Skip to main content
JCO Precision Oncology logoLink to JCO Precision Oncology
. 2019 Apr 25;3:PO.18.00247. doi: 10.1200/PO.18.00247

Precision Oncology in Sarcomas: Divide and Conquer

Roberto Carmagnani Pestana 1, Roman Groisberg 1, Jason Roszik 1, Vivek Subbiah 1,
PMCID: PMC7446356  PMID: 32914012

Abstract

Sarcomas are a heterogeneous group of rare malignancies that exhibit remarkable heterogeneity, with more than 50 subtypes recognized. Advances in next-generation sequencing technology have resulted in the discovery of genetic events in these mesenchymal tumors, which in addition to enhancing understanding of the biology, have opened up avenues for molecularly targeted therapy and immunotherapy. This review focuses on how incorporation of next-generation sequencing has affected drug development in sarcomas and strategies for optimizing precision oncology for these rare cancers. In a significant percentage of soft tissue sarcomas, which represent up to 40% of all sarcomas, specific driver molecular abnormalities have been identified. The challenge to evaluate these mutations across rare cancer subtypes requires the careful characterization of these genetic alterations to further define compelling drivers with therapeutic implications. Novel models of clinical trial design also are needed. This shift would entail sustained efforts by the sarcoma community to move from one-size-fits-all trials, in which all sarcomas are treated similarly, to divide-and-conquer subtype-specific strategies.

INTRODUCTION

Sarcomas are a heterogeneous group of mesenchymal malignancies that comprise less than 1% of adult and 12% of pediatric cancers.1,2 The WHO has defined more than 50 sarcoma subtypes, including a wide array of tumors that arise from adipose, muscular, bone, cartilage, and vascular tissues.3 Treatment options for patients with advanced soft tissue sarcomas (STSs) are limited, and a one-size-fits-all paradigm has prevailed despite the diverse nature of STSs. For metastatic STSs, anthracycline-based chemotherapy remains the backbone of first-line treatment regimens.4 However, efficacy is limited, with a median progression-free survival (mPFS) of approximately 6 months, and the incidence of treatment-associated adverse events (AEs) is high.5 Therefore, more individualized treatment options are critical to improve the survival and quality of life of this patient population.

Reflecting STS heterogeneity, multiple molecular pathways are implicated in the development and progression of these cancers. The characterization of specific genetic aberrations has led to the identification of novel diagnostic, prognostic, and predictive biomarkers.6 An understanding of the prevalence and function of specific genetic alterations in STS subtypes is critical to developing more-effective diagnostic tests and therapeutic approaches. Targeted therapies have the potential to produce significant tumor response by disrupting molecular pathways that drive oncogenesis, thus providing highly personalized treatment.7 A recent report by Lucchesi et al8 analyzed 584 patients with STS in the American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) database and identified that 41% of patients harbored a genetic alteration with potential to influence therapy. Accordingly, a single-center report by Boddu et al9 analyzed 114 patients with sarcoma and identified that 49% carried a mutation deemed actionable; 15 patients had next-generation sequencing (NGS)–guided therapies, with 26% of these achieving clinical benefit (Table 1). This review focuses on how incorporation of NGS has affected drug development in sarcomas and strategies for optimizing precision oncology for these rare cancers.

TABLE 1.

Studies That Evaluated NGS in Patients With Sarcoma

graphic file with name PO.18.00247t1.jpg

SARCOMA SUBTYPES

Traditionally, STSs have been classified on the basis of a presumptive tissue of origin or unique architectural pattern.3 However, molecular diagnostics have enhanced our understanding of the complex morphologic-genetic associations of STSs, establishing the foundation for a precision approach to therapy. Indeed, the current classification system was designed with meticulous correlation of recurrent molecular alterations with discrete histologic subtypes.15 Of the STS subtypes, the most prevalent in adults are undifferentiated/unclassified STS, undifferentiated pleomorphic sarcoma (UPS), liposarcoma, and leiomyosarcoma (Fig 1).

FIG 1.

FIG 1.

Prevalent sarcoma subtypes in the Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange database (n = 584). NOS, not otherwise specified.

A number of subtypes are associated with specific chromosomal translocations. Molecular approaches to detect the protein products of these fusion genes, such as fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction, can aid in the diagnosis of these tumors. For example, myxoid liposarcomas are characterized by reciprocal t(12;16)(q32;q16) translocation between the DDIT3(CHOP) and FUS genes, and synovial sarcomas demonstrate the translocation t(X;18)(p11.2;q11.2), which results in the fusion of the SS18 and SSX genes.16 A prospective, multicenter, French Sarcoma Group observational study demonstrated that diagnosis was modified after molecular genetics analysis for 53 (14%) of 384 patients with STS, and it has been argued that such testing should be considered mandatory.17

Previous studies have described the occurrence of potential driver mutations in a significant fraction of patients with sarcoma (Table 2), and these alterations are often type specific.8,18 For illustration, well-differentiated/dedifferentiated liposarcomas have a simple genomic profile characterized by amplification of the MDM2 and CDK4 genes.19 Alternatively, ALK fusions are observed in 50% of inflammatory myofibroblastic tumor (IMTs), whereas a subset of non-ALK–rearranged IMTs carry fusions in other tyrosine kinases, such as ROS1 or NTRK.20,21 These examples emphasize the heterogeneity of STSs and the importance of understanding the underlying biologic differences to enable rational therapeutic interventions.

TABLE 2.

Specific Targets and Associated Sarcoma Subtypes

graphic file with name PO.18.00247t2.jpg

CONVENTIONAL THERAPY

For most patients with advanced STS, systemic therapy is administered with palliative intent to alleviate symptoms and extend overall survival (OS). The majority of clinical trials that have assessed chemotherapy for advanced STSs have included heterogeneous populations, which complicates the assessment of clinical activity in specific subtypes.

Anthracyclines remain the most-used agents in the first-line treatment of metastatic STS. Several phase II/III trials have evaluated the efficacy of doxorubicin as a single agent, with objective response rates (ORRs) of approximately 10% to 30% and median OS of 8 to 17 months.123-125 In most prospective trials, anthracycline-based combination regimens, such as doxorubicin with ifosfamide, are associated with a higher ORR compared with single-agent doxorubicin, although in general, combinations have not prolonged OS.125 One exception is the combination of doxorubicin and olaratumab, which led to improved OS compared with doxorubicin as a single agent in a randomized phase II trial (26.5 v 14.7 months).5 Of note, these anthracycline-containing regimens are associated with considerable toxicity, with up to 70% of patients experiencing grade-3-or-higher AEs.125 The combination of gemcitabine and docetaxel is another widely used regimen for the treatment of STS.124,126 In a retrospective analysis by the French Sarcoma Group that involved 133 patients with STSs (leiomyosarcomas, [n = 76]; other subtypes, [n = 57]) treated with gemcitabine and docetaxel, the ORR was 18% and median OS was 12 months.127

Eribulin, trabectedin, and pazopanib are approved as later lines of treatment. Eribulin, an antimicrotubule agent, was shown to improve median OS in a multicenter phase III trial with dacarbazine as control.128 Of interest, benefit was limited to patients with liposarcoma.128 Trabectedin, a synthetic alkaloid derived from the Caribbean tunicate Ecteinascidia turbinata, was approved by the Food and Drug Administration (FDA) for patients with advanced liposarcoma and leiomyosarcoma who received prior anthracycline-based regimens on the basis of a phase II clinical trial that demonstrated its superiority to dacarbazine.129 Retrospective reports have shown particular benefit of therapy with trabectedin for myxoid liposarcoma (ORR, 51%; disease control rate [DCR], 90%), which achieved an mPFS of 14 months in patients who received multiple lines of treatment.130 Pazopanib is a multitargeted, small-molecule tyrosine kinase inhibitor (TKI). A randomized phase III trial compared pazopanib with placebo in patients with a variety of STS subtypes who had experienced disease progression after first-line anthracycline-containing therapy; a significantly prolonged mPFS in the pazopanib arm (4.6 v 1.6 months) was demonstrated, with benefit consistent across subgroups.131,132

SARCOMA FRUIT BOWL THEORY: DIVIDE AND CONQUER

Clinical trial design and, thus, treatment strategies need to expand as our capability of examining the genome, proteome, transcriptome, and immunophenotype of tumors continues to advance. With the increase in our understanding of the molecular underpinnings of sarcoma, the time is right to divide and conquer. This will allow us to move away from the one-size-fits-all or fruit bowl theory (the current paradigm) in which most patients with sarcoma receive conventional treatment that is not based on molecular testing to individual, customized therapy for specific sarcoma subtypes on the basis of biology (the future paradigm; Fig 2).133 The challenge that could be posed under this new model is performing studies in rare subsets of an already rare disease. However, as has been recently demonstrated in other rare tumor types that have led to FDA-approved therapies, it is possible to establish efficacy with low patient numbers. For instance, vemurafenib was approved recently in BRAFV600-positive Erdheim-Chester disease on the basis of results in fewer than 25 patients.134,135

FIG 2.

FIG 2.

Fruit bowl theory for customized therapy in sarcoma.

STRATEGIES TO BUILD EVIDENCE

The evaluation of targeted therapies can be challenging when the mutations are rare and present in different histologies. Therefore, innovative clinical trial designs are required to determine clinical benefit of new therapies in patients with rare STS subtypes.

Basket trials are an emerging model of trial design centered on the assumption that the existence of a specific molecular aberration or biomarker predicts the benefit of targeted therapies, regardless of cancer tissue of origin.136 The success of basket trials predominantly depends on the strength of the evidence that demonstrates tumor dependence on the targeted pathways and on reliable inhibition of the target by the drug.136

One example is the basket trial of vemurafenib for tumors that harbor a BRAFV600 mutation.71 Vemurafenib is an orally available TKI of BRAF, with higher selectivity for the BRAFV600 mutant form, that had been approved previously for patients with BRAFV600E mutation–positive metastatic melanoma.137 In this trial with six prespecified cohorts, there was one anecdotal response in a patient with clear-cell sarcoma. The ongoing National Cancer Institute Molecular Analysis for Therapy Choice trial is another ambitious illustration of novel trial strategies for the development of precision oncology; patients will be paired with a targeted drug that has demonstrated activity against their specific driver pathway abnormality, regardless of tumor histology.18

NGS IN SARCOMAS AND TARGETED THERAPY

Massively parallel NGS technology is rapidly being adopted by researchers around the world. In sarcomas, it is being used predominantly as a shotgun-screening tool to seek out novel, recurrent, and potentially actionable mutations. This technology is proving useful in the study of rare tumors because even a few patient samples can yield tremendous understanding of the disease on a molecular level. Some investigators are moving beyond searching for driver mutations and instead asking whether NGS can predict response or resistance to therapy; others are creating expression profiles that go beyond individual genes. NGS has the potential to become, in the coming years, an established platform of choice for researchers who are studying sarcomas.

To date, many investigators have attempted to identify recurrent aberrations using NGS. On the basis of the large variety of identified mutations in cancer-associated pathways, these are predominantly secondary mutations that occur later in the course of tumorigenesis. They may be responsible for accelerated growth in advanced disease but are unlikely to be the sole cause of the malignancy. Identification of these mutations is important, and with so much diversity in involved pathways, an individualized approach is necessary for proper sarcoma treatment.

The majority of mutations detected by NGS will not be drivers and, as such, will not translate into clinical benefit for patients. However, as a result of the decreasing costs of NGS-based commercially available genomic profiling, clinicians are identifying driver mutations and exploiting their therapeutic benefit. For patients with few treatment options, a rationally chosen clinical trial on the basis of NGS-derived data is the best chance for prolonged survival.

To demonstrate the ability of NGS to detect gene fusions, Qadir et al138 designed Child-Seq, a pilot platform that rapidly screens cancer tissue for particular gene fusions characteristic of specific sarcomas. Results demonstrated that the technique could reliably identify gene fusions in their corresponding tumor type without false positives. Although this platform was limited to four particular childhood sarcomas, it is legitimate to conceptualize a more robust tool that could identify dozens or even hundreds of fusions. In fact, currently there are commercially available tools for molecular tissue testing that include detection of fusions; identification of pathognomonic genetic alterations could enable a clinical diagnosis that is based solely on sequencing data. The potential to identify fusions by cell-free DNA testing also is promising. Shukla et al139 demonstrated the feasibility of detecting EWSR1 fusions in plasma derived cell-free DNA from patients with Ewing sarcoma and desmoplastic small round-cell tumor.

In general, NGS has proven to be a highly reliable technology. However, Varga et al140 reported a case of undifferentiated sarcoma in which a 236-gene NGS panel showed pathogenic mutations in BRCA2 and MLH1. The patient was counseled about the implications for herself and her family members. However, Sanger sequencing of the patient’s tumor as well as her father’s germline testing revealed that the MLH1 mutation was a variant of unknown significance rather than a pathogenic mutation. Additional discussions with the NGS laboratory revealed that its algorithms had flagged the mutation for review, but human error led to the release of this erroneous finding. Although this is a single case report, it highlights that even very robust technologies are subject to error.

NGS for Prediction

NGS has the potential to identify specific molecular subtypes with particular sensitivity or resistance to approved treatments. To date, however, there have been few examples of NGS being used as a predictive tool in sarcomas, and it important to recognize that the examples discussed herein are preliminary and should be interpreted with caution. Undoubtedly, as more patients undergo sequencing as part of their clinical care, patterns of response to therapy will emerge.

Koehler et al141 retrospectively reviewed 19 patients treated with pazopanib who also received NGS of their tumors. The authors found that the mPFS was significantly longer in patients with TP53 mutant advanced STS (208 days) than in those with TP53 wild-type tumors (136 days).

Lim et al142 performed NGS-based comprehensive genomic profiling on 39 paired samples from tumor and normal tissue from patients treated with everolimus. The patients’ tumor histology varied, but seven patients with sarcomas were included. The authors reported 22 patients with clinical benefit after treatment with everolimus, and 10 of these patients had aberrations in the mammalian target of rapamycin signaling pathway. Conversely, patients with mutations in chromatin remodeling genes and in receptor tyrosine kinase signaling universally did not respond to everolimus. These hypothesis-generating data reinforce the possibility of using NGS as a predictive tool in response to therapy.

Designing Clinical Trials on the Basis of NGS

Wang et al143 performed a retrospective study of 75 patients with sarcomas who were referred to the Clinical Center for Targeted Therapy at MD Anderson Cancer Center. Patients underwent commercially available NGS of their tumors. Only 54 patients enrolled in a trial, but 13 were treated in multiple trials, with 93 total treatments given. Although few responses were seen, patients who received a gene aberration targeted therapy had a better DCR than those who received other types of therapy, which translated into significantly longer mPFS (5.8 v 1.9 months) and OS (15.9 v 8.7 months).

Chang et al144 performed complete multiomics studies on 59 childhood tumors referred to the National Institutes of Health. The majority of these tumors were sarcomas. Multiomics studies included whole-exome sequencing; germline, whole-transcriptome sequencing; and single-nucleotide polymorphism array analysis of the tumor. Germline cancer-associated gene mutations were reported in seven patients. The authors found targeted therapy–matched clinical trials for 44% of the patients on the basis of their mutational status. One patient had an epithelioid IMT that was identified to contain an RANBP2-anaplastic lymphoma kinase (ALK) fusion; he experienced a complete response to crizotinib for 8 months.

Harris et al13 applied the same approach as Chang et al144 to 89 pediatric tumors. As expected in a pediatric population, sarcomas were the most common malignancy. Patients underwent targeted NGS, first of 41 and then of 275 cancer-related genes. Treatment recommendations were made by multidisciplinary expert panel. The panel was able to make a targeted therapy recommendation in 31% of patients. Only three patients received matched targeted therapy, but none had an objective response. This lack of response may have been due to the lower-quality preclinical evidence on which two of the recommendations were based.

Worst et al145 conducted the first true prospective NGS-based trial with therapeutic intent. The Individualized Therapy for Relapsed Malignancies in Childhood study was a multicenter German effort to identify therapeutic targets in individual pediatric cancers. As with Harris et al,13 a multidisciplinary expert panel wrote a final recommended target report. In this pilot study, 57 patients were enrolled from 20 centers; approximately one half had sarcomas. One half of the patients harbored a potentially druggable alteration, and 10 patients went on to receive targeted therapy. The estimated final cost was relatively modest at approximately 7,000 euros. However, because this was a pilot study designed to assess feasibility, there was no follow-up of patients who were treated with personalized targeted therapy and outcomes were not reported. The authors described one example of an IMT that was found to have a CARS-ALK fusion. The patient was treated with an ALK inhibitor with at least a partial response and continued with treatment 26 months later. We hope that future iterations of this trial will follow patients longitudinally and report on PFS and OS.

PRECISION IMMUNOTHERAPY FOR SARCOMAS

After more than a century since Coley’s report of complete regression of sarcomas secondary to severe episodes of erysipelas146, significant efforts have been made to incorporate immunotherapy in sarcoma treatment. Early results with the use of immune checkpoint inhibitors, however, have not been encouraging across all subtypes. Therefore, the development of immunotherapy for sarcomas also benefits from a precision oncology approach both in identifying predictive biomarkers and in developing strategies targeted to specific antigens.

In the phase II SARC028 trial (ClinicalTrials.gov identifier: NCT02301039) of pembrolizumab, an ORR of 18% was seen in STSs, with a 12-week PFS of 55%.92,147 Of note, this trial enrolled 40 patients with STS, including four tumor types; subgroup analysis of the results identified encouraging activity in UPS and dedifferentiated liposarcoma but not in other cohorts. The relevance of histology for immunotherapy efficacy in sarcomas has been highlighted by other reports. Combination of nivolumab and ipilimumab has provided promising efficacy in certain STS subtypes, with ORR and mPFS in the range of those produced by standard-of-care options; responses were observed in patients with leiomyosarcoma, myxofibrosarcoma, UPS, and angiosarcoma.148 Furthermore, activity in specific subtypes has been suggested by retrospective studies. In a case series, two patients with chordoma experienced responses to single-agent anti–programmed cell death ligand 1 (PD-L1) antibodies,149 and a review of patients enrolled in early-phase trials demonstrated that alveolar soft-part sarcoma was the most responsive subgroup to checkpoint blockade.91,150 The heterogeneity in the benefit of checkpoint inhibitors across sarcoma subtypes observed in these early-phase trials highlights the need for a precision approach to immunotherapy in these diverse tumors, and research to identify predictive biomarkers is warranted. One challenge is that at this time, the immune microenvironment of specific subtypes is not sufficiently characterized, and detailed characterization of the immune microenvironment in each subtype is a major task.

Examples of potential predictive biomarkers to allow for selection of sarcomas sensitive to immune checkpoint inhibition include microsatellite instability (MSI) status and PD-L1 expression in the tumor and microenvironment. In fact, one patient with sarcoma was included within the five trials that led to FDA approval of pembrolizumab for MSI-high tumors.151 A total of 149 patients were included in these trials, the majority (n = 90) with colorectal cancer, and results demonstrated an ORR of 39.6%, with 7.4% of patients achieving a complete response. In addition, responses were long lasting, with 78% lasting for 6 months or more.151 A recently published pan-cancer analysis identified that 5.7% of 785 STS cases analyzed were MSI high, which highlights the relevance of this indication for STS.152 With regard to PD-L1 status, a recent meta-analysis with data from 1,451 patients and 15 independent studies identified PD-L1 expression to be independently associated with poorer OS and event-free survival in bone and STSs.153 However, the predictive value of PD-L1 in selecting patients with sarcoma for immune checkpoint inhibition is still under evaluation, and clinical activity of checkpoint inhibitors in these trials were seen even in the absence of PD-L1 expression.92

Furthermore, cancer/testis antigens have been explored as potential targets for immunotherapy in STS and provide the utmost example of precision immunotherapy by targeting specific antigens. D’Angelo et al94 recently reported encouraging results with the adoptive transfer of NY-ESO-1 c259T cells in 12 patients with synovial sarcoma. The ORR in this study was 50%, including one complete response, and mPFS was 15 weeks. Moreover, treatment was safe, and there were no fatal AEs. These results are promising and relevant for a larger population because NY-ESO-1 is expressed in 70% to 80% of synovial sarcomas and is present in myxoid liposarcoma and malignant peripheral nerve sheath tumors.95,154 Expanding on this concept, adoptive cell therapy targeting MAGE-A3, MAGE-A4, and MAGE-A10 also are under evaluation in clinical trials.

GI STROMAL TUMOR: AN EXAMPLE OF THE PARADIGM SHIFT

Receptor tyrosine kinase activation in GI stromal tumor (GIST) is an emblematic example in which molecular characterization provides diagnostic, prognostic, and predictive information and enables improved outcomes.155 GISTs are intrinsically resistant to radiation and chemotherapy, being historically associated with poor prognosis because of the lack of effective treatment.156 For illustration, before the 1990s, recurrence rates reached 50% after resection, and median OS for metastatic GIST was approximately 9 months.157

In the 1990s, the landmark description of gain-of-function mutations in KIT in five patients with GIST inaugurated a new era.158 Since then, many lines of evidence have also supported the causative relationship between KIT and GIST oncogenesis and have demonstrated that 70% to 85% of GISTs carry a KIT mutation that renders constitutionally activated kinase.159 The most common mutation occurs in exon 11, which encodes the juxtamembrane domain that physiologically inhibits the kinase activation loop.159 In addition, mutations have been described in exons 8 and 9, which encode the extracellular domains, and less frequently in exons 13 and 17, which encode the kinase domains.159 Approximately 30% of KIT wild-type GISTs harbor activating mutations in platelet-derived growth factor-α (PDGFRα).67 The majority of these mutations occur in exon 18, although mutations in exons 12 and 14 also have been described.67 Moreover, 10% to 15% of GISTs are wild type for KIT, and PDGFRα-BRAF mutations have been identified in 7% to 15% of these tumors, most commonly the exon 15 V600E mutation, and a small subset of these tumors are associated with loss of function of the succinate dehydrogenase respiratory chain complex.31 Figure 3 illustrates the mutation landscape of patients with GIST in the AACR Project GENIE database.

FIG 3.

FIG 3.

Mutation landscape of patients with GI stromal tumor in the Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange database (n = 169).

The development of molecular-targeted therapy has dramatically changed the prognosis for GIST; currently, median OS for metastatic disease is 5 years, and 26% to 35% of patients survive for 9 years.160 Overall, trials of imatinib have demonstrated a DCR of 70% to 85% for KIT-mutated GIST, with an mPFS of 20 to 24 months.84 TKIs have revolutionized the treatment of localized GIST as well. Adjuvant therapy with imatinib for 3 years is the current standard of care for patients at high risk of recurrence, which is based on phase III data that demonstrated improved 5-year recurrence-free survival and OS rates compared with patients who received imatinib for 1 year (recurrence-free survival, 65.6% v 47.9%; OS, 92.0% v 81.7%).161

Subanalyses of clinical trials also have been able to identify predictive factors of imatinib resistance. Patients with KIT exon 11 mutations have been identified as sensitive to imatinib, whereas patients with a KIT exon 9 mutation, identified in 10% to 20%, respond poorly to imatinib at standard doses.155

Most patients, however, eventually will develop resistance to treatment. In spite of the exceptional advances with first-line imatinib, the results with targeted treatments for later lines of therapy have been disappointing, emphasizing the need for new therapeutic approaches. Imatinib dose escalation is one option for patients who have experienced disease progression on imatinib, and approximately 20% remain progression free for 1 year at the higher dose, especially among those with exon 9 mutations.162 Furthermore, additional TKIs are under development to optimize the treatment of imatinib-resistant GIST. Demetri et al163 demonstrated that sunitinib, an orally available multitarget TKI, was active in patients with GIST who had disease progression on imatinib, achieving an mPFS of 27.3 weeks compared with 6.4 weeks for placebo. In addition, regorafenib was recently approved by the FDA as a third-line therapy and has shown promising activity in patients with exon 17 mutations, which confer resistance to imatinib and sunitinib. Avapritinib is currently under evaluation after having demonstrated significant activity in patients with PDGFRα D842V exon 18 mutation.164,165

Available evidence indicates that KIT mutation is likely an early event in the development of GIST as suggested by the presence of activating KIT mutations in gastric precursor lesions. However, additional genetic mutations are needed for the progression of microscopic lesions to malignant GIST, and further characterization of these alterations may identify additional therapeutic targets. In a previous study, 74% of GISTs analyzed had at least one nondriver genetic abnormality.164 In that report, the most frequently mutated genes were TP53, RB1, SETD2, PTEN, MAX, PIK3CA, and TSC1, and the most prevalent copy number alteration in KIT-mutated GIST was CDKN2A deletion.164 In addition, activation of alternate receptor tyrosine kinases has been suggested as a mechanism of resistance to currently approved TKIs. For illustration, previous studies have identified that mutations that involve the RB1 gene are associated with high-risk tumors and aggressive clinical behavior and that activating mutations in the RAS and phosphatidylinositol 3-kinase pathways contribute to TKI resistance.166,167 Figure 4 illustrates co-occurring genetic alterations in KIT-mutated GIST in the AACR Project GENIE database.

FIG 4.

FIG 4.

Co-occurring alterations in KIT-mutated GI stromal tumor in the Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange database (n = 31).

In conclusion, sarcomas are a rare group of mesenchymal tumors in which the integration of NGS for diagnosis and management has provided informative evidence for precision medicine. In a significant percentage of STSs, which represent up to 40% of all sarcomas, specific driver molecular abnormalities have been identified. The challenge to evaluate these mutations across rare cancer subtypes requires the careful characterization of these genetic alterations to further define compelling drivers with therapeutic implications as well as novel models of clinical trial design. This shift would entail sustained efforts by the sarcoma community to move from one-size-fits-all trials to divide-and-conquer subtype-specific strategies.

Footnotes

Supported in part by National Cancer Institute grant P30CA016672 (used the Biostatistics Resource Group) and by the Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy (1U01CA180964).

AUTHOR CONTRIBUTIONS

Conception and design: Roberto Carmagnani Pestana, Roman Groisberg, Vivek Subbiah

Administrative support: Jason Roszik, Vivek Subbiah

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.

Vivek Subbiah

Consulting or Advisory Role: MedImmune

Research Funding: Novartis (Inst), GlaxoSmithKline (Inst), NanoCarrier (Inst), Northwest Biotherapeutics (Inst), Genentech (Inst), Roche (Inst), Berg Pharma (Inst), Bayer AG (Inst), Incyte (Inst), Fujifilm (Inst), PharmaMar (Inst), D3 Oncology Solutions (Inst), Pfizer (Inst), Amgen (Inst), AbbVie (Inst), Multivir (Inst), Blueprint Medicines (Inst), Loxo Oncology (Inst), Vegenics (Inst), Takeda Pharmaceuticals (Inst), Alfasigma (Inst), Agensys (Inst), Idera (Inst), Boston Biomedical (Inst), Inhibrx (Inst), Exelixis (Inst)

Travel, Accommodations, Expenses: PharmaMar, Bayer AG

No other potential conflicts of interest were reported.

REFERENCES

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
  • 2.Ward E, DeSantis C, Robbins A, et al. Childhood and adolescent cancer statistics, 2014. CA Cancer J Clin. 2014;64:83–103. doi: 10.3322/caac.21219. [DOI] [PubMed] [Google Scholar]
  • 3. Fletcher CDM, Bridge JA, Hogendoorn P, et al: WHO Classification of Tumours of Soft Tissue and Bone (ed 4). Geneva, Switzerland, WHO, 2013. [Google Scholar]
  • 4.Dufresne A, Brahmi M, Karanian M, et al. Using biology to guide the treatment of sarcomas and aggressive connective-tissue tumours. Nat Rev Clin Oncol. 2018;15:443–458. doi: 10.1038/s41571-018-0012-4. [DOI] [PubMed] [Google Scholar]
  • 5.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 1b and randomised phase 2 trial. Lancet. 2016;388:488–497. doi: 10.1016/S0140-6736(16)30587-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. doi: 10.1200/PO.18.00261. Wilky BA, Villalobos VM: Emerging role for precision therapy through next-generation sequencing for sarcomas. JCO Precis Oncol doi: 10.1200/PO.18.00261. [DOI] [PubMed] [Google Scholar]
  • 7.Cheng L, Lopez-Beltran A, Massari F, et al. Molecular testing for BRAF mutations to inform melanoma treatment decisions: A move toward precision medicine. Mod Pathol. 2018;31:24–38. doi: 10.1038/modpathol.2017.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lucchesi C, Khalifa E, Laizet Y, et al. Targetable alterations in adult patients with soft-tissue sarcomas: Insights for personalized therapy. JAMA Oncol. 2018;4:1398–1404. doi: 10.1001/jamaoncol.2018.0723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. doi: 10.1200/PO.18.00096. Boddu S, Walko CM, Bienasz S, et al: Clinical utility of genomic profiling in the treatment of advanced sarcomas: A single-center experience. JCO Precis Oncol doi: 10.1200/PO.18.00096. [DOI] [PubMed] [Google Scholar]
  • 10.Cote GM, He J, Choy E. Next-generation sequencing for patients with sarcoma: A single center experience. Oncologist. 2018;23:234–242. doi: 10.1634/theoncologist.2017-0290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Groisberg R, Hong DS, Holla V, et al. Clinical genomic profiling to identify actionable alterations for investigational therapies in patients with diverse sarcomas. Oncotarget. 2017;8:39254–39267. doi: 10.18632/oncotarget.16845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gounder MM, Ali SM, Robinson V, et al: Impact of next-generation sequencing (NGS) on diagnostic and therapeutic options in soft-tissue and bone sarcoma. J Clin Oncol 35, 2017 (suppl; abstr 11001) [Google Scholar]
  • 13.Harris MH, DuBois SG, Glade Bender JL, et al. Multicenter feasibility study of tumor molecular profiling to inform therapeutic decisions in advanced pediatric solid tumors: The Individualized Cancer Therapy (iCat) study. JAMA Oncol. 2016;2:608–615. doi: 10.1001/jamaoncol.2015.5689. [DOI] [PubMed] [Google Scholar]
  • 14.Jour G, Scarborough JD, Jones RL, et al. Molecular profiling of soft tissue sarcomas using next-generation sequencing: A pilot study toward precision therapeutics. Hum Pathol. 2014;45:1563–1571. doi: 10.1016/j.humpath.2014.04.012. [DOI] [PubMed] [Google Scholar]
  • 15.Doyle LA. Sarcoma classification: An update based on the 2013 World Health Organization Classification of Tumors of Soft Tissue and Bone. Cancer. 2014;120:1763–1774. doi: 10.1002/cncr.28657. [DOI] [PubMed] [Google Scholar]
  • 16.Mertens F, Antonescu CR, Mitelman F. Gene fusions in soft tissue tumors: Recurrent and overlapping pathogenetic themes. Genes Chromosomes Cancer. 2016;55:291–310. doi: 10.1002/gcc.22335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Italiano A, Di Mauro I, Rapp J, et al. Clinical effect of molecular methods in sarcoma diagnosis (GENSARC): A prospective, multicentre, observational study. Lancet Oncol. 2016;17:532–538. doi: 10.1016/S1470-2045(15)00583-5. [DOI] [PubMed] [Google Scholar]
  • 18.McNeil C. NCI-MATCH launch highlights new trial design in precision-medicine era. J Natl Cancer Inst. 2015;107:djv193. doi: 10.1093/jnci/djv193. [DOI] [PubMed] [Google Scholar]
  • 19.Somaiah N, Beird HC, Barbo A, et al. Targeted next generation sequencing of well-differentiated/dedifferentiated liposarcoma reveals novel gene amplifications and mutations. Oncotarget. 2018;9:19891–19899. doi: 10.18632/oncotarget.24924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lovly CM, Gupta A, Lipson D, et al. Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions. Cancer Discov. 2014;4:889–895. doi: 10.1158/2159-8290.CD-14-0377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. doi: 10.1097/PAS.0000000000000677. Alassiri A, Ali R, Shen Y, et al: ETV6-NTRK3 is expressed in a subset of ALK-negative inflammatory myofibroblastic tumors. Am J Surg Pathol 40:1051-1061, 2016. [DOI] [PubMed] [Google Scholar]
  • 22.Naing A, LoRusso P, Fu S, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res. 2012;18:2625–2631. doi: 10.1158/1078-0432.CCR-12-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Werner H, Idelman G, Rubinstein M, et al. A novel EWS-WT1 gene fusion product in desmoplastic small round cell tumor is a potent transactivator of the insulin-like growth factor-I receptor (IGF-IR) gene. Cancer Lett. 2007;247:84–90. doi: 10.1016/j.canlet.2006.03.027. [DOI] [PubMed] [Google Scholar]
  • 24.Kalebic T, Tsokos M, Helman LJ. In vivo treatment with antibody against IGF-1 receptor suppresses growth of human rhabdomyosarcoma and down-regulates p34cdc2. Cancer Res. 1994;54:5531–5534. [PubMed] [Google Scholar]
  • 25.Tarn C, Rink L, Merkel E, et al. Insulin-like growth factor 1 receptor is a potential therapeutic target for gastrointestinal stromal tumors. Proc Natl Acad Sci U S A. 2008;105:8387–8392. doi: 10.1073/pnas.0803383105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Subbiah V, Brown RE, Jiang Y, et al. Morphoproteomic profiling of the mammalian target of rapamycin (mTOR) signaling pathway in desmoplastic small round cell tumor (EWS/WT1), Ewing’s sarcoma (EWS/FLI1) and Wilms’ tumor (WT1) PLoS One. 2013;8:e68985. doi: 10.1371/journal.pone.0068985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Endo M, Yamamoto H, Setsu N, et al. Prognostic significance of AKT/mTOR and MAPK pathways and antitumor effect of mTOR inhibitor in NF1-related and sporadic malignant peripheral nerve sheath tumors. Clin Cancer Res. 2013;19:450–461. doi: 10.1158/1078-0432.CCR-12-1067. [DOI] [PubMed] [Google Scholar]
  • 28.Wagner AJ, Malinowska-Kolodziej I, Morgan JA, et al. Clinical activity of mTOR inhibition with sirolimus in malignant perivascular epithelioid cell tumors: Targeting the pathogenic activation of mTORC1 in tumors. J Clin Oncol. 2010;28:835–840. doi: 10.1200/JCO.2009.25.2981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Perez J, Decouvelaere AV, Pointecouteau T, et al. Inhibition of chondrosarcoma growth by mTOR inhibitor in an in vivo syngeneic rat model. PLoS One. 2012;7:e32458. doi: 10.1371/journal.pone.0032458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sanfilippo R, Dei Tos AP, Casali PG. Myxoid liposarcoma and the mammalian target of rapamycin pathway. Curr Opin Oncol. 2013;25:379–383. doi: 10.1097/CCO.0b013e32836227ac. [DOI] [PubMed] [Google Scholar]
  • 31.Wada R, Arai H, Kure S, et al. “Wild type” GIST: Clinicopathological features and clinical practice. Pathol Int. 2016;66:431–437. doi: 10.1111/pin.12431. [DOI] [PubMed] [Google Scholar]
  • 32.Gamie Z, Kapriniotis K, Papanikolaou D, et al. TNF-related apoptosis-inducing ligand (TRAIL) for bone sarcoma treatment: Pre-clinical and clinical data. Cancer Lett. 2017;409:66–80. doi: 10.1016/j.canlet.2017.08.036. [DOI] [PubMed] [Google Scholar]
  • 33.Subbiah V, Brown RE, Buryanek J, et al. Targeting the apoptotic pathway in chondrosarcoma using recombinant human Apo2L/TRAIL (dulanermin), a dual proapoptotic receptor (DR4/DR5) agonist. Mol Cancer Ther. 2012;11:2541–2546. doi: 10.1158/1535-7163.MCT-12-0358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Thomas D, Henshaw R, Skubitz K, et al. Denosumab in patients with giant-cell tumour of bone: An open-label, phase 2 study. Lancet Oncol. 2010;11:275–280. doi: 10.1016/S1470-2045(10)70010-3. [DOI] [PubMed] [Google Scholar]
  • 35.Chen Y, Di Grappa MA, Molyneux SD, et al. RANKL blockade prevents and treats aggressive osteosarcomas. Sci Transl Med. 2015;7:317ra197. doi: 10.1126/scitranslmed.aad0295. [DOI] [PubMed] [Google Scholar]
  • 36.Jones DH, Nakashima T, Sanchez OH, et al. Regulation of cancer cell migration and bone metastasis by RANKL. Nature. 2006;440:692–696. doi: 10.1038/nature04524. [DOI] [PubMed] [Google Scholar]
  • 37.Cupp JS, Miller MA, Montgomery KD, et al. Translocation and expression of CSF1 in pigmented villonodular synovitis, tenosynovial giant cell tumor, rheumatoid arthritis and other reactive synovitides. Am J Surg Pathol. 2007;31:970–976. doi: 10.1097/PAS.0b013e31802b86f8. [DOI] [PubMed] [Google Scholar]
  • 38.Tap WD, Wainberg ZA, Anthony SP, et al. Structure-guided blockade of CSF1R kinase in tenosynovial giant-cell tumor. N Engl J Med. 2015;373:428–437. doi: 10.1056/NEJMoa1411366. [DOI] [PubMed] [Google Scholar]
  • 39.Perez M, Muñoz-Galván S, Jiménez-García MP, et al. Efficacy of CDK4 inhibition against sarcomas depends on their levels of CDK4 and p16ink4 mRNA. Oncotarget. 2015;6:40557–40574. doi: 10.18632/oncotarget.5829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wallander ML, Tripp S, Layfield LJ. MDM2 amplification in malignant peripheral nerve sheath tumors correlates with p53 protein expression. Arch Pathol Lab Med. 2012;136:95–99. doi: 10.5858/arpa.2010-0729-OA. [DOI] [PubMed] [Google Scholar]
  • 41.Wang B, Fang L, Zhao H, et al. MDM2 inhibitor Nutlin-3a suppresses proliferation and promotes apoptosis in osteosarcoma cells. Acta Biochim Biophys Sin (Shanghai) 2012;44:685–691. doi: 10.1093/abbs/gms053. [DOI] [PubMed] [Google Scholar]
  • 42.Carol H, Reynolds CP, Kang MH, et al. Initial testing of the MDM2 inhibitor RG7112 by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer. 2013;60:633–641. doi: 10.1002/pbc.24235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kummar S, Allen D, Monks A, et al. Cediranib for metastatic alveolar soft-part sarcoma. J Clin Oncol. 2013;31:2296–2302. doi: 10.1200/JCO.2012.47.4288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Prochilo T, Savelli G, Bertocchi P, et al. Targeting VEGF-VEGFR pathway by sunitinib in peripheral primitive neuroectodermal tumor, paraganglioma and epithelioid hemangioendothelioma: Three case reports. Case Rep Oncol. 2013;6:90–97. doi: 10.1159/000348429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Valentin T, Fournier C, Penel N, et al. Sorafenib in patients with progressive malignant solitary fibrous tumors: A subgroup analysis from a phase II study of the French Sarcoma Group (GSF/GETO) Invest New Drugs. 2013;31:1626–1627. doi: 10.1007/s10637-013-0023-z. [DOI] [PubMed] [Google Scholar]
  • 46.Davis IJ, McFadden AW, Zhang Y, et al. Identification of the receptor tyrosine kinase c-Met and its ligand, hepatocyte growth factor, as therapeutic targets in clear cell sarcoma. Cancer Res. 2010;70:639–645. doi: 10.1158/0008-5472.CAN-09-1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jun HJ, Lee J, Lim DH, et al. Expression of MET in alveolar soft-part sarcoma. Med Oncol. 2010;27:459–465. doi: 10.1007/s12032-009-9234-8. [DOI] [PubMed] [Google Scholar]
  • 48.Taulli R, Scuoppo C, Bersani F, et al. Validation of met as a therapeutic target in alveolar and embryonal rhabdomyosarcoma. Cancer Res. 2006;66:4742–4749. doi: 10.1158/0008-5472.CAN-05-4292. [DOI] [PubMed] [Google Scholar]
  • 49.Torres KE, Zhu Q-S, Bill K, et al. Activated MET is a molecular prognosticator and potential therapeutic target for malignant peripheral nerve sheath tumors. Clin Cancer Res. 2011;17:3943–3955. doi: 10.1158/1078-0432.CCR-11-0193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang K, Zhuang Y, Liu C, et al. Inhibition of c-Met activation sensitizes osteosarcoma cells to cisplatin via suppression of the PI3K-Akt signaling. Arch Biochem Biophys. 2012;526:38–43. doi: 10.1016/j.abb.2012.07.003. [DOI] [PubMed] [Google Scholar]
  • 51.Fleuren ED, Roeffen MH, Leenders WP, et al. Expression and clinical relevance of MET and ALK in Ewing sarcomas. Int J Cancer. 2013;133:427–436. doi: 10.1002/ijc.28047. [DOI] [PubMed] [Google Scholar]
  • 52.Wagner AJ, Chugh R, Rosen LS, et al. A phase I study of the HSP90 inhibitor retaspimycin hydrochloride (IPI-504) in patients with gastrointestinal stromal tumors or soft-tissue sarcomas. Clin Cancer Res. 2013;19:6020–6029. doi: 10.1158/1078-0432.CCR-13-0953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Park H, Turkalo TK, Nelson K, et al. Ewing sarcoma EWS protein regulates midzone formation by recruiting aurora B kinase to the midzone. Cell Cycle. 2014;13:2391–2399. doi: 10.4161/cc.29337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nair JS, Schwartz GK. MLN-8237: A dual inhibitor of aurora A and B in soft tissue sarcomas. Oncotarget. 2016;7:12893–12903. doi: 10.18632/oncotarget.7335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Murahari S, Jalkanen AL, Kulp SK, et al. Sensitivity of osteosarcoma cells to HDAC inhibitor AR-42 mediated apoptosis. BMC Cancer. 2017;17:67. doi: 10.1186/s12885-017-3046-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sakimura R, Tanaka K, Yamamoto S, et al. The effects of histone deacetylase inhibitors on the induction of differentiation in chondrosarcoma cells. Clin Cancer Res. 2007;13:275–282. doi: 10.1158/1078-0432.CCR-06-1696. [DOI] [PubMed] [Google Scholar]
  • 57.Ou W-B, Zhu J, Eilers G, et al. HDACi inhibits liposarcoma via targeting of the MDM2-p53 signaling axis and PTEN, irrespective of p53 mutational status. Oncotarget. 2015;6:10510–10520. doi: 10.18632/oncotarget.3230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kashima K, Watanabe M, Sato Y, et al. Inhibition of metastasis of rhabdomyosarcoma by a novel neutralizing antibody to CXC chemokine receptor-4. Cancer Sci. 2014;105:1343–1350. doi: 10.1111/cas.12490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wakamatsu T, Naka N, Sasagawa S, et al. Deflection of vascular endothelial growth factor action by SS18-SSX and composite vascular endothelial growth factor- and chemokine (C-X-C motif) receptor 4-targeted therapy in synovial sarcoma. Cancer Sci. 2014;105:1124–1134. doi: 10.1111/cas.12469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Berning P, Schaefer C, Clemens D, et al. The CXCR4 antagonist plerixafor (AMD3100) promotes proliferation of Ewing sarcoma cell lines in vitro and activates receptor tyrosine kinase signaling. Cell Commun Signal. 2018;16:21. doi: 10.1186/s12964-018-0233-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ebb D, Meyers P, Grier H, et al. Phase II trial of trastuzumab in combination with cytotoxic chemotherapy for treatment of metastatic osteosarcoma with human epidermal growth factor receptor 2 overexpression: A report from the Children’s Oncology Group. J Clin Oncol. 2012;30:2545–2551. doi: 10.1200/JCO.2011.37.4546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nuciforo PG, Pellegrini C, Fasani R, et al. Molecular and immunohistochemical analysis of HER2/neu oncogene in synovial sarcoma. Hum Pathol. 2003;34:639–645. doi: 10.1016/s0046-8177(03)00238-7. [DOI] [PubMed] [Google Scholar]
  • 63.Scotlandi K, Manara MC, Hattinger CM, et al. Prognostic and therapeutic relevance of HER2 expression in osteosarcoma and Ewing’s sarcoma. Eur J Cancer. 2005;41:1349–1361. doi: 10.1016/j.ejca.2005.03.015. [DOI] [PubMed] [Google Scholar]
  • 64.Aloe L, Rocco ML, Balzamino BO, et al. Nerve growth factor: Role in growth, differentiation and controlling cancer cell development. J Exp Clin Cancer Res. 2016;35:116. doi: 10.1186/s13046-016-0395-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ugurel S, Mentzel T, Utikal J, et al. Neoadjuvant imatinib in advanced primary or locally recurrent dermatofibrosarcoma protuberans: A multicenter phase II DeCOG trial with long-term follow-up. Clin Cancer Res. 2014;20:499–510. doi: 10.1158/1078-0432.CCR-13-1411. [DOI] [PubMed] [Google Scholar]
  • 66.De Pas T, Toffalorio F, Colombo P, et al. Brief report: Activity of imatinib in a patient with platelet-derived-growth-factor receptor positive malignant solitary fibrous tumor of the pleura. J Thorac Oncol. 2008;3:938–941. doi: 10.1097/JTO.0b013e3181803f08. [DOI] [PubMed] [Google Scholar]
  • 67.Corless CL, Schroeder A, Griffith D, et al. PDGFRA mutations in gastrointestinal stromal tumors: Frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol. 2005;23:5357–5364. doi: 10.1200/JCO.2005.14.068. [DOI] [PubMed] [Google Scholar]
  • 68.Üren A, Merchant MS, Sun CJ, et al. Beta-platelet-derived growth factor receptor mediates motility and growth of Ewing’s sarcoma cells. Oncogene. 2003;22:2334–2342. doi: 10.1038/sj.onc.1206330. [DOI] [PubMed] [Google Scholar]
  • 69.Ehnman M, Missiaglia E, Folestad E, et al. Distinct effects of ligand-induced PDGFRα and PDGFRβ signaling in the human rhabdomyosarcoma tumor cell and stroma cell compartments. Cancer Res. 2013;73:2139–2149. doi: 10.1158/0008-5472.CAN-12-1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Knezevich SR, McFadden DE, Tao W, et al. A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet. 1998;18:184–187. doi: 10.1038/ng0298-184. [DOI] [PubMed] [Google Scholar]
  • 71.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 2015;373:726–736. doi: 10.1056/NEJMoa1502309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Moriceau G, Roelofs AJ, Brion R, et al. Synergistic inhibitory effect of apomine and lovastatin on osteosarcoma cell growth. Cancer. 2012;118:750–760. doi: 10.1002/cncr.26336. [DOI] [PubMed] [Google Scholar]
  • 73.Bell JB, Rink JS, Eckerdt F, et al. HDL nanoparticles targeting sonic hedgehog subtype medulloblastoma. Sci Rep. 2018;8:1211. doi: 10.1038/s41598-017-18100-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chan LH, Wang W, Yeung W, et al. Hedgehog signaling induces osteosarcoma development through Yap1 and H19 overexpression. Oncogene. 2014;33:4857–4866. doi: 10.1038/onc.2013.433. [DOI] [PubMed] [Google Scholar]
  • 75.Wang CYY, Wei Q, Han I, et al. Hedgehog and Notch signaling regulate self-renewal of undifferentiated pleomorphic sarcomas. Cancer Res. 2012;72:1013–1022. doi: 10.1158/0008-5472.CAN-11-2531. [DOI] [PubMed] [Google Scholar]
  • 76.Pressey JG, Anderson JR, Crossman DK, et al. Hedgehog pathway activity in pediatric embryonal rhabdomyosarcoma and undifferentiated sarcoma: A report from the Children’s Oncology Group. Pediatr Blood Cancer. 2011;57:930–938. doi: 10.1002/pbc.23174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ban J, Aryee DNT, Fourtouna A, et al. Suppression of deacetylase SIRT1 mediates tumor-suppressive NOTCH response and offers a novel treatment option in metastatic Ewing sarcoma. Cancer Res. 2014;74:6578–6588. doi: 10.1158/0008-5472.CAN-14-1736. [DOI] [PubMed] [Google Scholar]
  • 78.Ma Y, Ren Y, Han EQ, et al. Inhibition of the Wnt-β-catenin and Notch signaling pathways sensitizes osteosarcoma cells to chemotherapy. Biochem Biophys Res Commun. 2013;431:274–279. doi: 10.1016/j.bbrc.2012.12.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bi P, Yue F, Karki A, et al. Notch activation drives adipocyte dedifferentiation and tumorigenic transformation in mice. J Exp Med. 2016;213:2019–2037. doi: 10.1084/jem.20160157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Shang H, Braggio D, Lee YJ, et al. Targeting the Notch pathway: A potential therapeutic approach for desmoid tumors. Cancer. 2015;121:4088–4096. doi: 10.1002/cncr.29564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Butrynski JE, D’Adamo DR, Hornick JL, et al. Crizotinib in ALK-rearranged inflammatory myofibroblastic tumor. N Engl J Med. 2010;363:1727–1733. doi: 10.1056/NEJMoa1007056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Aggerholm-Pedersen N, Demuth C, Safwat A, et al. Dasatinib and doxorubicin treatment of sarcoma initiating cells: A possible new treatment strategy. Stem Cells Int. 2016;2016:9601493. doi: 10.1155/2016/9601493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Indovina P, Casini N, Forte IM, et al. SRC family kinase inhibition in Ewing sarcoma cells induces p38 MAP kinase-mediated cytotoxicity and reduces cell migration. J Cell Physiol. 2017;232:129–135. doi: 10.1002/jcp.25397. [DOI] [PubMed] [Google Scholar]
  • 84.Blanke CD, Rankin C, Demetri GD, et al. Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the Kit receptor tyrosine kinase: S0033. J Clin Oncol. 2008;26:626–632. doi: 10.1200/JCO.2007.13.4452. [DOI] [PubMed] [Google Scholar]
  • 85.Hwang J-A, Yang H-M, Hong D-P, et al. Gankyrin is a predictive and oncogenic factor in well-differentiated and dedifferentiated liposarcoma. Oncotarget. 2014;5:9065–9078. doi: 10.18632/oncotarget.2375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shukla N, Somwar R, Smith RS, et al. Proteasome addiction defined in Ewing’s sarcoma is effectively targeted by a novel class of 19S proteasome inhibitors. Cancer Res. 2016;76:4525–4534. doi: 10.1158/0008-5472.CAN-16-1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Skapek SX, Anderson JR, Hill DA, et al. Safety and efficacy of high-dose tamoxifen and sulindac for desmoid tumor in children: Results of a Children’s Oncology Group (COG) phase II study. Pediatr Blood Cancer. 2013;60:1108–1112. doi: 10.1002/pbc.24457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ioffe YJ, Li AJ, Walsh CS, et al. Hormone receptor expression in uterine sarcomas: Prognostic and therapeutic roles. Gynecol Oncol. 2009;115:466–471. doi: 10.1016/j.ygyno.2009.08.014. [DOI] [PubMed] [Google Scholar]
  • 89.Sheikh TN, Patwardhan PP, Cremers S, et al. Targeted inhibition of glutaminase as a potential new approach for the treatment of NF1 associated soft tissue malignancies. Oncotarget. 2017;8:94054–94068. doi: 10.18632/oncotarget.21573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Subbiah V, Westin SN, Wang K, et al. Targeted therapy by combined inhibition of the RAF and mTOR kinases in malignant spindle cell neoplasm harboring the KIAA1549-BRAF fusion protein. J Hematol Oncol. 2014;7:8. doi: 10.1186/1756-8722-7-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. doi: 10.1200/JGO.2017.009993. Conley AP, Trinh VA, Zobniw CM, et al: Positive tumor response to combined checkpoint inhibitors in a patient with refractory alveolar soft-part sarcoma: A case report. J Glob Oncol doi: 10.1200/JGO.2017.009993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tawbi HA, Burgess M, Bolejack V, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): A multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18:1493–1501. doi: 10.1016/S1470-2045(17)30624-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. doi: 10.1186/s40425-017-0263-0. Sindhu S, Gimber LH, Cranmer L, et al: Angiosarcoma treated successfully with anti-PD-1 therapy - a case report. J Immunother Cancer 5:58, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.D’Angelo SP, Melchiori L, Merchant MS, et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 c259T cells in synovial sarcoma. Cancer Discov. 2018;8:944–957. doi: 10.1158/2159-8290.CD-17-1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Shurell E, Vergara-Lluri ME, Li Y, et al. Comprehensive adipocytic and neurogenic tissue microarray analysis of NY-ESO-1 expression - a promising immunotherapy target in malignant peripheral nerve sheath tumor and liposarcoma. Oncotarget. 2016;7:72860–72867. doi: 10.18632/oncotarget.12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Roszik J, Wang W-L, Livingston JA, et al. Overexpressed PRAME is a potential immunotherapy target in sarcoma subtypes. Clin Sarcoma Res. 2017;7:11. doi: 10.1186/s13569-017-0077-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Iura K, Kohashi K, Hotokebuchi Y, et al. Cancer-testis antigens PRAME and NY-ESO-1 correlate with tumour grade and poor prognosis in myxoid liposarcoma. J Pathol Clin Res. 2015;1:144–159. doi: 10.1002/cjp2.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. doi: 10.1007/978-3-319-04843-7_16. Anderson PM, Subbiah V, Rohren E: Bone-seeking radiopharmaceuticals as targeted agents of osteosarcoma: Samarium-153-EDTMP and radium-223. Adv Exp Med Biol 804:291-304, 2014. [DOI] [PubMed] [Google Scholar]
  • 99.Subbiah V, Anderson PM, Kairemo K, et al. Alpha particle radium 223 dichloride in high-risk osteosarcoma: A phase I dose escalation trial. Clin Cancer Res. clincanres.3964.2018 [DOI] [PMC free article] [PubMed]
  • 100.Jones SE, Fleuren EDG, Frankum J, et al. ATR is a therapeutic target in synovial sarcoma. Cancer Res. 2017;77:7014–7026. doi: 10.1158/0008-5472.CAN-17-2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nieto-Soler M, Morgado-Palacin I, Lafarga V, et al. Efficacy of ATR inhibitors as single agents in Ewing sarcoma. Oncotarget. 2016;7:58759–58767. doi: 10.18632/oncotarget.11643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Stewart E, Goshorn R, Bradley C, et al. Targeting the DNA repair pathway in Ewing sarcoma. Cell Reports. 2014;9:829–841. doi: 10.1016/j.celrep.2014.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Yamasaki H, Miyamoto M, Yamamoto Y, et al. Synovial sarcoma cell lines showed reduced DNA repair activity and sensitivity to a PARP inhibitor. Genes Cells. 2016;21:852–860. doi: 10.1111/gtc.12387. [DOI] [PubMed] [Google Scholar]
  • 104.Laroche A, Chaire V, Le Loarer F, et al. Activity of trabectedin and the PARP inhibitor rucaparib in soft-tissue sarcomas. J Hematol Oncol. 2017;10:84. doi: 10.1186/s13045-017-0451-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.May CD, Garnett J, Ma X, et al. AXL is a potential therapeutic target in dedifferentiated and pleomorphic liposarcomas. BMC Cancer. 2015;15:901. doi: 10.1186/s12885-015-1916-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Fleuren EDG, Hillebrandt-Roeffen MHS, Flucke UE, et al. The role of AXL and the in vitro activity of the receptor tyrosine kinase inhibitor BGB324 in Ewing sarcoma. Oncotarget. 2014;5:12753–12768. doi: 10.18632/oncotarget.2648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zhang Y, Tang YJ, Man Y, et al. Knockdown of AXL receptor tyrosine kinase in osteosarcoma cells leads to decreased proliferation and increased apoptosis. Int J Immunopathol Pharmacol. 2013;26:179–188. doi: 10.1177/039463201302600117. [DOI] [PubMed] [Google Scholar]
  • 108.Hensel T, Giorgi C, Schmidt O, et al. Targeting the EWS-ETS transcriptional program by BET bromodomain inhibition in Ewing sarcoma. Oncotarget. 2016;7:1451–1463. doi: 10.18632/oncotarget.6385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Baker EK, Taylor S, Gupte A, et al. BET inhibitors induce apoptosis through a MYC independent mechanism and synergise with CDK inhibitors to kill osteosarcoma cells. Sci Rep. 2015;5:10120. doi: 10.1038/srep10120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Shen JK, Cote GM, Gao Y, et al. Targeting EZH2-mediated methylation of H3K27 inhibits proliferation and migration of synovial sarcoma in vitro. Sci Rep. 2016;6:25239. doi: 10.1038/srep25239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ciarapica R, Carcarino E, Adesso L, et al. Pharmacological inhibition of EZH2 as a promising differentiation therapy in embryonal RMS. BMC Cancer. 2014;14:139. doi: 10.1186/1471-2407-14-139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Tanaka M, Yamazaki Y, Kanno Y, et al. Ewing’s sarcoma precursors are highly enriched in embryonic osteochondrogenic progenitors. J Clin Invest. 2014;124:3061–3074. doi: 10.1172/JCI72399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang P, Yang X, Ma X, et al. Antitumor effects of pharmacological EZH2 inhibition on malignant peripheral nerve sheath tumor through the miR-30a and KPNB1 pathway. Mol Cancer. 2015;14:55. doi: 10.1186/s12943-015-0325-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Gounder MM, Stacchiotti S, Schöffski P, et al: Phase 2 multicenter study of the EZH2 inhibitor tazemetostat in adults with INI1 negative epithelioid sarcoma ( NCT02601950). J Clin Oncol 35, 2017 (suppl; abstr 11058) [Google Scholar]
  • 115.Van Tine BA, Bean GR, Boone P, et al. Using pegylated arginine deiminase (ADI-PEG20) for the treatment of sarcomas that lack argininosuccinate synthesase 1 expression. J Clin Oncol. 2013;31:10526. [Google Scholar]
  • 116.Toulmonde M, Penel N, Adam J, et al. Use of PD-1 targeting, macrophage infiltration, and IDO pathway activation in sarcomas: A phase 2 clinical trial. JAMA Oncol. 2018;4:93–97. doi: 10.1001/jamaoncol.2017.1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Barham W, Frump AL, Sherrill TP, et al. Targeting the Wnt pathway in synovial sarcoma models. Cancer Discov. 2013;3:1286–1301. doi: 10.1158/2159-8290.CD-13-0138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Dai G, Zheng D, Wang Q, et al. Baicalein inhibits progression of osteosarcoma cells through inactivation of the Wnt/β-catenin signaling pathway. Oncotarget. 2017;8:86098–86116. doi: 10.18632/oncotarget.20987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Pedersen EA, Menon R, Bailey KM, et al. Activation of Wnt/beta-catenin in Ewing sarcoma cells antagonizes EWS/ETS function and promotes phenotypic transition to more metastatic cell states. Cancer Res. 2016;76:5040–5053. doi: 10.1158/0008-5472.CAN-15-3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Singh S, Vinson C, Gurley CM, et al. Impaired Wnt signaling in embryonal rhabdomyosarcoma cells from p53/c-fos double mutant mice. Am J Pathol. 2010;177:2055–2066. doi: 10.2353/ajpath.2010.091195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Vijayakumar S, Liu G, Rus IA, et al. Wnt signaling is activated at high frequency and drives proliferation of multiple human sarcoma subtypes through a TCF/β-catenin target gene, CDC25A. Cancer Cell. 2011;19:601–612. doi: 10.1016/j.ccr.2011.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Subbiah V, Kurzrock R. Phase 1 clinical trials for sarcomas: The cutting edge. Curr Opin Oncol. 2011;23:352–360. doi: 10.1097/CCO.0b013e3283477a94. [DOI] [PubMed] [Google Scholar]
  • 123.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: 10.1200/JCO.1987.5.6.840. [DOI] [PubMed] [Google Scholar]
  • 124.Seddon B, Strauss SJ, Whelan J, et al. Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): A randomised controlled phase 3 trial. Lancet Oncol. 2017;18:1397–1410. doi: 10.1016/S1470-2045(17)30622-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.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: 10.1200/JCO.1993.11.7.1269. [DOI] [PubMed] [Google Scholar]
  • 126.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: 10.1200/JCO.2002.11.050. [DOI] [PubMed] [Google Scholar]
  • 127. doi: 10.1002/ijc.21867. Bay J-O, Ray-Coquard I, Fayette J, et al: Docetaxel and gemcitabine combination in 133 advanced soft-tissue sarcomas: A retrospective analysis. Int J Cancer 119:706-711, 2006 [Erratum: Int J Cancer 120:450, 2007] [DOI] [PubMed] [Google Scholar]
  • 128.Demetri GD, Schöffski P, Grignani G, et al. Activity of eribulin in patients with advanced liposarcoma demonstrated in a subgroup analysis from a randomized phase III study of eribulin versus dacarbazine. J Clin Oncol. 2017;35:3433–3439. doi: 10.1200/JCO.2016.71.6605. [DOI] [PubMed] [Google Scholar]
  • 129.Martin-Broto J, Pousa AL, de Las Peñas R, et al. Randomized phase II study of trabectedin and doxorubicin compared with doxorubicin alone as first-line treatment in patients with advanced soft tissue sarcomas: A Spanish Group for Research on Sarcoma Study. J Clin Oncol. 2016;34:2294–2302. doi: 10.1200/JCO.2015.65.3329. [DOI] [PubMed] [Google Scholar]
  • 130.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: 10.1016/S1470-2045(07)70175-4. [DOI] [PubMed] [Google Scholar]
  • 131.van der Graaf WTA, Blay J-Y, Chawla SP, et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2012;379:1879–1886. doi: 10.1016/S0140-6736(12)60651-5. [DOI] [PubMed] [Google Scholar]
  • 132. doi: 10.1158/1078-0432.CCR-17-0272. Subbiah V, Meyer C, Zinner R, et al: Phase Ib/II study of the safety and efficacy of combination therapy with multikinase VEGF inhibitor pazopanib and MEK inhibitor trametinib in advanced soft tissue sarcoma. Clin Cancer Res 23:4027-4034, 2017 [Erratum: Clin Cancer Res 24:500-501, 2018] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Subbiah V, Kurzrock R. Challenging standard-of-care paradigms in the precision oncology era. Trends Cancer. 2018;4:101–109. doi: 10.1016/j.trecan.2017.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. J Clin Oncol. 2018;36:7–13. doi: 10.1200/JCO.2017.73.6785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Diamond EL, Subbiah V, Lockhart AC, et al. Vemurafenib for BRAF V600-mutant Erdheim-Chester disease and Langerhans cell histiocytosis: Analysis of data from the histology-independent, phase 2, open-label VE-BASKET study. JAMA Oncol. 2018;4:384–388. doi: 10.1001/jamaoncol.2017.5029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Cunanan KM, Gonen M, Shen R, et al. Basket trials in oncology: A trade-off between complexity and efficiency. J Clin Oncol. 2017;35:271–273. doi: 10.1200/JCO.2016.69.9751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Qadir MA, Zhan SH, Kwok B, et al. ChildSeq-RNA: A next-generation sequencing-based diagnostic assay to identify known fusion transcripts in childhood sarcomas. J Mol Diagn. 2014;16:361–370. doi: 10.1016/j.jmoldx.2014.01.002. [DOI] [PubMed] [Google Scholar]
  • 139. doi: 10.1200/PO.16.00028. Shukla NN, Patel JA, Magnan H, et al: Plasma DNA-based molecular diagnosis, prognostication, and monitoring of patients with EWSR1 fusion-positive sarcomas. JCO Precis Oncol doi: 10.1200/PO.16.00028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Varga E, Chao EC, Yeager ND. The importance of proper bioinformatics analysis and clinical interpretation of tumor genomic profiling: A case study of undifferentiated sarcoma and a constitutional pathogenic BRCA2 mutation and an MLH1 variant of uncertain significance. Fam Cancer. 2015;14:481–485. doi: 10.1007/s10689-015-9790-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Koehler K, Liebner D, Chen JL. TP53 mutational status is predictive of pazopanib response in advanced sarcomas. Ann Oncol. 2016;27:539–543. doi: 10.1093/annonc/mdv598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Lim SM, Park HS, Kim S, et al. Next-generation sequencing reveals somatic mutations that confer exceptional response to everolimus. Oncotarget. 2016;7:10547–10556. doi: 10.18632/oncotarget.7234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wang Z, Shi N, Naing A, et al. Survival of patients with metastatic leiomyosarcoma: The MD Anderson Clinical Center for targeted therapy experience. Cancer Med. 2016;5:3437–3444. doi: 10.1002/cam4.956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Chang W, Brohl AS, Patidar R, et al. Multidimensional clinomics for precision therapy of children and adolescent young adults with relapsed and refractory cancer: A report from the Center for Cancer Research. Clin Cancer Res. 2016;22:3810–3820. doi: 10.1158/1078-0432.CCR-15-2717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Worst BC, van Tilburg CM, Balasubramanian GP, et al. Next-generation personalised medicine for high-risk paediatric cancer patients—The INFORM pilot study. Eur J Cancer. 2016;65:91–101. doi: 10.1016/j.ejca.2016.06.009. [DOI] [PubMed] [Google Scholar]
  • 146.Coley WB. 1910.
  • 147. Burgess MA, Bolejack V, Van Tine BA, et al: Multicenter phase II study of pembrolizumab (P) in advanced soft tissue (STS) and bone sarcomas (BS): Final results of SARC028 and biomarker analyses. J Clin Oncol 35, 2017 (suppl; abstr 11008) [Google Scholar]
  • 148.D’Angelo SP, Mahoney MR, Van Tine BA, et al. Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): Two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol. 2018;19:416–426. doi: 10.1016/S1470-2045(18)30006-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Migliorini D, Mach N, Aguiar D, et al. First report of clinical responses to immunotherapy in 3 relapsing cases of chordoma after failure of standard therapies. OncoImmunology. 2017;6:e1338235. doi: 10.1080/2162402X.2017.1338235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Groisberg R, Hong DS, Behrang A, et al. Characteristics and outcomes of patients with advanced sarcoma enrolled in early phase immunotherapy trials. J Immunother Cancer. 2017;5:100. doi: 10.1186/s40425-017-0301-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Keytruda [package insert]. Whitehouse Station, NJ, Merck, 2018.
  • 152. doi: 10.1200/JCO.18.00283. Latham A, Srinivasan P, Kemel Y, et al: Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer. J Clin Oncol 37:286-295, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zheng C, You W, Wan P, et al. Clinicopathological and prognostic significance of PD-L1 expression in sarcoma: A systematic review and meta-analysis. Medicine (Baltimore) 2018;97:e11004. doi: 10.1097/MD.0000000000011004. [DOI] [PMC free article] [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: 10.1002/ijc.1451. [DOI] [PubMed] [Google Scholar]
  • 155.Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol. 2003;21:4342–4349. doi: 10.1200/JCO.2003.04.190. [DOI] [PubMed] [Google Scholar]
  • 156. doi: 10.6004/jnccn.2010.0116. Demetri GD, von Mehren M, Antonescu CR, et al: NCCN task force report: Update on the management of patients with gastrointestinal stromal tumors. J Natl Compr Canc Netw 8:S1-S41, 2010; quiz S42-S44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.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: 10.1200/JCO.1999.17.1.150. [DOI] [PubMed] [Google Scholar]
  • 158.Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279:577–580. doi: 10.1126/science.279.5350.577. [DOI] [PubMed] [Google Scholar]
  • 159.Yan L, Zou L, Zhao W, et al. Clinicopathological significance of c-KIT mutation in gastrointestinal stromal tumors: A systematic review and meta-analysis. Sci Rep. 2015;5:13718. doi: 10.1038/srep13718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Patel S. Long-term efficacy of imatinib for treatment of metastatic GIST. Cancer Chemother Pharmacol. 2013;72:277–286. doi: 10.1007/s00280-013-2135-8. [DOI] [PubMed] [Google Scholar]
  • 161.Joensuu H, Eriksson M, Sundby Hall K, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: A randomized trial. JAMA. 2012;307:1265–1272. doi: 10.1001/jama.2012.347. [DOI] [PubMed] [Google Scholar]
  • 162.Yoo C, Ryu M-H, Ryoo B-Y, et al. Efficacy, safety, and pharmacokinetics of imatinib dose escalation to 800 mg/day in patients with advanced gastrointestinal stromal tumors. Invest New Drugs. 2013;31:1367–1374. doi: 10.1007/s10637-013-9961-8. [DOI] [PubMed] [Google Scholar]
  • 163.Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: A randomised controlled trial. Lancet. 2006;368:1329–1338. doi: 10.1016/S0140-6736(06)69446-4. [DOI] [PubMed] [Google Scholar]
  • 164. Kelly CM, Bowler TG, D’Angelo SP, et al: The clinical impact of performing routine next generation sequencing (NGS) in gastrointestinal stromal tumors (GIST). J Clin Oncol 35, 2017 (suppl; abstr 11010) [Google Scholar]
  • 165. Heinrich MC, Jones RL, von Mehren M, et al: Clinical activity of BLU-285 in advanced gastrointestinal stromal tumor (GIST). J Clin Oncol 35, 2017 (suppl; abstr 11011) [Google Scholar]
  • 166. Li B, Serrano Garcia C, Marino-Enriquez A, et al: Conjoined hyperactivation of the RAS and PI3K pathways in advanced GIST. J Clin Oncol 34, 2016 (suppl; abstr e22520) [Google Scholar]
  • 167.Merten L, Agaimy A, Moskalev EA, et al. Inactivating mutations of RB1 and TP53 correlate with sarcomatous histomorphology and metastasis/recurrence in gastrointestinal stromal tumors. Am J Clin Pathol. 2016;146:718–726. doi: 10.1093/ajcp/aqw193. [DOI] [PubMed] [Google Scholar]

Articles from JCO Precision Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES