS patients with up to two prior lines of therapy, the ORR for trabectedin was 8%, including one CR and two PRs, and two patients had minimal response (MR). All of the patients with objective responses had bulky disease and either LPS or LMS histology, including one patient with myxoid/round cell LPS (MRC-LPS) who achieved CR.82 In a second, multi-institutional phase II study of 54 STS patients with at least one prior line of therapy, there was again a low ORR of 3.7%, with only 2 PRs, as well as 4 MR and 2 SD. Outcomes did not vary with number of prior lines of therapy. The median PFS for all patients was 1.9 months, with 24% of patients progression free at 6 months. Meanwhile, the median PFS for patients with either PR or MR was 8.5 months.83 A third phase II study treated 104 patients with refractory STS across eight European institutions; among 99 evaluable patients there were 8 PRs and 45 SDs, for an ORR of 8% and DCR of 53.5%. Responses occurred in LMS, SS, LPS and UPS, with a median duration of response of 14 months. The median PFS for all patients was 3.8 months, with 29% of patients progression free at 6 months.3 Despite their low objective RR, these three phase II studies demonstrated trabectedin’s activity in refractory STS, and also emphasized the importance of PFS and SD as endpoints in second line and beyond.

A multi-institutional phase III study randomized 518 LPS and LMS patients 2:1 to receive either trabectedin or dacarbazine.84 All patients had progressed after either one anthracycline-based combination or an anthracycline plus a second-line regimen. Trabectedin significantly improved median PFS compared with dacarbazine (4.2 months versus 1.5 months), with a 45% reduction in progression or death [hazard ratio (HR) 0.55, 95% confidence interval (CI) 0.44–0.77, p < 0.001]. At 6 months, 37% of patients treated with trabectedin were progression free, compared with 14% of patients treated with dacarbazine. Although there was no difference in ORR (9.9% versus 6.9%, p = 0.33), DCR was higher in the trabectedin arm at 34% versus 19%, p < 0.001. It should be noted the greatest improvement in median PFS for trabectedin occurred in the subgroup of MRC-LPS (5.6 months versus 1.5 months). Trabectedin was well tolerated, with the most common grade 3–4 toxicities including myelosuppression and transient liver enzyme elevations.

Two studies evaluated trabectedin as first-line therapy for STS. Garcia-Carbonero et al. conducted a single-arm phase II study of trabectedin that resulted in one CR, five PRs and one MR, thus indicating the drug’s activity in the treatment-naïve patient.85 However, a subsequent phase IIb that randomized treatment-naïve patients to doxorubicin versus trabectedin failed to show any superiority to using trabectedin.86

In contrast to HDI or GD, responses to trabectedin appear specific not only to L-sarcoma histology subtypes, but also to molecular subtype. MRC-LPS has proven particularly sensitive to trabectedin, with retrospective studies showing ORR up to 50%, median PFS 14–17 months and 6 month PFS 88%.87,88 In addition, MRC-LPS patients who relapse after prior response are sensitive to rechallenge with trabectedin.89 This unexpectedly high antitumor activity has been attributed to the ability of trabectedin to modulate transcription of oncogenic fusion proteins, such as FUS-DDIT3, and thereby promote lipoblast differentiation.80 These findings led to further interest in trabectedin as a treatment option for other translocation-associated STSs, but subsequent studies showed little evidence of trabectedin having activity in SS, alveolar soft-part sarcoma or rhabdomyosarcoma.90,91

At our institution, we reserve trabectedin to the L-sarcomas, LMS and LPS, as second-line and beyond therapy, depending on patient characteristics and ease of administration. Trabectedin is dosed at 1.5 mg/m2 once every 3 weeks.92,93 It should be noted that trabectedin requires a continuous 24-hour infusion, and central venous access is recommended to avoid painful phlebitis. Dose-limiting toxicities include myelosuppression and reversible elevation of transaminases; patients with moderate hepatic impairment require dose reduction, while patients with severe hepatic impairment are not eligible for treatment.94 In addition to the L-sarcomas, trabectedin may have activity in other sarcomas, but this will require further investigation.95,96 A number of ongoing studies will assess the potential combination of trabectedin with other agents,97,98 as well as the possibility of DNA repair-based biomarkers to predict responsiveness to trabectedin.99,100

Pazopanib

Pazopanib is the only FDA approved oral agent for high grade STS, and we consider this an acceptable option for all STS subtypes after failing anthracycline, with the exception of LPS. Pazopanib is a multikinase tyrosine kinase inhibitor that targets multiple receptors, including vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGFR), and c-KIT. VEGF is overexpressed on tissue and serum samples across multiple STS subtypes, including UPS and LMS. Moreover, high VEGF expression levels are correlated with higher histologic grade, larger tumor size, higher stage, and worse prognosis,101104 thus supporting this as a therapeutic target for STS.

The EORTC 62043 single-arm study treated 142 advanced STS patients, who had up to two prior lines of chemotherapy, or who were ineligible for chemotherapy, with pazopanib 800 mg orally once daily.105 Among four different histology cohorts, three of them, LMS, SS and other sarcomas reached the primary endpoint of PFS at 12 weeks (PFR12w), while the LPS cohort was closed early, given it did not reach the interim analysis endpoint. Among LMS, SS and other sarcomas, the PFR12w rates were 44%, 49% and 39%, respectively. Among nine PRs, five occurred in patients with SS, and one with LMS, but there were no CRs.

The multi-institutional, phase III PALETTE study randomized 369 pretreated, metastatic STS patients from Europe, Asia, Australia and USA, 2:1 to pazopanib versus placebo.106 Patients were allowed up to four prior lines of therapy, with 99% receiving prior anthracyclines and 56% having at least two prior lines of therapy. Histology subtypes included SS, LMS and other sarcomas; LPS was not eligible. Pazopanib improved median PFS; 4.6 months versus 1.6 months (HR 0.31, 95% CI 0.24–0.40, p < 0.001), compared with placebo. Similar to the phase II study, rates of PR were very low (6% versus 0%), but rates of SD were higher for pazopanib, 67% versus 38%, compared with placebo.106 There was no significant difference in median OS of 12.5 months for pazopanib, compared with 10.7 months for placebo (HR 0.86, 95% CI 0.67–1.1, p = 0.25). The most common toxicities were hypertension, fatigue, diarrhea, nausea, and weight loss, while rare but significant toxicities associated with pazopanib included thromboembolism (5%), pneumothorax (3%) and decreased left ventricular ejection fraction (6.5%). Questionnaire-based, health-related quality of life (HRQoL) was not improved with pazopanib, although this did not correlate with significantly worse overall health status.107

In a retrospective analysis of both EORTC 62043 and PALETTE, a total of 77 patients had long-term responses with PFS >6 months and OS >18 months, including 12 patients who remained on treatment for more than 2 years.108 The investigators identified good performance status, low or intermediate histologic grade and normal hemoglobin as good prognostic factors. TP53 mutations may predict response to pazopanib, but further investigation is need to validate this, as well as other biomarkers.109,110 Hypertension has not proven a reliable biomarker to predict response to pazopanib.111

In 2012, pazopanib was FDA approved for the treatment of STS progressed on prior anthracycline-based therapy, with the exception of LPS histology. Additional studies confirmed that pazopanib has antitumor efficacy in multiple subtypes including LMS, SS, UPS and MPNST, but has minimal activity for LPS.112114 There are also limited data to suggest pazopanib has single-agent activity in other less common subtypes, such as clear cell sarcoma, solitary fibrous tumor, and hemangioendothelioma.115118

It is our practice to consider pazopanib in the second- or third-line setting for SS, UPS, or MPNST. For patients with LMS, we also offer pazopanib in the second, third, or even fourth line, depending on patient fitness and comorbidities. We do not use pazopanib for LPS, given its poor activity in studies thus far, although there is some interest in re-exploring the role of pazopanib for this subtype. At final analysis of EORTC 62043, an additional two LPS patients did meet the primary endpoint, with a resultant PFR12w of 26%, which would have met the study threshold for continuation, and based on these findings, pazopanib is approved for LPS in Japan.119 Because pazopanib is the only oral agent approved for high-grade STS and relatively well tolerated, it is an excellent alternative to other salvage therapies for the less robust patient. An ongoing phase II trial will examine the role of first-line pazopanib as an alternative to doxorubicin for elderly patients [ClinicalTrials.gov identifier: NCT01861951].120 Additional studies are also underway to explore the potential combination of pazopanib with PIK3/mTOR inhibitors121 and epigenetic-modifying agents.122,123

Eribulin

Eribulin is FDA approved for patients with LPS that progressed after treatment with anthracyclines. Eribulin is a synthetic analog of halichondrin B, which was isolated from the marine sponge, Halichondria okadai, and the Axinella family of sponges.124,125 Unlike other microtubule inhibitors (e.g. taxanes, vinca alkaloids), eribulin has a unique mechanism of action that sequesters tubulin into nonfunctioning aggregates.126 Eribulin also remodels the tumor vasculature, and reverses the epithelial–mesenchymal transition.127,128 Eribulin has activity in multiple solid tumors, and is FDA approved for breast cancer, in addition to LPS after prior anthracyclines.

In the phase II EORTC 62052 study, 128 STS patients, who progressed after one combination or up to two prior lines of therapy, were treated with eribulin 1.4 mg/m2, on days 1 and 8 every 3 weeks.129 There were four histology cohorts including SS, LMS, LPS, and other sarcomas. At the primary endpoint, 46.9% of LPS patients and 31.6% of LMS patients were progression free at 12 weeks, while among SS and other sarcomas, PFR12w was 21% and 19%, respectively. Despite the low ORR of 6% among LPS, there were 41% of patients with SD, for a DCR of 47%. The DCR for LMS, SS and other sarcomas were lower at 32%, 21%, and 19%, respectively. Median PFS for LPS, LMS, SS and other sarcomas were 2.6 months, 2.9 months, 2.6 months and 2.1 months, respectively. In a second phase II study, 51 patients in Japan with refractory STS, who failed at least one line of therapy, were treated with eribulin. At the primary endpoint, patients with LPS or LMS histology had a PFR12w of 60% compared with 31% in other sarcomas and 51% overall. Median PFS was 5.5 months for LPS or LMS, 2.0 months for other sarcomas, and 4.1 months overall. Meanwhile, median OS was 17 months for LPS or LMS, compared with 7.6 months in other sarcomas and 13.2 months overall.130

In a phase III randomized study across 22 countries, 452 patients with intermediate/high-grade LPS or LMS who had up to two lines of prior therapy were randomized to eribulin versus dacarbazine.131 Eribulin significantly improved median OS; 13.5 months versus 11.5 months, compared with dacarbazine (HR 0.77, 95% CI 0.62–0.95; p = 0.0169). Although not appropriately powered for this study, the median OS benefit of eribulin compared with dacarbazine was greater for LPS (15.6 months versus 8.4 months), as compared with LMS (12.7 months versus 13.0 months). Strikingly, there was no benefit on median PFS, which was 2.6 months (95% CI 1.9–2.8) for eribulin and 2.6 months (95% CI 1.8–2.7) for dacarbazine (HR 0.88; 95% CI 0.71–1.09, p = 0.23). PFR12w, DCR and HRQoL were not significantly different between treatment arms. The most common grade 3–4 toxicities related to eribulin were neutropenia (35%) and leukopenia (10%), while the most common toxicities overall included fatigue, nausea, neuropathy, elevated transaminases and alopecia.

It remains unclear why eribulin improved OS, but failed to improve the secondary endpoints of this study, including PFS, RR and DCR. Similar findings occurred in a breast cancer study, where eribulin resulted in improved OS, but not PFS, compared with other cytotoxic therapy.132 The investigators hypothesize that these results are due to the manner in which eribulin impacts the tumor microenvironment and induces vascular remodeling.127,128

We recommend eribulin 1.4 mg/m2, on days 1 and 8 every 3 weeks, as third-line and beyond therapy for LPS. Given the availability of other agents in this setting (e.g. trabectedin, pazopanib), we reserve this agent only for select, appropriately fit patients. Toxicities from eribulin include mucositis, myelosuppression, elevated transaminases, and sensory neuropathy.133 Nevertheless, eribulin is an important agent with a survival benefit in LPS, and should always remain within the consideration of the sarcoma oncologist.

Olaratumab: a novel agent for first-line therapy

In November 2016, the FDA granted accelerated approval of olaratumab in combination with doxorubicin for the treatment of anthracycline-naïve STS, based on a phase Ib/II study. Unlike the other aforementioned agents, olaratumab combined with doxorubicin was shown to improve OS in the anthracycline-naïve setting, and as such, may change the paradigm for management of advanced high-grade STS. Results from the confirmatory phase III study will be important towards the FDA granting full approval.

Olaratumab is a fully human recombinant IgG1 monoclonal antibody that blocks PDGF-AA and PDGF-BB from binding PDGFRα.134 PDGFRα overexpression has been found in a number of malignancies, including STS,135,136 glioblastoma,137 ovarian carcinoma,138 hepatocellular carcinoma,139 and metastatic medulloblastoma.140 PDGFR amplification and activating mutations have also been found in gliomas137,141 and gastrointestinal stromal tumor (GIST).142 Furthermore, PDGFR-ligand binding has been found to play a significant role in stemness, senescence and apoptosis in sarcoma, and is also associated with metastatic progression.143 In xenograft models of LMS and glioblastoma, olaratumab demonstrated antitumor activity via modulation of the PDGFR pathway, as well as downstream inhibition of Akt and MAPK.134 Olaratumab also delayed progression and reduced bone metastases in a prostate cancer xenograft.144

In the first-in-human phase I study, a total of 19 patients (none were STS) were treated with olaratumab. The maximum tolerated dose (MTD) was not reached, and the recommended phase II dose was 16 mg/kg weekly and 20 mg/kg biweekly. Twelve patients (63.2%) had a best response of stable disease.145 In a phase I study of 16 Japanese patients with advanced tumors treated with single-agent olaratumab, the best overall response was SD in seven patients (43.8%), including one patient with LMS originating in the inferior vena cava, with disease stabilization lasting 5.6 months.146

Olaratumab was given accelerated approval by the FDA in November 2016, based on a phase Ib/II study among anthracycline-naïve, advanced STS.147 The phase II trial randomized patients to olaratumab 15 mg/kg on days 1 and 8 and doxorubicin 75 m/gm2 on day 1, every 3 weeks, or doxorubicin alone at 75 mg/m2 on day 1, every 3 weeks. After eight cycles, patients on the olaratumab arm were allowed to continue olaratumab alone, while patients on the doxorubicin alone arm could receive olaratumab monotherapy after progression. The median PFS was not significantly improved with olaratumab at 6.6 months compared with 4.1 months for doxorubicin alone (HR 0.67, 95% CI 0.44–1.02, p = 0.0615). However, the median OS was 26.5 months with olaratumab, compared with 14.7 months in the doxorubicin alone arm, equivalent to an improvement of 11.8 months (HR 0.46, 95% CI 0.30–0.71, p = 0.0003). This benefit was seen regardless of STS subtype, PDGFR expression or number of lines of prior therapy. Best response with olaratumab included two patients with CR, but the ORR was not significantly different between both arms (18.2% versus 11.9%, p = 0.34). Overall toxicities related to olaratumab included fatigue, alopecia, neuropathy and headache, while grade 3–4 toxicities included neutropenia, mucositis, nausea, vomiting and diarrhea. The confirmatory phase III of olaratumab plus doxorubicin or doxorubicin alone has completed accrual and results are pending.

Olaratumab is the first novel agent that offers a change of strategy in the treatment of high-grade, advanced or metastatic, anthracycline-naïve STS. For patients with good performance status and STS histology who do not qualify for a clinical trial, the combination of doxorubicin on day 1 with olaratumab on days 1 and 8, every 3 weeks for eight cycles, should be considered. This should be followed by continuation with olaratumab alone. We routinely premedicate patients with dexamethasone and diphenhydramine and also administer granulocyte-colony stimulating factor with each of the first eight cycles.

The potential role of immunotherapy in STS

Although there are no approved agents using immunotherapy in sarcoma, this remains an area of active investigation and warrants mention. The role of immunotherapy in sarcoma dates back as far as the 19th century, when William Coley described a patient with the regression of sarcoma following clearance of a bacterial infection.148 More recently, the success of checkpoint inhibitors in solid tumors has led to interest in their use for treating sarcoma. While it is generally agreed that immune recognition is critical to harnessing immunotherapy in cancer, the optimal strategy for immunotherapy in STS is yet to be determined.

Overall tumor mutational burden has been identified as a predictor of responsiveness to checkpoint inhibition in multiple malignancies;149,150 while skeptics argue that sarcoma, as a whole, has a low mutational burden, this is likely dependent on the unique biology of each subtype.151 Another strategy has been to identify immunogenic antigens with expression specific to sarcoma. Characteristic translocations or fusion proteins, such as those found in SS and MRC-LPS, may represent one antigen for immune recognition.152 Another antigen is the cancer testis antigen, NY-ESO-1, which is expressed in up to 100% of MRC-LPS and 80% of SS, as well as other subtypes, including uterine LMS and osteosarcoma.153155 This led to a pilot study of checkpoint inhibition using the anti-CTLA-4 (cytotoxic T-lymphocyte antigen-4) antibody ipilimumab, 3 mg/kg every 3 weeks, in NY-ESO-1 expressing SS. Although no objective responses were seen among the six patients treated, ipilimumab was well tolerated with no serious adverse events reported.156 However, in another study using NY-ESO-1-engineered T cells, four out of six responses were noted in NY-ESO-1-expressing SS patients.157

Tumor-infiltrating lymphocytes (TILs) play an essential role in the immune response, and have been identified in multiple STS subtypes, including LPS, LMS, SS and MPNST,158160 although their presence alone does not result in tumor regression. Similar to CTLA-4, the programmed cell death 1 (PD-1) receptor and its programmed death ligand-1 (PD-L1) is another checkpoint molecule which has been unlocked with success in melanoma and non-small cell lung cancer, amongst others. PD-L1 tumor expression ranges from 12% to 58%, with notable variation by sarcoma subtype.160,161 In a retrospective study of 82 STS patients, PD-L1 expression was seen in 100% (7/7) patients with epithelioid sarcoma, 53% (10/19) of SS, 38% (12/32) of rhabdomyosarcoma, 33% (6/18) Ewing sarcoma, and 0% (0/6) mesenchymal chondrosarcoma.162 PD-1 positivity and PD-L1 expression in STS have been correlated with poor prognosis, advanced stage, higher histologic grade, distant metastasis, and degree of tumor differentiation and necrosis.161

SARC 028 is a multi-institutional, phase II study that treated 86 patients with STS and bone sarcoma with the anti-PD-1 therapy, pembrolizumab, 200 mg IV every 3 weeks [ClinicalTrials.gov identifier: NCT02301039]. Eligible subtypes included LMS, LPS, UPS and SS. Early interim analysis showed an 8-week ORR of 0% for LMS (0/10), LPS (0/9) and SS (0/10), but notably, an ORR of 22% (2/9) among UPS. PFS at 8 weeks (PFS8w) was 50%, 63%, 30%, and 67%, for LMS, LPS, SS and UPS, respectively. Among 24 patients who were evaluated at 24 weeks, one additional LPS and one additional UPS also achieved PR.163 Given the preliminary signal seen in dedifferentiated LPS and UPS, an expansion study is actively being planned.

A phase II, single-arm study assessed the impact of another anti-PD-1 therapy, nivolumab, in the treatment of uterine LMS with progression after at least one prior therapy.164 Among a total of 12 women with pretreated metastatic uterine LMS who received nivolumab 3 mg/kg IV every 2 weeks, the ORR was 0% (0/12).164 An ongoing phase II study [ClinicalTrials.gov identifier: NCT02500797] will randomize patients with advanced sarcoma to treatment with nivolumab with or without ipilimumab. Further investigation is needed to delineate the role of specific subsets of TILs in the STS microenvironment, and how to best utilize biomarkers such as NY-ESO-1 and PD-1/PD-L1, to select appropriate patients for therapy. While investigation into the role of immunotherapy for STS continues, the use of any checkpoint inhibition for STS should only be conducted in the context of a clinical trial at this time.

Conclusion

STS remains a major challenge to the medical oncologist, despite the advent of modern systemic treatment strategies, including targeted therapy and immunotherapy. Because STS is a relatively uncommon entity, small numbers of patients have limited the ability to conduct traditional clinical trials and advance drug development. Since 2007, three novel agents including trabectedin, pazopanib and eribulin have been approved for the treatment of high-grade STS in the second-line setting after progression on anthracyclines. Trabectedin has efficacy specific for the L-sarcomas, LMS and LPS. Pazopanib is an orally active VEGF-targeting agent, but not approved for LPS. Meanwhile, eribulin is approved only for LPS, and improves OS in the post-anthracycline setting. In November 2016, the FDA granted accelerated approval for olaratumab, which improves OS in advanced and metastatic, anthracycline-naïve high-grade STS, in combination with doxorubicin.

Further advances will require an improved understanding of the biological differences between STS subtypes, specific biomarkers to elucidate responses to treatment, and mechanisms of resistance. Innovative design of clinical trials is essential to maximize the impact of studies involving this rare disease entity and to understand the unique interplay between tumor, microenvironment and therapeutic interventions. The L-sarcoma trials using eribulin and trabectedin show proof of concept that randomized studies for specific subtypes are feasible and should be further pursued. Towards this aim, correlative endpoints, such as PFS and DCR, with informative biomarkers, will be increasingly important, so that active agents can be identified and future studies will be better informed. In addition to discovering new therapeutic agents, current and future trials to evaluate novel combinations, including immunotherapy combinations, will add to the growing number of options for STS.

Footnotes

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of interest statement: Gino In has no conflict of interests to declare. James Hu is a speaker for Esai Pharmaceuticals.

William Tseng has no conflict of interests to declare.

Contributor Information

Gino K. In, Division of Oncology, Norris Comprehensive Cancer Center, Los Angeles, CA, USA Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.

James S. Hu, Division of Oncology, Norris Comprehensive Cancer Center, Los Angeles, CA, USA Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.

William W. Tseng, Department of Surgery, Section of Surgical Oncology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.

References

  • 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016; 66: 7–30. [DOI] [PubMed] [Google Scholar]
  • 2. Frustaci S, Gherlinzoni F, De Paoli A, et al. Adjuvant chemotherapy for adult soft tissue sarcomas of the extremities and girdles: results of the Italian randomized cooperative trial. J Clin Oncol 2001; 19: 1238–1247. [DOI] [PubMed] [Google Scholar]
  • 3. Le Cesne A, Blay JY, Judson I, et al. Phase II study of ET-743 in advanced soft tissue sarcomas: a European Organisation for the Research and Treatment of Cancer (EORTC) soft tissue and bone sarcoma group trial. J Clin Oncol 2005; 23: 576–584. [DOI] [PubMed] [Google Scholar]
  • 4. Nielsen OS, Judson I, van Hoesel Q, et al. Effect of high-dose ifosfamide in advanced soft tissue sarcomas. A multicentre phase II study of the EORTC Soft Tissue and Bone Sarcoma Group. Eur J Cancer 2000; 36: 61–67. [DOI] [PubMed] [Google Scholar]
  • 5. Palumbo R, Palmeri S, Antimi M, et al. Phase II study of continuous-infusion high-dose ifosfamide in advanced and/or metastatic pretreated soft tissue sarcomas. Ann Oncol 1997; 8: 1159–1162. [DOI] [PubMed] [Google Scholar]
  • 6. Maki RG, Wathen JK, Patel SR, et al. Randomized phase II study of gemcitabine and docetaxel compared with gemcitabine alone in patients with metastatic soft tissue sarcomas: results of sarcoma alliance for research through collaboration study 002 [corrected]. J Clin Oncol 2007; 25: 2755–2763. [DOI] [PubMed] [Google Scholar]
  • 7. Hensley ML, Blessing JA, Degeest K, et al. Fixed-dose rate gemcitabine plus docetaxel as second-line therapy for metastatic uterine leiomyosarcoma: a Gynecologic Oncology Group phase II study. Gynecol Oncol 2008; 109: 323–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Harris SJ, Maruzzo M, Thway K, et al. Metastatic soft tissue sarcoma, an analysis of systemic therapy and impact on survival. 2015. ASCO Annual Meeting, 2015. [Google Scholar]
  • 9. 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 2012; 23: 2763–2770. [DOI] [PubMed] [Google Scholar]
  • 10. Ryan CW, Merimsky O, Agulnik M, et al. PICASSO III: a phase III, placebo-controlled study of doxorubicin with or without palifosfamide in patients with metastatic soft tissue sarcoma. J Clin Oncol 2016; 34: 3898–3905. [DOI] [PubMed] [Google Scholar]
  • 11. Zwelling LA, Silberman L, Estey E. Intercalator-induced, topoisomerase II-mediated DNA cleavage and its modification by antineoplastic antimetabolites. Int J Radiat Oncol Biol Phys 1986; 12: 1041–1047. [DOI] [PubMed] [Google Scholar]
  • 12. O’Bryan RM, Baker LH, Gottlieb JE, et al. Dose response evaluation of adriamycin in human neoplasia. Cancer 1977; 39: 1940–1948. [DOI] [PubMed] [Google Scholar]
  • 13. Borden EC, Amato DA, Rosenbaum C, et al. Randomized comparison of three adriamycin regimens for metastatic soft tissue sarcomas. J Clin Oncol 1987; 5: 840–850. [DOI] [PubMed] [Google Scholar]
  • 14. Judson I, Verweij J, Gelderblom H, et al. Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: a randomised controlled phase III trial. Lancet Oncol 2014; 15: 415–423. [DOI] [PubMed] [Google Scholar]
  • 15. Le Cesne A, Judson I, Crowther D, et al. Randomized phase III study comparing conventional-dose doxorubicin plus ifosfamide versus high-dose doxorubicin plus ifosfamide plus recombinant human granulocyte-macrophage colony-stimulating factor in advanced soft tissue sarcomas: a trial of the European Organization for Research and Treatment of Cancer/Soft Tissue and Bone Sarcoma Group. J Clin Oncol 2000; 18: 2676–2684. [DOI] [PubMed] [Google Scholar]
  • 16. Studzian K, Kinas R, Ciesielska E, et al. Effects of alkylating metabolites of ifosfamide and its bromo analogues on DNA of HeLa cells. Biochem Pharmacol 1992; 43: 937–943. [DOI] [PubMed] [Google Scholar]
  • 17. Blum RH, Edmonson J, Ryan L, et al. Efficacy of ifosfamide in combination with doxorubicin for the treatment of metastatic soft-tissue sarcoma. The Eastern Cooperative Oncology Group. Cancer Chemother Pharmacol 1993; 31(Suppl. 2): S238–S240. [PubMed] [Google Scholar]
  • 18. Edmonson JH, Ryan LM, Blum RH, et al. Randomized comparison of doxorubicin alone versus ifosfamide plus doxorubicin or mitomycin, doxorubicin, and cisplatin against advanced soft tissue sarcomas. J Clin Oncol 1993; 11: 1269–1275. [DOI] [PubMed] [Google Scholar]
  • 19. Santoro A, Tursz T, Mouridsen H, et al. Doxorubicin versus CYVADIC versus doxorubicin plus ifosfamide in first-line treatment of advanced soft tissue sarcomas: a randomized study of the European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. J Clin Oncol 1995; 13: 1537–1545. [DOI] [PubMed] [Google Scholar]
  • 20. Kroep JR, Ouali M, Gelderblom H, et al. First-line chemotherapy for malignant peripheral nerve sheath tumor (MPNST) versus other histological soft tissue sarcoma subtypes and as a prognostic factor for MPNST: an EORTC Soft Tissue and Bone Sarcoma Group study. Ann Oncol 2011; 22: 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Italiano A, Toulmonde M, Cioffi A, et al. Advanced well-differentiated/dedifferentiated liposarcomas: role of chemotherapy and survival. Ann Oncol 2012; 23: 1601–1607. [DOI] [PubMed] [Google Scholar]
  • 22. Antman K, Crowley J, Balcerzak SP, et al. An intergroup phase III randomized study of doxorubicin and dacarbazine with or without ifosfamide and mesna in advanced soft tissue and bone sarcomas. J Clin Oncol 1993; 11: 1276–1285. [DOI] [PubMed] [Google Scholar]
  • 23. Fayette J, Penel N, Chevreau C, et al. Phase III trial of standard versus dose-intensified doxorubicin, ifosfamide and dacarbazine (MAID) in the first-line treatment of metastatic and locally advanced soft tissue sarcoma. Invest New Drugs 2009; 27: 482–489. [DOI] [PubMed] [Google Scholar]
  • 24. Schoenfeld DA, Rosenbaum C, Horton J, et al. A comparison of adriamycin versus vincristine and adriamycin, and cyclophosphamide versus vincristine, actinomycin-D, and cyclophosphamide for advanced sarcoma. Cancer 1982; 50: 2757–2762. [DOI] [PubMed] [Google Scholar]
  • 25. Choi BW, Berger HJ, Schwartz PE, et al. Serial radionuclide assessment of doxorubicin cardiotoxicity in cancer patients with abnormal baseline resting left ventricular performance. Am Heart J 1983; 106: 638–643. [DOI] [PubMed] [Google Scholar]
  • 26. Ritchie JL, Singer JW, Thorning D, et al. Anthracycline cardiotoxicity: clinical and pathologic outcomes assessed by radionuclide ejection fraction. Cancer 1980; 46: 1109–1116. [DOI] [PubMed] [Google Scholar]
  • 27. Bristow MR, Mason JW, Billingham ME, et al. Dose–effect and structure–function relationships in doxorubicin cardiomyopathy. Am Heart J 1981; 102: 709–718. [DOI] [PubMed] [Google Scholar]
  • 28. Sleijfer S, Ouali M, van Glabbeke M, et al. Prognostic and predictive factors for outcome to first-line ifosfamide-containing chemotherapy for adult patients with advanced soft tissue sarcomas: an exploratory, retrospective analysis on large series from the European Organization for Research and Treatment of Cancer-Soft Tissue and Bone Sarcoma Group (EORTC-STBSG). Eur J Cancer 2010; 46: 72–83. [DOI] [PubMed] [Google Scholar]
  • 29. Penel N, Glabbeke MV, Mathoulin-Pelissier S, et al. Performance status is the most powerful risk factor for early death among patients with advanced soft tissue sarcoma: the European Organisation for Research and Treatment of Cancer-Soft Tissue and Bone Sarcoma Group (STBSG) and French Sarcoma Group (FSG) study. Br J Cancer 2011; 104: 1544–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Van Glabbeke M, van Oosterom AT, Oosterhuis JW, et al. Prognostic factors for the outcome of chemotherapy in advanced soft tissue sarcoma: an analysis of 2,185 patients treated with anthracycline-containing first-line regimens – a European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group Study. J Clin Oncol 1999; 17: 150–157. [DOI] [PubMed] [Google Scholar]
  • 31. Tap W, Papai Z, van Tine B, et al. Randomized phase III, multicenter, open-label study comparing evofosfamide (Evo) in combination with doxorubicin (D) vs. D alone in patients (pts) with advanced soft tissue sarcoma (STS): study TH-CR-406/SARC021. Paper presented at European Society for Medical Oncology (ESMO) 2016 Congress, 7–11 October 2016, Copenhagen, Denmark. [Google Scholar]
  • 32. De Sanctis R, Bertuzzi A, Basso U, et al. Non-pegylated liposomal doxorubicin plus ifosfamide in metastatic soft tissue sarcoma: results from a phase-II trial. Anticancer Res 2015; 35: 543–547. [PubMed] [Google Scholar]
  • 33. Judson I, Radford JA, Harris M, et al. Randomised phase II trial of pegylated liposomal doxorubicin (DOXIL/CAELYX) versus doxorubicin in the treatment of advanced or metastatic soft tissue sarcoma: a study by the EORTC Soft Tissue and Bone Sarcoma Group. Eur J Cancer 2001; 37: 870–877. [DOI] [PubMed] [Google Scholar]
  • 34. Skubitz KM. Phase II trial of pegylated-liposomal doxorubicin (Doxil) in sarcoma. Cancer Inves. 2003; 21: 167–176. [DOI] [PubMed] [Google Scholar]
  • 35. Kummar S, Allen D, Monks A, et al. Cediranib for metastatic alveolar soft part sarcoma. J Clin Oncol 2013; 31: 2296–2302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Schuetze SM, Bolejack V, Choy E, et al. Phase II study of dasatinib in patients with alveolar soft part sarcoma, chondrosarcoma, chordoma, epithelioid sarcoma, or solitary fibrous tumor. Cancer 2017; 123: 90–97. [DOI] [PubMed] [Google Scholar]
  • 37. Sethi TK, Keedy VL. Histology-specific uses of tyrosine kinase inhibitors in non-gastrointestinal stromal tumor sarcomas. Curr Treat Options Oncol 2016; 17: 11. [DOI] [PubMed] [Google Scholar]
  • 38. Stacchiotti S, Negri T, Zaffaroni N, et al. Sunitinib in advanced alveolar soft part sarcoma: evidence of a direct antitumor effect. Ann Oncol 2011; 22: 1682–1690. [DOI] [PubMed] [Google Scholar]
  • 39. Dufresne A, Cassier P, Couraud L, et al. Desmoplastic small round cell tumor: current management and recent findings. Sarcoma 2012; 2012: 5 pages. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. DeVito N, Henderson E, Han G, et al. Clinical characteristics and outcomes for Solitary Fibrous Tumor (SFT): a single center experience. PLoS One 2015;10: e0140362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Rosen G, Forscher C, Lowenbraun S, et al. Synovial sarcoma. Uniform response of metastases to high dose ifosfamide. Cancer 1994; 73: 2506–2511. [DOI] [PubMed] [Google Scholar]
  • 42. Le Cesne A, Antoine E, Spielmann M, et al. High-dose ifosfamide: circumvention of resistance to standard-dose ifosfamide in advanced soft tissue sarcomas. J Clin Oncol 1995; 13: 1600–1608. [DOI] [PubMed] [Google Scholar]
  • 43. Cerny T, Leyvraz S, von Briel T, et al. Saturable metabolism of continuous high-dose ifosfamide with mesna and GM-CSF: a pharmacokinetic study in advanced sarcoma patients. Swiss Group for Clinical Cancer Research (SAKK). Ann Oncol 1999; 10: 1087–1094. [DOI] [PubMed] [Google Scholar]
  • 44. Patel SR, Vadhan-Raj S, Papadopolous N, et al. High-dose ifosfamide in bone and soft tissue sarcomas: results of phase II and pilot studies – dose-response and schedule dependence. J Clin Oncol 1997; 15: 2378–2384. [DOI] [PubMed] [Google Scholar]
  • 45. McCune JS, Friedman DL, Schuetze S, et al. Influence of age upon ifosfamide-induced nephrotoxicity. Pediatr Blood Cancer 2004; 42: 427–432. [DOI] [PubMed] [Google Scholar]
  • 46. Rossi R, Godde A, Kleinebrand A, et al. Unilateral nephrectomy and cisplatin as risk factors of ifosfamide-induced nephrotoxicity: analysis of 120 patients. J Clin Oncol 1994; 12: 159–165. [DOI] [PubMed] [Google Scholar]
  • 47. Stohr W, Paulides M, Bielack S, et al. Ifosfamide-induced nephrotoxicity in 593 sarcoma patients: a report from the Late Effects Surveillance System. Pediatr Blood Cancer 2007; 48: 447–452. [DOI] [PubMed] [Google Scholar]
  • 48. Buesa JM, Lopez-Pousa A, Martin J, et al. Phase II trial of first-line high-dose ifosfamide in advanced soft tissue sarcomas of the adult: a study of the Spanish Group for Research on Sarcomas (GEIS). Ann Oncol 1998; 9: 871–876. [DOI] [PubMed] [Google Scholar]
  • 49. Zhang Y, Kawedia JD, Myers AL, et al. Physical and chemical stability of high-dose ifosfamide and mesna for prolonged 14-day continuous infusion. J Oncol Pharm Pract 2014; 20: 51–57. [DOI] [PubMed] [Google Scholar]
  • 50. Elias AD, Eder JP, Shea T, et al. High-dose ifosfamide with mesna uroprotection: a phase I study. J Clin Oncol 1990; 8: 170–178. [DOI] [PubMed] [Google Scholar]
  • 51. Yalcin B, Pamir A, Buyukcelik A, et al. High-dose ifosfamide with hematopoietic growth factor support in advanced bone and soft tissue sarcomas. Exp Oncol 2004; 26: 320–325. [PubMed] [Google Scholar]
  • 52. Antman KH, Elias A. Dana-Farber Cancer Institute studies in advanced sarcoma. Semin Oncol 1990; 17(1 Suppl. 2): 7–15. [PubMed] [Google Scholar]
  • 53. Lee SH, Chang MH, Baek KK, et al. High-dose ifosfamide as second- or third-line chemotherapy in refractory bone and soft tissue sarcoma patients. Oncology 2011; 80: 257–261. [DOI] [PubMed] [Google Scholar]
  • 54. Martin-Liberal J, Alam S, Constantinidou A, et al. Clinical activity and tolerability of a 14-day infusional ifosfamide schedule in soft-tissue sarcoma. Sarcoma 2013; 2013: 868973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Noujaim J, Constantinidou A, Messiou C, et al. Successful ifosfamide rechallenge in soft-tissue sarcoma. Am J Clin Oncol. Epub ahead of print 29 October 2015. DOI: 10.1097/COC.0000000000000243. [DOI] [PubMed] [Google Scholar]
  • 56. Sanfilippo R, Bertulli R, Marrari A, et al. High-dose continuous-infusion ifosfamide in advanced well-differentiated/dedifferentiated liposarcoma. Clin Sarcoma Res 2014; 4: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Patel SR, Gandhi V, Jenkins J, et al. Phase II clinical investigation of gemcitabine in advanced soft tissue sarcomas and window evaluation of dose rate on gemcitabine triphosphate accumulation. J Clin Oncol 2001; 19: 3483–3489. [DOI] [PubMed] [Google Scholar]
  • 58. Merimsky O, Meller I, Flusser G, et al. Gemcitabine in soft tissue or bone sarcoma resistant to standard chemotherapy: a phase II study. Cancer Chemother Pharmacol 2000; 45: 177–181. [DOI] [PubMed] [Google Scholar]
  • 59. Dileo P, Morgan JA, Zahrieh D, et al. Gemcitabine and vinorelbine combination chemotherapy for patients with advanced soft tissue sarcomas: results of a phase II trial. Cancer 2007; 109: 1863–1869. [DOI] [PubMed] [Google Scholar]
  • 60. Garcia-Del-Muro X, Lopez-Pousa A, Maurel J, et al. Randomized phase II study comparing gemcitabine plus dacarbazine versus dacarbazine alone in patients with previously treated soft tissue sarcoma: a Spanish Group for Research on Sarcomas study. J Clin Oncol 2011; 29: 2528–2533. [DOI] [PubMed] [Google Scholar]
  • 61. Losa R, Fra J, Lopez-Pousa A, et al. Phase II study with the combination of gemcitabine and DTIC in patients with advanced soft tissue sarcomas. Cancer Chemother Pharmacol 2007; 59: 251–259. [DOI] [PubMed] [Google Scholar]
  • 62. Edmonson JH, Ebbert LP, Nascimento AG, et al. Phase II study of docetaxel in advanced soft tissue sarcomas. Am J Clin Oncol 1996; 19: 574–576. [DOI] [PubMed] [Google Scholar]
  • 63. Van Hoesel QG, Verweij J, Catimel G, et al. Phase II study with docetaxel (Taxotere) in advanced soft tissue sarcomas of the adult. EORTC Soft Tissue and Bone Sarcoma Group. Ann Oncol 1994; 5: 539–542. [DOI] [PubMed] [Google Scholar]
  • 64. Hensley ML, Maki R, Venkatraman E, et al. Gemcitabine and docetaxel in patients with unresectable leiomyosarcoma: results of a phase II trial. J Clin Oncol 2002; 20: 2824–2831. [DOI] [PubMed] [Google Scholar]
  • 65. Pautier P, Floquet A, Penel N, et al. Randomized multicenter and stratified phase II study of gemcitabine alone versus gemcitabine and docetaxel in patients with metastatic or relapsed leiomyosarcomas: a Federation Nationale des Centres de Lutte Contre le Cancer (FNCLCC) French Sarcoma Group Study (TAXOGEM study). Oncologist 2012; 17: 1213–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Seddon BM, Whelan J, Strauss SJ, et al. GeDDiS: a prospective randomised controlled phase III trial of gemcitabine and docetaxel compared with doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft tissue sarcomas (EudraCT 2009–014907–29). 2015. ASCO Annual Meeting, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Herrera DAA, Kuk D, Keohan ML, et al. Outcomes of systemic therapy for patients with metastatic undifferentiated pleomorphic sarcoma (UPS). 2016. ASCO Annual Meeting, 2016. [Google Scholar]
  • 68. Hensley ML, Blessing JA, Mannel R, et al. Fixed-dose rate gemcitabine plus docetaxel as first-line therapy for metastatic uterine leiomyosarcoma: a Gynecologic Oncology Group phase II trial. Gynecol Oncol 2008; 109: 329–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Bay JO, Ray-Coquard I, Fayette J, et al. Docetaxel and gemcitabine combination in 133 advanced soft-tissue sarcomas: a retrospective analysis. Int J Cancer 2006; 119: 706–711. [DOI] [PubMed] [Google Scholar]
  • 70. Leu KM, Ostruszka LJ, Shewach D, et al. Laboratory and clinical evidence of synergistic cytotoxicity of sequential treatment with gemcitabine followed by docetaxel in the treatment of sarcoma. J Clin Oncol 2004; 22: 1706–1712. [DOI] [PubMed] [Google Scholar]
  • 71. Oosten AW, Seynaeve C, Schmitz PI, et al. Outcomes of first-line chemotherapy in patients with advanced or metastatic leiomyosarcoma of uterine and non-uterine origin. Sarcoma 2009; 2009: 348910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Lee EM, Rha SY, Lee J, et al. Phase II study of weekly docetaxel and fixed dose rate gemcitabine in patients with previously treated advanced soft tissue and bone sarcoma. Cancer Chemother Pharmacol 2012; 69: 635–642. [DOI] [PubMed] [Google Scholar]
  • 73. Pink D, Richter S, Gerdes S, et al. Gemcitabine and docetaxel for epithelioid sarcoma: results from a retrospective, multi-institutional analysis. Oncology 2014; 87: 95–103. [DOI] [PubMed] [Google Scholar]
  • 74. Van Kesteren C, de Vooght MM, Lopez-Lazaro L, et al. Yondelis (trabectedin, ET-743): the development of an anticancer agent of marine origin. Anticancer Drugs 2003; 14: 487–502. [DOI] [PubMed] [Google Scholar]
  • 75. Pommier Y, Kohlhagen G, Bailly C, et al. DNA sequence- and structure-selective alkylation of guanine N2 in the DNA minor groove by ecteinascidin 743, a potent antitumor compound from the Caribbean tunicate Ecteinascidia turbinata. Biochemistry 1996; 35: 13303–13309. [DOI] [PubMed] [Google Scholar]
  • 76. Zewail-Foote M, Hurley LH. Ecteinascidin 743: a minor groove alkylator that bends DNA toward the major groove. J Med Chem 1999; 42: 2493–2497. [DOI] [PubMed] [Google Scholar]
  • 77. D’Incalci M, Jimeno J. Preclinical and clinical results with the natural marine product ET-743. Expert Opin Investig Drugs 2003;12: 1843–1853. [PubMed] [Google Scholar]
  • 78. Germano G, Frapolli R, Simone M, et al. Antitumor and anti-inflammatory effects of trabectedin on human myxoid liposarcoma cells. Cancer Res 2010; 70: 2235–2244. [DOI] [PubMed] [Google Scholar]
  • 79. Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013; 23: 249–262. [DOI] [PubMed] [Google Scholar]
  • 80. Forni C, Minuzzo M, Virdis E, et al. Trabectedin (ET-743) promotes differentiation in myxoid liposarcoma tumors. Mol Cancer Ther 2009; 8: 449–457. [DOI] [PubMed] [Google Scholar]
  • 81. Le Cesne A, Cresta S, Maki RG, et al. A retrospective analysis of antitumour activity with trabectedin in translocation-related sarcomas. Eur J Cancer 2012; 48: 3036–3044. [DOI] [PubMed] [Google Scholar]
  • 82. Garcia-Carbonero R, Supko JG, Manola J, et al. Phase II and pharmacokinetic study of ecteinascidin 743 in patients with progressive sarcomas of soft tissues refractory to chemotherapy. J Clin Oncol 2004; 22: 1480–1490. [DOI] [PubMed] [Google Scholar]
  • 83. Yovine A, Riofrio M, Blay JY, et al. Phase II study of ecteinascidin-743 in advanced pretreated soft tissue sarcoma patients. J Clin Oncol 2004; 22: 890–899. [DOI] [PubMed] [Google Scholar]
  • 84. Demetri GD, von Mehren M, Jones RL, et al. Efficacy and safety of trabectedin or dacarbazine for metastatic liposarcoma or leiomyosarcoma after failure of conventional chemotherapy: results of a phase III randomized multicenter clinical trial. J Clin Oncol 2016; 34: 786–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Garcia-Carbonero R, Supko JG, Maki RG, et al. Ecteinascidin-743 (ET-743) for chemotherapy-naive patients with advanced soft tissue sarcomas: multicenter phase II and pharmacokinetic study. J Clin Oncol 2005; 23: 5484–5492. [DOI] [PubMed] [Google Scholar]
  • 86. Bui-Nguyen B, Butrynski JE, Penel N, et al. A phase IIb multicentre study comparing the efficacy of trabectedin to doxorubicin in patients with advanced or metastatic untreated soft tissue sarcoma: the TRUSTS trial. Eur J Cancer 2015; 51: 1312–1320. [DOI] [PubMed] [Google Scholar]
  • 87. Grosso F, Jones RL, Demetri GD, et al. Efficacy of trabectedin (ecteinascidin-743) in advanced pretreated myxoid liposarcomas: a retrospective study. Lancet Oncol 2007; 8: 595–602. [DOI] [PubMed] [Google Scholar]
  • 88. Grosso F, Sanfilippo R, Virdis E, et al. Trabectedin in myxoid liposarcomas (MLS): a long-term analysis of a single-institution series. Ann Oncol 2009; 20: 1439–1444. [DOI] [PubMed] [Google Scholar]
  • 89. Sanfilippo R, Grosso F, Virdis E, et al. Rechallenge with trabectedin in patients with responding myxoid liposarcoma. J Clin Oncol 2009; 27(Suppl. 15): abstract 10575. [Google Scholar]
  • 90. Kawai A, Araki N, Sugiura H, et al. Trabectedin monotherapy after standard chemotherapy versus best supportive care in patients with advanced, translocation-related sarcoma: a randomised, open-label, phase 2 study. Lancet Oncol 2015; 16: 406–416. [DOI] [PubMed] [Google Scholar]
  • 91. Blay JY, Leahy MG, Nguyen BB, et al. Randomised phase III trial of trabectedin versus doxorubicin-based chemotherapy as first-line therapy in translocation-related sarcomas. Eur J Cancer 2014; 50: 1137–1147. [DOI] [PubMed] [Google Scholar]
  • 92. Demetri GD, Chawla SP, von Mehren M, et al. Efficacy and safety of trabectedin in patients with advanced or metastatic liposarcoma or leiomyosarcoma after failure of prior anthracyclines and ifosfamide: results of a randomized phase II study of two different schedules. J Clin Oncol 2009; 27: 4188–4196. [DOI] [PubMed] [Google Scholar]
  • 93. Delaloge S, Yovine A, Taamma A, et al. Ecteinascidin-743: a marine-derived compound in advanced, pretreated sarcoma patients – preliminary evidence of activity. J Clin Oncol 2001; 19: 1248–1255. [DOI] [PubMed] [Google Scholar]
  • 94. Calvo E, Azaro A, Dirix L, et al. Trabectedin (T)-related liver toxicity: results of a pharmacokinetic study with T in patients with hepatic dysfunction (OVC1004) and experience from a phase III clinical trial (SAR3007). 2016. ASCO Annual Meeting, 2016. [Google Scholar]
  • 95. De Sanctis R, Marrari A, Marchetti S, et al. Efficacy of trabectedin in advanced soft tissue sarcoma: beyond lipo- and leiomyosarcoma. Drug Des Devel Ther 2015; 9: 5785–5791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Morioka H, Takahashi S, Araki N, et al. Results of sub-analysis of a phase II study on trabectedin treatment for extraskeletal myxoid chondrosarcoma and mesenchymal chondrosarcoma. BMC Cancer 2016; 16: 479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Herzog J, von Klot-Heydenfeldt F, Jabar S, et al. Trabectedin followed by irinotecan can stabilize disease in advanced translocation-positive sarcomas with acceptable toxicity. Sarcoma 2016; 2016: 6 pages. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Chu Q, Mita A, Forouzesh B, et al. Phase I and pharmacokinetic study of sequential paclitaxel and trabectedin every 2 weeks in patients with advanced solid tumors. Clin Cancer Res 2010; 16: 2656–2665. [DOI] [PubMed] [Google Scholar]
  • 99. Italiano A, Laurand A, Laroche A, et al. ERCC5/XPG, ERCC1, and BRCA1 gene status and clinical benefit of trabectedin in patients with soft tissue sarcoma. Cancer 2011; 117: 3445–3456. [DOI] [PubMed] [Google Scholar]
  • 100. Schoffski P, Taron M, Jimeno J, et al. Predictive impact of DNA repair functionality on clinical outcome of advanced sarcoma patients treated with trabectedin: a retrospective multicentric study. Eur J Cancer 2011; 47: 1006–1012. [DOI] [PubMed] [Google Scholar]
  • 101. Chao C, Al-Saleem T, Brooks JJ, et al. Vascular endothelial growth factor and soft tissue sarcomas: tumor expression correlates with grade. Ann Surg Oncol 2001; 8: 260–267. [DOI] [PubMed] [Google Scholar]
  • 102. Pakos EE, Goussia AC, Tsekeris PG, et al. Expression of vascular endothelial growth factor and its receptor, KDR/Flk-1, in soft tissue sarcomas. Anticancer Res 2005; 25: 3591–3596. [PubMed] [Google Scholar]
  • 103. Yoon SS, Segal NH, Olshen AB, et al. Circulating angiogenic factor levels correlate with extent of disease and risk of recurrence in patients with soft tissue sarcoma. Ann Oncol 2004; 15: 1261–1266. [DOI] [PubMed] [Google Scholar]
  • 104. Yudoh K, Kanamori M, Ohmori K, et al. Concentration of vascular endothelial growth factor in the tumour tissue as a prognostic factor of soft tissue sarcomas. Br J Cancer 2001; 84: 1610–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. 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 2009; 27: 3126–3132. [DOI] [PubMed] [Google Scholar]
  • 106. Van der Graaf WT, Blay JY, Chawla SP, et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase III trial. Lancet 2012; 379: 1879–1886. [DOI] [PubMed] [Google Scholar]
  • 107. Coens C, van der Graaf WT, Blay JY, et al. Health-related quality-of-life results from PALETTE: a randomized, double-blind, phase III trial of pazopanib versus placebo in patients with soft tissue sarcoma whose disease has progressed during or after prior chemotherapy-a European Organization for research and treatment of cancer soft tissue and bone sarcoma group global network study (EORTC 62072). Cancer 2015; 121: 2933–2941. [DOI] [PubMed] [Google Scholar]
  • 108. 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 2014; 25: 719–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Koehler K, Liebner D, Chen JL. TP53 mutational status is predictive of pazopanib response in advanced sarcomas. Ann Oncol 2016; 27: 539–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Dembla V, Fu S, Wheler J, et al. Outcomes of patients with advanced sarcoma enrolled in clinical trials of pazopanib in combination with histone deacetylase, mTOR, Her2, or MEK inhibitors. 2016. ASCO Annual Meeting, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Duffaud F, Sleijfer S, Litiere S, et al. Hypertension (HTN) as a potential biomarker of efficacy in pazopanib-treated patients with advanced non-adipocytic soft tissue sarcoma. A retrospective study based on European Organisation for Research and Treatment of Cancer (EORTC) 62043 and 62072 trials. Eur J Cancer 2015; 51: 2615–2623. [DOI] [PubMed] [Google Scholar]
  • 112. Benson C, Ray-Coquard I, Sleijfer S, et al. Outcome of uterine sarcoma patients treated with pazopanib: a retrospective analysis based on two European Organisation for Research and Treatment of Cancer (EORTC) Soft Tissue and Bone Sarcoma Group (STBSG) clinical trials 62043 and 62072. Gynecol Oncol 2016; 142: 89–94. [DOI] [PubMed] [Google Scholar]
  • 113. Yoo KH, Kim HS, Lee SJ, et al. Efficacy of pazopanib monotherapy in patients who had been heavily pretreated for metastatic soft tissue sarcoma: a retrospective case series. BMC Cancer 2015; 15: 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Nakamura T, Matsumine A, Kawai A, et al. The clinical outcome of pazopanib treatment in Japanese patients with relapsed soft tissue sarcoma: a Japanese Musculoskeletal Oncology Group (JMOG) study. Cancer 2016; 122: 1408–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Bally O, Tassy L, Richioud B, et al. Eight years tumor control with pazopanib for a metastatic resistant epithelioid hemangioendothelioma. Clin Sarcoma Res 2015; 5: 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Lee SJ, Kim ST, Park SH, et al. Successful use of pazopanib for treatment of refractory metastatic hemangiopericytoma. Clin Sarcoma Res 2014; 4: 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Maruzzo M, Martin-Liberal J, Messiou C, et al. Pazopanib as first line treatment for solitary fibrous tumours: the Royal Marsden Hospital experience. Clin Sarcoma Res 2015; 5: 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Outani H, Tanaka T, Wakamatsu T, et al. Establishment of a novel clear cell sarcoma cell line (Hewga-CCS), and investigation of the antitumor effects of pazopanib on Hewga-CCS. BMC Cancer 2014; 14: 455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Nakano K, Motoi N, Inagaki L, et al. Differences in the responses to pazopanib and the prognoses of soft tissue sarcomas by their histological eligibility for the PALETTE study. Jpn J Clin Oncol 2015; 45: 449–455. [DOI] [PubMed] [Google Scholar]
  • 120. Karch A, Koch A, Grunwald V. A phase II trial comparing pazopanib with doxorubicin as first-line treatment in elderly patients with metastatic or advanced soft tissue sarcoma (EPAZ): study protocol for a randomized controlled trial. Trials 2016; 17: 312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Kim HK, Kim SY, Lee SJ, et al. BEZ235 (PIK3/mTOR inhibitor) overcomes pazopanib resistance in patient-derived refractory soft tissue sarcoma cells. Transl Oncol 2016; 9: 197–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Fu S, Hou MM, Naing A, et al. Phase I study of pazopanib and vorinostat: a therapeutic approach for inhibiting mutant p53-mediated angiogenesis and facilitating mutant p53 degradation. Ann Oncol 2015; 26: 1012–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Tavallai S, Hamed HA, Grant S, et al. Pazopanib and HDAC inhibitors interact to kill sarcoma cells. Cancer Biol Ther 2014; 15: 578–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Jimeno A. Eribulin: rediscovering tubulin as an anticancer target. Clin Cancer Res 2009; 15: 3903–3905. [DOI] [PubMed] [Google Scholar]
  • 125. Bai RL, Paull KD, Herald CL, et al. Halichondrin B and homohalichondrin B, marine natural products binding in the vinca domain of tubulin. Discovery of tubulin-based mechanism of action by analysis of differential cytotoxicity data. J Biol Chem 1991; 266: 15882–15889. [PubMed] [Google Scholar]
  • 126. Jordan MA, Kamath K, Manna T, et al. The primary antimitotic mechanism of action of the synthetic halichondrin E7389 is suppression of microtubule growth. Mol Cancer Ther 2005; 4: 1086–1095. [DOI] [PubMed] [Google Scholar]
  • 127. Funahashi Y, Okamoto K, Adachi Y, et al. Eribulin mesylate reduces tumor microenvironment abnormality by vascular remodeling in preclinical human breast cancer models. Cancer Sci 2014; 105: 1334–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Yoshida T, Ozawa Y, Kimura T, et al. Eribulin mesilate suppresses experimental metastasis of breast cancer cells by reversing phenotype from epithelial–mesenchymal transition (EMT) to mesenchymal–epithelial transition (MET) states. Br J Cancer 2014; 110: 1497–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Schoffski P, Ray-Coquard IL, Cioffi A, et al. Activity of eribulin mesylate in patients with soft-tissue sarcoma: a phase II study in four independent histological subtypes. Lancet Oncol 2011; 12: 1045–1052. [DOI] [PubMed] [Google Scholar]
  • 130. Kawai A, Araki N, Naito Y, et al. 2017; 47: 137–144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Schoffski P, Chawla S, Maki RG, et al. Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: a randomised, open-label, multicentre, phase III trial. Lancet 2016; 387: 1629–1637. [DOI] [PubMed] [Google Scholar]
  • 132. Cortes J, O’Shaughnessy J, Loesch D, et al. Eribulin monotherapy versus treatment of physician’s choice in patients with metastatic breast cancer (EMBRACE): a phase III open-label randomised study. Lancet 2011; 377: 914–923. [DOI] [PubMed] [Google Scholar]
  • 133. Schöffski P, Maki RG, Italiano A, et al. Randomized, open-label, multicenter, phase III study of eribulin versus dacarbazine in patients (pts) with leiomyosarcoma (LMS) and adipocytic sarcoma (ADI). 2015. ASCO Annual Meeting, 29 May–2 June 2015, Chicago. [Google Scholar]
  • 134. Loizos N, Xu Y, Huber J, et al. Targeting the platelet-derived growth factor receptor alpha with a neutralizing human monoclonal antibody inhibits the growth of tumor xenografts: implications as a potential therapeutic target. Mol Cancer Ther 2005; 4: 369–379. [DOI] [PubMed] [Google Scholar]
  • 135. Adams SF, Hickson JA, Hutto JY, et al. PDGFR-alpha as a potential therapeutic target in uterine sarcomas. Gynecol Oncol 2007; 104: 524–528. [DOI] [PubMed] [Google Scholar]
  • 136. Bai Y, Li J, Fang B, et al. Phosphoproteomics identifies driver tyrosine kinases in sarcoma cell lines and tumors. Cancer Res 2012; 72: 2501–2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Fleming TP, Saxena A, Clark WC, et al. Amplification and/or overexpression of platelet-derived growth factor receptors and epidermal growth factor receptor in human glial tumors. Cancer Res 1992; 52: 4550–4553. [PubMed] [Google Scholar]
  • 138. Matei D, Emerson RE, Lai YC, et al. Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene 2006; 25: 2060–2069. [DOI] [PubMed] [Google Scholar]
  • 139. Stock P, Monga D, Tan X, et al. Platelet-derived growth factor receptor-alpha: a novel therapeutic target in human hepatocellular cancer. Mol Cancer Ther 2007; 6: 1932–1941. [DOI] [PubMed] [Google Scholar]
  • 140. MacDonald TJ, Brown KM, LaFleur B, et al. Expression profiling of medulloblastoma: PDGFRA and the RAS/MAPK pathway as therapeutic targets for metastatic disease. Nat Genet 2001; 29: 143–152. [DOI] [PubMed] [Google Scholar]
  • 141. Alentorn A, Marie Y, Carpentier C, et al. Prevalence, clinico-pathological value, and co-occurrence of PDGFRA abnormalities in diffuse gliomas. Neuro Oncol 2012; 14: 1393–1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003; 299: 708–710. [DOI] [PubMed] [Google Scholar]
  • 143. Ehnman M, Missiaglia E, Folestad E, et al. Distinct effects of ligand-induced PDGFRalpha and PDGFRbeta signaling in the human rhabdomyosarcoma tumor cell and stroma cell compartments. Cancer Res 2013; 73: 2139–2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Russell MR, Liu Q, Fatatis A. Targeting the {alpha} receptor for platelet-derived growth factor as a primary or combination therapy in a preclinical model of prostate cancer skeletal metastasis. Clin Cancer Res 2010; 16: 5002–5010. [DOI] [PubMed] [Google Scholar]
  • 145. Chiorean EG, Sweeney C, Youssoufian H, et al. A phase I study of olaratumab, an anti-platelet-derived growth factor receptor alpha (PDGFRalpha) monoclonal antibody, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014; 73: 595–604. [DOI] [PubMed] [Google Scholar]
  • 146. Doi T, Ma Y, Dontabhaktuni A, et al. Phase I study of olaratumab in Japanese patients with advanced solid tumors. Cancer Sci 2014; 105: 862–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Tap WD, Jones RL, Van Tine BA, et al. Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase Ib and randomised phase II trial. Lancet 2016; 388: 488–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Coley WB., II Contribution to the knowledge of sarcoma. Ann Surg 1891; 14: 199–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015; 350: 207–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013; 499: 214–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Tseng WW, Somaiah N, Engleman EG. Potential for immunotherapy in soft tissue sarcoma. Hum Vaccin Immunother 2014; 10: 3117–3124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Jungbluth AA, Chen YT, Stockert E, et al. Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer 2001; 92: 856–860. [DOI] [PubMed] [Google Scholar]
  • 154. Jungbluth AA, Antonescu CR, Busam KJ, et al. Monophasic and biphasic synovial sarcomas abundantly express cancer/testis antigen NY-ESO-1 but not MAGE-A1 or CT7. Int J Cancer 2001; 94: 252–256. [DOI] [PubMed] [Google Scholar]
  • 155. Pollack SM, Jungbluth AA, Hoch BL, et al. NY-ESO-1 is a ubiquitous immunotherapeutic target antigen for patients with myxoid/round cell liposarcoma. Cancer 2012; 118: 4564–4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Maki RG, Jungbluth AA, Gnjatic S, et al. A pilot study of anti-CTLA4 antibody ipilimumab in patients with synovial sarcoma. Sarcoma 2013; 2013: 168145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 2011; 29: 917–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Tseng WW, Malu S, Zhang M, et al. Analysis of the intratumoral adaptive immune response in well differentiated and dedifferentiated retroperitoneal liposarcoma. Sarcoma 2015; 2015: 547460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Shurell E, Singh AS, Crompton JG, et al. Characterizing the immune microenvironment of malignant peripheral nerve sheath tumor by PD-L1 expression and presence of CD8+ tumor infiltrating lymphocytes. Oncotarget 2016; 7: 64300–64308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. D’Angelo SP, Shoushtari AN, Agaram NP, et al. Prevalence of tumor-infiltrating lymphocytes and PD-L1 expression in the soft tissue sarcoma microenvironment. Hum Pathol 2015; 46: 357–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Kim JR, Moon YJ, Kwon KS, et al. Tumor infiltrating PD1-positive lymphocytes and the expression of PD-L1 predict poor prognosis of soft tissue sarcomas. PLoS One 2013; 8: e82870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Kim C, Kim EK, Jung H, et al. Prognostic implications of PD-L1 expression in patients with soft tissue sarcoma. BMC Cancer 2016; 16: 434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Tawbi HA-H, Burgess MA, Crowley J, et al. Safety and efficacy of PD-1 blockade using pembrolizumab in patients with advanced soft tissue (STS) and bone sarcomas (BS): results of SARC028—a multicenter phase II study. 2016. ASCO Annual Meeting, 2016. [Google Scholar]
  • 164. George S, Barysauskas CM, Solomon S, et al. Phase II study of nivolumab in metastatic leiomyosarcoma of the uterus. 2016. ASCO Annual Meeting, 2016, Chicago, IL. [Google Scholar]

Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES