51
|
Keenan TE, Li T, Vallius T, Guerriero JL, Tayob N, Kochupurakkal B, Davis J, Pastorello R, Tahara RK, Anderson L, Conway J, He MX, Shannon E, Godin RE, Sorger PK, D'Andrea A, Overmoyer B, Winer EP, Mittendorf EA, Van Allen EM, Shapiro GI, Tolaney SM. Clinical Efficacy and Molecular Response Correlates of the WEE1 Inhibitor Adavosertib Combined with Cisplatin in Patients with Metastatic Triple-Negative Breast Cancer. Clin Cancer Res 2021; 27:983-991. [PMID: 33257427 PMCID: PMC7887044 DOI: 10.1158/1078-0432.ccr-20-3089] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We report results from a phase II study assessing the efficacy of the WEE1 inhibitor adavosertib with cisplatin in metastatic triple-negative breast cancer (mTNBC). PATIENTS AND METHODS Patients with mTNBC treated with 0-1 prior lines of chemotherapy received cisplatin 75 mg/m2 i.v. followed 21 days later by cisplatin plus adavosertib 200 mg oral twice daily for five doses every 21 days. The study had 90% power to detect the difference between null (20%) and alternative (40%) objective response rates (ORR) with a one-sided type I error of 0.1: an ORR >30% was predefined as making the regimen worthy of further study. RNA sequencing and multiplex cyclic immunofluorescence on pre- and post-adavosertib tumor biopsies, as well as targeted next-generation sequencing on archival tissue, were correlated with clinical benefit, defined as stable disease ≥6 months or complete or partial response. RESULTS A total of 34 patients initiated protocol therapy; median age was 56 years, 2 patients (6%) had BRCA2 mutations, and 14 (41%) had one prior chemotherapy. ORR was 26% [95% confidence interval (CI), 13-44], and median progression-free survival was 4.9 months (95% CI, 2.3-5.7). Treatment-related grade 3-5 adverse events occurred in 53% of patients, most commonly diarrhea in 21%. One death occurred because of sepsis, possibly related to study therapy. Tumors from patients with clinical benefit demonstrated enriched immune gene expression and T-cell infiltration. CONCLUSIONS Among patients with mTNBC treated with 0-1 prior lines, adavosertib combined with cisplatin missed the prespecified ORR cutoff of >30%. The finding of immune-infiltrated tumors in patients with clinical benefit warrants validation.
Collapse
Affiliation(s)
- Tanya E Keenan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Tianyu Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Tuulia Vallius
- Breast Tumor Immunology Laboratory, Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Nabihah Tayob
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Bose Kochupurakkal
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Janae Davis
- Breast Tumor Immunology Laboratory, Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Ricardo Pastorello
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Rie K Tahara
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Leilani Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Jake Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Meng X He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Harvard Graduate Program in Biophysics, Boston, Massachusetts
| | - Erin Shannon
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | | | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Alan D'Andrea
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Beth Overmoyer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| |
Collapse
|
52
|
Hada T, Miyamoto M, Ishibashi H, Matsuura H, Sakamoto T, Kakimoto S, Iwahashi H, Tsuda H, Takano M. Survival and biomarker analysis for ovarian mucinous carcinoma according to invasive patterns: retrospective analysis and review literature. J Ovarian Res 2021; 14:33. [PMID: 33583413 PMCID: PMC7883414 DOI: 10.1186/s13048-021-00783-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/08/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In ovarian mucinous carcinoma, invasive pattern is the important factor but there were less reposts to investigate it. The aim of this study was to examine the association between prognosis and invasive patterns of ovarian mucinous carcinoma and to investigate the biomarkers of the diagnosis and prognosis immunochemically. Patients with ovarian mucinous carcinoma at our institution between 1984 and 2018 were identified. A pathological review was conducted using the 2020 World Health Organization criteria. The prognosis of infiltrative invasion and expansile invasion of ovarian mucinous carcinoma were retrospectively compared. In addition, immunohistochemical staining was conducted for all cases, and the immunohistochemical differences between the two invasive patterns were compared. RESULTS After the pathological review, 25 cases with infiltrative invasion and 24 cases with expansile invasion were included. Ovarian mucinous carcinoma with infiltrative invasion showed significantly worse progression-free survival (PFS, p < 0.01) and overall survival (OS, p < 0.01) than those with expansile invasion. Multivariate analysis demonstrated that the pattern of infiltrative invasion was a worse prognostic factor for PFS (hazard ratio 9.01, p < 0.01) and OS (hazard ratio 17.56, p < 0.01). Immunohistochemically, cytokeratin (CK) 5/6 (p = 0.01), cluster of differentiation (CD) 24 (p = 0.02), and epithelial growth factor receptor (EGFR) (p < 0.01) were statistically related to infiltrative invasion. The PFS (p = 0.04) and OS (p = 0.02) of cases with EGFR-positive OMC were worse than those with negative OMC. CONCLUSIONS Infiltrative invasion was observed to be a prognostic factor showing worse outcomes for ovarian mucinous carcinoma compared to expansile infiltration. CK5/6, CD24, and EGFR might be biomarkers of the diagnosis.
Collapse
Affiliation(s)
- Taira Hada
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Morikazu Miyamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan.
| | - Hiroki Ishibashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Hiroko Matsuura
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Takahiro Sakamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Soichiro Kakimoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Hideki Iwahashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Defense Medical College Hospital, 359-8513, Tokorozawa, Saitama, Japan
| | - Masashi Takano
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, 3-2, Namiki, Saitama, 359-8513, Tokorozawa, Japan
| |
Collapse
|
53
|
Elsholtz FHJ, Asbach P, Haas M, Becker M, Beets-Tan RGH, Thoeny HC, Padhani AR, Hamm B. Introducing the Node Reporting and Data System 1.0 (Node-RADS): a concept for standardized assessment of lymph nodes in cancer. Eur Radiol 2021; 31:6116-6124. [PMID: 33585994 PMCID: PMC8270876 DOI: 10.1007/s00330-020-07572-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/04/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
"Node-RADS" addresses the lack of consensus in the radiologic assessment of lymph node involvement by cancer and meets the increasing demand for structured reporting on the likelihood of disease involvement. Node Reporting and Data System 1.0 (Node-RADS) systematically classifies the degree of suspicion of lymph node involvement based on the synthesis of established imaging findings. Straightforward definitions of imaging findings for two proposed scoring categories "size" and "configuration" are combined into assessment categories between 1 ("very low likelihood") and 5 ("very high likelihood"). This scoring system is suitable for assessing likely involvement of lymph nodes on CT and MRI scans. It can be applied at any anatomical site, and to regional and non-regional lymph nodes in relation to a primary tumor location. Node-RADS will improve communication with referring physicians and promote the consistency of reporting for primary staging and in response assessment settings. KEY POINTS: • Node-RADS standardizes reporting of possible cancer involvement of regional and distant lymph nodes on CT and MRI. • Node-RADS proposes the scoring categories "size" and "configuration" for assigning the 5-point Node-RADS score from 1 ("very low likelihood") to 5 ("very high likelihood"). • Node-RADS aims to increase consensus among radiologists for primary staging and in response assessment settings.
Collapse
Affiliation(s)
- Fabian H J Elsholtz
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Patrick Asbach
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Haas
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Minerva Becker
- Division of Radiology, Department of Imaging and Medical Informatics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Harriet C Thoeny
- Department of Diagnostic and Interventional Radiology, Fribourg Cantonal Hospital, Faculty of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - Bernd Hamm
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
54
|
Qualitative and Quantitative Assessment of Nonlocal Means Reconstruction Algorithm in a Flexible PET Scanner. AJR Am J Roentgenol 2020; 216:486-493. [PMID: 33236947 DOI: 10.2214/ajr.19.22245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE. Flexible PET (fxPET) was designed to fit existing MRI systems. The newly modified nonlocal means (NLM) algorithm is combined with the 3D dynamic row-action maximum likelihood algorithm (DRAMA). We investigated qualitative and quantitative acceptability of fxPET images reconstructed by modified NLM compared with whole-body (WB) PET/CT images and conventional 3D DRAMA reconstruction alone. MATERIALS AND METHODS. Fifty-nine patients with known or suspected malignancies underwent WB PET/CT scanning approximately 1 hour after the injection of 18F-FDG, after which they underwent fxPET scanning. Two readers rated the quality of fxPET images by consensus. Detection rate (the proportion of lesions found on PET), maximal standardized uptake value (SUVmax), metabolic tumor volume (MTV), total lesion glycolysis (TLG), tumor-to-normal liver ratio (TNR), and background liver signal-to-noise ratio (SNR) were compared among the three datasets. RESULTS. Higher image quality was obtained by modified NLM reconstruction than by conventional reconstruction without statistical significance. The detection rate was comparable among three datasets. SUVmax was significantly higher, and MTV and TLG were significantly lower in the modified NLM dataset (p < 0.002) than in the other two datasets, with significantly positive correlations (p < 0.001; Spearman rank correlation coefficient, 0.87-0.99). The TNRs in modified NLM images were significantly larger than in the other datasets (p < 0.05). The background SNRs in modified NLM images were comparable with those in WB PET/CT images, and significantly higher than in the conventional fxPET images (p < 0.005). CONCLUSION. The modified NLM algorithm was clinically acceptable, yielding higher TNR and background SNR compared with conventional reconstruction. Image quality and the lesion detection rate were comparable in this population.
Collapse
|
55
|
Zhu Z, Li Q, Xu M, Qi Z. Effect of Whole-Brain and Intensity-Modulated Radiotherapy on Serum Levels of miR-21 and Prognosis for Lung Cancer Metastatic to the Brain. Med Sci Monit 2020; 26:e924640. [PMID: 33125362 PMCID: PMC7607665 DOI: 10.12659/msm.924640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background The goal of the present study was to explore the influence of whole-brain radiotherapy (WBRT) and intensity-modulated radiotherapy (IMRT) on serum levels of miR-21 and prognosis for lung cancer that has metastasized to the brain. Material/Methods Two hundred patients with lung cancer metastatic to the brain were randomized, half to the control group and half to the observation group. The observation group received WBRT and reduced-field IMRT (WBRT+RF-IMRT) and the control group received conventional-field IMRT (CF-IMRT). The total effective rate after treatment was determined. Serum levels of miR-21 were measured before and after radiotherapy with reverse transcriptase-polymerase chain reaction. In addition, tumor marker levels were measured with enzyme-linked immunosorbent assay. The relationship between miR-21 levels and tumor marker levels was assessed with a Pearson correlation coefficient test. Five-year survival was estimated with Kaplan-Meier curves. Results The total effective rate was higher in the observation group (86%) than in the control group (69%). Lower levels of miR-21 and tumor markers were seen in the observation group. Moreover, miR-21 levels were positively correlated with levels of tumor necrosis factor-α, neuron-specific enolase, SCC-Ag, and carcinoembryonic antigen. Low levels of miR-21 were associated with longer overall survival in patients with lung cancer metastatic to the brain. Conclusions WBRT+RF-IMRT is superior to CF-IMRT for lung cancer metastatic to the brain. MiR-21 may be a marker for prediction of the efficacy of radiotherapy in this disease setting.
Collapse
Affiliation(s)
- Zhensheng Zhu
- Department of Oncology, Jinan Hospital of Integrated Traditional Chinese and Western Medicine, Jinan, Shandong, China (mainland)
| | - Qiurong Li
- Department of Lung Disease Division, Jinan Hospital of Integrated Traditional Chinese and Western Medicine, Jinan, Shandong, China (mainland)
| | - Mingjuan Xu
- Department of Cardiology, Jinan People's Hospital, Jinan, Shandong, China (mainland)
| | - Zhongliang Qi
- Department of Oncology, Jinan Hospital of Integrated Traditional Chinese and Western Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|
56
|
Bielack SS, Cox MC, Nathrath M, Apel K, Blattmann C, Holl T, Jenewein R, Klenk U, Klothaki P, Müller-Abt P, Ortega-Lawerenz S, Reynolds M, Scheer M, Simon-Klingenstein K, Stegmaier S, Tupper R, Vokuhl C, von Kalle T. Rapid, complete and sustained tumour response to the TRK inhibitor larotrectinib in an infant with recurrent, chemotherapy-refractory infantile fibrosarcoma carrying the characteristic ETV6-NTRK3 gene fusion. Ann Oncol 2020; 30 Suppl 8:viii31-viii35. [PMID: 31738425 PMCID: PMC6859811 DOI: 10.1093/annonc/mdz382] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The ETV6-NTRK3 gene fusion is present in the majority of cases of infantile fibrosarcoma (IFS) and acts as a potent oncogenic driver. We report the very rapid, complete, and sustained response of an advanced, chemotherapy-refractory, recurrent IFS to targeted treatment with the oral tropomyosin receptor kinase (TRK) inhibitor larotrectinib. PATIENT AND METHODS A male infant born with a large congenital IFS of the tongue had the tumour surgically resected at age 4 days. Within 2 months, he developed extensive lymph node recurrence that progressed during two cycles of vincristine-doxorubicin-cyclophosphamide chemotherapy. At screening, a large right cervical mass was clinically visible. Magnetic resonance imaging (MRI) revealed bilateral cervical and axillary lymph node involvement as well as infiltration of the floor of the mouth. The largest lesion measured 5.5×4.5×4.4 cm (ca. 55 cm3). The patient started outpatient oral larotrectinib at 20 mg/kg twice daily at age 3.5 months. RESULTS After 4 days on treatment, the parents noted that the index tumour was visibly smaller and softer. The rapid tumour regression continued over the following weeks. On day 56 of treatment, the first scheduled control MRI showed the target lesion had shrunk to 1.2×1.2×0.8 cm (ca. 0.6 cm3), corresponding to a complete response according to the Response Evaluation Criteria In Solid Tumors version 1.1. This response was maintained over subsequent follow-up visits, and on day 112 at the second control MRI the target lymph node was completely normal. At last follow-up, the disease remained in complete remission after 16 months on larotrectinib, with negligible toxicity and no safety concerns. CONCLUSION(S) Selective TRK inhibition by larotrectinib offers a novel, highly specific and highly effective therapeutic option for IFS carrying the characteristic ETV6-NTRK3 gene fusion. Its use should be considered when surgery is not feasible. (NCT02637687).
Collapse
Affiliation(s)
- S S Bielack
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart.,Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - M C Cox
- Loxo Oncology, a wholly owned subsidiary of Eli Lilly and Company, South San Francisco, USA
| | - M Nathrath
- Department of Pediatric Hematology and Oncology, Klinikum Kassel, Kassel
| | - K Apel
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - C Blattmann
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - T Holl
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - R Jenewein
- Radiologic Institute, Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - U Klenk
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - P Klothaki
- Department of Pediatric Hematology and Oncology, Klinikum Kassel, Kassel
| | - P Müller-Abt
- Radiologic Institute, Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | | | - M Reynolds
- Loxo Oncology, a wholly owned subsidiary of Eli Lilly and Company, South San Francisco, USA
| | - M Scheer
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - K Simon-Klingenstein
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - S Stegmaier
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| | - R Tupper
- Loxo Oncology, a wholly owned subsidiary of Eli Lilly and Company, South San Francisco, USA
| | - C Vokuhl
- Institute of Pathology - Section Pediatric Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - T von Kalle
- Radiologic Institute, Center for Pediatric, Adolescent and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart - Olgahospital, Stuttgart
| |
Collapse
|
57
|
Differential Change in Size of Human Papillomavirus-Positive Cystic Versus Solid Squamous Cell Carcinoma Lymph Node Metastases in Response to Induction Chemotherapy. J Comput Assist Tomogr 2020; 44:389-392. [PMID: 32176158 DOI: 10.1097/rct.0000000000000998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE According to certain cancer treatment protocols, the response to induction chemotherapy of lymph node metastases based on radiographic measurements guides further management. The aim of this study is to verify the observation that cystic metastatic lymph nodes tend not to shrink as rapidly as solid metastatic lymph nodes in response to induction chemotherapy in patients with human papillomavirus-related oropharyngeal squamous cell carcinoma. METHODS The lymphadenopathy in a cohort of patients from a clinical trial with human papillomavirus-related oropharyngeal squamous cell carcinoma with both baseline and postinduction chemotherapy (carboplatin/paclitaxel/cetuximab) contrast-enhanced neck computed tomography was retrospectively reviewed. The appearance of the metastatic lymph nodes on computed tomography was characterized as cystic or solid. A cystic lymph node was defined as having a hypoattenuating component greater than 20% of the total volume. The rates of short-axis and volume changes of cystic and solid lymph nodes were compared using 1-tailed t test. RESULTS A total of 46 patients were included in this study, comprising 39 solid and 45 cystic lymph nodes. The rate of short-axis decrease was significantly greater for solid (1.33% per day) than cystic (1.08% per day) lymph nodes (P = 0.036). Likewise, the rate of volume decrease was significantly greater for solid (2.13% per day) than cystic (1.87% per day) lymph nodes (P = 0.014). CONCLUSIONS This study suggests that in patients with human papillomavirus-related oropharyngeal squamous cell carcinoma solid lymph node metastases generally decrease in size at a greater rate than cystic lymph nodes after induction chemotherapy.
Collapse
|
58
|
Lo YW, Lee JC, Hu YS, Li CY, Chen YL, Lin CS, Huang WS, Lin KH, Chen YW. The importance of optimal ROIs delineation for FBPA-PET before BNCT. Appl Radiat Isot 2020; 163:109219. [PMID: 32561058 DOI: 10.1016/j.apradiso.2020.109219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 11/27/2022]
Abstract
One of the eligible criteria for patients to receive boron neutron capture therapy (BNCT) is based on the tumour-to-normal ratio (T/N) measured by FBPA-PET. However, there is no standard protocol for normal region-of-interested delineation. With comparison of contralateral cerebrum, our study revealed the consistency (p < 0.05) and high feasibility using the cerebellum as an alternative normal tissue baseline because of its homogeneous uptake. Following RECIST version 1.1, the standard-operating-procedure (SOP) for the BNCT fulfilled the expected tumour response and tumour shrinkage rate (p < 0.05). Our modified procedure can provide more precise information for BNCT within a reasonable time.
Collapse
Affiliation(s)
- Yi-Wen Lo
- Integrated PET/MR Imaging Centre, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jia-Cheng Lee
- Division of Radiotherapy, Department of Oncology Medicine, Taipei Veterans General Hospital, Taiwan
| | - Yong-Sin Hu
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Ying Li
- Integrated PET/MR Imaging Centre, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Lun Chen
- Integrated PET/MR Imaging Centre, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Shuo Lin
- Division of Radiotherapy, Department of Oncology Medicine, Taipei Veterans General Hospital, Taiwan
| | - Wen-Sheng Huang
- Integrated PET/MR Imaging Centre, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ko-Han Lin
- Integrated PET/MR Imaging Centre, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Yi-Wei Chen
- Division of Radiotherapy, Department of Oncology Medicine, Taipei Veterans General Hospital, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
59
|
Bartsch F, Hahn F, Müller L, Baumgart J, Hoppe-Lotichius M, Kloeckner R, Lang H. Relevance of suspicious lymph nodes in preoperative imaging for resectability, recurrence and survival of intrahepatic cholangiocarcinoma. BMC Surg 2020; 20:75. [PMID: 32295646 PMCID: PMC7161232 DOI: 10.1186/s12893-020-00730-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is often diagnosed at an advanced stage resulting in a low resectability rate. Even after potentially curative resection the risk for tumor recurrence is high. Although the extent and value of lymphadenectomy is part of ongoing discussion, the role of preoperative imaging for assessment of suspicious lymph nodes (suspLN) has only been studied modestly. Aim of this study is to demonstrate the influence of suspicious lymph nodes in preoperative imaging on resectability, recurrence, and long-term outcome. METHODS All patients who underwent exploration for ICC between January 2008 and June 2018 were included. Preoperative imaging (CT or MRI) was analysed with focus on suspLN at the hepatoduodenal ligament, lesser curvature, interaortocaval, and superior to the diaphragm; suspLN were classified according to the universally accepted RECIST 1.1 criteria; histopathology served as gold standard. RESULTS Out of 187 patients resection was performed in 137 (73.3%), in 50 patients the procedure was terminated after exploration. Overall, suspLN were found preoperatively in 73/187 patients (39%). Comparing patients who underwent resection and exploration only, suspLN were significantly more common in the exploration group (p = 0.011). Regarding lymph node stations, significant differences could be shown regarding resectability: All tumors with suspLN superior to the diaphragm were irresectable. Preoperative imaging assessment showed a strong correlation with final histopathology, especially of suspLN of the hepatoduodenal ligament and the lesser curvature. Sensitivity of suspLN was 71.1%, specificity 90.8%. Appearance of tumor recurrence was not affected by suspLN (p = 0.289). Using a short-axis cut-off of <> 1 cm, suspLN had significant influence on recurrence-free survival (RFS, p = 0.009) with consecutive 1-, 3-, and 5-year RFS of 41, 21, and 15% versus 29, 0, and 0%, respectively. Similarly, 1-, 3- and 5-year overall survival (OS) was 75, 30, and 18% versus 59, 18, and 6%, respectively (p = 0.040). CONCLUSION Suspicious lymph nodes in preoperative imaging are predictor for unresectability and worse survival. Explorative laparoscopy should be considered, if distant suspicious lymph nodes are detected in preoperative imaging. Nevertheless, given a sensitivity of only 71.1%, detection of suspicious lymph nodes in the preoperative imaging alone is not sufficient to allow for a clear-cut decision against a surgical approach.
Collapse
Affiliation(s)
- Fabian Bartsch
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Felix Hahn
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas Müller
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Janine Baumgart
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Maria Hoppe-Lotichius
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Roman Kloeckner
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany.
| |
Collapse
|
60
|
Retroperitoneal and Pelvic Lymph Nodes in Children: What Is Normal? AJR Am J Roentgenol 2020; 214:1384-1388. [PMID: 32228324 DOI: 10.2214/ajr.19.22316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE. The purpose of this study was to evaluate size criteria for retroperitoneal and pelvic lymph nodes in healthy children. MATERIALS AND METHODS. We identified all trauma patients younger than 18 years old without underlying disease and with CT scans without abnormalities in the abdomen and pelvis during 2014-2015. Two pediatric radiologists reviewed the studies independently and recorded the number of retroperitoneal and pelvic lymph nodes with a long diameter 5 mm or greater and the size (two perpendicular diameters) of the largest lymph node in five anatomic locations. Discrepant results were reviewed in consensus. The relationship of short diameter to age and interobserver variability was evaluated. RESULTS. A total of 166 patients (86 boys) with a mean age of 7.2 years old (range, 0.1-18.0 years old) were identified. More than 95% of lymph nodes in the retroperitoneum and pelvis had a short diameter measuring at most 7 and 8 mm, respectively, by consensus. The size of the largest short diameter of lymph nodes did not vary with age. More than four lymph nodes were identified in any anatomic location in only three patients, by only one of the radiologists. Agreement for lymph nodes with largest diameter of 5 mm or greater between radiologists ranged from 70.5% to 97.6% for the five anatomic locations with poor interobserver agreement (κ, 0.2-0.3). CONCLUSION. The size and number of retroperitoneal and pelvic lymph nodes in children are less than in adults. A short diameter threshold of 7 mm (retroperitoneal) and 8 mm (pelvic) and more than four lymph nodes with long diameter of 5 mm or greater in one location may define disease.
Collapse
|
61
|
Charruf AZ, Ramos MFKP, Pereira MA, Dias AR, de Castria TB, Zilberstein B, Cecconelo I, Ribeiro U. Impact of neoadjuvant chemotherapy on surgical and pathological results of gastric cancer patients: A case-control study. J Surg Oncol 2020; 121:833-839. [PMID: 31943232 DOI: 10.1002/jso.25839] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Neoadjuvant chemotherapy (NACT) followed by radical surgery represents a treatment option for patients with advanced gastric cancer (GC). This case-control study aimed to evaluate the clinicopathological characteristics and surgical outcomes of GC patients who received NACT, and its impact on survival. METHODS We retrospectively reviewed all patients with GC who underwent gastrectomy. A total of 45 cases with NACT were matched with consecutive 45 patients who underwent upfront gastrectomy for the following characteristics: gender, age, gastrectomy type, lymphadenectomy extent, American Society of Anesthesiologists class, histological type, cT and cN. RESULTS NACT group had smaller tumors (4.9 vs 6.8 cm P = .006), lower lymphatic invasion rate (40% vs 73.3%, P = .001), lower venous invasion rate (18% vs 46.7%, P = .003) and lower perineural invasion rate (35% vs 77.8%, P < .0001). The ypTNM stage was lower in patients treated with NACT (P < .001). The major postoperative complication (POC) rate was lower in NACT patients (6.7% vs 24.4%, P = .02), as was hospital length of stay (10.8 vs 17 days, P = .005). CONCLUSIONS NACT allowed nodal and tumor downstaging. In addition, patients who underwent NACT had fewer POC and shorter length of hospital stay.
Collapse
Affiliation(s)
- Amir Zeide Charruf
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Marina Alessandra Pereira
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Andre Roncon Dias
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Tiago Biachi de Castria
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruno Zilberstein
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ivan Cecconelo
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ulysses Ribeiro
- Gastrointestinal Surgery Department, Instituto do Câncer - Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
62
|
Tanaka K, Matsumoto Y, Ishikawa H, Fukumitsu N, Numajiri H, Murofushi K, Oshiro Y, Okumura T, Satoh T, Sakurai H. Impact of RhoA overexpression on clinical outcomes in cervical squamous cell carcinoma treated with concurrent chemoradiotherapy. JOURNAL OF RADIATION RESEARCH 2020; 61:221-230. [PMID: 31976530 PMCID: PMC7246076 DOI: 10.1093/jrr/rrz093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/05/2019] [Indexed: 05/04/2023]
Abstract
The Rho-associated coiled-coil-containing protein kinase (ROCK) pathway is known to influence metastasis in several cancers; however, the impact of the pathway on clinical outcomes in patients undergoing radiotherapy remains unknown. In the present study, the expression of RhoA, RhoC, ROCK-1, ROCK-2 and p53 was immunohistochemically evaluated using biopsy specimens obtained from 49 patients with stage II-III cervical squamous cell carcinoma treated with concurrent chemoradiotherapy (CCRT). The relationship between the expression of these proteins and patient outcomes was investigated. RhoA overexpression was associated with significantly impaired disease-free survival and distant metastasis-free survival (P = 0.045 and P = 0.041, respectively) in stage III cancer patients. No differences in survival were observed based on the expression of the other proteins among stage III cancer patients. In stage II cancer patients, no differences in survival were noted based on the expression of any of the proteins. The expression of RhoA was able to successfully differentiate cervical cancer patients with distant metastasis after CCRT. This information may help stratify patients according to the risk of metastasis, thereby leading to the potential to provide individualized treatment.
Collapse
Affiliation(s)
- Keiichi Tanaka
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Yoshitaka Matsumoto
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
- Corresponding author. Proton Medical Research Center, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, 305-8576, Japan. Tel: +81-29-853-7100; Fax: +81-29-853-7103;
| | - Hitoshi Ishikawa
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Nobuyoshi Fukumitsu
- Department of Radiation Oncology, Kobe Proton Center, 1-6-8, Minatoshima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Haruko Numajiri
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Keiko Murofushi
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Yoshiko Oshiro
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Toshiyuki Okumura
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Toyomi Satoh
- Obstetrics & Gynecology of University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideyuki Sakurai
- Proton Medical Research Center, University of Tsukuba, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| |
Collapse
|
63
|
Samanci C, Onal Y, Sager S, Asa S, Ustabasioglu FE, Alis D, Akman C, Sonmezoglu K. Diagnostic Capabilities of MRI Versus 18F FDG PET-CT in Postoperative Patients with Thyroglobulin Positive, 131I-negative Local Recurrent or Metastatic Thyroid Cancer. Curr Med Imaging 2020; 15:956-964. [PMID: 32008523 DOI: 10.2174/1573405614666180718124739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND The detection of recurrence or metastasis might be challenging in patients, who underwent total thyroidectomy and radioactive iodine therapy for Differentiated Thyroid Carcinoma (DTC), with increased serum Thyroglobulin (Tg) levels and negative 131I whole body scan (131I-WBS) results. AIMS The purpose of this study was to compare the ability of Magnetic Resonance Imaging (MRI) and 18F-fluorodeoxyglucose Positron Emission Tomography/Computed Tomography (18F FDG PET-CT) to detect recurrence or cervical and upper mediastinal metastases in postoperative DTC patients who had negative 131I-WBS despite elevated serum Tg levels. STUDY DESIGN This study has a retrospective study design. METHODS We evaluated cervical and upper mediastinal MRI and 18F FDG PET-CT of 32 postoperative patients with DTC (26 patients with papillary thyroid carcinoma and 6 patients with follicular thyroid carcinoma). RESULTS We evaluated 44 lesions in 32 patients. For all lesions, the Positive Predictive Value, (PPV) Negative Predictive Value (NPV), sensitivity, specificity, and accuracy of MRI were 81.4%, 76.4%, 84.6%, 72.2%, and 79.5% respectively. The PPV, NPV, sensitivity, specificity, and accuracy of 18F FDG PET-CT were 100.0%, 85.7%, 88.4%, 100.0%, and 93.1%, respectively. CONCLUSION Although we could not replace 18F FDG PET-CT, MRI might be used as an adjunct to 18F FDG PET-CT for the evaluation of recurrent or cervical and upper mediastinal metastatic thyroid cancers; however, MRI is inadequate for the detection of metastases in small lymph nodes.
Collapse
Affiliation(s)
- Cesur Samanci
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yilmaz Onal
- Department of Radiology, İstanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sait Sager
- Department of Nuclear Medicine, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sertac Asa
- Department of Nuclear Medicine, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Fethi Emre Ustabasioglu
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Deniz Alis
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Canan Akman
- Department of Radiology, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Kerim Sonmezoglu
- Department of Nuclear Medicine, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
64
|
Ramanathan S, Tirumani SH, Ojili V. Nodal metastasis in gynecologic malignancies: Update on imaging and management. Clin Imaging 2020; 59:157-166. [DOI: 10.1016/j.clinimag.2019.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/25/2019] [Accepted: 11/14/2019] [Indexed: 01/18/2023]
|
65
|
mRECIST for HCC: Performance and novel refinements. J Hepatol 2020; 72:288-306. [PMID: 31954493 DOI: 10.1016/j.jhep.2019.09.026] [Citation(s) in RCA: 407] [Impact Index Per Article: 81.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023]
Abstract
In 2010, modified RECIST (mRECIST) criteria were proposed as a way of adapting the RECIST criteria to the particularities of hepatocellular carcinoma (HCC). We intended to overcome some limitations of RECIST in measuring tumour shrinkage with local and systemic therapies, and also to refine the assessment of progression that could be misinterpreted with conventional RECIST 1.1, due to clinical events related to the natural progression of chronic liver disease (development of ascites, enlargement of lymph nodes, etc.). mRECIST has served its purpose since being adopted or included in clinical practice guidelines (European, American and Asian) for the management of HCC; it has also been instrumental for assessing response and time-to-event endpoints in several phase II and III investigations. Nowadays, mRECIST has become the standard tool for measurement of radiological endpoints at early/intermediate stages of HCC. At advanced stages, guidelines recommend both methods. mRECIST has been proven to capture higher objective response rates in tumours treated with molecular therapies and those responses have shown to be independently associated with better survival. With the advent of novel treatment approaches (i.e. immunotherapy) and combination therapies there is a need to further refine and clarify some concepts around the performance of mRECIST. Similarly, changes in the landscape of standard of care at advanced stages of the disease are pointing towards progression-free survival as a potential primary endpoint in some phase III investigations, as effective therapies applied beyond progression might mask overall survival results. Strict recommendations for adopting this endpoint have been reported. Overall, we review the performance of mRECIST during the last decade, incorporating novel clarifications and refinements in light of emerging challenges in the study and management of HCC.
Collapse
|
66
|
Abstract
Background iRECIST for the objective monitoring of immunotherapies was published by the official RECIST working group in 2017. Main body Immune-checkpoint inhibitors represent one of the most important therapy advancements in modern oncology. They are currently used for treatment of multiple malignant diseases especially at advanced, metastatic stages which were poorly therapeutically accessible in the past. Promising results of recent studies suggest that their application will further grow in the near future, particularly when used in combination with chemotherapy. A challenging aspect of these immunotherapies is that they may show atypical therapy response patterns such as pseudoprogression and demonstrate a different imaging spectrum of adverse reactions, both of which are crucial for radiologists to understand. In 2017 the RECIST working group published a modified set of response criteria, iRECIST, for immunotherapy, based on RECIST 1.1 which was developed for cytotoxic therapies and adapted for targeted agents. Conclusion This article provides guidance for response assessment of oncologic patients under immunotherapy based on iRECIST criteria.
Collapse
Affiliation(s)
- Thorsten Persigehl
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
| | - Simon Lennartz
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University Cologne, Kerpener Straße 62, 50937, Cologne, Germany.,Else Kröner Forschungskolleg Clonal Evolution in Cancer, University Hospital Cologne, Weyertal 115b, 50931, Cologne, Germany
| | - Lawrence H Schwartz
- Department of Radiology, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, 10032, USA
| |
Collapse
|
67
|
Lalchandani UR, Sahai V, Hersberger K, Francis IR, Wasnik AP. A Radiologist's Guide to Response Evaluation Criteria in Solid Tumors. Curr Probl Diagn Radiol 2019; 48:576-585. [DOI: 10.1067/j.cpradiol.2018.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 01/09/2023]
|
68
|
Murez T, Fléchon A, Savoie PH, Rocher L, Camparo P, Morel-Journel N, Ferretti L, Sèbe P, Méjean A. [French ccAFU guidelines - Update 2018-2020: Testicular germ cell tumors]. Prog Urol 2019; 28 Suppl 1:R149-R166. [PMID: 31610870 DOI: 10.1016/j.purol.2019.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To update French guidelines concerning testicular germ cell cancer. METHODS Comprehensive Medline search between 2016 and 2018 upon diagnosis, treatment and follow-up of testicular germ cell cancer and treatments toxicities. Level of evidence was evaluated. RESULTS Testicular Germ cell tumor diagnosis is based on physical examination, biology tests (serum tumor markers AFP, hCGt, LDH) and radiological assessment (scrotal ultrasound and chest, abdomen and pelvis computerized tomography). Total inguinal orchiectomy is the first- line treatment allowing characterization of the histological type, local staging and identification of risk factors for micrometastases. In case of several therapeutic options, one must inform his patient balancing risks and benefits. Surveillance is usually chosen in stage I seminoma compliant patients as the evolution rate is low between 15 to 20 %. Carboplatin AUC7 is an alternative option. Radiotherapy indication should be avoided. In stage I non-seminomatous patients, either surveillance or risk-adapted strategy can be applied. Staging retroperitoneal lymphadenectomy has restricted indications. Metastatic germ cell tumors are usually treated by PEB chemotherapy according to IGCCCG prognostic classification. Lombo-aortic radiotherapy is still a standard treatment for stage IIA. Residual masses should be evaluated by biological and radiological assessment 3 to 4 weeks after the end of chemotherapy. Retroperitoneal lymphadenectomy is advocated for every non-seminomatous residual mass more than one cm. 18FDG uptake should be evaluated for each seminoma residual mass more than 3cm. CONCLUSIONS A rigorous use of classifications is mandatory to define staging since initial diagnosis. Applying treatments based on these classifications leads to excellent survival rates (99 % in CSI, 85 % in CSII+).
Collapse
Affiliation(s)
- T Murez
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, CHRU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34295 Montpellier cedex 5, France.
| | - A Fléchon
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'oncologie médicale, centre Léon-Bérard, 28, rue Laennec, 69008 Lyon, France
| | - P-H Savoie
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, hôpital d'instruction des armées Sainte-Anne, BP 600, 83800 Toulon cedex 09, France
| | - L Rocher
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service de radiologie, CHU Paris Sud, site Kremlin-Bicêtre, AP-HP, 94270 Le Kremlin-Bicêtre, France
| | - P Camparo
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Centre de pathologie, 51, rue de Jeanne-D'Arc, 80000 Amiens, France
| | - N Morel-Journel
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, centre hospitalier Lyon Sud (Pierre Bénite), HCL groupement hospitalier du Sud, 69495 Pierre Bénite cedex, France
| | - L Ferretti
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, MSP de Bordeaux-Bagatelle, 203, route de Toulouse, BP 50048, 33401 Talence cedex, France
| | - P Sèbe
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, groupe hospitalier Diaconesses Croix Saint-Simon, 125, rue d'Avron, 75020 Paris, France
| | - A Méjean
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, hôpital européen Georges-Pompidou, université Paris Descartes, AP-HP, 75015 Paris, France
| |
Collapse
|
69
|
Role of lymphadenectomy in endometrial cancer with nonbulky lymph node metastasis: Comparison of comprehensive surgical staging and sentinel lymph node algorithm. Gynecol Oncol 2019; 155:177-185. [PMID: 31604668 DOI: 10.1016/j.ygyno.2019.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To compare survival and progression outcomes between 2 nodal assessment approaches in patients with nonbulky stage IIIC endometrial cancer (EC). METHODS Patients with stage IIIC EC treated at 2 institutions were retrospectively identified. At 1 institution, a historical series (2004-2008) was treated with systematic pelvic and para-aortic lymphadenectomy (LND cohort). At the other institution, more contemporary patients (2006-2013) were treated using a sentinel lymph node algorithm (SLN cohort). Outcomes (hazard ratios [HRs]) within the first 5 years after surgery were compared between cohorts using Cox models adjusted for type of adjuvant therapy. RESULTS The study included 104 patients (48 LND, 56 SLN). The use of chemoradiotherapy was similar in the 2 cohorts (46% LND vs 50% SLN), but the use of chemotherapy alone (19% vs 36%) or radiotherapy alone (15% vs 2%) differed. Although there was evidence of higher risk of cause-specific death (HR, 2.10; 95% CI, 0.79-5.58; P = 0.14) and lower risk of para-aortic progression (HR, 0.27; 95% CI, 0.05-1.42; P = 0.12) for the LND group, the associations did not meet statistical significance. The risk of progression was not significantly different between the groups (HR, 1.27; 95% CI, 0.60-2.67; P =0 .53). In parsimonious multivariable models, high-risk tumor characteristics and nonendometrioid type were independently associated with lower cause-specific survival and progression-free survival. CONCLUSIONS In EC patients with nonbulky positive lymph nodes, use of the SLN algorithm with limited nodal dissection does not compromise survival compared with LND. Aggressive pathologic features of the primary tumor are the strongest determinants of prognosis.
Collapse
|
70
|
Watanabe M, Nakamoto Y, Nakamoto R, Ishimori T, Saga T, Togashi K. Performance Evaluation of a Newly Developed MR-Compatible Mobile PET Scanner with Two Detector Layouts. Mol Imaging Biol 2019; 22:407-415. [PMID: 31222564 DOI: 10.1007/s11307-019-01384-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE A mobile positron emission tomography (PET) scanner called flexible PET (fxPET), designed to fit existing magnetic resonance imaging (MRI) or computed tomography (CT) system, has been developed. The purpose of this study was to evaluate the image quality, lesion detection rate, and quantitative values of fxPET compared with conventional bismuth germanium oxide (BGO)-based PET/CT without time-of-flight capability. PROCEDURES Fifty-nine patients underwent whole-body (WB) PET/CT scans approximately 1 h after injection of 2-deoxy-2-[18F]fluoro-D-glucose, followed by the fxPET scans with detectors located above and below the patients (layout A) and with detectors closer to the patients (layout B). Two readers assessed the image quality using a 4-point grade for each layout and reached a consensus. We evaluated the differences and/or correlations between fxPET and WB PET/CT, including the lesion detection rates, the standardized uptake value (SUV), the metabolic tumor volume (MTV), the total lesion glycolysis (TLG), the tumor-to-normal liver ratio (TLR), and the background liver signal-to-noise ratio (SNR). RESULTS The image quality of layout B was better than layout A (p < 0.0001). Of 184 lesions, the detection rate of layout B was significantly higher than WB PET/CT (p = 0.041), while the detection rate of layout A was comparable to WB PET/CT. The SUVmax/mean/peak were larger, and the MTVs were smaller in fxPET than WB PET/CT, especially in the lesions smaller than 2 cm (p < 0.01). The SUVmax/mean/peak, the MTVs and the TLGs of fxPET had significant positive correlations with WB PET/CT (p < 0.0001). The TLRs were significantly larger (p < 0.0001), but the background SNRs were significantly lower in fxPET than WB PET/CT (p < 0.05). CONCLUSIONS The fxPET system yielded reasonable image quality and quantitative accuracy. Bringing the detectors closer to the patient yielded improved results.
Collapse
Affiliation(s)
- Masao Watanabe
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Ryusuke Nakamoto
- Department of Radiology, Shiga General Hospital, 5-4-30 Moriyama, Moriyama, Shiga, 524-8524, Japan
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tsuneo Saga
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
71
|
Šarenac T, Mikov M. Cervical Cancer, Different Treatments and Importance of Bile Acids as Therapeutic Agents in This Disease. Front Pharmacol 2019; 10:484. [PMID: 31214018 PMCID: PMC6558109 DOI: 10.3389/fphar.2019.00484] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/16/2019] [Indexed: 01/15/2023] Open
Abstract
Cervical cancer can be cured, because it has a long preinvasive period. Early diagnosis and treatment of cervical cancer at women are crucial for reducing of rate mortality. Today, there are many methods for detecting premalignant lesions and one of them is a conventional Papanicolaou test. Cervical cancer develops through a series of changes in the epithelium called cervical intraepithelial neoplasia (CIN). The biological and genetic characteristics of the cells at cancer in situ are irreversibly altered and abnormal cells have the potential to metastasize to others anatomical regions. Infection with human Papillomavirus, which is transmitted sexually, is considered the main cause and represent the necessary, but not the only factor for the development of cervical cancer. Types of high risk human Papillomavirus are often associated with invasive cervical cancer. The carcinogenic types of HPV 16 and 18 are responsible for 70% of cervical cancer and about 50% of CIN 3. Primary prevention of cervical cancer is aimed at reducing incidence, control of causes and risk factors. In this scientific work, in addition to explaining the various treatments necessary for the treatment of cervical carcinoma, we were discussed about the anticancer effects of the synthetic derivative of ursodeoxycholic acid, such as HS-1183, and synthetic derivatives of chenodeoxycholic acid such as HS-1199 and HS-1200. Also, the effects of bile acid complexes with metals such as platinum, zinc, nickel, and copper were considered in the effective treatment of cervical cancer. KEY POINTS • Lymphogenic spreading of cervical cancer occurs relatively early in the regional lymph nodes, while this sort of progression of cervical cancer is rarer in the juxtaregional (paraaortic), mediastinal and supraclavicular nodes. Clinically proven supraclavicular metastases are not a rarity. In stages IIb and IIIa with metastases in paraaortal nodes occur a 20% metastases at the neck lymph nodes. Hematogenic metastases are relatively rare and occur in the posterior phase. Distant metastases are detected in the lungs and liver. Preinvasive and microinvasive stages of cervical cancer are without symptoms. With deeper invasion of the strome, certain clinical symptoms such as prolonged menstruation, increased vaginal secretions, vaginal bleeding between the two periods, contact bleeding (after coitus), unilateral pelvic pain with spreading in hip joint (infiltration of the pelvic nerve plexus), dysuric disturbance, anemia, islet of the lower extremities. In order to diagnose the level spreading of primary lesion of cervical cancer most commonly are used the supplemental searches such as cytoscopy, rectoscopy, urography, irigography, lung and bone radiography, scintigraphy of the liver, kidney and bone, lymphography, CT (MR) of abdomen and pelvis, as well as laboratory analysis. Surgical treatment consists of transvaginal hysterectomy, transabdominal removal of the uterus (via laparotomy), bilateral adenectomy (removal of the ovaries and the fallopian tubes), upper and middle third of the vagina and lymphonodectomy of the regional lymph nodes. The most commonly used radiotherapy, intracavitary brachytherapy, manual afterloading technique and remote afterloading techniques. The synthetic derivatives of ursodeoxycholic acid and chenodeoxycholic acid such as HS-1183, HS-1199, and HS-1200 are used to treat cervical cancer. These derivatives of chenodeoxycholic acid and ursodeoxycholic acid are capable of inhibiting cell proliferation and inducing apoptosis in SiHa human cells of cervix. Platinum compounds are used as catalysts in cervical cancer therapy. Clinical use of platinum complexes for which the bile acids bind is based on the desire to achieve the death of tumor cells and the spectrum of drug activity in the treatment of cervical cancer. Bisursodeoxycholate (ethylenediamine) platinum (II) [Pt(UDC)2(en)] is characterized by important cytotoxicity against HeLa cervical carcinoma cells and this effect already being clearly detectable after 24 h.
Collapse
Affiliation(s)
- Tanja Šarenac
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | | |
Collapse
|
72
|
Zheng XH, Zhang W, Yang L, Du CX, Li N, Xing GS, Tian YT, Xie YB. Role of D2 gastrectomy in gastric cancer with clinical para-aortic lymph node metastasis. World J Gastroenterol 2019; 25:2338-2353. [PMID: 31148905 PMCID: PMC6529887 DOI: 10.3748/wjg.v25.i19.2338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Owing to the technical difficulty of pathological diagnosis, imaging is still the most commonly used method for clinical diagnosis of para-aortic lymph node metastasis (PALM) and evaluation of therapeutic effects in gastric cancer, which leads to inevitable false-positive findings in imaging. Patients with clinical PALM may have entirely different pathological stages (stage IV or not), which require completely different treatment strategies. There is no consensus on whether surgical intervention should be implemented for this group of patients. In particular, the value of D2 gastrectomy in a multidisciplinary treatment (MDT) approach for advanced gastric cancer with clinical PALM remains unknown. AIM To investigate the value of D2 gastrectomy in a MDT approach for gastric cancer patients with clinical PALM. METHODS In this real-world study, clinico-pathological data of all gastric cancer patients treated at the Cancer Hospital, Chinese Academy of Medical Sciences between 2011 and 2016 were reviewed to identify those with clinically enlarged PALM. All the clinico-pathological data were prospectively documented in the patient medical record. For all the gastric cancer patients with advanced stage disease, especially those with suspicious distant metastasis, the treatment methods were determined by a multidisciplinary team. RESULTS In total, 48 of 7077 primary gastric cancer patients were diagnosed as having clinical PALM without other distant metastases. All 48 patients received chemotherapy as the initial treatment. Complete or partial response was observed in 39.6% (19/48) of patients in overall and 52.1% (25/48) of patients in the primary tumor. Complete response of PALM was observed in 50.0% (24/48) of patients. After chemotherapy, 45.8% (22/48) of patients received D2 gastrectomy, and 12.5% (6/48) of patients received additional radiotherapy. The postoperative major complication rate and mortality were 27.3% (6/22) and 4.5% (1/22), respectively. The median overall survival and progression-free survival of all the patients were 18.9 and 12.1 mo, respectively. The median overall survival of patients who underwent surgical resection or not was 50.7 and 12.8 mo, respectively. The 3-year and 5-year survival rates were 56.8% and 47.3%, respectively, for patients who underwent D2 resection. Limited PALM and complete response of PALM after chemotherapy were identified as favorable factors for D2 gastrectomy. CONCLUSION For gastric cancer patients with radiologically suspicious PALM that responds well to chemotherapy, D2 gastrectomy could be a safe and effective treatment and should be adopted in a MDT approach for gastric cancer with clinical PALM.
Collapse
Affiliation(s)
- Xiao-Hao Zheng
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chun-Xia Du
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Gu-Sheng Xing
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan-Tao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yi-Bin Xie
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
73
|
Yoon JH, Yoon SH, Hahn S. Development of an algorithm for evaluating the impact of measurement variability on response categorization in oncology trials. BMC Med Res Methodol 2019; 19:90. [PMID: 31046712 PMCID: PMC6498480 DOI: 10.1186/s12874-019-0727-7] [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: 09/03/2018] [Accepted: 04/09/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Radiologic assessments of baseline and post-treatment tumor burden are subject to measurement variability, but the impact of this variability on the objective response rate (ORR) and progression rate in specific trials has been unpredictable on a practical level. In this study, we aimed to develop an algorithm for evaluating the quantitative impact of measurement variability on the ORR and progression rate. METHODS First, we devised a hierarchical model for estimating the distribution of measurement variability using a clinical trial dataset of computed tomography scans. Next, a simulation method was used to calculate the probability representing the effect of measurement errors on categorical diagnoses in various scenarios using the estimated distribution. Based on the probabilities derived from the simulation, we developed an algorithm to evaluate the reliability of an ORR (or progression rate) (i.e., the variation in the assessed rate) by generating a 95% central range of ORR (or progression rate) results if a reassessment was performed. Finally, we performed validation using an external dataset. In the validation of the estimated distribution of measurement variability, the coverage level was calculated as the proportion of the 95% central ranges of hypothetical second readings that covered the actual burden sizes. In the validation of the evaluation algorithm, for 100 resampled datasets, the coverage level was calculated as the proportion of the 95% central ranges of ORR results that covered the ORR from a real second assessment. RESULTS We built a web tool for implementing the algorithm (publicly available at http://studyanalysis2017.pythonanywhere.com/ ). In the validation of the estimated distribution and the algorithm, the coverage levels were 93 and 100%, respectively. CONCLUSIONS The validation exercise using an external dataset demonstrated the adequacy of the statistical model and the utility of the developed algorithm. Quantification of variation in the ORR and progression rate due to potential measurement variability is essential and will help inform decisions made on the basis of trial data.
Collapse
Affiliation(s)
- Jeong-Hwa Yoon
- Interdisciplinary Program in Medical Informatics, Seoul National University College of Medicine, Seoul, South Korea
| | - Soon Ho Yoon
- Department of Radiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Seokyung Hahn
- Medical Statistics Laboratory, Department of Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
74
|
Persigehl T, Poeppel TD, Sedlaczek O. [Radiological response assessment of modern immunotherapy using iRECIST]. Radiologe 2019; 57:826-833. [PMID: 28812098 DOI: 10.1007/s00117-017-0289-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CLINICAL/METHODICAL ISSUE Modern immunotherapies in oncology show tumor response patterns differing from conventional chemotherapies including initial pseudo-progression. STANDARD RADIOLOGICAL METHODS Response evaluation criteria in solid tumors (RECIST 1.1) represent the currently most used response criteria for conventional chemotherapy of solid tumors. However, atypical response patterns of immunotherapies are not correctly classified using RECIST 1.1 so that the effectiveness is also incorrectly interpreted. METHODICAL INNOVATIONS In order to correctly interpret these atypical response patterns, special immune-related response criteria in solid tumors (iRECIST) have been published. In contrast to RECIST 1.1 according to iRECIST an initially unconfirmed progressive disease (iUPD) requires confirmation (iCPD) in clinically stable patients by subsequent control imaging after 4-8 weeks. New lesions are separately assessed within iRECIST. PERFORMANCE The iRECIST procedure allows a standardized objective assessment of a possible pseudo-progression which can occur in up to 10% of cases depending on the immunomodulating drug and tumor entity. ACHIEVEMENTS In principle, iRECIST was developed only for usage in trials testing modern immunotherapeutics. PRACTICAL RECOMMENDATIONS The iRECIST procedure might also be helpful as an additional objective response criterium for clinical treatment decisions, taking the limitations into account.
Collapse
Affiliation(s)
- T Persigehl
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - T D Poeppel
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122, Essen, Deutschland
| | - O Sedlaczek
- Abteilung Diagnostische und Interventionelle Radiologie, NCT Heidelberg (DKFZ & Universitätsklinikum HD), Im Neuenheimer Feld 460, 69120, Heidelberg, Deutschland
| |
Collapse
|
75
|
Semantic segmentation and detection of mediastinal lymph nodes and anatomical structures in CT data for lung cancer staging. Int J Comput Assist Radiol Surg 2019; 14:977-986. [DOI: 10.1007/s11548-019-01948-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022]
|
76
|
Freedman RA, Gelman RS, Anders CK, Melisko ME, Parsons HA, Cropp AM, Silvestri K, Cotter CM, Componeschi KP, Marte JM, Connolly RM, Moy B, Van Poznak CH, Blackwell KL, Puhalla SL, Jankowitz RC, Smith KL, Ibrahim N, Moynihan TJ, O'Sullivan CC, Nangia J, Niravath P, Tung N, Pohlmann PR, Burns R, Rimawi MF, Krop IE, Wolff AC, Winer EP, Lin NU. TBCRC 022: A Phase II Trial of Neratinib and Capecitabine for Patients With Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. J Clin Oncol 2019; 37:1081-1089. [PMID: 30860945 PMCID: PMC6494354 DOI: 10.1200/jco.18.01511] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Evidence-based treatments for metastatic, human epidermal growth factor receptor 2 (HER2)-positive breast cancer to the CNS are limited. We previously reported modest activity of neratinib monotherapy for HER2-positive breast cancer brain metastases. Here we report the results from additional study cohorts. PATIENTS AND METHODS Patients with measurable, progressive, HER2-positive brain metastases (92% after receiving CNS surgery and/or radiotherapy) received neratinib 240 mg orally once per day plus capecitabine 750 mg/m2 twice per day for 14 days, then 7 days off. Lapatinib-naïve (cohort 3A) and lapatinib-treated (cohort 3B) patients were enrolled. If nine or more of 35 (cohort 3A) or three or more of 25 (cohort 3B) had CNS objective response rates (ORR), the drug combination would be deemed promising. The primary end point was composite CNS ORR in each cohort separately, requiring a reduction of 50% or more in the sum of target CNS lesion volumes without progression of nontarget lesions, new lesions, escalating steroids, progressive neurologic signs or symptoms, or non-CNS progression. RESULTS Forty-nine patients enrolled in cohorts 3A (n = 37) and 3B (n = 12; cohort closed for slow accrual). In cohort 3A, the composite CNS ORR = 49% (95% CI, 32% to 66%), and the CNS ORR in cohort 3B = 33% (95% CI, 10% to 65%). Median progression-free survival was 5.5 and 3.1 months in cohorts 3A and 3B, respectively; median survival was 13.3 and 15.1 months. Diarrhea was the most common grade 3 toxicity (29% in cohorts 3A and 3B). Neratinib plus capecitabine is active against refractory, HER2-positive breast cancer brain metastases, adding additional evidence that the efficacy of HER2-directed therapy in the brain is enhanced by chemotherapy. For optimal tolerance, efforts to minimize diarrhea are warranted.
Collapse
Affiliation(s)
| | | | - Carey K Anders
- 2 University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | | | | | | | | | - Beverly Moy
- 5 Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | - Nuhad Ibrahim
- 9 The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | - Nadine Tung
- 12 Beth Israel Deaconess Medical Center, Boston, MA
| | | | | | | | - Ian E Krop
- 1 Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | | |
Collapse
|
77
|
Downstaging of lymph node metastasis after neoadjuvant intraperitoneal and systemic chemotherapy in gastric carcinoma with peritoneal metastasis. Eur J Surg Oncol 2019; 45:1493-1497. [PMID: 30948161 DOI: 10.1016/j.ejso.2019.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/18/2018] [Accepted: 03/06/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The aim of the present study was to evaluate the clinical features and prognosis of lymph node metastasis (LNM) in gastric cancer patients with peritoneal metastasis (PM) after neoadjuvant intraperitoneal and systemic chemotherapy. METHODS A total of 69 gastric cancer patients with PM and LNM who received neoadjuvant intraperitoneal and systemic chemotherapy (NIPS) of intraperitoneal docetaxel (DXT) and cisplatin (CDDP); intravenous chemotherapy of DXT and CDDP and oral S-1in Kishiwada Tokushukai Hospital between January 2008 and February 2017. After surgical resection, the response of LNMs was studied to confirm the effect of NIPS on LNMs. RESULTS After NIPS, 197 lymph nodes (LNs) (42.5%) were graded as G3, the progression in LNMs were significantly better than in the primary tumors. Until the last follow-up, 1-year overall survival rate was 82.6%, and the median survival period was 22.0 ± 3.7 months. In the group of patients who had achieved a more than 50% G3 grade of the response of LNMs, the median survival period is 38 months; in the less than 50% G3 grade group, it is 14 months, that is a significantly different result. Multivariate analyses showed that the factors PCI, Post-therapeutic N status and response of the LNMs were found to be as independent prognostic factors. CONCLUSION Downstaging of LNMs were achieved in patients of gastric cancer with PM who received NIPS. Downstaging of LNMs after NIPS is related with the prognosis of gastric cancer and should be valued in subsequent surgery for gastric cancer with peritoneal and lymph nodes metastasis.
Collapse
|
78
|
Feng QX, Liu C, Qi L, Sun SW, Song Y, Yang G, Zhang YD, Liu XS. An Intelligent Clinical Decision Support System for Preoperative Prediction of Lymph Node Metastasis in Gastric Cancer. J Am Coll Radiol 2019; 16:952-960. [PMID: 30733162 DOI: 10.1016/j.jacr.2018.12.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE The aim of this study was to develop and validate a computational clinical decision support system (DSS) on the basis of CT radiomics features for the prediction of lymph node (LN) metastasis in gastric cancer (GC) using machine learning-based analysis. METHODS Clinicopathologic and CT imaging data were retrospectively collected from 490 patients who were diagnosed with GC between January 2002 and December 2016. Radiomics features were extracted from venous-phase CT images. Relevant features were selected, ranked, and modeled using a support vector machine classifier in 326 training and validation data sets. A model test was performed independently in a test set (n = 164). Finally, a head-to-head comparison of the diagnostic performance of the DSS and that of the conventional staging criterion was performed. RESULTS Two hundred ninety-seven of the 490 patients examined had histopathologic evidence of LN metastasis, yielding a 60.6% metastatic rate. The area under the curve for predicting LN+ was 0.824 (95% confidence interval, 0.804-0.847) for the DSS in the training and validation data and 0.764 (95% confidence interval, 0.699-0.833) in the test data. The calibration plots showed good concordance between the predicted and observed probability of LN+ using the DSS approach. The DSS was better able to predict LN metastasis than the conventional staging criterion in the training and validation data (accuracy 76.4% versus 63.5%) and in the test data (accuracy 71.3% versus 63.2%) CONCLUSIONS: A DSS based on 13 "worrisome" radiomics features appears to be a promising tool for the preoperative prediction of LN status in patients with GC.
Collapse
Affiliation(s)
- Qiu-Xia Feng
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Chang Liu
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Liang Qi
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Shu-Wen Sun
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Yang Song
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China
| | - Yu-Dong Zhang
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| | - Xi-Sheng Liu
- Department of Radiology, First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| |
Collapse
|
79
|
Prediction of Lymph Node Maximum Standardized Uptake Value in Patients With Cancer Using a 3D Convolutional Neural Network: A Proof-of-Concept Study. AJR Am J Roentgenol 2019; 212:238-244. [DOI: 10.2214/ajr.18.20094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
80
|
Chen CF, Zhang YL, Cai ZL, Sun SM, Lu XF, Lin HY, Liang WQ, Yuan MH, Zeng D. Predictive Value of Preoperative Multidetector-Row Computed Tomography for Axillary Lymph Nodes Metastasis in Patients With Breast Cancer. Front Oncol 2019; 8:666. [PMID: 30671386 PMCID: PMC6331431 DOI: 10.3389/fonc.2018.00666] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 12/17/2018] [Indexed: 02/05/2023] Open
Abstract
Introduction: Axillary lymph nodes (ALN) status is an essential component in tumor staging and treatment planning for patients with breast cancer. The aim of present study was to evaluate the predictive value of preoperative multidetector-row computed tomography (MDCT) for ALN metastasis in breast cancer patients. Methods: A total of 148 cases underwent preoperative MDCT examination and ALN surgery were eligible for the study. Logistic regression analysis of MDCT variates was used to estimate independent predictive factors for ALN metastasis. The prediction of ALN metastasis was determined with MDCT variates through receiver operating characteristic (ROC) analysis. Results: Among the 148 cases, 61 (41.2%) cases had ALN metastasis. The cortical thickness in metastatic ALN was significantly thicker than that in non-metastatic ALN (7.5 ± 5.0 mm vs. 2.6 ± 2.8 mm, P < 0.001). Multi-logistic regression analysis indicated that cortical thickness of >3 mm (OR: 12.32, 95% CI: 4.50–33.75, P < 0.001) and non-fatty hilum (OR: 5.38, 95% CI: 1.51–19.19, P = 0.009) were independent predictors for ALN metastasis. The sensitivity, specificity and AUC of MDCT for ALN metastasis prediction based on combined-variated analysis were 85.3%, 87.4%, and 0.893 (95% CI: 0.832–0.938, P < 0.001), respectively. Conclusions: Cortical thickness (>3 mm) and non-fatty hilum of MDCT were independent predictors for ALN metastasis. MDCT is a potent imaging tool for predicting ALN metastasis in breast cancer. Future prospective study on the value of contrast enhanced MDCT in preoperative ALN evaluation is warranted.
Collapse
Affiliation(s)
- Chun-Fa Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yu-Ling Zhang
- Department of Information, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ze-Long Cai
- Department of Medical Imaging, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Shu-Ming Sun
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xiao-Feng Lu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hao-Yu Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wei-Quan Liang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ming-Heng Yuan
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - De Zeng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
81
|
Beaumont H, Evans TL, Klifa C, Guermazi A, Hong SR, Chadjaa M, Monostori Z. Discrepancies of assessments in a RECIST 1.1 phase II clinical trial - association between adjudication rate and variability in images and tumors selection. Cancer Imaging 2018; 18:50. [PMID: 30537991 PMCID: PMC6288919 DOI: 10.1186/s40644-018-0186-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 12/04/2018] [Indexed: 02/05/2023] Open
Abstract
Background In imaging-based clinical trials, it is common practice to perform double reads for each image, discrepant interpretations can result from these two different evaluations. In this study we analyzed discrepancies that occurred between local investigators (LI) and blinded independent central review (BICR) by comparing reader-selected imaging scans and lesions. Our goal was to identify the causes of discrepant declarations of progressive disease (PD) between LI and BICR in a clinical trial. Methods We retrospectively analyzed imaging data from a RECIST 1.1-based, multi-sites, phase II clinical trial of 179 patients with adult small cell lung cancer, treated with Cabazitaxel compared to Topotecan. Any discrepancies in the determination of PD between LI and BICR readers were reviewed by a third-party adjudicator. For each imaging time point and reader, we recorded the selected target lesions, non-target lesions, and new lesions. Odds ratios were calculated to measure the association between discrepant declarations of PD and the differences in reviewed imaging scans (e.g. same imaging modality but with different reconstruction parameters) and selected lesions. Reasons for discrepancies were analyzed. Results The average number of target lesions found by LI and BICR was respectively 2.9 and 3.4 per patient (p < 0.05), 18.4% of these target lesions were actually non-measurable. LI and BICR performed their evaluations based on different baseline imaging scans for 59% of the patients, they selected at least one different target lesion in 85% of patients. A total of 36.7% of patients required adjudication. Reasons of adjudication included differences in 1) reporting new lesions (53.7%), 2) the measured change of the tumor burden (18.5%), and 3) the progression of non-target lesions (11.2%). The rate of discrepancy was not associated with the selection of non-measurable target lesions or with the readers’ assessment of different images. Paradoxically, more discrepancies occurred when LI and BICR selected exactly the same target lesions at baseline compared to when readers selected not exactly the same lesions. Conclusions For a large proportion of evaluations, LI and BICR did not select the same imaging scans and target lesions but with a limited impact on the rate of discrepancy. The majority of discrepancies were explained by the difference in detecting new lesions. Trial Registration ARD12166 (https://clinicaltrials.gov/ct2/show/NCT01500720).
Collapse
Affiliation(s)
- Hubert Beaumont
- Research & Clinical Development, Median Technologies, Les deux arcs - 1800 route des crêtes - Bat, B 06560, Valbonne, France.
| | - Tracey L Evans
- Department of medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Catherine Klifa
- Research & Clinical Development, Median Technologies, Les deux arcs - 1800 route des crêtes - Bat, B 06560, Valbonne, France
| | - Ali Guermazi
- Quantitative Imaging Center (QIC) Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sae Rom Hong
- Department of Radiology, Severance Hospital Yonsei University of Medicine, Seoul, South Korea
| | | | - Zsuzsanna Monostori
- Radiology, National Koranyi Institute of TB and pulmonology, Budapest, H-1121, Hungary
| |
Collapse
|
82
|
Abstract
Cross-sectional spinal imaging is common, and extraspinal findings are often incidentally identified during interpretation. Although some of these findings may cause symptoms that mimic a spinal disorder, the majority are entirely asymptomatic and incidental. It is essential that the radiologist not only identify those abnormalities that may have clinical significance but also recognize those that are clinically irrelevant and thereby prevent patients from being subjected to further unnecessary, expensive and potentially harmful interventions. This article focuses on those abnormalities that are commonly encountered and provides practical guidance for follow-up and management based on current recommendations.
Collapse
Affiliation(s)
- Prashant Raghavan
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 655 W. Baltimore St, Baltimore, MD 21201, USA.
| | - Jessica Record
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 655 W. Baltimore St, Baltimore, MD 21201, USA
| | - Lorenna Vidal
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 655 W. Baltimore St, Baltimore, MD 21201, USA
| |
Collapse
|
83
|
Pattern and impact of metastatic cardiophrenic lymph nodes in advanced epithelial ovarian cancer. Gynecol Oncol 2018; 152:76-81. [PMID: 30463683 DOI: 10.1016/j.ygyno.2018.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Cardiophrenic lymph nodes (CPLN) define FIGO stage IVB disease. We evaluate the pattern of CPLN metastases, their prognostic impact and the potential role of CPLN resection in patients with epithelial ovarian cancer (EOC). METHODS Analysis of 595 consecutive patients with EOC treated in the period 01/2011-05/2016. CT scans were re-reviewed by two radiologists. Positive CPLN were defined as ≥5 mm in the short-axis diameter. The role of CPLN resection was evaluated in a case-control matched-pair analysis. RESULTS Of 595 patients 458 had FIGO stage IIIB-IV disease. We excluded patients undergoing interval surgery (n = 54), without debulking surgery (n = 32) and without sufficient pre-operative imaging (n = 22), resulting in a study cohort of 350 patients. Of these, 133 (37.9%) had negative CPLN and 217 (62.0%) had radiologically positive CPLN. In patients with postoperative residual tumor, enlarged CPLN had no impact on survival. In patients with complete resection (n = 223), 98 (44.0%) had negative CPLN and a 5-year OS of 69% and a 5-year PFS of 41%; in contrast, in the 125 patients (56.0%) with positive CPLN, 5-year OS was 30% and 5-year PFS was 13%. In 52 patients we resected CPLN. The matched-pair case-control analysis did not demonstrate any significant impact on survival of CPLN resection. CONCLUSION CPLN metastases are associated with impaired PFS and OS in patients with macroscopically completely resected tumor. Intraabdominal residual tumor has a greater prognostic impact than positive CPLN. The impact of the resection of CPLN remains unclear.
Collapse
|
84
|
Abstract
PURPOSE The aim of this study was to determine the prognostic significance of PET/CT findings in women with cervical cancer and describe the normalization of lymph node SUVmax (nSUVmax). MATERIALS AND METHODS A retrospective review was performed of 113 patients with cervical cancer who underwent a PET/CT before receiving definitive therapy. SUVmax measurements were normalized to the SUV of the pelvic blood pool. Patient, tumor, and PET/CT data were correlated to extracervical recurrence-free survival (ecRFS) and lymph node pathology. RESULTS Of 113 patients, there were 23 (20%) extracervical recurrences. On univariate analysis, stage, histology, nSUVmax, and radiographic size of the primary tumor, and nSUVmax of the most hypermetabolic lymph node were significantly associated with ecRFS. On multivariable analysis, nSUVmax and radiographic size of the primary tumor remained associated with ecRFS (both P < 0.001). Sixty-six patients underwent pelvic, common iliac, and/or para-aortic nodal sampling. The sensitivity, specificity, false-negative, and false-positive rates of PET/CT for lymph node metastases were 53%, 75%, 6%, and 82%, respectively. On univariate analysis, nSUVmax, and radiographic size of the primary tumor, and nSUVmax of the most hypermetabolic lymph node, and radiographic size of the largest lymph node, were associated with the presence of at least one pathologically positive lymph node. On multivariable analysis, only the radiographic size of the largest lymph node remained significantly associated with lymph node metastases (P < 0.001). CONCLUSIONS The size and nSUVmax of the primary tumor were associated with ecRFS. PET/CT has a low false-negative rate but high false-positive rate for lymph node metastases.
Collapse
|
85
|
Murez T, Fléchon A, Savoie PH, Rocher L, Camparo P, Morel-Journel N, Ferretti L, Sèbe P, Méjean A. RETRACTED: Recommandations françaises du Comité de Cancérologie de l’AFU — Actualisation 2018—2020 : tumeurs germinales du testicule French ccAFU guidelines — Update 2018—2020: Testicular germ cell tumors. Prog Urol 2018; 28:S147-S164. [PMID: 30472999 DOI: 10.1016/j.purol.2018.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 11/30/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy).
Cet article est retiré de la publication à la demande des auteurs car ils ont apporté des modifications significatives sur des points scientifiques après la publication de la première version des recommandations.
Le nouvel article est disponible à cette adresse: doi:10.1016/j.purol.2019.01.009.
C’est cette nouvelle version qui doit être utilisée pour citer l’article.
This article has been retracted at the request of the authors, as it is not based on the definitive version of the text because some scientific data has been corrected since the first issue was published.
The replacement has been published at the doi:10.1016/j.purol.2019.01.009.
That newer version of the text should be used when citing the article.
Collapse
Affiliation(s)
- T Murez
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, CHRU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34295 Montpellier cedex 5, France.
| | - A Fléchon
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'oncologie médicale, centre Léon-Bérard, 28, rue Laennec, 69008 Lyon, France
| | - P-H Savoie
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, hôpital d'instruction des armées Sainte-Anne, BP 600, 83800 Toulon cedex 09, France
| | - L Rocher
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service de radiologie, CHU Paris Sud, site Kremlin-Bicêtre, AP-HP, 94270 Le Kremlin-Bicêtre, France
| | - P Camparo
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Centre de pathologie, 51, rue de Jeanne-D'Arc, 80000 Amiens, France
| | - N Morel-Journel
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, centre hospitalier Lyon Sud (Pierre Bénite), HCL groupement hospitalier du Sud, 69495 Pierre Bénite cedex, France
| | - L Ferretti
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, MSP de Bordeaux-Bagatelle, 203, route de Toulouse, BP 50048, 33401 Talence cedex, France
| | - P Sèbe
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, groupe hospitalier Diaconesses Croix Saint-Simon, 125, rue d'Avron, 75020 Paris, France
| | - A Méjean
- Comité de cancérologie de l'Association française d'urologie, groupe organes génitaux externes, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, hôpital européen Georges-Pompidou, université Paris Descartes, AP-HP, 75015 Paris, France
| |
Collapse
|
86
|
Soyama H, Miyamoto M, Takano M, Iwahashi H, Kato K, Sakamoto T, Kuwahara M, Ishibashi H, Matuura H, Yoshikawa T, Aoyama T, Tsuda H, Furuya K. A Pathological Study Using 2014 WHO Criteria Reveals Poor Prognosis of Grade 3 Ovarian Endometrioid Carcinomas. ACTA ACUST UNITED AC 2018; 32:597-602. [PMID: 29695566 DOI: 10.21873/invivo.11281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/10/2018] [Accepted: 02/13/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM The purpose of this study was to compare the clinical behavior of several grades of endometrioid carcinoma (EC) compared to high-grade serous carcinoma (HGSC), based on World Health Organization 2014 criteria. MATERIALS AND METHODS Clinicopathological features were compared between all grades of EC and HGSC, and between HGSC and either grade 1/2 or grade 3 EC. RESULTS Sixty-five patients with EC and 214 with HGSC were identified. Among patients with EC, 56 displayed 1/2 EC and nine had grade 3 EC. The progression-free (PFS) and overall (OS) survival of patients with grade 1/2 EC were better than of those of patients with HGSC; however, PFS and OS did not statistically differ between patients with grade 3 EC and those with HGSC. Grade 1/2 EC, but not grade 3, was a better prognostic factor compared with HGSC. CONCLUSION A grading system for EC would be beneficial for the accurate prognosis of ovarian cancer.
Collapse
Affiliation(s)
- Hiroaki Soyama
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Morikazu Miyamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Masashi Takano
- Department of Clinical Oncology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hideki Iwahashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Kento Kato
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Takahiro Sakamoto
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Mika Kuwahara
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hiroki Ishibashi
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hiroko Matuura
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Tomoyuki Yoshikawa
- Department of Clinical Oncology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Tadashi Aoyama
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College Hospital, Tokorozawa, Japan
| | - Kenichi Furuya
- Department of Obstetrics and Gynecology, National Defense Medical College Hospital, Tokorozawa, Japan
| |
Collapse
|
87
|
Kim H, Ro SM, Yang JH, Jeong JW, Lee JE, Roh SY, Kim IH. The neutrophil-to-lymphocyte ratio prechemotherapy and postchemotherapy as a prognostic marker in metastatic gastric cancer. Korean J Intern Med 2018; 33:990-999. [PMID: 29722249 PMCID: PMC6129621 DOI: 10.3904/kjim.2016.293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 01/18/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND/AIMS Markers of inflammation have been associated with outcomes in various cancers. The purpose of this study was to evaluate whether systemic inf lammatory markers and their f luctuations can predict survival and chemotherapy response in patients with metastatic gastric cancer (mGC). METHODS We retrospectively reviewed the records of 502 patients who received first-line palliative chemotherapy for mGC between 2007 and 2013. The neutrophil-to-lymphocyte ratio (NLR) and modified Glasgow prognostic score (mGPS) were assessed before and after chemotherapy to evaluate their association with survival. The NLR values were categorized into two groups based on a cut-off value of 3; mGPS values were classified as high versus low. RESULTS High prechemotherapy NLR was significantly associated with poor overall survival on univariate analysis (p = 0.002). On multivariate analysis, high prechemotherapy NLR (hazard ratio, 1.43; p < 0.001) was an independent prognostic factor for poor overall survival. However, the prechemotherapy mGPS was not significantly associated with survival. Continuously high NLR or a shift to high NLR postchemotherapy was associated with poor chemotherapy response as well as survival, while NLR reduction was associated with a good response (linear by linear association, p < 0.001) and a favorable prognosis. CONCLUSION Prechemotherapy NLR can be used as a prognostic factor in mGC, while the postchemotherapy NLR value may predict the chemotherapeutic response and prognosis. In contrast, mGPS has limited prognostic utility in mGC.
Collapse
Affiliation(s)
- Hyunho Kim
- Department of Internal Medicine, Korea Military Academy Hospital, Seoul, Korea
| | - Sang Mi Ro
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ji Hyun Yang
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Joon Won Jeong
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ji Eun Lee
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sang Young Roh
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
- Correspondence to In-Ho Kim, M.D. Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-6046 Fax: +82-2-599-3589 E-mail:
| |
Collapse
|
88
|
Armato SG, Nowak AK. Revised Modified Response Evaluation Criteria in Solid Tumors for Assessment of Response in Malignant Pleural Mesothelioma (Version 1.1). J Thorac Oncol 2018; 13:1012-1021. [DOI: 10.1016/j.jtho.2018.04.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/20/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022]
|
89
|
Jo J, Kim SH, Kim YJ, Lee J, Kim M, Keam B, Kim TM, Kim DW, Heo DS, Chung JH, Jeon YK, Lee JS. Efficacy of Pemetrexed-based Chemotherapy in Comparison to Non-Pemetrexed-based Chemotherapy in Advanced, ALK+ Non-Small Cell Lung Cancer. Yonsei Med J 2018; 59:202-210. [PMID: 29436187 PMCID: PMC5823821 DOI: 10.3349/ymj.2018.59.2.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/26/2017] [Accepted: 12/31/2017] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Previous retrospective studies suggest that anaplastic lymphoma kinase (ALK) mutation-positive (ALK+) non-small cell lung cancer (NSCLC) patients are sensitive to pemetrexed. To determine its efficacy, we retrospectively evaluated clinical outcomes of pemetrexed-based chemotherapy in patients with ALK+ NSCLC. MATERIALS AND METHODS We identified 126 patients with advanced, ALK+ NSCLC who received first-line cytotoxic chemotherapy. We compared response, progression-free survival (PFS), and overall survival (OS) rates according to chemotherapy regimens. Furthermore, we evaluated intracranial time to tumor progression (TTP) and proportion of ALK+ cells as prognostic factors. RESULTS Forty-eight patients received pemetrexed-based chemotherapy, while 78 received other regimens as first-line treatment. The pemetrexed-based chemotherapy group showed superior overall response (44.7% vs. 14.3%, p<0.001) and disease control (85.1% vs. 62.3%, p=0.008) rates. The pemetrexed-based chemotherapy group also exhibited longer PFS (6.6 months vs. 3.8 months, p<0.001); OS rates were not significantly different. The lack of exposure to second-generation ALK inhibitors and intracranial metastasis on initial diagnosis were independent negative prognostic factors of OS. Intracranial TTP was similar between the treatment groups (32.7 months vs. 35.7 months, p=0.733). Patients who harbored a greater number of ALK+ tumor cells (≥70%) showed prolonged OS on univariate analysis (not reached vs. 44.8 months, p=0.041), but not on multivariate analysis (hazard ratio: 0.19, 95% confidence interval: 0.03-1.42; p=0.106). CONCLUSION Pemetrexed-based regimens may prolong PFS in patients with ALK+ NSCLC as a first-line treatment, but are not associated with prolonged OS. Exposure to second-generation ALK inhibitors may improve OS rates in patients with ALK+ NSCLC.
Collapse
Affiliation(s)
- Jaemin Jo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Se Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Juhyun Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Jong Seok Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
90
|
Valpione S, Gremel G, Mundra P, Middlehurst P, Galvani E, Girotti MR, Lee RJ, Garner G, Dhomen N, Lorigan PC, Marais R. Plasma total cell-free DNA (cfDNA) is a surrogate biomarker for tumour burden and a prognostic biomarker for survival in metastatic melanoma patients. Eur J Cancer 2018; 88:1-9. [PMID: 29175734 PMCID: PMC5769519 DOI: 10.1016/j.ejca.2017.10.029] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/26/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Tumour burden is a prognostic biomarker in metastatic melanoma. However, tumour burden is difficult to measure and there are currently no reliable surrogate biomarkers to easily and reliably determine it. The aim of this study was to assess the potential of plasma total cell free DNA as biomarker of tumour burden and prognosis in metastatic melanoma patients. MATERIALS AND METHODS A prospective biomarker cohort study for total plasma circulating cell-free DNA (cfDNA) concentration was performed in 43 metastatic melanoma patients. For 38 patients, paired blood collections and scan assessments were available before treatment and at first response evaluation. Tumour burden was calculated as the sum of volumes from three-dimensional radiological measurements of all metastatic lesions in individual patients. RESULTS Baseline cfDNA concentration correlated with pre-treatment tumour burden (ρ = 0.52, P < 0.001). Baseline cfDNA levels correlated significantly with hazard of death and overall survival, and a cut off value of 89 pg/μl identified two distinct prognostic groups (HR = 2.22 for high cfDNA, P = 0.004). Patients with cfDNA ≥89 pg/μl had shorter OS (10.0 versus 22.7 months, P = 0.009; HR = 2.22 for high cfDNA, P = 0.004) and the significance was maintained when compared with lactic dehydrogenase (LDH) in a multivariate analysis. We also found a correlation between the changes of cfDNA and treatment-related changes in tumour burden (ρ = 0.49, P = 0.002). In addition, the ratio between baseline cfDNA and tumour burden was prognostic (HR = 2.7 for cfDNA/tumour volume ≥8 pg/(μl*cm3), P = 0.024). CONCLUSIONS We have demonstrated that cfDNA is a surrogate marker of tumour burden in metastatic melanoma patients, and that it is prognostic for overall survival.
Collapse
Affiliation(s)
- S Valpione
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK; Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - G Gremel
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - P Mundra
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - P Middlehurst
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - E Galvani
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - M R Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - R J Lee
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - G Garner
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - N Dhomen
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - P C Lorigan
- Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - R Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK.
| |
Collapse
|
91
|
Comparison of the RECIST 1.0 and RECIST 1.1 in patients treated with targeted agents: a pooled analysis and review. Oncotarget 2017; 7:13680-7. [PMID: 26885610 PMCID: PMC4924670 DOI: 10.18632/oncotarget.7322] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/29/2016] [Indexed: 11/25/2022] Open
Abstract
Patients treated with targeted agents were not included in the data warehouse when the RECIST 1.1 was revised in 2009. We conducted this pooled analysis to investigate the impact of the RECIST 1.1 on the assessment of tumor response in cancer patients treated with targeted agents. We surveyed MEDLINE, EMBASE and PubMed for articles with terms of the RECIST 1.0 or RECIST 1.1. We searched for all the references of relevant articles and reviews using the ‘related articles’ feature in the PubMed. There were six articles in the literature comparing the clinical impacts of the RECIST 1.0 and RECIST 1.1 in patients treated with targeted agents for advanced or metastatic cancer. A total of 322 patients were recruited from the six trials; 217 with non-small cell lung cancer, 23 with thyroid cancer, 20 with gastrointestinal stromal tumor, and 62 with renal cell carcinoma. Because of new lymph node criteria, eight patients (2.5%) had no target lesions when adopting the RECIST 1.1. The number of target lesions by the RECIST 1.1 was significantly lower than that by the RECIST 1.0 (P < 0.001). However, the RECIST 1.1 showed high concordance with the RECIST 1.0 in the assessment of best tumor responses (k = 0.908). Seventeen patients (5.6%) showed discrepancy in the best tumor response between the RECIST 1.0 and RECIST 1.1. This pooled study demonstrates that the RECIST 1.1 shows the highly concordant response assessment with the RECIST 1.0 in patients treated with targeted agents.
Collapse
|
92
|
Mine S, Watanabe M, Imamura Y, Okamura A, Kurogochi T, Sano T. Clinical Significance of the Pre-therapeutic Nodal Size in Patients Undergoing Neo-Adjuvant Treatment Followed by Esophagectomy for Esophageal Squamous Cell Carcinoma. World J Surg 2017; 41:184-190. [PMID: 27468743 DOI: 10.1007/s00268-016-3675-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The clinical significance of pre-therapeutic nodal size in patients with esophageal squamous cell carcinoma (ESCC) is not clear. We investigated whether nodal size was correlated with survival in patients undergoing neo-adjuvant treatment followed by esophagectomy for ESCC. METHODS In 2009-2013, 222 patients who underwent neo-adjuvant treatment followed by esophagectomy for ESCC were enrolled in this retrospective study. Nodal size was measured along the short axis of the largest node using pre-therapeutic CT images. Patients were then stratified based on this short axis, and nodal size was correlated with clinicopathological factors and survival. RESULTS Patients with larger nodes were likely to have deeper cT, higher cN status, and poorer survival. Among the clinical factors cT, cN, cM, and nodal size, only cT and nodal size were independent prognostic factors in multivariate analysis [hazard ratio (HR) 2.0, 95 % confidence interval (CI) 1.1-3.5, p = 0.025 and HR 1.5, 95 % CI 1-2.3, p = 0.036, respectively]. In addition, nodal size was significantly associated with hematological recurrence (p = 0.007), but not lymphatic relapse (p = 0.272). CONCLUSIONS The short axis of the largest node before neo-adjuvant treatment in patients with ESCC is a prognostic factor.
Collapse
Affiliation(s)
- Shinji Mine
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan. .,Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akihiko Okamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takanori Kurogochi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| |
Collapse
|
93
|
Kim IH, Lee JE, Yang JH, Jeong JW, Ro S, Lee MA. Clinical significance of changes in systemic inflammatory markers and carcinoembryonic antigen levels in predicting metastatic colorectal cancer prognosis and chemotherapy response. Asia Pac J Clin Oncol 2017; 14:239-246. [PMID: 29044941 DOI: 10.1111/ajco.12784] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
AIM Metastatic colorectal cancer (mCRC) is associated with poor prognosis, and biomarkers are required for predicting survival and chemotherapy response. This study aimed to evaluate the significance of changes in systemic inflammatory markers and carcinoembryonic antigen (CEA) levels in predicting mCRC prognosis and chemotherapy response. METHODS In this retrospective study, 503 patients who received first-line palliative chemotherapy for mCRC between 2008 and 2014 at a tertiary hospital in Korea were evaluated. Changes in neutrophil-to-lymphocyte ratio (NLR) and modified Glasgow prognostic score (mGPS) were divided into low-to-low, high-to-low, low-to-high and high-to-high groups. The CEA response was defined as CEA-complete response (CEA normalization), CEA-partial response (≥50% decrease in CEA levels), CEA-progressive disease (≥50% increase in CEA levels) and CEA-stable disease. Overall survival (OS) and progression-free survival (PFS) were evaluated according to NLR, mGPS and CEA levels. RESULTS High prechemotherapy NLR, mGPS and CEA levels independently predicted poor survival and chemotherapy response. Continuously high NLR or change to high NLR was also associated with poor OS and PFS; however, continuously low NLR or reduced NLR showed good prognosis. CEA response was also an independent prognostic marker for OS and PFS. High NLR and mGPS were correlated with elevated CEA levels. CONCLUSION Inflammatory marker levels were significantly associated with CEA levels. The prechemotherapy levels of systemic inflammatory markers and CEA were associated with OS or PFS. The change patterns in NLR and CEA levels can be utilized as prognostic and predictive markers for chemotherapy response.
Collapse
Affiliation(s)
- In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ji Eun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ji Hyun Yang
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Joon Won Jeong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Sangmi Ro
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Myung Ah Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.,Cancer Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
94
|
Utility of [ 18F]FSPG PET to Image Hepatocellular Carcinoma: First Clinical Evaluation in a US Population. Mol Imaging Biol 2017; 18:924-934. [PMID: 27677886 DOI: 10.1007/s11307-016-1007-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Non-invasive imaging is central to hepatocellular carcinoma (HCC) diagnosis; however, conventional modalities are limited by smaller tumors and other chronic diseases that are often present in patients with HCC, such as cirrhosis. This pilot study evaluated the feasibility of (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) positron emission tomography (PET)/X-ray computed tomography (CT) to image HCC. [18F]FSPG PET/CT was compared to standard-of-care (SOC) magnetic resonance imaging (MRI) and CT, and [11C]acetate PET/CT, commonly used in this setting. We report the largest cohort of HCC patients imaged to date with [18F]FSPG PET/CT and present the first comparison to [11C]acetate PET/CT and SOC imaging. This study represents the first in a US HCC population, which is distinguished by different underlying comorbidities than non-US populations. PROCEDURES xC- transporter RNA and protein levels were evaluated in HCC and matched liver samples from The Cancer Genome Atlas (n = 16) and a tissue microarray (n = 83). Eleven HCC patients who underwent prior MRI or CT scans were imaged by [18F]FSPG PET/CT, with seven patients also imaged with [11C]acetate PET/CT. RESULTS xC- transporter RNA and protein levels were elevated in HCC samples compared to background liver. Over 50 % of low-grade HCCs and ~70 % of high-grade tumors exceeded background liver protein expression. [18F]FSPG PET/CT demonstrated a detection rate of 75 %. [18F]FSPG PET/CT also identified an HCC devoid of typical MRI enhancement pattern. Patients scanned with [18F]FSPG and [11C]acetate PET/CT exhibited a 90 and 70 % detection rate, respectively. In dually positive tumors, [18F]FSPG accumulation consistently resulted in significantly greater tumor-to-liver background ratios compared with [11C]acetate PET/CT. CONCLUSIONS [18F]FSPG PET/CT is a promising modality for HCC imaging, and larger studies are warranted to examine [18F]FSPG PET/CT impact on diagnosis and management of HCC. [18F]FSPG PET/CT may also be useful for phenotyping HCC tumor metabolism as part of precision cancer medicine.
Collapse
|
95
|
Reduced dose CT with model-based iterative reconstruction compared to standard dose CT of the chest, abdomen, and pelvis in oncology patients: intra-individual comparison study on image quality and lesion conspicuity. Abdom Radiol (NY) 2017; 42:2279-2288. [PMID: 28417170 DOI: 10.1007/s00261-017-1140-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To compare image quality and lesion conspicuity of reduced dose (RD) CT with model-based iterative reconstruction (MBIR) compared to standard dose (SD) CT in patients undergoing oncological follow-up imaging. METHODS Forty-four cancer patients who had a staging SD CT within 12 months were prospectively included to undergo a weight-based RD CT with MBIR. Radiation dose was recorded and tissue attenuation and image noise of four tissue types were measured. Reproducibility of target lesion size measurements of up to 5 target lesions per patient were analyzed. Subjective image quality was evaluated for three readers independently utilizing 4- or 5-point Likert scales. RESULTS Median radiation dose reduction was 46% using RD CT (P < 0.01). Median image noise across all measured tissue types was lower (P < 0.01) in RD CT. Subjective image quality for RD CT was higher (P < 0.01) in regard to image noise and overall image quality; however, there was no statistically significant difference regarding image sharpness (P = 0.59). There were subjectively more artifacts on RD CT (P < 0.01). Lesion conspicuity was subjectively better in RD CT (P < 0.01). Repeated target lesion size measurements were highly reproducible both on SD CT (ICC = 0.987) and RD CT (ICC = 0.97). CONCLUSIONS RD CT imaging with MBIR provides diagnostic imaging quality and comparable lesion conspicuity on follow-up exams while allowing dose reduction by a median of 46% compared to SD CT imaging.
Collapse
|
96
|
Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, Burchill S, Boubaker A, McHugh K, Nuchtern JG, London WB, Seibel NL, Lindwasser OW, Maris JM, Brock P, Schleiermacher G, Ladenstein R, Matthay KK, Valteau-Couanet D. Revisions to the International Neuroblastoma Response Criteria: A Consensus Statement From the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol 2017; 35:2580-2587. [PMID: 28471719 PMCID: PMC5676955 DOI: 10.1200/jco.2016.72.0177] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose More than two decades ago, an international working group established the International Neuroblastoma Response Criteria (INRC) to assess treatment response in children with neuroblastoma. However, this system requires modification to incorporate modern imaging techniques and new methods for quantifying bone marrow disease that were not previously widely available. The National Cancer Institute sponsored a clinical trials planning meeting in 2012 to update and refine response criteria for patients with neuroblastoma. Methods Multidisciplinary investigators from 13 countries reviewed data from published trials performed through cooperative groups, consortia, and single institutions. Data from both prospective and retrospective trials were used to refine the INRC. Monthly international conference calls were held from 2011 to 2015, and consensus was reached through review by working group leadership and the National Cancer Institute Clinical Trials Planning Meeting leadership council. Results Overall response in the revised INRC will integrate tumor response in the primary tumor, soft tissue and bone metastases, and bone marrow. Primary and metastatic soft tissue sites will be assessed using Response Evaluation Criteria in Solid Tumors (RECIST) and iodine-123 (123I) -metaiodobenzylguanidine (MIBG) scans or [18F]fluorodeoxyglucose-positron emission tomography scans if the tumor is MIBG nonavid. 123I-MIBG scans, or [18F]fluorodeoxyglucose-positron emission tomography scans for MIBG-nonavid disease, replace technetium-99m diphosphonate bone scintigraphy for osteomedullary metastasis assessment. Bone marrow will be assessed by histology or immunohistochemistry and cytology or immunocytology. Bone marrow with ≤ 5% tumor involvement will be classified as minimal disease. Urinary catecholamine levels will not be included in response assessment. Overall response will be defined as complete response, partial response, minor response, stable disease, or progressive disease. Conclusion These revised criteria will provide a uniform assessment of disease response, improve the interpretability of clinical trial results, and facilitate collaborative trial designs.
Collapse
Affiliation(s)
- Julie R. Park
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Rochelle Bagatell
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Susan L. Cohn
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Andrew D. Pearson
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Judith G. Villablanca
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Frank Berthold
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Susan Burchill
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Ariane Boubaker
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Kieran McHugh
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Jed G. Nuchtern
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Wendy B. London
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Nita L. Seibel
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - O. Wolf Lindwasser
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - John M. Maris
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Penelope Brock
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Gudrun Schleiermacher
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Ruth Ladenstein
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Katherine K. Matthay
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| | - Dominique Valteau-Couanet
- Julie R. Park, Seattle Children’s Hospital and University of Washington School of Medicine, Seattle, WA; Rochelle Bagatell and John M. Maris, Children’s Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA; Susan L. Cohn, University of Chicago, Chicago, IL; Andrew D. Pearson, Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey; Susan Burchill, Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom; Judith G. Villablanca, Children’s Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles; Katherine K. Matthay, University of California San Francisco School of Medicine, San Francisco, CA; Frank Berthold, Children’s Hospital and University of Cologne, Köln, Germany; Ariane Boubaker, Institute of Radiology, Clinique de La Source, Lausanne, Switzerland; Jed G. Nuchtern, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX; Wendy B. London, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Harvard Medical School, Boston, MA; Nita L. Seibel and O. Wolf Lindwasser, National Cancer Institute, Bethesda, MD; Gudrun Schleiermacher, Institut Curie, Paris; Dominique Valteau-Couanet, Gustave Roussy, Villejuif, France; and Ruth Ladenstein, Children’s Cancer Research Institute, St Anna Children’s Hospital, Vienna, Austria
| |
Collapse
|
97
|
Jo J, Kwon HW, Park S, Oh DY, Cheon GJ, Bang YJ. Prospective Evaluation of the Clinical Implications of the Tumor Metabolism and Chemotherapy-Related Changes in Advanced Biliary Tract Cancer. J Nucl Med 2017; 58:1255-1261. [PMID: 28254865 DOI: 10.2967/jnumed.116.186239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/04/2017] [Indexed: 01/21/2023] Open
Abstract
Tumor metabolism measured by 18F-FDG PET has a diagnostic and prognostic role in several cancers. The clinical implication of tumor metabolism in biliary tract cancer (BTC) has not been studied well. Therefore, we evaluated the prognostic value of tumor metabolism and chemotherapy-related changes in advanced BTC patients. Methods: We prospectively enrolled advanced BTC patients before the initiation of palliative chemotherapy. Using 18F-FDG PET, we assessed the baseline SUVmax and monitored the changes in SUVmax during chemotherapy. We analyzed the associations between SUVmax, and clinicopathologic factors and clinical outcomes. Results: Seventy-five patients were enrolled. All patients received gemcitabine/cisplatin as first-line chemotherapy. Primary tumor site, histologic differentiation, molecular characteristics, laboratory findings, and disease extent were associated with the metabolic characteristics. The high-metabolism group showed worse survival outcome (hazard ratio [HR] = 4.09, P = 0.001 for progression-free survival; HR = 2.61, P = 0.019 for overall survival [OS]) than the low-metabolism group. The lesser reduction of SUVmax was also associated with worse outcome (HR = 3.35, P = 0.002 for progression-free survival; HR = 1.96, P = 0.082 for OS). When both baseline tumor metabolism and its chemotherapy-related changes were considered, patients with a low metabolism and more reduction in metabolism obtained the best OS (20.7 vs. 6.2 mo, P = 0.013). Conclusion: Tumor metabolic activity and the chemotherapy-related changes in the metabolism are associated with prognosis in advanced BTC patients.
Collapse
Affiliation(s)
- Jaemin Jo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Woo Kwon
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea; and
| | - Seongyeol Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea; and
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
98
|
Ribrag V. Toward common response evaluation criteria for solid tumors and lymphomas: RECIL and RECIST? Ann Oncol 2017; 28:1409-1411. [PMID: 28472399 DOI: 10.1093/annonc/mdx209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- V Ribrag
- Drug Development Department (DITEP), Gustave Roussy Institut, Villejuif, France
| |
Collapse
|
99
|
Younes A, Hilden P, Coiffier B, Hagenbeek A, Salles G, Wilson W, Seymour JF, Kelly K, Gribben J, Pfreunschuh M, Morschhauser F, Schoder H, Zelenetz AD, Rademaker J, Advani R, Valente N, Fortpied C, Witzig TE, Sehn LH, Engert A, Fisher RI, Zinzani PL, Federico M, Hutchings M, Bollard C, Trneny M, Elsayed YA, Tobinai K, Abramson JS, Fowler N, Goy A, Smith M, Ansell S, Kuruvilla J, Dreyling M, Thieblemont C, Little RF, Aurer I, Van Oers MHJ, Takeshita K, Gopal A, Rule S, de Vos S, Kloos I, Kaminski MS, Meignan M, Schwartz LH, Leonard JP, Schuster SJ, Seshan VE. International Working Group consensus response evaluation criteria in lymphoma (RECIL 2017). Ann Oncol 2017; 28:1436-1447. [PMID: 28379322 PMCID: PMC5834038 DOI: 10.1093/annonc/mdx097] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Indexed: 12/20/2022] Open
Abstract
In recent years, the number of approved and investigational agents that can be safely administered for the treatment of lymphoma patients for a prolonged period of time has substantially increased. Many of these novel agents are evaluated in early-phase clinical trials in patients with a wide range of malignancies, including solid tumors and lymphoma. Furthermore, with the advances in genome sequencing, new "basket" clinical trial designs have emerged that select patients based on the presence of specific genetic alterations across different types of solid tumors and lymphoma. The standard response criteria currently in use for lymphoma are the Lugano Criteria which are based on [18F]2-fluoro-2-deoxy-D-glucose positron emission tomography or bidimensional tumor measurements on computerized tomography scans. These differ from the RECIST criteria used in solid tumors, which use unidimensional measurements. The RECIL group hypothesized that single-dimension measurement could be used to assess response to therapy in lymphoma patients, producing results similar to the standard criteria. We tested this hypothesis by analyzing 47 828 imaging measurements from 2983 individual adult and pediatric lymphoma patients enrolled on 10 multicenter clinical trials and developed new lymphoma response criteria (RECIL 2017). We demonstrate that assessment of tumor burden in lymphoma clinical trials can use the sum of longest diameters of a maximum of three target lesions. Furthermore, we introduced a new provisional category of a minor response. We also clarified response assessment in patients receiving novel immune therapy and targeted agents that generate unique imaging situations.
Collapse
Affiliation(s)
| | - P. Hilden
- Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - B. Coiffier
- Hematology, Université Lyon-1, Lyon-Sud Charles Mérieux, Lyon, France
| | - A. Hagenbeek
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - G. Salles
- Hematology, Université Lyon-1, Lyon-Sud Charles Mérieux, Lyon, France
| | - W. Wilson
- Lymphoid Malignancies Branch, National Cancer Institute, Bethesda, USA
| | - J. F. Seymour
- Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - K. Kelly
- Pediatrics Department, Roswell-Park Cancer Institute, Buffalo, USA
| | - J. Gribben
- Department of Haemato-Oncology, Barts Cancer Institute, London, UK
| | - M. Pfreunschuh
- Department of Internal Medicine, Universität des Saarlandes, Homburg, Germany
| | - F. Morschhauser
- Department of Hematology, Université de Lille 2, Lille, France
| | - H. Schoder
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York
| | | | - J. Rademaker
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York
| | - R. Advani
- Department of Oncology, Stanford University, Stanford
| | | | | | | | - L. H. Sehn
- British Columbia Cancer Agency, Vancouver, Canada
| | - A. Engert
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | | | - P.-L. Zinzani
- Department of Hematology, University of Bologna, Bologna
| | - M. Federico
- Department of Diagnostic Medicine, University of Modena, Modena, Italy
| | - M. Hutchings
- Department of Hematology, University of Copenhagen, Denmark
| | - C. Bollard
- Children’s National Health System, Washington, USA
| | - M. Trneny
- Lymphoma and Stem Cell Transplantation Program, Charles University, Prague, Czech Republic
| | | | - K. Tobinai
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - J. S. Abramson
- Massachusetts General Hospital, Center for Lymphoma, Boston
| | - N. Fowler
- U.T. M.D.Anderson Cancer Center, Houston
| | - A. Goy
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack
| | - M. Smith
- Cleveland Clinic, Cleveland, USA
| | | | - J. Kuruvilla
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - M. Dreyling
- Medicine Clinic III, Ludwig Maximilian University, Munich, Germany
| | | | - R. F. Little
- Divisions of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - I. Aurer
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | | | - A. Gopal
- Fred Hutchinson Cancer Research Center, Seattle, USA
| | - S. Rule
- Haematology Department, Plymouth University, UK
| | | | - I. Kloos
- Servier, Neuilly sur Seine, France
| | - M. S. Kaminski
- University of Michigan Comprehensive Cancer Center, Ann Arbor, USA
| | - M. Meignan
- Nuclear Medicine, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - L. H. Schwartz
- Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York
| | - J. P. Leonard
- Weill Cornell Medicine and and New York Presbyterian Hospital, New York
| | - S. J. Schuster
- University of Pennsylvania School of Medicine, Philadelphia, USA
| | - V. E. Seshan
- Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| |
Collapse
|
100
|
Agarwal SK, Akali NR, Sarin D. Prospective analysis of 231 elective neck dissections in oral squamous cell carcinoma with node negative neck-To decide the extent of neck dissection. Auris Nasus Larynx 2017; 45:156-161. [PMID: 28662778 DOI: 10.1016/j.anl.2017.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/27/2017] [Accepted: 05/29/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate the incidence of level IIB and IV lymph node metastases in patients of oral carcinoma with N0 necks. This study will help to decide the extent and need of routine comprehensive removal of these specific lymph node groups in selective neck dissection. METHODS Prospective analytical study of 231 patients of oral carcinoma with N0 necks undergoing neck dissections were prospectively analyzed. The incidence of metastases at level IIB and IV were then observed. RESULTS 71 (30.73%) out of 231 cases had microscopic metastatic lymphadenopathy. Lymph node metastases from oral cancers were seen predominantly at levels IB and IIA. Metastases at levels IIB and IV were very rare (0.86% and 0%, respectively). Metastases at level IIB was associated with metastases at the level IIA in both cases (100%) and with level IB in 1 case (50%). 27 (11.68%) out of 231 cases had positive IIA nodes and conversely, only 7.4% (2/27) of all level IIA metastases had positive nodes at level IIB. CONCLUSION Selective neck dissection from I-III may be adequate for majority of patients with oral cancers. Level IIB and IV need not be dissected in N0 patients.
Collapse
Affiliation(s)
- Sangeet Kumar Agarwal
- Department of ENT and Head, Neck Surgery, Sir Ganga Ram Hospital, New Delhi-110060, India.
| | | | - Deepak Sarin
- Department of Head and Neck Oncosurgery Medanta Cancer Institute, Medanta-The Medicity Gurgaon, India
| |
Collapse
|