Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches

CANCER TREATMENT REVIEWS (2006) 32, 9–27 www.elsevierhealth.com/journals/ctrv ANTI-TUMOUR TREATMENT Adult soft tissue sarcomas: Conventional therap...

331KB Sizes 8 Downloads 115 Views

CANCER TREATMENT REVIEWS (2006) 32, 9–27

www.elsevierhealth.com/journals/ctrv

ANTI-TUMOUR TREATMENT

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches Simone Mocellin

a,c,

*, Carlo R. Rossi a, Alba Brandes b, Donato Nitti

a

a

Surgery Branch, Department of Oncological and Surgical Sciences, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy b Oncology Section, Department of Oncological and Surgical Sciences, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy c Istituto Oncologico Veneto - Istituto di Ricerca e Cura a Carattere Scientifico (IOV-IRCCS), via Gattamelata 64, 35128 Padova, Italy Received 12 July 2005; received in revised form 21 September 2005

KEYWORDS

Summary The therapeutic approach to soft tissue sarcomas (STS) has evolved over the past two decades based on the results from randomized controlled trials (RCT), which are guiding physicians in the treatment decision-making process. Despite significant improvements in the control of local disease, a significant number of patients ultimately die of recurrent/metastatic disease following radical surgery due to a lack of effective adjuvant treatments. In addition, the characteristic chemoresistance of STS has compromised the therapeutic value of conventional antineoplastic agents in cases of unresectable advanced/metastatic disease. Therefore, novel therapeutic strategies are urgently needed to improve the prognosis of patients with STS. Recent advances in STS biology are paving the way to the development of molecularly targeted therapeutic strategies, the efficacy of which relies not only on the knowledge of the molecular mechanisms underlying cancer development/progression but also on the personalization of the therapeutic regimen according to the molecular features of individual tumours. In this work, we review the state-of-the-art of conventional treatments for STS and summarize the most promising findings in the development of molecularly targeted therapeutic approaches. c 2005 Elsevier Ltd. All rights reserved.

Soft tissue sarcoma; Surgery; Chemotherapy; Radiotherapy; Molecular therapy; Targeted therapy; Gene therapy; Cancer vaccine



Introduction * Corresponding author. Tel.: +39 049 8211851; fax: +39 049 651891. E-mail address: [email protected] (S. Mocellin).



Soft tissue sarcomas (STS) are a heterogeneous group of rare mesenchymal tumours that account

0305-7372/$ - see front matter c 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.ctrv.2005.10.003

10 for roughly 1% of all adult malignancies.1 Currently, more than 50 histological types of STS have been identified, but the most common in adults are malignant fibrous histiocytoma (28%), leiomyosarcoma (12%), liposarcoma (15%), synovial sarcoma (10%), and malignant peripheral nerve sheath tumours (6%).2 Despite the variety of histological subtypes, STS present some common features. For instance, the clinical behaviour of most STS correlates with anatomic location, histological grade, and tumour size.3,4 Moreover, the prevalent metastatic pattern is hematogenous, with the lungs being the main target organ; by contrast lymphatic metastases are rare (<5%), except for few histological types (e.g., epithelioid sarcoma, synovial sarcoma, clear-cell sarcoma, angiosarcoma). However, the variability in patients’ clinical outcome underscores the heterogeneity of the biological aggressiveness of these tumours, the therapeutic management of which remains one of the most daunting tasks for oncologists. STS can occur anywhere in the body, but most originate in the extremities (60%), followed by the retroperitoneum (19%), abdominal/thoracic wall (15%), and head&neck (6%). As a corollary, radical resection can be functionally mutilating for patients and technically challenging for surgeons. Furthermore, most STS are characterized by relative resistance to chemotherapy, and the 5-year overall survival (OS) rate for STS of all stages is only 50–60%.5 Despite the improvements in the local control rate of limb STS treated with a multidisciplinary approach, a high frequency of recurrence/metastasis and poor OS remain significant problems, particularly in patients with highrisk STS (e.g., metastatic/recurrent disease, deep vs. superficial location, trunk/head&neck vs. extremity, tumour size >5 cm, intermediate-/ high- vs. low-grade).6 Nevertheless, recent advances in the biology of these tumours are paving the way to molecularly targeted therapeutic strategies that might change the natural history of STS.7,8 In this work, we reviewed the results of conventional treatments for STS and provide the reader with an overview on the most promising findings in the development of molecular therapeutic approaches to these tumours. To this aim, PubMed searches of the National Library of Medicine were performed with appropriate keywords, with the only restriction being English language. For ongoing clinical trials, the National Cancer Institute dedicated website (http://cancer.gov/clinicaltrials) was also searched.

S. Mocellin et al.

Standard treatments Extremity STS Surgery The surgical approach to STS has drastically changed over the last decades. In the 1970s, amputation was considered the standard treatment for limb STS and 50% of patients with extremity STS underwent amputation for local control of their tumours.9 However, large number of patients died of metastatic disease despite a local recurrence rate <15% after radical surgery. In 1982, a small randomized controlled trials (RCT) (n = 43) showed that limb-sparing surgery combined with radiotherapy could obtain a local recurrence rate of only 15%, with no difference in OS compared with amputation.10 In the 1980s, following the implementation of multimodality therapy (conservative surgery combined with radiotherapy or locoregional chemotherapy), wide peritumoural healthy tissue resections (compartmentectomy) compromising limb function were progressively abandoned, amputation rates dropped to less than 10%, and local recurrence rates remained low (5–15%).11 In the 1990s, improved selection of patients for adjuvant radiotherapy resulted from RCT, and technical advances in reconstructive surgery (e.g., pedicle or free tissue transfers, repair of bone defects with prostheses or autogenous bone grafts using micro-vascular techniques) allowed for the closure of large soft tissue defects,12 which contributed to minimize functional deficits in patients otherwise requiring amputation. The accumulated experience has allowed the definition of some practical guidelines. If applicable, the biopsy site/tract should be included en bloc with the resected specimen, as inadequate primary tumour resection can compromise the curative intent of surgery.13,14 STS are generally surrounded by a zone of compressed reactive tissue that forms a pseudocapsule, which may mistakenly be used by inexperienced surgeons to guide resection (enucleation). Microscopic extensions of tumour beyond the pseudocapsule (skip metastasis) must always be considered while planning surgery. Currently, wide local excision is the primary treatment strategy for extremity STS, with the goal to resect the tumour with a 1–2 cm margin of surrounding normal tissue. This strategy has been validated by long-term results.3,4 For instance, in a series of 77 patients treated with limb-sparing surgery without adjuvant treatments, the 10-year local recurrence rate was 7%, with a significant difference between patients who had a margin width greater or less than 1 cm (13% and 0%, respectively).15 These and other similar

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches findings indicate the importance of clinico-pathological (e.g., nomograms integrating standard prognostic factors) and molecular (e.g., detection of minimal residual disease by RT-PCR) criteria to select patients eligible for limb-sparing surgery without adjuvant treatment.16–21 Currently, patients who are candidates for this strategy are those with primary presentation and closest margin P1 cm, in whom local recurrence would not preclude subsequent limb salvage. When wide disease-free margins cannot be obtained owing to anatomic constraints, adjuvant radiotherapy is generally considered the standard approach for patients with primary tumours with no evidence of distant metastasis.4 After the NIH Consensus Conference in 1985, limb-sparing surgery for most patients with extremity STS was recommended.22 This is true also for most patients with STS of the hand and foot, which is a particularly challenging situation from the surgical viewpoint.23 As microscopically positive surgical margins imply an increased risk of local recurrence,24–27 amputation remains the only way to control local disease in the estimated 5–10% of patients whose tumour cannot be radically resected with a limb-sparing/function-preserving procedure at presentation, after neoadjuvant treatments, or following local recurrence.9,28 However, neither positive margins nor local recurrence appear to adversely affect OS.15,16,18,26,27,29 Thus, conservative surgery coupled with neoadjuvant/adjuvant treatments is generally recommended whenever achieving clear margins would require amputation or substantial functional compromise of an extremity.3 Radiotherapy The efficacy of radiotherapy in patients with STS amenable to conservative surgical resection is supported by two RCT30,31 and several single-institution studies.32–35 In the first RCT, 164 patients with completely resected extremity/trunk STS were either observed or underwent brachytherapy, a technique that consists of the direct application of radioactive seeds (iridium-192) into the tissue at risk of disease relapse (tumour bed) through catheters placed during surgery.30 Updated follow-up revealed that 10-year local disease-free survival (DFS) for the brachytherapy arm was 83% compared with 67% for patients treated with surgery alone (P = 0.39), with no difference in OS. However, patients with high-grade tumours had a 10-year local DFS of 90% with brachytherapy compared with 63% with surgery alone (P = 0.002). In the other RCT, 141 patients with limb STS were treated with limb-sparing surgery alone or combined with external-beam radiotherapy (EBRT); in addition, patients with high-grade tu-

11

mours (n = 91) also received systemic chemotherapy.31 The 10-year local control rate for all patients receiving radiotherapy was 98% (highgrade 0%, low-grade 5%), compared with 70% (high-grade 22%, low-grade 36%) for those not receiving radiotherapy (P = 0.0001), without a significant impact on OS. The relatively low local recurrence rate even in patients receiving surgery alone – coupled with no local recurrences if margins were >1 cm – suggests that selected patients could safely be treated with surgery alone and avoid long-term morbidity from radiotherapy.36,37 Despite the consideration that radiotherapy is effective for local disease control purposes in high-grade tumours with surgical margin less than 1 cm, several aspects of its role in the management of limb STS remain debated. For instance, small tumours (<5 cm) are less frequently associated with local recurrence, indicating that radiotherapy may not be necessary in these patients.24,38 Accordingly, in some series adjuvant radiotherapy does not improve the 5-year local DFS or OS rates in this cohort of patients.39,40 However, other authors have drawn opposite conclusions.26 The optimal mode (external-beam/brachytherapy) and timing (preoperative/intraoperative/postoperative) of radiotherapy is also yet to be defined. Postoperative radiotherapy planning is based on tumour grade, status of surgical margins, and institutional preferences. The entire surgical scar and drain sites should be included in the field so that a near-full dose is given to the superficial skin. Proponents of preoperative radiotherapy claim some advantages to this approach.41,42 First, the better oxygenation of an undisturbed tumour bed might enhance the antitumour effect of radiation. Second, preoperative radiation fields may be smaller than those in postoperative radiation fields, and this may result in an improved functional outcome. Third, by shrinking the tumour, radiotherapy can facilitate surgical resection. The main limits of preoperative radiotherapy are the difficulty of pathologically assessing surgical margins in irradiated specimens and the increased rate of wound complications. In fact, in the only RCT so far performed,43 wound complications were more common with preoperative compared with postoperative radiotherapy (35% vs. 17%; P = 0.01), with the risk exclusively confined to lower extremity lesions. In the same trial, at 3.3 years median follow-up local control (93%) was identical, and progression-free survival was not significantly different in the two treatment arms. Limb function status was the same for both groups 12 months after surgery, but after 2 years, rates of fibrosis (56% vs. 28%; P = 0.003) and oedema (24% vs. 7%; P = 0.01) were higher in the

12 postoperative arm because of greater irradiation volumes and doses. Complementary systemic chemotherapy Adjuvant chemotherapy Several RCT have shown that adjuvant systemic chemotherapy does not improve either DFS or OS in patients with STS.44 The Sarcoma Meta-analysis Collaboration evaluated the effect of adjuvant chemotherapy on localized, resectable STS in 1568 patients from 14 trials of doxorubicin-based adjuvant chemotherapy.45 At 9.4 years median follow-up, the time to local and distant recurrence was significantly better in patients receiving doxorubicin-based chemotherapy than in the control group. However, the OS rate advantage in the treatment group was only 4%, which was not significant (P = 0.12). In a subset analysis, patients with extremity tumours (n = 886) showed a 7% benefit in survival (P = 0.029). In a subsequent RCT (n = 104),46 patients with resectable high-grade/large (>5 cm) or recurrent limb STS treated with an intensified anthracycline/ifosfamide combination showed a better DFS and OS rates as compared to controls, with the absolute benefit for OS being 13% and 19% at 2 and 4 years, respectively, although there was no difference in distant relapse rates at 4 years. These results have been criticized on the basis of a retrospective analysis made on a larger series of patients (n = 674) treated with pre- or postoperative chemotherapy: in fact, in this study the clinical benefits associated with doxorubicin-based chemotherapy in patients with high-risk (stage-III) extremity STS appear to vanish after 1-year follow-up.47 An ongoing RCT is testing the efficacy of radical surgery plus intensified adjuvant chemotherapy in a large cohort of patients (Table 1). Although the efficacy of this strategy is yet to be demonstrated, outside clinical trials an anthracycline/ifosfamide combination is often recommended to young patients with large/high-grade extremity STS.48 Neoadjuvant chemotherapy There are some theoretical advantages to neoadjuvant systemic treatment: (1) to increase the chances of limb salvage by shrinking tumour size; (2) to assess the tumour response in situ using radiological imaging and pathological evaluation after surgical resection; (3) to spare patients who do not respond to preoperative therapy the prolonged toxicities of an ineffective adjuvant treatment; conversely, patients who respond to preoperative chemotherapy can be selected for postoperative treatment as a chemotherapysensitive subset.

S. Mocellin et al. Importantly, no increase in the postoperative complication rate has been reported following neoadjuvant systemic chemotherapy.49 Nevertheless, there is scarce clinical evidence supporting the usefulness of such an approach.50–52 Although correlated with favourable clinical outcome, treatment-induced complete tumour necrosis occurs in only a minority of patients (69/496, 14%).53 Moreover, in another retrospective analysis assessing the impact of preoperative chemotherapy on the extent of the surgical procedure in 65 patients with stage-II/III limb/retroperitoneal STS, only eight patients (12%) showed a response sufficient for their operation to be reduced, while in another 6 cases (9%) the disease progressed enough to require an increase in the magnitude of their operation; furthermore, limb salvage could not be performed in any of the nine patients who were determined to require amputation before chemotherapy.54 Neoadjuvant concomitant radio-chemotherapy By exploiting the radiosensitizing effects of some chemotherapeutic drugs, neoadjuvant treatments that combine chemotherapy with concurrent external-beam radiotherapy may theoretically increase the limb-sparing rate and improve DFS.55 The initial approach consisted of intra-arterial doxorubicin and high-dose-per-fraction radiotherapy.56,57 Then, several groups tested other radio-chemotherapy approaches using systemic chemotherapy and alternative chemotherapeutic agents.58–60 Remarkably, when intra-arterial polychemotherapy was adopted, the favourable clinical outcome led investigators to question the need for combining radiotherapy.61 Overall, the small number of patients and the heterogeneity of series so far reported do not permit to compare (even historically) this strategy with others. High toxicity rates have been reported and most series include patients with STS at highrisk of recurrence, but not limb-threatening (unresectable) tumours. Nevertheless, the encouraging results in terms of tumour response (>50%) and local control (<15%) rates justifies continued investigation of preoperative chemoradiation regimens for localized STS. Isolated limb perfusion A locoregional approach proposed for the treatment of limb-threatening malignancies is represented by the hyperthermic isolated limb perfusion (HILP), a technically demanding surgical procedure that allows tumour drug concentrations to reach 20-fold higher than those achievable with systemic chemotherapy.62 This drug-delivery system also exploits the antitumour synergism be-

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches Table 1

Ongoing clinical trials of conventional and molecularly targeted treatments for soft tissue sarcomas (STS)

Study design Therapeutic regimen Phase III Phase III Randomized Phase III Phase III Phase III Phase III Phase III Phase II Phase II Phase II Phase II Phase II Phase II

Phase II

Phase II Phase II Phase II Phase I Phase I Phase I Phase I

13

2

Systemic chemotherapy: DXR (75 mg/m ) + IFO (5 g/m2) vs. observation Systemic chemotherapy (etoposide + IFO + DXR) ± regional hyperthermia Systemic chemotherapy: gemcitabine ± docetaxel Radiotherapy Systemic chemotherapy: vincristine + Dactinomycin + cyclophosphamide + DXR ± IFO Tyrosine-kinase inhibitor (imatinib); doserandomized study Tyrosine-kinase inhibitor (imatinib) vs. placebo Tyrosine-kinase inhibitor (SU011248) vs. placebo

Clinical setting

Study ID/Ref.

Adjuvant treatment for high-grade/recurrent STS Neoadjuvant treatment for high-risk/recurrent STS Advanced/metastatic STS Neoadjuvant treatment for retroperitoneal STS Advanced/metastatic STS

EORTC-62931 (accrual closed) EORTC-62961 Ref.123 ECOG-Z9031 EORTC-62012

Advanced/metastatic GIST

SWOG-S0033 (accrual closed) Adjuvant treatment for GIST NCT00041197 Advanced/metastatic GIST NCT00085618, resistant to imatinib NCT00094029 Isolated limb infusion with DLocally advanced extremity NCT00004250 actinomycin + melphalan STS mTOR inhibitor (AP23573, rapamycin analog) Advanced/metastatic STS NCT00093080 (no GIST) mTOR inhibitor (CCI-779, rapamycin analog) Advanced/metastatic STS NCT00087074 (including GIST) Tyrosine-kinase inhibitor (imatinib) Neoadjuvant/adjuvant NCT00028002 treatment for GIST Tyrosine-kinase inhibitor (imatinib) Advanced/metastatic EORTC-62027, dermatofibrosarcoma SWOG-S0345 Tyrosine-kinase inhibitor (erlotinib) Advanced/metastatic SWOG-S0330 malignant peripheral nerve sheath tumour Tyrosine-kinase inhibitor (gefitinib) Advanced/metastatic EORTC-62022 synovial sarcoma expressing HER1 Antiangiogenic therapy (bevacizumab, antibody HIV-positive/negative NCI-03-C-0110E to VEGF) Kaposi’s sarcoma Gene therapy (oblimersen, antisense Advanced/metastatic GIST NCT00091078 oligonucleotide targeting Bcl-2) resistant to imatinib MMP inhibitors (indinavir, saquinavir) Stage I–III HIV-negative NCT00003419 Kaposi’s sarcoma Cancer vaccine (TERT peptide + GM-CSF) STS (including GIST) and NCT00069940 brain tumours Cancer vaccine (NY-ESO1 peptides + GM-CSF) Stage II–IV STS expressing NCT00027911 NY-ESO1 Antiangiogenic therapy (sorafenib, protein kinase Advanced/metastatic solid NCI-05-C-0022 inhibitor) tumours Antiangiogenic therapy (VEGF decoy receptor) Advanced/metastatic solid NCT0004526 tumours

DXR: doxorubicin; GIST: gastrointestinal stromal tumours; IFO: ifosfamide; GM-CSF: granulocyte-macrophage colony stimulating factor; MMP: matrix metallo-proteinase; VEGF: vascular endothelium growth factor.

tween heat and some antineoplastic agents (e.g., melphalan, doxorubicin and tumour-necrosis-factor, TNF) and maximizes the likelihood of sterilizing microscopic skip metastases throughout the limb. Despite these premises, clinical results with both single chemotherapeutic agents (e.g., melphalan, doxorubicin) and polychemotherapy regimens did not meet the expectations in patients with STS.63 By contrast, when TNF (a cytokine with

antitumour and antiangiogenic properties)64 is added to the drug regimen, the complete and overall tumour response rates range from 10% to 90% and from 50% to 100%, respectively, and limb-sparing surgery is feasible in 70–100% of cases.65–69 At 2 years median follow-up, local DFS is 80–90%, with a secondary amputation being necessary in up to 5% of cases because of either HILP-related local toxicity or tumour recurrence. Although the

14 fear of TNF systemic toxicity has been tempered by the use of real-time monitoring of perfusate/ plasma drug leakage,70 severe locoregional toxicity occurs on average in 10% of patients. Despite the lack of RCT, TNF-based HILP is generally considered one of the most effective neoadjuvant treatments in terms of local disease control and limb-sparing rates in STS patients otherwise candidate for amputation.71 However, there is no evidence that HILP affects patients’ OS.

Retroperitoneal STS The prognosis of patients with retroperitoneal STS – which are mainly represented by liposarcomas and leiomyosarcomas – is worse than that of patients with extremity STS, with the 5-year OS rate being 40–50% and 60–70%, respectively.72–76 Large tumour size, high histological grade, unresectability, as well as positive resection margins are strongly associated with a worse prognosis. Surgery Unlike limb STS, about 75% of patients with retroperitoneal STS ultimately die of locally recurrent disease without distant metastasis, underscoring the importance of trying to achieve a complete resection at presentation.3 In all published series the best chance of cure is achieved in patients with margin-free tumour resection.77 In the largest single institution series (n = 500), median OS was 103 months in patients who underwent complete resection and only 18 months in those who underwent either incomplete resection or observation without resection.72 However, due to their large size (mean diameter 15–20 cm) and locally advanced presentation (infiltration of contiguous organs/structures occurs in 50–75% of cases),4 complete resection is feasible in only about a half of cases, and may require major surgical interventions including the en bloc resection of bowel segments, kidney, spleen, liver segments, pancreas, inferior vena cava and so forth. Adjuvant and neoadjuvant treatments Even with aggressive surgical management, local recurrence occurs in 50–60% of cases. In order to reduce the risk of disease relapse after surgery, several radiotherapy- and/or chemotherapy-based trials have been performed: unfortunately, there is no consensus regarding the benefit of these additional complementary treatments.77,78 As regards radiotherapy, available results suggesting clinical benefit are difficult to interpret

S. Mocellin et al. due to small patient numbers, non-randomized trial design, and use of different sequences (preoperative vs. postoperative) and concurrent treatments.35,41,78–80 Although reasonable doses of radiation can be delivered to the retroperitoneal space with acceptable toxicity and a delay in time to recurrence is likely to occur, the effect on OS is not evident. Doses must be restricted because of sensitive normal structures within the target volume, particularly small bowel. Large field sizes contribute to toxicity, generally favouring the use of preoperative radiotherapy that presents some theoretical advantages: (1) the gross tumour volume can be defined, allowing accurate treatment planning; (2) tumours can displace radiosensitive viscera outside the radiation field;80 and (3) lower radiation doses may be equally effective preoperatively due to better oxygenation of undisturbed tumour tissue. Based on these premises, a RCT of neoadjuvant radiotherapy is underway. Among the most recent developments of radiation-based treatments, intensity modulated radiotherapy (IMRT)–an advanced form of threedimensional conformal radiotherapy – holds promise.81 With IMRT, radiation beams are not only shaped at their perimeters but also have variable intensity across their profiles, which should greatly improve tumour targeting in a complex anatomical region such as the retroperitoneal space. As regards systemic chemotherapy, no available data support its routine use in the adjuvant setting for the treatment of retroperitoneal STS. As mentioned above, the largest meta-analysis on RCT of doxorubicin-based adjuvant systemic chemotherapy for adult STS (regardless of tumour site) showed no improvement in OS despite a longer disease-free time in the treatment arms.45 Other chemotherapeutic regimens/strategies have been the subject of non-randomized trials, including the combination of neoadjuvant chemotherapy with regional hyperthermia82 (currently under evaluation in a phase III study) or radiotherapy.55,79 Intraoperative treatments As compared to EBRT, intraoperative radiotherapy (IORT) allows for the administration of higher radiation doses, since sensitive organs can be shielded and the tumour bed can be selectively targeted by the radiation beam. The results from small phase I–II trials,60,83–86 although encouraging, are difficult to interpret, because of the combination with other therapeutic modalities (EBRT, systemic chemotherapy). In a small (n = 35) RCT (surgery/ IORT/EBRT vs. surgery/EBRT), the use of IORT was associated with a lower locoregional recurrence rate, but did not affect OS.87

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches Cytoreductive surgery combined with hyperthermic intraperitoneal intraoperative chemotherapy (HIIC) has already shown some interesting results for the control of peritoneal carcinomatosis.88,89 To assess the antitumour synergism of doxorubicin, cisplatin and heat for the treatment of intraabdominal sarcomatosis from retroperitoneal and visceral (mostly GIST) STS, phase I and II trials were conducted.90,91 Unfortunately, no improvement in local DFS or OS could be suggested in these historical control studies. Nevertheless, HIIC might be useful for the administration of more active regimens.92 For instance, following the satisfactory results obtained in patients with extremity STS, the addition of TNF to conventional HIIC regimens might represent an appealing way to treat locally advanced intra-abdominal STS.93 Photodynamic therapy (PDT) combines a photosensitizer (e.g., porphyrin derivatives retained in tumour tissues more than in normal tissues) with a laser light (in a wavelength range matching the photosensitizer’s absorption spectrum) to generate reactive oxygen species that ultimately kill tumour cells.94 Given the limited penetrance of the laser, this approach can be used only for tumours of the body surfaces (e.g., esophagus, bronchus, skin, peritoneum). The implementation of PDT for the treatment of intraperitoneal malignancies, including STS, is in its infancy,95–97 and further evaluation is warranted to judge its efficacy.

Local relapse and distant metastasis Surgery and complementary treatments Several reports suggest that the early recognition and treatment of local or distant disease recurrence can prolong survival.98–102 An isolated local recurrence should be treated aggressively with margin-negative resection. This frequently requires amputation in patients with extremity tumours, although acceptable rates of local control can be achieved in these patients with function-preserving resection combined with additional radiotherapy, with or without chemotherapy.28,103–105 The preferred treatment for locally recurrent retroperitoneal STS is also surgery, when complete resection is feasible. In the largest series thus far published, investigators were able to adequately resect recurrent tumours in 57% of the patients.72 However, adequate resection was possible in only 20% after the second recurrence and in 10% after the third one. Complete pulmonary resection can achieve longterm survival in 15–40% of patients with lung metastases who have fewer than four pulmonary

15

nodules, long disease-free intervals, and no bronchial infiltration.101,102,106 Favourable prognostic factors are microscopically disease-free margins, young age (<40 years), and low histological grade. Unfortunately, most patients with recurrent/metastatic disease are not eligible for radical surgery or locoregional therapies and the only conventional therapeutic option for them is systemic chemotherapy: however, their prognosis is generally dismal (median OS: 10–12 months).107,108 Systemic chemotherapy Tumour response rates to chemotherapy in patients with stage-IV STS are generally disappointing.107,108 Doxorubicin and ifosfamide are the most active chemotherapeutic agents for metastatic adult STS with single-agent response rates of 16–36%.109 Combination regimens (usually containing anthracyclines) have response rates up to 35–60%, but have increased toxicity compared with single agents.4 In a large (n = 2185) retrospective analysis of patients with advanced/metastatic STS treated with anthracycline-containing regimens as first-line chemotherapy, OS time and tumour response rate were 51 weeks and 26%, respectively.110 At multivariate analysis, survival was favourably affected by low histological grade, long disease-free interval and young age, while tumour response was better in younger patients, high-grade tumours and liposarcoma. A recent meta-analysis of eight RCT (n = 2281) comparing doxorubicin-based combinations with doxorubicin as a single agent showed no differences in response rates (odds ratio, 0.79; P = 0.10) or survival (odds ratio, 0.84; P = 0.13).111 These trials generally involved insufficient numbers of patients to provide adequate power for testing responses to chemotherapy by histological subtype. Results from small series suggesting that synovial sarcoma may be particularly responsive to ifosfamide remain to be confirmed in larger studies.112,113 Non-controlled studies have suggested a direct proportionality between the dose of anthracyclines and ifosfamide and tumour response rates.114,115 However, the findings from two RCT oppose this hypothesis both in terms of tumour response and OS.116,117 Preliminary results of high-dose chemotherapy with autologous bone marrow/stem-cell support are controversial.118–121 Moreover, higher toxicity rates are reported and the results are often difficult to interpret due to the inclusion of pediatric patients.3 Favourable preliminary results obtained with novel drug/drug regimens (e.g., docetaxel alone or with gemcitabine, ifosfamide plus etoposide

16 and cisplatin, ecteinascidin)122–126 are under evaluation in ongoing RCT.

Molecularly targeted therapies Over the last two decades many insights regarding the molecular mechanisms underlying cancer development/progression have been confirmed in STS and a number of genetic (point mutations, deletions, amplifications) and chromosomal (translocations, aneuploidy) alterations specific to STS have been identified and linked to their pathogenesis.7,8,127 Overall, these molecular oncology findings are not only facilitating STS classification and prognosis prediction, but also fostering the development of molecularly targeted therapeutic strategies (Fig. 1). Gastrointestinal stromal tumours as a model Gastrointestinal STS account for 1–3% of all gastrointestinal cancers, up to 20% of small bowel malignancies and about 5% of all STS.5 As is true for non-gastrointestinal STS, high histological grade and large tumour size (>5 cm) adversely affect prognosis. Locoregional recurrence (peritoneal sarcomatosis) is the most common form of disease relapse, followed by liver metastasis. Both are associated with a dismal prognosis owing to the intrinsic aggressiveness of these tumours and the remarkable chemoresistance.128

S. Mocellin et al. On the basis of ultrastructural and immunohistological features, it is now accepted that most gastrointestinal tumours previously designated as STS (e.g., leiomyosarcomas) are GIST, which probably originate from the pacemaker cells of Cajal.129 To date, surgery is the primary treatment for GIST, as complete resection with negative margins, even in patients with locally advanced tumours, is associated with improved survival.130,131 The 5-year survival rate ranges from 20% to 44% for all patients with GIST and up to 75% for patients with early-stage tumours that have been completely excised. Recent insights into the pathogenesis of these tumours are giving new hope for improving their prognosis. GIST are characterized by the expression of c-Kit, a tyrosine-kinase receptor with cell growth regulatory functions that plays a key role in the pathogenesis of such tumours following the occurrence of activating mutations. The development of small-molecule inhibitors (e.g., tyrosinekinase inhibitor STI571, imatinib) initially tested in myeloid leukemia patients and then proven to mediate impressive tumour regressions in patients with GIST132 has rapidly become a paradigm of molecularly targeted anticancer therapy.133 A recent phase III RCT has demonstrated that patients with advanced GIST benefit significantly from imatinib treatment in terms of progression-free survival.134 Although many issues (e.g., acquired

Figure 1 Molecularly targeted therapeutic approaches to soft tissue sarcoma. TKR: tyrosine-kinase receptor; TNF: tumour necrosis factor; TNFR: TNF receptor; PDGF: platelet-derived growth factor; PDGFR: PDGF receptor; SCF: stem cell factor; EGF: epidermal growth factor; EGFR: EGF receptor; VEGF: vascular endothelial growth factor; VEGFR: VEGF receptor; MMP: matrix metallo-proteinase; mTOR: mammalian target of rapamycin; PI3K: phosphatidylinositol3-kinase; MDR-1: multi-drug resistance protein-1; TERT: telomerase reverse transcriptase; TCR: T-cell receptor.

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches drug resistance, impact on OS, role in the adjuvant/neoadjuvant setting, long-term toxicity) are still being addressed (Table 1), imatinib represents the first active systemic therapy for unresectable locally advanced/metastatic GIST. Among non-gastrointestinal STS not expressing c-Kit, only dermatofibrosarcoma has been shown to be responsive to imatinib.135 The reason for this responsiveness hinges upon the fact that this lowgrade tumour is usually associated with the chromosomal translocation t(17:22), which ultimately results in the constitutive overexpression of platelet-derived growth factor (PDGF)-b. Accordingly, patients with advanced dermatofibrosarcoma may be successfully treated with imatinib, which specifically inhibits the tyrosine-kinase activity of cKit, ABL (myeloid leukemia) and PDGF-receptor. Interestingly, imatinib appears to exert a synergistic effect with standard chemotherapeutic drugs,136 and this might enhance the antitumour activity of their combination.137 On the other side, reports of acquired resistance to imatinib138 have fostered the development and clinical testing of alternative small-molecule inhibitors.139 Antiangiogenic therapy Angiogenesis has become an attractive target for cancer therapy with the potential to be effective for a variety of tumours.140 The use of antiangiogenic agents in the treatment of STS is even more attractive since some of these tumours arise from endothelial cells, and may induce both a direct antitumour effect and an antiangiogenic effect.141 However, the findings from the small series so far published are conflicting. For instance, the smallmolecule tyrosine-kinase inhibitor SU5416, a potent inhibitor of VEGF-receptors, has demonstrated some anti-sarcoma activity, particularly in patients with Kaposi’s sarcoma.142,143 These results have not been confirmed in a subsequent phase II trial.144 In a pilot study with a humanized monoclonal antibody blocking the angiogenesis-related integrinaVb3,145 there were no responses in 15 patients with leiomyosarcoma.146 Moreover, in a recent phase III RCT (highly-active antiretroviral therapy ± IM862) the effectiveness of IM862 (a synthetic dipeptide with antiangiogenic properties) for the treatment of HIV-positive Kaposi’s sarcoma has been refuted.147 A humanized anti-VEGF antibody (bevacizumab), when used in combination with chemotherapy, was shown to significantly improve tumour response and survival rates in patients with metastatic colorectal cancer.148 STS can overexpress VEGF;149,150 noticeably, VEGF overexpression by Ewing’s sarcomas is linked to the activity of the EWS-ETS onco-

17

protein, a product of the sarcoma-specific t(11;22) translocation.151 Therefore, STS might be effectively targeted by anti-VEGF therapy; this is being tested in an ongoing trial. Sorafenib is a potent inhibitor of protein-kinases involved in tumour angiogenesis (e.g., Raf-1, VEGFR, PDGFR, Flt-3, c-Kit) and possesses broadspectrum antitumour activity in cancer xenograft models, where analysis of microvessel density demonstrated significant inhibition of neovascularization.152 This novel drug is under clinical evaluation for the treatment of advanced solid tumours in combination with bevacizumab. Another way to block the VEGF-signaling pathway is to prevent VEGF from binding to its cell-surface receptors by administering soluble decoy-receptors. In animal models, this VEGF ‘‘trap’’ effectively suppresses tumour growth and vascularization in vivo.153 An ongoing phase I study is validating the feasibility of soluble decoy-receptor strategy in patients with lymphoma and advanced solid tumours. Overall, the role of these and other antiangiogenic agents as single anti-sarcoma agents remains to be demonstrated. Favourable preliminary results are prompting investigators to explore the potential of standard or metronomic chemotherapy combined with antiangiogenic agents.154–156 Matrix metalloproteinase inhibitors Matrix-metallo-proteinases (MMP) are a family of zinc-dependent enzymes that mediate degradation of extracellular matrix, a crucial step in tumour invasion and metastasis.157 Although MMP are absent or expressed at low levels in normal tissues, they are upregulated in many tumours, including STS.158–160 After showing promise in preclinical studies, numerous MMP-inhibitors (e.g., marimastat, prinomastat, metastat) are being evaluated for therapy in a variety of cancers.161 In preclinical sarcoma models these drugs have demonstrated antitumour activity in vivo.159,160, 162,163 To date, patients with STS have been enrolled only in small pilot trials.164–166 When administered orally to patients with AIDS-related Kaposis’s sarcoma, Col-3 (an oral chemically modified tetracycline derivative with potent inhibitory effects on MMP activity/production) was well tolerated and showed some antitumour activity.167 A reduced incidence and regression of Kaposi’s sarcoma and other tumours has been reported in AIDS patients treated with antiretroviral combination therapies containing HIV protease inhibitors (e.g., indinavir, saquinavir).147,168 Although these drugs were designed to selectively inhibit the HIV protease activity, they can also induce regression

18 of experimental sarcoma by inhibiting the activation of MMP-2, which ultimately blocks tumour angiogenesis/invasiveness.168 A phase II trial is assessing the effect of this novel class of MMPinhibitors on the progression of Kaposi’s sarcoma in HIV-negative individuals. Inhibitors of growth-factor signaling pathways One of the most active and promising areas of investigation in the field of molecular oncology is the development of drugs inhibiting the components of growth-factor signaling pathways,169 such as the ErbB receptor family, a group of tyrosinekinase receptors including ErbB1 (also known as epidermal growth-factor receptor [EGFR] or HER1), ErbB2 (HER2 or HER2/neu), ErbB3 (or HER3) and ErbB4 (or HER4). EGFR is overexpressed, dysregulated or mutated in many malignancies, including STS.170,171 Two main classes of EGFR inhibitors have been developed including monoclonal antibodies that target the extracellular domain of EGFR (e.g., cetuximab), and smallmolecule tyrosine-kinase inhibitors that target the receptor catalytic domain of EGFR (e.g., gefitinib, erlotinib). These drugs are in advanced phases of clinical experimentation for some epithelial cancers172 and are being tested in STS patients as well. As regards HER2/neu, a humanized recombinant IgG1j blocking antibody (trastuzumab) has been approved for the treatment of breast carcinoma and is in advanced phase of clinical evaluation for head&neck and lung carcinomas.169 The role of HER2/neu as a suitable therapeutic target for STS is debated, as variable results have been reported regarding its expression,173,174 prognostic value,175–177 and pathogenetic function.178,179 A convergence point of activation of several growth-factor receptors is downstream activation of the phosphatidylinositol-3-kinase (PI3K)-AKT/ PKB pathway, which can exert potent anti-apoptotic effects and thus favour tumour survival.180 Another consequence of activation of growthfactor-receptor signaling and/or activation of AKT is activation of the mammalian target of rapamycin (mTOR) pathway, which profoundly affects cell growth by regulating protein translation. mTOR is the target of rapamycin and its analogues, which are currently in clinical trials for the treatment of different cancer types, including STS.181 Gene therapy Although safety concerns have tempered the enthusiasm surrounding gene therapy,182 advances in gene-delivery (e.g., development of third-generation lenti-viral vectors, adenoviral vectors, and

S. Mocellin et al. encapsulated methods of delivering naked DNA183) and gene knock-down (e.g., RNA interference 184) technology are providing the hope for this approach. Results from preclinical sarcoma models are encouraging. For instance, tumour cell transfection with wild-type p53, which is lost in many cases of STS, causes tumour regression in 40% of treated mice185 and restores cancer sensitivity to doxorubicin by downregulating multidrug-resistance protein-1 (MDR1).186 Sarcoma regression has also been reported following both suicide-gene strategy (e.g., adenovirus-associated vector-2 encoding thymidine-kinase)187 and oligo-DNA antisense therapy targeting sarcoma cell survival-/proliferationrelated genes (e.g., insulin-like growth-factor-I receptor;88 Mcl-1, an anti-apoptotic Bcl-2 family member189). More recently, tumour regression has also been reported following RNA interferencemediated knock-down of oncogenes such as Met (a tumour growth/metastasis promoter)190 and survivin (an anti-apoptotic protein correlated with poor prognosis of STS patients).191,192 The tumour-selective delivery of systemic gene therapy remains one of the major limitations of this approach. Isolated limb perfusion, which can efficiently control the leakage from the perfusion circuit to the systemic circulation, has been proposed as a potential gene delivery system for limb STS.62 In animals, this strategy has been already validated for the treatment of STS.193,194 Among other strategies aimed at making highdose chemotherapy more tolerable,195 ‘‘reverse’’ gene therapy might be exploited for the transfer of MDR-1 gene to normal hematopoietic stem-cells and subsequent transplantation, which would reduce the hematotoxicity of antineoplastic drugs.187 Some initial attempts of gene therapy for patients with STS have been reported. In a phase I study the maximum tolerated dose of antisense oligo-DNA specific for type-I protein-kinase-A (a tumour-promoting factor) was determined.196 Using intratumoural injections of a plasmid encoding interleukin-2, other investigators observed tumour stabilization in 6/15 patients.197 Oncolytic adenoviruses lacking the E1B55K gene product for p53 degradation selectively replicate in and lyse p53-deficient tumour cells.198 In clinical trials, these genetically engineered viruses have been administered (together with chemotherapy) to patients with STS via intratumoural injections, and shown some significant antitumour activity.199,200 Some investigators have set-up a transgene encoding human TNF under control of a radiationinduced promoter.201 In a feasibility study, this transgene was coupled with radiotherapy for the treatment of patients with limb STS: of the 13

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches evaluable patients, eleven (85%) showed objective tumour responses (2 complete and 9 partial).202 Of the 11 patients who then underwent surgery, 10 (91%) showed a pathological complete/partial response. Only the conduct of prospective controlled trials will determine the relative contribution of gene therapy to this high tumour response rate. Cancer vaccines Active specific immunotherapy theoretically embodies the ideal tumour-killing system for three main reasons: (1) unlike chemotherapy, which follows a log-kill kinetics, the immune system can hunt-down the minimal residual disease on a single-cell basis; (2) the extreme selectivity of cytotoxic Tlymphocytes towards tumour cells guarantees minimal/absent toxicity; (3) once appropriately trained, the immune system can memorize the targeted tumour, thus, ensuring further protection against disease recurrence. Despite these premises and many successes in animal models, the results of such biotherapy in the clinical setting have not met the expectations owing to the difficulty of appropriately manipulating the highly complex immune network.203 The molecular identification of tumour-associated antigens (TAA) coupled with recent advances in tumour immunology have renewed the enthusiasm for the development of anticancer vaccination strategies.204,205 STS might represent an ideal target for active specific immunization.206,207 TAA commonly expressed by most solid tumours are weakly immunogenic due to their self origin: by contrast, highly immunogenic epitopes can be generated from fusion proteins resulting from STS-specific chromosomal translocations.208,209 These chimeric proteins are exquisitely tumour-specific and -unlike self antigens210 – their use for vaccination is not expected to cause any autoimmune toxicity. Finally, purification of the fusion gene product could be used for anti-sarcoma vaccination, like the use of idiotype protein for vaccination against B-cell lymphomas.211 Although STS express several known TAA212–214 and tumour regression can be observed in sarcoma animal models,215–217 active specific immunotherapy is in its infancy for the treatment of these tumours, only a handful of patients being so far enrolled.218–221 Pilot studies testing the immunogenicity STSspecific fusion site-deriving peptides have been successfully performed in pediatric and adult patients.222–224 Most TAA so far utilized in the clinical setting play a non-vital role in the metabolism of malignant cells (e.g., gp100, MAGE, CEA). Accordingly,

19

selection of tumour cells downregulating these TAA can be the only effect of anticancer vaccination. A new class of TAA is represented by antigens that are required by cancer cells to survive such as survivin and telomerase (a cell survival-related ribonucleoprotein), which have been already tested as TAA in preclinical and clinical models of anticancer vaccination.205 As regards STS, the EWS/ETS chimeric protein identified in most Ewing’s sarcomas promotes the overexpression of telomerase, thus, representing an ideal target for immunotherapy of this tumour.225 A phase I trial is testing the hypothesis that patients with STS can mount an effective immune response against a telomerase-derived peptide.

Conclusions The conduct of RCT over the past two decades has led to significant progress in the therapeutic management of patients with STS. Disease rarity and multidisciplinary treatment require subjects with suspected/known STS be referred to specialized centers where a dedicated team of pathologists, surgeons, oncologists and radiotherapists exists in order to provide patients with the best chance of cure.226 Despite some significant advances in terms of local disease control, further work is necessary to prove that systemic chemotherapy can improve the survival of patients clinically free of disease. Moreover, the characteristic chemoresistance of most STS has so far undermined the clinical utility of conventional antineoplastic agents in the setting of unresectable advanced/metastatic disease, and some investigators believe that current conventional therapy has reached the limits of efficacy.227 Therefore, novel therapeutic strategies are urgently needed to improve the prognosis of these patients. Thanks also to the implementation of novel high-throughput technologies (e.g., DNA-array and proteomics platforms),228,229 recent advances in the elucidation of STS biology are paving the way to the development of molecularly targeted therapeutic strategies. The dissection of the molecular mechanisms underlying cancer development/progression will not only facilitate the discovery of novel tumour ‘‘Achille’s heels’’ potentially targetable by novel cancer-selective drugs, but also allow for the personalization of the therapeutic regimen according to the molecular features of individual tumours. Current criteria for the formulation of a patient’s prognosis and prediction of treatment responsiveness rely upon traditional clinico-pathological factors (e.g., age,

20 performance status size, margin, grade, histological type),110,230,231 which are inadequate to accurately identify tumours with greater intrinsic aggressiveness/treatment resistance.4 The better understanding of the cascade of molecular events underlying STS metastasis/invasiveness and treatment sensitivity is providing investigators with pathogenesis-based information. This is essential both for the identification of patients requiring adjuvant therapy and the selection of the therapeutic approach most likely to be effective in each given patient.232–234 Only the implementation of such data in clinical protocols and the conduct of large trials with the novel tumour-selective drugs will allow investigators to test the enormous potential of the molecularly targeted treatment of STS.

Acknowledgements We thank the authors whose work was not cited due to lengthy considerations.

References 1. Jemal A, Tiwari RC, Murray T, Ghafoor A, Samuels A, Ward E, et al. Cancer statistics, 2004. CA Cancer J Clin 2004;54:8–29. 2. Hogendoorn PC. Pathology of soft tissue sarcomas with emphasis on molecular diagnostic techniques. Eur J Cancer:S205–10. 3. Cormier JN, Pollock RE. Soft tissue sarcomas. CA Cancer J Clin 2004;54:94–109. 4. Borden EC, Baker LH, Bell RS, Bramwell V, Demetri GD, Eisenberg BL, et al. Soft tissue sarcomas of adults: state of the translational science. Clin Cancer Res 2003;9: 1941–56. 5. Brennan M, Alektiar K, Maki R. Sarcomas of the soft tissue and bone. In: De Vita V, Hellman S, Rosenberg S, editors. Cancer: principles and practice of oncology, vol. 2. Philadelphia: Lippincott Williams and Wilkins; 2001. p. 1841–935. 6. O’Sullivan B, Pisters PW. Staging and prognostic factor evaluation in soft tissue sarcoma. Surg Oncol Clin N Am 2003;12:333–53. 7. Helman LJ, Meltzer P. Mechanisms of sarcoma development. Nat Rev Cancer 2003;3:685–94. 8. Fletcher JA. Molecular biology and cytogenetics of soft tissue sarcomas: relevance for targeted therapies. Cancer Treat Res 2004;120:99–116. 9. Clark MA, Thomas JM. Amputation for soft-tissue sarcoma. Lancet Oncol 2003;4:335–42. 10. Rosenberg SA, Tepper J, Glatstein E, Costa J, Baker A, Brennan M, et al. The treatment of soft-tissue sarcomas of the extremities: prospective randomized evaluations of (1) limb-sparing surgery plus radiation therapy compared with amputation and (2) the role of adjuvant chemotherapy. Ann Surg 1982;196:305–15. 11. Brennan MF, Casper ES, Harrison LB, Shiu MH, Gaynor J, Hajdu SI. The role of multimodality therapy in soft-tissue sarcoma. Ann Surg 1991;214:328–36. Discussion 36–8.

S. Mocellin et al. 12. Tukiainen E, Bohling T, Huuhtanen R. Soft tissue sarcoma of the trunk and extremities. Scand J Surg 2003;92: 257–63. 13. Siebenrock KA, Hertel R, Ganz R. Unexpected resection of soft-tissue sarcoma. More mutilating surgery, higher local recurrence rates, and obscure prognosis as consequences of improper surgery. Arch Orthop Trauma Surg 2000;120: 65–9. 14. Zagars GK, Ballo MT, Pisters PW, Pollock RE, Patel SR, Benjamin RS. Surgical margins and resection in the management of patients with soft tissue sarcoma using conservative surgery and radiation therapy. Cancer 2003;97:2544–53. 15. Baldini EH, Goldberg J, Jenner C, Manola JB, Demetri GD, Fletcher CD, et al. Long-term outcomes after functionsparing surgery without radiotherapy for soft tissue sarcoma of the extremities and trunk. J Clin Oncol 1999;17: 3252–9. 16. Stojadinovic A, Leung DH, Hoos A, Jaques DP, Lewis JJ, Brennan MF. Analysis of the prognostic significance of microscopic margins in 2084 localized primary adult soft tissue sarcomas. Ann Surg 2002;235:424–34. 17. Kattan MW, Leung DH, Brennan MF. Postoperative nomogram for 12-year sarcoma-specific death. J Clin Oncol 2002;20:791–6. 18. McKee MD, Liu DF, Brooks JJ, Gibbs JF, Driscoll DL, Kraybill WG. The prognostic significance of margin width for extremity and trunk sarcoma. J Surg Oncol 2004;85: 68–76. 19. Kawaguchi N, Ahmed AR, Matsumoto S, Manabe J, Matsushita Y. The concept of curative margin in surgery for bone and soft tissue sarcoma. Clin Orthop:165–72. 20. Willeke F, Sturm JW. Minimal residual disease in soft-tissue sarcomas. Semin Surg Oncol 2001;20:294–303. 21. Nakasone J, Shimizu T, Gomyo H, Yoshimura Y, Wakabayashi S, Mukaiyama K, et al. Assessment of microinvasion with reverse transcriptase polymerase chain reaction in a case of synovial sarcoma. J Orthop Sci 2004;9:162–5. 22. Consensus Conference. Limb-sparing treatment of adult soft-tissue sarcomas and osteosarcomas. JAMA 1985;254: 1791–4. 23. Lin PP, Guzel VB, Pisters PW, Zagars GK, Weber KL, Feig BW, et al. Surgical management of soft tissue sarcomas of the hand and foot. Cancer 2002;95:852–61. 24. Pisters PW, Leung DH, Woodruff J, Shi W, Brennan MF. Analysis of prognostic factors in 1041 patients with localized soft tissue sarcomas of the extremities. J Clin Oncol 1996;14:1679–89. 25. Gerrand CH, Wunder JS, Kandel RA, O’Sullivan B, Catton RS, Bell RS, et al. Classification of positive margins after resection of soft-tissue sarcoma of the limb predicts the risk of local recurrence. J Bone Joint Surg Br 2001;83: 1149–55. 26. Fleming JB, Berman RS, Cheng SC, Chen NP, Hunt KK, Feig BW, et al. Long-term outcome of patients with American Joint Committee on Cancer stage IIB extremity soft tissue sarcomas. J Clin Oncol 1999;17:2772–80. 27. Rossi CR, Foletto M, Alessio S, Menin N, D’Amore E, Rigon A, et al. Limb-sparing treatment for soft tissue sarcomas: influence of prognostic factors. J Surg Oncol 1996;63:3–8. 28. Stojadinovic A, Jaques DP, Leung DH, Healey JH, Brennan MF. Amputation for recurrent soft tissue sarcoma of the extremity: indications and outcome. Ann Surg Oncol 2001;8:509–18. 29. Eilber FC, Rosen G, Nelson SD, Selch M, Dorey F, Eckardt J, et al. High-grade extremity soft tissue sarcomas: factors

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

predictive of local recurrence and its effect on morbidity and mortality. Ann Surg 2003;237:218–26. Pisters PW, Harrison LB, Leung DH, Woodruff JM, Casper MF, Brennan MF. Long-term results of a prospective randomized trial of adjuvant brachytherapy in soft tissue sarcoma. J Clin Oncol 1996;14:859–68. Yang JC, Chang AE, Baker AR, Sindelar WF, Danforth DN, Topalian SL, et al. Randomized prospective study of the benefit of adjuvant radiation therapy in the treatment of soft tissue sarcomas of the extremity. J Clin Oncol 1998;16:197–203. Sondak VK, Robertson JM, Sussman JJ, Saran PA, Chang AE, Lawrence TS. Preoperative idoxuridine and radiation for large soft tissue sarcomas: clinical results with five-year follow-up. Ann Surg Oncol 1998;5:106–12. Zagars GK, Ballo MT, Pisters PW, Pollock RE, Patel SR, Benjamin RS, et al. Prognostic factors for patients with localized soft-tissue sarcoma treated with conservation surgery and radiation therapy: an analysis of 225 patients. Cancer 2003;97:2530–43. Alektiar KM, Leung D, Zelefsky MJ, Healey JH, Brennan MF. Adjuvant brachytherapy for primary high-grade soft tissue sarcoma of the extremity. Ann Surg Oncol 2002;9:48–56. Strander H, Turesson I, Cavallin-Stahl E. A systematic overview of radiation therapy effects in soft tissue sarcomas. Acta Oncol 2003;42:516–31. Alektiar KM, Velasco J, Zelefsky MJ, Woodruff JM, Lewis MF, Brennan MF. Adjuvant radiotherapy for margin-positive high-grade soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 2000;48:1051–8. Khanfir K, Alzieu L, Terrier P, Le Pechoux C, Bonvalot S, Vanel D, et al. Does adjuvant radiation therapy increase loco-regional control after optimal resection of soft-tissue sarcoma of the extremities. Eur J Cancer 2003;39: 1872–80. Coindre JM, Terrier P, Bui NB, Bonichon F, Collin F, Le Doussal V, et al. Prognostic factors in adult patients with locally controlled soft tissue sarcoma. A study of 546 patients from the French Federation of Cancer Centers Sarcoma Group. J Clin Oncol 1996;14:869–77. Geer RJ, Woodruff J, Casper ES, Brennan MF. Management of small soft-tissue sarcoma of the extremity in adults. Arch Surg 1992;127:1285–9. Karakousis CP, Zografos GC. Radiation therapy for high grade soft tissue sarcomas of the extremities treated with limb-preserving surgery. Eur J Surg Oncol 2002;28:431–6. O’Sullivan B, Ward I, Catton C. Recent advances in radiotherapy for soft-tissue sarcoma. Curr Oncol Rep 2003;5:274–81. Pollack A, Zagars GK, Goswitz MS, Pollock RA, Feig BW, Pisters PW. Preoperative vs. postoperative radiotherapy in the treatment of soft tissue sarcomas: a matter of presentation. Int J Radiat Oncol Biol Phys 1998;42: 563–72. O’Sullivan B, Davis AM, Turcotte R, Bell R, Catton C, Chabot P, et al. Preoperative versus postoperative radiotherapy in soft-tissue sarcoma of the limbs: a randomised trial. Lancet 2002;359:2235–41. McCarter MD, Jaques DP, Brennan MF. Randomized clinical trials in soft tissue sarcoma. Surg Oncol Clin N Am 2002;11:11–22. Adjuvant chemotherapy for localised resectable soft-tissue sarcoma of adults: meta-analysis of individual data. Sarcoma Meta-analysis Collaboration. Lancet 1997;350: 1647–54. Frustaci S, Gherlinzoni F, De Paoli A, Bonetti M, Azzarelli A, Comandone A, et al. Adjuvant chemotherapy for adult

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

21

soft tissue sarcomas of the extremities and girdles: results of the Italian randomized cooperative trial. J Clin Oncol 2001;19:1238–47. Cormier JN, Huang X, Xing Y, Thall PF, Wang X, Benjamin RS, et al. Cohort analysis of patients with localized, highrisk, extremity soft tissue sarcoma treated at two cancer centers: chemotherapy-associated outcomes. J Clin Oncol 2004;22:4567–74. Bramwell VH. Adjuvant chemotherapy for adult soft tissue sarcoma: Is there a standard of care? J Clin Oncol 2001;19:1235–7. Meric F, Milas M, Hunt KK, Hess KR, Pisters PW, Hildebrandt G, et al. Impact of neoadjuvant chemotherapy on postoperative morbidity in soft tissue sarcomas. J Clin Oncol 2000;18:3378–83. Pisters PW, Patel SR, Varma DG, Cheng SC, Chen NP, Nguyen HT, et al. Preoperative chemotherapy for stage IIIB extremity soft tissue sarcoma: long-term results from a single institution. J Clin Oncol 1997;15:3481–7. Grobmyer SR, Maki RG, Demetri GD, Mazumdar M, Riedel E, Brennan MF, et al. Neo-adjuvant chemotherapy for primary high-grade extremity soft tissue sarcoma. Ann Oncol 2004;15:1667–72. Gortzak E, Azzarelli A, Buesa J, Bramwell VH, van Coevorden F, van Geel AN, et al. A randomised phase II study on neo-adjuvant chemotherapy for ‘high-risk’ adult soft-tissue sarcoma. Eur J Cancer 2001;37:1096–103. Eilber FC, Rosen G, Eckardt J, Forscher C, Nelson SD, Selch M, et al. Treatment-induced pathologic necrosis: a predictor of local recurrence and survival in patients receiving neoadjuvant therapy for high-grade extremity soft tissue sarcomas. J Clin Oncol 2001;19:3203–9. Meric F, Hess KR, Varma DG, Hunt KK, Pisters PW, Milas KM, et al. Radiographic response to neoadjuvant chemotherapy is a predictor of local control and survival in soft tissue sarcomas. Cancer 2002;95:1120–6. Pisters PW, Ballo MT, Patel SR. Preoperative chemoradiation treatment strategies for localized sarcoma. Ann Surg Oncol 2002;9:535–42. Wanebo HJ, Temple WJ, Popp MB, Constable W, Aron B, Cunningham SL. Preoperative regional therapy for extremity sarcoma. A tricenter update. Cancer 1995;75: 2299–306. Levine EA, Trippon M, Das Gupta TK. Preoperative multimodality treatment for soft tissue sarcomas. Cancer 1993;71:3685–9. Edmonson JH, Petersen IA, Shives TC, Mahoney MR, Rock MG, Haddock MG, et al. Chemotherapy, irradiation, and surgery for function-preserving therapy of primary extremity soft tissue sarcomas: initial treatment with ifosfamide, mitomycin, doxorubicin, and cisplatin plus granulocyte macrophage-colony-stimulating factor. Cancer 2002;94: 786–92. DeLaney TF, Spiro IJ, Suit HD, Gebhardt MC, Hornicek FJ, Mankin HJ, et al. Neoadjuvant chemotherapy and radiotherapy for large extremity soft-tissue sarcomas. Int J Radiat Oncol Biol Phys 2003;56:1117–27. Pisters PW, Ballo MT, Fenstermacher MJ, Feig BW, Hunt KA, Raymond KA, et al. Phase I trial of preoperative concurrent doxorubicin and radiation therapy, surgical resection, and intraoperative electron-beam radiation therapy for patients with localized retroperitoneal sarcoma. J Clin Oncol 2003;21:3092–7. Henshaw RM, Priebat DA, Perry DJ, Shmookler BM, Malawer MM. Survival after induction chemotherapy and surgical resection for high-grade soft tissue sarcoma. Is radiation necessary? Ann Surg Oncol 2001;8:484–95.

22 62. Eggermont AM, de Wilt JH, ten Hagen TL. Current uses of isolated limb perfusion in the clinic and a model system for new strategies. Lancet Oncol 2003;4:429–37. 63. Rossi CR, Mocellin S, Pilati P, Foletto M, Nitti D, Lise M. TNFalpha-based isolated perfusion for limb-threatening soft tissue sarcomas: state of the art and future trends. J Immunother 2003;26:291–300. 64. Mocellin S, Rossi CR, Pilati P, Nitti D. Tumor necrosis factor, cancer and anticancer therapy. Cytokine Growth Factor Rev 2005;16:35–53. 65. Eggermont AM, Schraffordt Koops H, Klausner JM, Kroon PM, Schlag PM, Lienard D, et al. Isolated limb perfusion with tumour necrosis factor and melphalan for limb salvage in 186 patients with locally advanced soft tissue extremity sarcomas. The cumulative multicenter European experience. Ann Surg 1996;224:756–64.. Discussion 64–5. 66. Rossi CR, Mocellin S, Pilati P, Foletto M, Campana L, Quintieri L, et al. Hyperthermic isolated perfusion with low-dose tumor necrosis factor a and doxorubicin for the treatment of limb-threatening soft tissue sarcomas. Ann Surg Oncol 2005;12(5):398–405. 67. Plaat BE, Molenaar WM, Mastik MF, Koudstaal J, van den Berg E, Koops HS, et al. Hyperthermic isolated limb perfusion with tumour necrosis factor-alpha and melphalan in patients with locally advanced soft tissue sarcomas: treatment response and clinical outcome related to changes in proliferation and apoptosis. Clin Cancer Res 1999;5:1650–7. 68. Lejeune FJ, Pujol N, Lienard D, Mosimann F, Raffoul W, Genton A, et al. Limb salvage by neoadjuvant isolated perfusion with TNFalpha and melphalan for non-resectable soft tissue sarcoma of the extremities. Eur J Surg Oncol 2000;26:669–78. 69. Noorda EM, Vrouenraets BC, Nieweg OE, van Coevorden F, van Slooten GW, Kroon BB. Isolated limb perfusion with tumour necrosis factor-alpha and melphalan for patients with unresectable soft tissue sarcoma of the extremities. Cancer 2003;98:1483–90. 70. Casara D, Rubello D, Pilati P, Scalerta R, Foletto M, Rossi CR. Optimized procedure of real-time systemic leakage monitoring during isolated limb perfusion using a hand held gamma probe and 99mTc-HSA. Nucl Med Commun 2004;25: 61–6. 71. Noorda EM, Vrouenraets BC, Nieweg OE, Van Coevorden F, Kroon BB. Isolated limb perfusion: what is the evidence for its use? Ann Surg Oncol 2004;11:837–45. 72. Lewis JJ, Leung D, Woodruff JM, Brennan MF. Retroperitoneal soft-tissue sarcoma: analysis of 500 patients treated and followed at a single institution. Ann Surg 1998;228: 355–65. 73. Stoeckle E, Coindre JM, Bonvalot S, Kantor G, Terrier P, Bonichon F, et al. Prognostic factors in retroperitoneal sarcoma: a multivariate analysis of a series of 165 patients of the French Cancer Center Federation Sarcoma Group. Cancer 2001;92:359–68. 74. Youssef E, Fontanesi J, Mott M, Kraut M, Lucas D, Mekhael H, et al. Long-term outcome of combined modality therapy in retroperitoneal and deep-trunk soft-tissue sarcoma: analysis of prognostic factors. Int J Radiat Oncol Biol Phys 2002;54:514–9. 75. Hassan I, Park SZ, Donohue JH, Nagorney DM, Kay PA, Nasciemento AG, et al. Operative management of primary retroperitoneal sarcomas: a reappraisal of an institutional experience. Ann Surg 2004;239:244–50. 76. Gronchi A, Casali PG, Fiore M, Mariani L, Lo Vullo S, Bertulli R, et al. Retroperitoneal soft tissue sarcomas: patterns of

S. Mocellin et al.

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

recurrence in 167 patients treated at a single institution. Cancer 2004;100:2448–55. Pisters PW, O’Sullivan B. Retroperitoneal sarcomas: combined modality treatment approaches. Curr Opin Oncol 2002;14:400–5. Pirayesh A, Chee Y, Helliwell T, Hershman M, Leinster S, Fordham M, et al. The management of retroperitoneal soft tissue sarcoma: a single institution experience with a review of the literature. Eur J Surg Oncol 2001;27:491–7. Gilbeau L, Kantor G, Stoeckle E, Lagarde P, Thomas L, Kind M, et al. Surgical resection and radiotherapy for primary retroperitoneal soft tissue sarcoma. Radiother Oncol 2002;65:137–43. Jones J, Catton C, O’Sullivan B, Couture J, Heisler R, Kandel R, et al. Initial results of a trial of preoperative external beam radiation therapy and postoperative brachytherapy for retroperitoneal sarcoma. Ann Surg Oncol 2002;9:346–54. Webb S. Advances in three-dimensional conformal radiation therapy physics with intensity modulation. Lancet Oncol 2000;1:30–6. Wendtner CM, Abdel-Rahman S, Krych M, Baumert J, Lindner LH, Baur A, et al. Response to neoadjuvant chemotherapy combined with regional hyperthermia predicts long-term survival for adult patients with retroperitoneal and visceral high-risk soft tissue sarcomas. J Clin Oncol 2002;20:3156–64. Petersen I, Haddock M, Donohue J, Nagorney D, Grill J, Sargent D, et al. Use of intraoperative electron beam radiotherapy in the management of retroperitoneal soft tissue sarcomas. Int J Radiat Oncol Biol Phys 2002;52: 469–75. Bobin JY, Al-Lawati T, Granero LE, Adham M, Romestaing O, Chapet O, et al. Surgical management of retroperitoneal sarcomas associated with external and intraoperative electron beam radiotherapy. Eur J Surg Oncol 2003;29: 676–81. Alektiar KM, Hu K, Anderson L, Brennan MF, Harrison LB. High-dose-rate intraoperative radiation therapy (HDRIORT) for retroperitoneal sarcomas. Int J Radiat Oncol Biol Phys 2000;47:157–63. Gieschen HL, Spiro IJ, Suit HD, Ott MJ, Rattner DW, Ancukiewicz M, et al. Long-term results of intraoperative electron beam radiotherapy for primary and recurrent retroperitoneal soft tissue sarcoma. Int J Radiat Oncol Biol Phys 2001;50:127–31. Sindelar WF, Kinsella TJ, Chen PW, DeLaney TF, Tepper JE, Rosenberg SA, et al. Intraoperative radiotherapy in retroperitoneal sarcomas. Final results of a prospective, randomized, clinical trial. Arch Surg 1993;128:402–10. Verwaal VJ, van Ruth S, de Bree E, van Sloothen GW, van Tinteren H, Boot H, et al. Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 2003;21:3737–43. Pilati P, Mocellin S, Rossi C, Foletto M, Campana L, Nitti D, et al. Cytoreductive surgery combined with hyperthermic intraperitoneal intraoperative chemotherapy for peritoneal carcinomatosis arising from colon adenocarcinoma. Ann Surg Oncol 2003;10:508–13. Rossi C, Foletto M, Mocellin S, Pilati P, De Simone M, Deraco M, et al. Hyperthermic intraoperative intraperitoneal chemotherapy with cisplatin and doxorubicin in patients who undergo cytoreductive surgery for peritoneal carcinomatosis and sarcomatosis. Cancer 2002;94: 492–9.

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches 91. Rossi CR, Deraco M, De Simone M, Mocellin S, Pilati P, Foletto M, et al. Hyperthermic intraperitoneal intraoperative chemotherapy after cytoreductive surgery for the treatment of abdominal sarcomatosis: clinical outcome and prognostic factors in 60 consecutive patients. Cancer 2004;100:1943–50. 92. Glehen O, Mohamed F, Gilly FN. Peritoneal carcinomatosis from digestive tract cancer: new management by cytoreductive surgery and intraperitoneal chemohyperthermia. Lancet Oncol 2004;5:219–28. 93. Bartlett DL, Buell JF, Libutti SK, Reed E, Lee KB, Figg WD, et al. A phase I trial of continuous hyperthermic peritoneal perfusion with tumour necrosis factor and cisplatin in the treatment of peritoneal carcinomatosis. Cancer 1998;83: 1251–61. 94. Brown SB, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol 2004;5:497–508. 95. Wilson JJ, Jones H, Burock M, Smith D, Fraker DL, Metz J, et al. Patterns of recurrence in patients treated with photodynamic therapy for intraperitoneal carcinomatosis and sarcomatosis. Int J Oncol 2004;24:711–7. 96. Bauer TW, Hahn SM, Spitz FR, Kachur A, Glatstein E, Fraker DL. Preliminary report of photodynamic therapy for intraperitoneal sarcomatosis. Ann Surg Oncol 2001;8: 254–9. 97. Hendren SK, Hahn SM, Spitz FR, Bauer TW, Rubin SC, Zhu T, et al. Phase II trial of debulking surgery and photodynamic therapy for disseminated intraperitoneal tumours. Ann Surg Oncol 2001;8:65–71. 98. Billingsley KG, Lewis JJ, Leung DH, Casper ES, Woodruff MF, Brennan MF. Multifactorial analysis of the survival of patients with distant metastasis arising from primary extremity sarcoma. Cancer 1999;85:389–95. 99. Zagars GK, Ballo MT, Pisters PW, Pollock RE, Patel SR, Benjamin RS. Prognostic factors for disease-specific survival after first relapse of soft-tissue sarcoma: analysis of 402 patients with disease relapse after initial conservative surgery and radiotherapy. Int J Radiat Oncol Biol Phys 2003;57:739–47. 100. Stojadinovic A, Yeh A, Brennan MF. Completely resected recurrent soft tissue sarcoma: primary anatomic site governs outcomes. J Am Coll Surg 2002;194:436–47. 101. Weiser MR, Downey RJ, Leung DH, Brennan MF. Repeat resection of pulmonary metastases in patients with softtissue sarcoma. J Am Coll Surg 2000;191:184–90. Discussion 90–1. 102. Abdalla EK, Pisters PW. Metastasectomy for limited metastases from soft tissue sarcoma. Curr Treat Options Oncol 2002;3:497–505. 103. Midis GP, Pollock RE, Chen NP, Feig BW, Murphy A, Pollack A, et al. Locally recurrent soft tissue sarcoma of the extremities. Surgery 1998;123:666–71. 104. Catton C, Davis A, Bell R, O’Sullivan B, Fornasier V, Wunder J, et al. Soft tissue sarcoma of the extremity. Limb salvage after failure of combined conservative therapy. Radiother Oncol 1996;41:209–14. 105. Popov P, Tukiainen E, Asko-Seljavaara S, Huuhtanen R, Virolainen M, Virkkunen P, et al. Soft-tissue sarcomas of the upper extremity: surgical treatment and outcome. Plast Reconstr Surg 2004;113:222–30. Discussion 31–2. 106. Temple LK, Brennan MF. The role of pulmonary metastasectomy in soft tissue sarcoma. Semin Thorac Cardiovasc Surg 2002;14:35–44. 107. Blay JY, van Glabbeke M, Verweij J, van Oosterom AT, Le Cesne A, Oosterhuis JW, et al. Advanced soft-tissue sarcoma: a disease that is potentially curable for a subset

108.

109. 110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121. 122.

23

of patients treated with chemotherapy. Eur J Cancer 2003;39:64–9. Benjamin RS, Rouesse J, Bourgeois H, van Hoesel QG. Should patients with advanced sarcomas be treated with chemotherapy? Eur J Cancer 1998;34:958–65. Spira AI, Ettinger DS. The use of chemotherapy in softtissue sarcomas. Oncologist 2002;7:348–59. Van Glabbeke M, van Oosterom AT, Oosterhuis JW, Mouridsen H, Crowther D, Somers R, et al. Prognostic factors for the outcome of chemotherapy in advanced soft tissue sarcoma: an analysis of 2185 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;17:150–7. Bramwell VH, Anderson D, Charette ML. Doxorubicin-based chemotherapy for the palliative treatment of adult patients with locally advanced or metastatic soft tissue sarcoma. Cochrane Database Syst Rev 2003:CD003293. Spillane AJ, A’Hern R, Judson IR, Fisher C, Thomas JM. Synovial sarcoma: a clinicopathologic, staging, and prognostic assessment. J Clin Oncol 2000;18:3794–803. Spurrell EL, Fisher C, Thomas JM, Judson IR. Prognostic factors in advanced synovial sarcoma: an analysis of 104 patients treated at the Royal Marsden Hospital. Ann Oncol. Patel SR, Vadhan-Raj S, Papadopolous N, Plager C, Burgess C, Hays C, 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;15:2378–84. Lopez M, Vici P, Di Lauro L, Carpano S. Increasing single epirubicin doses in advanced soft tissue sarcomas. J Clin Oncol 2002;20:1329–34. Worden FP, Taylor JM, Biermann JS, Sondak VK, Leu KM, Chugh R, et al. Randomized phase II evaluation of 6 g/m2 of ifosfamide plus doxorubicin and granulocyte colonystimulating factor (G-CSF) compared with 12 g/m2 of ifosfamide plus doxorubicin and G-CSF in the treatment of poor-prognosis soft tissue sarcoma. J Clin Oncol 2005;23:105–12. Le Cesne A, Judson I, Crowther D, Rodenhuis S, Keizer HJ, Van Hoesel Q, et al. Randomized phase III study comparing conventional-dose doxorubicin plus ifosfamide versus highdose 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: 2676–84. Bertuzzi A, Castagna L, Nozza A, Quagliuolo V, Siracusano M, Balzarotti M, et al. High-dose chemotherapy in poorprognosis adult small round-cell tumours: clinical and molecular results from a prospective study. J Clin Oncol 2002;20:2181–8. Sawyer M, Bramwell V. The treatment of distant metastases in soft tissue sarcoma. Semin Radiat Oncol 1999;9: 389–400. Blay JY, Bouhour D, Ray-Coquard I, Dumontet C, Philip T, Biron P. High-dose chemotherapy with autologous hematopoietic stem-cell transplantation for advanced soft tissue sarcoma in adults. J Clin Oncol 2000;18:3643–50. Reichardt P. High-dose chemotherapy in adult soft tissue sarcoma. Crit Rev Oncol Hematol 2002;41:157–67. Hensley ML, Maki R, Venkatraman E, Geller G, Lovegren M, Aghajanian C, et al. Gemcitabine and docetaxel in patients with unresectable leiomyosarcoma: results of a phase II trial. J Clin Oncol 2002;20:2824–31.

24 123. Leu KM, Ostruszka LJ, Shewach D, Zalupski M, Sondak V, Biermann JS, 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;22:1706–12. 124. Kostler WJ, Brodowicz T, Attems Y, Hejna M, Tomek S, Amann G, et al. Docetaxel as rescue medication in anthracycline- and ifosfamide-resistant locally advanced or metastatic soft tissue sarcoma: results of a phase II trial. Ann Oncol 2001;12:1281–8. 125. Papai Z, Bodoky G, Szanto J, Poller I, Rahoty P, Eckhardt S, et al. The efficacy of a combination of etoposide, ifosfamide, and cisplatin in the treatment of patients with soft tissue sarcoma. Cancer 2000;89:177–80. 126. Le Cesne A, Blay JY, Judson I, Van Oosterom A, Verweij J, Radford J, 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;23:576–84. 127. Mackall CL, Meltzer PS, Helman LJ. Focus on sarcomas. Cancer Cell 2002;2:175–8. 128. Plaat BE, Hollema H, Molenaar WM, Torn Broers GH, Pijpe MF, Mastik MF, et al. Soft tissue leiomyosarcomas and malignant gastrointestinal stromal tumours: differences in clinical outcome and expression of multidrug resistance proteins. J Clin Oncol 2000;18:3211–20. 129. Rossi CR, Mocellin S, Mencarelli R, Foletto M, Pilati P, Nitti D, et al. Gastrointestinal stromal tumours: from a surgical to a molecular approach. Int J Cancer 2003;107:171–6. 130. Joensuu H, Fletcher C, Dimitrijevic S, Silberman S, Roberts G, Demetri G. Management of malignant gastrointestinal stromal tumours. Lancet Oncol 2002;3:655–64. 131. DeMatteo RP, Lewis JJ, Leung D, Mudan SS, Woodruff JM, Brennan MF. Two hundred gastrointestinal stromal tumours: recurrence patterns and prognostic factors for survival. Ann Surg 2000;231:51–8. 132. Demetri GD, von Mehren M, Blanke CD, Van den Abbeele B, Eisenberg B, Roberts PJ, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumours. N Engl J Med 2002;347:472–80. 133. Druker BJ. Imatinib as a paradigm of targeted therapies. Adv Cancer Res 2004;91:1–30. 134. Verweij J, Casali PG, Zalcberg J, LeCesne A, Reichardt P, Blay JY, et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet 2004;364:1127–34. 135. McArthur G. Molecularly targeted treatment for dermatofibrosarcoma protuberans. Semin Oncol 2004;31:30–6. 136. Gonzalez I, Andreu EJ, Panizo A, Inoges S, Fontalba A, Fernandez-Luna JL, et al. Imatinib inhibits proliferation of Ewing tumour cells mediated by the stem cell factor/KIT receptor pathway, and sensitizes cells to vincristine and doxorubicin-induced apoptosis. Clin Cancer Res 2004;10: 751–61. 137. Scotlandi K, Manara MC, Strammiello R, Landuzzi L, Benini S, Perdichizzi S, et al. C-kit receptor expression in Ewing’s sarcoma: lack of prognostic value but therapeutic targeting opportunities in appropriate conditions. J Clin Oncol 2003;21:1952–60. 138. Chen LL, Trent JC, Wu EF, Fuller GN, Ramdas L, Zhang W, et al. A missense mutation in KIT kinase domain 1 correlates with imatinib resistance in gastrointestinal stromal tumours. Cancer Res 2004;64:5913–9. 139. Deininger MW, Druker BJ. SR Circumventing imatinib resistance. Cancer Cell 2004;6:108–10. 140. Cao Y. Antiangiogenic cancer therapy. Semin Cancer Biol 2004;14:139–45.

S. Mocellin et al. 141. Heymach JV. Angiogenesis and antiangiogenic approaches to sarcomas. Curr Opin Oncol 2001;13:261–9. 142. Stopeck A, Sheldon M, Vahedian M, Cropp G, Gosalia R, Hannah A. Results of a phase I dose-escalating study of the antiangiogenic agent, SU5416, in patients with advanced malignancies. Clin Cancer Res 2002;8:2798–805. 143. Kuenen BC, Tabernero J, Baselga J, Cavalli F, Pfanner E, Conte PF, et al. Efficacy and toxicity of the angiogenesis inhibitor SU5416 as a single agent in patients with advanced renal cell carcinoma, melanoma, and soft tissue sarcoma. Clin Cancer Res 2003;9:1648–55. 144. Heymach JV, Desai J, Manola J, Davis DW, McConkey DJ, Harmon D, et al. Phase II study of the antiangiogenic agent SU5416 in patients with advanced soft tissue sarcomas. Clin Cancer Res 2004;10:5732–40. 145. Gutheil JC, Campbell TN, Pierce PR, Watkins JD, Huse WD, Bodkin DJ, et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res 2000;6: 3056–61. 146. Patel SR, Jenkins J, Papadopolous N, Burgess MA, Plager C, Gutterman J, et al. Pilot study of vitaxin – an angiogenesis inhibitor-in patients with advanced leiomyosarcomas. Cancer 2001;92:1347–8. 147. Noy A, Scadden DT, Lee J, Dezube BJ, Aboulafia D, Tulpule A, et al. Angiogenesis inhibitor IM862 is ineffective against AIDS-Kaposi’s sarcoma in a phase III trial, but highlights sustained, potent effect of highly active antiretroviral therapy: from the AIDS Malignancy Consortium and IM862 Study Team. J Clin Oncol. 148. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumour growth and angiogenesis. J Clin Oncol. 149. Chao C, Al-Saleem T, Brooks JJ, Rogatko A, Kraybill WG, Eisenberg B. Vascular endothelial growth factor and soft tissue sarcomas: tumour expression correlates with grade. Ann Surg Oncol 2001;8:260–7. 150. Potti A, Ganti AK, Tendulkar K, Sholes K, Chitajallu S, Koch M, et al. Determination of vascular endothelial growth factor (VEGF) overexpression in soft tissue sarcomas and the role of overexpression in leiomyosarcoma. J Cancer Res Clin Oncol 2004;130:52–6. 151. Fuchs B, Inwards CY, Janknecht R. Vascular endothelial growth factor expression is up-regulated by EWS-ETS oncoproteins and Sp1 and may represent an independent predictor of survival in Ewing’s sarcoma. Clin Cancer Res 2004;10:1344–53. 152. Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, et al. BAY 43-9006 exhibits broad spectrum oral antitumour activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumour progression and angiogenesis. Cancer Res 2004;64: 7099–109. 153. Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, et al. VEGF-Trap: a VEGF blocker with potent antitumour effects. Proc Natl Acad Sci USA 2002;99:11393–8. 154. Tran HT, Blumenschein Jr GR, Lu C, Meyers CA, Papadimitrakopoulou V, Fossella FV, et al. Clinical and pharmacokinetic study of TNP-470, an angiogenesis inhibitor, in combination with paclitaxel and carboplatin in patients with solid tumours. Cancer Chemother Pharmacol 2004;54:308–14. 155. Vogt T, Hafner C, Bross K, Bataille F, Jauch KW, Berand A, et al. Antiangiogenetic therapy with pioglitazone, rofecoxib, and metronomic trofosfamide in patients with advanced malignant vascular tumours. Cancer 2003;98: 2251–6.

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches 156. Reichle A, Bross K, Vogt T, Bataille F, Wild P, Berand A, et al. Pioglitazone and rofecoxib combined with angiostatically scheduled trofosfamide in the treatment of faradvanced melanoma and soft tissue sarcoma. Cancer 2004;101:2247–56. 157. Hidalgo M, Eckhardt SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst 2001;93:178–93. 158. Saito T, Oda Y, Sakamoto A, Tamiya S, Iwamoto Y, Tsuneyoshi M. Matrix metalloproteinase-2 expression correlates with morphological and immunohistochemical epithelial characteristics in synovial sarcoma. Histopathology 2002;40:279–85. 159. Sanceau J, Truchet S, Bauvois B. Matrix metalloproteinase9 silencing by RNA interference triggers the migratoryadhesive switch in Ewing’s sarcoma cells. J Biol Chem 2003;278:36537–46. 160. Sato T, Koike L, Miyata Y, Hirata M, Mimaki Y, Sashida Y, et al. Inhibition of activator protein-1 binding activity and phosphatidylinositol 3-kinase pathway by nobiletin, a polymethoxy flavonoid, results in augmentation of tissue inhibitor of metalloproteinases-1 production and suppression of production of matrix metalloproteinases-1 and -9 in human fibrosarcoma HT-1080 cells. Cancer Res 2002;62: 1025–9. 161. Overall CM, Lopez-Otin C. Strategies for MMP inhibition in cancer: innovations for the post-trial era. Nat Rev Cancer 2002;2:657–72. 162. Albini A, Morini M, D’Agostini F, Ferrari N, Campelli F, Arena G, et al. Inhibition of angiogenesis-driven Kaposi’s sarcoma tumour growth in nude mice by oral N-acetylcysteine. Cancer Res 2001;61:8171–8. 163. Bremer C, Tung CH, Weissleder R. In vivo molecular target assessment of matrix metalloproteinase inhibition. Nat Med 2001;7:743–8. 164. Syed S, Takimoto C, Hidalgo M, Rizzo J, Kuhn JG, Hammond LA, et al. A phase I and pharmacokinetic study of Col-3 (Metastat), an oral tetracycline derivative with potent matrix metalloproteinase and antitumour properties. Clin Cancer Res 2004;10:6512–21. 165. Pantanowitz L, Dezube BJ, Pinkus GS, Tahan SR. Histological characterization of regression in acquired immunodeficiency syndrome-related Kaposi’s sarcoma. J Cutan Pathol 2004;31:26–34. 166. Rudek MA, Figg WD, Dyer V, Dahut W, Turner ML, Steinberg SM, et al. Phase I clinical trial of oral COL-3, a matrix metalloproteinase inhibitor, in patients with refractory metastatic cancer. J Clin Oncol 2001;19: 584–92. 167. Cianfrocca M, Cooley TP, Lee JY, Rudek MA, Scadden DT, Ratner L, et al. Matrix metalloproteinase inhibitor COL-3 in the treatment of AIDS-related Kaposi’s sarcoma: a phase I AIDS malignancy consortium study. J Clin Oncol 2002;20:153–9. 168. Monini P, Sgadari C, Toschi E, Barillari G, Ensoli B. Antitumour effects of antiretroviral therapy. Nat Rev Cancer 2004;4:861–75. 169. Harari PM. Epidermal growth factor receptor inhibition strategies in oncology. Endocr Relat Cancer 2004;11: 689–708. 170. Beech D, Pollock RE, Tsan R, Radinsky R. Epidermal growth factor receptor and insulin-like growth factor-I receptor expression and function in human soft-tissue sarcoma cells. Int J Oncol 1998;12:329–36. 171. Nielsen TO, Hsu FD, O’Connell JX, Gilks CB, Sorensen PH, Linn S, et al. Tissue microarray validation of epidermal growth factor receptor and SALL2 in synovial sarcoma with

172. 173.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183. 184.

185.

186.

187.

188.

189.

25

comparison to tumours of similar histology. Am J Pathol 2003;163:1449–56. El-Rayes BF, LoRusso PM. Targeting the epidermal growth factor receptor. Br J Cancer 2004;91:418–24. Allander SV, Illei PB, Chen Y, Antonescu CR, Bittner M, Ladanyi M, et al. Expression profiling of synovial sarcoma by cDNA microarrays: association of ERBB2, IGFBP2, and ELF3 with epithelial differentiation. Am J Pathol 2002;161:1587–95. Amir G, Issakov J, Meller I, Sucher E, Peyser A, Cohen IJ, et al. Expression of p53 gene product and cell proliferation marker Ki-67 in Ewing’s sarcoma: correlation with clinical outcome. Hum Pathol 2002;33:170–4. Foster H, Knox S, Ganti AK, Hebert BJ, Koch M, Tendulkar K, et al. HER-2/neu overexpression detected by immunohistochemistry in soft tissue sarcomas. Am J Clin Oncol 2003;26:188–91. Nuciforo PG, Pellegrini C, Fasani R, Maggioni M, Coggi G, Parafioriti A, et al. Molecular and immunohistochemical analysis of HER2/neu oncogene in synovial sarcoma. Hum Pathol 2003;34:639–45. Sato T, Peiper M, Heinecke A, Zurakowski D, Eisenberger S, Hosch S, et al. Expression of HER2/neu does not correlate with survival in soft tissue sarcoma. Onkologie 2003;26:268–71. Nanni P, Nicoletti G, De Giovanni C, Croci S, Astolfi A, Landuzzi L, et al. Development of rhabdomyosarcoma in HER-2/neu transgenic p53 mutant mice. Cancer Res 2003;63:2728–32. Ye D, Maitra A, Timmons CF, Leavey PJ, Ashfaq R, Ilaria Jr RL. The epidermal growth factor receptor HER2 is not a major therapeutic target in Ewing sarcoma. J Pediatr Hematol Oncol 2003;25:459–66. Bjornsti MA, Houghton PJ. Lost in translation: dysregulation of cap-dependent translation and cancer. Cancer Cell 2004;5:519–23. Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J, et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004;22:2336–47. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet 2003;4:346–58. El-Aneed A. Current strategies in cancer gene therapy. Eur J Pharmacol 2004;498:1–8. Kovar H, Ban J, Pospisilova S. Potentials for RNAi in sarcoma research and therapy: Ewing’s sarcoma as a model. Semin Cancer Biol 2003;13:275–81. Milas M, Yu D, Lang A, Ge T, Feig B, El-Naggar AK, et al. Adenovirus-mediated p53 gene therapy inhibits human sarcoma tumourigenicity. Cancer Gene Ther 2000;7: 422–9. Zhan M, Yu D, Lang A, Li L, Pollock RE. Wild type p53 sensitizes soft tissue sarcoma cells to doxorubicin by downregulating multidrug resistance-1 expression. Cancer 2001;92:1556–66. Fruehauf S, Veldwijk MR, Berlinghoff S, Basara N, Baum C, Flasshove M, et al. Gene therapy for sarcoma. Cells Tissue Organs 2002;172:133–44. Scotlandi K, Maini C, Manara MC, Benini S, Serra M, Cerisano V, et al. Effectiveness of insulin-like growth factor I receptor antisense strategy against Ewing’s sarcoma cells. Cancer Gene Ther 2002;9:296–307. Thallinger C, Wolschek MF, Maierhofer H, Skvara H, Pehamberger H, Monia BP, et al. Mcl-1 is a novel therapeutic target for human sarcoma: synergistic inhibition of

26

190.

191.

192.

193.

194.

195.

196.

197.

198.

199.

200.

201.

202.

203.

204.

S. Mocellin et al. human sarcoma xenotransplants by a combination of mcl-1 antisense oligonucleotides with low-dose cyclophosphamide. Clin Cancer Res 2004;10:4185–91. Shinomiya N, Gao CF, Xie Q, Gustafson M, Waters DJ, Zhang YW, et al. RNA interference reveals that ligandindependent met activity is required for tumour cell signaling and survival. Cancer Res 2004;64:7962–70. Wurl P, Kappler M, Meye A, Bartel F, Kohler T, Lautenschlager C, et al. Co-expression of survivin and TERT and risk of tumour-related death in patients with soft-tissue sarcoma. Lancet 2002;359:943–5. Kappler M, Bache M, Bartel F, Kotzsch M, Panian M, Wurl P, et al. Knockdown of survivin expression by small interfering RNA reduces the clonogenic survival of human sarcoma cell lines independently of p53. Cancer Gene Ther 2004;11:186–93. Lans TE, Ten Hagen TL, Van Horssen R, Wu PC, Van Tiel ST, Libutti SK, et al. Improved antitumour response to isolated limb perfusion with tumour necrosis factor after upregulation of endothelial monocyte-activating polypeptide II in soft tissue sarcoma. Ann Surg Oncol 2002;9: 812–9. de Wilt JH, Bout A, Eggermont AM, van Tiel ST, de Vries TL, ten Hagen TL, et al. Adenovirus-mediated interleukin 3 beta gene transfer by isolated limb perfusion inhibits growth of limb sarcoma in rats. Hum Gene Ther 2001;12:489–502. Pasetto LM, Basso U, Brandes AA. Improved tolerability of chemotherapy in soft tissue sarcomas: old and new strategies. Expert Rev Anticancer Ther 2003;3:167–78. Chen HX, Marshall JL, Ness E, Martin RR, Dvorchik B, Rizvi N, et al. A safety and pharmacokinetic study of a mixedbackbone oligonucleotide (GEM231) targeting the type I protein kinase A by two-hour infusions in patients with refractory solid tumours. Clin Cancer Res 2000;6:1259–66. Galanis E, Hersh EM, Stopeck AT, Gonzalez R, Burch P, Spier C, et al. Immunotherapy of advanced malignancy by direct gene transfer of an interleukin-2 DNA/DMRIE/DOPE lipid complex: phase I/II experience. J Clin Oncol 1999;17:3313–23. Ries S, Korn WM. ONYX-015: mechanisms of action and clinical potential of a replication-selective adenovirus. Br J Cancer 2002;86:5–11. Galanis E, Okuno SH, Nascimento AG, Lewis BD, Lee RA, Oliveira AM, et al. Phase I–II trial of ONYX-015 in combination with MAP chemotherapy in patients with advanced sarcomas. Gene Ther. Lu W, Zheng S, Li XF, Huang JJ, Zheng X, Li Z. Intra-tumour injection of H101, a recombinant adenovirus, in combination with chemotherapy in patients with advanced cancers: a pilot phase II clinical trial. World J Gastroenterol 2004;10:3634–8. Senzer N, Mani S, Rosemurgy A, Nemunaitis J, Cunningham C, Guha C, et al. TNFerade biologic, an adenovector with a radiation-inducible promoter, carrying the human tumour necrosis factor alpha gene: a phase I study in patients with solid tumours. J Clin Oncol 2004;22:592–601. Mundt AJ, Vijayakumar S, Nemunaitis J, Sandler A, Schwartz H, Hanna N, et al. A phase I trial of TNFerade biologic in patients with soft tissue sarcoma in the extremities. Clin Cancer Res 2004;10:5747–53. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004;10: 909–15. Mocellin S, Rossi CR, Nitti D. Cancer vaccine development: on the way to break immune tolerance to malignant cells. Exp Cell Res 2004;299:267–78.

205. Mocellin S, Mandruzzato S, Bronte V, Lise M, Nitti D. Part I: Vaccines for solid tumours. Lancet Oncol 2004;5:681–9. 206. Stebbing J, Bower M, Srivastava P. Kaposi’s sarcoma as a model for cancer immunotherapy. Trends Mol Med 2004;10:187–93. 207. Maki RG. Immunity against soft-tissue sarcomas. Curr Oncol Rep 2003;5:282–7. 208. Sato Y, Nabeta Y, Tsukahara T, Hirohashi Y, Syunsui R, Maeda A, et al. Detection and induction of CTLs specific for SYT-SSX-derived peptides in HLA-A24(+) patients with synovial sarcoma. J Immunol 2002;169:1611–8. 209. Worley BS, van den Broeke LT, Goletz TJ, Pendleton CD, Daschbach EM, Thomas EK, et al. Antigenicity of fusion proteins from sarcoma-associated chromosomal translocations. Cancer Res 2001;61:6868–75. 210. Ludewig B, Ochsenbein AF, Odermatt B, Paulin D, Hengartner H, Zinkernagel RM. Immunotherapy with dendritic cells directed against tumour antigens shared with normal host cells results in severe autoimmune disease. J Exp Med 2000;191:795–804. 211. Mocellin S, Semenzato G, Mandruzzato S, Riccardo Rossi C. Part II: Vaccines for haematological malignant disorders. Lancet Oncol 2004;5:727–37. 212. Alves PM, Faure O, Graff-Dubois S, Gross DA, Cornet S, Chouaib S, et al. EphA2 as target of anticancer immunotherapy: identification of HLA-A*0201-restricted epitopes. Cancer Res 2003;63:8476–80. 213. Lee SY, Obata Y, Yoshida M, Stockert E, Williamson B, Jungbluth AA, et al. Immunomic analysis of human sarcoma. Proc Natl Acad Sci USA 2003;100:2651–6. 214. Dalerba P, Frascella E, Macino B, Mandruzzato S, Zambon A, Rosolen A, et al. MAGE, BAGE and GAGE gene expression in human rhabdomyosarcomas. Int J Cancer 2001;93:85–90. 215. Daftarian P, Song GY, Ali S, Faynsod M, Longmate J, Diamond DJ, et al. Two distinct pathways of immuno-modulation improve potency of p53 immunization in rejecting established tumours. Cancer Res 2004;64:5407–14. 216. Espenschied J, Lamont J, Longmate J, Pendas S, Wang Z, Diamond DJ, et al. CTLA-4 blockade enhances the therapeutic effect of an attenuated poxvirus vaccine targeting p53 in an established murine tumour model. J Immunol 2003;170:3401–7. 217. Kusmartsev S, Cheng F, Yu B, Nefedova Y, Sotomayor E, Lush R, et al. All-trans-retinoic acid eliminates immature myeloid cells from tumour-bearing mice and improves the effect of vaccination. Cancer Res 2003;63:4441–9. 218. Chapman PB, Morrisey D, Panageas KS, Williams L, Lewis RJ, Israel RJ, et al. Vaccination with a bivalent G(M2) and G(D2) ganglioside conjugate vaccine: a trial comparing doses of G(D2)-keyhole limpet hemocyanin. Clin Cancer Res 2000;6:4658–62. 219. MahviDM, Shi FS, Yang NS, Weber S, Hank J, Albertini M, et al. Immunization by particle-mediated transfer of the granulocyte-macrophage colony-stimulating factor gene into autologous tumour cells in melanoma or sarcoma patients: report of a phase I/IB study. Hum Gene Ther 2002;13:1711–21. 220. Dillman RO, Wiemann M, Nayak SK, DeLeon C, Hood K, DePriest C. Interferon-gamma or granulocyte-macrophage colony-stimulating factor administered as adjuvants with a vaccine of irradiated autologous tumour cells from shortterm cell line cultures: a randomized phase 2 trial of the cancer biotherapy research group. J Immunother 2003;26: 367–73. 221. Dillman R, Barth N, Selvan S, Beutel L, de Leon C, DePriest C, et al. Phase I/II trial of autologous tumour cell linederived vaccines for recurrent or metastatic sarcomas. Cancer Biother Radiopharm 2004;19:581–8.

Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches 222. Matsuzaki A, Suminoe A, Hattori H, Hoshina T, Hara T. Immunotherapy with autologous dendritic cells and tumour-specific synthetic peptides for synovial sarcoma. J Pediatr Hematol Oncol 2002;24:220–3. 223. Dagher R, Long LM, Read EJ, Leitman SF, Carter CS, Tsokos M, et al. Pilot trial of tumour-specific peptide vaccination and continuous infusion interleukin-2 in patients with recurrent Ewing sarcoma and alveolar rhabdomyosarcoma: an inter-institute NIH study. Med Pediatr Oncol 2002;38: 158–64. 224. Kawaguchi S, Wada T, Ida K, Sato Y, Nagoya S, Tsukahara T, et al. Phase I vaccination trial of SYT-SSX junction peptide in patients with disseminated synovial sarcoma. J Transl Med 2005;3:1. 225. Takahashi A, Higashino F, Aoyagi M, Yoshida K, Itoh M, Kyo S, et al. EWS/ETS fusions activate telomerase in Ewing’s tumours. Cancer Res 2003;63:8338–44. 226. Fanucchi M. Update on the management of connective tissue malignancies. Semin Oncol 2004;31:16–9. 227. Weitz J, Antonescu CR, Brennan MF. Localized extremity soft tissue sarcoma: improved knowledge with unchanged survival over time. J Clin Oncol 2003;21:2719–25. 228. Mocellin S, Provenzano M, Rossi CR, Pilati P, Nitti D, Lise M. DNA array-based gene profiling: from surgical specimen to

229.

230.

231.

232.

233.

234.

27

the molecular portrait of cancer. Ann Surg 2005;241: 16–26. Mocellin S, Riccardo Rossi C, Traldi P, Nitti D, Lise M. Molecular oncology in the post-genomic era: the challenge of proteomics. Trends Mol Med 2004;10:24–32. Eilber FC, Eilber FR, Eckardt J, Rosen G, Riedel E, Maki RG, et al. The impact of chemotherapy on the survival of patients with high-grade primary extremity liposarcoma. Ann Surg 2004;240:686–95. Discussion 95–7. Stojadinovic A, Leung DH, Allen P, Lewis JJ, Jaques DP, Brennan MF. Primary adult soft tissue sarcoma: timedependent influence of prognostic variables. J Clin Oncol 2002;20:4344–52. Nielsen TO, West RB, Linn SC, Alter O, Knowling MA, O’Connell JX, et al. Molecular characterisation of soft tissue tumours: a gene expression study. Lancet 2002;359: 1301–7. Lee YF, John M, Falconer A, Edwards S, Clark J, Flohr P, et al. A gene expression signature associated with metastatic outcome in human leiomyosarcomas. Cancer Res 2004;64:7201–4. Oliveira AM, Fletcher CD. Molecular prognostication for soft tissue sarcomas: are we ready yet? J Clin Oncol 2004;22:4031–4.