1
|
Gennaro N, van der Loo I, Reijers SJM, van Boven H, Snaebjornsson P, Bekers EM, Bodalal Z, Trebeschi S, Schrage YM, van der Graaf WTA, van Houdt WJ, Haas RLM, Velichko YS, Beets-Tan RGH, Bruining A. Heterogeneity in response to neoadjuvant radiotherapy between soft tissue sarcoma histotypes: associations between radiology and pathology findings. Eur Radiol 2025; 35:1337-1350. [PMID: 39699680 DOI: 10.1007/s00330-024-11258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVE To investigate imaging biomarkers of tumour response by describing changes in imaging and pathology findings after neoadjuvant radiotherapy (nRT) and exploring their correlations. MATERIALS AND METHODS Tumour diameter, volume, and tumour-to-muscle signal intensity (SI) ratio were collected before and after radiotherapy in a cohort of 107 patients with intermediate/high-grade STS and were correlated with post-radiotherapy pathology findings (percentage of necrosis, viable cells, and fibrosis) using Spearman Rank test. Pathological complete response (pCR) was defined as no residual viable cells present, whereas the presence of < 10% viable cells was defined as near-complete pathologic response (near-pCR). RESULTS Median amount of necrosis, viable cells, and fibrosis after nRT were 10%, 30%, and 25%, respectively. 7% of patients achieved pCR and 22% near-pCR. No changes in tumour volume were found except for subtypes myxoid liposarcoma (mLPS) -Δ54.47%, undifferentiated pleomorphic sarcoma (UPS) +Δ24.22% and dedifferentiated liposarcoma (dLPS) +Δ35.91%. The median change of tumour-to-muscle SI ratio was -19.7% for the entire population, whereas it was -19.55% and -36.26% for UPS and mLPS, respectively. Correlations (positive and negative) were found between change in volume and the presence of necrosis or fibrosis (rs = 0.44; rs = -0.44), as well as between tumour-to-muscle SI ratio and viable cells (rs = 0.33) or fibrosis (rs = -0.28). CONCLUSION STS displays extensive heterogeneity in response patterns after nRT. In some subgroups, particularly UPS and mLPS, tumour size changes or tumour-to-muscle SI ratio are significantly linked with the percentage of viable cells, fibrosis, or necrosis. KEY POINTS Question How do primary soft tissue sarcomas (STS) respond to neoadjuvant therapy, and what correlations exist between pathological findings and imaging characteristics in assessing treatment response? Findings mLPS shrank post-nRT; undifferentiated pleomorphic and dLPSs enlarged. Volume increase correlated with higher necrosis and lower fibrosis; tumour-to-muscle intensity ratio correlated with viable cells. Clinical relevance These findings emphasise the extensive heterogeneity in STS response to nRT across different subtypes. Preoperative correlations between tumour volume and SI changes with necrosis, fibrosis, and viable cells can aid in more precise treatment assessment and prognostication.
Collapse
Affiliation(s)
- Nicolò Gennaro
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Iris van der Loo
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, University of Maastricht, Maastricht, The Netherlands
| | - Sophie J M Reijers
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hester van Boven
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elise M Bekers
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Zuhir Bodalal
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, University of Maastricht, Maastricht, The Netherlands
| | - Stefano Trebeschi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, University of Maastricht, Maastricht, The Netherlands
| | - Yvonne M Schrage
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Winan J van Houdt
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rick L M Haas
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiation Oncology, Leiden University, Leiden, The Netherlands
| | - Yury S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW-Research Institute for Oncology & Reproduction, University of Maastricht, Maastricht, The Netherlands
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Annemarie Bruining
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Valenzuela RF, Duran Sierra EDJ, Canjirathinkal MA, Amini B, Hwang KP, Ma J, Torres KE, Stafford RJ, Wang WL, Benjamin RS, Bishop AJ, Madewell JE, Murphy WA, Costelloe CM. Novel Use and Value of Contrast-Enhanced Susceptibility-Weighted Imaging Morphologic and Radiomic Features in Predicting Extremity Soft Tissue Undifferentiated Pleomorphic Sarcoma Treatment Response. JCO Clin Cancer Inform 2025; 9:e2400042. [PMID: 39841956 DOI: 10.1200/cci.24.00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025] Open
Abstract
PURPOSE Undifferentiated pleomorphic sarcomas (UPSs) demonstrate therapy-induced hemosiderin deposition, granulation tissue formation, fibrosis, and calcification. We aimed to determine the treatment-assessment value of morphologic tumoral hemorrhage patterns and first- and high-order radiomic features extracted from contrast-enhanced susceptibility-weighted imaging (CE-SWI). MATERIALS AND METHODS This retrospective institutional review board-authorized study included 33 patients with extremity UPS with magnetic resonance imaging and resection performed from February 2021 to May 2023. Volumetric tumor segmentation was obtained at baseline, postsystemic chemotherapy (PC), and postradiation therapy (PRT). The pathology-assessed treatment effect (PATE) in surgical specimens separated patients into responders (R; ≥90%, n = 16), partial responders (PR; 89%-31%, n = 10), and nonresponders (NR; ≤30%, n = 7). RECIST, WHO, and volume were assessed for all time points. CE-SWI T2* morphologic patterns and 107 radiomic features were analyzed. RESULTS A Complete-Ring (CR) pattern was observed in PRT in 71.4% of R (P = 7.71 × 10-6), an Incomplete-Ring pattern in 33.3% of PR (P = .2751), and a Globular pattern in 50% of NR (P = .1562). The first-order radiomic analysis from the CE-SWI intensity histogram outlined the values of the 10th and 90th percentiles and their skewness. R showed a 280% increase in 10th percentile voxels (P = .061) and a 241% increase in skewness (P = .0449) at PC. PR/NR showed a 690% increase in the 90th percentile voxels (P = .03) at PC. Multiple high-order radiomic texture features observed at PRT discriminated better R versus PR/NR than the first-order features. CONCLUSION CE-SWI morphologic patterns strongly correlate with PATE. The CR morphology pattern was the most frequent in R and had the highest statistical association predicting response at PRT, easily recognized by a radiologist not requiring postprocessing software. It can potentially outperform size-based metrics, such as RECIST. The first- and high-order radiomic analysis found several features separating R versus PR/NR.
Collapse
Affiliation(s)
| | | | | | - Behrang Amini
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ken-Pin Hwang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jingfei Ma
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keila E Torres
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Wei-Lien Wang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | |
Collapse
|
3
|
Al Shihabi A, Tebon PJ, Nguyen HTL, Chantharasamee J, Sartini S, Davarifar A, Jensen AY, Diaz-Infante M, Cox H, Gonzalez AE, Norris S, Sperry J, Nakashima J, Tavanaie N, Winata H, Fitz-Gibbon ST, Yamaguchi TN, Jeong JH, Dry S, Singh AS, Chmielowski B, Crompton JG, Kalbasi AK, Eilber FC, Hornicek F, Bernthal NM, Nelson SD, Boutros PC, Federman NC, Yanagawa J, Soragni A. The landscape of drug sensitivity and resistance in sarcoma. Cell Stem Cell 2024; 31:1524-1542.e4. [PMID: 39305899 DOI: 10.1016/j.stem.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 06/14/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024]
Abstract
Sarcomas are rare malignancies with over 100 distinct histological subtypes. Their rarity and heterogeneity pose significant challenges to identifying effective therapies, and approved regimens show varied responses. Novel, personalized approaches to therapy are needed to improve patient outcomes. Patient-derived tumor organoids (PDTOs) model tumor behavior across an array of malignancies. We leverage PDTOs to characterize the landscape of drug resistance and sensitivity in sarcoma, collecting 194 specimens from 126 patients spanning 24 distinct sarcoma subtypes. Our high-throughput organoid screening pipeline tested single agents and combinations, with results available within a week from surgery. Drug sensitivity correlated with clinical features such as tumor subtype, treatment history, and disease trajectory. PDTO screening can facilitate optimal drug selection and mirror patient outcomes in sarcoma. We could identify at least one FDA-approved or NCCN-recommended effective regimen for 59% of the specimens, demonstrating the potential of our pipeline to provide actionable treatment information.
Collapse
Affiliation(s)
- Ahmad Al Shihabi
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peyton J Tebon
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Huyen Thi Lam Nguyen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jomjit Chantharasamee
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sara Sartini
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ardalan Davarifar
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexandra Y Jensen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Miranda Diaz-Infante
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hannah Cox
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Summer Norris
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Nasrin Tavanaie
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Helena Winata
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sorel T Fitz-Gibbon
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Takafumi N Yamaguchi
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jae H Jeong
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarah Dry
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Arun S Singh
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bartosz Chmielowski
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joseph G Crompton
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Division of Surgical Oncology David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anusha K Kalbasi
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fritz C Eilber
- Division of Surgical Oncology David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Francis Hornicek
- Department of Orthopedic Surgery, University of Miami, Miami, FL, USA
| | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Scott D Nelson
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Noah C Federman
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jane Yanagawa
- Department of Surgery, Division of Thoracic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Hanslik N, Bourgier C, Thezenas S, Carrère S, Firmin N, Riou O, Azria D, Llacer-Moscardo C. [Predictive factors assessment of pathological response to neoadjuvant radiotherapy of soft tissue sarcomas]. Cancer Radiother 2023; 27:689-697. [PMID: 37813717 DOI: 10.1016/j.canrad.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 10/11/2023]
Abstract
PURPOSE Conserving surgery combined with radiotherapy in presence of local recurrence risk factors is standard treatment of soft tissue sarcomas, a group of rare and heterogeneous tumours. Radiotherapy is performed before or after surgery. In neoadjuvant setting, late radiation-induced toxicity is reduced and pathological response to radiotherapy could be achieved. A complete pathological response to radiotherapy has recently been shown to predict better survival. Our study aims at identifying predictive factors of pathological response to neoadjuvant radiotherapy (clinical, radiological or histological) of soft tissue sarcomas. PATIENTS AND METHODS Clinical, imaging (MRI: perilesional oedema, necrosis, tumour heterogeneity, vasculonervous relationships) and pathological (pathological subtype, tumour grade, anticipated/obtained resection quality) data were retrospectively collected. Tumour response (imaging and pathological), patient outcome, acute and late radiation-induced toxicity, predictive factors of pathological response to neoadjuvant radiotherapy were studied. The 2-test or exact-Fisher test (qualitative variables) and by Student's t-test or Kruskal-Wallis test (quantitative variables) were used for statistical analysis. RESULTS From April 2017 to April 2021, neoadjuvant radiotherapy (50Gy in 25 fractions) followed by surgical excision was performed to 36 consecutive patients with liposarcomas (n=17/36), or undifferentiated sarcomas (n=8/36). MRI response was complete in 1 patient, partial in 9 patients (n=9/36, 25%), stable in 21 patients (n=21/36, 58%) or in progression in 5 patients (n=5/36, 14%). Pathological response was observed in 22 patients (61%). No grade 3-4 acute radiation-induced toxicity was observed. Regarding late toxicity, 28% of patients had grade 1-2 oedema (n=10/36), 39% had a grade 1 fibrosis (n=14/36), and 30% grade 1 pain (n=11/36). No predictive factors of response to radiotherapy was statistically significant. CONCLUSIONS Neoadjuvant radiotherapy is well-tolerated. No clinical, radiological or pathological predictive factors was identified for radiotherapy tumour response.
Collapse
Affiliation(s)
- N Hanslik
- Fédération universitaire d'oncologie radiothérapie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France
| | - C Bourgier
- Fédération universitaire d'oncologie radiothérapie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France; IRCM, institut de recherche en cancérologie de Montpellier, Inserm U1194, université de Montpellier, avenue des Apothicaires, 34298 Montpellier cedex 05, France
| | - S Thezenas
- Unité de biostatistiques, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France
| | - S Carrère
- Service de chirurgie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France
| | - N Firmin
- Département d'oncologie, ICM, institut régional du Cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France
| | - O Riou
- Fédération universitaire d'oncologie radiothérapie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France
| | - D Azria
- Fédération universitaire d'oncologie radiothérapie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France; IRCM, institut de recherche en cancérologie de Montpellier, Inserm U1194, université de Montpellier, avenue des Apothicaires, 34298 Montpellier cedex 05, France
| | - C Llacer-Moscardo
- Fédération universitaire d'oncologie radiothérapie, ICM, institut régional du cancer Montpellier, rue Croix-Verte, 34298 Montpellier cedex 05, France.
| |
Collapse
|
5
|
Predicting Soft Tissue Sarcoma Response to Neoadjuvant Chemotherapy Using an MRI-Based Delta-Radiomics Approach. Mol Imaging Biol 2023:10.1007/s11307-023-01803-y. [PMID: 36695966 DOI: 10.1007/s11307-023-01803-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVES To evaluate the performance of machine learning-augmented MRI-based radiomics models for predicting response to neoadjuvant chemotherapy (NAC) in soft tissue sarcomas. METHODS Forty-four subjects were identified retrospectively from patients who received NAC at our institution for pathologically proven soft tissue sarcomas. Only subjects who had both a baseline MRI prior to initiating chemotherapy and a post-treatment scan at least 2 months after initiating chemotherapy and prior to surgical resection were included. 3D ROIs were used to delineate whole-tumor volumes on pre- and post-treatment scans, from which 1708 radiomics features were extracted. Delta-radiomics features were calculated by subtraction of baseline from post-treatment values and used to distinguish treatment response through univariate analyses as well as machine learning-augmented radiomics analyses. RESULTS Though only 4.74% of variables overall reached significance at p ≤ 0.05 in univariate analyses, Laws Texture Energy (LTE)-derived metrics represented 46.04% of all such features reaching statistical significance. ROC analyses similarly failed to predict NAC response, with AUCs of 0.40 (95% CI 0.22-0.58) and 0.44 (95% CI 0.26-0.62) for RF and AdaBoost, respectively. CONCLUSION Overall, while our result was not able to separate NAC responders from non-responders, our analyses did identify a subset of LTE-derived metrics that show promise for further investigations. Future studies will likely benefit from larger sample size constructions so as to avoid the need for data filtering and feature selection techniques, which have the potential to significantly bias the machine learning procedures.
Collapse
|
6
|
Castillo-Flores S, Gonzalez MR, Bryce-Alberti M, de Souza F, Subhawong TK, Kuker R, Pretell-Mazzini J. PET-CT in the Evaluation of Neoadjuvant/Adjuvant Treatment Response of Soft-tissue Sarcomas: A Comprehensive Review of the Literature. JBJS Rev 2022; 10:01874474-202212000-00003. [PMID: 36639875 DOI: 10.2106/jbjs.rvw.22.00131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
➢ In soft-tissue sarcomas (STSs), the use of positron emission tomography-computed tomography (PET-CT) through a standardized uptake value reduction rate correlates well with histopathological response to neoadjuvant treatment and survival. ➢ PET-CT has shown a better sensitivity to diagnose systemic involvement compared with magnetic resonance imaging and CT; therefore, it has an important role in detecting recurrent systemic disease. However, delaying the use of PET-CT scan, to differentiate tumor recurrence from benign fluorodeoxyglucose uptake changes after surgical treatment and radiotherapy, is essential. ➢ PET-CT limitations such as difficult differentiation between benign inflammatory and malignant processes, inefficient discrimination between benign soft-tissue tumors and STSs, and low sensitivity when evaluating small pulmonary metastases must be of special consideration.
Collapse
Affiliation(s)
- Samy Castillo-Flores
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Marcos R Gonzalez
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mayte Bryce-Alberti
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Felipe de Souza
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Ty K Subhawong
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Russ Kuker
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Pretell-Mazzini
- Division of Orthopedic Oncology, Miami Cancer Institute, Baptist Health System South Florida, Plantation, Florida
| |
Collapse
|
7
|
Boxberg M, Langer R, Woertler K, Knebel C, Rechl H, von Eisenhart-Rothe R, Weichert W, Combs SE, Hadjamu M, Röper B, Specht K. Neoadjuvant Radiation in High-Grade Soft-Tissue Sarcomas: Histopathologic Features and Response Evaluation. Am J Surg Pathol 2022; 46:1060-1070. [PMID: 35687332 DOI: 10.1097/pas.0000000000001922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this study, we sought to determine the prognostic value of both the European Organization for Research and Treatment of Cancer-Soft Tissue and Bone Sarcoma Group (EORTC-STBSG) score and the histologic parameters viable tumor, coagulative necrosis, hyalinization/fibrosis, and infarction in patients (n=64) with localized, nonmetastatic high-grade soft-tissue sarcomas after preoperative radiomonotherapy. A standardized macroscopic workup for pretreated surgical specimen including evaluation of a whole section of high-grade soft tissue sarcomas in the largest diameter, was used. Association with overall survival and disease-free survival was assessed. Limb salvage could be accomplished in 98.4% of patients. Overall, 90.6% tumors had negative resection margins. The median postoperative tumor diameter was 9 cm. Undifferentiated pleomorphic sarcoma (42.2%) and myxofibrosarcoma (17.2%) were the most common diagnoses. In all, 9.4% of patients had local recurrence despite clear resection margins, and 50% had distant metastases. Morphologic mapping suggests an overall heterogenous intratumoral response to radiotherapy, with significant differences among histologic subtypes. Complete regression (0% vital tumor cells) was not seen. Categorizing the results according to the proposed EORTC-STBSG 5-tier response score, <1% viable tumor cells were seen in 3.1%, ≥1% to <10% viable tumor cells in 20.4%, ≥10% to <50% viable tumor cells in 35.9% and ≥50% viable tumor cells in 40.6% of cases. Mean values for viable tumor cells were 40% (range: 1% to 100%), coagulative necrosis 5% (0% to 60%), hyalinization/fibrosis 25% (0% to 90%) and infarction 15% (0% to 79%). Hyalinization/fibrosis was a significant independent prognostic factor for overall survival (hazard ratio=4.4; P =0.047), while the other histologic parameters including the EORTC-STBSG score were not prognostic.
Collapse
Affiliation(s)
| | - Rupert Langer
- Institute of Clinical Pathology and Molecular Pathology, Johannes Kepler University and Kepler University Hospital Linz, Linz, Austria
| | | | | | | | | | | | - Stephanie E Combs
- Radiation Oncology, rechts der Isar Hospital, Technical University of Munich
- German Cancer Consortium (DKTK), Partner Site Munich
- Institute of Radiation Medicine (IRM), Helmholtz Munich, Ober-schleißheim, Germany
| | - Miriam Hadjamu
- Radiation Oncology, rechts der Isar Hospital, Technical University of Munich
- Ambulatory Health Care Centre "Radiotherapy" Harlaching/Bogenhausen, Munich
| | - Barbara Röper
- Radiation Oncology, rechts der Isar Hospital, Technical University of Munich
- Ambulatory Health Care Centre "Radiotherapy" Harlaching/Bogenhausen, Munich
| | - Katja Specht
- Institute of Pathology, Technical University of Munich
| |
Collapse
|
8
|
Soft Tissue Sarcomas: The Role of Quantitative MRI in Treatment Response Evaluation. Acad Radiol 2022; 29:1065-1084. [PMID: 34548230 DOI: 10.1016/j.acra.2021.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although curative surgery remains the cornerstone of the therapeutic strategy in patients with soft tissue sarcomas (STS), neoadjuvant radiotherapy and chemotherapy (NART and NACT, respectively) are increasingly used to improve operability, surgical margins and patient outcome. The best imaging modality for locoregional assessment of STS is MRI but these tumors are mostly evaluated in a qualitative manner. OBJECTIVE After an overview of the current standard of care regarding treatment for patients with locally advanced STS, this review aims to summarize the principles and limitations of (i) the current methods used to evaluate response to neoadjuvant treatment in clinical practice and clinical trials in STS (RECIST 1.1 and modified Choi criteria), (ii) quantitative MRI sequences (i.e., diffusion weighted imaging and dynamic contrast enhanced MRI), and (iii) texture analyses and (delta-) radiomics.
Collapse
|
9
|
Collier AB, Krailo MD, Dang HM, DuBois SG, Hawkins DS, Bernstein ML, Bomgaars LR, Reed DR, Gorlick RG, Janeway KA. Outcome of patients with relapsed or progressive Ewing sarcoma enrolled on cooperative group phase 2 clinical trials: A report from the Children's Oncology Group. Pediatr Blood Cancer 2021; 68:e29333. [PMID: 34496122 PMCID: PMC8541905 DOI: 10.1002/pbc.29333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/20/2021] [Indexed: 11/10/2022]
Abstract
SevenChildren's Oncology Group phase 2 trials for patients with relapsed/progressive solid tumors were analyzed to estimate the event-free survival (EFS) for relapsed/progressive Ewing sarcoma. One hundred twenty-eight Ewing sarcoma patients were enrolled and 124 events occurred. The 6-month EFS was 12.7%, demonstrating the poor outcome of these patients. Only docetaxel achieved its protocol-specified radiographic response rate for activity; however, the EFS for docetaxel was similar to other agents, indicating that a higher radiographic response rate may not translate into superior disease control. This EFS benchmark could be utilized as an additional endpoint in trials for recurrent Ewing sarcoma.
Collapse
Affiliation(s)
- Anderson B. Collier
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS
| | - Mark D. Krailo
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Ha M. Dang
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Steven G. DuBois
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School; Boston, MA
| | | | | | - Lisa R. Bomgaars
- Baylor College of Medicine/Dan L Duncan Comprehensive Cancer Center; Houston, TX
| | - Damon R. Reed
- Johns Hopkins All Children’s Hospital; St Petersburg, FL
| | | | - Katherine A. Janeway
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School; Boston, MA
| |
Collapse
|
10
|
Smrke A, Thway K, H Huang P, Jones RL, Hayes AJ. Solitary fibrous tumor: molecular hallmarks and treatment for a rare sarcoma. Future Oncol 2021; 17:3627-3636. [PMID: 34409859 DOI: 10.2217/fon-2021-0030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Solitary fibrous tumor (SFT) is a rare soft tissue sarcoma subtype which mainly affects adults in the fifth and sixth decades of life. Originally part of a spectrum of tumors called hemangiopericytomas, classification has been refined such that SFTs now represent a distinct subtype. The identification of NAB2-STAT6 fusion in virtually all SFTs has further aided to define this rare subgroup. SFTs have a spectrum of behavior from benign to malignant, with evidence suggesting risk of metastases related to age at diagnosis, extent of necrosis, mitotic rate and tumor size. The standard treatment for localized disease is surgical excision with or without radiotherapy. Retrospective and prospective evidence suggests antiangiogenic treatment is effective for unresectable disease. Further translational work is required to understand the biology driving the differential behavior and identify more effective treatments for patients with metastatic disease.
Collapse
Affiliation(s)
- Alannah Smrke
- Sarcoma Unit, Royal Marsden Hospital, 203 Fulham Road, London, SW3 6JJ, UK
| | - Khin Thway
- Sarcoma Unit, Royal Marsden Hospital, 203 Fulham Road, London, SW3 6JJ, UK.,The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Paul H Huang
- The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Robin L Jones
- Sarcoma Unit, Royal Marsden Hospital, 203 Fulham Road, London, SW3 6JJ, UK.,The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Andrew J Hayes
- Sarcoma Unit, Royal Marsden Hospital, 203 Fulham Road, London, SW3 6JJ, UK.,The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| |
Collapse
|
11
|
Gennaro N, Reijers S, Bruining A, Messiou C, Haas R, Colombo P, Bodalal Z, Beets-Tan R, van Houdt W, van der Graaf WTA. Imaging response evaluation after neoadjuvant treatment in soft tissue sarcomas: Where do we stand? Crit Rev Oncol Hematol 2021; 160:103309. [PMID: 33757836 DOI: 10.1016/j.critrevonc.2021.103309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Soft tissue sarcomas (STS) represent a broad family of rare tumours for which surgery with radiotherapy represents first-line treatment. Recently, neoadjuvant chemo-radiotherapy has been increasingly used in high-risk patients in an effort to reduce surgical morbidity and improve clinical outcomes. An adequate understanding of the efficacy of neoadjuvant therapies would optimise patient care, allowing a tailored approach. Although response evaluation criteria in solid tumours (RECIST) is the most common imaging method to assess tumour response, Choi criteria and functional and molecular imaging (DWI, DCE-MRI and 18F-FDG-PET) seem to outperform it in the discrimination between responders and non-responders. Moreover, the radiologic-pathology correlation of treatment-related changes remains poorly understood. In this review, we provide an overview of the imaging assessment of tumour response in STS undergoing neoadjuvant treatment, including conventional imaging (CT, MRI, PET) and advanced imaging analysis. Future directions will be presented to shed light on potential advances in pre-surgical imaging assessments that have clinical implications for sarcoma patients.
Collapse
Affiliation(s)
- Nicolò Gennaro
- Humanitas Research and Cancer Center, Dept. of Radiology, Rozzano, Italy; Humanitas University, Dept. of Biomedical Sciences, Pieve Emanuele, Italy; The Netherlands Cancer Institute, Dept. of Radiology, Amsterdam, the Netherlands.
| | - Sophie Reijers
- The Netherlands Cancer Institute, Dept. of Surgical Oncology, Amsterdam, the Netherlands
| | - Annemarie Bruining
- The Netherlands Cancer Institute, Dept. of Radiology, Amsterdam, the Netherlands
| | - Christina Messiou
- The Royal Marsden NHS Foundation Trust, Dept. Of Radiology Sarcoma Unit, Sutton, United Kingdom; The Institute of Cancer Research, Sutton, United Kingdom
| | - Rick Haas
- The Netherlands Cancer Institute, Dept. of Radiation Oncology, Amsterdam, the Netherlands; Leiden University Medical Center, Dept. of Radiation Oncology, the Netherlands
| | | | - Zuhir Bodalal
- The Netherlands Cancer Institute, Dept. of Radiology, Amsterdam, the Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Regina Beets-Tan
- The Netherlands Cancer Institute, Dept. of Radiology, Amsterdam, the Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands; Danish Colorectal Cancer Center South, Vejle University Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - Winan van Houdt
- The Netherlands Cancer Institute, Dept. of Surgical Oncology, Amsterdam, the Netherlands
| | - Winette T A van der Graaf
- The Netherlands Cancer Institute, Dept. of Medical Oncology, Amsterdam, the Netherlands; Erasmus MC Cancer Institute, Dept. of Medical Oncology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Schlicht SM. Nuclear Medicine and Molecular Imaging Techniques. Sarcoma 2021. [DOI: 10.1007/978-981-15-9414-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
13
|
Valenzuela RF, Kundra V, Madewell JE, Costelloe CM. Advanced Imaging in Musculoskeletal Oncology: Moving Away From RECIST and Embracing Advanced Bone and Soft Tissue Tumor Imaging (ABASTI) - Part I - Tumor Response Criteria and Established Functional Imaging Techniques. Semin Ultrasound CT MR 2020; 42:201-214. [PMID: 33814106 DOI: 10.1053/j.sult.2020.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
According to the Revised Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, the majority of bone metastases are considered to be nonmeasurable disease. Traditional response criteria rely on physical measurements. New criteria would be valuable if they incorporated newly developed imaging features in order to provide a more comprehensive assessment of oncological status. Advanced magnetic resonance imaging (MRI) sequences such as diffusion-weighted imaging (DWI) and perfusion-weighted imaging (PWI) with dynamic contrast-enhanced (DCE) perfusion imaging are reviewed in the context of the initial and post-therapeutic assessment of musculoskeletal tumors. Particular attention is directed to the pseudoprogression phenomenon in which a successfully treated tumor enlarges from the pretherapeutic baseline, followed by regression without a change in therapy.
Collapse
Affiliation(s)
- Raul Fernando Valenzuela
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas.
| | - Vikas Kundra
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| | - John E Madewell
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| | - Colleen M Costelloe
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| |
Collapse
|
14
|
Martin-Tellez KS, van Houdt WJ, van Coevorden F, Colombo C, Fiore M. Isolated limb perfusion for soft tissue sarcoma: Current practices and future directions. A survey of experts and a review of literature. Cancer Treat Rev 2020; 88:102058. [PMID: 32619864 DOI: 10.1016/j.ctrv.2020.102058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
Soft tissue sarcomas constitute 1% of adult malignant tumors. They are a heterogeneous group of more than 50 different histologic types. Isolated limb perfusion is an established treatment strategy for locally advanced sarcomas. Since its adoption for sarcomas in 1992, after the addition of TNFα, few modifications have been done and although indications for the procedure are essentially the same across centers, technical details vary widely. The procedures mainly involves a 60 min perfusion with melphalan and TNFα under mild hyperthermia, achieving a limb preservation rate of 72-96%; with an overall response rates from 72 to 82.5% and an acceptable toxicity according to the Wieberdink scale. The local failure rate is 27% after a median follow up of 14-31 months compared to 40% of distant recurrences after a follow up of 12-22 months. Currently there is no consensus regarding the benefit of ILP per histotype, and the value of addition of radiotherapy or systemic treatment. Further developments towards individualized treatments will provide a better understanding of the population that can derive maximum benefit of ILP with the least morbidity.
Collapse
Affiliation(s)
- K S Martin-Tellez
- Fellow of the European School of Soft Tissue Sarcoma, Department of Surgical Oncology, The American British Cowdray Medical Center ABC, Mexico city, Mexico.
| | - W J van Houdt
- Sarcoma Unit, Department of Surgical Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - F van Coevorden
- Sarcoma Unit, Department of Surgical Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - C Colombo
- Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - M Fiore
- Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
15
|
Quantitative magnetic resonance imaging (q-MRI) for the assessment of soft-tissue sarcoma treatment response: a narrative case review of technique development. Clin Imaging 2020; 63:83-93. [PMID: 32163847 DOI: 10.1016/j.clinimag.2020.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/18/2020] [Accepted: 02/25/2020] [Indexed: 11/20/2022]
Abstract
Soft-tissue sarcomas are a heterogeneous class of tumors that exhibit varying degrees of cellularity and cystic degeneration in response to neoadjuvant chemotherapy. This creates unique challenges in the radiographic assessment of treatment response when relying on conventional markers such as tumor diameter (RECIST criteria). In this case series, we provide a narrative discussion of technique development for whole tumor volume quantitative magnetic resonance imaging (q-MRI), highlighting cases from a small pilot study of 8 patients (9 tumors) pre- and post-neoadjuvant chemotherapy. One of the methods of q-MRI analysis (the "constant-cutoff" technique) was able to predict responders versus non-responders based on percent necrosis and viable tumor volume calculations (p = 0.05), respectively. Our results suggest that q-MRI of whole tumor volume contrast enhancement may have a role in tumor response assessment, although further validation is needed.
Collapse
|
16
|
Younis JA, Al Antably IM, Zamzam M, Salem HT, Zaki EM, Hassanian OA. Role of 18F-fluorodeoxyglucose positron emission tomography/computed tomography and magnetic resonance imaging in prediction of response to neoadjuvant chemotherapy in pediatric osteosarcoma. World J Nucl Med 2019; 18:378-388. [PMID: 31933554 PMCID: PMC6945349 DOI: 10.4103/wjnm.wjnm_52_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/12/2018] [Indexed: 11/04/2022] Open
Abstract
The aim of our study was to evaluate the role of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT) and magnetic resonance imaging (MRI) in prediction of response to neoadjuvant chemotherapy (NAC) in pediatric osteosarcoma (OS) patients compared to percentage of tumor necrosis after surgical excision of the tumor. Forty-six pediatric OS patients treated with neoadjuvant chemotherapy and surgery were underwent PET/CT and MRI before, after 3 cycles, and after the completion of neoadjuvant chemotherapy. Imaging parameters include maximum standardized uptake value (SUVmax1, 2, and 3), tumor liver ratio (TLR 1, 2, and 3), and MRI tumor volume (MRTV 1, 2, and 3) at initial assessment before starting NAC, after finishing three cycles and after finishing 6 cycles before tumor excision, respectively. Cutoff values of the PET/CT and MRI parameters were determined using receiver operating characteristic (ROC) curve analysis and percentage of tumor necrosis of postsurgical specimen. Fourteen patients were good responders (30.4%), with more than 90% tumor necrosis, while 31 patients were poor responders (67.4%). The results of one patient were missed. We noticed that higher sensitivity for detecting poor responders was detected by SUVmax3/1, TLR3/1, and MRTV2/1 ratio cutoff values, while higher specificity was detected by TRL2 and SUVmax3 cutoff values. ROC curve analysis of MRTV2/1 and MRTV3/1 ratio was fair in predicting poor responders. PET/CT parameters are capable of predicting histological response to NAC in OS patients with overall sensitivity and specificity higher than MRI parameters.
Collapse
Affiliation(s)
- Jehan Ahmed Younis
- Department of Oncology and Nuclear Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Manal Zamzam
- Department of Pediatric Oncology, National Cancer Institute, Children Cancer Hospital, Cairo, Egypt
| | - Hala Taha Salem
- Department of Pathology, National Cancer Institute, Children Cancer Hospital, Cairo, Egypt
| | - Eman Mohammed Zaki
- Department of Radiology, National Cancer Institute, Children Cancer Hospital, Cairo, Egypt
| | - Omneya Ahmed Hassanian
- Department of Statistics, National Cancer Institute, Children Cancer Hospital, Cairo, Egypt
| |
Collapse
|
17
|
Castello A, Lopci E. Response assessment of bone metastatic disease: seeing the forest for the trees RECIST, PERCIST, iRECIST, and PCWG-2. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:150-158. [PMID: 31286751 DOI: 10.23736/s1824-4785.19.03193-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor response is often used as a surrogate marker for survival practically in all clinical trials. Therefore, robust and valid response criteria during the course of trials are fundamental for the assessment of response to therapy. This aspect, however, becomes particularly challenging when it comes to bone metastases. In the era of targeted therapies and immune-checkpoint inhibitors (ICI), response assessment by morphologic-based criteria cannot detect the real tumor response and, consequently, fail to demonstrate the actual clinical benefit. This review will focus on some of the most common morphologic and metabolic response criteria and their application for bone lesions, highlighting relative strengths and weaknesses as well as potential future methods in the era of target therapies and immunotherapy with ICI.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy -
| |
Collapse
|
18
|
Winfield JM, Miah AB, Strauss D, Thway K, Collins DJ, deSouza NM, Leach MO, Morgan VA, Giles SL, Moskovic E, Hayes A, Smith M, Zaidi SH, Henderson D, Messiou C. Utility of Multi-Parametric Quantitative Magnetic Resonance Imaging for Characterization and Radiotherapy Response Assessment in Soft-Tissue Sarcomas and Correlation With Histopathology. Front Oncol 2019; 9:280. [PMID: 31106141 PMCID: PMC6494941 DOI: 10.3389/fonc.2019.00280] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/27/2019] [Indexed: 02/05/2023] Open
Abstract
Purpose: To evaluate repeatability of quantitative multi-parametric MRI in retroperitoneal sarcomas, assess parameter changes with radiotherapy, and correlate pre-operative values with histopathological findings in the surgical specimens. Materials and Methods: Thirty patients with retroperitoneal sarcoma were imaged at baseline, of whom 27 also underwent a second baseline examination for repeatability assessment. 14/30 patients were treated with pre-operative radiotherapy and were imaged again after completing radiotherapy (50.4 Gy in 28 daily fractions, over 5.5 weeks). The following parameter estimates were assessed in the whole tumor volume at baseline and following radiotherapy: apparent diffusion coefficient (ADC), parameters of the intra-voxel incoherent motion model of diffusion-weighted MRI (D, f, D*), transverse relaxation rate, fat fraction, and enhancing fraction after gadolinium-based contrast injection. Correlation was evaluated between pre-operative quantitative parameters and histopathological assessments of cellularity and fat fraction in post-surgical specimens (ClinicalTrials.gov, registration number NCT01902667). Results: Upper and lower 95% limits of agreement were 7.1 and -6.6%, respectively for median ADC at baseline. Median ADC increased significantly post-radiotherapy. Pre-operative ADC and D were negatively correlated with cellularity (r = -0.42, p = 0.01, 95% confidence interval (CI) -0.22 to -0.59 for ADC; r = -0.45, p = 0.005, 95% CI -0.25 to -0.62 for D), and fat fraction from Dixon MRI showed strong correlation with histopathological assessment of fat fraction (r = 0.79, p = 10-7, 95% CI 0.69-0.86). Conclusion: Fat fraction on MRI corresponded to fat content on histology and therefore contributes to lesion characterization. Measurement repeatability was excellent for ADC; this parameter increased significantly post-radiotherapy even in disease categorized as stable by size criteria, and corresponded to cellularity on histology. ADC can be utilized for characterizing and assessing response in heterogeneous retroperitoneal sarcomas.
Collapse
Affiliation(s)
- Jessica M. Winfield
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Aisha B. Miah
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Dirk Strauss
- Department of Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Khin Thway
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Department of Histopathology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - David J. Collins
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Nandita M. deSouza
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Martin O. Leach
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Veronica A. Morgan
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Sharon L. Giles
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Eleanor Moskovic
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Andrew Hayes
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Myles Smith
- Department of Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Shane H. Zaidi
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Daniel Henderson
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Christina Messiou
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| |
Collapse
|
19
|
Salah S, Lewin J, Amir E, Abdul Razak A. Tumor necrosis and clinical outcomes following neoadjuvant therapy in soft tissue sarcoma: A systematic review and meta-analysis. Cancer Treat Rev 2018; 69:1-10. [PMID: 29843049 DOI: 10.1016/j.ctrv.2018.05.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prognostic role of tumor necrosis following neoadjuvant therapy is established in bone sarcomas but remains unclear in soft tissue sarcomas (STS). METHODS We searched MEDLINE, MEDLINE in progress, EMBASE and Cochrane to identify studies that investigated neoadjuvant therapy in STS. Studies were required to report survival data based on extent of necrosis, or provided individual patient data allowing estimation thereof. Hazard ratios (HR) for relapse-free (RFS) and overall survival (OS) and odds ratios (OR) for recurrence at 3 years and for death at 5 years were pooled in a random effect meta-analysis. Associations between patient characteristics and attainment of ≥90% necrosis were explored. RESULTS 21 studies comprising 1663 patients were included. Extremity tumors were most common (n = 1554; 93%). Induction regimens included chemotherapy with radiotherapy (n = 924; 56%), chemotherapy alone (n = 412; 25%), radiotherapy alone (n = 78; 5%), isolated limb perfusion (ILP) (n = 231; 14%), and targeted therapy/radiotherapy (n = 18; 1%). Patients with <90% necrosis had higher hazard of recurrence (hazard ratio [HR] 1.47; 95% CI: 1.06-2.04; p = 0.02) and death (HR 1.86; 95% CI: 1.41-2.46; p < 0.001). Risk of recurrence at 3 years (OR = 3.35; 95% CI: 2.27-4.92; p < 0.001) and of death at 5 years (OR 2.60; 95% CI: 1.59-4.26; p < 0.001) were similarly increased. Compared to other modalities, ILP was associated with higher odds of achieving ≥90% necrosis (OR 12.1; 95% CI: 3.69-39.88; p < 0.001). CONCLUSION Tumour necrosis <90% following neoadjuvant therapy is associated with increased recurrence risk and inferior OS in patients with STS.
Collapse
Affiliation(s)
- Samer Salah
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medical Oncology, King Hussein Cancer Centre, Queen Rania Al Abdullah St 202, Amman, Jordan.
| | - Jeremy Lewin
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| | - Eitan Amir
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| | - Albiruni Abdul Razak
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
20
|
Role of chemotherapy in dedifferentiated liposarcoma of the retroperitoneum: defining the benefit and challenges of the standard. Sci Rep 2017; 7:11836. [PMID: 28928422 PMCID: PMC5605500 DOI: 10.1038/s41598-017-12132-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/05/2017] [Indexed: 11/08/2022] Open
Abstract
Benefit from chemotherapy for well-differentiated/de-differentiated (WD/DD) liposarcomas has been reported to be minimal, however traditional response criteria may not adequately capture positive treatment effect. In this study, we evaluate benefit from first-line chemotherapy and characterize imaging response characteristics in patients with retroperitoneal (RP) WD/DD liposarcoma treated at The University of Texas MD Anderson Cancer Center. Response was assessed using RECIST (Response Evaluation Criteria in Solid Tumors) and an exploratory analysis of vascular response was characterized. Among 82 patients evaluable for response to first-line therapy, 31 patients received neoadjuvant chemotherapy for localized/locally advanced disease; 51 received chemotherapy for unresectable recurrent/metastatic disease. Median overall survival from the start of chemotherapy was 29 months (95% CI 24-40 months). Response rates by RECIST: partial response (PR) 21% (17/82), stable disease (SD) 40%, and progression (PD) 39%. All RECIST responses were in patients receiving combination chemotherapy. A qualitative vascular response was seen in 24 patients (31%). Combination chemotherapy yields a response rate of 24% and a clinical benefit rate (CR/PR/SD > 6 months) of 44%, higher than previously reported in DD liposarcoma. A higher percentage of patients experience a vascular response with chemotherapy that is not adequately captured by RECIST in these large heterogeneous tumors.
Collapse
|
21
|
Choi H. Role of Imaging in Response Assessment and Individualised Treatment for Sarcomas. Clin Oncol (R Coll Radiol) 2017; 29:481-488. [PMID: 28506521 DOI: 10.1016/j.clon.2017.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/21/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
The first systematic response evaluation criteria were established by WHO, based on the tumor size changes shortly after the computed tomography (CT) technique became available to the daily practice. RECIST, a simplified version of WHO criteria, and its newer version, RECIST1.1 are the currently available international response evaluation criteria in solid tumors and remains based on tumor size changes. While the introduction of molecularly targeted drugs has significantly improved the survival in patient with sarcomas, the evaluation of tumor response has become more complicated. Increasing number of studies have reported the lack of shrinkage in responding tumors and raised concerns of significant underestimation of responses using RECIST. The first such observation was made on gastrointestinal stromal tumor (GIST) treated with imatinib. In GISTs responding to imatinib, the degree of contrast enhancement on CT typically decreases significantly compared with the baseline, and, regardless of whether tumors shrink, heterogeneous hyperattenuating tumors become homogeneous hypoattenuating tumors with a smaller enhancing solid component. In current oncology practice, CT is a widely accepted method of evaluating tumor response. CT images are relatively simple to acquire and can be reasonably reproduced with no significant technical obstacles. FDG-PET is highly sensitive and specific in identifying responding sarcomas. It has mostly been used as a problem solver and for those with marginally resectable GIST. More recently, the utility of whole body MRI is undergoing exploration. This article discusses the traditional size-based response evaluation criteria, and introduces new evidence based response evaluation based on changes in morphology in addition to changes in tumor size on CT images, and whole body imaging is introduced at the end.
Collapse
Affiliation(s)
- H Choi
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
22
|
Wulfkuhle JD, Spira A, Edmiston KH, Petricoin EF. Innovations in Clinical Trial Design in the Era of Molecular Profiling. Methods Mol Biol 2017; 1606:19-36. [PMID: 28501991 DOI: 10.1007/978-1-4939-6990-6_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Historically, cancer has been studied, and therapeutic agents have been evaluated based on organ site, clinical staging, and histology. The science of molecular profiling has expanded our knowledge of cancer at the cellular and molecular level such that numerous subtypes are being described based on biomarker expression and genetic mutations rather than traditional classifications of the disease. Drug development has experienced a concomitant revolution in response to this knowledge with many new targeted therapeutic agents becoming available, and this has necessitated an evolution in clinical trial design. The traditional, large phase II and phase III adjuvant trial models need to be replaced with smaller, shorter, and more focused trials. These trials need to be more efficient and adaptive in order to quickly assess the efficacy of new agents and develop new companion diagnostics. We are now seeing a substantial shift from the traditional multiphase trial model to an increase in phase II adjuvant and neoadjuvant trials in earlier-stage disease incorporating surrogate endpoints for long-term survival to assess efficacy of therapeutic agents in shorter time frames. New trial designs have emerged with capabilities to assess more efficiently multiple disease types, multiple molecular subtypes, and multiple agents simultaneously, and regulatory agencies have responded by outlining new pathways for accelerated drug approval that can help bring effective targeted therapeutic agents to the clinic more quickly for patients in need.
Collapse
Affiliation(s)
- Julia D Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, Institute for Advanced Biomedical Research, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, USA.
| | - Alexander Spira
- Virginia Cancer Specialists, 8503 Arlington Blvd, Suite 400, Fairfax, VA, 22031, USA
- Department of Surgery, Inova Fairfax Hospital Cancer Center, 3300 Gallows Road, Falls Church, VA, 22042, USA
| | - Kirsten H Edmiston
- Department of Surgery, Inova Fairfax Hospital Cancer Center, 3300 Gallows Road, Falls Church, VA, 22042, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, Institute for Advanced Biomedical Research, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, USA
| |
Collapse
|
23
|
Eary JF. Diagnostic Applications of Nuclear Medicine: Sarcomas. NUCLEAR ONCOLOGY 2017:1047-1064. [DOI: 10.1007/978-3-319-26236-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Lagmay JP, Krailo MD, Dang H, Kim A, Hawkins DS, Beaty O, Widemann BC, Zwerdling T, Bomgaars L, Langevin AM, Grier HE, Weigel B, Blaney SM, Gorlick R, Janeway KA. Outcome of Patients With Recurrent Osteosarcoma Enrolled in Seven Phase II Trials Through Children's Cancer Group, Pediatric Oncology Group, and Children's Oncology Group: Learning From the Past to Move Forward. J Clin Oncol 2016; 34:3031-8. [PMID: 27400942 DOI: 10.1200/jco.2015.65.5381] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The use of radiographic response as the primary end point in phase II osteosarcoma trials may limit optimal detection of treatment response because of the calcified tumor matrix. We performed this study to determine if time to progression could be used as an end point for subsequent studies. PATIENTS AND METHODS We performed a retrospective analysis of outcome for patients with recurrent/refractory osteosarcoma enrolled in one of seven phase II trials conducted by the Children's Oncology Group and predecessor groups from 1997 to 2007. All trials used RECIST or WHO radiographic response criteria and the primary end point of response rate. The following potential prognostic factors-age, trial, number of prior chemotherapy regimens, sex, and race/ethnicity-were evaluated for their impact on event-free survival (EFS). We used data from a phase II study (AOST0221) of patients with osteosarcoma who were given inhaled granulocyte-macrophage colony-stimulating factor with first pulmonary recurrence who had an EFS as well as biologic end point to determine the historical disease control rate for patients with fully resected disease. RESULTS In each included trial, the drugs tested were determined to be inactive on the basis of radiographic response rates. The EFS for 96 patients with osteosarcoma and measurable disease was 12% at 4 months (95% CI, 6% to 19%). There was no significant difference in EFS across trials according to number of prior treatment regimens or patient age, sex, and ethnicity. The 12-month EFS for the 42 evaluable patients enrolled in AOST0221 was 20% (95% CI, 10% to 34%). CONCLUSION The EFS was uniformly poor for children with recurrent/refractory osteosarcoma in these single-arm phase II trials. We have now constructed baseline EFS outcomes that can be used as a comparison for future phase II trials for recurrent osteosarcoma.
Collapse
Affiliation(s)
- Joanne P Lagmay
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY.
| | - Mark D Krailo
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Ha Dang
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - AeRang Kim
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Douglas S Hawkins
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Orren Beaty
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brigitte C Widemann
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Theodore Zwerdling
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Lisa Bomgaars
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Anne-Marie Langevin
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Holcombe E Grier
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brenda Weigel
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Susan M Blaney
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Richard Gorlick
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Katherine A Janeway
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| |
Collapse
|
25
|
Aghighi M, Boe J, Rosenberg J, Von Eyben R, Gawande RS, Petit P, Sethi TK, Sharib J, Marina NM, DuBois SG, Daldrup-Link HE. Three-dimensional Radiologic Assessment of Chemotherapy Response in Ewing Sarcoma Can Be Used to Predict Clinical Outcome. Radiology 2016; 280:905-15. [PMID: 26982677 DOI: 10.1148/radiol.2016151301] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose To compare the agreement of three-dimensional (3D) tumor measurements for therapeutic response assessment of Ewing sarcoma according to the Children's Oncology Group (COG) criteria, one-dimensional (1D) Response Evaluation Criteria in Solid Tumors (RECIST), and two-dimensional (2D) measurements defined by the World Health Organization (WHO) with tumor volume measurements as the standard of reference and to determine which method correlates best with clinical outcomes. Materials and Methods This retrospective study was approved by the institutional review board of three institutions. Seventy-four patients (mean age ± standard deviation, 14.5 years ± 6.5) with newly diagnosed Ewing sarcoma treated at three medical centers were evaluated. Primary tumor size was assessed on pre- and posttreatment magnetic resonance images according to 1D RECIST, 2D WHO, and 3D COG measurements. Tumor responses were compared with the standard of reference (tumor volume) on the basis of RECIST, COG, and WHO therapeutic response thresholds. Agreement between the percentage reduction measurements of the methods was assessed with concordance correlation, Bland-Altman analysis, and Spearman rank correlation. Agreement between therapeutic responses was assessed with Kendall tau and unweighted κ statistics. Tumor responses were compared with patient survival by using the log-rank test, Kaplan-Meier plots, and Cox regression. Results Agreement with the reference standard was significantly better for 3D measurement than for 1D and 2D measurements on the basis of RECIST and COG therapeutic response thresholds (concordance correlation of 0.41, 0.72, and 0.84 for 1D, 2D, and 3D measurements, respectively; P < .0001). Comparison of overall survival of responders and nonresponders demonstrated P values of .4133, .0112, .0032, and .0027 for 1D, 2D, 3D, and volume measurements, respectively, indicating that higher dimensional measurements were significantly better predictors of overall survival. Conclusion The 3D tumor measurements according to COG are better predictors of therapeutic response of Ewing sarcoma than 1D RECIST or 2D WHO measurements and show a significantly higher correlation with clinical outcomes. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Maryam Aghighi
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Justin Boe
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Jarrett Rosenberg
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Rie Von Eyben
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Rakhee S Gawande
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Philippe Petit
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Tarsheen K Sethi
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Jeremy Sharib
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Neyssa M Marina
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Steven G DuBois
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| | - Heike E Daldrup-Link
- From the Department of Radiology, Section of Pediatric Radiology (M.A., J.B., J.R., R.S.G., T.K.S., H.E.D.L.), and Department of Pediatric Hematology/Oncology (N.M.M.), Lucile Packard Children's Hospital, Stanford University, 725 Welch Rd, Stanford, CA 94305-5654; Department of Radiation and Oncology, Stanford University, Stanford, Calif (R.V.E.); Department of Pediatric and Prenatal Imaging, Hôpital de la Timone, Marseille, France (P.P.); Department of Pediatrics, University of California-San Francisco School of Medicine, San Francisco, Calif (J.S., S.G.D.); and UCSF Benioff Children's Hospital, San Francisco, Calif (J.S., S.G.D.)
| |
Collapse
|
26
|
Ronellenfitsch U, Dimitrakopoulou-Strauss A, Jakob J, Kasper B, Nowak K, Pilz LR, Attenberger U, Gaiser T, Egerer G, Fröhling S, Derigs HG, Schwarzbach M, Hohenberger P. Preoperative therapy with pazopanib in high-risk soft tissue sarcoma: a phase II window-of-opportunity study by the German Interdisciplinary Sarcoma Group (GISG-04/NOPASS). BMJ Open 2016; 6:e009558. [PMID: 26739732 PMCID: PMC4716254 DOI: 10.1136/bmjopen-2015-009558] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION For resectable soft tissue sarcoma (STS), radical surgery, usually combined with radiotherapy, is the mainstay of treatment and the only potentially curative modality. Since surgery is often complicated by large tumour size and extensive tumour vasculature, preoperative treatment strategies with the aim of devitalising the tumour are being explored. One option is treatment with antiangiogenic drugs. The multikinase inhibitor pazopanib, which possesses pronounced antiangiogenic effects, has shown activity in metastatic and unresectable STS, but has so far not been tested in the preoperative setting. METHODS AND ANALYSIS This open-label, multicentre phase II window-of-opportunity trial assesses pazopanib as preoperative treatment of resectable STS. Participants receive a 21-day course of pazopanib 800 mg daily during wait time for surgery. Major eligibility criteria are resectable, high-risk adult STS of any location, or metachronous solitary STS metastasis for which resection is planned, and adequate organ function and performance status. The trial uses an exact single-stage design. The primary end point is metabolic response rate (MRR), that is, the proportion of patients with >50% reduction of the mean standardised uptake value (SUVmean) in post-treatment compared to pre-treatment fluorodeoxyglucose positron emission tomography CT. The MRR below which the treatment is considered ineffective is 0.2. The MRR above which the treatment warrants further exploration is 0.4. With a type I error of 5% and a power of 80%, the sample size is 35 evaluable patients, with 12 or more responders as threshold. Main secondary end points are histopathological and MRI response, resectability, toxicity, recurrence-free and overall survival. In a translational substudy, endothelial progenitor cells and vascular epithelial growth factor receptor are analysed as potential prognostic and predictive markers. ETHICS AND DISSEMINATION Approval by the ethics committee II, University of Heidelberg, Germany (2012-019F-MA), German Federal Institute for Drugs and Medical Devices (61-3910-4038155) and German Federal Institute for Radiation Protection (Z5-22463/2-2012-007). TRIAL REGISTRATION NUMBER NCT01543802, EudraCT: 2011-003745-18; Pre-results.
Collapse
Affiliation(s)
- Ulrich Ronellenfitsch
- Division of Surgical Oncology and Thoracic Surgery, Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Jens Jakob
- Division of Surgical Oncology and Thoracic Surgery, Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bernd Kasper
- Medical Faculty Mannheim, Interdisciplinary Tumor Center, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kai Nowak
- Division of Surgical Oncology and Thoracic Surgery, Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lothar R Pilz
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ulrike Attenberger
- Medical Faculty Mannheim, Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Timo Gaiser
- Medical Faculty Mannheim, Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gerlinde Egerer
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Heidelberg, Germany Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hans-Günter Derigs
- Department of Hematology and Oncology, Klinikum Frankfurt-Höchst, Frankfurt am Main, Germany
| | | | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
27
|
Technical prerequisites and imaging protocols for CT perfusion imaging in oncology. Eur J Radiol 2015; 84:2359-67. [PMID: 26137905 DOI: 10.1016/j.ejrad.2015.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 06/11/2015] [Indexed: 12/29/2022]
Abstract
The aim of this review article is to define the technical prerequisites of modern state-of-the-art CT perfusion imaging in oncology at reasonable dose levels. The focus is mainly on abdominal and thoracic tumor imaging, as they pose the largest challenges with respect to attenuation and patient motion. We will show that low kV dynamic scanning in conjunction with detection technology optimized for low photon fluxes has the highest impact on reducing dose independently of other choices made in the protocol selection. We discuss, derived from relatively simple first principles, on what appropriate temporal sampling and total scan duration depend on and why optimized contrast medium injection protocols are also essential in limiting dose. Finally we will examine the possibility of simultaneously extracting standard morphological and functional information from one single 4D examination as a potential enabler for a more widespread use of dynamic contrast enhanced CT in oncology.
Collapse
|
28
|
Jakob J, Simeonova A, Kasper B, Ronellenfitsch U, Wenz F, Hohenberger P. Combined radiation therapy and sunitinib for preoperative treatment of soft tissue sarcoma. Ann Surg Oncol 2015; 22:2839-45. [PMID: 26085221 DOI: 10.1245/s10434-015-4680-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 01/29/2023]
Abstract
BACKGROUND Antiangiogenic substances and radiation therapy (RT) may have synergistic effects and improve irradiation efficacy. We present a cohort study evaluating the toxicity of combined sunitinib and RT as neoadjuvant treatment of extremity and retroperitoneal soft tissue sarcoma (STS). METHODS Sixteen patients with locally advanced extremity (6/16) or retroperitoneal (10/16) STS were treated with continuous-dosing sunitinib (15/16: 37.5 mg daily; 1/16: 25 mg daily) and standard RT (45-50.4 Gy) preoperatively. Surgery was scheduled 5-9 weeks following neoadjuvant treatment. The primary goal of the study was to determine combined treatment toxicity according to the Common Terminology Criteria for Adverse Events. Secondary goals were the evaluation of postoperative morbidity and treatment response. RESULTS Eight of 16 patients developed grade 3, and one patient developed grade 4, hematological toxicity. One patient experienced grade 3 hand-foot syndrome. The most frequent treatment toxicities of any grade were hematological (15/16) or dermatological (9/16). Three patients had partial response, 11 had stable disease, and 2 had progressive disease according to Response Evaluation Criteria in Solid Tumors (RECIST). Fourteen of 16 patients underwent surgery; tumors were not removed in two patients because of patient refusal or intercurrent metastatic disease. The proportion of tumor necrosis exceeded 90 % in 5 of 14 patients, and 4 patients had postoperative complications requiring reintervention. CONCLUSIONS Preoperative treatment with concurrent sunitinib and RT was tolerable, and postoperative morbidity did not increase. Combined treatment with RT and sunitinib was also feasible in patients with retroperitoneal STS, and warrants further investigation.
Collapse
Affiliation(s)
- Jens Jakob
- Department of Surgery, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,
| | | | | | | | | | | |
Collapse
|
29
|
Schuler MK, Platzek I, Beuthien-Baumann B, Fenchel M, Ehninger G, van den Hoff J. (18)F-FDG PET/MRI for therapy response assessment in sarcoma: comparison of PET and MR imaging results. Clin Imaging 2015; 39:866-70. [PMID: 26117565 DOI: 10.1016/j.clinimag.2015.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 05/06/2015] [Accepted: 05/26/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND (18)F-Fluorodeoxyglucose (FDG) positron emission tomography (PET) has proven to be of substantial benefit in imaging of sarcoma patients. We therefore investigated the feasibility and benefit of combined PET/magnetic resonance imaging (MRI). METHODS Twelve patients with sarcoma who underwent FDG PET/MRI for staging and response assessment after chemotherapy were included. RESULTS Based on contrast-enhanced MRI and application of Choi criteria, therapy response was classified as stable disease in 6/12 patients (50%) and as partial remission in 6/12 patients (50%). CONCLUSION In sarcoma patients, response assessment using Choi criteria based on contrast-enhanced MRI in comparison to FDG PET imaging only demonstrates slight correlation.
Collapse
Affiliation(s)
- Markus Kajo Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden.
| | - Ivan Platzek
- Department of Radiology, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden
| | - Bettina Beuthien-Baumann
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden
| | | | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden
| | | |
Collapse
|
30
|
Kim MN, Kim BK, Han KH, Kim SU. Evolution from WHO to EASL and mRECIST for hepatocellular carcinoma: considerations for tumor response assessment. Expert Rev Gastroenterol Hepatol 2015; 9:335-348. [PMID: 25370168 DOI: 10.1586/17474124.2015.959929] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Radiological response assessment criteria in hepatocellular carcinoma (HCC) have evolved to accurately evaluate tumor responses. The WHO criteria and the subsequent Response Evaluation Criteria in Solid Tumors (RECIST) evaluate change in tumor size; however, these criteria generally ignore tumor necrosis and therefore may underestimate treatment responses. Thus, a panel of experts of the European Association for the Study of Liver (EASL) amended the response criteria to take into account tumor necrosis. In 2010, the modified RECIST (mRECIST) was developed, which consider both the concept of tumor viability based on arterial enhancement and single linear summation, ultimately simplifying EASL criteria. Currently, the mRECIST represents the gold standard for radiologically evaluating tumor response during HCC treatment. Here, the authors review application and performance of mRECIST as well as other HCC response assessment criteria and discuss unmet and open issues regarding response evaluation for HCC treatments.
Collapse
Affiliation(s)
- Mi Na Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul 120-752, Korea
| | | | | | | |
Collapse
|
31
|
Oubel E, Bonnard E, Sueoka-Aragane N, Kobayashi N, Charbonnier C, Yamamichi J, Mizobe H, Kimura S. Volume-based response evaluation with consensual lesion selection: a pilot study by using cloud solutions and comparison to RECIST 1.1. Acad Radiol 2015; 22:217-25. [PMID: 25488429 DOI: 10.1016/j.acra.2014.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 09/05/2014] [Accepted: 09/20/2014] [Indexed: 12/11/2022]
Abstract
RATIONALE AND OBJECTIVES Lesion volume is considered as a promising alternative to Response Evaluation Criteria in Solid Tumors (RECIST) to make tumor measurements more accurate and consistent, which would enable an earlier detection of temporal changes. In this article, we report the results of a pilot study aiming at evaluating the effects of a consensual lesion selection on volume-based response (VBR) assessments. MATERIALS AND METHODS Eleven patients with lung computed tomography scans acquired at three time points were selected from Reference Image Database to Evaluate Response to therapy in lung cancer (RIDER) and proprietary databases. Images were analyzed according to RECIST 1.1 and VBR criteria by three readers working in different geographic locations. Cloud solutions were used to connect readers and carry out a consensus process on the selection of lesions used for computing response. Because there are not currently accepted thresholds for computing VBR, we have applied a set of thresholds based on measurement variability (-35% and +55%). The benefit of this consensus was measured in terms of multiobserver agreement by using Fleiss kappa (κfleiss) and corresponding standard errors (SE). RESULTS VBR after consensual selection of target lesions allowed to obtain κfleiss = 0.85 (SE = 0.091), which increases up to 0.95 (SE = 0.092), if an extra consensus on new lesions is added. As a reference, the agreement when applying RECIST without consensus was κfleiss = 0.72 (SE = 0.088). These differences were found to be statistically significant according to a z-test. CONCLUSIONS An agreement on the selection of lesions allows reducing the inter-reader variability when computing VBR. Cloud solutions showed to be an interesting and feasible strategy for standardizing response evaluations, reducing variability, and increasing consistency of results in multicenter clinical trials.
Collapse
Affiliation(s)
- Estanislao Oubel
- R&D Department, MEDIAN Technologies, Les Deux Arcs B, 1800 Route des Crêtes, Valbonne 06560, France.
| | - Eric Bonnard
- Radiology Department, Nice University Hospital, Nice, France
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Naomi Kobayashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Colette Charbonnier
- R&D Department, MEDIAN Technologies, Les Deux Arcs B, 1800 Route des Crêtes, Valbonne 06560, France
| | - Junta Yamamichi
- Global Healthcare IT Project, Medical Equipment Group, Canon Inc, Tokyo, Japan
| | - Hideaki Mizobe
- Global Healthcare IT Project, Medical Equipment Group, Canon Inc, Tokyo, Japan
| | - Shinya Kimura
- Radiology Department, Nice University Hospital, Nice, France
| |
Collapse
|
32
|
Bellera CA, Penel N, Ouali M, Bonvalot S, Casali PG, Nielsen OS, Delannes M, Litière S, Bonnetain F, Dabakuyo TS, Benjamin RS, Blay JY, Bui BN, Collin F, Delaney TF, Duffaud F, Filleron T, Fiore M, Gelderblom H, George S, Grimer R, Grosclaude P, Gronchi A, Haas R, Hohenberger P, Issels R, Italiano A, Jooste V, Krarup-Hansen A, Le Péchoux C, Mussi C, Oberlin O, Patel S, Piperno-Neumann S, Raut C, Ray-Coquard I, Rutkowski P, Schuetze S, Sleijfer S, Stoeckle E, Van Glabbeke M, Woll P, Gourgou-Bourgade S, Mathoulin-Pélissier S. Guidelines for time-to-event end point definitions in sarcomas and gastrointestinal stromal tumors (GIST) trials: results of the DATECAN initiative (Definition for the Assessment of Time-to-event Endpoints in CANcer trials)†. Ann Oncol 2014; 26:865-872. [PMID: 25070543 DOI: 10.1093/annonc/mdu360] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 07/23/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The use of potential surrogate end points for overall survival, such as disease-free survival (DFS) or time-to-treatment failure (TTF) is increasingly common in randomized controlled trials (RCTs) in cancer. However, the definition of time-to-event (TTE) end points is rarely precise and lacks uniformity across trials. End point definition can impact trial results by affecting estimation of treatment effect and statistical power. The DATECAN initiative (Definition for the Assessment of Time-to-event End points in CANcer trials) aims to provide recommendations for definitions of TTE end points. We report guidelines for RCT in sarcomas and gastrointestinal stromal tumors (GIST). METHODS We first carried out a literature review to identify TTE end points (primary or secondary) reported in publications of RCT. An international multidisciplinary panel of experts proposed recommendations for the definitions of these end points. Recommendations were developed through a validated consensus method formalizing the degree of agreement among experts. RESULTS Recommended guidelines for the definition of TTE end points commonly used in RCT for sarcomas and GIST are provided for adjuvant and metastatic settings, including DFS, TTF, time to progression and others. CONCLUSION Use of standardized definitions should facilitate comparison of trials' results, and improve the quality of trial design and reporting. These guidelines could be of particular interest to research scientists involved in the design, conduct, reporting or assessment of RCT such as investigators, statisticians, reviewers, editors or regulatory authorities.
Collapse
Affiliation(s)
- C A Bellera
- Clinical and Epidemiological Research Unit, Institut Bergonie, Comprehensive Cancer Centre, Bordeaux; Clinical Epidemiology Unit, INSERM CIC 14.01 (Clinical Epidemiology), Bordeaux.
| | - N Penel
- Department of Medical Oncology, Centre Oscar Lambret, Comprehensive Cancer Centre, Lille, France
| | - M Ouali
- Department of Biostatistics, European Organisation for Research and Treatment of Cancer, Brussels, Belgium; Biostatistics Unit, Institut Claudius Regaud, Comprehensive Cancer Centre, Toulouse
| | - S Bonvalot
- Department of Surgery, Institut Gustave Roussy, Comprehensive Cancer Centre, Villejuif, France
| | - P G Casali
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - O S Nielsen
- Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - M Delannes
- Department of Radiotherapy, Institut Claudius Régaud, Comprehensive Cancer Center, Toulouse
| | - S Litière
- Department of Biostatistics, European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - F Bonnetain
- Methodological and Quality of Life Unit in Oncology (EA3181), CHU Besançon, Besançon
| | - T S Dabakuyo
- Biostatistics and Quality of Life Unit (EA4184), Centre Georges-François Leclerc, Comprehensive Cancer Centre, Dijon, France
| | - R S Benjamin
- Division of Cancer Medicine and Sarcoma Center, The University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - J-Y Blay
- Department of Medical Oncology, Centre Léon Bérard, Comprehensive Cancer Centre, Lyon; Claude Bernard Lyon I University, Lyon; Medical Oncology Unit, Edouard Herriot Hospital, Lyon
| | - B N Bui
- Department of Medical Oncology, Institut Bergonié, Comprehensive Cancer Centre, Bordeaux
| | - F Collin
- Department of Biology and Pathology, Centre Georges-François Leclerc, Comprehensive Cancer Centre, Dijon, France
| | - T F Delaney
- Department of Radiation Oncology and Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, USA
| | - F Duffaud
- Department of Medical Oncology, La Timone Hospital University, Marseille, France
| | - T Filleron
- Biostatistics Unit, Institut Claudius Regaud, Comprehensive Cancer Centre, Toulouse
| | - M Fiore
- Department of Surgery and Sarcoma Unit, Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - H Gelderblom
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - S George
- Department of Medical Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - R Grimer
- Royal Orthopaedic Hospital NHS Trust, Birmingham, UK
| | - P Grosclaude
- Cancer Registry of Tarn, Institut Claudius Regaud, Comprehensive Cancer Centre, Toulouse, France
| | - A Gronchi
- Department of Surgery and Sarcoma Unit, Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - R Haas
- Department of Radiation Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - P Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, Mannheim University Medical Center, Mannheim
| | - R Issels
- Sarcoma Center, Ludwig-Maximilian University Munich, Munich; Department of Internal Medicine, Klinikum Grosshadern Medical Center, University of Munich, Munich, Germany
| | - A Italiano
- Department of Medical Oncology, Institut Bergonié, Comprehensive Cancer Centre, Bordeaux
| | - V Jooste
- Burgundy Digestive Cancer Registry, INSERM U866, University of Burgundy, Dijon, France
| | - A Krarup-Hansen
- Department of Oncology, Herlev Hospital-University Copenhagen, Herlev, Denmark
| | - C Le Péchoux
- Department of Radiotherapy, Institut Gustave Roussy, Comprehensive Cancer Centre, Villejuif, France
| | - C Mussi
- Department of Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - O Oberlin
- Department of Surgery and Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, Comprehensive Cancer Centre, Villejuif
| | - S Patel
- Division of Cancer Medicine and Sarcoma Center, The University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - S Piperno-Neumann
- Department of Medical Oncology, Institut Curie, Comprehensive Cancer Centre, Paris, France
| | - C Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - I Ray-Coquard
- Department of Medical Oncology, Centre Léon Bérard, Comprehensive Cancer Centre, Lyon
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - S Schuetze
- Department of Medical Oncology, University of Michigan, Ann Arbor, USA
| | - S Sleijfer
- Department of Medical Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | - E Stoeckle
- Department of Surgical Oncology, Institut Bergonié, Comprehensive Cancer Centre, Bordeaux, France
| | - M Van Glabbeke
- Department of Biostatistics, European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - P Woll
- Department of Oncology, Sheffield Cancer Research Centre, Weston Park Hospital, Sheffield, UK
| | - S Gourgou-Bourgade
- Montpellier Cancer Institute, Comprehensive Cancer Centre, Montpellier, France
| | - S Mathoulin-Pélissier
- Clinical and Epidemiological Research Unit, Institut Bergonie, Comprehensive Cancer Centre, Bordeaux; Clinical Epidemiology Unit, INSERM CIC 14.01 (Clinical Epidemiology), Bordeaux
| |
Collapse
|
33
|
Shen Y, Anderson A, Sinha R, Li Y. Joint modeling tumor burden and time to event data in oncology trials. Pharm Stat 2014; 13:286-93. [PMID: 25044957 DOI: 10.1002/pst.1629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 04/22/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
Abstract
The tumor burden (TB) process is postulated to be the primary mechanism through which most anticancer treatments provide benefit. In phase II oncology trials, the biologic effects of a therapeutic agent are often analyzed using conventional endpoints for best response, such as objective response rate and progression-free survival, both of which causes loss of information. On the other hand, graphical methods including spider plot and waterfall plot lack any statistical inference when there is more than one treatment arm. Therefore, longitudinal analysis of TB data is well recognized as a better approach for treatment evaluation. However, longitudinal TB process suffers from informative missingness because of progression or death. We propose to analyze the treatment effect on tumor growth kinetics using a joint modeling framework accounting for the informative missing mechanism. Our approach is illustrated by multisetting simulation studies and an application to a nonsmall-cell lung cancer data set. The proposed analyses can be performed in early-phase clinical trials to better characterize treatment effect and thereby inform decision-making.
Collapse
Affiliation(s)
- Ye Shen
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, 30602, GA, USA
| | | | | | | |
Collapse
|
34
|
Tumour volume changes following pre-operative radiotherapy in borderline resectable limb and trunk soft tissue sarcoma. Eur J Surg Oncol 2014; 40:394-401. [PMID: 24534361 DOI: 10.1016/j.ejso.2014.01.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/31/2013] [Accepted: 01/17/2014] [Indexed: 11/24/2022] Open
Abstract
AIMS To evaluate tumour volume changes after preoperative radiotherapy (PRT) for borderline operable soft tissue sarcomas (STS). MATERIALS AND METHODS A retrospective review was performed of 68 patients who received PRT between December 2004 and July 2011. Endpoints were radiological response, surgical margins, local control and survival. RESULTS Median tumour size was 12.5 cm. Tumour location was extremity (87%), trunk (12%), and neck (1%). Commonest histological subtypes were myxoid liposarcoma (32%) and myxofibrosarcoma (16%). The majority of patients (88%) received 50 Gy in 25 fractions. Post-radiotherapy imaging was available in 55 cases. By RECIST there was stable disease in 89%, partial response in 7% and progressive disease in 4%. Tumour volumes reduced in 80%. Median change in maximal tumour dimension was -13.6%; median change in volume was greater, at -33.3%. Tumour volumes increased in 11 cases (20%). However, surgical margins were clear in all 11 cases, with no local recurrences in this group. For the entire group, surgical margins were clear in 93%, and microscopically positive in 7%. Eight patients (12%) had local relapse at 2-24.8 months after surgery. Two year local relapse free survival was 87.5%; 2 year overall survival was 74.7%. CONCLUSION The majority of tumours showed reduction in volume. A small number of tumours increased in volume, but there was no definite relationship between volume increase and poor surgical outcomes or lower local control rates. Local control was equivalent to published series' of PRT. PRT is a reasonable approach in patients with borderline resectable tumours.
Collapse
|
35
|
Chmielowski B, Federman N, Tap WD. Clinical trial end points for assessing efficacy of novel therapies for soft-tissue sarcomas. Expert Rev Anticancer Ther 2014; 12:1217-28. [DOI: 10.1586/era.12.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Schmitt T, Kasper B. New medical treatment options and strategies to assess clinical outcome in soft-tissue sarcoma. Expert Rev Anticancer Ther 2014; 9:1159-67. [PMID: 19671035 DOI: 10.1586/era.09.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas Schmitt
- University of Heidelberg, Department of Internal Medicine V, Germany
| | | |
Collapse
|
37
|
Alvarez FJ, Hosoya K, Lara-Garcia A, Kisseberth W, Couto G. VAC protocol for treatment of dogs with stage III hemangiosarcoma. J Am Anim Hosp Assoc 2013; 49:370-7. [PMID: 24051260 DOI: 10.5326/jaaha-ms-5954] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hemangiosarcomas (HSAs) are aggressive tumors with a high rate of metastasis. Clinical stage has been considered a negative prognostic factor for survival. The study authors hypothesized that the median survival time (MST) of dogs with metastatic (stage III) HSA treated with a vincristine, doxorubicin, and cyclophosphamide (VAC) chemotherapy protocol would not be different than those with stage I/II HSA. Sixty-seven dogs with HSA in different anatomic locations were evaluated retrospectively. All dogs received the VAC protocol as an adjuvant to surgery (n = 50), neoadjuvant (n = 3), or as the sole treatment modality (n = 14). There was no significant difference (P = 0.97) between the MST of dogs with stage III and stage I/II HSA. For dogs presenting with splenic HSA alone, there was no significant difference between the MST of dogs with stage III and stage I/II disease (P = 0.12). The overall response rate (complete response [CR] and partial response [PR]) was 86%). No unacceptable toxicities were observed. Dogs with stage III HSA treated with the VAC protocol have a similar prognosis to dogs with stage I/II HSA. Dogs with HSA and evidence of metastases at the time of diagnosis should not be denied treatment.
Collapse
|
38
|
|
39
|
Peungjesada S, Chuang HH, Prasad SR, Choi H, Loyer EM, Bronstein Y. Evaluation of cancer treatment in the abdomen: Trends and advances. World J Radiol 2013; 5:126-42. [PMID: 23671749 PMCID: PMC3650203 DOI: 10.4329/wjr.v5.i3.126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 01/24/2013] [Accepted: 01/31/2013] [Indexed: 02/06/2023] Open
Abstract
Response evaluation in Oncology has relied primarily on change in tumor size. Inconsistent results in the prediction of clinical outcome when size based criteria are used and the increasing role of targeted and loco-regional therapies have led to the development of new methods of response evaluation that are unrelated to change in tumor size. The goals of this review are to expose briefly the size based criteria and to present the non-size based approaches that are currently applicable in the clinical setting. Other paths that are still being explored are not discussed in details.
Collapse
|
40
|
Schiavon G, Ruggiero A, Schöffski P, van der Holt B, Bekers DJ, Eechoute K, Vandecaveye V, Krestin GP, Verweij J, Sleijfer S, Mathijssen RHJ. Tumor volume as an alternative response measurement for imatinib treated GIST patients. PLoS One 2012; 7:e48372. [PMID: 23133631 PMCID: PMC3487791 DOI: 10.1371/journal.pone.0048372] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022] Open
Abstract
Background Assessment of tumor size changes is crucial in clinical trials and patient care. We compared imatinib-induced volume changes of liver metastases (LM) from gastro-intestinal stromal tumors (GIST) to RECIST and Choi criteria and their association with overall survival (OS). Methods LM from 84 GIST patients (training and validation set) were evaluated using manual and semi-automated Computed Tomography measurements at baseline, after 3, 6 and 12 months of imatinib. The ability of uni-dimensional (1D) and three-dimensional (3D) measurements to detect size changes (increase/decrease) ≥20% was evaluated. Volumetric response cut-offs were derived from minimally relevant changes (+20/−30%) by RECIST, considering lesions as spherical or ellipsoidal. Results 3D measurements detected size changes ≥20% more frequently than 1D at every time-point (P≤0.008). 3D and Choi criteria registered more responses than RECIST at 3 and 6 months for 3D-spheres (P≤0.03) and at all time-points for 3D-ellipsoids and Choi criteria (P<0.001). Progressive disease by 3D criteria seems to better correlate to OS at late time-points than other criteria. Conclusion Volume criteria (especially ellipsoids) classify a higher number of patients as imatinib-responders than RECIST. Volume discriminates size changes better than diameter in GIST and constitutes a feasible and robust method to evaluate response and predict patient benefit.
Collapse
Affiliation(s)
- Gaia Schiavon
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Liu K, Qian T, Tang L, Wang J, Yang H, Ren J. Decreased expression of microRNA let-7i and its association with chemotherapeutic response in human gastric cancer. World J Surg Oncol 2012; 10:225. [PMID: 23107361 PMCID: PMC3500711 DOI: 10.1186/1477-7819-10-225] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 10/15/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MicroRNA let-7i has been proven to be down-regulated in many human malignancies and correlated with tumor progression and anticancer drug resistance. Our study aims to characterize the contribution of miRNA let-7i to the initiation and malignant progression of locally advanced gastric cancer (LAGC), and evaluate its possible value in neoadjuvant chemotherapeutic efficacy prediction. METHODS Eighty-six previously untreated LAGC patients who underwent preoperative chemotherapy and radical resection were included in our study. Let-7i expression was examined for pairs of cancer tissues and corresponding normal adjacent tissues (NATs), using quantitative RT-PCR. The relationship of let-7i level to clinicopathological characteristics, pathologic tumor regression grades after chemotherapy, and overall survival (OS) was also investigated. RESULTS Let-7i was significantly down-regulated in most tumor tissues (78/86: 91%) compared with paired NATs (P < 0.001), and low levels of let-7i were significantly correlated with local invasion, lymphatic metastasis, and poor pathologic tumor response. Multivariate Cox regression analysis revealed that low let-7i expression was an unfavorable prognostic factor of OS (hazard ratio (HR) = 2.316, P = 0.024) independently of other clinicopathological factors, including tumor node metastasis (TNM) stage (HR = 3.226, P = 0.013), depth of infiltration (HR = 4.167, P < 0.001), and lymph node status (HR = 2.245, P = 0.037). CONCLUSIONS These findings indicate that let-7i may be a good candidate for use a therapeutic target and a potential tissue marker for the prediction of chemotherapeutic sensitivity and prognosis in LAGC patients.
Collapse
Affiliation(s)
- Kun Liu
- Department of General Surgery, Changzhou No. 2 Hospital, Nanjing Medical University, Nanjing, China
| | - Tao Qian
- Department of General Surgery, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Liming Tang
- Department of General Surgery, Changzhou No. 2 Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Wang
- Department of General Surgery, Changzhou No. 2 Hospital, Nanjing Medical University, Nanjing, China
| | - Haohua Yang
- Department of General Surgery, Changzhou No. 2 Hospital, Nanjing Medical University, Nanjing, China
| | - Jun Ren
- Department of General Surgery, Changzhou No. 2 Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Gámez Cenzano C, Sabaté Llobera A, Narváez García JA, Rodríguez Bel L, García del Muro FJ. [Positron emission tomography-computed tomography in tumors of the locomotor apparatus]. RADIOLOGIA 2012; 54 Suppl 1:3-13. [PMID: 22959330 DOI: 10.1016/j.rx.2012.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/26/2012] [Accepted: 07/09/2012] [Indexed: 12/23/2022]
Abstract
Positron emission tomography/computed tomography (PET/CT) is a hybrid imaging technique that combines the anatomic information from CT with the metabolic information acquired from PET after the administration of specific radiotracers, the most commonly used of which is F18-fluorodeoxyglucose (FDG). In oncology, this technique is based on the increased uptake of FDG by malignant lesions. In the locomotor apparatus, some uptake by bones and soft tissues is physiological or benign and this uptake must be differentiated from uptake by malignancies, whether primary or secondary. The most important limitations are active inflammatory or infectious processes, which are positive on PET images, and malignant lesions that are smaller than 1cm, cystic, necrotic, or low-grade, which are negative on PET images. PET/CT in the locomotor apparatus is especially useful for the detection of metastases from the most common tumors. It is also used for staging and monitoring the response to treatment of some hematological tumors like lymphoma, where it is fundamental to determine whether the bone marrow has been infiltrated, or myeloma. Lastly, although it is not yet an established indication, PET/CT is being increasingly used to study sarcomas, because it can provide additional information that can be useful for the characterization and grading of tumors, for guiding biopsies, for staging and re-staging, and for evaluating the response to neoadjuvant therapy as well as for evaluating new drugs in clinical trials.
Collapse
Affiliation(s)
- C Gámez Cenzano
- Unidad PET-IDI, Hospital Universitario de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, España.
| | | | | | | | | |
Collapse
|
43
|
Levine ZH, Pintar AL, Hagedorn JG, Fenimore CP, Heussel CP. Uncertainties in RECIST as a measure of volume for lung nodules and liver tumors. Med Phys 2012; 39:2628-37. [PMID: 22559633 DOI: 10.1118/1.3701791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE The authors wish to determine the extent to which the Response Evaluation Criteria in Solid Tumors (RECIST) and the criteria of the World Health Organization (WHO) can predict tumor volumes and changes in volume using clinical data. METHODS The data presented are a reanalysis of data acquired in other studies, including the public database from the Lung Image Database Consortium (LIDC) and from a study of liver tumors. RESULTS The principal result is that a given RECIST diameter predicts volume to a factor of 16 or 10 for the two data sets, respectively, by examining 95% prediction bounds and that changes in volume are predicted only little better: to within a factor of 7 for the liver data. The WHO criteria reduce the prediction bounds by a factor of 1.3 in all cases. Also, the RECIST threshold of 10 mm to measure a nodule corresponds to a transition zone width of a factor of more than 2 in volume for the nodules in the LIDC database. CONCLUSIONS While the RECIST diameter is certainly correlated with the volume, and similarly for changes in these quantities, the use of the diameter introduces additional variation assuming volume is the quantity of interest. Exactly how much this reduces the statistical power of clinical drug trials is a key open question for future research.
Collapse
Affiliation(s)
- Zachary H Levine
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | | | | | | | | |
Collapse
|
44
|
Grabellus F, Stylianou E, Umutlu L, Sheu SY, Lehmann N, Taeger G, Lauenstein TC. Size-based clinical response evaluation is insufficient to assess clinical response of sarcomas treated with isolated limb perfusion with TNF-α and melphalan. Ann Surg Oncol 2012; 19:3375-85. [PMID: 22622472 DOI: 10.1245/s10434-012-2408-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Indexed: 11/18/2022]
Abstract
BACKGROUND The clinical assessment of the response of sarcomas to preoperative treatment is usually defined using size-based evaluation standards. For nonresectable sarcomas, hyperthermic isolated limb perfusion with TNF-α and melphalan (TM-ILP) yields high response rates. Based on our experience, we assume that anatomic radiological response criteria are insufficient to assess the degree of regression after TM-ILP. METHODS The clinical response of 35 sarcomas to TM-ILP was assessed by unidimensional, bidimensional, and tridimensional size-based anatomical criteria, and responders were identified according to the established thresholds. The same tumors were investigated for pathological response according to the Salzer-Kuntschik regression scale (>90% devitalization) and reviewed for cystic degeneration, hemorrhage, and predominant necrotic or fibrosclerotic regression phenotype. RESULTS None of the clinical response criteria were able to reliably identify the pathologic responders. The extent of size changes showed no association with the pathological degree of regression. The number of clinical responders was low compared with the number of pathological responders (RECIST N = 1, WHO N = 3, volumetry N = 3, pathology N = 19). The occurrence of hemorrhage and/or cystic degeneration was more frequently observed in predominant necrotic sarcomas and was associated with an increase in tumor size after TM-ILP. Furthermore, we identified the fibrosclerotic phenotype of regression to be more significantly strongly associated with posttherapeutic shrinkage than necrosis. CONCLUSIONS Size-based clinical response evaluation is insufficient to assess clinical response in TM-ILP-treated sarcomas. The size changes of tumors after therapy reflect the type of regression rather than the extent of destruction.
Collapse
Affiliation(s)
- Florian Grabellus
- Institute of Pathology and Neuropathology, University Hospital of Essen and Sarcoma Center at the West German Cancer Center, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Liu K, Li G, Fan C, Zhou C, Li J. Adapted Choi response criteria for prediction of clinical outcome in locally advanced gastric cancer patients following preoperative chemotherapy. Acta Radiol 2012; 53:127-34. [PMID: 22156007 DOI: 10.1258/ar.2011.110273] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tumor response to chemotherapy has traditionally been assessed by using Response Evaluation Criteria in Solid Tumors (RECIST) based on changes in tumor size alone. However, adapted Choi criteria, which incorporate volumetric tumor attenuation in addition to tumor size, have been reported more predictive of chemotherapeutic efficacy than RECIST in some studies. PURPOSE To examine the usefulness of adapted Choi criteria in predicting clinical survival in locally advanced gastric cancer patients treated with cytotoxic drugs. MATERIAL AND METHODS A total of 48 histologically proven gastric cancer patients who received neoadjuvant chemotherapy and surgery were involved. Pre- and post-chemotherapy short-axis diameter and volumetric mean tumor attenuation of target lymph nodes on contrast-enhanced CT images were measured. Tumor response was assessed by using both RECIST and adapted Choi criteria, and was correlated with progression-free survival (PFS) and overall survival (OS). RESULTS Significant decrease was observed in the sum of short-axis diameters and tumor attenuation of metastatic lymph nodes between baseline and post-chemotherapy CT images. The inter-observer agreement for both parameters was good. The PFS and OS of 17 RECIST responders were identical with that of 28 adapted Choi responders (P = 0.855 and 0.913, respectively). PFS and OS of 31 RECIST non-responders were significantly prolonged compared to that of 20 adapted Choi non-responders (P = 0.018 and 0.042, respectively). To the 11 RECIST stable disease (SD) but adapted Choi partial response (PR) patients, the PFS and OS were similar to the survival of 17 RECIST PR patients (P = 0.785 and 0.838, respectively), but significantly prolonged compared to that of the 12 both RECIST and adapted Choi criteria SD patients (P < 0.001 and P = 0.004, respectively). CONCLUSION Adapted Choi criteria might be helpful to predict PFS and OS in locally advanced gastric cancer patients following chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Changsheng Zhou
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | | |
Collapse
|
46
|
Olofsson R, Bergh P, Berlin Ö, Engström K, Gunterberg B, Hansson M, Lindnér P, Mattsson J. Long-Term Outcome of Isolated Limb Perfusion in Advanced Soft Tissue Sarcoma of the Extremity. Ann Surg Oncol 2012; 19:1800-7. [DOI: 10.1245/s10434-011-2196-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Indexed: 11/18/2022]
|
47
|
Abstract
The accelerating science of molecular profiling has necessitated a rapid evolution in clinical trial design. Traditional clinical research begins with Phase I studies to characterize dose-limiting toxicities and defines maximally tolerated doses of drugs in small numbers of patients. Traditional Phase II studies test these drugs at the doses discovered during Phase I drug development in small numbers of patients evaluating efficacy and safety. Phase III studies test new therapies to demonstrate improved activity or improved tolerability compared with a standard of care regimen or a placebo. The rapid advances in the understanding of signal transduction, and the identification of new potential diagnostic and therapeutic targets, now require the design and implementation of molecular clinical trials that are very different than traditional Phase I, II, or III trials. The main differentiating factor is the use of a molecular end point to stratify a subset of patients to receive a specific treatment regimen. This chapter focuses on the issues surrounding (a) the definition of clinical end points and the assessment of tumor response; (b) clinical trial design models to define the targeted pathway; and (c) the need for appropriate biomarkers to monitor the response.
Collapse
|
48
|
Schmitt T, Lehner B, Kasper B, Bischof M, Roeder F, Dietrich S, Dimitrakopoulou-Strauss A, Strauss LG, Mechtersheimer G, Wuchter P, Ho AD, Egerer G. A phase II study evaluating neo-/adjuvant EIA chemotherapy, surgical resection and radiotherapy in high-risk soft tissue sarcoma. BMC Cancer 2011; 11:510. [PMID: 22152120 PMCID: PMC3248452 DOI: 10.1186/1471-2407-11-510] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 12/07/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The role of chemotherapy in high-risk soft tissue sarcoma is controversial. Though many patients undergo initial curative resection, distant metastasis is a frequent event, resulting in 5-year overall survival rates of only 50-60%. Neo-adjuvant and adjuvant chemotherapy (CTX) has been applied to achieve pre-operative cytoreduction, assess chemosensitivity, and to eliminate occult metastasis. Here we report on the results of our non-randomized phase II study on neo-adjuvant treatment for high-risk STS. METHOD Patients with potentially curative high-risk STS (size ≥ 5 cm, deep/extracompartimental localization, tumor grades II-III [FNCLCC]) were included. The protocol comprised 4 cycles of neo-adjuvant chemotherapy (EIA, etoposide 125 mg/m(2) iv days 1 and 4, ifosfamide 1500 mg/m2 iv days 1 - 4, doxorubicin 50 mg/m(2) day 1, pegfilgrastim 6 mg sc day 5), definitive surgery with intra-operative radiotherapy, adjuvant radiotherapy and 4 adjuvant cycles of EIA. RESULT Between 06/2005 and 03/2010 a total of 50 subjects (male = 33, female = 17, median age 50.1 years) were enrolled. Median follow-up was 30.5 months. The majority of primary tumors were located in the extremities or trunk (92%), 6% originated in the abdomen/retroperitoneum. Response by RECIST criteria to neo-adjuvant CTX was 6% CR (n = 3), 24% PR (n = 12), 62% SD (n = 31) and 8% PD (n = 4). Local recurrence occurred in 3 subjects (6%). Distant metastasis was observed in 12 patients (24%). Overall survival (OS) and disease-free survival (DFS) at 2 years was 83% and 68%, respectively. Multivariate analysis failed to prove influence of resection status or grade of histological necrosis on OS or DFS. Severe toxicities included neutropenic fever (4/50), cardiac toxicity (2/50), and CNS toxicity (4/50) leading to CTX dose reductions in 4 subjects. No cases of secondary leukemias were observed so far. CONCLUSION The current protocol is feasible for achieving local control rates, as well as OS and DFS comparable to previously published data on neo-/adjuvant chemotherapy in this setting. However, the definitive role of chemotherapy remains unclear in the absence of large, randomized trials. Therefore, the current regimen can only be recommended within a clinical study, and a possibly increased risk of secondary leukemias has to be taken into account. TRIAL REGISTRATION ClinicalTrials.gov NCT01382030, EudraCT 2004-002501-72.
Collapse
Affiliation(s)
- Thomas Schmitt
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Lehnhardt M, Schmitt T, Bischof M, Daigeler A, Egerer G. [Current state of neoadjuvant therapy of soft tissue sarcoma]. Chirurg 2011; 82:995-1000. [PMID: 22008846 DOI: 10.1007/s00104-011-2132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The treatment of soft tissue sarcoma is clinically challenging. Referral to an experienced center with an interdisciplinary team is strongly recommended. Neoadjuvant therapy, including irradiation and chemotherapy, has been applied to improve local control rates, eradicate micrometastases and assess chemosensitivity. However, the role of neoadjuvant therapy remains controversial, especially for systemic therapy, as the only available randomized trial failed to prove a benefit for survival. Nevertheless, on the basis of the current body of literature, neoadjuvant therapy can be considered on an individual basis for patients with high-risk tumors. Whenever possible, patients should be included in a clinical trial.
Collapse
Affiliation(s)
- M Lehnhardt
- Klinik für Hand-, Plastische- und Rekonstruktive Chirurgie, Schwerbrandverletztenzentrum, Klinik für Plastische Chirurgie der Ruprecht-Karls-Universität Heidelberg, BG-Unfallklinik Ludwigshafen, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Deutschland.
| | | | | | | | | |
Collapse
|